ISSN 0006-2979, Biochemistry (Moscow), 2025, Vol. 90, No. 11, pp. 1484-1503 © Pleiades Publishing, Ltd., 2025.
Russian Text © The Author(s), 2025, published in Biokhimiya, 2025, Vol. 90, No. 11, pp. 1579-1600.
1484
REVIEW
Circulating Tumor DNA And Its Potential Applications
for Assessing Effectiveness of Neoadjuvant Drug
Therapy in the Breast Cancer Patients
Tatiana M. Zavarykina
1,2,a
*, Irina V. Pronina
1,2
, Polina S. Mazina
1,2
,
Svetlana V. Khokhlova
2
, and Gennady T. Sukhikh
2
1
N. M. Emanuel Institute of Biochemical Physics, Russian Academy of Science,
119334 Moscow, Russia
2
B. I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology,
Ministry of Health of the Russian Federation, 117997 Moscow, Russia
a
e-mail: tpalievskaya@yandex.ru
Received July 18, 2025
Revised October 8, 2025
Accepted October 15, 2025
AbstractThe review discusses main characteristics and analytical features of the circulating tumor DNA
(ctDNA), which accounts for a minor fraction of the cell-free DNA (cfDNA) in cancer patients. Currently, ctDNA
is considered to be a promising biomarker for assessing treatment efficacy, prognosis, and disease monitoring
in oncology, including breast cancer (BC). A significant proportion of BC patients receive neoadjuvant drug
therapy, effectiveness of which largely determines necessity and extent of subsequent treatment. Determina-
tion of ctDNA could be the most sensitive method for evaluating response to neoadjuvant therapy, as it enables
real-time monitoring of molecular changes during the treatment, prediction of therapeutic response, and as-
sessment of recurrence risk. This approach could become an additional tool for personalization of BC therapy.
DOI: 10.1134/S0006297925602187
Keywords: circulating tumor DNA, breast cancer, neoadjuvant chemotherapy, pathologic response of tumor,
relapse
* To whom correspondence should be addressed.
INTRODUCTION
Breast cancer (BC) is the most common and so-
cially significant malignancy among women, requir-
ing substantial economic resources for the patient
treatment and rehabilitation. Development of cancer,
among other factors, is associated with accumulation
of somatic mutations in the tumor, particularly mu-
tations in the driver genes, which play a major role
in malignant transformation and serve as targets for
targeted therapies. Presence of tumor-associated dis-
turbances in the DNA isolated from blood plasma has
been reported in various types of cancer, including
breast cancer [1], lung cancer [2, 3], colorectal cancer
[4, 5], prostate cancer [6], gastric cancer [7], ovarian
cancer [8], and other malignancies [9-13]. Detection
of the circulating tumor DNA (ctDNA), which carries
genetic alterations unique to the tumor, is a basis for
a new form of liquid biopsy that is being integrated
into the clinical practice. Since most molecular aber-
rations identified in the plasma ctDNA reflect genet-
ic changes present in the primary tumor, the ctDNA
analysis serves as a convenient predictive and prog-
nostic biomarker, as well as a tool for monitoring dis-
ease progression [14-16].
DEVELOPMENT OF THE
CIRCULATING TUMOR DNA CONCEPT
The existence of extracellular nucleic acids was
first demonstrated in the mid-20th century, when
Mandel and Métais (1948) [17] detected and isolated
extracellular nucleic acids from human blood plasma.
In 1966, Tan etal. [18] performed quantitative analysis
of the cell-free DNA (cfDNA) levels in blood samples
CIRCULATING TUMOR DNA IN BC PATIENTS 1485
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
Fig. 1. Forms of cell-free DNA (cfDNA) in the bloodstream.
from the patients with systemic lupus erythematosus.
They were the first to confirm the hypothesis that
the disease-specific cfDNA can circulate in the blood-
stream. Elevated cfDNA levels in the samples of pa-
tient serum provided prospects of its molecular pro-
filing for prediction and management of autoimmune
disorders. Prognostic potential of cfDNA for assessing
cancer treatment efficacy was first demonstrated in
1977. In the study by Leon etal. [19], total cfDNA con-
centration in the serum of the patients with various
cancers was measured and found to be significantly
higher than in the healthy individuals. Furthermore,
the cfDNA concentration correlated with the tumor
response to neoadjuvant therapy, showing decrease
in the patients who responded positively to radiation
treatment [19]. These findings suggested that quanti-
tative cfDNA analysis could be employed to evaluate
efficacy of anticancer therapies, including comparison
of different treatment regimens. In 1999, Silva et  al.
[20] detected ctDNA in the plasma of patients with
BC, and its presence showed a statistically significant
correlation with the clinicopathological characteristics
of the patients.
Today, ctDNA is being actively investigated as a
potential biomarker for cancer diagnosis and as a
minimally invasive tool for assessing treatment effi-
cacy, prognosis, and disease monitoring in oncology.
CHARACTERISTICS OF ctDNA
There are various types of free-circulating nucleic
acids distinguished depending on the origin, such as
genomic DNA, mRNA, viral DNA and RNA, microRNA,
etc. Cell-free DNA (cfDNA) represents fragmented DNA
molecules circulating in biological fluids. It can exist
as a single- or double-stranded DNA with fragment
lengths ranging from 120 to 21,000  bp (on average
160-180  bp). In most cases, cfDNA circulates in the
bloodstream as part of nucleosomes – macromolecular
complexes of histones and DNA [21] – or within ves-
icles [22] (Fig.  1). In these forms, cfDNA is protected
from nuclease degradation and from eliciting immune
responses [22]. In addition, cfDNA can be bound to the
surface of blood cells, such as erythrocytes, through
specific membrane proteins.
In the healthy individuals, cfDNA originates from
several physiological processes, including apoptosis,
phagocytosis, DNA release from neutrophils, and enu-
cleation of erythroid precursors [23]. Plasma cfDNA
is actively secreted by leukocytes (55%), erythroid
precursors (30%), and lymphocytes (12%) [24]. Con-
centration of free-circulating cfDNA in the healthy
individuals typically ranges from 1 to 10  ng/mL [25,
26]. Due to the rapid nuclease degradation, half-life of
free-circulating cfDNA varies between 15  min and 2  h,
ZAVARYKINA et al.1486
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
which makes it suitable for use in modern medical
monitoring approaches [27].
In cancer patients, a fraction of cfDNA consists
of ctDNA, which accounts for approximately 0.01-1.0%
of the total cfDNA. Concentration of ctDNA depends
on the tumor stage and localization, ranging from
5 to 1500 ng/mL [25]. ctDNA could be released into
the bloodstream from the primary tumor cells, dis-
tant metastatic nodes, or circulating in the peripheral
blood tumor cells [28, 29]. Detection of tumor-specif-
ic mutations in cfDNA allows ctDNA to be identified
within the total pool of circulating nucleic acids. Both
qualitative and quantitative mutation composition of
ctDNA vary not only among the cancer types but
also between the patients with the same diagnosis.
For example, in the case of metastasis, ctDNA could
comprise more than 10% of cfDNA, whereas at ear-
ly stages of cancer or during minimal residual dis-
ease (MRD) monitoring, its fraction could be below
0.1% [30]. Although cfDNA is rapidly degraded in
bloodstream, a comparative study employing digital
PCR (dPCR) approach, targeted and whole-genome
sequencing (WGS), comparing different methods of
blood sample collection and storage for isolation
of cfDNA and analysis of ctDNA, demonstrated that
cfDNA remains stable for up to 24h in the EDTA-treat-
ed blood plasma samples at room temperature, up to
48 h at 4°C, and for extended periods when plasma
is appropriately processed and frozen [30]. Howev-
er, the reported acceptable processing intervals for
blood samples in EDTA tubes vary across studies,
ranging from 1-2  h after blood collection [31,  32] to
up to 24  h [30,  33,  34]. Therefore, in routine labora-
tory practice, blood samples collected in the EDTA
tubes for the ctDNA analysis are recommended to
be processed as soon as possible. The highest cfDNA
stability is achieved using specialized blood collection
tubes containing stabilizing reagents, whose perfor-
mance is comparable across different manufactur-
ers [30].
ctDNA has been detected in the patients with
early-stage BC [35-37] and could serve as a prog-
nostic biomarker [38-43]. It enables real-time mon-
itoring of the treatment response, including detec-
tion of minimal residual disease [38-44], and could
be applied to assess therapy efficacy in the patients
with BC [45-53].
DETECTION OF ctDNA
AS A VARIANT OF LIQUID BIOPSY
Analysis of ctDNA in the cancer patients – pri-
marily focused on identifying driver mutations that
determine tumor sensitivity to targeted therapy –
represents a form of liquid biopsy. Although ctDNA
is most commonly analyzed in the peripheral blood,
other biological fluids such as urine, saliva, pleural
or cerebrospinal fluid can also be used, depending
on the tumor type and localization [54]. Liquid bi-
opsy with ctDNA detection offers several advantages
over conventional tissue biopsy. Classical biopsy is an
invasive procedure that is not always feasible due to
the tumor localization. Moreover, it often fails to cap-
ture the complete molecular and genetic profile of the
malignancy because of tumor heterogeneity – that is,
coexistence of multiple tumor cell clones. Presence
of the so-called cancer stem cells contributes to the
ongoing evolution of genetic and biological tumor
properties during therapy and disease progression,
which is nearly impossible to monitor using conven-
tional tissue sampling. Performing repeated tissue
biopsies to assess tumor evolution and response to
therapy, is often complicated due to dispersion of tu-
mor foci, size or localization, and overall condition
of the patient. In particular, sampling from the meta-
static nodes may be technically challenging or risky.
Incontrast, liquid biopsy sampling for ctDNA analysis
is a simple, routine, and minimally invasive proce-
dure. Therefore, biological material can be collected
as frequently as needed to monitor disease dynamics
and to obtain real-time information about the tumor
[55, 56]. Thus, ctDNA analysis serves as a minimally
invasive alternative for the molecular characteriza-
tion of solid tumors.
METHODS FOR DETECTING
CIRCULATING TUMOR DNA
In most solid tumors, including BC, analysis
of ctDNA in blood samples is the most informative
and technically accessible approach. For quantitative
ctDNA analysis, blood plasma is preferred, as adher-
ence to the rigorously standardized pre-analytical
procedures involving double centrifugation minimiz-
es contamination with the genomic DNA from blood
cells, thereby increasing specificity and sensitivity of
the method [57].
Since ctDNA constitutes only a very small fraction
of the total cfDNA (Fig. 1), highly sensitive analytical
techniques are required for its detection. The most
common ctDNA detection approach relies on iden-
tifying tumor-specific mutations within the cfDNA
[58]. Quantitative ratio of normal cfDNA to ctDNA in
blood plasma of the cancer patients is expressed as
the mutant allele fraction (MAF). MAF of 0.1% means
that for every 999 molecules of cfDNA derived from
normal tissue, there is 1 molecule of ctDNA. It is
well established that the MAF values decline as tu-
mor size decreases [59]. For accurate quantitative and
qualitative analysis of ctDNA, various modifications
CIRCULATING TUMOR DNA IN BC PATIENTS 1487
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
of dPCR and next-generation sequencing (NGS) are
employed [60, 61].
The dPCR method allows detection of extreme-
ly small amounts of target DNA (in this case, ctDNA)
even in the presence of a large amount of other
DNA molecules [62], with sensitivity reaching MAF
values as low as 0.001% [63]. Major limitation of the
dPCR-based approaches is the requirement for prior
knowledge of potential mutations and their genomic
locations, which is necessary for primer and probe
design. This method efficiently detects the known
mutations, such as major driver mutations charac-
teristic of the primary tumor or variants associated
with therapeutic response in the specific tumor types
[64]. It correlates well with histopathological findings,
complements them, and provides additional informa-
tion for more precise tumor classification, recurrence
risk assessment, and therapy optimization.
Cancers lacking known mutations require a com-
bined methodological approach for ctDNA analysis,
involving prior identification of molecular–genetic
alterations in the primary tumor  [65]. This is partic-
ularly relevant for the triple-negative BC, in which
no driver mutations have been identified. Whole-ge-
nome sequencing (WGS)  [66] or whole-exome se-
quencing (WES)  [67] can be used to identify somatic
mutations in the tumor. Based on these findings, a
panel of most representative somatic mutations is
generated, including corresponding primer and probe
systems for their detection by dPCR [39].
NGS-based approaches enable detection of a
broad range of mutations – both previously described
and novel ones [68]. ctDNA analysis using NGS can be
performed in two formats: targeted sequencing of the
selected genes or gene panels, and whole-genome ap-
proaches [69]. Whole-genome sequencing allows iden-
tification of genetic alterations without prior analysis
of the primary tumor [58]. In particular, approaches
based on the copy number aberration analysis using
WGS data with different coverage depths have been
proposed [70]. Tumor-specific mutations were iden-
tified based on the ratio of detected variants at dif-
ferent depths of WGS coverage, without the need for
primary tumor analysis [70]. Some systems, such as
the Guardant Reveal multi-cancer assay  [71], evalu-
ate not only genetic but also epigenetic alterations,
namely, CpG methylation. Detection of ctDNA was
performed not by comparing tumor and leukocyte
DNA, but by comparing cfDNA sequencing results
with the reference databases  [72]. Despite numerous
advantages and substantial technological progress,
the NGS-based ctDNA analysis faces several challeng-
es. Detection of rare mutations present at very low
frequencies could be hampered by sequencing errors
[73]. Therefore, specialized bioinformatic approaches
and additional error-correction systems are required
for analysis of the sequencing data aimed at studying
ctDNA [1, 74-76].
An actively developing and increasingly clinically
implemented approach involves creation of the per-
sonalized marker panels for ctDNA analysis. In this
approach, both tumor biopsy material and blood sam-
ples are used for sequencing to identify somatic muta-
tions specific to the individual patient. Whole-genome
[66] or whole-exome [67] sequencing is typically ap-
plied, while targeted sequencing is used less frequent-
ly [77]. The phenomenon of clonal hematopoiesis [16,
78] should be considered when analyzing the paired
blood sample [79]. The resulting mutation panel, re-
flecting the most abundant mutations in the tumor, is
next used for ctDNA detection. An example of such
approach is the commercially available Signatera™
assay (Natera, USA), developed for monitoring MRD,
that is, molecular residual disease in BC and sever-
al other malignancies [80,  81]. Personalized systems
can also evaluate nucleotide substitutions, particular-
ly among the previously identified single-nucleotide
variants (SNVs) [82], and are capable of detecting in-
sertions and deletions [83]. The number of mutations
assessed across different systems ranges from 1-2 up
to 16 (in the Signatera™ assay). This approach enables
tracking of changes in the clonal composition of the
tumor, predicting therapeutic response. Furthermore,
detection of ctDNA in the patient plasma may indicate
the presence of micrometastases [83]. The CloneSight
system, developed in Spain, enables monitoring of BC
recurrence by detecting ctDNA at 6, 12, 18 months
and beyond after completion of therapy [84].
Combined approaches to ctDNA analysis are also
being actively developed [65]. These include the use
of ELISA or conventional molecular biology techniques
integrated with biosensor-based ctDNA detection plat-
forms, such as electrochemical and fluorescent sys-
tems. In these methods, particular attention is paid
to simplifying and accelerating sample preparation
by reducing quality requirements for the biological
material needed for analysis. Furthermore, electro-
chemical detection offers the potential for “point-of-
care” ctDNA testing, since this approach does not re-
quire expensive equipment and specialized laboratory
conditions. In the study by Wang et  al. [85], several
types of sensor elements are described, in which spe-
cific ctDNA-binding receptors – based on antibodies,
aptamers, enzymatic systems, etc. – are immobilized
on the sensor surface. Upon the ctDNA–receptor
binding, a signal is transmitted to the electrode, fol-
lowed by potentiometric or voltammetric analysis.
Photometric sensors could be designed based on the
same principle. Approaches for cfDNA detection in-
volving probe-mediated reactions in solution are also
under development. Such reactions are most often
based on redox reactions, with a resulting product
ZAVARYKINA et al.1488
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
Fig. 2. Methods for detecting circulating tumor DNA (ctDNA).
that could be detected electrochemically, photometri-
cally, or fluorimetrically [86]. The probes could include
single-stranded DNA molecules complementary to the
target ctDNA [87], RNA molecules [86], antibodies [88],
the latter allows identification of ctDNA methylation.
These studies focus on analyzing physical and chem-
ical properties, as well as on quantitative assessment
of the ctDNA content (Fig. 2) [89].
Due to the technical challenges in the develop-
ment of such systems, they have so far been imple-
mented only for the most frequently occurring driver
mutations in the genes such as PIK3CA [90, 91], KRAS
[92], EGFR [93], and MGMT [94].
At present, rapid progress in analytical technol-
ogies suggests that the diversity of approaches for
ctDNA analysis will significantly increase in the near
future, expanding the arsenal of available methods
for ctDNA detection.
POTENTIAL OF ctDNA ANALYSIS
FOR EVALUATING EFFICACY OF NEOADJUVANT
DRUG THERAPY IN BREAST CANCER PATIENTS
Clinicopathological prerequisites for ctDNA
analysis in neoadjuvant treatment of breast can-
cer patients. For the patients with BC, neoadjuvant
drug therapy constitutes an integral part of the man-
agement of both early-stage and locally advanced
disease. In these cases, achieving pathologic complete
response(pCR) is a crucial prognostic factor and is as-
sociated with improved overall survival(OS) and dis-
ease-free survival (DFS) [95-102]. This relationship is
most pronounced in the triple-negative breast cancer
(TNBC) and HER2-positive BC. Typically, neoadjuvant
therapy includes 6-8 cycles of chemotherapy, with
addition of the targeted therapy when indicated (an-
ti-HER2 therapy for HER2-positive tumors). Some on-
cologists believe that such treatment intensity may be
excessive for the early-stage patients with high che-
mosensitivity. Monitoring ctDNA levels during thera-
py may facilitate treatment personalization, enabling
optimization of the therapy duration for each patient.
The response to neoadjuvant chemotherapy
(NACT) provides prognostic information that comple-
ments standard clinicopathological parameters of the
primary tumor, such as molecular biological subtype,
disease stage, and malignancy degree [98, 99]. Cur-
rently, efficacy of NACT is assessed using clinical pa-
rameters and pathomorphological examination of the
resected tissue after surgery. Routine practice relies
on conventional clinical and instrumental methods
CIRCULATING TUMOR DNA IN BC PATIENTS 1489
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
(physical examination, palpation, ultrasound of breast
and regional zones, mammography, MRI of mammary
gland), which often fail to reflect the true therapeutic
effect. The rate of complete response detection may be
up to twofold lower when based on clinical and im-
aging assessment than when determined by pathomo-
rphological examination of postoperative tissue [103].
A key parameter for assessing NACT efficacy is
pCR, characterized by the absence of viable tumor
cells in both breast tissue and lymph nodes upon
pathomorphological examination. The standard meth-
od for evaluating the degree of treatment-induced
pathomorphosis is routine pathological/histological
examination, including determination of the ypT and
ypN categories (post-NACT pathomorphological stag-
ing of tumor to T and N categories) or assessment of
residual cancer burden (RCB). This method involves
manual review of serial histological sections by a pa-
thologist, which typically does not cover the entire
volume of resected tissue, thereby contributing to po-
tential errors due to heterogeneity of biological sam-
ple; furthermore, analytical precision varies widely
across laboratories. Nevertheless, information about
the degree of treatment-induced pathomorphosis is
crucial for determining post-neoadjuvant therapeutic
strategies in the TNBC and HER2-positive BC. A com-
bined analysis has shown that the patients achieving
pCR have a significant survival advantage compared
to those with residual disease after NACT (residual
tumor cells after treatment) [101, 102]. While pCR
is a strong predictor of low recurrence risk, studies
indicate that predicting early metastatic recurrence
in the patients with residual tumors remains less re-
liable [102, 104, 105]. For example, survival analysis
in the I-SPY2 Trial Consortium demonstrated that the
3-year DFS reached 95% among the patients achieving
pCR [102], whereas metastases occurred in 22% of pa-
tients without pCR.
In the United States, several prognostic gene ex-
pression panels associated with BC progression have
been developed, including MammaPrint (Agendia Inc.),
Oncotype DX (Genomic Health), and Prosigna (Vera-
cyte Inc.), which are designed to predict recurrence
and/or metastasis. Based on analysis of these mark-
ers, a decision can be made regarding the necessity of
adjuvant (postoperative) chemotherapy. These panels
have proven their utility for the luminal ER-positive/
HER2-negative tumors, which is the most common
subtype of BC [104]. However, these assays do not re-
flect dynamic state of the tumor, its response to NACT
(prediction of pCR), and therefore cannot be applied to
guide decisions regarding the timing of chemotherapy
completion.
Detection of ctDNA in blood may represent a
more sensitive approach for evaluating the effect of
NACT, showing a strong correlation with pathologi-
cal methods used to assess the degree of pathologi-
cal response. Development and implementation of a
reliable marker reflecting tumor pCR could become
an additional tool for therapy individualization and
could also help determine optimal timing for the
treatment completion. Moreover, a highly relevant
emerging direction in the early-stage BC management
is investigation of whether surgery could be safely
omitted in the patients who achieve pCR. In such
studies, pCR is typically confirmed by the tissue bi-
opsy; however, this approach cannot fully capture the
entire tumor burden. Analysis of ctDNA concentration
and characteristics in these cases may provide a more
informative, sensitive, and specific assessment, as it
reflects molecular tumor dynamics in real time. For
instance, in BC, ctDNA quantification enables a more
accurate evaluation of tumor response to therapy
and facilitates detection of minimal residual disease
(MRD) and tumor resistance to therapy, i.e., molecular
relapse [40, 41, 44, 105, 106]. ctDNA monitoring could
be used for the early detection of the disease recur-
rence [65] and, in the treatment of metastatic disease,
for selecting the most effective treatment regimens
[46, 47, 81]. In this review, we focus on the potential
of ctDNA analysis for assessing NACT efficacy in BC
patients.
CtDNA analysis for detection of minimal re-
sidual disease during primary treatment. Analy-
sis of ctDNA provides opportunities for detection of
MRD, that is, presence of ctDNA in the absence of
other clinical signs of recurrence in the patients who
have completed therapy. Assessing treatment response
based on MRD detection could become an important
component of personalized management strategies,
helping to guide further therapeutic decisions in sub-
sequent observation of BC patients. Feasibility of us-
ing ctDNA to determine MRD following NACT and
surgery has been demonstrated in several studies [1,
52, 105, 106]. Of particular importance is the dynamic
change in ctDNA levels relative to baseline. As early
as 2015, it was shown that ctDNA detection in the
serial samples obtained from the patients after NACT
and subsequent surgery was associated with early
recurrence [39]. These findings were later confirmed
by additional studies involving various subgroups
of BC patients.
CtDNA assessment during NACT. Monitoring
ctDNA during NACT enables prediction of treatment
response, showing correlation with pCR, as well as
with the risk of recurrence and metastasis. Moreover,
the ctDNA positivity at mid-NACT (12 weeks after
treatment initiation) was found to be strongly associ-
ated with the decreased OS and DFS (p =  0.0002 and
p =  0.0034, respectively) [81].
Using a personalized whole-genome approach, the
ctDNA analysis allows evaluation of the relationship
ZAVARYKINA et al.1490
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
between the ctDNA presence, NACT response, recur-
rence risk, metastasis development, and 3-year surviv-
al. In this study, individualized mutation panels were
designed for each patient based on the whole-exome
sequencing of tumor tissue. The early treatment pe-
riod of NACT (3 weeks after therapy initiation) was
identified as a significant time point for pCR predic-
tion (odds ratio (OR)  =  4.33; p =  0.012). After comple-
tion of NACT, all patients who achieved pCR were
ctDNA-negative. Among those who did not achieve
pCR, the ctDNA-positive patients exhibited a higher
risk of metastasis (OR  =  10.4). Patients who failed
to reach ctDNA negativity showed poor treatment
response and increased metastatic risk (OR  =  22.4;
95% CI; p <  0.001), whereas the ctDNA-negative state
was associated with improved survival even in the
absence of pCR [44]. These findings are consistent
with the results of previous works [105,  107] and
the study by Chen et  al. [81], which demonstrated
that the ctDNA absence at mid-NACT (12 weeks after
treatment initiation) was associated with pCR achieve-
ment (p =  0.02).
Subsequent studies revealed relationship be-
tween the ctDNA-negative status and pCR achieve-
ment in the TNBC patients [108], as well as a posi-
tive correlation between the ctDNA levels and RCB
[1, 109]. A correlation was also observed between the
ctDNA positivity and reduced DFS regarding distant
metastases, particularly at mid-NACT (12 weeks af-
ter treatment initiation) and after NACT but before
surgery (p <  0.0001) [108].
Personalized ctDNA monitoring during NACT may
provide a real-time assessment tool for treatment
response and predict pCR as a surrogate survival
marker [44, 104].
CtDNA assessment after NACT. CtDNA detection
after completion of NACT but before surgery could
be considered as a prognostic marker. The presence
of ctDNA has been shown to be associated with poor-
er DFS [76,  110]. One of the earliest studies demon-
strated an increased risk of early recurrence in the
ctDNA-positive patients both after surgery and during
subsequent observation of the patient, with hazard
ratios (HR) of 25.0 and 12.0, respectively. 96% of the
patients without disease recurrence had no detectable
ctDNA (p <  0.0038) [39].
This correlation was most prominent in the
study by Cailleux et  al. (HR  =  53; p <  0.01) [106].
In the ctDNA-positive patients after NACT who did
not achieve pCR, a higher risk of distant metastasis
was observed (HR  =  5.89 for hormone receptor-posi-
tive BC and HR = 3.79 for TNBC) [108]. These findings
were subsequently confirmed by additional studies
involving various BC patient subgroups [41, 76, 109].
Using common mutations in the signaling path-
ways associated with BC progression is suggested
to be a promising approach for ctDNA quantification
and treatment response evaluation. It was found that
in the BC patients carrying PIK3CA and/or TP53 mu-
tations in the tumor tissue, absence of these muta-
tions in the cfDNA analysis after NACT corresponded
to achievement of pCR in 93.33% of cases [111]. For the
HER2-positive BC subtype, the analysis of TP53 mu-
tations in cfDNA has been shown to be a marker of
resistance to anti-HER2 therapy with monoclonal an-
tibodies [112], as well as the presence of PIK3CA and/
or TP53 mutations in cfDNA has been identified as a
predictor of poor response to anti-HER2 treatment [64].
CtDNA assessment after NACT and surgery. For
the ctDNA-positive patients after NACT and surgical
treatment, an earlier disease recurrence (reduced RFS)
was observed [113, 114]. CtDNA monitoring in the BC
patients with residual tumors after NACT demonstrat-
ed that persistence of ctDNA positivity after NACT
and surgery was associated with the decreased RFS.
This approach could enable identification of the pa-
tient subgroups requiring more intensive therapy, im-
proving their survival outcomes [115]. Association of
ctDNA with the disease recurrence and progression
was also observed during the post-NACT observation
[72, 114].
Baseline ctDNA levels. It should be noted that
ctDNA serves both as a predictive and as prognostic
marker when analyzed during or after NACT, prior
to surgery, and throughout subsequent treatment or
observation. However, the pretreatment ctDNA lev-
els are not significant in the TNBC or heterogeneous
BC cohorts, as no association has been observed be-
tween the presence of ctDNA before NACT and RFS
[44, 105, 106]. At the same time, correlation between
the ctDNA levels and clinicopathological character-
istics of BC – such as tumor size, lymph node me-
tastasis, and tumor grade – has been reported [108].
However, in the HER2-positive subgroup, the ctDNA
positivity (based on detection of PIK3CA and/or TP53
point mutations) before initiation of anti-HER2 thera-
py was found to be associated with the lower rate of
pCR achievement [64].
Potential of ctDNA analysis in TNBC patients.
The triple-negative subtype represents the most ag-
gressive form of BC, lacking known targets for hor-
mone or targeted therapies, with chemotherapy con-
tinuing to be the standard therapeutic approach. In
recent years, growing interest has been directed to-
ward evaluating response to NACT in the TNBC pa-
tients through changes in the blood levels of ctDNA.
For the TNBC patients, detection of the ctDNA-posi-
tive status has been associated with high incidence
of metastatic disease (nearly 72%) [38]. The potential
of ctDNA analysis for this most unfavorable BC sub-
type has been demonstrated in the long-term study
with up to 12 years of observation, where the
CIRCULATING TUMOR DNA IN BC PATIENTS 1491
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
Table 1. Key studies on ctDNA analysis for assessing the efficacy of NACT
Patients in the study Main results Reference
Early-stage BC ctDNA positivity after NACT was associated with an increased
risk of early recurrence, both after surgery and during
subsequent observation, with hazard ratios (HR) of 25.0 and 12.0,
respectively (p <  0.0001);
ctDNA was undetectable in 96% of patients who remained
recurrence-free, both at the postoperative time point (p< 0.0038)
and during subsequent observation (p <  0.0001)
[39]
TNBC absence of ctDNA in the middle of NACT (12 weeks after
the initiation of therapy) was associated with achieving pCR (p =  0.02);
ctDNA positivity at mid-NACT (12 weeks after the initiation of therapy)
was strictly associated with the decreased OS and DFS (p =  0.0002
and p =  0.0034, respectively)
[81]
Stage I and II BC presence of ctDNA after completion of NACT and prior to surgery
was associated with shorter DFS
[111]
Early-stage BC presence of ctDNA after completion of NACT and prior to surgery
was associated with shorter DFS (HR  =  53; p <  0.01)
[106]
High-risk stage II
and III BC
before treatment, 73% of the patients were ctDNA-positive,
and this proportion decreased during therapy;
importantly, the early stage of NACT (3 weeks after treatment
initiation) was identified as a critical time point for predicting pCR
(odds ratio (OR) =  4.33; p =  0.012);
a ctDNA-negative status at this early stage correlated
with achieving pCR;
after completion of NACT, all patients who achieved pCR
were ctDNA-negative (n =  17; 100%);
among those who did not achieve pCR (n =  43), the ctDNA-positive
patients (14%) had an increased risk of metastasis (OR  =  10.4);
the patients who failed to reach ctDNA negativity demonstrated
poor response to treatment and higher risk of metastasis (OR  =  22.4;
95% CI, 2.5-201; p <  0.001), whereas ctDNA negativity was associated
with improved survival even in the patients who did not achieve pCR
[44]
Localized BC patients exhibited significantly higher levels of cfDNA
from mammary gland compared to the healthy donors
(p =  5.3  ×  10
−12
; AUCROC  =  91.25% (83.79–98.71%));
BC patients could be identified with 80% sensitivity
while maintaining 97% specificity;
presence of mammary gland specific cfDNA at the end
of chemotherapy reflected the presence of residual disease
[107]
Hormone receptor
(HR)-positive/
HER2-negative BC
and TNBC
ctDNA-negative status at 3 weeks after treatment initiation was
associated with achieving pCR in the TNBC patients;
correlation between the ctDNA levels during NACT and pCR
achievement was observed in TNBC, but not in the HR-positive BC;
ctDNA levels were also linked to clinicopathological features of BC
such as tumor size, lymph node metastasis, and histological grade;
among the patients who did not achieve pCR, the ctDNA positivity
after NACT was associated with higher risk of distant metastasis
(HR  =  5.89 for HR-positive BC and HR  =  3.79 for TNBC), whereas
the ctDNA negativity indicated a favorable prognosis even
in the patients with residual disease;
tumor mRNA analysis before treatment revealed correlation between
the ctDNA appearance in circulation, cell cycle, and immune-related
signaling pathways
[108]
ZAVARYKINA et al.1492
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
Table 1 (cont.)
Patients in the study Main results Reference
Stage II and III BC patients who achieved complete response exhibited
a more pronounced decrease in the ctDNA levels during
postoperativetherapy;
early changes in the ctDNA levels during treatment had significant
prognostic value for the BC patients;
correlation was observed between the early ctDNA decline
and longer DFS compared with the patients showing increase
in the ctDNA levels (HR  =  12.54; 95%  CI 2.084-75.42; p =  0.0063)
[116]
Hormone
receptor-positive/
HER2-negative BC
and TNBC
a positive correlation between the ctDNA levels and RCB
was established;
in TNBC, a moderate negative correlation between the cfDNA
concentration and RCB was detected at 3 weeks after treatment
initiation;
at the same time, ctDNA concentration showed a significant
positive correlation with RCB at all time points (correlation
coefficient R  >  0.3; p <  0.05);
in the HR-positive/HER2-negative BC, the cfDNA concentration
was not associated with the response to NACT; however, the patients
with high baseline cfDNA levels had significantly shorter distant
DFS compared to those with low cfDNA levels (HR = 2.12; p = 0.037);
in TNBC, survival differences between the patients with high and low
cfDNA levels at all time points were not statistically significant;
meanwhile, the ctDNA levels at all time points showed significant
correlation with the distant DFS in both subtypes
[109]
Early-stage BC positive correlation between the ctDNA levels and RCB was identified
detection of ctDNA at mid-therapy was significantly associated
with the higher RCB (OR =  0.062; 95%  CI 0.01-0.48; p =  0.0077),
allowing identification of the patients who would not achieve pCR
and would be classified as RCB II/III
[1]
Early-stage TNBC
with residual
disease after NACT
presence of ctDNA after completion of NACT and prior
to surgery was associated with the shorter distant DFS (median
distant DFS, 32.5 months vs. not reached; HR  =  2.99;
95% CI  1.38–6.48; p =  0.006);
detection of ctDNA was also associated with worse DFS
(HR  =  2.67; 95%  CI 1.28–5.57; p =  0.009) and OS (HR  =  4.16;
95%  CI 1.66–10.42; p =  0.002)
[76]
TNBC
and HER2-positive BC
in the BC patients carrying PIK3CA and/or TP53 mutations in the
tumor tissue, absence of these mutations in the ctDNA after NACT
corresponded to achievement of pCR in 93.33% of cases; moreover,
presence of mutations in the ctDNA allowed exclusion of several
patients from the pCR group with an accuracy of 89.47%
[111]
HER2-positive BC presence of mutations in the TP53 gene in the ctDNA
was evaluated as a potential marker of resistance to anti-HER2
monoclonal antibody therapy in the HER2-positive subtype of BC;
the patients harboring mutations in the TP53 gene who received
anti-HER2 therapy had significantly shorter DFS (p =  0.004);
in the patients with mutations in the TP53 gene, a trend
toward poorer prognosis under the anti-HER2 antibody
treatment was observed compared to those with wild-type TP53
in the phase II study (p =  0.15), and this trend was confirmed
in the combined analysis of the MutHER and SUMMIT
cohorts (p =  0.01)
[112]
CIRCULATING TUMOR DNA IN BC PATIENTS 1493
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
Table 1 (cont.)
Patients in the study Main results Reference
HER2-positive BC significance of the PIK3CA and/or TP53 mutations detected
in the ctDNA as predictors of poor response to anti-HER2 therapy was
demonstrated;
ctDNA positivity, defined by the presence of point mutations
in PIK3CA and/or TP53, was associated with the lower rate of pCR,
particularly at the stage prior to initiation of anti-HER2 therapy;
detection of ctDNA before NACT was associated with the reduced
likelihood of achieving pCR (HR =  0.15; 95%  CI 0.034-0.7; p =  0.0089),
but not with DFS;
notably, the patients with HER2-positive tumors and absence
of ctDNA prior to treatment exhibited the highest pCR rates;
whereas those with detectable ctDNA both before treatment
and at week 2 had the lowest pCR rates
[64]
High-risk BC in the patients, who remained ctDNA-positive after NACT
and surgery, earlier recurrence was reported (reflected
by decreased DFS);
the Invitae Personalized Cancer Monitoring™ (PCM) panel, used
for monitoring before and during neoadjuvant therapy, after
surgery, and throughout observation, demonstrated 100% specificity
and prognostic value;
all patients (10 of 61; 16%) with detectable ctDNA during
the monitoring period subsequently developed recurrence;
detection of ctDNA during monitoring was associated
with the markedly increased risk of future recurrence (HR =  37.2;
95%  CI  10.5-131.9; p <  0.0001), with a median lead time from ctDNA
detection to clinical recurrence of 11.7 months
[113]
Early-stage BC the patients who remained ctDNA-positive after NACT and surgery
experienced earlier recurrence (reflected by the reduced DFS);
correlation between the ctDNA detection and disease recurrence
or progression was observed, including during post-NACT observation;
preoperative ctDNA detection was significantly associated
with the decreased DFS (adjusted HR  =  3.09; 95%  CI 2.65-80.0;
p =  0.001);
after a median observation period consisted of 26.6 months among
11 patients, recurrence occurred in 5 cases, all of whom had detectable
ctDNA at the 2-4-week postoperative time point;
ctDNA clearance at this time point was associated with significantly
longer DFS (p =  0.0009), whereas persistent ctDNA positivity
after adjuvant therapy was observed in 36.4% (4 of 11)
of the patients with stage III disease;
during monitoring, ctDNA detection demonstrated sensitivity
of 90.9% and specificity of 98.8% for predicting recurrence,
with the median lead time of 9.7 months;
the patients with detectable ctDNA exhibited significantly shorter
progression-free survival than those without detectable ctDNA
(adjusted HR  =  207.05; 95%  CI 41.38-1000; p =  0.001);
therefore, the ctDNA status both before and after surgery could aid
in risk stratification for recurrence in breast cancer patients
[114]
Non-metastatic BC
with residual disease
monitoring of ctDNA in the BC patients with residual disease after
NACT demonstrated that persistent ctDNA positivity following NACT
and surgery was associated with the decreased DFS;
the 4-year DFS rate was 100% in the patients who were ctDNA-negative
before treatment, compared with only 67% in the ctDNA-positive
patients (p =  0.032)
[115]
ZAVARYKINA et al.1494
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
Table 1 (cont.)
Patients in the study Main results Reference
Early-stage BC association between ctDNA and disease recurrence or progression
was also observed during the long-term monitoring after NACT;
in this study, only plasma samples were analyzed, without prior
identification of tumor-specific mutations from the biopsy samples;
to detect ctDNA, a bioinformatic classifier was developed to identify
tumor-associated somatic variants and methylation profiles
for the specific cancer types using the 5-Mb NGS panels;
ctDNA was detected during or before distant recurrence
in 11 of 14 (79%) patients, with sensitivity of 85% (11 of 13)
among the samples collected within 2 years prior to recurrence;
lead time was estimated in 4 of 6 (67%) of the ctDNA-positive
samples collected before distant recurrence and ranged
from 3.4 to 18.5 months;
no ctDNA was detected in the samples from the patients
without recurrence (n = 13)
[72]
Primary BC prediction of recurrence with the lead time of up to 38 months
(median, 10.5 months; range, 0-38 months) was demonstrated,
with ctDNA positivity being associated with shorter distant DFS
(p <  0.0001) and OS (p <  0.0001) during the long-term monitoring
over 12 years; the most significant results were observed
in the patients with TNBC;
among all patients with recurrent TNBC (n =  7 of 23),
the ctDNA positivity was detected within a median lead time
of 8 months (range, 0-19 months), whereas in 16 TNBC patients
without recurrence, ctDNA remained undetectable throughout
a median observation period of 58 months (range, 8-99months)
[65]
Early-stage TNBC according to the results of the meta-analysis including
1202 patients with TNBC, ctDNA positivity detected after NACT,
either before or after surgery, was associated with the risk
of recurrence and OS (HR  =  3.26; 95%  CI 1.88-5.63)
in this BC subtype
[117]
Stage II-III early TNBC in the patients with early-stage TNBC, prognostic significance
of ctDNA was demonstrated, allowing identification
of the subgroups with high treatment sensitivity and increased risk
of disease recurrence;
a threshold value of the maximum allele frequency
of 1.1% at baseline stratifies patients according to recurrence risk,
as confirmed by both internal and external quality controls;
the systemic tumor burden model integrating baseline
and postoperative ctDNA represents an independent prognostic
model (p =  0.022);
combination of the systemic tumor burden with pathological response
allows identification of a subgroup of the patients with high likelihood
of cure and a subgroup of the patients with high-risk early-stage TNBC;
long-term ctDNA monitoring enables detection of the patients
with high risk of recurrence
[118]
most reliable results were obtained for TNBC [65].
Magbanua et al. [108] reported that correlation be-
tween the ctDNA levels during NACT and achieve-
ment of pCR was observed in TNBC, but not in the
hormone receptor-positive BC. Several studies have
also shown that the ctDNA-negative status correlates
with improved RFS and distant metastasis-free sur-
vival in the patients with TNBC [108, 116]. Meta-
analysis including 1,202 TNBC patients further con-
firmed correlation between the ctDNA positivity and
increased risk of recurrence and decreased OS in this
BC subgroup [117]. In another recent study involving
CIRCULATING TUMOR DNA IN BC PATIENTS 1495
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
patients with early-stage TNBC, ctDNA demonstrated
prognostic relevance, enabling identification of the
patients highly sensitive to therapy as well as those
at high risk of disease recurrence [118].
The key findings from the studies evaluating
ctDNA analysis for NACT efficacy and disease progno-
sis are summarized in Table  1. The accumulated body
of evidence has formed the basis for the development
of the commercial Signatera assay for MRD detection,
based on ctDNA analysis in blood plasma (Natera) of
the BC patients. This assay employs whole-exome –
and potentially whole-genome – sequencing of tumor
tissue, followed by ultra-deep sequencing of plasma
ctDNA and bioinformatic analysis. The method is al-
ready being used in clinical trials for ctDNA analy-
sis [65, 81].
CONCLUSION
Thus, persistence of ctDNA during neoadjuvant
therapy and after primary treatment is associated
with an increased risk of recurrence and an unfavor-
able prognosis. In recent years, post-neoadjuvant ther-
apy approaches have been supplemented with sever-
al highly effective agents. In particular, the CDK4/6
inhibitors are used in the hormone receptor-positive
HER2-negative BC, TD-M1 in the HER2-positive BC,
capecitabine and immunotherapy in TNBC, while
the PARP inhibitor olaparib has proven effective in
the BRCA-associated BC. However, despite their high
efficacy, these agents do not achieve complete cure
in all patients and are associated with considerable
toxicity. Quantitative and qualitative assessment of
ctDNA is important not only from the therapeutic
and prognostic perspective for optimizing treatment
strategies, but also from an economic standpoint,
since personalized therapy can markedly reduce
treatment costs. Detection of cfDNA after neoadju-
vant therapy and surgical treatment could help iden-
tify patient subgroups, based on the ctDNA biologi-
cal characteristics, for whom appropriate subsequent
systemic therapy is indicated.
The ctDNA analysis also enables regular molec-
ular monitoring throughout treatment and subse-
quent observation. Integration of ctDNA analysis, as
a form of liquid biopsy, into routine clinical practice
will make it possible to track real-time tumor re-
sponse, predict disease progression, and guide deci-
sions regarding treatment duration. Compared with
the conventional tissue biopsy, the ctDNA testing is
suggested to be a more convenient and minimally
invasive biomarker, and could become an additional
tool for personalized therapy optimization.
At present, investigation of ctDNA as a marker
of response to neoadjuvant therapy in the BC pa-
tients is limited only to clinical trials. Nevertheless,
the success of ctDNA-based approaches in colorectal
cancer – for which the biomarker has already been
incorporated into the ASCO (American Society of Clin-
ical Oncology) recommendations – and the growing
body of evidence from the ctDNA studies in BC sug-
gest that clinical implementation of ctDNA as a sensi-
tive predictive and prognostic BC biomarker is likely
in the near future.
Abbreviations
95% CI 95% confidence interval
BC breast cancer
cfDNA cell-free DNA
ctDNA circulating tumor DNA
DFS disease-free survival
dPCR digital PCR
HR hazard ratio, risk ratio
MAF mutant allele fraction
MRD minimal residual disease
NACT neoadjuvant chemotherapy
NGS next-generation sequencing
OS overall survival
RCB residual cancer burden
pCR pathologic complete response
TNBC triple-negative breast cancer
Contributions
G.T.S. supervised the work and performed review
of the final manuscript with subsequent approval;
T.M.Z. came up with the concept of the work, car-
ried out the literature search, prepared the manu-
script including structuration and editing; I.V.P. pro-
vided the section “Methods for detecting circulating
tumor DNA” and edited the manuscript; P.S.M. pro-
vided the section “Methods for detecting circulating
tumor DNA” as well as technical preparation of the
manuscript; S.V.Kh. provided the section “Potential
of ctDNA analysis for evaluating the efficacy of neo-
adjuvant drug therapy in breast cancer patients”
and participated in manuscript editing.
Funding
This study was financially supported by the Min-
istry of Health of the Russian Federation (project
no. 125050605836-5) and by the Ministry of Science
and Higher Education of the Russian Federation
(project no.122041400080-0).
Ethics approval and consent to participate
This work does not contain any studies involving hu-
man and animal subjects.
Conflict of interest
The authors of this work declare that they have
noconflicts of interest.
ZAVARYKINA et al.1496
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
REFERENCES
1. Zhou, Q., Gampenrieder, S.P., Frantal,S., Rinnerthal-
er, G., Singer, C. F., Egle, D., Pfeiler, G., Bartsch, R.,
Wette, V., Pichler, A., Petru, E., Dubsky, P. C., Bago-
Horvath,Z., Fesl,C., Rudas,M., Ståhlberg,A., Graf,R.,
Weber, S., Dandachi, N., Filipits, M., and Heitzer, E.
(2022) Persistence of ctDNA in patients with breast
cancer during neoadjuvant treatment is a Signifi-
cant predictor of poor tumor response, Clin. Cancer
Res., 28, 697-707, https://doi.org/10.1158/1078-0432.
CCR-21-3231.
2. Cheng,L., Gao,G., Zhao, C., Wang,H., Yao,C., Yu,H.,
Yao, J., Li, F., Guo, L., Jian, Q., Chen, X., Li, X., and
Zhou, C. (2023) Personalized circulating tumor DNA
detection to monitor immunotherapy efficacy and
predict outcome in locally advanced or metastatic
non-small cell lung cancer, Cancer Med., 12, 14317-
14326, https://doi.org/10.1002/cam4.6108.
3. Moiseenko,F., Stepanova,M., Volkov,N., Zhabina,A.,
Myslik, A., Meldo, A., Rysev, N., Krylova, D.,
Klimenko, V., Bogdanov, A., and Moiseenko, V.
(2020) Predictive value of dynamic circulating tu-
mor DNA assessment during osimertinib therapy
in patients with EGFR-mutated non-small cell lung
cancer, Problems Oncol., 66, 135-142, https://doi.org/
10.37469/0507-3758-2020-66-2-135-142.
4. Stasik, S., Mende, M., Schuster, C., Mahler, S.,
Aust, D., Tannapfel, A., Reinacher-Schick, A., Baret-
ton,G., Krippendorf,C., Bornhäuser,M., Ehninger,G.,
Folprecht, G., and Thiede, C. (2022) Sensitive quan-
tification of cell-free tumor DNA for early detection
of recurrence in colorectal cancer, Front. Genet., 12,
811291, https://doi.org/10.3389/fgene.2021.811291.
5. Polyanskaya, E. M., Fedyanin, M. Yu., Boyarskikh,
U.A., Kechin, A.A., Moroz, E.A., Khrapov, E.A., Osko-
robin, I.P., Shamovskaya, D.V., Aliev, V.A., Mamedli,
Z.Z., Tryakin, A.A., Filipenko, M.L., and Tyulyandin,
S.A. (2022) Prognostic value of circulating tumor DNA
in blood as a marker of minimal residual disease in
stage I-III colorectal cancer, Adv. Mol. Oncol., 9, 32-42,
https://doi.org/10.17650/2313-805X-2022-9-2-32-42.
6. Fei, X., Du, X., Gong, Y., Liu, J., Fan, L., Wang, J.,
Wang,Y., Zhu,Y., Pan,J., Dong,B., and Xue,W. (2023)
Early plasma circulating tumor DNA as a potential
biomarker of disease recurrence in non-metastat-
ic prostate cancer, Cancer Res. Treat., 55, 969-977,
https://doi.org/10.4143/crt.2022.1557.
7. Yang, J., Gong, Y., Lam, V. K., Shi, Y., Guan, Y.,
Zhang, Y., Ji, L., Chen, Y., Zhao, Y., Qian, F., Chen, J.,
Li,P., Zhang,F., Wang,J., Zhang,X., Yang,L., Kopetz,S.,
Futreal, P.A., Zhang,J., Yi, X., and Yu,P. (2020) Deep
sequencing of circulating tumor DNA detects mo-
lecular residual disease and predicts recurrence in
gastric cancer, Cell Death Disease, 11, 346, https://
doi.org/10.1038/s41419-020-2531-z.
8. Hou, J. Y., Chapman, J. S., Kalashnikova, E., Pier-
son, W., Smith-McCune, K., Pineda, G., Vattakalam,
R. M., Ross, A., Mills, M., Suarez, C. J., Davis, T., Ed-
wards, R., Boisen, M., Sawyer, S., Wu, H. T., Dash-
ner, S., Aushev, V. N., George, G. V., Malhotra, M.,
Zimmermann, B., and Ford, J. M. (2022) Circulating
tumor DNA monitoring for early recurrence detec-
tion in epithelial ovarian cancer, Gynecol. Oncol., 167,
334-341, https://doi.org/10.1016/j.ygyno.2022.09.004.
9. Fu, Y., Yang, Z., Hu, Z., Yang, Z., Pan, Y., Chen, J.,
Wang, J., Hu, D., Zhou, Z., Xu, L., Chen, M., and
Zhang, Y. (2022) Preoperative serum ctDNA predicts
early hepatocellular carcinoma recurrence and re-
sponse to systemic therapies, Hepatol. Int., 16, 868-
878, https://doi.org/10.1007/s12072-022-10348-1.
10. Le Guin, C. H. D., Bornfeld, N., Bechrakis, N. E.,
Jabbarli, L., Richly, H., Lohmann, D. R., and
Zeschnigk, M. (2021) Early detection of metastatic
uveal melanoma by the analysis of tumor-specific
mutations in cell-free plasma DNA, Cancer Med., 10,
5974-5982, https://doi.org/10.1002/cam4.4153.
11. Moss, E. L., Gorsia, D. N., Collins, A., Sandhu, P.,
Foreman, N., Gore, A., Wood, J., Kent, C., Silcock, L.,
and Guttery, D. S. (2020) Utility of circulating tumor
DNA for detection and monitoring of endometri-
al cancer recurrence and progression, Cancers, 12,
2231, https://doi.org/10.3390/cancers12082231.
12. Zhou, M., Bui, N., Rathore, R., Sudhaman,S., George,
G. V., Malashevich, A. K., Malhotra, M., Liu, M. C.,
Aleshin,A., and Ganjoo, K.N. (2022) Feasibility of lon-
gitudinal ctDNA assessment in patients with uterine
and extra-uterine leiomyosarcoma, Cancers, 15, 157,
https://doi.org/10.3390/cancers15010157.
13. Wang, X., Yu, N., Cheng, G., Zhang, T., Wang, J.,
Deng, L., Li, J., Zhao, X., Xu, Y., Yang, P., Bai, N.,
Li, Y., and Bi, N. (2022) Prognostic value of circulat-
ing tumour DNA during post-radiotherapy surveil-
lance in locally advanced esophageal squamous cell
carcinoma, Clin. Trans lat. Med., 12, e1116, https://
doi.org/10.1002/ctm2.1116.
14. Loupakis, F., Sharma, S., Derouazi, M., Murgioni, S.,
Biason, P., Rizzato, M. D., Rasola, C., Renner, D.,
Shchegrova,S., Koyen Malashevich,A., Malhotra,M.,
Sethi, H., Zimmermann, B. G., Aleshin, A., Moshkev-
ich, S., Billings, P. R., Sedgwick, J. D., Schirripa, M.,
Munari,G., Cillo, U., and Fassan,M. (2021) Detection
of molecular residual disease using personalized cir-
culating tumor DNA assay in patients with colorec-
tal cancer undergoing resection of metastases, JCO
Precis. Oncol., 5, PO.21.00101, https://doi.org/10.1200/
PO.21.00101.
15. Shaw, J. A., Page, K., Blighe,K., Hava,N., Guttery, D.,
Ward, B., Brown, J., Ruangpratheep, C., Stebbing, J.,
Payne, R., Palmieri, C., Cleator, S., Walker, R. A., and
Coombes, R. C. (2012) Genomic analysis of circulat-
ing cell-free DNA infers breast cancer dormancy,
CIRCULATING TUMOR DNA IN BC PATIENTS 1497
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
Genome Res., 22, 220-231, https://doi.org/10.1101/gr.
123497.111.
16. Chin, Y. M., Takahashi, Y., Chan, H. T., Otaki, M.,
Fujishima, M., Shibayama, T., Miki, Y., Ueno, T.,
Nakamura, Y., and Low, S. K. (2021) Ultradeep tar-
geted sequencing of circulating tumor DNA in plas-
ma of early and advanced breast cancer, Cancer Sci.,
112, 454-464, https://doi.org/10.1111/cas.14697.
17. Mandel,P., and Metais,P. (1948) Nuclear acids in hu-
man blood plasma [in French], Rep. Meet. Biol. Soc.
Subsid., 142, 241-243.
18. Tan, E. M., Schur, P. H., Carr, R. I., and Kunkel, H.G.
(1966) Deoxybonucleic acid (DNA) and antibodies to
DNA in the serum of patients with systemic lupus
erythematosus, J.Clin. Invest., 45, 1732-1740, https://
doi.org/10.1172/JCI105479.
19. Leon, S. A., Green, A., Yaros, M. J., and Shapiro, B.
(1975) Radioimmunoassay for nanogram quantities
of DNA, J. Immunol. Methods, 9, 157-164, https://
doi.org/10.1016/0022-1759(75)90106-4.
20. Silva, J.M., Dominguez,G., Garcia, J.M., Gonzalez,R.,
Villanueva, M. J., Navarro, F., Provencio, M., San
Martin,S., España,P., and Bonilla, F. (1999) Presence
of tumor DNA in plasma of breast cancer patients:
clinicopathological correlations, Cancer Res., 59,
3251-3256.
21. Roth, C., Pantel, K., Müller, V., Rack, B., Kasimir-
Bauer, S., Janni, W., and Schwarzenbach, H. (2011)
Apoptosis-related deregulation of proteolytic activi-
ties and high serum levels of circulating nucleosomes
and DNA in blood correlate with breast cancer pro-
gression, BMC Cancer, 11, 4, https://doi.org/10.1186/
1471-2407-11-4.
22. Marsman, G., Zeerleder, S., and Luken, B. M. (2016)
Extracellular histones, cell-free DNA, or nucle-
osomes: differences in immunostimulation, Cell
Death Disease, 7, e2518, https://doi.org/10.1038/cddis.
2016.410.
23. Suzuki, N., Kamataki, A., Yamaki, J., and Homma, Y.
(2008) Characterization of circulating DNA in healthy
human plasma, Clin. Chim. Acta, 387, 55-58, https://
doi.org/10.1016/j.cca.2007.09.001.
24. Moss, J., Magenheim, J., Neiman, D., Zemmour, H.,
Loyfer,N., Korach,A., Samet, Y., Maoz,M., Druid, H.,
Arner, P., Fu, K. Y., Kiss, E., Spalding, K. L., Landes-
berg, G., Zick, A., Grinshpun, A., Shapiro, A. M. J.,
Grompe,M., Wittenberg, A. D., Glaser, B., and Dor, Y.
(2018) Comprehensive human cell-type methylation
atlas reveals origins of circulating cell-free DNA in
health and disease, Nat. Commun., 9, 5068, https://
doi.org/10.1038/s41467-018-07466-6.
25. Thierry, A.R., El Messaoudi,S., Gahan, P.B., Anker,P.,
and Stroun, M. (2016) Origins, structures, and func-
tions of circulating DNA in oncology, Cancer Me-
tastas. Rev., 35, 347-376, https://doi.org/10.1007/
s10555-016-9629-x.
26. Gao, Y., Zhao, H., An, K., Liu, Z., Hai, L., Li, R.,
Zhou, Y., Zhao, W., Jia, Y., Wu, N., Li, L., Ying, J.,
Wang, J., Xu, B., Wu, Z., Tong, Z., He, J., and Sun, Y.
(2022) Whole- genome bisulfite sequencing analysis of
circulating tumour DNA for the detection and molec-
ular classification of cancer, Clin. Translat. Med., 12,
e1014, https://doi.org/10.1002/ctm2.1014.
27. Diehl,F., Schmidt, K., Choti, M.A., Romans,K., Good-
man, S., Li, M., Thornton, K., Agrawal, N., Sokoll, L.,
Szabo, S. A., Kinzler, K. W., Vogelstein, B., and Diaz,
L. A., Jr. (2008) Circulating mutant DNA to assess
tumor dynamics, Nat. Med., 14, 985-990, https://
doi.org/10.1038/nm.1789.
28. Crowley, E., Di Nicolantonio, F., Loupakis, F., and
Bardelli,A. (2013) Liquid biopsy: monitoring cancer-
genetics in the blood, Nat. Rev. Clin. Oncol., 10,
472-484, https://doi.org/10.1038/nrclinonc.2013.110.
29. Diaz, L.A.,Jr., and Bardelli,A. (2014) Liquid biopsies:
genotyping circulating tumor DNA, J.Clin. Oncol., 32,
579-586, https://doi.org/10.1200/JCO.2012.45.2011.
30. Risberg,B., Tsui, D. W. Y., Biggs, H., Ruiz-Valdepenas
Martin de Almagro, A., Dawson, S. J., Hodgkin, C.,
Jones, L., Parkinson, C., Piskorz, A., Marass, F.,
Chandrananda, D., Moore, E., Morris, J., Plagnol, V.,
Rosenfeld, N., Caldas, C., Brenton, J. D., and Gale, D.
(2018) Effects of collection and processing proce-
dures on plasma circulating cell-free DNA from
cancer patients, J. Mol. Diagn., 20, 883-892, https://
doi.org/10.1016/j.jmoldx.2018.07.005.
31. Markus, H., Contente-Cuomo, T., Farooq, M., Liang,
W. S., Borad, M. J., Sivakumar, S., Gollins, S., Tran,
N. L., Dhruv, H. D., Berens, M. E., Bryce, A., Seku-
lic, A., Ribas, A., Trent, J. M., LoRusso, P. M., and
Murtaza, M. (2018) Evaluation of pre-analytical fac-
tors affecting plasma DNA analysis, Sci. Rep., 8, 7375,
https://doi.org/10.1038/s41598-018-25810-0.
32. Zhao, Y., Li, Y., Chen, P., Li, S., Luo, J., and Xia, H.
(2019) Performance comparison of blood collec-
tion tubes as liquid biopsy storage system for min-
imizing cfDNA contamination from genomic DNA,
J.Clin. Lab. Anal., 33, e22670, https://doi.org/10.1002/
jcla.22670.
33. Meddeb, R., Pisareva, E., and Thierry, A. R. (2019)
Guidelines for the preanalytical conditions for ana-
lyzing circulating cell-free DNA, Clin. Chem., 65, 623-
633, https://doi.org/10.1373/clinchem.2018.298323.
34. Nesic, M., Bodker, J. S., Terp, S. K., and Dybkaer, K.
(2021) Optimization of preanalytical variables for
cfDNA processing and detection of ctDNA in ar-
chival plasma samples, Biomed. Res. Int., https://
doi.org/10.1155/2021/5585148.
35. Cohen, J. D., Li, L., Wang, Y., Thoburn, C., Afsari, B.,
Danilova, L., Douville, C., Javed, A. A., Wong, F.,
Mattox, A., Hruban, R. H., Wolfgang, C. L., Goggins,
M.G., Dal Molin,M., Wang, T.L., Roden,R., Klein, A.P.,
Ptak,J., Dobbyn,L., Schaefer,J., and Papadopoulos,N.
ZAVARYKINA et al.1498
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
(2018) Detection and localization of surgically re-
sectable cancers with a multi-analyte blood test,
Science, 359, 926-930, https://doi.org/10.1126/science.
aar3247.
36. Jiménez-Rodríguez, B., Alba-Bernal, A., López-
López,E., Quirós-Ortega, M.E., Carbajosa,G., Garrido-
Aranda, A., Álvarez, M., Godoy-Ortiz, A., Queipo-
Ortuño, M.I., Vicioso,L., Díaz-Córdoba,G., Roldán-Díaz,
M. D., Velasco-Suelto, J., Hernando, C., Bermejo, B.,
Julve-Parreño, A., Lluch, A., Pascual, J., Comino-
Méndez, I., and Alba, E. (2022) 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, 146, https://
doi.org/10.3390/ijms24010146.
37. Rodriguez, B. J., Córdoba, G. D., Aranda, A. G.,
Álvarez, M., Vicioso, L., Pérez, C. L., Hernando, C.,
Bermejo, B., Parreño, A. J., Lluch, A., Ryder, M. B.,
Jones, F. S., Fredebohm, J., Holtrup, F., Queipo-
Ortuño, M. I., and Alba, E. (2019) Detection of TP53
and PIK3CA mutations in circulating tumor DNA us-
ing next-generation sequencing in the screening pro-
cess for early breast cancer diagnosis, J.Clin. Med., 8,
1183, https://doi.org/10.3390/jcm8081183.
38. Turner, N. C., Swift, C., Jenkins, B., Kilburn, L.,
Coakley, M., Beaney,M., Fox,L., Goddard, K., Garcia-
Murillas, I., Proszek, P., Hall, P., Harper-Wynne, C.,
Hickish, T., Kernaghan, S., Macpherson, I.R., Okines,
A. F. C., Palmieri, C., Perry, S., Randle, K., Snow-
don, C., and c-TRAK TN investigators (2023) Results
of the c-TRAK TN trial: a clinical trial utilising ctDNA
mutation tracking to detect molecular residual dis-
ease and trigger intervention in patients with mod-
erate- and high-risk early-stage triple-negative breast
cancer, Ann. Oncol., 34, 200-211, https://doi.org/
10.1016/j.annonc.2022.11.005.
39. Garcia-Murillas, I., Schiavon, G., Weigelt, B., Ng, C.,
Hrebien, S., Cutts, R. J., Cheang, M., Osin, P.,
Nerurkar,A., Kozarewa,I., Garrido, J.A., Dowsett,M.,
Reis- Filho, J. S., Smith, I. E., and Turner, N. C. (2015)
Mutation tracking in circulating tumor DNA predicts
relapse in early breast cancer, Sci. Translat. Med.,
7, 302ra133, https://doi.org/10.1126/scitranslmed.
aab0021.
40. Beaver, J.A., Jelovac,D., Balukrishna,S., Cochran,R.,
Croessmann, S., Zabransky, D. J., Wong, H. Y., Toro,
P.V., Cidado,J., Blair, B.G., Chu,D., Burns,T., Higgins,
M. J., Stearns, V., Jacobs, L., Habibi, M., Lange, J.,
Hurley, P. J., Lauring, J., VanDenBerg, D., and Park,
B. H. (2014) Detection of cancer DNA in plasma of
patients with early-stage breast cancer, Clin. Cancer
Res., 20, 2643-2650, https://doi.org/10.1158/1078-0432.
CCR-13-2933.
41. Garcia-Murillas, I., Chopra, N., Comino- Méndez, I.,
Beaney, M., Tovey, H., Cutts, R. J., Swift, C.,
Kriplani, D., Afentakis, M., Hrebien, S., Walsh-
Crestani, G., Barry, P., Johnston, S. R. D., Ring, A.,
Bliss,J., Russell,S., Evans,A., Skene,A., Wheatley,D.,
Dowsett, M., and Turner, N. C. (2019) Assessment
of molecular relapse detection in early-stage breast
cancer, JAMA Oncol., 5, 1473-1478, https://doi.org/
10.1001/jamaoncol.2019.1838.
42. Dawson, S. J., Tsui, D. W., Murtaza, M., Biggs, H.,
Rueda, O. M., Chin, S. F., Dunning, M. J., Gale, D.,
Forshew,T., Mahler-Araujo,B., Rajan,S., Humphray,S.,
Becq,J., Halsall, D., Wallis, M., Bentley, D., Caldas,C.,
and Rosenfeld, N. (2013) Analysis of circulating tu-
mor DNA to monitor metastatic breast cancer, New
Eng. J. Med., 368, 1199-1209, https://doi.org/10.1056/
NEJMoa1213261.
43. Murtaza, M., Dawson, S. J., Pogrebniak, K., Rueda,
O. M., Provenzano, E., Grant, J., Chin, S. F., Tsui,
D. W. Y., Marass, F., Gale, D., Ali, H. R., Shah, P.,
Contente- Cuomo, T., Farahani, H., Shumansky, K.,
Kingsbury, Z., Humphray, S., Bentley, D., Shah, S. P.,
Wallis,M., and Caldas,C. (2015) Multifocal clonal evo-
lution characterized using circulating tumour DNA
in a case of metastatic breast cancer, Nat. Commun.,
6, 8760, https://doi.org/10.1038/ncomms9760.
44. Magbanua, M. J. M., Swigart, L. B., Wu, H. T.,
Hirst, G. L., Yau, C., Wolf, D. M., Tin, A., Salari, R.,
Shchegrova,S., Pawar,H., Delson, A.L., DeMichele,A.,
Liu, M.C., Chien, A.J., Tripathy,D., Asare,S., Lin, C.J.,
Billings,P., Aleshin,A., Sethi,H., and van‘tVeer, L.J.
(2021) Circulating tumor DNA in neoadjuvant-treat-
ed breast cancer reflects response and survival,
Ann. Oncol., 32, 229-239, https://doi.org/10.1016/
j.annonc.2020.11.007.
45. Yi, Z., Ma, F., Rong, G., Guan, Y., Li, C., and Xu, B.
(2020) Clinical spectrum and prognostic value of TP53
mutations in circulating tumor DNA from breast can-
cer patients in China, Cancer Commun., 40, 260-269,
https://doi.org/10.1002/cac2.12032.
46. Li, X., Lu, J., Zhang, L., Luo, Y., Zhao, Z., and Li, M.
(2020) Clinical implications of monitoring ESR1 mu-
tations by circulating tumor DNA in estrogen recep-
tor positive metastatic breast cancer: a pilot study,
Translat. Oncol., 13, 321-328, https://doi.org/10.1016/
j.tranon.2019.11.007.
47. Cristofanilli, M., Rugo, H. S., Im, S. A., Slamon, D. J.,
Harbeck,N., Bondarenko,I., Masuda,N., Colleoni,M.,
DeMichele,A., Loi,S., Iwata,H., O’Leary,B., André,F.,
Loibl, S., Bananis, E., Liu, Y., Huang, X., Kim, S.,
Lechuga Frean, M.J., and Turner, N.C. (2022) Overall
survival with palbociclib and fulvestrant in women
with HR+/HER2– ABC: updated exploratory analy-
ses of PALOMA-3, a double-blind, phase III random-
ized study, Clin. Cancer Res., 28, 3433-3442, https://
doi.org/10.1158/1078-0432.CCR-22-0305.
48. Berger, F., Marce, M., Delaloge, S., Hardy-Bessard,
A. C., Bachelot, T., Bièche, I., Pradines, A., De La
Motte Rouge, T., Canon, J. L., André, F., Arnould, L.,
CIRCULATING TUMOR DNA IN BC PATIENTS 1499
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
Clatot,F., Lemonnier,J., Marques,S., Bidard, F.C., and
PADA-1 investigators (2022) Randomised, open-label,
multicentric phase III trial to evaluate the safety
and efficacy of palbociclib in combination with en-
docrine therapy, guided by ESR1 mutation moni-
toring in oestrogen receptor-positive, HER2-nega-
tive metastatic breast cancer patients: study design
of PADA-1, BMJ Open, 12, e055821, https://doi.org/
10.1136/bmjopen-2021-055821.
49. Tolaney, S. M., Toi, M., Neven, P., Sohn, J., Grischke,
E. M., Llombart-Cussac, A., Soliman, H., Wang, H.,
Wijayawardana, S., Jansen, V. M., Litchfield, L. M.,
and Sledge, G. W. (2022) Clinical significance of
PIK3CA and ESR1 mutations in circulating tumor
DNA: analysis from the MONARCH 2 study of abemac-
iclib plus fulvestrant, Clin. Cancer Res., 28, 1500-1506,
https://doi.org/10.1158/1078-0432.CCR-21-3276.
50. Di Leo, A., Johnston, S., Lee, K. S., Ciruelos, E.,
Lønning, P.E., Janni,W., O’Regan,R., Mouret- Reynier,
M. A., Kalev, D., Egle, D., Csőszi, T., Bordonaro, R.,
Decker,T., Tjan-Heijnen, V.C.G., Blau,S., Schirone,A.,
Weber,D., El-Hashimy,M., Dharan,B., Sellami,D., and
Bachelot,T. (2018) Buparlisib plus fulvestrant in post-
menopausal women with hormone-receptor-positive,
HER2-negative, advanced breast cancer progressing
on or after mTOR inhibition (BELLE-3): a randomised,
double-blind, placebo-controlled, phase 3 trial,
Lancet Oncol., 19, 87-100, https://doi.org/10.1016/
S1470-2045(17)30688-5.
51. Sabatier, R., Vicier, C., Garnier, S., Guille, A., Carbuc-
cia, N., Isambert, N., Dalenc, F., Robert, M., Levy, C.,
Pakradouni,J., Adelaïde,J., Chaffanet,M., Sfumato,P.,
Mamessier, E., Bertucci, F., and Goncalves, A. (2022)
Circulating tumor DNA predicts efficacy of a dual
AKT/p70S6K inhibitor (LY2780301) plus paclitaxel
in metastatic breast cancer: plasma analysis of the
TAKTIC phase IB/II study, Mol. Oncol., 16, 2057-2070,
https://doi.org/10.1002/1878-0261.13188.
52. Lyu,D., Liu,B., Lan,B., Sun,X., Li,L., Zhai,J., Qian,H.,
and Ma, F. (2022) Clinical value of next-generation
sequencing in guiding decisions regarding endocrine
therapy for advanced HR-positive/HER-2-negative
breast cancer, Chinese J. Cancer Res., 34, 343-352,
https://doi.org/10.21147/j.issn.1000-9604.2022.04.03.
53. Yi,Z., Ma,F., Rong,G., Liu,B., Guan,Y., Li,J., Sun,X.,
Wang, W., Guan, X., Mo, H., Wang, J., Qian, H., and
Xu, B. (2021) The molecular tumor burden index
as a response evaluation criterion in breast can-
cer, Signal Transduct. Target. Ther., 6, 251, https://
doi.org/10.1038/s41392-021-00662-9.
54. Pantel, K., and Alix-Panabières, C. (2010) Circulat-
ing tumour cells in cancer patients: challenges and
perspectives, Trends Mol. Med., 16, 398-406, https://
doi.org/10.1016/j.molmed.2010.07.001.
55. Nikanjam,M., Kato,S., and Kurzrock,R. (2022) Liquid
biopsy: current technology and clinical applications,
J. Hematol. Oncol., 15, 131, https://doi.org/10.1186/
s13045-022-01351-y.
56. Sama, S., Le, T., Ullah, A., Elhelf, I. A., Kavuri, S. K.,
and Karim, N. A. (2022) The role of serial liquid bi-
opsy in the management of metastatic non-small cell
lung cancer (NSCLC), Clinics Practice, 12, 419-424,
https://doi.org/10.3390/clinpract12030046.
57. El Messaoudi, S., Rolet, F., Mouliere, F., and Thierry,
A. R. (2013) Circulating cell free DNA: preanalytical
considerations, Clin. Chim. Acta, 424, 222-230, https://
doi.org/10.1016/j.cca.2013.05.022.
58. Lin, C., Liu, X., Zheng, B., Ke, R., and Tzeng, C. M.
(2021) Liquid biopsy, ctDNA diagnosis through NGS,
Life, 11, 890, https://doi.org/10.3390/life11090890.
59. Pons-Belda, O.D., Fernandez-Uriarte,A., and Diaman-
dis, E.P. (2021) Can circulating tumor DNA support a
successful screening test for early cancer detection?
The grail paradigm, Diagnostics, 11, 2171, https://
doi.org/10.3390/diagnostics11122171.
60. Lu, L., Bi, J., and Bao, L. (2018) Genetic profiling of
cancer with circulating tumor DNA analysis, J. Gen-
et. Genomics, 45, 79-85, https://doi.org/10.1016/j.jgg.
2017.11.006.
61. Kastrisiou, M., Zarkavelis, G., Pentheroudakis, G.,
and Magklara, A. (2019) Clinical application of
next-generation sequencing as a liquid biopsy tech-
nique in advanced colorectal cancer: a trick or
a treat? Cancers, 11, 1573, https://doi.org/10.3390/
cancers11101573.
62. Yi, K., Wang, X., Filippov, S. K., and Zhang, H. (2023)
Emerging ctDNA detection strategies in clinical can-
cer theranostics, Smart Med., 2, e20230031, https://
doi.org/10.1002/SMMD.20230031.
63. Ou, C. Y., Vu, T., Grunwald, J. T., Toledano, M.,
Zimak, J., Toosky, M., Shen, B., Zell, J. A., Gratton,E.,
Abram, T. J., and Zhao, W. (2019) An ultrasensi-
tive test for profiling circulating tumor DNA using
integrated comprehensive droplet digital detec-
tion, Lab Chip, 19, 993-1005, https://doi.org/10.1039/
c8lc01399c.
64. Rothé, F., Silva, M. J., Venet, D., Campbell, C., Brad-
burry, I., Rouas, G., de Azambuja, E., Maetens, M.,
Fumagalli, D., Rodrik-Outmezguine,V., Di Cosimo, S.,
Rosa, D., Chia, S., Wardley, A., Ueno, T., Janni, W.,
Huober, J., Baselga, J., Piccart, M., Loi, S., and Ignati-
adis, M. (2019) Circulating tumor DNA in HER2-am-
plified breast cancer: a translational research sub-
study of the NeoALTTO phase III trial, Clin. Cancer
Res., 25, 3581-3588, https://doi.org/10.1158/1078-0432.
CCR-18-2521.
65. Shaw, J. A., Page, K., Wren, E., de Bruin, E. C.,
Kalashnikova,E., Hastings,R., McEwen,R., Zhang,E.,
Wadsley, M., Acheampong, E., Renner, D., Gleason,
K. L. T., Ambasager, B., Stetson, D., Fernandez-
Garcia, D., Guttery, D., Allsopp, R. C., Rodriguez, A.,
Zimmermann, B., Sethi, H., and Coombes, R. C.
ZAVARYKINA et al.1500
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
(2024) Serial postoperative circulating tumor DNA
assessment has strong prognostic value during long-
term follow-up in patients with breast cancer, JCO
Prec. Oncol., 8, e2300456, https://doi.org/10.1200/
PO.23.00456.
66. Reinert, T., Henriksen, T. V., Christensen, E., Shar-
ma, S., Salari, R., Sethi, H., Knudsen, M., Norden-
toft,I., Wu, H. T., Tin, A. S., Heilskov Rasmussen,M.,
Vang,S., Shchegrova,S., Frydendahl Boll Johansen,A.,
Srinivasan,R., Assaf,Z., Balcioglu,M., Olson,A., Dash-
ner, S., Hafez, D., and Lindbjerg Andersen, C. (2019)
Analysis of plasma cell-free DNA by ultradeep se-
quencing in patients with stagesI toIII colorectal can-
cer, JAMA Oncol., 5, 1124-1131, https://doi.org/10.1001/
jamaoncol.2019.0528.
67. Abbosh,C., Birkbak, N.J., Wilson, G.A., Jamal-Hanja-
ni,M., Constantin,T., Salari,R., Le Quesne,J., Moore,
D. A., Veeriah, S., Rosenthal, R., Marafioti, T., Kirki-
zlar, E., Watkins, T. B. K., McGranahan, N., Ward, S.,
Martinson, L., Riley, J., Fraioli,F., Al Bakir, M., Grön-
roos, E., and Swanton, C. (2017) Phylogenetic ctDNA
analysis depicts early-stage lung cancer evolution,
Nature, 545, 446-451, https://doi.org/10.1038/na-
ture22364.
68. Diao, Z., Han, D., Zhang, R., and Li, J. (2021) Metag-
enomics next-generation sequencing tests take the
stage in the diagnosis of lower respiratory tract
infections, J. Adv. Res., 38, 201-212, https://doi.org/
10.1016/j.jare.2021.09.012.
69. Bai,Y., Wang,Z., Liu,Z., Liang,G., Gu,W., and Ge,Q.
(2020) Technical progress in circulating tumor DNA
analysis using next generation sequencing, Mol.
Cell. Probes, 49, 101480, https://doi.org/10.1016/j.mcp.
2019.101480.
70. Santonja, A., Cooper, W.N., Eldridge, M.D., Edwards,
P. A. W., Morris, J. A., Edwards, A. R., Zhao, H.,
Heider, K., Couturier, D. L., Vijayaraghavan, A.,
Mennea, P., Ditter, E. J., Smith, C. G., Boursnell, C.,
Manzano García, R., Rueda, O. M., Beddowes, E.,
Biggs, H., Sammut, S. J., Rosenfeld, N., and Gale, D.
(2023) Comparison of tumor-informed and tu-
mor-naïve sequencing assays for ctDNA detection in
breast cancer, EMBO Mol. Med., 15, e16505, https://
doi.org/10.15252/emmm.202216505.
71. Willis,J., Lefterova, M.I., Artyomenko,A., Kasi, P.M.,
Nakamura,Y., Mody,K., Catenacci, D.V.T., Fakih,M.,
Barbacioru, C., Zhao, J., Sikora, M., Fairclough, S. R.,
Lee, H., Kim, K. M., Kim, S. T., Kim, J., Gavino, D.,
Benavides, M., Peled, N., Nguyen, T., and Odegaard,
J. I. (2019) Validation of microsatellite instability de-
tection using a comprehensive plasma-based geno-
typing panel, CLIN, Cancer Res., 25, 7035-7045, https://
doi.org/10.1158/1078-0432.CCR-19-1324.
72. Janni, W., Rack, B., Friedl, T. W. P., Hartkopf, A. D.,
Wiesmüller, L., Pfister, K., Mergel, F., Fink, A.,
Braun, T., Mehmeti, F., Uhl, N., De Gregorio, A.,
Huober,J., Fehm,T., Müller,V., Rich, T.A., Dustin,D.J.,
Zhang, S., and Huesmann, S. T. (2025) Detection of
minimal residual disease and prediction of recur-
rence in breast cancer using a plasma-only circu-
lating tumor DNA assay, ESMO Open, 10, 104296,
https://doi.org/10.1016/j.esmoop.2025.104296.
73. Parsons, H. A., Blewett, T., Chu, X., Sridhar, S.,
Santos, K., Xiong, K., Abramson, V. G., Patel, A.,
Cheng, J., Brufsky, A., Rhoades, J., Force, J., Liu, R.,
Traina, T.A., Carey, L.A., Rimawi, M.F., Miller, K.D.,
Stearns, V., Specht, J., Falkson, C., and Adalsteinsson,
V. A. (2023) Circulating tumor DNA association with
residual cancer burden after neoadjuvant chemo-
therapy in triple-negative breast cancer in TBCRC
030, Ann. Oncol., 34, 899-906, https://doi.org/10.1016/
j.annonc.2023.08.004.
74. Tie,J., Wang,Y., Cohen,J., Li,L., Hong,W., Christie,M.,
Wong, H. L., Kosmider, S., Wong, R., Thomson, B.,
Choi, J., Fox, A., Field, K., Burge, M., Shannon, J.,
Kotasek,D., Tebbutt, N.C., Karapetis,C., Underhill,C.,
Haydon, A., and Gibbs, P. (2021) Circulating tumor
DNA dynamics and recurrence risk in patients un-
dergoing curative intent resection of colorectal can-
cer liver metastases: a prospective cohort study, PLoS
Med., 18, e1003620, https://doi.org/10.1371/journal.
pmed.1003620.
75. Parsons, H. A., Rhoades, J., Reed, S. C., Gydush, G.,
Ram, P., Exman, P., Xiong, K., Lo, C. C., Li, T.,
Fleharty, M., Kirkner, G. J., Rotem, D., Cohen, O.,
Yu,F., Fitarelli-Kiehl, M., Leong, K.W., Hughes, M.E.,
Rosenberg, S. M., Collins, L. C., Miller, K. D., and
Adalsteinsson, V. A. (2020) Sensitive detection of
minimal residual disease in patients treated for ear-
ly-stage Breast cancer, Clin. Cancer Res., 26, 2556-2564,
https://doi.org/10.1158/1078-0432.CCR-19-3005.
76. Radovich,M., Jiang,G., Hancock, B.A., Chitambar,C.,
Nanda, R., Falkson, C., Lynce, F. C., Gallagher, C.,
Isaacs, C., Blaya, M., Paplomata, E., Walling, R.,
Daily, K., Mahtani, R., Thompson, M. A., Graham, R.,
Cooper, M.E., Pavlick, D.C., Albacker, L. A., Gregg,J.,
and Schneider, B. P. (2020) Association of circulating
tumor DNA and circulating tumor cells after neoad-
juvant chemotherapy with disease recurrence in pa-
tients with triple-negative breast cancer: preplanned
secondary analysis of the BRE12-158 randomized
clinical trial, JAMA Oncol., 6, 1410-1415, https://
doi.org/10.1001/jamaoncol.2020.2295.
77. Zaikova,E., Cheng, B.Y.C., Cerda,V., Kong,E., Lai,D.,
Lum,A., Bates,C., den Brok,W., Kono,T., Bourque,S.,
Chan,A., Feng,X., Fenton,D., Gurjal,A., Levasseur,N.,
Lohrisch, C., Roberts, S., Shenkier, T., Simmons, C.,
Taylor, S., and Gelmon, K. (2024) Circulating tu-
mour mutation detection in triple-negative breast
cancer as an adjunct to tissue response assessment,
NPJ Breast Cancer, 10, 3, https://doi.org/10.1038/
s41523-023-00607-1.
CIRCULATING TUMOR DNA IN BC PATIENTS 1501
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
78. Chan, H. T., Chin, Y. M., Nakamura, Y., and Low,
S. K. (2020) Clonal hematopoiesis in liquid biop-
sy: from biological noise to valuable clinical impli-
cations, Cancers, 12, 2277, https://doi.org/10.3390/
cancers12082277.
79. Tie, J., Cohen, J. D., Wang, Y., Li, L., Christie, M.,
Simons,K., Elsaleh,H., Kosmider,S., Wong,R., Yip,D.,
Lee,M., Tran,B., Rangiah,D., Burge,M., Goldstein,D.,
Singh, M., Skinner, I., Faragher, I., Croxford, M.,
Bampton,C., and Gibbs,P. (2019) Serial circulating tu-
mour DNA analysis during multimodality treatment
of locally advanced rectal cancer: a prospective bio-
marker study, Gut, 68, 663-671, https://doi.org/10.1136/
gutjnl-2017-315852.
80. Cutts, R., Ulrich, L., Beaney, M., Robert, M.,
Coakley, M., Bunce, C., Crestani, G. W., Hrebien, S.,
Kalashnikova, E., Wu, H. T., Dashner, S., Sethi, H.,
Aleshin, A., Liu, M., Ring, A., Okines, A., Smith,
I. E., Barry, P., Turner, N. C., and Garcia-Murillas, I.
(2024) Association of post-operative ctDNA detection
with outcomes of patients with early breast can-
cers, ESMO Open, 9, 103687, https://doi.org/10.1016/
j.esmoop.2024.103687.
81. Chen, J. H., Addanki, S., Roy, D., Bassett, R., Kalash-
nikova, E., Spickard, E., Kuerer, H. M., Meas, S.,
Sarli, V. N., Korkut, A., White, J. B., Rauch, G. M.,
Tripathy, D., Arun, B. K., Barcenas, C. H., Yam, C.,
Sethi, H., Rodriguez, A. A., Liu, M. C., Moulder, S. L.,
and Lucci,A. (2024) Monitoring response to neoadju-
vant chemotherapy in triple negative breast cancer
using circulating tumor DNA, BMC Cancer, 24, 1016,
https://doi.org/10.1186/s12885-024-12689-6.
82. Coakley, M., Villacampa, G., Sritharan, P., Swift, C.,
Dunne, K., Kilburn, L., Goddard, K., Pipinikas, C.,
Rojas, P., Emmett, W., Hall, P., Harper-Wynne, C.,
Hickish, T., Macpherson, I., Okines, A., Wardley, A.,
Wheatley, D., Waters, S., Palmieri, C., Winter, M.,
and Turner, N. C. (2024) Comparison of circulating
tumor DNA assays for molecular residual disease
detection in early-stage triple-negative breast cancer,
Clin. Cancer Res., 30, 895-903, https://doi.org/10.1158/
1078-0432.CCR-23-2326.
83. Coombes, R. C., Page, K., Salari, R., Hastings, R. K.,
Armstrong,A., Ahmed,S., Ali,S., Cleator,S., Kenny,L.,
Stebbing, J., Rutherford, M., Sethi, H., Boydell, A.,
Swenerton,R., Fernandez-Garcia,D., Gleason, K.L.T.,
Goddard, K., Guttery, D. S., Assaf, Z. J., Wu, H. T.,
and Shaw, J. A. (2019) Personalized detection of cir-
culating tumor DNA antedates breast cancer meta-
static recurrence, Clin. Cancer Res., 25, 4255-4263,
https://doi.org/10.1158/1078-0432.CCR-18-3663.
84. Comino-Méndez,I., Velasco-Suelto,J., Pascual,J., López-
López, E., Quirós-Ortega, M. E., Gaona-Romero, C.,
Martín-Muñoz, A., Losana, P., Heredia, Y., Alba, E.,
and Guerrero-Zotano,A. (2025) Identification of min-
imal residual disease using the clonesight test for
ultrasensitive ctDNA detection to anticipate late re-
lapse in early breast cancer, Breast Cancer Res., 27,
65, https://doi.org/10.1186/s13058-025-02016-7.
85. Wang, K., Peng, Z., Lin, X., Nian, W., Zheng, X., and
Wu,J. (2022) Electrochemical biosensors for circulat-
ing tumor DNA detection, Biosensors, 12, 649, https://
doi.org/10.3390/bios12080649.
86. Uygun, Z. O., Yeniay, L., and Sağın, F. G. (2020)
CRISPR- dCas9 powered impedimetric biosensor for
label-free detection of circulating tumor DNAs, Anal.
Chim. Acta, 1121, 35-41, https://doi.org/10.1016/j.aca.
2020.04.009.
87. Zhang, W., Dai, Z., Liu, X., and Yang, J. (2018)
High-performance electrochemical sensing of circu-
lating tumor DNA in peripheral blood based on po-
ly-xanthurenic acid functionalized MoS2 nanosheets,
Biosensors Bioelectronics, 105, 116-120, https://
doi.org/10.1016/j.bios.2018.01.038.
88. Povedano, E., Vargas, E., Montiel, V. R., Torrente-
Rodríguez, R.M., Pedrero,M., Barderas,R., Segundo-
Acosta, P. S., Peláez-García, A., Mendiola, M., Hardis-
son, D., Campuzano, S., and Pingarrón, J. M. (2018)
Electrochemical affinity biosensors for fast detection
of gene-specific methylations with no need for bisul-
fite and amplification treatments, Sci. Rep., 8, 6418,
https://doi.org/10.1038/s41598-018-24902-1.
89. Wang,L., Zhuang,Y., Yu,Y., Guo,Z., Guo,Q., Qiao,L.,
Wang, X., Liang, X., Zhang, P., Li, Q., Huang, C.,
Cong, R., Li, Y., Che, B., Xiong, H., Lin, G., Rao, M.,
Hu, R., Wang, W., Yang, G., and Lou, J. (2023) An
ultrasensitive method for detecting mutations from
short and rare cell-free DNA, Biosensors Bioelec-
tronics, 238, 115548, https://doi.org/10.1016/j.bios.
2023.115548.
90. Li, D., Chen, H., Fan, K., Labunov, V., Lazarouk, S.,
Yue,X., Liu,C., Yang,X., Dong,L., and Wang,G. (2021)
A supersensitive silicon nanowire array biosensor
for quantitating tumor marker ctDNA, Biosensors
Bioelectronics, 181, 113147, https://doi.org/10.1016/
j.bios.2021.113147.
91. Nguyen, A. H., and Sim, S. J. (2015) Nanoplasmon-
ic biosensor: detection and amplification of dual
bio-signatures of circulating tumor DNA, Biosensors
Bioelectronics, 67, 443-449, https://doi.org/10.1016/
j.bios.2014.09.003.
92. Miao, P., Chai, H., and Tang, Y. (2022) DNA hairpins
and dumbbell-wheel transitions amplified walk-
ing nanomachine for ultrasensitive nucleic acid
detection, ACS Nano, 16, 4726-4733, https://doi.org/
10.1021/acsnano.1c11582.
93. Liu,F., Peng,J., Lei, Y.M., Liu, R.S., Jin,L., Liang,H.,
Liu, H.F., Ma, S.Y., Zhang, X.H., Zhang, Y.P., Li, C.P.,
and Zhao, H. (2022) Electrochemical detection of
ctDNA mutation in non-small cell lung cancer based
on CRISPR/Cas12a system, Sensors Actuators B Chem.,
362, 131807, https://doi.org/10.1016/j.snb.2022.131807.
ZAVARYKINA et al.1502
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
94. Povedano, E., Montiel, V. R., Valverde, A., Navarro-
Villoslada,F., Yáñez-Sedeño,P., Pedrero,M., Montero-
Calle,A., Barderas,R., Peláez-García,A., Mendiola,M.,
Hardisson,D., Feliú,J., Camps,J., Rodríguez-Tomàs,E.,
Joven, J., Arenas, M., Campuzano, S., and Pingarrón,
J. M. (2019) Versatile electroanalytical bioplatforms
for simultaneous determination of cancer-related
DNA 5-methyl- and 5-hydroxymethyl-cytosines at glob-
al and gene-specific levels in human serum and tis-
sues, ACS Sensors, 4, 227-234, https://doi.org/10.1021/
acssensors.8b01339.
95. Wang, M., Hou, L., Chen, M., Zhou, Y., Liang, Y.,
Wang, S., Jiang, J., and Zhang, Y. (2017) Neoadjuvant
chemotherapy creates surgery opportunities for in-
operable advanced breast cancer, Sci. Rep., 7, 44673,
https://doi.org/10.1038/srep44673.
96. Derks, M. G. M., and van de Velde, C. J. H. (2018)
Neoadjuvant chemotherapy in breast cancer: more
than just downsizing, Lancet Oncol., 19, 2-3, https://
doi.org/10.1016/S1470-2045(17)30914-2.
97. Early Breast Cancer Trialists’ Collaborative Group
(EBCTCG) (2018) Long-term outcomes for neoadju-
vant versus adjuvant chemotherapy in early breast
cancer: meta-analysis of individual patient data
from ten randomised trials, Lancet Oncol., 19, 27-39,
https://doi.org/10.1016/S1470-2045(17)30777-5.
98. Von Minckwitz, G., Untch, M., Blohmer, J. U., Costa,
S. D., Eidtmann, H., Fasching, P. A., Gerber, B.,
Eiermann, W., Hilfrich, J., Huober, J., Jackisch, C.,
Kaufmann,M., Konecny, G. E., Denkert,C., Nekljudo-
va, V., Mehta, K., and Loibl, S. (2012) Definition and
impact of pathologic complete response on prognosis
after neoadjuvant chemotherapy in various intrinsic
breast cancer subtypes, J.Clin. Oncol., 30, 1796-1804,
https://doi.org/10.1200/JCO.2011.38.8595.
99. Symmans, W. F., Wei, C., Gould, R., Yu, X., Zhang, Y.,
Liu, M., Walls, A., Bousamra, A., Ramineni, M.,
Sinn, B., Hunt,K., Buchholz, T. A., Valero, V., Buzdar,
A.U., Yang,W., Brewster, A.M., Moulder,S., Pusztai,L.,
Hatzis, C., and Hortobagyi, G. N. (2017) Long-term
prognostic risk after neoadjuvant chemotherapy
associated with residual cancer burden and breast
cancer subtype, J.Clin. Oncol., 35, 1049-1060, https://
doi.org/10.1200/JCO.2015.63.1010.
100. Symmans, W. F., Peintinger, F., Hatzis, C., Rajan, R.,
Kuerer, H., Valero, V., Assad, L., Poniecka, A.,
Hennessy,B., Green,M., Buzdar, A.U., Singletary, S.E.,
Hortobagyi, G.N., and Pusztai,L. (2007) Measurement
of residual breast cancer burden to predict survival
after neoadjuvant chemotherapy, J.Clin. Oncol., 25,
4414-4422, https://doi.org/10.1200/JCO.2007.10.6823.
101. Cortazar, P., Zhang, L., Untch, M., Mehta, K., Costan-
tino, J. P., Wolmark, N., Bonnefoi, H., Cameron, D.,
Gianni, L., Valagussa, P., Swain, S. M., Prowell, T.,
Loibl, S., Wickerham, D. L., Bogaerts, J., Baselga, J.,
Perou, C., Blumenthal, G., Blohmer, J., Mamounas,
E. P., and von Minckwitz, G. (2014) Pathological
complete response and long-term clinical bene-
fit in breast cancer: the CTNeoBC pooled analysis,
Lancet, 384, 164-172, https://doi.org/10.1016/S0140-
6736(13)62422-8.
102. I-SPY2 Trial Consortium, Yee, D., DeMichele, A. M.,
Yau, C., Isaacs, C., Symmans, W. F., Albain, K. S.,
Chen, Y.Y., Krings,G., Wei,S., Harada,S., Datnow,B.,
Fadare, O., Klein, M., Pambuccian, S., Chen, B.,
Adamson, K., Sams, S., Mhawech-Fauceglia, P.,
Magliocco, A., and Berry, D. A. (2020) Association of
event-free and distant recurrence-free survival with
individual-level pathologic complete response in neo-
adjuvant treatment of stages 2 and 3 breast cancer:
three-year follow-up analysis for the I-SPY2 adaptive-
ly randomized clinical trial, JAMA Oncol., 6, 1355-
1362, https://doi.org/10.1001/jamaoncol.2020.2535.
103. Ignatova, E. O., Frolova, M. A., Petrovsky, A. B.,
Stenina, M. B., Glazkova, E. V., Krokhina, O. V., and
Tjulandin, C.A. (2016) Evaluation of efficacy and tox-
icity of neoadjuvant chemotherapy with dose-dense
doxorubicin, cisplatin, and paclitaxel in patients
with early triple-negative breast cancer, Malignant
Tumours, 4, 49-57, https://doi.org/10.18027/2224-
5057-2016-4-49-57.
104. DeVita, V. T., Lawrence, T. S., and Rosenberg, S. A.
(2023) Malignant tumors of the breast, in DeVita,
Hellman, and Rosenberg’s Cancer: Principles & Prac-
tice of Oncology, 12thEdn, Wolters Kluwer, The Neth-
erlands.
105. McDonald, B. R., Contente-Cuomo, T., Sammut, S. J.,
Odenheimer-Bergman, A., Ernst, B., Perdigones, N.,
Chin, S. F., Farooq, M., Mejia, R., Cronin, P. A.,
Anderson, K. S., Kosiorek, H. E., Northfelt, D. W.,
McCullough, A. E., Patel, B. K., Weitzel, J. N., Slavin,
T. P., Caldas, C., Pockaj, B.A., and Murtaza,M. (2019)
Personalized circulating tumor DNA analysis to de-
tect residual disease after neoadjuvant therapy in
breast cancer, Sci. Translat. Med., 11, eaax7392,
https://doi.org/10.1126/scitranslmed.aax7392.
106. Cailleux, F., Agostinetto,E., Lambertini, M., Rothé, F.,
Wu, H.T., Balcioglu,M., Kalashnikova,E., Vincent,D.,
Viglietti, G., Gombos, A., Papagiannis, A., Veys, I.,
Awada, A., Sethi, H., Aleshin, A., Larsimont, D.,
Sotiriou, C., Venet, D., and Ignatiadis, M. (2022) Cir-
culating tumor DNA after neoadjuvant chemother-
apy in breast cancer is associated with disease re-
lapse, JCO Precis. Oncol., 6, e2200148, https://doi.org/
10.1200/PO.22.00148.
107. Moss,J., Zick,A., Grinshpun,A., Carmon,E., Maoz,M.,
Ochana, B.L., Abraham,O., Arieli,O., Germansky,L.,
Meir, K., Glaser, B., Shemer,R., Uziely, B., and Dor, Y.
(2020) Circulating breast-derived DNA allows uni-
versal detection and monitoring of localized breast
cancer, Ann. Oncol., 31, 395-403, https://doi.org/
10.1016/j.annonc.2019.11.014.
CIRCULATING TUMOR DNA IN BC PATIENTS 1503
BIOCHEMISTRY (Moscow) Vol. 90 No. 11 2025
108. Magbanua, M. J. M., Brown Swigart, L., Ahmed, Z.,
Sayaman, R. W., Renner, D., Kalashnikova, E., Hirst,
G.L., Yau,C., Wolf, D.M., Li,W., Delson, A.L., Asare,S.,
Liu, M. C., Albain, K., Chien, A. J., Forero-Torres, A.,
Isaacs, C., Nanda, R., Tripathy, D., Rodriguez, A., and
van‘tVeer, L.J. (2023) Clinical significance and biol-
ogy of circulating tumor DNA in high-risk early-stage
HER2-negative breast cancer receiving neoadjuvant
chemotherapy, Cancer Cell, 41, 1091-1102.e4, https://
doi.org/10.1016/j.ccell.2023.04.008.
109. Magbanua, M.J.M., Ahmed,Z., Sayaman, R.W., Brown
Swigart, L., Hirst, G. L., Yau, C., Wolf, D. M., Li, W.,
Delson, A. L., Perlmutter, J., Pohlmann, P., Symmans,
W.F., Yee,D., Hylton, N.M., Esserman, L.J., DeMichele,
A. M., Rugo, H. S., and van ‘t Veer, L. J. (2024) Cell-
free DNA concentration as a biomarker of response
and recurrence in HER2-negative breast cancer re-
ceiving neoadjuvant chemotherapy, Clin. Cancer
Res., 30, 2444-2451, https://doi.org/10.1158/1078-0432.
CCR-23-2928.
110. Lin, P. H., Wang, M. Y., Lo, C., Tsai, L. W., Yen, T. C.,
Huang, T. Y., Huang, W. C., Yang, K., Chen, C.K., Fan,
S. C., Kuo, S. H., and Huang, C. S. (2021) Circulating
tumor DNA as a predictive marker of recurrence
for patients with stage II-III breast cancer treated
with neoadjuvant therapy, Front. Oncol., 11, 736769,
https://doi.org/10.3389/fonc.2021.736769.
111. Ciriaco, N., Zamora, E., Escrivá-de-Romaní, S.,
Miranda Gómez, I., Jiménez Flores, J., Saura, C.,
Sloane, H., Starus, A., Fredebohm, J., Georgieva, L.,
Speight, G., Jones, F., Ramón Y Cajal, S., Espino-
sa-Bravo, M., and Peg, V. (2022) Clearance of ctDNA
in triple-negative and HER2-positive breast can-
cer patients during neoadjuvant treatment is cor-
related with pathologic complete response, Ther.
Adv. Med. Oncol., 14, 17588359221139601, https://
doi.org/10.1177/17588359221139601.
112. Liu, B., Yi, Z., Guan, Y., Ouyang, Q., Li, C., Guan, X.,
Lv, D., Li, L., Zhai, J., Qian, H., Xu, B., Ma, F., and
Zeng, Y. (2022) Molecular landscape of TP53 muta-
tions in breast cancer and their utility for predicting
the response to HER-targeted therapy in HER2 ampli-
fication-positive and HER2 mutation-positive amplifi-
cation-negative patients, Cancer Med., 11, 2767-2778,
https://doi.org/10.1002/cam4.4652.
113. Garcia-Murillas, I., Cutts, R. J., Walsh-Crestani, G.,
Phillips,E., Hrebien,S., Dunne,K., Sidhu,K., Daber,R.,
Hubert, B., Graybill, C., DeFord, P. M., Wooten, D. J.,
Zhao, J., Ellsworth, R. E., Johnston, S. R. D., Ring, A.,
Russell, S., Evans, A., Skene, A., Wheatley, D., and
Turner, N.  C. (2025) Longitudinal monitoring of cir-
culating tumor DNA to detect relapse early and
predict outcome in early breast cancer, Breast Can-
cer Res. Treat., 209, 493-502, https://doi.org/10.1007/
s10549-024-07508-2.
114. Nguyen, S.T., Nguyen Hoang, V.A., Nguyen Trieu,V.,
Pham, T. H., Dinh, T. C., Pham, D. H., Nguyen, N.,
Vinh, D.N., Do, T. T.T., Nguyen, D.S., Nguyen, H.N.,
Giang, H., and Tu, L. N. (2025) Personalized muta-
tion tracking in circulating-tumor DNA predicts re-
currence in patients with high-risk early breast can-
cer, NPJ Breast Cancer, 11, 58, https://doi.org/10.1038/
s41523-025-00778-z.
115. Lee, T. H., Kim, H., Kim, Y. J., Park, W. Y., Park, W.,
Cho, W.K., and Kim,N. (2024) Implication of pre- and
post-radiotherapy ctDNA dynamics in patients with
residual triple-negative breast cancer at surgery af-
ter neoadjuvant chemotherapy: findings from a pro-
spective observational study, Cancer Res. Treat., 56,
531-537, https://doi.org/10.4143/crt.2023.996.
116. Wang, R., Wang, B., Zhang, H., Liao, X., Shi, B.,
Zhou,Y., Zhou,C., Yan,Y., Zhang,W., Wang,K., Ge,G.,
Ren, Y., Tang, X., Gan, B., He, J., and Niu, L. (2024)
Early evaluation of circulating tumor DNA as marker
of therapeutic efficacy and prognosis in breast cancer
patients during primary systemic therapy, Breast, 76,
103738, https://doi.org/10.1016/j.breast.2024.103738.
117. Zhang, D., Jahanfar, S., Rabinowitz, J. B., Dower, J.,
Song, F., Wu, C. H., Hu, X., Tracy, P., Basik, M.,
Medford, A., Lin, P. H., Huang, C. S., Bidard, F. C.,
Renault, S., Pai, L., Buss, M., Parsons, H. A., and
Schlam, I. (2025) Role of circulating tumor DNA in
early-stage triple-negative breast cancer: a systemat-
ic review and meta-analysis, Breast Cancer Res., 27,
38, https://doi.org/10.1186/s13058-025-01986-y.
118. Li,S., Li,Y., Wei,W., Gong,C., Wang,T., Li,G., Yao,F.,
Ou, J.H., Xu,Y., Wu,W., Jin,L., Rao,N., Nie,Y., Yu,F.,
Jia, W., Li, X. R., Zhang, J., Yang, H. W., Yang, Y.,
Wu, M., and Liu, Q. (2025) Dynamic ctDNA tracking
stratifies relapse risk for triple negative breast cancer
patients receiving neoadjuvant chemotherapy, Nat.
Commun., 16, 2786, https://doi.org/10.1038/s41467-
025-57988-z.
Publishers Note. Pleiades Publishing remains
neutral with regard to jurisdictional claims in published
maps and institutional affiliations. AI tools may have
been used in the translation or editing of this article.