You are on page 1of 15

Therapeutic Advances in Medical Oncology Review

Ther Adv Med Oncol


Circulating tumour cells: their utility in cancer (2010) 2(6) 351—365
DOI: 10.1177/
management and predicting outcomes 1758834010378414
! The Author(s), 2010.
Reprints and permissions:
Matthew G. Krebs, Jian-Mei Hou, Tim H. Ward, Fiona H. Blackhall and Caroline Dive http://www.sagepub.co.uk/
journalsPermissions.nav

Abstract: Recent advances in technology now permit robust and reproducible detection of
circulating tumour cells (CTCs) from a simple blood test. Standardization in methodology has
been instrumental in facilitating multicentre trials with the purpose of evaluating the clinical
utility of CTCs. We review the current body of evidence supporting the prognostic value of CTC
enumeration in breast, prostate and colorectal cancer, using standardized approaches, and
studies evaluating the correlation of CTC number with radiological outcome. The exploitation of
CTCs in cancer management, however, is now extending beyond prognostication. As technol-
ogies emerge to characterize CTCs at the molecular level, biological information can be
obtained in real time, with the promise of serving as a ‘surrogate tumour biopsy’. Current
studies illuminate the potential of CTCs as pharmacodynamic and predictive biomarkers and
potentially their use in revealing drug resistance in real time. Approaches for CTC charac-
terization are summarized and the potential of CTCs in cancer patient management exempli-
fied via the detection of epidermal growth factor receptor mutations from CTCs in patients with
non-small cell lung cancer. The opportunity to learn more about the biology of metastasis
through CTC analysis is now being realized with the horizon of CTC-guided development of
novel anticancer therapies.

Keywords: biomarker, circulating tumour cells, circulating tumour microemboli, clinical


trials, personalized medicine, pharmacodynamic biomarker, predictive biomarker, prognosis

Introduction biological significance is being revealed. This Correspondence to:


Professor Caroline Dive,
Metastatic disease is responsible for over 90% of review focuses on some of the more developed PhD
cancer deaths [Wittekind and Neid, 2005; Weiss, methods for CTC detection and the current Paterson Institute for
Cancer Research,
2000]. The hypothesis that circulating tumour body of evidence supporting the clinical utility Wilmslow Road,
cells (CTCs) are a fundamental prerequisite to of detecting and characterizing CTCs. Manchester, M20 4BX, UK
cdive@picr.man.ac.uk
metastasis was first proposed in the mid 19th
Matthew G. Krebs,
Century by Thomas Ashworth, an Australian Potential therapeutic uses of circulating MBChB
pathologist [Ashworth, 1869]. Today, the identi- tumour cells Jian-Mei Hou, PhD
Tim H. Ward, PhD
fication and molecular characterization of CTCs The potential applications of CTC enumeration Fiona H. Blackhall, PhD,
in cancer patients remains key to unprecedented and characterization are far-reaching and could FRCP
Clinical and Experimental
insights into the metastatic process and is antic- facilitate several key areas of cancer therapeutics Pharmacology Group,
ipated to unmask novel therapeutic targets for the (Table 1). Most clinical studies, so far, have Paterson Institute for
Cancer Research and
treatment of cancer. Progress, however, has been focused on CTC enumeration in guiding progno- School of Cancer and
hindered until quite recently by the technical sis in metastatic cancer patients and current Enabling Sciences,
University of Manchester,
challenges posed by CTC detection, analogous research is exploring the pharmacodynamic and Manchester Cancer
to looking for the proverbial ‘needle in a hay- predictive biomarker utility of CTCs. A much- Research Centre and
Manchester Academic
stack’. However, there have been major techno- anticipated application relates to the detection Health Sciences Centre,
logical advances, in recent years, leading to of CTCs in patients with early stage, resectable Manchester, UK

the genuine prospect of comprehensive interroga- cancers with the potential to discriminate selec-
tion of CTCs. Tumour cells can now be reliably tively for patients most likely to benefit from
identified in the peripheral bloodstream of can- adjuvant treatment and indeed to monitor the
cer patients with metastatic disease and their efficacy of therapy during the adjuvant course.

http://tam.sagepub.com 351
Therapeutic Advances in Medical Oncology 2 (6)

Table 1. Potential applications of circulating tumour cell (CTC) analysis.


Enumeration of CTCs Molecular characterisation of CTCs
 Guide prognosis  Surrogate for biological activity of underlying
 Assist in measuring response to anticancer tumour — ‘real-time biopsy’
therapy — predictive and/or pharmacodynamic  Elucidate prognostic and predictive molecular
biomarker features
 May lead to more accurate prognosis when  Detection of treatment-resistant profiles —ease
added to existing staging classifications of serial sampling
 Select patients for adjuvant chemotherapy  Improve understanding of mechanisms of
 Detect recurrent disease biological processes
 Aid diagnostic process  Discover and identify new targets for therapeu-
tic manipulation

However, most methods in current use are not traditional radiological methods, make a signifi-
sensitive enough for reliable enumeration of cant difference to patient outcomes. Early data to
CTCs in early stage cancer patients. In addition, support these potential clinical utilities of CTCs
CTC number surveillance, upon completion of are discussed herein, with specific focus on prog-
therapy, may detect relapse at a much earlier nostication, correlation of CTC number with
stage than traditional clinical or radiological radiological outcomes and a survey of approaches
parameters and this is under investigation. used to characterize CTCs at the molecular level.

The molecular characterization of CTCs offers a The metastatic process and difficulties
unique ability to assess genotypic and phenotypic presented for CTC detection
features of a cancer without the need for invasive In order to appreciate the challenges of CTC
biopsy. As our understanding of tumour biology, detection, it is important to consider the con-
the signalling pathways on which tumour forma- cepts and theories relating to the metastatic pro-
tion and maintenance depends and treatment cess itself. Paget first described the ‘seed and soil’
resistance mechanisms improves, it becomes theory of tumour invasion and dissemination in
increasingly clear that mechanism-based antican- 1889 to explain the nonrandom formation of
cer therapeutics must be tailored according to metastasis [Paget, 1889]. The intrinsic properties
individuals’ tumour characteristics: the concept of the tumour cells (seeds) and host microenvi-
of personalized medicine. Traditionally, treat- ronment (soil) are vital determinants of sites of
ment decisions have been empiric based on the formation. The anatomical structure of the vas-
histology of tumour biopsies, normally taken for culature is also thought to play a (more mechan-
diagnosis, that in some cases were procured sev- ical) role in the determination of the destination
eral months or even years prior to the patient of tumour cells that enter the circulation [Fidler
treatment decisions at hand. The characteriza- and Poste, 2008; Coman et al. 1951; Ewing,
tion of CTCs, derived from a ‘simple’ blood 1928]. The entire process of metastasis appears
test, have the potential to serve as an invaluable to be inefficient in that only the ‘decathlon cham-
‘real-time tumour biopsy’ permitting an up-to- pions’ of CTCs (as coined by Isaiah Fidler
date snapshot of tumour biology, ideally without [Fidler, 2003]) are successful in establishing dis-
the need for invasive tests. Furthermore, CTCs tant metastatic disease. In support of this hypoth-
can be analysed on a serial basis, permitting the esis, preclinical models demonstrate that within
identification of emergent treatment resistance 24 hours of intravenous administration of tumour
profiles in real time. If successful, this application cells, less than 0.1% cells remain viable and that
could herald a reduction in the incidence of less than 0.01% of these surviving CTCs can pro-
unnecessary toxicity experienced by a patient duce metastasis [Fidler, 1970]. This low ‘success
taking futile treatment regimens. However, well- rate’ could be explained by a stochastic/random
designed, prospective, randomized, multicentre survival and growth of a few neoplastic cells or,
clinical trials coupled with robust CTC method- more likely, by the selection of a unique subpop-
ologies will be needed to confirm that changes in ulation of cells with greater metastatic potential.
therapy based on CTC evaluation, imposed ear- Indeed, the hypothesis that some CTCs express
lier in a treatment course than possible with ‘tumour-initiating’ properties has been purported

352 http://tam.sagepub.com
MG Krebs, J-M Hou et al.

and perhaps it is these cells that are CTC cells circulating in the bloodstream from the vast
‘decathlon champions’ capable of seeding distant majority of red and white blood cells (in 10 ml of
metastatic disease [Theodoropoulos et al. 2010; blood there are an estimated 100 million leuco-
Aktas et al. 2009]. The identification of clusters cytes and 50 billion erythrocytes) but additionally
of contiguous tumour cells in the circulation of to identify that subpopulation of CTCs with
cancer patients, termed tumour microemboli the elite, lethal metastatic potential that are ulti-
[Friedl and Gilmour, 2009; Ilina and Friedl, mately responsible for mortality.
2009; Liotta et al. 1976], has given rise to an as
yet untested hypothesis that these cells, via Methods of CTC detection
cell—cell contact survival signaling, have a sur- A battery of novel CTC detection technologies
vival advantage over single CTCs and thus has emerged over the last few years, elegantly
might be more likely to contain the minority of summarized in recent reviews [Alunni-Fabbroni
culprit tumour cells that form metastasis. and Sandri, 2010; Pantel et al. 2009; Paterlini-
Brechot and Benali, 2007; Stebbing and Jiao,
Contemporary reviews of the metastatic process 2009]. In principle, methods can be divided
include the suggestion that a reversible epithelial- into nucleic-acid-based and cytometric
to-mesenchymal transition (EMT), describing a approaches.
major phenotypic change in a subset of cells
within the primary tumour, is essential for metas- Nucleic-acid-based methods were predominantly
tasis to proceed [Thiery and Sleeman, 2006; adopted throughout the 1990s following the
Yang et al. 2006; Thiery, 2003]. During EMT, development of the polymerase chain reaction
epithelial tumour cells lose cell-to-cell to contacts (PCR) and indeed can be very sensitive tech-
and develop a more motile and invasive mesen- niques, relying on the detection of specific
chymal phenotype, facilitating their entry into the DNA or RNA sequences differentially expressed
bloodstream, and revert back to an epithelial by tumour cells [Alunni-Fabbroni and Sandri,
phenotype upon extravasation in host tissue 2010; Paterlini-Brechot and Benali, 2007].
(the so-called mesenchymal-to-epithelial transi- However, in recent years there has been a prefer-
tion (MET)) [Christiansen and Rajasekaran, ential shift toward cytometric assays where cells
2006; Thiery and Sleeman, 2006]. Many CTC remain intact, hence morphology can be visual-
detection techniques depend on capture of CTCs ized, cells can be enumerated and further analysis
based on defined epithelial protein expression by techniques such as fluorescent in situ hybridi-
(e.g. EpCam and cytokeratins); thus, the very zation (FISH) or even DNA/RNA extraction are
process of CTC detection may be inherently practically or theoretically possible, contrasting
flawed if EMT has occurred. However, it is rec- with the more limited capabilities using nucleic-
ognized that EMT is not a homogenous ‘black acid-based methods (Table 2).
and white’ cellular scenario and it seems likely
that CTCs can express both epithelial and Cytometric approaches use immunostaining pro-
mesenchymal properties, to varying degrees, files to identify and characterize CTCs. These
giving rise to heterogeneous CTC populations assays need to be highly sensitive, highly specific
[Christiansen and Rajasekaran, 2006]. A recent and highly reproducible if they are to be useful in
in vivo study postulated a model of cell coopera- the clinical setting and used to make patient
tivity where mesenchymal expressing tumour treatment decisions. Most methods employ an
cells were responsible for breakdown of the extra- initial enrichment step to optimize the probability
cellular matrix and vascular wall invasion, which of rare cell detection, achievable through immu-
facilitated entry of both mesenchymal and nomagnetic separation, centrifugation or filtra-
‘passenger’ epithelial tumour cells into the circu- tion (Table 2). Cytometric-based techniques
lation [Tsuji et al. 2009]. Only the epithelial subsequently interrogate cells by fluorescence
expressing tumour cells had the capabilities to microscopy or immunohistochemistry.
subsequently form distant metastases in this
TM
model. The relationships between EMT and CellSearch system
collective cell migration are not yet clear. The most widely used cytometric CTC tech-
nology currently in clinical testing is the
TM
With this framework of metastatic tumour cell CellSearch platform (Veridex LLC,
behaviour in mind, the challenge for CTC Huntingdon Valley, PA, USA) and is the only
research is thus not only to discriminate tumour technology to have received FDA approval for

http://tam.sagepub.com 353
Therapeutic Advances in Medical Oncology 2 (6)

Table 2. Main advantages and disadvantages of techniques used for circulating tumour cell (CTC) enrichment and analysis
[Alunni-Fabbroni and Sandri, 2010; Paterlini-Brechot and Benali, 2007].
Advantages Disadvantages
Enrichment
Immunomagnetic Cells selected on basis of antigen expression Dependent on epithelial marker expression
separation (manual) using antibody coated magnetic beads. which tumour cells may or may not express;
Versatility for positive or negative cells may be lost in sample preparation
cell selection.
Immunomagnetic Semi-automated magnetic separation — easy Dependent on epithelial marker (EpCam)
separation to use. Blood samples stable for up to expression. Challenging to extract pure CTC
(CellSearch) 96 hours. Automation permits reproducible for further characterisation but enrichment
results between users, laboratories and good compared with other techniques.
samples thus robust for multisite trials. Expensive.
Centrifugation Simple method. Isolates mononuclear cells Poor enrichment for CTCs - high contamination
on the basis of differences in density gradient. with WBCs. Large number CTCs potentially
Nonexpensive. lost in processing
Filtration (ISET) Isolates cells on basis of size differences (CTC Small tumour cells may not be detected by this
large; WBC small). Effective method. Tumour method. Lower sensitivity compared with
cells amenable for molecular characterization. other techniques.
Analysis
Cytometric Cells directly visualized by IHC or IF - permits Detection depends on epithelial or tumour
evaluation of morphology and CTC marker expression. No pure CTC marker
enumeration. Cells can be extracted for exists. False-positive cells may be enriched.
molecular characterization, e.g. FISH, RT-PCR. Current techniques probably less sensitive
In combination this allows greater specificity than nucleic acid methods.
than nucleic acid methods.
Nucleic Acid Based Highly sensitive in particular with multimarker Cells cannot be visualized directly for
(RT-PCR) assay. Can be performed without enrichment morphology or enumeration. Clinical
step but at expense of sensitivity. Quantitative correlations reported but difficult to
RT-PCR permits relative quantification. standardize techniques across laboratories.
No transcripts purely specific for tumour cells
thus high false-positive rate.

FISH, fluorescent in situ hybridization; IF, immunofluorescence; IHC, immunohistochemistry; ISET, isolation by size of epithelial tumour cells;
RT-PCR, reverse transcription polymerase chain reaction; WBC, white blood cell.

the enumeration of CTC in whole blood in spe- patients. Unfortunately, many of these studies
cific cohorts of cancer patients [Miller et al. accrued small numbers of patients with insuffi-
2010]. The major advantage of this system is cient statistical power for translation to routine
its semi-automation and proven reproducibility, clinical practice. The inclusion within a single
reliability, sensitivity, linearity and accuracy study of patients with different disease stage
[Riethdorf et al. 2007; Allard et al. 2004]. and treatments also hindered the ability to draw
These are features crucial to any biomarker tech- robust conclusions. Other variables between
nology to ensure validity of results in clinical test- reported studies included: the timing of blood
ing across multiple sites and have so far been samples in relation to treatment interventions;
lacking with previous techniques. CellSearch whether or not the first few millilitres of blood
employs immunomagnetic bead-based separation drawn should be discarded on the basis that
to enrich for CTCs, and the platform character- this will avoid contamination with skin epithelial
istics have been described in detail previously cells; and, importantly, few studies reported con-
[Miller et al. 2010; Allard et al. 2004]. ditions for sample handling, sample transport
and the conditions and duration of sample stor-
Application of CTC technologies to age prior to analysis. All of these factors, if not
clinical testing standardized, are likely sources of variability.
Prior to the introduction of the CellSearch plat- Other biological factors have yet to be examined
form, a variety of combinations of enrichment in sufficient depth, such as the relationship
techniques and detection assays had been applied between the rate of cancer cell entry into the cir-
in an attempt to draw clinically relevant conclu- culation and circadian rhythms although studies
sions regarding the prevalence of CTCs in cancer in 8 prostate and 51 breast cancer patients

354 http://tam.sagepub.com
MG Krebs, J-M Hou et al.

suggested that multiple sampling over a 24-hour reproducible between duplicate measurements
period, within a given individual, did not fluctu- and between measurements performed in differ-
ate [Martin et al. 2009; Moreno et al. 2001]. ent laboratories, paving the way for further
Once the patients’ blood samples are presented studies aimed at determining the prognostic,
for evaluation, the choice of antigen for CTC pharmacodynamic and predictive value of CTC
capture in cytometric assays is critical and even enumeration in patients with advanced cancers.
if the same capture antigen is exploited across Three seminal studies in breast, prostate and
different studies and sites, if different antibodies colorectal cancer [Cohen et al. 2008; de Bono
are used they may have different sensitivity and et al. 2008; Cristofanilli et al. 2004] have subse-
specificities. Similarly, different reverse transcrip- quently shown that CTC#, over a specified cut-off
tion (RT)-PCR primers for target sequences in value, predicts for worse prognosis, and a change
nucleic-acid-based approaches, coupled with in CTC# following initiation of therapy demon-
subtle differences in assays, might amplify strates predictive value for survival outcome
cDNA with different efficiency with wide ranging (Tables 3 and 4). These studies have led to FDA
variation in PCR product. Criteria for the defini- approval of the CellSearch system as an aid to
tion of CTCs have also varied. The requirements prognosis and aid to monitoring patients on ther-
for exploratory CTC research and the application apy in these three clinical situations. Several smal-
of CTCs as biomarkers in clinical trials have dif- ler studies have supported these findings and our
ferent requirements, where standardization is own group has begun to investigate the role of
essential for the latter application. CTCs in the management of small cell lung
cancer (SCLC) and non-small cell lung cancer
CellSearch technology has addressed many of (NSCLC) [Krebs et al. 2010; Hou et al. 2009].
these variables with its semi-automation, stan-
dardized kits and user-proficiency assessments CTC number as a prognostic biomarker
and is consequently the predominate technology
for clinical testing. Its robust validation permits Breast cancer
for reliable comparisons of studies and, as such, The first prospective, multicentre prognostic
will remain a focus for this review. Many of the study using CellSearch was performed in patients
next-generation CTC technologies hold great with breast cancer. Out of 177 metastatic breast
promise for improving sensitivity and specificity cancer patients starting a new line of therapy, a
for CTC detection but validation studies are out- high CTC# at baseline defined as 5 CTCs
standing prior to their integration into large-scale compared with patients with <5 CTCs, was an
clinical studies. In this regard, the ‘CTC-chip’ is independent predictor for poor outcome in terms
a very promising approach based on microfluidics of progression-free survival (PFS; 2.7 versus 7.0
and immunomagnetic cell capture and its capa- months p < 0.0001) and overall survival (OS;
bility in lung cancer will be discussed [Nagrath 10.9 versus 21.9 months p < 0.0001) [Hayes
et al. 2007]. et al. 2006; Cristofanilli et al. 2005, 2004].
Assessment of CTC levels at any subsequent
Clinical studies using CellSearch technology time point (3 weeks onwards) reproducibly pre-
The first reported study using CellSearch dicted clinical outcome with the worst PFS and
assessed CTC numbers in 145 healthy women OS occurring in those patients with CTC counts
volunteers, 199 women with nonmalignant dis- 5 at any time point tested. Further analysis
ease and 964 patients with a range of metastatic determined that a dynamic change in CTC#
epithelial cancer types [Allard et al. 2004]. Of all from baseline, up to 15—20 weeks after initiation
healthy women donors and women with nonma- of therapy, predicted more discriminately for sur-
lignant disease, only one sample contained 2 vival outcome. For example, patients with persis-
CTCs (CTC number (#) is expressed per stan- tently elevated CTC counts (5 at baseline and
dard 7.5 ml blood). In contrast, out of 2183 sam- follow up) exhibited significantly worse OS com-
ples taken from 964 metastatic carcinoma pared with patients with persistently low CTC
patients CTC# ranged from 0 to 23,618 and counts (<5 at baseline and follow up).
36% of specimens had 2 CTCs. The presence Furthermore, patients who converted from an
varied widely in samples from different carcino- elevated CTC count (5) at baseline to a low
mas with frequency being highest in metastatic CTC count (<5) at follow up survived signifi-
prostate, unknown primary, ovary and breast car- cantly longer than those patients who converted
cinomas [Allard et al. 2004]. Results were highly from a low baseline CTC count to elevated CTC

http://tam.sagepub.com 355
356
Table 3. Summary of selected prognostic studies published to date using CellSearch technology.
Study and cancer Breast [Hayes Breast Castrate Castrate Castrate Castrate Colorectal Colorectal Small cell
type et al. 2006; [Nole et al. resistant resistant resistant resistant [Cohen [Tol et al. lung cancer
Cristofanilli 2008] prostate prostate prostate prostate et al. 2009, 2010] [Hou et al.
et al. 2005, cancer [de cancer cancer cancer 2008] 2009]
2004] Bono et al. [Scher et al. [Olmos [Danila
2008] 2009] et al. 2009] et al. 2007]
No of patients 177 80 219 156 119 112 413 451 50
% Positive for 49% (5) 61% (5) 58% (5) 55% (5) 50% (5) 62% (5) 26% (3) 29% (3) 86% (1)
CTCs at BL
(above cut-off)
Line of therapy Any 1st, 2nd or Any 1st Any Any 1st, 2nd or 1st 1st
Therapeutic Advances in Medical Oncology 2 (6)

3rd 3rd
CTC prognostic 5 5 5 no cut-off 5 no cut-off 3 3 no cut-off
cut-off

Median OS — — — — — — — — —
according to BL
CTC count:
<cut-off 18 m — 21.7 m — >30 m — 20.6 m 22.0 m —
cut-off 10.6 m — 11.5 m — 19.5 m — 9.4 m 13.7 m —
P value <0.001 — <0.0001 — <0.0001 — <0.0001 <0.0001 —

Median OS — — — — — — — — —
according to BL
and FU CTC
counts:
<cut-off at BL 22.6 m — >26 m — >30 m — 17.7 m 21.9 m —
<cut-off at FU
 cut-off at BL 19.8 m — 21.3 m — 21.4 m — 11.0 m 14.5 m —
<cut-off at FU
<cut-off at 10.6 m — 9.3 m — 11.2 m — 10.9 m 6.3 m —
BLcut-off at FU
cut-off at BL 4.1 m — 6.8 m — 9.2 m — 3.7 m 6.3 m —
cut-off at FU
Timing of FU Up to 15-20 — Up to 13-20 — After 1-2 — 3—5 weeks 1—2 weeks —
blood sampling: weeks weeks cycles of
novel agent
in phase 1
setting

(continued)

http://tam.sagepub.com
Table 3. Continued.
Study and cancer Breast [Hayes Breast Castrate Castrate Castrate Castrate Colorectal Colorectal Small cell
type et al. 2006; [Nole et al. resistant resistant resistant resistant [Cohen [Tol et al. lung cancer
Cristofanilli 2008] prostate prostate prostate prostate et al. 2009, 2010] [Hou et al.
et al. 2005, cancer [de cancer cancer cancer 2008] 2009]

http://tam.sagepub.com
2004] Bono et al. [Scher et al. [Olmos [Danila
2008] 2009] et al. 2009] et al. 2007]
Additional CTC values Patients CTC counts Higher Patients Higher CTC values CTC values Higher
comments assessed for with 5 predicted baseline with >50 baseline assessed assessed baseline
correlation CTC at OS better CTCs (as a CTC at CTCs (as a for correla- for correla- CTCs (as a
with radiologi- baseline or than PSA continuous baseline continuous tion with tion with continuous
cal response follow up decrement variable) had poorer variable) radiological radiological variable)
(see Table 4) compared algorithms predicted OS com- predicted response response predicted
to <5 CTC at all time for worse pared with for worse (see Table (see Table for worse
predicted points OS (HR patients OS. 4) 4) OS (HR 1.1,
worse PFS; 1.58, with 5—50 Predictive p ¼ 0.015)
S not p < 0.0001) or <5 CTCs model
reached (6.3 vs 21.1 improved
vs 30 with inclu-
months, sion of PSA
p < 0.001). and albu-
min levels

CTC, circulating tumour cell; HR, hazard ratio; m, months; OS, overall survival; PFS, progression free survival; FU, follow up; BL, baseline.
MG Krebs, J-M Hou et al.

357
358
Table 4. Summary of selected CellSearch-based studies comparing circulating tumour cell numbers with radiological outcomes and overall survival.

Study and cancer Type Breast [Budd et al. Breast CRC [Cohen et al. CRC [Tol et al. 2010] Breast [Nakamura Breast [Liu et al.
2006] [De Giorgi et al. 2008] et al. 2009] 2009]
2009]

Number of patients 138 102 364 307 107 68


Imaging technique CT PET-CT CT CT CT CT
Timing of imaging from 10 weeks 9—12 weeks 6—12 weeks 9 weeks 12 weeks 9—12 weeks
baseline
Timing of CTC from baseline 4 weeks 9—12 weeks Within 1 month 1—2 weeks 3—4 weeks 3—12 weeks
imaging/death
CTC cut off 5 5 3 3 5 5
Concordance (% patients with 76% 75% 78% 90% Not reported 70%
<cut off CTC and NPD by
imaging and patients
with  cut off CTC and PD
by imaging)
Median OS according to
Imaging and CTC
parameters:
1. NPD by Imaging and CTC 26.9 m Not reached 18.8 m 21.6 m Not reported Not reported
<cut off
Therapeutic Advances in Medical Oncology 2 (6)

2. PD by Imaging and CTC 19.9 m 14.7 m 8.3 m 7.1 m


<cut off
3. NPD by Imaging and CTC 15.3 m 4.9 m 7.1 m 9.4 m
cut off
4. PD by Imaging and CTC 6.4 m 9.8 m 3.1 m 3.4 m
cut off
Statistical significance
between groups:
1v2 0.0785 0.0018 <0.0001 <0.0001 Not reported Not reported
1v3 0.0389 <0.0001 <0.0001 <0.0001
1v4 <0.0001 <0.0001 <0.0001 <0.0001
2v3 0.7549 0.1073 0.4662 Not reported
2v4 0.0039 0.2244 0.0001 Not reported
3v4 0.0777 0.6441 0.0330 Not reported
Additional comments CTCs strongest CTCs strongest CTCs strong predic- CTCs at baseline In cases where If 5 CTCs detected
predictor for OS predictor for OS tor for OS in and at 1—2 CTCs increased at time of imag-
in multivariate in multivariate multivariate weeks, strongest at 3—4 weeks, ing, 3—5 weeks
analysis. analysis. In this analysis. predictor for PFS 63.6% patients prior to imaging
However, both series imaging However, both and OS in multi- showed PD at or 7—9 weeks
CTC values and helpful in deter- CTC values and variate analysis. time of CT scan. prior to imaging,
imaging were mining prognosis imaging were However, both In cases where Hazard Ratio for
useful in combi- in patients with useful in combi- CTC values and CTCs decreased PD at scan ¼ 6.3,
nation in predict- <5 CTCs. In nation in predict- imaging were by >90% at 3—4 3.1, 4.9 respec-
ing survival patients with  5 ing survival useful in combi- weeks, 85.7% tively (p < 0.001).
outcome. CTCs, difference outcome nation in predict- patients showed Study included
in survival was ing survival complete or par- patients on hor-
not significant outcome tial response at monal therapy as
between those imaging well as
with PD or NPD chemotherapy
by imaging

FU, follow up; OS, overall survival; PD, progressive disease; NPD, nonprogressive disease (defined as combination of complete response, partial response and stable disease); CTC,
circulating tumour cell; CRC, colorectal cancer.

http://tam.sagepub.com
MG Krebs, J-M Hou et al.

count at follow up (Tables 3 and 4). These were third-line therapy, patients with baseline CTC#
the first data to support CTCs as a potentially 3 had significantly worse median PFS (4.4
useful marker for determining response to versus 7.8 months, p ¼ 0.004) and OS (9.4
therapy. versus 20.6 months, p < 0.0001) compared with
patients with <3 CTCs [Cohen et al. 2009,
Metastatic castrate-resistant prostate cancer 2008]. CTC# at any subsequent blood draw,
Studies in metastatic castrate-resistant prostate up to 13—20 weeks after initiation of treatment,
cancer (mCRPC) have shown CTC# determined was consistently able to predict for worse PFS
by CellSearch to be prognostic. In 219 patients and OS on the basis of 3 CTCs. A dynamic
with mCRPC commencing a new line of chemo- change in CTC number from baseline to follow
therapy, baseline CTC# 5 predicted for signif- up predicted more discriminately for survival
icantly worse OS compared with patients with <5 outcome as seen for patients with breast and
CTCs (median OS 11.5 months versus 21.7 prostate cancer, e.g. patients with persistently
months respectively, p < 0.0001) [de Bono et al. elevated CTC counts (3 at baseline and
2008]. Blood samples were taken at monthly follow up) exhibited significantly worse OS com-
intervals following commencement of chemo- pared with patients with persistently low CTC
therapy and CTCs 5 at any subsequent time counts (<3 at baseline and follow up) (Table 3).
point consistently predicted for worse OS, in a
pattern similar to that seen in the breast cancer A second study in metastatic CRC assessed the
study (Table 3). prognostic value of CTCs in patients starting
first-line treatment with chemotherapy and tar-
Not only were CTCs a strong predictor for OS in geted agents [Tol et al. 2010]. The same patterns
multivariate analysis in this study but their pre- of results were identified (Table 3).
dictive value for survival was superior to that of
monitoring a reduction in prostate-specific anti- Lung cancer
gen (PSA) of >30% or >50%, a routinely tested Our group has shown that CTCs are detectable
tumour maker with potential predictive value for in much higher numbers in patients with SCLC
survival in mCRPC [de Bono et al. 2008]. compared with other disease types. In a cohort of
50 patients, the median number of CTCs
The use of a low cut-off value (5 CTCs) for detected was 28, ranging from 0 to 44,896 per
separating patients into ‘favourable’ and ‘unfa- 7.5 ml blood. Higher numbers of CTCs were
vourable’ prognostic groups has been questioned associated with worse survival in univariate anal-
by statistical considerations that challenge the ysis with >300 CTCs associated with signifi-
ability of CTC assays to accurately enumerate cantly worse survival compared with patients
small numbers of cells, with a risk of stratifying with <2 CTCs (4.5 months versus 14.8 months
patients into the wrong prognostic group [Allan respectively, p < 0.005) [Hou et al. 2009]. A per-
and Keeney, 2010; Tibbe et al. 2007]. An alter- sistently elevated CTC count after one cycle of
native approach is to consider CTC# as a con- chemotherapy was a stronger predictor for OS.
tinuous variable where one would hypothesize Studies are ongoing regarding the prognostic sig-
the higher the number of CTCs, the worse the nificance of CTC number in both SCLC and
prognosis. To this end, data from the above- NSCLC where in the latter the range of CTC#
detailed prostate cancer study were reanalysed is smaller than the former.
considering baseline and post-treatment CTC
values as continuous variables [Scher et al. Summary of prognostic studies
2009]. Results confirmed that the higher the Common to all of these studies is that CTC#
CTC#, the worse the OS for patients, irrespec- evaluated by CellSearch, prior to commencing a
tive of cut off (Table 3). Two smaller prostate new line of chemotherapy, has prognostic value
cancer studies exhibited similar results. [Olmos in breast, prostate and colorectal cancer, leading
et al. 2009; Danila et al. 2007] (Table 3). to FDA approval in these disease types.
Furthermore, changes in CTC number during
Colorectal cancer treatment predict survival outcome, highlighting
Studies in colorectal cancer (CRC) have also the potential to use CTCs as a surrogate end-
shown CTC# determined by CellSearch to be point biomarker. In the clinical setting this may
prognostic. In a study of 413 patients with influence changes in treatment strategy, within
metastatic CRC commencing first-, second- or just a few weeks of starting therapy, in cases

http://tam.sagepub.com 359
Therapeutic Advances in Medical Oncology 2 (6)

where CTCs remain above prognostic cut off or survival. For example, in a study of breast cancer,
demonstrate an increasing trend. Indeed, at least both CTC values at 4 weeks and imaging results at
one randomized study is currently underway to 10 weeks were independently predictive for sur-
test this hypothesis. The SWOG 0500 study ran- vival [Budd et al. 2006]. In combination, however,
domizes women with metastatic breast cancer to greater discrimination for survival was identified,
either continuation of the same chemotherapy or e.g. patients who exhibited PD by CT scan at 10
change in therapy on the basis of a persistently weeks, could be categorized into those with a very
elevated CTC count at 3 weeks (5 CTCs), in poor prognosis (5 CTCs; median OS 6.4
those with an initially elevated count (5 CTCs) months) and those with a better prognosis (<5
at baseline. Target accrual is 500 patients and CTCs; median OS 19.9 months, p ¼ 0.0039).
primary outcome measures are PFS and OS. Further details are summarized in Table 4.
Results are anticipated early 2012.
Studies in CRC showed similar results. In multi-
CTCs as a surrogate response biomarker variate analysis, CTCs were the most significant
Hand-in-hand with changes in CTC number factor for determining prognosis but the combi-
predicting survival outcome is the question of nation of CTCs and imaging provided additive
whether changes in CTC number correlate with data (Table 4) [Tol et al. 2010; Cohen et al.
traditional radiological outcomes, the ‘gold stan- 2008]. Such studies have not been reported in
dard’ for monitoring disease status. Several stud- prostate cancer as it is notoriously difficult to
ies have investigated this correlation, some of assess tumour response by standard imaging in
which were reported in parallel to the aforemen- this disease.
tioned prognostic studies (Table 4).
For future clinical use, the biological significance
The approach has been to take CTC samples at a of CTC# in predicting patient survival soon after
given time point after commencing a new line of commencing chemotherapy is encouraging and
therapy and to compare CTC values with may, in certain situations, be more informative
response to treatment according to standard-of- than response by imaging, e.g. with novel tar-
care imaging techniques. Studies correlated CTC geted therapies which are often cytostatic in
number (above and below a specified cut off) their effect. There is also a body of evidence to
with response to imaging according to Response support response by standard CT imaging as an
Evaluation Criteria in Solid Tumours (RECIST) unreliable surrogate for survival [Mandrekar et al.
and OS was calculated according to both param- 2010; Desar et al. 2009]. On the basis of data
eters independently or as combined modalities reported so far, a particularly pertinent clinical
(Table 4). setting to which CTCs could be applied is that
of phase 1 trials. CTC# may be used to predict
Although studies were slightly heterogeneous in the activity of a drug early in its development,
their methodological approaches with regard to potentially facilitating go/no-go decision-making
timing of CTC samples and imaging after com- and thus expediting the drug development pro-
mencing treatment, the results favoured CTCs cess and reducing expenditure by avoiding the
over conventional imaging to be the most signifi- all-too-frequently seen ‘drug fails late and expen-
cant factor in determining prognosis in multivari- sive’ scenario. The ability to ascertain drug—tar-
ate analysis. Concordance between CTCs and get expression and downstream measures of
imaging was reported in the range 70—90% drug—target hitting in CTCs are other exciting
although this should be interpreted with caution prospects beginning to be realized (as discussed
as patients with stable disease were grouped in the following).
together with patients with partial response (PR)
and complete response (CR), collectively termed CTCs as a real-time biopsy
nonprogressive disease (NPD). This means that The characterization of CTCs at the molecular
in cases where CTC# was reduced but stable dis- level will facilitate personalized medicine via
ease was seen by imaging, a positive correlation was analysis of pharmacodynamic and/or predictive
concluded, as opposed to drawing positive correla- biomarkers, and biomarkers of drug sensitiv-
tion only when reduction in CTC# was seen with a ity or inherent/acquired resistance. This is
true radiological response (i.e. PR or CR). already being realized, exemplified by studies of
Analysing CTCs and imaging, however, as com- human epidermal growth factor receptor-2
bined modalities enhanced the ability to predict (HER2) status in patients with breast cancer.

360 http://tam.sagepub.com
MG Krebs, J-M Hou et al.

Expression of HER2 has been detected in CTCs pancreatic (n ¼ 15), breast (n ¼ 10) and colon
in metastatic breast cancer patients in cases (n ¼ 10) [Nagrath et al. 2007].
where primary tumours were negative at original
diagnosis [Pestrin et al. 2009; Fehm et al. 2007; Using this technology, DNA was extracted from
Hayes et al. 2002]. This suggests change in bio- CTCs isolated from patients with NSCLC and
logical activity over time and may justify treat- EGFR-mutation analysis performed using the
ment with HER2-receptor antagonists, such as scorpion amplification refractory mutation
trastuzumab, in patients who previously would system (SARMS) [Maheswaran et al. 2008].
not have been eligible. Indeed one study CTCs were detected in 27/27 patients with a
showed 9 of 24 breast cancer patients whose pri- median number of 74 cells/ml. Twenty patients
mary tumour was HER2-negative each acquired with known EGFR-mutant primary tumours had
HER2 gene amplification in their CTCs during CTCs available for molecular analysis, from
cancer progression [Meng et al. 2004]. Four of whom EGFR mutations were detected in CTCs
the nine patients were treated with trastuzumab- from 19 patients (95%). In addition to the pri-
containing therapy, three of whom showed mary activating mutation, the resistance muta-
evidence of complete or partial response. tion T790M, predictive for EGFR tyrosine
kinase inhibitor resistance [Kobayashi et al.
An exemplary model for personalized medicine 2005], was detected in 11/20 patients. Serial
lies in the detection of epidermal growth factor molecular analysis of CTCs in four patients dem-
receptor (EGFR) mutations in patients with onstrated evolving genotypes with the emergence
NSCLC, predicting for response to EGFR tyro- of T790M drug-resistance mutation in all four
sine kinase inhibitors such as gefitinib and erloti- patients, consistent with development of clinical
nib [Sequist et al. 2007]. In a large phase 3 study drug resistance.
of East Asian patients with NSCLC, randomized
to gefitinib (250 mg/day) or carboplatin (area The potential to use this test in the clinical setting
under the curve 5 or 6 mg/ml/minute) with pac- is extremely promising for predicting both
litaxel (200 mg/m2) (CP), patients with EGFR response to therapy and emergence of resistance
mutations treated with gefitinib exhibited signif- genotypes. It exemplifies perfectly the crucial role
icantly longer PFS compared with patients trea- CTCs may play in tandem with development of
ted with CP (hazard ratio [HR] 0.48, 95% novel molecular agents and routine clinical
confidence interval [CI], 0.36—0.64, p < 0.001). management.
Conversely, patients negative for the EGFR
CTC characterization after enrichment using the
mutation exhibited significantly longer PFS if
CellSearch platform
they received CP chemotherapy versus gefitinib
Several other methods exist to characterize CTCs
(HR with gefitinib 2.85, 95% CI 2.05—3.98,
with the common goal of facilitating personalized
p < 0.001) [Mok et al. 2009]. Mutation analyses
medicine and to learn more about the biology of
were performed using tumour specimens; how-
metastasis. Our own group has characterized the
ever, EGFR-mutation analysis is now possible
malignant origin of CTCs isolated by CellSearch
in CTCs.
in patients with SCLC. The CellSearch system
has a channel, not required for CTC identifica-
CTC chip technology and EGFR tion, that can be used to detect an appropriately
mutation analysis fluorochrome labelled antibody for further CTC
Pre-eminent studies using a novel CTC technol- characterization. This was exploited in our stud-
ogy have demonstrated that EGFR mutations ies to examine a neuroendocrine marker, CD56
can be detected from CTCs [Sequist et al. expression in SCLC CTCs. The heterogeneity in
2009; Maheswaran et al. 2008; Nagrath et al. CTC expressions of CD56 broadly mirrored that
2007]. The ‘CTC chip’ isolates CTCs by passing found in matched tumour biopsy [Hou et al.
whole blood through a microchip containing 2009].
78,000 geometrically arranged microposts
coated in EpCam. Sensitivity and specificity is In a series of phase I studies, the CellSearch spare
reported as 99.1% and 100%, respectively, channel was used for the detection of insulin-
and CTCs were detected in 115/116 samples like growth factor-1 receptor (IGF-1R) in
taken from 68 patients with a range of tumours patients receiving a monoclonal antibody against
including NSCLC (n ¼ 55), prostate (n ¼ 26), IGF-1R either alone or in combination with

http://tam.sagepub.com 361
Therapeutic Advances in Medical Oncology 2 (6)

chemotherapy [de Bono et al. 2007]. Out of 26 learned of their biology. The genetic and epige-
patients with detectable CTCs, 23 had evidence netic determinants of tumour cell invasion, cell
of IGF-1R-positive CTCs. In patients with pros- survival within the circulation and ability to col-
tate cancer receiving anti-IGF-1R treatment and onize distant organs is an area of intense research
docetaxel, there was some evidence to support a [Nguyen et al. 2009]. CTC characterization of
greater and more rapid decline in PSA in those single cells, once achievable, will offer unprece-
patients with positive IGF-1R CTCs than dented opportunity to explore this further.
in those patients with no CTCs, suggesting a
potential predictive marker.
Conclusion
Revealing the genotype/phenotype of CTCs will Since their first description in 1869, a multitude
further our understanding of the heterogeneous of studies have provided evidence for CTCs in
populations of CTCs that are beginning to be the blood of cancer patients. Until recently
observed and potentially reveal the CTC ‘decath- there has been substantial variability in CTC
lon champions’ with the highest malignant poten- data using several different methods of detection
tial. To the best of the authors’ knowledge, only and analysis. CellSearch technology provides
one study, so far, has reported global gene robust, reproducible CTC enumeration and by
expression profiling from CTCs isolated by unifying methodology across the scientific com-
CellSearch [Smirnov et al. 2005]. This was per- munity meaningful interpretations are being
formed using samples from three patients, one drawn. The limitations of this approach, how-
with breast, one with prostate and one with colo- ever, are founded on the bias towards high
rectal cancer, each having >100 CTCs/7.5 ml enough levels of EpCam expression on CTCs,
sample. Differences in genetic profiles were and evidence is emerging to suggest that CTCs
seen between each of the patients and candidate are heterogeneous in this regard. Nevertheless,
tumour-specific genes were selected and prospec-
the prognostic utility of CTCs, enumerated
tively validated by qPCR in a cohort of 74 met-
using CellSearch, has been demonstrated for
astatic cancer patients and 50 healthy donors.
breast, prostate and colorectal cancer and our
The CTC gene signatures were successfully
own studies have shown prognostic significance
able to discriminate cancer patients from healthy
in SCLC. The potential for serial CTC enumer-
donors, and discriminate between the three dif-
ation to predict response to therapy has also been
ferent cancer types. This is promising initial data
demonstrated in breast, prostate and colorectal
but methods will need to advance further to reli-
cancer and a prospective evaluation of changing
ably isolate RNA from smaller numbers of CTCs
therapy based on CTC enumeration alone, early
and to improve purification from contaminating
white blood cells which overwhelm and dilute the in the treatment course, is underway in breast
tumour gene signatures. The move toward single cancer.
cell gene expression profiling from purified
CTCs is greatly anticipated. Whilst data are emerging on several cancer types
(breast, prostate, CRC and lung), there remains
FISH has also been performed in CTCs follow- little information regarding CTCs in many others
ing enrichment by CellSearch to confirm malig- (e.g. melanoma, pancreatic cancer, head and
nant origin or identify predictive aberrations. neck cancer, biliary cancer) and much more
Three studies in prostate cancer have assessed research across the cancer spectrum is required.
for FISH abnormalities in CTCs including The promise of CTC research in early stages of
amplification of androgen receptor (AR) and cancer is largely unmet, requiring more sensitive
MYC [Leversha et al. 2009]; gene copy number technologies. However, the ‘real-time biopsy’
for chromosomes 1, 7, 8 and 17 [Swennenhuis potential based on CTC molecular characteriza-
et al. 2009]; and fusions of TMPRSS2-ERG tion is intuitive and promising and remains a key
together with AR and phosphatase and tensin area for further intensive research. The envi-
homologue (PTEN) amplifications [Attard et al. sioned future is of a model synonymous to anti-
2009]. All have shown genetic aberrations of microbial therapy whereby a simple blood test
CTCs confirming their malignant origin. will permit molecular tumour characterization,
identification of treatment targets and aid in
Progress has been made in terms of ability to selection of the most appropriate targeted ther-
characterize CTCs but there is much still to be apy from an armamentarium of targeted agents.

362 http://tam.sagepub.com
MG Krebs, J-M Hou et al.

Funding Coman, D.R., de, L.R. and Mcc, U.M. (1951) Studies
Supported by Cancer Research UK grant C147/ on the mechanisms of metastasis; the distribution of
tumors in various organs in relation to the distribution
A12328. M.K. was supported by a Cancer of arterial emboli. Cancer Res 11: 648—651.
Research UK/AstraZeneca Clinical Pharmacology
Fellowship. Cristofanilli, M., Budd, G.T., Ellis, M.J., Stopeck, A.,
Matera, J., Miller, M.C. et al. (2004) Circulating
tumor cells, disease progression, and survival in met-
Conflict of interest statement astatic breast cancer. N Engl J Med 351: 781—791.
The authors declare no conflict of interest in the
Cristofanilli, M., Hayes, D.F., Budd, G.T., Ellis, M.J.,
preparation of this manuscript. Stopeck, A., Reuben, J.M. et al. (2005) Circulating
tumor cells: a novel prognostic factor for newly diag-
References nosed metastatic breast cancer. J Clin Oncol
Aktas, B., Tewes, M., Fehm, T., Hauch, S., 23: 1420—1430.
Kimmig, R. and Kasimir-Bauer, S. (2009) Stem cell
Danila, D.C., Heller, G., Gignac, G.A., Gonzalez-
and epithelial-mesenchymal transition markers are
Espinoza, R., Anand, A., Tanaka, E. et al. (2007)
frequently overexpressed in circulating tumor cells of
Circulating tumor cell number and prognosis in pro-
metastatic breast cancer patients. Breast Cancer Res
gressive castration-resistant prostate cancer. Clin
11: R46.
Cancer Res 13: 7053—7058.
Allan, A.L. and Keeney, M. (2010) Circulating tumor
cell analysis: technical and statistical considerations for de Bono, J.S., Attard, G., Adjei, A., Pollak, M.N.,
application to the clinic. J Oncol 2010: 426218. Fong, P.C., Haluska, P. et al. (2007) Potential appli-
cations for circulating tumor cells expressing the
Allard, W.J., Matera, J., Miller, M.C., Repollet, M., insulin-like growth factor-I receptor. Clin Cancer Res
Connelly, M.C., Rao, C. et al. (2004) Tumor cells 13: 3611—3616.
circulate in the peripheral blood of all major carcino-
mas but not in healthy subjects or patients with non- de Bono, J.S., Scher, H.I., Montgomery, R.B., Parker,
malignant diseases. Clin Cancer Res 10: 6897—6904. C., Miller, M.C., Tissing, H. et al. (2008) Circulating
tumor cells predict survival benefit from treatment in
Alunni-Fabbroni, M. and Sandri, M.T. (2010) metastatic castration-resistant prostate cancer. Clin
Circulating tumour cells in clinical practice: methods Cancer Res 14: 6302—6309.
of detection and possible characterization. Methods
50: 289—297. De Giorgi, U., Valero, V., Rohren, E., Dawood, S.,
Ueno, N.T., Miller, M.C. et al. (2009) Circulating
Ashworth, T. (1869) A case of cancer in which cells tumor cells and [18F]fluorodeoxyglucose positron
similar to those in the tumors were seen in the blood emission tomography/computed tomography for out-
after death. Aust Med J 14: 146—149. come prediction in metastatic breast cancer. J Clin
Oncol 27: 3303—3311.
Attard, G., Swennenhuis, J.F., Olmos, D., Reid, A.H.,
Vickers, E., A’Hern, R. et al. (2009) Characterization Desar, I.M., van Herpen, C.M., van Laarhoven, H.W.,
of ERG, AR and PTEN gene status in circulating Barentsz, J.O., Oyen, W.J., van der. and Graaf, W.T.
tumor cells from patients with castration-resistant (2009) Beyond RECIST: molecular and functional
prostate cancer. Cancer Res 69: 2912—2918. imaging techniques for evaluation of response to tar-
geted therapy. Cancer Treat Rev 35: 309—321.
Budd, G.T., Cristofanilli, M., Ellis, M.J., Stopeck, A.,
Borden, E., Miller, M.C. et al. (2006) Circulating Ewing, J. (1928) Neoplastic Diseases, 6th edn, WB
tumor cells versus imaging—predicting overall survival Saunders: Philadelphia, PA.
in metastatic breast cancer. Clin Cancer Res
12: 6403—6409. Fehm, T., Becker, S., Duerr-Stoerzer, S., Sotlar, K.,
Mueller, V., Wallwiener, D. et al. (2007)
Christiansen, J.J. and Rajasekaran, A.K. (2006) Determination of HER2 status using both serum
Reassessing epithelial to mesenchymal transition as a HER2 levels and circulating tumor cells in patients
prerequisite for carcinoma invasion and metastasis. with recurrent breast cancer whose primary tumor was
Cancer Res 66: 8319—8326. HER2 negative or of unknown HER2 status. Breast
Cohen, S.J., Punt, C.J., Iannotti, N., Saidman, B.H., Cancer Res 9: R74.
Sabbath, K.D., Gabrail, N.Y. et al. (2008) Relationship Fidler, I.J. (1970) Metastasis: quantitative analysis of
of circulating tumor cells to tumor response, progres- distribution and fate of tumor embolilabeled with 125
sion-free survival, and overall survival in patients with I-5-iodo-2’-deoxyuridine. J Natl Cancer Inst
metastatic colorectal cancer. J Clin Oncol 45: 773—782.
26: 3213—3221.
Fidler, I.J. (2003) The pathogenesis of cancer metas-
Cohen, S.J., Punt, C.J., Iannotti, N., Saidman, B.H., tasis: the ‘seed and soil’ hypothesis revisited. Nat Rev
Sabbath, K.D., Gabrail, N.Y. et al. (2009) Prognostic Cancer 3: 453—458.
significance of circulating tumor cells in patients with
metastatic colorectal cancer. Ann Oncol Fidler, I.J. and Poste, G. (2008) The ‘‘seed and soil’’
20: 1223—1229. hypothesis revisited. Lancet Oncol 9: 808.

http://tam.sagepub.com 363
Therapeutic Advances in Medical Oncology 2 (6)

Friedl, P. and Gilmour, D. (2009) Collective cell Meng, S., Tripathy, D., Shete, S., Ashfaq, R.,
migration in morphogenesis, regeneration and cancer. Haley, B., Perkins, S. et al. (2004) HER-2
Nat Rev Mol Cell Biol 10: 445—457. gene amplification can be acquired as breast can-
cer progresses. Proc Natl Acad Sci U S A
Hayes, D.F., Cristofanilli, M., Budd, G.T., Ellis, M.J., 101: 9393—9398.
Stopeck, A., Miller, M.C. et al. (2006) Circulating
tumor cells at each follow-up time point during ther- Miller, M.C., Doyle, G.V. and Terstappen, L.W.
apy of metastatic breast cancer patients predict pro- (2010) Significance of circulating tumor cells detected
gression-free and overall survival. Clin Cancer Res by the CellSearch system in patients with metastatic
12: 4218—4224. breast colorectal and prostate cancer. J Oncol
2010: 617421.
Hayes, D.F., Walker, T.M., Singh, B., Vitetta, E.S.,
Uhr, J.W., Gross, S. et al. (2002) Monitoring expres- Mok, T.S., Wu, Y.L., Thongprasert, S., Yang, C.H.,
sion of HER-2 on circulating epithelial cells in patients Chu, D.T., Saijo, N. et al. (2009) Gefitinib or
with advanced breast cancer. Int J Oncol carboplatin-paclitaxel in pulmonary adenocarcinoma.
21: 1111—1117. N Engl J Med 361: 947—957.

Hou, J.M., Greystoke, A., Lancashire, L., Cummings, Moreno, J.G., O’Hara, S.M., Gross, S., Doyle, G.,
J., Ward, T., Board, R. et al. (2009) Evaluation of cir- Fritsche, H., Gomella, L.G. et al. (2001) Changes in
culating tumor cells and serological cell death bio- circulating carcinoma cells in patients with metastatic
markers in small cell lung cancer patients undergoing prostate cancer correlate with disease status. Urology
chemotherapy. Am J Pathol 175: 808—816. 58: 386—392.

Ilina, O. and Friedl, P. (2009) Mechanisms of collec- Nagrath, S., Sequist, L.V., Maheswaran, S., Bell,
tive cell migration at a glance. J Cell Sci D.W., Irimia, D., Ulkus, L. et al. (2007) Isolation of
122: 3203—3208. rare circulating tumour cells in cancer patients by
microchip technology. Nature 450: 1235—1239.
Kobayashi, S., Boggon, T.J., Dayaram, T., Janne, P.A.,
Kocher, O., Meyerson, M. et al. (2005) EGFR muta- Nakamura, S., Yagata, H., Ohno, S., Yamaguchi, H.,
tion and resistance of non-small-cell lung cancer to Iwata, H., Tsunoda, N. et al. (2009) Multi-center study
gefitinib. N Engl J Med 352: 786—792. evaluating circulating tumor cells as a surrogate for
response to treatment and overall survival in metastatic
Krebs, M.G., Sloane, R., Priest, L., Lancashire, L., breast cancer. Breast Cancer [Epub Aug 1 2009].
Hou, J.M., Greystoke, A. et al. (2010) Evaluation and
prognostic significance of circulating tumor cells in Nguyen, D.X., Bos, P.D. and Massague, J. (2009)
patients with non small cell lung cancer (NSCLC). Metastasis: from dissemination to organ-specific
J Clin Oncol, in press. colonization. Nat Rev Cancer 9: 274—284.

Leversha, M.A., Han, J., Asgari, Z., Danila, D.C., Lin, Nole, F., Munzone, E., Zorzino, L., Minchella, I.,
O., Gonzalez-Espinoza, R. et al. (2009) Fluorescence Salvatici, M., Botteri, E. et al. (2008) Variation of
in situ hybridization analysis of circulating tumor cells circulating tumor cell levels during treatment of
in metastatic prostate cancer. Clin Cancer Res metastatic breast cancer: prognostic and therapeutic
15: 2091—2097. implications. Ann Oncol 19: 891—897.

Liotta, L.A., Saidel, M.G. and Kleinerman, J. (1976) Olmos, D., Arkenau, H.T., Ang, J.E., Ledaki, I.,
The significance of hematogenous tumor cell clumps Attard, G., Carden, C.P. et al. (2009) Circulating
tumour cell (CTC) counts as intermediate end points
in the metastatic process. Cancer Res 36: 889—894.
in castration-resistant prostate cancer (CRPC): a
Liu, M.C., Shields, P.G., Warren, R.D., Cohen, P., single-centre experience. Ann Oncol 20: 27—33.
Wilkinson, M., Ottaviano, Y.L. et al. (2009)
Paget, S. (1889) The distribution of secondary
Circulating tumor cells: a useful predictor of treatment
growths in cancer of the breast. Lancet 1: 571—573.
efficacy in metastatic breast cancer. J Clin Oncol
27: 5153—5159. Pantel, K., Alix-Panabieres, C. and Riethdorf, S.
(2009) Cancer micrometastases. Nat Rev Clin Oncol
Maheswaran, S., Sequist, L.V., Nagrath, S., Ulkus, L.,
6: 339—351.
Brannigan, B., Collura, C.V. et al. (2008) Detection of
mutations in EGFR in circulating lung-cancer cells. Paterlini-Brechot, P. and Benali, N.L. (2007)
N Engl J Med 359: 366—377. Circulating tumor cells (CTC) detection: clinical
impact and future directions. Cancer Lett
Mandrekar, S.J., Qi, Y., Hillman, S.L., Allen Ziegler, 253: 180—204.
K.L., Reuter, N.F., Rowland Jr, K.M. et al. (2010)
Endpoints in phase II trials for advanced non-small cell Pestrin, M., Bessi, S., Galardi, F., Truglia, M., Biggeri,
lung cancer. J Thorac Oncol 5: 3—9. A., Biagioni, C. et al. (2009) Correlation of HER2
status between primary tumors and corresponding
Martin, M., Garcia-Saenz, J.A., Maestro de las Casas, circulating tumor cells in advanced breast cancer
M.L., Vidaurreta, M., Puente, J., Veganzones, S. et al. patients. Breast Cancer Res Treat 118: 523—530.
(2009) Circulating tumor cells in metastatic breast
cancer: timing of blood extraction for analysis. Riethdorf, S., Fritsche, H., Muller, V., Rau, T.,
Anticancer Res 29: 4185—4187. Schindlbeck, C., Rack, B. et al. (2007) Detection of

364 http://tam.sagepub.com
MG Krebs, J-M Hou et al.

circulating tumor cells in peripheral blood of patients phenotype in peripheral blood of patients with breast
with metastatic breast cancer: a validation study of the cancer. Cancer Lett 288: 99—106.
CellSearch system. Clin Cancer Res 13: 920—928.
Thiery, J.P. (2003) Epithelial—mesenchymal transi-
Scher, H.I., Jia, X., de Bono, J.S., Fleisher, M., Pienta, tions in development and pathologies. Curr Opin Cell
K.J., Raghavan, D. et al. (2009) Circulating tumour Biol 15: 740—746.
cells as prognostic markers in progressive, castration-
resistant prostate cancer: a reanalysis of IMMC38 trial Thiery, J.P. and Sleeman, J.P. (2006) Complex net-
data. Lancet Oncol 10: 233—239. works orchestrate epithelial-mesenchymal transitions.
Nat Rev Mol Cell Biol 7: 131—142.
Sequist, L.V., Bell, D.W., Lynch, T.J. and Haber, D.A.
(2007) Molecular predictors of response to epidermal Tibbe, A.G., Miller, M.C. and Terstappen, L.W.
growth factor receptor antagonists in non-small-cell (2007) Statistical considerations for enumera-
lung cancer. J Clin Oncol 25: 587—595. tion of circulating tumor cells. Cytometry A
71: 154—162.
Sequist, L.V., Nagrath, S., Toner, M., Haber, D.A.
and Lynch, T.J. (2009) The CTC-chip: an exciting Tol, J., Koopman, M., Miller, M.C., Tibbe, A., Cats,
new tool to detect circulating tumor cells in lung A., Creemers, G.J. et al. (2010) Circulating tumour
cancer patients. J Thorac Oncol 4: 281—283. cells early predict progression-free and overall survival
in advanced colorectal cancer patients treated with
Smirnov, D.A., Zweitzig, D.R., Foulk, B.W., Miller, chemotherapy and targeted agents. Ann Oncol
M.C., Doyle, G.V., Pienta, K.J. et al. (2005) Global 21: 1006—1012.
gene expression profiling of circulating tumor cells.
Cancer Res 65: 4993—4997. Tsuji, T., Ibaragi, S. and Hu, G.F. (2009)
Epithelial—mesenchymal transition and cell coopera-
Stebbing, J. and Jiao, L.R. (2009) Circulating tumour tivity in metastasis. Cancer Res 69: 7135—7139.
cells as more than prognostic markers. Lancet Oncol
10: 1138—1139. Weiss, L. (2000) Metastasis of cancer: a conceptual
history from antiquity to the 1990s. Cancer Metastasis
Swennenhuis, J.F., Tibbe, A.G., Levink, R., Sipkema, Rev 19(I-XI): 193—383.
R.C. and Terstappen, L.W. (2009) Characterization of
circulating tumor cells by fluorescence in situ hybrid- Wittekind, C. and Neid, M. (2005) Cancer invasion
ization. Cytometry A 75: 520—527. and metastasis. Oncology 69(Suppl 1): 14—16.
Visit SAGE journals online
Theodoropoulos, P.A., Polioudaki, H., Agelaki, S., Yang, J., Mani, S.A. and Weinberg, R.A. (2006) http://tam.sagepub.com
Kallergi, G., Saridaki, Z., Mavroudis, D. et al. (2010) Exploring a new twist on tumor metastasis. Cancer Res
Circulating tumor cells with a putative stem cell 66: 4549—4552.

http://tam.sagepub.com 365

You might also like