cytogenomics of cancers from chromosome to sequence

14
Review Cytogenomics of cancers: From chromosome to sequence Alain Bernheim* Laboratoire de Ge ´nomique Cellulaire des Cancers, INSERM U985 and Molecular Pathology, Biopathology Department, Institut de Cance ´rologie Gustave Roussy, 39 rue Camille Desmoulins, 94805 Paris-Villejuif Cedex, France ARTICLE INFO Article history: Received 20 May 2010 Accepted 2 June 2010 Available online 11 June 2010 Keywords: Cancer Chromosome abnormalities Cytogenetics Oncogenesis ABSTRACT The role of acquired chromosomal rearrangements in oncogenesis (cytogenomics) and tu- mor progression is now well established. These alterations are multiple and diverse and the products of these rearranged genes play an essential role in the transformation and growth of cancer cells. The validity of this assumption is demonstrated by the development of specific inhibitors or antibodies that eliminate tumoral cells by targeting some of these changes. Imatinib, an inhibitor of the tyrosine kinase ABL, the prototype of these targeting drugs, is yielding complete remissions in most CML patients. Knowledge of chromosomal abnormalities is becoming an essential contribution to the diagnosis and prognosis of can- cers but also for monitoring minimal residual disease or relapse. The concept of the “cyto- genetic uniqueness” of each cancer has resulted in personalized treatment. This investigation will expound upon, besides the recurrent genomic alterations, the numerous products of perverted Darwinian selection at the cellular level. ª 2010 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved. 1. Introduction The Boveri hypothesis (Boveri, 1914) that cancer could be a chromosomal disease (Balmain, 2001) of the cell genome has been widely demonstrated by the cytogenetics and geno- mics of malignant diseases during the last fifty years. The seminal observations of the Ph chromosome in CML (Nowell and Hungerford, 1960; Rowley, 1973) and the correlation be- tween double minute chromosomes and genetic amplification are examples of how fruitful this approach has been. Acquired and monoclonal characteristics of most malignancies and the clonal evolution of tumor cell populations were described by cytogeneticists before the era of chromosome banding. Spe- cific chromosomal rearrangements, mainly translocations were described by banding and were found to be malignancy-specific (hematological or mesenchymal malig- nancies). Due to their presence in various cell types in bone marrow, they allowed us to demonstrate the coexistence of normal and malignant cells in the same tissue and led to the hypothesis of stem cell involvement in several malignancies (e.g. MDS or CML). Using the Ph chromosome as a marker of malignant cells, it was possible to show that it was present in all myeloid cells including B lymphocytes (Bernheim et al., 1981b). The recurrent breakpoints found in translocations in leu- kemia and lymphoma were enigmatic when gene mapping was in its infancy. The observation of variant translocations in Burkitt’s lymphoma allowed us to hypothesize that the spe- cific breakpoint of this proliferation was located on 8q24 (Bernheim et al., 1981a), near a “transforming gene” (Lenoir * Tel.: þ33 (0) 1 42 11 47 79; fax: þ33 (0) 1 42 11 52 60. E-mail address: [email protected] available at www.sciencedirect.com www.elsevier.com/locate/molonc 1574-7891/$ e see front matter ª 2010 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved. doi:10.1016/j.molonc.2010.06.003 MOLECULAR ONCOLOGY 4 (2010) 309 e322

Upload: alejandraoseguera

Post on 18-Nov-2015

6 views

Category:

Documents


0 download

DESCRIPTION

genetics

TRANSCRIPT

  • M O L E C U L A R O N C O L O G Y 4 ( 2 0 1 0 ) 3 0 9e3 2 2ava i lab le at www.sc ienced i rec t . com

    www.e lsev ie r . com/ loca te /moloncReview

    Cytogenomics of cancers: From chromosome to sequenceAlain Bernheim*

    Laboratoire de Genomique Cellulaire des Cancers, INSERM U985 and Molecular Pathology, Biopathology Department, Institut de Cancerologie

    Gustave Roussy, 39 rue Camille Desmoulins, 94805 Paris-Villejuif Cedex, FranceA R T I C L E I N F O

    Article history:

    Received 20 May 2010

    Accepted 2 June 2010

    Available online 11 June 2010

    Keywords:

    Cancer

    Chromosome abnormalities

    Cytogenetics

    Oncogenesis* Tel.: 33 (0) 1 42 11 47 79; fax: 33 (0) 1 4E-mail address: [email protected]

    1574-7891/$ e see front matter 2010 Federdoi:10.1016/j.molonc.2010.06.003A B S T R A C T

    The role of acquired chromosomal rearrangements in oncogenesis (cytogenomics) and tu-

    mor progression is now well established. These alterations are multiple and diverse and

    the products of these rearranged genes play an essential role in the transformation and

    growth of cancer cells. The validity of this assumption is demonstrated by the development

    of specific inhibitors or antibodies that eliminate tumoral cells by targeting some of these

    changes. Imatinib, an inhibitor of the tyrosine kinase ABL, the prototype of these targeting

    drugs, is yielding complete remissions in most CML patients. Knowledge of chromosomal

    abnormalities is becoming an essential contribution to the diagnosis and prognosis of can-

    cers but also for monitoring minimal residual disease or relapse. The concept of the cyto-

    genetic uniqueness of each cancer has resulted in personalized treatment. This

    investigation will expound upon, besides the recurrent genomic alterations, the numerous

    products of perverted Darwinian selection at the cellular level.

    2010 Federation of European Biochemical Societies.Published by Elsevier B.V. All rights reserved.1. Introduction malignancy-specific (hematological or mesenchymal malig-The Boveri hypothesis (Boveri, 1914) that cancer could be

    a chromosomal disease (Balmain, 2001) of the cell genome

    has been widely demonstrated by the cytogenetics and geno-

    mics of malignant diseases during the last fifty years. The

    seminal observations of the Ph chromosome in CML (Nowell

    and Hungerford, 1960; Rowley, 1973) and the correlation be-

    tween doubleminute chromosomes and genetic amplification

    are examples of how fruitful this approach has been. Acquired

    andmonoclonal characteristics of most malignancies and the

    clonal evolution of tumor cell populations were described by

    cytogeneticists before the era of chromosome banding. Spe-

    cific chromosomal rearrangements, mainly translocations

    were described by banding and were found to be2 11 52 60.

    ation of European Biochenancies). Due to their presence in various cell types in bone

    marrow, they allowed us to demonstrate the coexistence of

    normal and malignant cells in the same tissue and led to the

    hypothesis of stem cell involvement in several malignancies

    (e.g. MDS or CML). Using the Ph chromosome as a marker of

    malignant cells, it was possible to show that it was present

    in all myeloid cells including B lymphocytes (Bernheim

    et al., 1981b).

    The recurrent breakpoints found in translocations in leu-

    kemia and lymphoma were enigmatic when gene mapping

    was in its infancy. The observation of variant translocations

    in Burkitts lymphoma allowed us to hypothesize that the spe-

    cific breakpoint of this proliferation was located on 8q24

    (Bernheim et al., 1981a), near a transforming gene (Lenoirmical Societies. Published by Elsevier B.V. All rights reserved.

    mailto:[email protected]/locate/molonchttp://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003

  • M O L E C U L A R O N C O L O G Y 4 ( 2 0 1 0 ) 3 0 9e3 2 2310et al., 1981), because the term oncogenewas too precise to be

    used at the time. The correlation between the localization of

    the immunoglobulin genes on chromosomes 14, 2 or 22 and

    the 8q24 factor allowed us to put forward the hypothesis of

    a position effect due to the translocation of a promoter from

    the immunoglobulin gene to a transforming agent (Frohling

    and Dohner, 2008; Huret, 2010; Lenoir et al., 1982). Cytogenet-

    ics, molecular biology and gene mapping paths then con-

    verged and the following year enabled the identification of

    theMYC gene in Burkitts lymphoma. ThisMYC genewas dys-

    regulated by an immunoglobulin enhancer of new genes. Dur-

    ing the next twenty years, most recurrent balanced

    breakpoints were cloned (Huret, 2010; Mitelman et al., 2007;

    Rowley, 1998; Sandberg, 2002), whether they were dysregu-

    lated genes such as BCL2 in follicular lymphoma, or chimeric

    ones. Most of the important genes in malignancies were thus

    identified. This task continues to be a major issue, as we are

    well short of exhaustive knowledge of the somatic genetic al-

    terations in malignancy. These results radically changed our

    view of cancer. The abnormal chromosomes could often be

    qualified as markers of malignancy as they were observed

    exclusively in malignant cells. Their presence enhanced the

    quality of the diagnosis and allowed a clearer definition of

    the prognosis. Several diagnoses have become a routine

    task. Exquisitely sensitive procedures are used to detect and

    quantify residual disease. All types and sizes of chromosomal

    abnormalities can be found in all cancers (Fagin, 2002; Huret

    et al., 2001; Mitelman et al., 2003). More than 54,000 patients

    are reported in the Mitelman Catalog (Mitelman et al., 2003)

    and the WHO (World Health Organization) includes a growing

    number of genomic rearrangements in the definition ofmalig-

    nancies. More importantly, the products of these mutated or

    dysregulated genes have become the highly specific targets

    of new drugs (Druker, 2008; Thompson, 2009) which are radi-

    cally different from conventional chemotherapy.2. Origin of chromosomal rearrangements in cancer

    The human genome of a cell is far from stable. Chemical alter-

    ations, viral aggressions and ionizing radiations are ubiqui-

    tous. The process of replication is not absolutely faithful nor

    is chromosomal segregation at mitosis (even meiosis is not

    accurate due to gametic malsegregation: more than 60% of

    zygotes will have a chromosomal abnormality that will lead

    to an early abortion, which will mostly go unnoticed).

    In somatic cells, partial failure is the rule andnot the excep-

    tion. Fresh cells are constantly generated from stem cells and

    damagedcellsareeliminated.Overall control is thusadynamic

    process with multiple checkpoints at cellular and organism

    levels via homeostasis and immunological processes. The

    actual hypothesis of tumorigenesis is a mutation (including

    chromosomal rearrangements) (Schvartzman et al., 2010)

    that confers a selective advantage on a dividing cell. The

    monoclonal proliferation will be exposed to new mutations

    with a new round of selection (Gisselsson, 2001). The recursive

    multi-stage process will culminate with a significant tumor

    that will exhibit invasive and metastatic properties and often

    considerable clinical aggressiveness. This perverted Darwin-

    ian selection at the cellular level probably acts on larger cellpopulations than evolution on whole individuals in nature

    and the time required for the generation process is shorter.

    However, as tumordevelopment is theexceptionandgenerally

    occurs late in life, the process is poorly efficient and this is also

    due to very tight controls. Immunology plays a major role, as

    suggested by the frequency of Burkitts lymphoma or Kaposis

    sarcoma in AIDS patients.

    In several benign tumors such as lipoma (Mandahl, 2000) or

    uterus leiomyoma (Lynch and Morton, 2007) specific clonal

    chromosomal abnormalities can be observed. This suggests

    that all genomic rearrangements found in tumors, dont give

    complete proliferative dysregulation, neither invasive and

    metastatic characteristics.

    Hereditary deficiencies in repair processes such as in Xero-

    derma Pigmentosum, Fanconi anemia (Moldovan and

    DAndrea, 2009) and other chromosomal instability syn-

    dromes, often characterized by chromosomal breakages, are

    characterized by high level malignancies.

    Irradiation produces chromosomal breakage aswell asmu-

    tations and is known to be a potent mutagen. Numerous

    chemicals are clastogenics including free radicals, of various

    origins such as a chronic inflammation.3. Chromosomal rearrangements

    Several chromosome rearrangements (Albertson et al., 2003)

    are very specific and observed only in a particular disease

    (Mitelman et al., 2003). The translocations found in leukemias,

    lymphomas, sarcomas and some carcinomas belong to this

    category (Tables 1 and 2). They participate in the definition

    of the disease under the term genetic, as in the recent classi-

    fications of the WHO (Swerdlow et al., 2008). Other chromo-

    somal abnormalities simply indicate a cellular malignant

    process such as isochromosomes 8q and 17q in carcinomas

    (Huret, 2010) or trisomy 8 in acute myeloblastic leukemia

    (Huret, 2010). The pattern is a set of chromosomal abnor-

    malities that have no meaning when isolated, but is more or

    less associated with the diagnosis. Conventional chromo-

    somal abnormalities are only the tip of the iceberg. Many al-

    terations are too small to be seen by conventional

    techniques or even because mitosis are lacking, as in most

    solid tumors.4. From cytogenetics to cytogenomics

    As the human genome has been completely sequenced, genes

    along the 46 human chromosomes, which are conventionally

    known according to their appearance at the fleeting stage of

    mitosis duringmetaphase,with two clearly visible chromatids

    attached to centromeres and capped by a telomere, are now

    completely mapped. During interphase, the chromosomes

    are classically indistinct in the cell nucleus. The FISH painting

    technique has revealed that they maintain their individuality

    as chromosome territories. The position of the genes in these

    territories also seems to be important for their expression. A

    gene is inactive in an internal position, whereas a gene is ac-

    tive at the periphery or outside the territory.

    http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003

  • Table 1 e Selected chromosomal abnormalities in hematological malignancies.

    Disease Chromosomalabnormalities

    Genesa Type Targetedtherapyb

    Acute malignant iymphoid proliferation

    ALL L1/L2 Pre-B t(1;19)(q23;p13) PBX1-TCF3 I

    ALL L1/L2 B or biphenotypic t(9;22)(q34;q11) ABL-BCR I

    ALL L1/L2 biphenotypic t(4;11)(q21;q23) AF4-MLL I

    ALL L1/L2 (child) t(12;21)(p13;q22) TEL-AML1 I

    ALL L1/L2 50e60 chromosomes,

    hyper diploidy

    III

    ALL L1/L2 t(5;14)(q31;q32) IL3*IGH II

    ALL L1/L3 dup(6)(q22eq23) MYB III

    ALL L1/L2 del(9p),t(9p) ?CDKN2(p16) IV

    ALL L1/L3 del(9)(p13) PAX5 IV

    ALL L1/L2 t(9;12)(q34;p13) ABL-TEL I

    ALL L1/L2 t(11;V)(q23;V) MLL-V I

    ALL L1/L2 del(12p) ETV6? IV

    ALL L1/L3 episome(9q34.1)x NUP214-ABL1 I ImatinibB (ALL3, Burkitts leukemia/lymphoma) t(8;14)(q24;q32) IGH*MYC II

    B (ALL3, Burkitts leukemia/lymphoma) t(2;8)(p12;q24) IGK*MYCc II

    B (ALL3, Burkitts leukemia/lymphoma) t(8;22)(q24;q11) IGK*MYCc II

    Follicular lymphoma to large-cell diffuse

    lymphoma

    t(14;18)(q32;q21) and

    variants

    IGH*BCL2/IGK/IGL II

    Mantle-cell lymphoma t(11;14)(q13;q32) CCND1*IGH II

    Marginal zone lymphoma t(1;14)(p21;q32) BCL10*IGH II

    Marginal zone lymphoma 3 III

    Marginal zone lymphoma t(11;18)(q21;q21) BIRC3-MALT1 I

    Large-cell diffuse lymphoma t(3;14)(q27;q32), variants BCL6*IGH, BCL6*V II

    Large-cell diffuse lymphoma t(11;14)(q13;q32) CCND1*IGH II

    Anaplastic large-cell lymphoma t(2;5)(p23;q35), variants ALK-NPM1 I

    Chronic malignant lymphoid proliferation

    Lymphocytic B cell lymphoma, chronic

    lymphocytic leukemia

    t(11;14)(q13;q32) CCND1*IGH II

    Lymphocytic B cell lymphoma, chronic

    lymphocytic leukemia

    t(14;19)(q32;q13) IGH*BCL3 II

    Lymphocytic B cell lymphoma, chronic

    lymphocytic leukemia

    t(2;14)(p13;q32) BCL11A*IGH II

    Lymphocytic B cell lymphoma, chronic

    lymphocytic leukemia

    del(11)(q23.1) ATM IV

    Lymphocytic B cell lymphoma, chronic

    lymphocytic leukemia

    del(13)(q14) DLEU, miR-16-1 & 15a IV

    Prolymphocytic T leukemia inv(14)(q11q32) TCRA/TCR D* TCL1A II

    Prolymphocytic T leukemia t(14;14)(q11;q32) TCRA/TCR D* TCL1A II

    Prolymphocytic T leukemia t(7;14)(q35;q32.1) TCRB* TCL1A II

    Multiple myeloma t(11;14)(q13;q32) CCND1*IGH II

    Multiple myeloma t(4;14)(p16;q32) WHSC1-IGHG1 II

    Multiple myeloma del(6)(q21) IV

    del(13)(q14) DLEU, miR-16-1 & 15a IV

    Acute myeloid leukemia, myelodysplastic

    syndrome

    AML M2 t(8;21)(q22;q22) RUNX1-RUNX1T1 I

    AML M3 and microgranular variant t(15;17)(q22;q11e12) PML-RARA I Retinoid Acid

    AML M3 (atypical) t(11;17)(q23;q12) PLZF-RARA I Retinoid Acid

    AML M4Eo inv(16)(p13q22) ou CBFB-MYH11 I

    t(16;16)(p13;q22 CBFB-MYH11 I

    AML M5a and other AML t(9;11)(p22;q23) MLL-MLLT3 I

    AML M5a and other AML t(11q23;V) MLL multiple partners

    including MLL

    I

    Acute megakaryoblastic leukemia t(1;22)(p13;q13) RBM15-MKL1 I

    AML, MDS t(3;3)(q21;q26) or variants RPN1-EVI1 I

    AML, MDS t(3;5)(q25;q34) MLF1-NPM1 I

    AML, MDS t(5;12)(q33;p13) PDGFRB-ETV6 I

    AML, MDS 5/del(5q) RPS14 IVAML, MDS t(6;9)(p23;q34) DEK-NUP214 I

    (continued on next page)

    M O L E C U L A R O N C O L O G Y 4 ( 2 0 1 0 ) 3 0 9e3 2 2 311

    http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003

  • Table 1 (continued)

    Disease Chromosomalabnormalities

    Genesa Type Targetedtherapyb

    AML, MDS t(7;11)(p15;p15) HOXA9-NUP98 I

    AML, MDS 7 ou del(7q) Numerous genes IVAML, MDS 8 IIIAML, MDS t(8;16)(p11;p13) MOZ-CBP I

    AML, MDS t(9;12)(q34; p13) ETV6-ABL I

    AML, MDS t(12;13)(p13;q12.3) ETV6-CDX2 I

    AML, MDS t(12;22)(p13;q13) ETV6-NM1 I

    AML, MDS t(12;V)(p13;V), del(12p) ETV6L-V I

    AML, MDS t(16;21)(p11;q22) FUS-ERG I

    AML, MDS del(20q) IV

    Therapy-induced leukemia

    Alkylating agent- and

    irradiation-induced leukemia

    5 ou del(5q) IV

    Alkylating agent- and

    irradiation-induced leukemia

    7 ou del(7q) IV

    Anti topoisomerase

    II induced leukemia

    t(11q23;V) MLL-V I

    Chronic myeloid proliferation

    Chronic myeloid leukemia (CML) t(9;22)(q34;q11) BCR-ABL1 I Imatimib, 2nd generation TKI

    Lymphoblastic acutisation of CML t(9;22), 8,Ph, 19, i(17q) BCR-ABL1 I & III Imatimib, 2nd generation TKIPolycytemia vera 9p IIIPolycytemia vera del(20q) IV

    MDS/MPD t(8;9(p21;p24) PCM1-JAK2 I

    Chronic myelomonocytic leukemia t(5;12)(q33;p13) PDGFRB-TEL I Imatinib

    5q- syndrome del(5q) RPS14 IV

    a * dysregulation gene, -mean fusion gene, V rearrangement variants. For details see: http://atlasgeneticsoncology.org.b Targeted therapy against genes involved in genetic rearrangements.

    c miR-1204 and miR-1205 could also be dysregulated (Toujani et al., 2009).

    M O L E C U L A R O N C O L O G Y 4 ( 2 0 1 0 ) 3 0 9e3 2 2312A gene-rich chromosome such as chromosome 19 seems to

    fit into an internal position in the nucleus, whereas those lack-

    ing in genes are at the nuclear periphery, forming a protection

    against possible aggression mutagens. It is conceivable that

    the structural organization of chromosomes in the cell nu-

    cleus will become a major subject of study in the near future

    (Mani et al., 2009; Nikiforova et al., 2000).

    The emergence of molecular cytogenetics (Pleasance et al.,

    2010), whose main methods are in situ hybridization (FISH),

    comparative genomic hybridization (aCGH), and now whole

    genome sequencing, possibly associated with DNA stretching

    techniques (DNA combing) and chromosomemicrodissection,

    has greatly expanded the cytogenetics field metamorphosing

    it into cytogenomics (Beroukhim et al., 2010; Bignell et al.,

    2007).5. In situ hybridization (ISH)

    The molecular reassociation of chromosomal DNA with DNA

    probes in situ can be done at all stages of the cell cycle. Chem-

    ical labeling is now very diverse, allowing the detection of

    probes by fluorescence or increasingly by cytochemistry. Of-

    ten available commercially, these reagents cover a continu-

    ously growing spectrum of chromosomal rearrangements

    that are detected onmitotic chromosomes but also on cell nu-

    clei. Consequently, cytogenetic analysis is not limited to suc-

    cessful cell cultures. Adaptations are successfully done onparaffin sections, especially to detect gene amplification

    such as ERBB2-NEU, but also for single copy rearrangements.

    The human genome sequence, available on the Internet, of-

    fers tremendous opportunities to cytogeneticists as they can

    order probes next to a breakpoint, making investigations by

    FISH available for the entire genome.

    The use of three simultaneous florescence stains, one to la-

    bel each of the two probes and one for DNA staining, is now

    commonplace. Currently five fluorochromes are typically

    available to create more than 24 different color combinations

    simultaneously (SKY, multifish) that identify each chromo-

    some. The fluorescencemicroscope is nowpart of a computer-

    ized imaging system with automated scanning, ergonomic

    reading solutions and quality control which is necessary for

    clinical practice and incorporates these advances.6. CGH

    CGH(ComparativeGenomicHybridization)quantifies testDNA

    compared to control DNA, and this can be done at any point in

    the genome. Initially described by Kallioniemi et al. (1992), its

    principle is simple.ThepurifiedtumorDNAismarkedbyafluo-

    rochrome emitting in green (e.g., fluorescing), whereas normal

    DNA ismarkedby another fluorochrome emitting in the red (or

    vice versa). Originally both DNAwere cohybridized on awhole

    normal human genome, represented by normal humanmeta-

    phasic chromosomes, according to FISH procedures. The

    http://atlasgeneticsoncology.orghttp://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003

  • Table 2 e Selected examples of chromosomal rearrangements in solid tumors.

    Disease Chromosomalrearrangements

    Genea,b Type Targetedtherapyc

    Breast cancer amp(1)(q32.1) IKBKE IIIa

    Breast and various cancers amp(6)(q25.1) ESR1 IIIa Tamoxifen

    Breast cancer amp(17)(q21.1) ERBB2 (HER2) IIIa Trastuzumab, Lapatinib

    Breast cancer amp(20)(q12) NCOA3 IIIa

    Breast and various cancers t(12;15)(p13;q25) ETV6-NTRK3 I

    Colon cancer del(4)(q12) REST IV

    Colon cancer del(5)(q21eq22) APC IV

    Hepatocellular carcinoma amp(11)(q13eq22) BIRC2 IIIa

    Hepatocellular carcinoma amp(11)(q13eq22) YAP1 IIIa

    Lung cancer amp(1)(p34.2) MYCL1 IIIa

    Lung cancer (non-small-cell) inv(2)(p22ep21p23) EML4-ALK I

    Lung, head and neck cancers amp(3)(q26.3) DCUN1D1 IIIa

    Lung cancer (non-small-cell) amp(7)(p12) EGFR IIIa Cetuximab, Panitumumab, Gefitinib, Erlotinib

    Lung cancer (non-small-cell) amp(14)(q13) NKX2-1 IIIa

    Ovarian cancer amp(1)(q22) RAB25 IIIa

    Ovarian cancer amp(3)(q26.3) PIK3CA IIIa

    Ovarian, breast cancers amp(11)(q13.5) EMSY IIIa

    Ovarian, breast cancers amp(17)(q23.1) RPS6KB1 IIIa

    Prostate cancer amp(X)(q12) AR IIIa

    Prostate cancer del(21)(q22.3q22.3) TMPRSS2*ERG II

    Renal carcinoma papillary .7q31 METRenal carcinoma papillary .7q31 METRenal carcinoma papillary .17q ?Renal carcinoma papillary .17q ?Renal carcinoma papillary t(X;1)(p11;p34) PSF-TFE3

    Renal carcinoma papillary t(X;1)(p11.2;q21.2) PRCC-TFE3

    Thyroid cancer follicular t(2;3)(q12eq14;p25) PAX8-PPARG I

    Thyroid cancer papillary inv(10)(q11.2q11.2) RET-NCOA4 I

    Thyroid cancer papillary inv(10)(q11.2q21) RET-CCDC6 I

    Ewings sarcoma t(11;22)(q24.1eq24.3;q12.2) FLI1-EWSR1 I

    Ewings sarcoma t(21;22)(q22.3;q12.2) ERG-EWSR1 I

    Rhabdomyosarcoma (alveolar) t(1;13)(p36;q14) PAX7-FKHR I

    Rhabdomyosarcoma (alveolar) t(1;13)(p36;q14) PAX7-FKHR I

    Rhabdomyosarcoma (alveolar) t(2;13)(q37;q14) PAX3-FKHR I

    Chondrosarcoma (extrasqueletical) t(9;17)(q22;q11) RBP56-CHN I

    Chondrosarcomas (myxoid) t(9;22)(q22;q12) EWS-CHN I

    Desmoplastic tumors t(11;22)(p13;q12) WT1-EWS I

    Clear cell sarcomas t(12;22)(q13;q12) ATF1-EWS I

    Liposarcomas t(12;16)(q13;p11) CHOP-FUS I

    Liposarcomas (myxoid) t(12;16)(q13;p11) CHOP-FUS I

    Dermatofibrosarcomas protuberans t(17;22)(q22;q13) COL1A1-PDGFB I

    Alveolar soft part sarcomas der(17)t(X;17)(p11;q25) ASPSCR1-TFE3 I

    Synovialosarcomas t(X;18)(p11.2;q11.2) SYT-SSX1/SSX2-SYT I

    Malignant melanoma amp(3)(p14.2ep14.1) MITF IIIa

    Glioma amp(1)(q32) MDM4 IIIa

    Astrocytoma, glioblastoma .7 ? IIIAnaplastic oligodendroglioma del(19q) ? IV

    anaplastic oligodendroglioma del(1p) ? IV

    Medulloblastoma amp(2)(p24.1) MYCN IIIa

    Medulloblastoma del(6)(q23.1) WNT IV

    Medulloblastoma amp(8)(q24.2) MYC IIIa

    Medulloblastoma del(9)(p21) CDKN2A/CDKN2B IV

    Medulloblastoma i(17q) p53 III, IV

    Neuroblastoma amp(2)(p24.1) MYCN IIIa

    Neuroblastoma amp(2)(p23.1) ALK IIIa

    Neuroblastoma del(1p) ? IV

    Renal-cell cancer del(3p26ep25) VHL IV

    Retinoblastoma del(13)(q14.2) RB1 IV

    Retinoblastoma amp(1)(q32) MDM4 IIIa

    Retinoblastoma del(13)(q14) RB

    Testicular germ-cell tumor 12p ? IIITesticular germ-cell tumor 12p ? IIIWilms tumor del(11p) WT1 IV

    (continued on next page)

    M O L E C U L A R O N C O L O G Y 4 ( 2 0 1 0 ) 3 0 9e3 2 2 313

    http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003

  • Table 2 (continued)

    Disease Chromosomalrearrangements

    Genea,b Type Targetedtherapyc

    Wilms tumor del(X)(q11.1) FAM123B IV

    Various cancers 1q ? IIIVarious cancers 1q ? IIIVarious cancers del(3p) ? IV

    Various cancers amp(5)(p13) SKP2 IIIa

    Various cancers amp(5)(p13) SKP2 IIIa

    Various cancers amp(6)(p22) E2F3 IIIa

    Various cancers del(6q) ? IV

    Various cancers amp(7)(p12) EGFR IIIa Cetuximab, Panitumumab, Gefitinib, Erlotinib

    Various cancers amp(7)(q31) MET IIIa

    Various cancers amp(8)(p11.2) FGFR1 IIIa

    Various cancers amp(8)(q24.2) MYC IIIa

    Various cancers del(9)(p21) CDKN2A/CDKN2B IV

    Various cancers del(10)(q23.3) PTEN IV

    Various cancers amp(11)(q13) CCND1 II, IIIa

    Various cancers del(11)(q22eq23) ATM IV

    Various cancers del(11q) ? IV

    Various cancers amp(12)(p12.1) KRAS IIIa

    Various cancers amp(12)(q14.3) MDM2 IIIa

    Various cancers amp(12)(q14) CDK4 IIIa

    Various cancers amp(12)(q15) DYRK2 IIIa

    Various cancers amp(13)(q32) GPC5 IIIa

    Various cancers 17q ? IIIVarious cancers amp(17)(q21.1) ERBB2 (HER2) IIIa Trastuzumab, Lapatinib

    Various cancers del(17)(p13.1) TP53 IV

    Various cancers del(17)(q11.2) NF1 IV

    Various cancers amp(19)(q12) CCNE1 IIIa

    Various cancers amp(20)(q13) AURKA IIIa

    a a* dysregulation gene, -mean fusion gene, V rearrangement variants. For details see: http://atlasgeneticsoncology.orgb Often also mutated.

    c Targeted therapy against genes involved in genetic rearrangements.

    M O L E C U L A R O N C O L O G Y 4 ( 2 0 1 0 ) 3 0 9e3 2 2314unique chromosome sequences were simultaneously the tar-

    getsof the tumorandcontrolDNAs.Afterwashingandbanding

    withDAPI to generate banding, preparationswere analyzed on

    amicroscopicdigital imageanalyzer.Nowthe targetsareavery

    largenumber of BACsor of selectedoligonucleotides, sampling

    the total human genome (Pinkel and Albertson, 2005).

    The hybridization of tumor DNA to each point of the ge-

    nome is compared to that of control DNA. A loss of this region

    in the tumor genomewill be revealed by an excess signal from

    the normal DNA. Conversely, a gain will be detected by an ex-

    cess of tumorDNA,whichwill bemajor in the case of gene am-

    plification. The exact quantification is done by calculating the

    normalizedratiobetweentumorandnormalDNAfluorescence

    along each chromosome. Metaphasic chromosomes are cheap

    natural microchips covering the entire genome with a high

    level of integration, but with poor resolution. 1 Mb resolution

    has been achievedwith BAC CGH arrays. Now, oligonucleotide

    chipswithin the resolution rangeof a fewkb, from1 105 up toseveral million, are available and yield robust results.

    Paradoxically, CGH greatly simplifies the interpretation of

    anomalies in its standard format.

    Tumor DNA can be prepared from fresh or frozenmaterial.

    CGH is very sensitive to the presence of normal cells in the

    sample under analysis, which exert a dilution effect on tumor

    DNA. This makes CGH unreliable when there are fewer than

    60% of normal cells. Enrichment in tumor cells could bedone by cell sorting or laser microdissection, but it would

    probably be difficult to generalize those techniques.

    CGHcannotdetect balanced translocations and their equiv-

    alents, nor overall changes in ploidy (triploidy, tetraploidy).

    The analysis can be far from simple in some cancers, with

    the added complexity from inherited CNV. These CNV add

    complexity to interpretation. However, the simplest way to

    avoid it is tousenormalDNAfromeachpatient as controlDNA.

    An alternativemethodology is single copy number analysis

    derived from SNP analysis. This approach depicts the loss of

    heterozygozity (LOH) which occurs via acquired isodisomy,

    without copy number variation. The incidence reported is

    about 10% of CNA (copy number aberration). Currently, there

    is no system allowing the two analyses to be conducted rou-

    tinely on the same array in cancer.7. Sequence

    Next generation sequencing is rapidly progressing (Mardis,

    2009). Prices are dropping every year, making the objective

    of 1000$, or even less, for a full human genome fully conceiv-

    able. Bioinformatics is very demanding but this aspect will be

    solved in a few years. The first results of whole genome se-

    quencing (WGS) in tumors revealed that the diagnosis of bal-

    anced rearrangements could be achieved in the absence of

    http://atlasgeneticsoncology.orghttp://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003

  • e MET

    nce

    r,MYCN

    in

    blastoma

    rmutationofRB,loss

    of

    M O L E C U L A R O N C O L O G Y 4 ( 2 0 1 0 ) 3 0 9e3 2 2 315classic karyotypes that require cell culture. Several malignan-

    cies have recently been sequenced. Extremely divergent

    results have been obtained. Several solid tumors exhibit

    a very high number of mutations, up to more than 20,000,

    a similar number to that of genes (Pleasance et al., 2010). In

    contrast, AML, with a normal karyotype, exhibits a few muta-

    tions, most of which are in known oncogenes (Mardis et al.,

    2009).Table

    3e

    Chromosometypology

    abnorm

    alities.

    Abnorm

    ality

    type

    Involvedgenes

    Main

    functionaleffect

    Exampl

    TypeI

    Fusiongenesoriginating

    from

    transloca

    tionsoroth

    er

    chro

    moso

    malrearrangements

    Onco

    genes,

    kinase

    s,

    transc

    riptionfactors,etc

    Functiongain

    FLI1-EW

    Sin

    thet

    (11;22),MLLduplica

    tion

    TypeII

    Positioneffect

    originatingfrom

    transloca

    tionsoroth

    er

    chro

    moso

    malrearrangements

    Onco

    genes,

    oth

    ergenes

    Dysregulation

    IGH*BCL2in

    t(14;18),MYCin

    t(8;14)(q24;q32)

    TypeIII

    Copynumbergain

    ofallorpart

    ofach

    romoso

    me

    (IIIs)

    Onco

    genes,

    oth

    ergenes

    Genedosa

    ge

    effect

    (mayact

    ona

    modifiedgene)

    Duplica

    tionofth

    ePhin

    CML,of

    genebywhole

    trisomy

    7orse

    gmentalduplica

    tion

    TypeIIIa

    Geneamplifica

    tion

    (more

    than2,2e4co

    py

    perhaploid

    genome

    Onco

    genes,

    oth

    ergenes

    Genedosa

    geeffect

    (sometimesactingon

    amodifiedgene)

    EGFRin

    lung

    cance

    r,ERBB2/H

    ER2in

    breast

    ca

    neuro

    blastom

    a

    TypeIV

    Totalorpartial(IVs)

    loss

    ofach

    romoso

    me,Total

    orpartialacq

    uiredisodisomy,inactivationbymutation.

    Tumorsu

    ppressorgenes

    Loss

    offu

    nction,LOH

    InactivationofRBgenein

    retino

    via

    deletionofch

    romoso

    me13o

    CDKN2A

    invariousca

    nce

    rs8. Typology of genomic abnormalities (chromosomeand gene) (Table 3)

    Chromosomal abnormalities can be classified into four types,

    depending on their consequences.

    Type I refers to the generally balanced translocations or

    other structural rearrangements that fuse two genes, form-

    ing a hybrid gene. As the structure of eukaryotic genes is di-

    vided into exons and introns, this fusion enables the

    formation of chimeric proteins which acquire an oncogenic

    potential. This new hybrid gene is malignancy-specific as

    for example the FLI1-EWS fusion gene following the t(11;

    22) (q24, q12) translocation in Ewings sarcoma (Delattre

    et al., 1992, 1994; Riggi and Stamenkovic, 2007) or the

    EML4-ALK fusion gene following an inversion on 2p in lung

    carcinoma (Soda et al., 2007) (Tables 1 and 2). These anom-

    alies may be regarded as resulting in, strictly speaking,

    a gain of function. The genes involved often involve tran-

    scription factors but also tyrosine kinases. These often bal-

    anced abnormalities first appeared to be mainly confined

    to hematological malignancies and sarcomas but they are

    now often found in carcinomas. Anomalies of RET or of

    the PAX8-PPARG fusion gene in thyroid cancer (Nikiforova

    and Nikiforov, 2009; Pierotti, 2001), the t(X;1) translocation

    in cancers of the kidney (Argani et al., 2003; Camparo

    et al., 2008), the inv(2)(p21p23) fusing EML4 to ALK in lung

    cancers are good examples (Table 3). Cryptic genomic rear-

    rangements may contribute to gain of function: partial du-

    plication of a gene on itself as in MLL with the 50 part issometimes duplicated and activates this gene in leukemia

    secondary to chemotherapy with topoisomerase II inhibitors

    (Huret, 2009).

    The chromosomal balanced translocations, which give

    rise to chimeric genes, are often malignancy-specific and

    are still the main road to identifying the genes involved in

    cancer through positional cloning. Although a lot of rear-

    rangements are now well known, other less frequent ones

    have yet to be investigated in detail as they could help

    identify new genes. Several of the genes involved in fusion,

    are swingers as they can be associated with many other

    partner genes such as RET for example in thyroid cancer.

    The processes are recurrent as these partner genes are

    sometimes themselves associated with various partners.

    This concept of networking between various oncogenes,

    coupled with the fact that there are only 23,000 genes in

    humans, suggests that a limited number of genes are in-

    volved in oncogenesis.

    Type II concerns the case of dysregulation of a gene by

    position effect. The paradigm is Burkitts lymphoma charac-

    terized by the t(8;14) (q24, q32) or its variant, t(2; 8) (p12; q24)

    http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003

  • M O L E C U L A R O N C O L O G Y 4 ( 2 0 1 0 ) 3 0 9e3 2 2316

    http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003

  • M O L E C U L A R O N C O L O G Y 4 ( 2 0 1 0 ) 3 0 9e3 2 2 317or t(8; 22) (q24, q11) (Bernheim et al., 1981a). The MYC gene,

    localized to 8q24, is dysregulated under the influence of an im-

    munoglobulin heavy chain gene enhancer localized in 14q32.

    The same mechanism is thought to occur in variant forms

    with kappa (2p12) or lambda (22q11) light chains despite the

    distant 30 breakpoint of MYC and the possible involvement ofmiRNA (Figure 1) (Toujani et al., 2009). The MYC gene remains

    structurally intact, although it is the target of numerous punc-

    tual mutations. As it occupies the position of the variable part

    of the immunoglobulin genes, like them, it is submitted to

    hypermutation possibly induced by AID. These translocations

    on immunoglobulin geneshavebeena real key to thediscovery

    of a number of important genes such as BCL1 and BCL2. Iso-

    lated from the t(14; 18) (q32, q21) which is specific for non-

    Hodgkin centro-follicular lymphoma, BCL2 is a ubiquitous ap-

    optosis inhibitor.

    Type III, is the gain of genetic material.

    The gain of material is very frequent and may involve

    whole chromosomes as the result of malsegregation gener-

    ated by a pathological spindle. Segmental gains result from

    unbalanced structural chromosomal rearrangements. Their

    functional significance remains unknown but an increase in

    gene size with a gain of function is often seen. For example,

    the MET oncogene is overrepresented in sporadic papillary re-

    nal-cell tumors via a trisomy 7 and the protein is proportion-

    ally overexpressed. During acutisation in Chronic Myeloid

    Leukemia (CML), the Ph chromosomewith the BCR-ABL fusion

    gene, is often duplicated. Copy number abnormalities are very

    often segmental due to structural unbalanced chromosomal

    rearrangements such as the gain of part of 17q in

    neuroblastomas.

    Type IIIa corresponds to gene amplification which is a se-

    lective increase in DNA copy number (Sartorius). It can ex-

    hibit two cytogenetic aspects. Double minute (DM)

    chromosomes are self-replicating extra-chromosomal acen-

    tric rings or episomes that were initially shown in meta-

    phase and are now easily found in interphase cells.

    Generally, there is considerable heterogeneity from cell to

    cell in the number of DM, even if the mean number is

    high. In some instances, the DM can be expulsed from the

    nucleus and even from the cell (Valent et al., 2001). The

    other aspect of gene amplification is homogenously staining

    regions (HSR) where multiple copies of a genomic region are

    integrated into chromosome(s) in the form of block(s) of re-

    peat amplicons. They are produced by a break-fusion-bridge

    process that remains in the cycling process until the opti-

    mal amount of gene expression is achieved. MYCN onco-

    gene amplification in neuroblastoma which is oftenFigure 1 e Virtual cloning of 8q24 in Burkitt lymphoma showing MYC reg

    Ly 47 cell lines, characterized by a t(8;22)(q24;q11), the der(8)t(8;22) region

    chromosome 8 (labeled in green) appeared trisomic from pter to 8q24.2, 129

    appeared normal (BL84 Partial profiling aCGH), corresponding to the nor

    translocated to the der(22)(BL84 partial RHG-banding). The breakpoint wa

    mir-1205 and mir-1206 (Toujani et al., 2009). The chromosome 22 (labele

    modified. B) Then a complete loss of IGL (immunoglobulin light chain la

    rearrangement of IGL, while the t(8;22) occurred during one of them. The

    (immunoglobulin heavy chain) regions are also physiologically rearranged o

    21.58e49.05 Megabases, the chromosome 22 is trisomic.observed in tumor cells as DM is generally observed as

    HSR(s) in the culture-established cell lines generated from

    these tumor cells. Some human cell lines however harbor

    DM such as SW613 (Guillaud-Bataille et al., 2009), with

    a MYC amplification linked to tumorigenesis in mice, but

    also another population of DM derived from 14q24.1 that

    were purported to contribute to maintaining amplification

    in the form of DMs (Guillaud-Bataille et al., 2009).

    The amplification can involve a series of oncogenes,

    whose sequence does not appear to be impaired, such as

    ERBB2, MYCN, MYC, CCND1, GLI, MDM2, ALK (Huret, 2010;

    Santarius et al., 2010) but also gene fusion as in some alve-

    olar rhabdomyosarcoma with the t(1;13) translocation

    (Table 2). The number of copies of a DNA sequence that con-

    stitute genome amplification is variously described but gen-

    erally considered greater than 4-fold relative to a non-

    amplified marker in a haploid genome. In diploid cells this

    would be equivalent to at least 8 copies. For example, this

    definition is used for MYCN amplification which is associ-

    ated with a poor outcome in neuroblastoma (Schwab,

    2004). ERBB2 amplification which is deemed present when

    the mean FISH ratio of ERBB2_HER2 versus the centrometric

    probe of chromosome 7, calculated after examining 60 cells,

    is greater than 2 or if more than 6 copies/nucleus are found.

    This is required to be eligible for anti-ERBB2 therapeutic

    agents. Positive ERBB2-amplified cases are distinct from

    cases that are equivocal for ERBB2 gene amplification

    (Couturier et al., 2008). As the copy number in tissues is

    measured extensively by array- or PCR-based techniques,

    the criteria for genomic amplification are frequently

    wrongly based on thresholds that are specific for the normal

    experimental variations experienced, thus mixing gain and

    real genetic amplification.

    Type IV corresponds to the loss of genetic information, ei-

    ther through deletion, unbalanced translocation, or any other

    mechanism leading to the loss of heterozygosity (LOH), in-

    cluding acquired parental unidisomy, be it complete or seg-

    mental. Loss of function by point mutation is in this

    category (Albertson et al., 2003; Chin and Gray, 2008). These

    frequent rearrangements, diagnosed by the loss of heterozy-

    gosity (LOH), losses of material by microarrays, FISH or karyo-

    type, often involve tumor suppressor genes (e.g. RB, RET,

    BRCA1, P53 genes) (Huret, 2010). They are involved in predis-

    position to many types of cancer. Other losses of function

    can affect genome repair genes, caretaker or gatekeeper

    genes. These losses can affect non coding genes such as

    miRNA that are non randomly lost in the same malignancy

    (Calin and Croce, 2009; Croce, 2009).ion breakpoints (modified from Toujani et al., 2009). A) In BL84 and

    harboring the MYC locus was duplicated. For example on BL84, the

    .15 Megabases (BL84 partial RHG-banding), then its ratio on aCGH

    mal chromosome 8 and to the distal part of the chromosome 8

    s observed distal to MYC as expected and is mapped to PVT1, between

    d in blue) from the centromere to the long arm telomere is first non

    mbda) region is observed suggesting a physiological biallelic

    IGK (immunoglobulin light chain kappa) and the IGH

    n a single allele (A & B). After this lost region, from 22q11 to qter,

    http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003

  • M O L E C U L A R O N C O L O G Y 4 ( 2 0 1 0 ) 3 0 9e3 2 23189. Evolution of chromosomal abnormalities intumors

    Although the concept of primary abnormality and additional

    anomalies is very important, it needs to be revisited. There

    are numerous examples, such as leukemia, where a recurrent

    chromosomal abnormality such as the t(9;22)(q34;q11) trans-

    location in CML, was conjointly observed with the t(15;17)

    (q22;q21) in promyelocytic AML. Only the t(9;22) was observed

    after treatment. The two abnormalities were again present in

    the same cells at relapse (Castaigne et al., 1984). Another ex-

    ample is lymphomas, where a recurrent chromosomal abnor-

    mality such as the t(8; 14)(q24, q32) translocation in Burkitts

    lymphoma, is conjointly observed with the t(14;18) of follicu-

    lar lymphoma (Tomita et al., 2009). This suggests that these al-

    terations have cumulative effects, if not synergistic. They are

    commutative, as the order in which they occur does not al-

    ways appear to be important. In solid tumors, complex chro-

    mosomal abnormalities reflect the multistep process of

    oncogenesis and tumor progression. There are striking exam-

    ples of profiles that show themultiple subclones in a single tu-

    mor as for example in progression towards gene

    amplification. Cell tolerance to alterations in the genome

    means mayhem in genome maintenance. In some cases, the

    alteration is exclusively local, with most of the chromosomes

    exhibiting a normal profile. In others there are multiple rear-

    rangements culminating in genomic storms with highly

    rearranged chromosomal regions shown by aCGH. Telomeres

    also appear to play a key role in controlling the number of cell

    divisions, through gradual terminal truncation. The inactiva-

    tion of this control introduces a form of chromosomal insta-

    bility with telomeric fusions followed by random breaks

    inducing cell death, but it may also select the cell with dam-

    aged genomic material and allow it to recover telomerase

    function (OSullivan and Karlseder, 2010). Cancer cell lines

    with a highly rearranged karyotype can maintain stability

    throughout the cell culture passages, which indicates that

    rearranged chromosomes are not intrinsically unstable. It is

    possible that the dynamic process of Darwinian selection of

    the cancer genome leads to some form of stable pathologic

    disease. Some tumors, that have amutator phenotype, exhibit

    few visible chromosomal abnormalities.

    The evolution of the chromosomal abnormalities is classic

    during disease progression. It may occur as its natural history

    such as the evolution of an indolent follicular lymphomawith

    a single t(14;18) to an aggressive diffuse lymphoma with mul-

    tiple abnormalities added to the initial translocation (Yunis

    et al., 1989). A post-therapeutic relapse often keeps several

    chromosomal rearrangements from the initial proliferation,

    but some of the initial ones may be lost and new ones may

    be acquired. This can lead to a search for residual disease

    with wrongly targeted probes.10. Lesions and tumors secondary to physical orchemical mutagens

    Alkylating agents or radiotherapy currently used to treat

    cancers induce chromosomal breakage which is themanifestation of the genotoxic activity of such drugs. The

    doses commonly used are in a range that allows tolerable

    toxicity. Patients suffering from Fanconi disease who have

    a congenital deficit in DNA repair are highly susceptible to

    these agents which induce acute toxicity with extensive

    chromosomal breakage (Berger et al., 1980; Moldovan and

    DAndrea, 2009).

    In a few standard patients, secondary therapy-induced

    AML may occur five to seven years after successful treatment

    of the initial malignancy. It then has specific abnormalities

    such as 5/5q, 7/7q, 12p. Topoisomerase II-targeteddrugs (ATII), such as etoposide and anthracyclines, may in-

    duce the second type of t-AML. It occurs in amedian of 2 years

    and is not preceded by MDS. Cytogenetic analysis shows

    a high frequency of rearrangements of chromosome band

    11q23 but also recurrent balanced rearrangements such as t

    (8;21), t(15;17) and inv(16) (Qian et al., 2009).

    Secondary tumors can also occur in the absence of treat-

    ment but through irradiation accidents. Patients irradiated af-

    ter Chernobyl (Tronko et al., 2006) showed a specificity for

    radiation-induced rearrangements in thyroid tumors, particu-

    larly affecting RET (Nikiforov, 2006).11. Targeted therapies

    Chromosomal abnormalities in malignancy have been pivotal

    in the discovery of targeted therapy against cancer cells. One

    of the first successes came from the discovery that patients

    suffering from Fanconi anemia were highly sensitive to

    Endoxan, a bifunctional alkylating drug used in the late 70s

    for bonemarrow graft conditioning (Berger et al., 1980). A spe-

    cific reduced-dose cytotoxic conditioning was proposed for

    Fanconi patients allowing successful bone marrow grafts

    (Gluckman et al., 1980). That was the first application of ther-

    apy following cytogenetic findings.

    Retinoic acid in promyelocytic leukemia (PML), initially

    proposed in China, induced the maturation of leukemic cells

    and the patients achieved a complete remission (Wang and

    Chen, 2008). It was rapidly linked to the AML M3 specific t

    (15;17) and when the cloning of the breakpoints in PML and

    RARA (Retinoic Acid Receptor Alpha) genes was done, a phys-

    iopathological mechanism appeared.

    The paradigm of CML had become the rule in only ten

    years. This malignant proliferation is caused by the consti-

    tutively active chimeric tyrosine kinase BCR-ABL. This pro-

    tein is encoded by a BCR-ABL fusion gene caused by the t

    (9; 22) (q34, q11) translocation designated as the Philadel-

    phia chromosome or Ph (Nowell and Hungerford, 1960;

    Rowley, 1973).

    The Imatinib discovery programwas initiated with the aim

    of rationally developing a targeted anticancer approved drug.

    A lead compoundwas identified from a screen for inhibitors of

    protein kinase C. During the sophisticated optimization of the

    molecule structure (Buchdunger et al., 1996; Toledo et al.,

    1999), researchers observed that a modification had induced

    an inhibitory effect on tyrosine kinases. This property was

    then carefully enhanced while PKC inhibition was suppressed

    and molecule solubility was increased by other structural ad-

    justments in order to allow oral intake. Among the TKs tested,

    http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003

  • M O L E C U L A R O N C O L O G Y 4 ( 2 0 1 0 ) 3 0 9e3 2 2 319PDGFR and c-KIT were strongly inhibited by this small mole-

    cule as well as BCR-ABL in cell lines and animal models. In

    1998, the first patient with CML was treated. Imatinib was so

    efficient and devoid of major toxicity that the phase III trial

    was initiated after only two years and 10 months later the

    FDA approved Imatinib for Ph leukemia. As usual, the designprocess took around 8 years, but the clinical development was

    remarkably rapid (Druker, 2009; Sherbenou and Druker, 2007).

    Thus a second fusion gene, a concept initiated by somatic ge-

    neticists from gene mapping, either cytogeneticist or molecu-

    lar biologists, has been successfully targeted. The fact that

    amajor pharmaceutical laboratorymade such a breakthrough

    without losing money on a specific drug for a limited number

    of patients opened the way for targeted drugs against proteins

    specifically modified by genetic changes in cancer.

    Imatinib (Gleevec), induces apoptosis of Ph, BCR-ABLcells in patients, with clinically impressive results: most pa-

    tients are in complete cytogenetic remission. Molecular re-

    mission can occur but it is not the rule. This remission

    seems to persist under permanent treatment. There is some

    primary and secondary resistance caused by either overrepre-

    sentation of the Ph chromosome, additional chromosomal

    rearrangements or mutations at the active site. Those acute

    leukemias with the Philadelphia chromosome can also re-

    spond to the drug but results are less consistent and relapses

    are frequent. A second and even third generation of mole-

    cules, Dasatinib, Nilotinib, has been developed that could in-

    hibit most resistant ABL mutations and could help to

    achieve better patient compliance.

    Imatinib is active in other diseases with a constitutionally

    active tyrosine kinase. GIST (Gastro-Intestinal Stromal Tumor)

    with a KIT mutation respond to it (Duffaud and Le Cesne,

    2009). So does an infrequent myeloproliferative disorder, be-

    longing to idiopathic hypereosinophilia which is due to a fu-

    sion between FIP1L1 and PDGFRA resulting from an

    interstitial deletion in 4q12. Imatinib inhibits the chimeric

    gene by acting on PDGFRA (Cools et al., 2003). Dermatofibro-

    sarcomas protuberans, a slowly growing cutaneous tumor is

    characterized by a t(17;22) and a COL1A1-PDGFB fusion gene

    whose protein is inhibited by Imatimib (Duffaud and Le

    Cesne, 2009).

    Other targeted drugs against products of rearranged genes

    exist in solid tumors. The epidermal growth factor receptor 2

    ERBB2 (HER2), another tyrosine kinase gene, is often amplified

    in breast cancer in the formofHSRs (Andre et al., 2009). Among

    other tyrosine kinases, Lapatinib can inhibit ERBB2 in breast

    cancer. Trastuzamab, a humanized monoclonal antibody

    against ERRB2, has demonstrated activity. It is now currently

    prescribed to women with a gene amplification because sur-

    vival is longer. This amplification is diagnosed by immunohis-

    tochemistry and/or ISH (Couturier et al., 2008; Hanna, 2001).

    The epidermal growth factor receptor (EGFR) can be either

    amplified or mutated in a subset of non-small-cell lung can-

    cers (NSCLC) (Dahabreh et al., 2010; Sholl et al., 2009). Gefeti-

    nib, Erlotinib are inhibitors of EGFR and approved for use

    against those cancers. Cetuximab and Panitunmumab are

    monoclonal antibodies against EGFR. The first one has been

    approved for the treatment of head and neck cancer and

    both are indicated in colon cancer. Many other drugs are un-

    der development.12. Role of clinical cytogenomics in the diagnosis,prognosis and in monitoring residual disease

    Chromosomal rearrangements are now used clinically as

    markers of malignancy.

    It is well established that in leukemias and lymphomas,

    several rearrangements are highly associatedwith a particular

    type of leukemia (Huret, 2010). For example, a t(15;17) is seen

    exclusively in acute promyelocytic leukemia, and this anom-

    aly has become part of the definition of the entity. That chro-

    mosomal abnormalities play a prognostic role is now an

    integral part of common practice since most treatment proto-

    cols stratify patients according to their karyotype.

    The diagnosis of chromosome alterations is required for

    the typing and subtyping of sarcomas. The EML-ALK fusion

    gene is sought in lung cancer by FISH. The prognosis of neuro-

    blastoma is now stratified not only onMYCN amplification but

    on segmental rearrangements shown by aCGH. The presence

    of segmental copy number aberrations in neuroblastomas

    means stratification into a poor prognosis group (Janoueix-

    Lerosey et al., 2009). Cerebral tumors are also stratified follow-

    ing genomic investigations, like breast tumors, lung cancers

    and many other types of cancers.

    The diagnosis of residual disease which generally requires

    quantification can be assessed using two complementary

    methods. The first, quantitative real-time PCR, is a reliable

    and sensitive technique, and it yields statistical results. An-

    other approach is FISH which is less sensitive, but it provides

    a true estimate of malignant cells present. The metaphasic

    karyotype has proven clinically very relevant for monitoring

    CML. These methods are widely used to detect residual dis-

    eased cells in hematology. In solid tumors, the diagnosis of re-

    lapses can benefit from genomic knowledge of a primary

    tumor in order to design specific probes to show targeted chro-

    mosomal rearrangements.13. Databases

    Knowledge of cancer genetics is now too massive and is mov-

    ing too fast to be fully apprehended by a single individual. For

    example, more than 3000 genes are involved in cancer which

    is about 14% of the total number of existing genes. It requires

    a collective effort facilitated by Web tools that allow interac-

    tive consultation. For example, the Atlas of Genetics and Cyto-

    genetics in Oncology and Hematology (Huret, 2010), is a free

    Internet database that provides summary information and

    updates on genes, chromosomal abnormalities, and clinical

    entities in onco-hematology. It contains multiple links to the

    multiple genomic databases that are expanding like mush-

    rooms. Indexed by Current Content, it is considered a scien-

    tific journal of international repute and more than 1400

    international authors are involved in this work. With thou-

    sands of hits per day, it has been top ranked by Google for

    the words chromosome cancer.The NCBI/NCI (National Cancer Institute) had created the

    Cancer Chromosomes initiative (NCI and NCBI, 2001) which

    is based on three databases: the SKY/M-FICH & CGH database,

    the NCI Mitelman Database of Chromosome Aberrations in

    http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003

  • M O L E C U L A R O N C O L O G Y 4 ( 2 0 1 0 ) 3 0 9e3 2 2320Cancer which identifies the published cases (Mitelman et al.,

    2003), and the NCI Recurrent aberrations in Cancer database.

    Each site has its own characteristics and they are more com-

    plementary than competitive.

    Moreover, the human genome sequence has generated

    fantastic tools to investigate the genome of malignant cells.

    The identification of genes potentially involved in transloca-

    tion, the ability to generate highly specific oligonucleotide

    primers, genetic interpretation of array copy number analysis

    and whole genome sequencing analysis are among the possi-

    bilities now available.14. Prospects

    Genomic abnormalities are being increasingly used for the di-

    agnosis, prognosis, treatment and tomonitor patients in order

    to achieve personalized treatment for cancer. The information

    obtained from karyotype studies, high-resolution array CGH,

    mitoses and nuclei using FISH and from cells using PCR will

    be consistent but each techniquewill contribute its specificity:

    karyotype versatility, high-resolution aCGHdetection of unex-

    pected events, focused FISH analysis of tumor cells enabling

    the differentiation of the proportion of malignant cells and

    the very strong sensitivity of PCR. Whole genome sequencing

    will be increasingly used in anot toodistant future.Weare cur-

    rently experiencing a period of progressive generalization that

    is far fromsimple. Tumormaterial is not readily accessible and

    possible changes inpracticeswill beneeded.Malignant cell en-

    richment is often required. PCR may detect recurrent translo-

    cations for the diagnosis but it is currently incapable of

    diagnosing additional abnormalities thatmaybe innumerable.15. Conclusion

    Acquired chromosomal abnormalities are now considered as

    causal inmalignant proliferations. Recurrent chromosomal ab-

    normalities, their additive character combined with their mul-

    tiplicity in a single tumor are obvious paths in the multi-stage

    process of Darwinian oncogenesis. It allows specific chromo-

    somal signatures to be identified in more and more tumors

    coexisting with possible heterogeneity of the karyotype from

    one cell to another, or from a subclone to another. Genomic ab-

    normalities are known as the tip of the iceberg because many

    small rearrangementsgounnoticedandnewonesaredescribed

    each month. Considerable work is still needed in clinical solid

    tumor cytogenomics. This approach has proven successful

    since it has allowed (and still allows) the identification of a lot

    of genes involved in oncogenesis. The black box that remains

    the core of a cancer cell is now becoming dark grey. Beyond

    progress in concepts and classifications, the designation of tar-

    gets for new drugs is creating a new paradigm for cancer ther-

    apy. The pharmaceutical industry has proven all its skills in

    chemistry, leavinghope for anumerousprogeny to target onco-

    genes while finding suitable models for highly specific drugs

    with possibly, a more limited market than that observed con-

    ventionally. The monotherapy, circumvented by resistance,

    will be replaced by a combination of new and old therapies.Acknowledgement

    The author thanks Lorna Saint Ange for editing and Bernard

    Caillou for helpful discussions.R E F E R E N C E S

    Albertson,D.,Collins,C.,McCormick, F.,Gray, J., 2003.Chromosomeaberrations in solid tumors. Nat. Genet. 34, 369e376.

    Andre, F., Job, B., Dessen, P., Tordai, A., Michiels, S., Liedtke, C.,Richon, C., Yan, K., Wang, B., Vassal, G., Delaloge, S.,Hortobagyi, G.N., Symmans, W.F., Lazar, V., Pusztai, L., 2009.Molecular characterization of breast cancer with high-resolution oligonucleotide comparative genomichybridization array. Clin. Cancer Res. 15, 441e451.

    Argani, P., Lui, M.Y., Couturier, J., Bouvier, R., Fournet, J.C.,Ladanyi, M., 2003. A novel CLTC-TFE3 gene fusion in pediatricrenal adenocarcinoma with t(X;17)(p11.2;q23). Oncogene 22,5374e5378.

    Balmain, A., 2001. Cancer genetics: from Boveri and Mendel tomicroarrays. Nat. Rev. Cancer 1, 77e82.

    Berger, R., Bernheim, A., Gluckman, E., Gisselbrecht, C., 1980. Invitro effect of cyclophosphamide metabolites onchromosomes of Fanconi anaemia patients. Br. J. Haematol.45, 565e568.

    Bernheim, A., Berger, R., Lenoir, G., 1981a. Cytogenetic studies onAfrican Burkitts lymphoma cell lines: t(8;14), t(2;8) and t(8;22)translocations. Cancer. Genet. Cytogenet. 3, 307e315.

    Bernheim, A., Berger, R., Preudhomme, J.L., Labaume, S.,Bussel, A., Barot-Ciorbaru, R., 1981b. Philadelphiachromosome positive blood B lymphocytes in chronicmyelocytic leukemia. Leuk. Res. 5, 331e339.

    Beroukhim, R., Mermel, C.H., Porter, D., Wei, G., Raychaudhuri, S.,Donovan, J., Barretina, J., Boehm, J.S., Dobson, J., Urashima, M.,McHenry, K.T., Pinchback, R.M., Ligon, A.H., Cho, Y.J., Haery, L.,Greulich,H., Reich,M.,Winckler,W., Lawrence,M.S.,Weir, B.A.,Tanaka, K.E., Chiang, D.Y., Bass, A.J., Loo, A., Hoffman, C.,Prensner, J., Liefeld, T., Gao, Q., Yecies, D., Signoretti, S.,Maher, E., Kaye, F.J., Sasaki, H., Tepper, J.E., Fletcher, J.A.,Tabernero, J., Baselga, J., Tsao, M.S., Demichelis, F., Rubin, M.A.,Janne, P.A., Daly, M.J., Nucera, C., Levine, R.L., Ebert, B.L.,Gabriel, S., Rustgi, A.K., Antonescu, C.R., Ladanyi, M., Letai, A.,Garraway, L.A., Loda, M., Beer, D.G., True, L.D., Okamoto, A.,Pomeroy, S.L., Singer, S., Golub, T.R., Lander, E.S., Getz, G.,Sellers, W.R., Meyerson, M., 2010. The landscape of somaticcopy-number alteration across. Nature 463, 899e905.

    Bignell, G.R., Santarius, T., Pole, J.C., Butler, A.P., Perry, J.,Pleasance, E., Greenman, C., Menzies, A., Taylor, S., Edkins, S.,Campbell, P., Quail, M., Plumb, B., Matthews, L., McLay, K.,Edwards, P.A., Rogers, J., Wooster, R., Futreal, P.A., Stratton, M.R., 2007. Architectures of somatic genomic rearrangement insequence-level resolution. Genome Res. 17, 1296e1303.

    Boveri, T., 1914. Zur Frage der Entstehung Maligner Tumoren.Gustav Fisher, Jena.

    Buchdunger, E., Zimmermann, J., Mett, H., Meyer, T., Muller, M.,Druker, B.J., Lydon, N.B., 1996. Inhibition of the Abl protein-tyrosine kinase in vitro and in vivo by a 2-phenylaminopyrimidine derivative. Cancer Res. 56, 100e104.

    Calin, G.A., Croce, C.M., 2009. Chronic lymphocytic leukemia:interplay between noncoding RNAs and protein-coding genes.Blood 114, 4761e4770.

    Camparo, P., Vasiliu, V., Molinie, V., Couturier, J., Dykema, K.J.,Petillo, D., Furge, K.A., Comperat, E.M., Lae, M., Bouvier, R.,Boccon-Gibod, L., Denoux,Y., Ferlicot, S., Forest, E., Fromont, G.,Hintzy,M.C., Laghouati, M., Sibony, M., Tucker, M.L.,Weber, N.,

    http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003

  • M O L E C U L A R O N C O L O G Y 4 ( 2 0 1 0 ) 3 0 9e3 2 2 321Teh, B.T., Vieillefond, A., 2008. Renal translocation carcinomas:clinicopathologic, immunohistochemical, and gene expressionprofiling analysis of 31 caseswith a reviewof the literature. Am.J. Surg. Pathol. 32, 656e670.

    Castaigne, S., Berger, R., Jolly, V., Daniel, M.T., Bernheim, A.,Marty, M., Degos, L., Flandrin, G., 1984. Promyelocytic blastcrisis of chronic myelocytic leukemia with both t(9;22) and t(15;17) in M3 cells. Cancer 54, 2409e2413.

    Chin, L., Gray, J.W., 2008. Translating insights from the cancergenome into clinical practice. Nature 452, 553e563.

    Cools, J., DeAngelo, D., Gotlib, J., Stover, E., Legare, R., Cortes, J.,Kutok, J., Clark, J., Galinsky, I., Griffin, J., Cross, N.,Tefferi, A., Malone, J., Alam, R., Schrier, S., Schmid, J.,Rose, M., Vandenberghe, P., Verhoef, G., Boogaerts, M.,Wlodarska, I., Kantarjian, H., Marynen, P., Coutre, S.,Stone, R., Gilliland, D., 2003. A tyrosine kinase created byfusion of the PDGFRA and FIP1L1 genes as a therapeutictarget of imatinib in idiopathic hypereosinophilic syndrome.N. Engl. J. Med. 348, 1201e1204.

    Couturier, J., Vincent-Salomon, A., Mathieu, M.C., Valent, A.,Bernheim, A., 2008. Diagnosis of HER2 gene amplification inbreast carcinoma. Pathol. Biol. (Paris).

    Croce, C.M., 2009. Causes and consequences of microRNAdysregulation in cancer. Nat. Rev. Genet. 10, 704e714.

    Dahabreh, I.J., Linardou, H., Siannis, F., Kosmidis, P.,Bafaloukos, D., Murray, S., 2010. Somatic EGFR mutation andgene copy gain as predictive biomarkers for response totyrosine kinase inhibitors in non-small cell lung cancer. Clin.Cancer Res. 16, 291e303.

    Delattre, O., Zucman, J., Plougastel, B., Desmaze, C., Melot, T.,Peter, M., Kovar, H., Joubert, I., Dejong, P., Rouleau, G.,Aurias, A., Thomas, G., 1992. Gene fusion with an ETS DNA-binding domain caused by chromosome translocation inhuman tumours. Nature 359, 162e165.

    Delattre, O., Zucman, J., Melot, T., Garau, X.S., Zucker, J.M.,Lenoir, G.M., Ambros, P.F., Sheer, D., Turc-Carel, C., Triche, T.J., et al., 1994. The Ewing family of tumors e a subgroup ofsmall-round-cell tumors defined by specific chimerictranscripts. N. Engl. J. Med. 331, 294e299.

    Druker, B.J., 2008. Translation of the Philadelphia chromosomeinto therapy for CML. Blood 112, 4808e4817.

    Druker, B.J., 2009. Perspectives on the development of imatiniband the future of cancer research. Nat. Med. 15, 1149e1152.

    Duffaud, F., Le Cesne, A., 2009. Imatinib in the treatment of solidtumours. Target. Oncol. 4, 45e56.

    Fagin, J.A., 2002. Perspective: lessons learned frommolecular geneticstudiesof thyroidcancere insights intopathogenesisandtumor-specific therapeutic targets. Endocrinology 143, 2025e2028.

    Frohling, S., Dohner, H., 2008. Chromosomal abnormalities incancer. N. Engl. J. Med. 359, 722e734.

    Gisselsson, D., 2001. Chromosomal instability in cancer: causesand consequences. Atlas Genet. Cytogenet. Oncol. Haematol..

    Gluckman, E., Devergie, A., Schaison, G., Bussel, A., Berger, R.,Sohier, J., Bernard, J., 1980. Bone marrow transplantation inFanconi anaemia. Br. J. Haematol. 45, 557e564.

    Guillaud-Bataille, M., Brison, O., Danglot, G., Lavialle, C.,Raynal, B., Lazar, V., Dessen, P., Bernheim, A., 2009. Twopopulations of double minute chromosomes harbor distinctamplicons, the MYC locus at 8q24.2 and a 0.43-Mb region at14q24.1, in the SW613-S human carcinoma cell line.Cytogenet. Genome Res. 124, 1e11.

    Hanna, W., 2001. Testing for HER2 status. Oncology 61, 22e30.Huret, J., 2009. MLL (myeloid/lymphoid or mixed lineage

    leukemia). Atlas Genet. Cytogenet. Oncol. Haematol. URL:http://AtlasGeneticsOncology.org/Genes/MLL.html.

    Huret, J.L., 2010. Atlas Genet. Cytogenet. Oncol. Haematol..Huret, J.L., Dessen, P., Bernheim, A., 2001. Atlas Genet. Cytogenet.

    Oncol. Haematol. 29, 303e304. updated. Nucl. Acids Res.Janoueix-Lerosey, I., Schleiermacher, G., Michels, E., Mosseri, V.,Ribeiro, A., Lequin, D., Vermeulen, J., Couturier, J.,Peuchmaur, M., Valent, A., Plantaz, D., Rubie, H., Valteau-Couanet, D., Thomas, C., Combaret, V., Rousseau, R.,Eggert, A., Michon, J., Speleman, F., Delattre, O., 2009. Overallgenomic pattern is a predictor of outcome in neuroblastoma. J.Clin. Oncol. 27, 1026e1033.

    Kallioniemi, A., Kallioniemi, O.P., Sudar, D., Rutovitz, D., Gray, J.W., Waldman, F., Pinkel, D., 1992. Comparative genomichybridization for molecular cytogenetic analysis of solidtumors. Science 258, 818e821.

    Lenoir, G., Preudhomme, J.L., Bernheim, A., Berger, R., 1981.Correlation between variant translocation and the expressionof immunoglobulin light chains in Burkitt-type lymphomasand leukemias. C. R. Seances Acad. Sci. III 293, 427e429.

    Lenoir, G.M., Preudhomme, J.L., Bernheim, A., Berger, R., 1982.Correlation between immunoglobulin light chain expressionand variant translocation in Burkitts lymphoma. Nature 298,474e476.

    Lynch, A., Morton, C., 2007. Uterus: leiomyoma. Atlas Genet.Cytogenet. Oncol. Haematol..

    Mandahl, N., 2000. Soft tissue tumors: lipoma/benign lipomatoustumors. Atlas Genet. Cytogenet. Oncol. Haematol..

    Mani, R.S., Tomlins, S.A., Callahan, K., Ghosh, A., Nyati, M.K.,Varambally, S., Palanisamy, N., Chinnaiyan, A.M., 2009.Induced chromosomal proximity and gene fusions in prostatecancer. Science 326, 1230.

    Mardis, E.R., 2009. New strategies and emerging technologies formassively parallel sequencing: applications in medicalresearch. Genome Med. 1, 40.

    Mardis, E.R., Ding, L., Dooling, D.J., Larson, D.E., McLellan, M.D.,Chen, K., Koboldt, D.C., Fulton, R.S., Delehaunty, K.D.,McGrath, S.D., Fulton, L.A., Locke, D.P., Magrini, V.J., Abbott, R.M., Vickery, T.L., Reed, J.S., Robinson, J.S., Wylie, T., Smith, S.M., Carmichael, L., Eldred, J.M., Harris, C.C., Walker, J., Peck, J.B., Du, F., Dukes, A.F., Sanderson, G.E., Brummett, A.M.,Clark, E., McMichael, J.F., Meyer, R.J., Schindler, J.K., Pohl, C.S.,Wallis, J.W., Shi, X., Lin, L., Schmidt, H., Tang, Y., Haipek, C.,Wiechert, M.E., Ivy, J.V., Kalicki, J., Elliott, G., Ries, R.E.,Payton, J.E., Westervelt, P., Tomasson, M.H., Watson, M.A.,Baty, J., Heath, S., Shannon, W.D., Nagarajan, R., Link, D.C.,Walter, M.J., Graubert, T.A., DiPersio, J.F., Wilson, R.K., Ley, T.J., 2009. Recurring mutations found by sequencing an acutemyeloid leukemia genome. N. Engl. J. Med. 361, 1058e1066.

    Mitelman, F., Johansson, B., Mertens, F., 2003. Database ofChromosome Aberrations in Cancer. http://cgap.nci.nih.gov/Chromosomes/Mitelman.

    Mitelman, F., Johansson, B., Mertens, F., 2007. The impact oftranslocations and gene fusions on cancer causation. Nat. Rev.Cancer 7, 233e245.

    Moldovan, G.L., DAndrea, A.D., 2009. How the fanconi anemiapathway guards the genome. Annu. Rev. Genet. 43, 223e249.

    NCI, NCBI, 2001. NCI and NCBIs SKY/M-FISH and CGH Database.http://www.ncbi.nlm.nih.gov/sky/skyweb.cgi.

    Nikiforov, Y.E., 2006. Radiation-induced thyroid cancer: what wehave learned from chernobyl. Endocr. Pathol. 17, 307e317.

    Nikiforova, M.N., Nikiforov, Y.E., 2009. Molecular diagnostics andpredictors in thyroid cancer. Thyroid 19, 1351e1361.

    Nikiforova, M.N., Stringer, J.R., Blough, R., Medvedovic, M.,Fagin, J.A., Nikiforov, Y.E., 2000. Proximity of chromosomalloci that participate in radiation-induced rearrangements inhuman cells. Science 290, 138e141.

    Nowell, P.C., Hungerford, D.A., 1960. Chromosome studies onnormal and leukemic human leukocytes. J. Natl. Cancer Inst.25, 85e109.

    OSullivan, R.J., Karlseder, J., 2010. Telomeres: protectingchromosomes against genome instability. Nat. Rev. Mol. Cell.Biol. 11, 171e181.

    http://AtlasGeneticsOncology.org/Genes/MLL.htmlhttp://cgap.nci.nih.gov/Chromosomes/Mitelmanhttp://cgap.nci.nih.gov/Chromosomes/Mitelmanhttp://www.ncbi.nlm.nih.gov/sky/skyweb.cgihttp://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003

  • M O L E C U L A R O N C O L O G Y 4 ( 2 0 1 0 ) 3 0 9e3 2 2322Pierotti, M., 2001. Chromosomal rearrangements in thyroidcarcinomas: a recombination or death dilemma. Cancer Lett.166, 1e7.

    Pinkel, D., Albertson, D.G., 2005. Array comparative genomichybridization and its applications in cancer. Nat Genet 37(Suppl), S11eS17.

    Pleasance, E.D., Cheetham, R.K., Stephens, P.J., McBride, D.J.,Humphray, S.J., Greenman, C.D., Varela, I., Lin, M.L.,Ordonez, G.R., Bignell, G.R., Ye, K., Alipaz, J., Bauer, M.J.,Beare, D., Butler, A., Carter, R.J., Chen, L., Cox, A.J., Edkins, S.,Kokko-Gonzales, P.I., Gormley, N.A., Grocock, R.J.,Haudenschild, C.D., Hims, M.M., James, T., Jia, M.,Kingsbury, Z., Leroy, C., Marshall, J., Menzies, A., Mudie, L.J.,Ning, Z., Royce, T., Schulz-Trieglaff, O.B., Spiridou, A.,Stebbings, L.A., Szajkowski, L., Teague, J., Williamson, D.,Chin, L., Ross, M.T., Campbell, P.J., Bentley, D.R., Futreal, P.A.,Stratton, M.R., 2010. A comprehensive catalogue of somaticmutations from a human cancer genome. Nature 463,191e196.

    Qian, Z., Joslin, J.M., Tennant, T.R., Reshmi, S.C., Young, D.J.,Stoddart, A., Larson, R.A., Le Beau, M.M., 2009. Cytogenetic andgenetic pathways in therapy-related acute myeloid leukemia.Chem. Biol. Interact..

    Riggi, N., Stamenkovic, I., 2007. The biology of Ewing sarcoma.Cancer Lett. 254, 1e10.

    Rowley, J.D., 1973. Letter: a new consistent chromosomalabnormality in chronic myelogenous leukemia identified byquinacrine fluorescence and Giemsa staining. Nature 243,290e293.

    Rowley, J.D., 1998. The critical role of chromosome translocationsin human leukemias. Annu. Rev. Genet. 32, 495e519.

    Sandberg, A., 2002. Cytogenetics and molecular genetics of boneand soft-tissue tumors. Am. J. Med. Genet. 115, 189e193.

    Santarius, T., Shipley, J., Brewer, D., Stratton, M.R., Cooper, C.S.,2010. A census of amplified and overexpressed human cancergenes. Nat. Rev. Cancer 10, 59e64.

    Schvartzman, J.M., Sotillo, R., Benezra, R., 2010. Mitoticchromosomal instability and cancer: mouse modelling of thehuman disease. Nat. Rev. Cancer 10, 102e115.

    Schwab, M., 2004. MYCN in neuronal tumours. Cancer Lett. 204,179e187.

    Sherbenou, D.W., Druker, B.J., 2007. Applying the discovery of thePhiladelphia chromosome. J. Clin. Invest. 117, 2067e2074.

    Sholl, L.M., Yeap, B.Y., Iafrate, A.J., Holmes-Tisch, A.J., Chou, Y.P.,Wu, M.T., Goan, Y.G., Su, L., Benedettini, E., Yu, J., Loda, M.,Janne, P.A., Christiani, D.C., Chirieac, L.R., 2009. Lungadenocarcinoma with EGFR amplification has molecularfeatures in never-smokers. Cancer Res. 69, 8341e8348.

    Soda, M., Choi, Y.L., Enomoto, M., Takada, S., Yamashita, Y.,Ishikawa, S., Fujiwara, S., Watanabe, H., Kurashina, K.,Hatanaka, H., Bando, M., Ohno, S., Ishikawa, Y., Aburatani, H.,Niki, T., Sohara,Y., Sugiyama,Y.,Mano,H., 2007. Identificationofthe transforming EML4-ALK fusion cancer. Nature 448, 561e566.

    Swerdlow, S.H., Campo, E., Harris, N.L., Jaffe, E.S., Pileri, S.A.,Stein, H., Thiele, J., Vardiman, J.W., 2008. WHO Classificationof Tumours of Haematopoietic and Lymphoid Tissues, fourthed. WHO Press, Geneva.

    Thompson, C.B., 2009. Attacking cancer at its root. Cell 138,1051e1054.

    Toledo, L.M., Lydon, N.B., Elbaum, D., 1999. The structure-baseddesign of ATP-site directed protein kinase inhibitors. Curr.Med. Chem. 6, 775e805.

    Tomita, N., Tokunaka, M., Nakamura, N., Takeuchi, K., Koike, J.,Motomura, S., Miyamoto, K., Kikuchi, A., Hyo, R.,Yakushijin, Y., Masaki, Y., Fujii, S., Hayashi, T., Ishigatsubo, Y.,Miura, I., 2009. Clinicopathological features of lymphoma/leukemia patients carrying both BCL2 and MYCtranslocations. Haematologica 94, 935e943.

    Toujani, S., Dessen, P., Ithzar, N., Danglot, G.L., Richon, C.,Vassetzky, Y., Robert, T., Lazar, V., Bosq, J., Da Costa, L.,Perot, C., Ribrag, V., Patte, C., Wiels, J.e., Bernheim, A., 2009.High resolution genome-wide analysis of chromosomalalterations in Burkitts lymphoma. PLoS ONE 4, e7089.

    Tronko, M.D., Howe, G.R., Bogdanova, T.I., Bouville, A.C.,Epstein, O.V., Brill, A.B., Likhtarev, I.A., Fink, D.J., Markov, V.V.,Greenebaum, E., Olijnyk, V.A., Masnyk, I.J., Shpak, V.M.,McConnell, R.J., Tereshchenko, V.P., Robbins, J., Zvinchuk, O.V., Zablotska, L.B., Hatch, M., Luckyanov, N.K., Ron, E.,Thomas, T.L., Voilleque, P.G., Beebe, G.W., 2006. A cohortstudy of thyroid cancer and other thyroid diseases after thechornobyl accident: thyroid cancer in Ukraine detected duringfirst screening. J. Natl. Cancer Inst. 98, 897e903.

    Valent, A., Venuat, A.M., Danglot, G., Da Silva, J., Duarte, N.,Bernheim, A., Benard, J., 2001. Stromal cells and humanmalignant neuroblasts derived from bone marrow metastasismay share common karyotypic abnormalities: the case of theIGR-N-91 cell line. Med. Pediatr. Oncol. 36, 100e103.

    Wang, Z.Y., Chen, Z., 2008. Acute promyelocytic leukemia: fromhighly fatal to highly curable. Blood 111, 2505e2515.

    Yunis, J.J., Mayer, M.G., Arnesen, M.A., Aeppli, D.P., Oken, M.M.,Frizzera, G., 1989. bcl-2 and other genomic alterations in theprognosisof large-cell lymphoma.N.Engl. J.Med. 320, 1047e1054.

    http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003http://dx.doi.org/10.1016/j.molonc.2010.06.003

    Cytogenomics of cancers: From chromosome to sequenceIntroductionOrigin of chromosomal rearrangements in cancerChromosomal rearrangementsFrom cytogenetics to cytogenomicsIn situ hybridization (ISH)CGHSequenceTypology of genomic abnormalities (chromosome and gene) (Table 3)Evolution of chromosomal abnormalities in tumorsLesions and tumors secondary to physical or chemical mutagensTargeted therapiesRole of clinical cytogenomics in the diagnosis, prognosis and in monitoring residual diseaseDatabasesProspectsConclusionAcknowledgementReferences