Subido por rdgutirrrez

1-s2.0-S0009898123004758-main

Anuncio
Clinica Chimica Acta 552 (2024) 117673
Contents lists available at ScienceDirect
Clinica Chimica Acta
journal homepage: www.elsevier.com/locate/cca
Comparative study of droplet-digital PCR and absolute Q digital PCR for
ctDNA detection in early-stage breast cancer patients
Victoria Sánchez-Martín a, b, Esperanza López-López a, c, Diego Reguero-Paredes a,
Ana Godoy-Ortiz a, b, c, Maria Emilia Domínguez-Recio a, c, Begoña Jiménez-Rodríguez a, b, c,
Alfonso Alba-Bernal a, c, d, Maria Elena Quirós-Ortega a, c, d, María Dunia Roldán-Díaz a, c,
Jesús Velasco-Suelto a, c, Noelia Linares-Valencia a, c, Alicia Garrido-Aranda a, c, d,
Rocío Lavado-Valenzuela a, b, c, d, Martina Álvarez a, b, d, e, Javier Pascual a, b, c, d,
Emilio Alba a, b, c, d, e, *, Iñaki Comino-Méndez a, b, c, d, *
a
Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
Centro de Investigacion Biomedica en Red de Cancer (CIBERONC - CB16/12/00481), 28029, Madrid, Spain
The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), 29010, Malaga, Spain
d
Andalusia-Roche Network in Precision Medical Oncology, 41092, Sevilla, Spain
e
University of Málaga, Faculty of Medicine, 29010 Malaga, Spain
b
c
A R T I C L E I N F O
A B S T R A C T
Keywords:
Circulating tumor DNA
Digital PCR
Liquid biopsy
Breast cancer
Background: Analysis of circulating tumor DNA (ctDNA) is increasingly used for clinical decision-making in
oncology. However, ctDNA could represent ≤ 0.1 % of cell-free DNA in early-stage tumors and its detection
requires high-sensitive techniques such as digital PCR (dPCR).
Methods: In 46 samples from patients with early-stage breast cancer, we compared two leading dPCR assays for
ctDNA analysis: QX200 droplet digital PCR (ddPCR) system from Bio-Rad which is the gold-standard in the field,
and Absolute Q plate-based digital PCR (pdPCR) system from Thermo Fisher Scientific which has not been re­
ported before. We analyzed 5 mL of baseline plasma samples prior to any treatment.
Results: Both systems displayed a comparable sensitivity with no significant differences observed in mutant allele
frequency. In fact, ddPCR and pdPCR possessed a concordance > 90 % in ctDNA positivity. Nevertheless, ddPCR
exhibited higher variability and a longer workflow. Finally, we explored the association between ctDNA levels
and clinicopathological features. Significantly higher ctDNA levels were present in patients with a Ki67 score >
20 % or with estrogen receptor-negative or triple-negative breast cancer subtypes.
Conclusion: Both ddPCR and pdPCR may constitute sensitive and reliable tools for ctDNA analysis with an
adequate agreement in early-stage breast cancer patients.
1. Introduction
Liquid biopsy has emerged as a minimally invasive and promising
methodology in molecular oncology, offering crucial insights into
various tumor components released into the bloodstream [1]. Among
these components, circulating tumor DNA (ctDNA) found in the plasma
of cancer patients provides a valuable source for investigating genomic
aberrations that define the carcinogenic process[2]. Indeed, ctDNA has
found diverse applications in cancer research, including diagnosis, pa­
tient stratification, prediction of treatment response, efficacy moni­
toring, and post-treatment surveillance for minimal residual disease
(MRD) detection [3–7].
Abbreviations: BC, breast cancer; cfDNA, cell-free DNA; ctDNA, circulating tumor DNA; ddPCR, droplet digital PCR; ER, estrogen receptor; HER2, epidermal
growth factor receptor 2; MAF, mutant allele frequency; PCR, pathologic complete response; pdPCR, plate-based digital PCR; PR, progesterone receptor; TNBC, triple
negative breast cancer; Tm, melting temperature.
* Corresponding authors at: Clinical and Traslational Cancer Research Group, IBIMA Institute, Calle Severo Ochoa, 35, Parque Tecnologico de Andalucia (PTA),
Campanillas 29590 (Malaga), Spain.
E-mail addresses: [email protected] (E. Alba), [email protected] (I. Comino-Méndez).
https://doi.org/10.1016/j.cca.2023.117673
Received 20 September 2023; Received in revised form 17 November 2023; Accepted 19 November 2023
Available online 23 November 2023
0009-8981/© 2023 The Author(s). Published by Elsevier B.V. This is an open access article under the CC BY-NC license (http://creativecommons.org/licenses/bync/4.0/).
V. Sánchez-Martín et al.
Clinica Chimica Acta 552 (2024) 117673
However, it is essential to note that ctDNA fractions may fluctuate
based on the cancer stage, and early-stage tumors usually present ctDNA
levels below 0.1 % of cell-free DNA [8]. As a result, detecting ctDNA
requires highly sensitive technologies. Currently, digital PCR (dPCR)
stands as the gold-standard approach due to its exceptional sensitivity,
experimental simplicity, and favorable price-to-sample ratio [9].More­
over, next-generation sequencing (NGS) based procedures allow for a
comprehensive description of mutational profiles, but NGS remains
relatively expensive compared to other methods [10]. Several studies
have utilized Bio-Rad’s QX droplet digital PCR systems to analyze ctDNA
in early-stage breast cancer (BC). These investigations have demon­
strated the potential of ctDNA tracking in predicting future relapse,
monitoring treatment response, and assessing clinical outcomes
[11–14].
In the present work, we aimed to conduct a side-by-side evaluation of
two leading dPCR assays for ctDNA detection: the QX200 droplet digital
PCR system from Bio-Rad, which is the most used system to date, and the
Applied Biosystems QuantStudio Absolute Q digital PCR system from
Thermo Fisher Scientific which has not been previously reported in the
scientific literature. We tested 46 samples from patients with early-stage
BC, and we analyzed their baseline plasma samples prior to any treat­
ment separately with both dPCR systems. By comparing the perfor­
mance and sensitivity of these two dPCR systems, we aim to provide
valuable insights into their potential applications for ctDNA detection in
early-stage BC. This study aims to have a significant impact on ctDNAbased diagnostics and monitoring in clinical settings, ultimately lead­
ing to improved patient management and the development of person­
alized treatment strategies.
libraries were prepared using SureSelect Human All Exon v6 (Agilent,
5190–8863) with probes capturing the whole exome, and sequenced on
a DNB-seq platform (BGI Genomics, China).
Quality control of WES data was performed using fastQC (v0.11.9),
followed by trimming and quality filtering using Trim Galore (v0.6.7).
Pre-processed reads were mapped to the GRCh38 reference genome by
BWA-mem (v0.7.17). Data correction for technical biases and somatic
mutation calling were performed according to GATK’s best practices (htt
ps://software.broadinstitute.org/gatk/best-practices). The resulting
aligned SAM files were sorted by coordinate using Picard Sortsam
(Picard v2.26.10), and converted to BAM format with Samtools (v1.9).
Picard Mark Duplicates was run to mark duplicated reads from each
BAM file. GATK Base Recalibrator and Apply BQSR (GATK v4.2.2.0)
were used for base quality score recalibration. Somatic variants analysis
for each tumor sample was performed by GATK Mutect2 in matched
normal mode including a custom panel of normal non-cancer variations
which was previously built and a germline variant annotation file for the
GRCh38 reference genome obtained from the GATK resource bundle.
Reads counting summaries were obtained using GATK Get Pileup
Summaries and passed to GATK Caculate Contamination for contami­
nation calculation. The reported variants were filtered to get true so­
matic mutations using Filter Mutect Calls. Somatic variants were
annotated by ANNOVAR (v20200608) with custom made databases for
COSMIC v95 and TCGA mutation data retrieved from GDC data portal
[15].
After identifying somatic mutations, custom TaqMan™ SNP Geno­
typing Assays with FAM fluorophore mutant probes and VIC fluorophore
wild-type probes were designed and ordered (Thermo Fisher Scientific).
Finally, every tracking somatic mutation was re-validated using tumor
and germline DNA by droplet digital PCR (see below), optimizing the
annealing temperature for each assay (Table 1).
2. Materials and methods
2.1. Patients and samples
2.4. DNA extraction, quantification and sample preparation
In this prospective study, we enrolled a cohort of 23 patients starting
in 2020diagnosed with early-stage BC. Prior to cancer diagnosis, plasma
samples were collected just before any treatment. Here, we tested 2
samples per patient with a total of 46 samples analyzed. The patients
were recruited, and samples were collected at Hospitals Virgen de la
Victoria and Regional of Malaga, Spain. All patients provided informed
consent, and the study followed the principles of the Helsinki Declara­
tion and received approval from the local Ethical Committee. Human
rights were protected.
Cell-free DNA (cfDNA) was obtained from plasma samples using the
QIAamp Circulating Nucleic Acid Kit (Qiagen, 55114) following the
manufacturer’s instructions. Specifically, 5 mL of plasma were extracted
for use with the two dPCR platforms. The elution of cfDNA was per­
formed using 100 µL of AVE buffer, and the samples were subsequently
stored at − 20 ◦ C.
Germline DNA from PBMCs was used as a negative control for each
patient to ensure the reliability of the results and control for false pos­
itives and clonal hematopoiesis of indeterminate potential (CHIP)
events. DNA from PBMCs was extracted using the QIAamp DNA Blood
Mini Kit (Qiagen, 51104) following the manufacturer’s protocol, and the
samples were subsequently stored at − 20 ◦ C until use.
DNA quantification was performed using the RNAse P assay (Ther­
moFisher Scientific) [11].
For each patient sample, the entire cfDNA amount and an equivalent
amount of DNA from PBMCs were used as input in the subsequent an­
alyses. Preparations of cfDNA were dried down at 45 ◦ C using the
Eppendorf™ Concentrator plus (Thermo Fisher Scientific) and subse­
quently resuspended in 7 µL of nuclease-free water (Canvax Biotech,
E0320). The corresponding amount of germline DNA from PBMCs was
also dried down and resuspended in the same manner.
2.2. Blood sample processing
Blood samples from the study participants were collected in citrate
blood bags and processed within 2 h following venipuncture. The
plasma supernatant was isolated by centrifugation for 10 min at 3,000
rpm at room temperature and subsequently stored at − 80 ◦ C until the
extraction of cell-free DNA. For the isolation of PBMCs, a density
gradient centrifugation method was employed using Lymphoprep (Stem
Cell Technologies, 07801) following the manufacturer’s instructions.
The isolated PBMCs were stored at − 196 ◦ C until further use.
2.3. Whole exome sequencing
The identification of somatic mutations for each patient was
accomplished through whole exome sequencing (WES) of both formalinfixed paraffin-embedded (FFPE) or fresh frozen (FF) tumor tissue and
peripheral blood mononuclear cells (PBMCs). For tumor tissue DNA
extraction, the RecoverAll™ Total Nucleic Acid Isolation Kit (Thermo
Fisher Scientific, AM1975) was used following the manufacturer’s in­
structions. Germline DNA from PBMCs was isolated using the QIAamp
DNA Blood Mini Kit (Qiagen, 51104) following the manufacturer’s
protocol. To quantify DNA, the RNAse P assay (Thermo Fisher Scientific)
was employed, following a previously published protocol [11]. WES
2.5. Droplet digital PCR
Samples were screened using the customTaqMan™ SNP Genotyping
Assays with FAM fluorophore mutant probes and VIC fluorophore wildtype probes.The screening was conducted using the optimal conditions
that had been previously optimized.Droplet digital PCR (ddPCR) was
performed on a QX200 Droplet Digital PCR System (Bio-Rad).The cfDNA
samples were divided into 7 independent PCR reactions. Each PCR re­
action consisted of 1X ddPCR Supermix for probes (Bio-Rad, 1863024),
1X custom TaqMan ddPCR assays (ThermoFisher Scientific), and 1 µL of
2
V. Sánchez-Martín et al.
Clinica Chimica Acta 552 (2024) 117673
Table 1
Somatic mutations tracked and ctDNA detection results. This table presents the results for the analysis of 23 plasma samples from early-stage BC patients. Using Whole
Exome Sequencing (WES), one truncal somatic mutation was identified per patient. TaqMan assays were custom-designed to cover these mutations, and the respective
melting temperature (Tm) was optimized. The total FAM + VIC- compartments indicate the number of droplets or micro-chambers containing ctDNA, detected by
ddPCR or pdPCR, respectively. The last column displays the Mutant Allele Frequency (MAF) values (%).
Patient sample
BC-008
BC-009
BC-010
BC-014
BC-015
BC-016
BC-017
BC-021
BC-026
BC-031
BC-033
BC-035
BC-037
BC-041
BC-046
BC-053
BC-059
BC-060
BC-061
BC-064
BC-069
BC-082
BC-085
Somatic mutation tracked in ctDNA
Gene
Mutation
TP53
TCP11L2
DIDO1
PIK3CA
PIK3CA
TP53
FI3AI
TSPAN32
ERBB2
ADAM29
PIK3CA
TMEM205
IPO8
TP53
TP53
TP53
TP53
PIK3CA
TP53
HIPR1
GSAP
TP53
TP53
R248Q
E269X
R187H
E545K
E545K
C238Y
V297I
F37F
V777L
G414E
E545K
A183T
R53Q
R249S
R213X
E221X
R213X
H1047Y
R248Q
R252Q
F209F
G266R
R273C
Total FAM+VIC- compartments
Tm
60 ◦ C
60 ◦ C
60 ◦ C
64 ◦ C
64 ◦ C
62 ◦ C
60 ◦ C
64 ◦ C
60 ◦ C
64 ◦ C
64 ◦ C
64 ◦ C
60 ◦ C
64 ◦ C
60 ◦ C
60 ◦ C
60 ◦ C
60 ◦ C
60 ◦ C
60 ◦ C
60 ◦ C
60 ◦ C
60 ◦ C
MAF (%)
ddPCR
pdPCR
ddPCR
pdPCR
2
1
2
183
1
34
1
11
0
1
1
0
3
0
512
124
13
28
87
14
0
0
12
2
0
1
272
0
0
0
23
0
1
1
0
9
0
558
188
9
47
153
28
1
1
21
0.012
0.003
0.007
0.647
0.008
0.317
0.011
0.143
0.000
0.006
0.002
0.000
0.025
0.000
6.120
1.739
0.147
0.197
0.412
0.211
0.000
0.000
0.122
0.008
0.000
0.003
0.902
0.000
0.000
0.000
0.135
0.000
0.003
0.002
0.000
0.045
0.000
5.131
1.506
0.096
0.224
0.483
0.346
0.013
0.002
0.158
ddPCR, droplet digital PCR; MAF, mutant allele frequency; pdPCR, plate-based digital PCR; Tm, melting temperature.
cell-free DNA in a total volume of 20 µL. For germline DNA from PBMCs,
5U HindIII-HF (New England Biolabs, R3104) was added to fragment
genomic DNA. Droplets were generated using the Auto droplet generator
(Bio-Rad) following the manufacturer’s instructions. The assays were
conducted on 96-well plates with the following thermal cycling condi­
tions in a C1000 Touch™ thermal cycler (Bio-Rad): 95 ◦ C for 10 min, 40
cycles of 94 ◦ C for 30 sec and a specific annealing temperature for 60 sec,
98 ◦ C for 10 min. The temperature ramp increment was 2 ◦ C/sec for all
steps. The plates were read on the Bio-Rad QX-200 droplet reader (BioRad). Data analysis was performed using QuantaSoft v1.7 software (BioRad). Threshold gating was manually set for each patient in the previous
validation phase and maintained consistently for all plasma samples
from the same patient. Positive results required at least 2 droplets on the
mutant channel, and at least one non-template negative control was run
with every assay.
samples from the same patient. Positive results required at least 2 microchambers on the mutant channel, and at least one non-template control
was run with every assay.
2.7. Data analyses
The experiment was deemed invalid if positivity was detected in any
of the non-template controls. The number of FAM-positive (mutant)
droplets or micro-chambers and the mean of mutant copies per micro­
liter were directly obtained from the QX200 Droplet Digital PCR System
(Bio-Rad) or the Applied Biosystems QuantStudio Absolute Q Digital
PCR System (ThermoFisher Scientific), respectively. The mutant copies
per microliter were then transformed into mutant copies per eluate using
the following formula:
Mutant copies per eluate = Mutant copies per μL × μL of mix
2.6. Plate-based digital PCR
× number of reactions assayed
The plate-based digital PCR (pdPCR) assays were conducted under
the same conditions as ddPCR, with respect to TaqMan probes and
cycling conditions as previously described. Input samples were parti­
tioned into 7 independent PCR reactions, as the ddPCR setup. The
Applied Biosystems QuantStudio Absolute Q Digital PCR System
(ThermoFisher Scientific) was used for the pdPCR experiments. Each
pdPCR reaction consisted of 1X dPCR Master Mix (ThermoFisher Sci­
entific, A52490), 1X custom TaqMan ddPCR assays (ThermoFisher Sci­
entific), and 1 µL of cell-free DNA in a total volume of 9 µL. For germline
DNA from PBMCs, 5U HindIII-HF (New England Biolabs, R3104) was
added to fragment genomic DNA. The PCR reactions were loaded onto
MAP16 plates (ThermoFisher Scientific, A52688), and then, 15 µL of
Isolation Buffer (ThermoFisher Scientific, A52730) was transferred to
wells with the PCR mix. All the necessary steps for dPCR, including
compartmentalizing, thermal cycling, and data acquisition, were per­
formed on a single instrument, the Applied Biosystems QuantStudio
Absolute Q Digital PCR System (ThermoFisher Scientific). Threshold
gating was set individually and manually for each in the previous vali­
dation phase patient sample but consistently maintained for all plasma
Where the volume of the mix was 20 µL in the case of the Bio-Rad system
and 9 µL for the Absolute Q. Regardless of the platform, the assays were
partitioned into 7 reactions. The mutant copies per mL of plasma were
calculated as follows:
Mutant copies per mL of plasma
= Mutant copies per eluate/mL of plasma assayed
Where the volume of plasma employed was 5 mL in all the assays.
Finally, mutant allele frequency (MAF) was calculated as follows:
MAF (%) = Mutant copies per μL/Wild − type copies per μL × 100
Where both the values of mutant and wild-type copies per µL were
directly obtained from the corresponding platform.
2.8. Statistical analyses
Statistical analyses and graphical representations were performed
using R (v4.2.2) and GraphPad Prism 8. The agreement between
3
V. Sánchez-Martín et al.
Clinica Chimica Acta 552 (2024) 117673
platforms was assessed using the Cohen К statistic, and correlation was
calculated using the Spearman ρ coefficient. Differences between plat­
forms were evaluated using the Mann-Whitney-Wilcoxon test, and
analysis of variances was conducted with the Fisher test. For all tests, pvalues below 0.05 were considered statistically significant and denoted
as follows: *p < 0.05; **p < 0.01, and ***p < 0.001.
For the sliding window analysis, all samples with a positive call from
either platform were included. The samples were ranked from the
highest to the lowest mutant allele frequency (MAF), considering the
largest MAF value obtained from both technologies for each sample. A
sliding window of 5 patients was established, starting from the samples
with the highest MAF. Within each sliding window, the median MAF and
the percentage of concordant samples between platforms were calcu­
lated and plotted. For each new window, the sample with the highest
MAF was removed, and the sample with the next highest MAF was
included. These calculations were repeated for each window until the
last window containing the samples with the lowest MAF.
MAF in either method (Spearman ρ = -0.06, p-value > 0.05 for ddPCR
and Spearman ρ = 0.07, p-value > 0.05 for pdPCR) (Fig. 2C). Interest­
ingly, the number of compartments analyzed was significantly higher in
pdPCR than ddPCR (Wilcoxon W = 529, p-value < 0.001), and the
comparison of variances showed a significantly higher variability in
ddPCR compared to pdPCR (Fisher F = 0.0008, p-value < 0.001).
3.2. Concordance of ctDNA detection between ddPCR and pdPCR
We compared ctDNA positivity between ddPCR and pdPCR in 23
patients with early-stage BC. The results showed that ddPCR detected
ctDNA in 56.5 % (13/23) of patients, while pdPCR detected ctDNA in
47.8 % (11/23) (Fig. 3A). Despite this slight difference in ctDNA
detection rates, there was close agreement between the two platforms,
with a Cohen κ value of 0.83 (95 % CI, 0.60 – 1.00). Only 8.7 % (2/23) of
cases showed discordant ctDNA positivity. In particular, discordance
occurred in BC-010 and BC-016 samples, which were ctDNA positive by
ddPCR but ctDNA negative by pdPCR (Fig. 3B). Composite allele fraction
of MAF between the two methodologies was tightly correlated with a
Spearman ρ = 0.76 (p-value < 0.0001) (Fig. 3C). Analyzing patients
with a positive result from at least one platform using a sliding window
of 5 patients from high to low MAF, we observed that the percentage of
concordant cases varied with the MAF of windows (Fig. 3D). Total
concordance was reported for windows with MAFs ≤ 0.147 % and MAFs
≥ 0.902 %, while the percentage of concordant cases was 80 % for
windows with MAFs in the range 0.158 % − 0.403 %. No sharp dete­
rioration in concordance was observed in any of the analyzed windows.
3. Results
3.1. Comparing droplet and Plate-Based digital PCR for ctDNA analysis
A total of 23 patients with early-stage BC participated in this ctDNA
assessment using digital PCR (dPCR). Particularly, 2 samples per patient
were tested with a total of 46 samples analyzed. Cell-free DNA (cfDNA)
was isolated from 5 mL of plasma, quantified and subjected to dPCR for
ctDNA detection. We utilized two different platforms for analysis: the
QX200 droplet digital PCR system (ddPCR) and the Applied Biosystems
QuantStudio Absolute Q digital PCR system (pdPCR) (Fig. 1A).
ddPCR is based on water–oil emulsion technology, creating
nanoliter-sized droplets that act as partitions to separate DNA mole­
cules. On the other hand, pdPCR relies on microfluidics to compart­
mentalize DNA molecules into micro-chambers. Both platforms require
compartmentalization, PCR amplification, and data acquisition to
identify mutant DNA molecules in a sample. In terms of experimental
steps and time-to-perform comparison, ddPCR involves a droplet
generator for compartmentalization, a thermal cycler for PCR amplifi­
cation, and a droplet reader for data acquisition. In contrast, pdPCR with
Absolute Q allows all necessary steps to be conducted on a single in­
strument, making the workflow faster. In fact, pdPCR takes ~ 3 h to
complete the full protocol, while ddPCR requires ~ 4 h. (Fig. 1B).
In this comparative analysis, we studied baseline plasma samples
from 23 patients with early-stage BC using the two different method­
ologies. Whole-exome sequencing of tumor and germline DNA was
performed to identify somatic mutations, from which one truncal mu­
tation per patient was selected (Table 1). Custom TaqMan™ SNP Gen­
otyping Assays with FAM and VIC fluorophore probes for mutant and
wild-type alleles, respectively, were designed and optimized (Table 1).
In our cohort, both ddPCR and pdPCR detected a minimum of 1
mutant FAM-positive VIC-negative allele, with a maximum of 512 and
518 FAM-positive VIC-negative droplets and microchambers respec­
tively (Table 1). The determination of mutant allele frequency (MAF)
showed similar results for both platforms (Fig. 2A), with a minimum
MAF of 0.002 % in both methods for the BC-033 plasma sample
(Table 1). The mean MAF was 0.440 % (range 0 – 6.120) in ddPCR and
0.394 % (range 0– 5.131) in pdPCR, and there were no statistical dif­
ferences between the platforms (Wilcoxon W = 289, p-value > 0.05).
Next, we explored the potential role of input cfDNA for ddPCR and
pdPCR and its relation to MAF. The amount of input cfDNA was not
correlated with MAF in either platform, with a Spearman ρ = -0.12 (pvalue > 0.05) for ddPCR and a Spearman ρ = -0.30 (p-value > 0.05) for
pdPCR (Fig. 2B). Moreover, there were no significant differences in cfDNA
input between the two platforms (Wilcoxon W = 258, p-value > 0.05).
We further assessed the total number of compartments analyzed in
both methodologies and its potential association with MAF. The number
of droplets in ddPCR or micro-chambers in pdPCR did not correlate with
3.3. Association between ctDNA levels and clinicopathological
characteristics
In this study, we explored the correlation between ctDNA levels and
various clinicopathological features in early-stage BC patients. In
particular, the mean of MAF obtained from both ddPCR and pdPCR
platforms was considered. The clinicopathological characteristics
analyzed included tumor size, affected lymph nodes, tumor grade, Ki67
score, estrogen receptor (ER), progesterone receptor (PR) status and
epidermal growth factor receptor 2 (HER2) status, triple-negative breast
cancer (TNBC) subtype, and pathologic complete response (PCR) after
neoadjuvant chemotherapy (Fig. 4).The results showed that ctDNA
levels were not significantly influenced by tumor size, affected lymph
nodes, tumor grade, PR status, HER2 status, or PCR. However, we
observed significantly higher ctDNA levels in patients with ER-negative,
TNBC subtype and those with a Ki67 score above 20 %.
In this prospective study, patients are currently under clinical followup, and thus far, two of them have experienced relapse (BC-016 and BC046). Notably, the patient BC-046, who underwent neoadjuvant
chemotherapy, exhibited the highest MAF (6.12 %, as determined by
ddPCR) among the positive samples. When compared to the calculated
median of all positive samples (ddPCR values), this represented a sub­
stantial 50.16-fold change (see Fig. 2 and Table 1).
It is crucial to emphasize that the other relapsed patient (BC-016)
demonstrated ctDNA positivity at baseline through ddPCR but not
pdPCR (see Fig. 2 and Fig. 3). Additionally, it is noteworthy that the
time-to-relapse from the baseline sample extraction was shorter for the
patient with the highest ctDNA levels (BC-046), at 1.43 years, compared
to the other patient (BC-016), who experienced relapse after 2.08 years.
4. Discussion
To date, ddPCR has been exploited to detect ctDNA in early-stage BC
with high sensitivity [12–14,16–18].In fact, ddPCR remains as the gold
standard methodology for ctDNA analysis not only in BC, but also in
other types of cancer [19]. In this regard, a recent study compared the
performance of the ddPCR technology with a different novel array-based
digital PCR system for detecting specific mutations in lung and
4
V. Sánchez-Martín et al.
Clinica Chimica Acta 552 (2024) 117673
Fig. 1. Comparison of ddPCR and pdPCR for ctDNA analysis. A) Illustrative depiction of the experimental workflow employed in this comparative study. Beginning
with 5 mL of plasma, cfDNA extraction, quantification, and subsequent application for either ddPCR (with an anticipated analysis of 20,000 droplets per reaction) or
pdPCR (with an expected analysis of 20,000 micro-chambers per reaction).B) Schematic presentation of the experimental protocol involving ddPCR and pdPCR,
encompassing the subsequent stages: formulation and loading of reaction mix, compartmentalization, PCR amplification, data capture, and data analysis. cfDNA, cellfree DNA; ddPCR, droplet digital PCR; pdPCR, plate-based digital PCR.
5
V. Sánchez-Martín et al.
Clinica Chimica Acta 552 (2024) 117673
Fig. 2. Analysis of sensitivity and variability for ctDNA detection between ddPCR and pdPCR. A) Representation of the values of MAF (%) per patient that were
obtained both by ddPCR and pdPCR, rendering information about sensitivity. B) Graphical presentation of MAF (%) values versus the total cfDNA quantity extracted
from each plasma sample, assessing the influence of input cfDNA on MAF (%) values for each platform (ddPCR on the left, pdPCR on the right).C) Depiction of MAF
(%) values against the total count of compartments examined per plasma sample, aimed at assessing the influence of partitions on the resulting MAF (%) values using
both platforms (ddPCR on the left, pdPCR on the right). Positive outcomes were ascertained when a minimum of 2 compartments were identified on the mutant
channel. ddPCR, droplet digital PCR; pdPCR, plate-based digital PCR.
6
V. Sánchez-Martín et al.
Clinica Chimica Acta 552 (2024) 117673
Fig. 3. Concordance of ctDNA detection between ddPCR and pdPCR. A) Contingency table for positive and negative ctDNA detection per patient sample using ddPCR
and pdPCR. B) 2D plots from ddPCR and pdPCR discordant samples (BC-010 and BC-016). C) Composite allele fraction of the agreement on MAF between ddPCR and
pdPCR. D) Sliding window analysis of patients with positive ctDNA detection in at least 1 platform, showing variation in the percentage of concordant cases with
median MAF of the window (n = 5 in every sliding window). ddPCR, droplet digital PCR; pdPCR, plate-based digital PCR.
colorectal cancer patients. The authors revealed weak concordance be­
tween both platforms, particularly for EGFR or RAS mutations, espe­
cially in detecting ctDNA mutations with very low mutant allele
frequency (MAF). The array-based system demonstrated better sensi­
tivity in this context [20]. However, to the best of our knowledge, there
are no previous studies assaying and comparing ctDNA detection using
the novel Absolute Q pdPCR platform.
The present study aimed to compare two digital PCR platforms for the
analysis of ctDNA in early-stage BC patients. Our results showed that both
ddPCR and pdPCR displayed a comparable sensitivity for ctDNA detection,
with no significant differences observed in MAF between the two plat­
forms. It is important to note here that the amplification conditions were
optimized for ddPCR and turned out to be optimal also for pdPCR. How­
ever, ddPCR exhibited higher variability in the number of compartments
analyzed compared to pdPCR. This finding suggests that pdPCR may
provide a more consistent and reproducible measurement of ctDNA levels
in blood samples.
Regarding ctDNA positivity, we found a high level of concordance
between ddPCR and pdPCR, with only a small percentage of cases
showing discordance. Importantly, the discordant cases exhibited very
low MAF values, close to the minimum limit of detection for both
platforms. This observation highlights the importance of considering the
lower limit of detection when interpreting ctDNA results in clinical
practice, particularly in cases with low tumor burden. In such scenarios,
repeating the testing enhances overall sensitivity and is recommended to
prevent false-negative or false-positive results [21].
In addition to technical comparisons, we explored the association
between ctDNA levels and various clinicopathological characteristics.
We observed that ctDNA levels were not significantly influenced by
tumor size, affected lymph nodes, tumor grade, PR and HER2 status, or
pathologic complete response after neoadjuvant chemotherapy in our
sample cohort. However, intriguingly, we found distinct associations
between ctDNA levels and HR status as well as the Ki67 score, a marker
of cell proliferation.These findings partially overlap with results from
7
V. Sánchez-Martín et al.
Clinica Chimica Acta 552 (2024) 117673
Fig. 4. Association of ctDNA levels with clinicopathological characteristics. Plots representing the statistical analysis between mean ctDNA MAFs from ddPCR and
pdPCR platforms and A) tumor size, B) affected lymph nodes (according to TNM scale) C) differentiation grade, D) Ki67 score, E) ER status, F) PR status, G) HER2
status, H) TNBC subtype and I) PCR. ddPCR, droplet digital PCR; ER, estrogen receptor; HER2, epidermal growth factor receptor 2; PCR, pathologic complete response;
pdPCR, plate-based digital PCR; PR, progesterone receptor; TNBC, triple negative breast cancer.
previous studies, reinforcing the notion that baseline ctDNA analysis
offers insights into clinical features in early-stage BC patients [3]. For
example, the presence of elevated ctDNA levels has been notably linked
to specific BC subtypes, such as triple-negative breast cancer (TNBC), a
particularly aggressive subtype [22]. Aligning with our findings, Mag­
banua et al. reported higher ctDNA positivity rates in TNBC patients
both before and during neoadjuvant chemotherapy [23] a finding that
was also observed in other studies [24,25]. In terms of the ER status,
existing studies have shown that HR status can predict the detectability
of ctDNA in blood; however, these analyses did not delve into ER status
specifically [26]. However, our findings align with previous in­
vestigations in this regard [24,25]. Turning to the Ki67 score, divergent
findings are evident in the literature. Similar to our outcomes, baseline
ctDNA detection has been significantly associated with a Ki67 index
exceeding 20 % in early-stage BC patients [27]. In contrast, a distinct
study failed to establish a substantial connection between baseline
ctDNA positivity and the Ki67 index in early-stage BC [25].
Regarding disease relapse, two patients, BC-016 and BC-046, have
experienced recurrence to date. Intriguingly, the patient with the highest
ctDNA levels, BC-046, relapsed earlier and belonged to the TNBC subtype,
consistent with findings from prior studies [23,24]. This observation
suggests the possibility that patient BC-046 may have an undetectable but
active micro metastatic site. Therefore, these results once again empha­
sized the significance of baseline ctDNA measurements in predicting
8
V. Sánchez-Martín et al.
Clinica Chimica Acta 552 (2024) 117673
disease prognosis among early-stage BC patients.
Overall, our findings underscore the potential of ctDNA analysis to
propel personalized medicine forward, offering a non-invasive avenue to
gather critical disease-related insights.
Authors’ contribution
VS-M performed the comparative experiments and data analysis. ELL carried out the bioinformatic and statistical analyses. DR-P performed
the comparative experiments. AG-O, MED-R, and BJ-R recruited the
patients. AA-B, MEQ-O, MDR-D, JV-S, and NL-V processed the blood
samples. AG-A, RL-V, and MA managed the project and resources. VS-M
and IC-M wrote the manuscript. JP and EA reviewed the manuscript. ICM conceptualized and supervised the project, and reviewed and edited
the manuscript. All authors read and approved the final manuscript
5. Conclusions
Our study demonstrates the utility of the novel Absolute Q pdPCR
platform as a sensitive and reliable tool for ctDNA analysis in early-stage
cancers. Compared to ddPCR (Bio-Rad), both platforms exhibited a high
level of concordance in ctDNA detection. Absolute Q pdPCR surpassed
ddPCR in terms of reproducibility with a higher and less variable
number of compartments analyzed. In addition, Absolute Q pdPCR
allowed to perform all the required steps in a single instrument, short­
ening the workflow. Finally, ctDNA detection performed by both plat­
forms revealed associations with clinicopathological characteristics
providing valuable insights into the clinical implications of ctDNA
analysis in cancer management.
Ethics approval and consent.
This study included a cohort of 23 patients diagnosed with earlystage breast cancer at Hospitals Virgen de la Victoria and Regional of
Malaga, Spain. Prior to participation, all patients provided informed
consent, and the study adhered to the principles of the Helsinki Decla­
ration. Ethical approval was obtained from the local Ethical Committee.
References
[1] A. Alba-Bernal, et al., Challenges and achievements of liquid biopsy technologies
employed in early breast cancer, EBioMedicine 62 (2020.), 103100.
[2] J.C.M. Wan, et al., Liquid biopsies come of age: towards implementation of
circulating tumour DNA, Nat. Rev. Cancer 17 (4) (2017) 223–238.
[3] B. Jiménez-Rodríguez, et al., Development of a novel NGS methodology for
ultrasensitive circulating tumor DNA detection as a tool for early-stage breast
cancer diagnosis, Int. J. Mol. Sci. 24 (1) (2022) 146.
[4] J. Lv, et al., Improving on-treatment risk stratification of cancer patients with
refined response classification and integration of circulating tumor DNA kinetics,
BMC Med. 20 (1) (2022).
[5] C.A. Parkinson, et al., Exploratory Analysis of TP53 Mutations in Circulating
Tumour DNA as Biomarkers of Treatment Response for Patients with Relapsed
High-Grade Serous Ovarian Carcinoma: A Retrospective Study, PLoS Med. 13 (12)
(2016) e1002198.
[6] M. Murtaza, et al., Non-invasive analysis of acquired resistance to cancer therapy
by sequencing of plasma DNA, Nature 497 (7447) (2013) 108–112.
[7] T. Mok, et al., “Detection and Dynamic Changes of EGFR Mutations from
Circulating Tumor DNA as a Predictor of Survival Outcomes in NSCLC Patients
Treated with First-line Intercalated Erlotinib and Chemotherapy,” (in eng), Clin.
Cancer Res. 21 (14) (2015) 3196–3203.
[8] F. Diehl, et al., Circulating mutant DNA to assess tumor dynamics, Nat. Med. 14 (9)
(2008.) 985–990.
[9] U. Gezer, A.J. Bronkhorst, S. Holdenrieder, The Clinical Utility of Droplet Digital
PCR for Profiling Circulating Tumor DNA in Breast Cancer Patients, Diagnostics 12
(12) (2022) 3042.
[10] C. Lin, X. Liu, B. Zheng, R. Ke, and C. M. Tzeng, “Liquid Biopsy, ctDNA Diagnosis
through NGS,” (in eng), Life (Basel), vol. 11, no. 9, Aug 28 2021.
[11] I. Garcia-Murillas, et al., Mutation tracking in circulating tumor DNA predicts
relapse in early breast cancer, Sci. Transl. Med. 7 (302) (2015) 302ra133.
[12] F. Riva, et al., Patient-Specific Circulating Tumor DNA Detection during
Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer, Clin. Chem. 63 (3)
(2017) 691–699.
[13] I. Garcia-Murillas, et al., “Assessment of Molecular Relapse Detection in EarlyStage Breast Cancer”, JAMA Oncol. 5 (10) (2019) 1473.
[14] F. Rothé, et al., “Circulating Tumor DNA in HER2-Amplified Breast Cancer: A
Translational Research Substudy of the NeoALTTO Phase III Trial,” (in eng), Clin.
Cancer Res. 25 (12) (2019) 3581–3588.
[15] R.L. Grossman, et al., Toward a Shared Vision for Cancer Genomic Data, N. Engl. J.
Med. 375 (12) (2016.) 1109–1112.
[16] C. Bettegowda, et al., Detection of Circulating Tumor DNA in Early- and Late-Stage
Human Malignancies, Sci. Transl. Med. 6 (224) (2014).
[17] J. Phallen, et al., Direct detection of early-stage cancers using circulating tumor
DNA, Sci. Transl. Med. vol. 9 (403) (2017).
[18] F. Rothé, et al., Circulating Tumor DNA in HER2-Amplified Breast Cancer: A
Translational Research Substudy of the NeoALTTO Phase III Trial, Clin. Cancer Res.
25 (12) (2019) 3581–3588.
[19] D.F.K. Williamson, et al., “Detection of EGFR mutations in non-small cell lung
cancer by droplet digital PCR,” (in eng), PLoS One 17 (2) (2022) e0264201.
[20] S. Crucitta, et al., Comparison of digital PCR systems for the analysis of liquid
biopsy samples of patients affected by lung and colorectal cancer, Clin. Chim. Acta
541 (2023), 117239.
[21] S.A. Cohen, M.C. Liu, A. Aleshin, Practical recommendations for using ctDNA in
clinical decision making, Nature 619 (7969) (2023) 259–268.
[22] B.J. Rodriguez, et al., “Detection of TP53 and PIK3CA mutations in circulating
tumor DNA using next-generation sequencing in the screening process for early
breast cancer diagnosis,” (in eng), J. Clin. Med. vol. 8 (8) (2019).
[23] M.J.M. Magbanua, et al., “Clinical significance and biology of circulating tumor
DNA in high-risk early-stage HER2-negative breast cancer receiving neoadjuvant
chemotherapy,” (in eng), Cancer Cell 41 (6) (2023).
[24] I. Garcia-Murillas, et al., Assessment of molecular relapse detection in early-stage
breast cancer, JAMA Oncol. 5 (10) (2019) 1473–1478.
[25] S. Li, et al., “Circulating Tumor DNA Predicts the Response and Prognosis in
Patients With Early Breast Cancer Receiving Neoadjuvant Chemotherapy,” JCO
Precis Oncol. vol. 4 (2020).
[26] Y. Zhou, et al., Clinical factors associated with circulating tumor DNA (ctDNA) in
primary breast cancer, Mol. Oncol. 13 (5) (2019) 1033–1046.
[27] F. Cailleux, et al., “Circulating tumor DNA after neoadjuvant chemotherapy in
breast cancer is associated with disease relapse”, JCO Precis. Oncol. no. 6 (2022).
CRediT authorship contribution statement
Victoria Sánchez-Martín: Data curation, Formal analysis, Investi­
gation, Methodology, Resources, Supervision, Validation, Visualization,
Writing – original draft, Writing – review & editing. Esperanza LópezLópez: Data curation, Formal analysis, Investigation, Methodology,
Resources, Software, Visualization. Diego Reguero-Paredes: Investi­
gation, Methodology, Validation. Ana Godoy-Ortiz: Investigation,
Methodology, Resources. Maria Emilia Domínguez-Recio: Investiga­
tion, Methodology, Resources. Begoña Jiménez-Rodríguez: Investi­
gation,
Methodology.
Alfonso
Alba-Bernal:
Investigation,
Methodology. Maria Elena Quirós-Ortega: Investigation, Methodol­
ogy, Resources. María Dunia Roldán-Díaz: Investigation, Methodol­
ogy. Jesús Velasco-Suelto: Investigation, Methodology. Noelia
Linares-Valencia: Investigation, Methodology. Alicia Garrido-Ara­
nda: Investigation, Methodology. Rocío Lavado-Valenzuela: Investi­
gation, Methodology. Martina Álvarez: Investigation, Methodology,
Resources. Javier Pascual: Investigation, Methodology, Writing – re­
view & editing. Emilio Alba: Conceptualization, Funding acquisition,
Investigation, Resources, Supervision, Writing – original draft, Writing –
review & editing. Iñaki Comino-Méndez: Conceptualization, Data
curation, Formal analysis, Funding acquisition, Investigation, Method­
ology, Project administration, Resources, Software, Supervision, Vali­
dation, Visualization, Writing – original draft, Writing – review &
editing.
Declaration of Competing Interest
The authors declare that they have no known competing financial
interests or personal relationships that could have appeared to influence
the work reported in this paper.
Data availability
All data generated or analysed during this study are included in this
published article.
Acknowledgements
We would like to express our gratitude to all women who partici­
pated in this study.
9
Descargar