Open Access

Real‑world data indicated that neoadjuvant chemotherapy alone was associated with a higher risk of tumor recurrence in high‑risk breast cancer subgroup patients

  • Authors:
    • Zhensheng Li
    • Yue Li
    • Yunjiang Liu
    • Yuguang Shang
    • Yarong Zhou
    • Xiaohui Ji
    • Huina Han
    • Kaiye Du
    • Jun Zhang
  • View Affiliations

  • Published online on: March 30, 2023     https://doi.org/10.3892/ol.2023.13779
  • Article Number: 193
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Numerous clinical trials have reported equal effects of tumor control between neoadjuvant chemotherapy (NAC) and adjuvant chemotherapy (AC) in patients with breast cancer (BC). However, this conclusion has not been verified in practice. The present retrospective study evaluated if there were different risk profiles for NAC, AC and their combinative modes on disease‑free survival (DFS) in patients with BC using real‑world data. All women with primary unilateral Stage I‑III BC and first recurrence in 2008‑2018 at The Fourth Hospital of Hebei Medical University were retrospectively identified for enrollment. The four modes of chemotherapy administered for primary BC were classified as ‘None’, ‘NAC only’, ‘NAC+AC’ and ‘AC only’. One multivariate Cox model was used to estimate the adjusted Hazard Ratio (HR) and P‑value. Covariates included age, Easter Cooperative Oncology Group grade, T stage, N stage, pathology, grade, lymphovascular invasion (LVI), BC subtype, number of chemotherapy cycles and other therapies. Amongst 637 patients, who had a mean age of 48.2 years at BC diagnosis and 50.9 years at recurrence, the median DFS by the ‘None’ (n=27), ‘NAC only’ (n=47), ‘NAC+AC’ (n=118) and ‘AC only’ (n=445) modes were 31.4, 16.6, 22.6 and 28.4 months (P<0.001), respectively. Compared with the ‘AC only’, adjusted HR (P‑value) of the ‘None’, ‘NAC only’ and ‘NAC+AC’ modes on tumor recurrence were 1.182 (0.551), 1.481 (0.037) and 1.102 (0.523), respectively. The adjusted HR of ‘NAC only’ vs. ‘AC only’ modes were 1.448 (P=0.157) for locoregional recurrence and 2.675 (P=0.003) for distant recurrence. Stratified analyses further indicated that the ‘NAC only’ mode was associated with a higher recurrence risk in T3‑4, N2‑3, LVI‑positive, or HER2‑negative subgroup patients. In conclusion, NAC alone was associated with a higher risk of tumor recurrence in high‑risk BC subgroup patients in real‑world data. Patient selection of chemotherapy mode was involved in practice but could not fully explain this finding. The ‘inadequate’ NAC was highly likely to have accounted for this observation.

Introduction

Neoadjuvant chemotherapy (NAC) is being increasingly used for operable breast cancer (BC) which is either locally advanced or has some risk factors (13). NAC benefits include increased rate of breast-conserving surgery (BCS), higher chance of tumor completion resection, and most importantly, providing valuable data about tumor response to chemotherapy (46). For example, randomized clinical trials (RCTs) have previously reported that human epidermal growth factor receptor 2 positive (HER2+) or triple negative BC (TNBC) patients had a better prognosis as a result of the changes to the chemotherapy strategy, based on the pathologic complete response (pCR) status after NAC (79). However, the routine use of NAC instead of adjunct chemotherapy (AC) in these patients with BC is still controversial (10,11). Although overall survival (OS) following NAC and AC were comparable in these trials, certain concerns about NAC have been reported. For example, certain studies have reported that NAC is associated with higher risk of locoregional recurrence (LR), frequent inconsistent correlation between pCR status and OS, and increased complexity of surgery (4,6,12,13). Some researchers have hypothesized that NAC may delay tumor surgery and make it more conservative which can lead to a higher chance of tumor recurrence in certain subgroups of patients with BC, especially those with tumors insensitive to NAC (6,14).

In China, the use of NAC in practice has been slow. Many Chinese patients with BC have low desire for BCS and are more concerned about how NAC could delay surgery and increase tumor progression (15). Furthermore, chemotherapy side effects such as fatigue, nausea, vomiting, leukopenia, emotional stress and unsatisfactory tumor reduction could discourage the patient or surgeon from continuing the NAC regime or completing the cycles as planned and then pursuing the necessary AC after the surgery done. The aforementioned issues indicate why the application of NAC in clinical practice is often different from that in trials. As a result, a retrospective study to characterize the possible different risk profiles of NAC vs. AC vs. their combinative chemotherapy modes, on disease-free survival (DFS) in patients with BC using real-world data was designed. After considering the history of NAC administration, an analysis was performed of all patients with BC at the first tumor recurrence over an 11-year time period.

Patients and methods

Patients and study design

All women with unilateral primary TNM stage I–III BC (7th edition, AJCC/UICC) and first tumor recurrence in 2008–2018 at The Fourth Hospital of Hebei Medical University were enrolled in the present study (16). Patient inclusion criteria included: i) Age at diagnosis ≥18 years and diagnosed in 1997 or later; and ii) BC diagnosed pathologically as LR or presented clinically distant metastasis by computed tomography, magnetic resonance, bone scintigraphy or positron emission tomography imaging. In the present study, LR was defined as tumor recurrence at contralateral or ipsilateral breast, chest wall, or regional lymph node (LN) at axillary, supraclavicular, infraclavicular and internal mammary regions. Exclusion criteria included: i) Inflammatory BC; ii) contralateral BC diagnosed within 6 months following the primary BC diagnosis; iii) any recurrence diagnosed before the completion of chemotherapy or RT; and iv) any recurrence potentially from other cancers.

In the analysis, the primary BC diagnosis date was defined as the baseline. The earliest date of all tumor recurrences diagnosed in regular medical check-ups was regarded as the endpoint date. DFS in days was calculated as the number of days between the endpoint date and the baseline plus 1. Three time-to-event outcomes of; any tumor recurrence, LR and organ metastasis were analyzed.

The present study was approved by the Research Ethics Committee of the Fourth Hospital of Hebei Medical University (approval no. 2017-288). All participants provided written informed consent. All sensitive health information of participants was excluded from the study dataset.

Chemotherapy mode and regimen

While the National Comprehensive Cancer Network (NCCN) guidelines for BC treatments were followed, the actual timing and number of cycles of chemotherapy were at the discretion of the individual physician. The most common regimens of chemotherapy mainly consisted of the dose-dense doxorubicin and cyclophosphamide followed by paclitaxel; docetaxel and cyclophosphamide; cyclophosphamide, methotrexate and fluorouracil epirubicin and cyclophosphamide; or docetaxel, doxorubicin and cyclophosphamide. Trastuzumab was the only HER2 targeted therapy drug available to be administered. Due to the high out-of-pocket cost, trastuzumab was administered only in a small portion of HER2+ patients. Based on the timing of the chemotherapy cycles, patients were assigned to one of four modes, as follows: ‘None’, ‘NAC only’, ‘NAC+AC’ or ‘AC only’.

Statistical methods

Continuous and categorical variables were analyzed using descriptive statistics. Analysis of variance (ANOVA), χ2 or Fisher's exact tests, were used to perform statistical comparisons as appropriate. The Kaplan-Meier curve and log-rank test were used to analyze DFS rates. The Cox survival model was used to estimate the hazard ratio (HR), 95% confidence interval (CI) and P-value. Two-sided P<0.05 was considered to indicate a statistically significant difference. All statistical analyses were performed using SAS 9.4 for Windows (SAS Institute, Inc.).

Results

Characteristics at baseline

Table I presented the 637 patients who were enrolled after reviewing all hospital admissions (n=16,891) of patients with BC. Among the enrolled patients, 25.9% (n=165) received NAC. Many variables at baseline were significantly associated with the choice of chemotherapy modes. Because the number of chemotherapy cycles was not normally distributed, its three-level category was created for analysis. While the proportion of ‘1–4 cycles’ in ‘AC only’ patients was 23.1% (103/445), it was markedly lower, 7.9% (13/165) in combined NAC patients. Furthermore, the proportion of ‘1–4 cycles’ was significantly higher in ‘NAC only’ patients than in ‘NAC+AC’ patients [14.9% (7/47) vs. 5.1% (6/118), P=0.035]. The aforementioned results indicated that NAC patients had either less cycles of NAC alone or, more likely, did not attend for AC after surgery.

Table I.

Baseline characteristics of patients.

Table I.

Baseline characteristics of patients.

Chemotherapy mode

CharacteristicAllNoneNAC onlyNAC+ACAC only P-valuea
Patients (n, %)637 (100)27 (4.2)47 (7.4)118 (18.5)445 (69.9)
Mean age ± SD, years48.2±10.454.8±13.848.5±10.248.8±10.147.6±10.10.005
Diagnosis yearb, n (%) <0.001
  1997-2010251 (39.4)10 (4.0)5 (2.0)41 (16.3)195 (77.7)
  2011-2016386 (60.6)17 (4.4)42 (10.9)77 (19.9)250 (64.8)
ECOG grade, n (%) 0.040
  0617 (96.5)25 (4.0)46 (7.5)117 (19.0)429 (69.5)
  1-220 (3.5)2 (10.0)1 (5.0)1 (5.0)16 (80.0)
Tumor laterality, n (%) 0.230
  Left351 (55.1)14 (4.0)24 (6.8)75 (21.4)238 (67.8)
  Right286 (44.9)13 (4.5)23 (8.0)43 (15)207 (72.4)
Tumor surgery, n (%) 0.053
  Lumpectomy22 (3.5)3 (13.6)0 (0.0)2 (9.1)17 (77.3)
  Mastectomy615 (96.5)24 (3.9)47 (7.6)116 (18.9)428 (69.6)
LN procedure, n (%)
  ALND579 (90.9)16 (2.8)47 (8.1)113 (19.5)403 (69.6)<0.001
  SLND22 (3.5)0 (0.0)3 (13.6)14 (63.6)5 (22.7)<0.001
  IMLND17 (2.7)0 (0.0)4 (23.5)5 (29.4)8 (47.1)0.134
T stage, n (%) <0.001
  T1141 (22.1)6 (4.3)7 (5.0)10 (7.1)118 (83.7)
  T2338 (53.1)16 (4.7)10 (3.0)51 (15.1)261 (77.2)
  T3-4114 (17.9)0 (0.0)30 (26.3)55 (48.2)29 (25.4)
  Unknown44 (6.9)5 (11.4)0 (0.0)2 (4.5)37 (84.1)
N stage, n (%) <0.001
  N0236 (37)16 (6.8)3 (1.3)14 (5.9)203 (86.0)
  N1167 (26.2)6 (3.6)9 (5.4)26 (15.6)126 (75.4)
  N274 (11.6)1 (1.4)8 (10.8)14 (18.9)51 (68.9)
  N3141 (22.1)1 (0.7)27 (19.1)63 (44.7)50 (35.5)
  Unknown19 (3.0)3 (15.8)0 (0.0)1 (5.3)15 (78.9)
Tumor pathology, n (%) 0.345
  IDC529 (83.0)21 (4.0)42 (7.9)102 (19.3)364 (68.8)
  Other108 (17.0)6 (5.6)5 (4.6)16 (14.8)81 (75.0)
Tumor grade, n (%) 0.070
  I8 (1.3)0 (0.0)0 (0.0)1 (12.5)7 (87.5)
  II393 (61.7)17 (4.3)34 (8.7)86 (21.9)256 (65.1)
  III140 (22.0)4 (2.9)8 (5.7)15 (10.7)113 (80.7)
  Not reported96 (15.1)6 (6.3)5 (5.2)16 (16.7)69 (71.9)
LVI, n (%) <0.001
  Positive152 (23.9)3 (2.0)5 (3.3)43 (28.3)101 (66.4)
  Negative164 (25.7)4 (2.4)8 (4.9)21 (12.8)131 (79.9)
  Not reported321 (50.4)20 (6.2)34 (10.6)54 (16.8)213 (66.4)
Subtype, n (%) 0.036
  Luminal A258 (40.5)11 (4.3)14 (5.4)43 (16.7)190 (73.6)
  Luminal B132 (20.7)6 (4.5)16 (12.1)21 (15.9)89 (67.4)
  HER2-enriched97 (15.2)2 (2.1)9 (9.3)21 (21.6)65 (67.0)
  TNBC86 (13.5)2 (2.3)8 (9.3)22 (25.6)54 (62.8)
  Unclassified64 (10.0)6 (9.4)0 (0.0)11 (17.2)47 (73.4)
Chemotherapy, cycles, n (%) <0.001
  027 (4.2)27 (100)0 (0.0)0 (0.0)0 (0.0)
  1-4116 (18.2)0 (0.0)7 (6.0)6 (5.2)103 (88.8)
  ≥5494 (77.6)0 (0.0)40 (8.1)112 (22.7)342 (69.2)
Systemic therapy, n (%)
  ET232 (36.4)7 (3.0)12 (5.2)33 (14.2)180 (77.6)0.015
  RT264 (41.4)1 (0.4)25 (9.5)75 (28.4)163 (61.7)<0.001
  Trastuzumab12 (1.9)0 (0.0)0 (0.0)5 (41.7)7 (58.3)0.158

a Calculated using ANOVA, χ2 or Fisher's Exact test, on non-missing values, as appropriate.

b Use of Trastuzumab was started in 2011.NAC, neoadjuvant chemotherapy; AC, adjuvant chemotherapy; ECOG, Easter Cooperative Oncology Group; LN, lymph node; ALND, axillary lymph node dissection; SLND, supraclavicular lymph node dissection; IMLND, internal mammary lymph node dissection; IDC, invasive ductal carcinoma; LVI, lymphovascular invasion; HER2, human epidermal growth factor receptor 2; TNBC, triple negative breast cancer; ET, endocrine therapy; RT, radiotherapy; T, tumor; N, node.

Characteristics of tumor recurrence

The overall median DFS was 25.9 months (Table II). The DFS rates were significantly different among chemotherapy mode (P<0.001, Fig. 1A) and cycle (P=0.002, Fig. 1B) subgroups. Patients who had ‘NAC only’ or had ≥5 chemotherapy cycles had the lowest DFS. Given the selection of chemotherapy mode varied by factors such as tumor size, LN status and BCS pursuit, as considered by the oncologist, multivariate analysis was performed to verify the independence of these observed links.

Table II.

Characteristics of tumor recurrence (n=637).

Table II.

Characteristics of tumor recurrence (n=637).

Chemotherapy mode

CharacteristicAlNoneNAC onlyNAC+ACAC only P-valuea
Mean age ± SD, years50.9±10.657.9±13.250.1±10.151.0±10.150.5±10.40.001
Median DFS, months (95%CI)25.9 (24.1-27.8)31.4 (13.3-39.4)16.6 (13.2-19.3)22.6 (19.7-24.7)28.4 (26.1-31.2)<.001
Recurrence site, n (%)
  Ipsilateral region321 (50.4)16 (5.0)23 (7.2)51 (15.9)231 (72.0)0.293
  Contralateral region123 (19.3)4 (3.3)18 (14.6)26 (21.1)75 (61.0)0.004
  Any organ453 (71.1)20 (4.4)34 (7.5)82 (18.1)317 (70.0)0.959
Recurrence organ, n (%)
  Bone254 (39.9)11 (4.3)19 (7.5)37 (14.6)187 (73.6)0.218
  Liver156 (24.5)3 (1.9)12 (7.7)33 (21.2)108 (69.2)0.331
  Lung154 (24.2)9 (5.8)10 (6.5)30 (19.5)105 (68.2)0.652
  Brain35 (5.5)1 (2.9)3 (8.6)9 (25.7)22 (62.9)0.675
  Other107 (16.8)5 (4.7)5 (4.7)25 (23.4)72 (67.3)0.379
Recurrence type, n (%)
  Region only137 (21.5)6 (4.4)4 (2.9)23 (16.8)104 (75.9)0.074
  Organ only315 (49.5)10 (3.2)24 (7.6)67 (21.3)214 (67.9)
  Region and organ185 (29.0)11 (5.9)19 (10.3)28 (15.1)127 (68.6)

a P-value from ANOVA, χ2 test or log-rank test on non-missing values, as appropriate. NAC, neoadjuvant chemotherapy; AC, adjuvant chemotherapy; DFS, disease-free survival; CI, confidence interval.

Cox survival models

Table III shows the analysis results. The covariates were determined based on the model building process and literature review. Compared with the ‘AC only’ patients, the ‘NAC only’ patients were significantly associated with higher risks of any recurrence (HR 1.452, P=0.037) and organ metastasis (HR 2.675, P=0.003) after the adjustment of covariates. No significant difference in the risk of any recurrence, LR and organ metastasis were demonstrated among the ‘NAC+AC’ patients and the ‘AC only’ patients (all HR 0.945-1.141, P=0.523-0.765). No difference in the risks linked to the ‘1–4 cycles’ (vs. ‘≥5 cycles’) of events was demonstrated by multivariate analyses for any recurrence (HR 0.811, P=0.062), LR (HR 0.824, P=0.207), and organ metastasis (HR 0.817, P=0.439) (data not shown).

Table III.

Cox proportional hazard regression analysis of recurrence type.

Table III.

Cox proportional hazard regression analysis of recurrence type.

A, Any recurrence

Univariate Multivariatea
ChemotherapyHR95% CIP-valueHR95% CIP-value
None0.835(0.56-1.245)0.3761.182(0.682-2.047)0.551
NAC only2.378(1.752-3.228)<0.0011.452(1.022-2.062)0.037
NAC + AC1.388(1.131-1.703)0.0021.085(0.845-1.394)0.523
AC only1.000 ref.1.000 ref.

B, regional recurrence

Univariate Multivariatea


ChemotherapyHR95% CIP-valueHR95% CIP-value

None0.915(0.539-1.553)0.7421.060(0.507-2.216)0.878
NAC only2.278(1.474-3.522)<0.0011.448(0.867-2.419)0.157
NAC + AC1.161(0.855-1.575)0.3390.945(0.652-1.369)0.765
AC only1.000 ref.1.000 ref.

C, Organ metastasis

Univariate Multivariatea


ChemotherapyHR95% CIP-valueHR95% CIP-value

None0.679(0.244-1.893)0.4601.090(0.273-4.345)0.903
NAC only5.960(3.510-10.122)<0.0012.675(1.392-5.139)0.003
NAC + AC1.880(1.197-2.951)0.0061.141(0.656-1.983)0.641
AC only1.000 ref.1.000 ref.

a The final covariates included diagnosis age at diagnosis of primary BC, ECOG, T stage, N stage, tumor pathology, tumor grade, LVI, subtype, ET, RT, Trastuzumab, if calendar year of primary BC diagnosis was after 2010, number group of chemotherapy cycles administered. HR hazard ratio, CI confidence interval, NAC neoadjuvant chemotherapy, AC adjuvant chemotherapy, ref. reference, BC breast cancer, ET endocrine therapy, RT radiotherapy; ECOG, Easter Cooperative Oncology Group; LVI, lymphovascular invasion; ET, endocrine therapy; RT, radiotherapy; T, tumor; N, node.

Stratified Cox analysis

Fig. 2 demonstrated that the significantly higher risk of any recurrence linked to the ‘NAC only’ mode compared with the ‘AC only’ mode was particularly presented in certain subgroups of patients. Specifically, it was present in patients with BC characterized by diagnosis and treatment in earlier years of the study period (HR 3.638, P=0.015), stage T3–4 (HR 3.441, P<0.001), stage N2–3 (HR 1.741, P=0.022), lymphovascular invasion positive (LVI+) (HR 5.155, P=0.005) or HER2-negative (HR 1.697, P=0.039). There were similar results from the stratified Cox analysis of organ metastasis (data not shown).

Discussion

For numerous reasons, such as different populations and interventions, real-world data could provide risk profiles of variables on tumor recurrence different to those generated in RCTs. Through adjusted and stratified analyses, this retrospective study demonstrated that, compared with the most commonly used ‘AC only’ mode, the ‘NAC only’ mode was associated with a significantly higher risk of tumor recurrence, especially at distant organ. These links were more specifically presented in high-risk BC subgroup patients characterized by stage T3-4, stage N2-3, LVI+ or HER2-negative. It could be hypothesized that the ‘inadequate’ total cycles of chemotherapy in BC patients who had NAC could have played a role. Another factor could possibly be the circumstances-these ‘NAC+AC’ patients would become ‘NAC only’ patients if they abandoned the planned AC. Given the potential clinical value of chemotherapy modes, more studies on the real-world data are needed to confirm and explain this finding.

Many factors can influence the decision-making for initiation, elongation, and termination of NAC in BC patients in clinical practice. Under the NCCN guidelines, NAC is usually recommended to patients with operable BC with larger tumors, more metastatic LN, HER2+, TNBC or stronger desire for BCS (17). For most early TNBC patients, NAC is currently used as the standard treatment while AC is only used for some patients opting for upfront surgery (18,19). The present study focused on patients with BC with first tumor recurrence in the past. It was noted that, different from the current guideline, the AC mode was demonstrated to be administered markedly more often than the NAC mode 62.8% (54/86) compared with 9.3% (8/86) in TNBC patients (Table I, Fig. 2). Furthermore, a previous meta-analysis (9 studies, 2,109 patients) reported that the inclusion of platinum in NAC regimens led to a significantly higher rate of pCR (51.2% vs. 37.0%; OR 1.96, 95%CI 1.46-2.62) in TNBC patients, but no significant improvement in OS (HR 0.86, 95%CI 0.46-1.63) (20). Many factors are involved the decision-making of NAC against AC. Based on the results of the present study, the tumor recurrence risk profiles of chemotherapy modes should consider factors such as patient age, performance status, tumor feature, tumor response, adverse effects of prior chemotherapy and others. In the present study, data for pCR and adverse effects of chemotherapy were lacking for additional analysis. However, the consistent results of multivariate analyses indicated that the covariates assessed could not fully account for the study's results.

NAC and AC may have different drug mechanisms for reducing risk of tumor recurrence in BC patients (6). While surgery can cure the primary tumor, systemic treatments of NAC or AC are reported to be required to eliminate the occult tumor cells (6,21). Previous studies demonstrated that the effectiveness of AC varies with BC subtypes, metastatic LN status, dosage and number of cycles of chemotherapy regimens (2225). For example, only patients with estrogen receptor-positive (ER+) and LN-negative BC with high 21-gene recurrence scores have been reported to benefit from adding chemotherapy to ET based on prognosis (2628). In patients with ER-negative (ER-) or HER2+ BC, only having AC over a certain dosage of its regimens and number of cycles has been reported to lead to improved DFS and OS (23,29). The present study was not able to exclude the possibility that NAC and AC may have different mechanisms and effectiveness in reducing the tumor recurrence risk in certain BC subgroups.

Numerous recent studies have reported that the pCR status after a certain number of cycles of NAC could provide valuable guidance to the application of AC (9,30). For example, the KATHERINE trial included 1,486 patients with early HER2+ BC who had non-pCR after NAC and were randomized into the trastuzumab armor T-DM1 arm in AC (9). The trial reported that there was an 11.3% absolute gain in 3-year DFS (HR 0.50; 95%CI 0.39-0.64) in the T-DM1 arm, compared to the trastuzumab arm (9). The CREATE-X trial included 910 patients with early HER2-negative BC who had non-pCR after NAC reported that adding capecitabine for 6–8 cycles significantly improved the 5-year DFS (HR 0.70, 95%CI 0.53-0.92) and 5-year OS (HR 0.59, 95%CI 0.39-0.90) (7). This benefit was larger in the TNBC subgroup patients (n=286) with 5-year DFS (HR 0.58, 95% CI 0.39-0.87) and 5-year OS (HR 0.52, 95% CI 0.30-0.90) (7). Given these results, the adapted strategy of modifying AC based on the pCR status after NAC has become more widely used in recent years (18). In the present study, trastuzumab, being the only anti-HER2 target agent used, was administered to only a few HER2+ patients. As stated previously, how the AC was discontinued or changed after NAC and surgery could not be summarized for any possible analysis in the study. The ‘NAC only’ patients had significantly less cycles of chemotherapy compared with the ‘NAC+AC’ patients. Since the chemotherapy cycle number was not normally distributed, the three levels were used one covariate in subsequent analysis.

There are a limited number of published studies that explore the relationship of NAC and AC with DFS and OS (6,13,14,31). In 2008, one combined analysis of two RCTs [National Surgical Adjuvant Breast and Bowel Project (NSABP) B-18, B-27] reported that the NAC and AC approaches did not have different tumor recurrence rates and OS in BC patients (13). However, a 2005 meta-analysis of 9 RCTs (n=3,946) reported that compared with the AC, the NAC approach was associated with a higher LR [relative risk (RR) 1.22, P=0.015] and had similar rates of any recurrence and BC-related mortality (14). An Early Breast Cancer Trialists Collaborative Group meta-analysis which compared NAC and AC (10 RCTs, N=4,756, 9-year median follow-up) reported that NAC was significantly associated with a higher BCS rate (65 vs. 49%) and significantly higher LR rate (LRR; +5.5%; RR 1.37, 95% CI 1.17-1.61, P=0.0001) (6). To interpret these results with caution, certain researchers believed that it was the NAC-related more conservative surgery which contributed to a higher LRR (6,11). Recently, a retrospective analysis of19,151 stage II–III TNBC patients from the National Cancer Database reported that the NAC patients had a lower OS compared with the AC patients (73.6 vs. 76.8%, P<0.0001) (31). Furthermore, the subgroup of patients with pCR after the NAC were associated with a significantly improved 5-year OS compared with the non-PCR patients (86.2 vs. 62.3%, P<0.0001) (31). The authors concluded that NAC may be inferior to AC in TNBC for tumor control; however, the role of patient selection bias could not be completely ruled out (31). In the present retrospective study, a poorer PFS was associated with the ‘NAC only’ vs. ‘AC only’ groups in TNBC patients as well.

The potential role of many factors in causation meant it was challenging to explain the study findings. Numerous patient, disease and physician factors can influence the choice and continuity of NAC or AC in patients with BC. Through multivariate analyses that adjust these factors, the present study assessed the independent risk profiles of chemotherapy modes on DFS. This analysis strategy along with stratifying the population in the present study, demonstrated that compared with the traditional ‘AC only’ approach, the ‘NAC only’ approach was associated with a higher risk of tumor recurrence. Based on the results of the present study it was hypothesized that an independent link of ‘NAC only’ with a higher risk of recurrence in the real-world practice exists. It was hypothesized that this could be due to the possible unintentional use of ‘inadequate’ chemotherapy cycles in NAC patients and unrecognized difference of drug mechanism of NAC vs. AC regimen in medical and surgical practice. Furthermore, the present study indicated that the aforementioned differences could be more evident in subgroup patients with BC characterized as T2-3, N2-3, LVI+ or HER2-negative.

The strengths of the present study included the large sample size of real-world data about patients with BC with first tumor recurrence, a large number of covariates for model adjustment and the use of stratified analyses. The limitations of the present study included being unable to infer a cause-effect relationship from this retrospective observational study, the possible existence of unadjusted confounders, the lack of pCR and drug side effect data, and the lack of recorded reasons to add or discontinue the NAC or AC for additional analysis. Furthermore, the different BC populations and treatment administrations (e.g., BCS procedure, inclusion of platinum in TNBC NAC, trastuzumab availability, and pCR as guidance) mean it would be challenging to simply compare and apply the findings of the present study to patients with BC outside of China.

While the results of the present study require further evaluation, oncologists should be aware that patients with BC with a high risk of recurrence could benefit from the use of full cycles of NAC or adding AC after ‘inadequate’ NAC for improved tumor control.

NAC alone is potentially associated with a higher risk of tumor recurrence in high-risk BC subgroup patients based on real-world data from clinical practice. Patient selection of chemotherapy mode was involved in practice but could not fully explain this finding.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received

Availability of data and materials

The anonymised datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

ZSL designed and supervised this study. ZSL, YL and YJL were responsible for the data analysis and manuscript writing. YGS, YRZ, XHJ, HH and KYD participated in the data collection, data quality control, data interpretation, and analysis discussion. YJL and JZ contributed to the interpretation of data and study results as well as providing critical discussion and revision of the manuscript. ZSL and YL confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Research Ethics Committee of The Fourth Hospital of Hebei Medical University (approval no. 2017-288). All patients provided written informed consent before study enrollment.

Patient consent for publication

Not applicable.

Competing interests

The authors declared that they have no competing interests.

Authors' information

Dr Zhensheng Li: ORCID: 0000-0002-6928-0050. Dr Yunjiang Liu: ORCID: 0000-0001-7202-2004.

Glossary

Abbreviations

Abbreviations:

AC

adjunct chemotherapy

ALND

axillary lymph node dissection

ANOVA

analysis of variance

BC

breast cancer

BCS

breast-conserving surgery

CI

confidence interval

DFS

disease-free survival

ECOG

Easter Cooperative Oncology Group

ER

estrogen receptor

HER2

human epidermal growth factor receptor 2

HR

hazard ratio

LN

lymph node

LR

locoregional recurrence

LRR

locoregional recurrence rate

LVI

lymphovascular invasion

NAC

neoadjuvant chemotherapy

NCCN

National Comprehensive Cancer Network

OS

overall survival

pCR

pathologic complete response

RCT

randomized clinical trial

RR

relative risk

T-DM1

trastuzumab emtansine

TNBC

triple negative breast cancer

References

1 

Mougalian SS, Soulos PR, Killelea BK, Lannin DR, Abu-Khalaf MM, DiGiovanna MP, Sanft TB, Pusztai L, Gross CP and Chagpar AB: Use of neoadjuvant chemotherapy for patients with stage I to III breast cancer in the United States. Cancer. 121:2544–2552. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Caparica R, Brandão M and Piccart M: Systemic treatment of patients with early breast cancer: Recent updates and state of the art. Breast. 48 (Suppl 1):S7–S20. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Waks AG and Winer EP: Breast cancer treatment: A review. JAMA. 321:288–300. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Fisher B, Brown A, Mamounas E, Wieand S, Robidoux A, Margolese RG, Cruz AB Jr, Fisher ER, Wickerham DL, Wolmark N, et al: Effect of preoperative chemotherapy on local-regional disease in women with operable breast cancer: Findings from national surgical adjuvant breast and bowel project B-18. J Clin Oncol. 15:2483–2493. 1997. View Article : Google Scholar : PubMed/NCBI

5 

Moreno-Aspitia A: Neoadjuvant therapy in early-stage breast cancer. Crit Rev Oncol Hematol. 82:187–199. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Early Breast Cancer Trialists' Collaborative Group (EBCTCG), . Long-term outcomes for neoadjuvant versus adjuvant chemotherapy in early breast cancer: Meta-analysis of individual patient data from ten randomised trials. Lancet Oncol. 19:27–39. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Masuda N, Lee SJ, Ohtani S, Im YH, Lee ES, Yokota I, Kuroi K, Im SA, Park BW, Kim SB, et al: Adjuvant capecitabine for breast cancer after preoperative chemotherapy. N Engl J Med. 376:2147–2159. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Murphy BL, Day CN, Hoskin TL, Habermann EB and Boughey JC: Neoadjuvant chemotherapy use in breast cancer is greatest in excellent responders: Triple-negative and HER2+ subtypes. Ann Surg Oncol. 25:2241–2248. 2018. View Article : Google Scholar : PubMed/NCBI

9 

von Minckwitz G, Huang CS, Mano MS, Loibl S, Mamounas EP, Untch M, Wolmark N, Rastogi P, Schneeweiss A, Redondo A, et al: Trastuzumab emtansine for residual invasive HER2-positive breast cancer. N Engl J Med. 380:617–628. 2019. View Article : Google Scholar : PubMed/NCBI

10 

Vaidya JS, Massarut S, Vaidya HJ, Alexander EC, Richards T, Caris JA, Sirohi B and Tobias JS: Rethinking neoadjuvant chemotherapy for breast cancer. BMJ. 360:j59132018. View Article : Google Scholar : PubMed/NCBI

11 

Reyal F, Hamy AS and Piccart MJ: Neoadjuvant treatment: The future of patients with breast cancer. ESMO Open. 3:e0003712018. View Article : Google Scholar : PubMed/NCBI

12 

Fisher B, Bryant J, Wolmark N, Mamounas E, Brown A, Fisher ER, Wickerham DL, Begovic M, DeCillis A, Robidoux A, et al: Effect of preoperative chemotherapy on the outcome of women with operable breast cancer. J Clin Oncol. 16:2672–2685. 1998. View Article : Google Scholar : PubMed/NCBI

13 

Rastogi P, Anderson SJ, Bear HD, Geyer CE, Kahlenberg MS, Robidoux A, Margolese RG, Hoehn JL, Vogel VG, Dakhil SR, et al: Preoperative chemotherapy: updates of national surgical adjuvant breast and bowel project protocols B-18 and B-27. J Clin Oncol. 26:778–785. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Mauri D, Pavlidis N and Ioannidis JP: Neoadjuvant versus adjuvant systemic treatment in breast cancer: A meta-analysis. J Natl Cancer Inst. 97:188–194. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Wang J, Xiu BQ, Guo R, Yang BL, Zhang Q, Su YH, Li L, Ji WR, Zhang YY, Cao AY, et al: Current trend of breast cancer neoadjuvant treatment in China: A cross-sectional study. Zhonghua Zhong Liu Za Zhi. 42:931–936. 2020.(In Chinese). PubMed/NCBI

16 

Edge SB and Compton CC: The American joint committee on cancer: the 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol. 17:1471–1474. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Senkus E, Kyriakides S, Ohno S, Penault-Llorca F, Poortmans P, Rutgers E, Zackrisson S and Cardoso F; ESMO Guidelines Committee, . Primary breast cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 26 (Suppl 5):v8–v30. 2015. View Article : Google Scholar : PubMed/NCBI

18 

National Comprehensive Cancer Network (NCCN), . Clinical Practice Guideline in Oncology. Breast Cancer version 1.2021. https://www.nccn.org/professionals/physician_gls/pdf/breast.pdfMarch 20–2021

19 

Denduluri N, Chavez-MacGregor M, Telli ML, Eisen A, Graff SL, Hassett MJ, Holloway JN, Hurria A, King TA, Lyman GH, et al: Selection of optimal adjuvant chemotherapy and targeted therapy for early breast cancer: ASCO clinical practice guideline focused update. J Clin Oncol. 36:2433–2443. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Poggio F, Bruzzone M, Ceppi M, Pondé NF, La Valle G, Del Mastro L, de Azambuja E and Lambertini M: Platinum-based neoadjuvant chemotherapy in triple-negative breast cancer: A systematic review and meta-analysis. Ann Oncol. 29:1497–1508. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Association of Breast Surgery at Baso 2009, . Surgical guidelines for the management of breast cancer. Eur J Surg Oncol. 35 (Suppl 1):S1–S22. 2009. View Article : Google Scholar

22 

Early Breast Cancer Trialists' Collaborative Group (EBCTCG), . Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: An overview of the randomised trials. Lancet. 365:1687–1717. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Berry DA, Cirrincione C, Henderson IC, Citron ML, Budman DR, Goldstein LJ, Martino S, Perez EA, Muss HB, Norton L, et al: Estrogen-receptor status and outcomes of modern chemotherapy for patients with node-positive breast cancer. JAMA. 295:1658–1667. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Pondé NF, Zardavas D and Piccart M: Progress in adjuvant systemic therapy for breast cancer. Nat Rev Clin Oncol. 16:27–44. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Early Breast Cancer Trialists' Collaborative Group (EBCTCG), . Increasing the dose intensity of chemotherapy by more frequent administration or sequential scheduling: A patient-level meta-analysis of 37 298 women with early breast cancer in 26 randomised trials. Lancet. 393:1440–1452. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Harris L, Fritsche H, Mennel R, Norton L, Ravdin P, Taube S, Somerfield MR, Hayes DF and Bast RC Jr; American Society of Clinical Oncology, . American society of clinical oncology 2007 update of recommendations for the use of tumor markers in breast cancer. J Clin Oncol. 25:5287–5312. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Sparano JA and Paik S: Development of the 21-gene assay and its application in clinical practice and clinical trials. J Clin Oncol. 26:721–728. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Sparano JA, Gray RJ, Makower DF, Pritchard KI, Albain KS, Hayes DF, Geyer CE Jr, Dees EC, Perez EA, Olson JA Jr, et al: Prospective validation of a 21-gene expression assay in breast cancer. N Engl J Med. 373:2005–2014. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Cooke T, Reeves J, Lanigan A and Stanton P: HER2 as a prognostic and predictive marker for breast cancer. Ann Oncol. 12 (Suppl 1):S23–S28. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Caparica R, Lambertini M, Pondé N, Fumagalli D, de Azambuja E and Piccart M: Post-neoadjuvant treatment and the management of residual disease in breast cancer: State of the art and perspectives. Ther Adv Med Oncol. 11:17588359198277142019. View Article : Google Scholar : PubMed/NCBI

31 

Bagegni NA, Tao Y and Ademuyiwa FO: Clinical outcomes with neoadjuvant versus adjuvant chemotherapy for triple negative breast cancer: A report from the national cancer database. PLoS One. 14:e02223582019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2023
Volume 25 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li Z, Li Y, Liu Y, Shang Y, Zhou Y, Ji X, Han H, Du K and Zhang J: Real‑world data indicated that neoadjuvant chemotherapy alone was associated with a higher risk of tumor recurrence in high‑risk breast cancer subgroup patients. Oncol Lett 25: 193, 2023
APA
Li, Z., Li, Y., Liu, Y., Shang, Y., Zhou, Y., Ji, X. ... Zhang, J. (2023). Real‑world data indicated that neoadjuvant chemotherapy alone was associated with a higher risk of tumor recurrence in high‑risk breast cancer subgroup patients. Oncology Letters, 25, 193. https://doi.org/10.3892/ol.2023.13779
MLA
Li, Z., Li, Y., Liu, Y., Shang, Y., Zhou, Y., Ji, X., Han, H., Du, K., Zhang, J."Real‑world data indicated that neoadjuvant chemotherapy alone was associated with a higher risk of tumor recurrence in high‑risk breast cancer subgroup patients". Oncology Letters 25.5 (2023): 193.
Chicago
Li, Z., Li, Y., Liu, Y., Shang, Y., Zhou, Y., Ji, X., Han, H., Du, K., Zhang, J."Real‑world data indicated that neoadjuvant chemotherapy alone was associated with a higher risk of tumor recurrence in high‑risk breast cancer subgroup patients". Oncology Letters 25, no. 5 (2023): 193. https://doi.org/10.3892/ol.2023.13779