Breast cancer remains a leading cause of cancer-related death in women despite screening and therapeutic advances. Early detection allows for resection of local disease; however, patients can develop metastatic recurrences years after curative treatment. There is no reliable blood-based monitoring after curative therapy, and radiographic evaluation for metastatic disease is performed only in response to symptoms. Advances in circulating tumor DNA (ctDNA) assays have allowed for a potential option for blood-based monitoring. The detection of ctDNA in the absence of overt metastasis or recurrent disease indicates molecular evidence of cancer, defined as molecular residual disease (MRD). Multiple studies have shown that MRD detection is strongly associated with disease recurrence, with a lead time prior to clinical evidence of recurrence of many months. Importantly, it is still unclear whether treatment changes in response to ctDNA detection will improve outcomes. There are currently ongoing trials evaluating the efficacy of therapy escalation in the setting of MRD, and these studies are being conducted in all major breast cancer subtypes. Additional therapies under study include CDK4/6 inhibitors, PARP inhibitors, HER2-targeted therapies, and immunotherapy. This review will summarize the underlying scientific principles of various MRD assays, their known prognostic roles in early breast cancer, and the ongoing clinical trials assessing the efficacy of therapy escalation in the setting of MRD.

Over the past several decades, there have been great strides in improving outcomes in breast cancer. Screening has led to earlier diagnoses, and advances in surgery, radiation, and medical therapeutics have led to improved overall outcomes. Despite these advances, breast cancer remains the second leading cause of cancer-related death in women in the United States and a leading cause of cancer-related death in women globally (1). This discrepancy is in part due to the vast heterogeneity of disease behavior and varying risks of recurrence. After surgery, adjuvant therapies are generally applied to minimize this risk; these measures may include radiation, chemotherapy, endocrine therapy, immunotherapy, HER2-directed therapies, cyclin-dependent kinase 4/6 (CDK 4/6) inhibition, and/or PARP inhibition. An important limitation surrounding the decision to use adjuvant therapy, however, is the fact that it is based upon extrapolation of a patient's recurrence risk at a single point in time, at the time of resection. There is not a standard method to monitor for recurrence or persistence of microscopic disease after surgery and/or adjuvant therapy (2). As a result, some patients who may benefit from additional adjuvant therapy are undertreated, and similarly some patients who may not need systemic therapy are overtreated. This unmet clinical need has created a burgeoning field in solid tumor oncology: the detection and management of molecular residual disease (MRD) after curative treatment.

Adjuvant treatment decisions are based on breast cancer subtypes, along with clinical, pathologic, and/or genomic risk factors assessed at the time of diagnosis and/or surgery (3–7). Patients may receive various therapies, all of which are given based on metrics assessed at the time of surgery. Subsequent surveillance consists of annual mammograms in any remaining breast tissue and otherwise, no blood-based or radiographic studies are undertaken unless there is a clinical finding that prompts imaging (8–10). The tumor markers CA 15–3 (CA 27.29) and carcinoembryonic antigen (CEA) are not exclusively produced by cancer cells and have low sensitivity (40.3%–88.3%) and specificity (46.2%–47.3%), which are further decreased in low disease states, and their half-lives are days to weeks (11, 12).

There is an unmet need for better assessment of MRD given concern over both overtreatment and undertreatment in the adjuvant setting, and new assays allow for the sensitive detection of circulating tumor DNA (ctDNA) and strongly predict disease recurrence (13–15). At present, clinical trials are underway to determine whether intervening in the setting of detectable MRD improves the risk of disease recurrence. Ideally, patients with MRD would receive targeted care to minimize this high recurrence risk. On the other hand, many patients may receive prolonged adjuvant therapy, with the accompanying risks of side effects, despite having been cured. Similarly, MRD has not yet been used as a tool for therapy deescalation; this is a goal for future applications. As such, there is considerable interest in the detection of MRD via ctDNA and the potential interventions to help achieve cure or deep control prior to overt metastatic breast cancer (MBC); these data will be discussed in this review article.

The concept of molecular/minimal residual disease was initially established in hematologic malignancies such as leukemia, where the cancer resides in the bloodstream and bone marrow, and detection is central to diagnosis and evaluation for treatment response. After a patient receives induction chemotherapy, a bone marrow biopsy is performed to evaluate for persistent leukemia, or MRD, which would indicate a need for escalated therapy (16–18). Similarly, detectable MRD after consolidation chemotherapy would also mandate more aggressive therapy. Thus, MRD serves as a risk stratification tool in hematologic malignancies and is used to inform the decision of whether to escalate therapy. Although early technology limitations had previously not allowed for the application of this concept in solid tumors, new advances now allow us to study MRD via ctDNA in this disease type, including breast cancer.

Unlike leukemia, breast cancer does not reside in a continuous compartment like blood or bone marrow, and the area of disease is resected entirely. Blood does, however, serve as a reservoir in which evidence of persistent or recurrent breast cancer may reside. As cells die, they shed small amounts of DNA, called cell-free DNA (cfDNA). If there is tumor DNA within the cfDNA content, this is called ctDNA. The half-life of ctDNA is under 2 hours, which allows it to function as an up-to-date measure of tumor activity (19). Thus, ctDNA detection after tumor resection and/or adjuvant therapy is suggestive of incomplete cure and risk of recurrence (Fig. 1).

Figure 1.

ctDNA dynamics and breast cancer standard of care. A, A representative graph of ctDNA dynamics during primary breast cancer treatment up to the point of initial metastatic recurrence. The dotted red lines indicate molecular residual disease, where ctDNA is either not cleared after surgery, or molecular disease is detected on serial sampling during/after adjuvant therapy. B, Images representing the initial treatments for localized breast cancer followed by adjuvant therapy and surveillance breast imaging (if residual breast tissue), aligned with ctDNA dynamics figure above. (B adapted from an image created with BioRender.com.)

Figure 1.

ctDNA dynamics and breast cancer standard of care. A, A representative graph of ctDNA dynamics during primary breast cancer treatment up to the point of initial metastatic recurrence. The dotted red lines indicate molecular residual disease, where ctDNA is either not cleared after surgery, or molecular disease is detected on serial sampling during/after adjuvant therapy. B, Images representing the initial treatments for localized breast cancer followed by adjuvant therapy and surveillance breast imaging (if residual breast tissue), aligned with ctDNA dynamics figure above. (B adapted from an image created with BioRender.com.)

Close modal

Multiple studies have since shown that detection of ctDNA after surgery, either immediately or on subsequent serial sampling, is predictive of early relapse and an overall poorer prognosis; furthermore, ctDNA detection has shown a median lead time of 7.9 to 18.9 months prior to clinical recurrence of breast cancer (20–24). With the lack of routine imaging in breast cancer, the lead time over radiologic relapse has not been established in most studies. Of note, detection of higher levels of circulating tumor cells and specific disseminated tumor cells may also be classified as MRD, though these have unique limitations, including a need for more tumor content and invasive analysis, respectively. Thus, while these are additional tools for MRD evaluation, their detection is currently less clinically feasible than ctDNA at this time, and they will not be discussed in detail in this review (25, 26).

Although ctDNA as a concept has been known for many years, detection in early breast cancer has only recently been unfolding (27–30). One major issue is the overall ctDNA content is partially proportional to the tumor burden. While in MBC, ctDNA may make up almost the entire cfDNA content, levels in localized breast cancer may be substantially lower, frequently at 0.1% or lower, and the content in MRD is even smaller (13, 28). The detection of MRD would likely be clinically meaningful only if it could be identified prior to radiographic and/or clinical evidence of disease recurrence. This fact mandates exquisite sensitivity at very low allele frequencies.

Broadly speaking, there are two approaches to ctDNA evaluation in MRD: tumor-informed and tumor-agnostic detection (Fig. 2).

  • (i) Tumor-agnostic detection: in the advanced cancer setting, Foundation One CDx and Guardant 360 are both FDA-approved ctDNA assays that use targeted sequencing of known cancer-associated mutations via hybrid capture and next-generation sequencing (NGS). These assays are ideal when the ctDNA content is large enough for targeted sequencing and/or whole-exome sequencing (WES) and are most effective in metastatic disease (31). These advanced cancer genotyping assays do not have a role in the MRD setting, given the need for sufficient sensitivity and specificity for potential clinical use. More recent tumor-agnostic assays have been designed for use in the low-ctDNA MRD setting. The Guardant Reveal assay uses cancer-specific methylation signatures alongside genomic data to identify tumor-agnostic MRD in ctDNA, and in colon cancer has a reported sensitivity of 55% to 91%, where higher sensitivity is observed after serial sampling, and a specificity of 100% (32, 33). This assay assesses both genomic and epigenomic regions and is currently being studied in the Observation of ResiduAl Cancer with Liquid biopsy Evaluation (ORACLE) study (NCT05059444), which includes 1,000 patients with localized cancer after curative treatment; the study includes 11 solid tumors, including breast cancer, and is scheduled to complete in 2027 (34).

  • (ii) Tumor-informed detection: tumor-informed assays increase sensitivity in a low ctDNA context by assessing for a known set of tumor-associated alterations (31, 35). Instead of initially obtaining a comprehensive genomic snapshot, these assays prioritize a “yes/no” answer of detectability. A tumor-informed approach involves taking an excised tumor and performing ultradeep sequencing to identify alleles specific to that exact tumor and distinct from other cfDNA. Once these alleles are identified, individualized primers or probes are designed on the basis of the tumor-specific mutations and used in subsequent MRD blood testing. Sensitivity is further increased by serial sampling and by evaluating for multiple alleles (13, 14, 20). The Natera Signatera assay has received FDA breakthrough designation and uses a tumor-informed PCR-based approach. A patient's primary tumor undergoes ultradeep WES, and personalized primers are developed to target up to 16 variants. This assay can evaluate for single-nucleotide variants and short indels, and when more than 16 are identified, prioritization is based upon variant allele frequency (VAF) and context of the mutation in the setting of the cancer. To distinguish somatic clonal variants from germline mutations, the patient's white blood cells also undergo matched WES (Fig. 2C). This technique also allows for distinguishing mutations due to clonal hematopoiesis of indeterminate potential (CHIP), a key confounder in ctDNA analysis (36).

  • Coombes and colleagues used the Signatera assay to serially evaluate 49 patients with early-stage breast cancer after surgery and adjuvant therapy. Out of the 18 patients who relapsed, 16 had detectable ctDNA with a sensitivity of 55.6%, increased to 88.9% on serial testing. All presented with MRD prior to clinical progression, with a median lead time of 8.9 months (0.5–24 months). At the time of publication, there were 31 patients who had not relapsed, and none had evidence of MRD (specificity 100%; ref. 13).

  • (iii) Combined approaches: combining tumor-informed detection with tumor-agnostic deep sequencing may capitalize on the strengths of each assay. As proof of concept, Garcia-Murillas used digital PCR (dPCR) to detect early breast cancer recurrence, and subsequent plasma-based sequencing in five patients predicted the genomic makeup of the metastatic disease with better accuracy than genomic profiling of the primary tumor. Given that dPCR detects only a target allele, this was used as an initial detection tool, and once patients had detectable MRD, the plasma subsequently underwent high-depth targeted capture-based massively parallel sequencing (24).

Figure 2.

MRD assay type examples. A, A schematic of tumor-informed MRD assay development using PCR-based NGS. B, A schematic of tumor-agnostic MRD assay development using methylation patterns. Of note, assays may combine methylation patterns with genomic analysis to increase sensitivity. C, Some MRD assays perform sequencing of the buffy coat to distinguish germline variants, as well as clonal hematopoiesis of indeterminate potential (CHIP). (Adapted from an image created with BioRender.com.)

Figure 2.

MRD assay type examples. A, A schematic of tumor-informed MRD assay development using PCR-based NGS. B, A schematic of tumor-agnostic MRD assay development using methylation patterns. Of note, assays may combine methylation patterns with genomic analysis to increase sensitivity. C, Some MRD assays perform sequencing of the buffy coat to distinguish germline variants, as well as clonal hematopoiesis of indeterminate potential (CHIP). (Adapted from an image created with BioRender.com.)

Close modal

There are other technologies currently in development.

  • (i) The residual disease and recurrence (RaDaR) assay: The RaDaR assay is a tumor-informed PCR-based assay where personalized primers and matched buffy coat genotyping are used to distinguish clonal hematopoiesis of indeterminate potential (CHIP) from tumor mutations. This assay was validated in a retrospective pilot study of early-stage breast cancer, where the assay detected ctDNA as low as 0.0029%, which strongly associated with distant recurrence, with 12 of 13 distant recurrences with positive ctDNA (92%; ref. 37). In a separate retrospective study of early-stage breast cancer, MRD was detected in 17 of 17 (100%) patients that relapsed and in 0 of 5 patients that did not. These above studies have been presented as abstracts in major oncology meetings (37, 38). A large number of variants were sequenced, with a median of 41 (range 10–61) at a depth of 100,000× per locus. Interestingly, patients with brain-only metastases also had MRD detected on this assay; detection of central nervous system (CNS) disease has been a historic limitation of plasma-based assays (39, 40). VAF was very low, ranging from 0.0007% to 1.3%. Median lead time was just over 1 year, with a maximum of 2 years (38).

  • In the phase II Oxel study evaluating capecitabine, nivolumab, or the two drugs in combination as adjuvant therapy for triple negative breast cancer (TNBC), patients underwent MRD analysis via RaDaR, and 12 of 33 (36%) of patients were found to have MRD. Three patients had MRD clearance on serial testing, and one of these three recurred. The remaining 9 patients with MRD also experienced recurrence. Twenty-one patients were MRD negative at baseline, and one patient's samples turned positive after 6 weeks. Of the 12 patients who had ctDNA measured and had distant recurrence, 10 were MRD positive at baseline and an additional patient's sampling turned positive after 6 weeks. Baseline MRD was positive at a VAF range of 0.0012% to 3.6% and was associated with inferior distant disease free survival (DFS) at a median of 4.0 months versus not reached (P < 0.0001) and overall survival (OS; median not reached for either group; P = 0.0048; ref. 41).

  • (ii) Invitae Personalized Cancer Monitoring: Invitae PCM is a PCR-based tumor-informed assay that reports a flexible design, wherein between 18 and 50 variants are tracked after amplicon design. The assay has been studied in stage II–III breast cancer and shown to be associated with a median lead-time to clinical relapse of 13.7 months with detection of ctDNA in 10/13 patients that experienced relapse and no patients that had not relapsed by a median follow-up of 52.7 months (42). This assay is currently being used in the TRAK-ER trial (NCT04985266), which is evaluating the impact of fulvestrant and palbociclib in patients with ER+/HER2 breast cancer, detectable ctDNA, and no overt metastatic disease.

  • (iii) NeXT Personal: This assay combines a tumor-informed approach as well as surveillance for a panel of known clinically significant variants. This assay reports detection of stage II–IV cancer down to a limit of detection of 1 part per million, around 1×10−6 allele fraction (43). This platform uses tumor and normal whole-genome sequences (WGS), rather than WES, to design the bespoke ctDNA panels. The latter is made up of up to 1,800 somatic tumor variants in order to maximize sensitivity. In testing in healthy donor plasma, the specificity of the assay was reported to be 100% (44). A notable distinction of this assay is that the tumor-informed approach uses WGS rather than WES.

  • (iv) CAPP-Seq: Other assays under development utilize hybrid capture-based next-generation sequencing (NGS). For example, the Cancer Personalized Profiling by deep-Sequencing (CAPP-Seq) assay uses biotinylated probes against known mutations from The Cancer Genome Atlas (TCGA) and COSMIC and reports a detection limit of 0.02%. By also incorporating barcoding and error correction via polishing to account for artifacts, they report a limit of 0.001% (45). This technique has been validated in lung cancer, diffuse large B-cell lymphoma, and bladder cancer but not breast cancer at the time of this review's publication. It is commercially in use via the Avenio system (46). A similar technique using targeted error correction sequencing (TEC-Seq) was developed by Phallen and colleagues and also uses hybrid capture as the assay's central mechanism (47).

  • (v) MAESTRO: Minor allele enriched sequencing through recognition oligonucleotides (MAESTRO) utilizes massively parallel mutation enrichment and short probe panels in duplex sequencing to assess for a larger number of mutations (up to 10,000) at very low frequencies using a technique that minimizes excess sequencing of normal DNA. By using short probes in this hybridization-based capture technique, this approach reports a need for up to 100-fold fewer reads. Ostensibly, this approach would be effective in WES- or WGS-based tumor-informed MRD detection. This technique has been retrospectively assessed in samples from patients with breast cancer and shown increased MRD detection, given the feasibility of tracking a larger number of unique variants with an improved signal-to-noise ratio (48).

  • (vi) PhasED-Seq: Kurtz and colleagues describe a technique for utilizing two or more mutations that occur in cis, termed phased variants, in a technique named PhasED-Seq. The purpose of this technique is to improve sensitivity and specificity at very low tumor fractions. This assay also uses WGS rather than WES and has been studied primarily in B-cell lymphomas, where phased variants are more commonly described, but this technique has been applied to six solid tumors, including one breast cancer (31, 49). In its application in solid tumors, the assay is used in a tumor-informed approach, where tumors undergo upfront WGS from paired tumor/normal samples to first identify potentially trackable phased variants, and a bespoke panel is subsequently designed. In these six solid tumors, multiple phased variants were identified, and the background rate of noncandidate variants was found to be lower than in the case of SNVs, and 6 additional samples at very low tumor fractions were found to have MRD where prior SNV-based assays had returned negative MRD results (49).

  • (vii) Grail Galleri: Epigenetics is becoming an area of increased focus in cfDNA assays, particularly in the setting of very low ctDNA content, with interest regarding the efficacy of testing for methylation signatures in patients with little to no ctDNA. This approach is the cornerstone technology for the Grail Galleri assay, which is currently evaluating a generalized cancer screening test on the basis of cfDNA methylation patterns in patients without known cancer (NCT05155605; ref. 50).

Multiple studies have shown that MRD detection precedes clinical metastatic recurrence with the lead time increasing as the sensitivities of the assays increase (Table 1). In 2015, one of the first studies to show the clinical implications of MRD in breast cancer demonstrated that ctDNA detection after surgery predicted relapse with a median lead time of 7.9 months. WES of the primary tumor was used to design digital PCR (dPCR) probes, which bound to tumor-specific targets in plasma sampling to detect analytes at a VAF of 0.1%. On the basis of massive parallel sequencing on the primary breast tumor, at least one somatic mutation was noted in 43 of the 55 (78%) patients prospectively studied. Detectable MRD was associated with an HR of 25.1 [95% confidence interval (95% CI), 4.08–130.5; P < 0.0001]. Sensitivity was initially 50% and increased to 80% on serial testing, while specificity was around 96% (20). In a subsequent prospective, multicenter validation study in 101 women with early-stage breast cancer, ctDNA detection on serial plasma samples predicted relapse with a median lead time of 10.7 months (95% CI, 8.1−19.1 months) at a sensitivity of 75% and specificity of 92%, with the exception of brain-only metastases. Brain-only metastases were detected via ctDNA in only 1 of 6 patients (17%), suggesting that the blood–brain barrier may disrupt the otherwise continuous repository of cfDNA in nonprotected parts of the body (24). Olsson and colleagues used ctDNA-based cancer rearrangements to retrospectively show 86% of patients with eventual metastases had detectable ctDNA prior to recurrence, with an average lead time of 11 months (range, 0–37 months) and a serial sensitivity of 93% and specificity of 100% (23). Parsons and colleagues used a tumor-informed approach to serially evaluate for up to 488 mutations and found MRD detection at 1 year in stage 0–III breast cancer was strongly associated with distant recurrence (HR 20.8), with a median lead time of 18.9 months (range 3.4–39.2; ref. 14). Lipsyc-Sharf and colleagues prospectively showed that in patients with high-risk hormone receptor positive (HR+) breast cancer in the late adjuvant setting, ctDNA was detectable a median of 1 year before clinical metastasis (15).

Table 1.

Clinical studies and MRD detection lead times over breast cancer recurrence.

StudyctDNA assayCancer typeNumber patientsMedian lead timeaHR- RFSMedian follow-upSensitivitySpecificity
Garcia-Murillas et al. (2015) (20dPCR All subtypes 55 7.9 months Single timepoint: 25.1 (95% CI, 4.08–130.5) ∼2 years Post-surgery: 50% 96% 
— — — — — Serial: 12.0 (95% CI, 3.36–43.07) — Serial: 80% — 
Olsson et al. (2015) (23dPCR All subtypes 20 11 months N/A 9.2 years 93% 100% 
Garcia-Murillas et al. (2019) (24dPCR All subtypes Primary cohort: 101 Primary: 38.0 months Primary: 16.7 (95% CI, 3.5–80.5, P < .001) Primary: 35.5 months 88.4% 100% 
— — — Combined cohort: 144 Combined: 10.7 months Combined: 17.4 (95% CI, 6.3–47.8) Combined: 36.3 months — — 
Coombes et al. (2019) (13Signatera All subtypes 49 8.9 Postsurgery: 11.8 (95% CI, 4.3–32.5) ∼2 years (study midpoint) 89% 100% 
— — — — — Serial: 35.8 (95% CI, 8.0–161.3) — — — 
Parsons et al. (2020) (14Internal tumor-informed assay HR+/HER2- 35 18.9 months Postsurgery: 5.1 (95% CI, 2.0–12.7) 7.1 years Postsurgery: 23% Postsurgery: 96% 
— — — — — 1 year postsurgery: 20.8 (95% CI, 7.3–58.9) — 1-year: 19% 1-year: 100% 
Lipsyc-Sharf et al. (2022) (15RaDaR HR+/HER2- 85 12.4 months N/A 10.4 85.7% 97.4% 
StudyctDNA assayCancer typeNumber patientsMedian lead timeaHR- RFSMedian follow-upSensitivitySpecificity
Garcia-Murillas et al. (2015) (20dPCR All subtypes 55 7.9 months Single timepoint: 25.1 (95% CI, 4.08–130.5) ∼2 years Post-surgery: 50% 96% 
— — — — — Serial: 12.0 (95% CI, 3.36–43.07) — Serial: 80% — 
Olsson et al. (2015) (23dPCR All subtypes 20 11 months N/A 9.2 years 93% 100% 
Garcia-Murillas et al. (2019) (24dPCR All subtypes Primary cohort: 101 Primary: 38.0 months Primary: 16.7 (95% CI, 3.5–80.5, P < .001) Primary: 35.5 months 88.4% 100% 
— — — Combined cohort: 144 Combined: 10.7 months Combined: 17.4 (95% CI, 6.3–47.8) Combined: 36.3 months — — 
Coombes et al. (2019) (13Signatera All subtypes 49 8.9 Postsurgery: 11.8 (95% CI, 4.3–32.5) ∼2 years (study midpoint) 89% 100% 
— — — — — Serial: 35.8 (95% CI, 8.0–161.3) — — — 
Parsons et al. (2020) (14Internal tumor-informed assay HR+/HER2- 35 18.9 months Postsurgery: 5.1 (95% CI, 2.0–12.7) 7.1 years Postsurgery: 23% Postsurgery: 96% 
— — — — — 1 year postsurgery: 20.8 (95% CI, 7.3–58.9) — 1-year: 19% 1-year: 100% 
Lipsyc-Sharf et al. (2022) (15RaDaR HR+/HER2- 85 12.4 months N/A 10.4 85.7% 97.4% 

Abbreviation: RFS, recurrence free survival.

aIn cases where multiple HRs were reported (e.g., time dependent), the standard HR is listed.

Studies have also shown that residual disease after neoadjuvant therapy is associated with higher risk of recurrence. Radovich and colleagues showed that ctDNA and/or circulating tumor cell (CTC) detection after neoadjuvant chemotherapy in TNBC was associated with inferior DFS (HR, 2.67; 95% CI, 1.28–5.57; P = 0.009) and inferior OS (HR, 4.16; 95% CI, 1.66–10.42; P = 0.002; ref. 21). Riva and colleagues designed dPCR probes against TP53 mutations and found that persistent ctDNA positivity after 1 cycle of therapy was associated with worse DFS (P = 0.001) and OS (P = 0.006; ref. 51). Furthermore, in the adaptive I-SPY 2 trial, Magbanua and colleagues reported evidence that lack of ctDNA clearance was strongly associated with recurrence risk (HR, 10.4; 95% CI, 2.3–46.6; ref. 52).

Multiple studies are now underway evaluating interventions to change the above natural history. The c-TRAK-TN trial is a multi-center phase II trial evaluating the efficacy of pembrolizumab in the setting of MRD after treatment for early-stage TNBC. This study used tumor-informed sequencing and serial sampling at 3-month intervals for 12 months. Among 208 patients enrolled, 161 entered ctDNA surveillance, and 45 were entered into the pembrolizumab intervention. Of note, over half of patients with MRD in fact had metastatic disease on staging scans at the time of ctDNA detection, which highlights an important area of variation and limitation in MRD analyses; breast cancer does not undergo regular radiographic surveillance besides in residual breast tissue, so the distinction between clinical and radiographic recurrence remains to be further assessed. Among a small number of patients (n = 5) who started pembrolizumab for detectable MRD, none experienced prolonged ctDNA clearance, and 4 recurred; further study is underway (53).

The important question remains as to whether therapeutic intervention impacts the poor prognosis associated with MRD. Multiple studies in the metastatic setting have shown changes in ctDNA alleles can predict response (35, 54–60). As such, there are multiple observational and interventional clinical trials currently underway in the adjuvant treatment setting using MRD. Several major interventional trials are highlighted below based on drug class:

  • (i) CDK 4/6 inhibitors: the ongoing LEADER trial (NCT03285412) at our institution is a phase II study currently evaluating the impact of adding ribociclib to adjuvant endocrine therapy in patients with resected early-stage HR+ breast cancer who have detectable MRD (Fig. 3). The DARE study is similarly evaluating the effect of adding palbociclib to patients with MRD (NCT04567420). Trak-ER (NCT04985266) is a two-part trial where patients with high-risk clinical features and MRD are randomized to standard endocrine therapy versus palbociclib + fulvestrant.

  • (ii) PARP inhibitors: the ZEST trial (NCT04915755) is evaluating the efficacy of adjuvant niraparib in stage I–III HER2 disease; patients with HR+ breast cancer must also have a BRCA mutation, while in the TNBC arm, patients need only to be MRD+.

  • (iii) Antibody–drug conjugates: the ASPRIA study (NCT04434040) is assessing the addition of the programmed death cell ligand 1 (PD-L1) inhibitor atezolizumab in combination with the antibody–drug conjugate (ADC) sacituzumab govitecan in patients with TNBC and positive MRD.

  • (iv) Immunotherapy: the c-TRAK-TN trial (NCT03145961) is evaluating pembrolizumab given in the setting of high risk TNBC, as described above. The ASPRIA trial above is combining immunotherapy with an ADC, and the PERSEVERE trial below has multiple arms, which include the use of immunotherapy.

  • (v) Basket Trial: the PERSEVERE (NCT04849364) trial is using ctDNA to inform postneoadjuvant therapy in patients with MRD+ TNBC and pathologic residual disease. Patients may receive standard of care capecitabine or the PARP inhibitor talazoparib, the PD-L1 inhibitor atezolizumab, or the PI3K inhibitor inavolisib.

The list of these ongoing clinical trials is summarized in Table 2. The variety of trials in this setting reflects the promise of this new technology and area of needed clinical study.

Figure 3.

MRD interception clinical trial schema. A schema of clinical trial design based on MRD detection and early interception. IO, immunotherapy.

Figure 3.

MRD interception clinical trial schema. A schema of clinical trial design based on MRD detection and early interception. IO, immunotherapy.

Close modal
Table 2.

Prospective clinical trials in MRD and breast cancer.

Study namePopulationSubtypeStudy designInterventionctDNA assayNational clinical trial number
ORACLE Stage I–III All subtypes Observational N/A Guardant reveal NCT05059444 
SAFE-DE Stage I HER2+, TNBC Observational N/A Signatera NCT05058183 
HARMONY Stage II–III, postneoadjuvant HER2+ Observational N/A Todai OncoPanel NCT05433753 
LEADER T1c-T4c, any N, M0 HR+/HER2 Interventional Ribociclib added to ET Signatera NCT03285412 
TRAK-ER aHigh risk, early-stage HR+/HER2 Interventional Palbociclib added to ET Invitae PCM NCT04985266 
DARE Stage II–III HR+/HER2 Interventional Fulvestrant + palbociclib Signatera NCT04567420 
No abbreviation Stage I–III HER2+ Interventional Neratinib + T-DM1 RaDaR NCT05388149 
ZEST Stage I–III HER2-/BRCAmut or TNBC/BRCAwt Interventional Niraparib Signatera NCT04915755 
c-TRAK-TN bModerate/high risk, early stage TNBC Interventional Pembrolizumab Bio-Rad QX200 NCT03145961 
PERSEVERE Residual disease, postneoadjuvant TNBC Interventional Genomic target: Talazoparib + capecitabine; pembrolizumab + capecitabine; inavolisib + capecitabine → pembrolizumab; Talazoparib + capecitabine → pembrolizumab FoundationOneLiquid CDx NCT04849364 
— — — — No genomic target: Capecitabine + pembrolizumab or TPC — — 
ASPRIA Residual disease, postneoadjuvant TNBC Interventional Sacituzumab govitecan + atezolizumab Sysmex Inostics NCT04434040 
Study namePopulationSubtypeStudy designInterventionctDNA assayNational clinical trial number
ORACLE Stage I–III All subtypes Observational N/A Guardant reveal NCT05059444 
SAFE-DE Stage I HER2+, TNBC Observational N/A Signatera NCT05058183 
HARMONY Stage II–III, postneoadjuvant HER2+ Observational N/A Todai OncoPanel NCT05433753 
LEADER T1c-T4c, any N, M0 HR+/HER2 Interventional Ribociclib added to ET Signatera NCT03285412 
TRAK-ER aHigh risk, early-stage HR+/HER2 Interventional Palbociclib added to ET Invitae PCM NCT04985266 
DARE Stage II–III HR+/HER2 Interventional Fulvestrant + palbociclib Signatera NCT04567420 
No abbreviation Stage I–III HER2+ Interventional Neratinib + T-DM1 RaDaR NCT05388149 
ZEST Stage I–III HER2-/BRCAmut or TNBC/BRCAwt Interventional Niraparib Signatera NCT04915755 
c-TRAK-TN bModerate/high risk, early stage TNBC Interventional Pembrolizumab Bio-Rad QX200 NCT03145961 
PERSEVERE Residual disease, postneoadjuvant TNBC Interventional Genomic target: Talazoparib + capecitabine; pembrolizumab + capecitabine; inavolisib + capecitabine → pembrolizumab; Talazoparib + capecitabine → pembrolizumab FoundationOneLiquid CDx NCT04849364 
— — — — No genomic target: Capecitabine + pembrolizumab or TPC — — 
ASPRIA Residual disease, postneoadjuvant TNBC Interventional Sacituzumab govitecan + atezolizumab Sysmex Inostics NCT04434040 

Abbreviations: ET, endocrine therapy; T-DM1, trastuzumab emtansine; TPC, treatment of physician choice.

a≥4 lymph nodes; tumor size >5 cm; 1–3 axillary lymph nodes + tumor size >3 cm, histologic grade 3, Oncotype Dx ≥26, Prosigna score ≥60, EPclin risk score ≥4, or Mammaprint high risk; ≥15% residual risk of death within 10 years using NHS PREDICT, ≥1 positive lymph node after chemotherapy or tumor >3 cm after chemotherapy.

bResidual disease after neoadjuvant therapy, primary tumor size >2 cm, or primary node positivity.

Although there are advantages offered by MRD detection, there remain notable limitations providers must consider. First, most patients with breast cancer will be receiving adjuvant therapy regardless of MRD detection. Thus, the presence of MRD after surgery is likely most informative when a patient is already receiving or has received standard of care adjuvant therapy. It is important to establish via clinical trial whether modification of our adjuvant approach improves outcomes in this setting. Second, technical errors are potential pitfalls. In order to limit error and background noise, assays may undergo interventions such as barcoding, polishing, and free radical elimination (31). To evaluate for CHIP, some assays are now performing preemptive WES of white blood cells, while other assays instead rely on knowing the most common CHIP mutations and interpreting these findings with caution. MRD detection may also be less sensitive when patients are on active chemotherapy, or if recurrence is local rather than distant. Further study is also required to assess the sensitivity of plasma-based ctDNA analysis in patients with CNS-only recurrence, given anatomic limitations of the blood–brain barrier. Studies are also continuing to distinguish the lead times of clinical versus radiographic recurrence, given breast cancer does not undergo systemic radiographic surveillance after curative surgery.

While the above are technical limitations, there also remain patient-level limitations. Most importantly, there is no evidence-based therapy that improves outcomes for patients with breast cancer. These studies are currently underway (Table 2). Until an effective intervention is identified, the knowledge that a patient has MRD, a known indicator of increased risk of recurrence, risks being a source of stress without a clear therapeutic benefit. We also do not yet know when one should test for MRD, and given we know sequential testing improves sensitivity and specificity, how often (56).

Furthermore, increasingly sensitive assays may allow for future studies of therapy deescalation. Evidence suggests that patients with breast cancer do not all require the same level of intensive adjuvant therapy (61). A reliable test for molecular evidence of disease could potentially allow for serial monitoring in the setting of deescalated adjuvant therapy in otherwise low risk patient populations. In colorectal cancer, the DYNAMIC study showed ctDNA-guided chemotherapy deescalation led to patients receiving less chemotherapy with no difference in 2-year recurrence free survival (62). A key piece to underline in this trial is the fact that adjuvant chemotherapy for stage II colorectal cancer has not definitively been linked to improved OS, which is distinct from breast cancer, where adjuvant therapy in all three receptor-based subtypes has shown survival benefit (62–64). Thus, the threshold to study deescalation may be initially higher in breast cancer and mandate an exquisitely sensitive concurrent MRD assay.

There are multiple areas of potential clinical impact in the MRD space. By definition, the term MRD itself describes residual disease; thus this describes a state where there is detectable disease after definitive treatment. That being said, these assays have also shown prognostic significance in the neoadjuvant space, and there are also ongoing studies of assays similar to the tumor-agnostic MRD assays in the early detection space. Each clinical application holds potential to markedly impact patient care. In the adjuvant space, there would be great benefit in distinguishing patient groups at greatest risk of recurrence, as well as at lowest risk, to potentially further refine the adjuvant treatment landscape to be even more personalized to each patient's individual risk.

Conclusion

The evolving field of MRD is an exciting and potentially practice-changing space. Breast cancer is a unique disease in that in some cases it can lie dormant for many years and recur as metastatic disease many years after curative intervention. There remains a need for real-time monitoring for recurrence, and increasingly sensitive ctDNA assays are affording providers that opportunity. As these continue to improve, we in parallel are seeking to learn about the clinical actionability of MRD. As new information is identified through clinical trials, there is hope for a tool that allows for real-time clinical escalation and deescalation in contexts personalized to each patient.

A.J. Medford reports personal fees from Illumina, Natera, and Guardant Health outside the submitted work. L.M. Spring reports consultant/advisory board for Novartis, Puma, G1 Therapeutics, Daiichi Pharma, and AstraZeneca, and institutional research support from Phillips, Merck, Genentech, Gilead, and Eli Lilly. S.A. Hurvitz reports grants from Ambrx, Arvinas, AstraZeneca, Bayer, Cytomx, Dantari, G1 Therapeutics, Greenwich Life Sciences Inc., Eli Lilly, LOXO Oncology, Macrogenics, Radius, Sanofi, and Zymeworks outside the submitted work; and grants and other support from Daiichi-Sankyo, Genentech/Roche, Gilead, Novartis, OBI Pharma, Orinove, Orum, Pfizer, Phoenix Molecular Design, Pieris, PUMA, and SeaGen. N.C. Turner reports advisory board honoraria from AstraZeneca, Lilly, Pfizer, Roche/Genentech, Novartis, GlaxoSmithKline, Repare Therapeutics, Relay Therapeutics, Zentalis, Gilead, Inivata, Guardant, and Exact Sciences; and research funding from Astra Zeneca, Pfizer, Roche/Genentech, Merck Sharp & Dohme, Guardant Health, Invitae, Inivata, Personalis, and Natera. A. Bardia reports personal fees from Pfizer, Novartis, Genentech, Merck, Radius Health, Immunomedics/Gilead, Sanofi, Daiichi Pharma/AstraZeneca, Phillips, Eli Lilly, and Foundation Medicine during the conduct of the study; and grants from Genentech, Novartis, Pfizer, Merck, Sanofi, Radius Health, Immunomedics/Gilead, Daiichi Pharma/Astra Zeneca, and Eli Lilly outside the submitted work. No disclosures were reported by the other author.

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

1.
Siegel
RL
,
Miller
KD
,
Wagle
NS
,
Jemal
A
.
Cancer statistics, 2023
.
CA Cancer J Clin
2023
;
73
:
17
48
.
2.
Morganti
S
,
Marra
A
,
Crimini
E
,
D'Amico
P
,
Zagami
P
,
Curigliano
G
.
Refining risk stratification in HR-positive/HER2-negative early breast cancer: how to select patients for treatment escalation?
Breast Cancer Res Treat
2022
;
192
:
465
84
.
3.
Pan
H
,
Gray
R
,
Braybrooke
J
,
Davies
C
,
Taylor
C
,
McGale
P
, et al
.
20-Year risks of breast-cancer recurrence after stopping endocrine therapy at 5 years
.
N Engl J Med
2017
;
377
:
1836
46
.
4.
Kalinsky
K
,
Barlow
WE
,
Gralow
JR
,
Meric-Bernstam
F
,
Albain
KS
,
Hayes
DF
, et al
.
21-Gene assay to inform chemotherapy benefit in node-positive breast cancer
.
N Engl J Med
2021
;
385
:
2336
47
.
5.
Sparano
JA
,
Gray
RJ
,
Makower
DF
,
Pritchard
KI
,
Albain
KS
,
Hayes
DF
, et al
.
Adjuvant chemotherapy guided by a 21-gene expression assay in breast cancer
.
N Engl J Med.
2018
;
379
:
111
21
.
6.
Masuda
N
,
Lee
S-J
,
Ohtani
S
,
Im
Y-H
,
Lee
E-S
,
Yokota
I
, et al
.
Adjuvant capecitabine for breast cancer after preoperative chemotherapy
.
N Engl J Med.
2017
;
376
:
2147
59
.
7.
Von Minckwitz
G
,
Huang
C-S
,
Mano
MS
,
Loibl
S
,
Mamounas
EP
,
Untch
M
, et al
.
Trastuzumab emtansine for residual invasive HER2-positive breast cancer
.
N Engl J Med
2019
;
380
:
617
28
.
8.
Khatcheressian
JL
,
Hurley
P
,
Bantug
E
,
Esserman
LJ
,
Grunfeld
E
,
Halberg
F
, et al
.
Breast cancer follow-up and management after primary treatment: American society of clinical oncology clinical practice guideline update
.
J Clin Oncol
2013
;
31
:
961
5
.
9.
Brennan
ME
,
Houssami
N
.
Evaluation of the evidence on staging imaging for detection of asymptomatic distant metastases in newly diagnosed breast cancer
.
Breast
2012
;
21
:
112
23
.
10.
Schnipper
LE
,
Smith
TJ
,
Raghavan
D
,
Blayney
DW
,
Ganz
PA
,
Mulvey
TM
, et al
.
American society of clinical oncology identifies five key opportunities to improve care and reduce costs: the top five list for oncology
.
J Clin Oncol
2012
;
30
:
1715
24
.
11.
Harris
L
,
Fritsche
H
,
Mennel
R
,
Norton
L
,
Ravdin
P
,
Taube
S
, et al
.
American Society of clinical oncology 2007 update of recommendations for the use of tumor markers in breast cancer
.
J Clin Oncol
2007
;
25
:
5287
312
.
12.
Uygur MM, Gümüş M.
The utility of serum tumor markers CEA and CA 15-3 for breast cancer prognosis and their association with clinicopathological parameters
.
Cancer Treat Res Commun
2021
:
28
;
100402
.
13.
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
:
4255
63
.
14.
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
:
2556
64
.
15.
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
:
2408
19
.
16.
Schuurhuis
GJ
,
Heuser
M
,
Freeman
S
,
Béné
M-C
,
Buccisano
F
,
Cloos
J
, et al
.
Minimal/measurable residual disease in AML: a consensus document from the European LeukemiaNet MRD working party
.
Blood
2018
;
131
:
1275
91
.
17.
Gökbuget
N
,
Kneba
M
,
Raff
T
,
Trautmann
H
,
Bartram
C-R
,
Arnold
R
, et al
.
Adult patients with acute lymphoblastic leukemia and molecular failure display a poor prognosis and are candidates for stem cell transplantation and targeted therapies
.
Blood
2012
;
120
:
1868
76
.
18.
Brüggemann
M
,
Raff
T
,
Flohr
T
,
Gökbuget
N
,
Nakao
M
,
Droese
J
, et al
.
Clinical significance of minimal residual disease quantification in adult patients with standard-risk acute lymphoblastic leukemia
.
Blood
2006
;
107
:
1116
23
.
19.
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
:
985
90
.
20.
Garcia-Murillas
I
,
Schiavon
G
,
Weigelt
B
,
Ng
C
,
Hrebien
S
,
Cutts
RJ
, et al
.
Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer
.
Sci Transl Med
2015
;
7
:
302ra133
.
21.
Radovich
M
,
Jiang
G
,
Hancock
BA
,
Chitambar
C
,
Nanda
R
,
Falkson
C
, 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
5
.
22.
Chen
Y-H
,
Hancock
BA
,
Solzak
JP
,
Brinza
D
,
Scafe
C
,
Miller
KD
, 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
:
24
.
23.
Olsson
E
,
Winter
C
,
George
A
,
Chen
Y
,
Howlin
J
,
Tang
M-HE
, 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
:
1034
47
.
24.
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
:
1473
8
.
25.
Werner
S
,
Heidrich
I
,
Pantel
K
.
Clinical management and biology of tumor dormancy in breast cancer
.
Semin Cancer Biol
2022
;
78
:
49
62
.
26.
Ignatiadis
M
,
Reinholz
M
.
Minimal residual disease and circulating tumor cells in breast cancer
.
Breast Cancer Res
2011
;
13
:
222
.
27.
Schwarzenbach
H
,
Hoon
DSB
,
Pantel
K
.
Cell-free nucleic acids as biomarkers in cancer patients
.
Nat Rev Cancer
2011
;
11
:
426
37
.
28.
Pantel
K
,
Alix-Panabières
C
.
Liquid biopsy and minimal residual disease—latest advances and implications for cure
.
Nat Rev Clin Oncol
2019
;
16
:
409
24
.
29.
Heitzer
E
,
Haque
IS
,
Roberts
CES
,
Speicher
MR
.
Current and future perspectives of liquid biopsies in genomics-driven oncology
.
Nat Rev Genet
2019
;
20
:
71
88
.
30.
D'Amico
P
,
Corvaja
C
,
Gerratana
L
,
Reduzzi
C
,
Curigliano
G
,
Cristofanilli
M
.
The use of liquid biopsy in early breast cancer: clinical evidence and future perspectives
.
J Cancer Metastasis Treat;
2021
.
Available from
: https://jcmtjournal.com/article/view/3855.
31.
Moding
EJ
,
Nabet
BY
,
Alizadeh
AA
,
Diehn
M
.
Detecting liquid remnants of solid tumors: circulating tumor dna minimal residual disease
.
Cancer Discov
2021
;
11
:
2968
86
.
32.
Parikh
AR
,
Van Seventer
EE
,
Siravegna
G
,
Hartwig
AV
,
Jaimovich
A
,
He
Y
, et al
.
Minimal residual disease detection using a plasma-only circulating tumor DNA assay in patients with colorectal cancer
.
Clin Cancer Res
2021
;
27
:
5586
94
.
33.
Reinert
T
,
Henriksen
TV
,
Christensen
E
,
Sharma
S
,
Salari
R
,
Sethi
H
, et al
.
Analysis of plasma cell-free DNA by ultradeep sequencing in patients with stages I to III colorectal cancer
.
JAMA Oncol
2019
;
5
:
1124
31
.
34.
Janni
W
,
Huober
J
,
Braun
T
,
Müller
V
,
Fink
A
,
de Gregorio
A
, et al
.
Abstract 3403: multiomic, plasma-only circulating tumor DNA (ctDNA) assay identifies breast cancer patients with minimal residual disease (MRD) and predicts distant recurrence
.
Cancer Res
2022
;
82
:
3403
.
35.
Chan
JCH
,
Chow
JCH
,
Ho
CHM
,
Tsui
TYM
,
Cho
WC
.
Clinical application of circulating tumor DNA in breast cancer
.
J Cancer Res Clin Oncol
2021
;
147
:
1431
42
.
36.
Jaiswal
S
,
Fontanillas
P
,
Flannick
J
,
Manning
A
,
Grauman
PV
,
Mar
BG
, et al
.
Age-related clonal hematopoiesis associated with adverse outcomes
.
N Engl J Med
2014
;
371
:
2488
98
.
37.
Janni
W
,
Huober
J
,
Huesmann
S
,
Pipinikas
C
,
Braun
T
,
Müller
V
, et al
.
Abstract P2–01–07: detection of early-stage breast cancer recurrence using a personalised liquid biopsy-based sequencing approach
.
Cancer Res
2022
;
82
:
P2–01 –07–P2 –01–7
.
38.
Cutts
RJ
,
Coakley
M
,
Garcia-Murillas
I
,
Ulrich
L
,
Howarth
K
,
Emmett
W
, et al
. Abstract
536: molecular residual disease detection in early stage breast cancer with a personalized sequencing approach
.
Cancer Res
2021
;
81
:
536
.
39.
De Mattos-Arruda
L
,
Mayor
R
,
Ng
CKY
,
Weigelt
B
,
Martínez-Ricarte
F
,
Torrejon
D
, et al
.
Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma
.
Nat Commun
2015
;
6
:
8839
.
40.
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
:
224ra24
.
41.
Lynce
F
,
Mainor
C
,
Geng
X
,
Jones
G
,
Schlam
I
,
Wang
H
, et al
.
Abstract PD9–02: Peripheral immune subsets and circulating tumor DNA (ctDNA) in patients (pts) with residual triple negative breast cancer (TNBC) treated with adjuvant immunotherapy and/or chemotherapy (chemo): The OXEL study
.
Cancer Res
2022
;
82
:
PD9
02
.
42.
Garcia-Murillas
I
,
Walsh-Crestani
G
,
Phillips
E
,
Cutts
R
,
Hrebien
S
,
Dunne
K
, et al
.
Abstract P5–05–01: personalized cancer monitoring (PCM): a novel ctDNA tool to detect molecular residual disease in patients with early-stage breast cancer
.
Cancer Res
2023
;
83
:
P5-05-01
.
43.
Boyle
SM
,
Bartha
G
,
Lyle
J
,
Harris
J
,
Northcott
J
,
Norton
D
, et al
.
Abstract 5163: A high sensitivity, tumor-informed liquid biopsy platform, designed to detect minimal residual disease at part per million resolution
.
Cancer Res
2022
;
82
:
5163
.
44.
Pruess
M
,
Newburn
E
,
Boyle
SM
,
Norton
D
,
Pyke
RM
,
Navarro
F
, et al
.
P04.02 A high sensitivity, tumor-informed liquid biopsy platform, designed to detect minimum residual disease at part per million resolution
; 2022.
J Immunother Cancer
.
Available from
: https://jitc.bmj.com/content/10/Suppl_1/A21.1.
45.
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
:
548
54
.
46.
Lam
SN
,
Zhou
YC
,
Chan
YM
,
Foo
CM
,
Lee
PY
,
Mok
WY
, et al
.
Comparison of target enrichment platforms for circulating tumor DNA detection
.
Sci Rep
2020
;
10
:
4124
.
47.
Phallen
J
,
Sausen
M
,
Adleff
V
,
Leal
A
,
Hruban
C
,
White
J
, et al
.
Direct detection of early-stage cancers using circulating tumor DNA
.
Sci Transl Med
2017
;
9
:
eaan2415
.
48.
Gydush
G
,
Nguyen
E
,
Bae
JH
,
Blewett
T
,
Rhoades
J
,
Reed
SC
, et al
.
Massively parallel enrichment of low-frequency alleles enables duplex sequencing at low depth
.
Nat Biomed Eng
2022
;
6
:
257
66
.
49.
Kurtz
DM
,
Soo
J
,
Co Ting Keh
L
,
Alig
S
,
Chabon
JJ
,
Sworder
BJ
, et al
.
Enhanced detection of minimal residual disease by targeted sequencing of phased variants in circulating tumor DNA
.
Nat Biotechnol
2021
;
39
:
1537
47
.
50.
Beer
TM
,
McDonnell
CH
,
Nadauld
L
,
Liu
MC
,
Klein
EA
,
Reid
RL
, et al
.
A prespecified interim analysis of the PATHFINDER study: performance of a multicancer early detection test in support of clinical implementation
.
J Clin Oncol
39
:
15s
,
2021
(
suppl; abstr 3070
).
51.
Riva
F
,
Bidard
FC
,
Houy
A
,
Saliou
A
,
Madic
J
.
Patient-specific circulating tumor DNA detection during neoadjuvant chemotherapy in triple-negative breast cancer
.
Clin Chem
2017
;
63
:
691
9
.
52.
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
:
229
39
.
53.
Turner
N
,
Swift
C
,
Jenkins
B
,
Kilburn
L
,
Coakley
M
,
Beaney
M
, et al
.
Abstract GS3–06: primary results of the cTRAK TN trial: a clinical trial utilising ctDNA mutation tracking to detect minimal residual disease and trigger intervention in patients with moderate and high risk early stage triple negative breast cancer
.
Cancer Res
2022
;
82
:
GS3
06
.
54.
Baselga
J
,
Sellami
D
,
El-Hashimy
M
,
Dharan
B
,
Wang
A
,
Scheuer
N
, et al
.
Abstract A050: PIK3CA mutation status in tumor tissue and ctDNA as a biomarker for PFS in patients with HR+, HER2- ABC treated with buparlisib or placebo plus fulvestrant: results from the BELLE-2 and BELLE-3 randomized studies
.
Mol Cancer Ther
2018
;
17
:
A050
.
55.
André
F
,
Hurvitz
S
,
Fasolo
A
,
Tseng
L-M
,
Jerusalem
G
,
Wilks
S
, et al
.
Molecular alterations and everolimus efficacy in human epidermal growth factor receptor 2–overexpressing metastatic breast cancers: combined exploratory biomarker analysis from BOLERO-1 and BOLERO-3
.
J Clin Oncol
2016
;
34
:
2115
24
.
56.
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
:
896
.
57.
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
:
5172
7
.
58.
Bardia
A
,
Kaklamani
V
,
Wilks
S
,
Weise
A
,
Richards
D
,
Harb
W
, et al
.
Phase I study of elacestrant (RAD1901), a novel selective estrogen receptor degrader, in ER-positive, HER2-negative advanced breast cancer
.
J Clin Oncol
2021
;
39
:
1360
70
.
59.
Cescon
DW
,
Kalinsky
K
,
Parsons
HA
,
Smith
KL
,
Spears
PA
,
Thomas
A
, et al
.
Therapeutic targeting of minimal residual disease to prevent late recurrence in hormone-receptor positive breast cancer: challenges and new approaches
.
Front Oncol
2021
;
11
:
667397
.
60.
Velimirovic
M
,
Juric
D
,
Niemierko
A
,
Spring
L
,
Vidula
N
,
Wander
SA
, et al
.
Rising circulating tumor DNA as a molecular biomarker of early disease progression in metastatic breast cancer
.
JCO Precis Oncol
2020
;
1246
62
.
61.
Alvarado
M
,
Ozanne
E
,
Esserman
L
.
Overdiagnosis and overtreatment of breast cancer
.
Am Soc Clin Oncol Educ Book
2012
;
e40
5
.
62.
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
:
2261
72
.
63.
Zhu
N
,
Hu
H
,
Yuan
Y
.
Circulating tumor DNA analysis: potential to revise adjuvant therapy for stage II colorectal cancer
.
Signal Transduct Target Ther
2022
;
7
:
308
.
64.
André
T
,
Boni
C
,
Mounedji-Boudiaf
L
,
Navarro
M
,
Tabernero
J
,
Hickish
T
, et al
.
Oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment for colon cancer
.
N Engl J Med
2004
;
350
:
2343
51
.