ArticlePDF Available

Circulating cell-free DNA as a prognostic and predictive biomarker in non-small cell lung cancer

Authors:

Abstract and Figures

Circulating cell-free DNA (cfDNA), which can be obtained from plasma or serum by non-invasive procedures, has showed great potential to predict treatment response and survival for cancer patients. Several studies have assessed the prognostic and predictive value of cfDNA in non-small cell lung cancer (NSCLC). However, these studies were often small and reported varying results. To address this issue, a meta-analysis was carried out. A total of 22 studies involving 2518 patients were subjected to the final analysis. Our results indicated that NSCLC patients with higher cfDNA concentration had shorter median progression-free survival (PFS) and overall survival (OS) time. In addition, high levels of cfDNA were significantly associated with poor PFS (hazard ratio or HR, 1.32; 95% CI, 1.02-1.71) and OS (HR, 1.64; 95% CI, 1.26-2.15). With respect to tumor specific mutations, we failed to reveal significant differences for PFS (HR, 1.30; 95% CI, 0.66-2.56) and OS (HR, 1.05; 95% CI, 0.49-2.25) when NSCLC patients were grouped according to KRAS genotype detected in cfDNA. However, NSCLC patients which harbored EGFR activating mutation in cfDNA had a greater chance of response to EGFR-TKIs (odds ratio or OR, 1.96; 95% CI, 1.59-2.42). No significant publication bias was detected in this study. In conclusion, cfDNA could act as a prognostic and predictive biomarker for patients with NSCLC.
Content may be subject to copyright.
Oncotarget44583
www.impactjournals.com/oncotarget
www.impactjournals.com/oncotarget/ Oncotarget, Vol. 7, No. 28
Circulating cell-free DNA as a prognostic and predictive
biomarker in non-small cell lung cancer
Bo Ai1, Huiquan Liu2, Yu Huang2, Ping Peng2
1Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology,
Hubei, Wuhan 430030, People’s Republic of China
2Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei,
Wuhan 430030, People’s Republic of China
Correspondence to: Ping Peng, email: pengpingtjh@163.com
Keywords: circulating cell-free DNA, non-small cell lung cancer, prognosis, biomarker, meta-analysis
Received: February 28, 2016 Accepted: May 29, 2016 Published: June 15, 2016
ABSTRACT
Circulating cell-free DNA (cfDNA), which can be obtained from plasma or serum
by non-invasive procedures, has showed great potential to predict treatment response
and survival for cancer patients. Several studies have assessed the prognostic and
predictive value of cfDNA in non-small cell lung cancer (NSCLC). However, these
studies were often small and reported varying results. To address this issue, a
meta-analysis was carried out. A total of 22 studies involving 2518 patients were
subjected to the nal analysis. Our results indicated that NSCLC patients with higher
cfDNA concentration had shorter median progression-free survival (PFS) and overall
survival (OS) time. In addition, high levels of cfDNA were signicantly associated with
poor PFS (hazard ratio or HR, 1.32; 95% CI, 1.02-1.71) and OS (HR, 1.64; 95% CI,
1.26-2.15). With respect to tumor specic mutations, we failed to reveal signicant
differences for PFS (HR, 1.30; 95% CI, 0.66-2.56) and OS (HR, 1.05; 95% CI, 0.49-
2.25) when NSCLC patients were grouped according to KRAS genotype detected in
cfDNA. However, NSCLC patients which harbored EGFR activating mutation in cfDNA
had a greater chance of response to EGFR-TKIs (odds ratio or OR, 1.96; 95% CI,
1.59-2.42). No signicant publication bias was detected in this study. In conclusion,
cfDNA could act as a prognostic and predictive biomarker for patients with NSCLC.
INTRODUCTION
Lung cancer is the most commonly diagnosed
cancer as well as the leading cause of cancer-related deaths
in the world [1]. Non-small cell lung cancer (NSCLC)
accounts for approximately 80% cases of lung cancer [2].
Most NSCLC patients are diagnosed with advanced or
distant stages and they are ineligible for curative surgery
and often suffer a poor survival. Identifying biomarkers
related to treatment response and prognosis may be helpful
to improve the clinical outcome of patients with NSCLC.
Circulating cell-free DNA (cfDNA), which can
be isolated from the plasma or serum by non-invasive
procedures, has been proposed as an attractive biomarker
to estimate treatment response, detect drug resistance
and predict clinical outcome for cancer patients [3–7].
It has been experimentally evidenced that tumor cells
can release genomic DNA into the blood and circulating
DNA can reect the tumor burden and tumor biologic
characteristics [6, 8]. A series of studies have shown that
NSCLC patients have higher levels of cfDNA in the blood
compared with healthy controls or patients with benign
diseases [9–11]. The quantitative assay of cfDNA may be
a screening tool for NSCLC. It has been shown that the
diagnostic accuracy of quantitative analysis of cfDNA is
not lower than conventional serum biomarkers for lung
cancer screening [12]. Furthermore, cancer-associated
genetic alterations, such as point mutations, deletions, and
copy number variations, can be detected in cfDNA [13].
In NSCLC, many studies have investigated the diagnostic
accuracy of cfDNA for detecting epithermal growth factor
receptor (EGFR) mutation [14–16]. Two recent meta-
analyses demonstrated that cfDNA was a highly specic
and effective biomarker to measure EGFR mutation
status in NSCLC [17, 18]. These evidences suggested that
genotype in cfDNA could be a promising tumor biomarker
for NSCLC.
Oncotarget44584
www.impactjournals.com/oncotarget
A large number of studies had investigated the
predictive or prognostic value of cfDNA concentration
in NSCLC patients in recent years [19–22] (see Table 1
for references). However, these studies were often small
and reported varying results. Some of them showed that a
higher cfDNA concentration was associated with poorer
survival in NSCLC patients [19, 20], whereas other studies
failed to demonstrate such correlation [21, 22]. On the
other hand, several studies had analyzed the association
between genotype detected in cfDNA with treatment
response or survival in NSCLC [23–26]. Some of them
suggested that tumor specic mutations such as KRAS or
EGFR presented in cfDNA might be useful prognostic
and predictive biomarkers for NSCLC [23, 24]. However,
some others indicated that such gene mutations in cfDNA
had no predictive or prognostic value [25, 26].
As the existing studies are conicting in their
results, it is still difcult to determine the predictive
and prognostic role of cfDNA in patients with NSCLC.
Therefore, a meta-analysis aimed to address this issue was
carried out.
RESULTS
Search results
Figure 1 illustrated the process of study selection.
298 studies were initially found by our search strategy.
30 articles were reviewed in detail after the article titles
and abstracts were checked [9, 16, 19–46]. Eight studies
were excluded from the meta-analysis [39–46], leaving
22 studies that fullled the eligibility criteria [9, 16, 19–
38] (Table 1). Among the 8 excluded studies, 7 did not
provide sufcient data for extracting odds ratio (OR) or
hazard ratio (HR) [39–45], and other 1 study was excluded
because the same cohort of patients was used in other
selected study [46]. The total number of patients included
in this study was 2518, ranging from 22 [21, 37] to 446
[29] cases per study. 12 studies evaluated the prognostic
role of cfDNA concentration in NSCLC [9, 19-22, 27-33].
4 studies reported the prognostic role of KRAS genotype
detected in cfDNA for NSCLC [20, 23, 25, 34]]. Another
7 studies dealt with the predictive role of EGFR genotype
presented in cfDNA for NSCLC patients who were treated
with tyrosine kinase inhibitors of EGFR (EGFR-TKIs)
[16, 24, 26, 35–38].
Impact of cfDNA concentration on the survival
of NSCLC
Six studies reported the median progression-free
survival (PFS) time in NSCLC patients according to
different cfDNA concentrations (high or low) [19, 20, 27,
29, 30, 32]. As showed in Figure 2A, patients with high
levels of cfDNA usually had shorter PFS time than those
with low cfDNA concentrations. In addition, the pooled
HR for PFS was 1.32 (95% CI, 1.02-1.71; P = 0.038),
suggesting that high cfDNA concentration was a good
predictor of poor PFS (Figure 2B). For overall survival
(OS), 6 of 7 studies reported shorter median OS times in
NSCLC patients with higher cfDNA concentration (Figure
3A). Similar to the results of PFS, higher levels of cfDNA
indicated lower overall survival rates with a pooled HR
of 1.64 (95% CI, 1.26-2.15; P = 0.000) (Figure 3B).
However, high heterogeneities were presented in these
analyses (I2 = 73.6%; P = 0.000 for PFS; I2 = 75.5%; P =
0.000 for OS).
As clinical stages and therapeutic regimens are
correlated with patient’s prognosis, they may bring
heterogeneity to the overall analysis. Consequently,
we focused on these two confounding variables in our
subgroup analysis. As showed in Table 1, the majority of
studies considered patients with advanced clinical stages
(stage III-IV). Thus, we combined studies that focused
on NSCLC patients with advanced stages to have a more
homogenic group. The pooled HRs for PFS and OS were
1.29 (95% CI, 1.02-1.65; P = 0.035; I2 = 71.7%; Figure
4A) and 1.64 (95% CI, 1.19-2.25; P = 0.002; I
2
= 81.1%;
Figure 4B), respectively. We further performed another
subgroup analysis according to the therapeutic regimens.
As chemotherapy was the most commonly used treatment
method in these studies, we then limited the analysis to
studies considering patients treated with chemotherapy.
The signicant association could also be observed for both
PFS (HR, 1.41; 95% CI, 1.06-1.89; P = 0.020; I
2
= 76.0%;
Figure 5A) and OS (HR, 1.83; 95% CI, 1.31-2.54; P =
0.000; I2 = 82.3%; Figure 5B).
Impact of KRAS genotype detected in cfDNA on
the survival of NSCLC
The correlation between KRAS genotype detected in
cfDNA with survival in NSCLC patients was evaluated in
four studies. The combined HR for PFS was 1.30 (95%
CI, 0.66-2.56; P = 0.450), suggesting that there were
no signicant differences between patients with KRAS
mutation and those with wild-type genotype with respect
to PFS (Figure 6A). Moreover, our study failed to reveal
signicant difference for OS when NSCLC patients were
grouped according to KRAS genotype detected in cfDNA
(HR, 1.05; 95% CI, 0.49-2.25; P = 0.892; Figure 6B).
Thus, KRAS genotype detected in cfDNA might not be a
prognostic factor for survival in NSCLC patients.
Impact of EGFR genotype detected in cfDNA on
response to EGFR-TKIs
Seven studies evaluated whether EGFR genotype
detected in cfDNA could act as a predictor of response to
EGFR-TKIs. As showed in Figure 7, the pooled OR for
objective response rates (ORR) was 1.96 (95% CI, 1.59-
2.42; P = 0.000; I2 = 71.4%). Our results suggested that
Oncotarget44585
www.impactjournals.com/oncotarget
Table 1: Characteristics of studies included in this meta-analysis
First author Country No. Clinical
stage
Therapeutic
regimen
cfDNA
assessments cfDNA analysis Clinical factors
Catarino(2012)9Portugal 104 I-IV chemotherapy qPCR(hTERT)
quantication(H/L)
OS
Tissot(2015)19 France 218 III-IV chemotherapy PicoGreen
dsDNA Kit
quantication(H/L)
PFS, OS
Nygaard(2014)20 Denmark 58 III-IV chemotherapy ARMS-qPCR
quantication(H/L)
PFS, OS
KRAS
mutation(+/-) PFS, OS
Bortolin(2015)21 Italy 22 I
stereotactic
body
radiotherapy
qPCR(hTERT)
quantication(H/L)
PFS, OS
Li(2016)22 America 101 III-IV chemotherapy qPCR(β-Actin)
quantication(H/L)
PFS, OS
Wang(2014)27 China 134 III-IV EGFR-TKI ARMS/
Scorpion assay
quantication(H/L)
PFS, OS
EGFR
mutation(+/-) PFS, OS
Vinayanuwattikun
(2013)28 Thailand 58 III-IV chemotherapy qPCR(GAPDH)
quantication(H/L)
OS
Sirera(2011)29 Spain 446 III-IV chemotherapy qPCR(hTERT)
quantication(H/L)
PFS, OS
Lee(2011)30 Korea 134 III-IV EGFR-TKI or
chemotherapy qPCR(β-Actin)
quantication(H/L)
PFS, OS
Ludovini(2008)31 Italy 76 I-III surgery+
chemotherapy qPCR(hTERT)
quantication(H/L)
PFS, OS
Camps(2006)32 Spain 99 III-IV chemotherapy qPCR(hTERT)
quantication(H/L)
PFS, OS
Gautschi(2004)33 Switzerland 185 I-IV chemotherapy Fluorogenic
qPCR
quantication(H/L)
OS
Nygaard(2013)23 Denmark 246 II-IV chemotherapy ARMS-qPCR KRAS
mutation(+/-) PFS, OS
Camps(2005)25 Spain 67 III-IV chemotherapy RFLP-PCR KRAS
mutation(+/-) PFS, OS
Gautschi(2007)34 Switzerland 175 I-IV surgery+
chemotherapy RFLP-PCR KRAS
mutation(+/-) OS
Bai(2009)16 China 102 III-IV EGFR-TKI DHPLC EGFR
mutation(+/-) ORR
Kimura(2007)24 Japan 42 III-IV EGFR-TKI DNA
sequencing
EGFR
mutation(+/-) ORR
Douillard(2014)26 Multicenter 102 III-IV EGFR-TKI EGFR RGQ
PCR kit
EGFR
mutation(+/-) ORR
He(2009)35 China 45 I-IV EGFR-TKI Mutant-
enriched PCR
EGFR
mutation(+/-) ORR
(Continued )
Oncotarget44586
www.impactjournals.com/oncotarget
Figure 1: Flow diagram of study selection.
First author Country No. Clinical
stage
Therapeutic
regimen
cfDNA
assessments cfDNA analysis Clinical factors
Kimura(2006)36 Japan 27 III-IV EGFR-TKI DNA
sequencing
EGFR
mutation(+/-) ORR
Kim(2013)37 Korea 22 III-IV EGFR-TKI PNA-LNA PCR EGFR
mutation(+/-) ORR
Li(2014)38 China 55 III-IV EGFR-TKI ARMS-qPCR EGFR
mutation(+/-) ORR
Abbreviation: No., number; cfDNA, circulating cell-free DNA; qPCR, quantitative polymerase chain reaction; hTERT,
human telomerase reverse transcriptase; ARMS, amplication refractory mutation system; GAPDH, glyceraldehyde-
phosphate dehydrogenase; RFLP, restricted fragment length polymorphisms; DHPLC, denaturing high-performance liquid
chromatography; PNA-LNA, peptide nucleic acid-locked nucleic acid; EGFR-TKI, epidermal growth factor receptor-
tyrosine kinase inhibitor; H/L, high/low; +/-, mutation/wide-type; OS, overall survival; PFS, progression-free survival;
ORR, objective response rate.
Oncotarget44587
www.impactjournals.com/oncotarget
patients with EGFR activating mutation in cfDNA had a
greater chance of response to EGFR-TKIs. Thus, EGFR
genotype detected in cfDNA may be a good predictor of
response to EGFR-TKIs for NSCLC patients.
Publication bias
We assessed the publication bias by visually
assessing a funnel plot for asymmetry and by
quantitatively performing Begg’s test and Egger’s test. As
shown in Figure 8, there was no clear evidence of funnel
plot asymmetry by visual assessment. Both Begg’s test
and Egger’s test revealed that no publication bias was
found when OS was analyzed (Begg’s test, p = 0.266,
Egger’s test, p = 0.286). The Egger’s test revealed a slight
publication bias when PFS was analyzed (Begg’s test,
p = 0.119, Egger’s test, p = 0.035). Thus, no signicant
publication bias existed in this study.
DISCUSSION
Non-invasive approaches, usually based on plasma
or serum samples, have showed great potential for
treatment monitoring in NSCLC patients [47]. cfDNA,
as an easily acquired liquid biomarker and a potential
surrogate for the entire tumor genome, may provide
Figure 2: Progression-free survival (PFS) according to cfDNA concentration in NSCLC patients. A. Median PFS time
according to cfDNA concentration. B. Forest plot of hazard ratio (HR) for the impact of cfDNA concentration on PFS.
Oncotarget44588
www.impactjournals.com/oncotarget
complementary roles in predicting treatment response
and survival of NSCLC patients. Many studies have
investigated the usefulness of cfDNA as a screening tool
for NSCLC. However, the predictive or prognostic role
of cfDNA remains to be conrmed. In this study, we
provided the evidence that high levels of cfDNA were
signicantly associated with poor survival in NSCLC. In
addition, our study indicated that cfDNA could act as a
promising predictive factor for response to EGFR-TKIs in
NSCLC patients.
To the best of our knowledge, this is the rst
comprehensive meta-analysis to conrm the prognostic
role of cfDNA concentration in NSCLC. Our study
suggested that NSCLC patients with higher levels of
cfDNA tend to have shorter PFS and OS time. One
explanation for our results might be that total cfDNA was
able to reect the underlying tumor burden. Many studies
had indicated that tumor cell lysate is the main source
of the DNA found in plasma or serum [12]. Besides, the
amount of cfDNA in the blood was signicantly higher
in NSCLC patients than that in healthy controls [9, 48].
What’s more, cfDNA levels were associated with tumor
volume, tumor stage, lymph node involvement and tumor
responses [13]. Newman et al. found that levels of cfDNA
Figure 3: Overall survival (OS) according to cfDNA concentration in NSCLC patients. A. Median OS time according to
cfDNA concentration. B. Forest plot of hazard ratio (HR) for the impact of cfDNA concentration on OS.
Oncotarget44589
www.impactjournals.com/oncotarget
signicantly correlated with tumor volume and provided
earlier response assessment than radiographic approaches
[49]. Thus, patients with higher tumor load might have
more intensive cfDNA released to the blood and cfDNA
levels could reect the tumor burden. On the other hand,
cfDNA levels can be regulated by treatment-caused cell
death. In NSCLC patients, an obvious transient rise
in cfDNA concentrations occurred immediately after
treatment and then it was followed by a rapid decrease
[50]. It suggested that cell death caused by treatment could
release cfDNA, which decreased as the tumor regressed.
These observations revealed that cfDNA levels in plasma
or serum were able to reect the tumor load. Thus, cfDNA
can be a surrogate for tumor burden, making it become a
valuable prognostic factor for patients with NSCLC.
Targeted therapy based on molecular
characterizations has greatly inuenced the treatment
strategies in NSCLC. Gene mutation analyses are the
commonly used predictive biomarkers for selecting
NSCLC patients to receive targeted agents. However,
the current mutation analyses are often based on tumor
tissues and have many limitations. First, the accessibility
of tumor tissues is not always satisfactory as most
NSCLC patients are diagnosed with advanced stages and
Figure 4: Forest plot of hazard ratio (HR) for the impact of cfDNA concentration on progression-free survival (PFS)
and overall survival (OS) in NSCLC patients with advanced stages. A. The impact of cfDNA concentration on PFS. B. The
impact of cfDNA concentration on OS.
Oncotarget44590
www.impactjournals.com/oncotarget
unsuitable to provide tissues through invasive surgery
or biopsy. Second, surgery and biopsy are not without
clinical complications. The adverse events rate for
thoracic biopsy was reported to be approximately 20%
[51]. Furthermore, some percentages of NSCLC patients
will develop resistance to molecular-targeted agents
[52, 53]. Assessing treatment resistance in real time by
repeated surgery or biopsy is not feasible. Considering
these limitations, exploring convenient and less invasive
techniques to monitor the therapeutic response and
effects in NSCLC is urgently needed. Due to its nature
of minimal invasiveness, cfDNA is a promising source
for gene mutation analyses. In this study, we analyzed the
impact of KRAS and EGFR genotype presented in cfDNA
on the survival and response to EGFR-TKIs in NSCLC
patients. Approximately 15-25% of patients with NSCLC
have KRAS mutations, resulting in constitutive activation
of KRAS signaling pathways. Many studies reported that
KRAS mutation could predict the poor outcomes of EGFR-
TKIs treatment and chemotherapy, but several studies
argued that KRAS mutation was not associated with the
outcome of NSCLC patients [54]. A meta-analysis aimed
to clarify the prognostic and predictive value of KRAS
mutation in NSCLC was carried out recently [54]. Its
Figure 5: Forest plot of hazard ratio (HR) for the impact of cfDNA concentration on progression-free survival (PFS)
and overall survival (OS) in NSCLC patients treated with chemotherapy. A. The impact of cfDNA concentration on PFS. B.
The impact of cfDNA concentration on OS.
Oncotarget44591
www.impactjournals.com/oncotarget
results showed that KRAS mutation was signicantly
associated with worse OS and disease-free survival
(DFS) in early stage NSCLC, and with inferior outcomes
of EGFR-TKIs treatment and chemotherapy. However,
statistical differences in DFS and PFS of chemotherapy
and response rates to EGFR-TKIs or chemotherapy were
not met when EGFR mutant patients were excluded.
Our results indicated that KRAS mutations detected in
cfDNA might not be a prognostic factor for the survival of
NSCLC patients. One explanation might be that mutations
of KRAS and EGFR were generally mutually exclusive in
NSCLC [55, 56]. Most EGFR mutations were existed in
KRAS wild-type patients, which might bias the results
toward an overestimation of the prognostic and predictive
value of KRAS mutation. Another reason might be that the
amount of studies which assessed the prognostic value of
KRAS mutation presented in cfDNA in NSCLC was small.
Thus, the clinical signicance of KRAS mutation detected
in cfDNA is yet under debate. Future large-scaled trails are
still needed to improve our results.
Nowadays, EGFR-TKIs are the most successful
example of targeted therapy in NSCLC. EGFR gene
mutations are the standard biomarkers for selecting
NSCLC patients to receive EGFR-TKIs treatment. As
a high degree of correlation between EGFR mutations
detected in tumors and those presented in cfDNA has
been conrmed by two recent meta-analyses [17, 18],
EGFR mutation presented in cfDNA may also be useful
predictive markers for guiding NSCLC patients to receive
EGFR-TKIs. Indeed, several studies have analyzed the
association between cfDNA EGFR mutation status and
clinical outcomes. Goto et al. [44] found a signicant
correlation between cfDNA EGFR mutation status and
PFS. In cfDNA EGFR activating mutation subgroup,
NSCLC patients had longer PFS when they were treated
with getinib. Another research demonstrated that EGFR
Figure 6: Forest plot of hazard ratio (HR) for the impact of KRAS genotype detected in cfDNA on progression-free
survival (PFS) and overall survival (OS). A. The impact of KRAS genotype detected in cfDNA on PFS. B. The impact of KRAS
genotype detected in cfDNA on OS.
Oncotarget44592
www.impactjournals.com/oncotarget
mutation status in cfDNA was a good predictor for
PFS after EGFR-TKIs treatment [57]. Consistently, our
results showed that EGFR activating mutation in cfDNA
indicated a greater chance of response to EGFR-TKIs
in NSCLC patients. Thus, cfDNA EGFR mutation test
had a good ability to predict the efcacy of EGFR-TKIs
treatment. cfDNA might be a reliable material to guide
EGFR-TKIs treatment for NSCLC patients.
However, there were some limitations in our present
meta-analysis. Firstly, our analyses were based on the
literature, making our results less reliable than individual
patient data-based analysis. Secondly, a signicant
heterogeneity was presented in this study. When subgroup
analyses were performed in terms of clinical stages and
therapeutic regimens, the heterogeneity between studies
did not change obviously. The heterogeneity might partly
Figure 7: Forest plot of odds ratio (OR) for the impact of EGFR genotype detected in cfDNA on response to EGFR-
TKIs.
Figure 8: Funnel plot for the assessment of publication bias in this study. A. Funnel plot for 8 studies reporting progression-free
survival (PFS). B. Funnel plot for 11 studies reporting overall survival (OS).
Oncotarget44593
www.impactjournals.com/oncotarget
come from other variations, such as techniques that were
adopted to detect cfDNA. Future standardization of
cfDNA assessment would hopefully solve this problem.
Thirdly, studies that could not provide sufcient data for
extracting OR or HR were excluded. The exclusion of
these studies might make the pooled estimates differ from
their true value on some level.
In view of this study, our ndings suggested that
cfDNA could act as a predictive and prognostic biomarker
for patients with NSCLC. High levels of cfDNA were
signicantly associated with poor PFS and OS in NSCLC.
In addition, EGFR activating mutation status in cfDNA
indicated a greater chance of response to EGFR-TKIs. In
conclusion, cfDNA had a prognostic and predictive value
for NSCLC patients, which might help to dene high risk
patients and guide clinical decision making. However,
considering the limitations of a literature-based meta-
analysis, these results need to be validated and updated by
future large-scaled researches.
MATERIALS AND METHODS
Literature searches
Electronic searches for relevant articles in PubMed,
Embase, and Web of Science databases were conducted
in January 2016. The search strategy was generated by
combining key words related to cfDNA (‘circulating
cell-free DNA or ‘plasma cell-free DNA or ‘serum cell-
free DNA’) and NSCLC (‘non small cell lung cancer’ or
‘NSCLC’). Moreover, we manually searched the reference
lists of relevant articles for additional publications.
Inclusion criteria
Studies were included in this meta-analysis if they
met the following criteria: (1) all patients recruited in the
study were diagnosed with NSCLC; (2) the predictive
or prognostic value of cfDNA was evaluated; (3) only
English-language studies were included; (4) the HR or
OR and their corresponding 95% CIs were described or
could be statistically extracted; (5) When several studies
reported the same patient population, the newest or most
informative study was included.
Data extraction
Data extraction was performed independently by 2
reviewers and disagreements among them were resolved
by consensus. The following information was extracted
from each study: rst author’s name, publication year,
country of origin, number of patients, therapeutic
regimen, cfDNA assessment (methods), cfDNA analysis
(quantication and molecular characterization) and
clinical factors (PFS, OS and ORR).
Statistical analysis
HR and its 95% CIs were used to estimate the
prognostic value of cfDNA. OR and its 95% CIs were
adopted to describe the correlation between cfDNA status
and objective response rates. The individual HR or OR
estimates were combined into an overall HR or OR, and
the results were presented in the form of a forest plot.
Pooled effect sizes were considered to be signicantly
different if their 95% CIs did not include 1 (p < 0.05).
HR > 1 implied a poor survival and OR > 1 indicated a
greater chance of objective response. Median pooled PFS
and OS were summarized using descriptive statistics.
The heterogeneity between studies was assessed by the
Cochran Q test and I2 test. When Cochran Q test P value
was 0.10 and I2 test I2 value was 50%, statistically
signicant heterogeneity was considered to be present.
Fixed effects models were employed when heterogeneity
was absent; otherwise, random effects models were
adopted. Funnel plots, Begg’s test, and Egger’s test were
performed to detect publication bias. All analyses were
carried out by using Stata Statistical Software, version
12.0 (Stata Corporation, College Station, TX, USA).
CONFLICTS OF INTEREST
The authors declared no conicts of interest.
REFERENCES
1.
Torre LA, Bray F, Siegel RL, et al. Global cancer statistics,
2012. CA Cancer J Clin. 2015; 65:87-108.
2.
Howlader N, Noone AM, Krapcho M, et al. SEER Cancer
Statistics Review, 1975-2012, National Cancer Institute.
Bethesda, MD, http://seer.cancer.gov/csr/1975_2012/,
based on November 2014 SEER data submission, posted to
the SEER web site, April 2015.
3.
De Mattos-Arruda L, Caldas C. Cell-free circulating tumour
DNA as a liquid biopsy in breast cancer. Mol Oncol. 2015.
doi: 10.1016/j.molonc.2015.12.001. [Epub ahead of print].
4.
Gray ES, Rizos H, Reid AL et al. Circulating tumor DNA to
monitor treatment response and detect acquired resistance
in patients with metastatic melanoma. Oncotarget. 2015;
6:42008-42018. doi: 10.18632/oncotarget.5788.
5.
Carpinetti P, Donnard E, Bettoni F et al. The use of
personalized biomarkers and liquid biopsies to monitor
treatment response and disease recurrence in locally advanced
rectal cancer after neoadjuvant chemoradiation. Oncotarget.
2015; 6:38360-38371. doi: 10.18632/oncotarget.5256.
6.
Jiang T, Ren S, Zhou C. Role of circulating-tumor DNA
analysis in non-small cell lung cancer. Lung Cancer. 2015;
90:128-134.
7.
Xia S, Kohli M, Du M et al. Plasma genetic and genomic
abnormalities predict treatment response and clinical
Oncotarget44594
www.impactjournals.com/oncotarget
outcome in advanced prostate cancer. Oncotarget. 2015;
6:16411-16421. doi: 10.18632/oncotarget.3845.
8.
Stroun M, Maurice P, Vasioukhin V et al. The origin and
mechanism of circulating DNA. Ann N Y Acad Sci. 2000;
906:161-168.
9.
Catarino R, Coelho A, Araujo A et al. Circulating DNA:
diagnostic tool and predictive marker for overall survival of
NSCLC patients. PLoS One. 2012; 7:e38559.
10. Szpechcinski A, Chorostowska-Wynimko J, Struniawski R
et al. Cell-free DNA levels in plasma of patients with non-
small-cell lung cancer and inammatory lung disease. Br J
Cancer. 2015; 113:476-483.
11.
Szpechcinski A, Rudzinski P, Kupis W et al. Plasma
cell-free DNA levels and integrity in patients with chest
radiological ndings: NSCLC versus benign lung nodules.
Cancer Lett. 2016. doi: 10.1016/j.canlet.2016.02.002. [Epub
ahead of print].
12.
Zhang R, Shao F, Wu X et al. Value of quantitative analysis
of circulating cell free DNA as a screening tool for lung
cancer: a meta-analysis. Lung Cancer. 2010; 69:225-31.
13. Nie K, Jia Y, Zhang X. Cell-free circulating tumor DNA in
plasma/serum of non-small cell lung cancer. Tumour Biol.
2015; 36:7-19.
14.
Zhao X, Han RB, Zhao J et al. Comparison of epidermal
growth factor receptor mutation statuses in tissue and
plasma in stage I-IV non-small cell lung cancer patients.
Respiration. 2013; 85:119-125.
15.
Ishii H, Azuma K, Sakai K et al. Digital PCR analysis
of plasma cell-free DNA for non-invasive detection of
drug resistance mechanisms in EGFR mutant NSCLC:
Correlation with paired tumor samples. Oncotarget. 2015;
6:30850-30858. doi: 10.18632/oncotarget.5068.
16.
Bai H, Mao L, Wang HS et al. Epidermal growth factor
receptor mutations in plasma DNA samples predict tumor
response in Chinese patients with stages IIIB to IV non-
small-cell lung cancer. J Clin Oncol. 2009; 27:2653-2659.
17.
Qiu M, Wang J, Xu Y et al. Circulating tumor DNA is
effective for the detection of EGFR mutation in non-
small cell lung cancer: a meta-analysis. Cancer Epidemiol
Biomarkers Prev. 2015; 24:206-212.
18.
Luo J, Shen L, Zheng D. Diagnostic value of circulating
free DNA for the detection of EGFR mutation status in
NSCLC: a systematic review and meta-analysis. Sci Rep.
2014; 4:6269.
19.
Tissot C, Toffart AC, Villar S et al. Circulating free DNA
concentration is an independent prognostic biomarker in
lung cancer. Eur Respir J. 2015; 46:1773-1780.
20.
Dowler Nygaard A, Spindler KL, Pallisgaard N et al. Levels
of cell-free DNA and plasma KRAS during treatment of
advanced NSCLC. Oncol Rep. 2014; 31:969-974.
21.
Bortolin MT, Tedeschi R, Bidoli E et al. Cell-free DNA
as a prognostic marker in stage I non-small-cell lung
cancer patients undergoing stereotactic body radiotherapy.
Biomarkers. 2015; 20:422-428.
22.
Li BT, Drilon A, Johnson ML et al. A prospective study
of total plasma cell-free DNA as a predictive biomarker
for response to systemic therapy in patients with advanced
non-small-cell lung cancersdagger. Ann Oncol. 2016;
27:154-159.
23.
Nygaard AD, Garm Spindler KL, Pallisgaard N et al.
The prognostic value of KRAS mutated plasma DNA in
advanced non-small cell lung cancer. Lung Cancer. 2013;
79:312-317.
24.
Kimura H, Suminoe M, Kasahara K et al. Evaluation of
epidermal growth factor receptor mutation status in serum
DNA as a predictor of response to getinib (IRESSA). Br J
Cancer. 2007; 97:778-784.
25.
Camps C, Sirera R, Bremnes R et al. Is there a prognostic
role of K-ras point mutations in the serum of patients with
advanced non-small cell lung cancer? Lung Cancer. 2005;
50:339-346.
26. Douillard JY, Ostoros G, Cobo M et al. Getinib treatment
in EGFR mutated caucasian NSCLC: circulating-free tumor
DNA as a surrogate for determination of EGFR status. J
Thorac Oncol. 2014; 9:1345-1353.
27.
Wang S, Han X, Hu X et al. Clinical signicance of
pretreatment plasma biomarkers in advanced non-small
cell lung cancer patients. Clin Chim Acta. 2014; 430:63-70.
28.
Vinayanuwattikun C, Winayanuwattikun P, Chantranuwat
P et al. The impact of non-tumor-derived circulating nucleic
acids implicates the prognosis of non-small cell lung cancer.
J Cancer Res Clin Oncol. 2013; 139:67-76.
29.
Sirera R, Bremnes RM, Cabrera A et al. Circulating DNA
is a useful prognostic factor in patients with advanced non-
small cell lung cancer. J Thorac Oncol. 2011; 6:286-290.
30.
Lee YJ, Yoon KA, Han JY et al. Circulating cell-
free DNA in plasma of never smokers with advanced
lung adenocarcinoma receiving getinib or standard
chemotherapy as rst-line therapy. Clin Cancer Res. 2011;
17:5179-5187.
31.
Ludovini V, Pistola L, Gregorc V et al. Plasma DNA,
microsatellite alterations, and p53 tumor mutations are
associated with disease-free survival in radically resected
non-small cell lung cancer patients: a study of the perugia
multidisciplinary team for thoracic oncology. J Thorac
Oncol. 2008; 3:365-373.
32.
Camps C, Sirera R, Bremnes RM et al. Quantication in
the serum of the catalytic fraction of reverse telomerase:
a useful prognostic factor in advanced non-small cell lung
cancer. Anticancer Res. 2006; 26:4905-4909.
33.
Gautschi O, Bigosch C, Huegli B et al. Circulating
deoxyribonucleic Acid as prognostic marker in non-small-
cell lung cancer patients undergoing chemotherapy. J Clin
Oncol. 2004; 22:4157-4164.
34.
Gautschi O, Huegli B, Ziegler A et al. Origin and prognostic
value of circulating KRAS mutations in lung cancer
patients. Cancer Lett. 2007; 254:265-273.
Oncotarget44595
www.impactjournals.com/oncotarget
35.
He C, Liu M, Zhou C et al. Detection of epidermal growth
factor receptor mutations in plasma by mutant-enriched
PCR assay for prediction of the response to getinib in
patients with non-small-cell lung cancer. Int J Cancer. 2009;
125:2393-2399.
36.
Kimura H, Kasahara K, Kawaishi M et al. Detection of
epidermal growth factor receptor mutations in serum as
a predictor of the response to getinib in patients with
non-small-cell lung cancer. Clin Cancer Res. 2006;
12:3915-3921.
37.
Kim ST, Sung JS, Jo UH et al. Can mutations of EGFR
and KRAS in serum be predictive and prognostic markers
in patients with advanced non-small cell lung cancer
(NSCLC)? Med Oncol. 2013; 30:328.
38.
Li X, Ren R, Ren S et al. Peripheral blood for epidermal
growth factor receptor mutation detection in non-small cell
lung cancer patients. Transl Oncol. 2014; 7:341-8.
39.
Camps C, Jantus-Lewintre E, Cabrera A et al. The
identication of KRAS mutations at codon 12 in plasma
DNA is not a prognostic factor in advanced non-small cell
lung cancer patients. Lung Cancer. 2011; 72:365-369.
40.
Karachaliou N, Mayo-de las Casas C, Queralt C et al.
Association of EGFR L858R Mutation in Circulating Free
DNA With Survival in the EURTAC Trial. JAMA Oncol.
2015; 1:149-157.
41.
Wang Z, Chen R, Wang S et al. Quantication and dynamic
monitoring of EGFR T790M in plasma cell-free DNA
by digital PCR for prognosis of EGFR-TKI treatment in
advanced NSCLC. PLoS One. 2014; 9:e110780.
42.
Punnoose EA, Atwal S, Liu W et al. Evaluation of
circulating tumor cells and circulating tumor DNA in non-
small cell lung cancer: association with clinical endpoints
in a phase II clinical trial of pertuzumab and erlotinib. Clin
Cancer Res. 2012; 18:2391-2401.
43.
Zhuo ML, Wu MN, Zhao J et al. Epidermal growth
factor receptor genotype in plasma DNA and outcome
of chemotherapy in the Chinese patients with advanced
non-small cell lung cancer. Chin Med J (Engl). 2011;
124:3510-3514.
44.
Goto K, Ichinose Y, Ohe Y et al. Epidermal growth factor
receptor mutation status in circulating free DNA in serum:
from IPASS, a phase III study of getinib or carboplatin/
paclitaxel in non-small cell lung cancer. J Thorac Oncol.
2012; 7:115-121.
45.
Thress KS, Brant R, Carr TH et al. EGFR mutation detection
in ctDNA from NSCLC patient plasma: A cross-platform
comparison of leading technologies to support the clinical
development of AZD9291. Lung Cancer. 2015; 90:509-515.
46.
Nygaard AD, Holdgaard PC, Spindler KL et al. The
correlation between cell-free DNA and tumour burden was
estimated by PET/CT in patients with advanced NSCLC. Br
J Cancer. 2014; 110:363-368.
47.
Sun W, Yuan X, Tian Y et al. Non-invasive approaches
to monitor EGFR-TKI treatment in non-small-cell lung
cancer. J Hematol Oncol. 2015; 8:95.
48.
Ulivi P, Mercatali L, Casoni GL et al. Multiple marker
detection in peripheral blood for NSCLC diagnosis. PLoS
One. 2013; 8:e57401.
49.
Newman AM, Bratman SV, To J et al. An ultrasensitive
method for quantitating circulating tumor DNA with broad
patient coverage. Nat Med. 2014; 20:548-554.
50.
Szpechcinski A, Chorostowska-Wynimko J, Kupis W et al.
Quantitative analysis of free-circulating DNA in plasma of
patients with resectable NSCLC. Expert Opin Biol Ther.
2012; 12:S3-9.
51.
Overman MJ, Modak J, Kopetz S et al. Use of research
biopsies in clinical trials: are risks and benets adequately
discussed? J Clin Oncol. 2013; 31:17-22.
52.
Janne PA. Challenges of detecting EGFR T790M in
getinib/erlotinib-resistant tumours. Lung Cancer. 2008;
60:S3-9.
53.
Russo A, Franchina T, Ricciardi GR et al. A decade of EGFR
inhibition in EGFR-mutated non small cell lung cancer
(NSCLC): Old successes and future perspectives. Oncotarget.
2015; 6:26814-26825. doi: 10.18632/oncotarget.4254.
54.
Pan W, Yang Y, Zhu H et al. KRAS mutation is a weak, but
valid predictor for poor prognosis and treatment outcomes
in NSCLC: A meta-analysis of 41 studies. Oncotarget.
2016;7:8373-88. doi: 10.18632/oncotarget.7080.
55.
Dogan S, Shen R, Ang DC et al. Molecular epidemiology
of EGFR and KRAS mutations in 3,026 lung
adenocarcinomas: higher susceptibility of women to
smoking-related KRAS-mutant cancers. Clin Cancer Res.
2012; 18:6169-6177.
56.
Boch C, Kollmeier J, Roth A et al. The frequency of
EGFR and KRAS mutations in non-small cell lung cancer
(NSCLC): routine screening data for central Europe from a
cohort study. BMJ Open. 2013; 3:e002560.
57.
Weber B, Meldgaard P, Hager H et al. Detection of EGFR
mutations in plasma and biopsies from non-small cell lung
cancer patients by allele-specic PCR assays. BMC Cancer.
2014; 14:294.
... In metastatic breast cancer patients, changes of cfDNA concentration in plasma correlated better with changes in tumour burden than did the classical marker CA 15-3 [12]. Numerous studies have also attempted to use cfDNA concentration as a prognostic marker [13][14][15][16][17][18]. One meta-analysis showed that high cfDNA concentrations in blood were associated with worse cancer-specific survival in stage III and IV of non-small cell lung cancer patients [13]. ...
... Numerous studies have also attempted to use cfDNA concentration as a prognostic marker [13][14][15][16][17][18]. One meta-analysis showed that high cfDNA concentrations in blood were associated with worse cancer-specific survival in stage III and IV of non-small cell lung cancer patients [13]. Similar results were also obtained in studies conducted on other types of cancers [14,15]. ...
Article
Full-text available
Introduction Assessment of renal tumour masses is based on conventional imaging studies (computer tomography or magnetic resonance), which does not allow characterisation of the histopathological type. Moreover, the prediction of prognosis in localised and metastatic renal cell carcinoma requires improvement as well. Analysis of circulating free DNA (cfDNA) in blood is one of the variants of liquid biopsy that may improve diagnostics and prognosis issues of patients with renal tumour masses suspected to be renal cell carcinoma. The aim of the study was to assess the diagnostic and prognostic role of preoperative cfDNA concentration in the plasma samples of clear cell renal cell carcinoma (ccRCC) patients. Material and methods The preoperative plasma cfDNA concentration was assessed in ccRCC patients (n = 46) and healthy individuals (control group) (n = 17). The circulating free DNA concentration was reflected by the 90 bp DNA fragments determined by real-time polymerase chain reaction. Results The median cfDNA concentration was significantly higher in ccRCC patients (n = 46) compared to the control g roup (n = 17) (2588 ±2554 copies/ml vs. 960 ±490 copies/ml, p < 0.01). In multivariate analysis, the preoperative plasma cfDNA concentration was the significant factor increasing the probability of ccRCC detection (OR: 1.003; 95% CI: 1.001–1.005). The median cfDNA concentration depended on the stage of ccRCC; it was higher in metastatic ccRCC patients (n = 11) compared to non-metastatic ccRCC patients (n = 35) (3619 ±4059 copies/ml vs. 2473 ±1378 copies/ml, p < 0.03). Kaplan-Meier survival analysis demon-strated that patients with high cfDNA values (above 2913 copies/ml) had significantly worse cancer-specific survival (HR: 4.5; 95% CI: 1.3–16.9, log-rank Mantel-Cox test p = 0.015). Conclusions Preoperative plasma cfDNA concentration has diagnostic and prognostic potential in ccRCC pa-tients.
... In non-small cell lung cancer (NSCLC), a meta-analysis of 22 studies found that patients with elevated cfDNA concentration tend to have shorter PFS and OS [66]. A study within that meta-analysis of 218 patients found that individuals in the top third of cfDNA concentration before treatment showed a significantly shorter PFS and OS than patients in the lower two-thirds. ...
... The meta-analysis for CRC noted that due to a lack of CEA measurements comparative conclusions cannot be made, but cfDNA could be a viable prognostic marker [64]. Total cfDNA also showed promise in NSCLC as a prognostic marker [66,67,70]. Total cfDNA concentration may also have utility in monitoring patients after treatment to detect recurrence or disease progression, with total cfDNA elevation being detected earlier than serum biomarkers [50,59,60,65]. ...
Article
Full-text available
The detection of circulating tumor DNA (ctDNA) in liquid biopsy samples as an oncological marker is being used in clinical trials at every step of clinical management. As ctDNA-based liquid biopsy kits are developed and used in clinics, companies work towards increased convenience, accuracy, and cost over solid biopsies and other oncological markers. The technology used to differentiate ctDNA and cell-free DNA (cfDNA) continues to improve with new tests and methodologies being able to detect down to mutant allele frequencies of 0.001% or 1/100,000 copies. Recognizing this development in technology, the FDA has recently given pre-market approval and breakthrough device designations to multiple companies. The purpose of this review is to look at the utility of measuring total cfDNA, techniques used to differentiate ctDNA from cfDNA, and the utility of different ctDNA-based liquid biopsy kits using relevant articles from PubMed, clinicaltrials.gov, FDA approvals, and company newsletters. Measuring total cfDNA could be a cost-effective, viable prognostic marker, but various factors do not favor it as a monitoring tool during chemotherapy. While there may be a place in the clinic for measuring total cfDNA in the future, the lack of standardization means that it is difficult to move forward with large-scale clinical validation studies currently. While the detection of ctDNA has promising standardized liquid biopsy kits from various companies with large clinical trials ongoing, their applications in screening and minimal residual disease can suffer from lower sensitivity. However, researchers are working towards solutions to these issues with innovations in technology, multi-omics, and sampling. With great promise, further research is needed before liquid biopsies can be recommended for everyday clinical management.
... Apart from these distinctive variations, levels of tumor-derived cfDNA among patients with the same cancer type and stage significantly differ [42,170]. In metastatic breast cancer, cfDNA exhibits a stronger correlation with tumor size compared to circulating tumor cells and the common protein biomarker, Cancer Antigen 15-3 [171]. ...
Article
Full-text available
Background: Cancer, characterized by the rapid and abnormal growth of cells affecting any part of the body, stands as the leading cause of death worldwide. Cell-free DNA (cfDNA) has garnered significant attention as a non-invasive liquid biopsy approach for disease detection, therapy evaluation, and prognosis. This review aims to provide a comprehensive exploration of cfDNA within the realm of oncology. It encompasses its diagnostic and therapeutic applications while identifying areas necessitating standardization and optimization. Methods: We reviewed existing literature to delve into the biological properties of cfDNA, exploring genetic and epigenetic aberrations found in various bodily fluids. Additionally, we explored its correlation with circulating tumor DNA (ctDNA). The review also encompasses preanalytical procedures and emerging technologies geared towards maximizing the complete potential of cfDNA. Results: Genetic and epigenetic markers have been identified in cfDNA across plasma, serum, and urine, presenting promising diagnostic and prognostic applications. Furthermore, ctDNA preserves genomic profiles akin to those found in corresponding tumor tissues, enabling a nuanced evaluation of tumor heterogeneity and mutation burdens. However, despite its potential, current methodologies suffer from a lack of standardization and optimization, thereby restraining the complete clinical utility of cfDNA. Conclusions: Although cfDNA stands as a compelling avenue for non-invasive early cancer diagnosis and therapy evaluation, unlocking its maximum potential requires methodological refinement and a deeper understanding of its biological characteristics. This review advocates for targeted research to standardize and optimize cfDNA analytical techniques, thereby enhancing its role in oncology.
... First, the accuracy of dd-cfDNA testing varies based on the technology and methods employed [28]. Second, clinical scenarios that cause organ injury and DNA release, such as pulmonary embolism, infection, autoimmune conditions, ischemia, and concomitant cancer, may lead to elevated dd-cfDNA levels in the plasma [29,30]. Clinicians must consider these factors to ensure accurate dd-cfDNA interpretation. ...
Article
Full-text available
Heart failure, particularly in its advanced stages, significantly impacts quality of life. Despite progress in Guideline-Directed Medical Therapy (GDMT) and invasive treatments, heart transplantation (HT) remains the primary option for severe cases. However, complications such as graft rejection present significant challenges that necessitate effective monitoring. Endomyocardial biopsy (EMB) is the gold standard for detecting rejection, but its invasive nature, associated risks, and healthcare costs have shifted interest in non-invasive techniques. Donor-derived cell-free DNA (dd-cfDNA) has gained attention as a promising non-invasive biomarker for monitoring graft rejection. Compared to EMB, dd-cfDNA detects graft rejection early and enables clinicians to adjust immunosuppression promptly. Despite its advantages, dd-cfDNA testing faces challenges, such as the need for specialized technology and potential inaccuracies due to other clinical conditions. Additionally, dd-cfDNA cannot yet differentiate between types of graft rejection, and its effectiveness in chronic rejection remains unclear. Research is ongoing to set precise standards for dd-cfDNA levels, which would enhance its diagnostic accuracy and help in clinical decisions. The article also points to the future of HT monitoring, which may involve combining dd-cfDNA with other biomarkers and integrating artificial intelligence to improve diagnostic capabilities and personalize patient care. Furthermore, it emphasizes both global and racial inequalities in dd-cfDNA testing and the ethical issues related to its use in transplant medicine.
... The role of cfDNA and NLR in the prognosis of advanced NSCLC has been demonstrated separately [28][29][30].However, in this study, we identified a high-risk subgroup with both high cfDNA and NLR by combining the two biomarkers, and found that they had the highest risk of disease progression. The complementary effects of these two biomarkers suggest the potential of using cfDNA and NLR in combination to predict prognosis in NSCLC patients in clinical practice. ...
Article
Full-text available
Background: Some research found that elevated plasma cell-free DNA (cfDNA) concentrations and poor prognosis are associated in non-small cell lung cancer (NSCLC). However, more studies need to be carried out to verify this conclusion. Therefore, this study investigated the relationship between cfDNA concentration and treatment outcomes including prognosis in patients with advanced NSCLC. Methods: We retrospectively collected medical records and cfDNA data from 160 patients with advanced NSCLC. Progression-free survival (PFS) were calculated using the Kaplan-Meier method and were compared between groups using the log rank test. Cox regression analysis was used for estimating the independent predictors of PFS. And we used logistic regression to evaluate the relationship between baseline biomarkers and efficacy. In our study, BT1 cfDNA, BT2 cfDNA, and BT3 cfDNA were defined as cfDNA concentration before the first treatment (baseline cfDNA concentration), cfDNA concentration before the second treatment, and cfDNA concentration before the third treatment, respectively. Results: Patients with low cfDNA (BT1 cfDNA < 15 (ng/mL)) were reported a significantly prolonged median progression-free survival (mPFS) compared with patients with patients with high cfDNA (BT1 cfDNA ≥ 15(ng/mL)) (mPFS: 14.6 vs. 8.3 months, P = 0.002), as well as patients with neutrophil/lymphocyte ratio (NLR)<2.98 (mPFS: 13.1 vs. 7.9 months, P = 0.023). In addition, Cox proportional hazards regression analysis identified independent indicators associated with PFS including BT1 cfDNA ≥ 15 (ng/mL), NLR ≥ 2.98 and extrapulmonary metastasis. The best cut-off value for BT3 cfDNA for predicting disease progression is 41.46 (ng/mL) (Area Under the Curve (AUC): 0.652, 95%CI: 0.516-0.788), achieving 90.7% sensitivity and 37.5% specificity for the prediction of disease progression. BT3 cfDNA (OR = 6.08, 95% CI: 1.94-19.57, P = 0.002) was an independent factor for disease progression in patients with advanced NSCLC. Conclusions: BT1 cfDNA may be a biomarker to assess the prognosis of advanced NSCLC. Patients with advanced NSCLC with lower cfDNA and NLR before treatment had a better prognosis. Increased BT3 cfDNA concentration was an independent factor of disease progression in advanced NSCLC patients. These findings may assist in identifying high-risk patients and guiding treatment strategies.
... The ctDNA recapitulates with accuracy the tumor characteristics and appears to be a diagnostic tool for many different solid tumors, for example, colorectal [143,144], endometrial [145,146], ovarian [78], breast [147], non-small cell lung cancer (NSCLC) [148][149][150], oropharyngeal [151], pancreatic [152], prostate [153,154] cancers, and melanoma [155]. ...
Article
Full-text available
Liquid biopsy is a diagnostic repeatable test, which in last years has emerged as a powerful tool for profiling cancer genomes in real-time with minimal invasiveness and tailoring oncological decision-making. It analyzes different blood-circulating biomarkers and circulating tumor DNA (ctDNA) is the preferred one. Nevertheless, tissue biopsy remains the gold standard for molecular evaluation of solid tumors whereas liquid biopsy is a complementary tool in many different clinical settings, such as treatment selection, monitoring treatment response, cancer clonal evolution, prognostic evaluation, as well as the detection of early disease and minimal residual disease (MRD). A wide number of technologies have been developed with the aim of increasing their sensitivity and specificity with acceptable costs. Moreover, several preclinical and clinical studies have been conducted to better understand liquid biopsy clinical utility. Anyway, several issues are still a limitation of its use such as false positive and negative results, results interpretation, and standardization of the panel tests. Although there has been rapid development of the research in these fields and recent advances in the clinical setting, many clinical trials and studies are still needed to make liquid biopsy an instrument of clinical routine. This review provides an overview of the current and future clinical applications and opening questions of liquid biopsy in different oncological settings, with particular attention to ctDNA liquid biopsy.
... We found that combining cfDNA levels at T 0 and T 1 showed a significant contribution to PFS time. The therapeutic utility of detecting mutations in cfDNA has been correlated to tumour burden [41], PFS and OS in NSCLC patients [42]. ctDNA EGFR mutations (exon 19 detection or exon 21 (L858R) mutations) are used to identify patients who may benefit from EGFR-TKIs [43]. ...
Article
Full-text available
Background Non-small cell lung cancer (NSCLC) often presents at an incurable stage, and majority of patients will be considered for palliative treatment at some point in their disease. Despite recent advances, the prognosis remains poor, with a median overall survival of 12-18 months. Liquid biopsy-based biomarkers have emerged as potential candidates for predicting prognosis and response to therapy in NSCLC patients. This pilot study evaluated whether combining circulating tumour cells and clusters (CTCs) and cell-free DNA (cfDNA) can predict progression-free survival (PFS) in NSCLC patients. Methods CTC and cfDNA/ctDNA from advanced stage NSCLC patients were measured at study entry (T⁰) and 3-months post-treatment (T¹). CTCs were enriched using a spiral microfluidic chip and characterised by immunofluorescence. ctDNA was assessed using an UltraSEEK® Lung Panel. Kaplan-Meier plots were generated to investigate the contribution of the presence of CTC/CTC clusters and cfDNA for PFS. Cox proportional hazards analysis compared time to progression versus CTC/CTC cluster counts and cfDNA levels. Results Single CTCs were found in 14 out of 25 patients, while CTC clusters were found in 8 out of the 25 patients at T⁰. At T¹, CTCs were found in 7 out of 18 patients, and CTC clusters in 1 out of the 18 patients. At T⁰, CTC presence and the combination of CTC cluster counts with cfDNA levels were associated with shorter PFS, p=0.0261, p=0.0022, respectively. Conclusions Combining CTC cluster counts and cfDNA levels could improve PFS assessment in NSCLC patients. Our results encourage further investigation on the combined effect of CTC/cfDNA as a prognostic biomarker in a large cohort of advanced stage NSCLC patients.
... Numerous studies have found that quantification of baseline ctDNA predicted prognosis of patients harboring EGFR mutations. 17,18 Recent studies have shown that ctDNA clearance was of prognostic significance, in which molecular responders with undetectable ctDNA after targeted therapy had significantly longer survival than non-responders. 19,20 Therefore, molecular evaluation of ctDNA clearance helps to identify a group of patients that would benefit from EGFR-targeted therapy. ...
Article
Full-text available
Objective: Circulating tumor DNA (ctDNA) monitoring proves to be a promising approach to assess response and predict survival in epidermal growth factor receptor (EGFR)-mutated non-small-cell lung cancer (NSCLC) patients treated with tyrosine kinase inhibitors (TKIs). However, whether the dynamic changes in ctDNA EGFR mutation status have the same predictive value as ctDNA remains unknown. This study aims to explore the predictive value of dynamic changes in both ctDNA and ctDNA EGFR status. Methods: A retrospective analysis was performed using 91 ctDNA samples from a cohort of 28 patients who were diagnosed with EGFR-mutated NSCLC and treated with EGFR-TKIs, including 14 patients treated with first-/second-generation TKIs and 14 treated with osimertinib. Blood samples at baseline (BL), within 4 weeks after TKI initiation (Week4), within 12 weeks before progression (pre-PD), and at progression were collected. The relationship alternatives in ctDNA status, ctDNA EGFR status and response to EGFR-TKIs as well as progression-free survival (PFS) were analyzed. Results: We categorized 20 BL-ctDNA positive patients with available Week4-ctDNA into two groups: ctDNA-clearance (N = 7, 35%) and ctDNA-non-clearance (N = 13, 65%). The ctDNA-clearance group had better PFS than the ctDNA-non-clearance group (ctDNA-clearance vs. ctDNA-non-clearance, p = 0.091, hazard ratio [HR] = 0.42, 95% confidence interval [CI] = 0.15-1.19). According to Week4-EGFR status, we observed that PFS was significantly longer in EGFR-clearance patients than EGFR-non-clearance groups, (p = 0.011, HR = 0.23, 95% CI = 0.08-0.72). We then categorized patients into three subgroups according to Week4-ctDNA and Week4-EGFR status: non-clearance (N = 9), only-EGFR-clearance (concomitant alterations non-clearance) (N = 4), and all-clearance (N = 7). The nonclearance group had a significantly worse PFS than the all-clearance group (median PFS = 5.07 vs. 11.40 months, p = 0.029, HR = 3.45, 95% CI = 1.05-11.49). The only-EGFR-clearance group had a similar PFS to the all-clearance group (p = 0.607), which was longer than that of the non-clearance group (median PFS = 9.20 vs. 5.07 months, p = 0.060, HR = 0.25, 95% CI = 0.05-1.18). We found that the all-clearance group had a similar objective response rate (ORR) to the only-EGFR-clearance group (p = 1.000) and a higher ORR than the non-clearance group (p = 0.012). Conclusion: Monitoring of EGFR clearance in ctDNA is promising and cost-effective in assessing response and predicting survival in EGFR-mutated NSCLC patients treated with EGFR-TKIs, with similar predictive value to ctDNA surveillance.
Article
Non-small-cell lung cancer (NSCLC) is a major cause of cancer-related death worldwide. In recent years, the discovery of actionable molecular alterations has changed the treatment paradigm of the disease. Tissue biopsies have been the gold standard for the identification of targetable alterations but present several limitations, calling for alternatives to detect driver and acquired resistance alterations. Liquid biopsies reveal great potential in this setting and also in the evaluation and monitoring of treatment response. However, several challenges currently hamper its widespread adoption in clinical practice. This perspective article evaluates the potential and challenges associated with liquid biopsy testing, considering a Portuguese expert panel dedicated to thoracic oncology point of view, and providing practical insights for its implementation based on the experience and applicability in the Portuguese context.
Article
Cell-free DNA (cfDNA) are un-encapsulated DNA fragments present in biological fluids ranging in an average size of up to 200 base pairs. The novel use of cfDNA is a prime candidate in the diagnostic and prognostic approach to unveiling many inflammatory diseases, especially cancer. Moreover, their potential as biomarkers is due to their ubiquitous presence in the body, non-invasive nature, and aiding in a different autopsy method. This review will focus on the diagnostic and prognostic potential of cfDNA as non-invasive biomarkers in oncology.
Article
Full-text available
Mutation of oncogene KRAS is common in non-small cell lung cancer (NSCLC), however, its clinical significance is still controversial. Independent studies evaluating its prognostic and predictive value usually drew inconsistent conclusions. Hence, We performed a meta-analysis with 41 relative publications, retrieved from multi-databases, to reconcile these controversial results and to give an overall impression of KRAS mutation in NSCLC. According to our findings, KRAS mutation was significantly associated with worse overall survival (OS) and disease-free survival (DFS) in early stage resected NSCLC (hazard ratio or HR=1.56 and 1.57, 95% CI 1.39-1.76 and 1.17-2.09 respectively), and with inferior outcomes of epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) treatment and chemotherapy (relative risk or RR=0.21 and 0.66 for objective response rate or ORR, 95% CI 0.12-0.39 and 0.54-0.81 respectively; HR=1.46 and 1.30 for progression-free survival or PFS, 95%CI 1.23-1.74 and 1.14-1.50 respectively) in advanced NSCLC. When EGFR mutant patients were excluded, KRAS mutation was still significantly associated with worse OS and PFS of EGFR-TKIs (HR=1.40 and 1.35, 95 % CI 1.21-1.61 and 1.11-1.64). Although KRAS mutant patients presented worse DFS and PFS of chemotherapy (HR=1.33 and 1.11, 95% CI 0.97-1.84 and 0.95-1.30), and lower response rate to EGFR-TKIs or chemotherapy (RR=0.55 and 0.88, 95 % CI 0.27-1.11 and 0.76-1.02), statistical differences were not met. In conclusion, KRAS mutation is a weak, but valid predictor for poor prognosis and treatment outcomes in NSCLC. There's a need for developing target therapies for KRAS mutant lung cancer and other tumors.
Article
Full-text available
Objective: To evaluate whether plasma cell-free DNA (cfDNA) was related to clinical outcome in inoperable stage I non-small cell lung cancer (NSCLC) patients undergoing stereotactic body radiotherapy (SBRT). Materials and methods: Plasma cfDNA was assessed at baseline, before the last day and 45 days after the end of SBRT, in 22 NSCLC patients. Twenty-two healthy controls were also evaluated. Results: Plasma cfDNA was higher in patients than in controls. An association with unfavourable disease-free survival was found for continuous baseline cfDNA increments (HR = 5.9, 95%CI: 1.7-19.8, p = 0.04). Conclusion: Plasma cfDNA may be a promising prognostic biomarker in high-risk NSCLC patients.
Article
Full-text available
Objectives: To assess the ability of different technology platforms to detect epidermal growth factor receptor (EGFR) mutations, including T790M, from circulating tumor DNA (ctDNA) in advanced non-small cell lung cancer (NSCLC) patients. Materials and methods: A comparison of multiple platforms for detecting EGFR mutations in plasma ctDNA was undertaken. Plasma samples were collected from patients entering the ongoing AURA trial (NCT01802632), investigating the safety, tolerability, and efficacy of AZD9291 in patients with EGFR-sensitizing mutation-positive NSCLC. Plasma was collected prior to AZD9291 dosing but following clinical progression on a previous EGFR-tyrosine kinase inhibitor (TKI). Extracted ctDNA was analyzed using two non-digital platforms (cobas(®) EGFR Mutation Test and therascreen™ EGFR amplification refractory mutation system assay) and two digital platforms (Droplet Digital™ PCR and BEAMing digital PCR [dPCR]). Results: Preliminary assessment (38 samples) was conducted using all four platforms. For EGFR-TKI-sensitizing mutations, high sensitivity (78-100%) and specificity (93-100%) were observed using tissue as a non-reference standard. For the T790M mutation, the digital platforms outperformed the non-digital platforms. Subsequent assessment using 72 additional baseline plasma samples was conducted using the cobas(®) EGFR Mutation Test and BEAMing dPCR. The two platforms demonstrated high sensitivity (82-87%) and specificity (97%) for EGFR-sensitizing mutations. For the T790M mutation, the sensitivity and specificity were 73% and 67%, respectively, with the cobas(®) EGFR Mutation Test, and 81% and 58%, respectively, with BEAMing dPCR. Concordance between the platforms was >90%, showing that multiple platforms are capable of sensitive and specific detection of EGFR-TKI-sensitizing mutations from NSCLC patient plasma. Conclusion: The cobas(®) EGFR Mutation Test and BEAMing dPCR demonstrate a high sensitivity for T790M mutation detection. Genomic heterogeneity of T790M-mediated resistance may explain the reduced specificity observed with plasma-based detection of T790M mutations versus tissue. These data support the use of both platforms in the AZD9291 clinical development program.
Article
Full-text available
Background: While previous studies have reported on the prognostic value of total plasma cell-free DNA (cfDNA) in lung cancers, few have prospectively evaluated its predictive value for systemic therapy response. Patients and methods: We conducted a prospective study to evaluate the association between changes in total cfDNA and radiologic response to systemic therapy in patients with stage IIIB/IV non-small cell lung cancers (NSCLCs). Paired blood collections for cfDNA and computed tomography (CT) assessments by RECIST v1.0 were performed at baseline and 6-12 weeks after therapy initiation. Total cfDNA levels were measured in plasma using quantitative real-time polymerase chain reaction. Associations between changes in cfDNA and radiologic response, progression-free survival (PFS), and overall survival (OS) were measured using Kruskal-Wallis and Kaplan-Meier estimates. Results: A total of 103 patients completed paired cfDNA and CT response assessments. Systemic therapy administered included cytotoxic chemotherapy in 57% (59/103), molecularly targeted therapy in 17% (17/103), and combination therapy in 26% (27/103). Median change in cfDNA from baseline to response assessment did not significantly differ by radiologic response categories of progression of disease, stable disease and partial response (P=0.10). However, using radiologic response as continuous variable, there was a weak positive correlation between change in radiologic response and change in cfDNA (Spearman correlation coefficient 0.21, P=0.03). Baseline cfDNA levels were not associated with PFS (HR=1.06, 95% CI 0.93-1.20, P=0.41) or OS (HR=1.04, 95% CI 0.93-1.17, P=0.51), neither were changes in cfDNA. Conclusions: In this large prospective study, changes in total cfDNA over time did not significantly predict radiologic response from systemic therapy in patients with advanced NSCLC. Pre-treatment levels of total cfDNA were not prognostic of survival. Total cfDNA level is not a highly specific predictive biomarker and future investigations in cfDNA should focus on tumor specific genomic alterations using expanded capabilities of next generation sequencing.
Article
Full-text available
Neoadjuvant chemoradiotherapy (nCRT) followed by surgery is the mainstay treatment for locally advanced rectal cancer. Variable degrees of tumor regression are observed after nCRT and alternative treatment strategies, including close surveillance without immediate surgery, have been investigated to spare patients with complete tumor regression from potentially adverse outcomes of radical surgery. However, clinical and radiological assessment of response does not allow accurate identification of patients with complete response. In addition, surveillance for recurrence is similarly important for these patients, as early detection of recurrence allows salvage resections and adjuvant interventions. We report the use of liquid biopsies and personalized biomarkers for monitoring treatment response to nCRT and detecting residual disease and recurrence in patients with rectal cancer. We sequenced the whole-genome of four rectal tumors to identify patient-specific chromosomal rearrangements that were used to monitor circulating tumor DNA (ctDNA) in liquid biopsies collected at diagnosis and during nCRT and follow-up. We compared ctDNA levels to clinical, radiological and pathological response to nCRT. Our results indicate that personalized biomarkers and liquid biopsies may not be sensitive for the detection of microscopic residual disease. However, it can be efficiently used to monitor treatment response to nCRT and detect disease recurrence, preceding increases in CEA levels and radiological diagnosis. Similar good results were observed when assessing tumor response to systemic therapy and disease progression. Our study supports the use of personalized biomarkers and liquid biopsies to tailor the management of rectal cancer patients, however, replication in a larger cohort is necessary to introduce this strategy into clinical practice.
Article
Full-text available
Repeat tumor biopsies to study genomic changes during therapy are difficult, invasive and data are confounded by tumoral heterogeneity. The analysis of circulating tumor DNA (ctDNA) can provide a non-invasive approach to assess prognosis and the genetic evolution of tumors in response to therapy. Mutation-specific droplet digital PCR was used to measure plasma concentrations of oncogenic BRAF and NRAS variants in 48 patients with advanced metastatic melanoma prior to treatment with targeted therapies (vemurafenib, dabrafenib or dabrafenib/trametinib combination) or immunotherapies (ipilimumab, nivolumab or pembrolizumab). Baseline ctDNA levels were evaluated relative to treatment response and progression-free survival (PFS). Tumor-associated ctDNA was detected in the plasma of 35/48 (73%) patients prior to treatment and lower ctDNA levels at this time point were significantly associated with response to treatment and prolonged PFS, irrespective of therapy type. Levels of ctDNA decreased significantly in patients treated with MAPK inhibitors (p < 0.001) in accordance with response to therapy, but this was not apparent in patients receiving immunotherapies. We show that circulating NRAS mutations, known to confer resistance to BRAF inhibitors, were detected in 3 of 7 (43%) patients progressing on kinase inhibitor therapy. Significantly, ctDNA rebound and circulating mutant NRAS preceded radiological detection of progressive disease. Our data demonstrate that ctDNA is a useful biomarker of response to kinase inhibitor therapy and can be used to monitor tumor evolution and detect the early appearance of resistance effectors.
Article
Highlights * NSCLC patients present significantly higher plasma cfDNA levels than those with benign lung tumours. * Plasma cfDNA integrity is significantly altered in patients with NSCLC or benign lung tumours. * Diagnostic capacity of our cfDNA assay is comparable to values presented by conventional imaging modalities. * Plasma cfDNA quantification can be used to differentiate between NSCLC and benign lung tumours significantly. Effective discrimination between lung cancer and benign tumours is a common clinical problem in the differential diagnosis of solitary pulmonary nodules. The analysis of cell-free DNA (cfDNA) in blood may greatly aid the early detection of lung cancer by evaluating cancer-related alterations. The plasma cfDNA levels and integrity were analysed in 65 non-small cell lung cancer (NSCLC) patients, 28 subjects with benign lung tumours, and 16 healthy controls using real-time PCR. The NSCLC patients demonstrated significantly higher mean plasma cfDNA levels compared with those with benign tumours (p=0.0009) and healthy controls (p<0.0001). The plasma cfDNA integrity in healthy individuals was significantly different than that found in patients with NSCLC or benign lung tumours (p<0.0003). In ROC curve analysis, plasma cfDNA levels >2.8 ng/ml provided 86.4% sensitivity and 61.4% specificity in discriminating NSCLC from benign lung pathologies and healthy controls. cfDNA integrity showed better discriminatory power (91% sensitivity, 68.2% specificity). These data demonstrate that plasma cfDNA concentration and integrity analyses can significantly differentiate between NSCLC and benign lung tumours. The diagnostic capacity of the quantitative cfDNA assay is comparable to the values presented by conventional imaging modalities used in clinical practice.
Article
Recent developments in massively parallel sequencing and digital genomic techniques support the clinical validity of cell-free circulating tumour DNA (ctDNA) as a ‘liquid biopsy’ in human cancer. In breast cancer, ctDNA detected in plasma can be used to non-invasively scan tumour genomes and quantify tumour burden. The applications for ctDNA in plasma include identifying actionable genomic alterations, monitoring treatment responses, unravelling therapeutic resistance, and potentially detecting disease progression before clinical and radiological confirmation. ctDNA may be used to characterise tumour heterogeneity and metastasis-specific mutations providing information to adapt the therapeutic management of patients. In this article, we review the current status of ctDNA as a ‘liquid biopsy’ in breast cancer.
Article
Plasma circulating cell-free (cf)DNA is of interest in oncology because it has been shown to contain tumour DNA and may thus be used as liquid biopsy. In nonsmall cell lung cancer (NSCLC), cfDNA quantification has been proposed for the monitoring and follow-up of patients. However, available studies are limited and need to be confirmed by studies with larger sample sizes and including patients who receive more homogenous treatments. Our objective was to assess the predictive and prognostic value of plasma cfDNA concentration in a large series of patients with NSCLC and treated with a standard chemotherapy regimen. We included samples from lung cancer patients recruited into the Pharmacogenoscan study. The cfDNA of 218 patients was extracted and quantified by fluorometry before and after two or three cycles of platinum-based chemotherapy. The association between baseline and post-chemotherapy concentrations and treatment response, assessed by RECIST (response evaluation criteria in solid tumours) or patient survival was analysed. Patients with high cfDNA concentrations (highest tertile) at baseline had a significantly worse disease-free and overall survival than those with lower concentrations (lowest and middle tertiles) (median overall survival 10 months (95% CI 10.7–13.9) versus 14.2 months (95% CI 12.6–15.8), respectively; p=0.001). In multivariate analysis, increased baseline concentration of cfDNA was an independent prognostic factor. However, we did not find any association between cfDNA concentration and response to treatment. cfDNA may be a biomarker for the assessment of prognosis in NSCLC. However, total concentration of cfDNA does not appear to predict chemotherapy response.