* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Download Application and interpretation of FISH in biomarker studies Jane Bayani Mini-review
Gene therapy of the human retina wikipedia , lookup
Epigenetics of human development wikipedia , lookup
Genomic imprinting wikipedia , lookup
Gene therapy wikipedia , lookup
Gene expression programming wikipedia , lookup
X-inactivation wikipedia , lookup
Comparative genomic hybridization wikipedia , lookup
Neocentromere wikipedia , lookup
Gene expression profiling wikipedia , lookup
Vectors in gene therapy wikipedia , lookup
BRCA mutation wikipedia , lookup
Therapeutic gene modulation wikipedia , lookup
Site-specific recombinase technology wikipedia , lookup
Microevolution wikipedia , lookup
Cancer epigenetics wikipedia , lookup
Artificial gene synthesis wikipedia , lookup
Designer baby wikipedia , lookup
Polycomb Group Proteins and Cancer wikipedia , lookup
Nutriepigenomics wikipedia , lookup
Genome (book) wikipedia , lookup
Cancer Letters 249 (2007) 97–109 www.elsevier.com/locate/canlet Mini-review Application and interpretation of FISH in biomarker studies Jane Bayani a, Jeremy A. Squire a,b,* a b Division of Applied Molecular Oncology, Princess Margaret Hospital, University Health Network, 610 University Avenue, Room 9-717, Toronto, Ont., Canada M5G 2M9 Department of Laboratory Medicine and Pathobiology and the Department of Medical Biophysics, University of Toronto, Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, 610 University Avenue, Room 9-717, Toronto, Ont., Canada M5G 2M9 Abstract Emerging genomic and proteomic data is creating new opportunities to identify novel biomarkers that will have pathway-specific therapeutic impact on cancer progression. Molecular cytogenetic and fluorescence in situ hybridization (FISH) methods have been primarily used in discovery genetic research laboratories until recently. New automated analytical platforms based on FISH technologies and tissue microarray methods are providing a rapid means to determine the impact of consistent genomic aberrations in clinical trials, and in studies designed to investigate differential chemotherapeutic response. Ó 2007 Elsevier Ireland Ltd. All rights reserved. Keywords: Chromosomes; Cancer genes; Arrays 1. Cancer biology and biomarkers The ideal biomarker is one that most closely resembles the carcinogenic process it is modeling [1], with the goal of molecular therapeutics to target the underlying defects that lead to cancer initiation and progression based on our current understanding of cancer biology [2]. The current view of cancer biology has been reviewed by Hanahan and Weinberg [3], and include, (1) the ability for selfsufficiency in growth signals; (2) the insensitivity to anti-growth signals; (3) evading apoptosis; (4) * Corresponding author. Tel.: +1 416 946 4509; fax: +1 416 946 2840. E-mail address: [email protected] (J.A. Squire). sustained angiogenesis; (5) limitless replicative potential; (6) tissue invasion and metastasis. These capabilities are all believed be affected in all cancer types in some way, though the mechanisms of their actions may follow different paths. In addition, because cancer is a disease of genetic progression that is often associated with specific molecular, genetic and histological changes [1], the ability to develop biomarkers that can detect the critical components of these hallmarks of cancer together provides a powerful basis for diagnosing, monitoring and predicting outcome and response to treatment. In most tumours, a variety of chromosomal abnormalities characterized by changes in chromosomal structure and number can be observed. Some of these chromosomal abnormalities have provided 0304-3835/$ - see front matter Ó 2007 Elsevier Ireland Ltd. All rights reserved. doi:10.1016/j.canlet.2006.12.030 98 J. Bayani, J.A. Squire / Cancer Letters 249 (2007) 97–109 clues about basic genetic mechanisms and has led to very specific molecular diagnostic testing in clinical laboratories, while others have been found to have prognostic significance. The purpose of this review is to provide the reader with an appreciation of the contribution that molecular cytogenetics, and specifically Fluorescence in situ Hybridization (FISH), is making in the assessment of biomarkers in which chromosomal rearrangements are affecting the molecular biology and clinical response of tumours. [9]. Gene amplification occurs frequently in solid tumours but is seldom found in hematologic malignancies. In contrast, the cytogenetics of tumour suppressor genes are quite different to that associated with oncogenes, because tumour suppressor genes are subject to inactivation mechanisms [13]. When they are functioning normally, the proteins encoded by tumour suppressor genes inhibit cell proliferation. For these genes, loss or inactivation removes the normal constraints on cellular growth. The loss of such a gene often results in a tumour bearing a genomic deletion detectable by FISH (see below). 2. Chromosomal aberrations and cancer genes The molecular consequences of chromosome aberrations influence the functioning of two broad classes of cancer genes in human tumours: dominantly acting oncogenes and tumour suppressor genes. When an oncogene becomes inappropriately activated by a chromosomal mechanism, it can stimulate cells to continue to proliferate, leading to a tumour. There are at least three chromosomal mechanisms for activating oncogenes: (1) Fusion of the oncogene with a second gene at a site of translocation or inversion of chromosomes generating a chimeric gene and a new protein [4]. This mechanism is found predominantly in leukemias, lymphomas, sarcomas, and more recently prostate cancer [5–8]. (2) Juxtaposition of the oncogene to regulatory elements in immunoglobulin or T-cell receptor genes in B- and T-lymphocyte malignancies, respectively, leading to inappropriate expression of the oncogene [6]. This mechanism is one of the commonest methods of oncogene activation in hematologic malignancies. (3) Gene amplification [9] arising from an increase in the amount of DNA from a specific region of a chromosome. Such areas on chromosomes are referred to as homogeneously staining regions (HSR) or double minutes (DMs). HSRs are associated with extensive gene amplification, commonly associated with overexpression of oncogenes that may impact on drug response [10]. Double minutes (DMs) on the other hand are a related form of gene amplification that appear as paired extrachromosomal bodies of dark-staining material in a metaphase preparation. In some instances, the number of DMs can be very large, approaching several hundred in a cell in some neuroblastomas [11] and medulloblastomas [12]. Various mechanisms have been proposed for the amplification process, based on molecular analysis of the regions of amplified DNA (known as amplicons) in different cell types 3. Molecular cytogenetics and biomarker detection in tumours Improvements in cloning technologies and antibody conjugation in the late 1970s and early 1980s led to the introduction of FISH as early as 1977 [14]. By the late 1980s, FISH was being employed to detect specific chromosomal regions and loci [15] using recombinant libraries, enabling the chromosomal mapping of many genes [16–18] within the human genome. Later in 1992, Kallioniemi et al. [19] introduced Comparative Genomic Hybridization (CGH), the predecessor to today’s microarray CGH assays (aCGH) [20], which was a FISH-based method that could assess the net genomic gains and losses in a given DNA sample without the need for fresh (viable) material required for short-term culture in traditional metaphase analysis. The competitive hybridization strategy identified regions of net genomic loss and gain, revealing regions harboring potential tumour suppressor genes (CGH regions of genomic deletion) and oncogenes (CGH regions of genomic gain/amplification). The findings of both metaphase- and aCGH surveys resulted in the discovery of genomic signatures characteristic of specific cancers, from which specific genes/loci could be interrogated and validated by other molecular methods. However, because of the bulk extraction of the DNA, the information regarding karyotypic clonality and genomic heterogeneity was lost. The advancement in FISH-based technologies continued with improved flurochromes/dyes as well as in microscopy and imaging in the late 1990s culminating in multi-fluorochrome assays, termed MFISH [21] and in particular, Spectral Karyotyping (SKY) [22], which now allows the entire metaphase spread to be analyzed in 24-colours, revealing the chromosomal origins of structural rearrangements [23,24]. The NCBI hosts a public J. Bayani, J.A. Squire / Cancer Letters 249 (2007) 97–109 database containing SKY/M-FISH and CGH data from various neoplasms (http://www.ncbi.nlm.nih. gov/sky/) submitted by individuals. Together these newer molecular cytogenetic techniques provide a much more comprehensive survey of the genome revealing the true karyotypic complexities of neoplastic cells (Fig. 1). For a more comprehensive review of FISH-based techniques, the reader is referred to Speicher and Carter [25]. 4. Detection of specific biomarkers by interphase FISH analyses As outlined in Fig. 1, both metaphase and interphase FISH analysis possess advantages and disadvantages, and in an ideal situation should be performed in parallel, but to do so is expensive, labor-intensive and time-consuming. Fortunately, the loss of information regarding the context of the chromosomal aberration within the tumour karyotype derived from metaphase analysis is less important when the same information can be derived through interphase cytogenetics. Due to advances in the efficiency and stability of conjugat- 99 ing fluorochromes/dyes into the DNA probe, interphase FISH has developed into a sensitive, stabile and reproducible assay for the detection of genomic/gene deletion, amplification, specific translocations and global genomic instability by screening nuclei of a tumour. Thus interphase FISH has the compelling advantage for biomarker studies of being capable of examining hundreds of nuclei to survey numerical chromosomal content changes such as gene amplification (Fig. 2). Since it can be applied to both cytogenetic specimens and tissues embedded in paraffin section, the technique has been widely performed on Tissue Microarrays (TMAs) [26] which afford the high-throughput screening of many tumours within one experiment and permits comparative analysis with histology (Fig. 2). Interphase FISH-based methodologies can now be automated [27] (also see Metasystems http://www.metasystems.de/, Applied Imaging http://www.aicorp.com/index.htm) which renders then suitable for large-scale biomarker screens as part of clinical trials investigating the influence of chromosomal composition on therapeutic response [10]. In the sections below we summarize the four Fig. 1. Overview of metaphase and interphase cytogenetics. Metaphase cytogenetics requires the preparation of metaphase spreads from the test specimen,which can then be interrogated with probes, either in a whole genome-fashion, such as SKY, MBand or CGH, or through the use of locus-specific probes. Interphase Cytogenetics requires only interphase nuclei either from cytogenetic preparations or from paraffin such as TMAs or frozen sections, as well as cytological smears or touch-preparations. 100 J. Bayani, J.A. Squire / Cancer Letters 249 (2007) 97–109 Fig. 2. Interphase FISH analysis to archival material permits correlation to histological or immunological types. Shown is an example of the type information that can be generated from interphase FISH analysis when applied to a histological section or Tissue Microarrays (TMA). In one experiment, hundreds of tissue cores can be evaluated with specific probes to detect gene/loci deletions, amplifications, specific translocations, changes in chromosomal ploidy or gauge chromosomal instability, as it relates to such aspects such as histological subtype, tumour stage/grade, immunohistochemical makers; or to clinical parameters such as response to treatment, outcome, disease-free interval, time to recurrence. A general sequence of events during the progression of carcinogenesis is depicted with the loss of a tumour suppressor gene or formation of an oncogenic fusion/translocation event early in tumourigenesis, followed by chromosomal instability (CIN) and events such as gene amplification occurring later; which can all be identified through interphase FISH analysis. An example of the type of information generated by such analysis is shown at the bottom, where the distribution of centromere signals for chromosomes 2, 4, 7 and 8 were enumerated in 200 nuclei and graphically displayed. The distribution of centromere signals ranging from 2 to 9 per cells for the various chromosomes implies a high level of chromosomal instability and karyotypic heterogeneity. An interphase FISH image from this case shows signals for chromosomes 7 (red) and 8 (green). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this paper.) main classes of genomic aberrations to which interphase FISH-based biomarker analyses has been applied (Fig. 3). 5. Deletion studies The deletion of specific chromosomal loci have been associated with various cancers including oligodendrogliomas with deletions of 1p and 19q [28] and neuroblastomas with deletions of 1p36 [29]. More specific gene deletions of known tumour suppressor genes include TP53 [30], RB1 [31], TP16 [32] and have shown clinical correlation to parameters including outcome and risk of metastasis. Typical FISH-deletion studies employ the use of a locus- specific probe for the gene/locus of interest as well as the accompanying centromere which are labelled for a two-colour strategy. The presence or absence of the locus-specific gene in relation to the number of copies of the centromere probe determines the overall copy number status of that gene. Among the tumour suppressor genes, PTEN has recently shown the greatest promise as a biomarker with strong correlation to therapeutic treatment. The PTEN tumour suppressor gene is located at 10q23, whose function is impaired through somatic mutation, deletion or by epigenetic modification in many cancers [33]. PTEN dephosphorylates phosphotidylinositol 3,4,5-triphosphate (PIP3), a product of PI3K [34–36], leading to inactivation or J. Bayani, J.A. Squire / Cancer Letters 249 (2007) 97–109 101 Fig. 3. Examples of PTEN deletion in prostate cancer. Shown from left to right are normal prostate tissue, high-grade PIN and prostate carcinoma tissue sections hybridized with a probe specific for the PTEN gene (red) and for centromere 10 (green). In each case, the inset is an enlargement of a boxed nucleus. In the normal tissue, two signals for PTEN (red) and two signals for centromere 10 (green) are detected in the typical pattern for a normal diploid cell. In high-grade PIN, two centromere 10 signals remain, but only one signal for PTEN was detected indicating a loss of one homolog. In the prostate carcinoma, the same pattern for PTEN deletion was also detected. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this paper.) down-regulation of the PTEN/PI3K/AKT/PKB signaling pathway. Lack of PTEN in a cell leads to accumulation of PIP3 that activates the AKT/ PKB signaling oncoprotein [37–39]. Active AKT modulates a number of downstream targets, which have important roles in the regulation of apoptosis and the cell cycle, including BAD [40], CASP3 and CASP9 [41], MDM2 [42], p27 [43], and FOXO transcription factors [44]. Other AKT substrates include the mammalian target of rapamycin (mTOR) [45] and WNT/CTNNB1 [46], as well as other proteins implicated in regulation of cellular proliferation, differentiation and invasion. The loss of inhibition of these pathways by PTEN gene inactivation is associated with tumour progression in several types of cancer, including prostate cancer [47,48]. In recent years, CCI-779, a homolog of the macrolide antibiotic rapamycin, has been shown to exhibit activity against proliferation in tumour cell lines with defective PTEN both in vitro and in vivo [reviewed by Mills in reference 2]. The availability of such treatment has increased the practical important of determining PTEN status. Locus-specific probes for the PTEN gene and centromere 10 permit FISH-based methods to do this, and when applied to paraffin sections have the added advantage of being able to score many cells maintained within their histological context. Because traditional screening methods that rely on gene sequencing or investigation of LOH require bulk extraction of nucleic acids, small foci of cells containing the deletion may go undetected due to the ‘‘diluting effect’’ of contaminating surrounding normal cells. The utility of FISH in detecting PTEN deletions in pros- tate cancer has been demonstrated recently by our group [49]. In prostate cancer, PTEN decreased expression has been associated with high grade and advanced stage [50]. We identified deletions of PTEN in 23% of the high-grade prostatic intra-epithelial neoplasia (H-PIN) specimens examined and in 66% of the prostate carcinomas. Results with fish were consistent with immunohistochemical findings. 6. Amplification studies The amplification of oncogenes is another feature of many cancers, generally reflecting poor prognosis and clinical outcome [51]. Amplification of this type has included MYCC in osteosarcoma [52,53] and prostate cancer [54–57], MYCN in neuroblastoma [58,59] and medulloblastoma [60–62], EGFR in lung cancer [10,63,64] and glioblastomas [65,66], and HER2 in breast cancer [67–69]. The amplification of these and other genes enhance their biological roles, and lead to the six hallmarks of cancer outlined in the Introduction to this Review [3]. In many cases, gene amplification occurs as a late event in disease progression, often associated with advanced stage, high grade, aggressive or metastatic disease, with a poor clinical outcome [51]. Gene amplification when identified serves as an important prognostic marker. It may also serve to predict treatment efficacy and thereby aid in treatment selection, particularly for breast and lung cancer patients. The FISH strategy for detecting gene/locus-specific amplification is identical to the strategy for detecting deletions, although it is also important to distinguish gains in copy number due to ploidy versus. 102 J. Bayani, J.A. Squire / Cancer Letters 249 (2007) 97–109 Fig. 4. Interphase FISH patterns for gene amplification. Shown from left two right are the different patterns for gene amplification using a locus-specific probe for EGFR (red) and centromere 7 (green). Low-copy number gains of EGFR are shown in the left panel where anywhere from 2 to 3 copies of the gene may occur in the cell above the ploidy of the cell, established by the number of centromeres. When gains or amplifications of EGFR are also associated with a gain of the resident chromosome 7, there are generally equal numbers red to green signals, as shown in the middle panel. High-copy number gene amplification possesses the most distinctive interphase FISH pattern, with large clusters or ‘‘domains’’ of signal within the nucleus as shown in the right panel. In this case, the EGFR signal intensity and signal number greatly out-number the number of centromere 7 signals. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this paper.) that due to locus/gene-specific amplification that is independent of ploidy (Fig. 4). HER2 is one of the best known genes associated with gene amplification and cancer. It is a member of the receptor tyrosine kinase 1 (RTK1) family, shows both gene amplification and increased expression in breast cancer, and is associated with poor clinical outcome [70]. In 1998, treatment with trastuzumab became available and was found to be beneficial to those women with amplified Her2 [71]. In addition the combination of paclitaxel with trastuzumab, following adjuvant chemotherapy was found to significantly improve the outcome for women positive for Her2 amplification, implicating a role for trastuzumab for early stage breast cancer [72]. More recently, our group [10] contributed in a clinical trial that compared erlotinib with a placebo for non-small-cell lung cancer. The trial demonstrated a survival benefit for erlotinib. Tumour-biopsy samples from participants in this trial were used to investigate whether responsiveness to erlotinib and its impact on survival were associated with expression by the tumour of epidermal growth factor receptor (EGFR) and EGFR gene amplification and mutations. EGFR expression was evaluated by immunohistochemical, mutation and paraffin interphase-FISH analysis in non-small-cell lung cancer specimens. Using univariate analyses, survival was longer in the erlotinib group than in the placebo group when EGFR was expressed (hazard ratio for death, 0.68; P = 0.02) or where there were a high number of copies of EGFR (hazard ratio, 0.44; P = 0.008). Using multivariate analyses, adenocarcinoma (P = 0.01), never having smoked (P < 0.001), and expression of EGFR (P = 0.03) were associated with an objective response. In multivariate analysis, survival after treatment with erlotinib was not influenced by the status of EGFR expression, the number of EGFR copies, or EGFR mutation, suggesting that among patients with non-small-cell lung cancer who receive erlotinib, the presence of an EGFR mutation may increase responsiveness to the agent, but it is not indicative of a survival benefit. 7. Translocation studies One of the landmark discoveries of cytogenetics was the identification of the Ph chromosome [73,74] as a recurrent aberration in CML. Later, other recurrent translocations were identified, particularly among the hematologicial malignancies and sarcomas (http://atlasgeneticsoncology.org// index.html) and more recently in prostate carcinoma [8,75]. The Ph chromosome results from the translocation between chromosomes 9 and 22 that causes the fusion of the ABL (9q34) and BCR (22q11) genes [76]. Imatinib therapy leads to major cytogenetic remission in a majority of CML patients as assessed by FISH as well as RT-PCR methods [77]. Translocation FISH employs a two-colour approach, whereby the genes involved in the translocation event are differentially labelled so that when hybridized together, the co-localization of both probes indicates the presence of the translocation. This is typically referred to as ‘‘fusion’’ FISH. Another way of identifying translocations is through the disruption of the gene. By this J. Bayani, J.A. Squire / Cancer Letters 249 (2007) 97–109 approach, a FISH probe spanning the entire gene of interest or spanning the known breakpoint, is hybridized to the target specimen. The appearance of a ‘‘split’’ signal indicates the disruption of the gene. Although a single colour FISH approach can be used, typically these ‘‘break-apart’’ assays uses a two-colour probe set with one probe on the 5 0 side of the breakpoint and the other probe on the 3 0 side of the breakpoint. The BCR/ABL translocation makes use of the fusion-FISH approach which is routinely used in cancer cytogenetics laboratories for diagnosis and residual disease monitoring. Interestingly, atypical FISH patterns using the dual-colour probe strategy led to the identification of a submicroscopic deletion of the 5 0 region of ABL and the 3 0 region of the BCR genes on the 9q(+) chromosome by our group [78]. It was determined that the CML patients with deletions had a shorter survival time and a high relapse rate following bone marrow transplant. In addition, because deletions are associated with both Ph(+) CML and ALL, it seemed probable that other leukemiaassociated genomic rearrangements may also have submicroscopic deletions. This was confirmed by the detection of deletions of the 3 0 regions of the CBFB and the MLL genes in AML M4 patients with inv(16) and in patients with ALL and AML associated with MLL gene translocations, respectively. Recently, Tomlins et al. [8] made the exciting discovery of recurrent translocations in prostate cancer involving the TMPRSS2 gene on chromosome 21q21.2 to members of the ETS transcription factor genes ERG on chromosome 21q21.2 or ETV1 on 103 chromosome 7p21.2. The significance of such a discovery is the fact that this is the first example of a specific recurrent translocation in carcinomas. To date, specific translocations have been identified for the hematological malignancies [6] and for a handful of sarcomas [5]. The finding of a recurrent translocation in a carcinoma leads to the speculation that there may be others. This initial finding by Tomlins was confirmed by our laboratory [75] using a three-colour FISH strategy that confirmed the fusion event, and that the TMPRSS2/ERG fusion may be accompanied by a small hemizygous sequence deletion on chromosome 21 between ERG and TMPRSS2 genes (Fig. 5). Since these initial findings, other ETS family genes have been identified as translocation partners [7,79] The specific biological role of this translocation and its variants is still unclear, however studies have already been conducted to determine the relationship between the presence of the translocation, and its specific isoform and expression level, to clinical outcome [80]. Wang et al. [80] found that a significant variation in the alternatively spliced isoforms was expressed in different cancers and that expression of an isoform, in which the native ATG in exon 2 of the TMPRSS2 gene is in frame with exon 4 of the ERG gene, was associated with clinical and pathologic variables of aggressive disease. Expression of other isoforms, in which the native ERG ATG in exon 3 was the first in-frame ATG, was associated with seminal vesicle invasion, which is correlated with poor outcome following radical prostatectomy. Cancers not expressing these isoforms tended to express higher levels of fusion mRNAs, and in this Fig. 5. The TMPRSS2/ERG translocation in prostate cancer. Shown is an example of interphase FISH analysis on paraffin section using a three-colour FISH approach detecting the deletion of the 3 0 TMPRSS2 gene, resulting in the fusion of ERG to the 5 0 TMPRSS2 as described previously by Yoshimoto et al. [75]. In this strategy, the ERG gene is labelled in red, the 3 0 end of TMPRSS2 is labelled in green, and the 5 0 TMPRSS2 is labelled in aqua. In the normal chromosome 21, the genes are oriented as shown in the left ideogram from centromere to telomere and the resulting FISH pattern shows all three signals in close proximity to each other. The fusion results from the deletion of the green probe, bringing the red ERG gene adjacent to the blue 5 0 TMPRSS2. In this particular case, two fusions were detected in this cell, with one normal. 104 J. Bayani, J.A. Squire / Cancer Letters 249 (2007) 97–109 group, higher expression levels of fusion mRNA were present in cancers with early prostate-specific antigen recurrence. 8. Chromosomal instability (CIN) studies Chromosomal instability (CIN) refers to the rate of chromosomal gain and loss as well as the presence of structural rearrangements [81]. The presence of CIN is believed to be the driving force of carcinogenesis, although this is the subject of continuing debate [82–87]. However, it is evident that CIN provides the necessary genomic variation for cell selection and for cancer cells to develop a growth advantage [83]. The mechanisms that drive the formation of complex chromosomal aberrations or copy number changes are influenced by sequence structure, DNA conformation, aberrations in DNA damage repair as well as telomere and mitotic segregation dysfunction (reviewed by Bayani et al. [88]). Thus, the detection of CIN in cancer may be an important tool for assessing disease progression, aggressiveness and disease resistance (Figs. 2 and 6). In neuroblastoma for example, it has been demonstrated that the presence of a di-tetraploid was associated with a significantly worse prognosis and overall survival at 4 years in comparison with near triploid tumours [89]. In ovarian cancer it has been demonstrated that carcinoma develops apparently through 2 pathways – one characterized by +7, +8q, and +12, and one by 6q- and 1q-. Moreover, at least three phases of karyotypic evolution were also identified [90]. At the early stages, Phase I, the karyotypic evolution seems to proceed though step-wise acquisition of changes. The transition to Phase II showed signs of an increased chromosomal instability, most probably caused by extensive telomere crisis and the onset of breakage-fusion-bridge (BFB) cycles. This process was linked to the presence of imbalances characteristic for the 6q-/1qpathway. The transition to Phase III involved triploidization and was also linked to the presence of the 6q-/1q-pathway. Because of the metaphaseand array-CGH based findings in various neoplasms (http://www.progenetix.de/~pgscripts/progenetix/ Aboutprogenetix.html), genomic signatures with characteristic gains and losses of chromosomes/ chromosomal regions have enabled investigators to use these combinations of gain/loss/amplification to screen specimens for aspects such as cancer risk [91,92] prognosis [93], progression and metastasis[94–96], as well as residual disease [97]. In addition to changes in chromosome number and ploidy, the assessment of telomere length by FISH has become an increasingly useful tool, although there are currently few studies applied to large patient cohorts. Telomeres are complex nucleoprotein structures located at the ends of linear chromosomes and are critical for maintaining genome integrity [98]. In the absence of telomerase, telomeres progressively shorten. The loss of telomere capping function resulting in dysfunctional telomeres is one of the telomere-mediated mechanisms promoting genomic instability. Excessive telomere shortening has been shown to lead to BFB cycles and, eventually to generalized genome instability, leading to either cell death or crisis. Only cells that have acquired a telomere maintenance Fig. 6. Detecting CIN through interphase FISH. Shown in the left panel is an example of multi-colour interphase FISH analysis using a 4colour probe cocktail for centromere 3 (red), centromere 7 (green), centromere 17 (aqua) and a locus-specific probe for the gene p16 (yellow/gold), to detect levels of aneuploidy and intratumoural heterogenetity. The right panel shown the hybridization of a tissue section with telomere-specific probes (red), from which the signal intensities can be quantified (QFISH). The shortening or stabling of telomere length has been correlated to various stages of tumour progression and may have some bearing on response to treatment and outcome. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this paper.) J. Bayani, J.A. Squire / Cancer Letters 249 (2007) 97–109 mechanism can escape from this crisis. Telomere length analysis studies [99–101] have revealed a relationship between telomere length and karyotypic complexity, such that cells with shorter telomeres possessed more structurally complex chromosomal aberrations and chromosomal instability. Telomere FISH studies, also known as QFISH (Fig. 6), involve the use of telomere specific probes, which can be quantified for both average signal intensity within an interphase cell, or for individual signal intensity on a chromosome in a metaphase spread [102]. In a study of spontaneous tumour regression of pediatric low-grade gliomas (PLGG) [103], it was determined that younger PLGG patients, who exhibit more aggressive and frequently recurrent tumours, had significantly longer telomeres than older ones (P = .00014). Tumours with a terminal restriction fragment length <7.5 did not recur, whereas the presence of longer telomeres (>8.0) conferred a high likelihood of late recurrences in PLGG, implicating a plausible biologic mechanism to explain the tendency of PLGG to exhibit growth arrest and spontaneous regression. Telomere studies by our laboratory in prostate cancer [104] showed that a significant decrease in telomere length was shown in both HPIN and CaP in comparison with normal epithelium, and that elevated rates of aneusomy suggested that increased levels of chromosomal aberrations were associated with decreased telomere length. In addition, it was revealed that multiple foci of HPIN exhibited a heterogeneous overall reduction of telomere length, which was more evident in the histologic regions of the prostate containing CaP. This implicates telomere erosion as a consistent feature of Cap oncogenesis and a marker for disease progression and increasing genomic instability. 9. Conclusions and perspectives Treatment failure is often associated with tumours acquiring new genomic alterations that lead to the evolution of more aggressive malignant clones. Systematic molecular cytogenetic and FISH-based analyses can now contribute to the improved assessment of cell-to-cell variation and the complexity of numerical and structural alterations in clinical trials by using TMA and automated imaging platforms. The FISH technologies we have outlined above are highly specific and can be used to determine the clinical impact of a genomic change 105 that may influence a therapeutic pathway of response. Understanding the chromosomal mechanisms driving tumour progression, and relating these findings to the observed dynamic behaviour of cancer cells may enable a better prediction of patient outcome and response to treatment. Acknowledgements The authors wish to acknowledge Paula Marrano, Maisa Yoshimoto, Olga Ludkovski, Maria Zielenska, Anthony Joshua, Elena Kolomietz, Jana Paderova, and Shamini Selvarajah for their technical and conceptual contributions. This work was supported by the Ontario Cancer Biomarker Network with funds from the Ontario Cancer Research Network. References [1] G.J. Kelloff, S.M. Lippman, A.J. Dannenberg, C.C. Sigman, H.L. Pearce, B.J. Reid, E. Szabo, V.C. Jordan, M.R. Spitz, G.B. Mills, V.A. Papadimitrakopoulou, R. Lotan, B.B. Aggarwal, R.S. Bresalier, J. Kim, B. Arun, K.H. Lu, M.E. Thomas, H.E. Rhodes, M.A. Brewer, M. Follen, D.M. Shin, H.L. Parnes, J.M. Siegfried, A.A. Evans, W.J. Blot, W.H. Chow, P.L. Blount, C.C. Maley, K.K. Wang, S. Lam, J.J. Lee, S.M. Dubinett, P.F. Engstrom, F.L. Meyskens Jr., J. O’Shaughnessy, E.T. Hawk, B. Levin, W.G. Nelson, W.K. Hong, Progress in chemoprevention drug development: the promise of molecular biomarkers for prevention of intraepithelial neoplasia and cancer – a plan to move forward, Clin. Cancer. Res. 12 (2006) 3661–3697. [2] G.B. Mills, Y. Lu, E.C. Kohn, Linking molecular therapeutics to molecular diagnostics: inhibition of the FRAP/ RAFT/TOR component of the PI3K pathway preferentially blocks PTEN mutant cells in vitro and in vivo, Proc. Natl. Acad. Sci. USA 98 (2001) 10031–10033. [3] D. Hanahan, R.A. Weinberg, The hallmarks of cancer, Cell 100 (2000) 57–70. [4] T.H. Rabbitts, Chromosomal translocations in human cancer, Nature 372 (1994) 143–149. [5] J.L. Bennicelli, F.G. Barr, Chromosomal translocations and sarcomas, Curr. Opin. Oncol. 14 (2002) 412–419. [6] J.D. Rowley, Chromosome translocations: dangerous liaisons revisited, Nat. Rev. Cancer 1 (2001) 245–250. [7] S.A. Tomlins, R. Mehra, D.R. Rhodes, L.R. Smith, D. Roulston, B.E. Helgeson, X. Cao, J.T. Wei, M.A. Rubin, R.B. Shah, A.M. Chinnaiyan, TMPRSS2:ETV4 gene fusions define a third molecular subtype of prostate cancer, Cancer Res. 66 (2006) 3396–3400. [8] S.A. Tomlins, D.R. Rhodes, S. Perner, S.M. Dhanasekaran, R. Mehra, X.W. Sun, S. Varambally, X. Cao, J. Tchinda, R. Kuefer, C. Lee, J.E. Montie, R.B. Shah, K.J. Pienta, M.A. Rubin, A.M. Chinnaiyan, Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer, Science 310 (2005) 644–648. 106 J. Bayani, J.A. Squire / Cancer Letters 249 (2007) 97–109 [9] D.G. Albertson, Gene amplification in cancer, Trends Genet. 22 (2006) 447–455. [10] M.S. Tsao, A. Sakurada, J.C. Cutz, C.Q. Zhu, S. KamelReid, J. Squire, I. Lorimer, T. Zhang, N. Liu, M. Daneshmand, P. Marrano, G. da Cunha Santos, A. Lagarde, F. Richardson, L. Seymour, M. Whitehead, K. Ding, J. Pater, F.A. Shepherd, Erlotinib in lung cancer – molecular and clinical predictors of outcome, N. Engl. J. Med. 353 (2005) 133–144. [11] P.K. Leong, P. Thorner, H. Yeger, K. Ng, Z. Zhang, J. Squire, Detection of MYCN gene amplification and deletions of chromosome 1p in neuroblastoma by in situ hybridization using routine histologic sections, Lab. Invest. 69 (1993) 43–50. [12] M. Yoshimoto, J. Bayani, P.A. Nuin, N.S. Silva, S. Cavalheiro, J.N. Stavale, J.A. Andrade, M. Zielenska, J.A. Squire, S.R. de Toledo, Metaphase and array comparative genomic hybridization: unique copy number changes and gene amplification of medulloblastomas in South America, Cancer Genet. Cytogenet. 170 (2006) 40–47. [13] S.B. Baylin, DNA methylation and gene silencing in cancer, Nat. Clin. Pract. Oncol. 2 (Suppl. 1) (2005) S4–S11. [14] G.T. Rudkin, B.D. Stollar, High resolution detection of DNA-RNA hybrids in situ by indirect immunofluorescence, Nature 265 (1977) 472–473. [15] P. Lichter, T. Cremer, J. Borden, L. Manuelidis, D.C. Ward, Delineation of individual human chromosomes in metaphase and interphase cells by in situ suppression hybridization using recombinant DNA libraries, Hum. Genet. 80 (1988) 224–234. [16] J. Squire, E.F. Meurs, K.L. Chong, N.A. McMillan, A.G. Hovanessian, B.R. Williams, Localization of the human interferon-induced, ds-RNA activated p68 kinase gene (PRKR) to chromosome 2p21-p22, Genomics 16 (1993) 768–770. [17] S.N. Sait, N.J. Nowak, P. Singh-Kahlon, R. Weksberg, J. Squire, T.B. Shows, M.J. Higgins, Localization of Beckwith-Wiedemann and rhabdoid tumor chromosome rearrangements to a defined interval in chromosome band 11p15.5, Genes Chromosomes Cancer 11 (1994) 97–105. [18] G.E. Hannigan, J. Bayani, R. Weksberg, B. Beatty, A. Pandita, S. Dedhar, J. Squire, Mapping of the gene encoding the integrin-linked kinase, ILK, to human chromosome 11p15.5-p15.4, Genomics 42 (1997) 177–179. [19] A. Kallioniemi, O.P. Kallioniemi, D. Sudar, D. Rutovitz, J.W. Gray, F. Waldman, D. Pinkel, Comparative genomic hybridization for molecular cytogenetic analysis of solid tumors, Science 258 (1992) 818–821. [20] D. Pinkel, D.G. Albertson, Comparative genomic hybridization, Annu. Rev. Genomics. Hum. Genet. 6 (2005) 331–354. [21] M.R. Speicher, S. Gwyn Ballard, D.C. Ward, Karyotyping human chromosomes by combinatorial multi-fluor FISH, Nat. Genet. 12 (1996) 368–375. [22] E. Schrock, S. du Manoir, T. Veldman, B. Schoell, J. Wienberg, M.A. Ferguson-Smith, Y. Ning, D.H. Ledbetter, I. Bar-Am, D. Soenksen, Y. Garini, T. Ried, Multicolor spectral karyotyping of human chromosomes, Science 273 (1996) 494–497. [23] J.M. Bayani, J.A. Squire, Applications of SKY in cancer cytogenetics, Cancer Invest. 20 (2002) 373–386. [24] J. Bayani, J.A. Squire, Advances in the detection of chromosomal aberrations using spectral karyotyping, Clin. Genet. 59 (2001) 65–73. [25] M.R. Speicher, N.P. Carter, The new cytogenetics: blurring the boundaries with molecular biology, Nat. Rev. Genet. 6 (2005) 782–792. [26] T. Braunschweig, J.Y. Chung, S.M. Hewitt, Tissue microarrays: bridging the gap between research and the clinic, Expert Rev. Proteomics. 2 (2005) 325–336. [27] A.M. Grigoryan, E.R. Dougherty, J. Kononen, L. Bubendorf, G. Hostetter, O. Kallioniemi, Morphological spot counting from stacked images for automated analysis of gene copy numbers by fluorescence in situ hybridization, J. Biomed Opt. 7 (2002) 109–122. [28] K.A. Jaeckle, K.V. Ballman, R.D. Rao, R.B. Jenkins, J.C. Buckner, Current strategies in treatment of oligodendroglioma: evolution of molecular signatures of response, J. Clin. Oncol. 24 (2006) 1246–1252. [29] F. Westermann, M. Schwab, Genetic parameters of neuroblastomas, Cancer Lett. 184 (2002) 127–147. [30] S.R. Fink, S.A. Smoley, K.J. Stockero, S.F. Paternoster, E.C. Thorland, D.L. Van Dyke, T.D. Shanafelt, C.S. Zent, T.G. Call, N.E. Kay, G.W. Dewald, Loss of TP53 is due to rearrangements involving chromosome region 17p10 approximately p12 in chronic lymphocytic leukemia, Cancer Genet. Cytogenet. 167 (2006) 177–181. [31] D.D. Dao, J.R. Sawyer, J. Epstein, R.G. Hoover, B. Barlogie, G. Tricot, Deletion of the retinoblastoma gene in multiple myeloma, Leukemia 8 (1994) 1280–1284. [32] A. Namazie, S. Alavi, N. Aghamohammadi, M. Aghamohammadi, T.C. Calcaterra, M.B. Wang, E.S. Srivatsan, Fluorescence in situ hybridization for detecting TP16 MTS1/CDK41 gene deletions in squamous cell carcinoma of the head and neck, Cancer Genet. Cytogenet. 141 (2003) 49–55. [33] M. Cully, H. You, A.J. Levine, T.W. Mak, Beyond PTEN mutations: the PI3K pathway as an integrator of multiple inputs during tumorigenesis, Nat. Rev. Cancer 6 (2006) 184–192. [34] A. Besson, S.M. Robbins, V.W. Yong, PTEN/MMAC1/ TEP1 in signal transduction and tumorigenesis, Eur. J. Biochem. 263 (1999) 605–611. [35] R. Parsons, Human cancer, PTEN and the PI-3 kinase pathway, Semin. Cell Dev. Biol. 15 (2004) 171–176. [36] S. Ramaswamy, N. Nakamura, F. Vazquez, D.B. Batt, S. Perera, T.M. Roberts, W.R. Sellers, Regulation of G1 progression by the PTEN tumor suppressor protein is linked to inhibition of the phosphatidylinositol 3-kinase/ Akt pathway, Proc. Natl. Acad. Sci. USA 96 (1999) 2110– 2115. [37] T. Ohigashi, R. Mizuno, J. Nakashima, K. Marumo, M. Murai, Inhibition of Wnt signaling downregulates Akt activity and induces chemosensitivity in PTEN-mutated prostate cancer cells, Prostate 62 (2005) 61–68. [38] I. Sansal, W.R. Sellers, The biology and clinical relevance of the PTEN tumor suppressor pathway, J. Clin. Oncol. 22 (2004) 2954–2963. [39] I. Vivanco, C.L. Sawyers, The phosphatidylinositol 3Kinase AKT pathway in human cancer, Nat. Rev. Cancer 2 (2002) 489–501. [40] S.R. Datta, H. Dudek, X. Tao, S. Masters, H. Fu, Y. Gotoh, M.E. Greenberg, Akt phosphorylation of BAD J. Bayani, J.A. Squire / Cancer Letters 249 (2007) 97–109 [41] [42] [43] [44] [45] [46] [47] [48] [49] [50] [51] [52] [53] [54] couples survival signals to the cell-intrinsic death machinery, Cell 91 (1997) 231–241. M.H. Cardone, N. Roy, H.R. Stennicke, G.S. Salvesen, T.F. Franke, E. Stanbridge, S. Frisch, J.C. Reed, Regulation of cell death protease caspase-9 by phosphorylation, Science 282 (1998) 1318–1321. M. Ashcroft, R.L. Ludwig, D.B. Woods, T.D. Copeland, H.O. Weber, E.J. MacRae, K.H. Vousden, Phosphorylation of HDM2 by Akt, Oncogene 21 (2002) 1955–1962. J.R. Graff, B.W. Konicek, A.M. McNulty, Z. Wang, K. Houck, S. Allen, J.D. Paul, A. Hbaiu, R.G. Goode, G.E. Sandusky, R.L. Vessella, B.L. Neubauer, Increased AKT activity contributes to prostate cancer progression by dramatically accelerating prostate tumor growth and diminishing p27Kip1 expression, J. Biol. Chem. 275 (2000) 24500–24505. A. Brunet, A. Bonni, M.J. Zigmond, M.Z. Lin, P. Juo, L.S. Hu, M.J. Anderson, K.C. Arden, J. Blenis, M.E. Greenberg, Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor, Cell 96 (1999) 857–868. C.J. Richardson, M. Broenstrup, D.C. Fingar, K. Julich, B.A. Ballif, S. Gygi, J. Blenis, SKAR is a specific target of S6 kinase 1 in cell growth control, Curr. Biol. 14 (2004) 1540–1549. B. Stiles, M. Groszer, S. Wang, J. Jiao, H. Wu, PTENless means more, Dev. Biol. 273 (2004) 175–184. A. Di Cristofano, B. Pesce, C. Cordon-Cardo, P.P. Pandolfi, Pten is essential for embryonic development and tumour suppression, Nat. Genet. 19 (1998) 348–355. K.P. Porkka, T. Visakorpi, Molecular mechanisms of prostate cancer, Eur. Urol. 45 (2004) 683–691. M. Yoshimoto, J.C. Cutz, P.A. Nuin, A.M. Joshua, J. Bayani, A.J. Evans, M. Zielenska, J.A. Squire, Interphase FISH analysis of PTEN in histologic sections shows genomic deletions in 68% of primary prostate cancer and 23% of high-grade prostatic intra-epithelial neoplasias, Cancer Genet. Cytogenet. 169 (2006) 128–137. M.E. McMenamin, P. Soung, S. Perera, I. Kaplan, M. Loda, W.R. Sellers, Loss of PTEN expression in paraffin-embedded primary prostate cancer correlates with high Gleason score and advanced stage, Cancer Res. 59 (1999) 4291–4296. E. Gebhart, Double minutes, cytogenetic equivalents of gene amplification, in human neoplasia - a review, Clin. Transl. Oncol. 7 (2005) 477–485. J.A. Squire, J. Pei, P. Marrano, B. Beheshti, J. Bayani, G. Lim, L. Moldovan, M. Zielenska, High-resolution mapping of amplifications and deletions in pediatric osteosarcoma by use of CGH analysis of cDNA microarrays, Genes Chromosomes Cancer 38 (2003) 215–225. M. Zielenska, P. Marrano, P. Thorner, J. Pei, B. Beheshti, M. Ho, J. Bayani, Y. Liu, B.C. Sun, J.A. Squire, X.S. Hao, High-resolution cDNA microarray CGH mapping of genomic imbalances in osteosarcoma using formalin-fixed paraffin-embedded tissue, Cytogenet. Genome Res. 107 (2004) 77–82. M. Gallucci, R. Merola, A. Farsetti, G. Orlandi, S. Sentinelli, P. De Carli, C. Leonardo, P. Carlini, F. Guadagni, I. Sperduti, A.M. Cianciulli, Cytogenetic profiles as additional markers to pathological features in clinically localized prostate carcinoma, Cancer Lett. 237 (2006) 76–82. 107 [55] R.B. Jenkins, J. Qian, M.M. Lieber, D.G. Bostwick, Detection of c-myc oncogene amplification and chromosomal anomalies in metastatic prostatic carcinoma by fluorescence in situ hybridization, Cancer Res. 57 (1997) 524–531. [56] H. Sato, S. Minei, T. Hachiya, T. Yoshida, Y. Takimoto, Fluorescence in situ hybridization analysis of c-myc amplification in stage TNM prostate cancer in Japanese patients, Int. J. Urol. 13 (2006) 761–766. [57] Y. Wang, H. Xue, J.C. Cutz, J. Bayani, N.R. Mawji, W.G. Chen, L.J. Goetz, S.W. Hayward, M.D. Sadar, C.B. Gilks, P.W. Gout, J.A. Squire, G.R. Cunha, Y.Z. Wang, An orthotopic metastatic prostate cancer model in SCID mice via grafting of a transplantable human prostate tumor line, Lab. Invest. 85 (2005) 1392–1404. [58] P.F. Ambros, I.M. Ambros, R. Kerbl, A. Luegmayr, S. Rumpler, R. Ladenstein, G. Amann, H. Kovar, E. Horcher, B. De Bernardi, J. Michon, H. Gadner, Intratumoural heterogeneity of 1p deletions and MYCN amplification in neuroblastomas, Med. Pediatr. Oncol. 36 (2001) 1–4. [59] J.A. Squire, P. Thorner, P. Marrano, D. Parkinson, Y.K. Ng, B. Gerrie, S. Chilton-Macneill, M. Zielenska, Identification of MYCN Copy Number Heterogeneity by Direct FISH Analysis of Neuroblastoma Preparations, Mol. Diagn. 1 (1996) 281–289. [60] J.M. Lamont, C.S. McManamy, A.D. Pearson, S.C. Clifford, D.W. Ellison, Combined histopathological and molecular cytogenetic stratification of medulloblastoma patients, Clin. Cancer Res. 10 (2004) 5482–5493. [61] C.G. Eberhart, J. Kratz, Y. Wang, K. Summers, D. Stearns, K. Cohen, C.V. Dang, P.C. Burger, Histopathological and molecular prognostic markers in medulloblastoma: c-myc, N-myc, TrkC, and anaplasia, J. Neuropathol. Exp. Neurol. 63 (2004) 441–449. [62] N. Aldosari, S.H. Bigner, P.C. Burger, L. Becker, J.L. Kepner, H.S. Friedman, R.E. McLendon, MYCC and MYCN oncogene amplification in medulloblastoma. A fluorescence in situ hybridization study on paraffin sections from the Children’s Oncology Group, Arch. Pathol. Lab Med. 126 (2002) 540–544. [63] F. Cappuzzo, F.R. Hirsch, E. Rossi, S. Bartolini, G.L. Ceresoli, L. Bemis, J. Haney, S. Witta, K. Danenberg, I. Domenichini, V. Ludovini, E. Magrini, V. Gregorc, C. Doglioni, A. Sidoni, M. Tonato, W.A. Franklin, L. Crino, P.A. Bunn Jr., M. Varella-Garcia, Epidermal growth factor receptor gene and protein and gefitinib sensitivity in non-small-cell lung cancer, J. Natl. Cancer Inst. 97 (2005) 643–655. [64] F.R. Hirsch, M. Varella-Garcia, P.A. Bunn Jr., M.V. Di Maria, R. Veve, R.M. Bremmes, A.E. Baron, C. Zeng, W.A. Franklin, Epidermal growth factor receptor in nonsmall-cell lung carcinomas: correlation between gene copy number and protein expression and impact on prognosis, J. Clin. Oncol. 21 (2003) 3798–3807. [65] L.J. Layfield, C. Willmore, S. Tripp, C. Jones, R.L. Jensen, Epidermal growth factor receptor gene amplification and protein expression in glioblastoma multiforme: prognostic significance and relationship to other prognostic factors, Appl. Immunohistochem. Mol. Morphol. 14 (2006) 91–96. [66] B.K. Rasheed, R.N. Wiltshire, S.H. Bigner, D.D. Bigner, Molecular pathogenesis of malignant gliomas, Curr. Opin. Oncol. 11 (1999) 162–167. 108 J. Bayani, J.A. Squire / Cancer Letters 249 (2007) 97–109 [67] W. Hanna, Testing for HER2 status, Oncology 61 (Suppl. 2) (2001) 22–30. [68] J.S. Winston, J. Ramanaryanan, E. Levine, HER-2/neu evaluation in breast cancer are we there yet?, Am J. Clin. Pathol. 121 (Suppl.) (2004) S33–S49. [69] D.G. Hicks, R.R. Tubbs, Assessment of the HER2 status in breast cancer by fluorescence in situ hybridization: a technical review with interpretive guidelines, Hum. Pathol. 36 (2005) 250–261. [70] D.J. Slamon, G.M. Clark, S.G. Wong, W.J. Levin, A. Ullrich, W.L. McGuire, Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene, Science 235 (1987) 177–182. [71] J.S. Ross, J.A. Fletcher, K.J. Bloom, G.P. Linette, J. Stec, W.F. Symmans, L. Pusztai, G.N. Hortobagyi, Targeted therapy in breast cancer: the HER-2/neu gene and protein, Mol. Cell. Proteomics 3 (2004) 379–398. [72] E.H. Romond, E.A. Perez, J. Bryant, V.J. Suman, C.E. Geyer Jr., N.E. Davidson, E. Tan-Chiu, S. Martino, S. Paik, P.A. Kaufman, S.M. Swain, T.M. Pisansky, L. Fehrenbacher, L.A. Kutteh, V.G. Vogel, D.W. Visscher, G. Yothers, R.B. Jenkins, A.M. Brown, S.R. Dakhil, E.P. Mamounas, W.L. Lingle, P.M. Klein, J.N. Ingle, N. Wolmark, Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer, N. Engl. J. Med. 353 (2005) 1673–1684. [73] P.C. Nowell, D.A. Hungerford, Chromosome studies on normal and leukemic human leukocytes, J. Natl. Cancer Inst. 25 (1960) 85–109. [74] J.D. Rowley, Letter: A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining, Nature 243 (1973) 290–293. [75] M. Yoshimoto, A.M. Joshua, S. Chilton-Macneill, J. Bayani, S. Selvarajah, A.J. Evans, M. Zielenska, J.A. Squire, Three-color FISH analysis of TMPRSS2/ ERG fusions in prostate cancer indicates that genomic microdeletion of chromosome 21 is associated with rearrangement, Neoplasia 8 (2006) 465–469. [76] A.P. Landstrom, A. Tefferi, Fluorescent in situ hybridization in the diagnosis, prognosis, and treatment monitoring of chronic myeloid leukemia, Leuk. Lymphoma 47 (2006) 397–402. [77] J. Goldman, Monitoring minimal residual disease in BCRABL-positive chronic myeloid leukemia in the imatinib era, Curr. Opin. Hematol. 12 (2005) 33–39. [78] E. Kolomietz, J. Al-Maghrabi, S. Brennan, J. Karaskova, S. Minkin, J. Lipton, J.A. Squire, Primary chromosomal rearrangements of leukemia are frequently accompanied by extensive submicroscopic deletions and may lead to altered prognosis, Blood 97 (2001) 3581–3588. [79] M.J. Soller, M. Isaksson, P. Elfving, W. Soller, R. Lundgren, I. Panagopoulos, Confirmation of the high frequency of the TMPRSS2/ERG fusion gene in prostate cancer, Genes Chromosomes Cancer 45 (2006) 717–719. [80] J. Wang, Y. Cai, C. Ren, M. Ittmann, Expression of variant TMPRSS2/ERG fusion messenger RNAs is associated with aggressive prostate cancer, Cancer Res. 66 (2006) 8347– 8351. [81] H. Rajagopalan, M.A. Nowak, B. Vogelstein, C. Lengauer, The significance of unstable chromosomes in colorectal cancer, Nat. Rev. Cancer 3 (2003) 695–701. [82] J. Breivik, G. Gaudernack, Genomic instability, DNA methylation, and natural selection in colorectal carcinogenesis, Semin. Cancer Biol. 9 (1999) 245–254. [83] P. Duesberg, R. Li, A. Fabarius, R. Hehlmann, Aneuploidy and cancer: from correlation to causation, Contrib. Microbiol. 13 (2006) 16–44. [84] R. Li, A. Sonik, R. Stindl, D. Rasnick, P. Duesberg, Aneuploidy vs. gene mutation hypothesis of cancer: recent study claims mutation but is found to support aneuploidy, Proc. Natl. Acad. Sci. USA 97 (2000) 3236–3241. [85] L.A. Loeb, C.F. Springgate, N. Battula, Errors in DNA replication as a basis of malignant changes, Cancer Res. 34 (1974) 2311–2321. [86] I.M. Shih, W. Zhou, S.N. Goodman, C. Lengauer, K.W. Kinzler, B. Vogelstein, Evidence that genetic instability occurs at an early stage of colorectal tumorigenesis, Cancer Res. 61 (2001) 818–822. [87] I. Tomlinson, W. Bodmer, Selection, the mutation rate and cancer: ensuring that the tail does not wag the dog, Nat. Med. 5 (1999) 11–12. [88] J. Bayani, S. Selvarajah, G. Maire, B. Vukovic, K. AlRohmaih, M. Zielenska, J.A. Squire, Genomic Mechanisms and measurement of structural and numerical instability in cancer cells, Semin. Cancer Biol. 17 (2007) 5–18. [89] R. Ladenstein, I.M. Ambros, U. Potschger, G. Amann, C. Urban, F.M. Fink, K. Schmitt, R. Jones, M. Slociak, F. Schilling, J. Ritter, F. Berthold, H. Gadner, P.F. Ambros, Prognostic significance of DNA di-tetraploidy in neuroblastoma, Med. Pediatr. Oncol. 36 (2001) 83–92. [90] M. Hoglund, D. Gisselsson, G.B. Hansen, T. Sall, F. Mitelman, Ovarian carcinoma develops through multiple modes of chromosomal evolution, Cancer Res. 63 (2003) 3378–3385. [91] N. Sneige, B. Liu, G. Yin, Y. Gong, B.K. Arun, Correlation of cytologic findings and chromosomal instability detected by fluorescence in situ hybridization in breast fine-needle aspiration specimens from women at high risk for breast cancer, Mod. Pathol. 19 (2006) 622–629. [92] S. Krishnamurthy, L. Zhao, K. Hayes, A.B. Glassman, M. Cristofanilli, S.E. Singletary, K.K. Hunt, H.M. Kuerer, N. Sneige, Feasibility and utility of using chromosomal aneusomy to further define the cytologic categories in nipple aspirate fluid specimens: a preliminary study, Cancer 102 (2004) 322–327. [93] C. Mian, M. Lodde, E. Comploj, L. Lusuardi, S. Palermo, M. Mian, K. Maier, A. Pycha, Multiprobe fluorescence in situ hybridisation: prognostic perspectives in superficial bladder cancer, J. Clin. Pathol. 59 (2006) 984–987. [94] F. Farabegoli, D. Santini, C. Ceccarelli, M. Taffurelli, D. Marrano, N. Baldini, Clone heterogeneity in diploid and aneuploid breast carcinomas as detected by FISH, Cytometry 46 (2001) 50–56. [95] R. Spitz, D.R. Betts, T. Simon, M. Boensch, J. Oestreich, F.K. Niggli, K. Ernestus, F. Berthold, B. Hero, Favorable outcome of triploid neuroblastomas: a contribution to the special oncogenesis of neuroblastoma, Cancer Genet. Cytogenet. 167 (2006) 51–56. [96] S. Sen, H. Zhou, R.D. Zhang, D.S. Yoon, F. Vakar-Lopez, S. Ito, F. Jiang, D. Johnston, H.B. Grossman, A.C. Ruifrok, R.L. Katz, W. Brinkley, B. Czerniak, Amplification/ overexpression of a mitotic kinase gene in human bladder cancer, J. Natl. Cancer Inst. 94 (2002) 1320–1329. J. Bayani, J.A. Squire / Cancer Letters 249 (2007) 97–109 [97] T. Zellweger, G. Benz, G. Cathomas, M.J. Mihatsch, T. Sulser, T.C. Gasser, L. Bubendorf, Multi-target fluorescence in situ hybridization in bladder washings for prediction of recurrent bladder cancer, Int. J. Cancer 119 (2006) 1660–1665. [98] J.W. Shay, W.E. Wright, Telomeres and telomerase: implications for cancer and aging, Radiat. Res. 155 (2001) 188–193. [99] B. Vukovic, B. Beheshti, P.C. Park, G. Lim, J. Bayani, M. Zielenska, J.A. Squire, Correlating breakage-fusion-bridge events with the overall chromosomal instability and in vitro karyotypic evolution in prostate cancer, Cytogenet. Genome Res. (2007), in press. [100] D. Gisselsson, L. Gorunova, M. Hoglund, N. Mandahl, P. Elfving, Telomere shortening and mitotic dysfunction generate cytogenetic heterogeneity in a subgroup of renal cell carcinomas, Br. J. Cancer 91 (2004) 327–332. [101] D. Gisselsson, M. Lv, S.W. Tsao, C. Man, C. Jin, M. Hoglund, Y.L. Kwong, Y. Jin, Telomere-mediated 109 mitotic disturbances in immortalized ovarian epithelial cells reproduce chromosomal losses and breakpoints from ovarian carcinoma, Genes Chromosomes Cancer 42 (2005) 22–33. [102] J.N. O’Sullivan, J.C. Finley, R.A. Risques, W.T. Shen, K.A. Gollahon, A.H. Moskovitz, S. Gryaznov, C.B. Harley, P.S. Rabinovitch, Telomere length assessment in tissue sections by quantitative FISH: image analysis algorithms, Cytometry A 58 (2004) 120–131. [103] U. Tabori, B. Vukovic, M. Zielenska, C. Hawkins, I. Braude, J. Rutka, E. Bouffet, J. Squire, D. Malkin, The role of telomere maintenance in the spontaneous growth arrest of pediatric low-grade gliomas, Neoplasia 8 (2006) 136–142. [104] B. Vukovic, P.C. Park, J. Al-Maghrabi, B. Beheshti, J. Sweet, A. Evans, J. Trachtenberg, J. Squire, Evidence of multifocality of telomere erosion in high-grade prostatic intraepithelial neoplasia (HPIN) and concurrent carcinoma, Oncogene 22 (2003) 1978–1987.