Genomic copy number alterations associated with the early brain metastasis of non-small cell lung cancer

  • Authors:
    • Hye Won Lee
    • Ho Jun Seol
    • Yoon-La Choi
    • Hyun Jung Ju
    • Kyeung Min Joo
    • Young-Hyeh Ko
    • Jung-Il Lee
    • Do-Hyun Nam
  • View Affiliations

  • Published online on: October 15, 2012     https://doi.org/10.3892/ijo.2012.1663
  • Pages: 2013-2020
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Frequent early development of systemic metastasis leads to unfavourable clinical prognosis of non-small cell lung cancer (NSCLC). Although brain metastasis (BM) contributes significantly to morbidity and mortality of NSCLC, relevant driver mechanisms are largely unknown. To elucidate genetic alterations associated with early BM of NSCLC, we retrospectively collected 18 NSCLC cases with BM [12 adenocarcinomas (ADC) and 6 squamous cell carcinomas (SQCC)] whose surgical tissues of both primary and brain metastatic tumors were preserved as formaldehyde-fixed and paraffin-embedded (FFPE) pathological samples. When chromosomal copy number alterations (CNA) of those FFPE samples were analysed by the Molecular Inversion Probe (MIP) technology, the most frequent CNAs detected in primary lung ADCs were gains of 3q, 5p, 5q, 6p, 8q, 9p, 11p, 15q, 17q and losses of 10q and 22q whereas primary lung SQCCs revealed gains in 4q and 12q and loss in 9q. In particular, when comparative MIP was performed in primary 12 ADCs depending on the pattern of BM to uncover predetermining signatures that can predict the risk of BM, selectively amplified regions of primary lung ADCs (5q35, 10q23 and 17q23-24) were identified as significantly associated with the development of early BM within 3 months after first diagnosis of primary tumors. Those regions harbour several candidate genes including NeurL1B, ACTA2, FAS and ICAM2. Although more validation is needed, the genetic signatures elucidated in this study help to identify useful molecular markers defining an NSCLC patient subgroup at risk of early BM, guiding therapeutic decisions.

Introduction

Non-small cell lung cancer (NSCLC) including adenocarcinoma (ADC) and squamous cell carcinoma (SQCC) accounts for approximately 80–90% of all diagnosed lung cancers (1,2). Despite recent advances in understanding complex pathophysiology, uncovering predictive/prognostic markers, and developing novel technologies to aid diagnosis/treatment decision, NSCLC is still the major determinant of overall cancer mortality (37). The 5-year survival rate was 15% across all stages of the disease-ranging from 2.8% for patients with distant metastases to nearly 50% for those presenting with local disease. Because NSCLC is a heterogeneous disease entity, the accurate stratification of the patients with high risk of developing recurrence or distant metastasis would improve final prognosis.

While the brain is a major site of relapse contributing to the unfavorable prognosis of the NSCLC, the relevant molecular mechanisms are largely unknown. Lung ADC is known to establish distant macrometastasis within months of initial diagnosis (810). This short latency implies that metastatic competence would result from early oncogenic events that drive primary tumor growth rather than late-arising, rare genomic alterations specific for metastasis (11). Therefore, defining consistent chromosomal changes in the primary NSCLC could help to identify metastasis-specific signatures. Molecular inversion probe (MIP) technology is a high-throughput genotyping capable of providing copy number measurements with high precision and specificity, proven to be a valid array for analyzing copy number in formaldehyde-fixed and paraffin-embedded (FFPE) samples (1214). In this study, to delineate prognostic markers to stratify NSCLC patients at higher risk for developing brain metastasis (BM) and novel therapeutic candidates targeting BM, the patterns of copy number alterations (CNA) in primary lung NSCLC FFPE samples were analyzed by MIP technology.

Materials and methods

Pathology samples from NSCLS patients with brain metastases

Our study was reviewed and approved by the Institutional Review Board of the Samsung Medical Center (Seoul, Korea). All pathology samples and clinical data were obtained with written informed consent according to the institutional regulations. In the pathology database of the Samsung Medical Center (1994.12–2010.7), 160 NSCLC brain metastasis patients were identified. From them, 36 cases were selected; exclusion criteria, the biopsy of primary lung cancer without brain metastasis sample; inclusion criteria, harbouring visible tumors. A total of 37 formaldehyde-fixed and paraffin-embedded (FFPE) pathology samples were analyzed by this MIP analysis including 3 normal lung and 5 brain controls as follows: 20 ADCs of 8 paired cases (lung cancer and BM derived from same patient) and 4 only lung cancer cases; 9 SQCCs of 3 paired cases and 3 only lung cancers. Normal lung and brain tissues were obtained from the patients with other benign diseases.

All NSCLC patients were classified according to the standard World Health Organization (WHO) histological typing of lung carcinomas and the TNM (tumor-node-metastasis) staging system of the International Union Against Cancer (UICC). Clinicopathological data including age, gender, tumor stage and treatment history were obtained by a review of the medical records. Sites of distant metastasis and disease recurrence after treatment were traced by serial computed tomography (CT), magnetic resonance imaging (MRI) and positron emission tomography (PET). BM was defined as synchronous when metastasis was detected within 3 months of the initial diagnosis of primary tumor. The others were defined as metachronous.

Isolation of genomic DNA

Genomic DNA (gDNA) was extracted from three 5-μm-thick sections per FFPE block for each pathology sample. Deparaffinization and RNA extraction/purification was performed using a QIAamp DNA FFPE\Tissue kit (Qiagen) according the proposed protocol. The only change to the standard protocol was to increase the proteinase K digestion time (overnight). Extracted gDNA was stored at −20°C, and DNA quantity was analyzed by a Quant-iT Picogreen dsDNA kit (Invitrogen).

Molecular inversion probe (MIP) assay and data analysis

The MIP assay was performed using the OncoScan™ FFPE Express 330K MIP platform (Affymetrix). The MIP assay was performed as described previously (12,15). Briefly, gDNA samples (75 ng per each sample) were annealed with the 24,037 MIP probes (200 amol/μl per probe) in a 384-well plate on ice at 20°C for 4 minutes (min), at 95°C for 5 min, and then at 58°C overnight. The mixtures were circularized with the addition of 4 μl of the appropriate nucleotide at 58°C for 10 min (gap-fill ligation). Un-circularized MIP probes and gDNA were eliminated by addition of 4 μl of exonucleases (37°C for 15 min followed by heat inactivation). The circularized probes were linearized by restriction enzyme digest at 37°C for 15 min, and then amplified using a universal primer for 18 cycles [95°C for 20 seconds (sec), 64°C for 40 sec, and 72°C for 10 sec]. For the labeling reaction, the amplified products were further amplified with the label primers for 10 cycles. The MIP polymerase chain reaction (PCR) products were mixed with hybridization cocktail, denatured, and hybridized to 30K Universal Tag Arrays (Affymetrix) at 39°C for 16 h with two arrays for each allele. The hybridized arrays were washed on a standard Affymetrix fluidic station and stained with streptavidin-phycoerythrin (5 ng/ml, Invitrogen).

Copy number estimation was obtained from the barcode hybridization signals as described previously (12). The copy number changes of the NSCLCs were analyzed compared with the copy number of the normal non-neoplastic lung and brain samples. Data analysis including copy sum and copy contrast computation, allele ratio, sample normalization, data smoothing was performed by Nexus copy number analytics Version 5.1 Software (Biodiscovery), using algorithm SNP-FASST2 with sensitivity p<0.05.

Results

Detailed clinicopathological data of the 18 NSCLC patients with BM (12 ADCs and 6 SQCCs) are summarized in Table I. The median age of the NSCLC patients at the initial diagnosis of the primary cancers was 55 years (range 33–72 years). Synchronous (metastasis within 3 months of the initial diagnosis of primary NSCLC) and metachronous (metastasis after 3 months of the initial diagnosis of primary NSCLC) BM were 6 (33%) and 12 cases (67%), respectively. Median time between initial diagnosis of primary tumors and onset of overall BM was 257.5 days (8.6 months). The median lag (range) till BM for synchronous tumors was 0 (0–88) while for metachronous tumors the lag was 545 (180–2190) days.

Table I

Cliniopathological features of 18 non-small cell lung cancer (NSCLC) cases studied.

Table I

Cliniopathological features of 18 non-small cell lung cancer (NSCLC) cases studied.

Sample IDGender/agePattern of metastasisSubtypeTreatment for PTTreatment for BMExtracranial metastasisInterval between PT and BM (days)OSa (days)PFSb (days)KRAS mutationEGFR mutation
1M/62SADCLobectomy, RTTR, RT, GKRS (2)None0496110G12Vwt
2M/66SLobectomy, RTTR, RTNone010981098G12Dwt
3F/55SLobectomy, RTTR, RTNone015901590wtL858R
4M/33SNeoadjuvant CCRT, lobectomyTR (2), RT, GKRS (4)None8867070wtE746-A750 del
5F/55MLobectomy, CCRTTR, RTNone219099-wtwt
6F/66MLobectomy, RTTR, RTNone1230933271wtE746-A750 del
7F/55MLobectomy, RTTRAdrenal gland, liver124030-wtE746-A750 del
8F/46MLobectomy, CCRTTR, GKRS, RTT-spine78032-wtwt
9M/64MNeoadjuvant CCRT, lobectomyTR, GKRSNone1110922-wtwt
10M/51MLobectomy, RTTR, RTNone210254-wtwt
11M/45MLobectomyGKRS (2), TRNone310922-NANA
12M/56MLobectomy, CCRTTR, RT, GKRSNone830405-NANA
13M/67SSQCCLobectomyTRNone024-wtwt
14M/54SLobectomyTR (3), GKRS (4)None026331607wtwt
15F/47MLobectomy, CTx.TR, RTNone18069-wtwt
16M/72MLobectomy, CTx.TR, RTNone3101210-wtwt
16M/51MLobectomy, CCRTTR (2), RT, GKRS (2)None210470390wtwt
17F/52MLobectomy, CTx.TR, GKS (2), RTNone305136-wtwt

{ label (or @symbol) needed for fn[@id='tfn1-ijo-41-06-2013'] } S, synchronous; M, metachronous; PT, primary tumor; BM, brain metastasis; OS, overall survival; PFS, progression-free survival; aber, aberration; ADC, adenocarcinoma; SQCC, squamous cell carcinoma; CTx., chemotherapy; RT, radiation therapy; CCRT, concomitant chemo-radiation therapy; TR, tumor removal (times); GKRS, gamma knife radiosurgery (times); wt, wild-type; del, deletion; NA, not analyzed.

a OS, overall survival periods from the diagnosis of brain metastasis.

b PFS, progression-free survival periods from the diagnosis of brain metastasis.

Specific copy number alterations depending on distinct histologic-subtypes of non-small cell lung cancer

In this study, chromosomal aberration of 18 NSCLC lung samples (12 ADCs and 6 SQCCs) and 3 normal lung controls was analyzed by the MIP technology to identify chromosomal aberrations involved in the distinct pathogenesis. Common genomic aberrations were defined as continuous genomic regions where the average signal in tumors consistently and significantly differed from that of normal lung samples. Since there is growing evidence that lung ADC and SQCC have distinct oncogenic mutations and divergent therapeutic responses (16,17), 12 ADCs and 6 SQCCs were analyzed separately. A full view of the frequently detected significant chromosomal aberrations is presented in Fig. 1. The most frequent aberrations identified in primary ADCs (>40% of cases) were 3q, 5p, 5q, 6p, 8q, 9p, 11p, 15q and 17q for copy number gains and 10q and 22q for losses (Table II). On the other hand, primary SQCCs revealed frequent copy number gains in 4q and 12q and copy number loss in 9q (Table II). Previously, whole-genome array comparative genomic hybridization (aCGH) identified significantly different chromosomal signatures between two subtypes of the NSCLC (18). Compared with the previous results using different experimental techniques, similar patterns of CNAs were observed in this study, confirming the validity of the MIP technology (1922). However, several studies demonstrated additional CNAs including gains of 1q and losses of 9q and 10p in ADC and gains of 3q, 7p, 12p and 20q as well as losses of 2q, 3p, 16p and 17p in SQCC (21,2325).

Table II

Frequent regions of adenocarcinoma-specific and squamous cell carcinoma-specific copy number alteration.

Table II

Frequent regions of adenocarcinoma-specific and squamous cell carcinoma-specific copy number alteration.

NSCLC subtypeChromosomal aberrationRegion (start site-end site)Cytoband locationFrequency %P-value
Adenocarcinoma (n=12)CN gain chr3:173,271,977–173,427,105q26.3141.70.011
CN gain chr5:40,349,188–40,886,467p13.1750.018
CN gain chr5:150,438,972–150,523,583q33.1500.001
CN gain chr5:172,100,206–172,250,021q35.1–q35.2500.001
CN gain chr6:26,128,335–26,352,388p22.166.70.004
CN gain chr8:134,026,676–134,089,398q24.2258.30
CN gain chr9:43,997,314–44,825,192p11.2500
CN loss chr10:133,727,624–135,374,737q26.341.70.026
CN gain chr11:35,117,949–35,240,617p1341.70
CN gain chr15:83,849,162–83,995,470q25.341.70
CN gain chr17:45,586,978–45,621,111q21.33500.015
CN gain chr17:55,139,289–55,290,700q23.1500.015
CN loss chr22:47,207,744–47,732,937q13.32500
Squamous cell carcinoma (n=6)CN gain chr4:87,398,382–87,491,412q21.3500.028
CN loss chr9:139,183,759–140,273,252q34.366.70.042
CN gain chr12:74,190,395–74,668,717q21.266.70.026

a Regions were included if present in >50% of total adenocarcinoma-specific cases.

Continuous somatic evolution eventually giving rise to overt metastasis revealed the metastasis-specific related genes that mediate or impede metastatic progression without affecting primary malignancy (26). When patterns of CNAs in 11 BM (8 ADCs and 3 SQCCs) were compared with those of corresponding primary lung tumors, BM were found to carry the majority of genetic alterations present in the corresponding primary tumors (data not shown). However, in ADCs, BM harbored specific CNAs; new 11p and 15q gain (data not shown). In SQCCs, no BM specific CNAs were detected, likely because of the small number (n=3) of the SQCC cases. Therefore, BM specific CNAs were further analyzed in the ADCs.

Genetic signatures associated with early brain metastasis of lung adenocarcinoma

Although comparing the genetic alterations between primary and metastatic tumor would have functional implications, BM-associated genetic signatures in the primary lung ADC would have been more clinically relevant. Lung ADC is characterized by the early development of BM and the incidence of BM based on autopsy findings was as high as 50% in patients with lung ADC (8,27). Given that BM significantly worsens prognosis of lung ADC patients, lung ADC patients who are likely to develop BM need adjuvant treatments for BM. Primary lung ADCs with early development of BM (synchronous) would contain more CNAs predictive of metastatic potential or aggressive transformation. To uncover complex genetic ‘signatures’ that can predict the risk of BM, the copy number changes of 4 lung ADCs with synchronous BM were compared with those of 8 lung ADCs with metachronous BM (Table III and Fig. 2). Amplification in 5q35.1–2, 10q23.31, 17q23.3–24.1 and 17q24.1 was detected in 100, 75, 100 and 75% of the cases with synchronous BM, respectively, significantly more frequent than that of lung ADCs with metachronous BM (Table III). Differentially gained regions between primary ADCs with synchronous and metachronous BM were found to contain putative metastasis promoting genes, NeurL1B, ACTA2, FAS and ICAM2 (Table III).

Table III

The comparison of recurrent copy number alterations in primary lung adenocarcinomas derived from the patients with synchronous (n=4) and metachronous (n=8) brain metastasis.

Table III

The comparison of recurrent copy number alterations in primary lung adenocarcinomas derived from the patients with synchronous (n=4) and metachronous (n=8) brain metastasis.

EventChromosome cytobandStartEndRegion lengthGenesFrequency % synchronous BM (Total cases = 8)Frequency % BM metachronous (Total cases = 4)P-value
Gain5q35.117200853017210020691,676NeurL1B10012.50.01
5q35.217225002117227741527394ERGIC110012.50.01
10q23.31906827109073575353043ACTA27500.018
10q23.3190749267907510251758FAS7500.018
17q23.3594246195945500030381C17orf72, ICAM210000.002
17q24.1601558626020384147979LOC14688010012.50.01
17q24.1606351406066538230242RGS97500.018

a BM, brain metastasis.

Discussion

Although the BM affecting up to 25% of NSCLC during their lifetime negatively impacts survival, currently, there are no standard practice measures to reduce BM risk in NSCLC (28,29). Under the hypothesis that the genes residing in amplified or deleted regions in each subtype play an important role in the histology-specific pathogenesis of NSCLC, this study was designed to identify ‘meta-signatures’ that stratify NSCLC patients at higher risk for BM. The NSCLC harbours several histopathologically and molecularly distinct subtypes including ADC and SQCC. However, important molecular differences between primary lung ADC and SQCC have been identified, suggesting that future targeted therapies need to be histology-specific (8,16,17,3036). For example, KRAS and epidermal growth factor receptor (EGFR) gene mutations are found almost exclusively in ADCs rather than SQCCs. Therefore, primary lung ADCs were separately analyzed in this study.

Due to increasing incidence and substantial relapse rate, the pattern of CNAs was compared between primary ADCs derived from patients with synchronous and metachronous BM to find brain-specific meta-signature and putative targets associated with BM in ADC subtype. Several putative genes reported to be involved in tumorigenesis of various cancers were demonstrated in our study. For example, Neuralized-1B is the E3 ubiquitin ligase that is required for endocytosis regulating both the receptor and ligand side in Notch signaling (37,38). Relatively frequent deregulation of the Notch pathway and Notch ligand Jagged2/miR-200-dependent pathway in NSCLC indicates the significance and mechanisms underpinning Notch pathway activation in lung cancer (3941). In addition, Fas signalling exhibits tumor-promoting effects by increasing proliferation and invasiveness (4246). Fas can promote lung cancer growth by recruiting MDSC via cancer cell-derived PGE2 and signal for cell invasion via the glycogen synthase kinase 3β pathway (47,48). Human smooth muscle α-actin (SMA/ACTA2) has been used as one of mesenchymal cell-specific markers showing the epithelial-to-myofibroblast transition involving actin-skeleton remodeling and myogenic reprogramming. The appearance of dot-like α-SMA staining in cytokeratin positive cells may indicate the initial phase of the epithelial to mesenchymal transition (EMT) (4954). Finally, a recent study reported ICAM2 as a mediator for a survival signal sufficient to block apoptosis by activation of the PI3K/AKT pathway (55). As tumor progression and response to treatment are determined by numerous co-dependent prognostic factors, a multi-genetic approach to determining the optimal treatment for individual patients is more likely to be successful rather than a single prognostic biomarker.

Evidence that highly metastatic clones from primary tumor had a higher rate of genetic mutability than non-metastatic clones from the same tumor provided an early link between metastasis and intrinsic genetic instability such as mutations and chromosomal rearrangements (56). Although several high-throughput technologies have been developed to detect genomic CNAs in a variety of cancers until recently, high sensitivity to detect single copy number changes and the ability to test FFPE samples in which DNA is known to be degraded to different degrees is important (12). Analyzing FFPE samples allows the utilization of the vast majority of cancer samples since most cancer tissue samples are available as FFPE. For our FFPE NSCLC samples, the MIP technology was used, which has been validated to be able to substitute aCGH (1215,57,58). Because MIP uses probes that have a genomic footprint of ∼40 bp, this high density platform only requires small amounts of DNA and, consequently, is well-suited for the analysis of degraded DNA in FFPE tissues (59). The results of this study using the MIP technique showed differential genetic alteration between lung ADCs and SQCCs, correlating well with previous studies using aCGH. The similarities support the validity of the technique and the results from it. However, due to inadequate amount of extracted gDNA, quantitative-real-time-PCR (qRT-PCR) to validate copy number alterations identified by the MIP array could not be performed in our study.

In this study, we identified several genes associated with early BM of lung ADCs. Although our data suggested a new aspect of genomic alterations associated with BM of NSCLC, further biological studies to validate the role of identified candidates in brain-specific metastasis and testing the predictive power of our meta-signature in larger NSCLC samples would be required. In spite of several limitations of our study including very small number of NSCLC samples and absence of validation studies such as qRT-PCR, these genomic signatures may help to generate useful markers to refine prognosis and guide therapeutic decisions for improvement of prognosis and quality of life for patients with NSCLC.

Acknowledgements

This study was supported by a grant of the Korea Healthcare Technology R&D Project, Ministry for Health and Welfare Affairs, Republic of Korea (A092255).

References

1. 

Choi YW, Choi JS, Zheng LT, et al: Comparative genomic hybridization array analysis and real time PCR reveals genomic alterations in squamous cell carcinomas of the lung. Lung Cancer. 55:43–51. 2007. View Article : Google Scholar : PubMed/NCBI

2. 

Travis WD, Brambilla E, Muller-Hermelink HK and Harris CC: World Health Organization Classification of Tumors. Pathology and Genetics of the Lung, Pleura, Thymus and Heart. IARC Press; Lyon: 2004

3. 

Spira A and Ettinger DS: Multidisciplinary management of lung cancer. N Engl J Med. 350:379–392. 2004. View Article : Google Scholar : PubMed/NCBI

4. 

Salgia R, Hensing T, Campbell N, et al: Personalized treatment of lung cancer. Semin Oncol. 38:274–283. 2011. View Article : Google Scholar : PubMed/NCBI

5. 

Parkin DM, Bray FI and Devesa SS: Cancer burden in the year 2000. The global picture. Eur J Cancer. 37(Suppl 8): S4–S66. 2001.PubMed/NCBI

6. 

Jemal A, Siegel R, Ward E, Hao Y, Xu J and Thun MJ: Cancer statistics, 2009. CA Cancer J Clin. 59:225–249. 2009. View Article : Google Scholar

7. 

Moran C: Importance of molecular features of non-small cell lung cancer for choice of treatment. Am J Pathol. 178:1940–1948. 2011. View Article : Google Scholar : PubMed/NCBI

8. 

Hoffman PC, Mauer AM and Vokes EE: Lung cancer. Lancet. 355:479–485. 2000. View Article : Google Scholar : PubMed/NCBI

9. 

Hess KR, Varadhachary GR, Taylor SH, et al: Metastatic patterns in adenocarcinoma. Cancer. 106:1624–1633. 2006. View Article : Google Scholar : PubMed/NCBI

10. 

Feld R, Rubinstein LV and Weisenberger TH: Sites of recurrence in resected stage I non-small-cell lung cancer: a guide for future studies. J Clin Oncol. 2:1352–1358. 1984.PubMed/NCBI

11. 

Bernards R and Weinberg RA: A progression puzzle. Nature. 418:8232002. View Article : Google Scholar : PubMed/NCBI

12. 

Wang Y, Moorhead M, Karlin-Neumann G, et al: Allele quantification using molecular inversion probes (MIP). Nucleic Acids Res. 33:e1832005. View Article : Google Scholar : PubMed/NCBI

13. 

Johnson CE, Gorringe KL, Thompson ER, et al: Identification of copy number alterations associated with the progression of DCIS to invasive ductal carcinoma. Breast Cancer Res Treat. 133:889–898. 2011. View Article : Google Scholar : PubMed/NCBI

14. 

Schiffman JD, Wang Y, McPherson LA, et al: Molecular inversion probes reveal patterns of 9p21 deletion and copy number aberrations in childhood leukemia. Cancer Genet Cytogenet. 193:9–18. 2009. View Article : Google Scholar : PubMed/NCBI

15. 

Wang Y, Moorhead M, Karlin-Neumann G, et al: Analysis of molecular inversion probe performance for allele copy number determination. Genome Biol. 8:R2462007. View Article : Google Scholar : PubMed/NCBI

16. 

Travis WD and Rekhtman N: Pathological diagnosis and classification of lung cancer in small biopsies and cytology: strategic management of tissue for molecular testing. Semin Respir Crit Care Med. 32:22–31. 2011. View Article : Google Scholar : PubMed/NCBI

17. 

Travis WD, Rekhtman N, Riley GJ, et al: Pathologic diagnosis of advanced lung cancer based on small biopsies and cytology: a paradigm shift. J Thorac Oncol. 5:411–414. 2010. View Article : Google Scholar : PubMed/NCBI

18. 

Kang JU, Koo SH, Kwon KC, Park JW and Kim JM: Identification of novel candidate target genes, including EPHB3, MASP1 and SST at 3q26.2–q29 in squamous cell carcinoma of the lung. BMC Cancer. 9:2372009.PubMed/NCBI

19. 

Yokoi S, Yasui K, Iizasa T, Imoto I, Fujisawa T and Inazawa J: TERC identified as a probable target within the 3q26 amplicon that is detected frequently in non-small cell lung cancers. Clin Cancer Res. 9:4705–4713. 2003.PubMed/NCBI

20. 

Ubagai T, Matsuura S, Tauchi H, Itou K and Komatsu K: Comparative genomic hybridization analysis suggests a gain of chromosome 7p associated with lymph node metastasis in non-small cell lung cancer. Oncol Rep. 8:83–88. 2001.PubMed/NCBI

21. 

Kang JU, KS, Kwon KC, Park JW, Shin SY, Kim JM and Jung SS: High frequency of genetic alterations in non-small cell lung cancer detected by multi-target fluorescence in situ hybridization. J Korean Med Sci. 22:47–51. 2007. View Article : Google Scholar : PubMed/NCBI

22. 

Dehan E, Ben-Dor A, Liao W, et al: Chromosomal aberrations and gene expression profiles in non-small cell lung cancer. Lung Cancer. 56:175–184. 2007. View Article : Google Scholar : PubMed/NCBI

23. 

Chujo M, Noguchi T, Miura T, Arinaga M, Uchida Y and Tagawa Y: Comparative genomic hybridization analysis detected frequent overrepresentation of chromosome 3q in squamous cell carcinoma of the lung. Lung Cancer. 38:23–29. 2002. View Article : Google Scholar : PubMed/NCBI

24. 

Kang JU, Koo SH, Kwon KC, Park JW and Jung SS: Gain of the EGFR gene located on 7p12 is a frequent and early event in squamous cell carcinoma of the lung. Cancer Genet Cytogenet. 184:31–37. 2008. View Article : Google Scholar : PubMed/NCBI

25. 

Massion PP, Kuo WL, Stokoe D, et al: Genomic copy number analysis of non-small cell lung cancer using array comparative genomic hybridization: implications of the phosphatidylinositol 3-kinase pathway. Cancer Res. 62:3636–3640. 2002.

26. 

Kikuchi T, Daigo Y, Ishikawa N, et al: Expression profiles of metastatic brain tumor from lung adenocarcinomas on cDNA microarray. Int J Oncol. 28:799–805. 2006.PubMed/NCBI

27. 

Robnett TJ, Machtay M, Stevenson JP, Algazy KM and Hahn SM: Factors affecting the risk of brain metastases after definitive chemo-radiation for locally advanced non-small-cell lung carcinoma. J Clin Oncol. 19:1344–1349. 2001.PubMed/NCBI

28. 

Grinberg-Rashi H, Ofek E, Perelman M, et al: The expression of three genes in primary non-small cell lung cancer is associated with metastatic spread to the brain. Clin Cancer Res. 15:1755–1761. 2009. View Article : Google Scholar : PubMed/NCBI

29. 

Oh Y, Taylor S, Bekele BN, et al: Number of metastatic sites is a strong predictor of survival in patients with nonsmall cell lung cancer with or without brain metastases. Cancer. 115:2930–2938. 2009. View Article : Google Scholar : PubMed/NCBI

30. 

Tam IY, Chung LP, Suen WS, et al: Distinct epidermal growth factor receptor and KRAS mutation patterns in non-small cell lung cancer patients with different tobacco exposure and clinicopathologic features. Clin Cancer Res. 12:1647–1653. 2006. View Article : Google Scholar : PubMed/NCBI

31. 

Mok TS, Wu YL, Thongprasert S, et al: Gefitinib or carboplatinpaclitaxel in pulmonary adenocarcinoma. N Engl J Med. 361:947–957. 2009. View Article : Google Scholar : PubMed/NCBI

32. 

Shaw AT, Yeap BY, Mino-Kenudson M, et al: Clinical features and outcome of patients with non-small-cell lung cancer who harbor EML4-ALK. J Clin Oncol. 27:4247–4253. 2009. View Article : Google Scholar : PubMed/NCBI

33. 

Scagliotti G, Brodowicz T, Shepherd FA, et al: Treatment-byhistology interaction analyses in three phase III trials show superiority of pemetrexed in nonsquamous non-small cell lung cancer. J Thorac Oncol. 6:64–70. 2011.PubMed/NCBI

34. 

Chansky K, Sculier JP, Crowley JJ, Giroux D, Van Meerbeeck J and Goldstraw P: The International Association for the Study of Lung Cancer Staging Project: prognostic factors and pathologic TNM stage in surgically managed non-small cell lung cancer. J Thorac Oncol. 4:792–801. 2009. View Article : Google Scholar

35. 

Pisters KM, Evans WK, Azzoli CG, et al: Cancer Care Ontario and American Society of Clinical Oncology adjuvant chemotherapy and adjuvant radiation therapy for stages I–IIIA resectable non-small-cell lung cancer guideline. J Clin Oncol. 25:5506–5518. 2007.PubMed/NCBI

36. 

Bass AJ, Watanabe H, Mermel CH, et al: SOX2 is an amplified lineage-survival oncogene in lung and esophageal squamous cell carcinomas. Nat Genet. 41:1238–1242. 2009. View Article : Google Scholar : PubMed/NCBI

37. 

Rullinkov G, Tamme R, Sarapuu A, et al: Neuralized-2: expression in human and rodents and interaction with Delta-like ligands. Biochem Biophys Res Commun. 389:420–425. 2009. View Article : Google Scholar : PubMed/NCBI

38. 

Song R, Koo BK, Yoon KJ, et al: Neuralized-2 regulates a Notch ligand in cooperation with Mind bomb-1. J Biol Chem. 281:36391–36400. 2006. View Article : Google Scholar : PubMed/NCBI

39. 

Collins BJ, Kleeberger W and Ball DW: Notch in lung development and lung cancer. Semin Cancer Biol. 14:357–364. 2004. View Article : Google Scholar : PubMed/NCBI

40. 

Westhoff B, Colaluca IN, D’Ario G, et al: Alterations of the Notch pathway in lung cancer. Proc Natl Acad Sci USA. 106:22293–22298. 2009. View Article : Google Scholar : PubMed/NCBI

41. 

Yang Y, Ahn YH, Gibbons DL, et al: The Notch ligand Jagged2 promotes lung adenocarcinoma metastasis through a miR-200-dependent pathway in mice. J Clin Invest. 121:1373–1385. 2011. View Article : Google Scholar : PubMed/NCBI

42. 

Peter ME, Budd RC, Desbarats J, et al: The CD95 receptor: apoptosis revisited. Cell. 129:447–450. 2007. View Article : Google Scholar : PubMed/NCBI

43. 

Peter ME, Legembre P and Barnhart BC: Does CD95 have tumor promoting activities? Biochim Biophys Acta. 1755:25–36. 2005.PubMed/NCBI

44. 

Mitsiades CS, Poulaki V, Fanourakis G, et al: Fas signaling in thyroid carcinomas is diverted from apoptosis to proliferation. Clin Cancer Res. 12:3705–3712. 2006. View Article : Google Scholar : PubMed/NCBI

45. 

Barnhart BC, Legembre P, Pietras E, Bubici C, Franzoso G and Peter ME: CD95 ligand induces motility and invasiveness of apoptosis-resistant tumor cells. EMBO J. 23:3175–3185. 2004. View Article : Google Scholar : PubMed/NCBI

46. 

Chen L, Park SM, Tumanov AV, et al: CD95 promotes tumour growth. Nature. 465:492–496. 2010. View Article : Google Scholar : PubMed/NCBI

47. 

Zhang Y, Liu Q, Zhang M, Yu Y, Liu X and Cao X: Fas signal promotes lung cancer growth by recruiting myeloid-derived suppressor cells via cancer cell-derived PGE2. J Immunol. 182:3801–3808. 2009. View Article : Google Scholar : PubMed/NCBI

48. 

Kleber S, Sancho-Martinez I, Wiestler B, et al: Yes and PI3K bind CD95 to signal invasion of glioblastoma. Cancer Cell. 13:235–248. 2008. View Article : Google Scholar : PubMed/NCBI

49. 

Masszi A, Speight P, Charbonney E, et al: Fate-determining mechanisms in epithelial-myofibroblast transition: major inhibitory role for Smad3. J Cell Biol. 188:383–399. 2010. View Article : Google Scholar : PubMed/NCBI

50. 

Wang J, Zohar R and McCulloch CA: Multiple roles of alpha-smooth muscle actin in mechanotransduction. Exp Cell Res. 312:205–214. 2006. View Article : Google Scholar : PubMed/NCBI

51. 

Han M, Liu M, Wang Y, et al: Re-expression of miR-21 contributes to migration and invasion by inducing epithelial-mesenchymal transition consistent with cancer stem cell characteristics in MCF-7 cells. Mol Cell Biochem. 363:427–436. 2012. View Article : Google Scholar : PubMed/NCBI

52. 

Owens GK and Thompson MM: Developmental changes in isoactin expression in rat aortic smooth muscle cells in vivo. Relationship between growth and cytodifferentiation. J Biol Chem. 261:13373–13380. 1986.PubMed/NCBI

53. 

Pirozzi G, Tirino V, Camerlingo R, et al: Epithelial to mesenchymal transition by TGFbeta-1 induction increases stemness characteristics in primary non-small cell lung cancer cell line. PLoS One. 6:e215482011. View Article : Google Scholar : PubMed/NCBI

54. 

Valcz G, Sipos F, Krenacs T, et al: Increase of alpha-SMA(+) and CK (+) cells as an early sign of epithelial-mesenchymal transition during colorectal carcinogenesis. Pathol Oncol Res. 18:371–376. 2012.

55. 

Perez OD, Kinoshita S, Hitoshi Y, et al: Activation of the PKB/AKT pathway by ICAM-2. Immunity. 16:51–65. 2002. View Article : Google Scholar : PubMed/NCBI

56. 

Fidler IJ: The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited. Nat Rev Cancer. 3:453–458. 2003.

57. 

Ji H, Kumm J, Zhang M, et al: Molecular inversion probe analysis of gene copy alterations reveals distinct categories of colorectal carcinoma. Cancer Res. 66:7910–7919. 2006. View Article : Google Scholar : PubMed/NCBI

58. 

Lapuk A, Volik S, Vincent R, et al: Computational BAC clone contig assembly for comprehensive genome analysis. Genes Chromosomes Cancer. 40:66–71. 2004. View Article : Google Scholar : PubMed/NCBI

59. 

Wang Y, Carlton VE, Karlin-Neumann G, et al: High quality copy number and genotype data from FFPE samples using Molecular Inversion Probe (MIP) microarrays. BMC Med Genomics. 2:82009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December 2012
Volume 41 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lee HW, Seol HJ, Choi Y, Ju HJ, Joo KM, Ko Y, Lee J and Nam D: Genomic copy number alterations associated with the early brain metastasis of non-small cell lung cancer. Int J Oncol 41: 2013-2020, 2012
APA
Lee, H.W., Seol, H.J., Choi, Y., Ju, H.J., Joo, K.M., Ko, Y. ... Nam, D. (2012). Genomic copy number alterations associated with the early brain metastasis of non-small cell lung cancer. International Journal of Oncology, 41, 2013-2020. https://doi.org/10.3892/ijo.2012.1663
MLA
Lee, H. W., Seol, H. J., Choi, Y., Ju, H. J., Joo, K. M., Ko, Y., Lee, J., Nam, D."Genomic copy number alterations associated with the early brain metastasis of non-small cell lung cancer". International Journal of Oncology 41.6 (2012): 2013-2020.
Chicago
Lee, H. W., Seol, H. J., Choi, Y., Ju, H. J., Joo, K. M., Ko, Y., Lee, J., Nam, D."Genomic copy number alterations associated with the early brain metastasis of non-small cell lung cancer". International Journal of Oncology 41, no. 6 (2012): 2013-2020. https://doi.org/10.3892/ijo.2012.1663