Skip to main content
Molecular Oncology logoLink to Molecular Oncology
. 2024 Jun 12;19(7):1897–1917. doi: 10.1002/1878-0261.13671

Clinical applications of next‐generation sequencing‐based ctDNA analyses in breast cancer: defining treatment targets and dynamic changes during disease progression

Eva Valentina Klocker 1, Samantha Hasenleithner 2, Rupert Bartsch 3, Simon P Gampenrieder 4, Daniel Egle 5, Christian F Singer 6, Gabriel Rinnerthaler 1, Michael Hubalek 7, Katja Schmitz 8,9, Zsuzsanna Bago‐Horvath 10, Andreas Petzer 11, Sonja Heibl 12, Ellen Heitzer 13, Marija Balic 1,14,, Michael Gnant 15
PMCID: PMC12234384  PMID: 38867388

Abstract

The advancements in the detection and characterization of circulating tumor DNA (ctDNA) have revolutionized precision medicine and are likely to transform standard clinical practice. The non‐invasive nature of this approach allows for molecular profiling of the entire tumor entity, while also enabling real‐time monitoring of the effectiveness of cancer therapies as well as the identification of resistance mechanisms to guide targeted therapy. Although the field of ctDNA studies offers a wide range of applications, including in early disease, in this review we mainly focus on the role of ctDNA in the dynamic molecular characterization of unresectable locally advanced and metastatic BC (mBC). Here, we provide clinical practice guidance for the rapidly evolving field of molecular profiling of mBC, outlining the current landscape of liquid biopsy applications and how to choose the right ctDNA assay. Additionally, we underline the importance of exploring the clinical relevance of novel molecular alterations that potentially represent therapeutic targets in mBC, along with mutations where targeted therapy is already approved. Finally, we present a potential roadmap for integrating ctDNA analysis into clinical practice.

Keywords: breast cancer, circulating tumor DNA, liquid biopsy, precision medicine, targeted therapies, therapeutic targets


Circulating tumor DNA (ctDNA) offers a possibility for different applications in early and late stage breast cancer management. In early breast cancer tumor informed approaches are increasingly used for detecting molecular residual disease (MRD) and early recurrence. In advanced stage, ctDNA provides a possibility for monitoring disease progression and identifying resistance mechanisms. Figure created with BioRender.com.

graphic file with name MOL2-19-1897-g002.jpg


Abbreviations

AI

aromatase inhibitor

ASCO

American Society of Clinical Oncology

CDK4/6

cyclin‐dependent kinase 4/6

cfDNA

cell‐free DNA

CGP

comprehensive genomic profiling

ctDNA

circulating tumor DNA

ddPCR

digital droplet PCR

ER

estrogen receptor

ET

endocrine therapy

gBRCA1/2m

germline BRCA1/2 mutations

HER2

human epidermal growth factor receptor 2

HR

hormone receptor

ICI

immune checkpoint inhibitor

mBC

metastatic breast cancer

mFAST‐SeqS

modified Fast Aneuploidy Screening Test‐Sequencing System

MRD

molecular residual disease

MSI

microsatellite instability

NCCN

National Comprehensive Cancer Network

NGS

next‐generation sequencing

ngSERD

next‐generation oral selective estrogen receptor antagonist and degrader

PARPi

poly ADP‐ribose polymerase inhibitor

pCR

pathological complete response

PFS

progression‐free survival

PR

progesterone receptor

RCB

Residual Cancer Burden

Safe‐SeqS

Safe‐Sequencing System

sBRCA1/2m

somatic BRCA1/2 mutations

SERD

selective estrogen receptor degrader

SiMSen‐seq

simple, multiplexed, PCR‐based barcoding of DNA for sensitive mutation detection using Sequencing

SOC

standard of care

Tam‐Seq

tagged‐amplicon deep sequencing

TMB

tumor mutational burden

TNBC

triple‐negative breast cancer

TKI

tyrosine kinase inhibitor

VAF

variant allele fraction

WES

whole‐exome sequencing

WGS

whole‐genome sequencing

1. Introduction

The contemporary goal of precision medicine in patients with cancer is to identify biologically sensitive groups of tumors that will respond to targeted therapies. In this context, genomic sequencing of specific molecular alterations is increasingly performed in tissue samples, although tumor heterogeneity – both spatial and temporal – represents a major limitation of this approach [1]. An emerging alternative for capturing spatial sub‐clonal heterogeneity and clonal evolution during treatment, that allows for therapeutic clinical decision making, is the detection of circulating tumor DNA (ctDNA) via non‐invasive liquid biopsy [2, 3, 4].

Treatment choices in patients with breast cancer have historically been based on human epidermal growth factor receptor 2 (HER2) and hormone receptor (HR) status [5]. In the neoadjuvant setting, a major challenge is the prediction of response to systemic treatment before surgery in order to escalate treatment in non‐responders and de‐escalate treatment in responders, as appropriate [6, 7, 8, 9, 10]. To date, neither imaging methods including ultrasound [11] or MRI [12] nor image‐guided breast biopsies before surgery [13, 14, 15] have been able to adequately predict pathologic complete response (pCR). For this purpose, serial ctDNA analyses could provide a new approach to assess or predict tumor response early on during neoadjuvant treatment, thus ultimately guiding treatment decisions [10].

In metastatic BC (mBC), classical biomarkers such as HR and HER2 status are traditionally important for treatment decisions. Therefore, this review summarizes the status quo of the dynamic molecular characterization of mBC, especially in HR‐positive, HER2‐negative disease [16]. Moreover, we provide clinical practice guidance for the rapidly evolving field of molecular profiling of mBC and underline the importance of exploring the clinical relevance of novel molecular alterations that potentially represent therapeutic targets in mBC.

2. Tumor biology evolves over time

According to GLOBOCAN, in 2020, female breast cancer was the most commonly diagnosed cancer worldwide, with an estimated 2.3 million new cases and 685 000 deaths [17]. While breast cancer is mostly diagnosed at an early stage and associated with a 96% 5‐year survival probability in Europe [18], mBC still largely represents an incurable disease [19, 20]. Breast cancer can be subclassified into five intrinsic subtypes, that is, luminal A, luminal B, HER2‐enriched, basal, and normal‐like, which reflect fundamental differences at the molecular level and thus distinct clinical outcomes, mainly in patients with early breast cancer [21, 22, 23, 24].

Tumor (tissue) biopsy still represents the gold standard for diagnosis, classification, and treatment decisions. However, studies have found changes in subtypes at or after progression on anticancer therapies in up to 40% of tumors [25, 26, 27, 28, 29, 30, 31, 32, 33]. Importantly, beyond the HR and/or HER2 status detected in tissue, liquid biopsy provides a new concept to characterize and monitor the tumor genome and is increasingly being used as a tool to further guide clinical decision making [25, 29, 34, 35, 36].

After the diagnosis of metachronous metastatic (metastatic spread after 3 months after initial diagnosis) disease, patients often undergo initial confirmatory tissue biopsy including reassessment of the receptor subtype classification, which appears necessary due to a high rate of changes in HR and HER2 receptor status [25, 26, 27, 28, 29, 30, 31, 32, 33]. In addition, mBC could display major genomic differences compared to primary breast cancer as reported in sequencing studies of HR‐positive mBC [37]. In this context, the AURORA trial showed enriched alterations in several driver genes from metastatic lesions, including: ESR1, PTEN, CDH1, PIK3CA and RB1 mutations, MDM4 and MYC amplifications, ARID1A deletions, and clonality in genes like RB1 and ERBB2. Additionally, high tumor mutational burden (TMB) was reported to correlate with shorter time to relapse in patients with HR‐positive/HER2‐negative mBC [29, 38]. Furthermore, in HR‐positive mBC, mechanisms of acquired resistance to prior endocrine therapy (ET) were linked to ESR1 [39], MAPK pathway mutations [40, 41], and transcription factor mutations (e.g., ARID1A [40, 42, 43]). In contrast, in HER2‐positive and TNBC, no major genomic differences have been reported in primary vs. advanced settings [37, 40].

Although tissue may be analyzed using next‐generation sequencing (NGS), serial tissue biopsies are not routinely performed, given their invasiveness as well as anatomical constraints (e.g., bone metastasis). This further highlights the relevance of ctDNA analysis via NGS as an alternative. In this context, the phase 2a plasmaMATCH trial has paved the way for future personalized treatment approaches, since patients were selected for specific treatments based on the detection of mutations in ctDNA. Here, the sensitivity and specificity were comparable between tissue and ctDNA analysis for AKT1, HER2, and PIK3CA mutations [44]. Recently, we reported that elevated longitudinal trajectories of tumor fractions, as detected by the untargeted modified Fast Aneuploidy Screening Test‐Sequencing System (mFAST‐SeqS), were significantly associated with a higher progression risk in patients with HR‐positive/HER2‐negative mBC. The tumor agnostic, mFAST‐SeqS approach represents a simple and affordable method to estimate tumor fractions, that is, the proportion of cell‐free DNA (cfDNA) that is tumor‐derived ctDNA, by assessing chromosomal aneuploidy instead of mutations. After amplification and sequencing of uniquely mappable LINE1‐sequences across the genome, read counts are determined on a chromosome‐arm level and compared to a set of healthy controls. Deviations are represented as z‐scores and indicate gains and losses of chromosomal material. Finally, the squared sum of chromosome‐arm z‐scores is calculated, that is, the genome‐wide z‐score, which can be used as a surrogate for tumor fraction [45]. We have shown a very good correlation of mFAST‐SeqS based z‐scores with ‘ichorCNA’ tumor fractions [46]. Patients who developed progressive disease had higher baseline tumor fractions and showed constant increases over time. Furthermore, we have demonstrated the added value of evaluation of an untargeted assessment of tumor fractions along with cfDNA‐based mutational profiling [47].

3. Liquid biopsy

The broad concept of liquid biopsy encompasses the analysis of circulating nucleic acids, tumor cells, cell‐free RNA, extracellular vesicles, and tumor‐educated platelets that are released by primary or metastatic tumor lesions into the bloodstream or other body fluids. Thus, liquid biopsy represents a simple, non‐invasive method (e.g., based on blood sampling) providing a molecular footprint of the whole tumor entity [28, 35, 48, 49, 50, 51].

A number of processes involved in regular cell turnover, for example, necrosis or apoptosis, result in the release of cfDNA, whereas senescence prevents it [52, 53, 54, 55]. In the setting of cancer, a certain proportion of cfDNA is derived from tumor cells (i.e., primary and secondary tumor sites) and contains the full set of its genetic information. Thus, it is referred to as ctDNA [48, 49, 50, 51].

The amount of ctDNA ranges from 0.01% to 0.1% in early‐stage to 5–10% or even higher in advanced‐stage cancers where cancer cells are more abundant and undergoing more rapid cell division [51, 56]. While both cfDNA and ctDNA can include fragments of similar sizes, ctDNA fragments often exhibit a more pronounced peak at shorter lengths or a more diverse size distribution [57]. ctDNA varies with respect to fragment length, numbers, and variant allele frequency (VAF) of alterations and has a very short half‐life of < 120 min [58], despite the lack of comprehensive pharmacokinetic studies to accurately establish this. ctDNA assessment allows for a comprehensive genomic snapshot of the tumor mutational content, even in cases of tumor heterogeneity and cell plasticity during cancer progression [58, 59, 60]. The ctDNA dynamics along the clinical course of breast cancer including appropriate ctDNA assays and their sensitivity are depicted in Fig. 1.

Fig. 1.

Fig. 1

ctDNA dynamics along the clinical course of breast cancer. The blue line represents ctDNA variations over disease progression and the orange line represents the emergence of new mutations during treatment. ddPCR, digital droplet PCR; MRD, molecular residual disease; NGS, next‐generation sequencing; qPCR, quantitative PCR; WES, whole‐exome sequencing; WGS, whole‐genome sequencing. Adapted from [142]. Figure created with BioRender.com.

4. Preanalytical and analytical considerations

Plasma seems to be the ideal analyte for ctDNA collection and diagnostic purposes in solid tumors, including breast cancer [48, 49, 50]. There is a consensus to use specialized collection tubes, for example, PAXgene Blood circulating cfDNA tubes or those from commercial providers that contain an additive to stabilize blood cells, thus preventing cell lysis, particularly if plasma extraction cannot be done with 4–6 h after blood draw. After plasma separation, DNA extraction is performed using commercial kits specifically designed for cfDNA, and finally, the amount of cfDNA is quantified [61]. Importantly, the selection of the approach to library preparation dictates the final type of information obtained from the analysis.

Although there are several targeted sequencing approaches available, the most common ones are amplicon‐based (i.e., tagged‐amplicon deep sequencing (TAm‐Seq), Safe‐Sequencing System (Safe‐SeqS), and simple, multiplexed, PCR‐based barcoding of DNA for sensitive mutation detection using Sequencing (SiMSen‐seq)), and hybrid capture‐based (i.e., pan‐cancer ctDNA expanded liquid biopsy panel) methods. Briefly, the PCR amplicon‐based method uses specific primers to amplify and enrich only target genomic regions of interest. After amplification, sequencing adapters and barcode indexes are added to create the library for sequencing. In the hybrid capture method, genomic DNA is first fragmented followed by ligation of adapters and barcode indices to generate a genomic library which is further subjected to hybridization using long, biotinylated specific oligonucleotide baits (probes). The target sequences are captured using streptavidin magnetic beads followed by elution of the library which is further subjected to PCR amplification and deep sequencing [61, 62]. Overall, amplicon‐based sequencing requires lower amounts of DNA input, is generally more cost‐effective, and has higher on‐target rates. As a result, it is more often used in small‐scale experiments. In contrast, hybridization capture provides more uniform coverage of on‐target reads. The longer bait sequences allow for a greater level of specificity in region selection. Furthermore, it produces fewer PCR duplicates, making it particularly useful for samples where PCR artifacts are likely to occur, such as ctDNA samples. Therefore, it is the preferred methodology for large panels and whole‐exome sequencing [62, 63].

5. Choosing the right NGS‐based ctDNA assay

The choice of the right assay, which may be based on PCR, ddPCR, or NGS, first depends on the clinical question at hand, what type of information needs to be harvested from the sample, and the disease stage of the patient. In terms of NGS‐based approaches, the decision generally relies on the distinction between a targeted approach that focuses on particular predefined regions of the genome and an untargeted approach, such as whole‐exome sequencing (WES), which targets all human protein coding genes, or whole‐genome sequencing (WGS). Moreover, these assays can either be personalized based on existing sequencing data or tumor‐agnostic, which means that the assessment is performed without any prior knowledge of the tumor genome (or in the absence of any baseline data) [61].

In the setting of early disease and MRD (molecular residual disease) monitoring, where the likelihood of detecting ctDNA fragments must be maximized, tumor‐informed NGS assays designed to track patient‐specific mutations longitudinally are typically used, as they can achieve the required sensitivities to detect minute traces of ctDNA [64, 65, 66]. This approach entails WES or WGS of a baseline tissue or plasma sample and then subsequently designing patient‐specific panels to screen for dozens or up to thousands of mutations. These approaches enable the detection of ctDNA at levels below one part‐per‐million (< 0.0001%) (ppm) [66]. However, a variety of tumor‐agnostic approaches employing multi‐omics methods, such as ctDNA methylation or fragment‐omics, have also demonstrated promising performances, and many more are in development [67].

For patients with advanced cancer, the most predominant current clinical use case for ctDNA analysis is molecular profiling for the selection of targeted treatments. In this regard, the strategy is to maximize the identification of actionable alterations that can be matched to targeted therapies. Typically, pan‐cancer comprehensive genomic profiling (CGP) panels are employed to enable the detection of many clinically relevant biomarkers [61].

There are several commercially available kits enriching for tens or hundreds of genes. For example, the AVENIO ctDNA Expanded kit, that is based on the Cancer Personalized Profiling by deep Sequencing (CAPP‐Seq) [68], contains 77 genes, of which 17 biomarkers are recommended by the National Comprehensive Cancer Network (NCCN) guidelines and 60 biomarkers are currently under investigation in clinical trials. Other panels, such as the TruSight Oncology 500 ctDNA which enables in‐house CGP of ctDNA and targets 523 genes to assess DNA variants across major variant classes (SNV, MNV, indels, CNV, and gene rearrangements), also include the assessment of TMB or microsatellite instability (MSI). If there are no in‐house partner laboratories that carry out such analyses, samples can be sent to a commercial end‐to‐end provider to provide the molecular tumor board with all relevant information [61].

It is also worth mentioning that other ctDNA‐based approaches may have specific applications that do not require NGS. For example, the Therascreen® PIK3CA RGQ PCR kit, which received FDA approval for advanced‐stage HR‐positive/HER2 ‐negative breast cancer based on the findings of the phase III SOLAR‐1 trial, is a real‐time PCR test that screens for 11 therapy‐relevant PIK3CA mutations from plasma [69]. Tables 1, 2, 3 summarize available ctDNA assays to direct therapy in advanced disease, for MRD detection and early detection, respectively, while Fig. 2 depicts ctDNA analytical approaches.

Table 1.

Examples of commercially available ctDNA assays to direct therapy in advanced disease. WBC, white blood cells.

Test name Company FDA approval as companion diagnostic Number of genes in panel Complex biomarkers included Trials performed with the assay
Foundation Liquid CDx Foundation Medicine Yes 324 Yes

IMAGE trial

mTNBC, ≥ 1 treatment lines, genomic driven therapies, NGS in tissue vs. plasma [76]

Guardant360 CDx Guardant Health Yes 74 Yes

mBC, ctDNA

ESR1 status for treatment with Elacestrant (Emerald trial [77])

ESR1, HER2, AKT1, PTEN mutations for targeted therapies (plasmaMATCH [78])

PGDx elio Plasma Resolve Labcorp No, but has received Breakthrough Device Designation 33 No Metastatic colorectal cancer, ctDNA in combination with WBC for treatment response monitoring [79]
PGDx elio Plasma Complete Labcorp 521 Yes
Tempus xF+ Tempus No 523 Yes mBC NGS is tissue vs. ctDNA [80]
Circulogene TumorClear Circulogene No 88 Yes

Table 2.

Examples of commercially available ctDNA assays for MRD detection. Adapted from [74].

Test name Company FDA approval Tumor types Details Trials
RaDAR NeoGenomics No, but received Breakthrough Device Designation Breast, colon, lung, head & neck Tumor whole‐exome sequencing (WES) and monitoring of up to 48 tumor/patient‐specific mutations

High‐stage II‐III HR‐positive

mBC, ctDNA to detect MRD and development of metastasis [81]

Guardant Reveal Guardant Health No Early‐stage (II&III) CRC Tumor‐uninformed approach, integrates somatic alterations with an epigenomic cancer signature to identify the presence of methylation signatures associated with cancer vs. normal DNA

Colorectal Cancer

To detect MRD and correlation with recurrence [82]

brPROPHET™ Burning Rock Biotech No Early‐stage CRC Tumor WES followed by customized design of a patient‐specific panel consisting of 50 single‐nucleotide variants

Colorectal Cancer

Stages I–IV, detection of MRD [83]

PCM ArcherDx/Invitae No NSCLC Tumor WES followed by customized design of a patient‐specific panel consisting of 50 single‐nucleotide variants

NSCLC

Stages I–III

Detection of MRD and correlation with relapse [84]

Signatera™ Residual disease test (MRD) Natera™ No, but received Breakthrough Device Designation Colorectal, NSCLC, breast, and bladder cancer

Tumor WES of tissue sample and matched normal and selection of 16 patient‐specific, clonal somatic variants for design of custom 16‐plex PCR followed by NGS to monitor ctDNA longitudinally.

A positive Signatera™ result predicts relapse with overall positive predictive value more than 98%

High‐risk early stage HER2‐ negative BC

Detection of ctDNA clearance under neoadjuvant treatment, correlation of response, lack of ctDNA clearance was associated with reduced distant free survival [85]

Haystack MRD™ Quest Diagnostics™ No Solid tumors Tumor WES to identify individual‐specific mutations and selection of up to 50 to personalize the subsequent MRD assay.

Colorectal Cancer, Stage II

1 : 2 randomization

Intervention cohort: treatment was only given when positive ctDNA at week 4 or 7 was detected, recurrence‐free survival was not inferior to standard cohort [86]

Table 3.

Examples of commercially available ctDNA assays for early cancer detection.

Test name Company FDA approval Tumor types Details
CancerSEEK Exact Sciences No Multiple A multi‐biomarker class (mutations, methylation, proteins, and aneuploidy) and machine‐learning approach. CancerSEEK was used in DETECT‐A study [87]. Test is under further development. The features above describe current development goals
Epi proColon Epigenomics Yes Colorectal cancer Real‐time PCR detection of Septin9 methylation, which is altered in colorectal cancer tumor cells more often than in normal cells [88]
Epi proLung Epigenomics No Lung cancer Real‐time PCR assay for the qualitative detection of SHOX2 and PTGER4 gene methylation in DNA [89]
Shield Guardant Health No, but submitted application for Pre‐Market Approval Colorectal cancer Uses a multimodal approach, integrating genomics, epigenomics, and proteomics, to achieve high sensitivity and specificity in detecting early signs of colorectal cancer in average‐risk adults aged 45 and older (ECLIPSE: NCT04136002)
Galleri® GRAIL No Multi‐cancer (50+ types) Plasma cfDNA bisulfite sequencing targeting a panel of > 100 000 informative methylation regions. Cancer detection and tissue of origin (TOO) localization through a proprietary classifier. Recently tested in the large‐scale SYMPLIFY study in a symptomatic patient population [90]

Fig. 2.

Fig. 2

Tumor‐informed vs. tumor‐naive approaches. This figure gives an overview of strategies for liquid biopsy based in clinical practice. In orange the applications of tumor‐informed assays are highlighted, in green the use of tumor‐naïve assays is shown. Adapted from [143, 144]. Figure created with BioRender.com.

6. Early breast cancer – risk stratification and early assessment of disease progression

In patients with early breast cancer, ctDNA levels were shown to be elevated before and after surgery compared to those with benign breast lesions, and ctDNA levels correlated with nodal involvement and tumor size [82]. Additionally, detection of ctDNA during follow‐up in patients with early breast cancer was shown to be associated with a high risk of future relapse [83]. In terms of response rates to neoadjuvant systemic treatment and pCR, studies have shown lower rates of pCR in patients where ctDNA was detected before the administration of neoadjuvant anti‐HER2 therapies [84]. In a subanalysis of the NEOALLTO trial, ctDNA detection before neoadjuvant treatment was significantly associated with older age and ER negativity. The lowest pCR rate was detected in patients with detectable ctDNA before and during neoadjuvant treatment, whereas during neoadjuvant treatment, a rapid decrease in ctDNA levels (ctDNA clearance) was linked to response like pCR [85, 86]. Moreover, the detection and persistence of ctDNA during neoadjuvant systemic treatment was shown to predict poor response [10] and metastatic recurrence [87]. Presence of ctDNA towards the end of treatment is expected to reflect residual disease [74]. Magbanua et al. [76] evaluated ctDNA clearance under neoadjuvant treatment in 295 patients with early disease with the Signatera™ residual disease assay, a personalized tumor‐informed ctDNA test. Early ctDNA clearance was significantly associated to Residual Cancer Burden (RCB) 0 and pCR, respectively, in triple negative breast cancer (TNBC) but not for HR‐positive and HER2‐positive tumors. ctDNA persistence at the time of surgery was significantly associated with increased risk of relapse and worse overall survival. However, the data demonstrate that particularly in HR‐positive patients more sensitive tests are needed to decrease the false‐negative cases. Further clinical data are needed to establish the value of ctDNA in these groups. Nevertheless, this trial is so far the largest neoadjuvant trial demonstrating the potential of ctDNA testing at an early neoadjuvant stage. Another trial by Parson et al. [70] enrolled solely TNBC and demonstrated that tumor‐informed clearance of ctDNA was associated with pCR and RCBI or 0 and that five of six patients with relapse were ctDNA positive before surgery.

In patients with non‐metastatic (stage I–III) breast cancer, serial monitoring of ctDNA detected progression with a sensitivity of 86–93% and a specificity of 100% with an average lead time of 11 months (range, 0.5–37) even before recurrent disease was detected by imaging or clinical signs became apparent [75, 88, 89].

Despite these promising data, the primary concern is still the restricted sensitivity of existing assays in accurately identifying all patients at risk of relapse. Consequently, studies investigating the potential for treatment de‐escalation based on ctDNA are not yet poised for initiation. However, findings from all studies conducted in the neoadjuvant setting thus far, including our own research, consistently indicate that the persistence of ctDNA under neoadjuvant treatment carries an unfavorable prognosis. Early assessment in these cases may serve as a basis for treatment intensification, thus enhancing the prognosis for these patients. Many follow‐up studies evaluating ctDNA detection after treatment are underway. Turner et al. [90] examined a surveillance strategy and advocated for treatment escalation among patients with TNBC based on ctDNA positivity. In the intervention cohort, patients received pembrolizumab when ctDNA was detected. However, with only nine of 32 patients remaining in the intervention cohort after staging, and only five receiving pembrolizumab, the trial was underpowered and enriched for high‐risk patients. Only one patient experienced a drop in ctDNA levels under treatment. Many similar trials evaluating ctDNA detection after treatment are underway and we anticipate a substantial rise in evidence, with several methods outlined in Tables 1, 2, 3 showing encouraging outcomes for future clinical interventions [73, 91, 92].

7. Advanced breast cancer – detection of an increasing number of genome‐based therapeutic targets

While ctDNA testing may be used to identify disease relapse prior to imaging in early‐stage breast cancer as a future perspective, it is already becoming a crucial part of routine clinical practice in mBC. This is due to the high number of actionable mutations that can be detected, as well as the utility of ctDNA as a surrogate for tumor burden. Besides ER, progesterone receptor (PR), and HER2 status, there is an increasing number of other biomarkers known to predict drug benefits and affect patient management and treatment approaches [93, 94]. These include germline BRCA1/2 mutations (gBRCA1/2m) in HER2‐negative mBC, PD‐L1 in TNBC or PIK3CA in ER/PR‐positive, HER2‐negative mBC [95] and, more recently, ESR1 mutations. In this context, the development of endocrine resistance in mBC represents a challenging clinical scenario in which serial testing of ctDNA represents a vital tool for early detection of resistance mechanisms and prediction of progression‐free survival (PFS) [96, 97, 98].

In patients with locally advanced or metastatic ER‐positive/HER2‐negative breast cancer, ET with either aromatase inhibitors (AI) or fulvestrant plus a cyclin‐dependent kinase 4/6 (CDK4/6) inhibitor is recommended as first‐line standard of care (SOC). Progression after at least one line of ET or CDK4/6 inhibitor therapy is often associated with endocrine resistance, which includes development of acquired mutations in a variety of genes, including ERBB2, NF1, and estrogen receptor 1 ESR1 [95, 99].

7.1. ESR1 mutations

Mutations in the ESR1 gene that encodes for ERα are known drivers of resistance to first‐line AI, and approximately 40% of these patients acquire ESR1 mutations over time during the disease progression and evolution [100, 101, 102]. The most common ESR1 hotspot mutations are Y537S/N/C, D538G, and L536Y (see Fig. 3) [103]. A combined analysis of the phase III SoFEA and EFECT Trials, which enrolled HR‐positive mBC patients, demonstrated that patients with ESR1 mutation at baseline who had previously progressed on nonsteroidal AI therapy experienced inferior PFS and OS when treated with exemestane vs. fulvestrant 250 mg [72]. The PALOMA‐3 trial further investigated specific hotspot mutations, some of them assumed to be sub‐clonal in resistant cancer cells. Specifically, in 114 of 445 patients an ESR1 hotspot mutation was detected; and in 100 patients, a PIK3CA hotspot mutation was found at treatment initiation. On day 15 of treatment, ctDNA levels of ESR1 mutations and also the levels of PIK3CA mutations significantly declined, especially in the cohort receiving palbociclib. To measure the molecular response of ctDNA the ratio of mutant copies·mL−1 of plasma was compared between day 1 and day 15. In contrast to PI3KCA, the early dynamics of ESR1 mutations failed to predict PFS in this trial. Although there was a decrease in ESR1 mutant clones upon treatment with fulvestrant ± palbociclib, this did not correlate with long‐term improvement in PFS [104]. Nevertheless, one could speculate that decreasing levels of ctDNA correlated with some response to treatment, a concept which is now also evaluated in prospective clinical trials.

Fig. 3.

Fig. 3

List of currently clinically relevant ESR1 mutations in patients with endocrine resistant ER‐positive breast cancer (as analyzed in the current studies leading to approval or upcoming approval of SERDs in. development). ER, estrogen receptor; SERD, selective estrogen receptor degrader. Figure created with BioRender.com.

The PADA‐1 trial adopted a pioneering strategy by exchanging the endocrine partner of CDK4/6 inhibitor therapy upon the detection of an ESR1 mutation in patients with ER‐positive/HER2‐negative mBC, which led to almost doubling the PFS by switching from AI + palbociclib to fulvestrant + palbociclib [105]. A multiplex digital droplet PCR (ddPCR) based detection of ESR1 mutations was conducted analyzing only a subset of ESR1 hotspot mutations (Fig. 3). ESR1 mutations result in estrogen‐independent ER activation and thus, resistance to AI, but not to ER inhibitors, arises [100, 106]. However, as recent trials with oral selective estrogen receptor degraders (SERDs) have shown, fulvestrant has less activity in patients with ESR1 mutations. This was particularly shown in the SERENA 2 trial, where ctDNA‐based ESR1 clearance was earlier and more effective with camizestrant as with fulvestrant. The concept of PADA1 trial was novel, although the testing strategy in future trials may be improved, and treatments selection toward more effective strategies may be more appropriate. Currently ongoing clinical trials address this question. The EMERALD trial evaluated the efficacy and safety of the oral ERD elacestrant vs. standard‐of‐care ET (either AI or fulvestrant) in previously treated patient with ER‐positive/HER2‐negative mBC Elacestrant treatment resulted in a 30% reduction in the risk of progression or death compared to SOC in the overall cohort, as well as a 45% risk reduction in patients with ESR1 mutation [71]. However, some patients harboring ESR1 mutations do not benefit from elacestrant, which is indicated by an initial drop in PFS that may indicate ‘total endocrine resistance’ [71]. Therefore, it is important to identify further biomarkers for the selection of patients who will benefit from other treatment regimens, for example, chemotherapy or antibody drug conjugates. Whereas the EMERALD trial has been the first which led to approval of an oral SERD in palliative treatment, only a small proportion of patients will be selected for single agent treatment after progression on combinations with CDK4/6 inhibitors. Nevertheless, these data gave rise to an update of the American Society of Clinical Oncology (ASCO) guidelines that now include a testing recommendation for ESR1 mutations, as well as a recommendation of the oral SERD elacestrant for patients with mBC and detectable ESR1 mutations who experience disease progression on ET with or without prior CDK4/6 inhibitor treatment. Moreover, the updated ASCO guidelines state that retesting at subsequent disease progression is warranted to determine whether an ESR1 mutation has emerged [107]. The FDA has already approved elacestrant and the Guardant360 CDx assay as a companion diagnostic device in the beginning of 2023. The EMA has approved elacestrant in September 2023 for the treatment of postmenopausal women and men with ER‐positive/HER2‐negative, locally advanced, or mBC with an activating ESR1 mutation who have disease progression following at least 1 line of ET including a CDK4/6 inhibitor. This implicates an urgent need for reliable ESR1 testing.

In the SERENA‐2 trial, camizestrant, a next‐generation oral selective estrogen receptor antagonist and degrader (ngSERD), is being studied with promising results in post‐menopausal women with advanced ER‐positive/HER2‐negative BC. At the San Antonio Breast Cancer Symposium, Oliveira et al. [108] showed that treatment with camizestrant (both 75 and 150 mg) resulted in eradicated or near undetectable ESR1m ctDNA levels by day 1 of cycle 2. These lower levels were maintained for at least 6 months. These data suggest that an early intervention upon detection of an ESR1 mutation may provide the maximum patient benefit, as shown by a PFS of 12.9 months (range, 4.5–14.7) on camizestrant treatment vs. 2.2 months (range, 1.9–9.3) in fulvestrant‐treated patients with only a single ESR1 mutation [109]. This is being investigated further in the randomized, double‐blind, phase III SERENA‐6 trial. In that trial, the efficacy and safety of switching from an AI to camizestrant is assessed in patients with HR‐positive/HER2‐negative mBC on first‐line therapy upon detection of an ESR1 mutation in ctDNA, while maintaining treatment with the same CDK4/6 inhibitor [110]. In addition, these data underline the potential of ctDNA as a monitoring tool for treatment response.

Another approach to overcome the limited treatment options for patients with mBC with ESR1 mutations is lasofoxifene, a third‐generation selective estrogen receptor modulator. In the ELAINE‐1 study, lasofoxifene demonstrated antitumor activity compared to fulvestrant [111], and in ELAINE‐2, lasofoxifene showed antitumor activity in combination with abemaciclib [112]. Based on these data, patients with ESR1‐mutant, ER‐positive, HER2‐negative mBC will be randomized to either abemaciclib plus fulvestrant or abemaciclib plus lasofoxifene in the large, randomized phase III ELAINE‐3 trial (NCT05696626) that is using the FDAapproved Guardant360® CDx liquid biopsy NGS–based test as a companion diagnostic for the sequencing of 74 genes. Although the clinical need for liquid biopsy‐based testing is becoming increasingly apparent, none of the assays described or listed here are currently reimbursed in a routine clinical setting in Austria, where the authors are actively practicing. This applies to all mutations and targets discussed, including PIK3CA or ESR1 hotspot mutations, among others. The situation is particularly crucial for ESR1 testing, since the authors mandate ctDNA‐based detection of ESR1 mutation in order to reimburse elacestrant. Thus, there is no clear recommendation for a specific assay to be used, but rather an attempt of the authors to address this critical issue.

In addition to detecting ESR1 point mutations, liquid biopsies were shown to efficiently identify ESR1 fusions, particularly in metastatic ET‐resistant ER‐positive breast cancer [113]. Since not all ESR1 fusions are clinically actionable, Gou et al. [114] developed a 24‐gene expression signature specific for transcriptionally active ESR1 fusion proteins. Depending on the ESR1 fusion type, there are different strategies, for example, targeting of CDK4/6 if an active ligand‐binding/Activation Function 2 domain swap is detected, or downstream CDK4/6 inhibition if the ligand‐binding domain is absent, or administration of HER2‐directed antibody‐drug‐conjugates if a patient presents with an ESR1::CCDC170 fusion [115]. However, these are observations from case studies, and further strategies aimed at precision treatment of these resistance alterations is warranted.

7.2. PIK3CA mutations

In 28–46% of patients with HR‐positive/HER2‐negative mBC, mutations in the PI3KCA gene that encodes the p110α subunit of phosphatidylinositol‐3‐kinase (PI3K) are reported and these are associated with chemoresistance and poor prognosis [116, 117, 118]. The SOLAR‐1 trial that included HR‐positive/HER2‐negative mBC patients showed that the addition of alpelisib, an orally bioavailable α‐selective PI3K inhibitor, resulted in a significant and clinically meaningful PFS benefit in the presence of the PIK3CA mutation regardless of its location (E542X, E545X, or H1047X). Although the overall survival results did not cross the prespecified efficacy boundary, a 7.9‐month numeric improvement in median overall survival was reported in patients receiving fulvestrant plus alpelisib [118]. In the BYLieve trial, alpelisib plus fulvestrant also showed a significant impact on PFS in patients who had experienced progression on a CDK4/6 inhibitor plus an AI [119]. We have shown that the SiMSen‐Seq‐based detection of PIK3CA mutations in plasma exhibits advantageous concordance with the tissue analyses, and when combined with an untargeted, mutation‐independent approach for detecting ctDNA fractions, it enhances the performance of the SiMSen‐Seq test. This combinatory approach helps select suitable patients for alpelisib treatment [120]. Our current approaches are focusing on maintaining the sensitivity for PIK3CA mutational testing while also allowing for testing of additional targets including ESR1.

7.3. Protein kinase B ( AKT ) mutations

The protein kinase B (AKT) comprises three highly homologous isoforms: namely Akt1 (PKBα), Akt2 (PKBβ), and Akt3 (PKBγ) [121]. In addition to PIK3CA and ESR1, AKT is implicated in endocrine resistance in HR‐positive/HER2‐negative mBC. The phase III CAPItello‐291 trial investigated the efficacy and safety of the pan‐AKT inhibitor capivasertib + fulvestrant in patients with AI‐resistant HR‐positive/HER2‐negative mBC [122]. The dual primary endpoint consisting of investigator‐assessed PFS in the overall population (7.2 vs. 3.6 months) as well as in patients with AKT pathway‐altered tumors (7.3 vs. 3.1 months) was met. Besides PIK3CA mutations, AKT1 and PTEN mutations were associated with greater benefit from capivasertib then in the overall population. Although NGS was performed in tumor tissue [123], other trials have already identified AKT1 and PTEN mutations via ctDNA and allocated patients to an appropriate therapy [44]. Nevertheless, if included in the panel, this information may add information to clinical interpretation.

7.4. ERBB2 (HER2) mutations

Somatic activating mutations in the ERBB2 (HER2) gene are present in approximately 3–5% of patients with metastatic HR‐positive breast cancer and are associated with endocrine resistance due to crosstalk between HER2 and ER signaling pathways. In the SUMMIT trial, neratinib, an oral, irreversible, pan‐HER tyrosine kinase inhibitor (TKI), was administered in combination with fulvestrant and trastuzumab to patients with HR‐positive HER2‐mutant mBC after progression on CDK4/6 inhibitor therapy. The triplet combination showed encouraging clinical efficacy with an overall response rate of 39% (95% CI, 26–52) and a median PFS of 8.3 months (95% CI, 6.0–15.1) [124]. The value of liquid biopsy‐based detection of HER2 mutation was clearly suggested by plasmaMATCH trial [44].

7.5. BRCA mutations

Approximately, two thirds of the BRCA1/2 mutations found in breast cancer are germline mutations, whereas the remaining third are somatic [125]. While the efficacy of poly ADP‐ribose polymerase inhibitor (PARPi) therapy is currently under investigation for mBC with somatic BRCA1/2 mutations (sBRCA1/2m) by the Translational Breast Cancer Research Consortium (NCT03344965), there are data on germline mutations in BRCA1 or BRCA2 already, which are carried by approximately 5% of unselected patients with breast cancer [126] and are detected in up to 20% of patients with TNBC [127]. The findings of the phase III OlympiAD trial (olaparib vs. chemotherapy median PFS of 7.0 vs. 4.2 months; hazard ratio = 0.58, 95% CI: 0.43–0.80) showed that patients with HER2‐negative locally advanced breast cancer or mBC and gBRCA1/2m derive significant benefit from PARPi therapy, which also applied to the phase III EMBRACA study (talazoparib vs. chemotherapy, median PFS of 8.6 vs. 5.6 months; hazard ratio = 0.54, 95% CI: 0.41–0.71) [128, 129]. An investigator‐initiated phase II study revealed that olaparib is also effective in patients with sBRCA1/2m, with almost similar ORR (50%, 90% CI: 28–72) as reported in the OlympiAD and EMBRACA trials (59.9%, 95% CI: 52.0–67.4 and 62.2%, 95% CI: 55.8–69.0, respectively) and a median PFS of 6.3 months (90% CI, 4.4–not available) [128, 129, 130]. It is of importance that the majority of used NGS panels incorporate BRCA mutations, and in most cases, the allele frequency of mutations enables conclusions regarding the origin of mutations (germline vs. somatic). Also, other genes that play an important role in the management of patients with mBC, including TP53, GATA3, and ARID1A can be assessed. However, since these are not yet clinically established, we did not cover these alterations in detail [131].

8. TMB and MSI as predictive complex biomarkers for immunotherapy

High TMB defined as ≥ 10 mutations per megabase, leads to clinically meaningful benefit from treatment with pembrolizumab in several cancer types [132]. However, no patients with mBC had been included in this study. However, in an explorative analysis from the Impassion130 trial, higher TMB was associated with better OS in the PD‐L1 positive subgroup [133]. In both trials, TMB was tested in tumor tissue. It was shown that TMB in plasma, referred to as blood TMB, is highly correlative to tissue‐based TMB. However, data in breast cancer regarding blood TMB as a predictive marker for immune checkpoint inhibitor (ICI) are lacking. MSI is another biomarker for the clinical benefit of ICI [134]. Also, prospective data demonstrating liquid biopsy‐based detection of MSI for the prediction of utility of ICI may be promising. The assessment of these theragnostic markers – TMB and MSI – demonstrated feasibility in ctDNA assays, although studies in patients with breast cancer are still limited [135]. A list of mutations and matched therapies according to FDA and EMA approval is displayed in Table 4.

Table 4.

Mutation diagnosis – precision medicine for mBC targeted therapies.

Genomic finding Matched therapy
ESR1 mut Elacestrant* ,
PIK3CA mut Alpelisib (+ fulvestrant)* ,
HER2 mut Neratinib* , , Lapatinib* ,
BRCA1/2 Olaparib* , , Talazoparib* ,
NTRK fusion Larotrectinib* , , Entrectinib* ,
Microsatellite instability‐high or mismatch repair deficient Pembrolizumab* ,
Tumor mutational burden‐high ≥10 mut/Mb Pembrolizumab* ,
PDL‐L1 expression ≥1% Atezolizumab* ,
PD‐L1 combined positive score ≥ 10 Pembrolizumab* ,
RET fusion Selpercatinib*

*FDA approved.

EMA approved.

9. Outlook and considerations

Although the indications and guidelines for molecular profiling of patients with mBC outlined above appear straightforward, integrating molecular testing results from ctDNA into routine clinical practice requires a careful understanding of the associated technical caveats and clinical implications, which can differ among clinics. In terms of the test itself, it is recommended to utilize a well validated assay with known performance metrics, ensuring that molecular findings can be accurately interpreted [136]. In addition, the molecular test must align with the therapy indication, costs, and reimbursement thereof, which plays a major role in the selection of the assay. In this context, a genomic testing cost calculator has demonstrated that the use of NGS for up to 50 genes compared to sequential single‐gene testing (PCR for PIK3CA, IHC for HER2 amplification and MSI‐H, as well as FISH for NTRK fusions) led to a cost reduction of approximately 42% per correctly identified patient with mBC [137]. Ultimately, the clinical interpretation is not always binary, that is, a mutation was detected and therefore is matched to treatment, or a mutation was not detected and therefore no treatment is recommended. To this end, in the following, we take ESR1 testing as a use case and provide example profiling results and decision‐making thought processes to demonstrate the intricacies of any issues that may arise in the single biomarker indication setting (Fig. 4). Here, we cover six cases of patients with mBC using a liquid biopsy‐based CGP panel (77‐gene panel, assay limit of detection: VAF 0.5%) for the indication of ESR1 mutation testing for treatment with SERDs.

Fig. 4.

Fig. 4

Six examples for patients with mBC with ESR1 molecular testing results. Six patient examples are illustrated to demonstrate the spectrum of results that may be observed from routine molecular profiling of plasma DNA in the clinical setting. The same 77‐gene CGP panel was employed for all cases. This assay has been validated to reliably detect variants down to a VAF of 0.5%, indicating that any variant with a VAF below this percentage is below the technical LOD. In the left‐most panel, the alterations detected are listed alongside their VAFs, which are provided as a percentage in parentheses. An asterisk next to the VAF designates that this variant is below the assay LOD. In addition, the TF in plasma was estimated with a standard algorithm (ichorCNA), which has an LOD of 3%, and is provided in the box above the list of detected variants. In the middle column, a brief orientation and interpretation of the results is provided, which should help understand the technical and biological considerations that play a role in the clinical decision. The third column provides a brief justification of the treatment implications. As an official guideline or decision tree for the implementation of ESR1 mutation testing results is currently lacking, the justification here serves as more of a high‐level interpretation of the results for clinicians. Importantly, this highlights the utility of the molecular tumor board in performing an interdisciplinary, comprehensive evaluation of the results within the individual patient context to derive the most evidence‐based decision. CGP, comprehensive genomic profiling; LOD, limit of detection; mBC, metastatic breast cancer; TF, tumor fraction; VAF, variant allele frequency. Figure created with BioRender.com.

Considering the presumed future increase in molecular alterations, it can be assumed that NGS will reduce the financial burden of testing. Regarding the assay type, targeted sequencing approaches offer the benefit of being more cost‐effective, since only loci of interest or rather clinically relevant hotspot mutations are analyzed, considering that a large part of the genome is still undruggable [138]. Thus, it might be easier to push the reimbursement of liquid biopsies that are directly linked to specific treatments. With an emphasis on the value of molecular characterization for treatment decisions in breast cancer, we finally provide a potential roadmap depicting this integration into clinical practice in Fig. 5.

Fig. 5.

Fig. 5

Key steps for integrating liquid biopsy assays into routine clinical workflows (adapted from [50]).

10. Conclusions

Liquid biopsy, particularly the analysis of ctDNA, is under active investigation. This field offers a wide range of applications, from early detection to monitoring of metastatic disease, as well as selection of mutation‐directed therapies [139, 140, 141]. Minimally invasive ctDNA testing, compared to conventional tissue‐based molecular analyses, not only allows for multiple sampling but also enables the visualization of intricate subclonal variations within the spreading tumor. These real‐time insights into tumor dynamics and genetic alterations facilitate personalized therapy selection and enable early and intensified intervention or therapy de‐escalation strategies, with additional psychosocial impact on patient quality of life.

Some of these applications have become routine in clinical practice, such as the analysis of the PIK3CA mutational status, which can be performed based on both tissue and plasma samples. In view of this breakthrough and results from ESR1 trials, the 2023 ASCO guideline update strongly recommends ESR1 ctDNA‐based testing due to its superior sensitivity in the detection of subclonal mutations that have been acquired over time. It also suggests retesting upon disease progression, which demonstrates how the non‐invasive nature of this approach can transform standard clinical practices.

Conflict of interest

E.V. Klocker received personal fees/travel support from AstraZeneca, DaiichiSankyo, EliLilly, Gilead, Novartis, Roche, and PierreFabre. S. Hasenleithner is a Member of the Advisory Board of CureMatch. R. Bartsch. reports an advisory Role: AstraZeneca, Daiichi, Eisai, Eli‐Lilly, Gilead, Gruenenthal, MSD, Novartis, Pfizer, Pierre‐Fabre, Puma, Roche, Seagen, Stemline, Lecture Honoraria: AstraZeneca, BMS, Daichi, Eisai, Eli‐Lilly, Gilead, Gruenenthal, MSD, Novartis, Pfizer, Pierre‐Fabre, Roche, Seagen, Research Support: Daiichi, MSD, Novartis, Roche. S.P. Gampenrieder reports personal fees/travel support from MSD, Novartis, AstraZeneca, Lilly, Seagen, Daiichi Sankyo, Gilead, Pfizer, Stemline Therapeutics, Janssen, and research support from Roche, Daiichi Sankyo, Novartis, Pfizer, Caris Life Sciences, Lilly, Seagen, Gilead and AstraZeneca. D. Egle reports personal fees/travel support from Amgen, AstraZeneca, Daiichi‐Sankyo, Gilead, Lilly, Mennarini, MSD, Novartis, Pfizer, Roche, Sandoz, Seagen, Sirius Medical. C. F. Singer reports travel, research and unrestricted grants from Novartis, AstraZeneca, Daiichi, Amgen, Roche, Seagen und Gilead Sciences. G. Rinnerthaler reports Honoraria from Amgen, AstraZeneca, Daiichi Sankyo, Eli Lilly, Gilead, MSD, Novartis, Pfizer, Roche, Seagen, Stemline, BMS; Consulting or Advisory Role: Amgen, AstraZeneca, Daiichi Sankyo, Eli Lilly, Gilead, MSD, Novartis, Pfizer, Pierre Fabre, Roche, Stemline; Travel, Accommodations, Expenses: Amgen, Daiichi Sankyo, Eli Lilly, Gilead, Merck, Pfizer, Roche. M. Hubalek reports personal fees/travel support from Amgen, AstraZeneca, DaiichiSankyo, EliLilly, Menarini‐Stemline. K. Schmitz reports personal fees/travel support from Amgen, AstraZeneca, Bayer, Daiichi‐Sankyo, Diaceutics, Discovery Life Sciences, Incyte, Janssen, Merck, MSD, Novartis, Pfizer, PharmaMar, Roche, Sanofi, Servier, Stemline, Takeda and Targos GmbH. Z. Bago‐Horvath reports no conflicts of interest. A. Petzer reports honoraria/advisory role from Novartis, Amgen, Celgene‐BMS, Sandoz, Janssen, AstraZeneca, Abbvie, Takeda, Sanofi, Kite‐Gilead, Roche, Pfizer, Saegen, Daiichi Sankyo and travel support from Roche, Gilead, Daiichi Sankyo, Janssen, Eli Lilly, Pierre Fabre. S. Heibl reports her role in advisory boards and honoraria from Stemline, Daiichi‐Sankyo, Eli Lilly, Novartis, Amgen, Gilead, Roche. E. Heitzer has received unrelated funding from Illumina, Roche, Servier, Freenome, and PreAnalytiX, and received honoraria from Roche and AstraZeneca for advisory boards, not related to our study. M. Gnant reports personal fees/travel support from Amgen, AstraZeneca, DaiichiSankyo, EliLilly, Menarini‐Stemline, MSD, Novartis, PierreFabre, Veracyte; an immediate family member is employed by Sandoz. M. Balic reports personal fees/travel support from Amgen, AstraZeneca, DaiichiSankyo, EliLilly, Gilead, Menarini‐Stemline, MSD, Novartis, PierreFabre, Seagen, and research support from AstraZeneca, Daiichi, Novartis, Pierre Fabre, Pfizer, and Seagen.

Author contributions

MG, MB, EVK, SH, EH, and AF contributed to the conception and design. The manuscript was written by AF, EVK, and MB. All authors read and approved the final manuscript.

Acknowledgements

The Graphical Abstract and Figures 1, 2, 3, 4 have been created with BioRender.com.

Marija Balic and Michael Gnant contributed equally as last senior authors

References

  • 1. Berger MF, Mardis ER. The emerging clinical relevance of genomics in cancer medicine. Nat Rev Clin Oncol. 2018;15(6):353–365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Buono G, Gerratana L, Bulfoni M, Provinciali N, Basile D, Giuliano M, et al. Circulating tumor DNA analysis in breast cancer: is it ready for prime‐time? Cancer Treat Rev. 2019;73:73–83. [DOI] [PubMed] [Google Scholar]
  • 3. Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, et al. Detection of circulating tumor DNA in early‐ and late‐stage human malignancies. Sci Transl Med. 2014;6(224):224ra24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Loibl S, Poortmans P, Morrow M, Denkert C, Curigliano G. Breast cancer. Lancet. 2021;397(10286):1750–1769. [DOI] [PubMed] [Google Scholar]
  • 5. Harbeck N, Gnant M. Breast cancer. Lancet. 2017;389(10074):1134–1150. [DOI] [PubMed] [Google Scholar]
  • 6. Saura C, Hlauschek D, Oliveira M, Zardavas D, Jallitsch‐Halper A, de la Peña L, et al. Neoadjuvant letrozole plus taselisib versus letrozole plus placebo in postmenopausal women with oestrogen receptor‐positive, HER2‐negative, early‐stage breast cancer (LORELEI): a multicentre, randomised, double‐blind, placebo‐controlled, phase 2 trial. Lancet Oncol. 2019;20(9):1226–1238. [DOI] [PubMed] [Google Scholar]
  • 7. Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, et al. Pathological complete response and long‐term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet. 2014;384(9938):164–172. [DOI] [PubMed] [Google Scholar]
  • 8. Burstein HJ, Curigliano G, Loibl S, Dubsky P, Gnant M, Poortmans P, et al. Estimating the benefits of therapy for early‐stage breast cancer: the St. Gallen international consensus guidelines for the primary therapy of early breast cancer 2019. Ann Oncol. 2019;30(10):1541–1557. [DOI] [PubMed] [Google Scholar]
  • 9. Heil J, Kuerer HM, Pfob A, Rauch G, Sinn HP, Golatta M, et al. Eliminating the breast cancer surgery paradigm after neoadjuvant systemic therapy: current evidence and future challenges. Ann Oncol. 2020;31(1):61–71. [DOI] [PubMed] [Google Scholar]
  • 10. Zhou Q, Gampenrieder SP, Frantal S, Rinnerthaler G, Singer CF, Egle D, et al. Persistence of ctDNA in patients with breast cancer during Neoadjuvant treatment is a significant predictor of poor tumor response. Clin Cancer Res. 2021;28(4):697–707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Marinovich ML, Houssami N, Macaskill P, von Minckwitz G, Blohmer J‐U, Irwig L. Accuracy of ultrasound for predicting pathologic response during neoadjuvant therapy for breast cancer. Int J Cancer. 2015;136(11):2730–2737. [DOI] [PubMed] [Google Scholar]
  • 12. Gampenrieder SP, Peer A, Weismann C, Meissnitzer M, Rinnerthaler G, Webhofer J, et al. Radiologic complete response (rCR) in contrast‐enhanced magnetic resonance imaging (CE‐MRI) after neoadjuvant chemotherapy for early breast cancer predicts recurrence‐free survival but not pathologic complete response (pCR). Breast Cancer Res. 2019;21(1):19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. van Loevezijn AA, van der Noordaa MEM, van Werkhoven ED, Loo CE, Winter‐Warnars GAO, Wiersma T, et al. Minimally invasive complete response assessment of the breast after Neoadjuvant systemic therapy for early breast cancer (MICRA trial): interim analysis of a multicenter observational cohort study. Ann Surg Oncol. 2021;28(6):3243–3253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Tasoulis MK, Lee H‐B, Yang W, Pope R, Krishnamurthy S, Kim S‐Y, et al. Accuracy of post–Neoadjuvant chemotherapy image‐guided breast biopsy to predict residual cancer. JAMA Surg. 2020;155(12):e204103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Heil J, Pfob A, Sinn H‐P, Rauch G, Bach P, Thomas B, et al. Diagnosing pathologic complete response in the breast after Neoadjuvant systemic treatment of breast cancer patients by minimal invasive biopsy. Ann Surg. 2022;275(3):576–581. [DOI] [PubMed] [Google Scholar]
  • 16. Harbeck N, Penault‐Llorca F, Cortes J, Gnant M, Houssami N, Poortmans P, et al. Breast cancer. Nat Rev Dis Primers. 2019;5(1):66. [DOI] [PubMed] [Google Scholar]
  • 17. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–249. [DOI] [PubMed] [Google Scholar]
  • 18. Allemani C, Matsuda T, Di Carlo V, Harewood R, Matz M, Nikšić M, et al. Global surveillance of trends in cancer survival 2000–14 (CONCORD‐3): analysis of individual records for 37 513 025 patients diagnosed with one of 18 cancers from 322 population‐based registries in 71 countries. Lancet. 2018;391(10125):1023–1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Antonarelli G, Corti C, Tarantino P, Salimbeni BT, Zagami P, Marra A, et al. Management of patients with HER2‐positive metastatic breast cancer after trastuzumab deruxtecan failure. ESMO Open. 2023;8(4):101608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Miglietta F, Bottosso M, Griguolo G, Dieci MV, Guarneri V. Major advancements in metastatic breast cancer treatment: when expanding options means prolonging survival. ESMO Open. 2022;7(2):100409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, et al. Molecular portraits of human breast tumours. Nature. 2000;406(6797):747–752. [DOI] [PubMed] [Google Scholar]
  • 22. Sørlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA. 2001;98(19):10869–10874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Sørlie T, Tibshirani R, Parker J, Hastie T, Marron JS, Nobel A, et al. Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci USA. 2003;100(14):8418–8423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Balic M, Thomssen C, Gnant M, Harbeck N. St. Gallen/Vienna 2023: optimization of treatment for patients with primary breast cancer – a brief summary of the consensus discussion. Breast Care. 2023;18(3):213–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Perakis S, Speicher MR. Emerging concepts in liquid biopsies. BMC Med. 2017;15(1):75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Byler S, Goldgar S, Heerboth S, Leary M, Housman G, Moulton K, et al. Genetic and epigenetic aspects of breast cancer progression and therapy. Anticancer Res. 2014;34(3):1071–1077. [PubMed] [Google Scholar]
  • 27. Kim C, Lee J, Lee W, Kim A. Changes in intrinsic subtype of breast cancer during tumor progression in the same patient. Int J Clin Exp Pathol. 2015;8(11):15184–15190. [PMC free article] [PubMed] [Google Scholar]
  • 28. Sant M, Bernat‐Peguera A, Felip E, Margelí M. Role of ctDNA in breast cancer. Cancer. 2022;14(2):310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Aftimos P, Oliveira M, Irrthum A, Fumagalli D, Sotiriou C, Gal‐Yam EN, et al. Genomic and transcriptomic analyses of breast cancer primaries and matched metastases in AURORA, the breast international group (BIG) molecular screening initiative. Cancer Discov. 2021;11(11):2796–2811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Kotecha R, Tonse R, Rubens M, McDermott MW, Odia Y, Appel H, et al. Systematic review and meta‐analysis of breast cancer brain metastasis and primary tumor receptor expression discordance. Neurooncol Adv. 2021;3(1):vdab010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Priedigkeit N, Hartmaier RJ, Chen Y, Vareslija D, Basudan A, Watters RJ, et al. Intrinsic subtype switching and acquired ERBB2/HER2 amplifications and mutations in breast cancer brain metastases. JAMA Oncol. 2017;3(5):666–671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Hulsbergen AFC, Claes A, Kavouridis VK, Ansaripour A, Nogarede C, Hughes ME, et al. Subtype switching in breast cancer brain metastases: a multicenter analysis. Neuro Oncol. 2020;22(8):1173–1181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Klebe M, Fremd C, Kriegsmann M, Kriegsmann K, Albrecht T, Thewes V, et al. Frequent molecular subtype switching and gene expression alterations in lung and pleural metastasis from luminal A–type breast cancer. JCO Precis Oncol. 2020;4:848–859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Russo M, Bardelli A. Lesion‐directed therapies and monitoring tumor evolution using liquid biopsies. Cold Spring Harb Perspect Med. 2017;7(2):a029587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Pascual J, Attard G, Bidard F‐C, Curigliano G, Mattos‐Arruda LD, Diehn M, et al. ESMO recommendations on the use of circulating tumour DNA assays for patients with cancer: a report from the ESMO precision medicine working group. Ann Oncol. 2022;33(8):750–768. [DOI] [PubMed] [Google Scholar]
  • 36. Jørgensen CLT, Larsson A‐M, Forsare C, Aaltonen K, Jansson S, Bradshaw R, et al. PAM50 intrinsic subtype profiles in primary and metastatic breast cancer show a significant shift toward more aggressive subtypes with prognostic implications. Cancer. 2021;13(7):1592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Bertucci F, Ng CKY, Patsouris A, Droin N, Piscuoglio S, Carbuccia N, et al. Genomic characterization of metastatic breast cancers. Nature. 2019;569(7757):560–564. [DOI] [PubMed] [Google Scholar]
  • 38. Caballero C, Irrthum A, Goulioti T, Cameron D, Norton L, Piccart M. International research to address the challenges of metastatic breast cancer: the AURORA program (BIG 14‐01). NPJ Breast Cancer. 2023;9(1):42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Lefebvre C, Bachelot T, Filleron T, Pedrero M, Campone M, Soria J‐C, et al. Mutational profile of metastatic breast cancers: a retrospective analysis. PLoS Med. 2016;13(12):e1002201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Razavi P, Chang MT, Xu G, Bandlamudi C, Ross DS, Vasan N, et al. The genomic landscape of endocrine‐resistant advanced breast cancers. Cancer Cell. 2018;34(3):427–438.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Pearson A, Proszek P, Pascual J, Fribbens C, Shamsher MK, Kingston B, et al. Inactivating NF1 mutations are enriched in advanced breast cancer and contribute to endocrine therapy resistance. Clin Cancer Res. 2020;26(3):608–622. [DOI] [PubMed] [Google Scholar]
  • 42. Nagarajan S, Rao SV, Sutton J, Cheeseman D, Dunn S, Papachristou EK, et al. ARID1A influences HDAC1/BRD4 activity, intrinsic proliferative capacity and breast cancer treatment response. Nat Genet. 2020;52(2):187–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Xu G, Chhangawala S, Cocco E, Razavi P, Cai Y, Otto JE, et al. ARID1A determines luminal identity and therapeutic response in estrogen‐receptor‐positive breast cancer. Nat Genet. 2020;52(2):198–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Turner NC, Kingston B, Kilburn LS, Kernaghan S, Wardley AM, Macpherson IR, 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(10):1296–1308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Belic J, Koch M, Ulz P, Auer M, Gerhalter T, Mohan S, et al. Rapid identification of plasma DNA samples with increased ctDNA levels by a modified FAST‐SeqS approach. Clin Chem. 2015;61(6):838–849. [DOI] [PubMed] [Google Scholar]
  • 46. Suppan C, Brcic I, Tiran V, Mueller HD, Posch F, Auer M, et al. Untargeted assessment of tumor fractions in plasma for monitoring and prognostication from metastatic breast cancer patients undergoing systemic treatment. Cancer. 2019;11(8):1171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Dandachi N, Posch F, Graf R, Suppan C, Klocker EV, Müller HD, et al. Longitudinal tumor fraction trajectories predict risk of progression in metastatic HR+ breast cancer patients undergoing CDK4/6 treatment. Mol Oncol. 2020;15(9):2390–2400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Dang DK, Park BH. Circulating tumor DNA: current challenges for clinical utility. J Clin Invest. 2022;132(12):e154941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Heidrich I, Ačkar L, Mossahebi Mohammadi P, Pantel K. Liquid biopsies: potential and challenges. Int J Cancer. 2021;148(3):528–545. [DOI] [PubMed] [Google Scholar]
  • 50. Ignatiadis M, Sledge GW, Jeffrey SS. Liquid biopsy enters the clinic—implementation issues and future challenges. Nat Rev Clin Oncol. 2021;18(5):297–312. [DOI] [PubMed] [Google Scholar]
  • 51. Vlataki K, Antonouli S, Kalyvioti C, Lampri E, Kamina S, Mauri D, et al. Circulating tumor DNA in the Management of Early‐Stage Breast Cancer. Cells. 2023;12(12):1573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch R‐D, et al. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res. 2001;61(4):1659–1665. [PubMed] [Google Scholar]
  • 53. Rostami A, Lambie M, Yu CW, Stambolic V, Waldron JN, Bratman SV. Senescence, necrosis, and apoptosis govern circulating cell‐free DNA release kinetics. Cell Rep. 2020;31(13):107830. [DOI] [PubMed] [Google Scholar]
  • 54. Sanz‐Garcia E, Zhao E, Bratman SV, Siu LL. Monitoring and adapting cancer treatment using circulating tumor DNA kinetics: current research, opportunities, and challenges. Sci Adv. 2022;8(4):eabi8618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Stroun M, Lyautey J, Lederrey C, Olson‐Sand A, Anker P. About the possible origin and mechanism of circulating DNA. Clin Chim Acta. 2001;313(1–2):139–142. [DOI] [PubMed] [Google Scholar]
  • 56. Diaz LA, Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol. 2014;32(6):579–586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Udomruk S, Orrapin S, Pruksakorn D, Chaiyawat P. Size distribution of cell‐free DNA in oncology. Crit Rev Oncol Hematol. 2021;166:103455. [DOI] [PubMed] [Google Scholar]
  • 58. Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008;14(9):985–990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Pessoa LS, Heringer M, Ferrer VP. ctDNA as a cancer biomarker: a broad overview. Crit Rev Oncol Hematol. 2020;155:103109. [DOI] [PubMed] [Google Scholar]
  • 60. Liu B, Hu Z, Ran J, Xie N, Tian C, Tang Y, et al. The circulating tumor DNA (ctDNA) alteration level predicts therapeutic response in metastatic breast cancer: novel prognostic indexes based on ctDNA. Breast. 2022;65:116–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Hasenleithner SO, Speicher MR. A clinician's handbook for using ctDNA throughout the patient journey. Mol Cancer. 2022;21:81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Singh RR. Target enrichment approaches for next‐generation sequencing applications in oncology. Diagnostics. 2022;12(7):1539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Bewicke‐Copley F, Arjun Kumar E, Palladino G, Korfi K, Wang J. Applications and analysis of targeted genomic sequencing in cancer studies. Comput Struct Biotechnol J. 2019;17:1348–1359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Chen K, Shields MD, Chauhan PS, Ramirez RJ, Harris PK, Reimers MA, et al. Commercial ctDNA assays for minimal residual disease detection of solid tumors. Mol Diagn Ther. 2021;25(6):757–774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Parsons HA, Rhoades J, Reed SC, Gydush G, Ram P, Exman P, et al. Sensitive detection of minimal residual disease in patients treated for early‐stage breast cancer. Clin Cancer Res. 2020;26(11):2556–2564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Moding EJ, Nabet BY, Alizadeh AA, Diehn M. Detecting liquid remnants of solid tumors: circulating tumor DNA minimal residual disease. Cancer Discov. 2021;11(12):2968–2986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Dong Q, Chen C, Hu Y, Zhang W, Yang X, Qi Y, et al. Clinical application of molecular residual disease detection by circulation tumor DNA in solid cancers and a comparison of technologies: review article. Cancer Biol Ther. 2023;24(1):2274123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Newman AM, Bratman SV, To J, Wynne JF, Eclov NCW, Modlin LA, et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med. 2014;20(5):548–554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. André F, Ciruelos E, Rubovszky G, Campone M, Loibl S, Rugo HS, et al. Alpelisib for PIK3CA‐mutated, hormone receptor–positive advanced breast cancer. N Engl J Med. 2019;380(20):1929–1940. [DOI] [PubMed] [Google Scholar]
  • 70. Parsons HA, Beaver JA, Cimino‐Mathews A, Ali SM, Axilbund J, Chu D, et al. Individualized molecular analyses guide efforts (IMAGE): a prospective study of molecular profiling of tissue and blood in metastatic triple‐negative breast cancer. Clin Cancer Res. 2017;23(2):379–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Bidard F‐C, Kaklamani VG, Neven P, Streich G, Montero AJ, Forget F, et al. Elacestrant (oral selective estrogen receptor degrader) versus standard endocrine therapy for estrogen receptor–positive, human epidermal growth factor receptor 2–negative advanced breast cancer: results from the randomized phase III EMERALD trial. J Clin Oncol. 2022;40(28):3246–3256. 10.1200/JCO.22.00338 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Turner NC, Swift C, Kilburn L, Fribbens C, Beaney M, Garcia‐Murillas I, et al. ESR1 mutations and overall survival on Fulvestrant versus Exemestane in advanced hormone receptor–positive breast cancer: a combined analysis of the phase III SoFEA and EFECT trials. Clin Cancer Res. 2020;26(19):5172–5177. [DOI] [PubMed] [Google Scholar]
  • 73. Liu MC, MacKay M, Kase M, Piwowarczyk A, Lo C, Schaeffer J, et al. Longitudinal shifts of solid tumor and liquid biopsy sequencing concordance in metastatic breast cancer. JCO Precis Oncol. 2022;6:e2100321. 10.1200/PO.21.00321 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Gezer U, Bronkhorst AJ, Holdenrieder S. The clinical utility of droplet digital PCR for profiling circulating tumor DNA in breast cancer patients. Diagnostics. 2022;12(12):3042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Lipsyc‐Sharf M, de Bruin EC, Santos K, McEwen R, Stetson D, Patel A, et al. Circulating tumor DNA and late recurrence in high‐risk hormone receptor–positive, human epidermal growth factor receptor 2–negative breast cancer. J Clin Oncol. 2022;40(22):2408–2419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Magbanua MJM, Swigart LB, Ahmed Z, Sayaman RW, Renner D, Kalashnikova E, et al. Clinical significance and biology of circulating tumor DNA in high‐risk early‐stage HER2‐negative breast cancer receiving neoadjuvant chemotherapy. Cancer Cell. 2023;41(6):1091–1102.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Tie J, Cohen JD, Lahouel K, Lo SN, Wang Y, Kosmider S, et al. Circulating tumor DNA analysis guiding adjuvant therapy in stage II colon cancer. N Engl J Med. 2022;386(24):2261–2272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Lennon AM, Buchanan AH, Kinde I, Warren A, Honushefsky A, Cohain AT, et al. Feasibility of blood testing combined with PET‐CT to screen for cancer and guide intervention. Science. 2020;369(6499):eabb9601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Wu D, Zhou G, Jin P, Zhu J, Li S, Wu Q, et al. Detection of colorectal cancer using a simplified SEPT9 gene methylation assay is a reliable method for opportunistic screening. J Mol Diagn. 2016;18(4):535–545. [DOI] [PubMed] [Google Scholar]
  • 80. Dietrich D, Kneip C, Raji O, Liloglou T, Seegebarth A, Schlegel T, et al. Performance evaluation of the DNA methylation biomarker SHOX2 for the aid in diagnosis of lung cancer based on the analysis of bronchial aspirates. Int J Oncol. 2012;40(3):825–832. [DOI] [PubMed] [Google Scholar]
  • 81. Nicholson BD, Oke J, Virdee PS, Harris DA, O'Doherty C, Park JE, et al. Multi‐cancer early detection test in symptomatic patients referred for cancer investigation in England and Wales (SYMPLIFY): a large‐scale, observational cohort study. Lancet Oncol. 2023;24(7):733–743. [DOI] [PubMed] [Google Scholar]
  • 82. Agassi R, Czeiger D, Shaked G, Avriel A, Sheynin J, Lavrenkov K, et al. Measurement of circulating cell‐free DNA levels by a simple fluorescent test in patients with breast cancer. Am J Clin Pathol. 2015;143(1):18–24. [DOI] [PubMed] [Google Scholar]
  • 83. Garcia‐Murillas I, Chopra N, Comino‐Méndez I, Beaney M, Tovey H, Cutts RJ, et al. Assessment of molecular relapse detection in early‐stage breast cancer. JAMA Oncol. 2019;5(10):1473–1478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Rothé F, Silva MJ, Venet D, Campbell C, Bradburry I, Rouas G, 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(12):3581–3588. [DOI] [PubMed] [Google Scholar]
  • 85. Riva F, Bidard F‐C, Houy A, Saliou A, Madic J, Rampanou A, et al. Patient‐specific circulating tumor DNA detection during Neoadjuvant chemotherapy in triple‐negative breast cancer. Clin Chem. 2017;63(3):691–699. [DOI] [PubMed] [Google Scholar]
  • 86. Butler TM, Boniface CT, Johnson‐Camacho K, Tabatabaei S, Melendez D, Kelley T, et al. Circulating tumor DNA dynamics using patient‐customized assays are associated with outcome in neoadjuvantly treated breast cancer. Cold Spring Harb Mol Case Stud. 2019;5(2):a003772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Magbanua MJM, Swigart LB, Wu H‐T, Hirst GL, Yau C, Wolf DM, et al. Circulating tumor DNA in neoadjuvant‐treated breast cancer reflects response and survival. Ann Oncol. 2021;32(2):229–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Coombes RC, Page K, Salari R, Hastings RK, Armstrong A, Ahmed S, et al. Personalized detection of circulating tumor DNA antedates breast cancer metastatic recurrence. Clin Cancer Res. 2019;25(14):4255–4263. [DOI] [PubMed] [Google Scholar]
  • 89. Olsson E, Winter C, George A, Chen Y, Howlin J, Tang ME, et al. Serial monitoring of circulating tumor DNA in patients with primary breast cancer for detection of occult metastatic disease. EMBO Mol Med. 2015;7(8):1034–1047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Turner NC, Swift C, Jenkins B, Kilburn L, Coakley M, Beaney M, et al. Results of the c‐TRAK TN trial: a clinical trial utilising ctDNA mutation tracking to detect molecular residual disease and trigger intervention in patients with moderate‐ and high‐risk early‐stage triple‐negative breast cancer. Ann Oncol. 2023;34(2):200–211. [DOI] [PubMed] [Google Scholar]
  • 91. Janni W. Analysis of ctDNA for the detection of minimal residual disease (MRD) using a tissue‐free, multiomic assay in patients with early‐stage breast cancer. San Antonio Breast Cancer Symposium. 2023. Presented 06122023 (Abstract PS06‐06).
  • 92. Fernandez SV, Tan YF, Rao S, Fittipaldi P, Sheriff F, Borghaei H, et al. Validation of a molecular diagnostic test for circulating tumor DNA by next‐gen sequencing. Int J Mol Sci. 2023;24(21):15779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Chan A, Moy B, Mansi J, Ejlertsen B, Holmes FA, Chia S, et al. Final efficacy results of Neratinib in HER2‐positive hormone receptor‐positive early‐stage breast cancer from the phase III ExteNET trial. Clin Breast Cancer. 2021;21(1):80–91.e7. [DOI] [PubMed] [Google Scholar]
  • 94. Baselga J, Campone M, Piccart M, Burris HA, Rugo HS, Sahmoud T, et al. Everolimus in postmenopausal hormone‐receptor–positive advanced breast cancer. N Engl J Med. 2012;366(6):520–529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Gennari A, André F, Barrios CH, Cortés J, Azambuja E d, DeMichele A, et al. ESMO clinical practice guideline for the diagnosis, staging and treatment of patients with metastatic breast cancer. Ann Oncol. 2021;32(12):1475–1495. [DOI] [PubMed] [Google Scholar]
  • 96. Ghosh M, Naik R, Lingaraju SM, Susheela SP, Patil S, Srinivasachar GK, et al. Landscape of clinically actionable mutations in breast cancer ‘A cohort study’. Transl Oncol. 2021;14(1):100877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Cullinane C, Fleming C, O'Leary DP, Hassan F, Kelly L, O'Sullivan MJ, et al. Association of Circulating Tumor DNA with disease‐free survival in breast cancer. JAMA Netw Open. 2020;3(11):e2026921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Prat A, Brasó‐Maristany F, Martínez‐Sáez O, Sanfeliu E, Xia Y, Bellet M, et al. Circulating tumor DNA reveals complex biological features with clinical relevance in metastatic breast cancer. Nat Commun. 2023;14(1):1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Burstein HJ, Somerfield MR, Barton DL, Dorris A, Fallowfield LJ, Jain D, et al. Endocrine treatment and targeted therapy for hormone receptor–positive, human epidermal growth factor receptor 2–negative metastatic breast cancer: ASCO guideline update. J Clin Oncol. 2021;39(35):3959–3977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Brett JO, Spring LM, Bardia A, Wander SA. ESR1 mutation as an emerging clinical biomarker in metastatic hormone receptor‐positive breast cancer. Breast Cancer Res. 2021;23(1):85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Carausu M, Bidard F‐C, Callens C, Melaabi S, Jeannot E, Pierga J‐Y, et al. ESR1 mutations: a new biomarker in breast cancer. Expert Rev Mol Diagn. 2019;19(7):599–611. [DOI] [PubMed] [Google Scholar]
  • 102. Page K, Martinson LJ, Fernandez‐Garcia D, Hills A, Gleason KLT, Gray MC, et al. Circulating tumor DNA profiling from breast cancer screening through to metastatic disease. JCO Precis Oncol. 2021;5:1768–1776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Zundelevich A, Dadiani M, Kahana‐Edwin S, Itay A, Sella T, Gadot M, et al. ESR1 mutations are frequent in newly diagnosed metastatic and loco‐regional recurrence of endocrine‐treated breast cancer and carry worse prognosis. Breast Cancer Res. 2020;22(1):16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. O'Leary B, Hrebien S, Morden JP, Beaney M, Fribbens C, Huang X, et al. Early circulating tumor DNA dynamics and clonal selection with palbociclib and fulvestrant for breast cancer. Nat Commun. 2018;9(1):896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Bidard F‐C, Hardy‐Bessard A‐C, Dalenc F, Bachelot T, Pierga J‐Y, Rouge T d l M, et al. Switch to fulvestrant and palbociclib versus no switch in advanced breast cancer with rising ESR1 mutation during aromatase inhibitor and palbociclib therapy (PADA‐1): a randomised, open‐label, multicentre, phase 3 trial. Lancet Oncol. 2022;23(11):1367–1377. [DOI] [PubMed] [Google Scholar]
  • 106. Jeselsohn R, Yelensky R, Buchwalter G, Frampton G, Meric‐Bernstam F, Gonzalez‐Angulo AM, et al. Emergence of constitutively active estrogen receptor‐α mutations in pretreated advanced estrogen receptor–positive breast cancer. Clin Cancer Res. 2014;20(7):1757–1767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Burstein HJ, DeMichele A, Somerfield MR, Henry NL. Testing for ESR1 mutations to guide therapy for hormone receptor–positive, human epidermal growth factor receptor 2–negative metastatic breast cancer: ASCO guideline rapid recommendation update. J Clin Oncol. 2023;41(18):3423–3425. [DOI] [PubMed] [Google Scholar]
  • 108. Oliveira M, Pominchuk D, Nowecki Z, Hamilton E, Kulyaba Y, Andabekov T. Camizestrant, a next‐generation oral SERD, vs fulvestrant in post‐menopausal women with advanced ER‐positive HER2‐negative breast cancer: results of the randomized, multi‐dose phase 2 SERENA‐2 trial. San Antonio Breast Cancer Symposium. 2022. Presented 08122022 (Abstract GS3‐02). [DOI] [PubMed]
  • 109. Oliveira M, Pominchuk D, Hamilton EP, Kulyaba Y, Melkadze T, Neven P, et al. Clinical activity of camizestrant, a next‐generation SERD, versus fulvestrant in patients with a detectable ESR1 mutation: exploratory analysis of the SERENA‐2 phase 2 trial. J Clin Oncol. 2023;41(16_suppl):1066. [Google Scholar]
  • 110. Turner N, Huang‐Bartlett C, Kalinsky K, Cristofanilli M, Bianchini G, Chia S, et al. Design of SERENA‐6, a phase III switching trial of camizestrant in ESR1‐mutant breast cancer during first‐line treatment. Future Oncol. 2023;19(8):559–573. [DOI] [PubMed] [Google Scholar]
  • 111. Goetz MP, Plourde P, Stover DG, Bagegni N, Vidal GA, Brufsky A, et al. Open‐label, randomized study of lasofoxifene (LAS) vs fulvestrant (Fulv) for women with locally advanced/metastatic ER+/HER2‐ breast cancer (mBC), an estrogen receptor 1 (ESR1) mutation, and disease progression on aromatase (AI) and cyclin‐dependent kinase 4/6 (CDK4/6i) inhibitors. Ann Oncol. 2022;33(suppl_7):S808–S869. [Google Scholar]
  • 112. Damodaran S, Plourde PV, Moore HCF, Anderson IC, Portman DJ. Open‐label, phase 2, multicenter study of lasofoxifene (LAS) combined with abemaciclib (Abema) for treating pre‐ and postmenopausal women with locally advanced or metastatic ER+/HER2− breast cancer and an ESR1 mutation after progression on prior therapies. J Clin Oncol. 2022;40(16_suppl):1022. [Google Scholar]
  • 113. Hartmaier RJ, Trabucco SE, Priedigkeit N, Chung JH, Parachoniak CA, Vanden Borre P, et al. Recurrent hyperactive ESR1 fusion proteins in endocrine therapy‐resistant breast cancer. Ann Oncol. 2018;29(4):872–880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Gou X, Anurag M, Lei JT, Kim B‐J, Singh P, Seker S, et al. Transcriptional reprogramming differentiates active from inactive ESR1 fusions in endocrine therapy‐refractory metastatic breast cancer. Cancer Res. 2021;81(24):6259–6272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Brett JO, Ritterhouse LL, Newman ET, Irwin KE, Dawson M, Ryan LY, et al. Clinical implications and treatment strategies for ESR1 fusions in hormone receptor‐positive metastatic breast cancer: a case series. Oncologist. 2023;28(2):172–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Sobhani N, Roviello G, Corona SP, Scaltriti M, Ianza A, Bortul M, et al. The prognostic value of PI3K mutational status in breast cancer: a meta‐analysis. J Cell Biochem. 2018;119(6):4287–4292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Mosele F, Stefanovska B, Lusque A, Tran Dien A, Garberis I, Droin N, et al. Outcome and molecular landscape of patients with PIK3CA‐mutated metastatic breast cancer. Ann Oncol. 2020;31(3):377–386. [DOI] [PubMed] [Google Scholar]
  • 118. André F, Ciruelos EM, Juric D, Loibl S, Campone M, Mayer IA, 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(2):208–217. [DOI] [PubMed] [Google Scholar]
  • 119. Rugo HS, Lerebours F, Ciruelos E, Drullinsky P, Ruiz‐Borrego M, Neven P, et al. Alpelisib plus fulvestrant in PIK3CA‐mutated, hormone receptor‐positive advanced breast cancer after a CDK4/6 inhibitor (BYLieve): one cohort of a phase 2, multicentre, open‐label, non‐comparative study. Lancet Oncol. 2021;22(4):489–498. [DOI] [PubMed] [Google Scholar]
  • 120. Suppan C, Graf R, Jahn S, Zhou Q, Klocker EV, Bartsch R, 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(3):456–463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Basu A, Lambring CB. Akt isoforms: a family affair in breast cancer. Cancer. 2021;13(14):3445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Turner NC, Oliveira M, Howell SJ, Dalenc F, Cortes J, Gomez Moreno HL, et al. Capivasertib in hormone receptor–positive advanced breast cancer. N Engl J Med. 2023;388(22):2058–2070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Turner N, Oliveira M, Howell SJ, Dalenc F, Cortés J, Gomez H, et al. Abstract GS3‐04: GS3‐04 Capivasertib and fulvestrant for patients with aromatase inhibitor‐resistant hormone receptor‐positive/human epidermal growth factor receptor 2‐negative advanced breast cancer: results from the phase III CAPItello‐291 trial. Cancer Res. 2023;83(5_Supplement):GS3‐04. [Google Scholar]
  • 124. Jhaveri K, Eli LD, Wildiers H, Hurvitz SA, Guerrero‐Zotano A, Unni N, et al. Neratinib + fulvestrant + trastuzumab for HR‐positive, HER2‐negative, HER2‐mutant metastatic breast cancer: outcomes and biomarker analysis from the SUMMIT trial. Ann Oncol. 2023;34(10):885–898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Loboda AP, Adonin LS, Zvereva SD, Guschin DY, Korneenko TV, Telegina AV, et al. BRCA mutations—the Achilles heel of breast, ovarian and other epithelial cancers. Int J Mol Sci. 2023;24(5):4982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Tutt ANJ, Garber JE, Kaufman B, Viale G, Fumagalli D, Rastogi P, et al. Adjuvant Olaparib for patients with BRCA1‐ or BRCA2‐mutated breast cancer. N Engl J Med. 2021;384(25):2394–2405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Tung NM, Garber JE. BRCA1/2 testing: therapeutic implications for breast cancer management. Br J Cancer. 2018;119(2):141–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Litton JK, Rugo HS, Ettl J, Hurvitz SA, Gonçalves A, Lee K‐H, et al. Talazoparib in patients with advanced breast cancer and a germline BRCA mutation. N Engl J Med. 2018;379(8):753–763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Robson M, Im S‐A, Senkus E, Xu B, Domchek SM, Masuda N, et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N Engl J Med. 2017;377(6):523–533. [DOI] [PubMed] [Google Scholar]
  • 130. Tung NM, Robson ME, Ventz S, Santa‐Maria CA, Nanda R, Marcom PK, et al. TBCRC 048: phase II study of Olaparib for metastatic breast cancer and mutations in homologous recombination‐related genes. J Clin Oncol. 2020;38(36):4274–4282. [DOI] [PubMed] [Google Scholar]
  • 131. Kingston B, Cutts RJ, Bye H, Beaney M, Walsh‐Crestani G, Hrebien S, et al. Genomic profile of advanced breast cancer in circulating tumour DNA. Nat Commun. 2021;12(1):2423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Marabelle A, Fakih M, Lopez J, Shah M, Shapira‐Frommer R, Nakagawa K, et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open‐label, phase 2 KEYNOTE‐158 study. Lancet Oncol. 2020;21(10):1353–1365. [DOI] [PubMed] [Google Scholar]
  • 133. Emens LA, Molinero L, Adams S, Rugo HS, Schneeweiss A, Diéras V, et al. 296P tumour mutational burden and clinical outcomes with first‐line atezolizumab and nab‐paclitaxel in triple‐negative breast cancer: exploratory analysis of the phase III IMpassion130 trial. Ann Oncol. 2020;31:S360–S361. [Google Scholar]
  • 134. Maio M, Ascierto PA, Manzyuk L, Motola‐Kuba D, Penel N, Cassier PA, et al. Pembrolizumab in microsatellite instability high or mismatch repair deficient cancers: updated analysis from the phase II KEYNOTE‐158 study. Ann Oncol. 2022;33(9):929–938. [DOI] [PubMed] [Google Scholar]
  • 135. Venetis K, Cursano G, Pescia C, D'Ercole M, Porta FM, Blanco MC, et al. Liquid biopsy: cell‐free DNA based analysis in breast cancer. J Liq Biopsy. 2023;1:100002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Lockwood CM, Borsu L, Cankovic M, Earle JSL, Gocke CD, Hameed M, et al. Recommendations for cell‐free DNA assay validations: a joint consensus recommendation of the Association for Molecular Pathology and College of American pathologists. J Mol Diagn. 2023;25(12):876–897. [DOI] [PubMed] [Google Scholar]
  • 137. Stenzinger A, Cuffel B, Paracha N, Vail E, Garcia‐Foncillas J, Goodman C, et al. Supporting biomarker‐driven therapies in oncology: a genomic testing cost calculator. Oncologist. 2023;28(5):e242–e253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Finan C, Gaulton A, Kruger FA, Lumbers RT, Shah T, Engmann J, et al. The druggable genome and support for target identification and validation in drug development. Sci Transl Med. 2017;9(383):eaag1166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Balic M, Wimmer K, Suppan C, Silovski T, Brunner C. Expert discussion: ASCO 2022. Breast Care. 2022;17(4):430–436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Dandachi N, Balic M. Deciphering the molecular landscape of metastatic lobular breast cancer. EBioMedicine. 2022;86:104385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Paradiso A, Bravaccini S, Pronzato P. Biomarkers: which ones do we really need today? Breast Care. 2023;18(4):315–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Stejskal P, Goodarzi H, Srovnal J, Hajdúch M, van't Veer LJ, Magbanua MJM. Circulating tumor nucleic acids: biology, release mechanisms, and clinical relevance. Mol Cancer. 2023;22(1):15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Santonja A, Cooper WN, Eldridge MD, Edwards PAW, Morris JA, Edwards AR, et al. Comparison of tumor‐informed and tumor‐naïve sequencing assays for ctDNA detection in breast cancer. EMBO Mol Med. 2023;15(6):e16505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Hays P. Trends in precision oncology and precision medicine 2.0. Advancing healthcare through personalized medicine. Cham: Springer International Publishing; 2021. p. 419–480. [Google Scholar]

Articles from Molecular Oncology are provided here courtesy of Wiley

RESOURCES