Next Article in Journal
Immunotherapy for Management of Thymic Epithelial Tumors: A Double-Edged Sword
Next Article in Special Issue
Circulating Cell-Free DNA Assessment in Biofluids from Children with Neuroblastoma Demonstrates Feasibility and Potential for Minimally Invasive Molecular Diagnostics
Previous Article in Journal
Updated Functional Roles of NAMPT in Carcinogenesis and Therapeutic Niches
Previous Article in Special Issue
TLR4 and pSTAT3 Expression on Circulating Tumor Cells (CTCs) and Immune Cells in the Peripheral Blood of Breast Cancer Patients: Prognostic Implications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current and Developing Liquid Biopsy Techniques for Breast Cancer

1
Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan
2
Department of Medical Research, Chang Bing Show Chwan Memorial Hospital, Lukang Town, Changhua 505, Taiwan
3
Department of Biology, National Changhua University of Education, Changhua 500, Taiwan
4
Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan
5
Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan
6
School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
7
Department of Health Food, Chung Chou University of Science and Technology, Changhua 510, Taiwan
8
National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(9), 2052; https://doi.org/10.3390/cancers14092052
Submission received: 26 March 2022 / Revised: 13 April 2022 / Accepted: 15 April 2022 / Published: 19 April 2022
(This article belongs to the Special Issue Clinical Perspective and Translational Oncology of Liquid Biopsy)

Abstract

:

Simple Summary

Breast cancer is the most common cancer and leading cause of death worldwide. Therefore, it is important to diagnose and treat breast cancer early. Current diagnostic methods include mammography and tissue biopsy; however, they have limitations. Liquid biopsy is a less invasive tool for diagnosis. In this review, we summarize and focus on the recent discoveries on liquid biopsy and development of detection techniques.

Abstract

Breast cancer is the most commonly diagnosed cancer and leading cause of cancer mortality among woman worldwide. The techniques of diagnosis, prognosis, and therapy monitoring of breast cancer are critical. Current diagnostic techniques are mammography and tissue biopsy; however, they have limitations. With the development of novel techniques, such as personalized medicine and genetic profiling, liquid biopsy is emerging as the less invasive tool for diagnosing and monitoring breast cancer. Liquid biopsy is performed by sampling biofluids and extracting tumor components, such as circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), cell-free mRNA (cfRNA) and microRNA (miRNA), proteins, and extracellular vehicles (EVs). In this review, we summarize and focus on the recent discoveries of tumor components and biomarkers applied in liquid biopsy and novel development of detection techniques, such as surface-enhanced Raman spectroscopy (SERS) and microfluidic devices.

1. Introduction

Breast cancer is the most common female cancer in 2020, with an incidence of estimated 2.3 million, representing 11.7% of total cancer cases in the world, and the leading cause of cancer mortality in women, which was responsible for nearly 685,000 deaths worldwide [1]. In US, the American Cancer Society’s 2022 update estimated that approximately 287,850 new cases of breast cancer will be diagnosed in US women, with an estimated 43,250 deaths [2]. Based on immunohistochemistry classification, breast cancer is classified to five major molecular subtypes: luminal A (estrogen receptor (ER)+, progesterone receptor (PR)+, human epidermal growth factor receptor 2 (HER2)−, Ki-67 low), luminal B HER2− (ER+, PR+, HER2−, Ki-67 high), luminal B HER2+ (ER+, PR+, HER2+, Ki-67 high), HER2 (ER−, PR−, HER2+), and basal-like (triple-negative (TNBC), ER−, PR−, HER2−), which are related to the clinical outcomes [3,4].
Despite advances in diagnosis and treatments for breast cancer, the standard methods have several drawbacks, such as being invasive, expensive, not suitable for all patients, and low sensitivity and specificity [5]. Classical diagnostic/monitoring techniques include imaging (mammography, ultrasound, MRI, CT, PET, and X-ray) and tissue biopsy [6,7]. Mammography can lead to both false-positive and -negative results, unnecessary exposure to radiation, and the excessive use of biopsies, and it may fail to rapidly detect the changes in tumor burden [8]. Particularly, tissue biopsy is an invasive procedure that is neither extensive enough to capture the overall genomic landscape of breast tumors nor applicable for monitoring treatment response [9]. These limitations point to the urgent need for better and novel non-invasive methods for early detection, patient survival prediction, and treatment response monitoring. Recent advances in molecular testing and genomics have led the trend of personalized and precision medicine.
Liquid biopsy has attracted considerable attention and become an attractive alternative strategy, as it is a minimally invasive molecular procedure for advanced monitoring of cancer. It relies on the quantification of genetic materials derived from tumor cells and released into circulation, such as circulating tumor cells (CTCs), cell-free DNA (cfDNA)/circulating tumor DNA (ctDNA), circulating tumor RNA, extracellular vesicles (EVs), circulating tumor proteins, and tumor-educated platelets (TEPs) (Figure 1) by collecting body fluids, mostly peripheral blood [10,11]. In comparison with traditional tissue biopsy, liquid biopsy offers a number of notable advantages with easier and non-invasive sampling for serial evaluation [12]. A liquid biopsy, combined with highly sensitive molecular technologies and advance bioinformatics protocols, could reflect the intra-tumoral heterogeneity (spatial heterogeneity) and molecular evolution of a distant metastatic lesion (temporal heterogeneity), which is not possible for conventional tissue biopsies, as the biopsy specimen may not be representative of all the tumor cells [13,14,15,16]. Furthermore, it is possible for the early diagnosis and screening, prediction of prognosis, early relapse detection in localized and locally advance breast cancer, minimal residual disease (MRD) identification, and longitudinal monitoring of the disease progression and treatment response surveillance during adjuvant and neoadjuvant therapies upon sequential sampling, due to its minimally invasive nature [9,17,18] (Figure 1). Despite these, detection limits of liquid biopsy still exist. The low levels of CTCs and ctDNA found in early-stage breast cancer, along with the lack of ctDNA secreting from some tumors, can further complicate detection. Moreover, genetic patterns in primary tumors and metastases vary significantly from patient to patient [12,19]. More sensitive detection methods are urgently needed to improve the clinical application of liquid biopsy.
In this review, we discuss a variety of tumor components and biomarkers applied in liquid biopsy (both already applied in clinical practice and under research), as well as the current developed detection techniques for liquid biopsy in breast cancer, mainly focusing on recent studies.

2. Tumor Components

Liquid biopsy components, termed tumor circulome, including CTCs, cfRNA, ctDNA, TEPs, EVs, proteins, and metabolites, are secreted from tumor (apoptotic or necrotic) cells [20,21] (Figure 1). These tumor components present novel and minimally invasive biosources that are clinically implicated in precision medicine [22]. Notably, CTCs and ctDNA have been approved by the US Food and Drug Administration (FDA) as biomarkers in clinical use for cancer management [23].

2.1. Circulating Tumor Cells (CTCs)

CTCs are cancer cells compromising of a heterogeneous population with the majority of cells being highly differentiated, while others have stem cell-like properties (CSCs). They are released from primary and metastatic tumors into the circulation by trans-endothelial transition as single cells or clusters. These cells, which are able to adapt and survive by epithelial-to-mesenchymal transition (EMT) in the bloodstream and different tissues, can form new tumors or metastases [24,25,26,27]. Interacting with blood components, such as platelets, is critical for promoting tumor cells for subsequent metastasis [28], and interaction with immune cells results in evasion from immune surveillance and formation of metastases [29,30].
There are a considerable number of studies demonstrating CTC detection as an effective technique for the evaluating treatment efficacy, early diagnoses, metastatic progresses, recurrence, and prognosis [31,32,33], and it was correlated with unfavorable prognosis, shorter disease-free survival (DFS) and overall survival (OS), lack of treatment efficacy with poor recurrence-free survival (RFS), and tumor progression [34,35,36,37]. Several researches showed that CTC enumeration could be an independent prognostic tool for early breast cancer patients, particularly for metastatic breast cancer [38,39]. CTCs are substantially less abundant in the blood of patients with early stage of tumors [34,35,36,37]. Cristofallini et al. applied CTCs, detected by CellSearch system, to stratify patients into Stage IV aggressive with ≥5 CTCs/7.5 mL and Stage IV indolent with <5 CTCs/7.5 mL. In a pooled analysis of 2436 metastatic breast cancer patients, Stage IV indolent patients had significantly longer median OS (36.3 months) than Stage IV aggressive patients (16.0 months, p < 0.0001), independent of metastasis localization, tumor subtype, and molecular variables [40]. Therefore, this study further demonstrated CTC count is an important prognostic tool for metastatic breast cancer. More recently, 1933 HER2- metastatic breast cancer patients who participated in DETECT III and IV trials were screened, and it was confirmed that the CTC count has a high prognostic relevance [41]. Intriguingly, patients with ER- and PR+ tumors were more likely to harbor ≥1 CTC with strong HER2 staining, and it was significantly associated with shorter OS (median OS: 9.7 vs. 16.5 months in patients with CTCs with negative-to-moderate HER2 staining, p = 0.013). This study indicates that CTC detection, in patients with HER2- breast cancer, is a strong prognostic factor, and it remains the largest study conducted in HER2- metastatic breast cancer.
In addition to the prognostic value, in the STIC CTC randomized, multicenter prospective, noninferiority phase 3 trial, 755 hormone receptor (HR)+, HER2- metastatic breast cancer patients were allocated into either clinician-driven group, where the decision to administer hormone therapy or chemotherapy was made clinically without the CTC results, or a CTC-driven group, where endocrine therapy was administered if CTC <5/7.5 mL and chemotherapy administered if CTC ≥5/7.5 mL. Median progression-free survival (PFS) was significantly longer in the CTC-driven arm (15.5 months, 95% CI: 12.7–17.3), compared with the clinically-driven arm (13.9 months, 95% CI: 12.2–16.3) [33]. This key study demonstrated that CTC is promising to direct therapy. However, there is the need for more studies to validate this. Other studies also proved that CTCs can be applied in real-time monitoring treatment responses at different time points during the tumor progression and for the detection of relapses [42,43] (Figure 1). In another study of F.C. Bidard’s group, the CirCe01 trial evaluated the clinical utility of CTC-based monitoring of therapy [44]. In this prospective, multicentre, randomized phase III study (NCT01349842), patients with metastatic breast cancer, scheduled beyond the third line of chemotherapy, were randomized between the CTC-driven arm and standard arm. However, OS was not significantly different between two groups (p = 0.8). In subgroup analyses, patients with no CTC response who switched chemotherapy early nevertheless experienced longer median PFS and OS than those who did not.
Beside blood, as the most commonly studied and clinically used fluid in liquid biopsy, Malani et al. [45] recently applied the CTC count in cerebrospinal fluid (CSF) diagnose leptomeningeal metastases in HER2+ breast cancer patients. Their study also proved that CSF CTC enumeration could assess the tumor burden in the central nervous system during therapy for leptomeningeal metastasis and before detectable changes on MRI images or CSF cytology [45]. Importantly, these recent studies on CTC, as a liquid biopsy, confirmed its clinical value in prognosis and role in dynamic and real-time monitoring of treatment, although there is no current clinical application of CTC [39].

2.2. Cell-Free DNA (cfDNA) and Circulating Tumor DNA (ctDNA)

Like CTC, cfDNA and ctDNA play important roles in liquid biopsy. cfDNA refers to the double- or single-stranded fragmented DNA liberated into body fluids, such as blood, saliva, lymph, tear fluid, bile, urine, milk, sweat, mucous suspension, amniotic, cerebrospinal and pleural fluids, cervicovaginal secretion, and wound efflux, by both normal and tumor cells, whereas circulating tumor DNA (ctDNA) represents only a fraction of cfDNA derived from the tumor tissue [9,46,47]. Specific patterns of cfDNA can be analyzed ex vivo to characterize the targets of interest [48]. While cfDNA is present in healthy controls, its concentration is significantly lower in healthy subjects, compared to cancer patients, due to active nuclease degradation [49,50].
In addition to cfDNA gene sequence and mutation, cfDNA can be further analyzed for epigenetic alterations, such as DNA methylation, histone modifications, and expression of long and micro non-coding RNAs [51,52]. Methylation changes in DNA contribute to gene expression regulation and play a significant role in the etiology of early breast cancer [53,54]. The DNA methylation pattern is retained in the cfDNA released from its tissue origins of tumor cells [55,56]. Therefore, DNA methylation could serve as important biomarkers for diagnosis of cancer [57]. Indeed, DNA methylation has been assessed in cfDNA in several studies, both single and panels of genes have been demonstrated as diagnostic tools [58,59,60]. Furthermore, the methylation patterns of cfDNA could be also related to relapse, metastasis, and survival [5]. Panagopoulou et al. established a cfDNA methylation panel of five cancer-related genes (KLK10, SOX17, WNT5A, MSH2, and GATA3) and found that increased methylation of three or more and four or more genes (KLK10, SOX17, WNT5A, and MSH2) significantly correlated to OS (p = 0.042, 0.043, and 0.048) and the absence of pharmacotherapy response (p = 0.002), respectively. Subsequently, using machine learning combined clinical data and experimental findings, they developed multi-parametric prognostic signatures for the prediction of survival and treatment response to chemotherapy in metastatic breast cancer [19].
Correlations between elevated concentrations of cfDNA and tumor stage, tumor size, and nodal involvement were demonstrated [19]. In particular, Panagopoulou et al. showed that the metastatic breast cancer patients who had cfDNA levels > median value of 496.5 ng/mL had significantly shortened PFS, compared with those who had < median value of cfDNA (p = 0.036), indicating cfDNA quantification could serve as a prognostic marker for PFS. For the predictive value of cfDNA levels for the treatment response of metastatic breast cancer patients to first-line chemotherapy, the median value of cfDNA of the “non-responders” (970.0 ng/mL) was significantly higher than that of the “responders” (465.0 ng/mL, p = 0.026), thereby demonstrating cfDNA as a potent predictive marker for response to first-line chemotherapy [19]. The prognosis values of the combination of CTC and cfDNA were firstly evaluated by Ye et al. [61] by collecting blood samples from 117 metastatic breast cancer patients. High levels of CTC (CTC ≥ 5) and cfDNA, individually or jointly, had significantly higher risks of PFS and OS (CTC: p < 0.001 for PFS, p = 0.001 for OS; cfDNA: p = 0.001 for PFS, p = 0.002 for OS; joint effect: p < 0.001 for PFS, p = 0.002 for OS). In a similar result, Fernandez-Garcia et al. compared cfDNA and CTCs with conventional breast cancer blood biomarkers (CA15-3 and alkaline phosphatase (AP)) by analyzing blood samples from 194 metastatic breast cancer patients. Their results showed that both CTCs and total cfDNA levels are predictors for OS (p = 0.001 and 0.024, respectively), while only cfDNA correlated with PFS (p = 0.042), indicating their potential clinical application of liquid biopsy [62].
Generally, ctDNA can be released into the bloodstream by excretion and transport in exosomes or during the apoptosis and necrosis of tumor cells [47]. ctDNA is a small nucleic acid fragment of about 134–144 bp [50,63]. ctDNA is more abundant than CTCs, but it is more rapidly cleared from circulation, within hours, than CTCs. Moreover, ctDNA has been demonstrated to accurately represent the mutational profile of CTCs; Thierry et al. showed that ctDNA can capture the majority of mutations found in tissue biopsy, such as the PIK3CA and ESR1 mutations [64]. However, the evidence on the prognostic value of ctDNA in metastatic breast cancer is rather limited, especially compared with CTCs [65]. Specific somatic DNA mutations, loss of heterogeneity (LOH), and epigenetic alterations, such as methylations, are the valuable factors for precisely discriminating the cfDNA from normal cell and tumor cell [66]. LOH is a cross chromosomal event that results in the loss of one normal allele producing a locus with no normal function [67]. This is a common mechanism for cancer development as the inactivation of a tumor suppressor gene occurs [68]. ctDNA has been demonstrated to detect cancer in early stages [69,70], determine prognosis [13], real-time monitor treatment response [71], and determine therapeutic resistance [72], MRD after primary treatments, and relapse [73,74]. Minimally invasive serial measurement of ctDNA might, thus, monitor and predict treatment response, presenting an advantage over tissue biopsy [5,75,76,77] (Figure 1). Remarkably, increases in ctDNA levels could predict disease progression several months before standard imaging techniques [64]. However, ctDNA has not yet been validated to apply in clinical practice [78].
Prognostically, ctDNA detection was correlated with poor survival in early breast cancer [79,80,81,82]. As in early breast cancer, the quantity of ctDNA is associated with a worse outcome in metastatic breast cancer [75,76,77,83,84,85]. In both the INSPIRE phase II and LOTUS randomized phase II trials, ctDNA levels in TNBC were correlated with PFS, OS, and overall clinical response rate (ORR) [86].
In the aspect of recurrence, a prospective and multicenter study utilized serial plasma samples to assess patients with early-stage breast cancer [74]. Somatic mutations of primary tumors were identified by sequencing, and personalized tumor-specific digital polymerase chain reaction (digital PCR, dPCR) assays were applied to surveil these mutations. Plasma samples were collected every three months for the first-year follow-up and subsequently every six months. The results showed that the presence of ctDNA had a median lead time of 10.7 months before the development of clinical symptoms, indicating ctDNA could predict relapse. Moreover, the use of ctDNA could detect extracranial metastatic relapse in 96% of patients. This addressed that the use of ctDNA, as a surveillance technique, may improve survival.
A number of studies have evaluated ctDNA levels in both the neoadjuvant and adjuvant therapies [81,82,87,88,89]. In the phase 2 I-SPY 2 trial, Magbanua et al. examined the serial ctDNA test, in early breast cancer patients undertaking neoadjuvant chemotherapy, for predicting pathologic complete response (pCR) and risk of recurrence. Blood samples were collected at several time points, i.e., pretreatment, after therapy initiation, between regimens, or prior to surgery. Patients who remained ctDNA-positive after therapy initiation were significantly more likely to have residual disease after neoadjuvant chemotherapy (83% non-pCR) than those who were ctDNA-negative (52% non-pCR, p = 0.012). After neoadjuvant chemotherapy, the presence of ctDNA was associated with lower pCR rates, whereas ctDNA clearance after treatment was correlated with longer survival. Therefore, personalized monitoring of ctDNA during treatment may be a good predictor treatment response [81]. McDonald et al. also demonstrated nonmetastatic breast cancer patients with lower ctDNA concentrations achieve pCR than patients with higher ctDNA level after neoadjuvant therapy (p = 0.0057) [89], illustrating that personalized ctDNA panels could monitor breast cancer progression in the neoadjuvant setting. Most recently, Papakonstantinou et al. [90] performed a systematic review and meta-analysis to investigate the prognostic value of ctDNA in patients with early breast cancer treated with neoadjuvant therapy. The association between the detection of ctDNA, both at baseline and after completion of neoadjuvant therapy, and worse relapse-free survival (HR: 4.22, 95% CI: 1.29–13.82 and HR: 5.67, 95% CI: 2.73–11.75, respectively) and OS (HR: 19.1, 95% CI: 6.9–53.04 and HR: 4.00, 95% CI: 1.90–8.42, respectively) were observed, whereas the detection of ctDNA did not achieve a pCR. Therefore, this meta-analysis again supports the previous studies.
In metastatic breast cancer, Darrigues et al. also collected plasma samples of 61 patients at different time points, i.e., before treatment, at day 15, at day 30, and at disease progression, and proved that treatment with palbociclib and fulvestrant can be successfully monitored by serial ctDNA measurements before radiological evaluation [77]. However, more large, prospective, and randomized trials are needed. Interestingly, a study evaluated the predictive and prognostic values of ctDNA in 26 TNBC patients and observed a significant rise in ctDNA levels after neoadjuvant therapy was predictive of residual tumor and, thus, an incomplete pathologic response. This also indicated worse relapse-free survival (p = 0.046) and OS (p = 0.043) [79]. These studies support using serial monitoring of ctDNA for accurate assessment of tumor progression in real time, resulting therapeutic decision making. However, more clinical studies will be required before ctDNA monitoring can be implemented in a clinical setting [12,79].

2.3. Non-Coding RNAs

It is known that RNA, especially non-coding RNA (ncRNA), plays significant roles in the deregulation of cell function and cancer development. Like CTC and ctDNA, RNA can also be secreted from tumor cells into blood and other biological fluids of cancer patients and, thus, as a potential analyte in liquid biopsy [91,92]. However, RNA is less stable than CTC and DNA and hindered by the variability in the methodologies performed [93]. Despite these, there are growing evidences depicting the importance of circulating ncRNAs representing 80% of the total circulating RNA application in the field of oncology. They are involved in regulating transduction pathways, acting as tumor activators or suppressors [94]. There are a number of types of ncRNAs, including long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), microRNAs (miRNAs), and PIWI-interacting RNAs (piRNAs) [95,96].
microRNAs (miRNAs) are small ncRNAs (18~25 nt), capable of binding and regulating mRNA expression at the post-transcriptional level [97]. Additionally, miRNAs play important role in cellular communication, proliferation, programmed cell death, and differentiation [98]; thus, they have significant implications in cancer management [99] as potential biomarkers applied in liquid biopsy. miRNAs are derived from CTCs, cell-free miRNAs, apoptotic bodies, or from extracellular vesicles (EVs), either in their lumen or on their surface [100]. miRNAs are the most studied RNA types in tissues and the bloodstream, where several studies proved their clinical application in diagnosis, prognosis, detection of metastasis, and drug resistance [101,102,103,104]. However, little is known about their clinical utility as biomarkers in liquid biopsy, which requires more studies [101].

2.4. Extracellular Vesicles (EVs)

EVs refers to the cell-derived membranous vesicles released by all cells into the extracellular environment [105]. They play a role in intracellular communication among tumor cells [106]. EVs carry DNA, mRNA, ncRNA, lipids, metabolites, and proteins protecting and preventing degradation of their cargo from enzymes, such as plasma nucleases, and transferring their contents from a parental to different recipient cells [107,108]. Unlike CTCs, which are mostly released into blood, EVs exist in a variety of body fluids and can be more easily enriched for subsequent analysis than CTCs [109]. cfDNA is secreted into the bloodstream either as free DNA (unbound DNA), bound to protein or lipoprotein complexes (nucleosomes and vitrosomes) [110], or enclosed in EVs [111,112].
It has been proven that EVs, involve in the tumor development and initiating the formation of premetastasis niche, play a role in intracellular communication [113]. Tumor-derived vesicles also carry the molecular footprint reflecting the genetic status of parental tumor cells [114]. EVs have been demonstrated as diagnostic, prognostic, and therapeutic agents in clinical settings and have also been associated with drug resistance [115]. As a result, EVs are promising biomarkers in liquid biopsy. However, further studies are required to investigate their clinical validity in breast cancer [5].
EVs are generally heterogeneous and classified into microvesicles (MVs, also referred to as ectosomes or microparticles), exosomes, and apoptotic bodies, based on origin and size [116,117]. Apoptotic bodies are the largest vesicles (1~5 μm in diameter) derived from budding of apoptotic cells and usually contain nucleosomes, protecting tumor DNA and RNA from degradation by DNAses and RNAses [118,119].
The second largest EVs are microvesicles with large diameters (100–1000 nm) that are actively shed from protuberances in the plasma membrane [120,121]. Tumor-derived microvesicles (TDMs) contain DNA reflecting the genetic status of their original cell [5]; they also carry RNA that can be transferred to recipient cells [114]. It was found that the number of TDMs in the plasma of breast cancer patients was significantly associated to disease stages I-IV (p < 0.05 and p < 0.0001) [122], indicating a clinical value.
Exosomes, the best studied EVs, with small diameters (30–150 nm) derived from the endocytic pathway, are secreted upon fusion of multivesicular bodies (MVBs) with the plasma membrane [105,117,121]. Exosomes are secreted by almost all types of cells and can be transferred to recipient cells [123]. They also play critical roles in intercellular communication and can deliver their content to other cells in a paracrine fashion. Importantly, exosomes are also detected in biological fluids, including blood, saliva, urine, breast milk, and cerebrospinal fluid, indicating that they can act as mediators in long distance cellular signaling [124,125,126]. In particular, it has been demonstrated that exosomes contribute to cancer development and metastasis, preparation of the pre-metastatic niche, stem cell stimulation, apoptosis, angiogenesis, immunity, and drug resistance [117,127,128,129]. Tumor-derived exosomes also contain cancer-associated miRNA [130] and proteins [131] that could have diagnostic, prognostic, and therapy monitoring values. Exosomal miRNAs are also associated with tumor aggressiveness [132], angiogenesis [133], metastasis [134], and drug resistance [135] in breast cancer. Remarkably, it has been shown that tumor cells secrete more exosomes than normal cells in response to pathophysiological conditions, such as hypoxia in the tumor microenvironment [129]. Furthermore, exosomes from breast cancer patients contain distinct RNA and protein from healthy donors [136,137].
EVs represent one of the latest biomarkers in the liquid biopsy field; thus, the clinical application of EVs is still immature, and no standard detection method exists for breast cancer [23]. More clinical studies are required to confirm the clinical relevance of EVs, such as diagnosis and prognosis, and evaluate the sensitivity and specificity of EVs-based assays.

3. Biomarkers

Biomarkers, including DNA, miRNA, and EV, detected in blood or other body fluid, are important for early diagnosis and prognosis. In addition, the ability to monitor cancer progression and assess response to treatment is important for clinicians to determine the most effective therapy [12]. These can all be performed by liquid biopsy [138]. These are elucidated in detail in the following sections (Table 1); the sensitivities of these methods vary from 19% to 98%, and specificities vary from 64% to 100%.

3.1. Gene Mutation

3.1.1. For Diagnosis of Breast Cancer

A number of studies have shown that ctDNA PIK3CA mutations can be detected in breast cancer [177,178]. Rodriguez et al. [140] evaluated the utility of ctDNA in the diagnosis of early breast cancer patients by comparing the PIK3CA and TP53 mutations of fresh tissue biopsies and plasma samples using amplicon-based SafeSEQ (Sysmex Inostics) technology and NGS TruSeq custom amplicon low input panel (Illumina). Intriguingly, they found that plasma DNA sequencing permitted the identification of additional TP53 and PIK3CA mutations in ctDNA not detected in tumor biopsy sequencing. Furthermore, ctDNA detection was significantly correlated with younger age (p = 0.040), higher tumor grade (p = 0.041) and size (p = 0.033), immunohistochemical subtype (p = 0.025), BIRADS category (p = 0.004), and lymph node positivity (p < 0.001). This study addresses the fact that ctDNA analysis could be used in diagnosis of early breast cancer and highlights the importance of plasma ctDNA as an accurate alternative to tissue biopsy. More recently, Chin et al. showed the TP53, PIK3CA, and AKT1 mutations for ctDNA detection. ctDNA detection rates were 37% and 81% for stage I-III breast cancer and metastatic or recurrent breast cancer, respectively. Additionally, the ctDNA detection rate was correlated with disease stage (p = 0.00026), nodal spread (p = 0.00649), and distant metastases (p = 0.0005) [141].
Other studies have investigated additional mutations in ctDNA as a biomarker for early breast cancer detection. Bartnykaite et al. investigated the association between single nucleotide polymorphisms (SNPs) in MDM2 (rs2279744, rs937283, rs937282) and MDM4 (rs1380576, rs4245739) and breast cancer of the I–II stage. Their results showed that the rs937283 AG, rs937282 CG, rs1380576 CC, and rs4245739 AA genotypes were linked to HR+ breast cancer and suggested they may be useful diagnostic biomarker [142].

3.1.2. For Prognosis and Recurrence of Breast Cancer

It is important that early diagnosis of recurrence can decrease the mortality of breast cancer patients greatly. Consequently, there are a number of studies on the evaluation of biomarkers for the prognosis of breast cancer. Here, we only highlight the recent studies.
In a sub-study of the NeoALTTO phase 3 trial, a randomized, neoadjuvant dual HER2-targeted therapy study in early HER2+ breast cancer patients, Rothé and colleagues [144] found that the presence of PIK3CA and TP53 mutations, before neoadjuvant therapy, was correlated with low pCR (p = 0.0089). This result indicate that targeted treatment could be planned for patients carrying these mutations. Intriguingly, patients with HER2+ breast cancer and undetectable baseline ctDNA had the highest pCR, indicating the need to apply treatment de-escalation strategies.
Zhang et al. [143] developed a novel methodology to detect ctDNA by designing a panel based on COSMIC data, covering 136 genes and integrating with Breast Imaging Reporting and Data System classification (BI-RADS). Remarkably, the predictive value of this combination improved from 74.2% up to 92%. Thus, ctDNA detection is also a sensitive and specific marker indicating worse prognosis, and the combination of ctDNA with current imaging techniques might be applied to reduce surgical overtreatment.
MRD detection could be critical for assessing therapeutic response and guiding subsequent treatment decisions [78]. Hence, several trials specifically evaluated the value of ctDNA for detecting MRD in the post-neoadjuvant and post-surgery setting [88,179]. Parsons et al. developed an ultrasensitive patient-specific ctDNA panel for tracking up to 488 mutations having 100-fold more sensitivity than droplet dPCR (ddPCR). The clinical sensitivity for detecting MRD was 19% at 1-year postoperatively, which was strongly associated with distant recurrence (HR = 20.8; 95% CI: 7.3–58.9). Importantly, the median lead time from first positive test to recurrence was 18.9 months [88]. These patient-specific ctDNA mutations are attractive biomarkers in liquid biopsy and advance the field of precision medicine. However, it is important to avoid false-negative results, sequencing errors, and artifacts.
In addition, novel immunotherapies have emerged for cancer treatment. In a prospective clinical trial in 72 patients with metastatic breast cancer, no significant association was revealed between PD-L1 tumors and CTC expression. Triple negative (TN) phenotype, number of metastatic treatments, metastatic sites, ≥5 CTCs, and PD-L1(+)-CTCs were shown to be significantly correlated with PFS, whereas tissue PD-L1 expression was not. Therefore, unlike PD-L1(+) tumors, PD-L1 expression in CTCs was associated with survival in metastatic breast cancer, indicating a potential role of PD-L1(+)-CTCs as a stratifying factor for anti-PD-1/PD-L1 treatment for metastatic breast cancer patients [139].

3.1.3. For Predicting Treatment Response of Breast Cancer

In evaluating the predictive value of cfDNA in metastatic breast cancer patients, a large, phase III, ongoing PADA1 study analyzed the ESR1 mutation in 1017 ER+ HER2- patients treated with a first-line aromatase inhibitor plus palbociclib at regular intervals [151]. ESR1 mutations, detected in cfDNA, were associated with significantly shorter PFS on treatment with aromatase inhibitors + palbociclib than those with wild-type ESR1 (11.0 vs. 26.7 months). This indicated that the presence of the ESR1 mutation, at baseline, might promote resistance to aromatase inhibitors plus palbociclib.
The mutations in ctDNA may also be promising biomarkers for assessing treatment response or resistance [180]. For example, HER2 [82], ESR1 [181], and TP53 [182] in ctDNA could be applied to monitor therapy response. The ongoing plasmaMATCH trial, a multicenter, multicohort, phase IIa trial of ctDNA, tested on metastatic breast cancer patients from 18 UK hospitals, assesses the utility of ctDNA in predicting treatment response. A total of 1034 patients were recruited into four treatment cohorts, based on mutations identified in ctDNA—cohort A, with ESR1 mutations treated with extended dose of fulvestrant; cohort B, with HER2 mutations received oral neratinib and in case of ER+ breast cancer with a standard-dose of fulvestrant; cohort C, with AKT1 mutations in ER+ cancer treated with capivasertib + standard-dose of fulvestrant; and corhort D, with AKT1 mutations in ER- cancer or with PTEN mutations treated with capivasertib [146]. Long-term follow-up is ongoing, but preliminary results have shown positive response in cohorts B and C, with 25% and 22% response rates for neratinib and capivasertib, respectively, demonstrating the ctDNA’s utility to identify targetable mutations, PIK3CA, ESR1, HER2, PTEN, and AKT1 in metastatic patients and enables the selection of mutation-directed therapies (NCT03182634) [146].
Additionally, there are several studies that identified the PIK3CA mutation in ctDNA for treatment prediction. The phase III SOLAR-1 trial determined PIK3CA mutation-status using both tissue samples and ctDNA of 572 ER+ HER2- endocrine pretreated metastatic breast cancer patients. For the patients with ctDNA PIK3CA mutations treated with alpelisib and fulvestrant, there was a 45% risk reduction in PFS (11 vs. 5.7 months, HR: 0.65, 95% CI: 0.50–0.85, p < 0.001) [147,148]. The recently published paper for this trial showed an improvement of 7.9 months in OS for alpelisib treatment in the PIK3CA mutated group, but it was not statistically significant (HR: 0.86, 95%: CI 0.64–1.15, p = 0.15) [149].
In the retrospective study, Chen et al. revealed a direct correlation between ctDNA profiling, therapeutic response, and disease progression in breast cancer patients [145]. They identified multiple ctDNA mutations in HER2+ and HER2- breast cancer patients that reliably associated with treatment response and drug resistance. The ERBB2, TP53, EGFR, NF1, and SETD2 mutations were identified in HER2+ breast cancer as contributing to trastuzumab resistance, whereas in HER2- breast cancer patients with resistance to chemotherapy, genetic variations in the TP53, PIK3CA, and DNA damage repair genes were shown. The study also demonstrated that longitudinal ctDNA monitoring provides valuable insights for assessing therapy efficacy and therapeutic resistance.
The predictive value of ctDNA in drug efficacy was confirmed in the phase I/II BEECH trial, which included patients with ER+ metastatic breast cancer treated with paclitaxel and AKT-inhibitor capivasertib vs. paclitaxel and placebo [83]. Several specific mutations, such as AKT1, PIK3CA, ATM, TP53, ERB2 and ESR1, have been detected by ddPCR to evaluate ctDNA changes during the treatment. The median PFS in patients with suppressed ctDNA at 4 weeks and in patients with high ctDNA were 11.1 months and 6.4 months, respectively (p < 0.0001).
Apart from the PIK3CA and ESR1 mutations, other gene mutations were found to predict treatment response. In MONALEESA 2-, 3-, and 7-trials, the largest biomarker analysis of any CDK4/6 inhibitor in advanced breast cancer [150], blood samples from 1507 ER+ HER2-metastatic breast cancer patients were analyzed at baseline using NGS with a targeted panel of 557 genes. Gene changes in FRS2, PRKCA, MDM2, ERB2, AKT1, and BRCA1/2 were correlated with increased PFS benefit of ribociclib treatment; thus, they are potential biomarkers of response. However, the patients with alterations in CHD4, BCL11B, ATM, or CDKN2A/2B/2C genes had little or no benefit from ribociclib, indicating the biomarkers of resistance.
Therefore, these trials showed encouraging results that cfDNA and ctDNA in liquid biopsy may serve as early predictors for therapeutic response, and there are a number of gene mutations serving as biomarkers for this purpose.

3.2. miRNAs

3.2.1. For Diagnosis of Breast Cancer

Shimomura et al. [152] analyzed the sera of 1280 early breast cancer patients and demonstrated that the combination of five miRNAs (miR-1246, miR-1307-3p, miR-4634, miR-6861-5p, and miR-6875-5p) was able to detect early breast cancer with 97.6% sensitivity and 82.9% specificity. Similarly, Cui et al. [153] reanalyzed the dataset from Shimomura et al. [152] and revealed a panel of three miRNAs, miR-1246, miR-6756-5p, and miR-8073, in order to generate an neural network cascade model can successfully diagnose breast cancer. It showed 97.1% accuracy in 429 breast cancer patients and 895 healthy controls (AUC = 0.971, sensitivity = 96.7%, specificity = 97.2%). Therefore, these studies proved the feasibility of miRNA in liquid biopsy for early breast detection.
The novel method of machine learning models, incorporating a large set of miRNA expression profiles, have been developed for early detection of five types of cancers including breast cancer. For breast cancer, they demonstrated a 91% sensitivity and 90% specificity. This presents the promising value of liquid biopsy, combined with machine learning, that they are more sensitive even in the early stages of cancer, compared to other diagnostic methods, such as cfDNA diagnostics [183].
These studies confirm the great potential of miRNAs as diagnostic biomarkers for breast cancer. However, there is no best miRNA to be applied in the clinical setting yet. Hence, more accurate and robust studies for miRNA are required.

3.2.2. For Prognosis of Breast Cancer

miRNAs were also investigated as prognostic biomarkers in early breast cancer. By using Exiqon miRCURY microRNA RT-PCR panels, Huo et al. [154] investigated the expression levels of 11 miRNAs between patients with and without recurrence and identified seven miRNAs, including four upregulated (miR-21-5p, miR-194-5p, miR-205-5p, and miR-375) and three downregulated (miR-376c-3p, miR382-5p, and miR-411-5p) for recurrent patients. This seven-miRNA signature showed a better discriminatory capacity than individual miRNA and could be utilized as prognostic biomarkers for both HR+ and TNBC patients. Madhavan et al. [155] established a prognostic miRNA panel template (PROMPT), including 16 miRNAs, i.e., miR-141, miR-144, miR-193b, miR-200a, miR-200b, miR-200c, miR-203, miR-210, miR-215, miR-365, miR-375, miR-429, miR-486-5p, miR-801, miR-1260, and miR-1274a, correlated with OS and RFS. Therefore, these miRNAs could serve as prognostic biomarkers for metastatic breast cancer, which can assist making the decision of the treatment. Papadaki et al. [18] observed the expression levels of a different set of four miRNAs with an increase in the expression of miR-21 (p < 0.001), miR-23b (p = 0.028), and miR-200c (p < 0.001), as well as a decrease in miR-190 (p = 0.0032) that discriminated relapsed from non-relapsed patients. Thus, the combined expression of these four miRNAs could be prognostic biomarkers.
Obviously, there is increasing evidence that the use of miRNAs signatures as prognostic biomarkers is increasing, although no consensus has been reached for using in clinical setting.

3.2.3. For Predicting Treatment Response of Breast Cancer

Apart from the functions described, miRNAs can also be biomarkers for monitoring response in treatment. Hamam et al. proved miR-125b was correlated with chemotherapeutic resistance [156]. Furthermore, miR-155 levels decreased after surgery and four cycles of chemotherapy in breast cancer patients, indicating miR-155 can possibly monitor treatment response [157].

3.3. EVs

3.3.1. For Diagnosis of Breast Cancer

There are a number of studies identifying miRNAs derived from EVs for breast cancer detection. A recent study proved that a panel including miR-142-5p, miR-320a, and miR-4433b-5p has clinical value as breast cancer biomarker (AUC of 0.8387, sensitivity of 93.33%, and specificity of 68.75%) [158]. Zou et al. [160] focused on the members from the miR-532-502 cluster with tumor regulation roles. They analyzed the expression patterns of miRNAs in the miR-532-502 cluster in approximately 800 plasma and serum samples from breast cancer patients and healthy controls. Three miRNAs (miR-188-3p, miR-500a-5p, and miR-501-5p) in plasma and five miRNAs (miR-188-3p, miR-501-3p, miR-502-3p, miR-532-3p, and miR-532-5p) in serum were significantly increased in breast cancer patients. More recently, Zou et al. [161] investigated the expression of 12 miRNAs from 32 pairs of serum-derived exosomal samples from breast cancer patients and healthy controls and identified 10 miRNAs, i.e., let-7b-5p, miR-106a-5p, miR-19a-3p, miR-19b-3p, miR-25-3p, miR-425-5p, miR-451a, miR-92a-3p, miR-93-5p, and miR-16-5p, to be upregulated in breast cancer patients than controls.
Besides blood samples, Hirschfeld et al. analyzed the expression of 13 miRNAs derived from exosomes extracted from urine samples of 69 patients with breast cancer and 40 healthy controls [159]. They identified that a specific panel of four urine exosomal miRNAs, including miR-424, miR-423, miR-660, and let7-i, could be utilized as a highly specific combinatory biomarkers for detecting breast cancer (98.6% sensitivity and 100% specificity).
In the aspect of lncRNA, Zhong et al. discovered that serum exosomal levels of the lncRNA H19 were significantly more elevated in patients with breast cancer than healthy controls (p < 0.001), indicating as a novel biomarker for the diagnosis of breast cancer [162].
There are already some EV proteins, discovered from a large number of patient samples, showing good discriminative power. Thus, they are potential diagnostic biomarkers for breast cancer [23]. Recently, a prospective clinical pilot study revealed that there were increased levels of exosomal Hsp70 in plasma from breast cancer patients than healthy donors [163]. In addition, claudin-7 and claudin-7/CD81 levels in EVs showed no significant correlation with ER, PR, and HER2 status in breast cancer patients, indicating that claudin-7 might be a universal biomarker for the early diagnosis of breast cancer [164]. Vinik et al. [165] performed proteomic analysis of small EV and identified seven proteins, fibronectin, focal adhesion kinase 1 (FAK), dual-specificity mitogen-activated protein kinase kinase 1, β-Actin, p90RSK_pT573, N-cadherin, and proto-oncogene c-RAF, to discriminate breast cancer patients from healthy individuals (sensitivity: 94%, specificity: 82%), in which FAK and fibronectin revealed high early diagnostic accuracy. Furthermore, Li et al. [166] revealed the epidermal growth factor receptor (EGFR) as a potential biomarker candidate for the early diagnosis of breast cancer, with a sensitivity of >90% and different clinical stages of I–IV, although the AUC was ~0.7. Tian et al. [167] identified that the weighted sum of eight plasma EV protein markers, EV signature, including mucin-1, CA-125, carcinoembryonic antigen, HER2, EGFR, prostate-specific membrane antigen (PSMA), EpCAM, and vascular endothelial growth factor (VEGF) was able to discriminate metastatic breast cancer, non-metastatic breast cancer, and healthy donors, with a high accuracy of 91.1%.
Other studies proved the higher expression of serum exosomal annexin A2 (AnxA2) in breast cancer patients, compared to non-cancer females (p < 0.0001), especially for TNBC, rather than luminal and HER2+ breast cancer. In addition, high expression of exosomal AnxA2 levels in breast cancer was significantly correlated with tumor grade (p < 0.0001), poor OS (p = 0.0353), and DFS (p = 0.0301). Hence, in addition to the diagnostic biomarker, exosomal AnxA2 represents a promising prognostic biomarker and therapeutic target of TNBC [168].
Additionally, EV protein biomarkers from other sources were investigated. Takeuchi et al. [169] successfully applied tears, for the first time, to detect breast cancer-related small EVs and discovered that γ-glutamyltransferase 1, in small EVs from tears, could differentiate between breast cancer patients and healthy donors. Further analysis revealed a significant reduction in postoperative γ-glutamyltransferase 1 signals from tear small EVs of patients with stage I breast cancer, before and after total mastectomy, also revealed a significant reduction in postoperative γ-glutamyltransferase 1 signals. Because the sample size was small, further large studies are required to verify this diagnostic biomarker [23].

3.3.2. For Prognosis of Breast Cancer

In the study of Rodríguez-Martínez et al. [170], the miRNA expression in 53 patients was measured before and after neoadjuvant therapy, showing that levels of circulating exosomal miR-21 and miR-105 were significantly higher in metastatic patients, compared to local ones, as well as controls. In HER2+ cancers, the level of miR-21 decreased after treatment with trastuzumab in a neoadjuvant setting, suggesting that miR-21 levels could be the biomarker for monitoring the treatment response [170].
Most recently, three exosomal miRNAs, miR-30b, miR-328, and miR-423 before neoadjuvant chemotherapy predicted pCR. An increase in miR-127 correlated with pCR in TNBC. After the first neoadjuvant chemotherapy, exo-miR-141 was used to predict pCR, whereas non-pCR was predicted by miR-34a, exo-miR182, and exo-miR-183. The candidate miRNAs were significantly correlated with OS, subtype, and metastasis in breast cancer, indicating their promising role as predictive biomarkers of pCR in liquid biopsy [171]. However, this should be further validated by studies with large cohorts.
In the aspect of protein, the heat shock protein 70 was significantly upregulated in presurgery plasma small EVs from patients with recurrence [165]. Likewise, Rothammer et al. [172] demonstrated that heat shock protein 70 levels in serum from breast cancer patients who developed recurrence or metastases after radiotherapy were significantly higher than those who remained disease-free (p = 0.007). As a result, these confirmed that heat shock protein 70 in EVs could be the prognostic biomarkers for breast cancer.

3.3.3. For Predicting Treatment Response of Breast Cancer

Most studies investigated exosomal miRNAs, but there are still few studies demonstrating that exosomal mRNAs and lncRNAs might be potential biomarkers [184]. In HR+ advanced breast cancer, upregulated exosomal mRNAs encoding cell cycle-regulated thymidine kinase 1 (TK1) (p = 0.01) and cyclin-dependent kinase 9 (CDK9) (p = 0.03) correlated with poor clinical response to the CDK4/CDK6 inhibitor palbociclib [173]. Furthermore, it was found that lncRNA HOTAIR levels were elevated in circulating exosomes in breast cancer patients than those in healthy controls [174]. High exosomal HOTAIR levels were also correlated with poor prognosis; thus, it is the potential diagnostic and prognostic biomarker.
Keklikoglou et al. revealed increased levels of ANXA6 in plasma EVs, compared with pretreatment levels in five of six breast cancer patients undergoing neo-adjuvant treatment, decreased at the end of therapy with partial or complete remission [175]. This addresses the importance of EV proteins that might have the potential to be used as the biomarker clinically for breast cancer prognosis [23].

3.4. Proteins

Although proteins are mostly expressed on CTCs or enclosed in EVs, they can be freely circulated in blood stream. Recently, a novel biomarker, protein cellular communication network factor 1 (CNN1), was analyzed in the plasma of 544 breast cancer patients and 427 healthy controls by ELISA [176]. CCN1, formerly cysteine-rich angiogenic inducer 61, is an extracellular matrix-associated signaling protein of the CCN family and can regulate a broad range of cellular functions, such as cell adhesion, migration, and differentiation, by interacting with cell surface integrin receptors [185,186]. Importantly, CCN1 has been involved in breast cancer progression [176]. Remarkably, it showed the cancer detection specificity of 99.0% and sensitivity of 80.0%. Even 81.5% of small T1 cancers were CCN1+. These demonstrated that circulating protein CCN1 could be applied in the early detection of breast cancer, and it was suggested it could be, thus, included in liquid biopsy panels containing other DNA or proteins [176].

4. Detection Techniques

The detection methods for tumor components are well-established. There are three main types of detection methods: PCR-based techniques, targeted deep sequencing, and whole-genome sequencing (WGS) [12]. For PCR-based method, dPCR are extremely sensitive and can detect point mutations as low as 0.01% [187]; this is suitable for liquid biopsy as the concentrations of tumor components are always low. Furthermore, dPCR has been developed to ddPCR [188]. However, these techniques require prior mutational information of tumor cells [12]. Unknown mutations are screened by targeted DNA sequencing techniques, cancer personalized profiling by deep sequencing (CAPP-Seq), tagged-amplicon deep sequencing (Tam-Seq), the safe sequencing system (Safe-SeqS), and amplicon sequencing (AmpliSeq) by means of next-generation sequencing (NGS) [189]. Whole-genome (or exome) sequencing (WGS/WES) can provide a more comprehensive ctDNA profile, based on the somatic chromosomal aberrations, copy number variations, and detection of rearrangements; however, they have the disadvantage of decreased analytical sensitivity [15]. The recent development of detection techniques is elucidated in the details in the following section and Table 2.

4.1. Detection for CTCs

There is a variety of technologies for detecting CTC. The technique developed by Menarini Silicon Biosystems called CellSearch® is the gold standard and only technique approved by the FDA for isolating and detecting CTCs in metastatic breast, colon, and prostate cancers. This technique target the epithelial marker protein, EpCAM, on surface of CTCs. However, when CTCs do not express EpCAM in the status of EMT or the stem cell stage, they will not be detected. Several studies have confirmed that a count of ≥1 CTCs, in 7.5 mL of blood, by the CellSearch® system, at different time points, is correlated with worse PFS and OS, and recurrence [37,190,207]. Adnatest (QIAGEN®) is another commercial technique for CTCs analysis, which uses a combination of antibodies conjugated with magnetic beads for targeting epithelial markers and RT-PCR for detecting mRNAs biomarkers. Kasimir-Bauer et al. [191] utilized this technique to isolate CTCs and demonstrated the correlation between CTC and worse prognosis. In the study by Kwan et al. [192], a digital RNA signature and a technique called CTC-iChip were performed for CTC isolation and detection in early and metastatic breast cancer patients.
In addition, a nanotube-CTC-chip, a newly developed methodology to detect CTCs in early breast cancer patients, was used [193,194]. This technique utilizes label-free nanotube-antibody microarrays using breast cancer-specific antibodies, such as anti-EpCAM and anti-HER2. Remarkably, this technology was able to identify CTCs in the 100% of the studied breast cancer blood samples. Most recently, Abdulla et al. [195] developed a novel antibody functionalized microfluidic (AFM) chip for detection of CTCs in breast cancer patients’ whole blood. AFM chip can achieve capture efficiency of 99.5% and capture EpCAM, CK19, CD45, and DAPI rapidly, demonstrating AFM chip could be beneficial in clinical setting.

4.2. Detection for cfDNA

The targeted DNA sequencing techniques are very useful for analyzing a limited panel of potential mutations in biopsy samples [196,208]. The Oncomine Breast cfDNA (Thermofisher, Waltham, MA, USA) test, based on AmpliSeq technology, is applied in clinical practice to detect mutations in a limited number of genes from breast cancer patients [196]. More recently, Xie et al. [197] validated the newly developed dPCR detection on HER2 status of cfDNA in stage III/IV breast cancer by comparing with tissue biopsy by using immunohistochemistry (IHC)/fluorescence in situ hybridization (FISH). The sensitivity and specificity between dPCR in plasma and IHC/FISH in tissues were 43.75% and 84.38%, respectively, for 224 breast cancer patients. The overall concordance is 66.97%. Therefore, dPCR could be used as a companion diagnostic tool to detect plasma HER2 status.

4.3. Detection for ctDNA

Generally, there are two main techniques for detecting ctDNA, targeted and untargeted techniques. The targeted technique, referring to ddPCR or beads amplification magnetics PCR (BEAMing-PCR), is applied to detect previously determined tumor-specific mutations, such as PIK3CA and ESR1. The untargeted technique, on the other hand, refers to digital NGS (dNGS), such as genome-wide analysis of copy number aberrations (CNAs), WGS, WES, or array comparative genomic hybridization (CGH). These methods allow sequencing of large DNA or RNA fragments and are able to detect previously unknown genetic mutations [22]. Measuring ctDNA is more specific than measuring cfDNA; thus, developing tools for ctDNA detection are more clinically applicable [184].

4.3.1. ddPCR

Both ddPCR and BEAMing have been shown to be able to detect PIK3CA mutations in plasma ctDNA from patients with breast cancer [199,200]. The Therascreen PIK3CA RGQ PCR kit, detecting PIK3CA mutations on ctDNA, has been approved by the FDA. It is able to detect 11 mutations in the PIK3CA gene from patients with HR+, HER2-, and advanced or metastatic breast cancer. The presence of PIK3CA mutations is correlated, with response to treatment with PIQRAY® (alpelisib) [147]. Therefore, this kit may assist doctors stratify breast cancer patients who should be treated with PIQRAY®.
Recently, Wan et al. developed INtegration of VAriant Reads (INVAR), a novel technique for ctDNA detection. INVAR can detect as little as one mutant molecule per 100,000, thus significantly increasing the sensitivity. However, the median integrated mutant allele fraction (IMAF), obtained in early breast cancer, was 5.2 parts-per-million (ppm), much lower than that obtained in advanced melanoma with the 15,000 ppm, addressing the difficulties to detect ctDNA in localized breast tumors [202].

4.3.2. NGS

The first custom-built NGS-based ctDNA test, Signatera™, was launched in 2019, aiming to improve the detection of MRD after surgery, as well as earlier detection of recurrence. Signatera™ provides each individual with a customized blood test, tailored to match the clonal mutations by WES. Coombes et al. validated the clinical utility of Signatera™ on breast cancer patient samples [73]. Coombes et al. [73] performed WES of tumor tissues from 49 patients and designed personalized profiling targeting 16 patient-specific variants, targeted by multiplex sequencing of plasma ctDNA in the detection for recurrence. Importantly, ctDNA was detected in disease relapse in 89% of the relapsed patients, up to two years earlier than imaging with a specificity of 100%. Such early identification of relapse may provide more effective treatments.
McDonald et al. developed a targeted digital sequencing (TARDIS) of ctDNA might be a highly sensitive technique for predicting pCR to neoadjuvant treatment in early and locally advanced breast cancers [89]. TARDIS uses simultaneous deep sequencing of patient-specific panels, comprised of multiple tumor mutations as ctDNA biomarkers for monitoring disease in the pre-surgery setting, as well as to detect MRD [89]. By applying this sensitive technology, McDonald et al. analyzed plasma for personal mutations before neoadjuvant treatment and detected ctDNA in 100% of the samples. ctDNA was then monitored at different time points of neoadjuvant treatment and, expectedly, ctDNA levels were higher in patients with the residual disease, compared to those that displayed pCR. Therefore, TARDIS is a promising test demonstrating the ctDNA clinical relevance as a biomarker for NAT treatment response and MRD detection and surveillance in early breast cancer.
Most recently, PIK3CA hotspot mutations in HR+ metastatic breast cancer were analyzed by a newly developed and high-resolution SiMSen-Seq assay [201]. SiMSen-seq, a simple, multiplexed, PCR-based barcoding of DNA and sequencing, allows detection of extremely rare variant alleles at <0.1% frequency [209]. As a result, PIK3CA mutations were detected in 47.3% of plasma samples, with identical PIK3CA mutation detected in both tissue and plasma in 33.3% patients [201]. This implies detection of PIK3CA mutations in plasma using SiMSen-seq is feasible and shows concordance with tissue biopsy.
Both PCR and NGS-based techniques showed promising results; these might be the future trend in clinical practice.

4.4. Detection for miRNA

The current methods for detecting miRNA are reverse transcription-quantitative PCR (RT-qPCR), dPCR, microarray, and NGS. RT-qPCR is the gold standard method for quantifying small amounts of miRNA, showing good sensitivity, reproducibility, and accuracy. dPCR offer another technique for quantifying miRNA [210]. Microarray and NGS approaches are usually utilized for initial screening and obtain profiles of miRNAs, whereas RT-qPCR and dPCR are applied to validate previous results [211].
A newly developed technique for detecting miRNA is called surface-enhanced Raman spectroscopy (SERS) with seed-mediated grown Ag nanopillars (SMGAPs). The electrochemical reduction on the pre-distributed 40 nm gold nanoparticle seeds (sGNP) served as scaffolds for growth of silver ion, and a nanopillar-shaped silver structure was successfully grown on the substrate surface of gold. miR-21 and miR-155 were applied as the SERS diagnostic target. The limits of detection of each labeled target were 451 zmol and 1.65 amol, respectively. Hence, quantitative analysis of miRNA in urine was successful, compared to that of the healthy group [203].

4.5. Detection for Protein

Currently, commercialized EV ELISA kits have been available for quantifying common EV proteins, such as the tetraspanins CD63, CD9, or CD81. Mass spectrometry, on the other hand, is the core technique for characterizing protein. Recently, new techniques have been developed for quantifying proteins, such as microfluidics, SERS, high-resolution flow cytometry, antibody microarrays, electrochemical sensors, and DNA aptamers [212]. Furthermore, a localized fluorescent imaging method, termed digital profiling of proteins on individual EV (DPPIE), was recently developed for analysis of multiple proteins, CD63, EpCAM, and mucin-1, on individual EV [204]. High-dimensional data collected from each individual EV would provide more precise information than ELISA. The proportion of CD63/EpCAM/mucin-1 vesicle in patients with breast cancer was significantly higher than that of healthy control with overall accuracy of 91%. Additionally, high-resolution flow cytometry has been commercialized, and the standardization of assay is also being established [205]. With advance in nanotechnology, microfluidic devices are expected to gain better specificity and sensitivity [206]. Moreover, thermophoretic aptasensor (TAS) is rapid, sensitive, and low-cost to profile cancer-associated protein profiles of plasma EVs. This aptasensor method has several advantages that preseparation of EVs is not needed, the total detection time is short (within 3 h), and it has a low cost (less than $1) [167].This work addresses the promising clinical utility of EVs in the care of metastatic breast cancer. Combination of these diverse detection techniques on a microfluidic platform could achieve personalized assistance for the clinical application of EVs in breast cancer [213].

5. Current Challenges with Liquid Biopsy

Although liquid biopsy has many advantages over tissue biopsy, there are challenges for liquid biopsy that need to be solved before applying in clinical practice. The challenges for CTCs are isolation, detection limit and feasibility in clinical application. The number of CTCs in blood is low and profoundly diluted by blood cells, making their detection technically difficult particularly in early breast cancer [101,214]. Currently, the CellSearch® platform is the only technique approved by the FDA for isolating CTCs [215]. The isolation of CTCs by CellSearch® platform is based on the expression of EpCAM. However, EpCAM expression is downregulated in most aggressive cancer cells undergoing EMT [216,217] mentioned earlier in Section 4.1. Therefore, this makes CellSearch® platform face the serious limitation of leaving most CTCs undetected in advanced stage of breast cancer [218]. In addition, current biological techniques will inactivate CTCs, which greatly affects the application of CTCs in clinical setting. Thus, there is an urgent need to develop a mild and specific technique for isolating CTCs in vitro.
The difficulty for cfDNA detection is again its low amount in blood [219]. Importantly, the major challenge of utilizing cfDNA for diagnosing breast cancer is the need for a prior knowledge of tumor-specific variants [220]. An impediment of technical detection of ctDNA in early cancer stages is low concentration of ctDNA, relative to the total concentration of cfDNA [15,221]. Early stages of cancers have about <0.1% ctDNA (10 ctDNA copies per 5 mL), in contrast to ~1% in non-metastatic advanced cancer patients and in stage IV 100–1000 copies per 5 mL (up to 10%) were detected [222,223]. Consequently, technical detection of ctDNA in early cancer stages is strongly challenging, due to its extremely low concentration and requires ultra-sensitive technologies. Recently, Stetson et al. compared four commercial NGS gene panel assays for detecting mutations in ctDNAs. However, there were substantial variability among the ctDNA assays, with a range of sensitivity (38~89%) and positive predictive value (36~80%), particularly in the detection of allele frequency variants <1% [224]. These findings indicate that most NGS assay discordance is a result of technical variations; therefore, standardization of sample collection and analysis is urgently needed before ctDNA tests could be implemented in the clinical setting. Lastly, the utility of ctDNA for diagnosis is further complicated by false-positive readouts caused by clonal hematopoiesis of indeterminant potential (CHIP) mutations, which are somatic mutations in blood stem cells in healthy and elderly population [225,226]. CHIP mutations are considered background noise in liquid biopsy samples, and thus they can lead to in accurate diagnosis and subsequently inappropriate therapeutic treatment [227]. Therefore, it is significantly required to develop strategies for accurately identifying CHIP mutations to avoid false positives.
There are challenges in translating an accurate and reliable panel of circulating miRNA to clinical setting, due to their low amount, differences in the cohort size, collection, type of sample and processing, and current inability to detect novel miRNAs [156,210]. There is not a significant overlap in the miRNA panels across the different studies, reflecting the complicated miRNA expression in breast cancer patients [214]. Another variation is the isolation method different for miRNAs from different sources, such as EVs or cancer cells [210].
For exosome, it is required to standardize exosome extraction, which can exclude contaminants, such as lipoprotein particles and protein complexes [228]. Furthermore, the methods need to be fast and convenient for application in clinical practice; however, the current isolation method for exosome is ultracentrifugation, which is tedious [184].

6. Conclusions and Future Directions

Despite the recent advance in diagnosis and treatment for breast cancer, breast cancer is still the leading cause of death in women worldwide. Therefore, developing innovative technologies with the clinical potential to detect breast cancer at its early stage and predict treatment response is still highly required. Liquid biopsy has gained much attention as a non-invasive methodology, which serves to obtain key tumor information via blood-based biomarkers for cancer diagnosis and treatment monitoring. All the tumor components of liquid biopsy, including CTCs, cfDNA, ctDNA, miRNA, and EVs, have promising value in diagnosis, prognosis, and treatment prediction. Especially, the greatest advantage of liquid biopsy over tissue biopsy is the “real-time” longitudinal monitoring of disease progression and treatment response. Currently, only a liquid biopsy blood test, for detecting the PIK3CA mutation, is approved by the FDA for breast cancer that can be used in clinical practice. Most biomarkers described in this review are still under clinical trials or pilot studies (Table 1).
Despite the benefits mentioned above, the clinical application of tumor components in liquid biopsy remains to be fully established and requires performing multicentre clinical studies with a large cohort of breast cancer patients. Another major problem hampering their application in clinical practice is the lack of a standard procedure. Furthermore, it is difficult to detect circulating tumor in the early stage of breast cancer, as their amounts in the biofluids from patients are very low. Important advances in detection were developed, such as NGS techniques, which can detect ultra-low diluted tumor materials. Once standard and feasible methods are established, these technologies benefit both the patients and clinicians, as they are relatively inexpensive and noninvasive for diagnosing and monitoring early-stage cancers.
Most RNAs show promising utility in liquid biopsy are miRNAs. More characterization of other circulating RNA types, such as lncRNAs, will provide more options for liquid biopsy.
Finally, the combination of different tumor components, i.e., multi-omics approaches, should be considered, in order to fulfil the unmet need in clinical practice.

Author Contributions

Conceptualization, H.-J.W. and P.-Y.C.; writing—original draft preparation, H.-J.W.; writing—review and editing, P.-Y.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the Ministry of Science and Technology, Taiwan (MOST 106-2314-B-442-001-MY3, and MOST 109-2314-B-442-001, National Health Research Institutes, Taiwan (NHRI-109BCCO-MF-202015-01), and Show Chwan Memorial Hospital, Taiwan (SRD-109023, SRD-109034, SRD-109035, SRD-110008, SRD-110016, SRD-110017, SRD-110030, and SRD-110052).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
  3. Stastny, I.; Zubor, P.; Kajo, K.; Kubatka, P.; Golubnitschaja, O.; Dankova, Z. Aberrantly Methylated cfDNA in Body Fluids as a Promising Diagnostic Tool for Early Detection of Breast Cancer. Clin. Breast Cancer 2020, 20, e711–e722. [Google Scholar] [CrossRef]
  4. Harbeck, N.; Penault-Llorca, F.; Cortes, J.; Gnant, M.; Houssami, N.; Poortmans, P.; Ruddy, K.; Tsang, J.; Cardoso, F. Breast cancer. Nat. Rev. Dis. Primers 2019, 5, 66. [Google Scholar] [CrossRef] [PubMed]
  5. Panagopoulou, M.; Esteller, M.; Chatzaki, E. Circulating Cell-Free DNA in Breast Cancer: Searching for Hidden Information towards Precision Medicine. Cancers 2021, 13, 728. [Google Scholar] [CrossRef] [PubMed]
  6. Oeffinger, K.C.; Fontham, E.T.; Etzioni, R.; Herzig, A.; Michaelson, J.S.; Shih, Y.C.; Walter, L.C.; Church, T.R.; Flowers, C.R.; LaMonte, S.J.; et al. Breast Cancer Screening for Women at Average Risk: 2015 Guideline Update From the American Cancer Society. JAMA 2015, 314, 1599–1614. [Google Scholar] [CrossRef]
  7. Marinovich, M.L.; Bernardi, D.; Macaskill, P.; Ventriglia, A.; Sabatino, V.; Houssami, N. Agreement between digital breast tomosynthesis and pathologic tumour size for staging breast cancer, and comparison with standard mammography. Breast 2019, 43, 59–66. [Google Scholar] [CrossRef]
  8. Zubor, P.; Kubatka, P.; Kajo, K.; Dankova, Z.; Polacek, H.; Bielik, T.; Kudela, E.; Samec, M.; Liskova, A.; Vlcakova, D.; et al. Why the Gold Standard Approach by Mammography Demands Extension by Multiomics? Application of Liquid Biopsy miRNA Profiles to Breast Cancer Disease Management. Int. J. Mol. Sci. 2019, 20, 2878. [Google Scholar] [CrossRef] [Green Version]
  9. Alimirzaie, S.; Bagherzadeh, M.; Akbari, M.R. Liquid biopsy in breast cancer: A comprehensive review. Clin. Genet. 2019, 95, 643–660. [Google Scholar] [CrossRef]
  10. Palmirotta, R.; Lovero, D.; Cafforio, P.; Felici, C.; Mannavola, F.; Pelle, E.; Quaresmini, D.; Tucci, M.; Silvestris, F. Liquid biopsy of cancer: A multimodal diagnostic tool in clinical oncology. Ther. Adv. Med. Oncol. 2018, 10, 1758835918794630. [Google Scholar] [CrossRef]
  11. Poulet, G.; Massias, J.; Taly, V. Liquid Biopsy: General Concepts. Acta Cytol. 2019, 63, 449–455. [Google Scholar] [CrossRef] [PubMed]
  12. Croessmann, S.; Park, B.H. Circulating tumor DNA in early-stage breast cancer: New directions and potential clinical applications. Clin. Adv. Hematol. Oncol. 2021, 19, 155–161. [Google Scholar] [PubMed]
  13. Wang, R.; Li, X.; Zhang, H.; Wang, K.; He, J. Cell-free circulating tumor DNA analysis for breast cancer and its clinical utilization as a biomarker. Oncotarget 2017, 8, 75742–75755. [Google Scholar] [CrossRef] [PubMed]
  14. Appierto, V.; Di Cosimo, S.; Reduzzi, C.; Pala, V.; Cappelletti, V.; Daidone, M.G. How to study and overcome tumor heterogeneity with circulating biomarkers: The breast cancer case. Semin. Cancer Biol. 2017, 44, 106–116. [Google Scholar] [CrossRef] [PubMed]
  15. De Rubis, G.; Rajeev Krishnan, S.; Bebawy, M. Liquid Biopsies in Cancer Diagnosis, Monitoring, and Prognosis. Trends Pharmacol. Sci. 2019, 40, 172–186. [Google Scholar] [CrossRef]
  16. Beca, F.; Polyak, K. Intratumor Heterogeneity in Breast Cancer. Adv. Exp. Med. Biol. 2016, 882, 169–189. [Google Scholar]
  17. Fiala, C.; Diamandis, E.P. Utility of circulating tumor DNA in cancer diagnostics with emphasis on early detection. BMC Med. 2018, 16, 166. [Google Scholar] [CrossRef] [Green Version]
  18. Papadaki, C.; Stratigos, M.; Markakis, G.; Spiliotaki, M.; Mastrostamatis, G.; Nikolaou, C.; Mavroudis, D.; Agelaki, S. Circulating microRNAs in the early prediction of disease recurrence in primary breast cancer. Breast Cancer Res. 2018, 20, 72. [Google Scholar] [CrossRef] [Green Version]
  19. Panagopoulou, M.; Karaglani, M.; Balgkouranidou, I.; Biziota, E.; Koukaki, T.; Karamitrousis, E.; Nena, E.; Tsamardinos, I.; Kolios, G.; Lianidou, E.; et al. Circulating cell-free DNA in breast cancer: Size profiling, levels, and methylation patterns lead to prognostic and predictive classifiers. Oncogene 2019, 38, 3387–3401. [Google Scholar] [CrossRef]
  20. Siravegna, G.; Marsoni, S.; Siena, S.; Bardelli, A. Integrating liquid biopsies into the management of cancer. Nat. Rev. Clin. Oncol. 2017, 14, 531–548. [Google Scholar] [CrossRef]
  21. Bardelli, A.; Pantel, K. Liquid Biopsies, What We Do Not Know (Yet). Cancer Cell 2017, 31, 172–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Alix-Panabieres, C.; Pantel, K. Clinical Applications of Circulating Tumor Cells and Circulating Tumor DNA as Liquid Biopsy. Cancer Discov. 2016, 6, 479–491. [Google Scholar] [CrossRef] [Green Version]
  23. Li, D.; Lai, W.; Fan, D.; Fang, Q. Protein biomarkers in breast cancer-derived extracellular vesicles for use in liquid biopsies. Am. J. Physiol. Cell Physiol. 2021, 321, C779–C797. [Google Scholar] [CrossRef] [PubMed]
  24. Bidard, F.C.; Michiels, S.; Riethdorf, S.; Mueller, V.; Esserman, L.J.; Lucci, A.; Naume, B.; Horiguchi, J.; Gisbert-Criado, R.; Sleijfer, S.; et al. Circulating Tumor Cells in Breast Cancer Patients Treated by Neoadjuvant Chemotherapy: A Meta-analysis. J. Natl. Cancer Inst. 2018, 110, 560–567. [Google Scholar] [CrossRef] [PubMed]
  25. Micalizzi, D.S.; Maheswaran, S.; Haber, D.A. A conduit to metastasis: Circulating tumor cell biology. Genes Dev. 2017, 31, 1827–1840. [Google Scholar] [CrossRef] [PubMed]
  26. Gkountela, S.; Castro-Giner, F.; Szczerba, B.M.; Vetter, M.; Landin, J.; Scherrer, R.; Krol, I.; Scheidmann, M.C.; Beisel, C.; Stirnimann, C.U.; et al. Circulating Tumor Cell Clustering Shapes DNA Methylation to Enable Metastasis Seeding. Cell 2019, 176, 98–112.e14. [Google Scholar] [CrossRef] [Green Version]
  27. Yu, M.; Bardia, A.; Wittner, B.S.; Stott, S.L.; Smas, M.E.; Ting, D.T.; Isakoff, S.J.; Ciciliano, J.C.; Wells, M.N.; Shah, A.M.; et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science 2013, 339, 580–584. [Google Scholar] [CrossRef] [Green Version]
  28. Labelle, M.; Begum, S.; Hynes, R.O. Direct signaling between platelets and cancer cells induces an epithelial-mesenchymal-like transition and promotes metastasis. Cancer Cell 2011, 20, 576–590. [Google Scholar] [CrossRef] [Green Version]
  29. Mohme, M.; Riethdorf, S.; Pantel, K. Circulating and disseminated tumour cells—Mechanisms of immune surveillance and escape. Nat. Rev. Clin. Oncol. 2017, 14, 155–167. [Google Scholar] [CrossRef]
  30. Kitamura, T.; Qian, B.Z.; Pollard, J.W. Immune cell promotion of metastasis. Nat. Rev. Immunol. 2015, 15, 73–86. [Google Scholar] [CrossRef]
  31. De Toro, J.; Herschlik, L.; Waldner, C.; Mongini, C. Emerging roles of exosomes in normal and pathological conditions: New insights for diagnosis and therapeutic applications. Front. Immunol. 2015, 6, 203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Banys-Paluchowski, M.; Reinhard, F.; Fehm, T. Circulating Tumor Cells in Metastatic Breast Cancer: Clinical Applications and Future Possibilities. Appl. Sci. 2020, 10, 3311. [Google Scholar] [CrossRef]
  33. Bidard, F.C.; Jacot, W.; Kiavue, N.; Dureau, S.; Kadi, A.; Brain, E.; Bachelot, T.; Bourgeois, H.; Goncalves, A.; Ladoire, S.; et al. Efficacy of Circulating Tumor Cell Count-Driven vs Clinician-Driven First-line Therapy Choice in Hormone Receptor-Positive, ERBB2-Negative Metastatic Breast Cancer: The STIC CTC Randomized Clinical Trial. JAMA Oncol. 2021, 7, 34–41. [Google Scholar] [CrossRef] [PubMed]
  34. Rack, B.; Schindlbeck, C.; Juckstock, J.; Andergassen, U.; Hepp, P.; Zwingers, T.; Friedl, T.W.; Lorenz, R.; Tesch, H.; Fasching, P.A.; et al. Circulating tumor cells predict survival in early average-to-high risk breast cancer patients. J. Natl. Cancer Inst. 2014, 106, dju066. [Google Scholar] [CrossRef] [PubMed]
  35. Zhang, L.; Riethdorf, S.; Wu, G.; Wang, T.; Yang, K.; Peng, G.; Liu, J.; Pantel, K. Meta-analysis of the prognostic value of circulating tumor cells in breast cancer. Clin. Cancer Res. 2012, 18, 5701–5710. [Google Scholar] [CrossRef] [Green Version]
  36. Janni, W.J.; Rack, B.; Terstappen, L.W.; Pierga, J.Y.; Taran, F.A.; Fehm, T.; Hall, C.; de Groot, M.R.; Bidard, F.C.; Friedl, T.W.; et al. Pooled Analysis of the Prognostic Relevance of Circulating Tumor Cells in Primary Breast Cancer. Clin. Cancer Res. 2016, 22, 2583–2593. [Google Scholar] [CrossRef] [Green Version]
  37. Sparano, J.; O’Neill, A.; Alpaugh, K.; Wolff, A.C.; Northfelt, D.W.; Dang, C.T.; Sledge, G.W.; Miller, K.D. Association of Circulating Tumor Cells With Late Recurrence of Estrogen Receptor-Positive Breast Cancer: A Secondary Analysis of a Randomized Clinical Trial. JAMA Oncol. 2018, 4, 1700–1706. [Google Scholar] [CrossRef]
  38. Martos, T.; Casadevall, D.; Albanell, J. Circulating Tumor Cells: Applications for Early Breast Cancer. Adv. Exp. Med. Biol. 2020, 1220, 135–146. [Google Scholar]
  39. Cayrefourcq, L.; Alix-Panabieres, C. Clinical relevance of liquid biopsy in breast cancer: Update in 2020. Expert Rev. Mol. Diagn. 2020, 20, 913–919. [Google Scholar] [CrossRef]
  40. Cristofanilli, M.; Pierga, J.Y.; Reuben, J.; Rademaker, A.; Davis, A.A.; Peeters, D.J.; Fehm, T.; Nole, F.; Gisbert-Criado, R.; Mavroudis, D.; et al. The clinical use of circulating tumor cells (CTCs) enumeration for staging of metastatic breast cancer (MBC): International expert consensus paper. Crit. Rev. Oncol. Hematol. 2019, 134, 39–45. [Google Scholar] [CrossRef]
  41. Muller, V.; Banys-Paluchowski, M.; Friedl, T.W.P.; Fasching, P.A.; Schneeweiss, A.; Hartkopf, A.; Wallwiener, D.; Rack, B.; Meier-Stiegen, F.; Huober, J.; et al. Prognostic relevance of the HER2 status of circulating tumor cells in metastatic breast cancer patients screened for participation in the DETECT study program. ESMO Open 2021, 6, 100299. [Google Scholar] [CrossRef] [PubMed]
  42. Pachmann, K.; Schuster, S. The Value of Monitoring the Behavior of Circulating Tumor Cells at the End of Endocrine Therapy in Breast Cancer Patients. Cancers 2018, 10, 407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Medford, A.J.; Dubash, T.D.; Juric, D.; Spring, L.; Niemierko, A.; Vidula, N.; Peppercorn, J.; Isakoff, S.; Reeves, B.A.; LiCausi, J.A.; et al. Blood-based monitoring identifies acquired and targetable driver HER2 mutations in endocrine-resistant metastatic breast cancer. NPJ Precis Oncol. 2019, 3, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Cabel, L.; Berger, F.; Cottu, P.; Loirat, D.; Rampanou, A.; Brain, E.; Cyrille, S.; Bourgeois, H.; Kiavue, N.; Deluche, E.; et al. Clinical utility of circulating tumour cell-based monitoring of late-line chemotherapy for metastatic breast cancer: The randomised CirCe01 trial. Br. J. Cancer 2021, 124, 1207–1213. [Google Scholar] [CrossRef]
  45. Malani, R.; Fleisher, M.; Kumthekar, P.; Lin, X.; Omuro, A.; Groves, M.D.; Lin, N.U.; Melisko, M.; Lassman, A.B.; Jeyapalan, S.; et al. Cerebrospinal fluid circulating tumor cells as a quantifiable measurement of leptomeningeal metastases in patients with HER2 positive cancer. J. Neurooncol. 2020, 148, 599–606. [Google Scholar] [CrossRef] [PubMed]
  46. Kidess, E.; Jeffrey, S.S. Circulating tumor cells versus tumor-derived cell-free DNA: Rivals or partners in cancer care in the era of single-cell analysis? Genome Med. 2013, 5, 70. [Google Scholar] [CrossRef] [Green Version]
  47. Thierry, A.R.; El Messaoudi, S.; Gahan, P.B.; Anker, P.; Stroun, M. Origins, structures, and functions of circulating DNA in oncology. Cancer Metastasis Rev. 2016, 35, 347–376. [Google Scholar] [CrossRef] [Green Version]
  48. Gerner, C.; Costigliola, V.; Golubnitschaja, O. Multiomic Patterns in Body Fluids: Technological Challenge with a Great Potential to Implement the Advanced Paradigm of 3p Medicine. Mass Spectrom. Rev. 2020, 39, 442–451. [Google Scholar] [CrossRef]
  49. Schwarzenbach, H.; Hoon, D.S.; Pantel, K. Cell-free nucleic acids as biomarkers in cancer patients. Nat. Rev. Cancer 2011, 11, 426–437. [Google Scholar] [CrossRef]
  50. Stewart, C.M.; Kothari, P.D.; Mouliere, F.; Mair, R.; Somnay, S.; Benayed, R.; Zehir, A.; Weigelt, B.; Dawson, S.J.; Arcila, M.E.; et al. The value of cell-free DNA for molecular pathology. J. Pathol. 2018, 244, 616–627. [Google Scholar] [CrossRef]
  51. Tan, G.; Chu, C.; Gui, X.; Li, J.; Chen, Q. The prognostic value of circulating cell-free DNA in breast cancer: A meta-analysis. Medicine 2018, 97, e0197. [Google Scholar] [CrossRef] [PubMed]
  52. Romagnolo, D.F.; Daniels, K.D.; Grunwald, J.T.; Ramos, S.A.; Propper, C.R.; Selmin, O.I. Epigenetics of breast cancer: Modifying role of environmental and bioactive food compounds. Mol. Nutr. Food Res. 2016, 60, 1310–1329. [Google Scholar] [CrossRef] [Green Version]
  53. Williams, K.E.; Jawale, R.M.; Schneider, S.S.; Otis, C.N.; Pentecost, B.T.; Arcaro, K.F. DNA methylation in breast cancers: Differences based on estrogen receptor status and recurrence. J. Cell Biochem. 2019, 120, 738–755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Parashar, S.; Cheishvili, D.; Mahmood, N.; Arakelian, A.; Tanvir, I.; Khan, H.A.; Kremer, R.; Mihalcioiu, C.; Szyf, M.; Rabbani, S.A. DNA methylation signatures of breast cancer in peripheral T-cells. BMC Cancer 2018, 18, 574. [Google Scholar] [CrossRef] [PubMed]
  55. Fece de la Cruz, F.; Corcoran, R.B. Methylation in cell-free DNA for early cancer detection. Ann. Oncol. 2018, 29, 1351–1353. [Google Scholar] [CrossRef] [PubMed]
  56. Panagopoulou, M.; Karaglani, M.; Balgkouranidou, I.; Pantazi, C.; Kolios, G.; Kakolyris, S.; Chatzaki, E. Circulating cell-free DNA release in vitro: Kinetics, size profiling, and cancer-related gene methylation. J. Cell Physiol. 2019, 234, 14079–14089. [Google Scholar] [CrossRef]
  57. Pfeifer, G.P. Defining Driver DNA Methylation Changes in Human Cancer. Int. J. Mol. Sci. 2018, 19, 1166. [Google Scholar] [CrossRef] [Green Version]
  58. Qian, X.; Ruan, L. APC gene promoter aberrant methylation in serum as a biomarker for breast cancer diagnosis: A meta-analysis. Thorac. Cancer 2018, 9, 284–290. [Google Scholar] [CrossRef] [Green Version]
  59. Shan, M.; Yin, H.; Li, J.; Li, X.; Wang, D.; Su, Y.; Niu, M.; Zhong, Z.; Wang, J.; Zhang, X.; et al. Detection of aberrant methylation of a six-gene panel in serum DNA for diagnosis of breast cancer. Oncotarget 2016, 7, 18485–18494. [Google Scholar] [CrossRef] [Green Version]
  60. Salta, S.; Nunes, P.S.; Fontes-Sousa, M.; Lopes, P.; Freitas, M.; Caldas, M.; Antunes, L.; Castro, F.; Antunes, P.; Palma de Sousa, S.; et al. A DNA Methylation-Based Test for Breast Cancer Detection in Circulating Cell-Free DNA. J. Clin. Med. 2018, 7, 420. [Google Scholar] [CrossRef] [Green Version]
  61. Ye, Z.; Wang, C.; Wan, S.; Mu, Z.; Zhang, Z.; Abu-Khalaf, M.M.; Fellin, F.M.; Silver, D.P.; Neupane, M.; Jaslow, R.J.; et al. Association of clinical outcomes in metastatic breast cancer patients with circulating tumour cell and circulating cell-free DNA. Eur. J. Cancer 2019, 106, 133–143. [Google Scholar] [CrossRef] [PubMed]
  62. Fernandez-Garcia, D.; Hills, A.; Page, K.; Hastings, R.K.; Toghill, B.; Goddard, K.S.; Ion, C.; Ogle, O.; Boydell, A.R.; Gleason, K.; et al. Plasma cell-free DNA (cfDNA) as a predictive and prognostic marker in patients with metastatic breast cancer. Breast Cancer Res. 2019, 21, 149. [Google Scholar] [CrossRef] [PubMed]
  63. Siravegna, G.; Mussolin, B.; Venesio, T.; Marsoni, S.; Seoane, J.; Dive, C.; Papadopoulos, N.; Kopetz, S.; Corcoran, R.B.; Siu, L.L.; et al. How liquid biopsies can change clinical practice in oncology. Ann. Oncol. 2019, 30, 1580–1590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Rossi, G.; Mu, Z.; Rademaker, A.W.; Austin, L.K.; Strickland, K.S.; Costa, R.L.B.; Nagy, R.J.; Zagonel, V.; Taxter, T.J.; Behdad, A.; et al. Cell-Free DNA and Circulating Tumor Cells: Comprehensive Liquid Biopsy Analysis in Advanced Breast Cancer. Clin. Cancer Res. 2018, 24, 560–568. [Google Scholar] [CrossRef] [Green Version]
  65. Banys-Paluchowski, M.; Krawczyk, N.; Fehm, T. Liquid Biopsy in Breast Cancer. Geburtshilfe Frauenheilkd 2020, 80, 1093–1104. [Google Scholar] [CrossRef]
  66. Warton, K.; Mahon, K.L.; Samimi, G. Methylated circulating tumor DNA in blood: Power in cancer prognosis and response. Endocr. Relat. Cancer 2016, 23, R157-71. [Google Scholar] [CrossRef] [Green Version]
  67. Joseph, C.G.; Darrah, E.; Shah, A.A.; Skora, A.D.; Casciola-Rosen, L.A.; Wigley, F.M.; Boin, F.; Fava, A.; Thoburn, C.; Kinde, I.; et al. Association of the autoimmune disease scleroderma with an immunologic response to cancer. Science 2014, 343, 152–157. [Google Scholar] [CrossRef] [Green Version]
  68. Ganten, D.; Ruckpaul, K. Loss of Heterogeneity. In Encyclopedic Reference of Genomics and Proteomics in Molecular Medicine; Springer: Berlin/Heidelberg, Germany, 2006. [Google Scholar]
  69. Cohen, J.D.; Li, L.; Wang, Y.; Thoburn, C.; Afsari, B.; Danilova, L.; Douville, C.; Javed, A.A.; Wong, F.; Mattox, A.; et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science 2018, 359, 926–930. [Google Scholar] [CrossRef] [Green Version]
  70. Phallen, J.; Sausen, M.; Adleff, V.; Leal, A.; Hruban, C.; White, J.; Anagnostou, V.; Fiksel, J.; Cristiano, S.; Papp, E.; et al. Direct detection of early-stage cancers using circulating tumor DNA. Sci. Transl. Med. 2017, 9, eaan2415. [Google Scholar] [CrossRef] [Green Version]
  71. Thierry, A.R.; Mouliere, F.; El Messaoudi, S.; Mollevi, C.; Lopez-Crapez, E.; Rolet, F.; Gillet, B.; Gongora, C.; Dechelotte, P.; Robert, B.; et al. Clinical validation of the detection of KRAS and BRAF mutations from circulating tumor DNA. Nat. Med. 2014, 20, 430–435. [Google Scholar] [CrossRef]
  72. Misale, S.; Yaeger, R.; Hobor, S.; Scala, E.; Janakiraman, M.; Liska, D.; Valtorta, E.; Schiavo, R.; Buscarino, M.; Siravegna, G.; et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature 2012, 486, 532–536. [Google Scholar] [CrossRef] [Green Version]
  73. Coombes, R.C.; Page, K.; Salari, R.; Hastings, R.K.; Armstrong, A.; Ahmed, S.; Ali, S.; Cleator, S.; Kenny, L.; Stebbing, J.; et al. Personalized Detection of Circulating Tumor DNA Antedates Breast Cancer Metastatic Recurrence. Clin. Cancer Res. 2019, 25, 4255–4263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Garcia-Murillas, I.; Chopra, N.; Comino-Mendez, I.; Beaney, M.; Tovey, H.; Cutts, R.J.; Swift, C.; Kriplani, D.; Afentakis, M.; Hrebien, S.; et al. Assessment of Molecular Relapse Detection in Early-Stage Breast Cancer. JAMA Oncol. 2019, 5, 1473–1478. [Google Scholar] [CrossRef] [PubMed]
  75. Jacob, S.; Davis, A.A.; Gerratana, L.; Velimirovic, M.; Shah, A.N.; Wehbe, F.; Katam, N.; Zhang, Q.; Flaum, L.; Siziopikou, K.P.; et al. The Use of Serial Circulating Tumor DNA to Detect Resistance Alterations in Progressive Metastatic Breast Cancer. Clin. Cancer Res. 2021, 27, 1361–1370. [Google Scholar] [CrossRef] [PubMed]
  76. Aguilar-Mahecha, A.; Lafleur, J.; Brousse, S.; Savichtcheva, O.; Holden, K.A.; Faulkner, N.; McLennan, G.; Jensen, T.J.; Basik, M. Early, On-Treatment Levels and Dynamic Changes of Genomic Instability in Circulating Tumor DNA Predict Response to Treatment and Outcome in Metastatic Breast Cancer Patients. Cancers 2021, 13, 1331. [Google Scholar] [CrossRef]
  77. Darrigues, L.; Pierga, J.Y.; Bernard-Tessier, A.; Bieche, I.; Silveira, A.B.; Michel, M.; Loirat, D.; Cottu, P.; Cabel, L.; Dubot, C.; et al. Circulating tumor DNA as a dynamic biomarker of response to palbociclib and fulvestrant in metastatic breast cancer patients. Breast Cancer Res. 2021, 23, 31. [Google Scholar] [CrossRef]
  78. Sant, M.; Bernat-Peguera, A.; Felip, E.; Margeli, M. Role of ctDNA in Breast Cancer. Cancers 2022, 14, 310. [Google Scholar] [CrossRef]
  79. Cavallone, L.; Aguilar-Mahecha, A.; Lafleur, J.; Brousse, S.; Aldamry, M.; Roseshter, T.; Lan, C.; Alirezaie, N.; Bareke, E.; Majewski, J.; et al. Prognostic and predictive value of circulating tumor DNA during neoadjuvant chemotherapy for triple negative breast cancer. Sci. Rep. 2020, 10, 14704. [Google Scholar] [CrossRef]
  80. Radovich, M.; Jiang, G.; Hancock, B.A.; Chitambar, C.; Nanda, R.; Falkson, C.; Lynce, F.C.; Gallagher, C.; Isaacs, C.; Blaya, M.; et al. Association of Circulating Tumor DNA and Circulating Tumor Cells After Neoadjuvant Chemotherapy With Disease Recurrence in Patients With Triple-Negative Breast Cancer: Preplanned Secondary Analysis of the BRE12-158 Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1410–1415. [Google Scholar] [CrossRef]
  81. 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.; et al. Circulating tumor DNA in neoadjuvant-treated breast cancer reflects response and survival. Ann. Oncol. 2021, 32, 229–239. [Google Scholar] [CrossRef]
  82. Guan, X.; Liu, B.; Niu, Y.; Dong, X.; Zhu, X.; Li, C.; Li, L.; Yi, Z.; Sun, X.; Chen, H.; et al. Longitudinal HER2 amplification tracked in circulating tumor DNA for therapeutic effect monitoring and prognostic evaluation in patients with breast cancer. Breast 2020, 49, 261–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Hrebien, S.; Citi, V.; Garcia-Murillas, I.; Cutts, R.; Fenwick, K.; Kozarewa, I.; McEwen, R.; Ratnayake, J.; Maudsley, R.; Carr, T.H.; et al. Early ctDNA dynamics as a surrogate for progression-free survival in advanced breast cancer in the BEECH trial. Ann. Oncol. 2019, 30, 945–952. [Google Scholar] [CrossRef] [PubMed]
  84. Kruger, D.T.; Jansen, M.; Konings, I.; Dercksen, W.M.; Jager, A.; Oulad Hadj, J.; Sleijfer, S.; Martens, J.W.M.; Boven, E. High ctDNA molecule numbers relate with poor outcome in advanced ER+, HER2− postmenopausal breast cancer patients treated with everolimus and exemestane. Mol. Oncol. 2020, 14, 490–503. [Google Scholar] [CrossRef] [PubMed]
  85. O’Leary, B.; Cutts, R.J.; Huang, X.; Hrebien, S.; Liu, Y.; Andre, F.; Loibl, S.; Loi, S.; Garcia-Murillas, I.; Cristofanilli, M.; et al. Circulating Tumor DNA Markers for Early Progression on Fulvestrant With or Without Palbociclib in ER+ Advanced Breast Cancer. J. Natl. Cancer Inst. 2021, 113, 309–317. [Google Scholar] [CrossRef] [PubMed]
  86. Fiste, O.; Liontos, M.; Koutsoukos, K.; Terpos, E.; Dimopoulos, M.A.; Zagouri, F. Circulating tumor DNA-based predictive biomarkers in breast cancer clinical trials: A narrative review. Ann. Transl. Med. 2020, 8, 1603. [Google Scholar] [CrossRef] [PubMed]
  87. Riva, F.; Bidard, F.C.; Houy, A.; Saliou, A.; Madic, J.; Rampanou, A.; Hego, C.; Milder, M.; Cottu, P.; Sablin, M.P.; et al. Patient-Specific Circulating Tumor DNA Detection during Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer. Clin. Chem. 2017, 63, 691–699. [Google Scholar] [CrossRef] [Green Version]
  88. Parsons, H.A.; Rhoades, J.; Reed, S.C.; Gydush, G.; Ram, P.; Exman, P.; Xiong, K.; Lo, C.C.; Li, T.; Fleharty, M.; et al. Sensitive Detection of Minimal Residual Disease in Patients Treated for Early-Stage Breast Cancer. Clin. Cancer Res. 2020, 26, 2556–2564. [Google Scholar] [CrossRef] [Green Version]
  89. 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.; et al. Personalized circulating tumor DNA analysis to detect residual disease after neoadjuvant therapy in breast cancer. Sci. Transl Med. 2019, 11, eaax7392. [Google Scholar] [CrossRef]
  90. Papakonstantinou, A.; Gonzalez, N.S.; Pimentel, I.; Sunol, A.; Zamora, E.; Ortiz, C.; Espinosa-Bravo, M.; Peg, V.; Vivancos, A.; Saura, C.; et al. Prognostic value of ctDNA detection in patients with early breast cancer undergoing neoadjuvant therapy: A systematic review and meta-analysis. Cancer Treat. Rev. 2022, 104, 102362. [Google Scholar] [CrossRef]
  91. Weber, J.A.; Baxter, D.H.; Zhang, S.; Huang, D.Y.; Huang, K.H.; Lee, M.J.; Galas, D.J.; Wang, K. The microRNA spectrum in 12 body fluids. Clin. Chem. 2010, 56, 1733–1741. [Google Scholar] [CrossRef]
  92. Zeng, Z.; Chen, X.; Zhu, D.; Luo, Z.; Yang, M. Low Expression of Circulating MicroRNA-34c is Associated with Poor Prognosis in Triple-Negative Breast Cancer. Yonsei Med. J. 2017, 58, 697–702. [Google Scholar] [CrossRef] [PubMed]
  93. Yuan, T.; Huang, X.; Woodcock, M.; Du, M.; Dittmar, R.; Wang, Y.; Tsai, S.; Kohli, M.; Boardman, L.; Patel, T.; et al. Plasma extracellular RNA profiles in healthy and cancer patients. Sci. Rep. 2016, 6, 19413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Lanzos, A.; Carlevaro-Fita, J.; Mularoni, L.; Reverter, F.; Palumbo, E.; Guigo, R.; Johnson, R. Discovery of Cancer Driver Long Noncoding RNAs across 1112 Tumour Genomes: New Candidates and Distinguishing Features. Sci. Rep. 2017, 7, 41544. [Google Scholar] [CrossRef] [Green Version]
  95. Dvinge, H.; Guenthoer, J.; Porter, P.L.; Bradley, R.K. RNA components of the spliceosome regulate tissue- and cancer-specific alternative splicing. Genome Res. 2019, 29, 1591–1604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Liu, Y.; Dou, M.; Song, X.; Dong, Y.; Liu, S.; Liu, H.; Tao, J.; Li, W.; Yin, X.; Xu, W. The emerging role of the piRNA/piwi complex in cancer. Mol. Cancer 2019, 18, 123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Rupaimoole, R.; Calin, G.A.; Lopez-Berestein, G.; Sood, A.K. miRNA Deregulation in Cancer Cells and the Tumor Microenvironment. Cancer Discov. 2016, 6, 235–246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Ramassone, A.; Pagotto, S.; Veronese, A.; Visone, R. Epigenetics and MicroRNAs in Cancer. Int. J. Mol. Sci. 2018, 19, 459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Mandujano-Tinoco, E.A.; Garcia-Venzor, A.; Melendez-Zajgla, J.; Maldonado, V. New emerging roles of microRNAs in breast cancer. Breast Cancer Res. Treat. 2018, 171, 247–259. [Google Scholar] [CrossRef]
  100. Sohel, M.H. Extracellular/Circulating MicroRNAs: Release Mechanisms, Functions and Challenges. Achiev. Life Sci. 2016, 10, 175–186. [Google Scholar] [CrossRef] [Green Version]
  101. Alba-Bernal, A.; Lavado-Valenzuela, R.; Dominguez-Recio, M.E.; Jimenez-Rodriguez, B.; Queipo-Ortuno, M.I.; Alba, E.; Comino-Mendez, I. Challenges and achievements of liquid biopsy technologies employed in early breast cancer. EBioMedicine 2020, 62, 103100. [Google Scholar] [CrossRef]
  102. Mangolini, A.; Ferracin, M.; Zanzi, M.V.; Saccenti, E.; Ebnaof, S.O.; Poma, V.V.; Sanz, J.M.; Passaro, A.; Pedriali, M.; Frassoldati, A.; et al. Diagnostic and prognostic microRNAs in the serum of breast cancer patients measured by droplet digital PCR. Biomark Res. 2015, 3, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Kleivi Sahlberg, K.; Bottai, G.; Naume, B.; Burwinkel, B.; Calin, G.A.; Borresen-Dale, A.L.; Santarpia, L. A serum microRNA signature predicts tumor relapse and survival in triple-negative breast cancer patients. Clin. Cancer Res. 2015, 21, 1207–1214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Shaker, O.; Maher, M.; Nassar, Y.; Morcos, G.; Gad, Z. Role of microRNAs -29b-2, -155, -197 and -205 as diagnostic biomarkers in serum of breast cancer females. Gene 2015, 560, 77–82. [Google Scholar] [CrossRef] [PubMed]
  105. Hessvik, N.P.; Llorente, A. Current knowledge on exosome biogenesis and release. Cell Mol. Life Sci. 2018, 75, 193–208. [Google Scholar] [CrossRef] [Green Version]
  106. Vader, P.; Breakefield, X.O.; Wood, M.J. Extracellular vesicles: Emerging targets for cancer therapy. Trends Mol. Med. 2014, 20, 385–393. [Google Scholar] [CrossRef] [Green Version]
  107. Yanez-Mo, M.; Siljander, P.R.; Andreu, Z.; Zavec, A.B.; Borras, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [Green Version]
  108. Javeed, N.; Mukhopadhyay, D. Exosomes and their role in the micro-/macro-environment: A comprehensive review. J. Biomed. Res. 2017, 31, 386–394. [Google Scholar]
  109. Vasconcelos, M.H.; Caires, H.R.; Abols, A.; Xavier, C.P.R.; Line, A. Extracellular vesicles as a novel source of biomarkers in liquid biopsies for monitoring cancer progression and drug resistance. Drug Resist. Updates 2019, 47, 100647. [Google Scholar] [CrossRef]
  110. Snyder, M.W.; Kircher, M.; Hill, A.J.; Daza, R.M.; Shendure, J. Cell-free DNA Comprises an In Vivo Nucleosome Footprint that Informs Its Tissues-of-Origin. Cell 2016, 164, 57–68. [Google Scholar] [CrossRef] [Green Version]
  111. Wang, W.; Kong, P.; Ma, G.; Li, L.; Zhu, J.; Xia, T.; Xie, H.; Zhou, W.; Wang, S. Characterization of the release and biological significance of cell-free DNA from breast cancer cell lines. Oncotarget 2017, 8, 43180–43191. [Google Scholar] [CrossRef] [Green Version]
  112. Fernando, M.R.; Jiang, C.; Krzyzanowski, G.D.; Ryan, W.L. New evidence that a large proportion of human blood plasma cell-free DNA is localized in exosomes. PLoS ONE 2017, 12, e0183915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Becker, A.; Thakur, B.K.; Weiss, J.M.; Kim, H.S.; Peinado, H.; Lyden, D. Extracellular Vesicles in Cancer: Cell-to-Cell Mediators of Metastasis. Cancer Cell 2016, 30, 836–848. [Google Scholar] [CrossRef] [Green Version]
  114. Balaj, L.; Lessard, R.; Dai, L.; Cho, Y.J.; Pomeroy, S.L.; Breakefield, X.O.; Skog, J. Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nat. Commun. 2011, 2, 180. [Google Scholar] [CrossRef] [PubMed]
  115. Gyorgy, B.; Hung, M.E.; Breakefield, X.O.; Leonard, J.N. Therapeutic applications of extracellular vesicles: Clinical promise and open questions. Annu. Rev. Pharmacol. Toxicol. 2015, 55, 439–464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Thery, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [Green Version]
  117. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, 367. [Google Scholar] [CrossRef]
  118. Mittra, I.; Nair, N.K.; Mishra, P.K. Nucleic acids in circulation: Are they harmful to the host? J. Biosci. 2012, 37, 301–312. [Google Scholar] [CrossRef]
  119. Akers, J.C.; Gonda, D.; Kim, R.; Carter, B.S.; Chen, C.C. Biogenesis of extracellular vesicles (EV): Exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies. J. Neurooncol. 2013, 113, 1–11. [Google Scholar] [CrossRef] [Green Version]
  120. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [Green Version]
  121. van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
  122. Galindo-Hernandez, O.; Villegas-Comonfort, S.; Candanedo, F.; Gonzalez-Vazquez, M.C.; Chavez-Ocana, S.; Jimenez-Villanueva, X.; Sierra-Martinez, M.; Salazar, E.P. Elevated concentration of microvesicles isolated from peripheral blood in breast cancer patients. Arch. Med. Res. 2013, 44, 208–214. [Google Scholar] [CrossRef] [PubMed]
  123. Peinado, H.; Lavotshkin, S.; Lyden, D. The secreted factors responsible for pre-metastatic niche formation: Old sayings and new thoughts. Semin. Cancer Biol. 2011, 21, 139–146. [Google Scholar] [CrossRef] [PubMed]
  124. LeBleu, V.S.; Kalluri, R. Exosomes as a Multicomponent Biomarker Platform in Cancer. Trends Cancer 2020, 6, 767–774. [Google Scholar] [CrossRef] [PubMed]
  125. Zhou, B.; Xu, K.; Zheng, X.; Chen, T.; Wang, J.; Song, Y.; Shao, Y.; Zheng, S. Application of exosomes as liquid biopsy in clinical diagnosis. Signal Transduct. Target. Ther. 2020, 5, 144. [Google Scholar] [CrossRef]
  126. Logozzi, M.; Mizzoni, D.; Di Raimo, R.; Fais, S. Exosomes: A Source for New and Old Biomarkers in Cancer. Cancers 2020, 12, 2566. [Google Scholar] [CrossRef]
  127. Guo, Y.; Ji, X.; Liu, J.; Fan, D.; Zhou, Q.; Chen, C.; Wang, W.; Wang, G.; Wang, H.; Yuan, W.; et al. Effects of exosomes on pre-metastatic niche formation in tumors. Mol. Cancer 2019, 18, 39. [Google Scholar] [CrossRef] [Green Version]
  128. Dai, J.; Su, Y.; Zhong, S.; Cong, L.; Liu, B.; Yang, J.; Tao, Y.; He, Z.; Chen, C.; Jiang, Y. Exosomes: Key players in cancer and potential therapeutic strategy. Signal Transduct. Target. Ther. 2020, 5, 145. [Google Scholar] [CrossRef]
  129. McAndrews, K.M.; Kalluri, R. Mechanisms associated with biogenesis of exosomes in cancer. Mol. Cancer 2019, 18, 52. [Google Scholar] [CrossRef]
  130. Melo, S.A.; Sugimoto, H.; O’Connell, J.T.; Kato, N.; Villanueva, A.; Vidal, A.; Qiu, L.; Vitkin, E.; Perelman, L.T.; Melo, C.A.; et al. Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis. Cancer Cell 2014, 26, 707–721. [Google Scholar] [CrossRef] [Green Version]
  131. Lee, J.E.; Moon, P.G.; Cho, Y.E.; Kim, Y.B.; Kim, I.S.; Park, H.; Baek, M.C. Identification of EDIL3 on extracellular vesicles involved in breast cancer cell invasion. J. Proteom. 2016, 131, 17–28. [Google Scholar] [CrossRef]
  132. Eichelser, C.; Stuckrath, I.; Muller, V.; Milde-Langosch, K.; Wikman, H.; Pantel, K.; Schwarzenbach, H. Increased serum levels of circulating exosomal microRNA-373 in receptor-negative breast cancer patients. Oncotarget 2014, 5, 9650–9663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. O’Brien, K.; Rani, S.; Corcoran, C.; Wallace, R.; Hughes, L.; Friel, A.M.; McDonnell, S.; Crown, J.; Radomski, M.W.; O’Driscoll, L. Exosomes from triple-negative breast cancer cells can transfer phenotypic traits representing their cells of origin to secondary cells. Eur. J. Cancer 2013, 49, 1845–1859. [Google Scholar] [CrossRef] [PubMed]
  134. Tominaga, N.; Kosaka, N.; Ono, M.; Katsuda, T.; Yoshioka, Y.; Tamura, K.; Lotvall, J.; Nakagama, H.; Ochiya, T. Brain metastatic cancer cells release microRNA-181c-containing extracellular vesicles capable of destructing blood-brain barrier. Nat. Commun. 2015, 6, 6716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Chen, W.X.; Liu, X.M.; Lv, M.M.; Chen, L.; Zhao, J.H.; Zhong, S.L.; Ji, M.H.; Hu, Q.; Luo, Z.; Wu, J.Z.; et al. Exosomes from drug-resistant breast cancer cells transmit chemoresistance by a horizontal transfer of microRNAs. PLoS ONE 2014, 9, e95240. [Google Scholar] [CrossRef]
  136. Jia, Y.; Chen, Y.; Wang, Q.; Jayasinghe, U.; Luo, X.; Wei, Q.; Wang, J.; Xiong, H.; Chen, C.; Xu, B.; et al. Exosome: Emerging biomarker in breast cancer. Oncotarget 2017, 8, 41717–41733. [Google Scholar] [CrossRef] [Green Version]
  137. Meng, Y.; Sun, J.; Wang, X.; Hu, T.; Ma, Y.; Kong, C.; Piao, H.; Yu, T.; Zhang, G. Exosomes: A Promising Avenue for the Diagnosis of Breast Cancer. Technol. Cancer Res. Treat. 2019, 18, 1533033818821421. [Google Scholar] [CrossRef] [Green Version]
  138. Buono, G.; Gerratana, L.; Bulfoni, M.; Provinciali, N.; Basile, D.; Giuliano, M.; Corvaja, C.; Arpino, G.; Del Mastro, L.; De Placido, S.; et al. Circulating tumor DNA analysis in breast cancer: Is it ready for prime-time? Cancer Treat. Rev. 2019, 73, 73–83. [Google Scholar] [CrossRef] [Green Version]
  139. Jacot, W.; Mazel, M.; Mollevi, C.; Pouderoux, S.; D’Hondt, V.; Cayrefourcq, L.; Bourgier, C.; Boissiere-Michot, F.; Berrabah, F.; Lopez-Crapez, E.; et al. Clinical Correlations of Programmed Cell Death Ligand 1 Status in Liquid and Standard Biopsies in Breast Cancer. Clin. Chem. 2020, 66, 1093–1101. [Google Scholar] [CrossRef]
  140. Rodriguez, B.J.; Cordoba, G.D.; Aranda, A.G.; Alvarez, M.; Vicioso, L.; Perez, C.L.; Hernando, C.; Bermejo, B.; Parreno, A.J.; Lluch, A.; et al. Detection of TP53 and PIK3CA Mutations in Circulating Tumor DNA Using Next-Generation Sequencing in the Screening Process for Early Breast Cancer Diagnosis. J. Clin. Med. 2019, 8, 1183. [Google Scholar] [CrossRef] [Green Version]
  141. Chin, Y.M.; Takahashi, Y.; Chan, H.T.; Otaki, M.; Fujishima, M.; Shibayama, T.; Miki, Y.; Ueno, T.; Nakamura, Y.; Low, S.K. Ultradeep targeted sequencing of circulating tumor DNA in plasma of early and advanced breast cancer. Cancer Sci. 2021, 112, 454–464. [Google Scholar] [CrossRef]
  142. Bartnykaite, A.; Savukaityte, A.; Ugenskiene, R.; Dauksaite, M.; Korobeinikova, E.; Gudaitiene, J.; Juozaityte, E. Associations of MDM2 and MDM4 Polymorphisms with Early-Stage Breast Cancer. J. Clin. Med. 2021, 10, 886. [Google Scholar] [CrossRef]
  143. Zhang, X.; Zhao, W.; Wei, W.; You, Z.; Ou, X.; Sun, M.; Yin, Y.; Tang, X.; Zhao, Z.; Hu, C.; et al. Parallel Analyses of Somatic Mutations in Plasma Circulating Tumor DNA (ctDNA) and Matched Tumor Tissues in Early-Stage Breast Cancer. Clin. Cancer Res. 2019, 25, 6546–6553. [Google Scholar] [CrossRef] [Green Version]
  144. Rothe, F.; Silva, M.J.; Venet, D.; Campbell, C.; Bradburry, I.; Rouas, G.; de Azambuja, E.; Maetens, M.; Fumagalli, D.; Rodrik-Outmezguine, V.; et al. Circulating Tumor DNA in HER2-Amplified Breast Cancer: A Translational Research Substudy of the NeoALTTO Phase III Trial. Clin. Cancer Res. 2019, 25, 3581–3588. [Google Scholar] [CrossRef] [Green Version]
  145. Chen, Z.; Sun, T.; Yang, Z.; Zheng, Y.; Yu, R.; Wu, X.; Yan, J.; Shao, Y.W.; Shao, X.; Cao, W.; et al. Monitoring treatment efficacy and resistance in breast cancer patients via circulating tumor DNA genomic profiling. Mol. Genet. Genomic Med. 2020, 8, e1079. [Google Scholar] [CrossRef] [Green Version]
  146. Turner, N.C.; Kingston, B.; Kilburn, L.S.; Kernaghan, S.; Wardley, A.M.; Macpherson, I.R.; Baird, R.D.; Roylance, R.; Stephens, P.; Oikonomidou, O.; et al. Circulating tumour DNA analysis to direct therapy in advanced breast cancer (plasmaMATCH): A multicentre, multicohort, phase 2a, platform trial. Lancet Oncol. 2020, 21, 1296–1308. [Google Scholar] [CrossRef]
  147. Andre, F.; Ciruelos, E.; Rubovszky, G.; Campone, M.; Loibl, S.; Rugo, H.S.; Iwata, H.; Conte, P.; Mayer, I.A.; Kaufman, B.; et al. Alpelisib for PIK3CA-Mutated, Hormone Receptor-Positive Advanced Breast Cancer. N. Engl. J. Med. 2019, 380, 1929–1940. [Google Scholar] [CrossRef] [PubMed]
  148. Juric, D.; Ciruelos, E.; Rubovszky, G.; Campone, M.; Loibl, S.; Rugo, H.S.; Iwata, H.; Conte, P.; Mayer, I.A.; Kaufman, B.; et al. Abstract GS3-08: Alpelisib + fulvestrant for advanced breast cancer: Subgroup analyses from the phase III SOLAR-1 trial. Cancer Res. 2019, 79, GS3-08. [Google Scholar]
  149. Andre, F.; Ciruelos, E.M.; Juric, D.; Loibl, S.; Campone, M.; Mayer, I.A.; Rubovszky, G.; Yamashita, T.; Kaufman, B.; Lu, Y.S.; et al. Alpelisib plus fulvestrant for PIK3CA-mutated, hormone receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: Final overall survival results from SOLAR-1. Ann. Oncol. 2021, 32, 208–217. [Google Scholar] [CrossRef] [PubMed]
  150. Andre, F.; Su, F.; Solovieff, N.; Arteaga, C.L.; Hortobagyi, G.N.; Chia, S.K.L.; Neven, P.; Bardia, A.; Tripathy, D.; Lu, Y.S.; et al. Pooled ctDNA analysis of the MONALEESA (ML) phase III advanced breast cancer (ABC) trials. J. Clin. Oncol. 2020, 38, 1009. [Google Scholar] [CrossRef]
  151. Bidard, F.C.; Callens, C.; Dalenc, F.; Pistilli, B.; De La Motte Rouge, T.; Clatot, F.; D’hondt, V.; Teixeira, L.; Vegas, H.; Everhard, S.; et al. Prognostic impact of ESR1 mutations in ER+ HER2- MBC patients prior treated with first line AI and palbociclib: An exploratory analysis of the PADA-1 trial. J. Clin. Oncol. 2020, 38, 1010. [Google Scholar] [CrossRef]
  152. Shimomura, A.; Shiino, S.; Kawauchi, J.; Takizawa, S.; Sakamoto, H.; Matsuzaki, J.; Ono, M.; Takeshita, F.; Niida, S.; Shimizu, C.; et al. Novel combination of serum microRNA for detecting breast cancer in the early stage. Cancer Sci. 2016, 107, 326–334. [Google Scholar] [CrossRef] [PubMed]
  153. Cui, X.; Li, Z.; Zhao, Y.; Song, A.; Shi, Y.; Hai, X.; Zhu, W. Breast cancer identification via modeling of peripherally circulating miRNAs. PeerJ 2018, 6, e4551. [Google Scholar] [CrossRef] [PubMed]
  154. Huo, D.; Clayton, W.M.; Yoshimatsu, T.F.; Chen, J.; Olopade, O.I. Identification of a circulating microRNA signature to distinguish recurrence in breast cancer patients. Oncotarget 2016, 7, 55231–55248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Madhavan, D.; Peng, C.; Wallwiener, M.; Zucknick, M.; Nees, J.; Schott, S.; Rudolph, A.; Riethdorf, S.; Trumpp, A.; Pantel, K.; et al. Circulating miRNAs with prognostic value in metastatic breast cancer and for early detection of metastasis. Carcinogenesis 2016, 37, 461–470. [Google Scholar] [CrossRef] [Green Version]
  156. Hamam, R.; Hamam, D.; Alsaleh, K.A.; Kassem, M.; Zaher, W.; Alfayez, M.; Aldahmash, A.; Alajez, N.M. Circulating microRNAs in breast cancer: Novel diagnostic and prognostic biomarkers. Cell Death Dis. 2017, 8, e3045. [Google Scholar] [CrossRef] [Green Version]
  157. Cardoso, F.; Kyriakides, S.; Ohno, S.; Penault-Llorca, F.; Poortmans, P.; Rubio, I.T.; Zackrisson, S.; Senkus, E. Early breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-updagger. Ann. Oncol. 2019, 30, 1194–1220. [Google Scholar] [CrossRef] [Green Version]
  158. Ozawa, P.M.M.; Vieira, E.; Lemos, D.S.; Souza, I.L.M.; Zanata, S.M.; Pankievicz, V.C.; Tuleski, T.R.; Souza, E.M.; Wowk, P.F.; Urban, C.A.; et al. Identification of miRNAs Enriched in Extracellular Vesicles Derived from Serum Samples of Breast Cancer Patients. Biomolecules 2020, 10, 150. [Google Scholar] [CrossRef] [Green Version]
  159. Hirschfeld, M.; Rucker, G.; Weiss, D.; Berner, K.; Ritter, A.; Jager, M.; Erbes, T. Urinary Exosomal MicroRNAs as Potential Non-invasive Biomarkers in Breast Cancer Detection. Mol. Diagn Ther. 2020, 24, 215–232. [Google Scholar] [CrossRef]
  160. Zou, X.; Li, M.; Huang, Z.; Zhou, X.; Liu, Q.; Xia, T.; Zhu, W. Circulating miR-532-502 cluster derived from chromosome X as biomarkers for diagnosis of breast cancer. Gene 2020, 722, 144104. [Google Scholar] [CrossRef]
  161. Zou, X.; Xia, T.; Li, M.; Wang, T.; Liu, P.; Zhou, X.; Huang, Z.; Zhu, W. MicroRNA profiling in serum: Potential signatures for breast cancer diagnosis. Cancer Biomark 2021, 30, 41–53. [Google Scholar] [CrossRef]
  162. Zhong, G.; Wang, K.; Li, J.; Xiao, S.; Wei, W.; Liu, J. Determination of Serum Exosomal H19 as a Noninvasive Biomarker for Breast Cancer Diagnosis. Onco Targets Ther. 2020, 13, 2563–2571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Chanteloup, G.; Cordonnier, M.; Isambert, N.; Bertaut, A.; Hervieu, A.; Hennequin, A.; Luu, M.; Zanetta, S.; Coudert, B.; Bengrine, L.; et al. Monitoring HSP70 exosomes in cancer patients’ follow up: A clinical prospective pilot study. J. Extracell. Vesicles 2020, 9, 1766192. [Google Scholar] [CrossRef] [PubMed]
  164. Ortega, F.G.; Regiart, M.D.; Rodriguez-Martinez, A.; de Miguel-Perez, D.; Serrano, M.J.; Lorente, J.A.; Tortella, G.; Rubilar, O.; Sapag, K.; Bertotti, M.; et al. Sandwich-Type Electrochemical Paper-Based Immunosensor for Claudin 7 and CD81 Dual Determination on Extracellular Vesicles from Breast Cancer Patients. Anal. Chem. 2021, 93, 1143–1153. [Google Scholar] [CrossRef] [PubMed]
  165. Vinik, Y.; Ortega, F.G.; Mills, G.B.; Lu, Y.; Jurkowicz, M.; Halperin, S.; Aharoni, M.; Gutman, M.; Lev, S. Proteomic analysis of circulating extracellular vesicles identifies potential markers of breast cancer progression, recurrence, and response. Sci. Adv. 2020, 6, eaba5714. [Google Scholar] [CrossRef]
  166. Li, B.; Liu, C.; Pan, W.; Shen, J.; Guo, J.; Luo, T.; Feng, J.; Situ, B.; An, T.; Zhang, Y.; et al. Facile fluorescent aptasensor using aggregation-induced emission luminogens for exosomal proteins profiling towards liquid biopsy. Biosens Bioelectron 2020, 168, 112520. [Google Scholar] [CrossRef]
  167. Tian, F.; Zhang, S.; Liu, C.; Han, Z.; Liu, Y.; Deng, J.; Li, Y.; Wu, X.; Cai, L.; Qin, L.; et al. Protein analysis of extracellular vesicles to monitor and predict therapeutic response in metastatic breast cancer. Nat. Commun. 2021, 12, 2536. [Google Scholar] [CrossRef]
  168. Chaudhary, P.; Gibbs, L.D.; Maji, S.; Lewis, C.M.; Suzuki, S.; Vishwanatha, J.K. Serum exosomal-annexin A2 is associated with African-American triple-negative breast cancer and promotes angiogenesis. Breast Cancer Res. 2020, 22, 11. [Google Scholar] [CrossRef]
  169. Takeuchi, T.; Mori, K.; Sunayama, H.; Takano, E.; Kitayama, Y.; Shimizu, T.; Hirose, Y.; Inubushi, S.; Sasaki, R.; Tanino, H. Antibody-Conjugated Signaling Nanocavities Fabricated by Dynamic Molding for Detecting Cancers Using Small Extracellular Vesicle Markers from Tears. J. Am. Chem. Soc. 2020, 142, 6617–6624. [Google Scholar] [CrossRef]
  170. Rodriguez-Martinez, A.; de Miguel-Perez, D.; Ortega, F.G.; Garcia-Puche, J.L.; Robles-Fernandez, I.; Exposito, J.; Martorell-Marugan, J.; Carmona-Saez, P.; Garrido-Navas, M.D.C.; Rolfo, C.; et al. Exosomal miRNA profile as complementary tool in the diagnostic and prediction of treatment response in localized breast cancer under neoadjuvant chemotherapy. Breast Cancer Res. 2019, 21, 21. [Google Scholar] [CrossRef]
  171. Todorova, V.K.; Byrum, S.D.; Gies, A.J.; Haynie, C.; Smith, H.; Reyna, N.S.; Makhoul, I. Circulating Exosomal microRNAs as Predictive Biomarkers of Neoadjuvant Chemotherapy Response in Breast Cancer. Curr. Oncol. 2022, 29, 613–630. [Google Scholar] [CrossRef]
  172. Rothammer, A.; Sage, E.K.; Werner, C.; Combs, S.E.; Multhoff, G. Increased heat shock protein 70 (Hsp70) serum levels and low NK cell counts after radiotherapy-potential markers for predicting breast cancer recurrence? Radiat Oncol. 2019, 14, 78. [Google Scholar] [CrossRef] [PubMed]
  173. Del Re, M.; Bertolini, I.; Crucitta, S.; Fontanelli, L.; Rofi, E.; De Angelis, C.; Diodati, L.; Cavallero, D.; Gianfilippo, G.; Salvadori, B.; et al. Overexpression of TK1 and CDK9 in plasma-derived exosomes is associated with clinical resistance to CDK4/6 inhibitors in metastatic breast cancer patients. Breast Cancer Res. Treat. 2019, 178, 57–62. [Google Scholar] [CrossRef] [PubMed]
  174. Tang, S.; Zheng, K.; Tang, Y.; Li, Z.; Zou, T.; Liu, D. Overexpression of serum exosomal HOTAIR is correlated with poor survival and poor response to chemotherapy in breast cancer patients. J. Biosci. 2019, 44, 37. [Google Scholar] [CrossRef]
  175. Keklikoglou, I.; Cianciaruso, C.; Guc, E.; Squadrito, M.L.; Spring, L.M.; Tazzyman, S.; Lambein, L.; Poissonnier, A.; Ferraro, G.B.; Baer, C.; et al. Chemotherapy elicits pro-metastatic extracellular vesicles in breast cancer models. Nat. Cell Biol. 2019, 21, 190–202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Bartkowiak, K.; Heidrich, I.; Kwiatkowski, M.; Banys-Paluchowski, M.; Andreas, A.; Wurlitzer, M.; Geffken, M.; Voss, H.; Zeller, T.; Blankenberg, S.; et al. Circulating Cellular Communication Network Factor 1 Protein as a Sensitive Liquid Biopsy Marker for Early Detection of Breast Cancer. Clin. Chem. 2022, 68, 344–353. [Google Scholar] [CrossRef] [PubMed]
  177. 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.; et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N. Engl. J. Med. 2013, 368, 1199–1209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Garcia-Saenz, J.A.; Ayllon, P.; Laig, M.; Acosta-Eyzaguirre, D.; Garcia-Esquinas, M.; Montes, M.; Sanz, J.; Barquin, M.; Moreno, F.; Garcia-Barberan, V.; et al. Tumor burden monitoring using cell-free tumor DNA could be limited by tumor heterogeneity in advanced breast cancer and should be evaluated together with radiographic imaging. BMC Cancer 2017, 17, 210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Garcia-Murillas, I.; Schiavon, G.; Weigelt, B.; Ng, C.; Hrebien, S.; Cutts, R.J.; Cheang, M.; Osin, P.; Nerurkar, A.; Kozarewa, I.; et al. Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer. Sci. Transl. Med. 2015, 7, 302ra133. [Google Scholar] [CrossRef]
  180. Shoukry, M.; Broccard, S.; Kaplan, J.; Gabriel, E. The Emerging Role of Circulating Tumor DNA in the Management of Breast Cancer. Cancers 2021, 13, 3813. [Google Scholar] [CrossRef]
  181. Li, X.; Lu, J.; Zhang, L.; Luo, Y.; Zhao, Z.; Li, M. Clinical Implications of Monitoring ESR1 Mutations by Circulating Tumor DNA in Estrogen Receptor Positive Metastatic Breast Cancer: A Pilot Study. Transl. Oncol. 2020, 13, 321–328. [Google Scholar] [CrossRef]
  182. Yi, Z.; Ma, F.; Rong, G.; Guan, Y.; Li, C.; Xu, B. Clinical spectrum and prognostic value of TP53 mutations in circulating tumor DNA from breast cancer patients in China. Cancer Commun. 2020, 40, 260–269. [Google Scholar] [CrossRef] [PubMed]
  183. Matsuzaki, J.; Yamamoto, Y.; Yi, O.; Ayyar, S.; Miyajima, R.; Takeda, Y.; Timothy Nolan, T.; Kawai, N.; Kato, K.; Ota, N.; et al. Machine learning-based multiple cancer detections with circulating miRNA profiles in the blood. J. Clin. Oncol. 2021, 39, 3037. [Google Scholar] [CrossRef]
  184. Tellez-Gabriel, M.; Knutsen, E.; Perander, M. Current Status of Circulating Tumor Cells, Circulating Tumor DNA, and Exosomes in Breast Cancer Liquid Biopsies. Int. J. Mol. Sci. 2020, 21, 9457. [Google Scholar] [CrossRef] [PubMed]
  185. Lau, L.F. CCN1/CYR61: The very model of a modern matricellular protein. Cell Mol. Life Sci. 2011, 68, 3149–3163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Jun, J.I.; Lau, L.F. Taking aim at the extracellular matrix: CCN proteins as emerging therapeutic targets. Nat. Rev. Drug Discov. 2011, 10, 945–963. [Google Scholar] [CrossRef] [Green Version]
  187. Milbury, C.A.; Zhong, Q.; Lin, J.; Williams, M.; Olson, J.; Link, D.R.; Hutchison, B. Determining lower limits of detection of digital PCR assays for cancer-related gene mutations. Biomol. Detect. Quantif. 2014, 1, 8–22. [Google Scholar] [CrossRef] [Green Version]
  188. O’Leary, B.; Hrebien, S.; Beaney, M.; Fribbens, C.; Garcia-Murillas, I.; Jiang, J.; Li, Y.; Huang Bartlett, C.; Andre, F.; Loibl, S.; et al. Comparison of BEAMing and Droplet Digital PCR for Circulating Tumor DNA Analysis. Clin. Chem. 2019, 65, 1405–1413. [Google Scholar] [CrossRef]
  189. Koo, K.M.; Mainwaring, P.N. The role of circulating tumor DNA testing in breast cancer liquid biopsies: Getting ready for prime time. Breast Cancer Manag. 2020, 9, BMT34. [Google Scholar] [CrossRef] [Green Version]
  190. Hall, C.; Karhade, M.; Laubacher, B.; Anderson, A.; Kuerer, H.; DeSynder, S.; Lucci, A. Circulating Tumor Cells After Neoadjuvant Chemotherapy in Stage I-III Triple-Negative Breast Cancer. Ann. Surg. Oncol. 2015, 22 (Suppl. 3), S552–S558. [Google Scholar] [CrossRef]
  191. Kasimir-Bauer, S.; Bittner, A.K.; Konig, L.; Reiter, K.; Keller, T.; Kimmig, R.; Hoffmann, O. Does primary neoadjuvant systemic therapy eradicate minimal residual disease? Analysis of disseminated and circulating tumor cells before and after therapy. Breast Cancer Res. 2016, 18, 20. [Google Scholar] [CrossRef]
  192. Kwan, T.T.; Bardia, A.; Spring, L.M.; Giobbie-Hurder, A.; Kalinich, M.; Dubash, T.; Sundaresan, T.; Hong, X.; LiCausi, J.A.; Ho, U.; et al. A Digital RNA Signature of Circulating Tumor Cells Predicting Early Therapeutic Response in Localized and Metastatic Breast Cancer. Cancer Discov. 2018, 8, 1286–1299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Khosravi, F.; Trainor, P.J.; Lambert, C.; Kloecker, G.; Wickstrom, E.; Rai, S.N.; Panchapakesan, B. Static micro-array isolation, dynamic time series classification, capture and enumeration of spiked breast cancer cells in blood: The nanotube-CTC chip. Nanotechnology 2016, 27, 44LT03. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Loeian, M.S.; Mehdi Aghaei, S.; Farhadi, F.; Rai, V.; Yang, H.W.; Johnson, M.D.; Aqil, F.; Mandadi, M.; Rai, S.N.; Panchapakesan, B. Liquid biopsy using the nanotube-CTC-chip: Capture of invasive CTCs with high purity using preferential adherence in breast cancer patients. Lab Chip 2019, 19, 1899–1915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Abdulla, A.; Zhang, Z.; Ahmad, K.Z.; Warden, A.R.; Li, H.; Ding, X. Rapid and efficient capturing of circulating tumor cells from breast cancer Patient’s whole blood via the antibody functionalized microfluidic (AFM) chip. Biosens. Bioelectron. 2022, 201, 113965. [Google Scholar] [CrossRef] [PubMed]
  196. Heitzer, E.; Haque, I.S.; Roberts, C.E.S.; Speicher, M.R. Current and future perspectives of liquid biopsies in genomics-driven oncology. Nat. Rev. Genet. 2019, 20, 71–88. [Google Scholar] [CrossRef] [PubMed]
  197. Xie, S.; Wang, Y.; Gong, Z.; Li, Y.; Yang, W.; Liu, G.; Li, J.; Hu, X.; Wang, Y.; Tong, Y.; et al. Liquid Biopsy and Tissue Biopsy Comparison with Digital PCR and IHC/FISH for HER2 Amplification Detection in Breast Cancer Patients. J. Cancer 2022, 13, 744–751. [Google Scholar] [CrossRef] [PubMed]
  198. Freidin, M.B.; Freydina, D.V.; Leung, M.; Montero Fernandez, A.; Nicholson, A.G.; Lim, E. Circulating tumor DNA outperforms circulating tumor cells for KRAS mutation detection in thoracic malignancies. Clin. Chem. 2015, 61, 1299–1304. [Google Scholar] [CrossRef] [Green Version]
  199. Kodahl, A.R.; Ehmsen, S.; Pallisgaard, N.; Jylling, A.M.B.; Jensen, J.D.; Laenkholm, A.V.; Knoop, A.S.; Ditzel, H.J. Correlation between circulating cell-free PIK3CA tumor DNA levels and treatment response in patients with PIK3CA-mutated metastatic breast cancer. Mol. Oncol. 2018, 12, 925–935. [Google Scholar] [CrossRef] [Green Version]
  200. Nakai, M.; Yamada, T.; Sekiya, K.; Sato, A.; Hankyo, M.; Kuriyama, S.; Takahashi, G.; Kurita, T.; Yanagihara, K.; Yoshida, H.; et al. Use of Liquid Biopsy to Detect PIK3CA Mutation in Metastatic Breast Cancer. J. Nippon Med. Sch. 2022, 89, 66–71. [Google Scholar] [CrossRef]
  201. Suppan, C.; Graf, R.; Jahn, S.; Zhou, Q.; Klocker, E.V.; Bartsch, R.; Terbuch, A.; Kashofer, K.; Regitnig, P.; Lindenmann, J.; et al. Sensitive and robust liquid biopsy-based detection of PIK3CA mutations in hormone-receptor-positive metastatic breast cancer patients. Br. J. Cancer 2022, 126, 456–463. [Google Scholar] [CrossRef]
  202. Wan, J.C.M.; Heider, K.; Gale, D.; Murphy, S.; Fisher, E.; Mouliere, F.; Ruiz-Valdepenas, A.; Santonja, A.; Morris, J.; Chandrananda, D.; et al. ctDNA monitoring using patient-specific sequencing and integration of variant reads. Sci. Transl. Med. 2020, 12, eaaz8084. [Google Scholar] [CrossRef] [PubMed]
  203. Kim, J.; Park, J.; Ki, J.; Rho, H.W.; Huh, Y.M.; Kim, E.; Son, H.Y.; Haam, S. Simultaneous dual-targeted monitoring of breast cancer circulating miRNA via surface-enhanced Raman spectroscopy. Biosens. Bioelectron. 2022, 207, 114143. [Google Scholar] [CrossRef] [PubMed]
  204. Zhang, J.; Shi, J.; Zhang, H.; Zhu, Y.; Liu, W.; Zhang, K.; Zhang, Z. Localized fluorescent imaging of multiple proteins on individual extracellular vesicles using rolling circle amplification for cancer diagnosis. J. Extracell. Vesicles 2020, 10, e12025. [Google Scholar] [CrossRef] [PubMed]
  205. Welsh, J.A.; Tang, V.A.; van der Pol, E.; Gorgens, A. MIFlowCyt-EV: The Next Chapter in the Reporting and Reliability of Single Extracellular Vesicle Flow Cytometry Experiments. Cytometry A 2021, 99, 365–368. [Google Scholar] [CrossRef] [PubMed]
  206. Guo, S.C.; Tao, S.C.; Dawn, H. Microfluidics-based on-a-chip systems for isolating and analysing extracellular vesicles. J. Extracell. Vesicles 2018, 7, 1508271. [Google Scholar] [CrossRef] [Green Version]
  207. Trapp, E.; Janni, W.; Schindlbeck, C.; Juckstock, J.; Andergassen, U.; de Gregorio, A.; Alunni-Fabbroni, M.; Tzschaschel, M.; Polasik, A.; Koch, J.G.; et al. Presence of Circulating Tumor Cells in High-Risk Early Breast Cancer During Follow-Up and Prognosis. J. Natl. Cancer Inst. 2019, 111, 380–387. [Google Scholar] [CrossRef]
  208. Pantel, K.; Alix-Panabieres, C. Liquid biopsy and minimal residual disease—Latest advances and implications for cure. Nat. Rev. Clin. Oncol. 2019, 16, 409–424. [Google Scholar] [CrossRef]
  209. Stahlberg, A.; Krzyzanowski, P.M.; Egyud, M.; Filges, S.; Stein, L.; Godfrey, T.E. Simple multiplexed PCR-based barcoding of DNA for ultrasensitive mutation detection by next-generation sequencing. Nat. Protoc. 2017, 12, 664–682. [Google Scholar] [CrossRef]
  210. Ozawa, P.M.M.; Jucoski, T.S.; Vieira, E.; Carvalho, T.M.; Malheiros, D.; Ribeiro, E. Liquid biopsy for breast cancer using extracellular vesicles and cell-free microRNAs as biomarkers. Transl. Res. 2020, 223, 40–60. [Google Scholar] [CrossRef]
  211. Lee, I.; Baxter, D.; Lee, M.Y.; Scherler, K.; Wang, K. The Importance of Standardization on Analyzing Circulating RNA. Mol. Diagn Ther. 2017, 21, 259–268. [Google Scholar] [CrossRef]
  212. Cloet, T.; Momenbeitollahi, N.; Li, H. Recent advances on protein-based quantification of extracellular vesicles. Anal. Biochem. 2021, 622, 114168. [Google Scholar] [CrossRef] [PubMed]
  213. Malhotra, S.; Amin, Z.M.; Dobhal, G.; Cottam, S.; Nann, T.; Goreham, R.V. Novel devices for isolation and detection of bacterial and mammalian extracellular vesicles. Mikrochim. Acta 2021, 188, 139. [Google Scholar] [CrossRef] [PubMed]
  214. Seale, K.N.; Tkaczuk, K.H.R. Circulating Biomarkers in Breast Cancer. Clin. Breast Cancer 2021, 22, e319–e331. [Google Scholar] [CrossRef] [PubMed]
  215. Millner, L.M.; Linder, M.W.; Valdes, R., Jr. Circulating tumor cells: A review of present methods and the need to identify heterogeneous phenotypes. Ann. Clin. Lab Sci. 2013, 43, 295–304. [Google Scholar]
  216. Prieto-Garcia, E.; Diaz-Garcia, C.V.; Garcia-Ruiz, I.; Agullo-Ortuno, M.T. Epithelial-to-mesenchymal transition in tumor progression. Med. Oncol. 2017, 34, 122. [Google Scholar] [CrossRef] [PubMed]
  217. Nieto, M.A.; Huang, R.Y.; Jackson, R.A.; Thiery, J.P. Emt: 2016. Cell 2016, 166, 21–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Hyun, K.A.; Koo, G.B.; Han, H.; Sohn, J.; Choi, W.; Kim, S.I.; Jung, H.I.; Kim, Y.S. Epithelial-to-mesenchymal transition leads to loss of EpCAM and different physical properties in circulating tumor cells from metastatic breast cancer. Oncotarget 2016, 7, 24677–24687. [Google Scholar] [CrossRef] [Green Version]
  219. Lissa, D.; Robles, A.I. Methylation analyses in liquid biopsy. Transl. Lung Cancer Res. 2016, 5, 492–504. [Google Scholar] [CrossRef] [Green Version]
  220. Leary, R.J.; Sausen, M.; Kinde, I.; Papadopoulos, N.; Carpten, J.D.; Craig, D.; O’Shaughnessy, J.; Kinzler, K.W.; Parmigiani, G.; Vogelstein, B.; et al. Detection of chromosomal alterations in the circulation of cancer patients with whole-genome sequencing. Sci. Transl. Med. 2012, 4, 162ra154. [Google Scholar] [CrossRef] [Green Version]
  221. Matsutani, A.; Udagawa, C.; Matsunaga, Y.; Nakamura, S.; Zembutsu, H. Liquid biopsy for the detection of clinical biomarkers in early breast cancer: New insights and challenges. Pharmacogenomics 2020, 21, 359–367. [Google Scholar] [CrossRef]
  222. Diehl, F.; Schmidt, K.; Choti, M.A.; Romans, K.; Goodman, S.; Li, M.; Thornton, K.; Agrawal, N.; Sokoll, L.; Szabo, S.A.; et al. Circulating mutant DNA to assess tumor dynamics. Nat. Med. 2008, 14, 985–990. [Google Scholar] [CrossRef] [PubMed]
  223. Stewart, C.M.; Tsui, D.W.Y. Circulating cell-free DNA for non-invasive cancer management. Cancer Genet. 2018, 228–229, 169–179. [Google Scholar] [CrossRef]
  224. Stetson, D.; Ahmed, A.; Xu, X.; Nuttall, B.R.B.; Lubinski, T.J.; Johnson, J.H.; Barrett, J.C.; Dougherty, B.A. Orthogonal Comparison of Four Plasma NGS Tests With Tumor Suggests Technical Factors are a Major Source of Assay Discordance. JCO Precis. Oncol. 2019, 3, 1–9. [Google Scholar] [CrossRef] [PubMed]
  225. Steensma, D.P. Clinical Implications of Clonal Hematopoiesis. Mayo Clin. Proc. 2018, 93, 1122–1130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Bowman, R.L.; Busque, L.; Levine, R.L. Clonal Hematopoiesis and Evolution to Hematopoietic Malignancies. Cell Stem Cell 2018, 22, 157–170. [Google Scholar] [CrossRef] [Green Version]
  227. Chan, H.T.; Chin, Y.M.; Nakamura, Y.; Low, S.K. Clonal Hematopoiesis in Liquid Biopsy: From Biological Noise to Valuable Clinical Implications. Cancers 2020, 12, E2277. [Google Scholar] [CrossRef] [PubMed]
  228. Gandham, S.; Su, X.; Wood, J.; Nocera, A.L.; Alli, S.C.; Milane, L.; Zimmerman, A.; Amiji, M.; Ivanov, A.R. Technologies and Standardization in Research on Extracellular Vesicles. Trends Biotechnol. 2020, 38, 1066–1098. [Google Scholar] [CrossRef]
Figure 1. Comparison of liquid biopsy and tissue biopsy. Liquid biopsy is a minimally invasive method and relies on quantification of genetic materials derived from tumor cells and released into circulation, such as circulating tumor cells (CTCs), cell-free DNA (cfDNA)/circulating tumor DNA (ctDNA), circulating tumor RNA, extracellular vesicles (EVs), and circulating tumor proteins. Liquid biopsy allows for early diagnosis and screening, prediction of prognosis, early relapse detection in localized and locally advance breast cancer, minimal residual disease (MRD) identification, and longitudinal monitoring of the disease progression and treatment response. Therefore, liquid biopsy can be applied in as many time points as required during tumor progression and treatments, in order to detect recurrence and monitor response to treatment (green arrows). In contrast, tissue biopsy is an invasive procedure and not applicable for monitoring treatment response; subsequently, tissue biopsy is mainly applied at the time points for diagnosis and detection of recurrence during tumor progression (purple arrows).
Figure 1. Comparison of liquid biopsy and tissue biopsy. Liquid biopsy is a minimally invasive method and relies on quantification of genetic materials derived from tumor cells and released into circulation, such as circulating tumor cells (CTCs), cell-free DNA (cfDNA)/circulating tumor DNA (ctDNA), circulating tumor RNA, extracellular vesicles (EVs), and circulating tumor proteins. Liquid biopsy allows for early diagnosis and screening, prediction of prognosis, early relapse detection in localized and locally advance breast cancer, minimal residual disease (MRD) identification, and longitudinal monitoring of the disease progression and treatment response. Therefore, liquid biopsy can be applied in as many time points as required during tumor progression and treatments, in order to detect recurrence and monitor response to treatment (green arrows). In contrast, tissue biopsy is an invasive procedure and not applicable for monitoring treatment response; subsequently, tissue biopsy is mainly applied at the time points for diagnosis and detection of recurrence during tumor progression (purple arrows).
Cancers 14 02052 g001
Table 1. Biomarkers utilized for liquid biopsy.
Table 1. Biomarkers utilized for liquid biopsy.
BiomarkersClinical OutcomeSensitivity and SpecificityClinical TrialsReferences
CTC
For Prognosis
PD-L1 expression in CTCsPD-L1 expression in CTCs correlates with survival in metastatic breast cancer-A total of 72 patients with metastatic breast cancer
(prospective clinical trial (NCT02866149))
[139]
cfDNA/ctDNA
For diagnosis
ctDNA: PIK3CA and TP53Correlation between ctDNA detection with age, tumor grade and size, immunohistochemical subtype, BIRADS category, and lymph node positivity-A total of 29 patients[140]
ctDNA: the TP53, PIK3CA, and AKT1For the detection of early and advanced breast cancerctDNA detection rates: 37% for local or locally advanced breast cancer;
81% for metastatic or recurrent breast cancer
A total of 109 early and metastatic breast cancer patients[141]
ctDNA: SNPs in MDM2 and MDM4For the detection of early breast cancer-A total of 100 unrelated Lithuanian women[142]
For prognosis
ctDNA: a panel, based on COSMIC data, covering 136 genesServed as a predictor of worse prognosisPredictive value: 92%A total of 861 serial plasma and matched tissue specimens from 102 patients with early-stage breast cancer who need chemotherapy and 50 individuals with benign breast tumors[143]
ctDNA: PIK3CA and TP53 Absence of detectable PIK3CA and TP53 variants before neoadjuvant therapy was associated with high pCR rates-A total of 455 patients
(sub-study of the NeoALTTO phase 3 trial)
[144]
ctDNA panel: 488 mutationsDetecting MRD at 1-year postoperatively, which was positively associated with distant recurrenceSensitivity: 19%A total of 6 patients with ER+/HER2- metastatic breast cancer and 142 patients with stage 0 to III breast cancer[88]
ctDNA: TP53, PIK3CA, and DNA damage repair genesCorrelation between ctDNA profiling and therapeutic response and disease progression-A total of 19 HER2+ and 12 HER2- breast cancer patients[145]
For predicting treatment response
ctDNA: the PIK3CA, ESR1, HER2, PTEN, and AKT1Enables the selection of mutation-directed therapiesSensitivity: 93%A total of 1034 patients
(plasmaMATCH trial)
[146]
ctDNA: PIK3CA Treatment with alpelisib-fulvestrant prolonged progression-free survival among patients with PIK3CA-mutated, HR+, HER2- advanced breast cancer-A total of 572 patients
(341 patients with confirmed tumor-tissue PIK3CA mutations, SOLAR-1 trial)
[147,148,149]
ctDNA: AKT1, PIK3CA, ATM, TP53, ERB2, and ESR1Predict PFS in the treatment of paclitaxel and capivasertib-A total of 66 patients with ER+ metastatic breast cancer
(phase I/II BEECH trial)
[83]
ctDNA: FRS2, PRKCA, MDM2, ERB2, AKT1, and BRCA1/2Predicted a trend for increased PFS benefit of ribociclib treatment-A total of 1507 ER+ HER2- metastatic breast cancer patients
(MONALEESA 2-, 3-, and 7-trials)
[150]
ctDNA: ESR1 ESR1 mutations predicted significantly shorter PFS on treatment with aromatase inhibitors and palbociclib-A total of 1017 ER+ HER2- patients
(a large phase III PADA1 study)
[151]
cfDNA/ctDNA Methylation
For diagnosis
APC, FOXA1, and RASSF1AMethylation levels differed markedly in breast cancer patients in comparison to healthy controlsSensitivity: 81,82%
Specificity: 76,92%
A total of 137 cases of primary breast cancer tissues and 44 cases of plasma samples[60]
For prognosis
cfDNA methylation panel of five genes (KLK10, SOX17, WNT5A, MSH2, and GATA3)Methylation of ≥3 and ≥4 genes correlated to OS and no pharmacotherapy response, respectivelySensitivity: 80% specificity: 59%A total of 150 and 16 breast cancer patients under adjuvant and neoadjuvant therapy, respectively, 34 patients with metastatic disease and 35 healthy volunteers[19]
miRNA
For diagnosis
Combination of miR-1246, miR-1307-3p, miR-4634, miR-6861-5p, and miR-6875-5pDetect early breast cancerSensitivity: 97.6%
Specificity: 82.9%
The serum of 1280 patients with early breast cancer[152]
miR-1246, miR-6756-5p, and miR-8073For detection of breast cancer Sensitivity: 96.7%
Specificity: 97.2%
A total of 429 breast cancer patients and 895 healthy controls[153]
For prognosis
miR-21-5p, miR-194-5p, miR-205-5p, miR-375, miR-376c-3p, miR382-5p, and miR-411-5p Could be used as recurrence biomarkers for both hormonal positive and TNBC patientsSensitivity: 92.9%
Specificity: 77.4%
A total of 48 breast cancer patients[154]
A prognostic miRNA panel template (PROMPT): miRNAs, miR-141, miR-144, miR-193b, miR-200a, miR-200b, miR-200c, miR-203, miR-210, miR-215, miR-365, miR-375, miR-429, miR-486-5p, miR-801, miR-1260, and miR-1274a Associated with OS and RFSSensitivity: 77%
Specificity: 75%
A total of 237 metastatic breast cancer patients[155]
miR-21, miR-23b, miR-200c, and miR-190An increase in the expression of miR-21, miR-23b, and miR-200c, accompanied by a decrease in miR-190 in relapsed patients, compared to the non-relapsed onesSensitivity: 71.4%
Specificity 63.9%
A total of 49 relapsed and 84 non-relapsed localized breast cancer patients[18]
For predicting treatment response
miR-125bCorrelation between miR-125b and chemotherapeutic resistance--[156]
miR-155miR-155 serum levels decreased after surgery and four cycles of chemotherapy--[157]
EV
For diagnosis
Exosomoal miR-142-5p, miR-320a, and miR-4433b-5p For breast cancer diagnosisSensitivity: 93.33%
Specificity: 68.75%
A total of 31 breast cancer patients[158]
Exosomal miR-424, miR-423, miR-660, and let7-iFor breast cancer detectionSensitivity: 98.6%
Specificity: 100%
A total of 69 breast cancer patients and 40 healthy controls[159]
Exosomal miR-188-3p, miR-500a-5p, and miR-501-5p in plasma; exosomal miR-188-3p, miR-501-3p, miR-502-3p, miR-532-3p, and miR-532-5p in serum Upregulated in breast cancer patients-A total of 800 plasma and serum samples from breast cancer patients and healthy controls[160]
let-7b-5p, miR-106a-5p, miR-19a-3p, miR-19b-3p, miR-25-3p, miR-425-5p, miR-451a, miR-92a-3p, miR-93-5p, and miR-16-5pUpregulated in serum-derived exosomes in breast cancer patients, compared to controlsSpecificity: 94.9%
Sensitivity: 96.2%
A total of 32 pairs of breast cancer patients and healthy controls[161]
Exosomal lncRNA H19Exosomal levels of the lncRNA H19 were significantly higher in breast cancer patients than healthy controlsSensitivity: 87.0%
Specificity: 70.6%
A total of 50 patients[162]
Exosomal Hsp70Increased levels of exosomal Hsp70 in breast cancer patients, compared to healthy donors-A total of 40 patients and 14 healthy volunteers[163]
Claudin-7 and claudin-7/CD81 levels in EVs Claudin-7 might be a universal marker for the early diagnosis of breast cancerSensitivity: 95%
Specificity: 75.13%
A total of 60 breast cancer patients and 20 healthy volunteers[164]
Seven proteins (fibronectin, focal adhesion kinase 1 (FAK), dual-specificity mitogen-activated protein kinase kinase 1, β-Actin, p90RSK_pT573, N-cadherin, and proto-oncogene c-RAF) Distinguish patients (early patients accounted for nearly 70%) with breast cancer from healthy individualsSensitivity: 94%
Specificity: 82%
A total of 27 patients and 22 healthy controls[165]
EGFR in EVDiagnosing breast cancer patients with different clinical stages (I–IV)Sensitivity: 90%n = 49: 6 healthy control, 5 benign tumor, and 38 malignant tumor, including 13 with stage I, 14 with stage II, 5 with age III, 2 with stage IV, 4 without stage information[166]
Eight plasma EV protein markers (mucin-1, CA-125, carcinoembryonic antigen, HER2, EGFR, PSMA, EpCAM, and VEGF) Distinguish among metastatic breast cancer, nonmetastatic breast cancer, and healthy donorsOverall accuracy: 91.1%A total of 220 plasma samples from breast cancer patients[167]
Exosomal AnxA2Higher expression of serum exosomal AnxA2 in breast cancer patients compared to non-cancer females;
high expression of exosomal AnxA2 levels in was significantly associated with poor overall survival and poor disease-free survival
-A total of 169 breast cancer patients and 68 non-cancer females[168]
γ-glutamyltransferase 1 in EVsPatients with breast cancer had enhanced γ-glutamyltransferase 1 detection signals than those of healthy donors-Patients with breast cancer (five cases) and healthy donors (five cases)[169]
For prognosis
miR-21 and miR-105miR-21 and miR-105 were overexpressed in metastatic patients, compared to non-metastatic ones, as well as controls-A total of 53 patients[170]
Exosomal miR-30b, miR-328, and miR-423 Predicted pCR-A total of 20 breast cancer patients[171]
Heat shock protein 70 in small EVs Elevated in patients with recurrence or metastasis-
  • 27 patients;
  • Serum of 40 breast cancer patients
[165,172]
For predicting treatment response
Exosomal mRNAs encoding TK1 and CDK9 Elevated exosomal levels of mRNAs encoding TK1 and CDK9 were associated with poor clinical response to the CDK4/CDK6 inhibitor palbociclib--[173]
lncRNA HOTAIRPossible predictor of response to chemotherapy and tamoxifen treatment-A total of 15 breast cancer patients treated surgically, 15 healthy individuals, 25 patients received neoadjuvant chemotherapy before surgery, and 25 patients received tamoxifen
hormone treatment after surgery
[174]
ANXA6 in plasma EVs Reflect treatment response of neo-adjuvant treatment--[175]
Protein
CCN1For early cancer detectionSpecificity: 99.0%
Sensitivity: 80.0%
A total of 544 patients with breast cancer and 427 healthy controls[176]
Table 2. Detection techniques for liquid biopsy.
Table 2. Detection techniques for liquid biopsy.
Detection TechniquesTargetAdvantagesReferences
For CTC Detection
CellSearch®CTCs immunoisolation by positive selection targeting EpCAMGold standard and the only technique approved by the FDA for the isolation and detection of CTCs in metastatic breast, prostate, and colon cancer[37,190]
Adnatest (QIAGEN®)A combination of antibodies conjugated with magnetic beads for selecting tumor and epithelial markers and an RT-PCR for detecting breast cancer mRNAs biomarkersIsolate CTCs in the breast cancer neoadjuvant setting[191]
CTC-iChipa digital RNA signatureFor CTC isolation and detection in early and metastatic breast cancer patients[192]
Nanotube-CTC-chipBreast cancer-specific antibodies, such as anti-EpCAM and anti-her2Identify CTCs in the 100% of the studied breast cancer peripheral blood samples[193,194]
AFM chipEpCAM, CK19, CD45, and DAPIHighly efficient at rapidly capturing CTCs from cancer patients’ whole blood, without requiring extra equipment[195]
For cfDNA detection
The Oncomine Breast cfDNA (Thermofisher, Waltham, MA, USA) testDNADetect mutations in a limited number of genes from breast cancer patients[196]
dPCRcfDNA: HER2Could be used as a companion diagnostic tool to detect plasma HER2 status[197]
For ctDNA detection
ddPCR and the BEAMing technology PIK3CA mutations in plasma ctDNA from advanced breast cancer patientsAllow absolute quantification of allele frequencies as low as 0.01%[198,199,200]
PIK3CA RGQ PCR Kit 11 mutations in the PIK3CA gene from patients with advanced or metastatic breast cancer May help doctors identify
breast cancer patients who should be treated with PIQRAY®
[147]
NGS-based ctDNA test, Signatera™ctDNAFor the detection of MRD after surgery and earlier detection of disease recurrence[73]
TARDIS of ctDNAMultiple tumor mutations in ctDNAHighly sensitive method combining a targeted linear pre-amplification, followed by unique molecular identifiers (UMIs) ligation, targeted exponential PCR, and ultra-deep sequencing[89]
SiMSen-Seq assayPIK3CA mutations in ctDNAAllows detection of extremely rare variant alleles at <0.1% frequency and shows advantageous concordance with the tissue analyses[201]
INtegration of VAriant Reads (INVAR) ctDNA detection; up to a thousand loci for mutationsAs little as one mutant molecule per 100,000 can be detected, thus significantly increasing the ctDNA detection sensitivity[202]
For miRNA detection
SERS with SMGAPs miR-21 and miR-155SERS gives information for trace amount of material[203]
For protein detection
Localized fluorescent-imaging method Multiple proteins on individual EVsEnables the detection of multiple proteins on individual EVs[204]
High-resolution flow cytometryProteins on EVsImprove reporting and reliability of single EV flow cytometry experiments[205]
Microfluidic devicesProteins on EVsAchieve higher specificity and sensitivity[206]
The aptasensor methodProteins on EVsPre-separation of EVs is not needed, the total detection time is short (within 3 h), and it has a low cost (less than $1)[167]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wu, H.-J.; Chu, P.-Y. Current and Developing Liquid Biopsy Techniques for Breast Cancer. Cancers 2022, 14, 2052. https://doi.org/10.3390/cancers14092052

AMA Style

Wu H-J, Chu P-Y. Current and Developing Liquid Biopsy Techniques for Breast Cancer. Cancers. 2022; 14(9):2052. https://doi.org/10.3390/cancers14092052

Chicago/Turabian Style

Wu, Hsing-Ju, and Pei-Yi Chu. 2022. "Current and Developing Liquid Biopsy Techniques for Breast Cancer" Cancers 14, no. 9: 2052. https://doi.org/10.3390/cancers14092052

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop