This article reviews published clinical and investigational studies related to the neoadjuvant management of breast cancer and defines a comprehensive clinical and research position involving all forms of neoadjuvant therapies.
Keywords: Surgery, Chemotherapy, HER therapy, Hormonal therapy, Cost analysis
Abstract
Background.
Neoadjuvant treatment is increasingly one of the preferred therapeutic options for early breast cancer and may have some unique outcomes, such as identifying predictive and prognostic factors of response or increasing the knowledge of individual tumor biology.
Design.
A panel of experts from different specialties reviewed published clinical studies on the neoadjuvant management of breast cancer. Recommendations were made that emphasized the clinical multidisciplinary management and the investigational leverage in early breast cancer.
Results.
Neoadjuvant therapy has equivalent efficacy to adjuvant therapy, and it has some additional benefits that include increasing breast conservation, assessing tumor response, establishing prognosis based on the pathological response, and providing a “second opportunity” for nonresponding patients. Achieving pathological complete remission because of neoadjuvant therapy has been correlated with long‐term clinical benefit, particularly in HER2‐positive and triple‐negative breast cancer. In addition, the neoadjuvant setting is a powerful model for the development of new drugs and the identification of prognostic markers. Finally, neoadjuvant therapy has proven to be cost‐effective by reducing nondrug costs, avoiding radical surgery, and reducing hospital stays when compared with other treatment approaches.
Conclusion.
Neoadjuvant therapy has clinical benefits in early breast cancer and provides in vivo information of individual breast cancer biology while allowing the investigation of new treatment approaches. Access to neoadjuvant therapy should be an option available to all patients with breast cancer through multidisciplinary tumor management.
Implications for Practice.
Neoadjuvant treatment should be strongly considered as a therapeutic option for localized breast cancer and is a powerful tool for understanding breast cancer biology and investigating new treatment approaches.
摘要
背景。新辅助治疗越来越成为早期乳腺癌的治疗首选方案之一,并且可能具有一些独特的结果,例如识别缓解的预测和预后因素或增加个体肿瘤生物学的认识。
设计。来自不同专业的专家小组审查了关于乳腺癌新辅助治疗的已发表的临床研究。提出了在早期乳腺癌中强调临床多学科管理和研究的最大限度利用的建议。
结果。新辅助治疗具有与辅助治疗相同的功效,并且还具有一些额外的获益,包括增加保乳手术,评估肿瘤缓解,基于病理缓解确定预后,以及为无缓解的患者提供“第二次机会”。由于新辅助治疗而实现病理性完全缓解与长期临床获益相关,特别是在 HER2 阳性和三阴性乳腺癌中。此外,新辅助治疗是开发新药和确定预后标志物的有力模型。最后,与其他治疗方法相比,新辅助治疗通过降低非药物成本,避免根治性手术和减少住院时间而被证明具有成本效益。
结论。新辅助治疗在早期乳腺癌中具有临床益处,并提供个体乳腺癌生物学的体内信息,同时允许研究新的治疗方法。通过多学科肿瘤管理,所有乳腺癌患者应该都可以选择接受新辅助治疗。
实践意义:应强烈考虑将新辅助治疗作为局部乳腺癌的治疗方案,新辅助治疗是了解乳腺癌生物学和研究新治疗方法的有力工具。
Introduction
In early breast cancer, neoadjuvant therapy has become one of the preferred treatment options [1], [2], [3], [4], [5]. Its efficacy has been proven to be equivalent to adjuvant therapy, in terms of both overall survival (OS) and progression‐free survival (PFS), and it has also shown value from research and cost perspectives [6], [7]. In most specialist hospitals, it is standard practice to assess patients with early breast cancer in multidisciplinary committees before local treatment delivery, and this has been shown to extend the efficacy of neoadjuvant care [8].
Systemic medical therapy reduces the risk of distant metastasis and increases OS, when initiated either after surgery (adjuvant therapy) or before surgery (neoadjuvant therapy), because often the same drugs and regimens are employed [9]. In general, any patient who is a candidate for adjuvant systemic therapy can be considered for neoadjuvant therapy [1]. Neoadjuvant therapy has some particular advantages that distinguish it from adjuvant treatment. First, it reduces tumor size, which facilitates surgical resectability and increases breast‐conserving surgery (BCS) rates [10], and it also can eliminate axillary node metastasis that may be detected in sentinel node biopsy after neoadjuvant treatment [11]. Second, it enables an objective evaluation of treatment efficacy, allowing an in vivo test of cancer cells’ sensitivity to treatment. If no response occurs, the therapy can be modified, thus avoiding side effects from ineffective treatment [12]. Pathological complete response (pCR) is associated with longer PFS and OS, especially in the case of triple‐negative and HER2‐positive tumors [13], [14], [15], [16], [17]. Third, it allows the design of clinical trials with novel research approaches for adjuvant treatment, because there is no standard treatment after failure to respond to neoadjuvant therapy. Patients with residual disease after neoadjuvant therapy could benefit from clinical trials involving new treatments in the adjuvant setting. Fourth, it provides information about tumor biology, enabling prognostic and predictive biomarkers to be sought. Obtaining tumor specimens and blood samples from the patient before, during, and after neoadjuvant treatment can reveal biomarkers that might prove crucial to research. Lastly, clinical trials involving neoadjuvant treatment offer advantages compared with the adjuvant setting. For example, they require smaller patient populations, results are obtained early, pathological response is a reliable endpoint that can be related to long‐term prognosis, and such trials lend themselves to innovative designs for the development of new drugs.
In this study, a panel of experts was convened by the Spanish Society of Medical Oncology (SEOM) in order to review published clinical and investigational studies related to the neoadjuvant management of breast cancer and define a comprehensive clinical and research position involving all forms of neoadjuvant therapies. Recommendations are made emphasizing the clinical multidisciplinary management and the investigational leverage of the neoadjuvant approach in early breast cancer.
Overall Management of Neoadjuvant Therapy
Selecting Cases for Neoadjuvant Therapy
Relevant clinical practice guidelines for localized breast cancer (National Comprehensive Cancer Network [NCCN] [18], European Society for Medical Oncology [ESMO] [19], or SEOM [2]) recommend using neoadjuvant therapy depending on such aspects as tumor size and molecular features, nodal involvement, patient performance status, and comorbidities. Full clinical information must be provided to patients, explaining the advantages and disadvantages of BCS and neoadjuvant treatments in a comprehensible manner.
There are several clinical goals for neoadjuvant therapy: first, achieving a pCR (which has prognostic utility in HER‐2 and triple‐negative subtypes); second, increasing breast preservation rate (applicable to all tumors); third, providing a “second opportunity” for those patients not achieving a pCR (particularly patients with estrogen‐receptor [ER]‐negative tumors) by introducing a non‐cross‐resistant adjuvant therapy [20].
Extent.
For patients with operable breast cancer, including those who are candidates for surgery at diagnosis, neoadjuvant therapy followed by surgery has become an alternative option to initial surgery. There is no one single definition of a tumor size or clinical characteristic that can establish whether the patient will benefit from neoadjuvant therapy, although some centers favor those cases in which a breast carcinoma is either larger than 2 cm or has clinically involved axillary nodes (Table 1). However, when assessing clinical advantages and treatment, if the surgical options and/or the long‐term outcomes are improved, then the indication for neoadjuvant therapy is clear. Tumor characteristics and axillary status at diagnosis and before neoadjuvant therapy and response after neoadjuvant therapy are important in surgical decision making [21].
Table 1. Patient selection criteria for neoadjuvant therapy in early breast cancer.
Histological and Molecular Features.
The breast cancer subtypes that benefit most from neoadjuvant therapy are those that will show the greatest treatment response, which are triple‐negative and HER2‐positive tumors (Table 1). Up to 45% of triple‐negative tumors show a pCR after neoadjuvant chemotherapy. This is important because triple‐negative tumors with a pCR have a similar prognosis to other subtypes that also display a pCR. In contrast, triple‐negative tumors with residual disease have a high probability of recurrence [22], [23].
For HER2‐positive tumors, several studies have demonstrated a high pCR rate after neoadjuvant therapy that includes anti‐HER2 treatment, which can be up to 60% pCR with the newest drug regimens and is higher in ER‐negative than in ER‐positive cases. An association is seen between pCR and increased PFS and OS in HER2‐positive tumors [24].
Regarding luminal‐like tumors, neoadjuvant chemotherapy achieves a lower rate of pCR in comparison with other subtypes, with a pCR rate of around 10%–24% [25]. It has been shown that in this ER‐positive population, high Ki67 expression increases the probability of a pCR [26], [27].
Genomic platforms may be useful in defining which cases of hormone‐sensitive early breast cancer may benefit of the addition of chemotherapy to hormonal therapy. A very recent publication shows that adjuvant chemotherapy is not indicated in patients with hormone receptor‐positive, lymph node‐negative cancer who are at intermediate Oncotype Dx risk [28]. In these cases, the use of neoadjuvant chemotherapy would likewise not be of benefit.
Clinical Status.
The patient's general state of health and comorbidities will drive the indications for treatment. Age by itself is not a contraindication for surgery.
Accurate clinical staging at baseline is critical because in certain clinical situations, some or all forms of neoadjuvant systemic therapy might not be the best clinical choice [18]. Tumors with extensive involvement of carcinoma in situ, for example, in which the extent of the invasive component is not well defined, would better be staged surgically.
Patient Information.
Neoadjuvant therapy as a treatment option must always be thoroughly discussed with patients, in order to consider benefits and risks and the impact of neoadjuvant therapy on surgical options. Patients’ opinions and preferences about preserving the breast and the management of the axilla after neoadjuvant therapy must be carefully considered and recorded.
Importance of Multidisciplinary Breast Cancer Committees in Neoadjuvant Therapy
The best management of breast cancer requires a multidisciplinary approach to the disease. Accordingly, many hospitals have set up multidisciplinary breast cancer committees, which discuss the different features of each case in order to decide the best treatment option. In Spanish hospitals, 74% of breast cancer cases are discussed in multidisciplinary committees at the time of diagnosis [29].
Because the development and operation of committees differ between hospitals, and to ensure fairness of patient access to neoadjuvant treatment, it is essential to understand the advantages and implement all recommendations about multidisciplinary management of neoadjuvant therapy.
Selecting Drugs for Neoadjuvant Therapy
Neoadjuvant medical treatment in breast cancer can include chemotherapy, targeted treatments such as anti‐HER2 therapy, and hormone therapy.
Chemotherapy.
The choice of chemotherapy regimen will depend on the patient's clinical status and tumor subtype. Therapies with proven efficacy include doxorubicin and cyclophosphamide (AC); epirubicin and cyclophosphamide (EC); fluorouracil, epirubicin, and cyclophosphamide (FEC) or fluorouracil, doxorubicin, and cyclophosphamide (FAC); AC followed by weekly paclitaxel (AC → P); AC followed by docetaxel (AC → D); taxane, doxorubicin, and cyclophosphamide (TAC); or carboplatin and taxane (CbT) in triple‐negative tumors. The preplanned number of treatment cycles must be completed whenever possible if tolerated, even if the tumor is significantly reduced or disappears.
Using weekly paclitaxel has shown efficacy and a good toxicity profile [30]. Adding carboplatin can be considered in patients with a mutation in the BRCA gene, although clinical studies are inconclusive on this point [31], [32]. Regimens containing EC or AC may be dose‐dense in the case of high‐grade and hormone receptor‐negative tumors [33], [34]. There seems to be insufficient evidence for routinely substituting paclitaxel for nab‐paclitaxel as a component of neoadjuvant chemotherapy, given the differing results of the ETNA and the GBG69 trials [35], [36], [37].
A recent review shows that information provided by genomic platforms may be useful in predicting the response to chemotherapy, although platforms might not be useful in indicating anti‐HER2 therapy [38].
Anti‐HER2 Therapy.
In HER2‐positive breast tumors, neoadjuvant trastuzumab used in combination with standard chemotherapy is capable of inducing a 30% pCR rate [25], [39], [40].
More recently, a dual HER2 antibody blockade using trastuzumab plus pertuzumab combined with chemotherapy achieved pCR rates in the range of 50% to 60% (NeoSphere and TRYPHAENA studies) [41], [42], [43]. This pCR rate was confirmed in the GeparSepto [44], KRISTINE [45], Symphony [46], and BERENICE [47] studies.
Before surgery, the most common chemotherapy combinations used are AC or FEC for 3–4 months followed by weekly paclitaxel for 12 weeks, AC or FEC for 3–4 months followed by docetaxel for 3–4 months, and docetaxel and carboplatin for 6 months. After surgery, anti‐HER2 therapy is continued for 12 months.
The overall results of various studies involving lapatinib, a HER2 tyrosine kinase inhibitor, in neoadjuvant therapy do not support its use in this indication [47], [48], [49], [50], [51], [52].
Using anthracyclines in combination with trastuzumab may increase the risk of cardiac toxicity. However, no increase was seen in the BERENICE study, in which pertuzumab was added to the standard treatment, even when administered simultaneously with anthracyclines [53].
Hormone Therapy.
Neoadjuvant hormone therapy is indicated to reduce the extent of the tumor and allow appropriate surgery in patients who are eligible for neoadjuvant treatment but not with chemotherapy, such as elderly patients. In postmenopausal women, an aromatase inhibitor (such as anastrozole, letrozole, or exemestane) is preferred over tamoxifen [54], [55], [56]. Currently, there is no evidence that one aromatase inhibitor is more effective than any other [57]. In premenopausal patients, there is also little evidence on this point. Some authors have suggested using surgery as the first option, even if BCS is not feasible [58], [59].
The minimum duration of treatment is 3 to 4 months, and this period can be extended until maximal response is achieved prior to surgery [60], [61]. Some clinical trials have compared the use of neoadjuvant hormone treatment with the use of neoadjuvant chemotherapy in postmenopausal women with ER‐positive breast cancer and have reported similar results between the two neoadjuvant approaches. A phase II study carried out in 239 patients with ER‐positive and/or PR‐positive tumors found no differences between aromatase inhibitors (anastrozole or exemestane) and four cycles of doxorubicin‐paclitaxel chemotherapy either in response rate (64.5% vs. 63.6%, respectively; p > .5) or in BCS (33% vs. 24%, respectively; p = .058) [62]. In a phase II study by the Spanish Breast Cancer Research Group (GEICAM) in luminal tumors, exemestane was compared with chemotherapy (four cycles of epirubicin‐cyclophosphamide followed by four cycles of docetaxel). The authors reported that response rates showed nonsignificant differences (48% vs. 66%, respectively; p = .075) and that there were no differences in BCS rates between the two treatment arms (56% vs. 47%, respectively; p = .23) [63].
A limitation of neoadjuvant endocrine therapy is that pCR is uncommon and, thus, not an effective surrogate of clinical outcome [64]. Several phase II studies have evaluated the effects of neoadjuvant cyclin‐dependent kinase inhibitors in combination with endocrine therapy or endocrine therapy plus anti‐HER2 therapies in ER‐positive/HER2‐positive breast carcinoma [65], [66], [67]. Early evaluation of proliferation markers shows an effect of adding palbociclib, although pCRs are still limited.
Surgical Issues Regarding Neoadjuvant Treatment
Mammography and breast and axillary ultrasound are the most common imaging studies performed to assess tumor size and axillary status. In addition, magnetic resonance imaging has been shown to improve accuracy in estimating post‐chemotherapy residual disease [68]. Baseline characteristics are important, and assessing what type of surgery is planned is crucial for the decision‐making process after neoadjuvant therapy [69]. Initial tumor size should not influence the type of surgery that the patient is eligible to undergo until the response is assessed. Breast cancer surgery rates have not increased in correlation with the higher pCR rates, and, as seen when analyzed in the NeoALTTO trial, improvements in this approach are needed [70]. By reducing the size of the primary tumor with neoadjuvant chemotherapy, there is potential for improving the cosmetic result of surgery, even for patients who were candidates for BCS at presentation. Neoadjuvant therapy also downstages axillary lymph node involvement in a significant proportion of patients—close to 40% when treatment includes an anthracycline plus a taxane, and in more than half of patients with HER2‐positive breast cancer who receive chemotherapy plus anti‐HER2 therapy. This is clinically important because the use of sentinel lymph node biopsy after neoadjuvant therapy has been increasing. In those patients with clinically negative nodes at diagnosis, sentinel lymph node biopsy has proven to be accurate and to have high identification rates; patients with negative results can be spared a full axillary lymph node dissection. In patients with clinically positive nodes before neoadjuvant therapy that convert to negative, several randomized trials have reported a false negative rate of <10% when a dual tracer is used and more than two sentinel lymph nodes are excised. False negative rates can be lowered with the use of targeted axillary dissection [71]. Whether patients who are downstaged to a negative axilla can be safely spared an axillary lymph node dissection is still controversial, and further research is needed.
A recent meta‐analysis of neoadjuvant chemotherapy from 10 EBCTG trials performed before 2005 confirmed an improvement in BCS rate over adjuvant chemotherapy, without compromising on distant recurrence, breast cancer survival, or OS [72]. The authors observed a higher local recurrence rate with breast‐conserving therapy over mastectomy, which is one part of breast‐conserving procedures [73].
Standardization of Pathological Studies
Neoadjuvant treatment has an important impact for pathologists, as the handling and reporting of breast cancer specimens in this setting require specific considerations. Multicenter breast cancer clinical trial studies have indicated that there is a huge variation in the handling and reporting of specimens [74]. The pathologist's task in assessing post‐neoadjuvant therapy specimens may be further complicated by the fact that the definition of pCR is not uniformly standardized, which can create further challenging issues with the interpretation and reporting of the data. The Residual Disease Characterization Working Group on behalf of the Breast International Group‐North American Breast Cancer Group Collaboration has published recommendations for the standardization of the pathological evaluation and reporting of post‐neoadjuvant specimens in clinical trials of breast cancer [75]. These recommendations include information about pCR as defined by the U.S. Food and Drug Administration (FDA). A group of Spanish pathologists with expertise in neoadjuvant therapy for breast cancer has recently reported a consensus on these important issues with the aim of standardizing the handling, analysis, and reporting of breast cancer specimens, both in the context of clinical trials and in daily practice [76].
Neoadjuvant Therapy as a Model for Advancing New Drug Research
The Value of pCR as an Endpoint for New Drug Development
After Cortazar et al. published a pooled analysis of neoadjuvant clinical trial results in 2014 that included data from almost 12,000 patients, the predictive value of achieving pCR at the time of surgery after standard neoadjuvant therapy was firmly established for the various breast cancer subtypes [14]. This analysis found that achieving a pCR was associated with longer recurrence‐free survival (RFS) and OS, especially in triple‐negative tumors treated with chemotherapy (RFS: hazard ratio [HR] 0.24, 95% confidence interval [CI] 0.18–0.33; OS: HR 0.16, 95% CI 0.11–0.25) and in HER2‐positive, hormone receptor‐negative tumors treated with trastuzumab plus chemotherapy (RFS: HR 0.15, 95% CI 0.09–0.27; OS: HR 0.08, 95% CI 0.03–0.22). For other tumor types, the same trend was seen, although it did not attain statistical significance.
Based on the association between pCR and long‐term outcomes for patients with breast cancer, the FDA published guidelines in 2014 proposing the use of pCR as a valid outcome measure for obtaining accelerated approval of new drugs in development [77], [78]. In that document, the FDA clearly stated its intention to facilitate the development of new drugs to treat breast cancer using the neoadjuvant model as a platform for testing new medicines that might improve patient survival, especially in the case of more aggressive tumors. The FDA's proposed model for achieving drug approval requires a randomized study, with the primary objective of demonstrating superiority of the study drug in terms of pCR, and a subsequent confirmatory trial demonstrating a clinically and statistically significant benefit in RFS, PFS, or OS for the test drug.
This proposal became a reality after the pCR results obtained in the NeoSphere and TRYPHAENA studies [41], [42], [43], with the FDA granting approval for pertuzumab plus trastuzumab in the neoadjuvant treatment of patients with HER2‐positive breast cancer.
The Neoadjuvant Setting in Research
Using the neoadjuvant model in research has brought about a paradigm shift in the approval of new drugs for treating breast cancer. Traditionally, drugs were developed to treat metastatic disease, and it took years for them to be used in early treatment of the disease. The current aim is to enable patients to benefit as soon as possible from more effective treatments without losing sight of the necessary balance between safety and efficacy, when the disease is curable [78]. Clinical practice provides examples of drugs being developed in parallel in the neoadjuvant and metastatic settings. These include cyclin inhibitors, phosphoinositide 3‐kinase (PI3K) inhibitors, and the tyrosine kinase inhibitor neratinib.
Investigational Advantages of the Neoadjuvant Model.
Using the neoadjuvant model in research has three advantages: first, clinically objective results are obtained in a few months; second, the potential for obtaining evidence of efficacy by treating fewer patients than required in studies in the adjuvant setting; and third, the possibility of conducting in vivo studies of biomarkers that can be used as prognostic factors or predictors of response to a standard neoadjuvant therapy or in studies of short duration (window studies).
New predictive biomarkers can be identified from a baseline biopsy. One such example might be the identification of mutations in GATA3 and suppression of cell proliferation with aromatase inhibitors. This has been done in two phase II studies by massive sequencing of samples obtained from patients treated with aromatase inhibitors [79].
Early biomarkers of response can also be identified from the biopsy of a cancer specimen once treatment has started. Ki67 and the preoperative endocrine prognostic index (PEPI) are examples. After 2 weeks of exposure to endocrine therapy for hormone receptor‐positive tumors, Ki67 expression showed a statistically significant association with lower RFS (p = .004). This was not the case with the baseline biopsy [80]. Another study tested the effect of everolimus on Ki67 levels when added to letrozole in the neoadjuvant setting. Everolimus was found to increase the efficacy of letrozole by reducing Ki67 levels compared with placebo [81]. The results of this small study heralded the benefit of combined exemestane and everolimus in metastatic disease, as demonstrated years later in the BOLERO‐2 phase III registry study [82].
Residual tissue obtained during surgery on patients who fail to achieve pCR can be used to identify biomarkers of resistance. This is a valuable information source for discovering mechanisms of resistance to the treatment used, by identifying compensatory alternative pathways in residual cells. For example, the use of NanoString technology in triple‐negative tumors identified the DUSP4 gene, a negative regulator of ERK, low expression of which was associated with greater proliferation, worse survival, and greater sensitivity to MEK inhibition [83]. More recently, Karagiannis et al. observed an increase of the mammalian‐enabled protein (MENA), which regulates actin structure and cell motility, in 20 cases of residual tumor after therapy [84]. These findings help identify mechanisms of resistance to neoadjuvant chemotherapy, for which experimental drugs may prove effective in breaking [85]. Because only cases without a pCR having residual malignant tissue can be evaluated after neoadjuvant therapy, markers of response to therapy cannot be evaluated at the molecular level.
Investigational Limitations of the Neoadjuvant Model.
Despite the great advantages of using the neoadjuvant model for research, limitations also exist. Although an increase in pCR obtained by a cancer drug generally translates into a benefit in the adjuvant or metastatic setting, there are some exceptions, bevacizumab being one such case. The GeparQuinto study demonstrated an increase in pCR in triple‐negative tumors treated with neoadjuvant bevacizumab [86]. However, this benefit has not been achieved in the adjuvant setting, as confirmed by the BEATRICE phase III trial [53], [87]. Therefore, study results obtained in a certain indication cannot always be extended to other indications within the same disease.
The risk‐benefit balance of neoadjuvant treatment must be carefully evaluated. Neoadjuvant therapy should be used in high‐risk populations such as patients with triple‐negative or HER2‐positive disease or with luminal tumors in the presence of an additional risk factor such as high‐grade or high Ki67 [3].
Developing early pharmacodynamic or metabolic markers of response, as was suggested several years ago, may help in addressing some of these limitations. In recent research, Ki67 showed a significant drop in the NeoPredict trial [88] that can be used to adapt the therapies, and FDG PET/CT identified patients with an increased likelihood of pCR in the neoadjuvant NeoALTTO trial [89].
Future Directions of Neoadjuvant Therapy Evaluation
Currently, economic considerations are generally not part of the management of early breast cancer, particularly because there are many situations in which the cost cannot be estimated. In some homogeneous subsets of cases, however, economic analysis may be reasonable. From 2000 to 2015, several studies were published evaluating the cost‐effectiveness of drugs, of which only three were in the neoadjuvant setting (37 drugs were in the metastatic setting, and 101 were in the adjuvant setting) [90]. A substantial number of these studies have focused on HER2‐positive breast cancer. Table 2 highlights the most relevant of these studies.
Table 2. Summary of the most important cost‐effectiveness analyses on targeted HER2 therapies.
Abbreviations: ALY, adjusted life‐year; ICER, incremental cost‐effectiveness ratio; LY, life‐year; MBC, metastatic breast cancer; NHS, U.K. National Health Service; NICE, U.K. National Institute for Health and Care Excellence; PBAC, Australian Pharmaceutical Benefits Advisory Committee; pCODR, Pan‐Canadian Oncology Drug Review; QALY, quality‐adjusted life‐year.
Source: Nixon et al., 2016 [94].
The three published studies of neoadjuvant therapy analyzed the addition of pertuzumab in HER2‐positive tumors from the results of the TRYPHAENA and the NeoSphere trials. The Canadian article of Attard et al. described a favorable cost per quality‐adjusted life‐years (QALYs) [91]. The Spanish cost‐offset study of Albanell et al. quantified the benefit in avoiding costs related to disease relapse [92]. An appraisal by NICE in the U.K. recommends the use of neoadjuvant pertuzumab as an option for the neoadjuvant treatment of HER2‐positive breast cancer [93].
Recommendations
After reviewing the existing scientific evidence, this group of breast cancer experts, on behalf of SEOM, reached the following conclusions regarding the use of neoadjuvant therapy in the treatment of breast cancer:
Neoadjuvant systemic therapy involving chemotherapy, targeted therapy, and sometimes hormonal therapy is a preferred therapeutic option in early breast cancer before definitive breast surgery and has the same indications as adjuvant treatment.
Multidisciplinary committees play an essential role in the appropriate selection of patients eligible for neoadjuvant therapy.
Concerning chemotherapy, the recommended treatment regimen consists of anthracyclines followed by taxanes. A dose‐dense anthracycline regimen may be used in patients with high‐grade or hormone receptor‐negative tumors.
In patients with triple‐negative tumors and mutated BRCA, it is reasonable to supplement the taxane with carboplatin.
In patients with HER2‐positive tumors, neoadjuvant administration of dual anti‐HER2 therapy (trastuzumab and pertuzumab) is indicated together with chemotherapy.
In hormone receptor‐positive postmenopausal patients not eligible for chemotherapy, neoadjuvant endocrine therapy is an appropriate option.
Neoadjuvant treatment can significantly impact surgical treatment by downstaging the tumor without compromising locoregional control. It facilitates BCS and limited axillary surgery in selected patients.
The neoadjuvant model offers great advantages for new drug development in breast cancer and for individual investigations into the mechanisms of action of drugs.
pCR can be used as an endpoint for early drug approval.
Neoadjuvant therapies allow the early identification of efficacy markers.
Given the clinical benefit demonstrated by neoadjuvant therapy, future clinical trials should include specific evaluations of economic cost and cost‐offset analysis.
Acknowledgments
The authors wish to thank Dr. Fernando Sánchez Barbero from HealthCo S.L. (Madrid, Spain) for his help in the editing of the first draft of the manuscript. The authors acknowledge the members of the Spanish Society of Medical Oncology (SEOM) Board for their collaboration in the preparation of the manuscript. This position paper was commissioned by SEOM. Roche Farma provided financial support for medical writing services.
Author Contributions
Conception/design: Ramon Colomer, Cristina Saura, Pedro Sánchez‐Rovira, Tomás Pascual
Provision of study material or patients: Ramon Colomer, Cristina Saura, Pedro Sánchez‐Rovira, Tomás Pascual
Collection and/or assembly of data: Ramon Colomer, Cristina Saura, Pedro Sánchez‐Rovira, Tomás Pascual
Data analysis and interpretation: Ramon Colomer, Cristina Saura, Pedro Sánchez‐Rovira, Tomás Pascual, Isabel T. Rubio, Octavio Burgués, Lourdes Marcos, César A. Rodríguez, Miguel Martín, Ana Lluch
Manuscript writing: Ramon Colomer, Cristina Saura, Pedro Sánchez‐Rovira, Tomás Pascual, Isabel T. Rubio, Octavio Burgués, Lourdes Marcos, César A. Rodríguez, Miguel Martín, Ana Lluch
Final approval of manuscript: Ramon Colomer, Cristina Saura, Pedro Sánchez‐Rovira, Tomás Pascual, Isabel T. Rubio, Octavio Burgués, Lourdes Marcos, César A. Rodríguez, Miguel Martín, Ana Lluch
Disclosures
Ramón Colomer: Lilly, Merck Sharp & Dohme, Roche, Servier (C/A), Bristol‐Myers Squibb, Merck Sharp & Dohme, Roche, Pfizer, AstraZeneca, Astellas (RF), Fundacion Instituto Roche, Sanofi, Pfizer (H); Cristina Saura: AstraZeneca, Celgene, Daiichi Sankyo, Eisai, Roche, Genomic Health, Novartis, Pfizer, Pierre Fabre, Puma and Synthon (C/A), AstraZeneca, Roche, Genentech, Macrogenics, Novartis, Pfizer, Piqur Therapeutics, Puma and Synthon (RF); Pedro Sánchez‐Rovira: Roche, Celgene, Pfizer (C/A), Bristol‐Myers Squibb (RF); Tomás Pascual: Roche (C/A); Isabel T. Rubio: Roche (H); César A. Rodríguez: Roche, Novartis, Lilly, Pfizer, AMGEN, Merck Sharp & Dohme (C/A); Miguel Martín: AstraZeneca, Novartis, Roche‐Genentech, Pfizer, Glaxo, Pharmamar, Taiho Oncology and Lilly (C/A), Novartis and Roche (RF), Pfizer and Lilly (H); Ana Lluch: Novartis, Pfizer, Roche/Genentech, Eisai, Celgene (C/A), Amgen, Astra Zeneca, Boehringer‐Ingelheim, GSK, Novartis, Pfizer, Roche/Genentech, Eisai, Celgene, Pierre Fabre (RF). The other authors indicated no financial relationships.
(C/A) Consulting/advisory relationship; (RF) Research funding; (E) Employment; (ET) Expert testimony; (H) Honoraria received; (OI) Ownership interests; (IP) Intellectual property rights/inventor/patent holder; (SAB) Scientific advisory board
References
- 1.Kaufmann M, von Minckwitz G, Mamounas EP et al. Recommendations from an international consensus conference on the current status and future of neoadjuvant systemic therapy in primary breast cancer. Ann Surg Oncol 2012;19:1508–1516. [DOI] [PubMed] [Google Scholar]
- 2.García‐Sáenz JA, Bermejo B, Estévez LG et al. SEOM clinical guidelines in early‐stage breast cancer 2015. Clin Transl Oncol 2015;17:939–945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Makhoul I, Kiwan E. Neoadjuvant systemic treatment of breast cancer. J Surg Oncol 2011;103:348–357. [DOI] [PubMed] [Google Scholar]
- 4.Mittendorf EA, Buchholz TA, Tucker SL et al. Impact of chemotherapy sequencing on local‐regional failure risk in breast cancer patients undergoing breast‐conserving therapy. Ann Surg 2013;257:173–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Smith BL. Neoadjuvant versus adjuvant systemic therapy for operable breast cancer: Equivalent outcomes? Ann Surg 2013;257:180–181. [DOI] [PubMed] [Google Scholar]
- 6.Mauri D, Pavlidis N, Ioannidis JP. Neoadjuvant versus adjuvant systemic treatment in breast cancer: A meta‐analysis. J Natl Cancer Inst 2005;97:188–194. [DOI] [PubMed] [Google Scholar]
- 7.Rastogi P, Anderson SJ, Bear HD et al. Preoperative chemotherapy: Updates of National Surgical Adjuvant Breast and Bowel Project Protocols B‐18 and B‐27. J Clin Oncol 2008;26:778–785. [DOI] [PubMed] [Google Scholar]
- 8.Simmons CE, Hogeveen S, Leonard R et al. A Canadian national expert consensus on neoadjuvant therapy for breast cancer: Linking practice to evidence and beyond. Curr Oncol 2015;22:S43–S53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Thompson AM, Moulder‐Thompson SL. Neoadjuvant treatment of breast cancer. Ann Oncol 2012;23(suppl 10):x231–x236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Killelea BK, Yang VQ, Mougalian S et al. Neoadjuvant chemotherapy for breast cancer increases the rate of breast conservation: Results from the National Cancer Database. J Am Coll Surg 2015;220:1063–1069. [DOI] [PubMed] [Google Scholar]
- 11.Diaz‐Botero S, Espinosa‐Bravo M, Goncalves VR et al. Different prognostic implications of residual disease after neoadjuvant treatment: Impact of Ki 67 and site of response. Ann Surg Oncol 2016;23:3831–3837. [DOI] [PubMed] [Google Scholar]
- 12.Untch M, von Minckwitz G. Neoadjuvant chemotherapy: Early response as a guide for further treatment: Clinical, radiological, and biological. J Natl Cancer Inst Monogr 2011;2011:138–141. [DOI] [PubMed] [Google Scholar]
- 13.Berruti A, Amoroso V, Gallo F 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;32:3883–3891. [DOI] [PubMed] [Google Scholar]
- 14.Cortazar P, Zhang L, Untch M et al. Pathological complete response and long‐term clinical benefit in breast cancer: The CTNeoBC pooled analysis. Lancet 2014;384:164–172. [DOI] [PubMed] [Google Scholar]
- 15.von Minckwitz G, Untch M, Blohmer JU et al. Definition and impact of pathologic complete response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes. J Clin Oncol 2012;30:1796–1804. [DOI] [PubMed] [Google Scholar]
- 16.Rouzier R, Perou CM, Symmans WF et al. Breast cancer molecular subtypes respond differently to preoperative chemotherapy. Clin Cancer Res 2005;11:5678–5685. [DOI] [PubMed] [Google Scholar]
- 17.Esserman LJ, Berry DA, DeMichele A 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;30:3242–3249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.NCCN Clinical Practice Guidelines in Oncology . Breast Cancer Version 2‐2017. Plymouth Meeting, PA: National Comprehensive Cancer Network; 2017. [Google Scholar]
- 19.Senkus E, Kyriakides S, Ohno S et al. Primary breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow‐up. Ann Oncol 2015;26(suppl 5):v8–v30. [DOI] [PubMed] [Google Scholar]
- 20.Masuda N, Lee SJ, Ohtani S et al. Adjuvant capecitabine for breast cancer after preoperative chemotherapy. N Engl J Med 2017;376:2147–2159. [DOI] [PubMed] [Google Scholar]
- 21.Rubio IT, Esgueva‐Colmenarejo A, Espinosa‐Bravo M et al. Intraoperative ultrasound‐guided lumpectomy versus mammographic wire localization for breast cancer patients after neoadjuvant treatment. Ann Surg Oncol 2016;23:38–43. [DOI] [PubMed] [Google Scholar]
- 22.Carey LA, Dees EC, Sawyer L et al. The triple negative paradox: Primary tumor chemosensitivity of breast cancer subtypes. Clin Cancer Res 2007;13:2329–2334. [DOI] [PubMed] [Google Scholar]
- 23.Liedtke C, Mazouni C, Hess KR et al. Response to neoadjuvant therapy and long‐term survival in patients with triple‐negative breast cancer. J Clin Oncol 2008;26:1275–1281. [DOI] [PubMed] [Google Scholar]
- 24.Untch M, Fasching PA, Konecny GE et al. Pathologic complete response after neoadjuvant chemotherapy plus trastuzumab predicts favorable survival in human epidermal growth factor receptor 2‐overexpressing breast cancer: Results from the TECHNO trial of the AGO and GBG study groups. J Clin Oncol 2011;29:3351–3357. [DOI] [PubMed] [Google Scholar]
- 25.Buzdar AU, Valero V, Theriault RL et al. Pathological complete response to chemotherapy is related to hormone receptor status. Breast Cancer Res Treat 2003;82(suppl 1):302A. [Google Scholar]
- 26.Denkert C, Loibl S, Müller BM et al. Ki67 levels as predictive and prognostic parameters in pretherapeutic breast cancer core biopsies: A translational investigation in the neoadjuvant GeparTrio trial. Ann Oncol 2013;24:2786–2793. [DOI] [PubMed] [Google Scholar]
- 27.Klauschen F, Wienert S, Schmitt WD et al. Standardized Ki67 diagnostics using automated scoring – Clinical validation in the GeparTrio breast cancer study. Clin Cancer Res 2015;21:3651–3657. [DOI] [PubMed] [Google Scholar]
- 28.Sparano JA, Gray RJ, Makower DF et al. Adjuvant chemotherapy guided by a 21‐gene expression assay in breast cancer. N Engl J Med 2018;379:111–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Merck B, Ramos‐Rincón JM, Cansado P et al. EUSOMA requirements for breast cancer units in Spain [in Spanish]. Cir Esp 2005;77:221–225. [DOI] [PubMed] [Google Scholar]
- 30.Green MC, Buzdar AU, Smith T et al. Weekly paclitaxel improves pathologic complete remission in operable breast cancer when compared with paclitaxel once every 3 weeks. J Clin Oncol 2005;23:5983–5992. [DOI] [PubMed] [Google Scholar]
- 31.Sharma P, López‐Tarruella S, García‐Sáenz JA et al. Efficacy of neoadjuvant carboplatin plus docetaxel in triple‐negative breast cancer: Combined analysis of two cohorts. Clin Cancer Res 2017;23:649–657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Tutt A, Tovey H, Cheang MCU et al. Carboplatin in BRCA1/2‐mutated and triple‐negative breast cancer BRCAness subgroups: The TNT Trial. Nat Med 2018;24:628–637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Citron ML, Berry DA, Cirrincione C et al. Randomized trial of dose‐dense versus conventionally scheduled and sequential versus concurrent combination chemotherapy as postoperative adjuvant treatment of node‐positive primary breast cancer: First report of Intergroup Trial C9741/Cancer and Leukemia Group B Trial 9741. J Clin Oncol 2003;21:1431–1439. [DOI] [PubMed] [Google Scholar]
- 34.Untch M, von Minckwitz G, Konecny GE et al. PREPARE trial: A randomized phase III trial comparing preoperative, dose‐dense, dose‐intensified chemotherapy with epirubicin, paclitaxel, and CMF versus a standard‐dosed epirubicin‐cyclophosphamide followed by paclitaxel with or without darbepoetin alfa in primary breast cancer–outcome on prognosis. Ann Oncol 2011;22:1999–2006. [DOI] [PubMed] [Google Scholar]
- 35.Gianni L, Mansutti M, Anton A et al. Comparing neoadjuvant nab‐paclitaxel vs paclitaxel both followed by anthracycline regimens in women with ERBB2/HER2‐negative breast cancer‐the Evaluating Treatment with Neoadjuvant Abraxane (ETNA) trial: A randomized phase 3 clinical trial. JAMA Oncol 2018;4:302–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Sparano JA. Neoadjuvant systemic therapy for breast cancer: Searching for more effectively curative therapies. JAMA Oncol 2018;4:293–295. [DOI] [PubMed] [Google Scholar]
- 37.Untch M, Jackisch C, Schneeweiss A et al. Nab‐paclitaxel versus solvent‐based paclitaxel in neoadjuvant chemotherapy for early breast cancer (GeparSepto‐GBG 69): A randomised, phase 3 trial. Lancet Oncol 2016;17:345–356. [DOI] [PubMed] [Google Scholar]
- 38.Kwa M, Makris A, Esteva FJ. Clinical utility of gene‐expression signatures in early stage breast cancer. Nat Rev Clin Oncol 2017;14:595–610. [DOI] [PubMed] [Google Scholar]
- 39.Petrelli F, Borgonovo K, Cabiddu M et al. Neoadjuvant chemotherapy and concomitant trastuzumab in breast cancer: A pooled analysis of two randomized trials. Anticancer Drugs 2011;22:128–135. [DOI] [PubMed] [Google Scholar]
- 40.Piccart‐Gebhart M, Holmes AP, de Azambuja E et al. The association between event‐free survival and pathological complete response to neoadjuvant lapatinib, trastuzumab or their combination in HER2‐positive breast cancer. Survival follow‐up analysis of the NeoALTTO study (BIG 1‐06). Cancer Res 2013;73:S1–S01. [Google Scholar]
- 41.Gianni L, Pienkowski T, Im YH et al. Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2‐positive breast cancer (NeoSphere): A randomised multicentre, open‐label, phase 2 trial. Lancet Oncol 2012;13:25–32. [DOI] [PubMed] [Google Scholar]
- 42.Gianni L, Pienkowski T, Im YH et al. 5‐year analysis of neoadjuvant pertuzumab and trastuzumab in patients with locally advanced, inflammatory, or early‐stage HER2‐positive breast cancer (NeoSphere): A multicentre, open‐label, phase 2 randomised trial. Lancet Oncol 2016;17:791–800. [DOI] [PubMed] [Google Scholar]
- 43.Schneeweiss A, Chia S, Hickish T et al. Pertuzumab plus trastuzumab in combination with standard neoadjuvant anthracycline‐containing and anthracycline‐free chemotherapy regimens in patients with HER2‐positive early breast cancer: A randomized phase II cardiac safety study (TRYPHAENA). Ann Oncol 2013;24:2278–2284. [DOI] [PubMed] [Google Scholar]
- 44.Loibl S, Jackisch C, Schneeweiss A et al. Dual HER2‐blockade with pertuzumab and trastuzumab in HER2‐positive early breast cancer: A subanalysis of data from the randomized phase III GeparSepto trial. Ann Oncol 2017;28:497–504. [DOI] [PubMed] [Google Scholar]
- 45.Hurvitz SA, Martin M, Symmans WF et al. Pathologic complete response (pCR) rates after neoadjuvant trastuzumab emtansine (T‐DM1 [K]) + pertuzumab (P) vs docetaxel + carboplatin + trastuzumab + P (TCHP) treatment in patients with HER2‐positive (HER2+) early breast cancer (EBC) (KRISTINE). J Clin Oncol 2016;34(suppl 15):500A. [Google Scholar]
- 46.Beitsch P, Whitworth P, Baron P et al. Pertuzumab/trastuzumab/CT versus trastuzumab/CT therapy for HER2+ breast cancer: Results from the prospective Neoadjuvant Breast Registry Symphony Trial (NBRST). Ann Surg Oncol 2017;24:2539–2546. [DOI] [PubMed] [Google Scholar]
- 47.Untch M, Loibl S, Bischoff J et al. Lapatinib versus trastuzumab in combination with neoadjuvant anthracycline‐taxane‐based chemotherapy (GeparQuinto, GBG 44): A randomised phase 3 trial. Lancet Oncol 2012;13:135–144. [DOI] [PubMed] [Google Scholar]
- 48.Robidoux A, Tang G, Rastogi P et al. Lapatinib as a component of neoadjuvant therapy for HER2‐positive operable breast cancer (NSABP protocol B‐41): An open‐label, randomised phase 3 trial. Lancet Oncol 2013;14:1183–1192. [DOI] [PubMed] [Google Scholar]
- 49.Alba E, Albanell J, de la Haba J et al. Trastuzumab or lapatinib with standard chemotherapy for HER2‐positive breast cancer: Results from the GEICAM/2006‐14 trial. Br J Cancer 2014;110:1139–1147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Bonnefoi H, Jacot W, Saghatchian M et al. Neoadjuvant treatment with docetaxel plus lapatinib, trastuzumab, or both followed by an anthracycline‐based chemotherapy in HER2‐positive breast cancer: Results of the randomised phase II EORTC 10054 study. Ann Oncol 2015;26:325–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Nagayama A, Hayashida T, Jinno H et al. Comparative effectiveness of neoadjuvant therapy for HER2‐positive breast cancer: A network meta‐analysis. J Natl Cancer Inst 2014;106:dju203. [DOI] [PubMed] [Google Scholar]
- 52.de Azambuja E, Holmes AP, Piccart‐Gebhart M 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;15:1137–1146. [DOI] [PubMed] [Google Scholar]
- 53.Swain SM, Ewer MS, Viale G et al. Primary analysis of BERENICE: A phase II cardiac safety study of pertuzumab, trastuzumab, and neoadjuvant anthracycline‐based chemotherapy in patients with locally advanced, inflammatory, or early‐stage, unilateral, and invasive HER2‐positive breast cancer. Presented at: San Antonio Breast Cancer Symposium; December 2–6, 2016; San Antonio, TX; abstract P4‐21‐41.
- 54.Ellis MJ, Ma C. Letrozole in the neoadjuvant setting: The P024 trial. Breast Cancer Res Treat 2007;105:33–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Eiermann W, Paepke S, Appfelstaedt J et al. Preoperative treatment of postmenopausal breast cancer patients with letrozole: A randomized double‐blind multicenter study. Ann Oncol 2001;12:1527–1532. [DOI] [PubMed] [Google Scholar]
- 56.Smith IE, Dowsett M, Ebbs SR et al. Neoadjuvant treatment of postmenopausal breast cancer with anastrozole, tamoxifen, or both in combination: The Immediate Preoperative Anastrozole, Tamoxifen, or Combined with Tamoxifen (IMPACT) multicenter double‐blind randomized trial. J Clin Oncol 2005;23:5108–5116. [DOI] [PubMed] [Google Scholar]
- 57.Cataliotti L, Buzdar AU, Noguchi S et al. Comparison of anastrozole versus tamoxifen as preoperative therapy in postmenopausal women with hormone receptor‐positive breast cancer: The Pre‐Operative “Arimidex” Compared to Tamoxifen (PROACT) trial. Cancer 2006;106:2095–2103. [DOI] [PubMed] [Google Scholar]
- 58.Ellis MJ, Suman VJ, Hoog J et al. Randomized phase II neoadjuvant comparison between letrozole, anastrozole, and exemestane for postmenopausal women with estrogen receptor‐rich stage 2 to 3 breast cancer: Clinical and biomarker outcomes and predictive value of the baseline PAM50‐based intrinsic subtype–ACOSOG Z1031. J Clin Oncol 2011;29:2342–2349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Torrisi R, Bagnardi V, Pruneri G et al. Antitumour and biological effects of letrozole and GnRH analogue as primary therapy in premenopausal women with ER and PgR positive locally advanced operable breast cancer. Br J Cancer 2007;97:802–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Krainick‐Strobel UE, Lichtenegger W, Wallwiener D et al. Neoadjuvant letrozole in postmenopausal estrogen and/or progesterone receptor positive breast cancer: A phase IIb/III trial to investigate optimal duration of preoperative endocrine therapy. BMC Cancer 2008;8:62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Fontein DB, Charehbili A, Nortier JW et al. Efficacy of six month neoadjuvant endocrine therapy in postmenopausal, hormone receptor‐positive breast cancer patients–a phase II trial. Eur J Cancer 2014;50:2190–2200. [DOI] [PubMed] [Google Scholar]
- 62.Semiglazov VF, Semiglazov VV, Dashyan GA et al. Phase 2 randomized trial of primary endocrine therapy versus chemotherapy in postmenopausal patients with estrogen receptor‐positive breast cancer. Cancer 2007;110:244–254. [DOI] [PubMed] [Google Scholar]
- 63.Alba E, Calvo L, Albanell J et al. Chemotherapy (CT) and hormonotherapy (HT) as neoadjuvant treatment in luminal breast cancer patients: Results from the GEICAM/2006‐03, a multicenter, randomized, phase‐II study. Ann Oncol 2012;23:3069–3074. [DOI] [PubMed] [Google Scholar]
- 64.Guerrero‐Zotano AL, Arteaga CL. Neoadjuvant trials in ER(+) breast cancer: A tool for acceleration of drug development and discovery. Cancer Discov 2017;7:561–574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Chow LWC, Morita S, Chow CYC et al. Neoadjuvant palbociclib on ER+ breast cancer (N007): Clinical response and EndoPredict's value. Endocr Relat Cancer 2018;25:123–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Ma CX, Gao F, Luo J et al. NeoPalAna: Neoadjuvant palbociclib, a cyclin‐dependent kinase 4/6 Inhibitor, and anastrozole for clinical stage 2 or 3 estrogen receptor‐positive breast cancer. Clin Cancer Res 2017;23:4055–4065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Gianni L, Bisagni G, Colleoni M et al. Neoadjuvant treatment with trastuzumab and pertuzumab plus palbociclib and fulvestrant in HER2‐positive, ER‐positive breast cancer (NA‐PHER2): An exploratory, open‐label, phase 2 study. Lancet Oncol 2018;19:249–256.29326029 [Google Scholar]
- 68.Hylton NM, Blume JD, Bernreuter WK et al. Locally advanced breast cancer: MR imaging for prediction of response to neoadjuvant chemotherapy–results from ACRIN 6657/I‐SPY TRIAL. Radiology 2012;263:663–672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Houssami N, Macaskill P, von Minckwitz G et al. Meta‐analysis of the association of breast cancer subtype and pathologic complete response to neoadjuvant chemotherapy. Eur J Cancer 2012;48:3342–3354. [DOI] [PubMed] [Google Scholar]
- 70.Criscitiello C, Azim HA Jr, Agbor‐tarh D et al. Factors associated with surgical management following neoadjuvant therapy in patients with primary HER2‐positive breast cancer: Results from the NeoALTTO phase III trial. Ann Oncol 2013;24:1980–1985. [DOI] [PubMed] [Google Scholar]
- 71.Boughey JC, Suman VJ, Mittendorf EA et al. Sentinel lymph node surgery after neoadjuvant chemotherapy in patients with node‐positive breast cancer: The ACOSOG Z1071 (Alliance) clinical trial. JAMA 2013;310:1455–1461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Early Breast Cancer Trialists’ Collaborative Group . Long‐term outcomes for neoadjuvant versus adjuvant chemotherapy in early breast cancer: Meta‐analysis of individual patient data from ten randomised trials. Lancet Oncol 2018;19:27–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Derks MGM, van de Velde CJH. Neoadjuvant chemotherapy in breast cancer: More than just downsizing. Lancet Oncol 2018;19:2–3. [DOI] [PubMed] [Google Scholar]
- 74.Provenzano E, Vallier AL, Champ R et al. A central review of histopathology reports after breast cancer neoadjuvant chemotherapy in the neo‐tAnGo trial. Br J Cancer 2013;108:866–872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Provenzano E, Bossuyt V, Viale G et al. Standardization of pathologic evaluation and reporting of postneoadjuvant specimens in clinical trials of breast cancer: Recommendations from an international working group. Mod Pathol 2015;28:1185–1201. [DOI] [PubMed] [Google Scholar]
- 76.Burgués O, Lopez‐García MA, Pérez‐Míes B et al. The ever‐evolving role of pathologists in the management of breast cancer with neoadjuvant treatment: Recommendations based on the Spanish clinical experience. Clin Transl Oncol 2018;20:382–391. [DOI] [PubMed] [Google Scholar]
- 77.Guidance for Industry: Pathological Complete Response in Neoadjuvant Treatment of High‐Risk Early‐Stage Breast Cancer: Use as an Endpoint to Support Accelerated Approval. Silver Spring, MD: Center for Drug Evaluation and Research, Food and Drug Administration, U.S. Department of Health and Human Services; 2014.
- 78.Prowell TM, Pazdur R. Pathological complete response and accelerated drug approval in early breast cancer. N Engl J Med 2012;366:2438–2441. [DOI] [PubMed] [Google Scholar]
- 79.Ellis MJ, Ding L, Shen D et al. Whole‐genome analysis informs breast cancer response to aromatase inhibition. Nature 2012;486:353–360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Dowsett M, Smith IE, Ebbs SR et al. Prognostic value of Ki67 expression after short‐term presurgical endocrine therapy for primary breast cancer. J Natl Cancer Inst 2007;99:167–170. [DOI] [PubMed] [Google Scholar]
- 81.Baselga J, Semiglazov V, van Dam P et al. Phase II randomized study of neoadjuvant everolimus plus letrozole compared with placebo plus letrozole in patients with estrogen receptor‐positive breast cancer. J Clin Oncol 2009;27:2630–2637. [DOI] [PubMed] [Google Scholar]
- 82.Baselga J, Campone M, Piccart M et al. Everolimus in postmenopausal hormone‐receptor‐positive advanced breast cancer. N Engl J Med 2012;366:520–529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Balko JM, Cook RS, Vaught DB et al. Profiling of residual breast cancers after neoadjuvant chemotherapy identifies DUSP4 deficiency as a mechanism of drug resistance. Nat Med 2012;18:1052–1059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Karagiannis GS, Pastoriza JM, Wang Y et al. Neoadjuvant chemotherapy induces breast cancer metastasis through a TMEM‐mediated mechanism. Sci Transl Med 2017;9:eaan0026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.DeMichele A, Yee D, Esserman L. Mechanisms of resistance to neoadjuvant chemotherapy in breast cancer. N Engl J Med 2017;377:2287–2289. [DOI] [PubMed] [Google Scholar]
- 86.von Minckwitz G, Eidtmann H, Rezai M et al. Neoadjuvant chemotherapy and bevacizumab for HER2‐negative breast cancer. N Engl J Med 2012;366:299–309. [DOI] [PubMed] [Google Scholar]
- 87.Cameron D, Brown J, Dent R et al. Adjuvant bevacizumab‐containing therapy in triple‐negative breast cancer (BEATRICE): Primary results of a randomised, phase 3 trial. Lancet Oncol 2013;14:933–942. [DOI] [PubMed] [Google Scholar]
- 88.Urruticoechea A, Smith IE, Dowsett M. Proliferation marker Ki‐67 in early breast cancer. J Clin Oncol 2005;23:7212–7220. [DOI] [PubMed] [Google Scholar]
- 89.Gebhart G, Gámez C, Holmes E et al. 18F‐FDG PET/CT for early prediction of response to neoadjuvant lapatinib, trastuzumab, and their combination in HER2‐positive breast cancer: Results from Neo‐ALTTO. J Nucl Med 2013;54:1862–1868. [DOI] [PubMed] [Google Scholar]
- 90.Nerich V, Saing S, Gamper EM et al. Cost‐utility analyses of drug therapies in breast cancer: A systematic review. Breast Cancer Res Treat 2016;159:407–424. [DOI] [PubMed] [Google Scholar]
- 91.Attard CL, Pepper AN, Brown ST et al. Cost‐effectiveness analysis of neoadjuvant pertuzumab and trastuzumab therapy for locally advanced, inflammatory, or early HER2‐positive breast cancer in Canada. J Med Econ 2015;18:173–188. [DOI] [PubMed] [Google Scholar]
- 92.Albanell J, Ciruelos E, Colomer R et al. Adding pertuzumab in neoadjuvant treatment of patients with HER2+ breast cancer in Spain: A cost offsets study. Basic Clin Pharmacol Toxicol 2015;117(suppl 2):C070A. [Google Scholar]
- 93.Squires H, Pandor A, Thokala P et al. Pertuzumab for the neoadjuvant treatment of early‐stage HER2‐positive breast cancer: An Evidence Review Group perspective of a NICE single technology appraisal. Pharmacoeconomics 2018;36:29–38. [DOI] [PubMed] [Google Scholar]
- 94.Nixon N, Verma S. A value‐based approach to treatment of HER2‐positive breast cancer: Examining the evidence. Am Soc Clin Oncol Educ Book 2016;35:e56–e63. [DOI] [PubMed] [Google Scholar]