Open access

The Investigation of Gene Regulation and Variation in Human Cancers and Other Diseases

Written By

Shihori Tanabe and Sun Ha Jee

Submitted: 19 March 2012 Published: 12 December 2012

DOI: 10.5772/51271

From the Edited Volume

Latest Research into Quality Control

Edited by Isin Akyar

Chapter metrics overview

2,068 Chapter Downloads

View Full Metrics

1. Introduction

Dynamic regulation of genes is an important part of the cell life cycle in health and disease. The regulation includes the variety and alteration of genome and gene expression, and the concept such as quality of genome will be useful to predict and assess the developmental stages of the cells, disease status and drug sensitivity. Recent technologies and worldwide sequencing projects have revealed 26,383 annotated genes in the 2.91-Gigabase human genome [1,2]. The main molecular functions of the annotated genes, as categorized by Gene Ontology (GO), are enzyme, signal transduction, nucleic acid binding, cell adhesion, chaperone, cytoskeletal structural protein, extracellular matrix, immunoglobulin, ion channel, motor, structural protein of muscle, protooncogene, select calcium binding protein, intracellular transporter, and transporter [1,3]. Despite a wealth of knowledge, the function of 42% of the annotated genes remains unknown [1]. When the human genome sequence was published in 2001 [1], there were a predicted 39,114 genes, of which 59% were of unknown function. According to the International Human Genome Sequencing Consortium, the number of identified genes is approximately 32,000, of which 51% show a match within InterPro, a database that integrates diverse information about protein families, domains, and functional sites [2-5]. In 2001, InterPro combined sequence and pattern information from four databases (PRINTS, PROSITE, Pfam, Prosite Profile); however, it now includes information from an additional eight databases (SMART, ProDom, PIRSF, SUPERFAMILY, PANTHER, CATH-Gene3D, TIGRFAM, and HAMAP) [2,4-16]. In [2], the InterPro entries are collapsed into 12 broad categories: cellular processes, metabolism, DNA replication/modification, transcription/translation, intracellular signaling, cell–cell communication, protein folding and degradation, transport, multifunctional proteins, cytoskeletal/structural, defense and immunity, and miscellaneous function. The rate of single nucleotide polymorphism (SNP) variation has been reported as 1 in 1250 base pairs [1] and more than 1.4 million SNPs have been identified [2] (Table 1).

Size of the genome 2.91 Gbp [1]
Number of annotated genes 26,383 [1]
Main molecular functions of
annotated genes
enzyme, signal transduction, nucleic acid binding, cell adhesion, chaperone, cytoskeletal structural protein, extracellular matrix, immunoglobulin, ion channel, motor, structural protein of muscle, protooncogene, select calcium binding protein, intracellular transporter, transporter [1]
Percentage of annotated genes
with unknown function
42% [1]
Number of hypothetical and
annotated genes
39,114 [1]
Percentage of hypothetical and
annotated genes with unknown function
59% [1]
Number of identified genes approx. 32,000 [2]
Percentage of matches with
InterPro
51% [2]
Rate of SNP variation 1/1250 bp [1]
SNPs identified more than 1.4 million [2]

Table 1.

Genomic and gene characteristics revealed by the Human Genome Project.

Among the databases combined in InterPro (Table 2), PRINTS, PROSITE, and Pfam contain protein families in which the homology between each protein is predicted by the degree of sequence similarity [8]. The others—SMART, ProDom, PIRSF, SUPERFAMILY, PANTHER, CATH-Gene3D, TIGRFAM, and HAMAP [4-16]—have unique characteristics and URLs, and have been developed sharing information among each other and incorporating information from GO. In detail, PRINTS is a collection of diagnostic protein family “fingerprints”, which are groups of conserved motifs, evident in multiple sequence alignments [6]; PROSITE is a protein domain database for functional characterization and annotation that consists of documentation entries describing protein domains, families, and functional sites as well as associated patterns and profiles to identify them [7]; Pfam contains collections of protein families, each represented by multiple sequence alignments and hidden Markov models, available via servers in the UK, the USA, and Sweden [8]; SMART (Simple Modular Architecture Research Tool) is an online resource for the identification and annotation of protein domains and the analysis of protein domain architectures [9]; ProDom is a comprehensive set of protein domain families generated automatically from the UniProt database [10]; PIRSF is a classification system that reflects evolutionary relationships among full-length proteins and domains [11]; SUPERFAMILY is a database of structural and functional annotation for all proteins and genomes [12]; PANTHER is a classification system that classifies genes by their functions using published experimental evidence and evolutionary relationships to predict function even in the absence of direct experimental evidence [13]; CATH-Gene3D is a comprehensive database of protein domain assignments for sequences from the major sequence databases [14]; TIGRFAM is a collection of protein family definitions built to aid high-throughput annotation of specific protein functions [15]; and HAMAP is composed of two databases: the proteome database and the family database, and of an automatic annotation pipeline mainly focused on microbial proteomes [16]. Hidden Markov models are usually used for the database algorithm.

Database Name Context URL Reference
InterPro integrative predictive models of protein families, domain and functional sites of multiple databases such as PRINTS, PROSITE, Pfam, SMART, ProDom,
PIRSF, SUPERFAMILY, PANTHER, CATH-Gene3D, TIGRFAM, and HAMAP
http://www.ebi.ac.uk/interpro/ [4], [5]
PRINTS a collection of diagnostic protein family
"fingerprints" which are groups of conserved motifs, evident in multiple sequence alignments
http://www.bioinf.manchester.ac.uk/dbbrowser/PRINTS/index.php [6]
PROSITE a protein domain database for functional characterization and annotation which consists of documentation entries describing protein domains, families and functional sites as well as associated patterns and profiles to identify them http://prosite.expasy.org/ [7]
Pfam a database of collection of protein families, each represented by multiple sequence alignments and hidden Markov models, available via servers in the
UK, the USA and Sweden
http://pfam.sanger.ac.uk/http://pfam.janelia.org/http://pfam.sbc.su.se/ [8]
SMART an online resource for the identification and annotation of protein domains and the analysis of protein domain architectures, of which abbreviation
is Simple Modular Architecture Research Tool
http://smart.embl.de/ [9]
ProDom a comprehensive set of protein domain families automatically generated from the uniProt
knowledge Database
http://prodom.prabi.fr/prodom/current/html/home.php [10]
PIRSF the classification system which reflects evolutionary relationships of full-length proteins and domains http://pir.georgetown.edu/pirsf/ [11]
SUPERFAMILY a database of structural and functional annotation
for all proteins and genomes
http://supfam.org/SUPERFAMILY/ [12]
PANTHER the classification system which classifies genes by
their functions using published scientific experimental evidence and evolutionary relationships to predict function even in the absence of direct experimental evidence
http://www.pantherdb.org/ [13]
CATH-Gene3D a comprehensive database of protein domain assignments for sequences from the major sequence databases http://gene3d.biochem.ucl.ac.uk/ [14]
TIGRFAM a collection of protein family definitions built to aid
in high-throughput annotation of specific protein functions
http://www.jcvi.org/cgi-bin/tigrfams/index.cgi [15]
HAMAP a system which composed of two databases, the proteome database and the family database, and of an automatic annotation pipeline http://hamap.expasy.org/ [16]

Table 2.

Database information.

Advertisement

2. Gene regulation

2.1. Gene markers for cancer and cancer stem cells

Several molecular markers of cancer have been identified [17]. Metastatic cancer cells can transfer into bodily fluids through the cellular epithelia, which enables the detection of cancer markers in bodily fluids such as blood plasma, urine, or saliva [17]. The different types of cancer markers include genomic DNA point mutations, microsatellite alterations, promoter hypermethylation, viral sequences, aberrant chromosomal copy number, chromosomal translocations, deletions, or loss of heterozygosity, telomere extension, alterations in RNA or protein expression, and mitochondrial DNA mutations [17].

Molecular markers of cancer include TP53 (encoding p53), which has been shown to be mutated in head and neck, lung, colon, pancreatic, and bladder cancer [17,18]; colon, lung, esophagus, breast, liver, brain, reticuloendothelial tissue, and hematopoietic tissue cancers [19]; and bladder cancer [20]. Mutation of the epidermal growth factor receptor (EGFR) gene is an important predictive/prognostic factor for EGFR-tyrosine kinase inhibitor therapy in non-small cell lung cancer [21]. RAS oncogene mutations have been identified in colorectal tumors [22]. Microsatellites, which are tandem iterations of simple di-, tri-, or tetranucleotide repeats, have been reported to be unstable in some inherited diseases and in some types of cancer [23], including head and neck, lung, breast, and bladder cancer [17,23].

The expression levels of the cell cycle-related proteins p21 (CDKN1A), p53 (TP53), cyclin D1 (CCND1), and aurora kinase A (AURKA) may be used as prognostic markers to predict recurrence in stage II and stage III colon cancer [24]. In addition, markers of the epithelial–mesenchymal transition (EMT)–such as reduced expression of keratins, a switch from E-Cadherin to N-Cadherin, and enhanced migration in D492M cells—might be a useful marker in breast cancer [25]. Furthermore, expression of the stem cell markers cytokeratins 15 and 19 was altered in squamous cell carcinoma: cytokeratin 15 levels were decreased and the localization of cytokeratin 19 was altered [26]. KLK3, which encodes prostate-specific antigen, a member of the kallikrein family of serine proteases, is a biomarker for prostate cancer detection and disease monitoring [27,28]. Mitochondrial DNA mutations have been associated with bladder, head and neck, lung, colorectal, and pancreatic cancer [29-32] (Table 3).

Highly parallel identification of cancer-related genes using small hairpin RNA screening has revealed that the expression of known and putative oncogenes, such as EGFR, KRAS, MYC, BCR-ABL, MYB, CRKL, and CDK4 that are essential for cancer proliferation, is altered in cancer cells [33]. Other genes such as PTPN1, NF1, SMARCB1, and SMARCE1 have been identified as essential for the imatinib response of leukemia cells, and TOPOIIA expression is involved in resistance to etoposide, an anti-topoisomerase II agent, in small cell lung cancer [33-36].

Marker Cancer Type Reference
TP53 mutation

head and neck cancer [18]
bladder cancer [20]
lung cancer (small cell lung cancer and non-small cell lung cancer); breast, colon, esophagus, liver, bladder, ovary, and brain cancers; sarcomas, lymphomas, and leukemias [19]
EGFR mutation non-small cell lung cancer [21]
RAS mutation colorectal tumors [22]
DNA microsatellite alterations bladder cancer [23]
alteration in cell cycle mRNA
expression
colon cancer [24]
alteration in cytokeratin mRNA
expression
squamous cell carcinoma [26]
alteration in kallikrein mRNA
expression
prostate cancer [27]
mitochondrial DNA mutations
bladder cancer, head and neck cancer, lung cancer [29]
colorectal tumors [30], [32]
pancreatic cancer [31]

Table 3.

Genomic markers of cancer.

2.2. Genes related to cell proliferation

Cyclins, which regulate the cell cycle, play important roles in cell proliferation and the uncontrolled cell proliferation that is the most important factor in tumorigenesis [37]. Tumor cells accumulate mutations that result in constitutive mitogenic signaling and defective responses to anti-mitogenic signals that contribute to unscheduled proliferation [38]. In cancer, unscheduled proliferation, genomic instability, and chromosomal instability are the three major factors in cell cycle dysregulation [38]. Regulation of the cell cycle is mainly conducted by complexes of cyclins and cyclin-dependent kinases [38]. Cyclin D1 in cell migration and proliferation is temporo-spatially separated by its biphasic expression induced by thrombin, a G protein-coupled receptor agonist, which is mediated by nuclear factor of activated T cells c1 (NFATC1) and signal transducer and activator of transcription 3 (STAT3) [39]. Cyclin D1 regulates kinase activity and the G1–S phase transition in the cell cycle; deregulated cyclin D1 expression is well documented in breast, colon, and prostate cancers [39,40]. The expression of cyclin D1 is regulated by several factors including cytokines such as interleukin 3 and interleukin 6 via STAT3 and STAT5, or extracellular matrix factors such as collagen, fibronectin, and vitronectin, which activate focal adhesion kinase upon integrin clustering, and hepatocyte nuclear factor 6 [41]. Cyclin D1 is a crucial regulator of Wnt- and Notch-regulated development [41,42]. The binding of Wnt to its receptor, Frizzled, causes release of β-catenin to translocate from the cytoplasm to the nucleus, where it forms a complex with the ternary complex factor and/or the lymphoid enhancer-binding factor [41,43]. Cyclin D1 is induced by overexpression of β-catenin, which is a major component of adherens junctions that link the actin cytoskeleton to members of the cadherin family of transmembrane cell–cell adhesion receptors. It plays an important role in linking the cytoplasmic side of cadherin-mediated cell–cell contacts to the actin cytoskeleton [43]. Beta-catenin is upregulated in colorectal cancer, which is considered to trigger cyclin D1 gene expression followed by uncontrolled progression of the cell cycle [43]. In addition, β-catenin plays another role in signaling that involves transactivation, in complex with transcription factors of the lymphoid enhancing factor family in the nucleus [43]. The pathway involving β-catenin/LEF1 and elevation of cyclin D1 might be crucial for tumorigenesis [43]. Inhibiting EglN2, a member of the EglN (also called PHD or HPH) family of prolyl hydroxylases that regulates the heterodimeric transcription factor hypoxia-inducible factor (HIF), causes a decrease in the expression of its interaction partner cyclin D1 in cancer cells and impairs the cells’ ability to proliferate in vivo [44].

Progression of the eukaryotic cell cycle is driven by cyclin-dependent protein kinases (CDKs), which are binding partner of cyclins. The CDK oscillator acts as the primary organizer of the cell cycle [45]. Phosphorylation of cyclin-Cdk complexes is one of the primary mechanisms of cell cycle regulation [46]. Cyclins are degraded by ubiquitin-mediated proteolysis [46]. The ubiquitylation and degradation of cyclin 1 and cyclin 2 are mediated by the SCF complex, a multi-subunit ubiquitin ligase that contains Skp1, a member of the cullin family (Cdc53) and an F-box protein, as well as a RING-finger-containing protein [46]. CDKs including CDK1, CDK2, CDK4, CDK6, and CDK11 have various functions that have been investigated using loss-of-function, target validation, and gain-of-function mouse models [38]. CDK1 is a mitotic CDK, also known as cell division control protein 2 (CDC2). It is one of the master regulators of mitosis as it controls the centrosome cycle as well as mitotic onset; deficiency in CDK1 results in embryonic lethality in the first cell divisions [38,47]. CDK2, CDK4, and CDK6 are interphase CDKs that are not essential for the mammalian cell cycle; they are, however, required for the proliferation of specific cell types [38]. Deficiency in CDK2, CDK4, and CDK6 caused mid-gestation embryonic lethality because of hematopoietic defects [38,47].

2.3. Genes related to cell differentiation

Inhibitor of differentiation 1 (Id1) is associated with the induction of cell proliferation and invasion [48], as well as the invasive features of cancer and the EMT [48]. The HOX genes encode homeodomain-containing transcription factors involved in the regulation of cellular proliferation and differentiation during embryogenesis [49]. The expression of HOXA1, which plays an important role in proliferation, apoptosis, adhesion, invasion, the EMT, and anchorage-independent growth, was significantly increased in oral squamous cell carcinoma compared with in healthy oral mucosa [49], and it might be a useful prognostic marker for patients with this disease [49].

Wnt/β-catenin signaling controls skeletal development and differentiation [50]. The initiating step of skeletal development is mesenchymal condensation, during which mesenchymal progenitor cells are at least bipotentiate [50]. Osteochondral progenitor cells differentiate into osteoblasts instead of chondrocytes when Wnt/β-catenin signaling is activated [50]. In vitro models using human pluripotent stem cell-derived neural progenitor cells have been used to examine whether G11778A-mutated mitochondrial DNA, which is associated with Leber’s hereditary optic neuropathy, might be involved in the differentiation of neural progenitor cells into neurons, oligodendrocytes, and astrocytes [51]. The differentiation of neural progenitor cells can be visualized by staining for the neuronal marker class III beta-tubulin [51]. Alternative splicing of exons play an important role in cellular differentiation and pathogenesis [52]. Alternative splicing in colorectal cancer and renal cell cancer samples has been analyzed by the Bioinformatics Exon Array Tool (BEAT, http://beat.ba.itb.cnr.it/) using an Affymetrix GeneChip Exon Array [52]. When the dataset was analyzed using GO terms, the cell differentiation (GO:0030154)-related gene delta-like 1 (Drosophila) (DLL1) was found to be involved in colorectal cancer [52].

2.4. Genes related to apoptosis

Cell proliferation and death are regulated by various molecules. Recently, microRNAs have been revealed to play important roles during death receptor-mediated apoptosis (programmed cell death) [53]. Transfection with miR-133b caused a proapoptotic effect on tumor necrosis factor alpha (TNFα)-stimulated HeLa cells [53]: the expression of apoptosis regulatory proteins such as transgelin 2 (TAGLN2), myosin, heavy chain 9, non-muscle (MYH9), cytoskeleton-associated protein 4 (CKAP4), polypyrimidine tract binding protein 1 (PTBP1), glutathione-S-transferase pi 1 (GSTP1), and copine III (CPNE3) were down-regulated compared with in control cells [53]. The BCL protein family plays a major role in regulation of the apoptotic cascade [54]. BCL2-associated protein (BAX) promotes apoptosis and delays disease progression, and has been associated with longer disease-free survival in patients with a number of gastrointestinal cancers, such as esophageal, stomach, small intestine, and colon cancer; moreover, high BCL6 expression is correlated with worse prognosis in patients with other gastrointestinal tumors, such as esophageal adenocarcinoma [54]. There are two major cell death pathways that transduce the effects of various death inducers: the extrinsic death pathway that is mediated through cell death receptors of the TNF receptor family, such as the Fas receptor; and the intrinsic death pathway that proceeds through mitochondria [55]. The expression of apoptosis signal-regulating kinase (ASK1), which plays an important role as a mitogen-activated protein kinase kinase kinase in apoptosis signaling, is increased in gastric cancer [56]. Furthermore, the levels of cyclin D1 and phosphorylated JNK were higher in gastric cancer than in non-tumor epithelium [56]. ASK1 may play a role in the development of gastric cancer [56].

2.5. Detection of cell proliferation or apoptosis

Several methods have been suggested for the diagnosis of cancer [57]. Protein markers for cancer include prostate-specific antigen for prostate cancer, CA125 for ovarian cancer, carcinoembryonic antigen for colon cancer, human chorionic gonadotropin for trophoblastic cancer, and a-fetoprotein for hepatocellular carcinoma and germ cell tumors [57]. Assays to detect telomerase activity in clinical samples include the TRAP (telomere repeat amplification protocol) assay, which involves protein extraction and subsequent primer-directed PCR amplification of telomere extensions [57].

Assays for the detection of kinases that regulate cell growth, proliferation, differentiation, and metabolism have been developed [58]. The assay technology includes fluorescence polarization to detect protein phosphorylation, scintillation proximity to detect protein dephosphorylation by phosphatases, fluorescence resonance energy transfer to detect protein cleavage or modification, immunosorbent assays to detect phosphorylation state, luciferase-based ATP detection to detect the kinase-dependent depletion of ATP, luminescent oxygen channeling to detect phosphorylation, time-resolved fluorescence resonance energy transfer to detect phosphopeptide formation, and enzyme fragment complementation to detect molecular interactions with kinases [58,59]. Cell proliferation can also be determined by the tetrazolium hydroxide (XTT) cell proliferation assay, in which absorbance is measured by an ELISA reader under 490-nm-wavelength light (Biological Industries) [60].

Cell proliferation assays and apoptosis assays have been used to examine the effects of inhibitors on cancer cells [61]. The cell proliferation of Neuro-2A cells, neuroblastoma cells, can be determined using the CellTiter 96 Aqueous Non-Radioactive Cell Proliferation Assay reagent (Promega) [61]. A colony formation assay using Neuro-2A cells was used to determine the effect of an inhibitor of GSK-3β [61]. In this experiment, colonies were allowed to form for 10 days, after which the cells were fixed with 70% ethanol and stained with 1% methylene blue. Apoptosis was then measured by flow cytometry using an Annexin V-allophycocyanin (APC) /propidium iodide (PI) detection kit (BD PharMingen) [61]. Apoptosis was also determined using 4’6-diamidino-2-phenylindole (DAPI) staining, observing apoptotic nuclear morphology, and immunoblotting with antibodies to β-catenin, X-linked inhibitor of apoptosis, and BCL2 [61]. Cell cycle analysis using PI to quantify the proportions of cells in the G1/G0 or G2–M phases was used to examine cell cycle status [61].

Viable cells can be determined using MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) colorimetric assays [62]. Absorbance at 570 nm is used to detect the incorporation of MTT. Apoptosis can also be determined by caspase activation using an anti-poly ADP-ribose polymerase (PARP) antibody [62]. Viable cells can also be determined using a 3-(4,5-dimethyl-thiazol-2yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) kit (Promega) [63]. The terminal transferase dUTP nick end labeling (TUNEL) assay is commonly used to detect apoptosis [63]. Harvested cells are resuspended in DNA labeling solution consisting of TdT reaction buffer, TdT enzyme, and BrdUTP, then stained with PI to detect a fluorescein isothiocyanate-labeled anti-BrdU antibody [63]. Cell viability and proliferation assays were used to validate internal tandem duplication mutations in FLT3 as a therapeutic target for human acute myeloid leukemia [64]. Cell viability and proliferation can be determined using a Vi-cell XR automated cell viability analyzer (Beckman Coulter) [64].

Advertisement

3. Genomic variation in disease

3.1. Genome-wide association studies in cancer

Despite extensive research efforts for several decades, the genetic basis of common human diseases such as cancers remains largely unknown [65]. Genome-wide association studies (GWAS) have emerged as an important tool for the discovery of genomic regions that harbor genetic variants conferring risk for various cancers [66,67]. Family-based linkage studies and studies comprising tens of thousands of gene-based SNPs can also assay genetic variation across the genome [68], but the National Institutes of Health guidelines for GWAS require a sufficient density of genetic markers to capture a large proportion of the common variants in the study population, measured in enough individuals to provide sufficient power to detect variants of modest effect [67]. The recent success of GWAS can be attributed to the convergence of new technologies that can genotype hundreds of thousands of SNPs in hundreds or thousands of samples [66,69].

GWAS have been conducted in the five of the most common cancer types: breast, prostate, colorectal, lung, and melanoma (Table 4) and have identified more than 20 novel disease loci, confirming that susceptibility to these diseases is polygenic [70]. For many years, human genetics has been used to map rare mutations with large effect sizes in families or genetically homogeneous populations, such as BRCA1/BRCA2 mutations in Ashkenazi women with breast cancer and ovarian cancer [71]. A number of SNPs have now been associated with breast cancer; for example, a SNP in intron 2 of the FGFR2 gene, which encodes a receptor tyrosine kinase that is amplified and overexpressed in 5–10% of breast tumors [72,73], and SNPs on chromosomes 16q and 5q. The locus on 16q contains a gene TNRC9 and a hypothetical gene LOC643714. The function of TNRC9 is unknown but the presence of an HMG box motif suggests that it might act as a transcription factor. The 5q locus includes MAP3K1, which encodes a protein involved in signal transduction (but not previously known to be involved in cancer) and two other genes: MGC33648 and MIER3. In addition, several of the breast cancer loci appear to be associated with specific subtypes of the disease. In particular, the FGFR2 association is strongly associated with estrogen receptor-positive breast cancer, while the TNRC9 SNP is associated with both estrogen receptor-positive and -negative breast cancer [74,75]. It is surprising that none of the strongest associations map to regions harboring estrogen/progesterone genes in women of European background, particularly because a GWAS in Asian women reported a convincing association with markers near the estrogen receptor alpha (ESR1) gene [76]. In prostate cancer, the first and most important region to emerge was 8q24. This region was first associated with prostate cancer through linkage studies by the deCode group, was followed up by association analyses [77], and has been confirmed in subsequent GWAS [78-81]. Another signal, on chromosome 10q13, points to a variant in the promoter of the MSMB gene, which encodes the PSP94 protein; this is now under intense investigation as a biomarker for prostate cancer [80,81].

In general, the susceptibility alleles discovered thus far are common—that is, with a frequency in one or more population of >10%, and each allele confers a small contribution to the overall risk of the disease. For nearly all regions conclusively identified by GWAS, the effect sizes per allele are estimated at <1.3. It was not anticipated that GWAS in certain cancers would yield many novel regions when other cancers strongly associated with particular environmental exposures have yielded so few regions. For example, prostate cancer, breast cancer, and colon cancer have been associated with 29, 13, and 10 regions of the genome, respectively, while there are only three associated regions for lung cancer in smokers, and three for bladder cancer despite analysis of sufficiently large data sets [67]. Several GWAS for lung cancer have identified the same locus on 15q25, suggesting that this is an important susceptibility locus for this disease [82-87]. This locus contains the nicotinic acetylcholine receptor subunit genes CHRNA3 and CHRNA5, suggesting that susceptibility may be mediated through smoking behavior [86,87].

GWAS represent an important advance in discovering genetic variants influencing disease but have important limitations. There is a high potential for false-positive results, they do not yield information on gene function, they are insensitive to rare and structural variants, they require large sample sizes, and incur possible biases because of case and control selection and genotyping errors [88]. Clinicians and scientists must understand the unique aspects of these studies and be able to assess and interpret GWAS results for themselves and their patients. However, at present these studies mainly represent a valuable discovery tool for examining genomic function and clarifying pathophysiological mechanisms. However, through GWAS, the identification of variants, genes, and pathways involved in multiple cancers offers a potential route to new therapies, improved diagnosis, and better disease prevention [65].

Cancer type Reference Year Platform[SNP passing QC] Ethnic group Initial sample size Replication sample size
Cases Controls Ethnic groups Cases Controls
Breast cancer [89] 2012 Affymetrix
[555,525]
Korean 2,273 2,052 Korean 4,049 3,845
[90] 2012 Affymetrix
[690,947]
Chinese 2,918 2,324 Chinese 6,838 6,888
Han Chinese 1,297 1,585
Taiwan Chinese 1,066 1,065
Korean 5,038 6,869
Japanese 1,934 1,875
[91] 2012 Affymetrix
[613,031]
Chinese 1,950 - Chinese 4,160 -
[92] 2012 Illumina
[470,796]
Japanese 240 - Japanese 222 -
[93] 2011 Affymetrix
[684,457]
East Asian 2,062 2,066 East Asians 15,091 14,877
[94] 2011 Affymetrix
[782,838]
European 302 321 European 1,153 1,215
[95] 2011 Illumina
[~296,114]
British 1,694 2,365 British 7,317 8,124
European 1,145 1,142
[96] 2010 Illumina
[285,984]
Swedish & Finnish 617 4,583 European 1,001 7,604
[97] 2010 Affymetrix
[592,163]
European 899 804 European 1,264 1,222
[98] 2010 Illumina
[285,984]
European 2,702 5,726 European 7,386 7,576
[99] 2010 Illumina
[582,886]
UK 3,659 4,897 European 12,576 12,223
[100] 2010 Illumina
[528,252]
British 1,145 - British 4,335 -
[101] 2009 Illumina
[528,173]
1,145 1,142 8,625 9,657
[102] 2009 Affymetrix
[up to 607,728]
Chinese 1,505 1,522 Chinese 1,554 1,576
[103] 2008 Affymetrix
[200,220]
30 30 - - -
[104] 2008 Affymetrix
[492,900]
Ashkenazi Jewish 249 299 Ashkenazi Jewish 1,193 1,166
[105] 2007 Affymetrix
[70,897]
Framing-ham 1,345 - - - -
[106] 2007 Perlegen
[205,586]
390 634 26,646 24,889
[107] 2007 Illumina
[528,173]
1,145 1,142 1,176 2,072
Prostate cancer [108] 2012 Illumina
[509,916]
European 1,176 1,101 European 1,964 3,172
[109] 2012 Affymetrix
& Illumina
[1,117,531]
(imputed)
4,723 4,792 - - -
[110] 2011 NR
[2.6 million]
(imputed)
European 6,621 6,939 European
Japanese
Chinese
African American
22,957
285
135
112
  

7,140
23,234
298
135
298
  

5,455
[111] 2011 Illumina
[571,243]
European 2,782 4,458 European 8,217 6,732
[112] 2011 Illumina
[1,047,198]
African American 3,425 3,290 African American
Senegalese
Ghanaian
Barbadian
1,275
  

86
271
246
1,695
  

414
968
253
[113] 2011 Affymetrix
[387,384]
European 202 100 European 1,122 1,167
[114] 2010 Affymetrix
[419,613]
Caucasian 222 415 Caucasian 500 155
[115] 2010 Illumina
[510,687]
Japanese 1,583 3,386 Japanese 3,001 5,415
[116] 2009 Illumina
[541,129]
European 1,854 1,894 European, Chinese, Japanese, African American, Latino, and Hawaiian 19,879 18,761
[117] 2009 Illumina
[310,520]
Icelandic 1,968 35,382 European 11,806 12,387
[118] 2008 Illumina
[541,129]
European 1,854 1,894 3,268 3,366
[119] 2008 Illumina
[527,869]
1,172 1,157 3,941 3,964
[120] 2007 Affymetrix & Illumina
[60,275]
1,235 1,599 1,242 917
[121] 2007 Affymetrix
[70,897]
Framingham 1,345 - - -
[122] 2007 Illumina
[316,515]
1,453 3,064 East Asia 1,210 2,445
[123] 2007 Illumina
[538,548]
1,172 1,157 3,124 3,142
Colorectal cancer [124] 2011 Illumina
[378,739]
European 2,906 3,416 European 8,161 9,101
[125] 2010 Illumina
[~550,000]
(imputed)
European 3,334 4,628 European 14,851 15,569
[126] 2010 Affymetrix
[460,945]
German 371 1,263 German
Czech
4,121
794
7,344
815
[127] 2008 Illumina
[~548,586]
1,902 1,929 4,878 4,914
[128] 2008 Illumina
[541,628]
981 1,002 16,476 15,351
[129] 2008 Illumina
[547,647]
922 927 17,872 17,526
[130] 2007 Illumina
[547,647]
940 965 7,473 5,984
[131] 2007 Illumina
[547,647]
930 960 7,334 5,246
[132] 2007 Affymetrix & Illumina
[99,632]
1,257 1,336 6,223 6,443
Lung
cancer
[133] 2011 Affymetrix
[906,703]
Chinese 2,331 3,077 Chinese 6,313 6,409
[134] 2011 Illumina
[307,260]
White 327 European 587
[135] 2011 Illumina
[620,901]
(pooled)
Italian 600 - Italian 317 -
[136] 2010 Affymetrix
[265,996]
Han Chinese 245 - Han Chinese 305 -
[137] 2010 Affymetrix
[246,758]
Korean 621 1,541 Korean 804 1,470
[138] 2010 Illumina
[542,050]
377 377 511 1,007
[139] 2009 Illumina
[515,922]
European 5,739 5,848 European 7,561 13,818
[140] 2009 Illumina
[511,919]
European 1,952 1,438 European 5,608 6,767
[141] 2008 Illumina
[317,498]
1,154 1,138 2,724 3,694
[142] 2008 Illumina
[306,207]
10,995 smokers 4,848 smokers
[143] 2008 Illumina
[310,023]
1,926 2,522 2,513 4,752
[144] 2007 Affymetrix
[~116,204]
(pooled)
Italian 338 335 Norwegian 265 356
Melanoma [145] 2011 Illumina
[594,997]
European 2,804 7,618 European 5,551 7,449
[146] 2011 Illumina
[5,480,804
(imputed)]
European 2,168 4,387 European 5,193 15,144
[147] 2011 Illumina
[818,977]
European 1,804 1,026 European 6,483 23,324
[148] 2011 Illumina
[491,227]
European 156 2,150 NR - -
[149] 2009 Illumina
[~317,000]
European 1,539 3,917 European 2,312 1,867

Table 4.

Summary of GWAS for the five of the most common types of cancer.

3.2. Genetic risk score in cancer and diabetes

Type 2 diabetes mellitus and cancers are major health problems worldwide [150,151]. The recent increase in the prevalence of these diseases is largely attributable to environmental factors. However, convincing evidence shows that genetic factors may play an important role in these diseases [152,153]. Recent GWAS have led to the identification of a series of SNPs that are robustly associated with either the risk of diabetes or cancers [151,154-159]. For type 2 diabetes mellitus, common SNPs have been identified in the PPARG, KCNJ11, and TCF7L2 genes, and have been widely replicated in populations of various ethnicities [160-162]. Other potential new loci include HHEX, CDKAL1, CDKN2A/B, IGF2BP2, SLC30A8, and WFS1 [65,155-159,163,164]. A number of SNPs have been identified as associated with breast cancer risk, including FGFR2, CASP8, ERBB4, TAB2 , BARX2, TMEM45B, ESR1, FGFR2, TNRC9, MAP3K1, MGC33648, MIER3, and RAD51L1 [74,75,151] (Table 5).

Combining multiple loci with modest effects into a global genetic risk score (GRS) might improve the identification of those at risk for common complex diseases such as type 2 diabetes and cancers [165-167]. Several studies have developed methods to predict the risk of certain diseases, such as coronary heart disease, type 2 diabetes, and breast cancer, aggregating information from multiple SNPs into a single GRS [151,168,169]. For example, in the Atherosclerosis Risk in Communities study, the aggregation of multiple SNPs into a single GRS was responsible for improving the prediction of coronary heart disease incidence [168]. In a study that used a GRS to determine the risk of type 2 diabetes in US men and women, individuals in the highest quintile of GRS had a significantly increased risk of type 2 diabetes compared with those in the lowest quintile; however, the addition of a GRS to the conventional model consisting of lifestyle risk factors only increased the area under the curve by only 1% (AUC=0.78). In this instance, the GRS was determined to be useful only when combined with the body mass index or a family history of diabetes [169]. For breast cancer, a GRS was created using 14 SNPs previously associated with breast cancer, and was substantially more predictive of estrogen receptor-positive breast cancer than of estrogen receptor-negative breast cancer, particularly for absolute risk [151]. Further studies are needed to confirm whether a GRS improves disease risk prediction.

The GRS is calculated on the basis of reproducible tagging of SNP-associated loci reaching genome-wide levels of significance. The GRS can be created by two methods: a simple count method (count GRS) and a weighted method (weighted GRS) [169,170]. Both methods anticipate each SNP to be independently associated with risk. An additive genetic model is used for each SNP, applying a linear weighting of 0, 1, or 2 to genotypes containing 0, 1, or 2 risk alleles, respectively. This model is known to perform well even when the true genetic model is unknown or wrongly specified [171]. The count model assumes that each SNP in the panel contributes equally to the disease risk and is calculated by summing the values for each of the SNPs. The weighted GRS is calculated by multiplying each B-coefficient, the estimates resulting from an analysis carried out on variables that have been standardized, by the number of corresponding risk alleles (0, 1, or 2).

Disease Reference Year Ethnic group Participants No. of SNPs Genes found from GWAS
Type 2
diabetes
[165] 2008 Framingham 2,377 diabetic patients 18 NOTCH2 (rs10923931),
BCL11A (rs10490072),
THADA (rs7578597),
IGF2BP2 (rs1470579),
PPARg (rs1801282),
ADAMTS9 (rs4607103),
CDKAL (rs7754840),
VEGFA (rs9472138),
JAZF1 (rs86475),
SLC30A8 (rs13266634),
CDKNA/2B (rs10811661),
HHEX (rs1111875),
CDC123, CAMK1D (rs12779790),
TCF7L2 (rs7903146),
KCNJ11 (rs5219), INS (rs689),
DCD (rs1153188),
TSPAN8, LGR5 (rs7961581)
[169] 2009 European 2,809 diabetic
patients &
3,501 health
controls
10 WFS1 (rs10010131),
HHEX (rs1111875),
CDKAL1 (rs7756992),
IGF2BP2 (rs4402960),
SLC30A8 (rs13266634),
CDKN2A/B (rs10811661),
TCF7L2 (rs12255372),
PPARG (rs1801282),
KCNJ11 (rs5219)
Breast cancer [151] 2010 UK 10,306
breast cancer
patients &
10,393 controls
14 FGFR2 (rs2981582),
TNRC9 (rs3803662),
2q35 (rs13387042),
MAP3K1 (rs889312),
8q24 (rs13281615),
2p (rs4666451),
5pas (rs981782),
CASP8 (rs104548),
LSP1 (rs3817198),
5p (rs30099),
TGFB1 (rs198/2073),
ATM (rs1800054),
TNRC9 (rs8051542),
TNRC9 (rs12443621)

Table 5.

Studies using a genetic risk score for cancers and diabetes, comprising SNPs identified in GWAS.

3.3. Cancer Cell Line Encyclopedia

The Cancer Cell Line Encyclopedia (CCLE) has made predictive modeling of anticancer drug sensitivity a realistic proposition, by determining genomic markers of drug sensitivity in cancer cells [172,173]. The CCLE contains information from 947 human cancer cell lines including data on gene expression, chromosomal copy number, and massively parallel sequencing data. It has been used to identify genetic, lineage-specific, and gene expression-based predictors of drug sensitivity [172]. This has revealed, for example, that the plasma cell lineage is correlated with sensitivity to IGF1 receptor inhibitors, aryl hydrocarbon receptor (AHR) expression is associated with MEK inhibitor efficacy in NRAS-mutant lines, and SLFN11 expression is associated with sensitivity to topoisomerase inhibitors [172]. Genomic markers of drug sensitivity in cancer cells have also been systematically identified using the Genomics of Drug Sensitivity in Cancer database (http://www.cancerRxgene.org) [173]. These databases will enable to overview genome quality.

Advertisement

4. Conclusion

There are dramatic changes in the genomes of cancer cells, which vary according to cancer subtype. Integrative and wide investigations of cancer cell genomes have revealed mutations and alterations in gene expression that are associated with the disease. Databases that include abundant data related to gene and protein conformation, gene expression, and genomic mutations enable the construction of dynamic cellular simulations and disease models. New sequencing tools such as next-generation sequencing will reveal new horizons in the prediction of disease and drug sensitivity, which play an important role in personalized medicine. Appropriate translation of the abundance of information to clinical practice is one of most important future challenges for medicine. The quality of genome would be one of the important factors for detecting the development of the disease.

Advertisement

Acknowledgments

The authors are grateful to all those who helped with preparation of the manuscript. In particular, we thank Jaeseong Jo for his great assistance.

References

  1. 1. Venter J. C. Adams M. D. Myers E. W. Li P. W. Mural R. J. Sutton G. G. et al. 2001 The sequence of the human genome Science 291 5507 1304 1351
  2. 2. Lander E. S. Linton L. M. Birren B. Nusbaum C. Zody M. C. Baldwin J. et al. 2001 International Human Genome Sequencing Consortium Initial sequencing and analysis of the human genome Nature 409 6822 860 921
  3. 3. Ashburner M. Ball C. A. Blake J. A. Botstein D. Butler H. Cherry J. M. et al. 2000 Gene ontology: tool for the unification of biology. The Gene Ontology Consortium Nat. Genet. 25 1 25 29
  4. 4. Hunter S. Apweiler R. Attwood T. K. Bairoch A. Bateman A. Binns D. et al. 2009 InterPro: the integrative protein signature database Nucleic Acids Res. 37 D211 D215
  5. 5. Hunter S. Jones P. Mitchell A. Apweiler R. Attwood T. K. Bateman A. et al. 2012 InterPro in 2011: new developments in the family and domain prediction database Nucleic Acids Res. 40 D306 D312
  6. 6. Attwood T. K. Coletta A. Muirhead G. Pavlopoulou A. Philippou P. B. Popov I. et al. 2012 The PRINTS database: a fine-grained protein sequence annotation and analysis resource--its status in 2012 Database Oxford bas019
  7. 7. Sigrist C. J. A. Cerutti L. de Castro E. Langendijk-Genevaux P. S. Bulliard V. Bairoch A. et al. 2010 PROSITE, a protein domain database for functional characterization and annotation Nucleic Acids Res. 38 D161 D166
  8. 8. Punta M. Coggill P. C. Eberhardt R. Y. Mistry J. Tate J. Boursnell C. et al. 2012 The Pfam protein families database Nucleic Acids Res. 40 D290 D301
  9. 9. Letunic I. Doerks T. Bork P. 2012 SMART7: recent updates to the protein domain annotation resource Nucleic Acids Res. 40 D302 D305
  10. 10. Bru C. Courcelle E. Carrère S. Beausse Y. Dalmar S. Kahn D. 2005 The ProDom database of protein domain families: more emphasis on 3D Nucleic Acids Res. 33 D212 D215
  11. 11. Nikolskaya A. N. Arighi C. N. Huang H. Barker W. C. Wu C. H. 2007 PIRSF family classification system for protein functional and evolutionary analysis Evol. Bioinform Online 2 197 209
  12. 12. de Lima Morais. D. A. Fang H. Rackham O. J. L. Wilson D. Pethica R. Chothia C. et al. 2011 SUPERFAMILY 1.75 including a domain-centric gene ontology method Nucleic Acids Res. 39 D427 D434
  13. 13. Mi H. Dong Q. Muruganujan A. Gaudet P. Lewis S. Thomas P. D. 2010 PANTHER version 7: improved phylogenetic trees, orthologs and collaboration with the Gene Ontology Consortium Nucleic Acids Res. 38 D204 D210
  14. 14. Lees J. Yeats C. Perkins J. Sillitoe I. Rentzsch R. Dessailly B. H. et al. 2012 Gene3D: a domain-based resource for comparative genomics, functional annotation and protein network analysis Nucleic Acids Res. 40 D465 D471
  15. 15. Selengut J. D. Haft D. H. Davidsen T. Ganapathy A. Gwinn-Giglio M. Nelson W. C. et al. 2007 TIGRFAMs and Genome Properties: tools for the assignment of molecular function and biological process in prokaryotic genomes Nucleic Acids Res. 35 D260 D264
  16. 16. Lima T. Auchincloss A. H. Coudert E. Keller G. Michoud K. Rivoire C. et al. 2009 HAMAP: a database of completely sequenced microbial proteome sets and manually curated microbial protein families in UniProtKB/Swiss-Prot Nucleic Acids Res. 37 D471 D478
  17. 17. Sidransky D. 2002 Emerging molecular markers of cancer Nat. Rev. Cancer 2 3 210 219
  18. 18. Brennan J. A. Mao L. Hruban R. H. Boyle J. O. Eby Y. J. Koch W. M. et al. 1995 Molecular assessment of histopathological staging in squamous-cell carcinoma of the head and neck N. Engl. J. Med. 332 7 429 435
  19. 19. Hollstein M. Sidransky D. Vogelstein M. Harris C. C. 1991 p53 mutations in human cancers Science 253 5015 49 53
  20. 20. Sidransky D. Von Eschenbach A. Tsai Y. C. Jones P. Summerhayes I. Marshall F. et al. 1991 Identification of p53 gene mutations in bladder cancers and urine samples Science 252 5006 706 709
  21. 21. Rossi A. Galetta D. 2012 Biomarkers for the targeted therapies of non-small cell lung cancer Current Biomarker Findings 2 7 17
  22. 22. Sidransky D. Tokino T. Hamilton S. R. Kinzler K. W. Levin B. Frost P. et al. 1992 Identification of ras oncogene mutations in the stool of patients with curable colorectal tumors Science 256 5053 102 105
  23. 23. Gonzalez-Zulueta M. Ruppert J. M. Tokino K. Tsai Y. C. Spruck I. I. I. C. H. Miyano N. et al. 1993 Microsatellite instability in bladder cancer Cancer Res. 53 23 5620 5623
  24. 24. Belt E. J. T. Brosens R. P. M. Delis-van Diemen P. M. Bril H. Tijssen M. van Essen D. F. et al. 2012 Cell cycle proteins predict recurrence in stage II and III colon cancer Ann. Surg. Oncol. DOI: 10.1245/s10434-012-2216-7 Published online: 04 Feb 2012
  25. 25. Sigurdsson V. Hilmarsdottir B. Sigmundsdottir H. Fridriksdottir A. J. R. Ringnér M. Villadsen R. et al. 2011 Endothelial induced EMT in breast epithelial cells with stem cell properties PLoS One 6 9 e23833
  26. 26. Abbas O. Richards J. E. Yaar R. Mahalingam M. 2011 Stem cell markers (cytokeratin 15, cytokeratin 19 and p63) in in situ and invasive cutaneous epithelial lesions Mod. Pathol. 24 1 90 97
  27. 27. Kote-Jarai Z. Amin Al. Olama A. Leongamornlert D. Tymrakiewicz M. Saunders E. et al. 2011 Identification of a novel prostate cancer susceptibility variant in the KLK3 gene transcript Human Genet. 129 6 687 694
  28. 28. Schröder F. Hugosson J. Roobol M. J. Tammela T. L. J. Ciatto S. Nelen V. et al. 2012 ERSPC Investigators. Prostate-cancer mortality at 11 years of follow-up N. Engl. J. Med. 366 11 981 990
  29. 29. Fliss M. S. Usadel H. Caballero O. L. Wu L. Buta M. R. Eleff S. M. et al. 2000 Facile detection of mitochondrial DNA mutations in tumors and bodily fluid Science 287 5460 2017 2019
  30. 30. Polyak K. Li Y. Zhu H. Lengauer C. Willson J. K. V. Markowitz S. D. et al. 1998 Somatic mutations of the mitochondrial genome in human colorectal tumours Nat. Genet. 20 3 291 293
  31. 31. Lam E. T. Bracci P. M. Holly E. A. Chu C. Poon A. Wan E. et al. 2012 Mitochondrial DNA sequence variation and risk of pancreatic cancer Cancer Res. 72 3 686 695
  32. 32. Skonieczna K. Malyarchuk B. A. Grzybowski T. 2012 The landscape of mitochondrial DNA variation in human colorectal cancer on the background of phylogenetic knowledge Biochim. Biophys. Acta 1825 2 153 159
  33. 33. Luo B. Cheung H. W. Subramanian A. Sharifnia T. Okamoto M. Yang X. et al. 2008 Highly parallel identification of essential genes in cancer cells Proc. Natl. Acad. Sci. USA 105 51 20380 20385
  34. 34. Nitiss J. L. Liu Y. Hsiung Y. 1993 A temperature sensitive topoisomerase II allele confers temperature dependent drug resistance on amsacrine and etoposide: a genetic system for determining the targets of topoisomerase II inhibitors Cancer Res. 53 1 89 93
  35. 35. Osheroff N. 1989 Effect of antineoplastic agents on the DNA cleavage/religation reaction of eukaryotic topoisomerase II: inhibition of DNA religation by etoposide Biochemistry 28 15 6157 6160
  36. 36. Hande K. R. 1998 Etoposide: four decades of development of a topoisomerase II inhibitor European J. Cancer 34 1514 1521
  37. 37. Sherr C. J. 1996 Cancer cell cycles Science 274 5293 1672 1677
  38. 38. Malumbres M. Barbacid M. 2009 Cell cycle, CDKs and cancer: a changing paradigm Nat. Rev. Cancer 9 3 153 166
  39. 39. Kundumani-Sridharan V. Quyen D. V. Subramani J. Singh N. K. Chin Y. E. Rao G. N. 2012 Novel interactions between NFATc1 (nuclear factor of activated T cells c1) and STAT-3 (signal transducer and activator of transcription-3) mediate G protein-coupled receptor agonist, thrombin-induced biphasic expression of cyclin D1, with first phase influencing cell migration and second phase directing cell proliferation J. Biol. Chem. 287 27 22463 22482
  40. 40. Landis M. W. Pawlyk B. S. Li T. Sicinski P. Hinds P. W. 2006 Cyclin D1-dependent kinase activity in murine development and mammary tumorigenesis Cancer Cell 9 1 13 22
  41. 41. Klein E. A. Assoian R. K. 2008 Transcriptional regulation of the cyclin D1 gene at a glance J. Cell Sci. 121 Pt 23 3853 3857
  42. 42. Hsu W. Shakya R. Costantini F. 2001 Impaired mammary gland and lymphoid development caused by inducible expression of Axin in transgenic mice J. Cell Biol. 155 6 1055 1064
  43. 43. Shtutman M. Zhurinsky J. Simcha I. Albanese C. D’Amico M. Pestell R. et al. 1999 The cyclin D1 gene is a target of the beta-catenin/LEF-1 pathway Proc. Natl. Acad. Sci. USA 96 10 5522 5527
  44. 44. Zhang Q. Gu J. Li L. Liu J. Luo B. Cheung H. et al. 2009 Control of cyclin D1 and breast tumorigenesis by the EglN2 prolyl hydroxylase Cancer Cell 16 5 413 424
  45. 45. Coudreuse D. Nurse P. 2010 Driving the cell cycle with a minimal CDK control network Nature 468 7327 1074 1079
  46. 46. Bloom J. Cross F. R. 2007 Multiple levels of cyclin specificity in cell-cycle control Nat. Rev. Mol. Cell Biol. 8 2 149 160
  47. 47. Santamaría D. Barrière C. Cerqueira A. Hunt S. Tardy C. Newton K. et al. 2007 Cdk1 is sufficient to drive the mammalian cell cycle Nature 448 7155 811 815
  48. 48. Tobin N. P. Sims A. H. Lundgren K. L. Lehn S. Landberg G. 2011 Cyclin D1, Id1 and EMT in breast cancer BMC Cancer 11 417
  49. 49. Bitu C. Destro M. Carrera M. Silva S. Graner E. Kowalski L. P. Soares F. et al. 2012 HOXA1 is overexpressed in oral squamous cell carcinomas and its expression is correlated with poor prognosis BMC Cancer 12 146
  50. 50. Yang Y. 2012 Wnt signaling in development and disease Cell & Bioscience 2 14
  51. 51. Iyer S. Xiao E. Alsayegh K. Eroshenko N. Riggs M. J. Bennett J. P. et al. 2012 Mitochondrial gene replacement in human pluripotent stem cell-derived neural progenitors Gene Ther. 19 5 469 475
  52. 52. Consiglio A. Carella M. De Caro G. Delle Foglie. G. Giovannelli C. Grillo G. et al. 2012 BEAT: Bioinformatics Exon Array Tool to store, analyze and visualize Affymetrix GeneChip Human Exon Array data from disease experiments BMC Bioinformatics 13 Suppl 4 S21
  53. 53. Patron J. P. Fendler A. Bild M. Jung U. Müller H. Arntzen M. et al. 2012 MiR-133b targets antiapoptotic genes and enhances death receptor-induced apoptosis PLoS One 7 4 e35345
  54. 54. Alevizos L. Gomatos I. P. Smparounis S. Konstadoulakis M. M. Zografos G. 2012 Review of the molecular prolife and modern prognostic markers for gastric lymphoma: how do they affect clinical practice? Can. J. Surg. 55 2 117 124
  55. 55. Aoudjit F. Vuori K. 2012 Integrin signaling in cancer cell survival and chemoresistance Chemother. Res. Pract. 2012 283181
  56. 56. Hayakawa Y. Hirata Y. Nakagawa H. Sakamoto K. Hikiba Y. Kinoshita H. et al. 2011 Apoptosis signal-regulating kinase 1 and cyclin D1 compose a positive feedback loop contributing to tumor growth in gastric cancer Proc. Natl. Acad. Sci. USA 108 2 780 785
  57. 57. Cairns P. Sidransky D. 1999 Molecular methods for the diagnosis of cancer Biochim. Biophys. Acta 1423 2 C11 C18
  58. 58. Glickman J. F. Mc Gee J. Napper A. 2004 Assay development for protein kinase enzyme. Assay Guidance Manual [Internet]. Sittampalam GS, Weidner J, Auld D, et al., editors Bethesda (MD): Eli Lilly & Company and the National Center for Advancing Translational Sciences, 2012
  59. 59. Eglen R. M. Singh R. 2003 Beta galactosidase enzyme fragment complementation as a novel technology for high throughput screening Comb. Chem. High Throughput Screen. 6 4 381 387
  60. 60. Baran Y. Zencir S. Cakir Z. Ozturk E. Topcu Z. 2011 Imatinib-induced apoptosis: a possible link to topoisomerase enzyme inhibition J. Clin. Pharm. Ther. 36 6 673 679
  61. 61. Dickey A. Schleicher S. Leahy K. Hu R. Hallahan D. 2011 Thotala DK GSK-3β inhibition promotes cell death, apoptosis, and in vivo tumor growth delay in neuroblastoma Neuro-2A cell line J. Neurooncol. 104 1 145 153
  62. 62. Wang L. Hurley D. G. Watkins W. Araki H. Tamada Y. Muthukaruppan A. et al. 2012 Cell cycle gene networks are associated with melanoma prognosis PLoS One 7 4 e34247
  63. 63. Kim J. Chae M. Kim W. K. Kim Y. Kang H. S. Kim H. S. et al. 2011 Salinomycin sensitizes cancer cells to the effects of doxorubicin and etoposide treatment by increasing DNA damage and reducing p21protein Br. J. Pharmacol. 162 3 773 784
  64. 64. Smith C. C. Wang Q. Chin C. Salerno S. Damon L. E. Levis M. J. et al. 2012 Validation of ITD mutations in FLT3 as a therapeutic target in human acute myeloid leukaemia Nature 485 7397 260 263
  65. 65. Wellcome Trust Case Control Consortium. 2007 Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls Nature 447 7145 661 668
  66. 66. National Institutes of Health. 2007 Policy for sharing of data obtained in NIH supported or conducted genome-wide association studies (GWAS) Federal Regist. 72 166 49290 49297 http://www.grants.nih.gov/grants/guide/notice-files/NOT-OD-07-088.html accessed 3 July 2012
  67. 67. Chung C. C. Magalhaes W. C. Gonzalez-Bosquet J. Chanock S. J. 2010 Genome-wide association studies in cancer--current and future directions Carcinogenesis 31 1 111 120
  68. 68. Hampe J. Franke A. Rosenstiel P. Till A. Teuber M. Huse K. et al. 2007 A genome-wide association scan of nonsynonymous SNPs identifies a susceptibility variant for Crohn disease in ATG16L1 Nat. Genet. 39 2 207 211
  69. 69. Manolio T. A. 2010 Genomewide association studies and assessment of the risk of disease N. Engl. J. Med. 363 2 166 176
  70. 70. Easton D. F. Eeles R. A. 2008 Genome-wide association studies in cancer Hum. Mol. Genet. 17 R2 R109 R115
  71. 71. Miki Y. Swensen J. Shattuck-Eidens D. Futreal P. A. Harshman K. Tavtigian S. et al. 1994 A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1 Science 266 5182 66 71
  72. 72. Adnane J. Gaudray P. Dionne C. A. Crumley G. Jaye M. Schlessinger J. et al. 1991 BEK and FLG, two receptors to members of the FGF family, are amplified in subsets of human breast cancers Oncogene 6 4 659 663
  73. 73. Greenman C. Stephens P. Smith R. Dalgliesh G. L. Hunter C. Bignell G. et al. 2007 Patterns of somatic mutation in human cancer genomes Nature 446 7132 153 158
  74. 74. Garcia-Closas M. Hall P. Nevanlinna H. Pooley K. Morrison J. Richesson D. et al. 2008 Heterogeneity of breast cancer associations with five susceptibility loci by clinical and pathological characteristics PloS Genet. 4 4 e1000054
  75. 75. Antoniou A. C. Spurdle A. B. Sinilnikova O. M. Healey S. Pooley K. A. Schmutzler R. K. et al. 2008 CIMBA. Common breast cancer-predisposition alleles are associated with breast cancer risk in BRCA1 and BRCA2 mutation carriers Am. J. Hum. Genet. 82 4 937 948
  76. 76. Zheng W. Long J. Gao Y. T. Li C. Zheng Y. Xiang Y. B. et al. 2009 Genome-wide association study identifies a new breast cancer susceptibility locus at 6q25.1 Nat. Genet. 41 3 324 328
  77. 77. Amundadottir L. T. Sulem P. Gudmundsson J. Helgason A. Baker A. Agnarsson B. A. et al. 2006 A common variant associated with prostate cancer in European and African populations Nat. Genet. 38 6 652 658
  78. 78. Yeager M. Orr N. Hayes R. B. Jacobs K. B. Kraft P. Wacholder S. et al. 2007 Genome-wide association study of prostate cancer identifies a second risk locus at 8q24 Nat. Genet. 39 5 645 649
  79. 79. Gudmundsson J. Sulem P. Manolescu A. Amundadottir L. T. Gudbjartsson D. Helgason A. et al. 2007 Genome-wide association study identifies a second prostate cancer susceptibility variant at 8q24 Nat. Genet. 39 5 631 637
  80. 80. Eeles R. A. Kote-Jarai Z. Giles G. G. Olama A. A. Guy M. Jugurnauth S. K. et al. 2008 Multiple newly identified loci associated with prostate cancer susceptibility Nat. Genet. 40 30 316 321
  81. 81. Thomas G. Jacobs K. B. Yeager M. Kraft P. Wacholder S. Orr N. et al. 2008 Multiple loci identified in a genome-wide association study of prostate cancer Nat. Genet. 40 3 310 315
  82. 82. Chanock S. J. Hunter D. J. 2008 Genomics: when the smoke clears Nature 452 7187 537 538
  83. 83. Hung R. J. Mc Kay J. D. Gaborieau V. Boffetta P. Hashibe M. Zaridze D. et al. 2008 A susceptibility locus for lung cancer maps to nicotinic acetylcholine receptor subunit genes on 15q25 Nature 452 7187 633 637
  84. 84. Mc Kay J. D. Hung R. J. Gaborieau V. Boffetta P. Chabrier A. Byrnes G. et al. 2008 Lung cancer susceptibility locus at 5p15 Nat. Genet. 40 12 1404 1406
  85. 85. Wang Y. Broderick P. Webb E. Wu X. Vijayakrishnan J. Matakidou A. et al. 2008 Common 5p15and 6p21.33 variants influence lung cancer risk Nat. Genet. 40 12 1407 1409
  86. 86. Bierut L. J. Madden P. A. Breslau N. Johnson E. O. Hatsukami D. Pomerleau O. F. et al. 2007 Novel genes identified in a high-density genome wide association study for nicotine dependence Hum. Mol. Genet. 16 1 24 35
  87. 87. Caporaso N. Gu F. Chatterjee N. Sheng-Chih J. Yu K. Yeager M. et al. 2009 Genome-wide and candidate gene association study of cigarette smoking behaviors PloS One 4 2 e4653
  88. 88. Pearson T. A. Manolio T. A. 2008 How to interpret a genome-wide association study JAMA 299 11 1335 1344
  89. 89. Kim H. C. Lee J. Y. Sung H. Choi J. Y. Park S. K. Lee K. M. et al. 2012 A genome-wide association study identifies a breast cancer risk variant in ERBB4 at 2q34: results from the Seoul Breast Cancer Study Breast Cancer Res. 14 R56
  90. 90. Long J. Cai Q. Sung H. Shi J. Zhang B. Choi J. Y. et al. 2012 Genome-wide association study in east Asians identifies novel susceptibility loci for breast cancer PLoS Genet. 8 2 e1002532
  91. 91. Shu X. O. Long J. Lu W. Li C. Chen W. Y. Delahanty R. et al. 2012 Novel genetic markers of breast cancer survival identified by a genome-wide association study Cancer Res. 72 5 1182 1189
  92. 92. Kiyotani K. Mushiroda T. Tsunoda T. Morizono T. Hosono N. Kubo M. et al. 2012 A genome-wide association study identifies locus at 10q22 associated with clinical outcomes of adjuvant tamoxifen therapy for breast cancer patients in Japanese Hum. mol. genet. 21 7 1665 1672
  93. 93. Cai Q. Long J. Lu W. Qu S. Wen W. Kang D. et al. 2011 Genome-wide association study identifies breast cancer risk variant at 10q21.2: results from the Asia Breast Cancer Consortium Hum. Mol. Genet. 20 24 4991 4999
  94. 94. Sehrawat B. Sridharan M. Ghosh S. Robson P. Cass C. E. Mackey J. R. et al. 2011 Potential novel candidate polymorphisms identified in genome-wide association study for breast cancer susceptibility Hum. Genet. 130 4 529 537
  95. 95. Fletcher O. Johnson N. Orr N. Hosking F. J. Gibson L. J. Walker K. et al. 2011 Novel breast cancer susceptibility locus at 9q31.2: results of a genome-wide association study J. Natl. Cancer Inst. 103 5 425 435
  96. 96. Li J. Humphreys K. Darabi H. Rosin G. Hannelius U. Heikkinen T. et al. 2010 A genome-wide association scan on estrogen receptor-negative breast cancer Breast Cancer Res. 12 6 R93
  97. 97. Gaudet M. M. Kirchhoff T. Green T. Vijai J. Korn J. M. Guiducci C. et al. 2010 Common genetic variants and modification of penetrance of BRCA2-associated breast cancer PloS Genet. 6 10 e1001183
  98. 98. Li J. Humphreys K. Heikkinen T. Aittomäki K. Blomqvist C. Pharoah P. D. et al. 2011 A combined analysis of genome-wide association studies in breast cancer Breast Cancer Res Treat. 126 3 717 727
  99. 99. Turnbull C. Ahmed S. Morrison J. Pernet D. Renwick A. Maranian M. et al. 2010 Genome-wide association study identifies five new breast cancer susceptibility loci Nat. Genet. 42 6 504 507
  100. 100. Azzato E. M. Pharoah P. D. Harrington P. Easton D. F. Greenberg D. Caporaso N. E. et al. 2010 A genome-wide association study of prognosis in breast cancer Cancer Epidemiol. Biomarkers Prev. 19 4 1140 1143
  101. 101. Thomas G. Jacobs K. B. Kraft P. Yeager M. Wacholder S. Cox D. G. et al. 2009 A multistage genome-wide association study in breast cancer identifies two new risk alleles at 1p11and 14q24.1 (RAD51L1) Nat. Genet. 41 5 579 584
  102. 102. Zheng W. Long J. Gao Y. T. Li C. Zheng Y. Xiang Y. B. et al. 2009 Genome-wide association study identifies a new breast cancer susceptibility locus at 6q25.1 Nat. Genet. 41 3 324 328
  103. 103. Kibriya M. G. Jasmine F. Argos M. Andrulis I. L. John E. M. Chang-Claude J. et al. 2009 A pilot genome-wide association study of early-onset breast cancer Breast Cancer Res. Treat. 114 3 463 477
  104. 104. Gold B. Kirchhoff T. Stefanov S. Lautenberger J. Viale A. Garber J. et al. 2008 Genome-wide association study provides evidence for a breast cancer risk locus at 6q22.33 Proc. Natl. Acad. Sci. USA 105 11 4340 4345
  105. 105. Murabito J. M. Rosenberg C. L. Finger D. Kreger B. E. Levy D. Splansky G. L. et al. 2007 A genome-wide association study of breast and prostate cancer in the NHLBI’s Framingham Heart Study BMC Med. Genet. 8 Suppl 1 S6
  106. 106. Easton D. F. Pooley K. A. Dunning A. M. Pharoah P. D. Thompson D. Ballinger D. G. et al. 2007 Genome-wide association study identifies novel breast cancer susceptibility loci Nature 447 7148 1087 1093
  107. 107. Hunter D. J. Kraft P. Jacobs K. B. Cox D. G. Yeager M. Hankinson S. E. et al. 2007 A genome-wide association study identifies alleles in FGFR2 associated with risk of sporadic postmenopausal breast cancer Nat. Genet. 39 7 870 874
  108. 108. Tao S. Feng J. Webster T. Jin G. Hsu F. C. Chen S. H. et al. 2012 Genome-wide two-locus epistasis scans in prostate cancer using two European populations Hum. Genet. 131 1225 1234
  109. 109. Tao S. Wang Z. Feng J. Hsu F. C. Jin G. Kim S. T. et al. 2012 A genome-wide search for loci interacting with known prostate cancer risk-associated genetic variants Carcinogenesis 33 3 598 603
  110. 110. Kote-Jarai Z. Olama A. A. Giles G. G. Severi G. Schleutker J. Weischer M. et al. 2011 UK Genetic Prostate Cancer Study Collaborators/British Association of Urological Surgeons’ Section of Oncology; UK ProtecT Study Collaborators, The Australian Prostate Cancer BioResource; PRACTICAL Consortium. Seven prostate cancer susceptibility loci identified by a multi-stage genome-wide association study Nat. Genet. 43 8 785 791
  111. 111. Schumacher F. R. Berndt S. I. Siddiq A. Jacobs K. B. Wang Z. Lindstrom S. et al. 2011 Genome-wide association study identifies new prostate cancer susceptibility loci Hum. Mol. Genet. 20 19 3867 3875
  112. 112. Haiman C. A. Chen G. K. Blot W. J. Strom S. S. Berndt S. I. Kittles R. A. et al. 2011 Genome-wide association study of prostate cancer in men of African ancestry identifies a susceptibility locus at 17q21 Nat. Genet. 43 6 570 573
  113. 113. Fitz Gerald L. M. Kwon E. M. Conomos M. P. Kolb S. Holt S. K. Levine D. et al. 2011 Genome-wide association study identifies a genetic variant associated with risk for more aggressive prostate cancer Cancer Epidemiol. Biomarkers Prev. 20 6 1196 1203
  114. 114. Penney K. L. Pyne S. Schumacher F. R. Sinnott J. A. Mucci L. A. Kraft P. L. et al. 2010 Genome-wide association study of prostate cancer mortality Cancer Epidemiol. Biomarkers Prev. 19 11 2869 2876
  115. 115. Takata R. Akamatsu S. Kubo M. Takahashi A. Hosono N. Kawaguchi T. et al. 2010 Genome-wide association study identifies five new susceptibility loci for prostate cancer in the Japanese population Nat. Genet. 42 9 751 754
  116. 116. Eeles R. A. Kote-Jarai Z. Al Olama A. A. Giles G. G. Guy M. Severi G. et al. 2009 UK Genetic Prostate Cancer Study Collaborators/British Association of Urological Surgeons’ Section of Oncology; UK ProtecT Study Collaborators; PRACTICAL Consortium, Easton DF Identification of seven new prostate cancer susceptibility loci through a genome-wide association study Nat. Genet. 41 10 1116 1121
  117. 117. Gudmundsson J. Sulem P. Gudbjartsson D. F. Blondal T. Gylfason A. Agnarsson B. A. et al. 2009 Genome-wide association and replication studies identify four variants associated with prostate cancer susceptibility Nat. Genet. 41 10 1122 1126
  118. 118. Eeles R. A. Kote-Jarai Z. Giles G. G. AA Olama Guy. M. Jugurnauth S. K. et al. 2008 UK Genetic Prostate Cancer Study Collaborators; British Association of Urological Surgeons’ Section of Oncology; UK ProtecT Study Collaborators. Multiple newly identified loci associated with prostate cancer susceptibility Nat. Genet. 40 3 316 321
  119. 119. Thomas G. Jacobs K. B. Yeager M. Kraft P. Wacholder S. Orr N. et al. 2008 Multiple loci identified in a genome-wide association study of prostate cancer Nat. Genet. 40 3 310 315
  120. 120. Duggan D. Zheng S. L. Knowlton M. Benitez D. Dimitrov L. Wiklund F. et al. 2007 Two genome-wide association studies of aggressive prostate cancer implicate putative prostate tumor suppressor gene DAB2IP J. Natl. Cancer Inst. 99 24 1836 1844
  121. 121. Murabito J. M. Rosenberg C. L. Finger D. Kreger B. E. Levy D. Splansky G. L. et al. 2007 A genome-wide association study of breast and prostate cancer in the NHLBI’s Framingham Heart Study BMC Med. Genet. 8 Suppl 1 S6
  122. 122. Gudmundsson J. Sulem P. Manolescu A. Amundadottir L. T. Gudbjartsson D. Helgason A. et al. 2007 Genome-wide association study identifies a second prostate cancer susceptibility variant at 8q24 Nat. Genet. 39 5 631 637
  123. 123. Yeager M. Orr N. Hayes R. B. Jacobs K. B. Kraft P. Wacholder S. et al. 2007 Genome-wide association study of prostate cancer identifies a second risk locus at 8q24 Nat. Genet. 39 5 645 649
  124. 124. Peters U. Hutter C. M. Hsu L. Schumacher F. R. Conti D. V. Carlson C. S. et al. 2012 Meta-analysis of new genome-wide association studies of colorectal cancer risk Hum. Genet. 131 2 217 234
  125. 125. Houlston R. S. Cheadle J. Dobbins S. E. Tenesa A. Jones A. M. Howarth K. et al. 2010 Meta-analysis of three genome-wide association studies identifies susceptibility loci for colorectal cancer at 1q41, 3q26.2, 12q13.13 and 20q13.33 Nat. Genet. 42 11 973 977
  126. 126. Lascorz J. Försti A. Chen B. Buch S. Steinke V. Rahner N. et al. 2010 Genome-wide association study for colorectal cancer identifies risk polymorphisms in German familial cases and implicates MAPK signalling pathways in disease susceptibility Carcinogenesis 31 9 1612 1619
  127. 127. Houlston R. S. Webb E. Broderick P. Pittman A. M. Di Bernardo M. C. Lubbe S. et al. 2008 International Colorectal Cancer Genetic Association Consortium Meta-analysis of genome-wide association data identifies four new susceptibility loci for colorectal cancer Nat. Genet. 40 12 1426 1435
  128. 128. Tenesa A. Farrington S. M. Prendergast J. G. Porteous M. E. Walker M. Haq N. et al. 2008 Genome-wide association scan identifies a colorectal cancer susceptibility locus on 11q23 and replicates risk loci at 8q24 and 18q21 Nat. Genet. 40 5 631 637
  129. 129. Tomlinson I. P. Webb E. Carvajal-Carmona L. Broderick P. Howarth K. Pittman A. M. et al. 2008 A genome-wide association study identifies colorectal cancer susceptibility loci on chromosomes 10p14and 8q23.3 Nat. Genet. 40 5 623 630
  130. 130. Broderick P. Carvajal-Carmona L. Pittman A. M. Webb E. Howarth K. Rowan A. et al. 2007 CORGI Consortium. A genome-wide association study shows that common alleles of SMAD7 influence colorectal cancer risk Nat. Genet. 39 11 1315 1317
  131. 131. Tomlinson I. Webb E. Carvajal-Carmona L. Broderick P. Kemp Z. Spain S. et al. 2007 A genome-wide association scan of tag SNPs identifies a susceptibility variant for colorectal cancer at 8q24.2 Nat. Genet. 39 8 984 988
  132. 132. Zanke B. W. Greenwood C. M. Rangrej J. Kustra R. Tenesa A. Farrington S. M. et al. 2007 Genome-wide association scan identifies a colorectal cancer susceptibility locus on chromosome 8q24 Nat. Genet. 39 8 989 994
  133. 133. Hu Z. Wu C. Shi Y. Guo H. Zhao X. Yin Z. et al. 2011 A genome-wide association study identifies two new lung cancer susceptibility loci at 13q12.12 and 22q12.2 in Han Chinese Nat. Genet. 43 8 792 796
  134. 134. Wu X. Ye Y. Rosell R. Amos C. I. Stewart D. J. MA Hildebrandt et. al 2011 Genome-wide association study of survival in non-small cell lung cancer patients receiving platinum-based chemotherapy J. Natl. Cancer Inst. 103 10 817 825
  135. 135. Frullanti E. Galvan A. Falvella F. S. Manenti G. Colombo F. Vannelli A. et al. 2011 Multiple genetic loci modulate lung adenocarcinoma clinical staging Clin. Cancer Res. 17 8 2410 2416
  136. 136. Wu C. Xu B. Yuan P. Miao X. Liu Y. Guan Y. et al. 2010 Genome-wide interrogation identifies YAP1 variants associated with survival of small-cell lung cancer patients Cancer Res. 70 23 9721 9729
  137. 137. Yoon K. A. Park J. H. Han J. Park S. Lee G. K. Han J. Y. et al. 2010 A genome-wide association study reveals susceptibility variants for non-small cell lung cancer in the Korean population Hum. Mol. Genet. 19 24 4948 4954
  138. 138. Li Y. Sheu C. C. Ye Y. de Andrade M. Wang L. Chang S. C. et al. 2010 Genetic variants and risk of lung cancer in never smokers: a genome-wide association study Lancet Oncol. 11 4 321 330
  139. 139. Landi M. T. Chatterjee N. Yu K. Goldin L. R. Goldstein A. M. Rotunno M. et al. 2009 A genome-wide association study of lung cancer identifies a region of chromosome 5p15associated with risk for adenocarcinoma Am. J. Hum. Genet. 85 5 679 691
  140. 140. Broderick P. Wang Y. Vijayakrishnan J. Matakidou A. Spitz M. R. Eisen T. et al. 2009 Deciphering the impact of common genetic variation on lung cancer risk: a genome-wide association study Cancer Res. 69 19 6633 6641
  141. 141. Amos C. I. Wu X. Broderick P. Gorlov I. P. Gu J. Eisen T. et al. 2008 Genome-wide association scan of tag SNPs identifies a susceptibility locus for lung cancer at 15q25.1 Nat. Genet. 40 5 616 622
  142. 142. Thorgeirsson T. E. Geller F. Sulem P. Rafnar T. Wiste A. Magnusson K. P. et al. 2008 A variant associated with nicotine dependence, lung cancer and peripheral arterial disease Nature 452 7187 638 642
  143. 143. Hung R. J. Mc Kay J. D. Gaborieau V. Boffetta P. Hashibe M. Zaridze D. et al. 2008 A susceptibility locus for lung cancer maps to nicotinic acetylcholine receptor subunit genes on 15q25 Nature 452 7187 633 637
  144. 144. Spinola M. Leoni V. P. Galvan A. Korsching E. Conti B. Pastorino U. et al. 2007 Genome-wide single nucleotide polymorphism analysis of lung cancer risk detects the KLF6 gene Cancer Lett. 251 2 311 316
  145. 145. Barrett J. H. Iles M. M. Harland M. Taylor J. C. Aitken J. F. Andresen P. A. et al. 2011 GenoMEL Consortium. Genome-wide association study identifies three new melanoma susceptibility loci Nat. Genet. 43 11 1108 1113
  146. 146. Mac Gregor S. Montgomery G. W. Liu J. Z. Zhao Z. Z. Henders A. K. Stark M. et al. 2011 Genome-wide association study identifies a new melanoma susceptibility locus at 1q21.3 Nat. Genet. 43 11 1114 1118
  147. 147. Amos C. I. Wang L. E. Lee J. E. Gershenwald J. E. Chen W. V. Fang S. et al. 2011 GenoMEL Investigators. Genome-wide association study identifies novel loci predisposing to cutaneous melanoma Hum. Mol. Genet. 20 24 5012 5023
  148. 148. Teerlink C. Farnham J. Allen-Brady K. Camp N. J. Thomas A. Leachman S. et al. 2012 A unique genome-wide association analysis in extended Utah high-risk pedigrees identifies a novel melanoma risk variant on chromosome arm 10q Hum. Genet. 131 1 77 85
  149. 149. Bishop D. T. Demenais F. Iles M. M. Harland M. Taylor J. C. Corda E. et al. 2009 Genome-wide association study identifies three loci associated with melanoma risk Nat. Genet. 41 8 920 925
  150. 150. King H. Aubert R. E. Herman W. H. 1998 Global burden of diabetes, 1995-2025: prevalence, numerical estimates, and projections Diabetes Care 21 9 1414 1431
  151. 151. Reeves G. K. Travis R. C. Green J. Bull D. Tipper S. Baker K. et al. 2010 Incidence of breast cancer and its subtypes in relation to individual and multiple low-penetrance genetic susceptibility loci JAMA 304 4 426 434
  152. 152. Kaprio J. Tuomilehto J. Koskenvuo M. Romanov K. Reunanen A. Eriksson J. et al. 1992 Concordance for type 1 (insulin-dependent) and type 2 (non-insulin-dependent) diabetes mellitus in a population-based cohort of twins in Finland Diabetologia 35 11 1060 1067
  153. 153. Risch N. 1990 Linkage strategies for genetically complex traits. I. Multilocus models Am. J. Hum. Genet. 46 2 222 228
  154. 154. Grant S. F. Thorleifsson G. Reynisdottir I. Manolescu A. Sainz J. Helgason A. et al. 2006 Variant of transcription factor 7-like 2 (TCF7L2) gene confers risk of type 2 diabetes Nat. Genet. 38 3 320 323
  155. 155. Sladek R. Rocheleau G. Rung J. Dina C. Shen L. Serre D. et al. 2007 A genome-wide association study identifies novel risk loci for type 2 diabetes Nature 445 7130 881 885
  156. 156. Saxena R. Voight B. F. Lyssenko V. Burtt N. P. de Bakker P. I. Chen H. et al. 2007 Genome-wide association analysis identifies loci for type 2 diabetes and triglyceride levels Science 316 5829 1331 1336
  157. 157. Scott L. J. Mohlke K. L. Bonnycastle L. L. Willer C. J. Li Y. Duren W. L. et al. 2007 A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants Science 316 5829 1341 1345
  158. 158. Zeggini E. Weedon M. N. Lindgren C. M. Frayling T. M. Elliott K. S. Lango H. et al. 2007 Wellcome Trust Case Control Consortium (WTCCC). Replication of genome-wide association signals in UK samples reveals risk loci for type 2 diabetes Science 316 5829 1336 1341
  159. 159. Zeggini E. Scott L. J. Saxena R. Voight B. F. Marchini J. L. Hu T. et al. 2008 Meta-analysis of genome-wide association data and large-scale replication identifies additional susceptibility loci for type 2 diabetes Nat. Genet. 40 5 638 645
  160. 160. van Dam R. M. Hoebee B. Seidell J. C. MM Schaap de Bruin. T. W. Feskens E. J. 2005 Common variants in the ATP-sensitive K+ channel genes KCNJ11 (Kir6.2) and ABCC8 (SUR1) in relation to glucose intolerance: population-based studies and meta-analyses Diabet. Med. 22 5 590 598
  161. 161. Ludovico O. Pellegrini F. Di Paola R. Minenna A. Mastroianno S. Cardellini M. et al. 2007 Heterogeneous effect of peroxisome proliferator-activated receptor gamma2 Ala12 variant on type 2 diabetes risk Obesity (Silver Spring) 15 5 1076 1081
  162. 162. Cauchi S. El Achhab Y. Choquet H. Dina C. Krempler F. Weitgasser R. et al. 2007 TCF7L2 is reproducibly associated with type 2 diabetes in various ethnic groups: a global meta-analysis J. Mol. Med. 85 7 777 782
  163. 163. Steinthorsdottir V. Thorleifsson G. Reynisdottir I. Benediktsson R. Jonsdottir T. Walters G. B. et al. 2007 A variant in CDKAL1 influences insulin response and risk of type 2 diabetes Nat. Genet. 39 6 770 775
  164. 164. Sandhu M. S. Weedon M. N. Fawcett K. A. Wasson J. Debenham S. L. Daly A. et al. 2007 Common variants in WFS1 confer risk of type 2 diabetes Nat. Genet. 39 8 951 953
  165. 165. Meigs J. B. Shrader P. Sullivan L. M. Mc Ateer J. B. Fox C. S. Dupuis J. et al. 2008 Genotype score in addition to common risk factors for prediction of type 2 diabetes N. Engl. J. Med. 359 21 2208 2219
  166. 166. Weedon M. N. Mc Carthy M. I. Hitman G. Walker M. Groves C. J. Zeggini E. et al. 2006 Combining information from common type 2 diabetes risk polymorphisms improves disease prediction PloS. Med. 3 10 e374
  167. 167. Wray N. R. Goddard M. E. Visscher P. M. 2007 Prediction of individual genetic risk to disease from genome-wide association studies Genome Res. 17 10 1520 1528
  168. 168. Morrison A. C. Bare L. A. Chambless L. E. Ellis S. G. Malloy M. Kane J. P. et al. 2007 Prediction of coronary heart disease risk using a genetic risk score: the Atherosclerosis Risk in Communities Study Am. J. Epidemiol. 166 1 28 35
  169. 169. Cornelis M. C. Qi L. Zhang C. Kraft P. Manson J. Cai T. et al. 2009 Joint effects of common genetic variants on the risk for type 2 diabetes in U.S. men and women of European ancestry Ann. Intern. Med. 150 8 541 550
  170. 170. Ripatti S. Tikkanen E. Orho-Melander M. AS Havulinna Silander. K. Sharma A. et al. 2010 A multilocus genetic risk score for coronary heart disease: case-control and prospective cohort analyses Lancet 376 9750 1393 1400
  171. 171. Balding D. J. 2006 A tutorial on statistical methods for population association studies Nat. Rev. Genet. 7 10 781 791
  172. 172. Barretina J. Caponigro G. Stransky N. Venkatesan K. AA Margolin Kim. S. et al. 2012 The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity Nature 483 7391 603 607
  173. 173. Garnett N. J. Edelman E. J. Heidorn S. J. Greenman C. D. Dastur A. Lau K. W. et al. 2012 Systematic identification of genomic markers of drug sensitivity in cancer cells Nature 483 7391 570 575

Written By

Shihori Tanabe and Sun Ha Jee

Submitted: 19 March 2012 Published: 12 December 2012