Abstract
In the era of precision oncology, the management of triple-negative breast cancer (TNBC) is rapidly changing and becoming more complicated with a variety of chemotherapy, immunotherapy, and targeted treatment options. Currently, TNBC treatment is based on prognostic and predictive factors including immunohistochemical biomarkers [e.g. programmed death-ligand 1 (PD-L1)] and germline BRCA mutations. Given the current limitation of existing biomarkers, liquid biopsies may serve as clinically useful tools to determine treatment efficacy and response in both the (neo)adjuvant and metastatic settings, for detecting early relapse, and for monitoring clonal evolution during treatment. In this review, we comprehensively summarize current and future liquid biopsy applications. Specifically, we highlight the role of circulating tumor cell characterization, circulating tumor DNA, and other preclinical liquid biopsy technologies including circulating exosomes, RNA liquid biopsy, and circulating immune-based biomarkers. In the near future, these biomarkers may serve to identify early disease relapse, therapeutic targets, and disease clonality for patients with TNBC in the clinical setting.
Key words: liquid biopsy, triple-negative breast cancer, precision oncology, novel biomarkers, circulating tumor DNA, circulating tumor cells
Highlights
-
•
Liquid biopsy as a potential noninvasive prognostic and predictive tool for TNBC.
-
•
Liquid biopsy can detect early relapse and monitor metastatic disease biology.
-
•
Complementary role of circulating tumor DNA and circulating tumor cell enumeration in TNBC management is discussed.
-
•
Circulating exosomes, miRNA, immune-based biomarkers serve as potential novel useful tools.
Introduction
Current triple-negative breast cancer biomarkers are limited
Triple-negative breast cancer (TNBC) is characterized by the absence of estrogen receptor (ER), progesterone receptor, and human epidermal growth factor receptor 2 (HER2). TNBC represents ∼10%-15% of all BCs, is more common in women younger than 40 years, and is the most aggressive and highly heterogeneous of all BC subtypes.1
Despite considerable efforts to identify novel prognostic and predictive biomarkers in TNBC, only a few have been proven useful in clinical trials.2 These include the germline BRCA1/2 mutation, present in ∼10%-20% of TNBC cases and leading to homologous recombination deficiency3, 4, 5, 6; activation of phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR)-dependent pathway; and PD-L1. Among metastatic TNBC (mTNBC) cases, ∼20%-38% of patients express PD-L1.7,8 The prognostic and predictive roles of elevated tumor-infiltrating lymphocyte (TIL) counts are well established, with their presence being associated with a better prognosis,9, 10, 11 improved response to immunotherapy, and higher rates of pathologic complete response (pCR).10,12 Importantly, prognosis also depends on lymphocyte localization, with the best outcomes observed in patients with TILs infiltrating both the tumor epithelium and stroma. By contrast, TILs restricted to the tumor margins or absent altogether are associated with the worst prognosis.13 Besides, the International Immuno-Oncology Biomarker Working Group recently demonstrated the favorable prognostic role of TIL abundance in BC tissue in patients with early-stage TNBC who did not receive adjuvant or neoadjuvant chemotherapy (NAC).14
Recent advancements in noninvasive liquid biopsy have demonstrated its potential role as a predictive biomarker in TNBC. We analyzed current applications of liquid biopsy as prognostic and predictive biomarkers in TNBC across various settings, highlighting it potential as a potential noninvasive, clinically useful tool. In addition, we explore future liquid biopsy applications, including technologies such as circulating exosomes, RNA liquid biopsy, and circulating immune-based biomarkers. A summary of ongoing clinical trials exploring liquid biopsy applications in TNBC are reported in Figure 1 and Tables 1 and 2.
Figure 1.
Visual summary of liquid biopsy clinical trials in triple-negative breast cancer.
Table 1.
Liquid biopsy clinical trials in early TNBC
| Trial | Description | Aims in liquid biopsy | Timing |
|---|---|---|---|
| Serial ctDNA Monitoring During Adjuvant Capecitabine in Early TNBC ClinicalTrials.gov Identifier: NCT04768426 |
A phase II trial of ctDNA monitoring during adjuvant capecitabine in patients with TNBC and residual disease following standard NAC |
|
Starting date 3 February 2021 Estimated duration 5 years |
| Apollo ClinicalTrials.gov Identifier: NCT04501523 |
A prospective, phase II trial using ctDNA to initiate post-operation boost therapy after NAC in TNBC | To improve the outcome of patients with TNBC using ctDNA to identify those with high relapse risk. ctDNA-positive patients will be randomized to receive boost therapy or standard therapy indicated in NCCN guidelines after NAC | Starting date 3 August 2020 Estimated duration 7 years |
| PERSEVERE ClinicalTrials.gov Identifier: NCT04849364 |
A phase II circulating tumor DNA enriched, genomically directed postneoadjuvant trial for patients with residual TNBC | Patients with residual TNBC disease after preoperative therapy will be assigned to one of three arms based on plasma ctDNA positivity and genomic marker(s) | Starting date 24 August 2021 Estimated duration 13 years |
| ZEST ClinicalTrials.gov Identifier: NCT04915755 |
A randomized phase III double-blinded study comparing the efficacy and safety of niraparib with placebo in participants with either HER2− BRCA-mutated or TNBC with molecular disease based on the presence of circulating tumor DNA after definitive therapy | Efficacy and safety comparison of niraparib with placebo in participants with HER2− breast cancer susceptibility gene mutation (BRCAmut) or TNBC) with molecular disease (ctDNA) following surgery or completion of adjuvant therapy | Starting date 28 June 2021 Estimated duration 8 years |
| Safe-De ClinicalTrials.gov Identifier: NCT05058183 |
Safe de-escalation of chemotherapy for stage 1 BC |
|
Starting date 5 June 2023 Estimated duration 6 years |
| ASPRIA ClinicalTrials.gov Identifier: NCT04434040 |
A single-arm phase II trial of atezolizumab with Sacituzumab Govitecan to prevent recurrence in TNBC |
|
Starting date 2 July 2020 Estimated duration 5 years |
| TARMAC ClinicalTrials.gov Identifier: NCT04771871 |
One-stage phase II of treatment response and microRNA profiles in patients with TNBC receiving standard chemotherapy |
|
Starting date 29 November 2021 Estimated duration 2 years |
| Artemis ClinicalTrials.gov Identifier: NCT04803539 |
A prospective, phase II trial using ctDNA to initiate post-operation boost therapy after adjuvant chemotherapy in TNBC |
|
Starting date 1 April 2021 Estimated duration 7 years |
| BreastImmune03 ClinicalTrials.gov Identifier: NCT03818685 |
A multicenter, randomized, open-label phase II study to evaluate the clinical benefit of a post-operative treatment associating radiotherapy + nivolumab + ipilimumab versus radiotherapy + capecitabine for patients with TNBC with residual disease after NAC |
Secondary outcome measures:
|
Starting date 2 July 2019 Estimated duration 5 years |
| OXEL ClinicalTrials.gov Identifier: NCT03487666 |
A pilot study of immune checkpoint or capecitabine or combination therapy as adjuvant therapy for TNBC with residual disease following NAC | Secondary outcome measures:
|
Starting date 21 July 2018 Estimated duration 4 years |
| RESPONSE ClinicalTrials.gov Identifier: NCT05020860 |
A phase II trial to correlate early clinical response to pathologic outcome with neoadjuvant systemic therapy in patients with EBC |
|
Starting date 18 April 2023 Estimated duration 6 years |
|
ClinicalTrials.gov Identifier: NCT03872388 |
Atorvastatin in treating patients with stage IIb-III TNBC who did not achieve a pathologic complete response after receiving neoadjuvant chemotherapy |
|
Starting date 14 January 2019 Estimated duration 4 years |
|
ClinicalTrials.gov Identifier: NCT02945579 |
Eliminating surgery or radiotherapy after systemic therapy in treating patients with HER2+ or TNBC |
Other outcome measures:
|
Starting date 20 January 2017 Estimated duration 9 years |
CTC, circulating tumor cell; ctDNA, circulating tumor DNA; NAC, neoadjuvant chemotherapy; NCCN, National Comprehensive Cancer Network; pCR, pathologic complete response; TNBC, triple-negative breast cancer.
Table 2.
Liquid biopsy clinical trials in metastatic TNBC
| Trial | Description | Aims in liquid biopsy | Timing |
|---|---|---|---|
| GIM25 CAPT ClinicalTrials.gov Identifier: NCT05266937 |
A phase II trial of atezolizumab plus carboplatin plus paclitaxel as first-line therapy in patients with metastatic PD-L1-positive TNBC |
|
Starting date 3 July 2020 Estimated duration 4 years |
|
ClinicalTrials.gov Identifier: NCT03990896 |
A phase II clinical trial to evaluate the effectiveness of talazoparib as a potential treatment for metastatic breast cancer with a BRCA1 or BRCA2 mutation |
|
Starting date 18 November 2021 Estimated duration 3 year |
| EPIK-B3 ClinicalTrials.gov Identifier: NCT04251533 |
A phase III, multicenter, randomized, double-blind, placebo-controlled study to assess the efficacy and safety of alpelisib (BYL719) in combination with nab-paclitaxel in patients with advanced TNBC with either PIK3CA mutation or PTEN loss without PIK3CA mutation |
|
Starting date 8 June 2020 Estimated duration 7 years |
|
ClinicalTrials.gov Identifier: NCT04345913 |
A phase I/II trial evaluating the safety and efficacy of eribulin in combination with copanlisib in patients with mBC |
|
Starting date 1 March 2021 Estimated duration 3 years |
| NADiR ClinicalTrials.gov Identifier: NCT04837209 |
A phase II study of niraparib, dostarlimab, and radiotherapy in metastatic, PD-L1 negative or immunotherapy-refractory TNBC | Other outcome measures:
|
Starting date 21 July 2021 Estimated duration 8 years |
| 4CAST ClinicalTrials.gov Identifier: NCT04947189 |
A phase Ib dose exploration and dose expansion, open-label, single-center study evaluating the safety and efficacy of INO-464 in combination with chemotherapy in patients with mBC |
|
Starting date 1 November 2021 Estimated duration 4 years |
| PAveMenT ClinicalTrials.gov Identifier: NCT04360941 |
A phase Ib study of palbociclib and avelumab in metastatic AR+ TNBC |
|
Starting date 11 August 2020-08-11 Estimated duration 4 years |
|
ClinicalTrials.gov Identifier: NCT02971761 |
A phase 2 clinical trial of the combination of pembrolizumab and selective androgen receptor modulator GTX-024 in patients with metastatic androgen receptor-positive TNBC |
|
Starting date 1 June 2017 Estimated duration 5 years |
|
ClinicalTrials.gov Identifier: NCT04176848 |
A phase II study of CFI-400945 and durvalumab in patients with advanced/metastatic Triple Negative Breast Cancer (TNBC) |
Primary outcome measures: Objective response rate of CFI-400945 given with durvalumab using RECIST 1.1 (timeframe: 24 months) Secondary outcome measures:
|
Starting date 19 December 2019 Estimated duration 3 years |
| RADIOLA ClinicalTrials.gov Identifier: NCT05340413 |
Predicting olaparib sensitivity in patients with unresectable locally advanced/metastatic HER2-negative breast cancer with BRCA1, BRCA2, PALB2, RAD51C or RAD51D mutations or RAD51-foci low test |
Secondary outcome measures: Capacity of the ctDNA drop after 4 weeks of treatment to predict the efficacy of olaparib in both cohorts |
Starting date 25 March 2022 Estimated duration 2 years |
AR, androgen receptor; cfDNA, cell-free DNA; CTCAE, Common Terminology Criteria for Adverse Events; ctDNA, circulating tumor DNA; mBC, metastatic breast cancer; NGS, next-generation sequencing; PD-L1, programmed death-ligand 1; TEX, tumor-derived exosome; TNBC, triple-negative breast cancer; VAF, variant allele frequency.
Liquid biopsy
Liquid biopsy involves the analysis of cell-free DNA (cfDNA), circulating tumor cells (CTCs), and other components such as extracellular vesicles and RNA released from tumor cells into blood, urine, cerebrospinal fluid, or bone marrow. There has been an increasing focus on incorporating liquid biopsies into clinical practice for cancer management. Mutations in cfDNA serve as highly specific markers for cancer, and this gave rise to the term ‘circulating tumor DNA’ (ctDNA). Plasma ctDNA, obtained through blood sampling, provides genetic information from both the primary tumor and metastatic sites.15 Notably, the half-life of cfDNA in circulation ranges from 16 min to 2.5 h, making ctDNA analysis a real-time snapshot of the disease, useful for targeted treatment selection and monitoring.1,16,17 The number of CTCs can stratify patients with metastatic BC (mBC) into two prognostic groups: stage IV indolent or stage IV aggressive, based on a threshold of ≥5 CTCs per 7.5 ml. Patients in the stage IV indolent group exhibit a longer median overall survival (OS).18
CTCs in early TNBC
Trapp’s group evaluated the potential prognostic role of CTCs in the routine follow-up of patients with BC. In the adjuvant SUCCESS A trial, the presence of CTCs was assessed before and two years after chemotherapy in 1087 patients with high-risk BC.19 Two years post-chemotherapy, 198 patients were CTC positive, and this found to be an independent, statistically significant prognostic factor for poor OS and disease-free survival (DFS). CTC status at the 2-year follow-up was independent of baseline CTC status. Moreover, patients who were CTC positive both at baseline and the 2-year follow-up had the worst OS and DFS.20
Measuring treatment response in the neoadjuvant and adjuvant setting
Several studies have investigated the clinical validity of CTCs in assessing the efficacy of NAC and treatment response. In patients with nonmetastatic TNBC, the presence of one or more CTCs after completing NAC was associated with significantly decreased OS and relapse-free survival21 Bidard et al.22 presented the results of a meta-analysis based on 21 studies that detected CTCs by CELLSEARCH in 2030 patients with early BC (EBC) before NAC and surgery, 25.8% of whom had TNBC. The authors demonstrated the prognostic role of CTCs in patients with EBC treated with NAC, independent of tumor subtypes, thereby enhancing current prognostic models.
CTC detection after surgery or during adjuvant therapy has been associated with a poor prognosis. To investigate the role of CTC monitoring in early TNBC, 286 women were enrolled in the study23 Considering CTC levels after surgery, patients with >5 CTCs per 7.5 ml of blood experienced worse outcomes compared to patients with <5 CTCs. Specifically, patients with higher CTC counts had a recurrence rate of 22.4% seven days post-surgery.
CTCs In metastatic TNBC
The prognostic value of CTC enumeration has been demonstrated in metastatic BC (mBC) patients using several studies with the Food and Drug Administration (FDA)-cleared CELLSEARCH (Menarini Silicon Biosystems) methodology. However, multiple technologies are available for the detection and enumeration of CTCs.18,24
The molecular characteristics of CTCs contribute to the formation of metastatic lesions, enabling these cells to invade tissues, survive in circulation, and extravasate to remote locations.24 In TNBC, multicellular CTC clusters are associated with a worse outcome compared to single CTCs. Specifically, CTC clusters can promote the development of metastatic disease 20 to100 times more frequently than a single CTCs.25
The presence of CTCs in patients with TNBC varies by stage, with a higher number of CTCs observed in mBC patients. Patients with CTCs at or above the threshold of 5 per 7.5 ml of blood before treatment, and those who fail to clear the cells during treatment, have a significantly worse outcome compared to those who maintain a CTC count of less than 5 after starting systemic therapy.26 Another study showed that patients with CTCs ≥5 per 7.5 ml, and/or those who experienced an increase in CTC counts 3-5 weeks and/or 6-8 weeks after the start of treatment had decreased PFS and OS.27 However, the SWOG S0500 trial did not demonstrate improved clinical outcomes in any mBC subtypes, including TNBC, when using early CTC-guided treatment changes.28 In a retrospective analysis, investigators classified patients into three prognostic sub-groups based on to baseline CTC enumeration, confirming that patients with higher CTC counts per 7.5 ml of blood had worse outcomes.29
In the TBCRC 001 trial, a direct comparison of two CTC enumeration methods, CELLSEARCH and IE/FC, showed higher concordance between the techniques. CTC enumeration 7-14 days after treatment initiation was correlated with time-to-progression, suggesting that CTCs may serve as an early marker of response to targeted therapy and a more reliable indicator of progression risk compared to baseline counts.30
Detecting targetable alterations
Analysis of CTCs also provides insights into the molecular characteristics of patients with mTNBC. Abreu et al.31 immunoisolated CTCs from a cohort of 32 patients with stage III and IV TNBC were analyzed using CELLSEARCH technology. The samples were characterized with a panel of genes related to cancer aggressiveness and plasticity. The expression signature identified in CTCs was associated with a hybrid epithelial–mesenchymal transition (EMT) status and a stem-like phenotype. These cells were detected in 42% of patients, and 3 of these patients also had CTC clusters. All CTC-positive patients were metastatic at the time of sample collection, and 26% of these patients had >5 CTCs. No correlation was found between the number of CTCs and other clinicopathologic features. Patients with >5 CTCs had worse PFS and OS. Cellular plasticity in CTCs from patients with TNBC, indicated by the expression of hybrid EMT and stem cell markers, was associated with poor prognosis and increased aggressiveness of these tumors.31
PIK3CA is an actionable cancer gene in mBC, as previously demonstrated in hormone receptor positive (HR+) mBC.32 Pestrin et al.33 analyzed the PIK3CA mutational status within single CTCs isolated from 39 patients with mBC, 20 of whom had samples enriched with ≥5 CTCs. PIK3CA mutations were identified in six patients, and discordance between the PIK3CA status of the primary BC (wild type) and the matched CTC (exon 20 mutation) was observed in only one patient. This suggests a proof of concept for the potential utility of liquid biopsy in this setting.
Detecting tumor heterogeneity
The characteristics of CTCs change during tumor cell spreading, mainly due to the EMT process, which together with expression of stemness markers could facilitate chemotherapy resistance and promote their capacity to metastasize.31 Rothé et al.34 investigated mBC heterogeneity by analyzing CTCs matched with synchronous tumor biopsies from three patients with mBC. Considering tumor mutational burden (TMB) in tumor biopsies in the patient with TNBC, 38% of all single-nucleotide variants (SNVs) found in the CTC samples were also present in tumor biopsies. Interestingly, all SNVs in cancer driver genes identified in tumor biopsies were also detected in CTCs. Single CTC genomic analysis revealed some driver aberrations not found in the tumor sample and provided additional insights into the clonal/subclonal distribution of all aberrations identified.
ctDNA in early TNBC
Prognostic value
In locally advanced TNBC, there has been a continual escalation of treatment approaches, as supported by two landmark trials, the KEYNOTE-522 trial35 and the CREATE-X trial in the adjuvant setting.36 In this context, an emergent need is treatment tailoring in early TNBC according to the risk of recurrence,37 as already expressed in the St. Gallen International Expert Consensus Conference in 2017.38 ctDNA and CTCs were considered potentially useful biomarkers to guide treatment de-escalation.37
Notably, ctDNA may be detectable in early BC using high-sensitivity minimal residual disease (MRD) assays. The ctDNA detection after surgical resection reflects the persistence of micrometastatic residual disease, which is otherwise clinically undetectable.39,40 Therefore ctDNA could potentially serve as a marker of residual disease in patients with TNBC to guide therapeutic decisions after neoadjuvant therapy. Based on this potential, the Q-CROC-03 trial examined patients with TNBC undergoing NAC with biopsies carried out before chemotherapy and after chemotherapy as well as blood samples before, during, and after NAC to determine molecular factors of response or resistance to standard NAC. A slight increase in ctDNA levels was predictive of incomplete pCR, and the absence of ctDNA levels at this presurgical specimen was associated with long-term relapse-free survival and OS, with a prognostic value similar to pCR status.41
Measuring efficacy of neoadjuvant therapy
Further information can be obtained from liquid biopsy to anticipate NAC response. Magbanua et al.42 provided evidence regarding the role of ctDNA as a predictive biomarker for response and outcome in the I-SPY 2 trial, collecting blood samples before treatment (T0), 3 weeks after paclitaxel initiation (T1), between paclitaxel and anthracycline regimens (T2), and before surgery (T3). At T0, 73% of the patients had detectable ctDNA, which decreased over time (T1 35%; T2 14%; and T3 9%). Patients who remained ctDNA positive at T1 were significantly more likely to have residual disease after NAC (83% non-pCR) compared with those who cleared ctDNA (52% non-pCR). All patients who achieved pCR after NAC were ctDNA negative. In the non-pCR group, ctDNA-positive patients had a significantly increased risk of metastatic recurrence; notably, ctDNA-negative patients had excellent outcome, similar to those who achieved pCR. This suggests that rising ctDNA before surgery may be an indicator of increased risk of relapse and may characterize patients who might benefit from treatment intensification before surgery to achieve a pCR and prevent distant relapse.
Early relapse detection
ctDNA detection after surgery, along with CTC enumeration, may be used to detect MRD and assess which patients may later develop disease recurrence. In a cohort of 55 women with EBC at risk of cancer relapse treated with NAC independently of BC subtypes, plasma samples were collected at baseline, 2-4 weeks after the operation, and then every 6 months during follow-up. MRD was detectable before standard clinical relapse using mutation tracking in serial samples, which increased sensitivity for predicting clinical relapse with a median lead time of 7.9 months.43 Importantly, the genetic events of the primary tumor may differ from those found in the metastatic disease.44 To date, many companies are investing in the development of liquid biopsy platforms to detect ctDNA in the MRD setting.45 In this context, two different approaches exist: the tumor-informed approach looking for targeted variation [Signatera, Natera (platform)] and the tumor-uninformed approach searching de novo variations [Guardant Reveal, Guardant Health (assay)].46 The tumor-informed approach requires a priori knowledge of the tumor profile by tumor tissue analysis, and the MRD assay is patient specific, but this approach could potentially miss clonal variants different from the primary tumor sample. In the tumor-uninformed approach, a broad next-generation sequencing panel is used to look for MRD combined with methylation analysis, but background noise from nontumor-derived mutations could influence the results.45
Tumor-informed approaches with personalized PCR were utilized in the following trials to detect MRD starting from tumor tissue analysis. In a cohort of 170 patients with EBC, detection of MRD was associated with future clinical relapse for all major BC subtypes, with ctDNA detected before relapse in 22 of 23 patients.47 In a prospective trial of 42 patients with TNBC, ctDNA detection after NAC was associated with increased relapse risk, regardless of other clinicopathological features. Considering recurrent cases, ctDNA was detected 13 months before clinical diagnosis.43 In another trial, samples from 38 patients with early-stage TNBC with matched tumor and plasma were analyzed. Of the 33 patients who had a mutation identified in their primary tumor, mutations in ctDNA were detected in 4 patients, who experienced a rapid recurrence. Next-generation sequencing of ctDNA in patients with TNBC with residual disease after NAC could predict recurrence with high specificity but moderate sensitivity.48 With the cTRAK TN trial, Turner et al.49 confirmed that early ctDNA detection was associated with high rates of metastatic disease. The trial was unable to address the co-primary endpoint of pembrolizumab’s effect on ctDNA clearance due to only few patients receiving the programmed cell death protein 1 (PD-1) inhibitor treatment. However, the trial demonstrated the clinical critical need to have more frequent timepoints and higher sensitivity assays in ctDNA detection. Given the timing of plasma sampling and assay sensitivity, the cTRAK TN trial did not validate the approach that ctDNA MRD could be detected with sufficient lead time before radiographic evidence of disease to impact clinical management. This demonstrates the urgent need to optimize sampling timepoints and assay performance, and also, in parallel, develop more effective therapies for micrometastatic disease in the future. This is perhaps a minor setback in the field, but the assay utilized was far from optimal.
ctDNA in metastatic TNBC
Prognostic value
In a retrospective trial of 164 patients with mTNBC, the presence of a ctDNA fraction >10% was associated with worse outcomes, regardless of clinicopathological data.50 The strong prognostic tumor-agnostic impact of ctDNA was demonstrated in terms of calculating the circulating tumor fraction.51 It was also demonstrated that similar patterns of chromosomal alterations exist in primary and mTNBC, suggesting that most somatic copy number alterations occur early in tumorigenesis in TNBC.52
Detecting targetable alterations
Plasma ctDNA genotyping can identify mutations in potentially ‘actionable’ cancer genes.53 In a cohort of patients with mBC, considering all subtypes, with identified BRCA1/2 mutations using cfDNA genotyping, 13.5% of patients had BRCA1/2 somatic mutations, while 4% had known germline mutations. Sensitivity to poly (ADP-ribose) polymerase inhibitors (PARPis) in CTC-derived models was also explored. Some cell lines with somatically acquired driver variants showed increased sensitivity to PARPi, but others did not, suggesting differential response to PARPi based on BRCA1/2 mutations detected using cfDNA.54 This is further being evaluated in a phase II clinical trial to assess talazoparib efficacy in 30 patients with mBC presenting with pathogenic somatic BRCA1/2 mutations detected in cfDNA.55
In the plasmaMATCH trial,56 ctDNA was used to direct therapy in patients with mBC to screen for rare actionable mutations. Among the 1051 patients, 17% of patients had an mTNBC, with a small subset of patients receiving neratinib for a HER2 mutation or capivasertib for an AKT pathway activating mutation. In cohort E, including patients with TNBC without targetable mutation and treated with olaparib plus ceralasertib (ataxia telangiectasia and Rad3-related kinase - inhibitor), response rate to treatment did not meet prespecified criteria for efficacy.57 However, this trial demonstrated sufficient promise for ctDNA testing to guide selection of targeted therapies.56
Detecting tumor heterogeneity
Liquid biopsy could also detect intrapatient variability with respect to genomic alterations of primary and metastatic lesions. In a large clinically annotated cohort of patients with mBC who underwent ctDNA evaluation, the genomic landscape of patients with mTNBC (22% of population) was described, demonstrating genetic heterogeneity of mBC in blood.58 The mutant allele frequency (MAF) of the highest variant in the TNBC cohort was significantly higher compared with HR+ or HER2 positive (HER2+) patients. The most common SNVs were TP53 and PIK3CA, and the most frequent copy number variants were MYC, CCNE1, and PIK3CA. Confirming these data, Page et al.59 reported SNVs detected in ESR1 in three patients with mTNBC to provide evidence for small ER+ clones as a potential resistance mechanism. These data support the concept of increasing tumor heterogeneity in mTNBC compared with other mBC subtypes, with liquid biopsy serving as a potentially useful clinical tool. Data from a retrospective cohort of patients with mBC highlight the role of ctDNA in monitoring tumor genetic evolution. Increasing MAF (≥3.3) and number of alterations (≥4) were associated with disease progression.16
Complementary role of CTDNA and CTC enumeration
Early relapse detection in early TNBC
Detection of ctDNA and CTCs in patients with early-stage TNBC after NAC was independently associated with disease recurrence and could represent a stratification factor for future postneoadjuvant trials. In the BRE12-158 trial that randomized 196 patients with early-stage TNBC with residual disease after NAC to receive postneoadjuvant genomically directed therapy versus treatment of physician’s choice, blood samples were collected at the time of treatment assignment. Overall, the presence of ctDNA and CTC after NAC was associated with significantly inferior distant DFS and OS.60
However, there is still an unmet need to establish a CTC threshold for recurrence, improve MRD detection sensitivity, and determine the optimal timepoints for blood sample collection.61 In 2022, the ESMO Precision Medicine Working Group could not recommend the use of molecular residual disease/molecular relapse detection in clinical practice due to the lack of validated assays for both ctDNA and CTC detection.62
Treatment response monitoring and tumor heterogeneity in mTNBC
Recently, the clinical value of concomitant analysis of CTCs and ctDNA was investigated in a large prospective biomarker study (COMET) conducted in 189 patients with HER2 negative (HER2−) mBC treated with first-line chemotherapy, including 23% of patients with mTNBC. To understand the complementary contribution of CTCs and ctDNA, both biomarkers were analyzed before treatment and after 4 weeks of first-line chemotherapy. The CTC enumeration at 4 weeks (≥5 CTC/7.5 ml) after treatment initiation determined the best prognostic model for PFS with ctDNA variant allele frequency (VAF) at baseline, grade, and tumor subtype. CTCs and ctDNA provided complementary clinical information.63 However, their role as monitoring tools needs further prospective validation, as underlined in the ESMO recommendations.62 In addition, Venesio et al.53 demonstrated the complementary role of ctDNA and CTCs in describing cancer heterogeneity due to substantial differences between primary tumor and metastatic/recurrent disease.
Immunotherapy and liquid biopsy
The need to develop tumor-based predictive biomarkers is still unmet for patients with TNBC treated with immunotherapy.2 T-cell expansion and ctDNA analysis are promising tools, while the prognostic and predictive value of CTC enumeration is being explored with respect to PD-L1 expression on CTCs.64 TMB represents a pan-cancer feature associated with benefits from immunotherapy.2,64 It is defined by the number of somatic mutations per DNA megabase pair (mut/Mb) and may serve as a surrogate marker for the production of neoantigens, which can lead to increased T-cell infiltration.2,65 In BC, the role of TMB in tumor immunogenicity remains unclear, as it is generally low to intermediate, and the proportion of patients with high TMB (≥10 mut/Mb, TMB-H) is different across BC groups.65, 66, 67 Tumor whole exome sequencing or gene panel sequencing data from 3969 patients with primary or mBC were analyzed to evaluate the frequency, mutational patterns, and genomic profile of hypermutated BC. Hypermutation was identified in 5% of all cases of BC with enrichment in metastatic tumors (mBC 8.4% versus primary BC 2.9%). The median TMB was significantly higher in TNBC (1.8 mut/Mb) compared with HR+ (1.1 mut/Mb) or HER2+ BC (1.3 mut/Mb). According to preliminary data from this study, hypermutated BC would be more likely to benefit from PD-1 inhibitors.66 However, TMB-H has not been consistently demonstrated as a predictive factor for immune checkpoint inhibitors (ICIs) across all cancer types. This is due to variability in how different tumors are classified as TMB-H according to cancer type,68 despite the FDA’s tissue-agnostic approval for this threshold.69 Only a small proportion of TNBC cases (8%-10%) have a high TMB. Bianchini et al.2 suggested improving the predictive value of TMB in TNBC by considering specific mutational signatures, the number of clonal mutations, dinucleotide variants, indels, or additional genomic alterations.
Liquid-based TMB is still under investigation; several studies have explored the role of ctDNA in determining blood-based TMB as a potential clinically actionable biomarker.70 Considering both pretreatment and on-treatment VAF in blood samples across 16 advanced-stage tumor types from three phase I/II trials of durvalumab (with or without the anti-CTLA-4 therapy tremelimumab), the ‘molecular response’ was defined using ctDNA to predict long-term survival and identified early responders among patients with initially radiologically stable disease.71 In the INSPIRE trial, a correlation was demonstrated between baseline ctDNA concentration and clinical response, clinical and survival benefit, with a stronger association utilizing ctDNA dynamics during immunotherapy treatment.72 In the NIMBUS trial patients with HER2− mBC with tissue TMB-H experienced benefit from nivolumab plus ipilimumab therapy and investigators identified a subgroup of patients with TMB ≥14 Mut/Mb with higher ORR, underlining the unmet need of the optimal TMB cut-off in predicting benefit to ICI treatment in mBC.73
In the SAFIR02 BREAST IMMUNO study, CD274, the gene encoding PD-L1, emerged as a new biomarker related to PD-L1 expression. This cancer cell-extrinsic feature is associated with greater benefit from ICIs in patients with mTNBC, presenting new challenges and opportunities for liquid biopsy applications.74
Emerging or preclinical liquid biopsy biomarkers
RNA liquid biopsy targets
MicroRNA (miRNA) is a unique miRNA signature secreted into the bloodstream from the tumor’s primary site that could be used as a liquid biopsy target. The benefits of using miRNA-based assays include insight into specific genetic information throughout tumorigenesis in multiple bodily fluids due to their stability throughout the circulation.75 These targets have been investigated in several preclinical trials of patients with BC but have not been extensively evaluated in clinical trials. One of the earliest studies attempted to find a unique miRNA signature in the serum of patients with BC independent of subtype, and reported changes in miRNA expression in patients diagnosed with BC compared with healthy controls.76 These data supported the preclinical findings of varying RNA expression in multiple types of BC cell lines in vitro, including an increase in specific miRNA in TNBC cell lines.77 Assessment of cell-bound miRNA showed unique miRNA signatures in peripheral blood mononuclear cells (PBMCs) due to their supportive effect in the tumor microenvironment.78 There were different miRNA expressions of PBMCs between triple-positive and TNBC cells. The miRNA in the PMBCs were different from the miRNA profile from the tumor site. Cell-free miRNA signatures were different from each other in luminal BC and TNBC, creating the possibility of differentiating tumors by liquid biopsy.79 Alterations in miRNA from tumor and tumor supporting cells are also observed, but there is no consensus on which miRNA changes are specific to define a new TNBC diagnosis.
Another potential application of miRNA liquid biopsies is to serve as a prognostic biomarker after diagnosis. For example, an increase in total serum RNA in nonmetastatic BC was found compared with mBC. In addition, metastatic tumors were more likely to express a different miRNA signature compared with nonmetastatic cancer80 and changes in miRNA expression could be used to predict OS in patients with TNBC.81 The miRNA profile secreted into the bloodstream is likely different, limiting the utility of these signatures in a clinical-based liquid biopsy without additional validation. Besides, miRNA expression was associated with a decrease in OS and an increase in tumor reoccurrence.82 Multiple miRNA signatures have been identified as potential prognostic biomarkers but without a defined miRNA profile that predicts TNBC progression, limiting their current clinical utility.83
Circulating exosomes and liquid biopsy
Another potential biomarker is circulating exosomes, which are exocytosed vesicles secreted by tumor cells into the bloodstream. One major advantage of developing an exosome-based assay for cancer diagnosis and monitoring is the enhanced exosome stability in a variety of environments, such as blood, urine, or saliva.84 These vesicles package a variety of genetic material and proteins, potentially allowing for analysis of multiple targets from a single sample that mimics the tumor at its origin. Exosomes isolated from patients with BC independent of subtypes have a higher concentration of CD45+ leukocyte-derived microparticles, carcinoembryonic antigen, and CA15-3 compared with healthy people,85 as well as an increase in the expression of antiapoptotic proteins and their splice variants; additionally, the circulating expression of these proteins correlated with the expression at the primary tumor site.86 Exosome-bound miRNAs have an advantage over free miRNAs due to further enhanced stability, increased quantity, and more literature supporting their use as a biomarker.87 Eichelser et al.88 found an increase in specific miRNA signatures and surface proteins on exosomes in sera collected from patients with HR− BC. Considering 18 different miRNA compounds between TNBC and HER2+ malignancies, 9 had a differential expression between patients with BC and healthy people. Elevated miRNA levels in sera collected from TNBC and HER2+ BC correlated with patients who were more likely to have pCR.89 Patients with TNBC had upregulation of specific exosomal miRNA compared with healthy controls. Besides, there was a difference in the miRNA expression of patients who had a BC recurrence after treatment and those who did not develop recurrence.90 Exosomal miRNA may be helpful in predicting metastasis: exosome miRNA expression changes between patients with mBC and patients without metastasis.91 Exosomes can also contain tumor-associated DNA; however, the benefit of measuring tumor DNA in exosomes remains unclear. ctDNA was more sensitive to detecting the presence of DNA expression compared with exosomal DNA.92 Exosome-based liquid biopsies are currently being evaluated due to their potential role in TNBC diagnosis prognostication and therapeutic monitoring.93
Circulating immune-based biomarkers as liquid biopsy targets
A common feature of the tumor microenvironment is the presence of immunomodulators and tumor-associated immune cells. Thus monitoring these changes in the immune response may allow clinicians to monitor the host response against BC.94,95 Liquid biopsies may enable the investigation of PD-L1 expression heterogeneity in TNBC between the primary site and distant metastatic sites.96,97 Increased PD-L1 expression on the surface of CTCs was useful in predicting reduced OS in patients with mBC.98 Other immunogenic proteins have shown differences in patients diagnosed with cancer as well. Increased levels of early myeloid-derived suppressive cells (eMDSC) as well pro-eMDSC proteins in patients with TNBC were associated with a worse response to NAC.99 Previous studies using all subtypes of BC samples have shown an increase in CD117- and CD11b-expressing granulocytes at the time of diagnosis with BC.100 Analysis of the serum samples after surgical resection and radiotherapy showed decreased expression, suggesting that reducing the tumor burden reduces the measurable immune response. In addition, a decreased serum concentration of myeloid-derived suppressive cells, PD-L1-expressing T lymphocytes, and regulatory T cells was associated with a clinical benefit across all BC subtypes.101 Tumor-associated macrophages (TAMs) are recognized as having a significant role in tumorigenesis and immune evasion in BC.102 Concentrations of circulating CD163-positive TAMs have potential as a future serum immune cell biomarker.103 Monitoring serum TAM concentrations could demonstrate immune response to ongoing cancer therapies and help guide treatment with antimacrophage antibody therapies in patients with TNBC.
Conclusions
Liquid biopsy has the potential to be a useful tool in clinical practice to assess treatment response in all settings, including detecting early relapse and monitoring disease biology in the metastatic setting. We presented the current state of the art and future applications of liquid biopsy in TNBC. There are still significant unmet clinical needs to validate the role of ctDNA and CTCs in identifying therapeutic targets and clonality in TNBC serum markers in the metastatic setting. In addition, further research is needed to explore the role of CTC characterization and other liquid biopsy technologies, such as RNA liquid biopsy, circulating exosomes, and circulating immune-based biomarkers.
Acknowledgments
Funding
This work was supported by the Italian Ministry of Health–Ricerca Corrente (no grant number).
Disclosure
AAD received honoraria for advisory boards from Pfizer and Biotheranostics, all outside the submitted work. LG reports receiving consulting and advisory board honoraria from AstraZeneca, Daiichi Sankyo, Menarini Stemline, Eli Lilly, GlaxoSmithKline, Incyte, Novartis, Pfizer, and research funding from Menarini Silicon Biosystems, all outside the submitted work. FP reports receiving grants and research support from AstraZeneca, Eisai, and Roche; and receipt of honoraria or consultation fees from Amgen, AstraZeneca, Daiichi Sankyo, Celgene, Eisai, Eli Lilly, Gilead, GSK, Ipsen, MSD, Novartis, Pierre-Fabre, Pfizer, Roche, Seagen, Takeda, Menarini, and Viatris, all outside the submitted work. RM, JS, LP, and SB have declared no conflicts of interest.
References
- 1.Vagia E., Mahalingam D., Cristofanilli M. The landscape of targeted therapies in TNBC. Cancers (Basel) 2020;12:916. doi: 10.3390/cancers12040916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Bianchini G, De Angelis C, Licata L, Gianni L. Treatment landscape of triple-negative breast cancer - expanded options, evolving needs. Nat Rev Clin Oncol. 2022 Feb;19(2):91-113. 10.1038/s41571-021-00565-2. Epub 2021 Nov 9. PMID: 34754128. [DOI] [PubMed]
- 3.Robson M., Im S.-A., Senkus E., et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N Engl J Med. 2017;377:523–533. doi: 10.1056/NEJMoa1706450. [DOI] [PubMed] [Google Scholar]
- 4.Litton J.K., Rugo H.S., Ettl J., et al. Talazoparib in patients with advanced breast cancer and a germline BRCA mutation. N Engl J Med. 2018;379:753–763. doi: 10.1056/NEJMoa1802905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Diéras V., Han H.S., Kaufman B., et al. Veliparib with carboplatin and paclitaxel in BRCA-mutated advanced breast cancer (BROCADE3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2020;21:1269–1282. doi: 10.1016/S1470-2045(20)30447-2. [DOI] [PubMed] [Google Scholar]
- 6.Tutt A.N.J., Garber J.E., Kaufman B., et al. Adjuvant olaparib for patients with BRCA1 – or BRCA2-mutated breast cancer. N Engl J Med. 2021;384:2394–2405. doi: 10.1056/NEJMoa2105215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Davis A.A., Patel V.G. The role of PD-L1 expression as a predictive biomarker: an analysis of all US Food and Drug Administration (FDA) approvals of immune checkpoint inhibitors. J Immunother Cancer. 2019;7:1–8. doi: 10.1186/s40425-019-0768-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Sabatier R., Finetti P., Mamessier E., et al. Prognostic and predictive value of PDL1 expression in breast cancer. Oncotarget. 2015;6:5449–5464. doi: 10.18632/oncotarget.3216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Loi S., Drubay D., Adams S., et al. Tumor-infiltrating lymphocytes and prognosis: a pooled individual patient analysis of early-stage triple-negative breast cancers. J Clin Oncol. 2019;37:559–569. doi: 10.1200/JCO.18.01010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ibrahim E.M., Al-Foheidi M.E., Al-Mansour M.M., Kazkaz G.A. The prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancer: a meta-analysis. Breast Cancer Res Treat. 2014;148:467–476. doi: 10.1007/s10549-014-3185-2. [DOI] [PubMed] [Google Scholar]
- 11.Loi S., Michiels S., Salgado R., et al. Tumor infiltrating lymphocytes are prognostic in triple negative breast cancer and predictive for trastuzumab benefit in early breast cancer: results from the FinHER trial. Ann Oncol. 2014;25:1544–1550. doi: 10.1093/annonc/mdu112. [DOI] [PubMed] [Google Scholar]
- 12.de Jong V.M.T., Wang Y., Ter Hoeve N.D., et al. Prognostic value of stromal tumor-infiltrating lymphocytes in young, node-negative, triple-negative breast cancer patients who did not receive (neo)adjuvant systemic therapy. J Clin Oncol. 2022;40:2361–2374. doi: 10.1200/JCO.21.01536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Gruosso T., Gigoux M., Manem V.S.K., et al. Spatially distinct tumor immune microenvironments stratify triple-negative breast cancers. J Clin Invest. 2019;129:1785–1800. doi: 10.1172/JCI96313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Leon-Ferre R.A., Jonas S.F., Salgado R., et al. Tumor-infiltrating lymphocytes in triple-negative breast cancer. J Am Med Assoc. 2024;331:1135–1144. doi: 10.1001/jama.2024.3056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Diaz L.A., Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol. 2014;32:579–586. doi: 10.1200/JCO.2012.45.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Jacob S., Davis A.A., Gerratana L., et al. The use of serial circulating tumor DNA to detect resistance alterations in progressive metastatic breast cancer. Clin Cancer Res. 2021;27:1361–1370. doi: 10.1158/1078-0432.CCR-20-1566. [DOI] [PubMed] [Google Scholar]
- 17.Gerratana L., Davis A.A., Zhang Q., et al. Longitudinal dynamics of circulating tumor cells and circulating tumor DNA for treatment monitoring in metastatic breast cancer. JCO Precis Oncol. 2021:943–952. doi: 10.1200/PO.20.00345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Cristofanilli M., Pierga J.Y., Reuben J., et al. The clinical use of circulating tumor cells (CTCs) enumeration for staging of metastatic breast cancer (MBC): International Expert Consensus paper. Crit Rev Oncol Hematol. 2019;134:39–45. doi: 10.1016/j.critrevonc.2018.12.004. [DOI] [PubMed] [Google Scholar]
- 19.Friedl T.W.P., Fehm T., Müller V., et al. Prognosis of patients with early breast cancer receiving 5 years vs 2 years of adjuvant bisphosphonate treatment: a phase 3 randomized clinical trial. JAMA Oncol. 2021;7:1149–1157. doi: 10.1001/jamaoncol.2021.1854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Trapp E., Janni W., Schindlbeck C., et al. Presence of circulating tumor cells in high-risk early breast cancer during follow-up and prognosis. J Natl Cancer Inst. 2019;111:380–387. doi: 10.1093/jnci/djy152. [DOI] [PubMed] [Google Scholar]
- 21.Hall C., Karhade M., Laubacher B., et al. Circulating tumor cells after neoadjuvant chemotherapy in stage I-III triple-negative breast cancer. Ann Surg Oncol. 2015;22:552–558. doi: 10.1245/s10434-015-4600-6. [DOI] [PubMed] [Google Scholar]
- 22.Bidard F.C., Michiels S., Riethdorf S., et al. Circulating tumor cells in breast cancer patients treated by neoadjuvant chemotherapy: a meta-analysis. J Natl Cancer Inst. 2018;110:560–567. doi: 10.1093/jnci/djy018. [DOI] [PubMed] [Google Scholar]
- 23.Zhang Y., Lv Y., Niu Y., Su H., Feng A. Role of circulating tumor cell (CTC) monitoring in evaluating prognosis of triple-negative breast cancer patients in China. Med Sci Monit. 2017;23:3071–3079. doi: 10.12659/MSM.902637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ivanova E., Ward A., Wiegmans A.P., Richard D.J. Circulating tumor cells in metastatic breast cancer: from genome instability to metastasis. Front Mol Biosci. 2020;7:134. doi: 10.3389/fmolb.2020.00134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Schuster E., Taftaf R., Reduzzi C., et al. Better together: circulating tumor cell clustering in metastatic cancer. Trends Cancer. 2021;7:1020–1032. doi: 10.1016/j.trecan.2021.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Liu M.C., Shields P.G., Warren R.D., et al. Circulating tumor cells: a useful predictor of treatment efficacy in metastatic breast cancer. J Clin Oncol. 2009;27:5153–5159. doi: 10.1200/JCO.2008.20.6664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bidard F.C., Peeters D.J., Fehm T., et al. Clinical validity of circulating tumour cells in patients with metastatic breast cancer: a pooled analysis of individual patient data. Lancet Oncol. 2014;15:406–414. doi: 10.1016/S1470-2045(14)70069-5. [DOI] [PubMed] [Google Scholar]
- 28.Smerage J.B., Barlow W.E., Hortobagyi G.N., et al. Circulating tumor cells and response to chemotherapy in metastatic breast cancer: SWOG S0500. J Clin Oncol. 2014;32:3483–3489. doi: 10.1200/JCO.2014.56.2561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Paoletti C., Miao J., Dolce E.M., et al. Circulating tumor cell clusters in patients with metastatic breast cancer: a SWOG S0500 translational medicine study. Clin Cancer Res. 2019;25:6089–6097. doi: 10.1158/1078-0432.CCR-19-0208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Magbanua M.J.M., Carey L.A., DeLuca A., et al. Circulating tumor cell analysis in metastatic triple-negative breast cancers. Clin Cancer Res. 2015;21:1098–1105. doi: 10.1158/1078-0432.CCR-14-1948. [DOI] [PubMed] [Google Scholar]
- 31.Abreu M., Cabezas-Sainz P., Pereira-Veiga T., et al. Looking for a better characterization of triple-negative breast cancer by means of circulating tumor cells. J Clin Med. 2020;9:353. doi: 10.3390/jcm9020353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.André F., Ciruelos E., Rubovszky G., et al. SOLAR-1 Study Group Alpelisib for PIK3CA-mutated, hormone receptor-positive advanced breast cancer. N Engl J Med. 2019;380:1929–1940. doi: 10.1056/NEJMoa1813904. [DOI] [PubMed] [Google Scholar]
- 33.Pestrin M., Salvianti F., Galardi F., et al. Heterogeneity of PIK3CA mutational status at the single cell level in circulating tumor cells from metastatic breast cancer patients. Mol Oncol. 2015;9:749–757. doi: 10.1016/j.molonc.2014.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Rothé F., Venet D., Peeters D., et al. Interrogating breast cancer heterogeneity using single and pooled circulating tumor cell analysis. NPJ Breast Cancer. 2022;8:1–8. doi: 10.1038/s41523-022-00445-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Schmid P., Cortes J., Pusztai L., et al. Pembrolizumab for early triple-negative breast cancer. N Engl J Med. 2020;382:810–821. doi: 10.1056/NEJMoa1910549. [DOI] [PubMed] [Google Scholar]
- 36.Masuda N., Lee S.-J., Ohtani S., et al. Adjuvant capecitabine for breast cancer after preoperative chemotherapy. N Engl J Med. 2017;376:2147–2159. doi: 10.1056/NEJMoa1612645. [DOI] [PubMed] [Google Scholar]
- 37.Abuhadra N., Stecklein S., Sharma P., Moulder S. Early-stage triple-negative breast cancer: time to optimize personalized strategies. Oncologist. 2022;27:30–39. doi: 10.1093/oncolo/oyab003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Curigliano G., Burstein H.J., Winer E.P., et al. Vol. 28. Ann Oncol; 2017. pp. 1700–1712. (De-escalating and escalating treatments for early-stage breast cancer: the St. Gallen International Expert Consensus Conference on the Primary Therapy of Early Breast Cancer 2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Riva F., Bidard F.C., Houy A., et al. Patient-specific circulating tumor DNA detection during neoadjuvant chemotherapy in triple-negative breast cancer. Clin Chem. 2017;63:691–699. doi: 10.1373/clinchem.2016.262337. [DOI] [PubMed] [Google Scholar]
- 40.Siravegna G., Sartore-Bianchi A., Nagy R.J., et al. Plasma HER2 (ERBB2) copy number predicts response to HER2-targeted therapy in metastatic colorectal cancer. Clin Cancer Res. 2019;25:3046–3053. doi: 10.1158/1078-0432.CCR-18-3389. [DOI] [PubMed] [Google Scholar]
- 41.Cavallone L., Aguilar-Mahecha A., Lafleur J., et al. Prognostic and predictive value of circulating tumor DNA during neoadjuvant chemotherapy for triple negative breast cancer. Sci Rep. 2020;10:1–13. doi: 10.1038/s41598-020-71236-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Magbanua M.J.M., Swigart L.B., Wu H.T., et al. Circulating tumor DNA in neoadjuvant-treated breast cancer reflects response and survival. Ann Oncol. 2021;32:229–239. doi: 10.1016/j.annonc.2020.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Ortolan E., Appierto V., Silvestri M., et al. Blood-based genomics of triple-negative breast cancer progression in patients treated with neoadjuvant chemotherapy. ESMO Open. 2021;6:100086. doi: 10.1016/j.esmoop.2021.100086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Garcia-Murillas I., Schiavon G., Weigelt B., et al. Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer. Sci Transl Med. 2015;7 doi: 10.1126/scitranslmed.aab0021. [DOI] [PubMed] [Google Scholar]
- 45.Chen K., Shields M.D., Chauhan P.S., et al. Commercial ctDNA assays for minimal residual disease detection of solid tumors. Mol Diagn Ther. 2021;25:757–774. doi: 10.1007/s40291-021-00559-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Krebs M.G., Malapelle U., André F., et al. Practical considerations for the use of circulating tumor DNA in the treatment of patients with cancer: a narrative review. JAMA Oncol. 2022;8:1830–1839. doi: 10.1001/jamaoncol.2022.4457. [DOI] [PubMed] [Google Scholar]
- 47.Garcia-Murillas I., Chopra N., Comino-Méndez I., et al. Assessment of molecular relapse detection in early-stage breast cancer. JAMA Oncol. 2019;5:1473–1478. doi: 10.1001/jamaoncol.2019.1838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Chen Y.H., Hancock B.A., Solzak J.P., et al. Next-generation sequencing of circulating tumor DNA to predict recurrence in triple-negative breast cancer patients with residual disease after neoadjuvant chemotherapy. NPJ Breast Cancer. 2017;3:1–6. doi: 10.1038/s41523-017-0028-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Turner N.C., Swift C., Jenkins B., et al. c-TRAK TN investigators 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;34:200–211. doi: 10.1016/j.annonc.2022.11.005. [DOI] [PubMed] [Google Scholar]
- 50.Stover D.G., Parsons H.A., Ha G., et al. Association of cell-free DNA tumor fraction and somatic copy number alterations with survival in metastatic triple-negative breast cancer. J Clin Oncol. 2018;36:543–553. doi: 10.1200/JCO.2017.76.0033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Reichert Z.R., Morgan T.M., Li G., et al. Prognostic value of plasma circulating tumor DNA fraction across four common cancer types: a real-world outcomes study. Ann Oncol. 2023;34:111–120. doi: 10.1016/j.annonc.2022.09.163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Gao R., Davis A., McDonald T.O., et al. Punctuated copy number evolution and clonal stasis in triple-negative breast cancer. Nat Genet. 2016;48:1119–1130. doi: 10.1038/ng.3641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Venesio T., Siravegna G., Bardelli A., Sapino A. Liquid biopsies for monitoring temporal genomic heterogeneity in breast and colon cancers. Pathobiology. 2018;85:146–154. doi: 10.1159/000473882. [DOI] [PubMed] [Google Scholar]
- 54.Vidula N., Dubash T., Lawrence M.S., et al. Identification of somatically acquired BRCA1/2 mutations by cfDNA analysis in patients with metastatic breast cancer. Clin Cancer Res. 2020;26:4852–4862. doi: 10.1158/1078-0432.CCR-20-0638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Vidula N., Blouch E., Horick N.K., et al. Phase II multicenter study of talazoparib for somatic BRCA1/2 mutant metastatic breast cancer. J Clin Oncol. 2021;39 [Google Scholar]
- 56.Turner N.C., Kingston B., Kilburn L.S., et al. Circulating tumour DNA analysis to direct therapy in advanced breast cancer (plasmaMATCH): a multicentre, multicohort, phase 2a, platform trial. Lancet Oncol. 2020;21:1296–1308. doi: 10.1016/S1470-2045(20)30444-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Ring A.E., Moretti L., Afshari-Mehr A., et al. Results from plasmaMATCH trial cohort E: a phase II trial of olaparib and ceralasertib in patients with triple-negative advanced breast cancer (CRUK/15/010) J Clin Oncol. 2022;40:1024. doi: 10.1158/1078-0432.CCR-23-1696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Davis A.A., Jacob S., Gerratana L., et al. Landscape of circulating tumour DNA in metastatic breast cancer. EBioMedicine. 2020;58:102914. doi: 10.1016/j.ebiom.2020.102914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Page K., Martinson L.J., Fernandez-Garcia D., et al. Circulating tumor DNA profiling from breast cancer screening through to metastatic disease. JCO Precis Oncol. 2021;5 doi: 10.1200/PO.20.00522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Radovich M., Jiang G., Hancock B.A., et al. Association of circulating tumor DNA and circulating tumor cells after neoadjuvant chemotherapy with disease recurrence in patients with triple-negative breast cancer: preplanned secondary analysis of the BRE12-158 randomized clinical trial. JAMA Oncol. 2020;6:1410–1415. doi: 10.1001/jamaoncol.2020.2295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Davis A.A., Cristofanilli M. Minimal residual disease in patients with nonmetastatic triple-negative breast cancer: positive, negative, or a more complex issue? JAMA Oncol. 2020;6:1332–1333. doi: 10.1001/jamaoncol.2020.2285. [DOI] [PubMed] [Google Scholar]
- 62.Pascual J., Attard G., Bidard F.C., 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;33:750–768. doi: 10.1016/j.annonc.2022.05.520. [DOI] [PubMed] [Google Scholar]
- 63.Bortolini Silveira A., Bidard F.C., Tanguy M.L., et al. Multimodal liquid biopsy for early monitoring and outcome prediction of chemotherapy in metastatic breast cancer. NPJ Breast Cancer. 2021;7:115. doi: 10.1038/s41523-021-00319-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Kilgour E., Rothwell D.G., Brady G. Liquid biopsy-based biomarkers of treatment response and resistance. Cancer Cell. 2020;37:485–495. doi: 10.1016/j.ccell.2020.03.012. [DOI] [PubMed] [Google Scholar]
- 65.Chic N., Brasó-Maristany F., Prat A. Biomarkers of immunotherapy response in breast cancer beyond PD-L1. Breast Cancer Res Treat. 2022;191:39–49. doi: 10.1007/s10549-021-06421-2. [DOI] [PubMed] [Google Scholar]
- 66.Barroso-Sousa R., Jain E., Cohen O., et al. Prevalence and mutational determinants of high tumor mutation burden in breast cancer. Ann Oncol. 2020;31:387–394. doi: 10.1016/j.annonc.2019.11.010. [DOI] [PubMed] [Google Scholar]
- 67.Barroso-Sousa R., Forman J., Collier K., et al. Multidimensional molecular profiling of metastatic triple-negative breast cancer and immune checkpoint inhibitor benefit. JCO Precis Oncol. 2022;6 doi: 10.1200/PO.21.00413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.McGrail D.J., Pilié P.G., Rashid N.U., et al. High tumor mutation burden fails to predict immune checkpoint blockade response across all cancer types. Ann Oncol. 2021;32:661–672. doi: 10.1016/j.annonc.2021.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.FDA FDA approves pembrolizumab for adults and children with TMB-H solid tumors. https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-pembrolizumab-adults-and-children-tmb-h-solid-tumors Available at.
- 70.Cabel L., Proudhon C., Romano E., et al. Clinical potential of circulating tumour DNA in patients receiving anticancer immunotherapy. Nat Rev Clin Oncol. 2018;15:639–650. doi: 10.1038/s41571-018-0074-3. [DOI] [PubMed] [Google Scholar]
- 71.Zhang Q., Luo J., Wu S., et al. Prognostic and predictive impact of circulating tumor dna in patients with advanced cancers treated with immune checkpoint blockade. Cancer Discov. 2020;10:1842–1853. doi: 10.1158/2159-8290.CD-20-0047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Bratman S.V., Yang S.Y.C., Iafolla M.A.J., et al. Personalized circulating tumor DNA analysis as a predictive biomarker in solid tumor patients treated with pembrolizumab. Nat Cancer. 2020;1:873–881. doi: 10.1038/s43018-020-0096-5. [DOI] [PubMed] [Google Scholar]
- 73.Barroso-Sousa R., Li T., Reddy S., et al. Abstract GS2-10: Nimbus: a phase 2 trial of nivolumab plus ipilimumab for patients with hypermutated HER2-negative metastatic breast cancer (MBC) Cancer Res. 2022;82:GS2–GS10. [Google Scholar]
- 74.Bachelot T., Filleron T., Bieche I., et al. Durvalumab compared to maintenance chemotherapy in metastatic breast cancer: the randomized phase II SAFIR02-BREAST IMMUNO trial. Nat Med. 2021;27:250–255. doi: 10.1038/s41591-020-01189-2. [DOI] [PubMed] [Google Scholar]
- 75.Schwarzenbach H., Nishida N., Calin G.A., Pantel K. Clinical relevance of circulating cell-free microRNAs in cancer. Nat Rev Clin Oncol. 2014;11:145–156. doi: 10.1038/nrclinonc.2014.5. [DOI] [PubMed] [Google Scholar]
- 76.Cuk K., Zucknick M., Heil J., et al. Circulating microRNAs in plasma as early detection markers for breast cancer. Int J Cancer. 2013;132:1602–1612. doi: 10.1002/ijc.27799. [DOI] [PubMed] [Google Scholar]
- 77.Iorio M.V., Ferracin M., Liu C.G., et al. MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 2005;65:7065–7070. doi: 10.1158/0008-5472.CAN-05-1783. [DOI] [PubMed] [Google Scholar]
- 78.Mishra S., Srivastava A.K., Suman S. Circulating miRNAs revealed as surrogate molecular signatures for the early detection of breast cancer. Cancer Lett. 2015;369:67–75. doi: 10.1016/j.canlet.2015.07.045. [DOI] [PubMed] [Google Scholar]
- 79.Qattan A., Intabli H., Alkhayal W. Robust expression of tumor suppressor miRNA’s let-7 and miR-195 detected in plasma of Saudi female breast cancer patients. BMC Cancer. 2017;17:1–10. doi: 10.1186/s12885-017-3776-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Roth C., Rack B., Müller V., Janni W., Pantel K., Schwarzenbach H. Circulating microRNAs as blood-based markers for patients with primary and metastatic breast cancer. Breast Cancer Res. 2010;12:R90. doi: 10.1186/bcr2766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Lü L., Mao X., Shi P., et al. MicroRNAs in the prognosis of triple-negative breast cancer: a systematic review and meta-analysis. Medicine (Baltimore) 2017;96:e7085. doi: 10.1097/MD.0000000000007085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Sahlberg K.K., Bottai G., Naume B., et al. A serum MicroRNA signature predicts tumor relapse and survival in triple-negative breast cancer patients. Clin Cancer Res. 2015;21:1207–1214. doi: 10.1158/1078-0432.CCR-14-2011. [DOI] [PubMed] [Google Scholar]
- 83.Hong H.C., Chuang C.H., Huang W.C., et al. A panel of eight microRNAs is a good predictive parameter for triple-negative breast cancer relapse. Theranostics. 2020;10:8771. doi: 10.7150/thno.46142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Zhou B., Xu K., Zheng X., et al. Application of exosomes as liquid biopsy in clinical diagnosis. Signal Transduct Target Ther. 2020;5:1–14. doi: 10.1038/s41392-020-00258-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Toth B., Nieuwland R., Liebhardt S., et al. Circulating microparticles in breast cancer patients: a comparative analysis with established biomarkers. Anticancer Res. 2008;28:1107–1112. [PubMed] [Google Scholar]
- 86.Khan S., Bennit H.F., Turay D., et al. Early diagnostic value of survivin and its alternative splice variants in breast cancer. BMC Cancer. 2014;14:176. doi: 10.1186/1471-2407-14-176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Kamal NNSBNM, Shahidan W.N.S. Non-exosomal and exosomal circulatory microRNAs: which are more valid as biomarkers? Front Pharmacol. 2020;10:1500. doi: 10.3389/fphar.2019.01500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Eichelser C., Stückrath I., Müller V., et al. Increased serum levels of circulating exosomal microRNA-373 in receptor-negative breast cancer patients. Oncotarget. 2014;5:9650–9663. doi: 10.18632/oncotarget.2520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Stevic I., Müller V., Weber K., et al. Specific microRNA signatures in exosomes of triple-negative and HER2-positive breast cancer patients undergoing neoadjuvant therapy within the GeparSixto trial. BMC Med. 2018;16:1–16. doi: 10.1186/s12916-018-1163-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Wu H., Wang Q., Zhong H., et al. Differentially expressed microRNAs in exosomes of patients with breast cancer revealed by next-generation sequencing. Oncol Rep. 2020;43:240. doi: 10.3892/or.2019.7401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Rodríguez-Martínez A., de Miguel-Pérez D., Ortega F.G., et al. Exosomal miRNA profile as complementary tool in the diagnostic and prediction of treatment response in localized breast cancer under neoadjuvant chemotherapy. Breast Cancer Res. 2019;21:21. doi: 10.1186/s13058-019-1109-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Ruhen O., Mirzai B., Clark M.E., et al. Comparison of circulating tumour DNA and extracellular vesicle DNA by low-pass whole-genome sequencing reveals molecular drivers of disease in a breast cancer patient. Biomedicines. 2020;9:1–9. doi: 10.3390/biomedicines9010014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Goh C.Y., Wyse C., Ho M., et al. Exosomes in triple negative breast cancer: garbage disposals or trojan horses? Cancer Lett. 2020;473:90–97. doi: 10.1016/j.canlet.2019.12.046. [DOI] [PubMed] [Google Scholar]
- 94.Cortes J., Cescon D.W., Rugo H.S., et al. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet. 2020;396:1817–1828. doi: 10.1016/S0140-6736(20)32531-9. [DOI] [PubMed] [Google Scholar]
- 95.Schmid P., Adams S., Rugo H.S., et al. Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N Engl J Med. 2018;379:2108–2121. doi: 10.1056/NEJMoa1809615. [DOI] [PubMed] [Google Scholar]
- 96.Raimondi L., Raimondi F.M., Lazzeroni R., Di Benedetto L., Cimino G., Spinelli G.P. 23P Prediction of response to atezolizumab plus nab-paclitaxel in unresectable locally advanced triple-negative breast cancer (TNBC): The clinical usefulness of PD-L1 mRNA expression in plasma-derived exosomes. Ann Oncol. 2021;32:S10. [Google Scholar]
- 97.Li M., Li A., Zhou S., et al. Heterogeneity of PD-L1 expression in primary tumors and paired lymph node metastases of triple negative breast cancer. BMC Cancer. 2018;18:1–9. doi: 10.1186/s12885-017-3916-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Jacot W., Mazel M., Mollevi C., et al. Clinical correlations of programmed cell death ligand 1 status in liquid and standard biopsies in breast cancer. Clin Chem. 2020;66:1093–1101. doi: 10.1093/clinchem/hvaa121. [DOI] [PubMed] [Google Scholar]
- 99.Salvador-Coloma C., Santaballa A., Sanmartín E., et al. Immunosuppressive profiles in liquid biopsy at diagnosis predict response to neoadjuvant chemotherapy in triple-negative breast cancer. Eur J Cancer. 2020;139:119–134. doi: 10.1016/j.ejca.2020.08.020. [DOI] [PubMed] [Google Scholar]
- 100.Cattin S., Fellay B., Calderoni A., et al. Circulating immune cell populations related to primary breast cancer, surgical removal, and radiotherapy revealed by flow cytometry analysis. Breast Cancer Res. 2021;23:1–13. doi: 10.1186/s13058-021-01441-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Palazón-Carrión N., Jiménez-Cortegana C., Sánchez-León M.L., et al. Circulating immune biomarkers in peripheral blood correlate with clinical outcomes in advanced breast cancer. Sci Rep. 2021;11:1–12. doi: 10.1038/s41598-021-93838-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Santoni M., Romagnoli E., Saladino T., et al. Triple negative breast cancer: key role of tumor-associated macrophages in regulating the activity of anti-PD-1/PD-L1 agents. Biochim Biophys Acta Rev Cancer. 2018;1869:78–84. doi: 10.1016/j.bbcan.2017.10.007. [DOI] [PubMed] [Google Scholar]
- 103.Matsumoto H., Koo S.L., Dent R. Role of inflammatory infiltrates in triple negative breast cancer. J Clin Pathol. 2015;68:506–510. doi: 10.1136/jclinpath-2015-202944. [DOI] [PubMed] [Google Scholar]

