Purpose:

We evaluated the prognostic and predictive value of circulating tumor cells (CTCs) hormone receptor–positive (HR+) metastatic breast cancer (MBC) patients randomized to letrozole alone or letrozole plus bevacizumab in the first-line setting (CALGB 40503).

Experimental Design:

Blood samples were collected at pretreatment and three additional time points during therapy. The presence of ≥5 CTCs per 7.5 mL of blood was considered CTC positive. Association of CTCs with progression-free survival (PFS) and overall survival (OS) was assessed using Cox regression models.

Results:

Of 343 patients treated, 294 had CTC data and were included in this analysis. Median follow-up was 39 months. In multivariable analysis, CTC-positive patients at baseline (31%) had significantly reduced PFS [HR, 1.49; 95% confidence interval (CI), 1.12–1.97] and OS (HR, 2.08; 95% CI, 1.49–2.93) compared with CTC negative. Failure to clear CTCs during treatment was associated with significantly increased risk of progression (HR, 2.2; 95% CI, 1.58–3.07) and death (HR, 3.4; 95% CI, 2.36–4.88). CTC-positive patients who received only letrozole had the worse PFS (HR, 2.3; 95% CI, 1.54–3.47) and OS (HR, 2.6; 95% CI, 1.59–4.40). Median PFS in CTC-positive patients was significantly longer (18.0 vs. 7.0 months) in letrozole plus bevacizumab versus letrozole arm (P = 0.0009). Restricted mean survival time analysis further revealed that addition of bevacizumab was associated with PFS benefit in both CTC-positive and CTC-negative patients, but OS benefit was only observed in CTC-positive patients.

Conclusions:

CTCs were highly prognostic for the addition of bevacizumab to first-line letrozole in patients with HR+ MBC in CALGB 40503. Further research to determine the potential predictive value of CTCs in this setting is warranted.

Translational Relevance

Prognostic and predictive biomarkers are needed for robust estimation of risk of progression and death in hormone receptor–positive (HR+) metastatic breast cancer (MBC). Blood-based biomarkers, for example, circulating tumor cells (CTC), offer a minimally invasive approach for assessing prognosis and monitoring of disease burden and therapeutic response. Our findings demonstrate that CTCs are robust prognostic markers in postmenopausal women with HR+ MBC who received letrozole (an aromatase inhibitor) with or without bevacizumab (an antibody to VEGF-A). Results of exploratory analysis suggest a potential survival benefit from adding bevacizumab to letrozole in poor prognosis patients as defined by CTC positivity at baseline. Pending validation, CTCs may serve as predictive markers of benefit from bevacizumab treatment and may aid in the selection of patients in future clinical trials that investigate the efficacy of bevacizumab in MBC.

Hormone receptor–positive (HR+) breast cancer represents approximately 70% of all breast cancers (1, 2). The standard of care for metastatic breast cancer (MBC) includes sequential endocrine therapy (ET) alone or in combination with targeted agents (1, 2). In the Cancer and Leukemia Group B (CALGB, now part of Alliance for Clinical Trials in Oncology) 40503 trial, the addition of bevacizumab (an antibody to VEGF-A) to letrozole (an aromatase inhibitor) prolonged progression-free survival (PFS) but not overall survival (OS) in postmenopausal women with HR+ MBC (3). The mechanisms involved in resistance to ET are not fully understood and remain an active area of research (4). Biomarkers that can identify patients whose tumors are more likely to respond or develop resistance to ET and ET combinations are an unmet need.

Blood-based biomarkers, for example, circulating tumor cells (CTC), offer a minimally invasive approach for assessing prognosis and monitoring of disease burden in MBC (5, 6). Increased levels of CTCs prior to treatment is highly prognostic for disease progression and death (6–8). Moreover, failure to clear CTCs early in treatment is associated with poor response to therapy (6, 9–13). In principle, CTCs can facilitate monitoring of disease status and tumor response, and thus enable the potential use of more effective therapy earlier in the disease course.

Efforts to demonstrate the clinical utility of CTCs have been actively pursued (8). For example, the STIC CTC trial recently examined the potential role of CTCs in early treatment modification in HR+ HER2-negative MBC (14). Investigators found that switching to chemotherapy in patients with high levels of CTCs prior to treatment (≥5 CTC/7.5 mL of blood) resulted in significant improvements in PFS compared with patients who received standard hormone therapy (14). Smerage and colleagues conducted a single, prospective, randomized study (SWOG0500 trial) to examine whether serial monitoring of CTCs could guide treatment decisions in MBC (15). While the study failed to demonstrate that treatment modification based on CTC response at first follow-up could improve outcomes, it did confirm previous observations showing that patients who have high CTC counts at baseline (≥5 CTC/7.5 mL of blood) had worse outcomes, regardless of treatment, compared with those with <5 CTCs (15).

In this study, we hypothesized that CTCs could serve as a prognostic and predictive marker in HR+ MBC treated with letrozole or letrozole +bevacizumab in the first-line setting. To address this hypothesis, we performed an ancillary study in the CALGB 40503 trial to evaluate whether baseline and changes in serial CTC levels were associated with PFS and OS, and whether baseline CTCs could predict benefit from the addition of bevacizumab to letrozole (16).

Patients

This is a preplanned study to examine the clinical significance of CTCs in the CALGB 40503 trial (NCT00601900; ref. 3). This trial was a multicenter randomized phase III study that compared the efficacy of letrozole alone with letrozole given in combination with bevacizumab (antibody against VEGF-A) as first-line endocrine-based therapy in postmenopausal women with HR+ advanced breast cancers. Patients who received more than one prior chemotherapy for MBC were not eligible. Prior adjuvant or neoadjuvant chemotherapy was allowed. The study design and efficacy results have been previously reported (3). Patients were enrolled between December 2008 and December 2011. The institutional review boards at the NCI and at each site approved the study. All participants provided a written informed consent that included the use of collected specimens. The study was performed in accordance with the Declaration of Helsinki.

Enumeration of CTCs

Blood was collected at 4 time points: baseline and before every third bevacizumab cycle (3-week cycles): 2 (T1), 3 (T2), and 4 (T3) or approximately 21-day intervals in the letrozole only arm. Samples were drawn into CellSave preservative tubes (Menarini Silicon Biosystems, LLC) at each participating site and shipped to the University of California San Francisco (John W. Park Laboratory) for analysis. CTC enumeration was performed by investigator (J.H. Scott) who was blinded to the clinical data.

CTCs were enumerated within 96 hours using the CellSearch system (Menarini Silicon Biosystems, LLC) following the manufacturer's instructions without modification (17). Briefly, 7.5 mL of blood was subjected to immunomagnetic enrichment to capture EPCAM-positive cells using the CellSearch Circulating Tumor Cell Kit. This was followed by immunofluorescence microscopy to enumerate CTCs, which were defined as nucleated (DAPI-positive) cells of epithelial origin (cytokeratin-positive and CD45-negative). Samples with ≥5 CTCs per 7.5 mL of blood were considered CTC positive.

Study design: clinical data and endpoints

The primary endpoint of the study was PFS, defined as the interval between study entry and first documented disease progression or death without progression. A secondary endpoint was OS, defined as time from study entry to death from any cause. Event-free patients were censored at their last clinical evaluation. Stratification factors (disease measurability and disease-free interval), age, and HER2 status were included as covariables in the multivariable models. Survival analysis was performed on follow-up data available as of July 31, 2019.

Statistical analysis

Patient and tumor characteristics were summarized according to CTC status and the proportions between groups (CTC-positive vs. CTC-negative) were compared using Pearson χ2 test.

Survival curves were estimated by the Kaplan–Meier method and compared using the log-rank test (18). Multivariable Cox regression models adjusted for known prognostic factors were used to estimate HRs and 95% confidence intervals (CI). Restricted mean survival times and differences were calculated for different time points (19). The prognostic effect of changes in CTC status between baseline and other time points was tested using a time-dependent Cox model. Data collection and statistical analyses were conducted by the Alliance Statistics and Data Center. Statistical analysis was performed using R (version 3.6.0) and SAS software (version 9.4).

Patient characteristics

Of the 391 patients randomized in the CALGB 40503 (3), 48 were excluded due to missing adverse events, treatment, or disease evaluation data (Fig. 1A). Of the remaining 343, 3 had no stratification data and 46 did not have pretreatment CTC data and were excluded from the current analysis. The baseline analysis cohort consisted of 294 patients, of whom, 154 received letrozole + bevacizumab and 140 received letrozole only. No significant differences in characteristics were observed between patients in the original study cohort and those in this study (Supplementary Table S1). The proportion of patients with impaired functioning (Eastern Cooperative Oncology Group performance status 1) was higher in the group that was excluded from this study.

Figure 1.

CTC analysis in CALGB 40503. A, CONSORT flow chart showing the number of patients included and excluded from the study. B, Study schema and sample collection. Arrows indicate time points for blood collection. C, Number of patients with CTC data and percentages of CTC-positive samples at each time point. Let, letrozole; Let + Bev, letrozole + bevacizumab.

Figure 1.

CTC analysis in CALGB 40503. A, CONSORT flow chart showing the number of patients included and excluded from the study. B, Study schema and sample collection. Arrows indicate time points for blood collection. C, Number of patients with CTC data and percentages of CTC-positive samples at each time point. Let, letrozole; Let + Bev, letrozole + bevacizumab.

Close modal

A summary of the patient and tumor characteristics of the study cohort by baseline CTC status is shown in Table 1. The median age was 58 years old; 52% received both letrozole and bevacizumab; 99% were ER positive and 19% were HER2 positive; 63% had measurable disease; and 49% received prior hormone therapy. The treatment arms were balanced within the CTC-positive and CTC-negative patient groups.

Table 1.

Patient and tumor characteristics according to circulating tumor cells (CTC) status at baseline.

CTC-negative (N = 202)CTC-positive (N = 92)Total (N = 294)χ2P
Treatment    0.58 
 Letrozole/Bevacizumab 108 (53.5%) 46 (50.0%) 154 (52.4%)  
 Letrozole 94 (46.5%) 46 (50.0%) 140 (47.6%)  
Measurable disease    0.84 
 No 75 (37.1%) 33 (35.9%) 108 (36.7%)  
 Yes 127 (62.9%) 59 (64.1%) 186 (63.3%)  
Age    0.48 
 Median 57.9 55.9 57.7  
 Range (24.7–85.3) (31.6–82.1) (24.7–85.3)  
ECOG performance status    0.96 
 Missing  
 0 131 (65.5%) 60 (65.2%) 191 (65.4%)  
 1 69 (34.5%) 32 (34.8%) 101 (34.6%)  
Disease-free interval    0.17 
 ≤ 24 months 99 (49.0%) 53 (57.6%) 152 (51.7%)  
 >24 months 103 (51.0%) 39 (42.4%) 142 (48.3%)  
Estrogen receptor    0.50 
 Missing  
 Negative 1 (0.5%) 0 (0.0%) 1 (0.3%)  
 Positive 199 (99.5%) 92 (100.0%) 291 (99.7%)  
Progesterone receptor    0.69 
 Missing  
 Negative 35 (17.5%) 20 (21.7%) 55 (18.8%)  
 Positive 163 (81.5%) 71 (77.2%) 234 (80.1%)  
 Unknown 2 (1.0%) 1 (1.1%) 3 (1.0%)  
HER2    0.69 
 Missing  
 Positive 35 (17.5%) 20 (21.7%) 55 (18.8%)  
 Negative 163 (81.5%) 71 (77.2%) 234 (80.1%)  
 Unknown 2 (1.0%) 1 (1.1%) 3 (1.0%)  
Prior chemotherapy    0.65 
 No 122 (60.4%) 53 (57.6%) 175 (59.5%)  
 Yes 80 (39.6%) 39 (42.4%) 119 (40.5%)  
Any prior endocrine therapy    0.66 
 No 102 (50.5%) 49 (53.3%) 151 (51.4%)  
 Yes 100 (49.5%) 43 (46.7%) 143 (48.6%)  
Prior aromatase inhibitor    0.60 
 No 155 (76.7%) 68 (73.9%) 223 (75.9%)  
 Yes 47 (23.3%) 24 (26.1%) 71 (24.1%)  
Prior tamoxifen    0.85 
 No 134 (66.3%) 60 (65.2%) 194 (66.0%)  
 Yes 68 (33.7%) 32 (34.8%) 100 (34.0%)  
Metastatic site    <0.01 
 Missing  
 Bone only 87 (43.5%) 56 (60.9%) 143 (49.0%)  
 Visceral only 53 (25.5%) 22 (23.9%) 75 (25.7%)  
 Bone + Visceral 60 (30.0%) 14 (15.2%) 74 (25.3%)  
No. of metastatic sites    0.39 
 Missing  
 1–2 146 (73.3%) 63 (68.5%) 209 (71.8%)  
 ≥3 53 (26.6%) 29 (31.5%) 82 (28.2%)  
CTC-negative (N = 202)CTC-positive (N = 92)Total (N = 294)χ2P
Treatment    0.58 
 Letrozole/Bevacizumab 108 (53.5%) 46 (50.0%) 154 (52.4%)  
 Letrozole 94 (46.5%) 46 (50.0%) 140 (47.6%)  
Measurable disease    0.84 
 No 75 (37.1%) 33 (35.9%) 108 (36.7%)  
 Yes 127 (62.9%) 59 (64.1%) 186 (63.3%)  
Age    0.48 
 Median 57.9 55.9 57.7  
 Range (24.7–85.3) (31.6–82.1) (24.7–85.3)  
ECOG performance status    0.96 
 Missing  
 0 131 (65.5%) 60 (65.2%) 191 (65.4%)  
 1 69 (34.5%) 32 (34.8%) 101 (34.6%)  
Disease-free interval    0.17 
 ≤ 24 months 99 (49.0%) 53 (57.6%) 152 (51.7%)  
 >24 months 103 (51.0%) 39 (42.4%) 142 (48.3%)  
Estrogen receptor    0.50 
 Missing  
 Negative 1 (0.5%) 0 (0.0%) 1 (0.3%)  
 Positive 199 (99.5%) 92 (100.0%) 291 (99.7%)  
Progesterone receptor    0.69 
 Missing  
 Negative 35 (17.5%) 20 (21.7%) 55 (18.8%)  
 Positive 163 (81.5%) 71 (77.2%) 234 (80.1%)  
 Unknown 2 (1.0%) 1 (1.1%) 3 (1.0%)  
HER2    0.69 
 Missing  
 Positive 35 (17.5%) 20 (21.7%) 55 (18.8%)  
 Negative 163 (81.5%) 71 (77.2%) 234 (80.1%)  
 Unknown 2 (1.0%) 1 (1.1%) 3 (1.0%)  
Prior chemotherapy    0.65 
 No 122 (60.4%) 53 (57.6%) 175 (59.5%)  
 Yes 80 (39.6%) 39 (42.4%) 119 (40.5%)  
Any prior endocrine therapy    0.66 
 No 102 (50.5%) 49 (53.3%) 151 (51.4%)  
 Yes 100 (49.5%) 43 (46.7%) 143 (48.6%)  
Prior aromatase inhibitor    0.60 
 No 155 (76.7%) 68 (73.9%) 223 (75.9%)  
 Yes 47 (23.3%) 24 (26.1%) 71 (24.1%)  
Prior tamoxifen    0.85 
 No 134 (66.3%) 60 (65.2%) 194 (66.0%)  
 Yes 68 (33.7%) 32 (34.8%) 100 (34.0%)  
Metastatic site    <0.01 
 Missing  
 Bone only 87 (43.5%) 56 (60.9%) 143 (49.0%)  
 Visceral only 53 (25.5%) 22 (23.9%) 75 (25.7%)  
 Bone + Visceral 60 (30.0%) 14 (15.2%) 74 (25.3%)  
No. of metastatic sites    0.39 
 Missing  
 1–2 146 (73.3%) 63 (68.5%) 209 (71.8%)  
 ≥3 53 (26.6%) 29 (31.5%) 82 (28.2%)  

CTC positivity and clinicopathologic variables

Evaluation for CTCs was performed in 7.5 mL of blood at 4 time points using CellSearch (Fig. 1B). Of the 294 patients, 92 (31.3%) were CTC positive at baseline. CTC positivity was significantly associated with bone-only metastasis (P < 0.01; Table 1). The overall CTC-positive rates decreased over time (T1: 23% of 233; T2: 20% of 172, T3: 15% of 196; Fig. 1C).

Prognostic value of CTCs

We examined the association of CTC levels at baseline and serial CTC measurements with clinical outcomes. The median follow-up time for the patients in this study was 39 months. The first three analyses below were performed irrespective of arm assignment.

CTCs at baseline

CTC-positive patients had a significantly shorter median PFS (13.6 months, 95% CI, 8.4–16.9; Fig. 2A) and OS (32.5 months, 95% CI, 26.6–36.1; Fig. 2B) compared with CTC-negative patients (PFS: 16.9 months, 95% CI, 14.1–19.3; OS: 47.5 months, 95% CI, 42.9–50.2). In a multivariable Cox regression analysis that included other prognostic variables, CTCs remained a significant negative prognostic factor for PFS (HR, 1.79; 95% CI, 1.35–2.36; Fig. 2C; Supplementary Table S2) and OS (HR, 2.72; 95% CI, 1.98–3.73; Fig. 2D; Supplementary Table S2).

Figure 2.

Prognostic impact of baseline CTCs and changes in CTC status from baseline and first follow-up. Kaplan–Meier curves for PFS (A) and OS (B) in CTC-positive and CTC-negative patients at baseline (T0). Forest plot of PFS and OS. The dashed vertical line represents a HR of 1.0 (i.e., no difference in survival between the group shown vs. its reference group). Kaplan–Meier curves for PFS (C) and OS (D) of patients according to CTC status at baseline (T0) and at 3 weeks after initiation of therapy (T1).

Figure 2.

Prognostic impact of baseline CTCs and changes in CTC status from baseline and first follow-up. Kaplan–Meier curves for PFS (A) and OS (B) in CTC-positive and CTC-negative patients at baseline (T0). Forest plot of PFS and OS. The dashed vertical line represents a HR of 1.0 (i.e., no difference in survival between the group shown vs. its reference group). Kaplan–Meier curves for PFS (C) and OS (D) of patients according to CTC status at baseline (T0) and at 3 weeks after initiation of therapy (T1).

Close modal

Changes in CTC status from baseline to T1

We assessed whether change in CTC status from baseline to the first time point (T1) during therapy was associated with patient outcome. We identified 4 groups according to serial CTC status: patients who were positive at baseline and remained (i) positive (42 of 219, 19%) or (ii) became negative (37 of 219, 17%) at the T1 measurement, and patients who were negative at baseline and (iii) became positive at T1 (11 of 219, 5%); or (iv) remained negative at T1 (129 of 219, 59%). There were significant differences in the PFS (P = 0.02; Fig. 2D) and OS among the four groups (P < 0.01; Fig. 2E). Multivariable Cox regression analysis revealed that patients who remained CTC-positive at T1 had a significant increased risk of progression (HR, 2.15; 95% CI, 1.43–3.23) and death (HR, 2.7; 95% CI, 1.66–4.38) compared with those who remained CTC negative at T1 (Supplementary Table S3). Similarly, patients who became CTC positive at T1 had a significant increased risk of death compared with those who remained CTC negative (HR, 3.2; 95% CI, 1.57–6.51).

Changes in CTC status over follow-up

We assessed whether change in CTC status throughout therapy was associated with patient outcome. In this analysis, the CTC status was treated as a time-dependent variable. At baseline, patients were classified as CTC positive or CTC negative. At each time point, patients were reassigned to CTC positive or CTC negative if their status changed. Multivariable Cox regression analysis revealed that patients with CTC-positive status at any time (baseline or at a follow-up time point) had significant increased risk of progression (HR, 2.2; 95% CI, 1.58–3.07) and death (HR, 3.4; 95% CI, 2.36–4.88) compared with patients who were CTC negative (Supplementary Table S4).

Predictive value of CTCs

CTC status at baseline by arm

Next, we stratified patients into 4 groups according to CTC status (at baseline) and treatment arm (Table 2). Patients who were CTC positive and received letrozole only had the worse PFS (adjusted likelihood ratio P < 0.01) and OS (adjusted likelihood ratio P < 0.01; Fig. 3A).

Table 2.

Survival of patients according to circulating tumor cell status and treatment arm.

EndpointCTC status at baselineTreatment armTotalNumber of eventsMedian survival in months (95% CI)Adjusted HR (95% CI)Adjusted likelihood-ratio P
PFS <0.01 
 CTC-negative Let+bev 108 70 18.4 (15.0–23.5)  
 CTC-negative Let 94 74 14.7 (11.4–18.9) 1.44 (1.02–2.02)  
 CTC-positive Let+bev 46 42 18.0 (13.6–23.7) 1.44 (0.98–2.13)  
 CTC-positive Let 46 38 7.0 (2.8–10.9) 2.31(1.54–3.47)  
OS <0.01 
 CTC-negative Let+bev 108 35 49.1 (42.4-NE)  
 CTC-negative Let 94 44 45.0 (40.1–50.1) 1.29 (0.82–2.03)  
 CTC-positive Let+bev 46 34 33.6 (26.6–40.0) 2.20 (1.37–3.55)  
 CTC-positive Let 46 28 27.1 (20.6–36.1) 2.64 (1.59–4.40)  
EndpointCTC status at baselineTreatment armTotalNumber of eventsMedian survival in months (95% CI)Adjusted HR (95% CI)Adjusted likelihood-ratio P
PFS <0.01 
 CTC-negative Let+bev 108 70 18.4 (15.0–23.5)  
 CTC-negative Let 94 74 14.7 (11.4–18.9) 1.44 (1.02–2.02)  
 CTC-positive Let+bev 46 42 18.0 (13.6–23.7) 1.44 (0.98–2.13)  
 CTC-positive Let 46 38 7.0 (2.8–10.9) 2.31(1.54–3.47)  
OS <0.01 
 CTC-negative Let+bev 108 35 49.1 (42.4-NE)  
 CTC-negative Let 94 44 45.0 (40.1–50.1) 1.29 (0.82–2.03)  
 CTC-positive Let+bev 46 34 33.6 (26.6–40.0) 2.20 (1.37–3.55)  
 CTC-positive Let 46 28 27.1 (20.6–36.1) 2.64 (1.59–4.40)  

Abbreviations: Bev, bevacizumab; CTC, circulating tumor cell; Let, letrozole.

Figure 3.

Patient survival according to CTC status at baseline and changes in CTC status from baseline and first follow-up stratified according to treatment arm. Kaplan–Meier curves for PFS (A) and OS (B) in CTC-positive and CTC-negative patients at baseline (T0) grouped according to treatment arm. Kaplan–Meier curves showing PFS (C and E) and OS (D and F) for patients stratified on the basis of CTC status at baseline (T0) and at 3 weeks after initiation of therapy (T1) randomized to treatment arms: letrozole (Let; C and D) and letrozole + bevacizumab (Let+Bev; E and F).

Figure 3.

Patient survival according to CTC status at baseline and changes in CTC status from baseline and first follow-up stratified according to treatment arm. Kaplan–Meier curves for PFS (A) and OS (B) in CTC-positive and CTC-negative patients at baseline (T0) grouped according to treatment arm. Kaplan–Meier curves showing PFS (C and E) and OS (D and F) for patients stratified on the basis of CTC status at baseline (T0) and at 3 weeks after initiation of therapy (T1) randomized to treatment arms: letrozole (Let; C and D) and letrozole + bevacizumab (Let+Bev; E and F).

Close modal

For CTC-negative patients, there was no significant difference in median PFS (letrozole: 14.7 months vs. letrozole + bevacizumab: 18.4 months, adjusted likelihood ratio P = 0.18) and OS (letrozole: 45 months vs. letrozole + bevacizumab: 49.1 months, adjusted likelihood ratio P = 0.2) between treatment arms (Fig. 3B).

For CTC-positive patients, there was no significant difference in the median OS between arms (letrozole: 27.1 months vs. letrozole + bevacizumab: 33.6 months, adjusted likelihood ratio P = 0.5). Interestingly, median PFS was significantly longer in the letrozole + bevacizumab (18.0 months) versus letrozole (7.0 months; adjusted likelihood ratio P = 0.009).

We evaluated whether CTCs at baseline were predictive of treatment efficacy. The tests for interaction between baseline CTCs (positive vs. negative) and bevacizumab (yes vs. no) were not statistically significant for PFS (P = 0.87) or OS (P = 0.99).

Changes in CTC status from baseline to T1

We assessed whether change in CTC status from baseline to the first time point (T1) during therapy was associated with patient outcome in each of the treatment arms. We observed that patients in the letrozole arm who failed to clear CTCs (CTC+CTC+) had the worse PFS (adjusted likelihood ratio P = 0.05; Fig. 3C) and OS (adjusted likelihood-ratio P < 0.01; Fig. 3D). This observation was less apparent in the letrozole + bevacizumab arm. Patients who remained CTC-negative (CTCCTC) had the most favorable PFS (adjusted likelihood-ratio P < 0.01, Fig. 3E). Patients who were CTC negative at baseline and became CTC positive at T1 (CTCCTC+) had the worse OS (adjusted likelihood ratio P < 0.01; Fig. 3F). Interestingly, OS of patients who were initially CTC positive and either became negative (CTC+CTC) or remained positive (CTC+CTC+) was better compared with CTCCTC+ group.

PFS and OS benefit

We calculated the restricted mean survival time differences at 6, 12, 18, and 24 months between patients who received letrozole + bevacizumab versus letrozole(Table 3). Results revealed significant PFS benefit with the addition of bevacizumab for both CTC-positive and CTC-negative patients. For example, at 24 months, disease progression was, on average, delayed by 5.9 months (95% CI, 2.4–9.4) and 2.5 months (95% CI, 0.1–5) in CTC-positive and CTC-negative patients, respectively.

Table 3.

Restricted mean survival time (RMST) difference between patients in different arms (Let+bevacizumabminus Let) at specified time points in CTC-positive and CTC-negative patients.

End pointFollow-up time point (months)RSMT difference: Let+bevacizumabminus letrozole(months)95% CIPRMST difference: Let+bevacizumabminus letrozole(months)95% CIP
  CTC-positive at baseline CTC-negative at baseline 
PFS 
 1.0 0.4–1.7 <0.01 0.4 0.0–0.8 0.04 
 12 3.0 1.3–4.6 <0.01 1.4 0.3–2.4 0.01 
 18 4.7 2.1–7.3 <0.01 2.1 0.3–3.8 0.02 
 24 5.9 2.4–9.4 <0.01 2.5 0.1–5.0 0.04 
OS 
 0.2 −0.2–0.5 0.32 0.0 −0.1–0.1 0.74 
 12 0.8 −0.2–1.8 0.14 −0.1 −0.5–0.3 0.58 
 18 2.1 0.3–3.8 0.02 0.1 −0.6–0.8 0.80 
 24 3.0 0.4–5.6 0.02 0.3 −0.9–1.6 0.59 
End pointFollow-up time point (months)RSMT difference: Let+bevacizumabminus letrozole(months)95% CIPRMST difference: Let+bevacizumabminus letrozole(months)95% CIP
  CTC-positive at baseline CTC-negative at baseline 
PFS 
 1.0 0.4–1.7 <0.01 0.4 0.0–0.8 0.04 
 12 3.0 1.3–4.6 <0.01 1.4 0.3–2.4 0.01 
 18 4.7 2.1–7.3 <0.01 2.1 0.3–3.8 0.02 
 24 5.9 2.4–9.4 <0.01 2.5 0.1–5.0 0.04 
OS 
 0.2 −0.2–0.5 0.32 0.0 −0.1–0.1 0.74 
 12 0.8 −0.2–1.8 0.14 −0.1 −0.5–0.3 0.58 
 18 2.1 0.3–3.8 0.02 0.1 −0.6–0.8 0.80 
 24 3.0 0.4–5.6 0.02 0.3 −0.9–1.6 0.59 

Note: CTC status was determined at baseline.

Significant differences in mean OS of 2.1 months (95% CI, 0.3–3.8) and 3.0 months (95% CI, 0.4–5.6) were observed at 18 and 24 months, respectively, in CTC-positive patients who received letrozole + bevacizumab versus those who received letrozole alone. (Table 3). No significant differences were observed in earlier time points. Among the CTC-negative patients, there was no significant difference in mean OS between arms at all time points examined.

The CALGB 40503 trial was conducted to examine the efficacy of bevacizumab in extending PFS and OS when added to first-line letrozole in HR+ MBC (3). The study was activated in 2008 soon after the FDA granted accelerated approval of bevacizumab (in combination with first-line chemotherapy) for treatment of HER2-negative MBC. This approval was revoked in 2011 because of lack of evidence in prolonged OS, and while improvement in PFS was observed, the toxicities associated with bevacizumab remained significant (20). Subsequently, CALBG 40503 reported results that were consistent with this assessment (3). In contrast, a neoadjuvant study in early-stage breast cancer (NSABP B40: chemotherapy with or without bevacizumab in treating women with stage I, stage II, or stage IIIA breast cancer that can be removed by surgery) showed improvement in OS (21). These findings suggest that there may be subgroup(s) of patients who may benefit from bevacizumab treatment, and a biomarker which can identify this subset is clearly warranted.

We performed a prospective CTC study in patients enrolled in CALGB 40503. We enumerated CTCs in serially collected blood samples and evaluated the prognostic impact of these cells. Results of our study showed that baseline levels of CTCs were highly prognostic for both PFS and OS. Furthermore, we found that changes in CTC status between baseline and other time points were prognostic, that is, failure to clear CTCs (being consistently CTC positive) or a switch from CTC negative to CTC positive were associated with poor outcomes. These findings are consistent with results from previous studies (6–13).

In CALGB 40503, serial CTC information was not used to guide therapy. A clinical study in the first-line setting for treatment of HR+ MBC did show that changing to chemotherapy in patients with persistent increase in CTCs led to improvements in patient outcomes (15). In contrast, a previous study in unselected patients with MBC (all comers) failed to demonstrate that early change of chemotherapy regimen can improve survival in patients who failed to clear CTCs during first follow-up.

We examined the predictive value of CTCs by evaluating differences in survival of patients according to CTC status (i.e., CTC-positive or -negative at baseline) and treatment received. Comparison of the median PFS and OS revealed longer PFS and OS among CTC-positive patients who received bevacizumab compared with CTC-positive patients who only received letrozole. Furthermore, our exploratory analysis revealed significantly longer mean PFS (at all time points) and OS (at 18 and 20 months) in CTC-positive patients in the letrozole + bevacizumab versus the letrozole-only arm. In contrast, there was no significant difference in mean OS in CTC-negative patients between the two arms.

A large study by Cristofanilli and colleagues showed that patients with MBC with ≥5 CTCs per 7.5 mL blood (stage IVaggressive) have significantly worse OS compared with those with <5 (stage IVindolent; ref. 8). In our study, baseline CTCs in postmenopausal women with HR-positive MBC were highly prognostic not only for OS but also PFS. Taken together, these studies confirm CTCs as a strong negative prognostic factor in MBC regardless of breast cancer subtype.

Interesting but counterintuitive observations have been made regarding the clinical impact of CTC levels during treatment with bevacizumab (22–24). For example, Bidard and colleagues found that only baseline CTCs, but not on-treatment levels, were associated with progression in patients with MBC treated with bevacizumab and chemotherapy (24). More interestingly, Gazzaniga and colleagues noted that more than half of metastatic colorectal cancer patients who progressed on bevacizumab had undetectable CTCs (23). The investigators speculated that treatment with bevacizumab facilitated epithelial-to-mesenchymal transition in CTCs, and thus became undetectable by CellSearch, an epithelial-based assay (23), and because bevacizumab affects vessel endothelium (25), impairment of CTC intravasation was proposed to explain why CTC counts considerably decreased during treatment (24) even in patients who do not respond to bevacizumab (23).

In contrast to the observations made in previous studies (22–24), our study showed significant differences in PFS and OS of patients based on baseline CTCs and at first follow-up. Interestingly, patients who were CTC positive at baseline regardless of CTC status at T1 had comparable OS with those who were consistently CTC negative especially in the first 1.5 years of follow-up (Fig. 3F). In addition, these patients (CTC+CTC+ and CTC+CTC) had significantly longer OS compared with those who were initially CTC negative and became positive (CTCCTC+). The latter group was, however, very small (n = 5). Collectively, these observations suggest that serial analysis of CTCs may help identify groups of patients who could potentially benefit most from bevacizumab treatment.

Clinical studies have also examined circulating endothelial cells (CECs) as potential biomarkers of bevacizumab efficacy. Recent report by Vasseur and colleagues showed that high levels of CECs at baseline, but not during treatment, were associated with reduced PFS in patients with HER2-negative MBC treated with chemotherapy and bevacizumab (26). Contradictory results from previous studies from the same group (24, 27) as well as others (28), particularly on the direction of prognostic significance of CECs, have highlighted the need to further examine the clinical impact of these cells.

Our findings demonstrate the potential application of CTCs for patient stratification in clinical studies that investigate benefit from bevacizumab treatment, and potentially the benefit of adding other targeted agents to endocrine therapy in the metastatic setting. Understanding CTCs and treatment interactions could eventually help guide patient randomization and risk stratification to facilitate accurate testing of efficacy of novel agents for treatment of MBC. For example, inclusion of CTCs as a stratification factor can facilitate the enrichment of patients with specific risks and could in turn better identify patients most likely to benefit or not benefit from added therapy.

Our results suggest that elevated levels of CTCs at baseline may be predictive of benefit from bevacizumab treatment. Highly vascularized tumors may have the potential to better respond to bevacizumab (29). Moreover, it is hypothesized that extensive vascularization may promote the shedding of CTCs into the blood (30). We therefore speculate that increased CTC levels in the blood due to high vascularity may explain the associated benefit of bevacizumab in CTC-positive patients.

A limitation of the study was the modest sample size particularly in the CTC-positive subset (92/294 = 31%), and thus, validation in a larger cohort is warranted.

In summary, our findings demonstrate that CTCs are robust prognostic markers in postmenopausal women with HR+ MBC treated with letrozole or letrozole + bevacizumab in the first-line setting. Our results also suggest a potential OS benefit from adding bevacizumab to letrozole in patients with poor prognosis MBC as defined by CTC positivity at baseline. If confirmed, CTCs may be useful as predictive markers for treatment benefit from bevacizumab and may aid in patient selection for future clinical trials investigating the efficacy of bevacizumab and, importantly, other targeted agents in HR+ MBC.

K.V. Ballman reports grants from NCI during the conduct of the study and personal fees from Takeda (DSMB member), Johnson and Johnson (expert witness), Janssen Oncology (expert witness), and Eli Lilly (expert witness) outside the submitted work. J.W. Park reports personal fees from Genentech (honoraria) outside the submitted work. A. Partridge reports other from Novartis (travel support x 1 2019) outside the submitted work. L. Carey reports other from Roche (research funding for clinical trials and time spent as advisor) outside the submitted work. H.S. Rugo reports grants from Pfizer (funding for sponsored studies to UCSF), Novartis (funding for sponsored studies to UCSF), Lilly (funding for sponsored studies to UCSF), Genentech/Roche (funding for sponsored studies to UCSF), Macrogenics (funding for sponsored studies to UCSF), OBI (funding for sponsored studies to UCSF), Merck (funding for sponsored studies to UCSF), Eisai (funding for sponsored studies to UCSF), Immunomedics (funding for sponsored studies to UCSF), Daiichi (funding for sponsored studies to UCSF), Seattle Genetics (funding for sponsored studies to UCSF), Odonate (funding for sponsored studies to UCSF); nonfinancial support from Daiichi (travel support for educational meeting), Mylan (travel support for educational meeting), Pfizer (travel support for educational meeting), Merck (travel support for educational meeting), AstraZeneca (travel support for educational meeting), Novartis (travel support for educational meeting), Macrogenics (travel support for educational meeting), Novartis (travel support for educational meeting), and other from Samsung (limited consulting) and Puma (limited consulting) outside the submitted work. No potential conflicts of interest were disclosed by the other authors.

The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.

M.J.M. Magbanua: Conceptualization, data curation, software, formal analysis, supervision, validation, investigation, visualization, methodology, writing-original draft, project administration, writing-review and editing. O. Savenkov: Data curation, software, formal analysis, validation, investigation, visualization, methodology, writing-original draft, writing-review and editing. E.J. Asmus: Data curation, software, formal analysis, investigation, visualization, methodology. K.V. Ballman: Formal analysis, visualization, writing-original draft, writing-review and editing. J.H. Scott: Formal analysis, investigation, methodology. J.W. Park: Conceptualization, resources, supervision, funding acquisition, writing-review and editing. M. Dickler: Conceptualization, resources, funding acquisition, investigation. A. Partridge: Resources, funding acquisition, investigation, writing-review and editing. L.A. Carey: Resources, funding acquisition, investigation, writing-review and editing. E.P. Winer: Resources, funding acquisition, investigation, writing-review and editing. H.S. Rugo: Conceptualization, resources, supervision, funding acquisition, investigation, writing-review and editing.

Research reported in this publication was supported by the NCI of the NIH under award numbers U10CA180821 and U10CA180882 (to the Alliance for Clinical Trials in Oncology), U24CA196171, UG1CA233180, UG1CA233373, U10CA180820 (ECOG-ACRIN), and UG1CA233329, U10CA180888 (SWOG). Also supported in part by funds from Genentech. The authors thank the patients for participating in this trial. We wish to thank Bryan P. Schneider, M.D. (U10CA180820) and Debasish Tripathy, M.D. (U10CA180888) for championing the study in ECOG-ACRIN and SWOG National Clinical Trials Network, respectively. We also thank the following groups who provided support to Alliance for Clinical Trials in Oncology and Alliance Foundation Trials programs as listed in https://acknowledgments.alliancefound.org.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Zelnak
AB
,
O'Regan
RM
. 
Optimizing endocrine therapy for breast cancer
.
J Natl Compr Canc Netw
2015
;
13
:
e56
64
.
2.
Rugo
HS
,
Rumble
RB
,
Macrae
E
,
Barton
DL
,
Connolly
HK
,
Dickler
MN
, et al
Endocrine therapy for hormone receptor-positive metastatic breast cancer: american society of clinical oncology guideline
.
J Clin Oncol
2016
;
34
:
3069
103
.
3.
Dickler
MN
,
Barry
WT
,
Cirrincione
CT
,
Ellis
MJ
,
Moynahan
ME
,
Innocenti
F
, et al
Phase III trial evaluating letrozole as first-line endocrine therapy with or without bevacizumab for the treatment of postmenopausal women with hormone receptor-positive advanced-stage breast cancer: CALGB 40503 (alliance)
.
J Clin Oncol
2016
;
34
:
2602
9
.
4.
Szostakowska
M
,
Trebinska-Stryjewska
A
,
Grzybowska
EA
,
Fabisiewicz
A
. 
Resistance to endocrine therapy in breast cancer: molecular mechanisms and future goals
.
Breast Cancer Res Treat
2019
;
173
:
489
97
.
5.
Lee
JS
,
Magbanua
MJ
,
Park
JW
. 
Circulating tumor cells in breast cancer: applications in personalized medicine
.
Breast Cancer Res Treat
2016
;
160
:
411
24
.
6.
Bidard
FC
,
Peeters
DJ
,
Fehm
T
,
Nole
F
,
Gisbert-Criado
R
,
Mavroudis
D
, 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
14
.
7.
Cristofanilli
M
,
Budd
GT
,
Ellis
MJ
,
Stopeck
A
,
Matera
J
,
Miller
MC
, et al
Circulating tumor cells, disease progression, and survival in metastatic breast cancer
.
N Engl J Med
2004
;
351
:
781
91
.
8.
Cristofanilli
M
,
Pierga
JY
,
Reuben
J
,
Rademaker
A
,
Davis
AA
,
Peeters
DJ
, 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
.
9.
Hartkopf
AD
,
Wagner
P
,
Wallwiener
D
,
Fehm
T
,
Rothmund
R
. 
Changing levels of circulating tumor cells in monitoring chemotherapy response in patients with metastatic breast cancer
.
Anticancer Res
2011
;
31
:
979
84
.
10.
Hayes
DF
,
Cristofanilli
M
,
Budd
GT
,
Ellis
MJ
,
Stopeck
A
,
Miller
MC
, et al
Circulating tumor cells at each follow-up time point during therapy of metastatic breast cancer patients predict progression-free and overall survival
.
Clin Cancer Res
2006
;
12
:
4218
24
.
11.
Liu
MC
,
Shields
PG
,
Warren
RD
,
Cohen
P
,
Wilkinson
M
,
Ottaviano
YL
, et al
Circulating tumor cells: a useful predictor of treatment efficacy in metastatic breast cancer
.
J Clin Oncol
2009
;
27
:
5153
9
.
12.
Wallwiener
M
,
Riethdorf
S
,
Hartkopf
AD
,
Modugno
C
,
Nees
J
,
Madhavan
D
, et al
Serial enumeration of circulating tumor cells predicts treatment response and prognosis in metastatic breast cancer: a prospective study in 393 patients
.
BMC Cancer
2014
;
14
:
512
.
13.
Magbanua
MJ
,
Carey
LA
,
DeLuca
A
,
Hwang
J
,
Scott
JH
,
Rimawi
MF
, et al
Circulating tumor cell analysis in metastatic triple-negative breast cancers
.
Clin Cancer Res
2015
;
21
:
1098
105
.
14.
Bidard
F-C
,
Jacot
W
,
Dureau
S
,
Brain
E
,
Bachelot
T
,
Bourgeois
H
, et al
Abstract GS3–07: Clinical utility of circulating tumor cell count as a tool to chose between first line hormone therapy and chemotherapy for ER+ HER2- metastatic breast cancer: results of the phase III STIC CTC trial
.
Cancer Res
2019
;
79
:
GS3–07-GS3
.
15.
Smerage
JB
,
Barlow
WE
,
Hortobagyi
GN
,
Winer
EP
,
Leyland-Jones
B
,
Srkalovic
G
, et al
Circulating tumor cells and response to chemotherapy in metastatic breast cancer: SWOG S0500
.
J Clin Oncol
2014
;
32
:
3483
89
.
16.
Ballman
KV
. 
Biomarker: predictive or prognostic?
J Clin Oncol
2015
;
33
:
3968
71
.
17.
Riethdorf
S
,
Fritsche
H
,
Muller
V
,
Rau
T
,
Schindlbeck
C
,
Rack
B
, et al
Detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer: a validation study of the cellsearch system
.
Clin Cancer Res
2007
;
13
:
920
8
.
18.
Mantel
N
. 
Evaluation of survival data and two new rank order statistics arising in its consideration
.
Cancer Chemother Rep
1966
;
50
:
163
70
.
19.
Royston
P
,
Parmar
MK
. 
The use of restricted mean survival time to estimate the treatment effect in randomized clinical trials when the proportional hazards assumption is in doubt
.
Stat Med
2011
;
30
:
2409
21
.
20.
D'Agostino
RB
 Sr
. 
Changing end points in breast-cancer drug approval–the Avastin story
.
N Engl J Med
2011
;
365
:
e2
.
21.
Bear
HD
,
Tang
G
,
Rastogi
P
,
Geyer
CE
 Jr.
,
Liu
Q
,
Robidoux
A
, et al
Neoadjuvant plus adjuvant bevacizumab in early breast cancer (NSABP B-40 [NRG Oncology]): secondary outcomes of a phase 3, randomised controlled trial
.
Lancet Oncol
2015
;
16
:
1037
48
.
22.
Tol
J
,
Koopman
M
,
Miller
MC
,
Tibbe
A
,
Cats
A
,
Creemers
GJ
, et al
Circulating tumour cells early predict progression-free and overall survival in advanced colorectal cancer patients treated with chemotherapy and targeted agents
.
Ann Oncol
2010
;
21
:
1006
12
.
23.
Gazzaniga
P
,
Raimondi
C
,
Gradilone
A
,
Di Seri
M
,
Longo
F
,
Cortesi
E
, et al
Circulating tumor cells, colon cancer and bevacizumab: the meaning of zero
.
Ann Oncol
2011
;
22
:
1929
30
.
24.
Bidard
FC
,
Mathiot
C
,
Degeorges
A
,
Etienne-Grimaldi
MC
,
Delva
R
,
Pivot
X
, et al
Clinical value of circulating endothelial cells and circulating tumor cells in metastatic breast cancer patients treated first line with bevacizumab and chemotherapy
.
Ann Oncol
2010
;
21
:
1765
71
.
25.
Ferrara
N
,
Kerbel
RS
. 
Angiogenesis as a therapeutic target
.
Nature
2005
;
438
:
967
74
.
26.
Vasseur
A
,
Cabel
L
,
Tredan
O
,
Chevrier
M
,
Dubot
C
,
Lorgis
V
, et al
Prognostic value of CEC count in HER2-negative metastatic breast cancer patients treated with bevacizumab and chemotherapy: a prospective validation study (UCBG COMET)
.
Angiogenesis
2020
;
23
:
193
202
.
27.
Pierga
JY
,
Bidard
FC
,
Autret
A
,
Petit
T
,
Andre
F
,
Dalenc
F
, et al
Circulating tumour cells and pathological complete response: independent prognostic factors in inflammatory breast cancer in a pooled analysis of two multicentre phase II trials (BEVERLY-1 and -2) of neoadjuvant chemotherapy combined with bevacizumab
.
Ann Oncol
2017
;
28
:
103
9
.
28.
Calleri
A
,
Bono
A
,
Bagnardi
V
,
Quarna
J
,
Mancuso
P
,
Rabascio
C
, et al
Predictive potential of angiogenic growth factors and circulating endothelial cells in breast cancer patients receiving metronomic chemotherapy plus bevacizumab
.
Clin Cancer Res
2009
;
15
:
7652
7
.
29.
Ferrara
N
. 
Microvascular density as a predictive biomarker for bevacizumab survival benefit in ovarian cancer: back to first principles?
J Natl Cancer Inst
2017
;
109
(11), doi: 10.1093/jnci/djx067.
30.
Kim
MY
,
Oskarsson
T
,
Acharyya
S
,
Nguyen
DX
,
Zhang
XH
,
Norton
L
, et al
Tumor self-seeding by circulating cancer cells
.
Cell
2009
;
139
:
1315
26
.

Supplementary data