Skip to main content
Epigenetics logoLink to Epigenetics
. 2012 Jun 1;7(6):559–566. doi: 10.4161/epi.20219

Key epigenetic changes associated with lung cancer development

Results from dense methylation array profiling

Heather H Nelson 1,*, Carmen J Marsit 2, Brock C Christensen 2,3, EA Houseman 3, Milica Kontic 4, Joseph L Wiemels 5, Margaret R Karagas 6, Margaret R Wrensch 7, Shichun Zheng 7, John K Wiencke 7, Karl T Kelsey 2,3
PMCID: PMC3398985  PMID: 22522909

Abstract

Epigenetic alterations are a common event in lung cancer and their identification can serve to inform on the carcinogenic process and provide clinically relevant biomarkers. Using paired tumor and non-tumor lung tissues from 146 individuals from three independent populations we sought to identify common changes in DNA methylation associated with the development of non-small cell lung cancer. Pathologically normal lung tissue taken at the time of cancer resection was matched to tumorous lung tissue and together were probed for methylation using Illumina GoldenGate arrays in the discovery set (n = 47 pairs) followed by bisulfite pyrosequencing for validation sets (n = 99 pairs). For each matched pair the change in methylation at each CpG was calculated (the odds ratio), and these ratios were averaged across individuals and ranked by magnitude to identify the CpGs with the greatest change in methylation associated with tumor development. We identified the top gene-loci representing an increase in methylation (HOXA9, 10.3-fold and SOX1, 5.9-fold) and decrease in methylation (DDR1, 8.1-fold). In replication testing sets, methylation was higher in tumors for HOXA9 (p < 2.2 × 10−16) and SOX1 (p < 2.2 × 10−16) and lower for DDR1 (p < 2.2 × 10−16). The magnitude and strength of these changes were consistent across squamous cell and adenocarcinoma tumors. Our data indicate that the identified genes consistently have altered methylation in lung tumors. Our identified genes should be included in translational studies that aim to develop screening for early disease detection.

Keywords: DNA Methylation, goldengate, lung cancer, molecular epidemiology, pyrosequencing

Introduction

Lung cancer remains a significant worldwide public health concern and non-small cell lung cancer (NSCLC) accounts for approximately 70% of lung cancer diagnoses. In the United States in 2012, it is estimated that over 160,000 deaths will be attributed to lung cancer, which represents almost 28% of all cancer-related deaths in the US1 The two main forms of NSCLC, adenocarcinoma and squamous cell carcinoma, are both highly linked to tobacco smoking exposure. While we know that smoking causes most lung cancer, it is important to remember that smoking cessation reduces, but does not eliminate, risk. It is therefore essential to continue to identify tumor-specific molecular alterations so that effective screening, chemoprevention and curative therapies may be developed. Alterations to the epigenetics of tumors, including DNA methylation, are particularly appealing as they represent a readily detectable and potentially reversible alteration in malignancy.

Along with genetic alterations, epigenetic alterations are recognized as causal in carcinogenesis. DNA methylation is a mechanism of stable control of transcription: regulatory CpG clusters are common, often occur in tumor suppressor genes and are thought to remain largely unmethylated in noncancerous cells. In tumors, the classic example is gene promoter-based hypermethylation of CpGs that is associated with aberrant, stable gene silencing. Approximately half of all human genes contain regulatory CpG islands.2,3 However, compared with non-tumor cells, tumors may also exhibit losses of methylation at certain CpG loci, which may result in gene activation. Recently, the simultaneous resolution of hundreds of specific, phenotypically defined cancer-related CpG methylation marks has become technologically feasible, allowing for rapid, high-throughput epigenetic profiling of human tissue CpG methylation.4

Investigations of lung cancer dominated the early research in aberrant DNA methylation at tumor suppressor loci, particularly investigations of the CDKN2A and RASSF1 genes.5-8 Panels of candidate genes have been assembled and associated with clinical outcome,9,10 indicating that molecular profiling of epigenetic alterations in lung cancer will have clinical utility, although the translation of these markers to the clinic has been limited. Identifying additional lung cancer specific markers may provide improved utility and further define specific pathways altered in this disease, which can be used as pathways for new personalized and targeted treatment strategies. Therefore, it is critical to utilize high-throughput and genome-wide approaches to define key epigenetic alterations in NSCLC to foster translational studies aiming to develop screenings for early detection of disease and strategies for personalized medicine. Dense methylation profiling studies that include lung tissues are now emerging,4,11-16 and we hypothesized that there are a limited number of defining epigenetic events that are common to NSCLC tumors. To study this, we used an array-based approach, comparing matched pairs of tumor and adjacent non-tumor tissue to identify crucial epigenetic alterations, and followed these discovery-based approaches with in-depth, quantitative validation of the novel markers in independent tumor series.

Results

Discovery set tissue pairs (n = 47 pairs) were measured for CpG methylation using the Illumina Goldengate array. Eight CpG loci did not pass QA and were removed, leaving 1413 autosomal CpG loci associated with 773 cancer-related genes for analysis. Among the 47 NSCLC cases in the discovery set there were 22 adenocarcinomas and 25 squamous cell carcinomas.

CpG loci were ranked according to the magnitude of change in methylation. Given the bounded nature of the β value this was calculated as the Odds Ratio (OR), and is analogous to the fold change in methylation (increase or decrease in methylation in tumors relative to the methylation present in normal lung tissue). There were 107 CpG loci with a greater than 2-fold increase in methylation in tumors, and 43 loci with a greater than 2-fold decrease in methylation in tumors relative to the normal lung epigenome (Fig. 1 and Table 1). More specifically, among adenocarcinomas there were 128 CpG loci with greater than 2-fold increased methylation, and 57 loci with greater than 2-fold decreases in methylation. There were slightly fewer dramatic changes among squamous cell carcinomas with 95 CpG loci demonstrating greater than 2-fold increases in methylation and 40 loci with greater than 2-fold decreases in methylation. Within each histology, rank-ordered lists of ORs for methylation change were generated and there was marked similarity in the top 10 genes for adenocarcinoma and squamous cell carcinoma (Table 2). Among all 47 tissue pairs, there were several genes with CpG having at least a 5-fold increase in methylation, including homeobox A9 (HOXA9, 10.3-fold), T-cell acute lymphocytic leukemia 1 (TAL1, 7.9-fold), 5-hydroxytryptamine (serotonin) receptor 1B (HTR1B, 6.2-fold), SRY (sex determining region Y)-box 1 (SOX1, 5.9-fold), v-mos Moloney murine sarcoma viral oncogene homolog (MOS, 5.7-fold) and homeobox A11 (HOXA11, 5.0-fold). The CpGs demonstrating the greatest decreases in methylation in tumor compared with non-tumor were associated with genes, including protein tyrosine phosphatase, non-receptor type 6 (PTPN6, 5.3-fold), nidogen 1 (NID1, 3.3-fold), deleted in liver cancer 1 (DLC1, 2.9-fold), discoidin domain receptor tyrosine kinase 1 (DDR1, 2.9-fold) and nitric oxide synthase 3 (NOS3, 2.9-fold) (Table 1). Ingenuity Pathways Analysis of genes whose CpGs had at least 2-fold changes in methylation revealed that the top cellular networks for increased methylation were related to development and cell death, and the top cellular networks for decreased methylation were related to cell death and cancer (Table S1).

graphic file with name epi-7-559-g1.jpg

Figure 1. Mean fold change in methylation between tumor and normal for 47 matched tissue pairs and all 1413 autosomal CpGs by chromosome. Positive fold change values indicate increased methylation in tumor relative to normal tissue and negative fold change values indicate decreased methylation in tumor relative to normal tissue. Horizontal dotted lines indicate 2-fold and 5-fold changes in mean methylation between tumor and normal lung. Genes with CpG loci chosen for replication in independent populations (HOXA9, SOX1, DDR1) are shown.

Table 1. CpGs with greater than 2-fold increase or decrease in tumor methylation relative to non-tumor lung, discovery set (n = 47 pairs), from Illumina GoldenGate methylation array.

Gene-loci with increased methylation in tumors
Gene-loci with decreased methylation in tumors
   GENE CpG* OR** (fold change)    GENE CpG* OR** (fold change)
   HOXA9
E252
10.3
   PTPN6
E171
5.3
   TAL1
P594
7.9
   NID1
P677
3.3
   HOXA9
P1141
6.2
   DLC1
P695
2.9
   HTR1B
P222
6.2
   DDR1
P332
2.9
   SOX1
P1018
5.9
   NOS3
P38
2.9
   MOS
E60
5.7
   PTPN6
P282
2.9
   HOXA11
P698
5
   TMPRSS4
E83
2.7
   TPEF
S88
5
   AATK
E63
2.6
   MYOD1
E156
4.5
   GABRA5
P862
2.6
   HCK
P858
4.3
   SERPINB5
P19
2.6
   DLK1
E227
4
   IFNG
E293
2.5
   HS3ST2
E145
4
   SFN
P248
2.4
   IPF1
P750
4
   MUC1
P191
2.4
   DBC1
P351
3.9
   TNFSF10
P2
2.4
   MDR1
S300
3.6
   ITK
P114
2.4
   TWIST1
E117
3.6
   CASP8
E474
2.3
   WT1
E32
3.6
   EMR3
P39
2.3
   NPY
P295
3.6
   CPA4
E20
2.3
   SOX17
P303
3.6
   S100A2
P1186
2.3
   FRZB
E186
3.6
   PTPRH
E173
2.3
   SOX1
P294
3.5
   ZMYND10
P329
2.2
   ASCL2
P360
3.5
   MUC1
E18
2.2
   CDH13
E102
3.5
   GML
P281
2.2
   PDGFRA
P1429
3.3
   MKRN3
E144
2.2
   HTR1B
E232
3.2
   USP29
E274
2.2
   MT1A
P49
3.2
   TNF
P158
2.2
   RARA
P1076
3.1
   ZIM3
P718
2.1
   HOXA9
P303
3
   HDAC1
P414
2.1
   WT1
P853
3
   PLA2G2A
P528
2.1
   DCC
P471
3
   SPP1
P647
2.1
   CDH13
P88
3
   GABRA5
P1016
2.1
   SOX17
P287
2.9
   IL16
P93
2.1
   MYH11
P22
2.9
   NID1
P714
2.1
   MMP2
P303
2.9
   CYP2E1
P416
2.1
   RARB
E114
2.9
   RUNX3
E27
2
   HOXA11
E35
2.9
   BRCA1
P835
2
   IRF5
E101
2.9
   NBL1
P24
2
   TAL1
E122
2.9
   CLDN4
P1120
2
   EYA4
P794
2.9
   PI3
P1394
2
   IGF2AS
P203
2.8
   AIM2
P624
2
   PAX6
P50
2.8
   AFF3
P122
2
   STAT5A
E42
2.8
   MC2R
E455
2
   FGF2
P229
2.7
   MEST
P4
2
   TERT
P360
2.7
 
 
 
   CALCA
E174
2.6
 
 
 
   SPARC
P195
2.6
 
 
 
   NEFL
P209
2.6
 
 
 
   ASCL2
E76
2.6
 
 
 
   NTRK2
P395
2.6
 
 
 
   NTSR1
P318
2.6
 
 
 
   ERBB2
P59
2.6
 
 
 
   GSTM1
P266
2.6
 
 
 
   IGFBP3
P423
2.6
 
 
 
   AGTR1
P154
2.5
 
 
 
   FLT3
E326
2.5
 
 
 
   DCC
P177
2.5
 
 
 
   DIO3
P674
2.5
 
 
 
   MME
P388
2.5
 
 
 
   CDKN2A
S188
2.5
 
 
 
   PODXL
P1341
2.5
 
 
 
   FGF3
E198
2.5
 
 
 
   TWIST1
P44
2.5
 
 
 
   RASSF1
P244
2.5
 
 
 
   FGF3
P171
2.5
 
 
 
   GALR1
E52
2.4
 
 
 
   IRAK3
P13
2.4
 
 
 
   SLIT2
P208
2.4
 
 
 
   DCC
E53
2.4
 
 
 
   P2RX7
P119
2.4
 
 
 
   GFI1
P45
2.3
 
 
 
   FGF12
P210
2.3
 
 
 
   ADCYAP1
P398
2.3
 
 
 
   PROK2
P390
2.3
 
 
 
   EYA4
E277
2.3
 
 
 
   GFI1
E136
2.3
 
 
 
   MT1A
E13
2.3
 
 
 
   HOXB2
P488
2.3
 
 
 
   SFRP1
E398
2.3
 
 
 
   KDR
P445
2.2
 
 
 
   PITX2
E24
2.2
 
 
 
   AGTR1
P41
2.2
 
 
 
   PENK
P447
2.2
 
 
 
   ESR1
P151
2.2
 
 
 
   RASSF1
E116
2.2
 
 
 
   HOXA5
P1324
2.2
 
 
 
   NTRK3
P752
2.2
 
 
 
   HOXA5
E187
2.2
 
 
 
   MOS
P27
2.1
 
 
 
   NPR2
P618
2.1
 
 
 
   JAK3
P156
2.1
 
 
 
   TJP2
P330
2.1
 
 
 
   TCF4
P317
2.1
 
 
 
   HOXC6
P456
2.1
 
 
 
   NTRK3
E131
2.1
 
 
 
   NTRK2
P10
2.1
 
 
 
   MAF
P826
2.1
 
 
 
   COL1A2
P48
2.1
 
 
 
   TPEF
S36
2.1
 
 
 
   IRAK3
E130
2.1
 
 
 
   TAL1
P817
2
 
 
 
   CFTR
P115
2
 
 
 
   CHGA
E52
2
 
 
 
   TMEFF2
P152
2
 
 
 
   THY1
P149
2
 
 
 
   CCNA1
E7
2
 
 
 
   PYCARD
P150
2
 
 
 
   GUCY2D E419 2      
*

Location relative to transcription start site in bases, p = promoter, E = exonic. **OR: [(tumor β/1-tumor β) / (normal tissue β/1-normal tissue β)]

Table 2. Rank order of top differentially methylated CpGs between tumor and normal lung tissue by histology.

 
Rank
GENE_CpG Overall Adenocarcinomas Squamous cell carcinomas
HOXA9_E252
1
1
1
TAL1_P594
2
2
6
HOXA9_P1141
3
12
2
HTR1B_P222
4
7
5
SOX1_P1018
5
9
7
MOS_E60
6
5
11
HOXA11_P698
7
21
8
TPEF_S88
8
10
12
MYOD1_E156
9
11
17
HCK_P858 10 35 10

In the replication data sets there were significant differences in methylation comparing the matched normal and tumor tissues. In replication set 1, HOXA9 had a 2.1-fold methylation increase in tumors, which was comparable to the 3.5-fold methylation increase observed for tumors in replication set 2 (p < 7.3 × 10−9 for set 1 and p < 2.1 × 10−9 for set 2, Table 3). Similarly, in tumors compared with matched non-tumor, SOX1 had a 2.4-fold methylation increase in set 1 and a 3.1-fold methylation increase in replication set 2 (p < 5.2 × 10−8 and p < 1.5 × 10−10, respectively, Table 3). Finally, there were consistent results for decreased methylation at DDR1 in lung tumors relative to the normal lung genome. In replication set 1, there was a 1.4-fold decrease in methylation of DDR1 (p < 2.6 × 10−11) and, in replication set 2, there was a 1.4-fold decrease in DDR1 methylation (p < 1.5 × 10−11, Table 3). For all three genes a majority of tumor specimens were abnormally methylated (outside 1 standard deviation of the normal tissue mean): 73% and 64% of tumors were hypermethylated at SOX1 and HOXA9, respectively, and 73% of tumors were hypomethylated at DDR1.

Table 3. Mean CpG methylation and standard deviation at three genes in two independent tissue sets.

 
Replication set 1 (n = 55)
Replication set 2 (n = 54)
Pooled data (n = 99)
  Normal Tumor p value Normal Tumor p value Normal Tumor p value
HOXA9
16.6 ± 3.2
33.9 ± 18.3
7.3 × 10−9
17.9 ± 8.0
36.5 ± 17.1
2.1 × 10−9
17.1 ± 8.0
35.1 ± 17.6
2.2 × 10−16
SOX1
11.6 ± 3.2
23.9 ± 13.8
5.2 × 10−8
12.3 ± 4.5
31.3 ± 16.7
1.5×10−10
11.9 ± 3.9
27.4 ± 15.7
2.2 × 10−16
DDR1 54.6 ± 7.1 37.4 ± 13.1 2.6 × 10−11 51.1 ± 5.6 35.5 ± 12.3 1.5 × 10−11 52.9 ± 6.6 36.4 ± 12.7 2.2 × 10−16

Pooling the replication data and examining methylation changes according to histology revealed no significant differences. For squamous cell carcinoma there was a 3.5-fold increase in HOXA9 methylation, a 3.1-fold increase in SOX1 methylation, and a 1.5-fold decrease in DDR1 methylation. For adenocarcinoma these changes were a 2.1-fold increase in HOXA9, a 2.3-fold increase in SOX1 and a 1.4-fold decrease in DDR1 methylation. There was a higher percentage of squamous cell carcinomas with SOX1 hypermethylation than adenocarcinomas (83% and 65% of tumors). For HOXA9, hypermethylation was relatively constant for adenocarcinoma (68%) and squamous cell carcinoma (72%), whereas DDR1 hypomethylation was slightly higher in squamous cell carcinoma (78%) than adenocarcinomas (73%). None of these differences by histology were statistically significant.

Finally, for two of our genes, one hypomethylated in tumors (DDR1) and one hypermethylated in tumors (SOX1), we measured gene expression using RT-PCR (Fig. S1). We observed a higher level of DDR1 expression among tumors, consistent with the observed gene hypomethylation, and decreased SOX1 expression, again consistent with the hypermethylation observed in tumors.

Discussion

We used methylation arrays to discover common epigenetic alterations that define NSCLC by comparing patient-matched tumor and non-tumor tissues in three independent populations. By describing tumor-specific epigenetic alterations in NSCLC common to squamous cell and adenocarcinomas we are fostering the development of novel detection and screening strategies. We identified several genes with CpGs that have altered methylation in tumors compared with non-tumor tissue, and validated three genes; HOXA9, SOX1 and DDR1 in two replication sets from independent patient populations that have consistently and significantly altered DNA methylation in NSCLC compared with non-tumor lung tissue.

The homeobox (HOX) family of genes encodes transcription factors that are differentially expressed spatially and temporally during embryonic development, and HOXA9 is part of the HOXA family of transcription factors on chromosome 7. The homeobox genes have been described as being hypermethylated in lung cancer cell lines, and the HOXA9 gene has been specifically shown to be hypermethylated in primary lung cancers.15,17 Although we focused on HOXA9 in our replication sets, among the nine HOXA gene CpGs (from HOXA5, HOXA9 and HOXA11) measured on the array in the discovery pairs, seven (78%) demonstrated over 2-fold increases in methylation in tumors relative to non-tumors. The strong overrepresentation of HOXA gene CpGs with NSCLC-specific increases in methylation suggests that, in general, these genes are excellent targets for the development of lung cancer biomarkers. Other HOX family gene members that have been reported to have increased methylation in NSCLC include HOXC9 and HOXA1. Anglim et al. observed a significantly increased prevalence of HOXC9 methylation in squamous cell lung tumors, though the increase was of moderate magnitude, 66% of tumors compared with 60% of adjacent lung tissue samples.11 In adenocarcinomas, Tsou et al. reported significantly increased HOXA1 methylation compared with non-tumor lung.16 Additional findings that are consistent with ours are included in the original description of the GoldenGate methylation array.4 In the original description of the array, Bibikova et al. measured methylation in two independent sets (n = 11 and n = 12 pairs) of lung adenocarcinomas and normal lung tissues, identified 55 CpG loci with both statistically significant and a large magnitude of increased methylation in tumors, and five of these CpGs were in HOXA family genes.4

Similar to the homeobox genes, SOX1 encodes a transcription factor important in development. Although reports of SOX1 methylation in NSCLC are lacking, it has been reported to have increased methylation in malignant ovarian tumors compared with benign disease.18 Additional evidence for the potential utility of SOX1 as a cancer biomarker comes from Apostolidou et al.19 who reported a significantly increased prevalence of SOX1 methylation in high-grade squamous intraepithelial lesions compared with nonspecific cytological alterations in cervical cell suspension samples. Interestingly, in the same report, these authors reported significantly increased HOXA11 methylation in high-grade lesions and that both SOX1 and HOXA11 discriminated high-grade intraepithelial lesions from controls with high sensitivity and specificity. Consistent with the observed increases in methylation of developmentally important HOXA family and SOX1 transcription factors, Ingenuity cellular networks analysis revealed cellular development as a function common to the top three cellular networks with genes having at least 2-fold increases in methylation. In addition, cell death was a common function between both of the top two networks associated with 2-fold increases or decreases in gene methylation, suggesting that there is widespread epigenetic dysregulation of genes that participate in cell death processes.

Most classical investigations of tumor methylation have focused on increased methylation of tumor suppressor genes, though decreased gene-promoter methylation is also common in tumors and may be critically informative in biomarker development. For example, using the GoldenGate array, we have shown that the majority of significant methylation alterations (727 of 969, 75% with Q < 0.05) between non-tumor pleura and mesotheliomas are instances of decreased methylation.20 In the discovery set of lung tumor and non-tumor pairs described here, nearly 30% of CpGs with changes in methylation (of at least 2-fold) were decreases in methylation, including DDR1. DDR1 encodes a receptor tyrosine kinase normally expressed in epithelial cells, including the lung, and it is overexpressed in lung tumors,21-23 consistent with our finding of DNA methylation loss at this gene.

There is great interest in the development of lung cancer screening biomarkers that can identify early signs of disease, including tissue-associated biomarkers that could augment differential diagnosis following spiral CT. Examples include identifying tumor-specific genetic or epigenetic alterations in sputum, lung lavage or cell-free DNA in serum. Serum-based detection of epigenetic alterations associated with tumors has been proposed as a viable strategy in lung cancer screening.24 And, in fact, methylated circulating DNA in cancer patients has been previously associated with disease. For example, lung cancer cases have significantly more serum-derived DNA than controls (p < 0.0001).25 In non-small cell lung cancer patients, Esteller et al. showed that when a patient’s tumor was positive for methylation (at one of four investigated genes) 73% of the time methylated DNA was also detectable in the serum.26 Other work in lung cancer has been similar: in 2002, An et al. showed that 88% of patients (n = 64/73) with CDKN2A methylation in tumors also had methylation in serum-derived DNA.27 That same year Usadel and colleagues found that 47% (n = 42/89) of lung cancer cases had APC methylation in serum DNA.28 Finally, Ramirez et al. demonstrated a high within-person correlation between lung tumor methylation and sera-derived DNA methylation (p < 0.001) at the DAPK and RASSF1 genes,29 and Hsu et al. reported 75–86% concordance of serum and lung tumor methylation.30

Clearly, a major limitation of serum-based epigenetic lung cancer screening biomarkers is that no single methylation markers will capture every tumor. Distinct tumor histologies, different etiologic exposures or differences in underlying susceptibility may all contribute to heterogeneity in the pattern of methylation alterations associated with the tumor phenotype. However, between histologic subtypes and across nearly 50 tumors, our approach was able to identify 150 CpGs with statistically significant, at least 2-fold changes in the magnitude of methylation, and allows for the identification of common tagging marks to maximize biomarker sensitivity. Another potential limitation for epigenetic biomarkers of NSCLC is sub-optimal specificity, and candidate genes are often altered in many malignancies. Both HOXA9 and SOX1 are transcription factors known to be important in embryonic development and epigenetic alterations in these genes may not be specific to NSCLC. In light of the cancer stem cell hypothesis and studies in ovarian and cervical cancer discussed above, additional research replicating our other 150 preliminary CpG biomarker candidates may be necessary to proffer epigenetic biomarkers that are highly specific to NSCLC. Nonetheless, our discovery approach allows us to build on the innovative concept that a panel of genes targeted for epigenetic alteration can define a more sensitive and specific screening strategy for this disease.

One possible limitation of our approach is that the matched non-tumor lung tissue may not be entirely epigenetically normal. In an attempt to limit any field cancerization effect, the non-tumorous lung was sampled distant from the tumor mass being resected. However, as this tissue is taken from diseased individuals, most with a smoking history, it is plausible that the normal tissue could have pre-neoplastic change. On the other hand, identifying CpG loci that are altered in tumor tissue relative to non-tumor lung with potential pre-neoplastic epigenetic alterations increases the potential utility of these biomarkers for screening applications as they reflect changes that occur in the transition from pre-neoplasia to malignancy.

We identified 150 CpG loci associated with 120 genes that have a significant and at least 2-fold magnitude difference in methylation between tumor and matched non-tumor tissue. We then replicated the results for HOXA9, SOX1 and DDR1 in 99 additional paired tumor and non-tumor tissues from two independent populations. Our approach provides further proof of concept that strategies in the development of early detection biomarkers for NSCLC that include panels of markers will outperform single-marker approaches; and our results strongly indicate that CpGs in HOXA9, SOX1 and DDR are highly attractive as members of such a biomarker panel. Importantly, the discovery of a panel of epigenetic signals that can be combined to develop a sensitive and specific biomarker panel may also provide critical insight into cellular pathways where dysregulation is causal in this disease, further elucidate the mechanisms underlying lung cancer development and enable development of strategies for therapeutic intervention that successfully target the causal pathways.

Patients and Methods

Tissues

Subject demographic, tumor histology and smoking status are in Table 4.

Table 4. Subject demographic, tumor histology and smoking status count data.

  Discovery set (n = 47) Replication set 1 (n = 55) Replication set 2 (n = 54)
Age, mean (SD)
68.7 (9.1)
72.4 (10.3)
59.9 (6.5)
Sex
 
 
 
Female
21
34
18
Male
26
21
36
Histology
 
 
 
Adenocarcinoma
22
38
20
Squamous cell carcinoma
25
17
34
Smoking status
 
 
 
Never
2
 
2
Former
27
 
10
Current 15   41
*

These are counts, not percents

Discovery set

Paired tissues were derived from a surgical case series at the Massachusetts General Hospital from 1993 to 1996, as described in Wiencke et al.31 and Nelson et al.32 Patients undergoing surgical resection for non-small cell lung cancer consented to tissue collection and a small amount of tumor specimen and normal lung tissue distant to the tumor were collected during surgery and immediately snap frozen for research purposes. From the surgical series, a random subset of 47 patients with matched normal-tumor specimens was used.

Replication set #1

A set of paired lung tumor-normal pairs were obtained from the Brown Center for Cancer Research Molecular Pathology Core tissue bank. These were obtained from the pathology surgical suite and snap frozen.

Replication set #2

Tissues were obtained from a surgical series at the University of Belgrade Medical School Clinic for Pulmonology. This case-series of NSCLC was initiated in 2008 and is comprised of surgical patients with no pre-surgical chemotherapy or radiotherapy. Demographic data are obtained through patient interview, and clinical information derived from chart review. Tumor and normal tissue are obtained in the operating room and flash-frozen.

Methylation analysis

Preparation of DNA and Illumina GoldenGate array

DNA from fresh frozen tissue samples was isolated with QIAamp DNA mini kit (Qiagen), and modified with sodium bisulfite using the EZ DNA Methylation kit (Zymo Research). Illumina GoldenGate methylation bead arrays were processed at the University of California San Francisco Institute for Human Genetics, Genomics Core Facility as described by Bibikova and colleagues.4 Illumina GoldenGate array methylation data are publicly available on the GEO archive under accession number GSE27902.

Bisulfite Pyrosequencing

Three loci were chosen for analysis in the validation tissues using bisulfite pyrosequencing. Assays were designed using Pyromark Assay Design 2.0 (Qiagen), and pyrosequencing data were collected on the Pyromark Q96MD. For the HOXA9 gene, 1 CpG (the CpG measured by the array) was evaluated, for SOX1, 4 CpGs and for DDR1, 3 CpGs (both groups included the array CpG) were assessed. The average methylation across CpGs for each gene was calculated.

mRNA analysis

RNA was extracted from 30 mg of frozen lung tissue the RNeasy Mini Kit (Qiagen) and quantified with a NanoDrop Spectrophotometer. RNA was converted to cDNA with Invitrogen SuperScript III Reverse Transcriptase according to the manufacturer's protocol and included an Rnase inhibitor (Roche). Expression of the DDR1 and SOX1 mRNA transcripts was measured using commercially available Integrated DNA Technologies (IDT) PrimeTime qPCR primer/probe sets [IDT Assay names: DDR1 -Hs.PT.49a.19433871.g, (exon 6–7), SOX1 - Hs.PT.49a.2697171.g (exon 1)].

All reactions were run in triplicate on a BioRad CFX Connect Real Time PCR system with GAPDH serving as a referent [GAPDH - Hs.PT.49a.20047924.g (exon1)]. A pooled sample of RNAs was run on each plate to allow normalization using CFX Manager Software (version 2.1) and relative expression was then determined using the ΔΔCt method.

Data analysis

Illumina BeadStudio Methylation software was used for data set assembly. Fluorescent signals for methylated (Cy5) and unmethylated (Cy3) alleles give methylation level: β = [max(Cy5, 0)] /(|Cy3| + |Cy5| + 100) with ~30 replicate bead measurements per locus. For each matched tumor-normal paper the magnitude of methylation change was calculated using the odds ratio (OR: [(tumor β/1-tumor β) / (normal tissue β/1-normal tissue β)]). The OR is analogous to measuring the fold change in methylation, and was applied to these data as β is a restricted value between 0 and 1. For each locus the OR was averaged across all individuals. If the OR was less than one the number was inverted to obtain the relative fold decrease in methylation. Ingenuity Pathways Analysis software was used to determine the top networks enriched with genes whose CpGs had at least 2-fold increases or decreases (separately) in methylation using all autosomal array genes as the referent.

Supplementary Material

Additional material
epi-7-559-s01.pdf (259.1KB, pdf)

Funding

NIH R01ES006717 and R01CA126831 (JKW); NIH P20RR018728 (CJM); NIH R01CA52689 and P50097257 (MRW); NIH R01CA57494 and P42ES007373 (MRK); NIH R01CA078609, R01CA121147, R01CA126939 and R01CA100679 (KTK); NIH P30CA077598 (HNIH P30CA077598 (H.H.N.)HN).

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Footnotes

References

  • 1.American Cancer Society. Cancer Facts & Figures 2012. Atlanta: American Cancer Society; 2012. [Google Scholar]
  • 2.Bird A. DNA methylation patterns and epigenetic memory. Genes Dev. 2002;16:6–21. doi: 10.1101/gad.947102. [DOI] [PubMed] [Google Scholar]
  • 3.Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nat Rev Genet. 2002;3:415–28. doi: 10.1038/nrg816. [DOI] [PubMed] [Google Scholar]
  • 4.Bibikova M, Lin Z, Zhou L, Chudin E, Garcia EW, Wu B, et al. High-throughput DNA methylation profiling using universal bead arrays. Genome Res. 2006;16:383–93. doi: 10.1101/gr.4410706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kim DH, Nelson HH, Wiencke JK, Zheng S, Christiani DC, Wain JC, et al. p16(INK4a) and histology-specific methylation of CpG islands by exposure to tobacco smoke in non-small cell lung cancer. Cancer Res. 2001;61:3419–24. [PubMed] [Google Scholar]
  • 6.Marsit CJ, Kim DH, Liu M, Hinds PW, Wiencke JK, Nelson HH, et al. Hypermethylation of RASSF1A and BLU tumor suppressor genes in non-small cell lung cancer: implications for tobacco smoking during adolescence. Int J Cancer. 2005;114:219–23. doi: 10.1002/ijc.20714. [DOI] [PubMed] [Google Scholar]
  • 7.Belinsky SA, Nikula KJ, Palmisano WA, Michels R, Saccomanno G, Gabrielson E, et al. Aberrant methylation of p16(INK4a) is an early event in lung cancer and a potential biomarker for early diagnosis. Proc Natl Acad Sci U S A. 1998;95:11891–6. doi: 10.1073/pnas.95.20.11891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Dammann R, Li C, Yoon JH, Chin PL, Bates S, Pfeifer GP. Epigenetic inactivation of a RAS association domain family protein from the lung tumour suppressor locus 3p21.3. Nat Genet. 2000;25:315–9. doi: 10.1038/77083. [DOI] [PubMed] [Google Scholar]
  • 9.Brock MV, Hooker CM, Ota-Machida E, Han Y, Guo M, Ames S, et al. DNA methylation markers and early recurrence in stage I lung cancer. N Engl J Med. 2008;358:1118–28. doi: 10.1056/NEJMoa0706550. [DOI] [PubMed] [Google Scholar]
  • 10.Buckingham L, Penfield Faber L, Kim A, Liptay M, Barger C, Basu S, et al. PTEN, RASSF1 and DAPK site-specific hypermethylation and outcome in surgically treated stage I and II nonsmall cell lung cancer patients. Int J Cancer. 2010;126:1630–9. doi: 10.1002/ijc.24896. [DOI] [PubMed] [Google Scholar]
  • 11.Anglim PP, Galler JS, Koss MN, Hagen JA, Turla S, Campan M, et al. Identification of a panel of sensitive and specific DNA methylation markers for squamous cell lung cancer. Mol Cancer. 2008;7:62. doi: 10.1186/1476-4598-7-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Ehrich M, Field JK, Liloglou T, Xinarianos G, Oeth P, Nelson MR, et al. Cytosine methylation profiles as a molecular marker in non-small cell lung cancer. Cancer Res. 2006;66:10911–8. doi: 10.1158/0008-5472.CAN-06-0400. [DOI] [PubMed] [Google Scholar]
  • 13.Feng Q, Hawes SE, Stern JE, Wiens L, Lu H, Dong ZM, et al. DNA methylation in tumor and matched normal tissues from non-small cell lung cancer patients. Cancer Epidemiol Biomarkers Prev. 2008;17:645–54. doi: 10.1158/1055-9965.EPI-07-2518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Field JK, Liloglou T, Warrak S, Burger M, Becker E, Berlin K, et al. Methylation discriminators in NSCLC identified by a microarray based approach. Int J Oncol. 2005;27:105–11. doi: 10.3892/ijo.27.1.105. [DOI] [PubMed] [Google Scholar]
  • 15.Rauch T, Wang Z, Zhang X, Zhong X, Wu X, Lau SK, et al. Homeobox gene methylation in lung cancer studied by genome-wide analysis with a microarray-based methylated CpG island recovery assay. Proc Natl Acad Sci U S A. 2007;104:5527–32. doi: 10.1073/pnas.0701059104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Tsou JA, Galler JS, Siegmund KD, Laird PW, Turla S, Cozen W, et al. Identification of a panel of sensitive and specific DNA methylation markers for lung adenocarcinoma. Mol Cancer. 2007;6:70. doi: 10.1186/1476-4598-6-70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Weisenberger DJ, Trinh BN, Campan M, Sharma S, Long TI, Ananthnarayan S, et al. DNA methylation analysis by digital bisulfite genomic sequencing and digital MethyLight. Nucleic Acids Res. 2008;36:4689–98. doi: 10.1093/nar/gkn455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Su HY, Lai HC, Lin YW, Chou YC, Liu CY, Yu MH. An epigenetic marker panel for screening and prognostic prediction of ovarian cancer. Int J Cancer. 2009;124:387–93. doi: 10.1002/ijc.23957. [DOI] [PubMed] [Google Scholar]
  • 19.Apostolidou S, Hadwin R, Burnell M, Jones A, Baff D, Pyndiah N, et al. DNA methylation analysis in liquid-based cytology for cervical cancer screening. Int J Cancer. 2009;125:2995–3002. doi: 10.1002/ijc.24745. [DOI] [PubMed] [Google Scholar]
  • 20.Christensen BC, Houseman EA, Godleski JJ, Marsit CJ, Longacker JL, Roelofs CR, et al. Epigenetic profiles distinguish pleural mesothelioma from normal pleura and predict lung asbestos burden and clinical outcome. Cancer Res. 2009;69:227–34. doi: 10.1158/0008-5472.CAN-08-2586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ford CE, Lau SK, Zhu CQ, Andersson T, Tsao MS, Vogel WF. Expression and mutation analysis of the discoidin domain receptors 1 and 2 in non-small cell lung carcinoma. Br J Cancer. 2007;96:808–14. doi: 10.1038/sj.bjc.6603614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Rikova K, Guo A, Zeng Q, Possemato A, Yu J, Haack H, et al. Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell. 2007;131:1190–203. doi: 10.1016/j.cell.2007.11.025. [DOI] [PubMed] [Google Scholar]
  • 23.Yang SH, Baek HA, Lee HJ, Park HS, Jang KY, Kang MJ, et al. Discoidin domain receptor 1 is associated with poor prognosis of non-small cell lung carcinomas. Oncol Rep. 2010;24:311–9. doi: 10.3892/or_00000861. [DOI] [PubMed] [Google Scholar]
  • 24.Heller G, Zielinski CC, Zöchbauer-Müller S. Lung cancer: from single-gene methylation to methylome profiling. Cancer Metastasis Rev. 2010;29:95–107. doi: 10.1007/s10555-010-9203-x. [DOI] [PubMed] [Google Scholar]
  • 25.Yoon KA, Park S, Lee SH, Kim JH, Lee JS. Comparison of circulating plasma DNA levels between lung cancer patients and healthy controls. J Mol Diagn. 2009;11:182–5. doi: 10.2353/jmoldx.2009.080098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Esteller M, Sanchez-Cespedes M, Rosell R, Sidransky D, Baylin SB, Herman JG. Detection of aberrant promoter hypermethylation of tumor suppressor genes in serum DNA from non-small cell lung cancer patients. Cancer Res. 1999;59:67–70. [PubMed] [Google Scholar]
  • 27.An Q, Liu Y, Gao Y, Huang J, Fong X, Li L, et al. Detection of p16 hypermethylation in circulating plasma DNA of non-small cell lung cancer patients. Cancer Lett. 2002;188:109–14. doi: 10.1016/S0304-3835(02)00496-2. [DOI] [PubMed] [Google Scholar]
  • 28.Usadel H, Brabender J, Danenberg KD, Jerónimo C, Harden S, Engles J, et al. Quantitative adenomatous polyposis coli promoter methylation analysis in tumor tissue, serum, and plasma DNA of patients with lung cancer. Cancer Res. 2002;62:371–5. [PubMed] [Google Scholar]
  • 29.Ramirez JL, Sarries C, de Castro PL, Roig B, Queralt C, Escuin D, et al. Methylation patterns and K-ras mutations in tumor and paired serum of resected non-small-cell lung cancer patients. Cancer Lett. 2003;193:207–16. doi: 10.1016/S0304-3835(02)00740-1. [DOI] [PubMed] [Google Scholar]
  • 30.Hsu HS, Chen TP, Hung CH, Wen CK, Lin RK, Lee HC, et al. Characterization of a multiple epigenetic marker panel for lung cancer detection and risk assessment in plasma. Cancer. 2007;110:2019–26. doi: 10.1002/cncr.23001. [DOI] [PubMed] [Google Scholar]
  • 31.Wiencke JK, Thurston SW, Kelsey KT, Varkonyi A, Wain JC, Mark EJ, et al. Early age at smoking initiation and tobacco carcinogen DNA damage in the lung. J Natl Cancer Inst. 1999;91:614–9. doi: 10.1093/jnci/91.7.614. [DOI] [PubMed] [Google Scholar]
  • 32.Nelson HH, Christiani DC, Mark EJ, Wiencke JK, Wain JC, Kelsey KT. Implications and prognostic value of K-ras mutation for early-stage lung cancer in women. J Natl Cancer Inst. 1999;91:2032–8. doi: 10.1093/jnci/91.23.2032. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Additional material
epi-7-559-s01.pdf (259.1KB, pdf)

Articles from Epigenetics are provided here courtesy of Taylor & Francis

RESOURCES