ArticlePDF Available

Circulating Tumor Cells: Not All Detected Cells Are Bad and Not All Bad Cells Are Detected

Authors:

Abstract

More than 90% of cancer deaths result from the development of hematogenously disseminated metastasis. The presence of circulating tumor cells (CTCs) in patients with cancer was first reported in 1869. 1 More recently, methods have been developed to detect, isolate, and characterize CTCs in multiple different malignancies that arise in solid organs. 2 The most widely used method, designated CellSearch (Veridex, Raritan, NJ), relies on immunomagnetic capture of CTCs using antibodies against the epithelial cell adhesion molecule (EpCam), which is expressed on the cell surface of many epithelial malignancies, followed by additional characterization with 4,6-diamidino-2-phenylindole staining (to
REFERENCES
1. Lemos BD, Storer BE, Iyer JG, et al: Pathologic nodal evaluation improves
prognostic accuracy in Merkel cell carcinoma: Analysis of 5823 cases as the basis
of the first consensus staging system. J Am Acad Dermatol 63:751-761, 2010
2. Rockville Merkel Cell Carcinoma Group: Merkel cell carcinoma: Recent
progress and current priorities on etiology, pathogenesis, and clinical manage-
ment. J Clin Oncol 27:4021-4026, 2009
3. Feng H, Shuda M, Chang Y, et al: Clonal integration of a polyomavirus in
human Merkel cell carcinoma. Science 319:1096-1100, 2008
4. Touze´ A, Le Bidre E, Laude H, et al: High levels of antibodies against
Merkel cell polyomavirus identify a subset of patients with Merkel cell carcinoma
with better clinical outcome. J Clin Oncol 29:1612-1619, 2011
5. Paulson KG, Iyer JG, Tegeder AR, et al: Transcriptome-wide studies of Merkel
cell carcinoma and validation of intratumoral CD8
lymphocyte invasion as an
independent predictor of survival. J Clin Oncol 29:1539-1546, 2011
6. Zur Hausen H: The search for infectious causes of human cancers: Where
and why. Virology 392:1-10, 2009
7. Bodily J, Laimins LA: Persistence of human papillomavirus infection: Keys
to malignant progression. Trends Microbiol 19:33-39, 2011
8. Gjoerup O, Chang Y: Update on human polyomaviruses and cancer. Adv
Cancer Res 106:1-51, 2010
9. Shuda M, Arora R, Kwun HJ, et al: Human Merkel cell polyomavirus
infection I: MCV T antigen expression in Merkel cell carcinoma, lymphoid tissues
and lymphoid tumors. Int J Cancer 125:1243-1249, 2009
10. Houben R, Shuda M, Weinkam R, et al: Merkel cell polyomavirus-infected
Merkel cell carcinoma cells require expression of viral T antigens. J Virol
84:7064-7072, 2010
11. Carter JJ, Paulson KG, Wipf GC, et al: Association of Merkel cell
polyomavirus-specific antibodies with Merkel cell carcinoma. J Natl Cancer Inst
101:1510-1522, 2009
12. Tolstov YL, Pastrana DV, Feng H, et al: Human Merkel cell polyomavirus
infection II: MCV is a common human infection that can be detected by
conformational capsid epitope immunoassays. Int J Cancer 125:1250-1256, 2009
13. Pastrana DV, Tolstov YL, Becker JC, et al: Quantitation of human serore-
sponsiveness to Merkel cell polyomavirus. PLoS Pathog 5:e1000578, 2009
14. Paulson KG, Carter JJ, Johnson LG, et al: Antibodies to merkel cell
polyomavirus T antigen oncoproteins reflect tumor burden in merkel cell carci-
noma patients. Cancer Res 70:8388-8397, 2010
15. Engels EA, Frisch M, Goedert JJ, et al: Merkel cell carcinoma and HIV
infection. Lancet 359:497-498, 2002
16. Kaae J, Hansen AV, Biggar RJ, et al: Merkel cell carcinoma: Incidence,
mortality, and risk of other cancers. J Natl Cancer Inst 102:793-801, 2010
17. Sihto H, Kukko H, Koljonen V, et al: Clinical factors associated with Merkel cell
polyomavirus infection in Merkel cell carcinoma. J Natl Cancer Inst 101:938-945,
2009
18. Bhatia K, Goedert JJ, Modali R, et al: Immunological detection of viral large
T antigen identifies a subset of Merkel cell carcinoma tumors with higher viral
abundance and better clinical outcome. Int J Cancer 127:1493-1496, 2010
18a. Laude HC, Jonchère B, Maubec E, et al: Distinct merkel cell polyomavirus
molecular features in tumour and non tumour specimens from patients with
merkel cell carcinoma. PLoS Pathog 6:e1001076, 2010
19. Lowy DR, Munger K: Prognostic implications of HPV in oropharyngeal
cancer. N Engl J Med 363:82-84, 2010
20. Campo MS, O’Neil BW, Barron RJ, et al: Experimental reproduction of the
papilloma-carcinoma complex of the alimentary canal in cattle. Carcinogenesis
15:1597-1601, 1994
DOI: 10.1200/JCO.2010.34.0745; published online ahead of print at
www.jco.org on March 21, 2011
■■■
Circulating Tumor Cells: Not All Detected Cells Are
Bad and Not All Bad Cells Are Detected
Max S. Wicha and Daniel F. Hayes, The University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
See accompanying articles on pages 1547 and 1556
More than 90% of cancer deaths result from the development
of hematogenously disseminated metastasis. The presence of cir-
culating tumor cells (CTCs) in patients with cancer was first re-
ported in 1869.
1
More recently, methods have been developed to
detect, isolate, and characterize CTCs in multiple different malig-
nancies that arise in solid organs.
2
The most widely used method,
designated CellSearch (Veridex, Raritan, NJ), relies on immuno-
magnetic capture of CTCs using antibodies against the epithelial
cell adhesion molecule (EpCam), which is expressed on the cell
surface of many epithelial malignancies, followed by additional
characterization with 4,6-diamidino-2-phenylindole staining (to
demonstrate that the detected event is a nucleated cell), and by immu-
nofluorescence analysis with antibodies against cytokeratin (to dem-
onstrate it is epithelial) and CD45 (to demonstrate it is not a
leukocyte).
2
Numerous studies have shown that the presence of ele-
vated CTC levels, as determined by CellSearch, is negatively correlated
with prognosis in patients with metastatic cancers of the breast,
3,4
prostate,
5
and colon.
6
These studies have demonstrated the analytic and clinical
validity, as defined by the Evaluation of Genomic Applications in
Practice and Prevention Initiative of the Centers for Disease Con-
trol,
7
of counting CTCs in these common solid malignancies.
However, the clinical utility of monitoring CTC levels remains
controversial. Although the US Food and Drug Administration has
cleared the CellSearch assay for clinical use, the American Society
of Clinical Oncology Tumor Marker Guidelines Committee has
not recommended incorporation of CTC levels by any method into
standard care of patients with metastatic breast cancer.
8
Nonethe-
less, studies published subsequent to that analysis seem to support
a limited role for evaluation of CTC levels. In patients with meta-
static breast, colon, and prostate cancers, these assays might be
used to determine if a patient who has been receiving a given
regimen for some time has progressive disease and would be better
treated with an alternative regimen. Furthermore, the Southwest
Oncology Group is conducting a prospective randomized clinical
trial (S0500; A Randomized Phase III Trial to Test the Strategy of
Changing Therapy Versus Maintaining Therapy for Metastatic
Breast Cancer Patients Who Have Elevated Circulating Tumor Cell
Levels at First Follow-Up Assessment) that is designed to test the
clinical utility of changing therapy for patients with metastatic
breast cancer who have not cleared CTCs after only one cycle of a
new, first-line chemotherapy.
2
The use of any assay for CTCs in early-stage solid malignan-
cies has been limited by the poor performance characteristics in
Editorials
1508 Published by the American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY
Downloaded from jco.ascopubs.org on February 20, 2013. For personal use only. No other uses without permission.
Copyright © 2011 American Society of Clinical Oncology. All rights reserved.
this setting: low sensitivity and poor specificity. Only approxi-
mately 10% of patients with stage I and II breast cancer have 1
cell/23 mL whole blood, a lower cutoff in a larger volume than was
chosen to be optimal in the metastatic setting (5 CTCs/7.5 mL
whole blood). Nonetheless, a recently reported study suggests that,
using this less stringent cut point, patients with positive findings do
have a modestly but statistically significant worse prognosis.
9
Recent studies have suggested that the CellSearch technique
may underestimate the number of EpCam-expressing cells. In this
regard, more efficient capture of these cells can be facilitated by use
of alternative solid-phase capture strategies, which are either based
on immune-capture that uses anti-EpCam or that exploits physical
characteristics that might distinguish cancer from normal hema-
topoietic cells, such as size or migration differences.
10
An alterna-
tive approach to enumeration of CTCs is to estimate their presence
by use of reverse transcriptase polymerase chain reaction to detect
epithelial transcripts, which presumably should not be present in
normal hematopoietic cells. This strategy has been applied in
breast, lung, prostate, and colon cancers,
11
and has been found to
be more sensitive than immune-capture techniques and to predict
prognosis in early-stage breast and colon cancers as well as in
metastatic prostate cancers.
12-14
Taken together, these studies suggest an exciting, but yet
unproven, potential for detection, enumeration, and monitoring
of CTCs in patients with a variety of solid tumors, either in the
early-stage or late-stage disease settings. However, none of the
assays are ideal; they lack both sensitivity and specificity. In partic-
ular, specificity may not be just technical, but biologic as well.
Technical nonspecificity implies that the assay detects an entity
that, after more careful scrutiny, is not found to be a cancer cell.
Perhaps more importantly, biologic nonspecificity implies that the
assay detects an entity that by all criteria is a cancer cell, but which
lacks the ability to invade, proliferate, and cause a metastasis.
Therefore, it would be of great value to not only enumerate CTCs
but to characterize them as well. In this regard, several authors have
reported the ability to analyze CTCs for a number of markers,
including among others HER2, insulin-like growth factor receptor-1,
urokinase-type plasminogen activator, BCL2, and the apoptotic
marker M30.
10
Cancer Stem Cells
It has long been appreciated that cancers are composed of heter-
ogeneous populations of cells. Evidence is rapidly accumulating that
many, if not most, cancers contain populations of cells that display
stem-cell properties.
15
These cancer stem cells (CSCs), by virtue of
their relative resistance to classical therapies, including radiation and
cytotoxic chemotherapy, may contribute to treatment failure and
relapse.
16,17
Furthermore, in preclinical models, CSCs have been dem-
onstrated to be the cells with the greatest invasive and metastatic
capacity.
18
If this is the case in patients with cancer, then one would
predict that CTCs may be enriched for CSCs compared with the
primary tumors from which they originated. Indeed, in women with
metastatic breast cancer, CTCs were found to be highly enriched for
cells that expressed the breast CSC markers CD44
/CD24
–19
or alde-
hyde dehydrogenase (ALDH1).
20
Interestingly, micrometastasis de-
tected in the bone marrow of these patients showed a similar CSC
profile, which suggests an important functional role of circulating
tumor stem cells (CTSCs) in mediating micrometastasis.
21
Although
micrometastases are enriched for cells expressing CSC markers, mac-
rometastases more closely resemble the profile of the primary tumor,
which suggests that micrometastases are initiated by cancer stem cells
that can then self-renew as well as generate bulk tumor populations at
distant sites.
CTSCs As Prognostic Biomarkers
If CTSCs are the cells that are responsible for the generation of
metastatic disease, then one would predict that assessment of these
cells for biologically important markers, such as those that confer
cell stemness, would provide more clinically relevant prognostic
and predictive information than simple enumeration—in other
words, biologic specificity. Indeed, one of the major limitations of
the CellSearch assay is the reliance of this technique on the expres-
sion of EpCam for cellular detection. Many CSCs display proper-
ties of epithelial mesenchymal transition (EMT) in which
expression of cell surface EpCam is downregulated.
22
The EpCam-
negative EMT-like state has been associated with increased meta-
static capacity in preclinical models and metastatic lesions in
patients.
23,24
These observations additionally support develop-
ment of methodology that is not dependent on EpCam expression,
not simply to increase sensitivity, but to provide the opportunity to
detect CTSCs that express the EMT phenotype—in other words, to
enhance biologic specificity.
Quantitative polymerase chain reaction (qPCR) might repre-
sent such a technique. However, a number of validated CSC mark-
ers, such as CD44 and ALDH, are also expressed in hematopoietic
stem cells, and therefore blind qPCR of whole blood will be fraught
with false-positive findings. To circumvent this problem, several
investigators have used negative selection with antibodies against
the hematopoietic-specific antigen CD45 followed by qPCR or cell
analysis on the remaining cell population using stem cell markers.
Although one must have significant concerns about the ability to
completely eliminate contamination of leukocytes with such a
strategy, this technique has been used to demonstrate an increased
proportion of CD44
or ALDH
CTCs in women with metastatic
breast cancer.
19,20
An alternative approach is the use of markers such as CD133.
CD133 has been reported to be expressed in a number of solid
tumor CSCs, but not by hematopoietic stem cells. In this issue of
the Journal of Clinical Oncology, Iinuma et al
25
report use of a
PCR-based assay to detect mRNAs for CD133 in combination with
those for cytokeratin (CK) and carcinoembryonic antigens (CEA)
in the CTCs of patients with colorectal cancer. The authors inves-
tigated this assay in a multi-institutional study involving 735 pa-
tients, including a training set of 420 patients from whom archived
specimens were retrospectively available and 315 patients who
were prospectively enrolled in a validation set. These investigators
report that the overall survival and disease-free survival of patients
with detectable CEA/CK/CD133
mRNA were significantly worse
than those of patients whose blood was negative for expression of
these markers (P.003).
Importantly, the authors
25
report that the assay was not prog-
nostic in patients with Dukes’ stage A or Dukes’ stage B cancer who
were determined to have favorable prognoses on the basis of
clinical and pathologic features. The assay was prognostic in
patients with Dukes’ stage B cancer who were separated out
because of a less favorable prognosis on the basis of clinical and
Editorials
www.jco.org Published by the American Society of Clinical Oncology 1509
Downloaded from jco.ascopubs.org on February 20, 2013. For personal use only. No other uses without permission.
Copyright © 2011 American Society of Clinical Oncology. All rights reserved.
pathologic features (termed as separated patients in the study)
and in patients with Dukes’ stage C cancer. Clinically, these
findings are important. They suggest that patients with Dukes’
stage A and favorable Dukes’ stage B colorectal cancer have such
a low probability of recurrence that, even with this ultrasensi-
tive assay, one cannot find a group for whom adjuvant chemo-
therapy might be appropriate. In separated patients with Dukes’
stage B, none of whom received adjuvant chemotherapy, the
assay appears to identify a group of patients who, if chemother-
apy is effective, might benefit from it.
The results for Dukes’ stage C cancer raise a worrisome concern.
In addition to identifying a group of patients with poor prognosis in
the absence of therapy, does this assay also predict resistance to the
therapy itself? This hypothesis is completely plausible, given that a
major hallmark of cancer stem cellness is resistance to noxious stimuli,
such as chemotherapy.
16,17
One cannot answer this question without a
concurrent, randomly assigned, untreated control group, but the large
separation in the survival curves in patients with Dukes’ stage C can-
cer, all of whom received adjuvant chemotherapy, suggests that pa-
tients with circulating CEA/CK/CD133–positive cells may have
resistant disease. However, this concern is lessened by the observation
that the CSC marker CD133 seemed to be necessary to classify poor
prognosis in the untreated patients with Dukes’ stage B, but that the
CEA/CK assay was as prognostic as the CEA/CK/CD133 assay in the
group with Dukes’ stageC.
There are a number of limitations of this study, several of which
are acknowledged by the authors.
25
The use of the PCR assay does not
permit analysis at the single cell level, a feature that is important in CSC
analysis. Furthermore, the authors did not use negative selection to
remove hematopoietic cells. In fact, the authors acknowledge that
because CD133 is expressed in endothelial progenitor cells,
26
which
are known to be present in the circulation, they cannot conclusively
determine the cellular origin of CD133 mRNA. The development of
alternative strategies that permit isolation of individual CTSCs might
obviate some of these concerns. Approaches that use negative selec-
tion of hematopoietic cells followed by capture with CSC antigens
represent one such approach. Alternative approaches that are not
dependent on EpCam include cellular isolation on the basis of size or
microfluidic properties.
27
In conclusion, we are impressed by the rigorous technical and
clinical efforts the authors have applied to this prospectively
performed study of CTCs.
25
However, because of the limitations dis-
cussed above, and because therapy was applied according to standard
of care and not within a prospective randomized trial, these results
must be considered level of evidence II or III and not ready for use
when making clinical decisions.
28
The assay was not helpful in select-
ing patients with a favorable stage (Dukes’ stage A and favorable
Dukes’ stage B) who might benefit from therapy. One cannot deter-
mine whether separated patients with Dukes’ stage B with a positive
assay result will benefit from therapy, and likewise, from these results,
one cannot differentiate patients with Dukes’ stage C cancer who
might not have needed therapy.
Nevertheless, this study by Iinuma et al
25
does establish the
highly reproducible analytic validity of this assay in patients with
colorectal cancers. Moreover, the study raises fascinating hypoth-
eses that could and should be addressed in either archived speci-
mens from previously conducted trials or from ongoing or newly
designed randomized trials to determine the clinical utility of this
assay; it could have enormous utility for directing adjuvant therapy
for patients with this disease.
In addition, these results have implications for future clinical
trials of new therapeutic agents. To the extent that cancers follow a
stem cell model, current clinical response criteria may not accu-
rately predict patient outcome. The Response Evaluation Criteria
in Solid Tumors Group (RECIST) currently used to assess efficacy
in most clinical trials largely reflect the effects of therapeutic agents
on bulk tumor populations rather than the rarer CSC populations
that drive tumor growth and metastasis. In addition, pharmacody-
namic end points of targeted therapeutic agents are currently as-
sessed on bulk cell populations. This highlights the importance of
being able to assess the therapeutic effects of treatments on CSCs.
The utility of such an approach has been demonstrated in tissue
samples obtained before and after therapy in women undergoing
neoadjuvant therapy for locally advanced breast cancer.
16
How-
ever, the application of such an approach is limited by the technical
difficulty of obtaining pre- and post- treatment biopsies from
patients with metastatic solid malignancies. The ability to readily
obtain samples of CTSCs may obviate these difficulties and provide
the equivalent of multiple serial biopsies. In addition, the develop-
ment of technologies capable of reliably isolating and molecularly
characterizing CTSCs should facilitate the development of CSC-
targeted therapeutics.
AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST
Although all authors completed the disclosure declaration, the following
author(s) indicated a financial or other interest that is relevant to the subject
matter under consideration in this article. Certain relationships marked
with a “U” are those for which no compensation was received; those
relationships marked with a “C” were compensated. For a detailed
description of the disclosure categories, or for more information about
ASCO’s conflict of interest policy, please refer to the Author Disclosure
Declaration and the Disclosures of Potential Conflicts of Interest section in
Information for Contributors.
Employment or Leadership Position: None Consultant or Advisory
Role: Max S. Wicha, OncoMed Pharmaceuticals (C), Pfizer (C), Dompe
(C); Daniel F. Hayes, OncoMed Pharmaceuticals (C), Pfizer (C), Dompe
(U) Stock Ownership: Max S. Wicha, OncoMed Pharmaceuticals
Honoraria: None Research Funding: Max S. Wicha, Dompe; Daniel F.
Hayes, Veridex, Pfizer, Novartis Expert Testimony: None Other
Remuneration: None
AUTHOR CONTRIBUTIONS
Manuscript writing: All authors
Final approval of manuscript: All authors
REFERENCES
1. Ashworth TR: A case of cancer in which cells similar to those in the
tumours were seen in the blood after death. Aust Med J 14:146-149, 1869
2. Hayes DF, Smerage J: Is there a role for circulating tumor cells in the
management of breast cancer? Clin Cancer Res 14:3646-3650, 2008
3. Cristofanilli M, Budd GT, Ellis MJ, et al: Circulating tumor cells, disease
progression, and survival in metastatic breast cancer. N Engl J Med 351:781-791,
2004
4. Liu MC, Shields PG, Warren RD, et al: Circulating tumor cells: A useful
predictor of treatment efficacy in metastatic breast cancer. J Clin Oncol 27:5153-
5159, 2009
5. de Bono JS, Scher HI, Montgomery RB, et al: Circulating tumor cells
predict survival benefit from treatment in metastatic castration-resistant prostate
cancer. Clin Cancer Res 14:6302-6309, 2008
6. Cohen SJ, Punt CJ, Iannotti N, et al: Relationship of circulating tumor cells
to tumor response, progression-free survival, and overall survival in patients with
metastatic colorectal cancer. J Clin Oncol 26:3213-3221, 2008
Editorials
1510 Published by the American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY
Downloaded from jco.ascopubs.org on February 20, 2013. For personal use only. No other uses without permission.
Copyright © 2011 American Society of Clinical Oncology. All rights reserved.
7. Teutsch SM, Bradley LA, Palomaki GE, et al: The Evaluation of Genomic
Applications in Practice and Prevention (EGAPP) Initiative: Methods of the
EGAPP Working Group. Genet Med 11:3-14, 2009
8. Harris L, Fritsche H, Mennel R, et al: American Society of Clinical Oncology
2007 update of recommendations for the use of tumor markers in breast cancer.
J Clin Oncol 25:5287-5312, 2007
9. Rack B, Schindlbeck C, Andergassen U, et al: Prognostic role of circulating
tumor cells in the peripheral blood of primary breast cancer patients. Cancer Res
70:93, 2010 (suppl 24; abstr S6-5)
10. Hayes DF, Smerage JB: Circulating tumor cells. Prog Mol Biol Transl Sci
95:95-112, 2010
11. Pantel K, Alix-Panabie` res C: Circulating tumour cells in cancer patients:
Challenges and perspectives. Trends Mol Med 16:398-406, 2010
12. Ignatiadis M, Kallergi G, Ntoulia M, et al: Prognostic value of the molecular
detection of circulating tumor cells using a multimarker reverse transcription-PCR
assay for cytokeratin 19, mammaglobin A, and HER2 in early breast cancer. Clin
Cancer Res 14:2593-2600, 2008
13. Uen YH, Lin SR, Wu DC, et al: Prognostic significance of multiple molecular
markers for patients with stage II colorectal cancer undergoing curative resec-
tion. Ann Surg 246:1040-1046, 2007
14. Halabi S, Small EJ, Hayes DF, et al: Prognostic significance of reverse
transcriptase polymerase chain reaction for prostate-specific antigen in meta-
static prostate cancer: A nested study within CALGB 9583. J Clin Oncol
21:490-495, 2003
15. Boman BM, Wicha MS: Cancer stem cells: A step toward the cure. J Clin
Oncol 26:2795-2799, 2008
16. Li X, Lewis MT, Huang J, et al: Intrinsic resistance of tumorigenic breast
cancer cells to chemotherapy. J Natl Cancer Inst 100:672-679, 2008
17. Creighton CJ, Li X, Landis M, et al: Residual breast cancers after conven-
tional therapy display mesenchymal as well as tumor-initiating features. Proc Natl
Acad SciUSA106:13820-13825, 2009
18. Charafe-Jauffret E, Ginestier C, Iovino F, et al: Breast cancer cell lines
contain functional cancer stem cells with metastatic capacity and a distinct
molecular signature. Cancer Res 69:1302-1313, 2009
19. Aktas B, Tewes M, Fehm T, et al: Stem cell and epithelial-mesenchymal
transition markers are frequently overexpressed in circulating tumor cells of
metastatic breast cancer patients. Breast Cancer Res 11:R46, 2009
20. Fehm T, Hoffmann O, Aktas B, et al: Detection and characterization of
circulating tumor cells in blood of primary breast cancer patients by RT-PCR and
comparison to status of bone marrow disseminated cells. Breast Cancer Res
11:R59, 2009
21. Balic M, Lin H, Young L, et al: Most early disseminated cancer cells
detected in bone marrow of breast cancer patients have a putative breast cancer
stem cell phenotype. Clin Cancer Res 12:5615-5621, 2006
22. Mani SA, Guo W, Liao MJ, et al: The epithelial-mesenchymal transition
generates cells with properties of stem cells. Cell 133:704-715, 2008
23. Ota I, Li XY, Hu Y, et al: Induction of a MT1-MMP and MT2-MMP-
dependent basement membrane transmigration program in cancer cells by
Snail1. Proc Natl Acad SciUSA106:20318-20323, 2009
24. Bonnomet A, Brysse A, Tachsidis A, et al: Epithelial-to-mesenchymal
transitions and circulating tumor cells. J Mammary Gland Biol Neoplasia 15:261-
273, 2010
25. Iinuma H, Watanabe T, Mimori K, et al: Clinical significance of circulating
tumor cells, including cancer stem-like cells, in peripheral blood for recurrence
and prognosis in patients with Dukes’ stage B and C colorectal cancer. J Clin
Oncol 29:1547-1555, 2011
26. Spring H, Schu¨ ler T, Arnold B, et al: Chemokines direct endothelial progenitors into
tumor neovessels. Proc Natl Acad SciUSA102:18111-18116, 2005
27. Zheng S, Lin H, Liu JQ, et al: Membrane microfilter device for selective
capture, electrolysis and genomic analysis of human circulating tumor cells.
J Chromatogr A 1162:154-161, 2007
28. Simon RM, Paik S, Hayes DF: Use of archived specimens in evaluation
of prognostic and predictive biomarkers. J Natl Cancer Inst 101:1446-1452,
2009
DOI: 10.1200/JCO.2010.34.0026; published online ahead of print at
www.jco.org on March 21, 2011
■■■
Editorials
www.jco.org Published by the American Society of Clinical Oncology 1511
Downloaded from jco.ascopubs.org on February 20, 2013. For personal use only. No other uses without permission.
Copyright © 2011 American Society of Clinical Oncology. All rights reserved.
... Another category of cancer cells that requires identification is the circulating tumour cell (CTC) population, responsible for tumour spread and the initiation of metastatic disease. CTCs are epithelial malignant cells separated from the primary tumour, which underwent the epithelial-mesenchymal transition (EMT), and through intravasation into the blood stream they reached distant anatomic sites in order to extravasate and to form micrometastases [43]. The composition of the CTC population is highly heterogenous, consisting of subpopulations of various phenotypes, including cells with stem-like properties [43]. ...
... CTCs are epithelial malignant cells separated from the primary tumour, which underwent the epithelial-mesenchymal transition (EMT), and through intravasation into the blood stream they reached distant anatomic sites in order to extravasate and to form micrometastases [43]. The composition of the CTC population is highly heterogenous, consisting of subpopulations of various phenotypes, including cells with stem-like properties [43]. Several studies found correlations between the amount and type of CTC identified in the blood and clinical outcome of cancer patients, showing that the detection of CTCs with stem-like properties could provide prognostic and predictive information alike, which would assist with treatment adaptation and personalisation [44,45]. ...
Article
Full-text available
Cancer stem cells are known to play a key role in tumour development, proliferation, and metastases. Their unique properties confer resistance to therapy, often leading to treatment failure. It is believed that research into the identification, targeting, and eradication of these cells can revolutionise oncological treatment. Based on the principle that what cannot be seen, cannot be targeted, a primary step in cancer management is the identification of these cells. The current review aims to encompass the state-of-the-art functional imaging techniques that enable the identification of cancer stem cells via various pathways and mechanisms. The paper presents in vivo molecular techniques that are currently available or await clinical implementation. Challenges and future prospects are highlighted to open new research avenues in cancer stem cell imaging.
... CTC detection has gradually become a research hotspot in the diagnosis and treatment of malignant tumors due to its minimally invasive, high sensitivity, and high speci city advantages. In clinical practice, it has become an important means of non-invasive real-time monitoring of tumor progression (9,10). Previous studies have shown that LDCT combined with CTC is a reliable method for early lung cancer screening (11). ...
Preprint
Full-text available
Background Exploring the clinical application value of combining circulating tumor cell (CTC) with artificial intelligence in predicting the pathological nature of pulmonary nodules. Constructing a prediction model based on factors related to lung cancer to provide reliable prediction criteria for clinical doctors to predict the pathological nature of pulmonary nodules, in order to guide clinical doctors in judging the benign and malignant nature and infiltration degree of pulmonary nodules (PN). Methods This study included a total of 76 patients with PN who underwent surgical treatment. Based on preoperative imaging of the patients, an artificial intelligence imaging system called "United Imaging Intelligence" was used to classify the pulmonary nodules into three levels of "low risk", "medium risk", and "high risk", and the preoperative CTC level of the patients was recorded. Multiple logistic regression analysis was used to analyze the risk factors affecting the nature of the PN and to construct relevant column charts. Receiver operating characteristic (ROC) curves were used to analyze the diagnostic value of artificial intelligence and CTC levels for the nature of PN lesions. Results The artificial intelligence model for grouping benign and malignant PN and the difference in CTC levels have statistical significance (P < 0.05). The results of multifactor logistic regression analysis showed that artificial intelligence high-risk grouping, CTC level, and age are independent risk factors affecting the nature of PN (P < 0.05). We also constructed a column chart to guide clinical doctors in treatment. The area under the curve (AUC) for the artificial intelligence risk grouping and CTC level diagnosis of malignant PN were 78.9% and 74.3%, respectively. Conclusion Artificial intelligence model combined with CTC detection helps improve the accuracy of lung nodule characterization diagnosis and assists in guiding clinical decisions.
... Breast cancer is a heterogeneous disease and a major cause of mortality among women worldwide [1]. Ineffective utilization of expensive cancer screening methods and lack of diagnostic assays based on molecular markers for detecting the early and curable stages of breast cancer is the primary cause of mortality and poor survival among breast cancer patients in most developing countries [2,3]. Breast cancer can be classified into different types based on its hereditary/receptor status [4] and into different grades and stages based on the extent of tumor progression. ...
Research
Full-text available
Background Invasive ductal carcinoma (IDC) is the most common form of breast cancer which accounts for 85% of all breast cancer diagnoses. Non-invasive and early stages have a better prognosis than late-stage invasive cancer that has spread to lymph nodes. The involvement of microRNAs (miRNAs) in the initiation and progression of breast cancer holds great promise for the development of molecular tools for early diagnosis and prognosis. Therefore, developing a cost effective, quick and robust early detection protocol using miRNAs for breast cancer diagnosis is an imminent need that could strengthen the health care system to tackle this disease around the world. Methods We have analyzed putative miRNAs signatures in 100 breast cancer samples using two independent high fidelity array systems. Unique and common miRNA signatures from both array systems were validated using stringent double-blind individual TaqMan assays and their expression pattern was confirmed with tissue microarrays and northern analysis. In silico analysis were carried out to find miRNA targets and were validated with q-PCR and immunoblotting. In addition, functional validation using antibody arrays was also carried out to confirm the oncotargets and their networking in different pathways. Similar profiling was carried out in Brca2/p53 double knock out mice models using rodent miRNA microarrays that revealed common signatures with human arrays which could be used for future in vivo functional validation. Results Expression profile revealed 85% downregulated and 15% upregulated microRNAs in the patient samples of IDC. Among them, 439 miRNAs were associated with breast cancer, out of which 107 miRNAs qualified to be potential biomarkers for the stratification of different types, grades and stages of IDC after stringent validation. Functional validation of their putative targets revealed extensive miRNA network in different oncogenic pathways thus contributing to epithelial-mesenchymal transition (EMT) and cellular plasticity. Conclusion This study revealed potential biomarkers for the robust classification as well as rapid, cost effective and early detection of IDC of breast cancer. It not only confirmed the role of these miRNAs in cancer development but also revealed the oncogenic pathways involved in different progressive grades and stages thus suggesting a role in EMT and cellular plasticity during breast tumorigenesis per se and IDC in particular. Thus, our findings have provided newer insights into the miRNA signatures for the classification and early detection of IDC.
... It was found that most CTCs degenerate, die or become dormant in the circulation, while a very small number of CTC cells form micro-metastases in other organs (Wicha and Hayes 2011). PSA is currently used as an important evaluation criterion in prostate cancer screening and treatment. ...
Article
Full-text available
Purpose Prostate cancer (PCa) is one of the most common malignancies in men and one of the leading causes of cancer-related deaths; circulating tumor cells (CTC) are malignant cells that have broken off from original tumor or metastatic sites and extravasated into the blood vessels either naturally or maybe as a consequence of surgical procedures. This study aims to explore the feasibility of liquid biopsy technique to diagnose prostate cancer. Method We constructed an assay platform integrating magnetic separation and fluorescence in situ hybridization (FISH) to effectively capture prostate cancer CTCs and evaluate the distribution between healthy volunteers and prostate cancer patients, respectively. Results There was a significant difference in the number of CTCs between the healthy population and prostate cancer patients (P < 0.001). The results of the study showed that the CTCs capture identification system has good sensitivity and specificity in identifying prostate cancer patients. Conclusion The CTCs test allows us to accurately identify patients who are at high risk for prostate cancer, allowing for early intervention and treating patients effectively.
... However, several challenges still exist. CTC assays currently lack adequate sensitivity, especially in early-stage solid malignancies, due to low numbers of detectable CTCs [125]. One important area of research is the development of more sensitive and specific methods for detecting and analyzing CTCs. ...
Article
Full-text available
Circulating tumor cells (CTCs) are tumor cells shed from the primary tumor into circulation, with clusters of CTCs responsible for cancer metastases. CTC detection and isolation from the bloodstream are based on properties distinguishing CTCs from normal blood cells. Current CTC detection techniques can be divided into two main categories: label dependent, which depends upon antibodies that selectively bind cell surface antigens present on CTCs, or label-independent detection, which is detection based on the size, deformability, and biophysical properties of CTCs. CTCs may play significant roles in cancer screening, diagnosis, treatment navigation, including prognostication and precision medicine, and surveillance. In cancer screening, capturing and evaluating CTCs from peripheral blood could be a strategy to detect cancer at its earliest stage. Cancer diagnosis using liquid biopsy could also have tremendous benefits. Full utilization of CTCs in the clinical management of malignancies may be feasible in the near future; however, several challenges still exist. CTC assays currently lack adequate sensitivity, especially in early-stage solid malignancies, due to low numbers of detectable CTCs. As assays improve and more trials evaluate the clinical utility of CTC detection in guiding therapies, we anticipate increased use in cancer management.
... Nonsmall cell lung cancer (NSCLC) is the most common cause of superior vena cava (SVC) syndrome, accounting for around half of all cases. Due to restriction of blood flow through the superior vena cava, this usually manifests as gradual face/neck swelling with dilated neck veins and upper extremity swelling [21]. ...
Article
Full-text available
Lung cancer is a diagnosis that has risen to the top of the global cancer death toll. Adenocarcinoma, squamous cell carcinoma, and large cell carcinoma are all types of non-small cell lung cancer. This exercise examines the diagnosis and treatment of non-small cell lung cancer and emphasizes the importance of the interprofessional team inpatient care. This review article aims to: review the etiology of non-small cell lung cancer, describe the appropriate steps for non-small cell lung cancer evaluation, outline non-small cell lung cancer management options, and summarize the importance of collaboration and communication among the interprofessional team to improve care coordination for non-small cell lung cancer patients. Following the diagnosis, an interprofessional approach with medical oncology, radiation oncology, thoracic surgery, and pathology should be used to maximize the patient's treatment plan based on their TNM staging at the time of diagnosis.
... In addition, in a cohort of metastatic patients, the number of stem-like CTC subsets was found to be more associated with decreased OS and an increased number of metastatic sites than with the number of CTC [10]. Due to the potential relationship between stem cells and tumor growth, metastasis, and drug resistance, CTCs with stem cell phenotype and function may be a subpopulation worthy of further study [11][12][13][14]. ...
Article
Full-text available
Despite increasing interest in the study of circulating tumor cells (CTC) subsets, especially epithelial-mesenchymal transition (EMT) and stem cells subsets of CTC that play a key role in tumor recurrence and metastasis, there is no evidence from meta-analyses that shows the correlation between stem-like CTCs and prognosis in cancer patients. Thus, we performed a meta-analysis to assess its prognostic value. Sixteen articles were screened by searching the PubMed, Embase, Cochrane, China National Knowledge Internet (CNKI) and Wanfang databases. The hazard ratio (HR) and 95% confidence interval (95% CI) extracted from each article were summarized. Patients with positive stem-like CTCs in peripheral blood had significantly shorter overall survival (OS, HR: 2.58, 95% CI 1.76–3.79, P < 0.00001), progression-free survival (PFS, HR: 2.21, 95% CI 1.26–3.89, P = 0.006) and disease-free survival (DFS, HR: 2.53, 95% CI: 1.12–5.70, P = 0.03). This study provides the first meta-analysis evidence for the prognostic value of stem-like CTCs, demonstrating that these cells are associated with poor prognosis in cancer patients. Systematic review registration CRD42022322062
Chapter
Cancer stem cells (CSCs) are a specific type of cancer cell that can both self-renew and differentiate, playing a key role in cancer development and progression. Existing clinical biomarkers, however, are insufficient as robust biomarkers to be used in clinical practice for cancer patients due to their limited confirmation and contested prognostic relevance. Therefore, there is a large pool of potential biomarkers that have not been thoroughly explored. CSC surface biomarkers have recently been the focus of many studies in the clinical practice of cancer patients due to their potential utility in characterising the aetiology of cancer initiation, development, and metastasis. In this chapter, we discuss the most popular surface biomarkers of CSCs and provide their potential utility in cancer diagnosis and prognosis.KeywordsCancer stem cellsBiomarkersDiagnosisPrognosisOverall survival
Thesis
Der schwerwiegendste Aspekt maligner Tumore ist ihre Fähigkeit Fernmetastasen in lebenswichtigen Organen zu bilden. Bei der Mehrheit neudiagnostizierter solider maligner Tumore liegen zum Zeitpunkt der Diagnose keine Metastasen vor. Dennoch erleiden bis zu 50% der Patienten ein Rezidiv. Dies beruht auf der Disseminierung von Tumorzellen, die dann Metastasen im Sekundärgewebe bilden können. Jedoch können nicht alle disseminierten Tumorzellen zu Metastasen in distanten Organen heranwachsen. Eine kleine Subpopulation von Zellen, die ein hohes Selbsterneuerungspotential besitzt, scheint für die Entstehung von Tumoren und Rezidive verantwortlich zu sein. Diese Subpopulation wird als Krebsstammzellen oder Tumor-initiierende Zellen bezeichnet. Im ersten Teil der vorliegenden Arbeit konnte mit einem neuartigen 3D-Zellkultursystem die aggressivste Subpopulation der CETCs, die zirkulierenden Krebsstammzellen, die in-vitro zu Tumorsphären heranwachsen, identifiziert und weiter charakterisiert werden. Immunfluorimetrische, als auch genetische Untersuchungen konnten zeigen, dass sie Stammzelleigenschaften besitzen, die typisch für das Mammakarzinom sind. Die Anzahl an cCSCs spiegelte direkt die Aggressivität des Tumors wider und konnte das Risiko für Fernmetastasen bestimmen. Die Entwicklung eines patient-derived Xenograft Modells konnte zudem die Tumorigenität der in-vitro kultivierten Tumorsphären erstmals beweisen. Nach ihrer Applikation auf die Chorioallantoismembran des Hühnerembryos konnten sie innerhalb kürzester Zeit einen Tumor generieren, der pathologisch dem Primärtumor entsprach. Diese neu entwickelten Modelle konnten die Existenz der Krebsstammzellen nicht nur im Tumorgewebe sondern auch im peripheren Blut der Krebspatienten bestätigen und deren klinische Relevanz verdeutlichen. Dies eröffnet einen Weg Tumor-initiierende Zellen in unterschiedlichen Tumorentitäten zu identifizieren, molekular zu charakterisieren und zielgerichtete Therapien zu entwickeln. CETCs sind einer Reihe unterschiedlicher physiologischer und immunologischer Hürden ausgesetzt, die deren Überleben im peripheren Blut erschweren. Um sich vor der Eliminierung durch das Immunsystem erfolgreich zu schützen, nutzen die CETCs eine Vielzahl unterschiedlicher Strategien. Im zweiten Teil der vorliegenden Arbeit wurden die unterschiedlichen Mechanismen untersucht, mit denen sich die CETCs vor der Immunüberwachung tarnen können. Im Fokus der Immunonkologie stehen die sogenannten checkpoints des Immunsystems, die von den T-Zellen exprimiert werden und normalerweise eine überbordende Immunreaktion verhindern. Tumorzellen können sich diesen Mechanismus der Immunevasion zu Nutze machen. In dieser Arbeit konnte mit Hilfe immunfluorimetrischer und molekularzytogenetischer Analysen gezeigt werden, dass die CETCs bestimmte checkpoints wie PD-L1 und B7-H3 in einem hohen Prozentsatz auf ihrer Zelloberfläche exprimieren. Dies ermöglicht ihr Überleben im peripheren Blut und fördert gleichzeitig die metastatische Ausbreitung des Tumors. Eine Blockade dieser checkpoints bereits in frühen Stadien könnte klinisch relevant sein, um die Immunantwort, die sich gegen die Tumorzellen richtet, zu reaktivieren. Das Monitoring PD-L1 und B7-H3 positiver CETCs könnte insbesondere nach der operativen Entfernung des Tumors für die Überwachung der Immuntherapie nützlich sein. Ein weiterer Mechanismus, der die CETCs im peripheren Blut vor immunologischen Angriffen, den Scherkräften des Blutes und der Apoptose schützt, ist deren Interaktion mit anderen Komponenten des Blutes. Jüngste Studien deuten darauf hin, dass Thrombozyten eine entscheidende Rolle dabei spielen, das Überleben der CETCs im Blutkreislauf zu fördern. Die Tumorzell-induzierte Thrombozytenaggregation schützt die CETCs vor den Scherkräften des Blutes und gleichzeitig vor einer immunologischen Eliminierung. Dies kann jedoch auch der Grund dafür sein, dass die Antikörper die Oberflächenantigene der Tumorzellen nicht erreichen. In dieser Arbeit konnte die Interaktion zwischen den CETCs und den Thrombozyten zu drei verschiedenen Zeitpunkten nach der Blutentnahme bestätigt werden. Die Tumorzellen bildeten im Blut Aggregate mit den Thrombozyten, wodurch ihr Überleben im peripheren Blut gefördert, jedoch gleichzeitig deren Nachweis unmittelbar nach der Blutentnahme erschwert wurde. Durch die Lagerung der Blutproben über Nacht bei Raumtemperatur kam es zur Ablösung der Thrombozyten von den CETCs, wodurch sie mittels Immunfluoreszenz identifiziert werden konnten. Die Wirkung von Thrombozytenaggregationshemmern könnte einen inhibierenden Einfluss auf den Metastasierungsprozess haben. CETCs stellen wertvolle tumorspezifische Biomarker dar, die die verbliebene minimale Resterkrankung nach einer scheinbar erfolgreich abgeschlossenen Krebstherapie widerspiegeln. Zudem können sie zeitnah die Effizienz systemischer Therapien abbilden, wodurch sie eine prognostische Relevanz haben. Aufgrund der hohen intra- und intertumoralen Heterogenität, aber auch aufgrund möglicher Veränderungen der Tumorzelleigenschaften im Krankheitsverlauf ist die longitudinale Überwachung der zirkulierenden Tumorzellen von großer Bedeutung. Frühe Resistenzen, bzw. frühzeitige Rezidive lassen sich somit leicht verfolgen. Im dritten Teil der vorliegenden Arbeit konnte mit der maintrac® Methode veranschaulicht werden, dass die Quantifizierung der CETCs und vor allem der cCSCs ein geeignetes Stadien-unabhängiges Verfahren darstellt, um die Effizienz der aktuellen Therapie, wie der Immuntherapie, der Radiotherapie und der endokrinen Therapie zu überwachen. Patientinnen mit einem erhöhten Risiko für eine Therapieresistenz und ein Rezidiv könnten somit frühzeitig identifiziert und die Behandlungsstrategie angepasst werden. Die molekularbiologische Charakterisierung der Zellen hinsichtlich therapierelevanter Marker wie PD-L1, B7-H3 und AR bzw. ER könnte bei der Patientenstratifizierung für eine personalisierte Therapie behilflich sein.
Article
Full-text available
Epithelial-to-mesenchymal transition (EMT) phenomena endow epithelial cells with enhanced migratory and invasive potential, and as such, have been implicated in many physiological and pathological processes requiring cell migration/invasion. Although their involvement in the metastatic cascade is still a subject of debate, data are accumulating to demonstrate the existence of EMT phenotypes in primary human tumors, describe enhanced metastatic potential of EMT derivatives in animal models, and report EMT attributes in circulating tumor cells (CTCs). The relationships between EMT and CTCs remain largely unexplored, and we review here in vitro and in vivo data supporting a putative role of EMT processes in CTC generation and survival.
Article
Full-text available
The ability of carcinoma cells arising at primary sites to cross their underlying basement membrane (BM), a specialized form of extracellular matrix that subtends all epithelial cells, and to access the host vasculature are central features of the malignant phenotype. The initiation of the invasive phenotype has been linked to the aberrant expression of zinc-finger transcriptional repressors, like Snail1, which act by triggering an epithelial-mesenchymal cell-like transformation (EMT-like) via the regulation of largely undefined, downstream effectors. Herein, we find that Snail1 induces cancer cells to (i) degrade and perforate BM barriers, (ii) initiate angiogenesis, and (iii) and intravasate vascular networks in vivo via a matrix metalloproteinase (MMP)-dependent process. Unexpectedly, the complete Snail1 invasion program can be recapitulated by expressing directly either of the membrane-anchored metalloproteinases, MT1-MMP or MT2-MMP. The pro-invasive, angiogenic, and metastatic activities of MT1-MMP and MT2-MMP are unique relative to all other metalloproteinase family members and cannot be mimicked in vivo by the secreted MMPs, MMP-1, MMP-2, MMP-3, MMP-7, MMP-9, or MMP-13. Further, siRNA-specific silencing of MT1-MMP and MT2-MMP ablates completely the ability of Snail1 to drive cancer cell BM invasion, induce angiogenesis, or trigger intravasation. Taken together, these data demonstrate that MT1-MMP and MT2-MMP cooperatively function as direct-acting, pro-invasive factors that confer Snail1-triggered cells with the key activities most frequently linked to morbidity and mortality in cancer.
Article
Full-text available
The development of tumor biomarkers ready for clinical use is complex. We propose a refined system for biomarker study design, conduct, analysis, and evaluation that incorporates a hierarchal level of evidence scale for tumor marker studies, including those using archived specimens. Although fully prospective randomized clinical trials to evaluate the medical utility of a prognostic or predictive biomarker are the gold standard, such trials are costly, so we discuss more efficient indirect “prospective–retrospective” designs using archived specimens. In particular, we propose new guidelines that stipulate that 1) adequate amounts of archived tissue must be available from enough patients from a prospective trial (which for predictive factors should generally be a randomized design) for analyses to have adequate statistical power and for the patients included in the evaluation to be clearly representative of the patients in the trial; 2) the test should be analytically and preanalytically validated for use with archived tissue; 3) the plan for biomarker evaluation should be completely specified in writing before the performance of biomarker assays on archived tissue and should be focused on evaluation of a single completely defined classifier; and 4) the results from archived specimens should be validated using specimens from one or more similar, but separate, studies.
Article
The prognostic relevance of disseminated tumor cells in bone marrow of breast cancer patients at the time of primary diagnosis and during recurrence-free follow-up has been confirmed by large pooled analyses. Furthermore, circulating tumor cells (CTC) in peripheral blood have been shown as predictor of shortened progression-free and overall survival in metastatic disease. In view of the lack of data in early breast cancer, we evaluated whether the presence of CTC before the start of systemic adjuvant treatment increases the risk of subsequent relapse and death. Patients and Methods The SUCCESS A-Study is an open-label randomized controlled, phase III study comparing the disease-free survival after randomisation in patients treated with 3 cycles of Epirubicin (100 mg/m²)-Fluorouracil(500)- Cyclophosphamide (500, FEC)-chemotherapy, followed by 3 cycles of Docetaxel (100 mg/mg², D) versus 3 cycles of FEC, followed by 3 cycles of Gemcitabine (1,000mg/m² d1,8)-Docetaxel (75 mg/m²)(DG). In 2026 patients CTC were analyzed using the CellSearchSystem (Veridex, USA). 23 ml of peripheral blood were drawn after R0-resection of the primary tumor but before the start of adjuvant systemic treatment. After immunomagnetic enrichment with an anti-Epcam-antibody, cells were labelled with anti-Ck8/18/19 and anti-CD45 antibodies to distinguish between epithelial cells and leukocytes. Patients were followed for a median of 35 months. Univariate and multivariable proportional hazards models were estimated to assess the prognostic significance of CTC for disease-free and overall survival. Patients with evidence of at least 1 CTC were counted as positive. Results In 21.5% of patients (n = 435) CTC were detected before the start of systemic treatment (median 1.3, range 1 - 827). Patients with CTC before treatment were more frequently node-positive (P<0.001), but no correlation to tumor size, grading and HR-Status could be found. 114 recurrences occurred and 66 patients died of their disease. The presence of CTC before systemic treatment predicted poor disease-free (p < 0.0001), distant disease-free survival (p < 0.001) and overall survival (p = 0.0002). In multivariate analysis, detection of CTC before treatment was confirmed as independent predictor for both disease-free (HR 1.88) and overall survival (HR 1.91) next to tumor size, grading, lymph node involvement and hormone receptor status (p for all < 0.05). Outcome of patients was correlated to the number of CTC. Prognosis was worst in patients with 5 CTC or more with a four-fold increased risk for recurrence and and a three-fold increased risk for death (HR 4.04 for DFS and 3.05 for OAS; p < 0.05). This is the first study to prospectively demonstrate the prognostic relevance of CTC in peripheral blood of early breast cancer patients before the start of systemic treatment in a large patient cohort. CTC detection could serve as clinically useful prognostic marker and treatment monitoring tool and should be tested as indicator for secondary adjuvant treatment interventions within clinical trials. Citation Information: Cancer Res 2010;70(24 Suppl):Abstract nr S6-5.
Article
Using multiple genetic markers, including cancer stem-like cells, we evaluated the clinical significance of circulating tumor cells (CTCs) as a prognostic factor for overall survival (OS) and disease-free survival (DFS) in the peripheral blood (PB) of patients with colorectal cancer (CRC) who had undergone curative surgery. In a multi-institutional study, 735 patients with CRC were assigned to a retrospective training set (n = 420) or prospective validation set (n = 315). CTCs that expressed carcinoembryonic antigen (CEA), cytokeratin (CK) 19, CK20, and/or CD133 (CEA/CK/CD133) mRNA in PB were detected using real-time reverse transcription polymerase chain reaction assay. In the training sets, OS and DFS of patients who were positive for CEA/CK/CD133 were significantly worse than those of patients who were negative for these markers (P < .001). At each staging analysis, OS and DFS of patients with Dukes' stage B or C cancer who were positive for CEA/CK/CD133 were significantly worse than those of patients who were negative for these markers (P < .003 and P < .001 in Dukes' stage B; P < .001 in Dukes' stage C). In contrast, in patients with Dukes' stage A, no significant differences were seen between patients who were positive for these markers and those who were negative. Cox multivariate analysis demonstrated that CEA/CK/CD133 was a significant prognostic factor for OS (hazard ratio [HR], 3.84; 95% CI, 2.41 to 6.22; P < .001) and DFS (HR, 3.02; 95% CI, 1.83 to 5.00; P < .001). In particular, in patients with Dukes' stage B and C cancer, CEA/CK/CD133 demonstrated significant prognostic value. In validation sets, similar results were confirmed in patients with Dukes' stage B and C cancer. In patients with Dukes' stage B and C CRC who require adjuvant chemotherapy, detection of CEA/CK/CD133 mRNA in PB is a useful tool for determining which patients are at high risk for recurrence and poor prognosis.
Article
Circulating tumor cells (CTCs) can be separated and characterized from normal hematopoietic cellular constituents by a variety of methods. Different strategies have included separation by physical characteristics, such as size or weight, or by biological characteristics, such as expression of epithelial or cancer-specific markers. Of the latter, rtPCR for epithelial-related gene message, such as cytokeratin, and immunoseparation techniques using monoclonal antibodies against epithelial cellular adhesion molecule, have gained the most widespread use in investigational and standard clinical application to date. Detection and monitoring of CTCs might be useful for screening, prognosis, prediction of response to therapy, or monitoring clinical course in patients with primary or metastatic cancer. Currently, monitoring patients with metastatic disease is the most practical application of CTCs. In this regard, several studies have demonstrated that approximately 50-70% of patients with metastatic breast, colon, and prostate cancers have elevated CTC levels, when evaluated using a highly automated immunomagnetic CTC assay system, designated CellSearch®. These studies demonstrate that elevated CTC levels prior to initiation of a new systemic therapy are associated with a worse prognosis than those that do not, and that persistently elevated or subsequent rising CTC levels strongly suggest that the therapeutic regimen with which the patient is being treated is not working. Similar results have been shown with rtPCR assays, although they are not as widely available for routine clinical use. New areas of research are directed toward developing more sensitive means of CTC detection and generating a variety of methods to characterize the molecular and biologic nature of CTCs, such as the status of hormone receptors, epidermal, and other growth factor receptor family members, and indications of stem-cell characteristics.
Article
Ultrasensitive methods have been recently developed to detect circulating tumour cells (CTCs) in the peripheral blood and disseminated tumour cells (DTCs) in the bone marrow (BM) of cancer patients. Studies with these new methods indicate that BM is a common homing organ and a reservoir for DTCs derived from various organ sites including breast, prostate, lung and colon. Peripheral blood analyses, however, are more convenient for patients than invasive BM sampling and many research groups are currently assessing the clinical utility of CTCs for prognosis and monitoring response to systemic therapies. Moreover, molecular analyses of CTCs/DTCs have provided new insights into the biology of metastasis with important implications for the clinical management of cancer patients.