You are on page 1of 11

Published OnlineFirst May 29, 2019; DOI: 10.1158/1078-0432.

CCR-18-3720

Translational Cancer Mechanisms and Therapy Clinical


Cancer
Research
Molecular Classification of Epithelial Ovarian
Cancer Based on Methylation Profiling: Evidence
for Survival Heterogeneity
Clara Bodelon1, J. Keith Killian2, Joshua N. Sampson1, William F. Anderson1,
Rayna Matsuno3,4, Louise A. Brinton1, Jolanta Lissowska5, Michael S. Anglesio6,7,
David D.L. Bowtell8,9, Jennifer A. Doherty10, Susan J. Ramus8,11, Aline Talhouk6,
Mark E. Sherman1,12, and Nicolas Wentzensen1

Abstract
Purpose: Ovarian cancer is a heterogeneous disease that can expression data for 39 genes were available for 61 high-
be divided into multiple subtypes with variable etiology, grade serous carcinomas (HGSC).
pathogenesis, and prognosis. We analyzed DNA methylation Results: Consensus nonnegative matrix factorization clus-
profiling data to identify biologic subgroups of ovarian cancer tering of the 1,000 most variable CpG sites showed four major
and study their relationship with histologic subtypes, copy clusters among all epithelial ovarian cancers. We observed
number variation, RNA expression data, and outcomes. statistically significant differences in survival (log-rank test, P
Experimental Design: A total of 162 paraffin-embedded ¼ 9.1  107) and genomic instability across these clusters.
ovarian epithelial tumor tissues, including the five major Within HGSC, clustering showed three subgroups with sur-
epithelial ovarian tumor subtypes (high- and low-grade vival differences (log-rank test, P ¼ 0.002). Comparing models
serous, endometrioid, mucinous, and clear cell) and with and without methylation subgroups in addition to pre-
tumors of low malignant potential were selected from two viously identified gene expression subtypes suggested that the
different sources: The Polish Ovarian Cancer study, and methylation subgroups added significant survival information
the Surveillance, Epidemiology, and End Results Residual (P ¼ 0.007).
Tissue Repository (SEER RTR). Analyses were restricted to Conclusions: DNA methylation profiling of ovarian cancer
Caucasian women. Methylation profiling was conducted identified novel molecular subgroups that had significant
using the Illumina 450K methylation array. For 45 tumors survival difference and provided insights into the molecular
array copy number data were available. NanoString gene underpinnings of ovarian cancer.

Introduction effective screening strategy, and most cancers are detected at


advanced stages limiting curative therapies. Ovarian cancer is a
Ovarian cancer is the fifth leading cause of cancer-related death
histologically heterogeneous set of diseases with approximately
among women in the United States (1). Currently, there is no
90% of cases being of epithelial origin. The major histotypes of
malignant epithelial ovarian cancers (EOC) include serous (high-
and low-grade), endometrioid, clear cell, and mucinous (2).
1
Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland. Serous tumors account for 60%–70% of EOC, endometrioid
2
Center for Cancer Research (CCR), NCI, NIH, Bethesda, Maryland. 3Foundation
approximately 15%, clear cells for 5%, and mucinous for approx-
Medicine Inc., Cambridge, Massachusetts. 4University of California, San Diego,
California. 5M. Sklodowska-Curie Memorial Cancer Center and Institute of
imately 10% (3–7), although these rates vary globally and have
Oncology, Warsaw, Poland. 6Department of Pathology and Laboratory Medi- changed over time (8). Of all these, high-grade serous carcinomas
cine, University of British Columbia, Vancouver, Canada. 7Department of Obstet- (HGSC) represent the most common histotype and are associated
rics and Gynaecology, University of British Columbia, Vancouver, Canada. with poor prognosis (3, 5). Ovarian tumors of low malignant
8
The Kinghorn Cancer Center, Garvan Institute of Medical Research, Sydney, potential (LMP) or borderline tumors account for 15% of all
Australia. 9Peter MacCallum Cancer Center, Melbourne, Australia. 10Department
epithelial neoplasms, and are generally considered apart from
of Population Health Sciences, Huntsman Cancer Institute, University of Utah,
Salt Lake City, Utah. 11School of Women's and Children's Health, University of
extraovarian metastasis because of differences in proposed path-
New South Wales, Sydney, Australia. 12Mayo Clinic, Jacksonville, Florida. ogenesis and highly favorable clinical outcomes in the absence of
invasive implants (9, 10).
Note: Supplementary data for this article are available at Clinical Cancer
Research Online (http://clincancerres.aacrjournals.org/). Several studies have shown that the major histologic
subtypes of EOC have different risk factors (11), genetic
Corresponding Author: Clara Bodelon, NCI, 9609 Medical Center Drive,
susceptibilities (12–14), and clinical response to therapy (15, 16).
Bethesda, MD 20892. Phone: 240-276-7327; Fax: 240-276-7838; E-mail:
clara.bodelon@nih.gov These histotypes also differ at the molecular level. Serous tumors,
mostly high grade, were the only histologic subtypes included
Clin Cancer Res 2019;XX:XX–XX
in The Cancer Genome Atlas (TCGA; ref. 17). Results from this
doi: 10.1158/1078-0432.CCR-18-3720 large-scale genomics analysis showed that these tumors have
2019 American Association for Cancer Research. near ubiquitous TP53 mutations (96% of tumors) and a high

www.aacrjournals.org OF1

Downloaded from clincancerres.aacrjournals.org on July 29, 2019. © 2019 American Association for Cancer
Research.
Published OnlineFirst May 29, 2019; DOI: 10.1158/1078-0432.CCR-18-3720

Bodelon et al.

Materials and Methods


Translational Relevance Study population
Ovarian cancer is a heterogeneous disease with several Subjects included in this analysis were selected from two
histologic subtypes with different etiology, pathogenesis, and studies, the Polish Ovarian Cancer Study (POCS; ref. 24) and
prognosis. Identifying key molecular characteristics that differ the NCI's Surveillance Epidemiology and End Results (SEER)
across these subtypes could refine tumor classification beyond Residual Tissue Repository (RTR) study (25). Briefly, POCS was
histology alone and inform about potential treatments and a population-based case–control study that included eligible
etiology, and help in the development of new approaches for cases diagnosed between June 1, 2001 and December 30, 2003,
prevention and early detection. In this analysis of epithelial residents in Warsaw or Lodz, Poland, and were aged 20–74 at
ovarian tumor DNA methylation, that included high- and low- the time of diagnosis. Incident ovarian cancer cases were
grade serous, endometrioid, mucinous, and clear cell, and ascertained through a rapid identification system coordinated
tumors of low malignant potential, we showed that there are by five participating Polish hospitals, which cover approxi-
four methylation-defined subgroups of epithelial ovarian can- mately 85% of all cases diagnosed in the two cities (24). In
cer with survival differences, similar to histologic subtypes. addition, cancer registries in Warsaw and Lodz were used to
Within high-grade serous carcinomas, we found methylation identify cases missed by the rapid identification system. Med-
subgroups that added significant survival information to pre- ical records were reviewed for pathology, treatment, and out-
viously identified gene expression subtypes. comes information. An experienced gynecologic pathologist
(M. Sherman) reviewed the hematoxylin and eosin (H&E)
slides to determine the pathologic diagnosis used for this
analysis. Formalin-fixed paraffin-embedded (FFPE) tissue
level of copy number alterations. TCGA analyses confirmed four blocks were available for a subset of ovarian cancer cases
transcriptional subtypes that were previously described for (55%). Between two and four 1-mm cores were obtained
HGSC (18). Large-scale integrated characterization of the other from these patients with cancer. The POCS was reviewed
subtypes is mostly lacking. Mutations in BRAF, KRAS, PTEN, and and approved by the institutional review boards (IRB) of the
B-catenin have been shown in endometrioid, clear cell, low-grade U.S. NCI, the M. Sklodowska Curie Institute of Oncology
serous, mucinous EOC (19). and Cancer Center in Warsaw, and the Institute of Occupa-
There is increasing evidence that the different histotypes of tional Medicine in Lodz. This study is covered by Single
EOC have different cells of origin, including that many HGSCs Project Assurances (SPA) in Warsaw (S-009741-04) and
originate from secretory cells in the fimbria of the fallopian Lodz (S-017191-01). All participants provided written
tubes, while endometrioid and clear cell carcinomas appear to informed consent for use of clinical data and archival tissue
arise from endometriosis, which itself may represent explanted specimens. FFPE tumor blocks from 32 patients that did not
menstrual endometrium (20). The origin of mucinous tumors receive chemotherapy were included in the current analysis
is less well understood. It has been suggested that a possible (Supplementary Table S1).
origin is in epithelial nests located in the tubo-peritoneal The SEER RTR study included FFPE tissue blocks from pri-
junction (19). Of note, historically, many tumors classified as mary tumors in the Hawaii and Iowa Tumor Registries (25).
ovarian mucinous carcinomas represent metastases to the ova- Ovarian cancer cases in the Hawaii Tumor Registry were diag-
ry, often from an occult gastrointestinal primary (21). Large- nosed from 1983 through 2004, which represented 38% of all
scale identification of molecular features in tumors may help to ovarian tumors in the Hawaii catchment area during that
identify key molecular characteristics that differ across histo- period. Ovarian cancer cases in the Iowa Tumor Registry were
types, refine tumor classification beyond histology alone, and diagnosed from 1987 through 2003, representing 4% of all
inform about potential treatments and etiology, which could ovarian tumors in the Iowa catchment area during that period.
lead to development of new approaches for prevention and Tumors were restricted to those from European ancestry wom-
early detection of EOC. en. Other demographic characteristics available, in addition to
DNA methylation is a key epigenetic regulator of gene expres- race, were age and year at diagnosis and year of death. Tumor
sion across a variety of cellular processes (22). There is growing characteristics included histology and stage from the American
interest in the role of DNA methylation in carcinogenesis and in Joint Committee on Cancer, and grade (25). Because SEER's
defining molecular subtypes that can assist in elucidating the RTR data were anonymized, the NIH's Office of Human Sub-
etiology, clinical characteristics, and outcomes of EOC. Genome- jects Research (OHSR) designated the project as exempt from
wide methylation studies have focused mostly on a single sub- IRB approval (OHSR #4081); nonetheless, IRB approvals were
type (14, 17, 23) with different studies using different technol- provided at the Universities of Hawaii and Iowa. We retrieved
ogies, experimental conditions, and analytical techniques, which the available FFPE tissue blocks for primary invasive ovarian
makes it difficult to compare results across the different histo- carcinomas in the Hawaii and Iowa Discard Tumor Registries.
types. Therefore, a more comprehensive study involving epigen- Tumor blocks from 144 Caucasian patients from the SEER RTRs
ome-wide assessment of DNA methylation is needed to better [60 (42%) from the Hawaii Tumor Registry and 84 (58%) from
understand methylation status across different EOCs. the Iowa Tumor Registry] were included in this analysis (Sup-
In two population-based studies, we analyzed DNA methyla- plementary Table S1). H&E slides from all tumors went through
tion in the major histologies of ovarian cancer to define subtypes expert pathology reviewed by MES and the reviewed diagnosis
of EOC and compare these subtypes with the degree of chromo- was used in this analysis. Tumors included in the analysis did
somal copy number alterations. Furthermore, we compared DNA not receive neoadjuvant chemotherapy prior to their collection
methylation within HGSC to previously identified subgroups based on review of pathology reports. Mortality data were
based on gene expression. coded by year.

OF2 Clin Cancer Res; 2019 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on July 29, 2019. © 2019 American Association for Cancer
Research.
Published OnlineFirst May 29, 2019; DOI: 10.1158/1078-0432.CCR-18-3720

Methylation and Ovarian Cancer Survival

Methylation array logarithm ratios (log2 ratios) of the two intensity channels were
DNA extraction and genome-wide DNA methylation assays then computed. For the subject with duplicate runs, the log2 ratios
using the Infinium HumanMethylation450 BeadChips (Illu- of the two runs were averaged for analysis. The log2 ratios were
mina) were performed as described previously (26, 27). Briefly, subjected to the circular binary segmentation (CBS) algo-
FFPE tissue cores were deparaffinized using mineral oil and lysed rithm (32) by means of the Bioconductor package DNAcopy.
in Qiagen ATL buffer/proteinase K. NanoDrop UV absorbance Copy numbers (double deletions, losses, neutral, gains, and
and PicoGreen dsDNA fluorometry (Invitrogen) were used to amplifications) that were significantly altered across all samples
measures DNA levels in the lysates. Fifty microliters of isolated, were determined using the GISTIC 2.02 algorithm using the
purified DNA was sodium bisulfite–treated following the Zymo default parameters (33) in the NIH High-Performance Comput-
Research EZ-96 Methylation Gold kit. Converted DNA was further ing Biowulf cluster computing (http://hpc.nih.gov).
treated with the Illumina FFPE Restoration solution and 250 ng of
restored DNA was then run on the Infinium HumanMethyla-
NanoString assay
tion450 BeadChip according to the standard protocol. Raw data
FFPE tumor cores from the HGSC methylation array set (N ¼
were extracted using the Genome Studio v2011.1.
70) were sent to the international ovarian tumor tissue analysis
Four samples were run in triplicate and one cell line (GIST 28T)
(OTTA) consortium for gene expression assays of 518 genes on the
was included in quadruplicate across the two methylation plates
NanoString platform analysis. Tissues were deparaffinized with
for quality controls. The average correlation across these replicates
xylene and processed with the Qiagen miRNeasy FFPE protocol
was 0.984. A plot showing the intraclass-correlation coefficient
(Qiagen) as per the manufacturer's recommendation, including
against the variance of the beta values is shown in the Supple-
DNase 1 treatment and with an extended proteinase K digest
mentary Fig. S1. Fourteen samples were excluded on the basis of
(55 C digest period extended to 45 minutes). RNA was quanti-
the lack of a bimodal distribution of the beta values, as expected
tated on Nanodrop spectrophotometer. Nine samples failed
for this array. All these excluded samples were from the SEER
quality controls, leaving 61 samples for analyses.
Hawaii Tumor Registry collected prior to 1993. Raw intensity data
Specimens were processed following NanoString gene expres-
were imported into the R programming software using the minfi
sion codeset procedures (NanoString). Briefly, 500 ng of tRNA
Bioconductor package (28). Data were preprocessed for back-
from each sample was mixed with a custom codeset (NanoString)
ground subtraction and controls normalization using the prepro-
and hybridization buffer (NanoString). Specimens were hybrid-
cess Illumina function. Subset-quantile within array normaliza-
ized in a Tetrad 2 Thermal Cycler (Bio-Rad) overnight for 16 or
tion (SWAN) was performed to correct for type I and II probe
20 hours and then processed on the nCounter Prep Station
bias (29). Probes that were cross-reactive or located at single-
(NanoString). Cartridges were then imaged on nCounter Digital
nucleotide polymorphisms were excluded (30). Probes that had a
Analyzer (NanoString). Data was normalized to housekeeping
detection P > 0.01, which indicated that they were indistinguish-
genes (RPL19, ACTB, PGK1, SDHA, and POLR1B) and prepro-
able from the background, were also excluded. Analyses were
cessed using three pooled ovarian cancer specimens, run on the
restricted to probes in the 22 autosomes. A total of 362,498 CpG
same codeset-lot, and using the same protocols, as references in
sites remained for analysis.
accordance with a single sample classification scheme outlined
previously (34).
Array copy number assay
A subset of invasive tumors for which enough DNA was
extracted that was not required for the methylation arrays (N ¼ Statistical analysis
47) were analyzed for chromosomal copy number alterations Analyses were performed using the R software (version 3.1.1)
using a commercial 180 K-feature array comparative genomic and appropriate Biconductor packages.
hybridization (aCGH) assay (Agilent Inc.). We oversampled rare Consensus nonnegative matrix factorization (NMF) clustering
subtypes (clear cell, mucinous, and endometrioid) for this anal- algorithm (35) was used for unsupervised class discovery using
ysis (see Supplementary Tables S1 and S2). Briefly, purified DNA, the 1,000 most variable methylation probes according to the
prior to bisulfite conversion, was labeled with Cy5 and Promega median absolute deviation (MAD). To identify the number of
Male reference DNA was labeled with Cy3 using the Agilent stable clusters (rank) in our dataset, we performed 100 indepen-
Genomic DNA ULS Labeling Kit with subsequent purification of dent NMF runs for ranks between 2 and 7 using the Lee and Seung
DNA on Kreatech genomic purification columns to remove excess method (36). Previous studies have suggested that 30–50 runs are
Cy3/Cy5. Equal amounts of Cy5-tumor DNA and Cy3-reference sufficient to assess cluster stability (37, 38). For each rank, the
DNA were combined and hybridized to Agilent 4  180K CGH algorithm calculates how frequently two samples are grouped
arrays for at least 40 hours. Slides were washed and scanned in the together in repeated clustering runs. The resulting pairwise con-
Agilent Scanner C. Image data are extracted using the Agilent sensus rates (cophenetic correlation coefficient) are used for
Feature Extraction software and visual inspection of results is assessment of cluster stability. To estimate the optimal number
performed using the Nexus 6.0 (Biodiscovery) software. of clusters, we followed the approach proposed by Brunet and
Background correction and normalization of the aCGH raw colleagues (37) by selecting the rank for which the cophenetic
signal intensities were done using the Bioconductor limma pack- correlation coefficient starts to decrease, and therefore the stability
age with the minimum method for background correction and the of the cluster starts to decrease. This approach has been previously
print-tip loess normalization, which takes into account signal used in analyses of ovarian cancer samples (17, 18, 39–41). In
intensity and spatial position on the array (31). Diagnostic plots addition, we performed 2,000 bootstrap runs on resampled data
were used to assure that signal biases were not present in the data to compute confidence intervals (CI) around each cophenetic
after normalization. Two samples were excluded on the basis of coefficient and to assess whether the optimal rank choice was
this analysis, leaving data on 45 samples for analysis. Binary statistically significantly different to other ranks. The choice of

www.aacrjournals.org Clin Cancer Res; 2019 OF3

Downloaded from clincancerres.aacrjournals.org on July 29, 2019. © 2019 American Association for Cancer
Research.
Published OnlineFirst May 29, 2019; DOI: 10.1158/1078-0432.CCR-18-3720

Bodelon et al.

optimal rank was robust to the number of independent NMF runs, Almost all LMP serous tumors (97%) were in C2. C3 was pre-
the number of probes, and the NMF algorithm used. dominantly comprised of clear cell and endometrioid tumors
Overall survival was computed as the number of years between (74%). Finally, C4 was predominantly comprised of HGSC, high-
the year of diagnosis and the year of death from all causes, the date grade endometrioid carcinomas, and mucinous tumors. The
of last follow-up, or 5-year censored survival data. Kaplan–Meier concordance between methylation subgroups and histologic sub-
curves compared overall survival according to subgroups or types was approximately 67% (Supplementary Table S5). The
histotype and a log-rank test was used to assess the survival mean age at diagnosis of women was 64.0 years old in C1,
distributions across the subgroups. For univariate and multivar- 49.6 years old in C2, 55.9 years old in C3, and 59.2 years old
iate (adjusted for age, stage, grade, and study) survival analysis, a in C4 (C1 vs. C2, P ¼ 1.42  108; C1 vs. C3, P ¼ 0.003; C2 vs. C3,
Cox proportional hazards regression model was used, and HRs P ¼ 0.029; C2 vs. C4, P ¼ 0.002; other comparisons were not
and 95% CIs were estimated. Year of diagnosis was not associated statistically significant).
with survival. Kaplan–Meier curves of the methylation-based subtypes
To quantify the level of genomic instability in each sample, we showed significantly different overall survival outcomes
computed the fraction of the genome that was altered. This was for the different subtypes (log-rank test, P ¼ 9.1  107;
defined as the percentage of regions with gains or losses, based on Fig. 1D). The survival pattern of each of the methylation-
the GISTIC results, compared with the covered genome. We called based subtypes resembled that of the major histologic group
this measure the genomic instability index (GII) of the sam- predominantly represented in each methylation-based subtype
ple (42, 43). The Wilcoxon rank-sum test was used to compare (Fig. 1D and E). Compared with C2, the risk of mortality of
the distribution of the GII values across different groups. women in C1 had a 3-fold higher risk of death (HR ¼ 3.17; 95%
NanoString data for 39 genes were used to classify HGSC CI, 1.11–9.02; P ¼ 0.031; Table 1). In unadjusted analysis, the
samples according to previously defined gene expression sub- risk of mortality of women in cluster C1 was 6-fold higher
groups (18) using subtypes scores (44). Briefly, subtype scores compared with women in C2 (HR ¼ 6.29; 95% CI, 2.78–14.23;
were computed using a weighted score of the genes differentially P ¼ 9.99  106; Supplementary Table S6). Similarly, women
expressed in a specific subgroup. Genes upregulated in the sub- in C3 had higher risk of death compared with those in C2 (HR
group were allocated a weight of 1, while genes downregulated in ¼ 3.39; 95% CI, 1.38–8.31; P ¼ 0.008). Women in C3 were also
the subgroup were allocated a weight of 1. The subtype score was at higher risk compared with patients in cluster C2, but it was
computed as the sum of the subtype-specific genes weighted by not statistically significant (Supplementary Table S6). However,
the directionality of the gene (44, 45). Because the gene expression when analyses were adjusted for either histology or other
classification approach we used does not take into account batch clinical characteristics (importantly stage and grade), estimates
variability, we confirmed that the gene expression predicted were greatly reduced and none of the associations remained
clusters were validated against previously identified clusters by significant, suggesting that methylation clusters capture several
correlating cluster centroids. of the histologic or clinical phenotypes.
Finally, we assessed whether DNA methylation subgroups
added survival information to the previously defined gene expres- Genomic instability index across all EOC tumors
sion subgroups. Using a likelihood ratio test, we compared a Copy number was measured in a subset of the EOCs (N ¼ 45;
predictor model with gene expression subgroups, age, and stage, Supplementary Table S2) comprising serous (N ¼ 15), endome-
and stratified the baseline hazard by study to a model that had the trioid (N ¼ 20), clear cell (N ¼ 3), and mucinous (N ¼ 7) tumors
same variables and the methylation subgroups. (copy number information was not available for serous LMP
All statistical tests were two-sided and a P value of less than 0.05 tumors). We computed the fraction of genome that is altered
was considered statistically significant. using the GII for each sample (see Materials and Methods; Fig. 2).
The GII was associated with grade, with lower-grade samples
having lower values of GII and high-grade samples having higher
Results values of GII [mean GII in low-grade tumors: 0.016 (SD ¼ 0.018),
Our analysis included 162 EOC tumors covering the major mean GII in high-grade tumors: 0.050 (0.020); Wilcoxon test P ¼
histotypes of EOC (Supplementary Table S1). The majority of 1.03  105]. Next, we compared the distribution of the GII values
tumors were from the SEER RTR (N ¼ 130, 80%). Principal across the methylation-based groups and histologies. The average
component analysis was performed on the methylation beta GII of methylation cluster C1 was 0.061 (SD ¼ 0.016), of C2 was
values to explore whether methylation clustered according to 0.042 (0.030), of C3 was 0.021 (0.017), and of C4 was 0.044
factors such as study (POCS vs. SEER RTR), study site (Warsaw, (0.021). Compared with the GII values in methylation cluster C3,
Lodz, Hawaii, Iowa), year of diagnosis, age at diagnosis, or batch GII values in the methylation-based subtypes C1 and C4 were
(Supplementary Fig. S2). None of these factors were related to significantly higher (C1 vs. C3: Wilcoxon test P ¼ 7.75  105;
methylation differences. C4 vs. C3: Wilcoxon test P ¼ 0.0037). The relationship between
the GII values and histologies was as expected, with serous tumors
Methylation-based across all EOC tumors having, on average, higher GII values than the other subtypes:
Clustering analysis of the 1,000 most variable methylation mean GII of serous tumors: 0.055 (SD ¼ 0.024), mean GII of
probes across all EOC tumors identified four stable, methyla- endometrioid tumors: 0.025 (0.020); mean GII of mucinous
tion-based subtypes based on the decrease of the cophenetic tumors: 0.040 (0.021); and mean GII of clear cell tumors:
coefficient (C1, C2, C3, and C4; Fig. 1A and B; Supplementary 0.051 (0.009). Compared with the GII values of endometrioid
Table S3). C1 was predominantly comprised of HGSC tumors tumors, GII values in serous and clear cell tumors were signifi-
(88%; Fig. 1C; Supplementary Table S4); C2 was predominantly cantly higher (serous vs. endometrioid: Wilcoxon test P ¼ 8.2 
represented by LMPs (66%) and 27% of invasive serous tumors. 104; clear cell vs. endometrioid: Wilcoxon test P ¼ 0.035).

OF4 Clin Cancer Res; 2019 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on July 29, 2019. © 2019 American Association for Cancer
Research.
Published OnlineFirst May 29, 2019; DOI: 10.1158/1078-0432.CCR-18-3720

Methylation and Ovarian Cancer Survival

A B C
Rank = 2 Rank = 3 Rank = 4
1 1 1

1.00
0.8 0.8 0.8
Histology
0.6 0.6 0.6
Clear cell
0.4 0.4 0.4 0.8 Endometrioid
0.2 0.2 0.2 Mucinous

Cophenetic coefficient
0.95
0 0 0
Serous
0.6 Serous LMP

0.4 Grade
High

0.90
Rank = 5 Rank = 6 Rank = 7 Low
1 1 1 0.2
0.8 0.8 0.8 Clusters
0.6 0.6 0.6 C1
C2

0.85
0.4 0.4 0.4
C3
0.2 0.2 0.2
C4
0 0 0

2 3 4 5 6 7
Rank

D E
Kaplan−Meier curves (clusters) Kaplan−Meier curves (histologies)
1.0

1.0
Proportion of women alive

Proportion of women alive


0.8

0.8
0.6

0.6
0.4

0.4

Log−rank test P = 3.28e−06


Clear cell
0.2

0.2

C1 Endometrioid
C2 Mucinous
C3 Log−rank test P = 9.1e−07 Serous
C4 Serous LMP
0.0

0.0

0 10 20 30 40 50 60 0 10 20 30 40 50 60
Time since diagnosis (months) Time since diagnosis (months)

Figure 1.
Unsupervised nonnegative matrix factorization of the 1,000 most variable probes based on the MAD. A, Consensus matrices for rank values from 2 to 7 based on
100 independent NMF runs. B, Cophenetic correlation coefficient for rank values from 2 to 7 with 95% CIs computed by bootstrap. C, Methylation profiles of the
clusters. D, Survival curves for the methylation-derived clusters: C1 (N ¼ 51), C2 (N ¼ 44), C3 (N ¼ 34), and C4 (N ¼ 33). E, Survival for the different histologies:
clear cell (N ¼ 7), endometrioid (N ¼ 33), mucinous (N ¼ 16), serous (N ¼ 76), and serous LMP (N ¼ 30).

Methylation-based across in HGSC tumors ative (N5) subtypes had the worst survival and the immunore-
Within HGSC, unsupervised clustering of the 1,000 most active (N2) subtype had the most favorable survival. We studied
variable methylation probes found three stable methylation- the relationship between the 1,000 CpG sites used to define the
based subtypes (M1, M2, M3; Fig. 3; Supplementary Table S7). methylation-based subgroups and expression of the 39 genes
Overall survival was statistically significantly different between (Supplementary Fig. S3). None of the 1,000 CpG sites used to
these methylation subtypes of HGSC, with one subtype having define the methylation signatures were located in the 39 genes.
worse survival than the other two (log-rank test, P ¼ 0.0017). However, we observed five statistically significant correlations
Compared with patients with tumor samples in the HGSC M1/M3 after adjusting P values by the Bonferroni correction (P values
subtypes, patients in the HGSC M2 subtype had significantly smaller than 1.28  106; Supplementary Fig. S4). They were all
worse survival after adjusting for age, stage, and study (HR ¼ trans/distal associations. Next, we investigated the relationship
2.21; 95% CI, 1.12–4.36; P ¼ 0.028; Table 1). between gene expression signatures and methylation signatures.
The survival patterns observed in the methylation subtypes for all
Gene expression subgroups of HGSC the HGSC samples were retained in the subset of tumors with gene
For a subset of HGSC tumors (N ¼ 61), expression data for expression data (Fig. 4). Approximately, half of the subjects in the
39 genes were used to classify previously identified gene expres- gene expression subtype N5 were in the methylation-based cluster
sion subtypes of HGSC (Fig. 4). Survival difference, across the M2. Most subjects in the NanoString-based clusters N1 and N4
gene expression subgroups, was consistent with previous reports were in the methylation-based cluster M3. Although the N2 group
(log-rank test, P ¼ 0.023): the mesenchymal (N1) and prolifer- contained the highest proportion of subjects with the methyla-
tion-based cluster M1, approximately half of those in N2 were in
Table 1. HRs for mortality risk comparison of HGSC methylation clusters the methylation-based cluster M3 (Fig. 4; Supplementary Table
M1/M3 vs. M2 HR (95% CI) P S7). We observed that the NanoString and methylation clusters
Unadjusted 2.76 (1.48–5.12) 0.001 were significantly associated with each other (Fisher exact test, P ¼
Adjusteda 2.21 (1.12–4.36) 0.022 0.0037). Next, we investigated whether the methylation based-
NOTE: Sample size of methylation clusters: M1/M3 (N ¼ 52) and M2 (N ¼ 18). classification could add prognostic information to the gene
a
Adjusted for age, stage, and study. expression subtypes. A likelihood ratio test comparing survival

www.aacrjournals.org Clin Cancer Res; 2019 OF5

Downloaded from clincancerres.aacrjournals.org on July 29, 2019. © 2019 American Association for Cancer
Research.
Published OnlineFirst May 29, 2019; DOI: 10.1158/1078-0432.CCR-18-3720

Bodelon et al.

0.15
A B C1
C2
C3
C4

Chr
1

0.10
Status
Clear cell
1 Figure 2.

GII
Endometrioid
0.5 Mucinous A, Genomic profile of the

0.05
2
Serous segmented data, ordered by GII.
Cluster B, GII distribution by clusters
0 C1 indicated by dots: C1 (N ¼ 8), C2 (N

0.00
3
C2 ¼ 7), C3 (N ¼ 15), and C4 (N ¼ 15);
4
C3 0 10 20 30 40 segments indicate the mean values.
−0.5 C4
5
Subject Wilcoxon rank-test P (C1 vs. C3) ¼
Grade 7.75  105. Wilcoxon rank-test P
6 High
−1 (C3 vs. C4) ¼ 0.0037. Other
Low
7 comparisons were not statistically
8 GII
C significant. C, GII distribution by
0.09 histologies indicated by dots:
9
clear cell (N ¼ 3), endometrioid

0.15
0 Serous Mucinous
10 Endometrioid Clear cell (N ¼ 20), mucinous (N ¼ 7), and
serous (N ¼ 15); segments indicate
11

0.10
the mean values. Wilcoxon rank-
12
test P (serous vs. endometrioid) ¼

GII
13 8.2  104. Wilcoxon rank-test

0.05
14 P (endometrioid vs. clear cell) ¼
15
16 0.035. Other comparisons were not
17
0.00 statistically significant.
18
19
20 0 10 20 30 40
21
22
Subject

models with and without HGSC methylation subgroups (M1/ endometrioid and clear cell ovarian tumors, which, again, could
M3 vs. M2) suggested that adding the methylation subgroups reflect their cell of origin. Methylation subgroups could also reflect
added prognostic information (P ¼ 0.0072; Table 2). other etiologic factors, as endometrioid and clear cell tumors seem
to share many risk factors (11).
We related the EOC methylation subgroups to other molecular
Discussion features. In particular, we looked at the level of genomic instability
EOC is a heterogeneous disease with different histotypes exhi- for each of the methylation-based subgroups. This measure was
biting distinct clinical and genetic features. Most previous studies related to the grade of the tumor. We found that the fraction of the
of DNA methylation in EOC have focused on a single histotype or altered genome differed across the methylation subgroups with
differences between tumor and putative normal tis- EOC methylation subgroup C1, which had most of the high-grade
sues (17, 23, 46). However, the relationship between methylation tumors, having a higher average fraction of the altered genome
patterns and the different pathologic histotypes is not well under- compared with other methylation subgroups.
stood (47). In our analysis, we included the major histologic Previous studies have suggested that HGSCs are a heteroge-
subtypes, which led to the identification of four EOC methylation neous group of tumors at the molecular level (17). We found three
subgroups with shared molecular phenotypes and a statistically HGSC methylation-based subgroups with differences in survival.
significant survival difference. These EOC methylation subgroups The TCGA also had methylation information to characterize
may provide an alternative classification of ovarian tumors to HGSC. However, it used a lower density methylation array (the
histologic subtypes. The methylation-based subgroups could Illumina Infinium Human-Methylation27 BeadChip with
represent the origin of the tumors. In particular, evidence seems approximately 27,000 CpG sites) and, while the authors found
to suggest that the majority of HGSCs originate from tubal four methylation groups, they concluded that the clusters dem-
intraepithelial carcinoma (TIC; ref. 48), while a subset of HGSCs onstrated only modest stability with different clustering yielding
may have a truly ovarian origin (48, 49) as is still presumed for varying methylation subgroups (17). We found robust subgroups
LMP serous tumors (50). We found that one of the methylation regarding the number of probes, clustering methods, and repeated
subgroups (EOC methylation subgroup C1) was predominately clustering runs. The majority of the probes used to define the
represented by HGSCs, potentially being tumors of tubal origin, methylation subgroups in our study were part of the Methyla-
while another subgroup (EOC methylation subgroup C2) was tion450 BeadChip, but not present on the Methylation27 Bead-
predominantly represented by serous LMP and a small subset of Chip. However, our methylation groups need to be replicated.
HGSCs, possibly representing tumors of ovarian origin. Thus, In addition to the methylation subgroups, gene expression
while the HGSCs in both groups were histologically similar, the subtypes were consistent with previous reports (17, 18, 44). These
difference in methylation patterns among them could provide gene expression subtypes showed a statistically significant differ-
additional insights into etiology or carcinogenesis. Endometrioid ence in survival, with previously identified groups that demon-
and clear cell ovarian tumors are thought to have a common strated more favorable survival also showing better survival in our
origin from orthotopic or ectopic endometrial tissue (6). EOC study and vice versa. Previous studies that have multiple sub-
methylation subgroup C3 was predominately represented by groups based on different genomic characteristics, such as gene

OF6 Clin Cancer Res; 2019 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on July 29, 2019. © 2019 American Association for Cancer
Research.
Published OnlineFirst May 29, 2019; DOI: 10.1158/1078-0432.CCR-18-3720

Methylation and Ovarian Cancer Survival

A Rank = 2 Rank = 3 Rank = 4


B
1 1 1

0.8 0.8 0.8

1.00
0.6 0.6 0.6

0.4 0.4 0.4

0.2 0.2 0.2

Cophenetic coefficient
0.95
0 0 0

0.90
Rank = 5 Rank = 6 Rank = 7
1 1 1

0.8 0.8 0.8

0.85
0.6 0.6 0.6

0.4 0.4 0.4

0.2 0.2 0.2

0 0 0
2 3 4 5 6 7
Rank

Figure 3.
Unsupervised nonnegative matrix
factorization based on the 1,000 most C D
variable probes based on the MAD.
A, Consensus matrices for k ¼ 2 to Consensus
k ¼ 7. B, Cophenetic correlation M1
0.8 M2
coefficient for k ¼ 2 to k ¼ 7. M3
C, Methylation profiles of the clusters. 0.6
Kaplan−Meier curves (clusters)

1.0
D, Survival curves for the
0.4

Proportion of women alive


methylation-derived clusters:
M1 (N ¼ 16), M2 (N ¼ 18), and

0.8
0.2
M3 (N ¼ 36).

0.6
0.4
0.2

M1
M2
M3 Log−rank test P = 0.00171
0.0

0 10 20 30 40 50 60
Time since diagnosis (months)

expression and methylation, have not formally looked at the risk of mortality in our study, in agreement with the results of
relationship between the different genomic-based subgroups. Wang and colleagues (51). HRs ranged from 3.10 to 5.25 (P values
Here, we were interested in understanding what additional sur- ranged from 0.11 to 0.062).
vival information methylation subtypes could add to the gene Most previous studies of methylation in EOC have used fresh-
expression subtypes. While methylation subgroups were associ- frozen ovarian tumors (17, 23). However, the vast majority of
ated with gene expression subgroups, we observed that methyl- epidemiologic and clinical studies have only archival FFPE tumor
ation further indicated survival differences beyond current clinical specimens; it is therefore important to investigate whether char-
and molecular factors. This may be important for the classification acterization using methylation is robust in FFPE tumors. During
of HGSC, a better understanding of their etiology, as well as for formalin-fixation, DNA can be damaged or suffer from cross-
finding markers for early detection, and/or clinical outcomes. linking between biomolecules (DNA to DNA, DNA to proteins,
A recent paper found that hypermethylation of six adjacent etc.). The varying age of the tumors and the degree or method of
CpG loci close to the TAP1 promoter region in 6p21.3 was fixation can also affect the number of artefacts in FFPE sam-
associated with shorter time to recurrence in HGSC (51). Five of ples (52, 53). While these artifacts can have an impact on several
the six CpG sites were included in our analyses (cg02181920, technologies, such as gene expression or sequencing, DNA meth-
cg06473288, cg24111025, cg25042789, and cg26033526). ylation seems to be robust after DNA restoration (54, 55) and
Hypermethylation at these five sites was associated with higher produces accurate, reproducible results. We used tumors from two

www.aacrjournals.org Clin Cancer Res; 2019 OF7

Downloaded from clincancerres.aacrjournals.org on July 29, 2019. © 2019 American Association for Cancer
Research.
Published OnlineFirst May 29, 2019; DOI: 10.1158/1078-0432.CCR-18-3720

Bodelon et al.

A B Kaplan−Meier curves (clusters)


C
Kaplan−Meier curves (clusters) COL1A2 Methylation
M1

1.0
SPARC 2
1.0

M2
COL3A1
M3

Proportion of women alive


S100A6
Proportion of women alive

FN1 0 Nanostring
N1

0.8
TAGLN
0.8

EZR
N2
N4
SORL1 −2 N5
SLC40A1

0.6
MEST
0.6

ASRGL1 −4
ISG15
CTHRC1

0.4
0.4

DCN
FOLR1
CYP4B1
KIF1A

0.2
0.2

UCHL1
IDO1
N1 M1
N2 M2
PPL
N4 Log−rank test P = 0.000796
Log−rank test P = 0.0227 M3 PDZK1IP1

0.0
N5
0.0

NRIP1
SALL2

0 10 20 30 40 50 60 0 10 20 30 40 50 60 PRTFDC1
CXCL9

Time since diagnosis (months) Time since diagnosis (months) IGHM


IGJ
CXCL10
CXCL11
SLAMF7
ADAMDEC1
CXCL17
SCGB1D2
TSPAN8
C10orf116
MYCN
HMGA2
CD55
ZNF423

Figure 4.
A, Survival curves for gene expression subtypes [N1: mesenchymal (N ¼ 12); N2: immunoreactive (N ¼ 13); N4: differentiated (N ¼ 17); N5: proliferative (N ¼ 19)].
B, Survival curves for methylation clusters among patients with gene expression data: M1 (N ¼ 16), M2 (N ¼ 14), and M3 (N ¼ 31). C, Gene expression heatmap
showing the relationship between methylation subgroups and gene expression subtypes.

distinct studies conducted in two different countries during ovarian cancers (11). Understanding the relationship between
different periods, and we did not observe differences in methyl- risk and genetic factors with molecular subgroups could provide
ation by study. This provides evidence that methylation can additional information that could be crucial for improving pre-
robustly be run in collaborative efforts, such as consortia, where vention, early detection and therapies, especially for HGSC, which
studies are likely conducted in different locations with potentially is the most fatal subtype. We found that HGSC was not a single
variable collection protocols. In our experience, these factors did group at the methylation level. It will be important to understand
not affect the methylation signal. This is important as the classi- the relationship of these HGSC methylation subgroups in relation
fication found in our study could be applied to other epidemi- to risk and genetic factors.
ologic and clinical studies. BRCA status was unknown in our study participants. BRCA
Recent consortial epidemiologic studies have tried to clarify the carriers tend to be diagnosed with HGSC at a younger age than the
etiologic heterogeneity of EOC (11, 56, 57). These studies ana- population average. We explored whether the women diagnosed
lyzed risk factors according to histologic subtypes. Most of the with HGSC before the age of 55 (N ¼ 21) were grouped in specific
known risk factors were more strongly associated with nonserous clusters. We did not observe any specific clustering across those
represented by the different histologic subtypes (C1–C4), but 18
Table 2. HRs and likelihood ratio test comparing survival models of HGSC with of the 21 women were in HGSC cluster M3. In fact, these 18
gene expression clusters and with and without methylation subtypes women represented half of the women in cluster M3. While
N HR (95% CI)a P HR (95% CI)b P promoter methylation of BRCA1 in clusters M1–M3 was not
Gene expression but not methylation subtypes in the model
significantly different, it is possible that these are set of tumors
Gene expression clusters
N1 12 2.38 (0.94–6.05) 0.068 1.60 (0.58–4.45) 0.37
with other BRCA-like properties. Future studies with BRCA status
N2 13 0.75 (0.24–2.28) 0.61 0.81 (0.25–2.59) 0.72 should explore this further.
N4 17 1.00 (ref) — 1.00 (ref) — Our study has several strengths. It is the most comprehensive
N5 19 2.43 (1.02–5.81) 0.045 1.66 (0.62–4.39) 0.31 study of methylation of ovarian cancer to date, as we included the
Gene expression and methylation subtypes in the model major histologic subtypes of ovarian cancer and examined their
Gene expression clusters
shared methylation phenotypes. We used the same laboratory and
N1 12 2.21 (0.87–5.64) 0.097 1.56 (0.57–4.29) 0.39
N2 13 0.72 (0.23–2.20) 0.56 0.71 (0.22–2.31) 0.57
analytical methods for all EOC tumors, allowing for a valid
N4 17 1.00 (ref) — 1.00 (ref) — comparison across subtypes. We also related our methylation-
N5 19 1.44 (0.54–3.86) 0.47 1.13 (0.4–3.13) 0.82 based subgroups to other genomic features, such as copy number
Methylation clusters and gene expression. To date, this has only been done in TCGA,
M1/M3 47 1.00 (ref) - 1.00 (ref) - mostly for HGSC (some low-grade serous carcinomas might have
M2 14 3.05 (1.35–6.88) 0.007 3.12 (1.40–6.98) 0.006
been included as well; ref. 17). However, our study also suffers
Likelihood ratio test 0.0090 0.0072
comparing the
from few limitations, including sample size. Ovarian cancer is a
two models rare disease and most individual studies have small sample sizes.
a
Unadjusted for other variables. We were able to pool data from two distinct studies, and meth-
b
Additional adjusting variables in the model: age, stage and study. ylation showed robust signals independently of the study and
study period. However, larger studies should be conducted in the

OF8 Clin Cancer Res; 2019 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on July 29, 2019. © 2019 American Association for Cancer
Research.
Published OnlineFirst May 29, 2019; DOI: 10.1158/1078-0432.CCR-18-3720

Methylation and Ovarian Cancer Survival

future to validate our results. While we did not have a replication Acquisition of data (provided animals, acquired and managed patients,
set, we evaluated several independent genomic platforms in the provided facilities, etc.): C. Bodelon, W.F. Anderson, R. Matsuno,
J. Lissowska, M.S. Anglesio, D.D.L. Bowtell, J.A. Doherty, S.J. Ramus,
same specimens, including copy number and NanoString with
M. Sherman, N. Wentzensen
strong prior data in relation to ovarian cancer. NanoString-based Analysis and interpretation of data (e.g., statistical analysis, biostatistics,
clustering analysis of HGSC was aligned with previous classifica- computational analysis): C. Bodelon, J.N. Sampson, L.A. Brinton, J.A. Doherty,
tions of gene expression. Another limitation of our study is the A. Talhouk, M. Sherman, N. Wentzensen
lack of treatment information. The majority of the tumors includ- Writing, review, and/or revision of the manuscript: C. Bodelon, J.K. Killian,
ed in our analyses were from SEER population–based registries for W.F. Anderson, R. Matsuno, L.A. Brinton, J. Lissowska, M.S. Anglesio,
D.D.L. Bowtell, J.A. Doherty, S.J. Ramus, A. Talhouk, M. Sherman,
which specific treatment information is not available. It is possible
N. Wentzensen
that intrinsic characteristics of the tumors could result in better Administrative, technical, or material support (i.e., reporting or organizing
response to a specific therapy treatment or, independently of data, constructing databases): C. Bodelon, J. Lissowska, S.J. Ramus,
treatment, the molecular characteristics described in our analysis M. Sherman, N. Wentzensen
provide a survival advantage to the patient. Future studies with Study supervision: C. Bodelon, W.F. Anderson, J. Lissowska
treatment information will be able to study this question in detail. Others (pathology and development of resources): M. Sherman
In conclusion, in this comprehensive analysis of ovarian tumor
Acknowledgments
DNA methylation, we showed that there are four methylation- This research was supported by the Intramural Research Program of the NCI.
defined subgroups of EOC with survival differences, similar to In addition, S.J. Ramus acknowledges support by the grant "Identifying Prog-
histologic subtypes. Moreover, within HGSCs, we found meth- nostic Markers and Therapeutic Targets for Serous Ovarian Cancer" (R01-
ylation subgroups that added significant survival information to CA172404), J.A. Doherty acknowledges support by the grant "Epidemiologic
expression-derived molecular subgroups. factors and survival by molecular subtypes of ovarian cancer" (R01-CA168758),
and M.S. Anglesio acknowledges support by the Canadian Institutes of Health
Research (CIHR) Proof of Principle (phase I) grant "PrOTYPE: An Enabling
Disclosure of Potential Conflicts of Interest Technology to Improve Ovarian Cancer Care" and funding from the Janet D.
J.K. Killian is an employee of Foundation Medicine. No potential conflicts of Cottrelle Foundation Scholar Program administered by the BC Cancer
interest were disclosed by the other authors. Foundation.

The costs of publication of this article were defrayed in part by the payment
Authors' Contributions of page charges. This article must therefore be hereby marked advertisement
Conception and design: C. Bodelon, J.K. Killian, W.F. Anderson, A. Talhouk, in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
M. Sherman, N. Wentzensen
Development of methodology: C. Bodelon, J.K. Killian, J.N. Sampson, Received November 13, 2018; revised March 18, 2019; accepted May 23,
M.S. Anglesio, M. Sherman, N. Wentzensen 2019; published first May 29, 2019.

References
1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin 11. Wentzensen N, Poole EM, Trabert B, White E, Arslan AA, Patel AV,
2015;65:5–29. et al. Ovarian cancer risk factors by histologic subtype: an analysis
2. K€obel M, Bak J, Bertelsen BI, Carpen O, Grove A, Hansen ES, et al. Ovarian from the Ovarian Cancer Cohort Consortium. J Clin Oncol 2016;34:
carcinoma histotype determination is highly reproducible, and is 2888–98.
improved through the use of immunohistochemistry. Histopathology 12. Goode EL, Chenevix-Trench G, Song H, Ramus SJ, Notaridou M, Law-
2014;64:1004–13. renson K, et al. A genome-wide association study identifies susceptibility
3. Seidman JD, Horkayne-Szakaly I, Haiba M, Boice CR, Kurman RJ, Ronnett loci for ovarian cancer at 2q31 and 8q24. Nat Genet 2010;42:874–9.
BM. The histologic type and stage distribution of ovarian carcinomas of 13. Bolton KL, Tyrer J, Song H, Ramus SJ, Notaridou M, Jones C, et al. Common
surface epithelial origin. Int J Gynecol Pathol 2004;23:41–4. variants at 19p13 are associated with susceptibility to ovarian cancer.
4. Cho KR, Shih Ie M. Ovarian cancer. Annu Rev Pathol 2009;4:287–313. Nat Genet 2010;42:880–4.
5. Koonings PP, Campbell K, Mishell DR Jr, Grimes DA. Relative frequency of 14. Shen H, Fridley BL, Song H, Lawrenson K, Cunningham JM, Ramus SJ, et al.
primary ovarian neoplasms: a 10-year review. Obstet Gynecol 1989;74: Epigenetic analysis leads to identification of HNF1B as a subtype-specific
921–6. susceptibility gene for ovarian cancer. Nat Commun 2013;4:1628.
6. McCluggage WG. Morphological subtypes of ovarian carcinoma: a review 15. Raja FA, Chopra N, Ledermann JA. Optimal first-line treatment in ovarian
with emphasis on new developments and pathogenesis. Pathology 2011; cancer. Ann Oncol 2012;23:x118–27.
43:420–32. 16. Coward JI, Middleton K, Murphy F. New perspectives on targeted therapy
7. Trabert B, Coburn SB, Mariani A, Yang HP, Rosenberg PS, Gierach GL, in ovarian cancer. Int J Womens Health 2015;7:189–203.
et al. Reported incidence and survival of fallopian tube carcinomas: a 17. Cancer Genome Atlas Research Network. Integrated genomic analyses of
population-based analysis from the North American Association of ovarian carcinoma. Nature 2011;474:609–15.
Central Cancer Registries. J Natl Cancer Inst 2018;110:750–7. 18. Tothill RW, Tinker AV, George J, Brown R, Fox SB, Lade S, et al. Novel
8. Sung PL, Chang YH, Chao KC, Chuang CM, Task Force on Systematic molecular subtypes of serous and endometrioid ovarian cancer linked to
Review, Meta-analysis of Ovarian Cancer. Global distribution pattern of clinical outcome. Clin Cancer Res 2008;14:5198–208.
histological subtypes of epithelial ovarian cancer: a database analysis and 19. Kurman RJ, Shih Ie M. Molecular pathogenesis and extraovarian origin of
systematic review. Gynecol Oncol 2014;133:147–54. epithelial ovarian cancer–shifting the paradigm. Hum Pathol 2011;42:
9. Gardner GJ, Birrer MJ. Ovarian tumors of low malignant potential: can 918–31.
molecular biology solve this enigma? J Natl Cancer Inst 2001;93: 20. Kurman RJ, Shih Ie M. The origin and pathogenesis of epithelial ovarian
1122–3. cancer: a proposed unifying theory. Am J Surg Pathol 2010;34:433–43.
10. Zanetta G, Rota S, Chiari S, Bonazzi C, Bratina G, Mangioni C. Behavior of 21. Zaino RJ, Brady MF, Lele SM, Michael H, Greer B, Bookman MA. Advanced
borderline tumors with particular interest to persistence, recurrence, and stage mucinous adenocarcinoma of the ovary is both rare and highly lethal.
progression to invasive carcinoma: a prospective study. J Clin Oncol 2001; Cancer 2011;117:554–62.
19:2658–64. 22. Jones PA, Baylin SB. The epigenomics of cancer. Cell 2007;128:683–92.

www.aacrjournals.org Clin Cancer Res; 2019 OF9

Downloaded from clincancerres.aacrjournals.org on July 29, 2019. © 2019 American Association for Cancer
Research.
Published OnlineFirst May 29, 2019; DOI: 10.1158/1078-0432.CCR-18-3720

Bodelon et al.

23. Cicek MS, Koestler DC, Fridley BL, Kalli KR, Armasu SM, Larson MC, et al. 41. Bosquet JG, Marchion DC, Chon H, Lancaster JM, Chanock S. Analysis of
Epigenome-wide ovarian cancer analysis identifies a methylation profile chemotherapeutic response in ovarian cancers using publicly available
differentiating clear-cell histology with epigenetic silencing of the HERG high-throughput data. Cancer Res 2014;74:3902–12.
Kþ channel. Hum Mol Genet 2013;22:3038–47. 42. Curtis C, Shah SP, Chin SF, Turashvili G, Rueda OM, Dunning MJ, et al. The
24. Garcia-Closas M, Brinton LA, Lissowska J, Richesson D, Sherman ME, genomic and transcriptomic architecture of 2,000 breast tumours reveals
Szeszenia-Dabrowska N, et al. Ovarian cancer risk and common variation novel subgroups. Nature 2012;486:346–52.
in the sex hormone-binding globulin gene: a population-based case- 43. Bodelon C, Vinokurova S, Sampson JN, den Boon JA, Walker JL,
control study. BMC Cancer 2007;7:60. Horswill MA, et al. Chromosomal copy number alterations and HPV
25. Matsuno RK, Sherman ME, Visvanathan K, Goodman MT, Hernandez BY, integration in cervical precancer and invasive cancer. Carcinogenesis
Lynch CF, et al. Agreement for tumor grade of ovarian carcinoma: analysis 2016;37:188–96.
of archival tissues from the surveillance, epidemiology, and end results 44. Leong HS, Galletta L, Etemadmoghadam D, George J, Australian Ovarian
residual tissue repository. Cancer Causes Control 2013;24:749–57. Cancer Society, Kobel M, et al. Efficient molecular subtype classification of
26. Killian JK, Kim SY, Miettinen M, Smith C, Merino M, Tsokos M, et al. high-grade serous ovarian cancer. J Pathol 2015;236:272–7.
Succinate dehydrogenase mutation underlies global epigenomic diver- 45. Helland Å, Anglesio MS, George J, Cowin PA, Johnstone CN, House CM,
gence in gastrointestinal stromal tumor. Cancer Discov 2013;3:648–57. et al. Deregulation of MYCN, LIN28B and LET7 in a molecular subtype
27. Killian JK, Walker RL, Bilke S, Chen Y, Davis S, Cornelison R, et al. Genome- of aggressive high-grade serous ovarian cancers. PLoS One 2011;6:
wide methylation profiling in archival formalin-fixed paraffin-embedded e18064.
tissue samples. Methods Mol Biol 2012;823:107–18. 46. Kolbe DL, DeLoia JA, Porter-Gill P, Strange M, Petrykowska HM, Guirguis
28. Aryee MJ, Jaffe AE, Corrada-Bravo H, Ladd-Acosta C, Feinberg AP, Hansen A, et al. Differential analysis of ovarian and endometrial cancers identifies a
KD, et al. Minfi: a flexible and comprehensive Bioconductor package for the methylator phenotype. PLoS One 2012;7:e32941.
analysis of Infinium DNA methylation microarrays. Bioinformatics 2014; 47. Earp MA, Cunningham JM. DNA methylation changes in epithelial ovarian
30:1363–9. cancer histotypes. Genomics 2015;106:311–21.
29. Maksimovic J, Gordon L, Oshlack A. SWAN: Subset-quantile Within Array 48. Erickson BK, Conner MG, Landen CN Jr. The role of the fallopian
Normalization for Illumina Infinium HumanMethylation450 BeadChips. tube in the origin of ovarian cancer. Am J Obstet Gynecol 2013;209:
Genome Biol 2012;13:R44. 409–14.
30. Chen YA, Lemire M, Choufani S, Butcher DT, Grafodatskaya D, Zanke BW, 49. Przybycin CG, Kurman RJ, Ronnett BM, Shih I-M, Vang R. Are all pelvic
et al. Discovery of cross-reactive probes and polymorphic CpGs in the (nonuterine) serous carcinomas of tubal origin? Am J Surg Pathol 2010;34:
Illumina Infinium HumanMethylation450 microarray. Epigenetics 2013; 1407–16.
8:203–9. 50. Shih Ie M, Kurman RJ. Molecular pathogenesis of ovarian borderline
31. Smyth GK, Speed T. Normalization of cDNA microarray data. Methods tumors: new insights and old challenges. Clin Cancer Res 2005;11:
2003;31:265–73. 7273–9.
32. Venkatraman ES, Olshen AB. A faster circular binary segmentation algo- 51. Wang C, Cicek MS, Charbonneau B, Kalli KR, Armasu SM, Larson MC, et al.
rithm for the analysis of array CGH data. Bioinformatics 2007;23:657–63. Tumor hypomethylation at 6p21.3 associates with longer time to recur-
33. Mermel CH, Schumacher SE, Hill B, Meyerson ML, Beroukhim R, Getz G. rence of high-grade serous epithelial ovarian cancer. Cancer Res 2014;74:
GISTIC2.0 facilitates sensitive and confident localization of the targets of 3084–91.
focal somatic copy-number alteration in human cancers. Genome Biol 52. Kerick M, Isau M, Timmermann B, Sultmann H, Herwig R, Krobitsch S, et al.
2011;12:R41. Targeted high throughput sequencing in clinical cancer settings: formal-
34. Talhouk A, Kommoss S, Mackenzie R, Cheung M, Leung S, Chiu DS, et al. dehyde fixed-paraffin embedded (FFPE) tumor tissues, input amount and
Single-patient molecular testing with NanoString nCounter data using a tumor heterogeneity. BMC Med Genomics 2011;4:68.
reference-based strategy for batch effect correction. PLoS One 2016;11: 53. Schweiger MR, Kerick M, Timmermann B, Albrecht MW, Borodina T,
e0153844. Parkhomchuk D, et al. Genome-wide massively parallel sequencing of
35. Gaujoux R, Seoighe C. A flexible R package for nonnegative matrix formaldehyde fixed-paraffin embedded (FFPE) tumor tissues for copy-
factorization. BMC Bioinformatics 2010;11:1–9. number- and mutation-analysis. PLoS One 2009;4:e5548.
36. Lee DD, Seung HS. Algorithms for non-negative matrix factorization. 2001. 54. Dumenil TD, Wockner LF, Bettington M, McKeone DM, Klein K, Bowdler
https://collaborate.princeton.edu/en/publications/algorithms-for-non-negative- LM, et al. Genome-wide DNA methylation analysis of formalin-fixed
matrix-factorization paraffin embedded colorectal cancer tissue. Genes Chromosomes Cancer
37. Brunet J-P, Tamayo P, Golub TR, Mesirov JP. Metagenes and molecular 2014;53:537–48.
pattern discovery using matrix factorization. Proc Natl Acad Sci U S A 2004; 55. de Ruijter TC, de Hoon JPJ, Slaats J, de Vries B, Janssen MJFW, van Wezel T,
101:4164–9. et al. Formalin-fixed, paraffin-embedded (FFPE) tissue epigenomics using
38. Hutchins LN, Murphy SM, Singh P, Graber JH. Position-dependent motif Infinium HumanMethylation450 BeadChip assays. Lab Invest 2015;95:
characterization using non-negative matrix factorization. Bioinformatics 833–42.
2008;24:2684–90. 56. Ose J, Poole EM, Schock H, Lehtinen M, Arslan AA, Zeleniuch-Jacquotte
39. Konecny GE, Wang C, Hamidi H, Winterhoff B, Kalli KR, Dering J, et al. A, et al. Androgens are differentially associated with ovarian cancer
Prognostic and therapeutic relevance of molecular subtypes in high-grade subtypes in the Ovarian Cancer Cohort Consortium. Cancer Res 2017;
serous ovarian cancer. J Natl Cancer Inst 2014;106:pii: dju249. 77:3951–60.
40. Way GP, Rudd J, Wang C, Hamidi H, Fridley BL, Konecny GE, et al. 57. Cuellar-Partida G, Lu Y, Dixon SC, Australian Ovarian Cancer Society,
Comprehensive cross-population analysis of high-grade serous ovarian Fasching PA, Hein A, et al. Assessing the genetic architecture of epithelial
cancer supports no more than three subtypes. G3 2016;6:4097–103. ovarian cancer histological subtypes. Hum Genet 2016;135:741–56.

OF10 Clin Cancer Res; 2019 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on July 29, 2019. © 2019 American Association for Cancer
Research.
Published OnlineFirst May 29, 2019; DOI: 10.1158/1078-0432.CCR-18-3720

Molecular Classification of Epithelial Ovarian Cancer Based


on Methylation Profiling: Evidence for Survival
Heterogeneity
Clara Bodelon, J. Keith Killian, Joshua N. Sampson, et al.

Clin Cancer Res Published OnlineFirst May 29, 2019.

Updated version Access the most recent version of this article at:
doi:10.1158/1078-0432.CCR-18-3720

Supplementary Access the most recent supplemental material at:


Material http://clincancerres.aacrjournals.org/content/suppl/2019/05/29/1078-0432.CCR-18-3720.DC1

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Subscriptions Department at pubs@aacr.org.

Permissions To request permission to re-use all or part of this article, use this link
http://clincancerres.aacrjournals.org/content/early/2019/07/16/1078-0432.CCR-18-3720.
Click on "Request Permissions" which will take you to the Copyright Clearance Center's
(CCC)
Rightslink site.

Downloaded from clincancerres.aacrjournals.org on July 29, 2019. © 2019 American Association for Cancer
Research.

You might also like