pantel ea - 2012 - circulating cancer cells and dna - v2

Upload: clark-xingzhi-zhang

Post on 02-Apr-2018

217 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    1/20

    Circulating Tumor Cells anCirculating Tumor DNA

    Catherine Alix-Panabieres,1,2,3

    Heidi Schwarzenbach,4 and Klaus Pantel4

    1University Medical Center, Saint-Eloi Hospital, Institute of Research in BiotherapyLaboratory of Rare Human Circulating Cells, Montpellier, France; 2University MedCenter, Laboratory of Cell and Hormonal Biology, Arnaud de Villeneuve Hospital,

    Montpellier, France; 3University Institute of Clinical Research UM1 EA2415 Epidemiology, Biostatistics & Public Health; email: c-panabieres@chu-montpellier.

    4Institute of Tumor Biology, University Medical Center, Hamburg-Eppendorf,20246 Hamburg, Germany; email: [email protected]

    Annu. Rev. Med. 2012. 63:199215

    First published online as a Review in Advance on

    November 2, 2011The Annual Review of Medicine is online atmed.annualreviews.org

    This articles doi:10.1146/annurev-med-062310-094219

    Copyright c 2012 by Annual Reviews.All rights reserved

    0066-4219/12/0218-0199$20.00

    Keywords

    tumor cell dissemination, cell-free tumor DNA, allelic imbalance

    genetic alterations, epigenetic alterations

    Abstract

    Solidtumors derived fromepithelial tissues(carcinomas) are responfor 90% of all new cancers in Europe, and the main four tumor

    ties are breast, prostate, lung, and colon cancer. Present tumor stais mainly based on local tumor extension, metastatic lymph nod

    volvement, and evidence of overt distant metastasis obtained by iing technologies. However, these staging procedures are not sen

    enough to detect early tumor cell dissemination as a key event in tu

    progression. Many teams have therefore focused on the developme

    sensitive assays that allow the specific detection of single tumor cesmall amounts of cell-free tumor DNA in the peripheral blood ofcer patients. These methods allow the detection and characteriz

    of early metastatic spread and will provide unique insights into thology of metastatic progression of human tumors, including the ef

    of therapeutic interventions.

    199

    Click here for quick links to

    Annual Reviews content online,

    including:

    Other articles in this volume

    Top cited articles

    Top downloaded articles

    Our comprehensive search

    FurtherANNUAL

    REVIEWS

  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    2/20

    Circulating tumorcells (CTCs): cellsshed by the primarytumor into thebloodstream very earlyin tumor development

    Disseminated tumorcells (DTCs): CTCsthat have left the bloodcirculation and homedinto secondary organs

    MRD: minimalresidual disease

    BM: bone marrow

    Circulating cell-freetumor DNA: DNAreleased by apoptoticand necrotic cells of

    the primary tumorinto the bloodcirculation early intumor development

    INTRODUCTION

    Early during the formation and growth of aprimary tumor (e.g., breast, colon, or prostate

    cancer), cells are shed from the primary tumorand circulate through the bloodstream. These

    circulating tumor cells (CTCs) are very het-erogeneous and can be enriched and detected

    via different technologies based on their physi-cal and biological properties. CTC analyses are

    considered a real-time liquid biopsy in cancer

    patients.The prognosis of carcinoma patients, even

    with small primary tumors, is mainly deter-mined by the blood-borne dissemination of

    tumor cells from the primary site to distantorgans such as bone marrow, liver, lungs, or

    brain, and the subsequent outgrowth of thesecells in their new microenvironment (1, 2).

    Disseminated tumor cells (DTCs) are consid-ered micrometastases. They can remain in a

    dormant state for many years after completeresection of the primary tumor before giving

    rise to macrometastasis (3, 4). DTCs recircu-

    lating through the bloodstream may colonizeother distant organs, giving rise to secondary

    metastases. Interestingly, DTCs can even re-turn to the primary tumor, a process termed

    tumor self-seeding or cross-seeding, giving riseto aggressive metastatic variants. These DTCs

    could thereby potentially contribute to the de-velopment of local relapses (5, 6), although this

    provocative hypothesis requires support fromstudies in cancer patients.

    Minimal residual disease (MRD), i.e., the

    presence of DTCs, is undetectable by high-resolution imaging technologies. However,

    DTCs can now be identified in the bone mar-row (BM), lymph nodes, or circulating blood,

    using sensitive and specific assays (1, 4). BM iseasily accessible by needle aspiration through

    the iliac crest, and it plays the most prominentroleamongthedistantorgansasindicatororgan

    for MRD thus far. BM appears to be a commonhoming organ for DTCs derived from carcino-

    mas of different organs (7) and also might bea reservoir for DTCs with the capacity to re-

    enter other distant organs.

    For the follow-up of cancer patients, s

    quential analyses are pivotal. Because BM nedle aspiration is far more invasive than sam

    pling of peripheral blood, research groups acurrently evaluating the clinical utility of t

    mor cells in the blood rather than BM to asseprognosis and monitor systemic therapy (4).

    number of innovative technologies to impromethods for CTC detection with extraordina

    ily high sensitivity have recently been deveoped, including CTC microchips, filtration d

    vices, quantitative RT-PCR assays, and aut

    mated microscopic systems (1, 4). However, tspecificity and clinical utility of these metho

    still have to be demonstrated in large prospetive multicenter studies to reach the high lev

    of evidence required for introduction into cliical practice.

    Concentrations of circulating cell-free tmor DNA are high in cancer patients com

    pared to healthy individuals. Early in tumor dvelopment, apoptotic and necrotic cells of t

    primary tumor release DNA into the bloo

    stream (Figure 1). In the peripheral bloothis cell-free DNA circulates predominan

    in the form of nucleosomes, indicating thit retains at least some features of the n

    clear chromatin. This DNA can be extractfrom blood, and its genetic and epigenetic a

    terations can be determined (8). Epigene

    modifications include DNA methylation anconfiguration changes in chromatin histoproteins (9). In chromosomal regions of tumo

    associated genes, epigenetic modifications maffect important regulatory mechanisms f

    the pathogenesis of malignant transformatio

    DNA methylation of the cytosine base in Cpdinucleotides, which are found as isolated

    clustered CpG islands, induces gene represion by inhibiting the access of transcriptio

    factors to their binding sites. Inactivation

    tumor suppressor genes by promoter hypemethylation is thought to play a crucial role tumorigenesis.

    However, the aberrations of the cell-frDNA in blood do not always match those

    the primary tumor (8). This discrepancy m

    be ascribed at least partly to CTCs or DTC

    200 Alix-Panabieres Schwarzenbach Pantel

  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    3/20

    CTC

    + DNA

    Primary tumoror metastasis

    Extraction ofcell-free DNA

    b Capillary gel electrophoresis

    c DNA sequencing

    Blood

    vesse

    l

    1

    2

    0

    600

    1,200

    0150

    LOH120 130

    220 230

    Wild type DNA

    Microsatellite PCRusing cell-ree DNA

    or detection o LOH

    Methylation-sensitivePCR using bisulfte-converted cell-ree

    DNA or detection omethylated DNA

    Cell-ree blood DNA

    Length of the PCR product (bp)

    F

    luorescence

    intensity

    600

    1,200T TAT T T CG G GT CGAT T TAG AG

    TGAG GGAT GAT TA AAGGTTTTTT

    a

    Figure 1

    Detection of genetically and epigenetically altered DNA in blood. High levels of cell-free tumor DNA circulate in the blood of cpatients. (a) This tumor DNA found in blood can be released from either the primary tumor or (micro)metastasis, or apocirculating tumor cells (CTCs). This DNA can be extracted from blood, and the genetic and epigenetic alterations can be determTo detect loss of heterozygosity (LOH) on cell-free DNA, extracted DNA is amplified in a polymerase chain reaction (PCR)-bafluorescence microsatellite analysis using a gene-specific primer set binding to tumor suppressor genes. The fluorescence-labeledproducts can be separated by capillary gel electrophoresis and detected by a fluorescence laser. In the diagram ( b), the abscissa inthe length of the PCR product; the ordinate gives information on the fluorescence intensity represented as peaks. The upper anparts of the diagram show the PCR products derived from wild-type DNA (from leukocytes) and plasma DNA, respectively. Asdepicted by the two peaks of the amplified wild-type DNA, both alleles are intact, whereas the lower peak of the PCR product dfrom the plasma DNA shows LOH (arrow). (c) To detect cell-free methylated DNA, extracted DNA is denatured and treated wisodium bisulfite. In a methylation-sensitive PCR, the modified DNA is amplified with gene-specific primers. Because sodium biconverts unmethylated cytosine residues into uracil, in contrast to methylated cytosine, the methylation pattern can be determinDNA sequencing.

    www.annualreviews.org Circulating Tumor Cells and Tumor DNA 201

    Circulating cancer cell-freeDNA can be released from (1)he primary tumor or(micro)metastasis or (2)apoptotic circulating tumorcells (CTCs)

  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    4/20

    which can also release their DNA into the blood

    circulation (Figure 1).This review focuses on the detection and

    further characterization of individual CTCsand circulating cell-free DNA as real-time liq-

    uid biopsies that can help identify therapeutictargets and potential mechanisms of resistance

    to therapy. This strategy might contribute tothe development of improved individualized

    targeted treatment of cancer patients.

    CIRCULATING TUMOR CELLS

    Technologies Used forCTC Detection

    The detection of CTCs in peripheral blood of

    cancer patients holds great promise, and manyexciting technologies have been developed over

    the past few years. However, detecting CTCsremains technically challenging. CTCs occur

    at very low concentrations of one tumor cell

    in the background of millions of blood cells.Their identification and characterization re-

    quire extremely sensitive and specific analyticalmethods, which are usually a combination of

    enrichment and detection procedures. Below,we briefly introduce some of the key tech-

    nologies of CTC detection, although a com-prehensive overview of all existing assays and

    publications would be beyond the scope of thisarticle.

    CTC enrichment. CTC enrichment includesalargepaneloftechnologiesbasedonthediffer-

    ent properties of CTCs that distinguish themfrom the surrounding normal hematopoietic

    cells (Figure 2), including physical proper-

    ties (size, density, electric charges, deformabil-ity) and biological properties (surface protein

    expression, viability, and invasion capacity).

    Physical properties have the advantage thatthey allow CTC separation without labeling.Methods based on physical properties include

    density gradient centrifugation (Ficoll, Onco-Quick); filtration through special filters, e.g.,

    theISET(IsolationbySizeofEpithelialTumorCells) (10, 11) or a novel three-dimensional

    microfilter (12); a new versatile labelfr

    biochip using the unique differences in size andeformability of cancer cells (larger and stiff

    than blood cells) (13, 14); a microfluidic devicombining multi-orifice flow fractionati

    (MOFF) and the dielectrophoretic (DEcell separation technique (15); and a diele

    trophoretic fieldflow fractionation (DEFFF) device that allows isolation of viab

    CTCs by different response to DEP due to dference in size and membrane properties (16

    Biological properties are mainly used in im

    munological procedureswith antibodiesagaineither tumor-associated antigens (positive s

    lection) or the common leukocyte antigCD45 (negative selection). Immunomagne

    systems target an antigen with an antibody this coupled to a magnetic bead, and the antige

    antibody complex is subsequently isolated vexposure to a magnetic field. Positive selectio

    is usually carried out with antibodies againthe epithelial cell adhesion molecule (EpCAM

    and subsequent immunocytological detectio

    of CTCs is performed with antibodies to ctokeratins,the intermediatefilaments of epith

    lial cells (1). Among the current EpCAM-bastechnologies, the FDA-approved CellSearch

    system has gained considerable attention ovthe past seven years (4) and is the gold sta

    dard for all new CTC detection metho

    (17, 18).At present, there is a focus on the develo

    ment of microfluidic devices (chips), whi

    can handle very small blood volumes. A mcrofluidic platform called a CTC-chip consi

    of an array of anti-EpCAM antibody-coatmicroposts (1922). The high CTC counts

    nonmetastatic cancer patients and the freque

    detection of positive events in healthy controwarrant further investigations on the specifici

    of this assay. Recently, a new CTC-chip call

    Ephesia, which uses columns of biofunctioalized superparamagnetic beads self-assemblin a microfluidic channel onto an array

    magnetic traps (23), and another microfluidsystem using high-throughput selection, en

    meration, and electrokinetic manipulation low-abundance CTCs have been introduc

    202 Alix-Panabieres Schwarzenbach Pantel

  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    5/20

    Blood vessel

    a Physical properties

    Size/deformability Density Electric charges Marker proteins

    Invasive capacity

    EpCAM

    CD45

    CAM

    CD45+

    EpCAM+ EpCAM+

    EpCAM+

    CD45+

    EpCAM+

    MagSweeper

    Positive Positive Mic

    Positive

    N

    Ep

    PositiveGlyco A

    Fluo-CAM

    b Biological properties

    Leukocytes

    RBC

    CTC

    +

    + + +

    + + +

    + + +

    +

    +++

    +

    Figure 2

    Enrichment of circulating tumor cells (CTCs) from the peripheral blood of cancer patients is based on physical or biological proof CTCs. (a) Physical properties include size (membrane filter devices), deformability (microfluidic system in a chip), density (Ficentrifugation), and electric charge (dielectrophoresis). (b) Biological properties include expression of cell surface markers and incapacity. Cell surface markers include an epithelial cell adhesion molecule (EpCAM) for positive selection and CD45 for negativselection; anti-EpCAM or anti-CD45 antibodies conjugated with magnetic beads, used to enrich CTCs in a magnetic field; andanti-EpCAM antibodies on microposts or columns of nanobeads. Invasive capacity refers to adherence and invasion of fluorescematrix. Abbreviations: glyco A, glycophorin A (a 131-amino-acid protein present at the extracellular surface of the human red blcell); CAM, cell adhesion matrix; fluo-CAM, fluorescent cell adhesion matrix; RBC, red blood cells.

    (24); validation of these assays is still ongoing.Microdevices can handle cell numbers and sam-

    ple volumes at least 10 times smaller (

  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    6/20

    Epithelial-mesenchymaltransition (EMT):trans-differentiation ofepithelial cells intomesenchymal cells,

    often activated duringcancer invasion andmetastasis. Epithelialcells lose their cellularpolarity, and theirepithelial cellcell andcellmatrix adhesioncontacts areremodeled. E-cadherinand some cytokeratinsare downregulated;markers ofmesenchymal cells

    such as vimentin andN-cadherin areupregulated

    samples derived from patients with metastatic

    disease, which contained 12 23 CEpC per9 ml of blood, whereas such cells were not

    identified in any samples from healthy donors.Through the use of a 3D nanostructured

    substrate or nanofly paper technologya silicon-nanowire (SiNW) array coated with

    anti-EpCAM antibodiesCTCs can also becaptured efficiently (26). A specialized laser

    scanning cytometer (MAINTRACTM) providesanother EpCAM-based approach, which com-

    bines the cell-sorting speed of flow cytometry

    with the power to analyze the morphologicalproperties of a single positive cell (27). As al-

    most all patient samplesyield an extraordinarilyhigh number of CTCs with this method (50 to

    3105 or three log units higher than observedwith the other approaches), careful validation

    studies on the specificity of this technology arewarranted.

    Importantly, a subset of epithelial can-cer cells is likely to undergo epithelial-

    mesenchymal transition (EMT) prior to en-tering the peripheral circulation. EMT is a

    key developmental program that is often ac-tivated during cancer invasion and metasta-

    sis. It is thought to be linked to the gain of

    cancer stem cell properties. During EMT, E-cadherin and some cytokeratins are downreg-

    ulated, whereas markers of mesenchymal cells

    such as vimentin and N-cadherin are upregu-lated. By undergoing this transition, tumor cellsmay escape detection by conventional methods

    (28). Although there is an ongoing debate onthe relevance of EMT in cancer patients as op-

    posed to experimental studies in model systems(29), recent work suggested that EMT might

    particularly affect tumor cells with stem cell

    like properties (30, 31). Therefore, in recentyears, there has been great interest in investi-

    gating EMT markers in CTCs (32, 33). Inter-

    estingly, it has been reported that the presenceof mesenchymal markers on CTCs more accu-rately predicted worse prognosis than the ex-

    pression of cytokeratins alone, demonstratingthat current assays based on epithelial antigens

    may miss the most aggressive CTC subpopula-tion (3436). Thus, there is an urgent need for

    optimizing CTC detection methods throu

    the inclusion of markers that are not repressduring EMT but still allow the analyst to disti

    guish CTCs from the surrounding blood celFor example, vimentin, the mesenchymal inte

    mediate filament frequently expressed in carcnoma cellsthathave undergone an EMT, is al

    expressed in blood cells and thereforecannotused as a CTC marker.

    Another way to overcome the problem false negative findings due to EMT-like CTC

    is a functional cell separation method called t

    collagen adhesion matrix (CAM) assay, whihas been reported in breast, prostate, and ova

    ian cancer. CAM ingestion and epithelial immunostaining identifies CTCs based on the

    invasive properties in vitro (37).Besides the choice of the appropriate CT

    marker,thelimitedbloodsamplevolumesavaable from cancer patients may impose a ser

    ous limitation on the detection of rare evensuch as CTCs. A completely new concept

    enrichment and detection of CTCs has ther

    fore been introduced and validated in a moumodel (38). As CTCs express folate and urok

    nase plasminogen activator (uPA) receptothey can be dually targeted in the bloodstrea

    with folate-conjugated nanotubes and manetic uPA-conjugated nanoparticles and subs

    quently detected with two-color photoacous

    flow cytometry. Future studies on humans wtell us whether this new platform can diagnotumor cell dissemination in cancer patients.

    CTC detection. The CellSearch R syste

    and the two CTC chips described above uthe same identification step: cells are fluore

    cently stained for cytokeratins CK8, CK1

    and CK19, the common leukocyte antigCD45, and a nuclear dye (4,6-diamino-

    phenylindole; DAPI). Through multicolor im

    age analysis with a fluorescent microscopCTCs are defined as CK+/CD45/DAPcells.

    In order to detect only viable CTCs, thfunctional EPISPOT assay (for EPithelial Im

    munoSPOT), which can be added to any kinof enrichment step, was introduced for CT

    204 Alix-Panabieres Schwarzenbach Pantel

  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    7/20

  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    8/20

    Lung-

    CTC

    Bone-C

    TC

    Primary-

    CTC

    Primary-

    CTC

    Liver-CT

    C

    Primarydissemination

    DTC

    DTC

    Distantorgans

    Primary tumor Local relapse

    Secondarydissemination

    Blood

    vesse

    l

    Blood

    vesse

    l

    Figure 3

    Editing of the circulating tumor cell (CTC) pool by the microenvironment of secondary metastatic sites.Tumor cells leave the primary tumor and circulate through the bloodstream. Each time the CTCs reach anew niche (distant organs, e.g., bone, liver, or lung in breast cancer), they undergo an organ-specificmimetism and may leave this site with a new organ-specific signature. Abbreviation: DTC, disseminatedtumor cell.

    Osteomimetism:DTCs acquire bone-like properties to adapt

    to the bonemicroenvironment,adopting anosteoblast-likephenotype and alteringthe functions ofosteoblasts andosteoclasts

    clinical implications because niche signals mayregulate tumor dormancy and sensitivity tochemotherapy. In this context, it is important

    to mention that DTCs established from BM ofcancer patients express a particular set of stress

    proteins of the unfolded protein response thatenable cells to survive the hypoxic conditions

    present in the niche areas and render them

    more resistant to chemotherapy (49). More-over, a transcriptome analysis of osteotropic

    breast tumor cells found in the BM has revealed

    an osteoblast-like phenotype: these tumor cellsunderwent an osteomimetism in the bone byexpressing a pool of genes normally expressed

    by osteoclasts or osteoblasts (5052). We canspeculate that such an adaptation may take

    place each time a CTC reaches a new niche.Thus, it is conceivable that CTCs may acquire

    an organ-mimetic phenotype, and CTexpressing lung-, bone-, and liver-specigenes after a particular niche adaptation mig

    recirculate back into the blood (Figure 3).

    Molecular Characterization of CTCs

    Further characterization of CTCs is pivotal

    provide insights into the complex biology tumor cell spread, with important implicatio

    for defining therapeutic targets and elim

    nating MRD. Because of the higher numbof detectable CTCs, the majority of resuconcerning genetic aberrations publish

    thus far are derived from investigations CTCs from patients with castration-resista

    prostate cancer (CRPC). Genotyping usioligonucleotide-array comparative genom

    206 Alix-Panabieres Schwarzenbach Pantel

  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    9/20

    hybridization revealed copy-number profiles

    in CTCs from CRPC patients similar to thoseobserved in paired tumor tissues (53). By

    multicolor fluorescence in situ hybridization(FISH), homogeneity in ERG oncogene

    rearrangement status in CTCs from CRPCpatients was observed in contrast to significant

    heterogeneity of androgen receptor (AR)copy-number gain and PTENloss (54). Swen-

    nenhuis et al. reported that the majority ofCTCs in hormone-refractory prostate cancer

    are aneuploid. Furthermore, heterogeneous

    copy numbers of chromosomes 1, 7, 8, and 17were observed (55). In a considerable number

    of CTC samples from patients with CRPC,amplifications of the AR gene locus could be

    detected (56, 57). Very recently, mutations inAR were identified in CTC-enriched periph-

    eral blood samples from CRPC patients byapplying Transgenomics WAVE R denaturing

    high-performance liquid chromatographytechnology followed by direct sequencing

    (58). These studies demonstrate that CTCsdetected with the CellSearch R system have

    tumor-specific genomic characteristics butcan still show a marked genetic heterogeneity

    depending on the genomic aberration analyzed.

    Global gene expression profiles identifiedfor CTCs from patients with metastatic breast

    cancer might be useful to distinguish normal

    donors from cancer patients (59). One of thekey questions is whether CTCs exhibit a can-cer stem cell phenotype (see above). TWIST1,

    a transcription factor pivotal for metastasis bypromoting EMT, was part of the gene expres-

    sion signature identified in EpCAM-enrichedcells from BM of breast cancer patients af-

    ter chemotherapy. TWIST1 expression was as-

    sociated with distant metastasis and local tu-mor progression (60). Using the AdnaTest

    TumorStemCell/AdnaTest EMT RT-PCT

    assay, Aktas et al. also identified featurescharacteristic for stem cells and EMT in amajor proportion of CTCs from metastatic

    breast cancer patients (61). A subpopulation ofCTCs with the putative stem cell phenotypes

    CD44+/CD24/low or ALDH1high/CD24/low

    was recently reported in a subpopulation of

    patients with metastatic breast cancer using

    triple-marker immunofluorescence microscopy(62). In a subset of breast cancer stem cells with

    thepotential to self-renew, Notch seems to rep-resent a genetic biomarker that is frequently

    coexpressed with the HER2 oncogene (63). In arecent study, Gazzaniga et al. identified a puta-

    tive drug-resistance profile of CTCs with pre-dictive value for response to chemotherapy, in-

    dependentofthetumortypeandthestageofthedisease, probably relevant for the individualiza-

    tion of chemotherapy in cancer patients (64).

    Clinical Relevance of CTC Analyses

    Encouraging results concerning an associa-

    tion between CTC detection and metastaticprogression in patients with metastatic breast,

    prostate, and colorectal cancer have been re-cently published using the CellSearch R system

    (56, 6570). Results indicated that CTCs in pe-ripheral blood of metastatic breast cancer pa-

    tients at any time during therapy directly re-flect the patients response, or lack of response,

    to therapy (71) and are therefore superior oradditive to conventional imaging methods (66,

    72, 73). The randomized trial SWOG S0500,

    led by the Southwest Oncology Group (74),which is expected to enroll 500 patients with

    metastatic breast cancer, is now prospectively

    addressing the clinical utility of CTC measure-ments in metastatic breast cancer patients. Thekey question for clinical use is whether the

    change in therapeutic interventions based onCTC counts will result in a measurable benefit

    (e.g., longer progression-free survival) for thecancer patient.

    A challenging task for new techniques in-

    tended to analyze CTCs/DTCs is to enabledetection and monitoring of MRD in patients

    with nonmetastatic cancer. Recently, promis-

    ing results derived from patients with non-metastatic breast cancer enrolled in neoadju-vant treatment studies. After an extension of

    the median follow-up to 36 months, CTC de-tection before chemotherapy became an in-

    dependent prognostic factor for both distantmetastasis-free survival and overall survival

    www.annualreviews.org Circulating Tumor Cells and Tumor DNA 207

  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    10/20

    Loss ofheterozygosity(LOH): a commonform of allelicimbalance, which isused to identify

    genomic regions thatharbor tumorsuppressor genes andto characterize tumorstages and progression

    Microsatellite: ashort, highly repetitiveDNA sequence of 26base pairs occurring asa repetition of di-, tri-,or tetranucleotides. Inthe genome,microsatellites are

    widely spread andusually lie innoncoding regions

    (75). Interestingly, there was no significant cor-

    relation between changes in CTC counts andresponse of the primary tumor to chemother-

    apy, which is the current standard method toassess therapeutic efficacy in neoadjuvant ther-

    apies. Thus, CTCs may have a different re-sponse pattern than primary tumor cells, and

    measuring changes in CTC counts may pro-vide additional information on the response

    of an individual patient. Follow-up analysesof two German trials in breast cancer us-

    ing the CellSearch R technologythe GEPAR-

    Quattro trial on neoadjuvant chemotherapyand additional trastuzumab treatment (if in-

    dicated), and the SUCCESS trial on adjuvantchemotherapyare still ongoing and will show

    whether the observed decreases in CTC rateswill be associated with an improved survival

    rate (76, 77). As recently described by Xenidiset al., patients with detectable CK19 mRNA

    post chemotherapy had a significantly reducedoverall and disease-free survival (78).

    DTCs/CTCs capable of surviving chemo-therapy probably persist in a dormant, non-

    proliferating state over many years (79, 80).

    Targeted anticancer therapies that are moreeffective than current therapies and less harm-

    ful to normal cells are being developed to killthese dormant cells specifically, or at least

    block their outgrowth into overt metastases.

    Currently, the choice of a targeted therapy foran individual patient is made upon analyzingthe primary tumor for the expression and/or

    genomic status of a specific molecular target.However, this is often hampered by the hetero-

    geneity and plasticity of individual tumor cellsin this tissue (81). Several studies have shown

    that metastatic cells may have phenotypic and

    genotypic characteristics that are distinct fromthe bulk of the primary tumor (82), which can

    be explained by the facts that (a) the metastatic

    subclone within the primary tumor might besmall and easily missed and (b) metastatic cellsmay gain additional genomic characteristics

    over time and develop independently from theprimary tumor (83). Thus, the direct analysis

    of metastatic cells may provide important addi-tional information for stratification of patients

    to expensive therapies with considerable si

    effects. For example, information regardithe expression of the estrogen receptor or th

    HER2 oncogene on CTCs might be helpffor stratification and monitoring of endocri

    therapy or therapy with trastuzumab (humanti-HER2 antibodies), which are curren

    used in breast cancer only based on the analyof the primary tumors. However, an increasin

    number of reports indicates a clear discrepanbetween the primary tumor status of these ta

    gets and their expression on CTCs and DTC

    (76, 8486).

    CIRCULATING TUMOR DNA

    Investigations on combined analyses of CTCand cell-free circulating tumor DNA have ju

    begun and showed a potential relationship btween tumor DNA in blood serum/plasm

    CTCs in blood, and DTCs in BM of cancer ptients (8792), suggesting circulating cell-fr

    tumor DNA as a new biomarker for metastaspread in solid tumors.

    In patients with primary head andneck squmous cell carcinoma, allelic imbalance, such

    loss of heterozygosity (LOH), in serum DN

    measured by a PCR-based fluorescence mcrosatellite analysis has for the first time be

    reported to be helpful in identifying patients

    risk for distant metastasis. Since these patienwith tumor DNA in their serum had a highrate of distantmetastasis,CTCs may contribu

    to the presence of serum tumor DNA (89).In prostate cancer patients, the presen

    of CTCs was detected by an epithelial immunospot assay and significantly correlat

    with tumor stage, increasing Gleason scor

    and frequencies of LOH at three markeencoding the cytoskeletal protein demat

    the inhibitor of the cyclin-dependent kina

    CDKN2/p16, and BRCA1 (90). These findinshowed a relationship between the presenof CTCs and the circulating tumor-associat

    DNA in blood. Therefore, circulating cell-frtumor DNA might become a valuable sour

    of information on metastatic progression prostate cancer and contribute to a bett

    208 Alix-Panabieres Schwarzenbach Pantel

  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    11/20

    understanding of the early steps of the

    metastatic cascade in this carcinoma type.In breast cancer, CTCs and tumor-specific

    alterations in cell-free plasma DNA were com-pared as markers for subclinical metastases. In

    the study of Shaw et al., CTCs, DNA concen-tration/integrity, and evidence ofHER2 ampli-

    fication were measured in patients with indeter-minate early or metastatic disease. CTCs and

    plasma DNA analyses together helped to iden-tify patients with metastatic disease (91).

    The presence of CTCs pointed not only

    to genetic aberrations but also to epigeneticalterations of cell-free DNA. In stage IV

    melanoma patients before administration ofbiochemotherapy, the number of CTCs de-

    tected in blood significantly correlated with themethylation of cell-free RASSF1A and RAR-

    2 DNA molecules (87). To assess the presenceof CTCs, a multimarker quantitative real-time

    RT-PCR assay was performed in this study.Fordetectionof thecell-free, methylatedDNA,

    the PCR products of sodium bisulfitemodifiedDNA were analyzed by capillary array elec-

    trophoresis. CTCs and methylated RASSF1A

    and RAR-2 on cell-free DNA were associ-ated with the outcome of patients. Patients with

    CTCs and these methylated markers in theirblood showed significantly poorer response to

    biochemotherapy and shorter time to progres-

    sion and overall survival. These findings indi-cate that a combined assessment of cell-freemethylated DNA and CTCs in blood may be

    a useful determinant of disease status and ef-ficacy of systemic therapy of melanoma (87).

    Also, in breast cancer patients, the high in-cidence of cell-free methylated DNA corre-

    lated with the occurrence of CTCs in the pe-

    ripheral blood (92). Based on the relationshipof cell-free, methylated adenomatous polypo-

    sis coli (APC), Ras association domain family

    protein 1A (RASSF1A), and estrogen receptor1 (ESR1) DNA molecules with CTCs, it wasdeduced that CTCs are a potential source of

    circulating tumor-specific DNA, and that highnumbers of CTCs and methylated DNA in

    the blood are both indicators of more aggres-sive breast tumors (92). Moreover, in another

    study, the detection of methylated APC and

    glutathione s-transferase pi1 (GSTP1) DNA inserum of breast cancer patients correlated with

    the presence of CTCs, which were measured bya modified immunomagnetic Adna test. Both

    methylated DNA and CTCs correlated witha more aggressive tumor and advanced disease

    (88).Taken together, blood may be a reservoir

    collecting DNA from different sources, includ-ing CTCs and occult micrometastatic deposits

    in secondary organs. Combining these DNA

    analyses with the screening for CTCs mayprovide additional information for molecular

    staging of tumors and monitoring of tumorprogression.

    CONCLUSIONS

    A considerable number of rare-cell detection

    techniques have been developed during recentyears and are being continuously improved

    by several working groups. DTCs in BM andof CTCs in blood of cancer patients can be

    detected years before the occurrence of distantovert metastases. Nevertheless, analysis of

    DTCs/CTCs is still not part of routine tumor

    staging in clinical practice. This is mainly dueto the low number of these cells detectable with

    the currently available methods, limiting their

    value as a liquid biopsy especially in patientswith early-stage tumors. The new approacheshave to be evaluated for reproducibility,

    sensitivity, and specificity in order to becomeapplicable for clinical practice. In addition, we

    need to identify the most aggressive subset ofCTCs that are the metastasis-initiating cells

    (93). Therefore, we need to develop better

    strategies that are also able to isolate andidentify EMT-like subpopulations of tumor

    cells with downregulated epithelial-specific

    protein expression. Moreover, it might becomepossible to identify the tissue origin of CTCsby the detection of organ-specific metastatic

    signatures in these cells using expressionprofiling, which would help to localize small,

    occult metastatic lesions and guide furtherdiagnostic and therapeutic strategies.

    www.annualreviews.org Circulating Tumor Cells and Tumor DNA 209

  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    12/20

    Information about CTC/DTC status may

    be used to assess the individual prognosis ofcancer patients and to decide who should re-

    ceivesystemictherapiesaimedtopreventrecur-rences and metastatic relapses. Furthermore,

    CTC/DTC measurements within clinical tri-als might serve as important biomarkers for

    real-time monitoring of the efficacy of systemictherapies in individual cancer patients, and

    might thereby support accelerating drug devel-opmentanddefining subpopulations of patients

    with the highest treatment benefit. Moreover,

    CTC/DTC analysis for downstream compo-nents within signal transducing pathways that

    influence new targeted therapies (e.g., KRASmutations in EGFR-targetedtherapies or PI3K

    mutations in HER2-targeted therapies) mightprovide new insights into the complex mecha-

    nisms of drug resistance.Tumor-associated nucleic acids may be

    enriched in the blood of patients with tumors,and these cell-free nucleic acids may reflect

    MRD. It is likely that at least part of thecell-free DNA found in the blood of cancer

    patients is derived from CTCs or DTCs

    present in secondary organs, which explai

    the discrepancies between the genetic patterobserved in the blood and in the autologo

    primary tumors. The investigation of tumospecific genetic and epigenetic profiles reli

    on plasma and serum, which are easily obtainfor the development of clinical assays. Alo

    or in combination with the analyses of CTCcirculating cell-free DNA assays might allo

    monitoring of metastatic progression aidentification of mutations relevant for t

    response of patients to targeted therapies (20

    However, the analysis of circulating cell-frtumor DNA is also challenging and requir

    stringent technical quality control measur(8).

    Independent of the technical approach usethe key question to be addressed in clinic

    intervention trials is how the assessment CTCs and/or circulating cell-free tumor DN

    will guide therapy toward a more efficieelimination of metastatic cells. Detection an

    eradication of metastatic cells at an earlitime point clearly has the potential to decreacancer mortality.

    SUMMARY POINTS

    1. CTCs can be enriched using extremely sensitive and specific technologies based on their

    physical and biological properties.

    2. CTCs are usually detected by immunocytochemical or RT-PCR-based assays for

    epithelial-specific proteins or mRNA species. Cytokeratins are the most widely usedCTC markers.

    3. CTCs can be characterized at the molecular level by detecting chromosomal anomalies(i.e., gain, loss, mutations of genes) or gene expression signatures.

    4. EMT- and stem celllike phenotypes are two key characteristics of the more aggressivesubsets of CTCs.

    5. Each time CTCs reach a new distant organ (e.g., bone, liver, lung), they may acquire an

    organ-specific phenotype following particular niche adaptation.

    6. CTCs are clinically relevant because they are (a) an independent prognostic factor incancer patients and (b) a predictive biomarker for treatment efficacy.

    7. Circulating cell-free tumor DNA might become a biomarker for metastatic spread in

    solid cancer. Blood is a compartment collecting tumor DNA shed by apoptotic/necrotictumor cells derived from the primary tumor as well as from occult (micro)metastatic

    deposits, DTCs, and CTCs.

    210 Alix-Panabieres Schwarzenbach Pantel

  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    13/20

    FUTURE ISSUES

    1. Future CTC assays need to be evaluated for reproducibility, sensitivity, and specificity

    in order to become applicable for clinical practice. Better strategies that are also ableto isolate and detect EMT-like subsets of CTCs with downregulated epithelial-specific

    protein expression must be developed.

    2. Identifying the tissue origin of CTCs by the detection of organ-specific metastatic sig-

    natures in these cells using expression profiling of single cells or CTC-enriched cellfractions may help clinicians in the future to localize small, occult metastatic lesions and

    guide further diagnostic and therapeutic strategies.

    3. Understanding new signal-transducing pathways at the CTC/DTC level should allow

    the discovery of new targeted therapies and specific mechanisms of drug resistance.

    4. Developing standardized methodologies for circulating cell-free DNA analyses and val-

    idating these assays in large prospective clinical studies are indispensable to implementthis interesting approach in the clinical management of cancer patients.

    DISCLOSURE STATEMENTK.P. has received a research grant and speakers honoraria from Veridex. C.A.P. has received

    honoraria from Veridex.

    LITERATURE CITED

    1. Pantel K, Brakenhoff RH, Brandt B. 2008. Detection, clinical relevance, and specific biological properties

    of disseminating tumour cells. Nat. Rev. Cancer8:32940

    2. Hanahan D, Weinberg RA. 2011. Hallmarks of cancer: the next generation. Cell144:64674

    3. Aguirre-Ghiso JA. 2007. Models, mechanisms and clinical evidence for cancer dormancy.Nat. Rev. Cancer

    7:83446

    4. Pantel K, Alix-Panabieres C, Riethdorf S. 2009. Cancer micrometastases. Nat. Rev. Clin. Oncol. 6:339515. Kim MY, Oskarsson T, Acharyya S, et al. 2009. Tumor self-seeding by circulating cancer cells. Cell

    139:131526

    6. Aguirre-Ghiso JA. 2010. On the theory of tumor self-seeding: implications for metastasis progression in

    humans. Breast Cancer Res. 12:304

    7. Pantel K, Brakenhoff RH. 2004. Dissecting the metastatic cascade. Nat. Rev. Cancer4:44856

    8. A comprehens

    summary of cell-

    nucleic acids.

    8. Schwarzenbach H, Hoon DS, Pantel K. 2011. Cell-free nucleic acids as biomarkers in cancer

    patients. Nat. Rev. Cancer11:42637

    9. Cedar H, Bergman Y. 2009. Linking DNA methylation and histone modification: patterns and paradigms.

    Nat. Rev. Genet. 10:295304

    10. Vona G, Sabile A, Louha M, et al. 2000. Isolation by size of epithelial tumor cells: a new method for the

    immunomorphological and molecular characterization of circulating tumor cells. Am. J. Pathol. 156:57

    6311. Pinzani P, Salvadori B, Simi L, et al. 2006. Isolation by size of epithelial tumor cells in peripheral blood

    of patients with breast cancer: correlation with real-time reverse transcriptase-polymerase chain reaction

    results and feasibility of molecular analysis by laser microdissection. Hum. Pathol. 37:71118

    12. Zheng S, Lin HK, Lu B, et al. 2011. 3D microfilter device for viable circulating tumor cell (CTC)

    enrichment from blood. Biomed. Microdevices13:20313

    13. Tan SJ, Lakshmi RL, Chen P, et al. 2010. Versatile label free biochip for the detection of circulating

    tumor cells from peripheral blood in cancer patients. Biosens. Bioelectron. 26:17015

    www.annualreviews.org Circulating Tumor Cells and Tumor DNA 211

  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    14/20

    14. Bhagat AA, Hou HW, Li LD, et al. 2011. Pinched flow coupled shear-modulated inertial microfluid

    for high-throughput rare blood cell separation. Lab Chip 11:187078

    15. Moon HS, Kwon K, Kim SI, et al. 2011. Continuous separation of breast cancer cells from blood samp

    using multi-orifice flow fractionation (MOFF) and dielectrophoresis (DEP). Lab Chip 11:111825

    16. Gascoyne PR, Noshari J, Anderson TJ, et al. 2009. Isolation of rare cells from cell mixtures by diele

    trophoresis. Electrophoresis30:138898

    17. Andreopoulou E, Yang LY, Rangel KM, et al. 2011. Comparison of assay methods for detection

    circulating tumor cells (CTCs) in metastatic breast cancer (MBC): AdnaGen AdnaTest BreastCanc

    Select/Detect versus Veridex CellSearch system. Int. J. Cancer. Epub ahead of print18. Hofman V, Ilie MI, Long E, et al. 2011. Detection of circulating tumor cells as a prognostic factor

    patients undergoing radical surgery for non-small cell lung carcinoma: comparison of the efficacy of t

    CellSearch assay and the isolation by size of epithelial tumor cell method. Int. J. Cancer129:165160

    19. Nagrath S, Sequist LV, Maheswaran S, et al. 2007. Isolation of rare circulating tumour cells in canc

    patients by microchip technology. Nature 450:123539

    20. Maheswaran S, Sequist LV, Nagrath S, et al. 2008. Detection of mutations in EGFR in circulating lun

    cancer cells. N. Engl. J. Med. 359:36677

    21. Stott SL, Lee RJ, Nagrath S, et al. 2010. Isolation and characterization of circulating tumor cells fro

    patients with localized and metastatic prostate cancer. Sci. Transl. Med. 2:25ra23

    22. Stott SL, Hsu CH, Tsukrov DI, et al. 2010. Isolation of circulating tumor cells using a microvorte

    generating herringbone-chip. Proc. Natl. Acad. Sci. USA 107:1839297

    23. Saliba AE, Saias L, Psychari E, et al. 2010. Microfluidic sorting and multimodal typing of cancer cells

    self-assembled magnetic arrays. Proc. Natl. Acad. Sci. USA 107:1452429

    24. Dharmasiri U, Njoroge SK, Witek MA, et al. 2011. High-throughput selection, enumeration, electro

    netic manipulation, and molecular profiling of low-abundance circulating tumor cells using a microflui

    system. Anal. Chem. 83:23019

    25. Talasaz AH, Powell AA, Huber DE, et al. 2009. Isolating highly enriched populations of circulati

    epithelial cells and other rare cells from blood using a magnetic sweeper device. Proc. Natl. Acad. Sci. US

    106:397075

    26. Wang S, Owens GE, Tseng HR. 2011. Nano fly paper technology for the capture of circulating tum

    cells. Methods Mol. Biol. 726:14150

    27. Pachmann K, Camara O, Kavallaris A, et al. 2008. Monitoring the response of circulating epithelial tum

    cells to adjuvant chemotherapy in breast cancer allows detection of patients at risk of early relapse. J. Cl

    Oncol. 26:120815

    28. Muller V, Alix-Panabieres C, Pantel K. 2010. Insights into minimal residual disease in cancer patienimplications for anti-cancer therapies. Eur. J. Cancer46:118997

    29. Ledford H. 2011. Cancer theory faces doubts. Nature 472:273

    30. Mani SA, Guo W, Liao MJ, et al. 2008. The epithelial-mesenchymal transition generates cells wi

    properties of stem cells. Cell133:70415

    31. Mego M, Mani SA, Cristofanilli M. 2010. Molecular mechanisms of metastasis in breast cancerclini

    applications. Nat. Rev. Clin. Oncol. 7:693701

    32. Mego M, Mani SA, Lee BN, et al. 2011. Expression of epithelial-mesenchymal transition-inducing tra

    scription factors in primary breast cancer: the effect of neoadjuvant therapy. Int. J. Cancer. Epub ahead

    print

    33. Hou JM, Krebs M, Ward T, et al. 2011. Circulating tumor cells as a window on metastasis biology

    lung cancer. Am. J. Pathol. 178:98996

    34. Gradilone A, Raimondi C, Nicolazzo C, et al. 2011. Circulating tumor cells lacking cytokeratin in brecancer: the importance of being mesenchymal. J. Cell Mol. Med. 15:106670

    35. Raimondi C, Gradilone A, Naso G, et al. 2011. Epithelial-mesenchymal transition and stemness featu

    in circulating tumor cells from breast cancer patients. Breast Cancer Res. Treat. 130:44955

    36. Konigsberg R, Obermayr E, Bises G, et al. 2011. Detection of EpCAM positive and negative circulati

    tumor cells in metastatic breast cancer patients. Acta Oncol. 50:70010

    37. Lu J, Fan T, Zhao Q, et al. 2010. Isolation of circulating epithelial and tumor progenitor cells with

    invasive phenotype from breast cancer patients. Int. J. Cancer126:66983

    212 Alix-Panabieres Schwarzenbach Pantel

  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    15/20

    38. Galanzha EI, Shashkov EV,KellyT, et al. 2009. In vivo magnetic enrichment andmultiplex photoacoustic

    detection of circulating tumour cells. Nat. Nanotechnol. 4:85560

    39. First demons

    that cytokeratins

    released by viabl

    and used as mark

    EPISPOT assay.

    39. Alix-Panabieres C, Vendrell JP, Slijper M, et al. 2009. Full-length cytokeratin-19 is released by

    human tumor cells: a potential role in metastatic progression of breast cancer. Breast Cancer Res.

    11:R39

    40. Alix-Panabieres C, Vendrell JP, Pelle O, et al. 2007. Detection and characterization of putative metastatic

    precursor cells in cancer patients. Clin. Chem. 53:53739

    41. Krivacic RT, Ladanyi A, Curry DN, et al. 2004. A rare-cell detector for cancer. Proc. Natl. Acad. Sci. USA

    101:10501442. Somlo G, LauSK, Frankel P, et al. 2011. Multiple biomarkerexpression on circulating tumor cells in com-

    parison to tumor tissues from primary and metastatic sites in patients with locally advanced/inflammatory,

    and stage IV breast cancer, using a novel detection technology. Breast Cancer Res. Treat. 128:15563

    43. Ntouroupi TG, Ashraf SQ, McGregor SB, et al. 2008. Detection of circulating tumour cells in peripheral

    blood with an automated scanning fluorescence microscope. Br. J. Cancer99:78995

    44. Deng G, Herrler M, Burgess D,et al.2008.Enrichment with anti-cytokeratin alone orcombinedwithanti-

    EpCAM antibodies significantly increases the sensitivity for circulating tumor cell detection in metastatic

    breast cancer patients. Breast Cancer Res. 10:R69

    45. Scholtens TM, Schreuder F, Ligthart ST, et al. 2011. CellTracks TDI: An image cytometer for cell

    characterization. Cytometry A. 79:20313

    46. Markou A, Strati A, Malamos N, et al. 2011. Molecular characterization of circulating tumor cells in breast

    cancer by a liquid bead array hybridization assay. Clin. Chem. 57:4213047. Agrawal B, Krantz MJ, Parker J, et al. 1998. Expression of MUC1 mucin on activated human T cells:

    implications for a role of MUC1 in normal immune regulation. Cancer Res. 58:407981

    48. Schuettpelz LG, Link DC. 2011. Niche competition and cancer metastasis to bone. J. Clin. Invest.

    121:125355

    49. Experimenta

    shows that prost

    tumor cells comp

    the same niches

    hematopoietic st

    cells in the bone

    marrow.

    49. Shiozawa Y, Pedersen EA, Havens AM, et al. 2011. Human prostate cancer metastases target

    the hematopoietic stem cell niche to establish footholds in mouse bone marrow. J. Clin. Invest.

    121:1298312

    50. Bellahcene A, Bachelier R, Detry C, et al. 2007. Transcriptome analysis reveals an osteoblast-like pheno-

    type for human osteotropic breast cancer cells. Breast Cancer Res. Treat. 101:13548

    51. Garcia T, Jackson A, Bachelier R, et al. 2008. A convenient clinically relevant model of human breast

    cancer bone metastasis. Clin. Exp. Metastasis25:3342

    52. Le Gall C, Bellahcene A, Bonnelye E, et al. 2007. A cathepsin K inhibitor reduces breast cancer induced

    osteolysis and skeletal tumor burden. Cancer Res. 67:9894902

    53. Paris PL, Kobayashi Y, Zhao Q, et al. 2009. Functional phenotyping and genotyping of circulating tumor

    cells from patients with castration resistant prostate cancer. Cancer Lett. 277:16473

    54. Attard G, Swennenhuis JF, Olmos D, et al. 2009. Characterization of ERG, AR and PTEN gene status

    in circulating tumor cells from patients with castration-resistant prostate cancer. Cancer Res. 69:291218

    55. Swennenhuis JF, Tibbe AG, Levink R, et al. 2009. Characterization of circulating tumor cells by fluores-

    cence in situ hybridization. Cytometry A 75:52027

    56. Shaffer DR, Leversha MA, Danila DC, et al. 2007. Circulating tumor cell analysis in patients with pro-

    gressive castration-resistant prostate cancer. Clin. Cancer Res. 13:202329

    57. Leversha MA, Han J, Asgari Z, et al. 2009. Fluorescence in situ hybridization analysis of circulating tumor

    cells in metastatic prostate cancer. Clin. Cancer Res. 15:20919758. Jiang Y, Palma JF, Agus DB, et al. 2010. Detection of androgen receptor mutations in circulating tumor

    cells in castration-resistant prostate cancer. Clin. Chem. 56:149295

    59. Smirnov DA,Foulk BW,Doyle GV,et al.2006. Globalgene expression profiling of circulating endothelial

    cells in patients with metastatic carcinomas. Cancer Res. 66:291822

    60. Watson MA, Ylagan LR, Trinkaus KM, et al. 2007. Isolation and molecular profiling of bone marrow

    micrometastases identifies TWIST1 as a marker of early tumor relapse in breast cancer patients. Clin.

    Cancer Res. 13:50019

    www.annualreviews.org Circulating Tumor Cells and Tumor DNA 213

  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    16/20

    61. Aktas B, Tewes M, Fehm T, et al. 2009. Stem cell and epithelial-mesenchymal transition markers a

    frequently overexpressed in circulating tumor cells of metastatic breast cancer patients. Breast Cancer R

    11:R46

    62. Theodoropoulos PA, Polioudaki H, Agelaki S, et al. 2010. Circulating tumor cells with a putative ste

    cell phenotype in peripheral blood of patients with breast cancer. Cancer Lett. 288:99106

    63. Reuben JM, Lee BN, Li C, et al. 2010. Circulating tumor cells and biomarkers: implications for perso

    alized targeted treatments for metastatic breast cancer. Breast J. 16:32730

    64. Gazzaniga P, Naso G, Gradilone A, et al. 2010. Chemosensitivity profile assay of circulating cancer ce

    prognostic and predictive value in epithelial tumors. Int. J. Cancer126:24374765. First study

    demonstrating

    convincing data for the

    prognostic relevance of

    CTCs in breast cancer

    patients.

    65. Cristofanilli M, Budd GT, Ellis MJ, et al. 2004. Circulating tumor cells, disease progression, a

    survival in metastatic breast cancer. N. Engl. J. Med. 351:78191

    66. Cohen SJ, Punt CJ, Iannotti N, et al. 2008. Relationship of circulating tumor cells to tumor respon

    progression-free survival, and overall survival in patients with metastatic colorectal cancer. J. Clin. Onc

    26:321321

    67. de Bono JS, Attard G, Adjei A, et al. 2007. Potential applications for circulating tumor cells expressi

    the insulin-like growth factor-I receptor. Clin. Cancer Res. 13:361116

    68. First study

    demonstrating that

    CTC counts are

    complementary to

    serum markers for

    measuring therapyresponse.

    68. de Bono JS, Scher HI, Montgomery RB, et al. 2008. Circulating tumor cells predict survival bene

    from treatment in metastatic castration-resistant prostate cancer. Clin. Cancer Res. 14:63029

    69. Danila DC,Heller G, Gignac GA, et al. 2007. Circulating tumor cell numberand prognosis in progress

    castration-resistant prostate cancer. Clin. Cancer Res. 13:705358

    70. Liu MC, Shields PG, Warren RD, et al. 2009. Circulating tumor cells: a useful predictor of treatme

    efficacy in metastatic breast cancer. J. Clin. Oncol. 27:515359

    71. Hayes DF, Cristofanilli M, Budd GT, et al. 2006. Circulating tumor cells at each follow-up time poi

    during therapy of metastatic breast cancer patients predict progression-free and overall survival. Cl

    Cancer Res. 12:421824

    72. Budd GT,Cristofanilli M, Ellis MJ,et al.2006. Circulating tumor cells versus imagingpredicting over

    survival in metastatic breast cancer. Clin. Cancer Res. 12:64039

    73. De Giorgi U, Valero V, Rohren E, et al. 2009. Circulating tumor cells and [18F]fluorodeoxygluco

    positron emission tomography/computed tomography for outcome prediction in metastatic breast canc

    J. Clin. Oncol. 27:330311

    74. National Cancer Institute. 2011. Treatment decision making based on blood levels of tumor ce

    in women with metastatic breast cancer receiving chemotherapy. SWOG-S0500. http://clinicaltria

    gov/ct2/show/NCT00382018

    75. Bidard FC, Mathiot C, Delaloge S, et al. 2010. Single circulating tumor cell detection and overall surviin nonmetastatic breast cancer. Ann. Oncol. 21:72933

    76. Study develops

    scoring system for

    HER2 status of CTCs

    and demonstratesstriking heterogeneity

    of CTCs and primary

    breast tumors.76. Riethdorf S, Muller V, Zhang L, et al. 2010. Detection and HER2 expression of circulating tum

    cells prospective monitoring in breast cancer patients treated in the neoadjuvant GeparQuatt

    Trial. Clin. Cancer Res. 16:263445

    77. Rack B, Schindlbeck C, Andergassen U, et al. 2010. Use of circulating tumor cells (CTC) in periphe

    blood of breast cancer patients before and after adjuvant chemotherapy to predict risk for relapse: t

    SUCCESS Trial. J. Clin. Oncol. 28:7s

    78. Study shows that

    CTCs that survive

    chemotherapy are

    indicators of poor

    prognosis in breast

    cancer.

    78. Xenidis N, Ignatiadis M, Apostolaki S, et al. 2009. Cytokeratin-19 mRNA-positive circulati

    tumor cells after adjuvant chemotherapy in patients with early breast cancer. J. Clin. Onc

    27:217784

    79. Study shows

    significant correlation

    between detection of

    CTCs and DTCs in

    bone marrow, as well as

    low proliferative activity

    of CTCs.

    79. Muller V, Stahmann N, Riethdorf S, et al. 2005. Circulating tumor cells in breast cancer: co

    relation to bone marrow micrometastases, heterogeneous response to systemic therapy and loproliferative activity. Clin. Cancer Res. 11:367885

    80. Pantel K, Schlimok G, Braun S, et al. 1993. Differential expression of proliferation-associated molecu

    in individual micrometastatic carcinoma cells. J. Natl. Cancer Inst. 85:141924

    81. Stoecklein NH, Klein CA. 2010. Genetic disparity between primary tumours, disseminated tumour ce

    and manifest metastasis. Int. J. Cancer126:58998

    82. PantelK, Alix-PanabieresC. 2010.Circulating tumour cellsin cancer patients:challenges and perspectiv

    Trends Mol. Med. 16:398406

    214 Alix-Panabieres Schwarzenbach Pantel

    http://clinicaltrials.gov/ct2/show/NCT00382018http://clinicaltrials.gov/ct2/show/NCT00382018http://clinicaltrials.gov/ct2/show/NCT00382018http://clinicaltrials.gov/ct2/show/NCT00382018
  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    17/20

    83. Klein CA. 2009. Parallel progression of primary tumours and metastases. Nat. Rev. Cancer9:30212

    84. Fehm T, Hoffmann O, Aktas B, et al. 2009. Detection and characterization of circulating tumor cells in

    blood of primary breast cancer patients by RT-PCR and comparison to status of bonemarrowdisseminated

    cells. Breast Cancer Res. 11:R59

    85. Braun S, Schlimok G, Heumos I, et al. 2001. ErbB2 overexpression on occult metastatic cells in bone

    marrow predicts poor clinical outcome of stage IIII breast cancer patients. Cancer Res. 61:189095

    86. Fehm T, Muller V, Aktas B, et al. 2010. HER2 status of circulating tumor cells in patients with metastatic

    breast cancer: a prospective, multicenter trial. Breast Cancer Res. Treat. 124:40312

    87. Koyanagi K, Mori T, ODay SJ,et al.2006.Association of circulating tumor cells with serumtumor-relatedmethylated DNA in peripheral blood of melanoma patients. Cancer Res. 66:611117

    88. Matuschek C, Bolke E, Lammering G, et al. 2010. Methylated APC and GSTP1 genes in serum DNA

    correlate with the presence of circulating blood tumor cells and are associated with a more aggressive and

    advanced breast cancer disease. Eur. J. Med. Res. 15:27786

    89. Nawroz-Danish H, EisenbergerCF, YooGH, et al.2004.Microsatelliteanalysisof serumDNA in patients

    with head and neck cancer. Int. J. Cancer111:96100

    90. For the first t

    relationship betw

    the occurrence o

    and circulating tu

    associated DNA

    blood is describe

    prostate cancer p

    90. Schwarzenbach H, Alix-Panabieres C, Muller I, et al. 2009. Cell-free tumor DNA in blood plasma

    as a marker for circulating tumor cells in prostate cancer. Clin. Cancer Res. 15:103238

    91. Shaw JA, Brown J, Coombes RC, et al. 2011. Circulating tumor cells and plasma DNA analysis in patients

    with indeterminate early or metastatic breast cancer. Biomark Med. 5:8791

    92. Van derAuwera I, Elst HJ,Van LaereSJ, etal. 2009. Thepresence of circulating totalDNA andmethylated

    genes is associated with circulating tumour cells in blood from breast cancer patients. Br. J. Cancer100:127786

    93. Wicha MS, Hayes DF. 2011. Circulating tumor cells: not all detected cells are bad and not all bad cells

    are detected. J. Clin. Oncol. 29:150811

    RELATED RESOURCES

    1. Circulating Nucleic Acids in Plasma and Serum. Proc. Int. Conf. Circulating Nucleic Acids

    in Plasma and Serum, 6th, Hong Kong, Nov. 911, 2009. http://www.springer.com/biomed/cancer/book/978-90-481-9381-3

    2. Website of the Institute of Tumor Biology, UKE, Hamburg. http://www.uke.uni-hamburg.de/institute/tumorbiologie

    3. Website of the EU sixth FP Network Group DISMAL. http://www.dismal-project.eu/

    www.annualreviews.org Circulating Tumor Cells and Tumor DNA 215

    http://www.springer.com/biomed/cancer/book/978-90-481-9381-3http://www.springer.com/biomed/cancer/book/978-90-481-9381-3http://www.uke.uni-hamburg.de/institute/tumorbiologiehttp://www.uke.uni-hamburg.de/institute/tumorbiologiehttp://www.dismal-project.eu/http://www.dismal-project.eu/http://www.uke.uni-hamburg.de/institute/tumorbiologiehttp://www.uke.uni-hamburg.de/institute/tumorbiologiehttp://www.springer.com/biomed/cancer/book/978-90-481-9381-3http://www.springer.com/biomed/cancer/book/978-90-481-9381-3
  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    18/20

    Annual Review

    Medicine

    Volume 63, 201Contents

    Huntingtons Disease: Advocacy Driving Science

    Nancy S. Wexler p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 1

    Direct-to-Consumer Genetic Testing: Perceptions, Problems,

    and Policy Responses

    Timothy Caulfield and Amy L. McGuire p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p23

    Human Genome Sequencing in Health and Disease

    Claudia Gonzaga-Jauregui, James R. Lupski, and Richard A. Gibbsp p p p p p p p p p p p p p p p p p p p p

    35The Genetic Architecture of Schizophrenia: New Mutations

    and Emerging Paradigms

    Laura Rodriguez-Murillo, Joseph A. Gogos, and Maria Karayiorgou p p p p p p p p p p p p p p p p p p p p63

    CCR5 Antagonism in HIV Infection: Current Concepts

    and Future Opportunities

    Timothy J. Wilkin and Roy M. Gulick p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p81

    New Paradigms for HIV/AIDS Vaccine Development

    Louis J. Picker, Scott G. Hansen, and Jeffrey D. Lifson p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p95

    Emerging Concepts on the Role of Innate Immunity in the Preventionand Control of HIV Infection

    Margaret E. Ackerman, Anne-Sophie Dugast, and Galit Alter p p p p p p p p p p p p p p p p p p p p p p p p p 113

    Immunogenetics of Spontaneous Control of HIV

    Mary Carrington and Bruce D. Walker p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 131

    Recent Progress in HIV-Associated Nephropathy

    Christina M. Wyatt, Kristin Meliambro, and Paul E. Klotman p p p p p p p p p p p p p p p p p p p p p p p p 147

    Screening for Prostate Cancer: Early Detection or Overdetection?

    Andrew J. Vickers, Monique J. Roobol, and Hans Lilja p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 161

    Targeting Metastatic Melanoma

    Keith T. Flaherty p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 171

    Nanoparticle Delivery of Cancer Drugs

    Andrew Z. Wang, Robert Langer, and Omid C. Farokhzad p p p p p p p p p p p p p p p p p p p p p p p p p p p p 185

    v

  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    19/20

  • 7/27/2019 Pantel EA - 2012 - Circulating Cancer Cells and DNA - V2

    20/20

    Eosinophilic Esophagitis: Rapidly Advancing Insights

    J. Pablo Abonia and Marc E. Rothenberg p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 421

    Physician Workforce Projections in an Era of Health Care Reform

    Darrell G. Kirch, Mackenzie K. Henderson, and Michael J. Dill p p p p p p p p p p p p p p p p p p p p p p p 435

    Reducing Medical Errors and Adverse Events

    Julius Cuong Pham, Monica S. Aswani, Michael Rosen, HeeWon Lee,

    Matthew Huddle, Kristina Weeks, and Peter J. Pronovostp p p p p p p p p p p p p p p p p p p p p p p p p p p p

    447

    Relationships Between Medicine and Industry: Approaches to the

    Problem of Conflicts of Interest

    Raymond Raad and Paul S. Appelbaum p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 465

    Wireless Technology in Disease Management and Medicine

    Gari D. Clifford and David Clifton p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 479

    Geographic Variation in Health Care

    Tom Rosenthal p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 493

    Deep Brain Stimulation for Intractable Psychiatric DisordersWayne K. Goodman and Ron L. Alterman p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 511

    Contemporary Management of Male Infertility

    Peter J. Stahl, Doron S. Stember, and Marc Goldstein p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 525

    Indexes

    Cumulative Index of Contributing Authors, Volumes 5963 p p p p p p p p p p p p p p p p p p p p p p p p p p p 541

    Cumulative Index of Chapter Titles, Volumes 5963 p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 545

    Errata

    An online log of corrections to Annual Review of Medicine articles may be found at

    http://med.annualreviews.org/errata.shtml