Severi et al. - Online Resource METHODS Subjects and samples

advertisement
Severi et al. - Online Resource
METHODS
Subjects and samples
Subjects were selected from participants in the Melbourne Collaborative Cohort Study, a
prospective cohort study of 41,514 volunteers (24,469 women) aged between 27 and 76 years at
baseline (99.3% of whom were aged 40-69)35. At baseline attendance, participants completed
questionnaires that measured demographic characteristics and lifestyle factors including diet.
Height and weight were directly measured and a blood sample was collected and stored. For a
large proportion of individuals (75%) only dried blood spots (i.e. DBS) on Guthrie cards were
available while for others buffy coat or lymphocyte samples were available.
Nested case-control study
To estimate the association between aberrant DNA methylation from samples collected at
baseline and breast cancer risk we conducted a nested case-control study and used the Infinium
HM450 produced by Illumina to quantify DNA methylation in more than 480,000 CpG sites
covering up to 96% of CpG islands as well as a large number of non-island CpG sites 22,23.
The study sample included women in the nested case-control study that we designed to
estimate the association between mammographic density and breast cancer risk 36. In the
mammographic density study, cases were women with a first diagnosis of ductal carcinoma in
situ (DCIS; 120 cases) or invasive adenocarcinoma of the breast (International Classification of
Diseases for Oncology, C50.0-C50.9; 680 cases) occurring between baseline interview up until
31 December 2007 and ascertained by record linkage to the population-based Victorian Cancer
Registry (VCR), and to the Australian Cancer Database. For each breast cancer case, we
randomly selected 4 women as controls from among those who had not been diagnosed with
breast cancer at the age of diagnosis of the case (reference age). The controls were also matched
on year of birth, year of baseline attendance and country of origin (classified as Australia/New
1
Severi et al. - Online Resource
Zealand/UK/others, Italy or Greece). To be eligible for the methylation case-control study
women in the mammographic density case-control study had to have at least one mammogram
already retrieved at the time the study sample was assembled (first trimester 2010) and available
blood collected at baseline interview. The sample included 498 invasive breast cancer cases and
one control per case randomly selected from the four controls matching, when possible, on type
of biospecimen (DBS, buffy coat, or lymphocyte).
Tumour characteristics
Tumour characteristics were extracted from the VCR database; these include tumour grade,
size, nodal status, oestrogen receptor status (ER), progesterone receptor status (PR) and
epidermal growth factor 2 status (Her2). Additional assessment of ER, PR, HER2, EGRF1 and
CK56 status using immunohistochemistry techniques was performed by a single pathologist
(CM) 37. For the analyses presented in this paper, we used our immunohistochemistry measures
of ER, PR and HER2 and given the good agreement with the results from the original pathology
report 37, when archival tumour tissue was not available, ER, PR and HER2 status was assigned
according to the histopathology reports held at the VCR.
Biospecimens, DNA extraction and DNA methylation measures
In 485 matching sets both case and control had the same biospecimen available (373 DBS,
103 lymphocytes, 9 buffy coats); of the remaining 13 sets, in 5 the biospecimens were buffy coat
and lymphocytes and in 8 DBS and lymphocytes. This resulted in 754 samples from DBS (76%),
219 from lymphocytes (22%) and 23 from buffy coats (2%).
DNA extraction from lymphocytes and buffy coats were performed using Qiagen mini spin
columns (Hilden, Germany) while dried blood spot DNA was extracted using a method
developed in-house 38: twenty 3·2 mm diameter archived blood spot punches were added to 200
ul phosphate buffered saline and homogenised using the TissueLyser (Qiagen). The resulting
2
Severi et al. - Online Resource
supernatant was transferred to a clean 1·5 ml tube and DNA extracted using Qiagen mini spin
columns according to the manufacturer’s protocol. The quality and quantity of DNA was
assessed using the Quant-iT™ Picogreen® dsDNA assay measured on the Qubit® Fluorometer
(Life Technologies, Grand Island, NY).
Bisulfite conversion (EZ DNA Methylation-Gold kit, Zymo Research, Irvine, CA), quality
control analyses and the Infinium HM450 DNA methylation assays were performed at the
Australian Genome Research Facility as per the manufacturers’ instructions. The bisulfite
conversion control dashboard on the Illumina arrays was primarily used to check success of the
conversion. Also, a bioanalyser run was performed on a subset of samples in each batch to further
check the success of the conversion.
Samples were distributed into 96-well plates and processed by Infinium HM450 in chips of
12 arrays (8 chips per plate) with case-control pairs arranged consecutively on the same chip.
Data processing
Methylation data were normalised to the internal built-in controls as provided by the
standard Illumina software and subset-quantile within array normalization (SWAN) for type I and
II probe bias correction 39. We excluded the 416 CpGs sites on the Y chromosome and the 65
corresponding to single nucleotide polymorphisms. We assigned the methylation measures at
CpG sites with a detection p-value higher than 0·01 as missing. Samples for which more than 5%
of the CpG measures were missing were considered as “failed” and excluded from further
analyses as were the CpG sites whose measures were missing for more than 20% of the samples.
3
Severi et al. - Online Resource
References for Online Resource
1.
Das PM, Singal R. DNA methylation and cancer. Journal of clinical oncology : official
journal of the American Society of Clinical Oncology. Nov 15 2004;22(22):4632-4642.
2.
Kulis M, Esteller M. DNA methylation and cancer. Advances in genetics. 2010;70:27-56.
3.
Phillips T. The role of methylation in gene expression. Nature Education. 2008;1(1).
4.
Wilson AS, Power BE, Molloy PL. DNA hypomethylation and human diseases.
Biochimica et biophysica acta. Jan 2007;1775(1):138-162.
5.
Hinshelwood RA, Clark SJ. Breast cancer epigenetics: normal human mammary epithelial
cells as a model system. Journal of molecular medicine. Dec 2008;86(12):1315-1328.
6.
Jovanovic J, Ronneberg JA, Tost J, Kristensen V. The epigenetics of breast cancer.
Molecular oncology. Jun 2010;4(3):242-254.
7.
Kristiansen S, Jorgensen LM, Guldberg P, Soletormos G. Aberrantly methylated DNA as
a biomarker in breast cancer. The International journal of biological markers. Apr-Jun
2013;28(2):141-150.
8.
Szyf M. DNA methylation signatures for breast cancer classification and prognosis.
Genome medicine. Mar 30 2012;4(3):26.
9.
Fang F, Turcan S, Rimner A, et al. Breast cancer methylomes establish an epigenomic
foundation for metastasis. Science translational medicine. Mar 23 2011;3(75):75ra25.
10.
Zhuang J, Jones A, Lee SH, et al. The dynamics and prognostic potential of DNA
methylation changes at stem cell gene loci in women's cancer. PLoS genetics. Feb
2012;8(2):e1002517.
11.
Fackler MJ, Malone K, Zhang Z, et al. Quantitative multiplex methylation-specific PCR
analysis doubles detection of tumor cells in breast ductal fluid. Clinical cancer research.
Jun 1 2006;12(11 Pt 1):3306-3310.
4
Severi et al. - Online Resource
12.
Hoque MO, Feng Q, Toure P, et al. Detection of aberrant methylation of four genes in
plasma DNA for the detection of breast cancer. Journal of clinical oncology. Sep 10
2006;24(26):4262-4269.
13.
Horvath S, Zhang Y, Langfelder P, et al. Aging effects on DNA methylation modules in
human brain and blood tissue. Genome biology. Oct 3 2012;13(10):R97.
14.
Lim U, Flood A, Choi SW, et al. Genomic methylation of leukocyte DNA in relation to
colorectal adenoma among asymptomatic women. Gastroenterology. Jan 2008;134(1):4755.
15.
Pufulete M, Al-Ghnaniem R, Leather AJ, et al. Folate status, genomic DNA
hypomethylation, and risk of colorectal adenoma and cancer: a case control study.
Gastroenterology. May 2003;124(5):1240-1248.
16.
Hsiung DT, Marsit CJ, Houseman EA, et al. Global DNA methylation level in whole
blood as a biomarker in head and neck squamous cell carcinoma. Cancer epidemiology,
biomarkers & prevention. Jan 2007;16(1):108-114.
17.
Moore LE, Pfeiffer RM, Poscablo C, et al. Genomic DNA hypomethylation as a
biomarker for bladder cancer susceptibility in the Spanish Bladder Cancer Study: a casecontrol study. The lancet oncology. Apr 2008;9(4):359-366.
18.
Brennan K, Garcia-Closas M, Orr N, et al. Intragenic ATM methylation in peripheral
blood DNA as a biomarker of breast cancer risk. Cancer research. May 1
2012;72(9):2304-2313.
19.
Choi JY, James SR, Link PA, et al. Association between global DNA hypomethylation in
leukocytes and risk of breast cancer. Carcinogenesis. Nov 2009;30(11):1889-1897.
20.
Delgado-Cruzata L, Wu HC, Perrin M, et al. Global DNA methylation levels in white
blood cell DNA from sisters discordant for breast cancer from the New York site of the
Breast Cancer Family Registry. Epigenetics. Aug 2012;7(8):868-874.
5
Severi et al. - Online Resource
21.
Xu X, Gammon MD, Hernandez-Vargas H, et al. DNA methylation in peripheral blood
measured by LUMA is associated with breast cancer in a population-based study. FASEB
journal. Jun 2012;26(6):2657-2666.
22.
Sandoval J, Heyn H, Moran S, et al. Validation of a DNA methylation microarray for
450,000 CpG sites in the human genome. Epigenetics. Jun 2011;6(6):692-702.
23.
Clark C, Palta P, Joyce CJ, et al. A comparison of the whole genome approach of MeDIPseq to the targeted approach of the Infinium HumanMethylation450 BeadChip((R)) for
methylome profiling. PloS one. 2012;7(11):e50233.
24.
Houseman EA, Accomando WP, Koestler DC, et al. DNA methylation arrays as surrogate
measures of cell mixture distribution. BMC bioinformatics. 2012;13:86.
25.
Chen YA, Lemire M, Choufani S, et al. Discovery of cross-reactive probes and
polymorphic CpGs in the Illumina Infinium HumanMethylation450 microarray.
Epigenetics. Feb 2013;8(2):203-209.
26.
Laird PW. The power and the promise of DNA methylation markers. Nature reviews.
Cancer. Apr 2003;3(4):253-266.
27.
Woo HD, Kim J. Global DNA hypomethylation in peripheral blood leukocytes as a
biomarker for cancer risk: a meta-analysis. PloS one. 2012;7(4):e34615.
28.
Ostertag EM, Kazazian HH, Jr. Biology of mammalian L1 retrotransposons. Annual
review of genetics. 2001;35:501-538.
29.
Hoffmann MJ, Schulz WA. Causes and consequences of DNA hypomethylation in human
cancer. Biochemistry and cell biology. Jun 2005;83(3):296-321.
30.
Horard B, Eymery A, Fourel G, et al. Global analysis of DNA methylation and
transcription of human repetitive sequences. Epigenetics. Jul 1 2009;4(5):339-350.
6
Severi et al. - Online Resource
31.
Wu HC, Wang Q, Yang HI, Tsai WY, Chen CJ, Santella RM. Global DNA methylation
levels in white blood cells as a biomarker for hepatocellular carcinoma risk: a nested casecontrol study. Carcinogenesis. Jul 2012;33(7):1340-1345.
32.
Xu Z, Bolick SC, DeRoo LA, Weinberg CR, Sandler DP, Taylor JA. Epigenome-wide
association study of breast cancer using prospectively collected sister study samples.
Journal of the National Cancer Institute. May 15 2013;105(10):694-700.
33.
Michailidou K, Hall P, Gonzalez-Neira A, et al. Large-scale genotyping identifies 41 new
loci associated with breast cancer risk. Nature genetics. Apr 2013;45(4):353-361,
361e351-352.
34.
Severi G, Byrnes GB, Hopper JL. Five genetic variants associated with prostate cancer.
The New England journal of medicine. Jun 19 2008;358(25):2739-2740; author reply
2741.
35.
Giles GG, English DR. The Melbourne Collaborative Cohort Study. IARC scientific
publications. 2002;156:69-70.
36.
Baglietto L, Krishnan K, Stone J, et al. Associations of Mammographic Dense and
Nondense Areas and Body Mass Index With Risk of Breast Cancer. American journal of
epidemiology. Oct 28 2013.
37.
Krishnan K, Bassett JK, Macinnis RJ, et al. Associations between weight in early
adulthood, change in weight and breast cancer risk in postmenopausal women. Cancer
epidemiology, biomarkers & prevention. May 29 2013.
38.
Joo JE, Wong EM, Baglietto L, et al. The use of DNA from archival dried blood spots
with the Infinium HumanMethylation450 array. BMC biotechnology. 2013;13:23.
39.
Maksimovic J, Gordon L, Oshlack A. SWAN: Subset-quantile within array normalization
for illumina infinium HumanMethylation450 BeadChips. Genome biology.
2012;13(6):R44.
7
Severi et al. - Online Resource
Online Resource Table 1
Characteristics of the study sample
Age at blood collection
Cases
Controls
N=420
N=420
Mean: 56
Mean=56
SD=8
SD=8
Range=(38 to 70)
Range=(38 to 70)
Mean=56
Biospecimen
Country of Origin
Guthrie Card
325
320
Lymphocytes
84
92
Buffy Coats
11
8
355
356
65
64
Australia/NZ/UK
Italy/Greece
Reference age (*)
Mean: 64
SD: 8
Range: 44 to 83
Time from blood
collection to reference age
Grade
<5years
131
5-9years
148
10 years or more
141
Well differentiated
Moderately differentiated
179
Highly differentiated
120
Missing
Size
Nodal Status
ER
PR
HER2
Subtype
87
34
Less than 2 cm
286
2 cm or more
123
Missing
11
Negative
398
Positive
86
Missing
36
Negative
98
Positive
297
Missing
25
Negative
173
Positive
217
Missing
30
Negative
281
Positive
109
Missing
30
Luminal A
221
Luminal B
87
HER2+
20
Triple Negative
54
Missing
38
(*) Age at diagnosis for the cases and age of the case in the matching set for the controls
8
Severi et al. - Online Resource
Online Resource Table 2
Proportion of the total variance explained by the first 10
principal components and their correlation with global methylation overall and by CpG
genomic feature
PC1
PC2
PC3
PC4
PC5
PC6
PC7
PC8
PC9
PC10
Proportion
0·274
0·062
0·023
0·017
0·015
0·009
0·008
0·006
0·005
0·004
Cumulative Proportion
0·274
0·336
0·359
0·376
0·391
0·400
0·408
0·414
0·419
0·423
All
0·56
0·67
0·31
0·15
-0·30
-0·03
-0·09
0·02
-0·02
-0·02
Island/Shore
0·79
0·15
0·30
0·33
-0·36
0·07
-0·08
-0·04
-0·05
0·00
Shelf/None
0·22
0·89
0·30
-0·04
-0·20
-0·10
-0·06
0·04
0·00
-0·02
Functional Promoter
0·38
-0·25
0·19
0·43
-0·65
-0·01
-0·26
-0·14
-0·05
0·07
Non Functional Promoter
0·54
0·73
0·32
0·10
-0·23
-0·03
-0·05
0·03
-0·02
-0·03
Repetitive Elements
0·33
0·88
0·21
0·02
-0·22
-0·06
-0·07
0·01
0·00
-0·04
Non Repetitive Elements
0·62
0·58
0·34
0·20
-0·32
-0·02
-0·09
0·02
-0·02
-0·01
Total variance explained
Correlations
9
Severi et al. - Online Resource
Online Resource Table 3
Relative risk of breast cancer for 1 standard deviation of the first,
second, fifth and eight principal components
Overall
PC1
PC2
PC5
By time since blood collection
RR (95% CI)
p
0·74 (0·56-0·99)
0·04
0·66 (0·51-0·86)
0·83 (0·62-1·09)
0·002
0·18
RR (95% CI)
p
<5 years
0·44 (0·25-0·79)
0·006
5-9 years
0·99 (0·61-1·61)
0·98
10 years or more
0·82 (0·49-1·37)
0·44
test for linear trend
0·21
<5 years
0·45 (0·27-0·74)
0·002
5-9 years
0·66 (0·43-1·02)
0·06
10 years or more
0·83 (0·51-1·34)
0·44
test for linear trend
0·1
<5 years
1·2 (0·73-1·95)
0·48
5-9 years
0·73 (0·45-1·2)
0·22
0·61 (0·35-1·09)
0·1
test for linear trend
0·05
<5 years
1·83 (1·11-3·01)
0·02
5-9 years
1·10 (0·76-1·60)
0·61
10 years or more
1·04 (0·66-1·64)
0·86
test for linear trend
0·02
10 years or more
PC8
1·27 (1·00-1·62)
0·05
10
Severi et al. - Online Resource
Online Resource Figure 1
Difference in epigenome-wide methylation between cases and
controls by time between blood collection and diagnosis (number of sets in each time frame in
red)
11
Severi et al. - Online Resource
Online Resource Figure 2
Score plots of principle components analysis showing the first
component (PC1, x-axis) versus the second component (PC2, y-axis). Incident breast cancer
cases are shown in red with controls in black
12
Download