Purpose:

While various studies have highlighted the prognostic significance of pathologic complete response (pCR) after neoadjuvant chemotherapy (NAT), the impact of additional adjuvant therapy after pCR is not known.

Experimental Design:

PubMed was searched for studies with NAT for breast cancer and individual patient-level data was extracted for analysis using plot digitizer software. HRs, with 95% probability intervals (PI), measuring the association between pCR and overall survival (OS) or event-free survival (EFS), were estimated using Bayesian piece-wise exponential proportional hazards hierarchical models including pCR as predictor.

Results:

Overall, 52 of 3,209 publications met inclusion criteria, totaling 27,895 patients. Patients with a pCR after NAT had significantly better EFS (HR = 0.31; 95% PI, 0.24–0.39), particularly for triple-negative (HR = 0.18; 95% PI, 0.10–0.31) and HER2+ (HR = 0.32; 95% PI, 0.21–0.47) disease. Similarly, pCR after NAT was also associated with improved survival (HR = 0.22; 95% PI, 0.15–0.30). The association of pCR with improved EFS was similar among patients who received subsequent adjuvant chemotherapy (HR = 0.36; 95% PI, 0.19–0.67) and those without adjuvant chemotherapy (HR = 0.36; 95% PI, 0.27–0.54), with no significant difference between the two groups (P = 0.60).

Conclusions:

Achieving pCR following NAT is associated with significantly better EFS and OS, particularly for triple-negative and HER2+ breast cancer. The similar outcomes with or without adjuvant chemotherapy in patients who attain pCR likely reflects tumor biology and systemic clearance of micrometastatic disease, highlighting the potential of escalation/deescalation strategies in the adjuvant setting based on neoadjuvant response.

See related commentary by Esserman, p. 2771

Translational Relevance

Prior studies have highlighted the prognostic significance of pathologic complete response (pCR) after neoadjuvant chemotherapy in breast cancer. However, the clinical impact of adjuvant chemotherapy following pCR is not known. In the largest individual patient-level meta-analysis to date on the topic (N = 27,895), we demonstrated pCR was strongly associated with improved event-free and overall survival, and the receipt of additional cytotoxic chemotherapy following surgery did not further improve outcomes. The study results support the use of escalation/deescalation strategies in the adjuvant setting based on neoadjuvant response and has broad implications for the drug approval process.

Neoadjuvant chemotherapy (NAT) is increasingly being utilized as the first-line therapy for the management of high-risk localized breast cancer. Studies have demonstrated no difference in survival between adjuvant or neoadjuvant setting (1, 2). NAT for breast cancer is an established therapeutic option for selected high-risk, locally advanced, or unresectable breast cancers, or to improve eligibility for breast conserving surgery (BCS; ref. 2). Because the primary tumor remains intact during therapy, the neoadjuvant treatment strategy allows for monitoring of treatment response and discontinuing of therapy in the event of disease progression.

From a research perspective, the neoadjuvant setting has become recognized as a human in vivo system to evaluate predictive biomarkers, surrogate endpoints, and the efficacy of therapies including novel agents (3). The neoadjuvant therapy model provides a potential efficient trial design to explore the efficacy of novel therapies utilizing pathologic complete response (pCR) as a surrogate marker for disease free-survival and overall survival (3).

Yet, the prognostic significance of pCR after neoadjuvant chemotherapy remains somewhat controversial. While pCR demonstrates sensitivity to agents received in the neoadjuvant setting, true demonstration of treatment efficacy is dependent on its ability to predict long-term outcomes of recurrence and death, and this issue has not been completely settled in the literature. In a pooled analysis of 12 clinical trials by Cortazar and colleagues, the authors demonstrated that pCR is associated with improved event-free survival (EFS), but the association between the magnitude of treatment-induced pCR change and corresponding improvement in EFS could not be established (i.e., delta pCR and delta EFS; ref. 4). Similarly, a meta-regression of 29 randomized prospective studies of NAT demonstrated pCR to be a strong prognostic factor, but the magnitude of relationship between pCR and EFS varied by type of NAT (5). However, most neoadjuvant trials are powered to detect a difference in pCR among regimens, and likewise are not powered for long-term outcomes. Furthermore, these studies did not evaluate the impact of pCR on the utility of adjuvant therapy, which could potentially influence the clinical outcomes in patients with localized breast cancer. In addition, clinical subtype of breast cancer is an important factor to consider given differences in tumor biology as well as targeted therapy usage. The association of pCR with improved long-term outcomes is recognized for HER2-positive (HER2+) breast cancer and triple-negative (TN) breast cancer (TNBC; ref. 6), but is less understood for hormone receptor–positive (HR+)/HER2 breast cancer, where pCR is less common and adjuvant endocrine therapy is the mainstay of systemic therapy.

The objective of this study was to conduct a comprehensive meta-analysis of studies on NAT for localized breast cancer using extracted patient-level data to ascertain the potential association between pCR and subsequent breast cancer recurrence as well as survival, with careful consideration of tumor subtype, and the relationship between pCR and adjuvant treatment in modulating clinical outcomes.

Identification of studies

On the basis of the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement (7), a librarian-led systematic search restricted to English of PubMed was initially performed in September 2016 to identify potentially eligible studies. Meeting abstracts were excluded. The search strategy keywords included “breast cancer,” “neoadjuvant therapy,” “preoperative therapy,” “pathologic complete response,” “survival”, and “recurrence.” The detailed search strategy (eAppendix 1) and the flow diagram detailing study selection (Supplementary Fig. S1) are available in the Supplementary Data. We also reviewed reference lists of eligible studies, manuscripts citing the selected studies, and relevant reviews to identify additional publications. If it was determined that more than one publication reported on the same trial or patient cohort, the outcomes from the most recent publication were included.

Eligibility criteria

Inclusion criteria were clinical trials, prospective cohort studies, or retrospective cohort studies that reported pCR results after neoadjuvant chemotherapy as well as breast cancer recurrence and/or survival stratified by the presence or absence of pCR, with a total sample size of 25 patients or greater. Publications were included regardless of neoadjuvant regimen received. Studies including any neoplasm other than female breast cancer were excluded, as well as studies analyzing unresectable or metastatic breast cancer. Endocrine therapy–based neoadjuvant studies and neoadjuvant studies with radiation were also excluded. Studies were excluded from subanalyses based on receptor subtypes if HER2 status was unknown. Furthermore, only those publications where individual patient-level data were extractable either in the form of Kaplan–Meier (KM) curves with event data and/or survival estimates (e.g., median survival, and/or another landmark event such as 5-year survival with event data) were included.

Data extraction

For each selected manuscript, the following information was recorded by two independent reviewers: primary author name, year of publication, sample size, duration of follow-up, definition of pCR, patient and tumor characteristics, neoadjuvant regimen, adjuvant regimen if applicable, number of patients achieving a pCR, and number of outcome events by pCR status. When available, outcomes based on the major breast cancer subtypes were extracted. We obtained the individual patient data (IPD) used in our analysis from the selected manuscripts by one of two methods. If available, method one used the KM curves [extracted from the manuscript using the DigitizeIt (c) software; ref. 8] to reconstruct the IPD data via the method of Guyot and colleagues (9). Alternatively, if KM curves were not available, method two used either a measure of median survival or a landmark event and assumed an exponential distribution to impute the IPD from the metric. Identical methods to recover the IPD data were used for both the pCR and non-pCR groups within each study to reduce bias.

Evaluation of bias

A broad inclusion criterion was utilized as detailed above. All manuscripts were peer-reviewed publications and abstracts were therefore not included. Studies were allowable regardless of industry sponsorship. Demographic information on the study population and treatment details for each included manuscript was extracted and is presented in Table 1 and Supplementary Table S1 for comparison. A broad global population was represented.

Table 1.

Patient and study characteristics of included manuscripts.

First authorYearStudy typeEvaluable sample sizeSubtypes includedDefinition pCRMeasure of recurrenceRecurrence median follow-up (mo)Survival median follow-up (mo)Survival median follow-up (mo)
Kuerer 1999 Pooled (NRCT) 372 All ypT0/is ypN0 DFS 58.00 58.00 58.00 
Chollet 2002 Pooled (NRCT) 396 All ypT0 ypN0 DFS 96.00 96.00 96.00 
Dieras 2004 RCT 200 All ypT0/is ypN0 DFS 31.00 NA NA 
Lee 2004 NRCT 57 All ypT0/is ypN0 DFS 48.00 48.00 48.00 
Ring 2004 Retrospective 435 All ypT0/is ypN0 DFS 53.00 53.00 53.00 
Abrial 2005 Retrospective 651 All ypT0/is ypN0 DFS 91.20 91.20 91.20 
Guarneri 2006 Retrospective 1,163 All ypT0/is ypN0 PFS 107.00 118.00 118.00 
Hurley 2006 NRCT 48 HER2+ ypT0/is ypN0 PFS 43.00 43.00 43.00 
Andre 2007 Retrospective 534 All ypT0/is ypN0 RFS 31.20 31.20 31.20 
Eralp 2008 Retrospective 102 All ypT0/is ypN0 DFS 43.00 NA NA 
Liedtke 2008 Retrospective 1,118 All ypT0/is ypN0 NA NA 36.00 36.00 
Al-Tweigeri 2009 NRCT 59 All ypT0/is ypN0 DFS 60.00 60.00 60.00 
Frasci 2009 NRCT 74 TNBC ypT0/is ypN0 DFS 41.00 NA NA 
Chang 2010 NRCT 71 All ypT0/is ypN0 RFS 22.80 NA NA 
Chen 2010 Retrospective 225 All ypT0/is ypN0 DFS 32.50 32.50 32.50 
Jinno 2010 NRCT 71 All ypT0 ypN0 DFS 29.00 NA NA 
Kim 2010 Retrospective 257 All ypT0/is ypN0 DFS 21.30 NA NA 
Masuda 2010 Retrospective 33 All ypT0/is ypN0 DFS 24.00 NA NA 
Arun 2011 Retrospective 317 All (BRCA+ enriched) ypT0/is ypN0 RFS 38.40 38.40 38.40 
Fasching 2011 Retrospective 520 All ypT0 ypN0 DDFS 33.60 33.60 33.60 
Wu 2011 Retrospective 249 All ypT0/is ypN0 DFS 48.20 48.20 48.20 
Esserman 2012 NRCT 172 All ypT0/is ypN0 RFS 46.80 NA NA 
Im 2012 NRCT 53 HER2 ypT0/is ypN0 RFS 40.00 NA NA 
Melichar 2012 Retrospective 318 All ypT0/is ypN0 RFS 68.00 68.00 68.00 
Yoo 2012 Retrospective 276 All ypT0/is ypN0 PFS 32.30 32.30 32.30 
Zhang 2012 Retrospective 102 HER2 ypT0 ypN0 DFS 25.90 NA NA 
Guarneri 2013 Retrospective 107 All ypT0/is ypN0 DFS ND NA NA 
Guiu 2013 Retrospective 348 All ypT0/is ypN0 DFS 84.00 NA NA 
Hurley 2013 Retrospective 144 TNBC ypT0/is ypN0 PFS 48.00 45.60 45.60 
Krishnan 2013 Retrospective 365 All ypT0/is ypN0 DFS 49.00 49.00 49.00 
Marme 2013 Pooled (NRCT) 149 All ypT0 ypN0 DFS 82.80 82.80 82.80 
Natoli 2013 Retrospective 80 HER2 ypT0/is ypN0 DFS 32.00 NA NA 
Cortazar 2014 Pooled (RCT/NRCT) 11,955 All ypT0/is ypN0 EFS 64.80 64.44 64.44 
de Azambuja 2014 RCT 419 HER2 ypT0/is ypN0 EFS 45.30 45.30 45.30 
Groheux 2014 Retrospective 74 TNBC ypT0/is ypN0 EFS 31.00 NA NA 
Kawajiri 2014 Retrospective 90 All ypT0/is ypN0 DFS 53.00 53.00 53.00 
Takada 2014 Retrospective 764 HER2 ypT0/is ypN0 DFS 42.00 NA NA 
Tanioka 2014 Retrospective 366 HER2 ypT0/is ypN0 RFS 55.00 55.00 55.00 
Wang 2014 Retrospective 309 All ypT0/is ypN0 DFS 60.00 NA NA 
Al-Tweigeri 2015 NRCT 80 All ypT0/is ypN0 DFS 43.00 43.00 43.00 
Bear 2015 RCT 1,186 HR+/HER2-, TNBC ypT0/is ypN0 DFS 56.40 56.40 56.40 
Gonzalez-Angulo 2015 Retrospective 589 HER2 ypT0/is ypN0 RFS 45.00 45.00 45.00 
Ko 2015 Retrospective 174 All ypT0/is ypN0 RFS 54.80 NA NA 
Liu 2015 Retrospective 108 HER2 ypT0/is ypN0 EFS 32.00 NA NA 
Mayer 2015 Pooled (NRCT) 80 HER2 ypT0/is ypN0 RFS 105.60 NA NA 
Villarreal-Garza 2015 Retrospective 244 HER2 ypT0/is ypN0 DFS 47.00 47.00 47.00 
Zelnak 2015 NRCT 27 HER2 ypT0/is ypN0 DFS 52.20 NA NA 
Gianni 2016 RCT 417 HER2 ypT0/is ypN0 PFS 60.00 NA NA 
Li 2016 Retrospective 186 TNBC ypT0/is ypN0 RFS 48.10 NA NA 
Shao 2016 Retrospective 50 TNBC ypT0/is ypN0 PFS 54.50 54.50 54.50 
Villarreal-Garza 2016 Retrospective 1,639 All ypT0/is ypN0 DFS 50.80 50.80 50.80 
Zhang 2016 RCT 87 TNBC ypT0/is ypN0 RFS 55.00 55.00 55.00 
First authorYearStudy typeEvaluable sample sizeSubtypes includedDefinition pCRMeasure of recurrenceRecurrence median follow-up (mo)Survival median follow-up (mo)Survival median follow-up (mo)
Kuerer 1999 Pooled (NRCT) 372 All ypT0/is ypN0 DFS 58.00 58.00 58.00 
Chollet 2002 Pooled (NRCT) 396 All ypT0 ypN0 DFS 96.00 96.00 96.00 
Dieras 2004 RCT 200 All ypT0/is ypN0 DFS 31.00 NA NA 
Lee 2004 NRCT 57 All ypT0/is ypN0 DFS 48.00 48.00 48.00 
Ring 2004 Retrospective 435 All ypT0/is ypN0 DFS 53.00 53.00 53.00 
Abrial 2005 Retrospective 651 All ypT0/is ypN0 DFS 91.20 91.20 91.20 
Guarneri 2006 Retrospective 1,163 All ypT0/is ypN0 PFS 107.00 118.00 118.00 
Hurley 2006 NRCT 48 HER2+ ypT0/is ypN0 PFS 43.00 43.00 43.00 
Andre 2007 Retrospective 534 All ypT0/is ypN0 RFS 31.20 31.20 31.20 
Eralp 2008 Retrospective 102 All ypT0/is ypN0 DFS 43.00 NA NA 
Liedtke 2008 Retrospective 1,118 All ypT0/is ypN0 NA NA 36.00 36.00 
Al-Tweigeri 2009 NRCT 59 All ypT0/is ypN0 DFS 60.00 60.00 60.00 
Frasci 2009 NRCT 74 TNBC ypT0/is ypN0 DFS 41.00 NA NA 
Chang 2010 NRCT 71 All ypT0/is ypN0 RFS 22.80 NA NA 
Chen 2010 Retrospective 225 All ypT0/is ypN0 DFS 32.50 32.50 32.50 
Jinno 2010 NRCT 71 All ypT0 ypN0 DFS 29.00 NA NA 
Kim 2010 Retrospective 257 All ypT0/is ypN0 DFS 21.30 NA NA 
Masuda 2010 Retrospective 33 All ypT0/is ypN0 DFS 24.00 NA NA 
Arun 2011 Retrospective 317 All (BRCA+ enriched) ypT0/is ypN0 RFS 38.40 38.40 38.40 
Fasching 2011 Retrospective 520 All ypT0 ypN0 DDFS 33.60 33.60 33.60 
Wu 2011 Retrospective 249 All ypT0/is ypN0 DFS 48.20 48.20 48.20 
Esserman 2012 NRCT 172 All ypT0/is ypN0 RFS 46.80 NA NA 
Im 2012 NRCT 53 HER2 ypT0/is ypN0 RFS 40.00 NA NA 
Melichar 2012 Retrospective 318 All ypT0/is ypN0 RFS 68.00 68.00 68.00 
Yoo 2012 Retrospective 276 All ypT0/is ypN0 PFS 32.30 32.30 32.30 
Zhang 2012 Retrospective 102 HER2 ypT0 ypN0 DFS 25.90 NA NA 
Guarneri 2013 Retrospective 107 All ypT0/is ypN0 DFS ND NA NA 
Guiu 2013 Retrospective 348 All ypT0/is ypN0 DFS 84.00 NA NA 
Hurley 2013 Retrospective 144 TNBC ypT0/is ypN0 PFS 48.00 45.60 45.60 
Krishnan 2013 Retrospective 365 All ypT0/is ypN0 DFS 49.00 49.00 49.00 
Marme 2013 Pooled (NRCT) 149 All ypT0 ypN0 DFS 82.80 82.80 82.80 
Natoli 2013 Retrospective 80 HER2 ypT0/is ypN0 DFS 32.00 NA NA 
Cortazar 2014 Pooled (RCT/NRCT) 11,955 All ypT0/is ypN0 EFS 64.80 64.44 64.44 
de Azambuja 2014 RCT 419 HER2 ypT0/is ypN0 EFS 45.30 45.30 45.30 
Groheux 2014 Retrospective 74 TNBC ypT0/is ypN0 EFS 31.00 NA NA 
Kawajiri 2014 Retrospective 90 All ypT0/is ypN0 DFS 53.00 53.00 53.00 
Takada 2014 Retrospective 764 HER2 ypT0/is ypN0 DFS 42.00 NA NA 
Tanioka 2014 Retrospective 366 HER2 ypT0/is ypN0 RFS 55.00 55.00 55.00 
Wang 2014 Retrospective 309 All ypT0/is ypN0 DFS 60.00 NA NA 
Al-Tweigeri 2015 NRCT 80 All ypT0/is ypN0 DFS 43.00 43.00 43.00 
Bear 2015 RCT 1,186 HR+/HER2-, TNBC ypT0/is ypN0 DFS 56.40 56.40 56.40 
Gonzalez-Angulo 2015 Retrospective 589 HER2 ypT0/is ypN0 RFS 45.00 45.00 45.00 
Ko 2015 Retrospective 174 All ypT0/is ypN0 RFS 54.80 NA NA 
Liu 2015 Retrospective 108 HER2 ypT0/is ypN0 EFS 32.00 NA NA 
Mayer 2015 Pooled (NRCT) 80 HER2 ypT0/is ypN0 RFS 105.60 NA NA 
Villarreal-Garza 2015 Retrospective 244 HER2 ypT0/is ypN0 DFS 47.00 47.00 47.00 
Zelnak 2015 NRCT 27 HER2 ypT0/is ypN0 DFS 52.20 NA NA 
Gianni 2016 RCT 417 HER2 ypT0/is ypN0 PFS 60.00 NA NA 
Li 2016 Retrospective 186 TNBC ypT0/is ypN0 RFS 48.10 NA NA 
Shao 2016 Retrospective 50 TNBC ypT0/is ypN0 PFS 54.50 54.50 54.50 
Villarreal-Garza 2016 Retrospective 1,639 All ypT0/is ypN0 DFS 50.80 50.80 50.80 
Zhang 2016 RCT 87 TNBC ypT0/is ypN0 RFS 55.00 55.00 55.00 

Abbreviations: DDFS, distant disease free survival; DFS, disease-free survival; ER+, estrogen receptor-positive; mo, months; NA, not applicable; NRCT, non-randomized clinical trial; PFS, progression-free survival; RCT, randomized clinical trial; RFS, relapse free survival.

Endpoints

The primary clinical outcomes were breast cancer recurrence and overall survival. Results were examined in the overall study population and in subanalyses based on tumor subtype and treatment characteristics. Overall survival (OS) results were used to determine survival. A variety of endpoints were used among the manuscripts to describe breast cancer recurrence, including event-free survival (EFS), progression-free survival, recurrence-free survival, relapse-free survival, disease-free survival, and distant disease-free survival. These endpoints were treated as equivalent for the aggregate analyses and EFS is used throughout this article as a representative term, as done in previous meta-analyses (6). Endpoints with local recurrence only were excluded. The number of patients with and without a pCR (both breast and lymph nodes) in each manuscript was extracted. Allowable definitions of pCR were ypT0 ypN0 (no invasive or noninvasive residual in breast or nodes) and ypT0/is ypN0 (no invasive residual in breast or nodes; noninvasive breast residuals allowed), as suggested by FDA guidelines (10). If results were available for both of the allowable pCR definitions, ypT0/is ypN0 was utilized. Studies only listing pCR breast (with no information on lymph nodes) were excluded as well as studies utilizing the Sataloff criteria for pathologic tumor status given this definition allows minimal residual disease in the breast (11). Studies were considered to have used adjuvant chemotherapy if the majority (≥90%) of patients received adjuvant chemotherapy, and studies were considered to have not used adjuvant chemotherapy if the minority (<10%) of patients received adjuvant chemotherapy.

Statistical analysis

HRs measuring the association between pCR and OS or EFS, were estimated using Bayesian piece-wise exponential proportional hazards hierarchical models (considering dispersed prior distributions) using pCR as a predictor, together with their 95% probability intervals (95% PI, the Bayesian equivalent of a confidence interval). A piece-wise exponential model assumes the hazard of an event to be constant within pre-specified time intervals (12). Following Broglio and colleagues, in our analyses, we assumed that the hazard of OS or recurrence remained constant within each of 22 intervals of follow-up, the first 2 spanning 6 months and the remaining each spanning 12 (6). Random effects were considered when pooling data across multiple studies to account for heterogeneity. More details, including specification of the prior distributions and the computational strategy adopted to fit the model, are provided in the Supplementary Data (eAppendix 2). In addition, to explore the effect of pCR on OS or EFS in selected subgroups, we performed several stratified analyses using the model described above.

A total of 3,209 citations with associated abstracts were reviewed. Of these, a total of 166 were selected for full review. From these, 107 were excluded for not meeting eligibility criteria and 12 were excluded because individual patient-level data could not be extracted using the described methods. An additional five manuscripts were identified through reviewing reference lists of eligible studies, manuscripts citing the selected studies, and relevant reviews. Ultimately, 52 studies met the criteria for inclusion (Fig. 1; refs. 4, 13–62).

Figure 1.

Selection of studies for meta-analysis. On the basis of the search criteria, 3,209 citations with associated abstracts were reviewed. Of these, a total of 166 were selected for full review and ultimately 52 studies met the criteria for inclusion.

Figure 1.

Selection of studies for meta-analysis. On the basis of the search criteria, 3,209 citations with associated abstracts were reviewed. Of these, a total of 166 were selected for full review and ultimately 52 studies met the criteria for inclusion.

Close modal

Study characteristics

The selected studies were published from 1999 to 2016. The study sample size available for analysis ranged from 27 to 11,955, and featured a broad global patient population, including Europe, the United States, Mexico, Kuwait, Saudi Arabia, China, Japan, and Korea. Summary details on the selected studies are shown in Supplementary Table S1 and a detailed list of each study is shown in Table 1. Further details on each individual study and the associated patient population can be found in Supplementary Table S2. The CTNeoBC FDA meta-analysis (4), a pooled analysis of 12 randomized control trials (RCT), was treated as a single study for this analysis given most of its studies did not make extractable IPD publicly available. The 52 studies included in our analysis represent 27,895 total evaluable patients, with 14,254 (51.1%) from RCTs, 1,709 patients (6.1%) from nonrandomized clinical trials, and 11,932 patients (42.8%) from retrospective cohort studies. The overall pCR rate based on all 52 studies was 21.1% (range: 10.1%–74.2%), with the highest rates of pCR seen in HER2+ tumors at 36.4% (range: 17.5%–74.2%) and TN tumors at 32.6% (range: 20.3%–62.2%), with HR+/HER2 tumors the lowest at 9.3% (range: 5.5%–31.3%).

EFS and overall survival

Overall, patients who had pCR, as compared with absence of pCR, had significantly better EFS (HR = 0.31; 95% PI, 0.24–0.39, n = 26,378) as outlined in Fig. 2A. Similarly, patients who had pCR, as compared with absence of pCR, had significantly better OS (HR = 0.22; 95% PI, 0.15–0.30, n = 23,329) as outlined in Fig. 2B.

Figure 2.

Association of pCR with (A) event free survival and (B) overall survival. Forest plot of the overall HR estimate with the 95% PI for the association of pCR with the long-term outcomes EFS (A) and overall survival (OS; B), as compared with residual disease (RD). For comparison, the raw study-specific HR estimates are reported. The location of the box indicates the estimated HR for that study; the size of the box represents the relative number of events per study. HR and 95% PI for overall effects are also reported. The dashed line oriented at 1 represents the null of no difference.

Figure 2.

Association of pCR with (A) event free survival and (B) overall survival. Forest plot of the overall HR estimate with the 95% PI for the association of pCR with the long-term outcomes EFS (A) and overall survival (OS; B), as compared with residual disease (RD). For comparison, the raw study-specific HR estimates are reported. The location of the box indicates the estimated HR for that study; the size of the box represents the relative number of events per study. HR and 95% PI for overall effects are also reported. The dashed line oriented at 1 represents the null of no difference.

Close modal

Trial data versus retrospective data

The association of pCR with significantly improved EFS remained when only clinical trials were considered (HR = 0.30, 95% PI: 0.20–0.46, n = 15,873; Supplementary Fig. S1). Similarly, when only clinical trials were considered, the association of pCR with significantly improved OS was also observed (HR 0.31, 95% PI: 0.14–0.68, n = 14,431, Supplementary Fig. S2).

Role of duration of follow-up

The median follow-up time among all studies was 48 months (range 21.3–107) for EFS and 49.9 months (range 31.2–118) for OS. Among the subset of studies with 5 years or more of follow-up, the association of pCR with improved EFS (HR 0.45, 95% PI: 0.26–0.76, n = 15,449) remained (Supplementary Fig. S3).

Clinical outcomes among major breast cancer subtypes

We evaluated the association between pCR and clinical outcomes by three major clinical subtypes of breast cancer. The association of pCR with better EFS was statistically significant in patients with TNBC (HR = 0.18; 95% PI, 0.10–0.31; n = 2,039), HER2+ breast cancer (HR = 0.31; 95% PI, 0.21–0.50; n = 5,711), and trended toward significance for HR+ breast cancer (HR = 0.15; 95% PI, 0.02–1.10; n = 3,385) as outlined in Supplementary Fig. S4A–S4C. Similarly, the association of pCR with significantly improved survival was seen in TNBC (HR = 0.20; 95% PI, 0.07–0.41, n = 778) and HER2+ breast cancer (HR = 0.13; 95% PI, 0.04–0.35, n = 1,654) as outlined in Supplementary Fig. S5A and S5B. A significant relationship between pCR and improved survival was also noted in HR+ breast cancer (HR = 0.0003, 95% PI, 2.70E−11–0.81, n = 1,872) as outlined in Supplementary Fig. S5C, but wide probability intervals were observed.

In addition, we constructed model-based survival curves to evaluate the temporal relationship between pCR and EFS, overall and by breast cancer subtypes. As demonstrated in Fig. 3AD, patients who had a pCR achieved a 5-year EFS of 88% (95% PI, 85%–91%) while those without pCR had a 5-year EFS of 67% (95% PI: 63%–71%). Among patients with TNBC, patients with pCR had a 5-year EFS of 90% (95% PI, 81%–95%) while those without pCR had a 5-year EFS of 57% (95% PI, 41%–70%). For HER2+ subgroup, patients with pCR had a 5-year EFS of 86% (95% PI, 74%–94%), while those without pCR had a 5-year EFS of 63% (95% PI, 43%–78%). Among HR+ subgroup, those with pCR had a 5-year EFS of 97% (95% PI, 87%–100%), while those without a pCR had a 5-year EFS of 88% (95% PI, 75%–95%). Similar results were observed for OS. As demonstrated in Supplementary Fig. S6A–S6D, patients who experienced pCR achieved a 5-year OS of 94% (95% PI, 90%–96%), while those without a pCR achieved a 5-year OS of 75% (95% PI, 65%–82%). Among TN patients with pCR, the 5-year OS was 84% (95% PI, 60%–97%), while those without pCR had a 5-year OS of 47% (95% PI, 13%–77%). For HER2+ patients, those who experienced pCR achieved a 5-year OS of 95% (95% PI, 89%–99%), while those without a pCR achieved a 5-year OS of 76% (95% PI, 63%–88%). Among HR+ patients, those who experienced pCR achieved a 5-year OS of 98% (95% PI, 86%–100%), while those without a pCR achieved a 5-year OS of 82% (95% PI, 3%–97%).

Figure 3.

A–D, Relationship between pCR and EFS overall and among the major breast cancer subtypes. KM curves depicting the relationship between pCR and EFS overall (A), in TNBC (B), HER2+ breast cancer (C), and hormone receptor–positive breast cancer (D), based on HR data from the studies. The blue line represents the pCR group and the orange line represents the residual disease group. The shaded regions represent the 95% pointwise PI for their respective color (blue line, pCR group; orange line, RD group).

Figure 3.

A–D, Relationship between pCR and EFS overall and among the major breast cancer subtypes. KM curves depicting the relationship between pCR and EFS overall (A), in TNBC (B), HER2+ breast cancer (C), and hormone receptor–positive breast cancer (D), based on HR data from the studies. The blue line represents the pCR group and the orange line represents the residual disease group. The shaded regions represent the 95% pointwise PI for their respective color (blue line, pCR group; orange line, RD group).

Close modal

Among HER2+ patients, we also evaluated the role of neoadjuvant and adjuvant anti-HER2 therapy. For HER2+ patients receiving neoadjuvant anti-HER2 therapy, those with pCR had improved EFS compared with those with RD (HR = 0.33; 95% PI, 0.19–0.61; n = 4,636), as seen in Supplementary Fig. S7A. Similarly, among patients who did not receive neoadjuvant ani-HER2 therapies, those with pCR experienced improved outcomes compared with those with RD (HR = 0.19; 95% PI, 0.03–0.83; n = 213) as demonstrated in Supplementary Fig. S7B. In the adjuvant setting, patients receiving adjuvant anit-HER2 therapy who had a pCR experienced superior EFS compared with those with RD (HR = 0.38; 95% PI, 0.21–0.68; n = 1,962), as seen in Supplementary Fig. S8A. For HER2+ patients who did not receive adjuvant anti-HER2 therapy, EFS was greater in the pCR group compared with the RD group (HR = 0.12; 95% PI, 0–1.66; n = 133), although sample size was limited and results were not significant (Supplementary Fig. S8B).

Role of adjuvant cytotoxic chemotherapy

We then evaluated the association between pCR and clinical outcomes by adjuvant chemotherapy usage. Among patients who received additional cytotoxic chemotherapy in the adjuvant setting, pCR remained associated with significantly improved EFS (HR = 0.36; 95% PI, 0.19–0.67; n = 1,601), as seen in Fig. 4A and B and Supplementary Fig. S9A. Similarly, among patients who did not receive cytotoxic chemotherapy in the adjuvant setting, pCR remained associated with significantly improved EFS (HR = 0.36; 95% PI, 0.27–0.54; n = 18,462), as outlined in Fig. 4A and B and Supplementary Fig. S9A. Similar results were observed in terms of overall survival (Supplementary Fig. S10A and S10B). As evident from model-based survival curves (Fig. 4A), patients who had pCR and received adjuvant chemotherapy achieved a 5-year EFS of 86% (95% PI, 74%–93%), which was similar to the 5-year EFS of 88% (95% PI, 81%–92%) among those who had pCR, but received no adjuvant chemotherapy. In statistical comparison of pCR with EFS among the adjuvant chemotherapy group versus the no adjuvant chemotherapy group, no significant difference was seen between the two groups using a paired t test (difference in log-HR: 0.02, 95% PI: −0.75–0.73; P = 0.60).

Figure 4.

A and B, Impact of adjuvant chemotherapy on the relationship between pCR and EFS. A, KM curves depicting the relationship between pCR and EFS based on receipt of chemotherapy. The color blue represents the patient subpopulation where 10% or less of the patients received adjuvant chemotherapy. The color orange represents the patient subpopulation where 90% or more of the patients received adjuvant chemotherapy. The shaded regions represent the 95% pointwise PI for their respective color. B, The left forest plot is representative of the populations with at least 90% of patients receiving adjuvant chemotherapy while the forest plot on the right is representative of the populations with at most 10% of patients receiving adjuvant chemotherapy, with both comparing pCR to residual disease (RD). The HR estimate with the 95% PI are shown overall. For comparison, the raw study-specific HR estimates are reported. The location of the box indicates the estimated HR for that study; the size of the box represents the relative number of events per study. The dashed line oriented at 1 represents the null of no difference.

Figure 4.

A and B, Impact of adjuvant chemotherapy on the relationship between pCR and EFS. A, KM curves depicting the relationship between pCR and EFS based on receipt of chemotherapy. The color blue represents the patient subpopulation where 10% or less of the patients received adjuvant chemotherapy. The color orange represents the patient subpopulation where 90% or more of the patients received adjuvant chemotherapy. The shaded regions represent the 95% pointwise PI for their respective color. B, The left forest plot is representative of the populations with at least 90% of patients receiving adjuvant chemotherapy while the forest plot on the right is representative of the populations with at most 10% of patients receiving adjuvant chemotherapy, with both comparing pCR to residual disease (RD). The HR estimate with the 95% PI are shown overall. For comparison, the raw study-specific HR estimates are reported. The location of the box indicates the estimated HR for that study; the size of the box represents the relative number of events per study. The dashed line oriented at 1 represents the null of no difference.

Close modal

To our knowledge, this study, including a total of 52 studies representing 27,895 patients, is the largest meta-analysis exploring the significance of pCR following NAT. This is the first study to specifically explore the role of adjuvant cytotoxic chemotherapy following pCR after neoadjuvant treatment, and we notably found this did not further improve outcomes. The results of this comprehensive meta-analysis overall suggest pCR is a strong surrogate endpoint for TNBC and HER2+ breast cancer. Our results are consistent with other smaller studies and support the FDA's decision to use pCR rate as a surrogate marker of efficacy from neoadjuvant treatment, particularly for TNBC and HER2+ breast cancer (10, 63–65).

The study results have major implications for the field. Advances in adjuvant treatment have significantly improved breast cancer outcomes. However, as the bar is progressively set higher, it becomes more difficult to demonstrate therapeutic improvement in the adjuvant setting, with long follow-up required to see the number of events needed from a statistical standpoint. Between the expense of such trials, the large number of patients needed, and the need to assess therapies more efficiently in the era of targeted therapy, large adjuvant trials are becoming increasingly recognized as impractical in breast cancer (66). The neoadjuvant setting has therefore become recognized as an efficient model for drug development and is utilized by the FDA (3, 66). I-SPY (Investigation of Serial Studies to Predict Your Therapeutic Response with Imaging and Molecular Analysis) 2, a multicenter, randomized, phase II trial with multiple arms and pCR as the primary endpoint, utilizes an adaptive strategy for matching targeted therapies for breast cancer with the patients most likely to benefit from them (67). One of the major strengths of the I-SPY2 approach is its ability to triage promising new therapies and novel combinations in a relatively short time frame (68). Our results support this approach, and continued exploration of novel neoadjuvant clinical trial designs is needed to advance the field. The rapidly evolving field of blood-based biomarkers may also change the interpretation of pCR in the future through the identification of minimal residual disease with circulating tumor DNA, offering a number of avenues for novel trial design (69).

The potential role of adjuvant therapy after neoadjuvant therapy in influencing the relationship between pCR and survival was carefully considered in our study. Our results demonstrate the survival benefit is maintained whether adjuvant chemotherapy was received, with a similar magnitude, though it should be noted that this was not a randomized trial between adjuvant chemotherapy (vs. not) and there could be inherent differences between the two groups. Nevertheless, the results are based on adequately powered meta-analysis of multiple studies and represent the best evidence to date. The finding possibly reflects tumor biology wherein tumors sensitive to NAT in breast and lymph nodes are also typically sensitive to the therapy in micrometastatic sites. Presence of complete response in breast and axilla is likely associated with response in micrometastatic sites, minimizing the magnitude of benefit from additional adjuvant therapy. Given the potential toxicity associated with chemotherapy, one could potentially consider abbreviating adjuvant chemotherapy in patients who attain pCR in both breast and axilla after NAT. However, these findings are hypothesis generating and further research is needed before it can be incorporated in clinical practice. For example, the ongoing DAPHNe study (NCT03716180) and the planned HER2Compass trial will evaluate omitting adjuvant cytotoxic chemotherapy for HER2+ patients who achieve a pCR after neoadjuvant paclitaxel, trastuzumab, and pertuzumab. Conversely, there are a number of studies exploring additional adjuvant therapies for patients with TNBC with residual disease following neoadjuvant chemotherapy, and the use of adjuvant capecitabine for such patients has become a favored approach based on the improved overall survival results observed in the CREATE-X trial (70). However, it must be recognized that the strongest data for use of pCR as a surrogate exists for neoadjuvant cytotoxic therapies and the anti-HER2 antibodies trastuzumab and pertuzumab. More research, such as that being done in I-SPY2, is needed to understand the prognostic significance of pCR following the use of neoadjuvant targeted therapies and immunotherapy agents.

The results for TNBC and HER2+ breast cancer demonstrating significant improvement in long-term outcomes with achievement of pCR supports clinical trials triaging novel therapies for further development based on pCR. For HER2+ breast cancer, neoadjuvant trials are now exploring regimens featuring HER2-directed agents only based on pCR as primary endpoint (71). In TNBC, the addition of a platinum agent to anthracyline/taxane-based treatment has been shown to increase pCR rates, but at the expense of greater toxicity (72–74). While trials studying the addition of neoadjuvant platinum among patients with TNBC have had divergent long-term outcomes, these trials were not powered for survival (75, 76).

Major strengths of the present study are its large size and the inclusion of both trial and cohort studies, representing a more realistic experience and providing external validity. In addition, results were highly significant despite the inclusion of a variety of neoadjuvant regimens, suggesting the path taken to attain a pCR may not be critical. Although trial-level analyses, which allow comparison of different treatments, have not validated pCR as a surrogate endpoint for improved long-term outcomes (4, 5), it is important to note that most neoadjuvant trials are powered for pCR and not for measures of long-term outcomes. Further research into this question is needed and will benefit from improved access to direct patient-level data in publications on this topic. Predictive data regarding pCR as a surrogate endpoint could be more thoroughly assessed if more publications included a breakdown of the long-term data by pCR status and treatment arm.

This meta-analysis has several potential limitations. First, the analysis is subject to variable reporting and study specific outcome definitions used across studies. For example, a variety of endpoints were used to represent breast cancer recurrence. Some of these endpoints include local recurrences, which are potentially curable, while other utilized endpoints focused on distant events only. Endpoints with local recurrence only were excluded. The definition of hormone receptor positivity also varied among some studies and over the years in which the studies were undertaken. The definition of pCR also varied at times between studies, and we included ypT0 ypN0 and ypT0/is ypN0 as allowable definitions. However, each study's definition for pCR and long-term outcomes were consistently used for both the pCR and the non-pCR groups, minimizing bias. Some meta-analyses on this topic have included studies that only considered pCR in the breast and/or allowed minimal residual disease in the breast, which we were careful to exclude given these definitions do not confer the same survival advantage (77). Second, there was heterogeneity in the type of neoadjuvant therapies employed and the study results are broadly based on NAT in general rather than a specific therapeutic regimen. Third, a number of analyses of interest, such as exploring the relationship between molecular breast cancer subtypes and pCR with corresponding long-term outcomes, could not be performed on the basis of the data available. Among HR+ tumors, pCR rates are higher and the relationship with long-term outcomes is stronger among grade 3 tumors compared with lower grade tumors (4). A pooled analysis by the German Breast Group based on 6,377 patients receiving neoadjuvant anthracycline-taxane–based chemotherapy in seven randomized trials suggested pCR is a suitable surrogate endpoint of recurrence for patients with luminal B/HER2-negative, HER2-positive (nonluminal), and TN disease, but not for those with luminal B/HER2-positive or luminal A tumors (77). While luminal A versus B classification could not be evaluated in this study, our results support these findings, with the greatest absolute benefit of pCR being observed in HER2+ and TN tumors. However, a trend toward significance for HR+ tumors was observed, likely driven by higher grade and/or luminal B subtypes, where the recurrence risk tends to be earlier while late recurrences are more often seen with luminal A tumors. Fourth, for HR+ breast cancer, an alternative surrogate endpoint, such as the residual cancer burden (RCB) index, may be more appropriate as pCR rates are low, but was not evaluated in this study to maintain homogeneity in assessment of the primary endpoint (pCR in breast and nodes; ref. 78). Finally, the median follow-up time for the study overall was only 4 years, which is short for the natural history of certain subtypes of breast cancer (HR+), but one would have expected the bias to be toward the null if recurrence events did not lead to mortality in a shorter time frame.

In conclusion, the study results comprehensively demonstrate that pCR after neoadjuvant chemotherapy is associated with significantly better EFS and overall survival. This study highlights the impact of adjuvant therapy in modulating relationship between pCR and outcomes and provides guidance for clinical trials evaluating neoadjuvant therapies for patients with localized breast cancer.

L.M. Spring is a paid consultant for Novartis, Puma, and Lumicell, and reports receiving other commercial research support from Merck and Tesaro. B. M. Alexander is an employee of Foundation Medicine and holds ownership interest (including patents) in Roche. A. Bardia is a paid consultant for Immunomedics, Novartis, Radius Health, Pfizer, Genentech, Sanofi, Merck, and Daichii/AstraZeneca. No potential conflicts of interest were disclosed by the other authors.

Conception and design: L.M. Spring, R.A. Greenup, K.L. Reynolds, A. Bardia

Development of methodology: L.M. Spring, G. Fell, A. Arfe, L. Trippa, A. Bardia

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): L.M. Spring, G. Fell, A. Arfe, R.A. Greenup, K.L. Reynolds, B.L. Smith, B. Moy, A. Bardia

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): L.M. Spring, G. Fell, A. Arfe, B. Moy, S.J. Isakoff, G. Parmigiani, L. Trippa, A. Bardia

Writing, review, and/or revision of the manuscript: L.M. Spring, G. Fell, A. Arfe, C. Sharma, R.A. Greenup, K.L. Reynolds, B.L. Smith, B.M. Alexander, B. Moy, S.J. Isakoff, G. Parmigiani, L. Trippa, A. Bardia

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): L.M. Spring, G. Fell, C. Sharma, B.M. Alexander, A. Bardia

Study supervision: B.M. Alexander, A. Bardia

The study itself had no specific funding. We acknowledge support for individual investigators from NCI grant KL2 TR001100 (to L. Spring) and grant K12 CA087723 (to A. Bardia) and a grant from Susan G Komen (CCR15224703, to A. Bardia).

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.
Mauri
D
,
Pavlidis
N
,
Ioannidis
JPA
. 
Neoadjuvant versus adjuvant systemic treatment in breast cancer: a meta-analysis
.
J Natl Cancer Inst
2005
;
97
:
188
94
.
2.
Rastogi
P
,
Anderson
SJ
,
Bear
HD
,
Geyer
CE
,
Kahlenberg
MS
,
Robidoux
A
, et al
Preoperative chemotherapy: updates of national surgical adjuvant breast and bowel project protocols B-18 and B-27
.
J Clin Oncol
2008
;
26
:
778
85
.
3.
Bardia
A
,
Baselga
J
. 
Neoadjuvant therapy as a platform for drug development and approval in breast cancer
.
Clin Cancer Res
2013
;
19
:
6360
70
.
4.
Cortazar
P
,
Zhang
L
,
Untch
M
,
Mehta
K
,
Costantino
JP
,
Wolmark
N
, et al
Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis
.
Lancet North Am Ed
2014
;
384
:
164
72
.
5.
Berruti
A
,
Amoroso
V
,
Gallo
F
,
Bertaglia
V
,
Simoncini
E
,
Pedersini
R
, et al
Pathologic complete response as a potential surrogate for the clinical outcome in patients with breast cancer after neoadjuvant therapy: a meta-regression of 29 randomized prospective studies
.
J Clin Oncol
2014
;
32
:
3883
91
.
6.
Broglio
KR
,
Quintana
M
,
Foster
M
,
Olinger
M
,
McGlothlin
A
,
Berry
SM
, et al
Association of pathologic complete response to neoadjuvant therapy in HER2-positive breast cancer with long-term outcomes: a meta-analysis
.
JAMA Oncol
2016
;
2
:
751
60
.
7.
Moher
D
,
Liberati
A
,
Tetzlaff
J
,
Altman
DG
,
PRISMA Group
. 
Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement
.
Int J Surg
2010
;
8
:
336
41
.
8.
DigitizeIt - Plot Digitizer Software
. 
Digitize graphs, charts and math data
.
Available from:
https://www.digitizeit.de/.
9.
Guyot
P
,
Ades
A
,
Ouwens
MJ
,
Welton
NJ
. 
Enhanced secondary analysis of survival data: reconstructing the data from published Kaplan-Meier survival curves
.
BMC Med Res Method
2012
;
12
:
9
.
10.
US Department of Health and Human Services Food and Drug Administration
,
Center for Drug Evaluation and Research (CDER)
. 
Guidance for industry: pathological complete response in neoadjuvant treatment of high-risk early-stage breast cancer: use as an endpoint to support accelerated approval
.
Available from:
http://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/ucm305501.pdf.
11.
Penault-Llorca
F
,
Abrial
C
,
Raoelfils
I
,
Cayre
A
,
Mouret-Reynier
M-A
,
Leheurteur
M
, et al
Comparison of the prognostic significance of Chevallier and Sataloff's pathologic classifications after neoadjuvant chemotherapy of operable breast cancer
.
Hum Pathol
2008
;
39
:
1221
8
.
12.
Crowther
MJ
,
Riley
RD
,
Staessen
JA
,
Wang
J
,
Gueyffier
F
,
Lambert
PC
. 
Individual patient data meta-analysis of survival data using Poisson regression models
.
BMC Med Res Methodol
2012
;
12
:
34
.
13.
Kuerer
HM
,
Newman
LA
,
Smith
TL
,
Ames
FC
,
Hunt
KK
,
Dhingra
K
, et al
Clinical course of breast cancer patients with complete pathologic primary tumor and axillary lymph node response to doxorubicin-based neoadjuvant chemotherapy
.
J Clin Oncol
1999
;
17
:
460
9
.
14.
Chollet
P
,
Amat
S
,
Cure
H
,
de Latour
M
,
Le Bouedec
G
,
Mouret-Reynier
M-A
, et al
Prognostic significance of a complete pathological response after induction chemotherapy in operable breast cancer
.
Br J Cancer
2002
;
86
:
1041
6
.
15.
Diéras
V
,
Fumoleau
P
,
Romieu
G
,
Tubiana-Hulin
M
,
Namer
M
,
Mauriac
L
, et al
Randomized parallel study of doxorubicin plus paclitaxel and doxorubicin plus cyclophosphamide as neoadjuvant treatment of patients with breast cancer
.
J Clin Oncol
2004
;
22
:
4958
65
.
16.
Lee
YJ
,
Doliny
P
,
Gomez-Fernandez
C
,
Powell
J
,
Reis
I
,
Hurley
J
. 
Docetaxel and cisplatin as primary chemotherapy for treatment of locally advanced breast cancers
.
Clin Breast Cancer
2004
;
5
:
371
6
.
17.
Ring
AE
,
Smith
IE
,
Ashley
S
,
Fulford
LG
,
Lakhani
SR
. 
Oestrogen receptor status, pathological complete response and prognosis in patients receiving neoadjuvant chemotherapy for early breast cancer
.
Br J Cancer
2004
;
91
:
2012
7
.
18.
Abrial
SC
,
Penault-Llorca
F
,
Delva
R
,
Bougnoux
P
,
Leduc
B
,
Mouret-Reynier
M-A
, et al
High prognostic significance of residual disease after neoadjuvant chemotherapy: a retrospective study in 710 patients with operable breast cancer
.
Breast Cancer Res Treat
2005
;
94
:
255
63
.
19.
Guarneri
V
,
Broglio
K
,
Kau
S-W
,
Cristofanilli
M
,
Buzdar
AU
,
Valero
V
, et al
Prognostic value of pathologic complete response after primary chemotherapy in relation to hormone receptor status and other factors
.
J Clin Oncol
2006
;
24
:
1037
44
.
20.
Hurley
J
,
Doliny
P
,
Reis
I
,
Silva
O
,
Gomez-Fernandez
C
,
Velez
P
, et al
Docetaxel, cisplatin, and trastuzumab as primary systemic therapy for human epidermal growth factor receptor 2–positive locally advanced breast cancer
.
J Clin Oncol
2006
;
24
:
1831
8
.
21.
Andre
F
,
Mazouni
C
,
Liedtke
C
,
Kau
S-W
,
Frye
D
,
Green
M
, et al
HER2 expression and efficacy of preoperative paclitaxel/FAC chemotherapy in breast cancer
.
Breast Cancer Res Treat
2007
;
108
:
183
90
.
22.
Liedtke
C
,
Mazouni
C
,
Hess
KR
,
André
F
,
Tordai
A
,
Mejia
JA
, et al
Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer
.
J Clin Oncol
2008
;
26
:
1275
81
.
23.
Eralp
Y
,
Smith
TL
,
Altundağ
K
,
Kau
S-W
,
Litton
J
,
Valero
V
, et al
Clinical features associated with a favorable outcome following neoadjuvant chemotherapy in women with localized breast cancer aged 35 years or younger
.
J Cancer Res Clin Oncol
2008
;
135
:
141
8
.
24.
Frasci
G
,
Comella
P
,
Rinaldo
M
,
Iodice
G
,
Di Bonito
M
,
D'Aiuto
M
, et al
Preoperative weekly cisplatin–epirubicin–paclitaxel with G-CSF support in triple-negative large operable breast cancer
.
Ann Oncol
2009
;
20
:
1185
92
.
25.
Al-Tweigeri
TA
,
Ajarim
DS
,
Alsayed
AA
,
Rahal
MM
,
Alshabanah
MO
,
Tulbah
AM
, et al
Prospective phase II study of neoadjuvant doxorubicin followed by cisplatin/docetaxel in locally advanced breast cancer
.
Med Oncol
2009
;
27
:
571
7
.
26.
Chang
HR
,
Glaspy
J
,
Allison
MA
,
Kass
FC
,
Elashoff
R
,
Chung
DU
, et al
Differential response of triple-negative breast cancer to a docetaxel and carboplatin-based neoadjuvant treatment
.
Cancer
2010
;
116
:
4227
37
.
27.
Chen
XS
,
Wu
JY
,
Huang
O
,
Chen
CM
,
Wu
J
,
Lu
JS
, et al
Molecular subtype can predict the response and outcome of Chinese locally advanced breast cancer patients treated with preoperative therapy
.
Oncol Rep
2010
;
23
:
1213
20
.
28.
Jinno
H
,
Sakata
M
,
Hayashida
T
,
Takahashi
M
,
Mukai
M
,
Ikeda
T
, et al
A phase II trial of capecitabine and docetaxel followed by 5-fluorouracil/epirubicin/cyclophosphamide (FEC) as preoperative treatment in women with stage II/III breast cancer
.
Ann Oncol
2010
;
21
:
1262
6
.
29.
Kim
SI
,
Sohn
J
,
Koo
JS
,
Park
SH
,
Park
HS
,
Park
BW
. 
Molecular subtypes and tumor response to neoadjuvant chemotherapy in patients with locally advanced breast cancer
.
Oncology
2010
;
79
:
324
30
.
30.
Arun
B
,
Bayraktar
S
,
Liu
DD
,
Gutierrez Barrera
AM
,
Atchley
D
,
Pusztai
L
, et al
Response to neoadjuvant systemic therapy for breast cancer in BRCA mutation carriers and noncarriers: a single-institution experience
.
J Clin Oncol
2011
;
29
:
3739
46
.
31.
Wu
J
,
Li
S
,
Jia
W
,
Su
F
. 
Response and prognosis of taxanes and anthracyclines neoadjuvant chemotherapy in patients with triple-negative breast cancer
.
J Cancer Res Clin Oncol
2011
;
137
:
1505
10
.
32.
Fasching
PA
,
Heusinger
K
,
Haeberle
L
,
Niklos
M
,
Hein
A
,
Bayer
CM
, et al
Ki67, chemotherapy response, and prognosis in breast cancer patients receiving neoadjuvant treatment
.
BMC Cancer
2011
;
11
:
486
.
33.
Masuda
H
,
Masuda
N
,
Kodama
Y
,
Ogawa
M
,
Karita
M
,
Yamamura
J
, et al
Predictive factors for the effectiveness of neoadjuvant chemotherapy and prognosis in triple-negative breast cancer patients
.
Cancer Chemother Pharmacol
2010
;
67
:
911
7
.
34.
Esserman
LJ
,
Berry
DA
,
DeMichele
A
,
Carey
L
,
Davis
SE
,
Buxton
M
, et al
Pathologic complete response predicts recurrence-free survival more effectively by cancer subset: results from the I-SPY 1 TRIAL—CALGB 150007/150012, ACRIN 6657
.
J Clin Oncol
2012
;
30
:
3242
9
.
35.
Im
S-A
,
Lee
KS
,
Ro
J
,
Lee
ES
,
Kwon
Y
,
Ahn
J-H
, et al
Phase II trial of preoperative paclitaxel, gemcitabine, and trastuzumab combination therapy in HER2 positive stage II/III breast cancer: the Korean Cancer Study Group BR 07-01
.
Breast Cancer Res Treat
2012
;
132
:
589
600
.
36.
Yoo
C
,
Ahn
J-H
,
Jung
KH
,
Kim
S-B
,
Kim
H-H
,
Shin
HJ
, et al
Impact of immunohistochemistry-based molecular subtype on chemosensitivity and survival in patients with breast cancer following neoadjuvant chemotherapy
.
J Breast Cancer
2012
;
15
:
203
10
.
37.
Melichar
B
,
Hornychová
H
,
Kalábová
H
,
Bašová
H
,
Mergancová
J
,
Urminská
H
, et al
Increased efficacy of a dose-dense regimen of neoadjuvant chemotherapy in breast carcinoma: a retrospective analysis
.
Med Oncol
2012
;
29
:
2577
85
.
38.
Zhang
G-C
,
Qian
X-K
,
Guo
Z-B
,
Ren
C-Y
,
Yao
M
,
Li
X-R
, et al
Pre-treatment hormonal receptor status and Ki67 index predict pathologic complete response to neoadjuvant trastuzumab/taxanes but not disease-free survival in HER2-positive breast cancer patients
.
Med Oncol
2012
;
29
:
3222
31
.
39.
Marmé
F
,
Aigner
J
,
Lorenzo Bermejo
J
,
Sinn
P
,
Sohn
C
,
Jäger
D
, et al
Neoadjuvant epirubicin, gemcitabine and docetaxel for primary breast cancer: Long-term survival data and major prognostic factors based on two consecutive neoadjuvant phase I/II trials
.
Int J Cancer
2013
;
133
:
1006
15
.
40.
Krishnan
Y
,
Alawadhi
SA
,
Sreedharan
PS
,
Gopal
M
,
Thuruthel
S
. 
Pathological responses and long-term outcome analysis after neoadjuvant chemotheraphy in breast cancer patients from Kuwait over a period of 15 years
.
Ann Saudi Med
2013
;
33
:
443
50
.
41.
Guarneri
V
,
Dieci
MV
,
Barbieri
E
,
Piacentini
F
,
Omarini
C
,
Ficarra
G
, et al
Loss of HER2 positivity and prognosis after neoadjuvant therapy in HER2-positive breast cancer patients
.
Ann Oncol
2013
;
24
:
2990
4
.
42.
Natoli
C
,
Vici
P
,
Sperduti
I
,
Grassadonia
A
,
Bisagni
G
,
Tinari
N
, et al
Effectiveness of neoadjuvant trastuzumab and chemotherapy in HER2-overexpressing breast cancer
.
J Cancer Res Clin Oncol
2013
;
139
:
1229
40
.
43.
Hurley
J
,
Reis
IM
,
Rodgers
SE
,
Gomez-Fernandez
C
,
Wright
J
,
Leone
JP
, et al
The use of neoadjuvant platinum-based chemotherapy in locally advanced breast cancer that is triple negative: retrospective analysis of 144 patients
.
Breast Cancer Res Treat
2013
;
138
:
783
94
.
44.
Guiu
S
,
Arnould
L
,
Bonnetain
F
,
Dalban
C
,
Favier
L
,
Desmoulins
I
, et al
Pathological response and survival after neoadjuvant therapy for breast cancer: a 30-year study
.
Breast
2013
;
22
:
301
8
.
45.
de Azambuja
E
,
Holmes
AP
,
Piccart-Gebhart
M
,
Holmes
E
,
Di Cosimo
S
,
Swaby
RF
, et al
Lapatinib with trastuzumab for HER2-positive early breast cancer (NeoALTTO): survival outcomes of a randomised, open-label, multicentre, phase 3 trial and their association with pathological complete response
.
Lancet Oncol
2014
;
15
:
1137
46
.
46.
Tanioka
M
,
Sasaki
M
,
Shimomura
A
,
Fujishima
M
,
Doi
M
,
Matsuura
K
, et al
Pathologic complete response after neoadjuvant chemotherapy in HER2-overexpressing breast cancer according to hormonal receptor status
.
Breast
2014
;
23
:
466
72
.
47.
Takada
M
,
Ishiguro
H
,
Nagai
S
,
Ohtani
S
,
Kawabata
H
,
Yanagita
Y
, et al
Survival of HER2-positive primary breast cancer patients treated by neoadjuvant chemotherapy plus trastuzumab: a multicenter retrospective observational study (JBCRG-C03 study)
.
Breast Cancer Res Treat
2014
;
145
:
143
53
.
48.
Kawajiri
H
,
Takashima
T
,
Aomatsu
N
,
Kashiwagi
S
,
Noda
S
,
Onoda
N
, et al
Prognostic significance of pathological complete response following neoadjuvant chemotherapy for operable breast cancer
.
Oncol Lett
2014
;
7
:
663
8
.
49.
Wang
J
,
Xu
B
,
Yuan
P
,
Li
Q
,
Zhang
P
,
Cai
R
, et al
HER2 as a predictive factor for successful neoadjuvant anthracycline chemotherapy of locally advanced and early breast cancer
.
Int J Biol Markers
2014
;
29
:
e187
92
.
50.
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
.
51.
Gonzalez-Angulo
AM
,
Parinyanitikul
N
,
Lei
X
,
Mittendorf
EA
,
Zhang
H
,
Valero
V
, et al
Effect of adjuvant trastuzumab among patients treated with anti-HER2-based neoadjuvant therapy
.
Br J Cancer
2015
;
112
:
630
5
.
52.
Zelnak
AB
,
Nikolinakos
P
,
Srinivasiah
J
,
Jonas
W
,
Pippas
A
,
Liu
Y
, et al
High pathologic complete response in Her2-positive, early-stage breast cancer to a novel nonanthracycline neoadjuvant chemotherapy
.
Clin Breast Cancer
2015
;
15
:
31
6
.
53.
Ko
ES
,
Han
H
,
Han
B-K
,
Kim
SM
,
Kim
RB
,
Lee
G-W
, et al
Prognostic significance of a complete response on breast MRI in patients who received neoadjuvant chemotherapy according to the molecular subtype
.
Korean J Radiol
2015
;
16
:
986
.
54.
Villarreal-Garza
C
,
Soto-Perez-de-Celis
E
,
Sifuentes
E
,
Ruano
S
,
Baez-Revueltas
B
,
Lara-Medina
F
, et al
Outcomes of Hispanic women with lymph-node positive, HER2 positive breast cancer treated with neoadjuvant chemotherapy and trastuzumab in Mexico
.
Breast
2015
;
24
:
218
23
.
55.
Groheux
D
,
Giacchetti
S
,
Delord
M
,
de Roquancourt
A
,
Merlet
P
,
Hamy
AS
, et al
Prognostic impact of 18F-FDG PET/CT staging and of pathological response to neoadjuvant chemotherapy in triple-negative breast cancer
.
Eur J Nucl Med Mol Imaging
2014
;
42
:
377
85
.
56.
Mayer
EL
,
Gropper
AB
,
Harris
L
,
Gold
JM
,
Parker
L
,
Kuter
I
, et al
Long-term follow-up after preoperative trastuzumab and chemotherapy for HER2-overexpressing breast cancer
.
Clin Breast Cancer
2015
;
15
:
24
30
.
57.
Zhang
P
,
Yin
Y
,
Mo
H
,
Zhang
B
,
Wang
X
,
Li
Q
, et al
Better pathologic complete response and relapse-free survival after carboplatin plus paclitaxel compared with epirubicin plus paclitaxel as neoadjuvant chemotherapy for locally advanced triple-negative breast cancer: a randomized phase 2 trial
.
Oncotarget
2016
;
7
:
60647
56
.
58.
Li
J
,
Chen
S
,
Chen
C
,
Di
G
,
Liu
G
,
Wu
J
, et al
Pathological complete response as a surrogate for relapse-free survival in patients with triple negative breast cancer after neoadjuvant chemotherapy
.
Oncotarget
2017
;
8
:
18399
408
.
59.
Shao
Z
,
Chaudhri
S
,
Guo
M
,
Zhang
L
,
Rea
D
. 
Neoadjuvant chemotherapy in triple negative breast cancer: an observational study
.
Oncol Res
2016
;
23
:
291
302
.
60.
AL-Tweigeri
T
,
AlSayed
A
,
Alawadi
S
,
Ibrahim
M
,
Ashour
W
,
Jaafar
H
, et al
A multicenter prospective phase II trial of neoadjuvant epirubicin, cyclophosphamide, and 5-fluorouracil (FEC100) followed by cisplatin–docetaxel with or without trastuzumab in locally advanced breast cancer
.
Cancer Chemother Pharmacol
2015
;
77
:
147
53
.
61.
Liu
S
,
Duan
X
,
Xu
L
,
Ye
J
,
Cheng
Y
,
Liu
Q
, et al
Nuclear Gli1 expression is associated with pathological complete response and event-free survival in HER2-positive breast cancer treated with trastuzumab-based neoadjuvant therapy
.
Tumor Biol
2015
;
37
:
4873
81
.
62.
Villarreal-Garza
C
,
Bargallo-Rocha
JE
,
Soto-Perez-de-Celis
E
,
Lasa-Gonsebatt
F
,
Arce-Salinas
C
,
Lara-Medina
F
, et al
Real-world outcomes in young women with breast cancer treated with neoadjuvant chemotherapy
.
Breast Cancer Res Treat
2016
;
157
:
385
94
.
63.
Mieog
JS
,
van der Hage
JA
,
van de Velde
CJ
. 
Neoadjuvant chemotherapy for operable breast cancer
.
Br J Surg
2007
;
94
:
1189
200
.
64.
Kong
X
,
Moran
MS
,
Zhang
N
,
Haffty
B
,
Yang
Q
. 
Meta-analysis confirms achieving pathological complete response after neoadjuvant chemotherapy predicts favourable prognosis for breast cancer patients
.
Eur J Cancer
2011
;
47
:
2084
90
.
65.
Prowell
TM
,
Pazdur
R
. 
Pathological complete response and accelerated drug approval in early breast cancer
.
N Engl J Med
2012
;
366
:
2438
41
.
66.
Berry
DA
,
Hudis
CA
. 
Neoadjuvant therapy in breast cancer as a basis for drug approval
.
JAMA Oncol
2015
;
1
:
875
6
.
67.
Barker
A
,
Sigman
C
,
Kelloff
G
,
Hylton
N
,
Berry
D
,
Esserman
L
. 
I-SPY 2: an adaptive breast cancer trial design in the setting of neoadjuvant chemotherapy
.
Clin Pharmacol Ther
2009
;
86
:
97
100
.
68.
Carey
LA
,
Winer
EP
. 
I-SPY 2 — toward more rapid progress in breast cancer treatment
.
N Engl J Med
2016
;
375
:
83
4
.
69.
Earl
H
,
Provenzano
E
,
Abraham
J
,
Dunn
J
,
Vallier
A-L
,
Gounaris
I
, et al
Neoadjuvant trials in early breast cancer: pathological response at surgery and correlation to longer term outcomes – what does it all mean?
BMC Med
2015
;
13
:
234
.
70.
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
.
71.
Hurvitz
SA
,
Martin
M
,
Symmans
WF
,
Jung
KH
,
Huang
C-S
,
Thompson
AM
, et al
Neoadjuvant trastuzumab, pertuzumab, and chemotherapy versus trastuzumab emtansine plus pertuzumab in patients with HER2-positive breast cancer (KRISTINE): a randomised, open-label, multicentre, phase 3 trial
.
Lancet Oncol
2018
;
19
:
115
26
.
72.
Sikov
WM
,
Berry
DA
,
Perou
CM
,
Singh
B
,
Cirrincione
CT
,
Tolaney
SM
, et al
Impact of the addition of carboplatin and/or bevacizumab to neoadjuvant once-per-week paclitaxel followed by dose-dense doxorubicin and cyclophosphamide on pathologic complete response rates in stage II to III triple-negative breast cancer: CALGB 40603 (Alliance)
.
J Clin Oncol
2015
;
33
:
13
21
.
73.
von Minckwitz
G
,
Schneeweiss
A
,
Loibl
S
,
Salat
C
,
Denkert
C
,
Rezai
M
, et al
Neoadjuvant carboplatin in patients with triple-negative and HER2-positive early breast cancer (GeparSixto; GBG 66): a randomised phase 2 trial
.
Lancet Oncol
2014
;
15
:
747
56
.
74.
Loibl
S
,
O'Shaughnessy
J
,
Untch
M
,
Sikov
WM
,
Rugo
HS
,
McKee
MD
, et al
Addition of the PARP inhibitor veliparib plus carboplatin or carboplatin alone to standard neoadjuvant chemotherapy in triple-negative breast cancer (BrighTNess): a randomised, phase 3 trial
.
Lancet Oncol
2018
;
19
:
497
509
.
75.
Sikov
WM
,
Berry
DA
,
Perou
CM
,
Singh
B
,
Cirrincione
CT
,
Tolaney
SM
, et al
Abstract S2-05: event-free and overall survival following neoadjuvant weekly paclitaxel and dose-dense AC +/- carboplatin and/or bevacizumab in triple-negative breast cancer: outcomes from CALGB 40603 (Alliance)
.
Cancer Res
2016
;
76
(
4_suppl
):
S2-05
.
76.
von Minckwitz
G
,
Loibl
S
,
Schneeweiss
A
,
Salat
CT
,
Rezai
M
,
Zahm
D-M
, et al
Abstract S2-04: early survival analysis of the randomized phase II trial investigating the addition of carboplatin to neoadjuvant therapy for triple-negative and HER2-positive early breast cancer (GeparSixto)
.
Cancer Res
2016
;
76)
(
4_suppl
):
S2-04
.
77.
von Minckwitz
G
,
Untch
M
,
Blohmer
J-U
,
Costa
SD
,
Eidtmann
H
,
Fasching
PA
, et al
Definition and impact of pathologic complete response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes
.
J Clin Oncol
2012
;
30
:
1796
804
.
78.
Symmans
WF
,
Wei
C
,
Gould
R
,
Yu
X
,
Zhang
Y
,
Liu
M
, et al
Long-term prognostic risk after neoadjuvant chemotherapy associated with residual cancer burden and breast cancer subtype
.
J Clin Oncol
2017
;
35
:
1049
60
.

Supplementary data