ArticlePDF Available

Concordance of anaplastic lymphoma kinase (ALK) gene rearrangements between circulating tumor cells and tumor in non-small cell lung cancer

Authors:

Abstract and Figures

Anaplastic lymphoma kinase (ALK) gene rearrangement in non-small cell lung cancer (NSCLC) is routinely evaluated by fluorescent in-situ hybridization (FISH) testing on biopsy tissues. Testing can be challenging however, when suitable tissue samples are unavailable. We examined the relevance of circulating tumor cells (CTC) as a surrogate for biopsy-based FISH testing. We assessed paired tumor and CTC samples from patients with ALK rearranged lung cancer (n = 14), ALK-negative lung cancer (n = 12), and healthy controls (n = 5) to derive discriminant CTC counts, and to compare ALK rearrangement patterns. Blood samples were enriched for CTCs to be used for ALK FISH testing. ALK-positive CTCs counts were higher in ALK-positive NSCLC patients (3-15 cells/1.88 mL of blood) compared with ALK-negative NSCLC patients and healthy donors (0-2 cells/1.88 mL of blood). The latter range was validated as the 'false positive' cutoff for ALK FISH testing of CTCs. ALK FISH signal patterns observed on tumor biopsies were recapitulated in CTCs in all cases. Sequential CTC counts in an index case of lung cancer with no evaluable tumor tissue treated with crizotinib showed six, three and eleven ALK-positive CTCs per 1.88 mL blood at baseline, partial response and post-progression time points, respectively. Furthermore, ALK FISH rearrangement suggestive of gene copy number increase was observed in CTCs following progression. Recapitulation of ALK rearrangement patterns in the tumor on CTCs, suggested that CTCs might be used to complement tissue-based ALK testing in NSCLC to guide ALK-targeted therapy when suitable tissue biopsy samples are unavailable for testing.
Content may be subject to copyright.
Oncotarget23251
www.impactjournals.com/oncotarget
www.impactjournals.com/oncotarget/ Oncotarget, Vol. 7, No. 17
Concordance of anaplastic lymphoma kinase (ALK) gene
rearrangements between circulating tumor cells and tumor in
non-small cell lung cancer
Chye Ling Tan1,*, Tse Hui Lim1,*, Tony KH Lim1, Daniel Shao-Weng Tan2, Yong Wei
Chua1, Mei Kim Ang2, Brendan Pang3, Chwee Teck Lim4,5, Angela Takano1, Alvin
Soon-Tiong Lim1, Man Chun Leong6 and Wan-Teck Lim2,7
1 Department of Pathology, Singapore General Hospital, Singapore
2 Department of Medical Oncology, National Cancer Center Singapore, Singapore
3 Department of Molecular Oncology, National University Health System Singapore, Singapore
4 Faculty of Engineering, Department of Biomedical Engineering, National University of Singapore, Singapore
5 Mechanobiology Institute, National University of Singapore, Singapore
6 Clearbridge Biomedics Pte Ltd, Singapore
7 Institute of Molecular and Cell Biology, Singapore
* These authors have contributed equally to this work
Correspondence to: Wan-Teck Lim, email: dmolwt@nccs.com.sg
Keywords: ALK-gene rearrangement, circulating tumor cells, uorescent in-situ hybridization, lung cancer, molecular diagnosis
Received: November 14, 2015 Accepted: February 28, 2016 Published: March 16, 2016
ABSTRACT
Anaplastic lymphoma kinase (ALK) gene rearrangement in non-small cell lung
cancer (NSCLC) is routinely evaluated by uorescent in-situ hybridization (FISH)
testing on biopsy tissues. Testing can be challenging however, when suitable tissue
samples are unavailable. We examined the relevance of circulating tumor cells (CTC)
as a surrogate for biopsy-based FISH testing. We assessed paired tumor and CTC
samples from patients with ALK rearranged lung cancer (n = 14), ALK-negative lung
cancer (n = 12), and healthy controls (n = 5) to derive discriminant CTC counts,
and to compare ALK rearrangement patterns. Blood samples were enriched for
CTCs to be used for ALK FISH testing. ALK-positive CTCs counts were higher in ALK-
positive NSCLC patients (3–15 cells/1.88 mL of blood) compared with ALK-negative
NSCLC patients and healthy donors (0–2 cells/1.88 mL of blood). The latter range
was validated as the ‘false positive’ cutoff for ALK FISH testing of CTCs. ALK FISH
signal patterns observed on tumor biopsies were recapitulated in CTCs in all cases.
Sequential CTC counts in an index case of lung cancer with no evaluable tumor tissue
treated with crizotinib showed six, three and eleven ALK-positive CTCs per 1.88 mL
blood at baseline, partial response and post-progression time points, respectively.
Furthermore, ALK FISH rearrangement suggestive of gene copy number increase
was observed in CTCs following progression. Recapitulation of ALK rearrangement
patterns in the tumor on CTCs, suggested that CTCs might be used to complement
tissue-based ALK testing in NSCLC to guide ALK-targeted therapy when suitable tissue
biopsy samples are unavailable for testing.
INTRODUCTION
Lung cancer accounts for about 13% of all cancer
diagnoses and remains the leading cause of death by
cancer in the world [1], with almost 70% of patients
diagnosed with locally advanced or metastatic disease at
presentation [1, 2]. Non-small cell lung cancer (NSCLC)
accounts for approximately 85% of all cases of lung
cancer and is associated with poor prognosis [2]. The
5-year overall survival rate for NSCLC across all stages
is only 21% and is even lower (~5%) for stages IIIB and
IV [1, 3].
Oncotarget23252
www.impactjournals.com/oncotarget
Oncogenic ‘driver mutations’ have now been
identied in various subsets of NSCLC [4-6]. Of these
drivers, somatic mutations in epidermal growth factor
receptor (EGFR) and anaplastic lymphoma kinase
(ALK) [5-7] are the most frequently described. In 2007,
researchers identied the presence of a chimeric ALK
protein with broblast-transforming properties that was
formed following fusion of the echinoderm microtubule-
associated protein-like 4 (EML4) and ALK genes [6].
EML4-ALK subverts intracellular signaling pathways to
promote tumor cell survival and growth [8]. The overall
incidence of ALK gene rearrangement in NSCLC ranges
between 0.4% and 13.4%, and is similar in both Asian and
Western populations [9]. This discovery resulted in the
accelerated development and approval by the U.S. Food
and Drug Administration (FDA) of the ALK-targeting
tyrosine kinase inhibitors (TKIs) crizotinib (Xalkori®,
Pzer, New York, USA) in 2011, and ceritinib (Zykadia™,
Novartis, Basel, Switzerland) in 2014 to treat patients with
metastatic NSCLC who express the abnormal ALK gene
[10, 11].
The true therapeutic benet of novel molecules
targeting the mutant ALK fusion protein in NSCLC relies
on identifying the right patient population for treatment,
and on detecting the emergence of tumor resistance. The
American Society of Clinical Oncology (ASCO) endorsed
the joint College of American Pathologists (CAP)/
International Association for the Study of Lung Cancer
(IASLC)/Association for Molecular Pathology (AMP)
clinical practice guideline on EGFR and ALK molecular
testing for patients with lung cancer, which holds that an
ALK uorescent in-situ hybridization (FISH) assay using
dual-labeled break-apart probes is the preferred testing
methodology to detect ALK gene rearrangement [12].
The accuracy of testing nonetheless depends on the
quality of tumor biopsies. Approximately 50% of NSCLC
patients who undergo re-biopsy for determination of
resistance after rst-line chemotherapy have insufcient/
non-diagnostic biopsy specimens or cytology samples
available for molecular testing [13]. Biopsy is invasive and
repeating the procedure is not always feasible due to safety
concerns and general unwillingness of patients, among
other reasons [14]. Furthermore, lung adenocarcinomas
are heterogeneous with a diverse and ever-evolving
genetic and epigenetic makeup that contributes towards
treatment resistance [15]. These barriers to biopsy
collectively pose a challenge to track oncogene activity in
real-time over the course of treatment. There is a need for
a minimally invasive assay for tumor molecular proling
and continuous treatment monitoring in order to provide
timely and tailored cancer treatment.
Circulating tumor cells (CTCs) released from
the primary tumor site into the circulation represent a
potential means of non-invasively isolating tumor cells for
ALK FISH testing and other molecular characterizations.
Recent data supports the role of these renegade cells as
seeds of cancer metastases [16, 17]. They may recapitulate
the phenotypic heterogeneity and molecular signatures of
the primary tumor, as well as that of metastatic lesions
[18-20]. While their presence and prevalence in blood are
often associated with poor prognosis [21], CTCs may hold
further relevance as an alternative tumor source, which
can complement existing tissue-based diagnostic tests,
especially when biopsy material is absent or inadequate. In
patients with lung adenocarcinoma, hypothesis-generating
studies have strongly suggested that ALK status could be
determined based on testing of CTCs, with comparable
results as testing of tumor tissues [22, 23].
The key challenge of CTC-based testing is
the enrichment and isolation of these cells within an
acceptable timeframe. Various technical approaches
have been used to isolate these CTCs [19, 24, 25].
They can be broadly categorized into antibody afnity-
based, imaging-based and size-based techniques [26].
The only current US FDA-approved CTC capturing
technology utilizes EpCAM immunomagnetic means to
isolate EpCAM-positive CTCs for prognostic purposes
and would inadvertently miss out on EpCAM-negative
CTCs [27, 28]. As a predictive biomarker for treatment
monitoring and molecular analysis, it is pertinent to ensure
reliable and reproducible isolation of CTCs of different
phenotypic and molecular subtypes for various pre-and
post-treated patient cohorts [25]. An increasing body
of evidence suggests that non-immunomagnetic-based
CTC technologies can reliably retrieve a comprehensive
population of CTCs for molecular subtyping [19, 28].
Here, we evaluated the feasibility of an antibody-
independent CTC isolation system using lung
adenocarcinomas that have been tested ALK positive as a
model to examine the concordance patterns between CTCs
and tumor tissue, and to determine whether CTCs were
reproducibly detectable in circulation. We further explored
the potential use of CTCs in lung cancer, as a surrogate
for molecular testing of the primary tumor for ALK gene
rearrangement.
RESULTS
Study group
We prospectively recruited 27, mostly late-stage
NSCLC patients, 14 of whom had ALK-rearranged and
12 had wild-type ALK, determined from the initial biopsy
diagnoses. One patient in the cohort, who was not from
Singapore, had an unknown ALK status due to incomplete
referral records. Sixty percent of patients were males. All
ALK-positive patients were non-smokers. Five healthy
donors (three males and two females), aged between 18-
55 years old with no history of cancer, were also recruited
into the control cohort. As ALK translocation in NSCLC
Oncotarget23253
www.impactjournals.com/oncotarget
patients is strongly correlated with a non-smoker or
light smoker status [29], these healthy donors were non-
smokers. The clinicopathologic features of the study group
are summarized in Table 1.
Histopathological analysis of tumor tissues
In the ALK-positive group, three out of 14 tumor
tissues exhibited morphology that is associated with ALK
rearrangements [30]. Histological preparations showed
solid adenocarcinoma with signet ring cells (Figure
1A and 1B), and cribrifom adenocarcinoma with focal
squamoid cells (not shown). Immunohistochemical (IHC)
studies showed strong and diffuse nuclear reaction for
Thyroid Transcription Factor-1 (TTF-1). This nding
conrmed the diagnosis of adenocarcinoma of lung origin
in this particular setting (Figure 1C). Some tumors also
showed focal reaction to periodic acid-Schiff with diastase
(PAS-D) within mucin vacuoles, which is a general
feature of adenocarcinomas, as opposed to squamous cell
carcinomas (Figure 1D) [30].
Concordance in ALK rearrangement pattern
between CTCs and tumor
Following FISH testing on all tumor samples in the
cohort, it was found that ALK-positive tumors harbored
ALK rearrangements with various patterns of abnormality
(Table 2). The majority of the tumor samples harbored
the one fusion (F) and one split orange (R) and green (G)
signal (Figure 2A). The tumor from Patient P5 presented
various ALK rearrangement patterns such as 1F1R1G,
2F1R, 2F2R, 1F1R and 1F2R.
FISH testing was subsequently performed on CTCs
that were enriched and isolated from the matched blood
samples. Data showed that ALK rearrangement patterns
(majority 1F1R1G) observed in primary tumor tissues
were recapitulated on most of the ALK-positive CTCs,
giving an overall concordance rate of over 90% based on
the 1F1R1G fusion pattern (Table 2). In Patient P5 (Table
2), the CTCs were able to recapitulate three out of ve
ALK rearrangement patterns observed in the tumor tissue.
We further observed an overexpression of vimentin
in the tumor samples, along with the control bronchiolar
epithelium (Figure 2B). However, loss of E-cadherin was
not obvious in these samples.
The number of ALK-positive rearranged CTCs
Table 1: Clinicopathological characteristics of patients enrolled in this study
Patient characteristics Cases (%)
N = 27
Age, years 32–76
Sex
Male 16 (59.3%)
Female 11 (40.7%)
Smoking history
Non-smoker 16 (59.3%)
Smoker 5 (18.5%)
Ex-smoker 5 (18.5%)
No info 1 (3.7%)
Clinical staging
IB 1 (3.7%)
IIIA 1 (3.7%)
IIIB 2 (7.4%)
IV 23 (85.2%)
Histological subtype
ALK-positive 14 (51.9%)
Adenocarcinoma (NSCLC) 11 (40.7%)
Unknown subtype (NSCLC) 3 (11.1%)
ALK-negative 12 (44.4%)
Adenocarcinoma (NSCLC) 5 (18.5%)
Unknown subtype (NSCLC) 7 (25.9%)
ALK status unknown 1 (3.7%)
Adenocarcinoma (NSCLC) 1 (3.7%)
Abbreviations: ALK, anaplastic lymphoma kinase; NSCLC, non-small cell lung cancer
Oncotarget23254
www.impactjournals.com/oncotarget
Table 2: Concordance of ALK rearrangement patterns between CTC and tumor in patients with ALK-positive NSCLC
Case number of
patients with ALK-
positive NSCLC
ALK rearranged/ total
cells scored (Tumor)
ALK rearrangement patterns
(% of tumor cells observed with respective patterns)
Tumor CTC
P1 61/100 1F1R1G (100%) 1F1R1G (100%)
P2 45/100 1F1R1G (100%) 1F1R1G (100%)
P3 79/100 1F1R1G (100%) 1F1R1G (100%)
P4 30/100 1F1R1G (100%) 1F1R1G (100%)
P5 61/100
1F1R1G (4.9%)
2F1R (34.4%)
2F2R (3.3%)
1F1R (49.2%)
1F2R (8.2%)
1F1R1G (50%)
2F1R (37.5%)
2F2R (12.5%)
P6 72/100 1F1R1G (29.2%)
1F1R (70.8%) 1F1R1G (100%)
P7 81/100 1F1R (100%) 1F1R (75%)
1F1R1G (25%)
P8 55/100 1F1R1G (100%) 1F1R1G (100%)
P9 77/100 1F1R (31.2%)
1F1R1G (68.8%) 1F1R (100%)
P10 100/100 1F1R (100%) 1F1R (50%)
1F1R1G (50%)
P11 62/100 1F1R1G (100%) 1F1R1G (100%)
P12 77/100 1F1R1G (100%) 1F1R1G (100%)
P13 45/100 1F1R1G (100%) 1F1R1G (100%)
P14 Not available Not available 1F1R1G (100%)
Abbreviations: ALK, anaplastic lymphoma kinase; CTC, circulating tumor cells; NSCLC, non-small cell lung cancer.
Figure 1: Representative appearance of NSCLC adenocarcinoma with signet ring cells features. A. H&E stain showing
solid nests of tumor cells B. Solid with signet ring cells (arrow ) C. Thyroid transcription factor-1 (TTF-1) IHC stain showing strong nuclear
reaction in the signet ring cells. D. Solid tumor showing focal positive reaction for (PAS-D) within mucin vacuoles (arrow).
Oncotarget23255
www.impactjournals.com/oncotarget
Figure 2: High concordance of ALK FISH rearrangements patterns between CTCs and tumors in NSCLC
adenocarcinoma patients. A. Representative ALK FISH rearrangement patterns in CTCs and tumors showing 1F1R1G rearrangement
patterns. Yellow, red and green arrows represent fusion (F), orange (R) and green (G) uorescent signals. B. Representative vimentin (upper
panel) and E-cadherin (lower panel) IHC in tumor and control bronchiolar epithelium (black arrow).
Oncotarget23256
www.impactjournals.com/oncotarget
retrieved from ALK-positive patients was signicantly
enriched compared with ALK-negative patients (p <
0.0001) and healthy donors (p = 0.0003) (Figure 3).
Establishment and validation of ALK break-apart
probes cutoff in ALK-negative samples
ALK testing by FISH in NSCLC tumor tissues
without ALK rearrangement may detect rearrangement-
positive patterns (i.e. split patterns or isolated 3’
patterns) in a fraction of cells [31-33], likely because of
truncation artefact caused by tissue sectioning, or perhaps
a stochastic genomic alteration that does not indicate a
specic gene fusion. ALK FISH testing in formalin-xed,
parafn-embedded (FFPE) NSCLC tumor tissues has a
‘false positive’ cutoff value of 15% to allow for the best
separation between ALK-rearranged and ALK wild-type
cells [31, 33]. However, it is not possible to apply this
guideline in the ALK FISH testing on CTCs because the
number of CTCs in any given blood sample would be too
low.
In our study, we established and validated the ‘false
positive’ cutoff for ALK FISH in CTCs using 12 blood
samples from NSCLC ALK-negative patients and ve
blood samples from healthy donors (Supplementary Data).
Results from the ALK-negative NSCLC cohort scored a
median of two or less positive cells (range 0-2 cells/1.88
mL blood). The result concurred with the numbers
observed for healthy blood samples. In fact, no statistical
difference in ALK-positive cell counts was observed
between ALK-negative NSCLC cohort and healthy donors
(p = 0.0973) (Figure 3). This data established the ‘false-
positive’ cutoff for ALK break-apart probes in CTCs at
two cells per 1.88 mL blood.
Potential clinical applications
Sequential CTC enumeration and FISH was
performed on blood samples from a patient with no
accessible tissue for ALK FISH testing. The index case
was a never smoker male diagnosed with NSCLC. A
transthoracic needle aspiration biopsy was performed on
the right hilar mass to obtain a specimen for histological
analysis. The hematoxylin and eosin (H&E) stain
showed one small cluster of NSCLC cells with strong
nuclear reaction for TTF-1 favoring adenocarcinoma.
Unfortunately, his diagnostic tissue was exhausted and no
further molecular proling could be performed. He did not
Figure 3: Number of cells with ALK rearrangements in ALK-positive NSCLC patients is signicantly higher compared
to ALK-negative and healthy donors. Graph represents statistical analyses of the data on Table S1 using the non-parametric two-
tailed t-test. NS represents not signicant while p value <0.05 were considered signicant.
Oncotarget23257
www.impactjournals.com/oncotarget
respond to EGFR tyrosine kinase inhibitor (TKI) therapy
and had a short duration of response to pemetrexed and
cisplatin. Re-biopsy of the lung and liver tumors was
considered but the patient declined due to the risk of
bleeding. He consented to blood sampling instead; the
sample was subsequently processed as described in the
Methods section.
At baseline, six CTCs displaying a 1F1R1G
pattern were isolated and met the necessary cutoffs for
ALK-positivity (Figure 4A). A trial of crizotinib was
commenced. Conrmatory scans done 3 months after
completion of treatment demonstrated good partial
response in the liver and minor response in the primary
lung tumor, based on RECIST criteria (Figure 4A).
Sequential CTC counts dropped to three cells displaying
the similar ALK rearranged pattern as the baseline. He
continued on crizotinib but unfortunately, his disease
progressed in the liver and the brain 5 months after
treatment initiation (Figure 4A). A post-progression blood
sample showed additional ALK rearrangement patterns
present in his CTCs, which differed from the baseline
patterns. New ALK rearrangement patterns such as 2R2G
and 1F1R appeared, in addition to 1F1R1G, which was
previously present (Figure 4B). The number of ALK-
positive CTCs also increased from three to eleven CTCs
per 1.88 mL of blood post-progression.
DISCUSSION
We successfully captured CTCs using an antibody-
independent CTC isolation system. CTCs were enriched
from the blood samples collected from 27 NSCLC
patients, 14 of whom were ALK-positive. Three of
the cases exhibited solid with signet ring cells pattern
associated with ALK positivity [34-37]. Overall, CTCs
isolated from the ALK-positive patient cohort were above
detectable levels, even among previously treated patients.
The presence of ALK rearrangement in CTCs was
previously analyzed and reported by French groups using
the Isolation by Size of Epithelial Tumor (ISET) system,
Figure 4: An index case suggests that ALK-rearranged CTCs could have clinical application as a diagnostic biomarker
to monitor crizotinib treatment and response. A. CT scan taken at baseline, partial response and progression time points showing
presence of metastatic tumor in liver (arrow). CTC counts and ALK rearrangement patterns for each time point is indicated in the lower
panel. B. Representative images showing 1F1R1G, 2R2G and 1F1R ALK rearrangement patterns following progression on crizotinib
treatment. Yellow, red and green arrows represent fusion (F), orange (R) and green (G) uorescent signals.
Oncotarget23258
www.impactjournals.com/oncotarget
which is also an antibody-free system (Rarecells, Paris,
France) [22, 23]. They characterized 18 ALK-positive
lung tumor and CTC samples showing high concordance
in ALK rearrangement among European patient cohort.
In agreement with our data reported in this study, ‘false-
positive’ signals were similarly observed on CTCs
from ALK-negative samples and a 4 cell/mL cutoff was
eventually established [22, 38].
In a similarly designed study, Pailler et al. [22]
described high concordance in ALK rearrangement
patterns between the CTC and tumor biopsies in 18 ALK-
positive and 14 ALK-negative patients with metastatic
NSCLC. This percentage of concordance is in agreement
with our own results. They reported that all ALK-positive
NSCLC patients in their cohort had 4 or more ALK-
rearranged CTCs per mL of blood. The study did not
include healthy donors to establish a ‘false positive’ cutoff
for ALK FISH testing of CTCs. They further reported that
CTCs harboring the 1F1R1G ALK rearrangement patterns
is associated with epithelial-mesenchymal transition
(EMT) phenotype [22].
Another study had reported that the EMT phenotype
(represented by loss of E-cadherin and expression of
vimentin) was more common in ALK-rearranged tumors
than other genotypes (38.9%, 19.1%, 26.9% and 14.6% of
ALK-rearranged, EGFR-mutated, K-ras mutated and triple
negative tumors, respectively; p = 0.015) [39]. Separately,
expression of vimentin alone was detected in 49.30%
of ALK-rearranged tumors while loss of E-cadherin was
detected in 71.30% [39]. In our study, we also observed
an overexpression of vimentin in the tumor samples in
comparison with the control bronchiolar epithelial tissue.
However, the loss of E-cadherin was not obvious in our
tumor samples, which suggested that some of the tumor
cells retained their epithelial characteristics within a
heterogeneous population of cells. The predominance of
this particular ALK rearrangement pattern in our patient’s
CTCs is therefore consistent with the observation above
suggesting that these tumors and their CTCs may be
favoring the EMT pathway.
This study further highlights the utility of antibody-
independent microuidic isolation systems for the
isolation and downstream characterization of CTCs
compared with immunomagnetic antibody-dependent
systems. While the numbers of CTCs isolated here are
small and may present substrate limitations to downstream
characterization of CTC, it should be noted that the current
numbers were derived from <2 mL of blood, as opposed to
existing systems which use up to 7.5 mL of blood or more.
In addition, we have previously demonstrated that there is
an association between CTC number and the volume of
blood processed [19]. This suggests a limitation that can
be easily overcome.
The index case presented here raises the possibility
that CTC enumeration based on ALK FISH may be
associated with treatment response with crizotinib by
imaging. The appearance of additional ALK rearrangement
patterns following progression with crizotinib treatment
exhibited a double split in both ALK alleles giving rise to
the 2R2G ALK rearrangement pattern. The additional copy
of the oncogenic ALK may have contributed to disease
progression despite treatment with an ALK inhibitor. This
observation is worthy of further inquiry, because while
the presence of ALK copy number gain is correlated with
crizotinib resistance, as previously reported by Doebele
et al. [40], and in vitro studies have identied potential
resistance mutations in the ALK gene, for example
L1196M, G1269A, S1206Y and G1202R [40, 41], limited
analysis of post-progression biopsies of tumors from a
phase 1 study of LDK378 suggested that these secondary
resistance mutations or gene amplication do not account
for a majority of resistance cases [11]. Hence, further work
with paired re-biopsies and sequential CTC collection may
assist understanding of resistance mechanisms in ALK-
driven tumors.
Conclusions drawn from our study are limited by
its relative small patient population, as was Pallier’s study
[22]. Nonetheless, the converging trend of both studies’
ndings is indicative of the utility and potential of CTCs
as an alternate target of ALK testing in lung cancer and
informs the development of CTC-based technology. More
importantly, these studies provide the basis for subsequent,
large-scale validation studies.
In summary, high concordance of ALK
rearrangement patterns in CTCs and tumors as assessed by
ALK FISH testing indicates that CTCs may have utility as
a non-invasive surrogate diagnostic tool and may be useful
in the longitudinal follow-up for resistance proling. The
availability of a non-invasive tool would improve efforts
to guide ALK-rearranged targeted treatment in NSCLC,
especially in cases without tissue availability. Further
efforts at downstream CTC characterization and culture
following enrichment are ongoing.
MATERIALS AND METHODS
Patient recruitment and blood samples
Patients with conrmed NSCLC were recruited into
this trial. They were naïve for ALK-targeted TKI treatment,
but may have received other forms of chemotherapy. Once
informed consent was secured from these patients, their
blood samples were processed for CTC analysis. The
clinical sample collection protocols were reviewed and
approved by SingHealth Centralised Institutional Review
Board. Clinicopathological information was also recorded
for these patients. Blood samples from healthy donors
were used as controls in this study.
Oncotarget23259
www.impactjournals.com/oncotarget
Histopathology
H&E was done using the Leica ST5010 XL
automated stainer (Leica Biosystems, Wetzlar, Germany)
while periodic PAS-D staining was done using the Ventana
BenchMark Special Stains automated stainer (Ventana
Medical Systems Inc, Tucson, Arizona, USA), following
the respective standard protocols. Histological diagnoses
were made based on the World Health Organization
(WHO) classication [42]. IHC labeling was performed
on the Ventana BenchMark Ultra autostainer (Ventana
Medical Systems Inc, Tucson, Arizona, USA) using
the UltraView detection kit and proprietary Standard
CC1 (SC1) pre-treatment sets. The antibodies used with
their dilution and pre-treatments were as follows: TTF-
1 (Novacastra NCL-TTF-1, clone SPT24, SC1, dilution
1:30), vimentin (DAKO M0725, clone V9, SC1, 1:100)
and E-cadherin (Dako M3612, clone MCH-38, SC1,
1:30) antibodies. Histopathology data was reviewed by
pathologists who had been accredited by the College of
American Pathologists (CAP).
CTC enrichment
Peripheral blood was collected using K2 EDTA
vacutainer® blood collection tube (BD, Singapore)
and processed within 24 hours. Subsequently, 7.5 mL
of whole blood was incubated with red blood cell
(RBC) lysis buffer (G-Biosciences, USA) according
to manufacturer’s recommendations. Lysed RBCs in
the supernatant were discarded after centrifugation.
Remaining cell pellet containing CTCs was resuspended
in ClearCell
®
resuspension buffer prior to CTC enrichment
using the ClearCell® FX system (Clearbridge BioMedics,
Singapore), according to manufacturer’s instruction.
The ClearCell® FX system is an automated CTC
enrichment system driven by the CTChip® FR1, a
microuidic biochip to isolate CTCs based on size,
deformability and inertia. The isolation principle takes
advantage of the inherent Dean vortex ows present in
curvilinear channels for CTC enrichment, termed Dean
Flow Fractionation (DFF) [43]. The enriched CTC sample
output was equally divided into four portions.
Fluorescent in-situ hybridization
Four µm thick FFPE tumor tissue sections were
mounted on positively charged slides and deparafnized.
FISH was subsequently performed using the US
FDA-approved Vysis ALK Break Apart FISH Probe Kit
(Abbott Molecular, Abbott Park, Des Plaines, IL, USA).
The 5’ ALK probe was labeled with SpectrumGreen™
(G) and the 3’ ALK probe with SpectrumOrange™ (R).
ALK FISH for FFPE tissues were considered positive if
at least 15 % of the tumor cells showed abnormal break
apart signals as detailed in the IVD Vysis ALK Break
Apart FISH Probe Kit and by Camidge et al. [33]. A cell is
interpreted as having a split pattern (ALK-positive) when
the 5’ (G) and 3’ (R) signals are separated by two or more
signal diameters. Cells lacking both uorescent signals
were not evaluated.
One portion (one-quarter) of the enriched CTCs
was xed in Shandon CytoSpinTM Collection Fluid
(ThermoFisher Scientic, USA) overnight at 4°C. The
sample was deposited onto positively charged glass slides
by cytospin (800 rpm, 5 mins). All the cells on the slides
were analyzed for the ALK break-apart signal at 1000X
magnication. The scorers analyzing the ALK break-apart
signal on CTCs were blinded to the ALK rearrangement
patterns on the tumor samples, as well as whether the cell
isolated was from patients or healthy controls.
The remaining three portions of the enriched CTCs
were stored at 4°C under validated conditions for future
molecular testing.
Data analysis
Statistical analyses of the data were performed in
GraphPad Prism version 5.00 (GraphPad Software, San
Diego, CA, USA). A non-parametric two-tailed, t-test
(Mann-Whitney) was used for computing statistical
signicances. p value of less than 0.05 were considered
signicant.
ACKNOWLEDGMENTS
The authors wish to acknowledge Clearbridge
BioMedics, Singapore for providing the CTC capturing
technology and technical support. The authors would also
like to express sincere gratitude to patients and healthy
donors for participating in this study.
FUNDING
This study was supported by National Medical
Research Council grants (CSA040/2012 and CS-
IRG1225/2009).
CONFLICTS OF INTEREST
Chwee Teck Lim is an advisor with Clearbridge
Biomedics, Singapore. Man Chun Leong is an employee
of Clearbridge Biomedics, Singapore. All other authors
declare that they do not have any conicts.
Oncotarget23260
www.impactjournals.com/oncotarget
Editorial note
This paper has been accepted based in part on peer-
review conducted by another journal and the authors’
response and revisions as well as expedited peer-review
in Oncotarget.
REFERENCES
1. American Cancer Society. Cancer Facts & Figures 2015.
Atlanta: American Cancer Society; 2015. Accessed on
12 October 2015. Available from: http://www.cancer.org/
research/cancerfactsstatistics/cancerfactsgures2015/index
2. Molina JR, Yang P, Cassivi SD, Schild SE, Adjei AA.
Non-small cell lung cancer: epidemiology, risk factors,
treatment, and survivorship. Mayo Clin Proc. 2008; 83:
584-594.
3. Non-small cell lung cancer survival rate by stage. Am.
Cancer Soc. Atlanta; 2015. Accessed on 14 March 2015.
Available from: http://www.cancer.org/cancer/lungcancer-
non-smallcell/detailedguide/non-small-cell-lung-cancer-
survival-rates
4. Lira ME, Kim TM, Huang D, Deng S, Koh Y, Jang B, Go
H, Lee SH, Chung DH, Kim WH, Schoenmakers EFPM,
Choi Y La, Park K, et al. Multiplexed gene expression and
fusion transcript analysis to detect ALK fusions in lung
cancer. J Mol Diagn. 2013; 15: 51-61.
5. Paez JG, Jänne PA, Lee JC, Tracy S, Greulich H, Gabriel
S, Herman P, Kaye FJ, Lindeman N, Boggon TJ, Naoki K,
Sasaki H, Fujii Y, et al. EGFR mutations in lung cancer:
correlation with clinical response to getinib therapy.
Science. 2004; 304: 1497-500.
6. Soda M, Choi YL, Enomoto M, Takada S, Yamashita
Y, Ishikawa S, Fujiwara S, Watanabe H, Kurashina
K, Hatanaka H, Bando M, Ohno S, Ishikawa Y, et al.
Identication of the transforming EML4-ALK fusion gene
in non-small-cell lung cancer. Nature. 2007; 448: 561-566.
7. Sequist L V, Lynch TJ. EGFR tyrosine kinase inhibitors in
lung cancer: an evolving story. Annu Rev Med. 2008; 59:
429-442.
8. Roskoski R Jr. Anaplastic lymphoma kinase (ALK):
structure, oncogenic activation, and pharmacological
inhibition. Pharmacol Res. 2013; 68: 68-94.
9. Solomon B, Varella-Garcia M, Camidge DR. ALK gene
rearrangements: a new therapeutic target in a molecularly
dened subset of non-small cell lung cancer. J Thorac
Oncol. 2009; 4: 1450-1454.
10. Forde PM, Rudin CM. Crizotinib in the treatment of non-
small-cell lung cancer. Expert Opin Pharmacother. 2012;
13: 1195-1201.
11. Shaw AT, Kim D-W, Mehra R, Tan DSW, Felip E, Chow
LQM, Camidge DR, Vansteenkiste J, Sharma S, De Pas
T, Riely GJ, Solomon BJ, Wolf J, et al. Ceritinib in ALK-
rearranged non-small-cell lung cancer. N Engl J Med. 2014;
370: 1189-1197.
12. Lindeman NI, Cagle PT, Beasley MB, Chitale DA, Dacic
S, Giaccone G, Jenkins RB, Kwiatkowski DJ, Saldivar
JS, Squire J, Thunnissen E, Ladanyi M. Molecular testing
guideline for selection of lung cancer patients for EGFR and
ALK tyrosine kinase inhibitors: guideline from the College
of American Pathologists, International Association for
the Study of Lung Cancer, and Association for Molecular
Pathology. J Thorac Oncol. 2013; 8: 823-859.
13. Reck M, Hermes A, Tan E-H, Felip E, Klughammer B,
Baselga J. Tissue sampling in lung cancer: a review in light
of the MERIT experience. Lung Cancer. 2011; 74: 1-6.
14. Chouaid C, Dujon C, Do P, Monnet I, Madroszyk A, Le
Caer H, Auliac JB, Berard H, Thomas P, Lena H, Robinet
G, Baize N, Bizieux-Thaminy A, et al. Feasibility and
clinical impact of re-biopsy in advanced non small-cell
lung cancer: a prospective multicenter study in a real-world
setting (GFPC study 12-01). Lung Cancer. 2014; 86: 170-
173.
15. De Bruin E, McGranahan N, Yates L, Jamal-Hanjani
M, Salm M, Mitter R, Sha S, Murugaesu N, Rowan
A, Gerlinger M, Wedge D, Horswell S, Varela I, et al.
Intratumor heterogeneity in non-small cell lung cancer
inferred by multi-region exome sequencing. [abstract]. In:
Proceedings of the 105th Annual Meeting of the American
Association for Cancer Research; 2014 Apr 5-9; San Diego,
CA. Philadelphia (PA): AACR; Cancer Res 2014; 74(19
Suppl): Abstract nr 983.
16. Fidler IJ. The pathogenesis of cancer metastasis: the “seed
and soil” hypothesis revisited. Nat Rev Cancer. 2003; 3:
453-458.
17. Mendoza M, Khanna C. Revisiting the seed and soil in
cancer metastasis. Int J Biochem Cell Biol. 2009; 41: 1452-
1462.
18. Alix-Panabier̀es C, Pantel K. Circulating tumor cells: liquid
biopsy of cancer. Clin Chem. 2013; 59: 110-118.
19. Khoo BL, Warkiani ME, Tan DSW, Bhagat AAS, Irwin
D, Lau DP, Lim AST, Lim KH, Krisna SS, Lim WT, Yap
YS, Lee SC, Soo RA, et al. Clinical validation of an ultra
high-throughput spiral microuidics for the detection and
enrichment of viable circulating tumor cells. PLoS One.
2014; 9: e99409.
20. Bednarz-Knoll N, Alix-Panabières C, Pantel K. Clinical
relevance and biology of circulating tumor cells. Breast
Cancer Res. 2011; 13: 228.
21. Müller V, Riethdorf S, Rack B, Janni W, Fasching PA,
Solomayer E, Aktas B, Kasimir-Bauer S, Pantel K, Fehm
T. Prognostic impact of circulating tumor cells assessed
with the CellSearch SystemTM and AdnaTest BreastTM
in metastatic breast cancer patients: the DETECT study.
Breast Cancer Res. 2012; 14: R118.
22. Pailler E, Adam J, Barthélémy A, Oulhen M, Auger N,
Valent A, Borget I, Planchard D, Taylor M, André F, Soria
JC, Vielh P, Besse B, et al. Detection of circulating tumor
Oncotarget23261
www.impactjournals.com/oncotarget
cells harboring a unique ALK rearrangement in ALK-
positive non-small-cell lung cancer. J Clin Oncol. 2013; 31:
2273-2281.
23. Ilie M, Long E, Butori C, Hofman V, Coelle C, Mauro
V, Zahaf K, Marquette CH, Mouroux J, Paterlini-Bréchot
P, Hofman P. ALK-gene rearrangement: a comparative
analysis on circulating tumour cells and tumour tissue from
patients with lung adenocarcinoma. Ann Oncol. 2012; 23:
2907-2913.
24. Naoe M, Ogawa Y, Morita J, Omori K, Takeshita K,
Shichijyo T, Okumura T, Igarashi A, Yanaihara A,
Iwamoto S, Fukagai T, Miyazaki A, Yoshida H. Detection
of circulating urothelial cancer cells in the blood using the
CellSearch system. Cancer. 2007; 109: 1439-1445.
25. Vona G, Sabile A, Louha M, Sitruk V, Romana S, Schütze
K, Capron F, Franco D, Pazzagli M, Vekemans M, Lacour
B, Bréchot C, Paterlini-Bréchot P. Isolation by size of
epithelial tumor cells. Am J Pathol. 2000; 156: 57-63.
26. Yu M, Stott S, Toner M, Maheswaran S, Haber DA.
Circulating tumor cells: approaches to isolation and
characterization. J Cell Biol. 2011; 192: 373-382.
27. Chinen LTD, de Carvalho FM, Rocha BMM, Aguiar CM,
Abdallah EA, Campanha D, Mingues NB, de Oliveira TB,
Maciel MS, Cervantes GM, Dettino ALA, Soares FA,
Paterlini-Bréchot P, et al. Cytokeratin-based CTC counting
unrelated to clinical follow up. J Thorac Dis. 2013; 5: 593-
599.
28. Hofman V, Ilie MI, Long E, Selva E, Bonnetaud C, Molina
T, Vénissac N, Mouroux J, Vielh P, Hofman P. Detection
of circulating tumor cells as a prognostic factor in patients
undergoing radical surgery for non-small-cell lung
carcinoma: comparison of the efcacy of the CellSearch
AssayTM and the isolation by size of epithelial tumor cell
method. Int J Cancer. 2011; 129: 1651-1660.
29. Yang P, Kulig K, Boland JM, Erickson-Johnson MR,
Oliveira AM, Wamper J, Jatoi A, Deschamps C, Marks
R, Fortner C, Stoddard S, Nichols F, Molina J, et al. Worse
disease-free survival in never-smokers with ALK+ lung
adenocarcinoma. J Thorac Oncol. 2012; 7: 90-97.
30. Srodon M, Westra WH. Immunohistochemical staining for
thyroid transcription factor-1: a helpful aid in discerning
primary site of tumor origin in patients with brain
metastases. Hum Pathol. 2002; 33: 642-645.
31. Yoshida A, Tsuta K, Nitta H, Hatanaka Y, Asamura H,
Sekine I, Grogan TM, Fukayama M, Shibata T, Furuta K,
Kohno T, Tsuda H. Bright-eld dual-color chromogenic in
situ hybridization for diagnosing echinoderm microtubule-
associated protein-like 4-anaplastic lymphoma kinase-
positive lung adenocarcinomas. J Thorac Oncol. 2011; 6:
1677-1686.
32. Yoshida, A, Varella-Garcia M. Fluorescence in situ
Hybridization (FISH). In: Tsao, Ming Sound, Hirsch, Fred
R YY, editor. IASLC ATLAS ALK Test Lung Cancer. 10th
ed. Internation Association for the study of Lung Cancer,
Aurora,Co; 2013. page 23-4. Accessed on 12 October 2015.
Available from: https://www.iaslc.org/publications/iaslc-
atlas-alk-testing-lung-cancer
33. Camidge DR, Kono SA, Flacco A, Tan AC, Doebele
RC, Zhou Q, Crino L, Franklin WA, Varella-Garcia M.
Optimizing the detection of lung cancer patients harboring
anaplastic lymphoma kinase (ALK) gene rearrangements
potentially suitable for ALK inhibitor treatment. Clin
Cancer Res. 2010; 16: 5581-5590.
34. Klempner SJ, Cohen DW, Costa DB. ALK translocation
in non-small cell lung cancer with adenocarcinoma and
squamous cell carcinoma markers. J Thorac Oncol. 2011;
6: 1439-1440.
35. Qu Y, Che N, Zhao D, Zhang C, Su D, Zhou L, Zhang
L, Wang C, Zhang H, Wei L. The clinicopathological
signicance of ALK rearrangements and KRAS and EGFR
mutations in primary pulmonary mucinous adenocarcinoma.
Tumour Biol. 2015; 36: 6417-6424.
36. Nishino M, Klepeis VE, Yeap BY, Bergethon K, Morales-
Oyarvide V, Dias-Santagata D, Yagi Y, Mark EJ, Iafrate
AJ, Mino-Kenudson M. Histologic and cytomorphologic
features of ALK-rearranged lung adenocarcinomas. Mod
Pathol. 2012; 25: 1462-1472.
37. Shaw AT, Yeap BY, Mino-Kenudson M, Digumarthy SR,
Costa DB, Heist RS, Solomon B, Stubbs H, Admane S,
McDermott U, Settleman J, Kobayashi S, Mark EJ, et al.
Clinical features and outcome of patients with non-small-
cell lung cancer who harbor EML4-ALK. J Clin Oncol.
2009; 27: 4247-4253.
38. Faugeroux V, Pailler E, Auger N, Taylor M, Farace F.
Clinical utility of circulating tumor cells in ALK-positive
non-small-cell lung cancer. Front Oncol. 2014; 4: 281.
39. Kim H, Jang SJ, Chung DH, Yoo SB, Sun P, Jin Y, Nam
KH, Paik JH, Chung JH. A comprehensive comparative
analysis of the histomorphological features of ALK-
rearranged lung adenocarcinoma based on driver oncogene
mutations: frequent expression of epithelial-mesenchymal
transition markers than other genotype. PLoS One. 2013; 8:
e76999.
40. Doebele RC, Pilling AB, Aisner DL, Kutateladze TG, Le
AT, Weickhardt AJ, Kondo KL, Linderman DJ, Heasley
LE, Franklin WA, Varella-Garcia M, Camidge DR.
Mechanisms of resistance to crizotinib in patients with ALK
gene rearranged non-small cell lung cancer. Clin Cancer
Res. 2012; 18: 1472-1482.
41. Katayama R, Shaw AT, Khan TM, Mino-Kenudson M,
Solomon BJ, Halmos B, Jessop NA, Wain JC, Yeo AT,
Benes C, Drew L, Saeh JC, Crosby K, et al. Mechanisms
of acquired crizotinib resistance in ALK-rearranged lung
cancers. Sci Transl Med. 2012; 4: 120ra17.
42. Travis WD, Brambilla E, Burke, AP, Marx A, Nicholson
A. Lyon: IARC Press; 2015 World Health Organization
Classication of Tumours of the Lung, Pleura, Thymus and
Heart.
Oncotarget23262
www.impactjournals.com/oncotarget
43. Hou HW, Warkiani ME, Khoo BL, Li ZR, Soo R a, Tan
DS-W, Lim W-T, Han J, Bhagat AAS, Lim CT. Isolation
and retrieval of circulating tumor cells using centrifugal
forces. Sci Rep. 2013; 3: 1259.

Supplementary resource (1)

... Interestingly, in cases where no oncogenic drivers were found in the tissue-based examination, the assessment of CTCs was also able to identify EGFR mutations [163]. In addition to EGFR mutations, ALK rearrangements can be detected with a high sensitivity and high concordance with tissue [164] when analyzing CNVs, even though this is with a less broad range of mutations than with NGS-based methods in ctDNA [162,165,166]. The expression of MET in CTCs has been shown to correlate with their expression in the primary tumor tissues of NSCLC patients [167], expanding the number of molecular drivers that can be observed in CTCs. ...
Article
Full-text available
Simple Summary Liquid biopsy has recently been introduced as a novel method in cancer diagnostics. It is less invasive for patients than conventional tissue biopsies, as the assay material is drawn from peripheral blood. The detected DNA fragments, as well as the cells and extracellular vesicles, can be used as biomarkers for cancer. Not all biomarkers are equally reliable in cancer diagnostics. In this review, we give an overview of the lung cancer biomarkers identified in liquid biopsy assays and discuss the differences, current applications, and future perspectives of liquid biopsies in lung cancer. Abstract As lung cancer has the highest cancer-specific mortality rates worldwide, there is an urgent need for new therapeutic and diagnostic approaches to detect early-stage tumors and to monitor their response to the therapy. In addition to the well-established tissue biopsy analysis, liquid-biopsy-based assays may evolve as an important diagnostic tool. The analysis of circulating tumor DNA (ctDNA) is the most established method, followed by other methods such as the analysis of circulating tumor cells (CTCs), microRNAs (miRNAs), and extracellular vesicles (EVs). Both PCR- and NGS-based assays are used for the mutational assessment of lung cancer, including the most frequent driver mutations. However, ctDNA analysis might also play a role in monitoring the efficacy of immunotherapy and its recent accomplishments in the landscape of state-of-the-art lung cancer therapy. Despite the promising aspects of liquid-biopsy-based assays, there are some limitations regarding their sensitivity (risk of false-negative results) and specificity (interpretation of false-positive results). Hence, further studies are needed to evaluate the usefulness of liquid biopsies for lung cancer. Liquid-biopsy-based assays might be integrated into the diagnostic guidelines for lung cancer as a tool to complement conventional tissue sampling.
... ALK-rearranged CTC levels during the treatment monitoring of five patients with crizotinib presented different response patterns [203]. Moreover, in another small study, it was also shown that CTCs recapitulate the ALK rearrangement status of tumor tissue, and, therefore, CTCs represent a suitable alternative to tissue biopsy for guiding treatment [204]. In a study of 39 ALK-rearranged NSCLC patients treated with crizotinib, Pailler et al. found aberrant ALK copy number gain in CTCs and correlated the dynamic changes in the levels of these CTCs with PFS. ...
Article
Full-text available
The treatment of non-small cell lung cancer (NSCLC) has recently evolved with the introduction of targeted therapy based on the use of tyrosine kinase inhibitors (TKIs) in patients with certain gene alterations, including EGFR, ALK, ROS1, BRAF, and MET genes. Molecular targeted therapy based on TKIs has improved clinical outcomes in a large number of NSCLC patients with advanced disease, enabling significantly longer progression-free survival (PFS). Liquid biopsy is an increasingly popular diagnostic tool for treating TKI-based NSCLC. The studies presented in this article show that detection and analysis based on liquid biopsy elements such as circulating tumor cells (CTCs), cell-free DNA (cfDNA), exosomes, and/or tumor-educated platelets (TEPs) can contribute to the appropriate selection and monitoring of targeted therapy in NSCLC patients as complementary to invasive tissue biopsy. The detection of these elements, combined with their molecular analysis (using, e.g., digital PCR (dPCR), next generation sequencing (NGS), shallow whole genome sequencing (sWGS)), enables the detection of mutations, which are required for the TKI treatment. Despite such promising results obtained by many research teams, it is still necessary to carry out prospective studies on a larger group of patients in order to validate these methods before their application in clinical practice.
... In patients with lung adenocarcinoma, anaplastic large-cell lymphoma (ALK) gene rearrangement and ALK protein expression in CTCs were concordant with findings from tumor tissue, 138 which has been confirmed by other researchers. 139,140 A rearrangement of repressor of silencing 1 (ROS1) is another example of chromosomal aberrations detectable in CTCs, with biopsy-confirmed gene fusion in NSCLC patients. 141 Dynamic changes in the number of CTCs with aberrant ALK-fluorescence in situ hybridization patterns, such as ALK copy number gain, might serve as predictive markers of the response to treatment, as these aberrations are considered to be one of the mechanisms underlying acquired resistance to crizotinib (an ALK and ROS1 inhibitor). ...
Article
Full-text available
Lung cancer is one of the most common neoplasms and the leading cause of cancer-related deaths worldwide. Despite recent progress in understanding the pathomechanisms of lung cancer, it is frequently associated with late diagnosis, high incidence of metastases and poor response to treatment. Thus, there is extensive research in the field of biomarkers that aims to optimize management of lung cancer. The aim of this study was to review the current perspectives of a wide spectrum of circulating molecules that seem promising as new potential biomarkers of lung cancer. Among these, biochemical (active proteins), immunological (immunocompetent cells, cytokines, chemokines, and antibodies) and genetic (circulating tumor DNA, cell-free DNA and microRNA) markers are presented and discussed. The use of these markers would support the early detection of lung cancer and might be used for predicting disease progression, response of the disease to targeted therapies, monitoring the course of treatment, and developing individualized diagnostic and therapeutic strategies. Special attention was given to potential markers of nervous system involvement in the course of lung cancer, due to its prevalence and devastating impact. Limitations of the potential biomarkers are also outlined and future directions of investigations in this field highlighted, with the aim of improving the accuracy and practical utility of these biomarkers.
... Another opportunity for liquid biopsies involves CTCs that are cancer cells shed from the tumor into the bloodstream (41). Several studies have shown a high rate of concordance in gene mutations between circulating tumor cells and DNA from tumor tissue, making CTCs a potentially suitable candidate as a cancer biomarker (42)(43)(44). Clinically, the abundance of CTCs has been associated with a worse prognosis in prostate, lung, and other cancers. These clinical observations are corroborated by experimental findings that have demonstrated the potential of CTCs to form metastatic clusters (45). ...
Article
Although radiologic imaging and histological assessment of tumor tissues are classic approaches for diagnosis and monitoring of treatment response, they have many limitations. These include challenges in distinguishing benign from malignant masses, difficult access to the tumor, high cost of the procedures, and tumor heterogeneity. In this setting, liquid biopsy has emerged as a potential alternative for both diagnostic and monitoring purposes. The approaches to liquid biopsy include cell-free DNA/circulating tumor DNA (cfDNA/ctDNA), long and micro non-coding RNAs, proteins/peptides, carbohydrates/lectins, lipids, and metabolites. Other approaches include detection and analysis of circulating tumor cells (CTCs), extracellular vesicles (EVs), and tumor-activated platelets. Ultimately, reliable use of liquid biopsies requires bioinformatics and statistical integration of multiple data sets in order to achieve approval in a Clinical Laboratory Improvement Amendments (CLIA) setting. This review provides a balanced and critical assessment of recent discoveries regarding tumor-derived biomarkers in liquid biopsies along with the potential and pitfalls for cancer detection and longitudinal monitoring
... Currently, NSCLC is primarily treated with minimally invasive procedures, including video-assisted thoracoscopic surgery and smaller resection of the primary lesion [6,7]. Lobectomy and lymph node dissection are the standard treatment approaches for early-stage NSCLC. ...
Article
Full-text available
Background: Surgical treatment is the first choice for non-small-cell lung cancer. To date, there are only few studies on the changes in laboratory indexes in two types of surgery, namely, thoracoscopic lobectomy and segmental pneumonectomy. Aim: To investigate the clinical impact of thoracoscopic lobectomy and segmentectomy in patients with early-stage non-small-cell lung cancer. Methods: We retrospectively reviewed the medical records of 94 patients with early-stage NSCLC in our hospital from October 2017 to October 2019. The patients were divided into two groups. The patients in control and observation groups received thoracoscopic lobectomy and thoracoscopic segmentectomy, respectively. The perioperative indicators, complications, lung function, T cell subsets, tumor markers, follow-up of tumor recurrence rate, and survival rate were compared between two groups. Results: The operation time of the observation group was longer, and the chest drainage volume was less at 24-48 h after the operation, and the chest tube indwelling time and postoperative hospital stay were shorter than those of the control group. No significant differences in complication probability were observed between two groups. The levels of FEV1, FVC, and MVV in the two groups were lower than those before the operation at 3 days after surgery, but the FEV1, FVC, and MVV levels in the observation group were higher than those in the control group. The CD3+, CD4+, and CD4+/CD8+ levels in the two groups were lower than those before the operation at 24 h and 72 h after the operation, but CD3+, CD4+, and CD4+/CD8+ levels in the observation group were higher than those of the control group. Conclusion: Thoracoscopic lobectomy and segmental resection have similar clinical effects in the treatment of early-stage NSCLC patients, but segmental resection can preserve healthy lung tissue as much as possible, with less trauma, protect lung function, and promote postoperative recovery.
... Ainsi, lorsque des clones de CTC portant ces mutations sont identifiés, les patients pourraient être pris en charge différemment pour réajuster le traitement, notamment en les traitant avec des thérapies d'inhibiteurs de tyrosine kinase de troisième génération plus rapidement. Il est également possible d'utiliser des méthodes de FISH afin de détecter des amplifications ou des translocations de gènes d'intérêt dans les cancers, comme c'est le cas du réarrangement du gène ALK dans le NSCLC où l'étude des CTC complémente l'analyse de la tumeur et met en évidence les clones minoritaires(Tan et al. 2016;Pailler et al. 2013). De même pour l'amplification du gène HER2 chez les patientes atteintes de cancer du sein, où l'analyse FISH des CTC peut aider au choix du traitement(Frithiof, Aaltonen, and Ryden 2016), ou encore la perte de PTEN et le gain du gène MYC dans le cancer de la prostate(Sun et al. 2011). ...
Thesis
Le Carcinome à cellules de Merkel (CCM) est une tumeur cutanée rare et agressive, de mauvais pronostic. La détection précoce de la tumeur primaire et des rechutes est un enjeu majeur pour améliorer la survie des patients. Les Cellules tumorales circulantes (CTC) peuvent se détacher des tumeurs solides, passer dans le sang et vont pouvoir former des métastases. Leur détection, la biopsie liquide, peut apporter de nombreuses informations cliniques pertinentes notamment pour identifier précocement les rechutes.Le but de cette thèse a été l’identification des CTC dans le CCM grâce notamment à la mise au point d’une nouvelle méthode de détection. Cette méthode dite R-D a été comparée au CellSearch, Gold-Standard pour la détection des CTC chez 28 patients atteints de CCM. Ces deux méthodes apparaissent complémentaires dans la détection des CTC et les résultats sont corrélées au stade de la maladie. Un seuil de détection associé à la présence de métastases a été défini et doit être valider cliniquement pour la détection précoce de la dissémination de la maladie. Dans une seconde partie, l’objectif a été de rechercher la présence du polyomavirus du CCM (MCPyV), impliqué dans 80% des cas de CCM, dans les CTC individuelles ainsi que dans les cellules tumorales issues de biopsie tissulaire. Sa présence s’est révélée hétérogène à l’échelle cellulaire dans ces deux types d’échantillons, donnée non connue à ce jour. Enfin, nous avons tenté de mettre en culture in-vitro des CTC issues de prélèvements de patients, sans parvenir, pour le moment, à identifier les conditions idéales.Ce travail fournit des bases solides pour de futures études cliniques incluant des cohortes plus larges, afin notamment de déterminer l’intérêt clinique des CTC comme marqueur pronostique et évolutif du CCM, mais également pour des études biologiques sur les mécanismes oncogéniques (dépendant notamment du MCPyV).
... Advancements in cell enrichment techniques have generated a plethora of data detailing the molecular and functional characteristics of CTCs. In response to treatment for multiple types of solid cancers, expression patterns of CTC biomarkers shift dynamically which suggests that CTC biomarker analysis can be an effective measure of therapeutic response [16][17][18][19]. This is particularly significant because through epigenetic mutations and selective pressure from treatment, tumor cells that are in a constant state of genetic evolution can develop subclones that are resistant to front-line therapies [7,[20][21][22]. ...
Article
Full-text available
Direct assessment of patient samples holds unprecedented potential in the treatment of cancer. Circulating tumor cells (CTCs) in liquid biopsies are a rapidly evolving source of primary cells in the clinic and are ideal candidates for functional assays to uncover real-time tumor information in real-time. However, a lack of routines allowing direct and active interrogation of CTCs directly from liquid biopsy samples represents a bottleneck for the translational use of liquid biopsies in clinical settings. To address this, we present a workflow for using a microfluidic vortex-assisted electroporation system designed for the functional assessment of CTCs purified from blood. Validation of this approach was assessed through drug response assays on wild-type (HCC827 wt) and gefitinib-resistant (HCC827 GR6) non-small cell lung cancer (NSCLC) cells. HCC827 cells trapped within microscale vortices were electroporated to sequentially deliver drug agents into the cytosol. Electroporation conditions facilitating multi-agent delivery were characterized for both cell lines using an automatic single-cell image fluorescence intensity algorithm. HCC827 GR6 cells spiked into the blood to emulate drug-resistant CTCs were able to be collected with high purity, demonstrating the ability of the device to minimize background cell impact for downstream sensitive cell assays. Using our proposed workflow, drug agent combinations to restore gefitinib sensitivity reflected the anticipated cytotoxic response. Taken together, these results represent a microfluidics multi-drug screening panel workflow that can enable functional interrogation of patient CTCs in situ, thereby accelerating the clinical standardization of liquid biopsies.
Chapter
Comprehensive tumour characterisation is indispensable for patients to receive targeted therapy. The use of liquid biopsy, particularly circulating tumour cells (CTC), has shown great promise in the treatment and management of cancer patients. An in-depth understanding of CTCs at the cellular and molecular level can provide clues as to the mechanisms of cancer dissemination and the pathways responsible for conferring intrinsic and acquired resistance to therapeutic agents. Herein, we discuss the current methods of CTC isolation and analysis at the single-cell resolution for therapeutic applications in the management of cancer.KeywordsCirculating tumour cellsSingle-cell analysisCTC isolationTargeted therapy
Thesis
Full-text available
Lung cancer is the leading cause of cancer-related death worldwide. The majority of patients are diagnosed at advanced and currently incurable stages. Non-small cell lung cancer (NSCLC) represents around 85% of cases. Alterations in the DNA damage response (DDR) and resulting genomic instability (GI) contribute to NSCLC etiology and progression. However, their therapeutic exploitation is disappointing. Circulating tumor cells (CTCs) that dissociate from the primary tumor or its metastases harbor distinct biological properties that allow them to travel through circulation and colonize distant organs. Further insight into CTC biology and identification of their vulnerabilities would provide a rationale for the targeting of the most aggressive tumor clones that fuel metastatic progression.We hypothesized that the DDR and genome integrity maintenance dysfunctions are critical processes in CTC metastatic potency and NSCLC progression. The main aim of my PhD project was to perform a molecular and functional characterization of NSCLC CTCs to elucidate the mechanistic basis of their tumorigenic potential and identify novel therapeutic targets. Using CTCs from 55 patients with advanced-stage NSCLC, our laboratory has established four CTC-derived xenografts (CDX) (GR-CDXL1, GR-CDXL2, GR-CDXL3 and GR-CDXL4) and three CDX-derived cell lines. Preliminary analyses have shown that the four CDX recapitulated patient tumor histology and response to platinum-based chemotherapy, which validated the clinical relevance of our models. To determine the extent to which the CDX was representative of the corresponding patient tumor biopsy, whole-exome sequencing (WES) was performed on the CDX (GR-CDXL1, GR-CDXL2, GR-CDXL3, GR-CDXL4), CDX-derived cell lines, corresponding patient tumor biopsy and single CTCs. The four major goals of my thesis were as follows: (i) identification of genomic alterations and in-depth comparative genomic analysis of the CDX, CDX-derived cell lines, corresponding patient tumor biopsy and CTCs using WES analysis data, (ii) functional characterization of DDR activity and chromosomal instability (CIN) events in the CDX-derived cell lines, (iii) in vitro drug assays targeting DDR defects, (iv) 3D modeling of the metastatic potency of our CDX-derived cell lines in ovo in the chick embryo chorioallantoic membrane (CAM) and in vivo in immunodeficient mice and pharmacological targeting of metastasis.Genomic analysis by WES has shown considerable mutational landscape similarity between the CDX, the cell lines, the patient biopsies and single CTCs. WES and reconstruction of phylogenetic trees of the CDX and the CDX-derived cell lines revealed truncal alterations in key DDR and genome integrity-related genes prevalent across models and assessed as therapeutic targets in vitro, in ovo and in vivo. GR-CDXL1 presented homologous recombination deficiency linked to bi-allelic BRCA2 mutation and FANCA deletion and unrepaired DNA lesions post-mitosis. GR-CDXL1 cells were sensitive to PARP inhibitor (PARPi) olaparib, despite chemoresistance, which challenges the current clinical rationale claiming that chemosensitive NSCLC patients should respond to PARPi. Targeting CIN through centrosome clustering inhibition in GR-CDXL3 impeded tumor growth in ovo and in vivo. In GR-CDXL4, olaparib sensitivity was dictated by SLFN11 overexpression, which also corresponded with increased neuroendocrine marker expression at patient disease progression, suggesting a predictive value of SLFN11 in histological transformation of NSCLC into SCLC.This study unravels distinct DDR profiles as a central mechanism underpinning CTC metastatic potency. It also suggests SLFN11 overexpression as a potential biomarker of sensitivity to PARPi in NSCLC independently of BRCAness. Overall, our models provide a robust platform for drug testing of DDR-targeted strategies to expand patient categories that may benefit from precision medicine in metastatic NSCLC.
Article
Full-text available
The advent of rationally targeted therapies such as small-molecule tyrosine kinase inhibitors (TKIs) has considerably transformed the therapeutic management of a subset of patients with non-small-cell lung cancer (NSCLC) harboring defined molecular abnormalities. When such genetic molecular alterations are detected the use of specific TKI has demonstrated better results (overall response rate, progression free survival) compared to systemic therapy. However, the detection of such molecular abnormalities is complicated by the difficulty in obtaining sufficient tumor material, in terms of quantity and quality, from a biopsy. Here, we described how circulating tumor cells (CTCs) can have a clinical utility in anaplastic lymphoma kinase (ALK) positive NSCLC patients to diagnose ALK-EML4 gene rearrangement and to guide therapeutic management of these patients. The ability to detect genetic abnormalities such ALK rearrangement in CTCs shows that these cells could offer new perspectives both for the diagnosis and the monitoring of ALK-positive patients eligible for treatment with ALK inhibitors.
Article
Full-text available
Background Circulating tumor cells (CTCs) are cancer cells that can be isolated via liquid biopsy from blood and can be phenotypically and genetically characterized to provide critical information for guiding cancer treatment. Current analysis of CTCs is hindered by the throughput, selectivity and specificity of devices or assays used in CTC detection and isolation. Methodology/Principal Findings Here, we enriched and characterized putative CTCs from blood samples of patients with both advanced stage metastatic breast and lung cancers using a novel multiplexed spiral microfluidic chip. This system detected putative CTCs under high sensitivity (100%, n = 56) (Breast cancer samples: 12–1275 CTCs/ml; Lung cancer samples: 10–1535 CTCs/ml) rapidly from clinically relevant blood volumes (7.5 ml under 5 min). Blood samples were completely separated into plasma, CTCs and PBMCs components and each fraction were characterized with immunophenotyping (Pan-cytokeratin/CD45, CD44/CD24, EpCAM), fluorescence in-situ hybridization (FISH) (EML4-ALK) or targeted somatic mutation analysis. We used an ultra-sensitive mass spectrometry based system to highlight the presence of an EGFR-activating mutation in both isolated CTCs and plasma cell-free DNA (cf-DNA), and demonstrate concordance with the original tumor-biopsy samples. Conclusions/Significance We have clinically validated our multiplexed microfluidic chip for the ultra high-throughput, low-cost and label-free enrichment of CTCs. Retrieved cells were unlabeled and viable, enabling potential propagation and real-time downstream analysis using next generation sequencing (NGS) or proteomic analysis.
Article
Full-text available
Non-small-cell lung cancer (NSCLC) harboring the anaplastic lymphoma kinase gene (ALK) rearrangement is sensitive to the ALK inhibitor crizotinib, but resistance invariably develops. Ceritinib (LDK378) is a new ALK inhibitor that has shown greater antitumor potency than crizotinib in preclinical studies. In this phase 1 study, we administered oral ceritinib in doses of 50 to 750 mg once daily to patients with advanced cancers harboring genetic alterations in ALK. In an expansion phase of the study, patients received the maximum tolerated dose. Patients were assessed to determine the safety, pharmacokinetic properties, and antitumor activity of ceritinib. Tumor biopsies were performed before ceritinib treatment to identify resistance mutations in ALK in a group of patients with NSCLC who had had disease progression during treatment with crizotinib. A total of 59 patients were enrolled in the dose-escalation phase. The maximum tolerated dose of ceritinib was 750 mg once daily; dose-limiting toxic events included diarrhea, vomiting, dehydration, elevated aminotransferase levels, and hypophosphatemia. This phase was followed by an expansion phase, in which an additional 71 patients were treated, for a total of 130 patients overall. Among 114 patients with NSCLC who received at least 400 mg of ceritinib per day, the overall response rate was 58% (95% confidence interval [CI], 48 to 67). Among 80 patients who had received crizotinib previously, the response rate was 56% (95% CI, 45 to 67). Responses were observed in patients with various resistance mutations in ALK and in patients without detectable mutations. Among patients with NSCLC who received at least 400 mg of ceritinib per day, the median progression-free survival was 7.0 months (95% CI, 5.6 to 9.5). Ceritinib was highly active in patients with advanced, ALK-rearranged NSCLC, including those who had had disease progression during crizotinib treatment, regardless of the presence of resistance mutations in ALK. (Funded by Novartis Pharmaceuticals and others; ClinicalTrials.gov number, NCT01283516.).
Article
®orld Health Organization (WHO) Classifications of Tumors have historically provided a global standard for tumor diagnosis. Familiarity with these classifications is essential for clinicians because they provide the foundation for accurate patient diagnosis and optimal medical management. In addition, the diagnostic terminology and criteria recommended by the WHO should be incorporated into study design of clinical trials and molecular studies such as The Cancer Genome Atlas (TCGA). WHO Classifications are periodically updated to incorporate important advances to make them relevant to current knowledge and clinical practice. The 2015 WHO Classification of Tumors of the Lung, Pleura, Thymus and Heart has just been published 1 and the Journal of Thoracic Oncology will be publishing a series of articles summarizing key points about the new classification outlining the major changes from the 2004 classification. 2–4 † The International Association for Research on Cancer (IARC) under the leadership of Dr. Hiroko Ohgaki coordinated this project and was the publisher of this book. IARC hosted a consensus meeting in Lyon, France, April 24–26, 2014, where all major changes in the classification from the 2004 book were discussed and approved (Figure). The 2015 WHO Classification features the incorporation of many exciting new advances in thoracic tumor diagnosis and classification. It was formulated by a multidisciplinary group of international experts with broad international representation. The primary changes in the lung classification, which are highlighted in the first paper in this series, 3 relate to lung cancer where over the past decade remarkable progress in genetics and therapy has had a major impact on tumor classification. These most significant changes are consequent to the 2011 IASLC/ATS/ERS Classification 5 including recommendations for routine molecular testing and use of immunohistochemistry, a new approach to small biopsies and cytology and a novel way of subtyping of surgically resected lung adenocarcinomas that has provided a powerful new tool for identifying prognostic and molecular correlations. Large scale genomic studies from the TCGA 6,7 and Clinical Lung Cancer Genome Project 8 provided a strong genetic foundation for reclassification of squamous cell carcinoma, adenocarcinoma and large cell carcinoma. These changes have major clinical relevance as histologic type and genetics are now driving personalized medicine for lung cancer patients. New concepts in adenocarcinoma classification with lepidic versus invasive patterns are also having an impact on the approach to tumor size measurement for TNM staging of small tumors ≤3cm and therefore the surgical management of patients. Support from the IASLC, through their Pathology Committee and by providing multidisciplinary input, contributed greatly to the development of this classification. 5,9
Article
Lung cancer is the most common cancer worldwide, and a leading cause of cancer-related death. Despite improvements in molecular diagnosis and targeted therapies, the 5-years overall survival remains poor. To obtain a better insight into the genetic architecture of the most common type of lung cancer, non-small lung cancer (NSCLC), we performed multi-region DNA sequencing on 7 resected NSCLC samples. Ultra-deep sequencing of a comprehensive cancer gene panel and whole-exome sequencing revealed intratumor heterogeneity in all samples, with several putative tumor driver mutations present in some but not all regions of a tumor. Phylogenetic tree analyses based on non-synonymous mutations revealed a branched evolution pattern in all tumors. An adenosquamous tumor showed striking intratumor heterogeneity, with only a third of all non-synonymous mutations shared across all tumor regions, and clear separation of adenocarcinoma and squamous cell carcinoma tumor regions in the remaining two-third of the mutations. Our multi-region exome sequencing data also revealed regional differences in DNA copy number alterations. Some tumors, relatively homogeneous in terms of mutations, displayed high levels of intratumor heterogeneity in terms of DNA copy number changes, indicating the presence of distinct patterns of intratumor heterogeneity that might contribute to disease progression in different tumours. Overall, our multi-region deep exome sequencing data revealed intratumor heterogeneity in NSCLC, demonstrating branched evolution, both in terms of non-synoymous mutations and DNA copy number alterations, which has important implications for our understanding of the clonal evolution of NSCLC. Citation Format: Elza De Bruin, Nicholas McGranahan, Lucy Yates, Mariam Jamal-Hanjani, Max Salm, Richard Mitter, Seema Shafi, Nirupa Murugaesu, Andrew Rowan, Marco Gerlinger, David Wedge, Stuart Horswell, Ignacio Varela, Warren Tom, Chaitali Parikh, Timothy Harkins, Clarence Lee, Nik Matthews, Aengus Stewart, Peter Campbell, Charles Swanton. Intratumor heterogeneity in non-small cell lung cancer inferred by multi-region exome sequencing. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 983. doi:10.1158/1538-7445.AM2014-983
Article
Primary pulmonary mucinous adenocarcinoma (PPMA) is one of the important subtypes of lung adenocarcinoma. Detection of anaplastic lymphoma receptor tyrosine kinase (ALK) rearrangements and of KRAS and epidermal growth factor receptor (EGFR) mutations will help in diagnosing and predicting treatment outcome. The aim of this study was to investigate the clinicopathological significance of ALK rearrangements, KRAS and EGFR mutations in PPMA. ALK expression was detected immunohistochemically. KRAS and EGFR mutations were determined by the amplification refractory mutation system. Seventy-three patients of PPMA were enrolled. ALK rearrangements were detected in 34.2 % of patients and were more frequent in upper/middle lobe, stage III-IV, lymphatic permeation-positive patients and non-smokers. ALK rearrangements were significantly increased in the solid tumor predominant with mucin production subtype, and in special tissue structures, including signet ring cells, cribriform, and micropapillary patterns. KRAS mutations were observed in 23.3 % of patients and were more prevalent in invasive mucinous adenocarcinoma and lower lobe tumors. Only one case of ALK rearrangements harbored KRAS mutation, and no cases manifested with the coexistence of ALK rearrangements and EGFR mutations. KRAS and EGFR co-mutation was detected in one case. PPMA patients with ALK rearrangements or KRAS mutation represent a unique subtype in NSCLC. The results provide basis data for target therapy screening of PPMA patients.
Article
Objectives When advanced non-small-cell lung cancer (NSCLC) progresses during first-line treatment, re-biopsy may be indicated to detect a possible new biological profile (comparison to initial status, emergence of resistance biomarkers, or assessment of new biomarkers). The aim of this pragmatic prospective multicenter study was to assess the feasibility and clinical utility of re-biopsy in advanced NSCLC in a real-world setting. Methods The main inclusion criteria were advanced NSCLC with an indication for repeat biopsy identified by the patient's clinician. The primary outcome was the percentage of successful procedures. Secondary outcomes were the type of procedure, new biological status, tolerability of the procedure, and clinical utility (treatment modification). Results From May 2012 to May 2013, 18 centers enrolled 100 patients (males: 44%; median age: 64.8 years; PS 0/1: 88%; adenocarcinoma: 89%; EGFR mutated: 50%; no initial biological profile: 16.4%). Re-biopsy was not possible in 19.5% of cases and provided no or too few tumor cells in 25.6% of cases. Repeat biopsy was useful for guiding treatment in 30.4% (25/82) of cases. Complications were infrequent (2 cases of moderate bleeding and 1 case of pneumothorax). Conclusion Re-biopsy of advanced NSCLC is feasible in the real-world setting, with acceptable adverse events. Guidelines are needed on the indications of re-biopsy, the choice of procedure, the sampling site, and laboratory analysis.