Вы находитесь на странице: 1из 16

Lung (2015) 193:157–171

DOI 10.1007/s00408-015-9697-7

REVIEW

Circulating Tumor Cells in Lung Cancer: Detection Methods


and Clinical Applications
Na Yu • Jia Zhou • Fang Cui • Xiaokui Tang

Received: 16 August 2014 / Accepted: 9 February 2015 / Published online: 19 February 2015
Ó Springer Science+Business Media New York 2015

Abstract Circulating tumor cells (CTCs) are tumor cells Despite undergoing standardized operative treatments,
that have disseminated from primary and metastatic sites, and many patients with early-stage lung cancer relapse be-
circulate in the bloodstream. Advanced immunological and cause of distant organ or tissue metastasis, presumably
molecular-based methods can be used to detect and analyze due to the undetected spread of the cancer elsewhere in
the cells with the characteristics of tumor cells, and can be the body at the time of initial treatment. Currently, there
detected and analyzed in the blood of cancer patients. The are no sensitive methods or clinical indexes to recognize
most commonly used methods in lung cancer combine the occult micrometastatic tumor cells in patients with lo-
processes of immunomagnetic enrichment and immunocy- calized lung cancer. In patients with late-stage lung can-
tochemical detection, morphology-based enrichment coupled cer, the survival time of this malignant disease is only
with reverse transcriptase polymerase chain reaction (RT- minimally prolonged by chemotherapy and/or targeted
PCR), and RT-PCR alone. CTC analysis is considered a liq- drug therapy. Analyzing the genotype and phenotype of
uid biopsy approach for early diagnosis, risk stratification, tumor cells may provide essential information that could
evaluation of curative efficacy, and early detection of lung prolong survival time and optimize the therapeutic regi-
cancer relapse. In this review, we discuss the present tech- men of lung cancer patients.
niques for analyzing CTCs, and the restrictions of using these Circulating tumor cells are tumor cells shed from
methods in lung cancer. We also review the clinical studies in primary and metastatic sites that circulate in the pe-
lung cancer and discuss the underlying associations between ripheral blood, and can be detected by many advanced
these studies and their future applications to this disease. immunological and molecular-based methods. The Cell-
SearchTM system is the only diagnostic assay approved
Keywords Circulating tumor cells  Detection methods  by the US Food and Drug Administration to monitor
Clinical significance  Lung cancer  Review patients with metastatic breast, colorectal, and prostate
cancers. In lung cancer patients, CTCs can be recognized
Introduction in the peripheral blood depending on the characteristics
of tumor cells and the advanced analysis techniques
Lung cancer is the most frequently diagnosed cancer and used. These cells are identified not only in patients with
the leading cause of cancer-related deaths worldwide. distant metastases, but also in patients with localized
early tumors. It is vitally important to develop and op-
timize CTC detection methods, and to summarize the
N. Yu  J. Zhou  X. Tang (&)
future management of malignant disease, especially with
Department of Respiratory Disease, The First Affiliated Hospital
of Chongqing Medical University, #1 Youyi Rd., Yu-zhong regard to risk stratification of adjuvant factors, real-time
District, Chongqing 400016, China monitoring of treatment efficacy, and the development of
e-mail: txk1200@126.com new targeted strategies and resistance mechanisms. This
review focuses on the current approaches for CTC de-
F. Cui
Department of Clinical Laboratory, The First Affiliated Hospital tection and analysis, and their clinical applications in
of Chongqing Medical University, Chongqing 400016, China lung cancer.

123
158 Lung (2015) 193:157–171

Techniques to Detect CTCs buoyant density, which results in the separation of different
cell types into distinct layers. Lighter particles including
CTC detection methods comprise the following two parts: tumor and mononuclear cells and plasma remain at the top,
enrichment (isolation) approaches (morphological and im- and heavier particles including erythrocytes and neu-
munological techniques) and detection (identification) ap- trophils migrate to the bottom. Density gradient separation
proaches (cytometric and nucleic acid techniques). The of CTCs has been performed in lung cancer using Ficoll–
advantages and disadvantages of the enrichment and de- Hypaque or Oncoquick methods to separate CTCs and
tection methods in lung cancer are discussed below and mononuclear cells from other blood cells. Compared to the
summarized in Table 1. The optimal method should be conventional Ficoll procedure, Oncoquick was developed
highly sensitive, reproducible, and easy to implement in a to avoid the cross contamination of different layers using a
clinical setting. porous membrane; it was also shown to have higher sen-
sitivity when used for the enrichment of lung cancer cells
[1]. RosetteSepTM (Stem Cell Technologies) uses negative
Enrichment Methods selection to isolate cells from whole blood, and as such,
that has improved specificity compared to selection using
Enrichment methods are based on the morphological traditional density gradient sedimentation. In this tech-
aspects of CTCs (size or density), or on the selection of nique, bispecific antibodies cross-link white blood cells to
tumor cells using immunoseparation techniques. Most of red blood cells to increase their density, thereby allowing
these methods include an initial enrichment step such as their removal by density gradient centrifugation [2].
density gradient centrifugation, size-based filtration, or
immunomagnetic separation to enhance the probability of Size-Based Cell Separation
rare cell detection.
Enrichment approaches in which cells are collected by
size-based filtration are also used. Most peripheral blood
Morphology-Based Enrichment Methods leukocytes range from 8 to 11 microns in size, and are in fact
smaller than CTCs derived from epithelial cancers. Thus,
Density-Based Cell Separation they can easily be removed from the sample by filtration
through a polycarbonate pore membrane. The most com-
The basis of density-based cell separation is the differential monly used membrane microfilter devices have been applied
migration of cells during centrifugation according to their using several different filtration-based approaches, such as

Table 1 Main advantages and disadvantages of techniques used for CTCs enrichment and detection in lung cancer
Technique Advantages Disadvantages

Enrichment
Immunomagnetic separation Semi-automatic magnetic separation method—easy to EpCAM antibody dependent. Difficult to extract pure
(CellSearch) use. Highly reproducibility. High specificity. Fully CTC for further analysis. Low sensitivity.
validated. FDA-approved method Expensive
Density gradient Simple method. No EpCAM antibody dependent. Poor enrichment. Low specificity and sensitivity.
centrifugation (Ficoll– Isolates mononuclear cells based on different May lost large number of CTCs in processing
Hypaque or OncoQuick) density gradient. Non-expensive
Size-based filtration (ISET) Isolates cells based on different size (CTC larger than Small tumor cells may be omitted. Low sensitivity
WBC). No EpCAM antibody dependent. Effective and specificity
method. Tumor cells could be used for the
subsequently analysis. Isolates CTM
Detection
Cytometric Able to assess cell enumeration, morphology and Detection depends on epithelial or tumor biomarker
characteristics. Cells also can be isolated for the expression. No specific CTC marker exists. False-
subsequently molecular characterization positive cells may be assessed. Low sensitivity
Nucleic acid based (RT-PCR) Antibody independent. High sensitivity and Difficult to produce. Unable to assess cell
specificity, particular in multimarker assay. enumeration, morphology and characteristics. The
Enrichment step is not necessary. Quantitative false-positive is high
analysis
CTC circulating tumor cell, EpCAM epithelial cell adhesion molecule, CTM circulating tumor microemboli, ISET isolation by size of epithelial
tumor cells

123
Lung (2015) 193:157–171 159

isolation by size of epithelial tumor cells (ISET) [3]. Among EpCAM are currently the most widely used markers for the
the direct methods used to detect CTCs, ISET technology detection of tumor cells of epithelial origin. Antibodies
allows the substantial enrichment of CTCs in lung cancer against epithelial-specific proteins can indicate if and how
[4]. Moreover, because these cells are available for cy- many CTCs are present, and leukocyte-specific antibodies
topathological analysis, this method may provide specificity can be used to avoid false positives. Staining can be per-
and allow the development of an immunocytochemical ap- formed using primary antibodies with detectable tags (such
proach for identifying different CTC phenotypes. as a fluorescent molecule), or with tagged secondary anti-
bodies that bind to the primary antibody. These labeling
Immunomagnetic Separation applications use either enzymatic color reactions or
fluorescence-based techniques for visualization with a light
One of the most widely applied techniques is immuno- or fluorescence microscope. Several immunofluorescence-
logical enrichment using specific markers that are ex- based techniques are currently used with the aim of for-
pressed on the surface of CTCs; it involves a magnetic mulating a cut-off value of detection. Automated scanning
immunobead and a ferrofluid-based system. There are two devices [automated cellular imaging system (ACIS)] sti-
features that are used in current immunological isolation mulate the detection of CTC while decreasing inter-ob-
procedures: epithelial cell-specific markers [epithelial cell server discrepancy, and include fiber-optic array scanning
adhesion molecule (EpCAM) and cytokeratins (CK)], technology, laser-scanning cytometer, and flow cytometry.
which are generally expressed by tumor cells of epithelial
origin, and tumor-specific markers, which are expressed by Nucleic Acid-Based Techniques
certain types of cancer [5]. These approaches use anti-
bodies that bind target proteins present at the cell surface, Free circulating DNA can be released from either the pri-
and encompass both positive and negative selection mary or metastatic tumors, or apoptotic CTCs. Thus, the
strategies. Due to the absence of strictly tumor-specific detection of circulating DNA in the plasma could only
antigens, epithelial-specific proteins such as EpCAM and mean the presence of circulating nucleic acids, and not
CK have been employed for the positive selection of CTCs, CTCs, suggesting that circulating DNA in the bloodstream
and antibodies against leukocyte-specific surface antigens does not definitely point to the existence of CTCs. An al-
such as CD45 have been used to deplete leukocytes from ternative nucleic acid-based strategy is detection of tumor-
the blood sample for negative selection. Immunomagnetic associated mRNA by reverse transcriptase polymerase
isolation is performed with monoclonal antibodies coupled chain reaction (RT-PCR), which is one of the currently
to magnetic microbeads; a strong permanent magnet is most widely used methods for detecting CTCs in patients
used to isolate CTCs from a leukocyte background by with lung cancer [6, 7]. Recently, several studies have used
magnetic force. RT-PCR and variations of this technique to detect CTCs in
lung cancer patients by assessing tumor-associated markers
(such as CEA mRNA, CK19 mRNA, LUNX mRNA) ex-
pression in their peripheral blood, and the detection rates
Detection range from 3.63 to 100 % (Table 2). The advantage of
RNA-based approaches for CTC detection is that the
CTC detection methods can be summarily classified into degradation of RNA released from cells in clinical samples
two groups: cytometric-based and nucleic acid-based ap- is very rapid; therefore, the origin of detectable RNA
proaches. Cytometric-based techniques use immunocyto- transcripts in a blood sample is considered to be viable
chemical approaches used to analyze and characterize tumor cells. Total RNA is extracted from cells by standard
different tumor cells; nucleic acid-based techniques detect methods, and transcribed into cDNA using reverse tran-
DNA or RNA sequences, which have distinct expression scriptase. Subsequently, PCR amplification is performed
patterns in tumor cells and normal blood components. with the cDNA using primers specific to the transcript of
interest.
Immunocytochemical Approaches To increase sensitivity, an additional PCR reaction with
several primers can be performed on the amplified product
In immunocytochemistry, the cells to be detected are at- of the first reaction (nested PCR). Both positive and
tached to a solid support to allow easy handling. After negative controls are used in these reactions. In addition,
fixation, the cells are coated with one or several antibodies. housekeeping genes, such as glyceraldehyde-3-phosphate
The choice of appropriate markers is a challenge, because dehydrogenase (GAPDH) and b-actin, are used as internal
antigens solely expressed by CTCs are very rare. Anti- controls to optimize the quality of RNA and to normalize
bodies specific to epithelial antigens such as CK and the expression levels of target genes.

123
Table 2 CTC enrichment and detection platforms in lung cancer
160

CTC CTC CTC detection CTC Tumor Blood Total CTC counts CTC detection Number of Ref
enrichment enrichment techniques detection stage volume number rate % controls

123
platforms markers markers (ml) of patients (% positivity)

CellSearch Veridex EpCAM CellSpotter analyzer CK 8/18/19, CD45 stages IIIB–IV 7.5 37 C2 per 7.5 ml blood 9/37 (24.32 %) NA [39]
CellSearch Veridex EpCAM Cell track Analyzer II cytokeratin, CD45 stage IV, 10 24 1–24 per 7.5 ml blood 8/24 (33.3 %) NA [32]
recurrence
CellSearch Veridex EpCAM CellTracks Analyzer II CK 8/18/19, CD45 Metastatic NSCLC 20 33 1–19 per 7.5 ml blood 12/33 (36.36 %) NA [40]
CellSearch Veridex EpCAM CellTracks technology cytokeratin, CD45 stage IIIA 7.5 9 C1 per 7.5 ml blood 1/9 (11.11 %) NA [45]
CellSearch Veridex EpCAM Cell track Analyzer II cytokeratin, CD45 SCLC, stages I–IV 7.5 125 C1 per 7.5 ml blood 38/125 (30.4 %) 3/25 (12.0 %) [22]
(florescent (6/9 SCLC, nonmalignant
microscope) 32/116 NSCLC) patients
CellSearch Veridex EpCAM CellSpotter Analyzer cytokeratin 8/18/ stage I 7.5 9 10 per 7.5 ml blood 1/9 (11.11 %) 0/4 healthy [20]
(fluorescent 19, CD45 (before surgery) volunteers
microscope) 1–4 per 7.5 ml blood 3/9 (33.33 %)
( immediately
after surgery)
0/9 (0 %) (10 days
after surgery)
CellSearch Veridex EpCAM CellTracks technology cytokeratin, CD45 advanced NSCLC 10 37 C1 per 7.5 ml blood 28/37 (75.68 %) NA [33]
CellSearch Veridex EpCAM Cell track Analyzer II cytokeratin, CD45 stages I–IV 7.5 30 1–16 per 7.5 ml blood 5/30 (16.67 %) NA [46]
(florescent
microscope)
CellSearch Veridex EpCAM CellTracks technology cytokeratin 8/18/ stage IV 7.5 21 1–21 per 7.5 ml blood 9/21 (42.86 %) NA [34]
19, CD45
CellSearch Veridex EpCAM CellTracks technology cytokeratin, CD45 SCLC 10 51 C2 per 7.5 ml blood 35/51 (68.6 %) NA [23]
(baseline)
49 C2 per 7.5 ml blood 13/49 (26.5 %)
(posttreatment)
37 C2 per 7.5 ml blood 25/37 (67.6 %)
(relapse)
49 C8 per 7.5 ml blood 7/49 (14.3 %)
(posttreatment)
37 C8 per 7.5 ml blood 13/37 (35.1 %)
(relapse)
CellSearch Veridex EpCAM CellTracks technology cytokeratin 8/18/ SCLC 7.5 97 C50 per 7.5 ml blood 41/97 (42.26 %) NA [24]
19, CD45 (baseline)
IMS CD45 ICC Cytokeratin 18, 19 Stages I–IV 7.5 47 C2 per 7.5 ml blood 40/47 (85.11 %) 1/18 (5.56 %) [41]
mRNA healthy donors
and 1/13
(7.69 %)
nonmalignant
pulmonary
tuberculosis
patients
Lung (2015) 193:157–171
Table 2 continued
CTC CTC CTC detection CTC Tumor Blood Total CTC counts CTC detection Number of Ref
enrichment enrichment techniques detection stage volume number rate % controls
platforms markers markers (ml) of (% positivity)
patients

IMS Cytokeratin 7/8 ICC Pan-cytokeratin Stages I–IV 4 51 NA 26/51 (51.0 %) (6/ 0/20 patients with [47]
18 stage I, 20/33 BLD and 0/20
Lung (2015) 193:157–171

stage II–IV healthy


NSCLC) volunteers
IMS Cytokeratin 7/8 ICC Pan-cytokeratin Stages I–IV 4 35 NA 15/35 (42.9 %) (5/ 0/40, 20 patients [48]
15 stage I, 10/20 with BLD and 20
stage II–IV healthy
NSCLC) volunteers
IMS CD45 Immunofluorescence CK 18 SCLC, stages I–IV 7.5 42 C3 per 7.5 ml blood 20/42 (47.62 %) 0/10 nonmaligant [5]
analysis pulmonary
disease patients
Density gradient Marker independent ICC (fluorescent CK 1/4/5/6/8/10/ stages I–IV 6 –10 71 [2 per 1 9 107 45/71 (63.38 %) NA [49]
centrifugation microscope) 13/18/19, CD45
Density gradient Pan-cytokeratin ICC Lunx and SP-A Stages I–IV 10 32 NA 21/32 65.63 %) 0/32, 16 patients [50]
centrifugation antibody with BLD and 16
and IMS healthy
volunteers
Density gradient CK7/8 antibody ICC CK7/8 Stages I–IV 10 55 1CTC per 5 9 107 29/55 (52.7 %) (7/ 0/50, 25 patients [15]
centrifugation mononuclear cells 21 stage I, 22/34 with BLD and 25
and IMS stage II–IV healthy
NSCLC) volunteers
Density gradient Marker independent RT-PCR Cytokeratin 19 Stages I–IV 10 55 NA 29/55 (52.7 %) (7/ 6/25 (24 %) [15]
centrifugation mRNA 21 stage I patients with
NSCLC, 22/34 BLD and 0/25
stage II–IV healthy
NSCLC) volunteers
LUNX mRNA Stages I–IV 31/55 (56.4 %) (8/ 5/25 (20 %)
21 stage I patients with
NSCLC, 23/34 BLD and 0/25
stage II–IV healthy
NSCLC) volunteers
Density gradient Marker independent Quantitative RT-PCR Survivin mRNA NA 5 68 NA 28/68 (41.18 %) 30 healthy [16]
centrifugation hTERT mRNA 42/68 (61.76 %) volunteers used
to define a cut-off
Cytokeratin-7 28/68 (41.18 %) value for the
mRNA normalization of
TTF-1 mRNA 24/68 (35.29 %) mRNA
combined 56/68 (82.35 %) (at expression
least one marker
positive)
161

123
Table 2 continued
162

CTC CTC CTC detection CTC Tumor Blood Total CTC counts CTC detection Number of Ref
enrichment enrichment techniques detection stage volume number rate % controls

123
platforms markers markers (ml) of (% positivity)
patients

Density gradient Marker Quantitative RT-PCR EpCAM, MUC1 Stage IA–IIIA 6 61 NA 24/61(39.3 %) 20 healthy [17]
centrifugation independent mRNA (preoperation) volunteers used
16/61(26.2 %) as a calibrator for
(postoperation) normalizing the
mRNA amounts
Density gradient Marker Quantitative RT-PCR Cytokeratin 19 NA 6 55 NA 52/55 (94.5 %) 4/40 (25 %) benign [18]
centrifugation independent mRNA (before the lung disease
initiation of
chemotherapy)
Density gradient Marker Quantitative RT-PCR LUNX mRNA Stages I–IIIA 6 68 NA 40/68 (58.8 %) 42 patients with [35]
centrifugation independent (preoperation) beninge lung
22/68 (32.4 %) diseases used to
(postoperation) set range, defined
as negative
60 20/60 (33.3 %)
(after the
completion of
adjuvant
chemotherapy )
Ficoll–Hypaque Marker ICC Pan-cytokeratin Stages I–III 10 45 The number of cells 11/45 (24.4 %) NA [51]
independent positive for
cytokeratins ranged
from 2 to 10
Ficoll gradient Marker RT-PCR hnRNP-B1 mRNA NA 10 18 NA 14/18 (77.8 %) 0/12 healthy [52]
centrifugation independent Her2/neu mRNA 8/18 (44.4 %) volunteers
Ficoll-Paque Marker RT-PCR Cytokeratin 19 Stages II–IV 5 15 NA 12/15 (80 %) NA [26]
independent mRNA (before
radiotherapy)
8/15 (53 %) (after
radiotherapy)
Ficoll–Hypaque EpCAM antibody Quantitative RT-PCR Cytokeratin 7 Stages IIIB–IV 8 46 NA 14/46 (30.4 %) 6/46 (13.0 %) [8]
and MACS mRNA healthy
volunteers
Cytokeratin 19 4/46 (8.7 %) 0/46 healthy
mRNA volunteers
EGP mRNA 14/46 (30.4 %) 2/46 (4.3 %)
healthy
volunteers
FN1 mRNA 7/46 (15.2 %) 0/46 healthy
volunteers
Lung (2015) 193:157–171
Table 2 continued
CTC CTC CTC detection CTC Tumor Blood Total CTC counts CTC detection Number of Ref
enrichment enrichment techniques detection stage volume number rate % controls
platforms markers markers (ml) of patients (% positivity)

OncoQuick Marker RT-PCR LUNX mRNA Stages I–IV 10 –20 24 NA 10/24 (41.7 %) (stage NA [1]
independent I–IV NSCLC)
MUC1 mRNA 5/24 (20.8 %) (stage I–
Lung (2015) 193:157–171

IV NSCLC)
Cytokeratin 19 5/24 (20.8 %) (stage I–
mRNA IV NSCLC)
combined 14/24 (58.3 %) (stage
I-IV NSCLC)
StemCell CD45, RT-PCR CLCA2, HMGB3, NA 10 108 (88 NA 49/108 (45.4 %) (at 0/25 healthy [29]
Technologies CD66b, L587S, ASH1 NSCLC least one marker volunteers
CD36 mRNA and 20 positive)
antibodies SCLC)
RosetteSep Human CD45, c - FAST CK 8/18 Stages I–IV 18 –20 17 9–485 per 10 ml blood 16/17 (94.12 %) NA [2]
H2AX
ISET Marker ICC vimentin, pan- Metastatic NSCLC 10 6 5–88 per 5 ml blood 6/6 (100 %) 0/6 healthy [3]
independent cytokeratin volunteers
RT-PCR marker RT-PCR CEA mRNA Stages I–IIIA 5 103 NA 62/103 (60.2 %) 0/15 patients with [25]
independent (preoperation) interstitial
pulmonary
fibrosis who
underwent an
open-lung biopsy
and 0/32 healthy
subjects served as
controls
RT-PCR Marker RT-PCR SCG3 mRNA SCLC, stages I–IV NA 111 NA 51/111 (45.95 %) (36/ NA [53]
independent 67 SCLC, 15/44
stages I–IV NSCLC)
RT-PCR Marker RT-PCR CEA mRNA Stage I NA 29 NA 11/29 (37.9 %) 0/32 healthy [36]
independent (preoperation, by volunteers
video-assisted
lobectomy ) (stage I
NSCLC)
21/29 (72.4 %) (during
and after operation,
by video-assisted
lobectomy)
57 NA 22/57 (38.6 %)
(preoperation, by
open lobectomy)
24/57 (42.1 %) (during
and after operation,
by open lobectomy)
163

123
Table 2 continued
164

CTC CTC CTC detection CTC Tumor Blood Total CTC counts CTC detection Number of Ref
enrichment enrichment techniques detection stage volume number rate % controls

123
platforms markers markers (ml) of patients (% positivity)

RT-PCR Marker RT-PCR Cytokeratin 19 SCLC, stages I–IV 3 125 (102 NA 57/125 (45.6 %) 5/71 (7 %) healthy [30]
independent mRNA NSCLC, volunteers
PTHrP mRNA 23 SCLC) 81/125 (64.8 %) 5/71 (7 %) healthy
volunteers
LUNX mRNA 35/125 (28 %) 4/71 (5.6 %)
healthy
volunteers
RT-PCR Marker RT-PCR EGFR mRNA Metastatic lung 5 30 NA 17/30 (56.7 %) 4/38 (10.5 %) [54]
independent cancer healthy
volunteers
RT-PCR Marker RT-PCR Cytokeratin 19 Stages I–IV 4 35 NA 20/35 (57.1 %) (7/15 8/20 (40 %) [48]
independent mRNA stage I, 13/20 stage patients with
II–IV NSCLC) BLD and 6/20
(30 %) healthy
volunteers
CEA mRNA 18/35 (51.4 %) (6/15 6/20 (30 %)
stage I, 12/20 stage patients with
II–IV NSCLC) BLD and 5/20
(25 %) healthy
volunteers
RT-PCR Marker RT-PCR CEA mRNA Stage I–IIIA NA 103 NA 62/103 (60.2 %) no healthy [37]
independent (preoperation) volunteers were
29/103 (28.2 %) positive
(postoperation)
Quantitative RT- Marker Quantitative RT-PCR Cytokeratin-7, NA 10 22 NA 22/22 (100 %) (at least 19 healthy [55]
PCR independent EGFR, SCCA, one marker positive) volunteers used
SFTPB mRNA to set range,
defined as
negative
Quantitative RT- Marker Quantitative RT-PCR KRT19 mRNA Stages I–III 2.5 55 NA 21/55 (38.18 %) 12 healthy [56]
PCR independent CEACAM5 mRNA 2/55 (3.63 %) volunteers used
to set range, at
DSG3 mRNA 2/55 (3.63 %) very low or
SFTPA mRNA 2/55 (3.63 %) nondetectable
SFTPC mRNA 2/55 (3.63 %) levels
combined 26/55 (47.27 %) (at
least one marker
positive)
Lung (2015) 193:157–171
Table 2 continued
CTC CTC CTC detection CTC Tumor Blood Total CTC counts CTC detection Number of Ref
enrichment enrichment techniques detection stage volume number rate % controls
platforms markers markers (ml) of (% positivity)
patients

Nested RT-PCR Marker Nested RT-PCR Cytokeratin 19 Stages I–IV 3 83 NA 30/83 (36.1 %) (5/30 15 healthy volunteers [57]
independent mRNA stage I–II, 25/53 used to set range,
Lung (2015) 193:157–171

stage III–IV NSCLC) defined as negative


Cytokeratin 20 34/83 (41.0 %) (8/30
mRNA stage I–II, 26/53
stage III–IV NSCLC)
CEA mRNA 40/83 (48.2 %) (12/30
stage I–II, 28/53
stage III–IV NSCLC)
combined 61/83 (73.5 %) (18/30
stage I–II, 43/53
stage III–IV NSCLC)
(at least one marker
positive)
Nested RT-PCR Marker Nested RT-PCR TTF-1 mRNA Stages I–III NA 61 NA 22/61 (36.1 %) NA [27]
independent (presurgery)
48 18/48 (37.5 %)
(postsurgery)
Cytokeratin 19 61 26/61 (42.6 %)
mRNA (presurgery)
48 12/48 (25.0 %)
(postsurgery)
Nested RT-PCR Marker Nested RT-PCR SP-D mRNA 26 metastatic and 5 63 NA 33/63 (52.4 %) (24/26 0/30, 15 patients with [58]
independent 37 without with metastases, 9/37 BLD and 15 healthy
metastatic lung without metastases) volunteers
cancer
Nested RT-PCR Marker Nested RT-PCR BJ-TSA-9 mRNA Stage I–IV NSCLC 5 120 NA 68/120 (56.7 %) 1/106 (0.94 %) patients [59]
independent with BLD and 0/80
healthy volunteers
Cytokeratin 19 69/120 (57.5 %) 7/106 (6.6 %) pateints
mRNA with BLD and 3/80
healthy volunteers
Pre-proGRP 42/120 (35.0 %) 6/106 (5.66 %) pateints
mRNA with BLD and 0/80
healthy volunteers
SCC mRNA 29/120 (24.2 %) 9/106 (8.49 %) pateints
with BLD and 3/80
(3.75 %) healthy
volunteers
LUNX mRNA 22/120 (18.3 %) 7/106 (6.6 %) pateints
with BLD and 1/80
(1.25 %) healthy
volunteers
165

123
166 Lung (2015) 193:157–171

[47]

ICC immunocytochemistry, ISET isolation by size of epithelial tumour cells, IMS immunomagnetic separation, MACS magnetic-activated cell sorting system, FAST fiber-optic array scanning
Ref
The sensitivities of these approaches can be evaluated
by analyzing the serial dilutions of a lung cancer cell line in
the peripheral blood of a healthy volunteer. With standard

with BLD and 0/20

with BLD and 0/20


4/20 (20 %) patients

2/20 (10 %) patients


healthy volunteers

healthy volunteers
methods, samples considered positive or negative are based
(% positivity)

on the presence or absence of a PCR product; further


Number of

quantitative analysis cannot be performed. However, the


controls

application of real-time RT-PCR methods now makes the


quantification of cell numbers in clinical samples possible
by comparison with standard curves [8]. Another advan-
stage I, 21/33 stage

stage I, 23/33 stage


28/51 (54.9 %) (7/18

31/51 (60.9 %) (8/18

tage of real-time PCR is that a normal range can be


II–IV NSCLC)

II–IV NSCLC)
CTC detection

established if the assay has been performed for several


controls. Subsequently, a cut-off value for positivity can be
rate %

recognized to improve the specificity of the test.


In addition, fluorescence in situ hybridization (FISH) is
another very common nucleic acid-based technique to ana-
lyze CTCs with prior enrichment for changes in individual
genes, including gene copy number, gene rearrangement, and/
CTC counts

or gene deletion [9, 10]. FISH has previously been considered


as a cogent technique to assess genomic mutations in the
clinic in primary and/or metastatic lesions [11]. This tech-
NA

nique can assess the genomic characteristics of individual


patients
number

CTCs with visual confirmation. After the enrichment step


Total

(ISET or EpCAM-independent immunomagnetic separation),


51
of

ALK gene rearrangement, chromosome 7 and chromosome 8


volume

centromere probes can be detected in CTCs of patients with


Blood

(ml)

NSCLC using FISH analyses, facilitating both diagnostic


4

testing and monitoring of target treatment [9, 10, 12, 13]. Due
to the results of FISH analysis that may not truly represent
cellular phenotype and/or target molecule expression at the
Stages I–IV

protein level, a combination of different detection methods


Tumor
stage

may improve the accuracy of testing results.

Methods for Detecting CTCs in Lung Cancer


Cytokeratin 19

LUNX mRNA
detection

mRNA
markers

Currently, the most widely used methods for detecting


CTC

CTCs in lung cancer combine the processes of immuno-


technology, BLD benign lung diseases, NA not available

magnetic enrichment and immunocytochemical detection


(CellSearchTM System), morphology-based enrichment (-
CTC detection

Nested RT-PCR

density-based cell separation and/or size based on cell


separation) coupled with RT-PCR, and RT-PCR alone
techniques

(Table 2). The CellSearchTM system is the only FDA-ap-


proved device for the enumeration of CTCs in the whole
blood of patients with breast, colorectal, and prostate
independent
enrichment

cancers. Compared to RT-PCR, the major advantages of


the CellSearchTM system are its semi-automation feature,
markers

Marker
CTC

and its high reproducibility, accuracy, linearity, and re-


liability [14]. Although these features are vital to any
Table 2 continued

method to ensure that the results can be validated by dif-


Nested RT-PCR

ferent investigators, such features have been lacking in


enrichment
platforms

previous techniques. Because of the low numbers of CTCs


present in circulating blood, their short half-life, and a
CTC

paucity of functional markers available for their

123
Lung (2015) 193:157–171 167

identification, the sensitivity of the CellSearchTM system is are significantly higher numbers of CTCs in cancer patients
very low. In small-cell lung cancer (SCLC) patients, the than in benign patients [22], which may be a useful char-
detection rates of the CellSearchTM system range from 14.3 acteristic for the early detection of lung cancer. Among
to 68.6 %; in non-small-cell lung cancer (NSCLC) pa- primary lung cancer patients, SCLC patients showed a
tients, the detection rates range from 0 to 75.7 % (Table 2). higher CTC enumeration than NSCLC [22–24]. When the
Tissue-specific mRNA may be present although protein- detection rate of CTC was compared with clinical stage,
based assays are negative, and RT-PCR is the most sensi- the higher stage of lung cancer had a higher enumeration of
tive method for detecting tumor-associated molecular CTC [22, 25]. Kurusu et al. [25] indicated that the preop-
markers. This sensitivity is remarkably higher than protein- erative blood samples of patients with clinical stages IIA,
based assays that involve less sensitive antibody analysis IIB, and IIIA disease (50, 73, and 100 %, respectively) had
methods. Paradoxically, RT-PCR is also limited by its a higher incidence of positive carcinoembryonic antigen
extremely high sensitivity. Sample contamination and il- (CEA) mRNA than those with stages IA and IB (41 and
legitimate transcription [low-level omnipresent transcrip- 36 %, respectively). However, some studies have indicated
tion of any gene in any cell type or pseudogenes (genes that that there is no significant difference between CTC status
lack intrinsic sequences)] may lead to false-positive PCR and clinical stages [26–28]. Wendel et al. [28] evaluated
amplification results that are indistinguishable from PCR the association between CTCs and clinical stage in blood
products derived from intact mRNA. In order to increase samples from chemotherapy-naı̈ve NSCLC patients using a
the specificity of RT-PCR in lung cancer, some researchers fluid phase biopsy approach and found no significant dif-
have applied density gradient centrifugation [15–18] or ference in CTC number among patients with stage IV,
immunomagnetic separation [8] to enrich epithelial cells. stage III, and stages I/II disease. The presence of CTC in
This enriched RT-PCR assay has a higher detection rate of the blood before therapy does not mean the existence of
CTCs than standard RT-PCR, which lacks an enrichment advanced or incurable disease; we should detect CTC se-
step (Table 2). The main advantages and disadvantages of rially to monitor individual treatment.
techniques used for CTC enrichment and detection in lung Early studies using RT-PCR analyses to detect known
cancer are listed in Table 1. biomarkers (e.g., survivin, hTERT, TTF-1, CK19, PTHrP,
LUNX, CEA mRNA) indicated that patients with
metastatic lung cancer had a significantly higher detection
Clinical Significance rate than primary lung cancer patients without distant
metastasis [16, 25, 29, 30]. Katseli et al. evaluated the
Large numbers of tumor cells may shed into the circula- association between the mRNA expression of CK19/
tion, even in the early stages of tumor formation [19]. Yet, PTHrP/LUNX and distant metastases [30]. The authors
only a minority of these dispersed cells can develop into found that detection of CK19 mRNA positively correlated
overt metastasis. The existence of CTCs in the bloodstream with metastasis to the lymph nodes, brain, bones, and
should not be considered as a definitive indicator of adrenal glands; detection of PTHrP mRNA closely corre-
metastatic disease. To successfully develop metastasis lated with the presence of bone metastasis; detection of
successfully, CTCs must arrest blood supply, extravasate LUNX mRNA correlated with lymph node involvement.
into the target organ parenchyma, and proliferate. Tight However, the sensitivity of RT-PCR analyses for predic-
vascular wall barriers, adverse living conditions in distant tion of distant metastasis was not 100 %, suggesting that
organs, and the rate-limiting acquisition of organ imaging examinations [whole-body positron emission to-
colonization functions are some of the impediments to mography (PET) scanning, brain magnetic resonance
formation of distant metastasis. These limited metastatic imaging, computed tomography (CT)] are still necessary,
factors could account for the observation that, despite the even when no CTCs are detected. The molecular evidence
fact that tumor cell shedding occurs during surgical re- of recurrence in lung cancer patients after different treat-
section of primary lung cancer, CTCs disappear from the ment regimens has been controversial, due to the fact that
circulation after surgery [20]. The detection and enu- CTCs can shed into the vasculature following treatment
meration of CTCs in lung cancer may serve as an important (surgery or radiation). Thus, more studies are needed to
biomarker of clinical stage, and prognosis and treatment monitor the early recurrence of lung cancer patients.
predictions [21].
Detection of Early Relapse
Detection of Lung Cancer and Distant Metastasis
Only a few studies have evaluated the association between
It is likely that the diagnostic value of CTC will play an CTC and early recurrence in patients with lung cancer. In a
important role in detecting lung cancer. For example, there preliminary study by Sawabata and colleagues [20],

123
168 Lung (2015) 193:157–171

peripheral blood from nine patients undergoing a lobecto- shown that although the median survival time tends to be
my with stage I (IA in 6 cases and IB in 3 cases) NSCLC shorter in CTC-positive patients than in CTC-negative
was evaluated using the CellSearchTM System. Peripheral patients, the difference was not significant [39, 40]. When
blood samples were collected both during and after the comparing the results from studies that used samples col-
thoracotomy. One patient was positive for CTC before lected before and after treatments, we found that the sur-
surgery, while three were positive immediately after sur- vival outcome was worse for patients with persistently
gery. However, 10 days after surgery, the CTCs disap- positive CTC at different time points [35–37]. In addition,
peared in all of these patients. Lung cancer cells may shed the survival outcome for patients at pre- and post-treatment
into the circulation during surgery, but due to the afore- time points with persistently negative CTC or for patients
mentioned living conditions, CTCs only persist for a short who converted from a CTC-positive to CTC-negative sta-
time. The small number of events presented in the study tus was associated with a longer OS and/or PFS [18]. Due
means that the results must be considered preliminary, and to the heterogeneity among these studies, further studies
validation is warranted. Nonetheless, the findings suggest with validated standard techniques should be performed to
the clinical significance of CTCs in lung cancer. Patients evaluate whether treatment-associated variation in CTCs
with undetectable levels of circulating lung cancer cells are related to the prognosis of lung cancer. If such a cor-
had a lower risk of recurrence and mortality; thus, it re- relation could be established, novel or established adjuvant
mains to be determined if these individuals have a better therapies could be supervised, and the dose and duration
prognosis, ignoring the influence of other factors, and could be individualized. Although the same method was
should have standard adjuvant therapy withdrawn. Based used to detect CTCs in lung cancer in the aforementioned
on these studies, patients with detectable levels of circu- studies, the thresholds (1, 2, 5, 8, 50/7.5 ml) and results
lating lung cancer cells may have a higher risk of recur- differed. This illustrates that the existence of CTCs alone
rence, and as such, should be offered adjuvant therapy, does not have prognostic significance, but rather, other
while ignoring other prognostic factors. factors are also important for predicting the outcome of
lung cancer patients, such as the experimental conditions
Prognostic Value of CTCs and epithelial-to-mesenchymal transition (EMT).

Many researchers have evaluated the prognostic value of Assessment of Therapy


CTCs in lung cancer; however, the conclusions of these
studies are controversial. A meta-analysis suggested that Some studies evaluated the correlation between CTC enu-
the appearance of CTCs in peripheral blood is associated meration and radiography (CT scanning or fluorodeoxyglu-
with poor prognosis in lung cancer patients [31]. Mean- cose-PET), and suggested that CTCs may be related to
while, many studies showed that patients with positive disease response rate [33, 41]. Wu et al. [41] conducted a
CTC had a significantly shorter survival time [overall small-scale study to evaluate the correlation between CTC
survival (OS) and/or progression-free survival (PFS)] than number and radiographic appearance (CT scan). CTC
those without [16–18, 23, 24, 27, 32–38]. Naito et al. [23] number was determined in 12 patients before and after 2
indicated that when using 8 CTCs per 7.5 ml blood as the courses of first-line chemotherapy, followed by CT ex-
threshold count, patients with C8 CTCs had a significantly amination. Eight of these patients were classified as having
shorter survival time than the remaining patients with \8 stable disease by CT scanning, and among those patients,
CTCs after therapy in SCLC. Hou et al. [24] analyzed the five patients had a reduced CTC count, two patients main-
association between CTC enumeration and prognosis of tained a CTC count of 0, and one patient had a slightly
patients with SCLC, and drew the same conclusion using increased CTC count from 2 to 3 after treatment. For partial
50 CTCs per 7.5 ml blood as the cut-off value. Krebs et al. response patients, the CTC count dropped markedly from 52
[38] investigated CTC levels before and after chemother- to 0 in one patient and another two patients had a CTC count
apy using the CellSearchTM System in NSCLC patients. that remained below 2. The only patient whose CTC enu-
When using 5 CTCs per 7.5 ml blood as the threshold meration dramatically increased from 2 to 12 after therapy
count, patients with \5 CTCs had a mean PFS of was confirmed to have progressive disease by CT scanning.
6.8 months, an OS of 8.1 months, and 29 % died in Punnoose and co-workers [33] also evaluated the association
6 months, whereas patients with C5 CTCs showed PFS of between CTC count and fluorodeoxyglucose-PET imaging
2.4 months, OS of 4.3 months, and 93 % died in 6 months. in patients with NSCLC after the combination of erlotinib
These data indicate that the number of CTCs may be and pertuzumab therapy. The authors found that patients
closely related to the prognosis of patients with NSCLC; who showed partial or complete responses had a trend to-
the higher the number of CTCs, the shorter the survival wards higher baseline CTC counts than patients with stable
time of NSCLC patients. However, other studies have disease or progressive disease. Meanwhile, the decreased

123
Lung (2015) 193:157–171 169

CTC counts upon treatment were significantly associated Conclusion and Future Perspectives
with disease response rate; though, one study showed that
CTC count was not associated with disease response rate Despite the presence of some ambiguous results, pre-
[40]. Due to the difference in results, more studies are liminary studies suggest that analyses of CTCs may be
needed to determine if there is indeed a correlation between important in the diagnosis and treatment of lung cancer [2,
CTC count and disease response. Using a standardized 9, 20, 23, 24, 36, 38, 42]. CTC analyses can be considered
imaging method to assess the therapeutic effect may im- liquid biopsies without invasive tissue biopsies. In addition,
prove the reliability of these results. analyses of CTCs may provide broad information for
clinical lung cancer management, including detection, di-
Mutation Analysis agnosis, and therapy monitoring. It is possible to determine
the changes in CTC number during therapy by repeated
Somatic mutations of cancer genes are considered re- blood sample analyses. However, there are also some
markable markers because they are cancer cell specific, and challenges in this field. In current studies, patients show
such genomic alterations are not affected by changes in high variability in sensitivity and specificity which is il-
protein marker expression. Recent studies showed that the lustrated by the heterogeneity of study populations and
mutations in the epidermal growth factor receptor (EGFR) techniques used. Current cytometric-based and nucleic
gene was concordant both in CTCs of blood samples and acid-based analytical techniques to detect circulating lung
matched primary tumor biopsies in patients with NSCLC cancer cells are highly sensitive; however, the absence of
[33, 42]; therefore, EGFR mutation analysis is essential to lung cancer-associated biomarkers might increase the
guide targeted therapy. Maheswaran et al. [42] evaluated false-positive results in patients with lung cancer. During
the presence of CTCs and EGFR mutations in patients with cancer invasion and metastasis, some cells undergo a
NSCLC. In total, 20 patients had known EGFR mutation morphological switch from EMT. When using epithelial-
status based on their tumor tissue specimens, the EGFR based methods to detect CTCs, false-negative results will
mutation status in CTCs was identical to the mutations in increase in patients with lung cancer [3]. In this respect,
patients’ tumor biopsies in 19 of 20 cases (95 %). T790 M detecting markers of EMT (cytokeratins, E-cadherin, vi-
resistance mutations were detected in two of six patients mentin, neural cadherin) may complement existing ep-
(33 %) who had a response to EGFR tyrosinase kinase ithelial-based methods, and may improve the positive
inhibitor (TKI) therapy and in 9 of 14 patients (64 %) who detection rate of lung cancer. Due to the abovementioned
had progression on EGFR tyrosine kinase inhibitor therapy. reasons, the clinical applications of CTCs have been lim-
Meanwhile, Punnose et al. [33] also analyzed CTCs for ited in lung cancer.
EGFR mutational status by quantitative PCR; despite the An animal model study suggests that multiple mechan-
very low sensitivity, the detected mutational status in CTCs isms associated with tumor cell detachment, adhesion, and
was concordant with tumor biopsies. This indicates that the suppression of immunogenicity are responsible for the
detection of CTCs and EGFR mutations may play an im- ability of CTC to enter and survive in the circulation [43];
portant role in improving the treatment regimens of lung it is essential for the formation of distant metastases and to
cancer. predict recurrent or latent disease; another study indicates
Crizotinib, the ALK inhibitor, is becoming cumulatively that natural killer (NK) cells play an important role in
important in targeted therapies for NSCLC patients; thus, it preventing metastasis formation because they are able to
is necessary to evaluate the presence of ALK gene rear- decelerate the growth of the primary tumor and kill most of
rangement in these patients. Pailler et al. [9] evaluated 18 those CTCs in the blood [44]; it may be useful to improve
ALK-positive and 14 ALK-negative patients with NSCLC and characterize individual treatment of cancer patients.
using a filtration enrichment technique and filter-adapted The abovementioned data about animal model may provide
fluorescent in situ hybridization. All ALK-positive patients with clues for the future study of CTC in lung cancer. In
had four or more ALK-rearranged CTCs per 1 ml of blood, order to improve the clinical value of CTC investigations in
whereas no or only one ALK-rearranged CTC was detected the future, many more studies with standard techniques and
in ALK-negative patients. ALK-rearranged CTCs were investigations on animal models are needed to establish the
detected in five patients during crizotinib treatment. The clinical significance of CTC in patients with lung cancer.
levels of CTCs harboring ALK rearrangement and single
ALK native copies decreased in four patients during the Acknowledgments This work was supported by Chongqing Sci-
ence and Technology Commission (cstc2013yykfA10002). The re-
treatment. Evaluating the presence of ALK gene rear- cipient of this Grant is Xiaokui Tang.
rangements in CTCs may provide new insights into
monitoring quantitative and qualitative changes in patients Conflicts of interest No potential conflicts of interest were
with NSCLC undergoing crizotinib therapy. disclosed.

123
170 Lung (2015) 193:157–171

References 20. Sawabata N et al (2007) Circulating tumor cells in peripheral


blood caused by surgical manipulation of non-small-cell lung
1. Mitas M et al (2003) Lunx is a superior molecular marker for cancer: pilot study using an immunocytology method. Gen
detection of non-small cell lung cancer in peripheral blood. J Mol Thorac Cardiovasc Surg 55(5):189–192
Diagn 5(4):237–242 21. Truini A et al (2014) Clinical applications of circulating tumor
2. Martin OA et al (2014) Mobilization of Viable Tumor Cells Into cells in lung cancer patients by Cell Search System. Front Oncol
the Circulation During Radiation Therapy. Int J Radiat Oncol 4:242
Biol Phys 88(2):395–403 22. Tanaka F et al (2009) Circulating tumor cell as a diagnostic marker
3. Lecharpentier A et al (2011) Detection of circulating tumour cells in primary lung cancer. Clin Cancer Res 15(22):6980–6986
with a hybrid (epithelial/mesenchymal) phenotype in patients 23. Naito T et al (2012) Prognostic impact of circulating tumor cells
with metastatic non-small cell lung cancer. Br J Cancer 105(9): in patients with small cell lung cancer. J Thorac Oncol 7(3):
1338–1341 512–519
4. Vona G et al (2000) Isolation by size of epithelial tumor cells : a 24. Hou JM et al (2012) Clinical significance and molecular char-
new method for the immunomorphological and molecular char- acteristics of circulating tumor cells and circulating tumor mi-
acterization of circulating tumor cells. Am J Pathol 156(1):57–63 croemboli in patients with small-cell lung cancer. J Clin Oncol
5. Chen Q et al (2013) Lung cancer circulating tumor cells isolated 30(5):525–532
by the EpCAM-independent enrichment strategy correlate with 25. Kurusu Y, Yamashita J, Ogawa M (1999) Detection of circulating
Cytokeratin 19-derived CYFRA21-1 and pathological staging. tumor cells by reverse transcriptase-polymerase chain reaction in
Clin Chim Acta 419:57–61 patients with resectable non-small-cell lung cancer. Surgery
6. Nezos A et al (2009) Molecular markers detecting circulating 126(5):820–826
melanoma cells by reverse transcription polymerase chain reac- 26. Ge M et al (2005) Fluctuation of circulating tumor cells in pa-
tion: methodological pitfalls and clinical relevance. Clin Chem tients with lung cancer by real-time fluorescent quantitative-PCR
Lab Med 47(1):1–11 approach before and after radiotherapy. J Cancer Res Ther
7. Armakolas A et al (2010) Detection of the circulating tumor cells 1(4):221–226
in cancer patients. Future Oncol 6(12):1849–1856 27. Yoon SO et al (2011) TTF-1 mRNA-positive circulating tumor
8. Devriese LA et al (2012) Circulating tumor cell detection in cells in the peripheral blood predict poor prognosis in surgically
advanced non-small cell lung cancer patients by multi-marker resected non-small cell lung cancer patients. Lung Cancer
QPCR analysis. Lung Cancer 75(2):242–247 71(2):209–216
9. Pailler E et al (2013) Detection of circulating tumor cells har- 28. Wendel M et al (2012) Fluid biopsy for circulating tumor cell
boring a unique ALK rearrangement in ALK-positive non-small- identification in patients with early-and late-stage non-small cell
cell lung cancer. J Clin Oncol 31(18):2273–2281 lung cancer: a glimpse into lung cancer biology. Phys Biol
10. Ilie M et al (2012) ALK-gene rearrangement: a comparative 9(1):016005
analysis on circulating tumour cells and tumour tissue from pa- 29. Hayes DC et al (2006) Multigene real-time PCR detection of
tients with lung adenocarcinoma. Ann Oncol 23(11):2907–2913 circulating tumor cells in peripheral blood of lung cancer patients.
11. Wolff AC et al (2007) American Society of Clinical Oncology/ Anticancer Res 26(2B):1567–1575
College of American Pathologists guideline recommendations for 30. Katseli A et al (2013) Multiplex PCR-based detection of circu-
human epidermal growth factor receptor 2 testing in breast can- lating tumor cells in lung cancer patients using CK19, PTHrP,
cer. J Clin Oncol 25(1):118–145 and LUNX specific primers. Clin Lung Cancer 14(5):513–520
12. Chen YY, Xu GB (2014) Effect of circulating tumor cells com- 31. Ma XL et al (2012) Meta-analysis of circulating tumor cells as a
bined with negative enrichment and CD45-FISH identification in prognostic marker in lung cancer. Asian Pac J Cancer Prev
diagnosis, therapy monitoring and prognosis of primary lung 13(4):1137–1144
cancer. Med Oncol 31(12):240 32. Isobe K et al (2012) Clinical significance of circulating tumor
13. Ning N et al (2014) Improvement of specific detection of circu- cells and free DNA in non-small cell lung cancer. Anticancer Res
lating tumor cells using combined CD45 staining and fluores- 32(8):3339–3344
cence in situ hybridization. Clin Chim Acta 433:69–75 33. Punnoose EA et al (2012) Evaluation of circulating tumor cells
14. Allard WJ et al (2004) Tumor cells circulate in the peripheral and circulating tumor DNA in non-small cell lung cancer: asso-
blood of all major carcinomas but not in healthy subjects or ciation with clinical endpoints in a phase II clinical trial of per-
patients with nonmalignant diseases. Clin Cancer Res tuzumab and erlotinib. Clin Cancer Res 18(8):2391–2401
10(20):6897–6904 34. Nemunaitis J et al (2009) Phase II trial of Belagenpumatucel-L, a
15. Li Q et al (2011) Detection of micrometastases in peripheral TGF-beta2 antisense gene modified allogeneic tumor vaccine in
blood of non-small cell lung cancer with a refined immuno- advanced non small cell lung cancer (NSCLC) patients. Cancer
magnetic nanoparticle enrichment assay. Int J Nanomed Gene Ther 16(8):620–624
6:2175–2181 35. Li J et al (2014) LUNX mRNA-positive cells at different time
16. Yu Y et al (2013) Combination of four gene markers to detect points predict prognosis in patients with surgically resected
circulating tumor cells in the peripheral blood of patients with nonsmall cell lung cancer. Transl Res 163(1):27–35
advanced lung adenocarcinoma using real-time PCR. Oncol Lett 36. Yamashita JI et al (2000) Detection of circulating tumor cells in
5(4):1400–1406 patients with non-small cell lung cancer undergoing lobectomy
17. Zhu WF et al (2014) Prognostic value of EpCAM/MUC1 mRNA- by video-assisted thoracic surgery: a potential hazard for intra-
positive cells in non-small cell lung cancer patients. Tumour Biol operative hematogenous tumor cell dissemination. J Thorac
35(2):1211–1219 Cardiovasc Surg 119(5):899–905
18. Shi WL et al (2013) CK-19 mRNA-positive cells in peripheral 37. Yamashita J et al (2002) Preoperative evidence of circulating
blood predict treatment efficacy and survival in small-cell lung tumor cells by means of reverse transcriptase-polymerase chain
cancer patients. Med Oncol 30(4):755 reaction for carcinoembryonic antigen messenger RNA is an in-
19. Pantel K, Brakenhoff RH (2004) Dissecting the metastatic cas- dependent predictor of survival in non-small cell lung cancer: a
cade. Nat Rev Cancer 4(6):448–456 prospective study. J Thorac Cardiovasc Surg 124(2):299–305

123
Lung (2015) 193:157–171 171

38. Krebs MG et al (2011) Evaluation and prognostic significance of 49. Nair VS et al (2013) An observational study of circulating tumor
circulating tumor cells in patients with non-small-cell lung can- cells and (18)F-FDG PET uptake in patients with treatment-naive
cer. J Clin Oncol 29(12):1556–1563 non-small cell lung cancer. PLoS One 8(7):e67733
39. Juan O et al (2014) Prognostic significance of circulating tumor 50. Wang Y et al (2012) Detection of micrometastases in lung cancer
cells in advanced non-small cell lung cancer patients treated with with magnetic nanoparticles and quantum dots. Int J Nanomed
docetaxel and gemcitabine. Clin Transl Oncol 16(7):637–643 7:2315–2324
40. Hirose T et al (2012) Relationship of circulating tumor cells to the 51. Pirozzi G et al (2013) Prognostic value of cancer stem cells,
effectiveness of cytotoxic chemotherapy in patients with metastatic epithelial-mesenchymal transition and circulating tumor cells in
non-small-cell lung cancer. Oncol Res 20(2–3):131–137 lung cancer. Oncol Rep 29(5):1763–1768
41. Wu C et al (2009) Preliminary investigation of the clinical sig- 52. Fleischhacker M et al (2001) Detection of amplifiable messenger
nificance of detecting circulating tumor cells enriched from lung RNA in the serum of patients with lung cancer. Ann N. Y. Acad
cancer patients. J Thorac Oncol 4(1):30–36 Sci 945:179–188
42. Maheswaran S et al (2008) Detection of mutations in EGFR in 53. Moss AC et al (2009) SCG3 transcript in peripheral blood is a
circulating lung-cancer cells. N Engl J Med 359(4):366–377 prognostic biomarker for REST-deficient small cell lung cancer.
43. Howard EW et al (2008) Decreased adhesiveness, resistance to Clin Cancer Res 15(1):274–283
anoikis and suppression of GRP94 are integral to the survival of 54. De Luca A et al (2000) Detection of circulating tumor cells in
circulating tumor cells in prostate cancer. Clin Exp Metastasis carcinoma patients by a novel epidermal growth factor receptor
25(5):497–508 reverse transcription-PCR assay. Clin Cancer Res 6(4):1439–1444
44. Brodbeck T et al (2014) Perforin-dependent direct cytotoxicity in 55. Xi L et al (2007) Optimal markers for real-time quantitative re-
natural killer cells induces considerable knockdown of sponta- verse transcription PCR detection of circulating tumor cells from
neous lung metastases and computer modelling-proven tumor cell melanoma, breast, colon, esophageal, head and neck, and lung
dormancy in a HT29 human colon cancer xenograft mouse cancers. Clin Chem 53(7):1206–1215
model. Mol Cancer 13:244 56. Nordgard O et al (2013) Novel molecular tumor cell markers in
45. Tarumi S et al (2013) Innovative method using circulating tumor regional lymph nodes and blood samples from patients under-
cells for prediction of the effects of induction therapy on locally going surgery for non-small cell lung cancer. PLoS One 8(5):
advanced non-small cell lung cancer. J Cardiothorac Surg 8:175 e62153
46. Okumura Y et al (2009) Circulating tumor cells in pulmonary 57. Guo Y, Wang J, Huang P (2009) Clinical significance of CK20,
venous blood of primary lung cancer patients. Ann Thorac Surg CK19, CEA mRNAs in peripheral blood from lung cancer pa-
87(6):1669–1675 tients. Zhongguo Fei Ai Za Zhi 12(9):1013–1017
47. Huang TH et al (2007) Clinical significance of enrichment and 58. Qi ZL et al (2002) Expression and clinical significance of sur-
detection of circulating tumor cells in NSCLC patients with im- factant protein D mRNA in peripheral blood of lung cancer pa-
munomagnetic beads. Zhonghua Zhong Liu Za Zhi 29(9): tients. Ai Zheng 21(7):772–775
676–680 59. Qu YM et al (2010) Clinical significance of expressions of tumor
48. Wang XG, Hu H, Liu YN (2004) Detection of circulating tumor markers in peripheral blood in non-small cell lung cancer.
cells by nano-immunomagnetic enrichment in early lung cancer. Zhonghua Yi Xue Za Zhi 90(28):1958–1962
Zhonghua Yi Xue Za Zhi 84(16):1393–1395

123
Reproduced with permission of the copyright owner. Further reproduction prohibited without
permission.

Вам также может понравиться