Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Jan 1.
Published in final edited form as: Surg Oncol Clin N Am. 2018 Jan;27(1):95–120. doi: 10.1016/j.soc.2017.08.005

Molecular subtypes and local-regional control of breast cancer

Simona Maria Fragomeni 1, Andrew Sciallis 2, Jacqueline S Jeruss 2,3
PMCID: PMC5715810  NIHMSID: NIHMS899872  PMID: 29132568

Synopsis

In the era of personalized medicine, there has been significant progress regarding the molecular analysis of breast cancer subtypes. The major classification schemes are based on gene expression profiling are often referred to as intrinsic subtypes and include:luminal A, luminal B, HER2 enriched, and basal-like.

Research efforts have focused on how classification of these subtypes could provide information on prognosis and influence treatment planning. Cancer subtype stratification has become a critical component of disease characterization.

While much is known about the impact of different molecular subtypes on disease-specific survival, more recent studies have also investigated the role of the different molecular subtypes on local-regional recurrence. This is an area of active study, and in recent years there has been significant progress. The goal of this review is to describe outcomes among disease subtypes to aid in optimal surgical decision-making to improve local-regional control.

Keywords: Breast cancer, Molecular subtypes, Local-regional recurrence, Gene signatures, Immunohistochemical surrogates

Introduction

Breast cancer is a heterogeneous disease that affects one anatomic site yet is phenotypically variable1,2. The identification of different biological subtypes occurs primarily through the use of techniques including immunohistochemistry3 and gene expression profiling1. To date, several studies have shown that the different biological subtypes are associated with variations in treatment response and disease-specific outcomes410. Currently, decision-making for individual patients is based on several factors including tumor morphology and grade classification, tumor size, presence of lymph node metastases, and expression of ER, PR and HER2. While knowledge of these factors aids in treatment planning, there is a clear need to enhance the understanding of both prognostic and predictive markers that will facilitate customized treatment. The advent of novel technologies to aid in the identification of new markers will also be critical.

Through molecular analysis of breast cancers with gene expression profiling, both Perou and Sorlie showed that breast cancer could be sub-classified into different subtypes1,2. Broadly, these subtypes include: luminal ER positive (luminal A and luminal B), HER2 enriched, and basal-like (Table 1). Gene expression profiling can be costly, time consuming, and depending on the platform may require a fresh tumor biopsy sample that has not been fixed in formalin11,12. Given these constraints, gene expression profiling can be difficult to implement on a broad scale. Nevertheless, several groups including the American Society of Clinical Oncology (ASCO), the National Comprehensive Cancer Network (NCCN), and the St. Gallen Group have issued guidelines and recommendations supporting the implementation of molecular analysis as useful for risk stratification and for treatment planning1315.

Table 1.

Classification of molecular subtypes and correlation with biomarker staining on immunohistochemistry

Molecular subtype ER PR HER 2
Luminal A positive and/or positive negative
Luminal B positive and/or   positive
negative*
or negative
Luminal B positive and/or   positive
negative **
or positive
HER2 negative negative positive
Triple negative or basal-like negative negative negative
*

(PR<20% + Ki 67>14%)

**

(Any PR + any Ki 67)

ER – estrogen receptor; HER2 – human epidermal growth factor receptor 2; PR – progesterone receptor

To facilitate the implementation of breast cancer subtype classification, efforts have been made to utilize immunohistochemical analysis to create approximated subtypes. While taking this approach may enable stratification of breast cancers into subgroups that have outcomes comparable to those defined by gene expression profiling, there is not precise overlap16. Furthermore, in addition to the evaluation of standard biomarkers that can be assessed with immunohistochemistry (ER, PR and HER2), the contribution of other factors, such as proliferative rate and expression of cytokeratins, may also be important. For example, the St. Gallen 2013 classification included the evaluation of Ki67 (a marker of cell proliferation)17 and a cutoff of PR of less than 20% as factors associated with the luminal B, HER2-negative subtype15.

The different molecular subtypes reflect the biological diversity of breast cancer. In a time in which medicine is moving towards a personalized approach, a critical goal is the correlation of the different disease subtypes with clinical outcomes and targeted therapeutics. Several studies have evaluated the variance in systemic disease recurrence and survival among the intrinsic subtypes1821. To this end, additional work has pointed toward the growing significance of molecular subtypes in the risk of local-regional recurrence along with clinical and pathological features5.

Local-regional recurrence is described as ipsilateral, in-breast recurrence after lumpectomy, chest wall recurrence after mastectomy, or recurrence in the ipsilateral axillary, or supraclavicular lymph nodes (less commonly infraclavicular and/or internal mammary nodes)22. Overall, among patients with stage I–II breast cancer, approximately 10–15% will develop a local recurrence after breast conserving surgery and radiation therapy, while 10–20% of patients with stage I–IIIA disease will experience a chest wall recurrence after mastectomy22. To date, several factors have been associated with aggressive cancer biology and the increased risk for local-regional recurrence. These factors include:

  • Lymphovascular invasion23

  • Young age24

  • Increasing tumor size25

  • Close or involved margin status26,27

  • Positive nodal status28,29

  • High tumor grade23,30

  • Extensive intraductal component31

  • Multifocal/multicentric disease32

  • Negative hormone receptor status33,34

  • Lack of adjuvant systemic therapy3537

It follows that local-regional recurrence may be associated with a more aggressive tumor biology. The importance of adequate local disease control has been highlighted by findings from randomized studies showing the impact of local-regional recurrence on survival3840. Data from the Early Breast Cancer Trialists’ Collaborative Group (EBCTCG) overview analysis has demonstrated that for every four local recurrences prevented approximately one death may be avoided41.

Defining the biological characteristics of breast cancer facilitates: 1) characterization of different tumor histological types, 2) understanding of disease prognosis and 3) systemic treatment planning. Typically, immunohistochemistry is used to characterize intracellular or cell surface cancer-related protein expression. Among several factors, the most frequently evaluated immunohistochemical breast cancer prognostic and predictive markers include: ER, PR, HER2 and Ki67.

ER/PR

It has been known for some time that estrogen plays an important stimulatory role in the normal breast and in the development and progression of breast cancer. Moreover, the estrogen receptor (ER) is one of the most important prognostic biomarkers in breast cancer. ER belongs to a group of nuclear hormone receptors that act as transcription factors. There are two isoforms: ERα (the clinically measured isoform) and ERβ. Patients whose tumors are ER-positive benefit from endocrine therapy targeting ER (such as tamoxifen and aromatase inhibitors), and treatment can reduce local and distant recurrence and mortality42,43. However, ER-positive breast cancers do not respond as well to cytotoxic chemotherapy and are less likely to achieve a pathologic complete response (pCR) when compared to patients with ER-negative breast cancer who receive neoadjuvant chemotherapy44,45. The progesterone receptor (PR) also manifests as two major isoforms (PR-A and PR-B) and plays a role in downstream ER signaling46. It is likely that the PR acts as a driver for the development of breast cancer, that may be most impactful in postmenopausal women47. The ER is thought to regulate PR expression, and the presence of PR expression is considered indicative of a functional estrogen-ER axis48. In the majority of cases, PR expression correlates with ER expression, and from a practical standpoint, robust PR expression in the absence of ER may necessitate repeat testing. The presence of PR expression carries prognostic significance in early breast cancer with ER- and PR-positive cases having the best outcome49. Additionally, PR expression correlates with tumor responsiveness to endocrine therapy even when PR expression is low (i.e. ≥ 1% of tumor cell nuclei)50. However, in the setting of an ER-positive breast cancer, PR assessment may not add significant predictive information. That being said, breast cancers that are both ER- and PR-positive may derive greater benefit from endocrine therapy than ER-positive/PR-negative tumors51.

Currently, the evaluation of ER and PR receptor expression is standard practice and most often performed using immunohistochemistry. ER and PR immunoexpression manifests as nuclear staining, and heterogeneous (“physiologic”) expression is typically observed in normal breast ductal epithelium. Up to 80% of breast cancers are ER-positive52 and 55–65% are positive for PR expression53. The American Society of Clinical Oncology (ASCO) and College of American Pathologists (CAP) have provided recommendations regarding ER/PR measurement and reporting54. Per these recommendations, pathology reports for ER and PR results should include the following details: percentage and/or proportion of tumor cells staining positively in a tissue section or cytology preparation, the overall intensity of staining (weak, moderate, strong), and an interpretation as to hormone receptor status. As to the latter, ER and PR expression is considered positive when ≥ 1% of tumor cells stain positively, negative when < 1%, and uninterpretable when pre-analytic variables preclude an accurate assessment. These include the use of fixatives other than 10% neutral buffered formalin during tissue processing, duration of fixation less than 6 hours or more than 72 hours, delay in fixation/cold ischemic time more than 1 hour, decalcification of specimens using acids, and inappropriate intrinsic/extrinsic assay controls. Some institutions provide a composite score combining percentage and intensity of tumor cell hormone receptor expression.

The majority of pathology laboratories utilize immunohistochemical methods to determine ER and PR status. Testing is often performed on biopsy material prior to surgery as there is excellent agreement in hormone receptor expression between biopsy and resection specimens55. This enables clinicians, oncologists, surgeons, and patients to possess vital prognostic and therapeutic information prior to establishing a treatment plan. However, there is institutional variability in the manner by which ER and PR immunoexpression is evaluated. At some centers, pathologists utilize digital image analysis (DIA) in which slides are scanned and converted into high-resolution computer images so that quantitative immunohistochemistry can be performed56. DIA is being utilized with greater frequency owing to established intra- and inter-observer variability in scoring results by pathologists57. Finally, repeat testing of ER and PR on excision specimens in cases where prior core biopsy showed weak or equivocal expression is also undertaken at some institutions.

Androgen Receptor

Another hormone receptor currently being studied in breast cancer research, is the androgen receptor (AR). Like ER and PR, immunohistochemical AR expression is nuclear and usually strong and diffuse when present (i.e., present in > 80% of tumor cell nuclei). Expression is seen in the majority of ER-positive breast cancers as well as subsets of triple negative and HER2-positive carcinomas. In the spectrum of triple negative breast cancer (TNBC), tumors with robust AR expression often manifest as apocrine carcinoma (“carcinoma with apocrine differentiation”), a ductal tumor where the invasive cells exhibit extensive apocrine cell change (> 90% of tumor cells exhibiting apocrine features)58. The majority of pathologists view AR immunoexpression in 1% of tumor cell nuclei as AR-positive. Molecular studies using RNA expression profiling have revealed several potentially clinically relevant breast cancer subtypes. One of these is known as the molecular apocrine group, defined as tumors that are ER-negative but AR-positive59. These tumors often exhibit an expression profile that overlaps with luminal, basal-like, and occasionally HER2-positive groups. Interestingly, many of these tumors, but not all, show some degree of apocrine differentiation on histopathology.

From a clinical perspective, AR-positive breast cancers appear to behave favorably when compared to AR-negative tumors, regardless of ER status60. Among TNBCs, the significance of AR expression on prognosis and clinical course continues to be investigated. In some studies, AR-positive TNBC has a better prognosis when compared to AR-negative TNBC, however others have found a decreased survival for patients with early stage disease61. Further confounding the prognostic significance of AR expression is the presence of HER2 overexpression in molecular apocrine cases. The favorable clinical course of AR-positive TNBC may be related to the genetic overlap with luminal tumors. Because the AR can be targeted via aromatase inhibition, there has been considerable clinical interest in interrogating TNBC for AR expression, however there are no guidelines regarding routine testing for AR for all patients with breast cancer.

HER2

HER2 (human epidermal growth factor receptor 2, erbB-2) is a transmembrane tyrosine kinase receptor that regulates cell growth, proliferation, and survival through several different signaling pathways such as the mTOR and RAS/RAF/MEK/ERK pathways62. HER2 gene amplification is observed in 15–30% of breast cancers and is a strong prognostic biomarker for an aggressive clinical course63.

Importantly, HER2 gene overexpression is also a strong predictive marker of response to anti-HER2 therapy, which includes humanized monoclonal antibodies that bind to the extracellular domain of the HER2 receptor (trastuzumab and pertuzumab), small molecular receptor tyrosine kinase inhibitors (lapatanib), and an antibody drug conjugate of trastuzumab (ado-trastuzumab emtansine; T-DM1). Immunohistochemistry for HER2 protein overexpression has been developed, and testing for this protein is standard for invasive breast carcinomas whether primary or metastatic. Overexpression of the HER2 protein typically occurs secondary to HER2/neu gene amplification64. As per the most recent 2013 ASCO/CAP guideline recommendations65 regarding HER2 testing in breast carcinoma, the threshold for a positive test on immunohistochemistry is strong circumferential staining in > 10% of invasive tumor cells (score 3+), while criteria for a negative result includes (A) no staining observed or incomplete, faint/barely perceptible membrane staining in ≤ 10% of invasive tumor cells (score 0), or (B) incomplete, faint/barely perceptible membrane staining in > 10% of tumor cells (score 1+). An equivocal result (score 2+), is rendered when there is either incomplete and/or weak-to-moderate circumferential membrane staining in > 10% of invasive tumor cells or there is complete, intense, circumferential membrane staining in ≤ 10% of invasive tumor cells. Breast cancers with an equivocal result for HER2 overexpression are reflexed to either fluorescence in situ hybridization (FISH), chromogenic in situ hybridization (CISH), or silver-enhanced in situ hybridization (SISH) assays in order to determine HER2 gene amplification. Immunohistochemistry and gene amplification assays usually yield similar results, as gene amplification induces immunohistochemically detected protein overexpression in 95% of cases. Most institutions utilize FISH, however CISH and SISH techniques are increasingly being utilized as they may be implemented using a light microscopy. Moreover, probes for HER2 as well as centromere 17 (centromere enumeration probe, CEP17) are employed so that the ratio of HER2 signals to CEP17 can be calculated. A positive (amplified) test result is rendered when the average HER2 copy number ≥ 6.0 signals/cell or HER2/CEP17 ratio ≥ 2.0, while a negative (not amplified) result is found when the average HER2 copy number < 4.0 and the HER2/CEP17 ratio < 2.0. As for HER2 immunohistochemistry, an equivocal result is observed with a HER2/CEP17 ratio < 2.0 and the average HER2 copy number is ≥ 4.0 but < 6.0.

Ki67

Ki-67 is a nuclear proliferation marker expressed in all phases of the cell cycle except G066. In general, breast cancers expressing high levels of Ki67 correlates with worse outcomes67,68. Meta-analyses by de Azambuja et al. in 2007 and Stuart-Harris et al. in 2008, showed that Ki67 was associated with prognosis, disease-free survival, and overall-survival68,69. In 2009, the St. Gallen Consensus stratified tumors according to the Ki67 value as low, < 15%, intermediate, 16%–30%, and highly proliferative, > 30%, to help identify patients that could potentially benefit from treatment with chemotherapy or endocrine therapy. Additional studies reported a Ki67 level above 10%–14%70,71 and 20% (St. Gallen 201315), as characteristic of high-risk for poor outcomes. The implementation of Ki67 has been complex as some studies have used 10%, 14% or 20% cutpoints for treatment recommendations72,73. Accordingly, Maggie et al. defined a Ki67 cutoff to distinguish luminal A and B subtypes using immunohistochemistry74. This work defined the luminal A subtype as ER and/or PR-positive, HER2 negative, and Ki67 low (i.e., a Ki67 index of <14%) and the luminal B subtype as ER and/or PR-positive, HER2-negative, and Ki67 high (i.e., a Ki67 index of ≥14%) and showed that stratification using the immunohistochemical panel of four biomarkers (i.e., ER, PR, HER2, and Ki67) was statistically significant74. ASCO guidelines have not included Ki67 in the list of routinely assessed markers for breast cancer prognosis75. In Europe, the St. Gallen International Expert Consensus recommended the use of proliferation markers, such as Ki67, in addition to standard parameters (stage, grade, hormone receptors status) when choosing systemic treatment regimens15. A role for Ki67 was also found in the neoadjuvant setting, in particular for patients undergoing preoperative endocrine therapy76, indicating how a variation in Ki67 expression may predict long-term outcomes. Nishimura and colleagues, in 2010, investigated the role of Ki67 in neoadjuvant systemic treatment and found that marker values before starting treatment could help to predict response, while those measured after chemotherapy could help predict disease-free survival77. Additionally, measurement of Ki67 is cost effective when compared to multigene assays. At some centers, Ki67 is included in surgical pathology reports along with other standard markers, and can aid in the identification of luminal B tumors not identified by ER, PR and HER2.

Molecular subtypes

Luminal A

Frequency: 30–40% of all invasive breast cancers. Gene expression profiling (GEP): PIK3CA mutations, MAP3KI mutations, ESR1 high expression, XBP1 high expression, GATA3 mutations, FOXA1 mutations; gain of 1q, 8q, loss of 8p, 16q. Morphology: Grading 1 or 2; most are well differentiated carcinomas of no special type (NST), classical lobular carcinomas, tubular, mucinous, neuroendocrine and cribriform carcinomas78. Immunohistochemical profile (IHC): ER-positive, PR high expression (≥20%), HER2-negative, and low Ki-6779,80. Several studies showed that a Ki-67 of 14% was the cutoff point to separate luminal A from luminal B subtypes74. More recently, this cutoff was changed in 20%81.

Luminal B (HER2-negative)

Frequency: 20% to 30% of all invasive breast cancer. GEP: TP53 mutations, PIK3CA mutations, Cyclin D1 amplification, MDM2 amplification, ATM loss, enhanced genomic instability, focal amplifications (e.g. 8p12, 11q13). Morphology: Grading 2 or 3; less well differentiated cancers, mostly invasive ductal carcinomas NST and also some invasive micropapillary carcinomas78. Immunohistochemical profile: ER-positive, lower PR expression (<20%)15, HER2-negative and higher level of Ki67 labeling index (>14 or 20%)52,74. Correlation to Oncotype results: high recurrence score is suggestive of luminal B like subtype (St. Gallen 2013)15. Furthermore, on a molecular basis, it seems that luminal A and luminal B breast cancers have specific gene profiles that lead to oncogenic proliferation.

HER 2

Frequency: 12% to 20% of all invasive breast cancers. GEP: HER2 amplification, TP53 mutations, PIK3CA mutations, FGFR4 high expression, EGFR high expression, APOBEC mutations, cyclin D1 amplification, high genomic instability1,2,8285. Morphology: Grading 2 or 3; infiltrating carcinoma NST, apocrine and pleomorphic lobular carcinomas. Classification: in the study from Fountzilas et al.,86 hormone receptor status was shown to affect survival, metastatic spread, and treatment response. Therefore, on immunohistochemistry, HER2-positive tumors can be divided in two subtypes: HER2-enriched subtype (ER and/or PR-negative/HER2-positive)8688 andluminal HER2 subtype (ER and/or PR-positive, HER2-positive); and further divided into two phenotypes based on PR expression: ER-positive, PR-positive, HER2-positive and ER-positive, PR-negative, HER2-positive. There is preclinical evidence for crosstalk between the HER2 and ER signaling pathways with a negative effect on the response to endocrine therapy8993.

Triple negative breast cancer (TNBC)

Representing 15–20% of all invasive breast cancers, TNBC is defined by the absence of expression of ER, PR, and HER2. Despite its simple definition, it is a morphologically, genetically, and clinically heterogeneous category of breast cancer. Most TNBC manifest as invasive ductal carcinoma of no special type (NST), however the category also includes variants such as metaplastic carcinoma, carcinoma with medullary features (formerly known as medullary carcinoma), carcinoma with apocrine features (formerly known as apocrine carcinoma), secretory carcinoma (formerly known as juvenile breast cancer), and adenoid cystic carcinoma58. Gene-expression profiling has enabled the subdivision of these cancers into different, prognostically significant subtypes that, in some cases, correlate with a particular pathologic variant. The most common, and best characterized, molecular subtypes of TNBC are basal-like 1 (BL1), basal-like 2 (BL2), immunomodulatory (IM), mesenchymal (M), mesenchymal stem like (MSL), and luminal androgen receptor (LAR)94. BL1 breast cancers usually manifest clinically and pathologically as invasive ductal carcinoma NST and have high Ki67 proliferative indices while their expression profile is enriched in genes associated with basal cytokeratin genes (including KRT5), cell cycle and DNA replication, and DNA damage response pathways. BL2 tumors also manifest as invasive ductal carcinoma NST and also show increased basal cytokeratin gene expression, TP63, and growth factor signaling (e.g. EGF and IGFR1 pathways). In general, the immunomodulatory subtype of TNBC overlaps with carcinoma with medullary features, as both often contain a brisk lymphocytic infiltrate. Mesenchymal and mesenchymal stem-like subtypes may present clinically as metaplastic carcinoma, defined as breast cancer with epithelial and mesenchymal differentiation sometimes taking the form of heterologous elements like bone and cartilage. These tumors are enriched in genes important for cell differentiation and growth factor signaling pathways. Finally, the LAR subtype often presents as carcinoma with apocrine differentiation and correlates with the molecular apocrine type. As expected, LAR shows increased expression of luminal cytokeratin genes (including KRT7 and KRT1) and enriched in AR mRNA and protein expression, possibly explaining their responsiveness to hormone therapies.

Gene Expression Testing

The information obtained from microarray based gene expression profiling has revealed further information about breast cancer as a group of different diseases with different biological characteristics and behaviors95,96. A key finding, in addition to the validation of ER-positive and ER-negative cancers as different diseases from a molecular point of view, was that the expression of proliferation-related genes was also associated with prognosis of ER-positive tumors12,97. Through the advent of methods for gene expression testing, a prognostic multi-gene assessment has also been developed and validated in clinical trials, and integrated into clinical practice98. Through the work of Perou1 and Sorlie2, a group of ‘intrinsic genes’ was identified, revealing four molecular subtypes of breast cancer1 — luminal A and B, HER2-enriched and basal-like. The groups of genes primarily responsible for the segregation of molecular subtypes are genes related to expression of ER, regulation of proliferation, HER2, and genes mapping to chromosome 171,2. The prognostic signatures currently available provide clinicians with data complementary to factors with known prognostic significance including tumor size and nodal status (Table 2). To date, these gene signatures are more informative for patients with ER-positive disease, but have a more limited role for patients with ER-negative tumors. One of the main objectives of molecular subtyping was the ability to identify factors to aid in the discrimination between patients a with favorable versus an unfavorable prognosis to guide therapeutic decision-making96. In recent years, several prognostic signatures have been identified96 that revealed overlapping groups of patients with a poor prognosis99, primarily characterized by cancers with high expression of proliferation-related genes95,99. Given the significance of proliferation-related gene expression for prognosis for both ER-positive and ER-negative disease, these factors are active targets for cytotoxic chemotherapies95,100. In terms of outcome prediction generated from molecular subtyping, the insights obtained appear to extend beyond the short-term time-frame (e.g. 10 years instead of 5)101,102 serving as complementary to the parameters routinely used in clinical practice96,99.

Table 2.

Commercially available gene signatures

Test Method Result Clinical application Level of evidence
MammaPrint®
  • Microarray

  • 70 gene

  • Fresh tissue, frozen material and alternatively formalin fixed and paraffin embedded (FFPE)

Distant metastasis at 5 years (without adjuvant treatment):
  • Low risk 13%

  • High risk 56%

Prognosis of N0, <5 cm diameter, stage I/II disease, age <61 years II
Oncotype DX™
  • qRT – PCR

  • 21-gene

  • Fresh frozen tissue and/or FFPE

Risk of 10 years distant recurrence:
  • Low (<18)

  • Intermediate (18–30)

  • High (≥31)

Prediction of recurrence risk in ER+ and N0 disease treated with tamoxifen I

70 Gene Signature

MammaPrint (Agendia; Amsterdam, Netherlands) is a microarray-based test, primarily implemented in Europe, that can be used for risk stratification (i.e. low or high) for both ER-positive and ER-negative breast cancer patients. It was based on data derived from stage I and II patients younger than 55 years of age, with node-negative tumors less than 5 cm. The test can be performed with fresh or formalin fixed tumor samples. Much of the data related to this test was obtained from retrospective studies and the findings were then confirmed prospectively by the RASTER study103. The results of the prospective MINDACT trial recently published showed survival rates of ~95% at 5 years for low risk patients that did not receive chemotherapy.104

21 Gene Recurrence Score Assay

In the United States, implementation of the Oncotype DX assay has been widespread for patients with node-negative hormone receptor-positive breast cancer105. It is performed using a reverse transcription polymerase chain reaction (RT-PCR) on RNA isolated from paraffin-embedded breast cancer tissue, measuring the expression of 21 genes (16 cancer related genes and 5 reference genes). The Oncotype DX 21 gene recurrence score (RS) assay has been defined as a continuous variable (ranging from 0 to 100). This assay can be considered as an independent prognostic factor in node-negative, ER-positive breast cancer, measuring the risk of distant relapse at 10 years. With values ranging from 0 to 100, patients are stratified into 3 groups: 1) low risk (RS <18), 2) intermediate risk (RS 18–31), and 3) high risk (RS ≥31). These groups are associated with 10-year relapse rates of 7%, 14%, and 30%, respectively. For patients with ER-positive tumors, those who fall in the high risk group may have greater benefits from the use of chemotherapy105. Appropriate management of patients in the intermediate-risk group will be revealed through results of the TAILORx study (NCT00310180). Patients (ER-positive and node-negative) enrolled in TAILORx were assigned to low risk (RS <11), intermediate risk (RS 11–25), and high-risk groups (RS >25). The main aim of the study was to evaluate the risk of relapse after surgery for the intermediate risk group. Patients in this risk category were randomly assigned to hormone treatment alone or in combination with chemotherapy. The trial completed enrollment and results are pending. Emerging data from several studies highlight how Oncotype DX may also play a role in different patient scenarios including those with positive lymph nodes, ER and HER2-positive disease, and patients treated with aromatase inhibitors instead of tamoxifen106,107.

Based on the available evidence, guidelines have been developed for the use of for Oncotype DX clinically.NCCN guidelines suggest that Oncotype DX can be used as a predictor of recurrence and can be used as a guide for treatment decision making in ER-positive, node-negative breast cancer based on level I evidence14.

ASCO guidelines suggest it can be used as a tumor marker for risk of recurrence108.

In Europe, the latest St. Gallen International Expert Consensus recommended that the validated assay should be used to clarify the indication for chemotherapy and added to other known prognostic and predictive factors15. Along with the ability of the recurrence score to aid in predicting the response to adjuvant chemotherapy107,109, additional studies have shown how the recurrence score can be predictive of local-regional and distant recurrence105,110. While evidence suggests that the recurrence score obtained with the Oncotype DX correlates with traditional clinicopathologic data, the information obtained regarding prognosis from tumor size, lymph node status, and histologic grade remain as independent variables109,111.

Clinical Outcomes: Local-regional recurrence (LRR)

Significant progress has been made in terms of systemic therapy and targeted treatments, improving outcomes for breast cancer patients. Optimal local-regional control continues to be relevant in terms of clinical outcomes. The risk of a local-regional recurrence has been correlated to different parameters (both pathologic and clinical) with the aim to guide treatment decisions.

Risk factors for LRR

Breast conserving therapy (BCT)

Age

Studies have found that age is a risk factor for local-regional recurrence after BCT, with 10-year local recurrence rates reportedly higher for younger patients112,113. One relevant study showing the impact of age was the EORTC 22881-10882 trial114. A tumor bed boost used in addition to standard whole breast irradiation resulted in a reduction in local recurrence from 23.9% to 13.5% in patients aged 40 years or younger when compared to a decrease from 7.3% to 3.8% in patients older than 60 years114. Accordingly, clinicians should consider patient age when considering treatment planning in patients undergoing breast conserving surgery.

Margins

Positive margins (defined as ink on invasive carcinoma or ductal carcinoma in situ) are associated with a 2-fold increase in the risk of local recurrence when compared with negative margins (> 2 mm). This was an independent risk factor, not mitigated by radiation boost or other characteristics. There is no evidence that more widely free margins reduced this risk even in cases with other risk factors including: young age, unfavorable biology, lobular cancers, or cancers with an extensive intraductal component (EIC)115.

Extensive intraductal component (EIC)

EIC is defined as an infiltrating ductal cancer in which more than 25% of the tumor volume is DCIS and DCIS extends beyond the invasive cancer into surrounding normal breast parenchyma. Invasive breast carcinoma is accompanied by EIC in 15%–30% of patients14. It can be considered as a risk factor for local recurrence.114,116,117

Mastectomy

Several studies have identified risk factors for local recurrence after mastectomy (Jagsi el al. and the Breast Cancer Cooperative Group (DBCG82) b and c studies). Jagsi et al. demonstrated how tumor size > 2 cm, margins < 2 mm, premenopausal status, and lymphovascular invasion can be considered as independent prognostic factors for LRR. Ten-year LRR was 1.2% for those patients with no risk factors, and as high as 40.6% for those with 3 risk factors118. This retrospective analysis suggested the benefit of post-mastectomy radiation therapy (PMRT) to reduce the risk of recurrence for patients with node-negative disease and high-risk factors. In the Breast Cancer Cooperative Group (DBCG82) b and c studies, tumor size, histology positive for ductal carcinoma, high tumor grade, invasion of the pectoralis major fascia, several positive nodes, and extracapsular spread were identified as risk factors for LRR119,120. Abdulkarim et al. reported an increased risk of local-regional recurrence in women with node-negative, triple-negative breast cancer with tumors less than or equal to 5 cm when mastectomy was performed without radiation. This prospective study further identified that risk factors for LRR include node-positive disease, tumors greater than 2 cm, and lymphovascular invasion121.

Additional risk factors

Large tumor size, poor tumor differentiation, nodal status, multifocal/multicentric disease, and adjuvant treatment have all been found to be relevant to the assessment of the risk for local-regional recurrence122.

Lymphovascular invasion

LVI is a factor associated with a higher risk of recurrence. In patients treated with mastectomy, this parameter is one of the risk factors to be considered when offering postmastectomy radiation therapy14,123.

Receptor status

ER, PR and HER2 status, known as predictors of response to targeted treatment, also have a prognostic role after breast conserving therapy and mastectomy42. As described in the analysis of the Danish 82 b and c trials, triple negative disease was associated with higher local recurrence rates after mastectomy6.

BRCA1 and 2 mutation carriers

Patients with sporadic breast cancer treated with breast conserving surgery have an approximate 10% risk of ipsilateral-breast tumor recurrence. Although some studies have reported a risk reduction of contralateral breast cancers in BRCA mutation carriers who take tamoxifen or undergo oophorectomy, contralateral prophylactic mastectomy can decrease the risk to less than 10%124. Risk reduction strategies in BRCA1/2 mutation carriers, including bilateral prophylactic mastectomy, oophorectomy and/or the use of tamoxifen are being implemented in many counries124,125. BRCA1 mutation cancers are more likely to have estrogen receptor negative breast cancer and are also more likely to develop tumors with a higher histologic grade.

There are several studies showing that the different intrinsic subtypes have differences in overall prognosis2 and different rates of local-regional disease recurrence (Table 3).

Table. 3.

Local-regional recurrence by molecular subtype*

Molecular subtype Frequency (%)

Luminal A 0.8 – 8
Luminal B 1.5 – 8.7
HER2** 1.7 – 9.4
Triple negative    3 – 17
*

Patients treated with upfront surgery

**

Luminal HER2-positive and HER2-enriched

Intrinsic subtypes, what role?

Luminal A

The luminal A molecular subtype is generally associated with a highly favorable prognosis126 and typically shows less frequent and less extensive lymph nodal involvement127,128. This subtype also tends to have a more indolent course with a slower evolution over time when compared with the other molecular subtypes129. Additionally, a positive hormone receptor status is both a favorable prognostic factor and also predictive of response to endocrine therapy42,130,131. In terms of LRR several retrospective studies have shown similar outcomes with percentages ranging between 0.8 and 8%4,5,9,132,133.

Luminal B

The luminal B molecular subtype is associated with a more intermediate prognosis when compared with the luminal A molecular subtype126. The risk of local-regional recurrence for luminal B tumors, as described in literature, ranges from 1.5 and 8.7% with a peak of incidence in the first 5 years after diagnosis4,5,9,132,133.

Luminal tumor considerations

The luminal molecular subtypes have lower local-regional recurrence rates, with bothconservative surgery and mastectomy134. In the study by Lowery et al. it was estimated that the rate of LRR for both luminal A and B subtypes after breast conserving surgery and radiation was approximately 5%, even in the setting of noncompliance with endocrine therapy134. It is important for clinicians to reinforce that endocrine therapy reduces the local-regional recurrence and mortality rates by more than 50% in ER-positive breast cancer patients between 5 and 10 years from diagnosis42.

HER2-positive disease

Historically, HER2 overexpression has been associated with a higher frequency of local-regional recurrence based on studies where patients were not treated with HER2-targeted therapy, ranging between 4 and 15%4,5,9,132,133. In the meta-analysis from Lowery et al., 12,592 patients were evaluated for local-regional recurrence after breast conserving surgery (BCS) with radiation therapy and mastectomy. The results showed increased rates of LRR for patients with HER2-positive tumors, with higher rates of recurrence observed after BCS versus mastectomy. In this study, less than 6% of the HER2-positive patients were treated with trastuzumab. A subgroup analysis of 9,306 patients from nine studies showed a lower rate of LRR after mastectomy for luminal HER2-negative tumors, when compared to luminal HER2-positive cancers (7.5% vs. 9.4%) although in this analysis the use of trastuzumab was also low. In this subgroup analysis, no difference was observed in the risk of LRR after BCS for luminal HER2-negative tumors compared with luminal HER2-positive (RR 0.8 – 95% CI) tumors134. Overall, both luminal HER2-negative and luminal HER2-positive cancers showed a positive trend toward lower LRR rates when compared to ER-negative/HER2-positive and TNBC subtypes134. More recently, patients with HER2-positive tumors receive targeted therapies, including trastuzumab and pertuzumab, and this has modified the natural course of this disease subtype, resulting in improved outcomes126. HER2-positivity is a predictive factor for response to trastuzumab135 and also a prognostic factor for LRR131,136138. In astudy by Panoff et al.,139 patients with HER2-positive tumors who underwent mastectomy and received trastuzumab, had a LLR of 1.7%. This finding was supported by an analysis of six studies from Yin et al., who also showed trastuzumab treatment resulted in a decrease in LRR by 50%140. It has been suggested that breast cancer cells with radiation damage could be more vulnerable to injury when deprived of the mitogenic cell signaling provided by the HER2 pathway activation141. Further to this point, a study conducted in T1 (a or b) HER2-positive breast cancer patients showed a risk of LRR of 2 – 5.7% for those who did not receive trastuzumab, and 0% for those patients treated with trastuzumab +/− chemotherapy at 5 years post-diagnosis142.

TNBC

TNBC is associated with an unfavorable prognosis when compared to the other breast cancer subtypes secondary to a higher risk of disease recurrence126,143,144. For TNBC, the involvement of regional lymph nodes is associated with a poor prognosis, without a direct relationship to the number of involved nodes135. The TNBC subtype has been associated with a higher risk of local-regional relapse10 and contralateral disease, as shown by Bessonova et al.145 and Malone et al.143, and also systemic relapse126. Peak incidence for recurrence in TNBC has been reported in the first 1–3 years, as reported in studies by Jatoi et al.129 and Kumar et al.146. In the Lowery meta-analysis, the TNBC subtype was associated with increased LRR rates, after BCS and mastectomy, ranging from 3 and 17%134. Other studies have reported that TNBC is not associated with higher local-regional recurrence risk147,148. Abdulkarim et al.121 reported no difference in local recurrence events, while Wang et al147 found patients with TNBC undergoing mastectomy to have inferior outcomes. Several other studies have described the TNBC subtype as having the highest risk for LRR4,5,9,10,22,132,133. Moreover, after the diagnosis of a local-regional recurrence, this molecular subtype is associated with a high incidence of distant metastases and cancer related mortality146.

Another topic of debate is the apparent radiation resistance of this molecular subtype, which may be impacted by ER negativity, shorter cell cycle duration, and less time for DNA damage repair. Kyndi et al. reported a smaller reduction in LRR rates after PMRT in TNBC, and no overall survival benefit6, thought to be secondary to radio-resistance associated hormone receptor negativity146. Some studies are evaluating ways to improve radio-sensitivity through use of cisplatin-based chemoradiation in the setting of TNBC126.

LLR and molecular subtypes in the neoadjuvant setting

Huang et al. reported in the neoadjuvant setting that multifocal/multicentric disease, number of positive axillary nodes, axillary dissection with <10 nodes, lymphovascular invasion, extracapsular extension, skin or nipple involvement, and ER-negative disease were significantly associated with LRR. Patients who achieved a pathologic complete response (pCR) had a lower rate of LRR (2% vs. 12%)149. The study by Levy et al. evaluated the long-term local-regional control rates after breast-conserving therapy for patients undergoing surgery before or after chemotherapy150. In this study, ER and/or PR-negative disease were associated with higher rates of LLR, and the 10 year LRR rate was not related to surgical treatment approach150. Mamounas and colleagues also reported that after neoadjuvant treatment, surgical approach was not related to a different incidence in LRR151. Thus, for patients treated with neoadjuvant therapy, as for patients treated with surgery first, clinical stage, nodal involvement and histology can be considered prognostic factors for LRR. Two large trials, NSABP B-18 and B-27, have also shown that a pCR (defined in these studies as the absence of invasive tumor in the breast), after NCT can be considered as a prognostic factor for LRR151. von Minckwitz and colleagues studied 6,377 patients after neoadjuvant treatment, and found pCR was associated with better outcomes in luminal B/HER2-negative, HER2-positive (non-luminal), and TNBC subtypes, but not for patients with luminal B/HER2-positive or luminal A cancers152. Liedtke et al. found that patients experiencing a pCR had excellent outcomes regardless of receptor status but, when comparing TNBC patients who did not achieve a pCR to other subtypes, TNBC was associated with a lower overall and post-recurrence survival rate, with a risk of relapse and death found to be higher in the first 3 years after diagnosis. Despite this, when TNBC patients achieve a pCR, they have improved survival153. In a series published by Swisher et al. at the MD Anderson Cancer Center, the LRR-free survival rate for TNBC patients with residual disease post-treatment was estimated at approximately 89.9% versus 98.6% for patients with a pCR154. This study concluded that TNBC was an independent predictive factor for LLR154. The role of pCR on LRR was not observed for patients with HER2-positive subtypes154. Similar findings were reported in the studies of Peterson et al. and Kiess et al.155,156. Taken together, these findings point toward the role of molecular subtypes in the locoregional recurrence and could help to facilitate patient selection for optimal treatment planning.

LLR and Axillary Disease

Many changes have occurred in the management of the axilla over the last several years. Two trials that have proposed treatment changes include the AMAROS trial and ACOSOG Z0011. The AMAROS trial, was a multicenter phase 3 trial in which patients were enrolled with T1 and T2 breast cancers (both unifocal and multifocal) a clinically negative axilla and positive sentinel nodes. Patients were treated with BCS or mastectomy and were assigned to axillary node dissection versus radiation treatment (breast tangential fields, axillary and supraclavear nodes). The study endpoints were to evaluate the axillary recurrence, DFS, OS and quality of life in terms of arm lymphedema and motility. The authors showed additional metastases were found in 33% of cases and no differences were identified in terms of DFS and OS between the two treatment groups, with less morbidity reported in the axillary RT group (lymphedema 11% vs. 23%)157.

The American College of Surgeons (ACOSOG) Z0011 trial enrolled clinically node-negative women with T1 and T2 breast cancer with 1–2 positive sentinel nodes who underwent BCS and breast irradiation (breast tangential fields). This study showed that 27% or patients in the axillary dissection arm had additional positive non-sentinel nodes. There was no difference in regional recurrence and no difference in overall and disease-free survival between those who had completion axillary dissection versus those undergoing sentinel node dissection alone Nodal recurrences were reported in 1% of patients158,159. These results suggest that regional control can be achieved by radiation and systemic therapy, regardless of cancer subtypes160.

Ongoing trials and future perspectives

Currently, PMRT is not recommended for patients at low risk for LRR (<10%)161. The St. Gallen recommendations indicate PMRT should be considered in cases where the 10-year LRR rate is 20% or higher162,163. Therefore, the need to quantify the LRR rate is essential, and molecular subtyping could be a key part of this evaluation.

Currently there are several trials examining the need for radiation therapy based on disease subtype. IDEA study (Individualized Decisions for Endocrine Therapy Alone). In this prospective multicenter study, after the confirmation of a stage 1 invasive breast cancer, after breast conserving surgery, hormone receptor-positive, HER2-negative patients with an Oncotype-DX recurrence score less than or equal to 18 could be included. The enrolled patients receive endocrine therapy alone without radiotherapy164. PRECISION trial (Profiling Early Breast Cancer for Radiotherapy Omission). This trial is a prospective study performed on unifocal T1 (≤ 2 CM), ER-positive (≥ 10%), PR-positive, HER2-negative, grade 1 or 2, pN0 or pN0(i+) associated with a PAM 50 (Prosigna) low risk score. All the participants undergo omission of radiation therapy and will receive adjuvant endocrine treatment165. LUMINA. This is a prospective single arm study aims to evaluate the risk of ipsilateral breast cancer recurrence after breast conserving therapy without radiation. Patients included in the study are 55 years of age, tumors less or equal to 2 cm, ductal, tubular or mucinous disease, ER and PR-positive, HER2-negative, node-negative, with negative margins166.

SUPREMO trial

A randomized phase III trial assessing the role of chest wall irradiation in women with intermediate risk breast cancer following mastectomy. This is a trial from the Medical Research Council/European Organization for Research and Treatment of Cancer (MRC/EORTC) not yet recruiting167.

Conclusions

Luminal A cancers are associated with a better prognosis and a LRR that occurs more frequently after 5 or 10 years from diagnosis. Although the expression of ER and PR receptors overlaps between luminal A and luminal B cancers, it is critical to identify luminal B breast cancers because this subgroup is associated with a worse prognosis and could benefit from additional local and systemic treatment. HER2-positive cancers have improved outcomes in both local and systemic control with targeted therapies. TNBC remains a clinical challenge and several clinical trials are ongoing in attempts to identify mechanisms to help improve outcomes for this patient subgroup. The increasing prognostic and predictive information obtained from both prospective and retrospective studies on molecular subtypes will allow clinicians to customize treatment in terms of surgery, regional and systemic therapy, and follow-up. Implementing knowledge about tumor biology allows clinicians to distinguish high risk versus favorable, low risk disease subtypes through several different criteria. Further investigations will be necessary to potentially change guidelines regarding treatments offered for the local-regional control of breast cancer.

Keypoints.

  • The analysis of cancer gene expression patterns expands the understanding of breast cancer as a heterogeneous group of diseases.

  • The presence or absence of estrogen, progesterone, and HER2/neu receptor (ER/PR/HER2) expression is key to molecular subtype stratification.

  • Immunohistochemical techniques are widely applied to identify the markers of the different molecular subtypes.

  • Gene expression profiling techniques, including commercially available tools such as Oncotype Dx and MammaPrint, are considered complementary to the known prognostic factors.

  • According to the currently available data, the different molecular subtypes are associated with different patterns of local-regional recurrence and response to treatment.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflicts of interest: The authors have nothing to disclose.

References

  • 1.Perou CM, Sorlie T, Eisen MB, et al. Molecular portraits of human breast tumours. Nature. 2000;406(6797):747–752. doi: 10.1038/35021093. [DOI] [PubMed] [Google Scholar]
  • 2.Sorlie T, et al. Perou CM, Tibshirani R, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci. 2001;98(19):10869–10874. doi: 10.1073/pnas.191367098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Nielsen TO, Hsu FD, Jensen K, et al. Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res. 2004;10(16):5367–5374. doi: 10.1158/1078-0432.CCR-04-0220. [DOI] [PubMed] [Google Scholar]
  • 4.Millar EKA, Graham PH, O’Toole SA, et al. Prediction of local recurrence, distant metastases, and death after breast-conserving therapy in early-stage invasive breast cancer using a five-biomarker panel. J Clin Oncol. 2009;27(28):4701–4708. doi: 10.1200/JCO.2008.21.7075. [DOI] [PubMed] [Google Scholar]
  • 5.Voduc KD, Cheang MCU, Tyldesley S, Gelmon K, Nielsen TO, Kennecke H. Breast cancer subtypes and the risk of local and regional relapse. J Clin Oncol. 2010;28(10):1684–1691. doi: 10.1200/JCO.2009.24.9284. [DOI] [PubMed] [Google Scholar]
  • 6.Kyndi M, Sørensen FB, Knudsen H, et al. Estrogen receptor, progesterone receptor, HER-2, and response to postmastectomy radiotherapy in high-risk breast cancer: the Danish Breast Cancer Cooperative Group. J Clin Oncol. 2008;26(9):1419–1426. doi: 10.1200/JCO.2007.14.5565. [DOI] [PubMed] [Google Scholar]
  • 7.Gabos Z, Thoms J, Ghosh S, et al. The association between biological subtype and locoregional recurrence in newly diagnosed breast cancer. Breast Cancer Res Treat. 2010;124(1):187–194. doi: 10.1007/s10549-010-1135-1. [DOI] [PubMed] [Google Scholar]
  • 8.Metzger-Filho O, Sun Z, Viale G, et al. Patterns of recurrence and outcome according to breast cancer subtypes in lymph node-negative disease: Results from international breast cancer study group trials VIII and IX. J Clin Oncol. 2013;31(25):3083–3090. doi: 10.1200/JCO.2012.46.1574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Arvold ND, Taghian AG, Niemierko A, et al. Age, breast cancer subtype approximation, and local recurrence after breast-conserving therapy. J Clin Oncol. 2011;29(29):3885–3891. doi: 10.1200/JCO.2011.36.1105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Haffty BG, Yang Q, Reiss M, et al. Locoregional relapse and distant metastasis in conservatively managed triple negative early-stage breast cancer. J Clin Oncol. 2006;24(36):5652–5657. doi: 10.1200/JCO.2006.06.5664. [DOI] [PubMed] [Google Scholar]
  • 11.Arpino G, Generali D, Sapino A, et al. Corrigendum to “Gene expression profiling in breast cancer: A clinical perspective” [The Breast 22 (2013) 109–120] Breast. 2016;25(2):86. doi: 10.1016/j.breast.2013.01.016. [DOI] [PubMed] [Google Scholar]
  • 12.Reis-Filho JS, Pusztai L. Gene expression profiling in breast cancer: Classification, prognostication, and prediction. Lancet. 2011;378(9805):1812–1823. doi: 10.1016/S0140-6736(11)61539-0. [DOI] [PubMed] [Google Scholar]
  • 13.ASCO Guidelines on Breast Cancer. https://www.asco.org/practice-guidelines/quality-guidelines/guidelines/breast-cancer. Accessed January 17, 2017.
  • 14.Gradishar WJ, Robert CH, Anderson BO, et al. NCCN Guidelines Version 1.2016 Breast Cancer Panel Members. Natl Compr Cancer Netw. 2016 [Google Scholar]
  • 15.Goldhirsch A, Winer EP, Coates AS, et al. Personalizing the treatment of women with early breast cancer: Highlights of the st gallen international expert consensus on the primary therapy of early breast Cancer 2013. Ann Oncol. 2013;24(9):2206–2223. doi: 10.1093/annonc/mdt303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Cheang MC, Martin M, Nielsen TO, et al. Defining breast cancer intrinsic subtypes by quantitative receptor expression. Oncologist. 2015;20(5):474–482. doi: 10.1634/theoncologist.2014-0372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gerdes J, Schwab U, Lemke H, Stein H. Production of a mouse monoclonal antibody reactive with a human nuclear antigen associated with cell proliferation. Int J cancer. 1983;31(1):13–20. doi: 10.1002/ijc.2910310104. [DOI] [PubMed] [Google Scholar]
  • 18.Hennigs A, Riedel F, Gondos A, et al. Prognosis of breast cancer molecular subtypes in routine clinical care: A large prospective cohort study. BMC Cancer. 2016;16(1):734. doi: 10.1186/s12885-016-2766-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Li Z, Hu P, Tu J, Yu N. Luminal B breast cancer : patterns of recurrence and clinical outcome. Oncotarget. 2016;7(40):1–10. doi: 10.18632/oncotarget.11344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ehinger A, Malmström P, Bendahl P-O, et al. Histological grade provides significant prognostic information in addition to breast cancer subtypes defined according to St Gallen 2013. Acta Oncol (Madr) 2017;56(1):68–74. doi: 10.1080/0284186X.2016.1237778. [DOI] [PubMed] [Google Scholar]
  • 21.García Fernández A, Chabrera C, García Font M, et al. Differential patterns of recurrence and specific survival between luminal A and luminal B breast cancer according to recent changes in the 2013 St Gallen immunohistochemical classification. Clin Transl Oncol. 2015;17(3):238–246. doi: 10.1007/s12094-014-1220-8. [DOI] [PubMed] [Google Scholar]
  • 22.Freedman GM, Fowble BL. Local recurrence after mastectomy or breast-conserving surgery and radiation. Oncology (Williston Park) 2000;14(11):1561–1581. 1582, 1582–1584. [PubMed] [Google Scholar]
  • 23.Magee B, Swindell R, Harris M, Banerjee SS. Prognostic factors for breast recurrence after conservative breast surgery and radiotherapy: Results from a randomised trial. Radiother Oncol. 1996;39(3):223–227. doi: 10.1016/0167-8140(96)01747-1. [DOI] [PubMed] [Google Scholar]
  • 24.Bouvet M, Babiera GV, Tucker SL, McNeese MD, Singletary SE. Does Breast Conservation Therapy in Young Women with Breast Cancer Adversely Affect Local Disease Control and Survival Rate? The M. D. Anderson Cancer Center Experience. Breast J. 1997;3(4):169–175. [Google Scholar]
  • 25.Van Dongen JA, Voogd AC, Fentiman IS, et al. Long-term results of a randomized trial comparing breast-conserving therapy with mastectomy: European organization for research and treatment of cancer 10801 trial. J Natl Cancer Inst. 2000;92(14):1143–1150. doi: 10.1093/jnci/92.14.1143. [DOI] [PubMed] [Google Scholar]
  • 26.Jager JJ, Volovics L, Schouten LJ, et al. Loco–regional recurrences after mastectomy in breast cancer: prognostic factors and implications for postoperative irradiation. Radiother Oncol. 1999;50(3):267–275. doi: 10.1016/s0167-8140(98)00118-2. [DOI] [PubMed] [Google Scholar]
  • 27.Obedian E, Haffty BG. Negative margin status improves local control in conservatively managed breast cancer patients. Cancer J Sci Am. 2000;6(1):28–33. [PubMed] [Google Scholar]
  • 28.Touboul E, Buffat L, Belkacémi Y, et al. Local recurrences and distant metastases after breast-conserving surgery and radiation therapy for early breast cancer. Int J Radiat Oncol Biol Phys. 1999;43(1):25–38. doi: 10.1016/s0360-3016(98)00365-4. [DOI] [PubMed] [Google Scholar]
  • 29.Pisansky TM, Ingle JN, Schaid DJ, et al. Patterns of tumor relapse following mastectomy and adjuvant systemic therapy in patients with axillary lymph node-positive breast cancer. Impact of clinical, histopathologic, and flow cytometric factors. Cancer. 1993;72(4):1247–1260. doi: 10.1002/1097-0142(19930815)72:4<1247::aid-cncr2820720418>3.0.co;2-s. [DOI] [PubMed] [Google Scholar]
  • 30.O’Rourke S, Galea MH, Morgan D, et al. Local recurrence after simple mastectomy. Br J Surg. 1994;81(3):386–389. doi: 10.1002/bjs.1800810321. [DOI] [PubMed] [Google Scholar]
  • 31.Voogd AC, Peterse JL, Crommelin MA, et al. Histological determinants for different types of local recurrence after breast-conserving therapy of invasive breast cancer. Eur J Cancer. 1999;35(13):1828–1837. doi: 10.1016/s0959-8049(99)00220-8. [DOI] [PubMed] [Google Scholar]
  • 32.Wilson LD, Beinfield M, McKhann CF, Haffty BG. Conservative surgery and radiation in the treatment of synchronous ipsilateral breast cancers. Cancer. 1993;72(1):137–142. doi: 10.1002/1097-0142(19930701)72:1<137::aid-cncr2820720126>3.0.co;2-e. [DOI] [PubMed] [Google Scholar]
  • 33.Borger J, Kemperman H, Hart A, Peterse H, Van Dongen J, Bartelink H. Risk Factors in Breast-Conservation Therapy. J Clin Oncol. 1994;12:653–660. doi: 10.1200/JCO.1994.12.4.653. [DOI] [PubMed] [Google Scholar]
  • 34.Recht A, Gray R, Davidson NE, et al. Locoregional failure 10 years after mastectomy and adjuvant chemotherapy with or without tamoxifen without irradiation: Experience of the Eastern Cooperative Oncology Group. J Clin Oncol. 1999;17(6):1689–1700. doi: 10.1200/JCO.1999.17.6.1689. [DOI] [PubMed] [Google Scholar]
  • 35.Harris JR, Schnitt SJ, Park CC, et al. Outcome at 8 Years After Breast-Conserving Surgery and Radiation Therapy for Invasive Breast Cancer: Influence of Margin Status and Systemic Therapy on Local Recurrence. J Clin Oncol. 2000;18(8):1668–1675. doi: 10.1200/JCO.2000.18.8.1668. [DOI] [PubMed] [Google Scholar]
  • 36.Fisher B, Dignam J, Bryant J, et al. Five versus more than five years of tamoxifen therapy for breast cancer patients with negative lymph nodes and estrogen receptor-positive tumors. J Natl Cancer Inst. 1996;88(21):1529–1542. doi: 10.1093/jnci/88.21.1529. [DOI] [PubMed] [Google Scholar]
  • 37.Fisher B, Anderson S, Redmond CK, Wolmark N, Wickerham DL, Cronin WM. Reanalysis and results after 12 years of follow-up in a randomized clinical trial comparing total mastectomy with lumpectomy with or without irradiation in the treatment of breast cancer. N Engl J Med. 1995;333(22):1456–1461. doi: 10.1056/NEJM199511303332203. [DOI] [PubMed] [Google Scholar]
  • 38.Fisher B, Anderson S, Bryant J, et al. Twenty-Year Follow-up of a Randomized Trial Comparing Total Mastectomy, Lumpectomy, and Lumpectomy plus Irradiation for the Treatment of Invasive Breast Cancer. N Engl J Med. 2002;347(16):1233–1241. doi: 10.1056/NEJMoa022152. [DOI] [PubMed] [Google Scholar]
  • 39.Overgaard M, Jensen M-B, Overgaard J, et al. Postoperative radiotherapy in high-risk postmenopausal breast-cancer patients given adjuvant tamoxifen: Danish Breast Cancer Cooperative Group DBCG 82c randomised trial. Lancet. 1999;353(9165):1641–1648. doi: 10.1016/S0140-6736(98)09201-0. [DOI] [PubMed] [Google Scholar]
  • 40.Ragaz J, Olivotto IA, Spinelli JJ, et al. Locoregional Radiation Therapy in Patients With High-Risk Breast Cancer Receiving Adjuvant Chemotherapy: 20-Year Results of the British Columbia Randomized Trial. JNCI J Natl Cancer Inst. 2005;97(2):116–126. doi: 10.1093/jnci/djh297. [DOI] [PubMed] [Google Scholar]
  • 41.Abe O, Abe R, Enomoto K, et al. Effects of radiotherapy and of differences in the extent of surgery for early breast cancer on local recurrence and 15-year survival: An overview of the randomised trials. Lancet. 2005;366(9503):2087–2106. doi: 10.1016/S0140-6736(05)67887-7. [DOI] [PubMed] [Google Scholar]
  • 42.Early Breast Cancer Trialists’ Collaborative Group (EBCTCG) Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005;365(9472):1687–1717. doi: 10.1016/S0140-6736(05)66544-0. [DOI] [PubMed] [Google Scholar]
  • 43.Early Breast Cancer Trialists’ Collaborative Group (EBCTCG) EBCTCG. Davies C, Godwin J, et al. Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: Patient-level meta-analysis of randomised trials. Lancet. 2011;378(9793):771–784. doi: 10.1016/S0140-6736(11)60993-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Colleoni M, Viale G, Zahrieh D, et al. Chemotherapy Is More Effective in Patients with Breast Cancer Not Expressing Steroid Hormone Receptors: A Study of Preoperative Treatment. Clin Cancer Res. 2004;10(19):6622–6628. doi: 10.1158/1078-0432.CCR-04-0380. [DOI] [PubMed] [Google Scholar]
  • 45.Ring A, Smith I, Ashley S, Fulford L, Lakhani S. Oestrogen receptor status, pathological complete response and prognosis in patients receiving neoadjuvant chemotherapy for early breast cancer. Br J Cancer. 2004;91(12):2012–2017. doi: 10.1038/sj.bjc.6602235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Lange CA. Challenges to defining a role for progesterone in breast cancer. Steroids. 2008;73(9–10):914–921. doi: 10.1016/j.steroids.2007.12.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Chlebowski RT, H S. Influence of estrogen plus progestin on breast cancer and mammography in healthy postmenopausal women: The women’s health initiative randomized trial. JAMA. 2003;289(24):3243–3253. doi: 10.1001/jama.289.24.3243. [DOI] [PubMed] [Google Scholar]
  • 48.Clarke RB. Steroid receptors and proliferation in the human breast. Steroids. 2003;68:789–794. doi: 10.1016/s0039-128x(03)00122-3. [DOI] [PubMed] [Google Scholar]
  • 49.Pichon M-F, Pallud C, Brunet M, Milgrom E. Relationship of Presence of ProgesteroneReceptors to Prognosis in Early Breast Cancer. CANCER Res. 1980 Sep;40:3357–3360. [PubMed] [Google Scholar]
  • 50.Mohsin SK, Weiss H, Havighurst T, et al. Progesterone receptor by immunohistochemistry and clinical outcome in breast cancer: a validation study. Mod Pathol. 2004;17(12):1545–1554. doi: 10.1038/modpathol.3800229. [DOI] [PubMed] [Google Scholar]
  • 51.Bardou VJ, Arpino G, Elledge RM, Osborne CK, Clark GM. Progesterone receptor status significantly improves outcome prediction over estrogen receptor status alone for adjuvant endocrine therapy in two large breast cancer databases. J Clin Oncol. 2003;21(10):1973–1979. doi: 10.1200/JCO.2003.09.099. [DOI] [PubMed] [Google Scholar]
  • 52.Harvey JM, Clark GM, Osborne CK, Allred DC. Estrogen receptor status by immunohistochemistry is superior to the ligand-binding assay for predicting response to adjuvant endocrine therapy in breast cancer. J Clin Oncol. 1999;17(5):1474–1481. doi: 10.1200/JCO.1999.17.5.1474. [DOI] [PubMed] [Google Scholar]
  • 53.Schiff R, Osborn CK, F S. Clinical Aspects of Estrogen and Progesterone Receptors. 2012 [Google Scholar]
  • 54.Hammond MEH, Hayes DF, Dowsett M, et al. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. Arch Pathol Lab Med. 2010;134(7):e48–72. doi: 10.5858/134.7.e48. [DOI] [PubMed] [Google Scholar]
  • 55.Harris GC, Denley HE, Pinder SE, et al. Correlation of histologic prognostic factors in core biopsies and therapeutic excisions of invasive breast carcinoma. Am J Surg Pathol. 2003;27(1):11–15. doi: 10.1097/00000478-200301000-00002. [DOI] [PubMed] [Google Scholar]
  • 56.Hamilton PW, Bankhead P, Wang Y, et al. Digital pathology and image analysis in tissue biomarker research. Methods. 2014;70(1):59–73. doi: 10.1016/j.ymeth.2014.06.015. [DOI] [PubMed] [Google Scholar]
  • 57.Stålhammar G, Fuentes Martinez N, Lippert M, et al. Digital image analysis outperforms manual biomarker assessment in breast cancer. Mod Pathol. 2016;2(4):1–12. doi: 10.1038/modpathol.2016.34. [DOI] [PubMed] [Google Scholar]
  • 58.Lakhani SR, Ellis IO, Schnitt SJ, et al. WHO Classification of Tumours of the Breast. 2012 [Google Scholar]
  • 59.Farmer P, Bonnefoi H, Becette V, et al. Identification of molecular apocrine breast tumours by microarray analysis. Oncogene. 2005;24(29):4660–4671. doi: 10.1038/sj.onc.1208561. [DOI] [PubMed] [Google Scholar]
  • 60.Vera-Badillo FE, Templeton AJ, De Gouveia P, et al. Androgen receptor expression and outcomes in early breast cancer: A systematic review and meta-analysis. J Natl Cancer Inst. 2014;106(1):djt319–djt319. doi: 10.1093/jnci/djt319. [DOI] [PubMed] [Google Scholar]
  • 61.Lehmann-Che J, Hamy A-S, Porcher R, et al. Molecular apocrine breast cancers are aggressive estrogen receptor negative tumors overexpressing either HER2 or GCDFP15. Breast Cancer Res. 2013;15(3):R37. doi: 10.1186/bcr3421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Wieduwilt MJ, Moasser MM. The epidermal growth factor receptor family: Biology driving targeted therapeutics. Cell Mol Life Sci. 2008;65(10):1566–1584. doi: 10.1007/s00018-008-7440-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science. 1987;235(4785):177–182. doi: 10.1126/science.3798106. [DOI] [PubMed] [Google Scholar]
  • 64.Pegram MD, Konecny G, Slamon DJ. The molecular and cellular biology of HER2/neu gene amplification/overexpression and the clinical development of herceptin (trastuzumab) therapy for breast cancer. Cancer Treat Res. 2000;103:57–75. doi: 10.1007/978-1-4757-3147-7_4. [DOI] [PubMed] [Google Scholar]
  • 65.Wolff A, Hammond M, Hicks D, et al. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. J Clin Oncol. 2013;31(31):3997–4013. doi: 10.1200/JCO.2013.50.9984. [DOI] [PubMed] [Google Scholar]
  • 66.Gerdes J, Li L, Schlueter C, et al. Immunobiochemical and molecular biologic characterization of the cell proliferation-associated nuclear antigen that is defined by monoclonal antibody Ki-67. Am J Pathol. 1991;138(4):867–873. [PMC free article] [PubMed] [Google Scholar]
  • 67.Trihia H, Murray S, Price K, et al. Ki-67 expression in breast carcinoma: its association with grading systems, clinical parameters, and other prognostic factors–a surrogate marker? Cancer. 2003;97(5):1321–1331. doi: 10.1002/cncr.11188. [DOI] [PubMed] [Google Scholar]
  • 68.de Azambuja E, Cardoso F, de Castro G, et al. Ki-67 as prognostic marker in early breast cancer: a meta-analysis of published studies involving 12,155 patients. Br J Cancer. 2007;96(10):1504–1513. doi: 10.1038/sj.bjc.6603756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Stuart-Harris R, Caldas C, Pinder SE, Pharoah P. Proliferation markers and survival in early breast cancer: a systematic review and meta-analysis of 85 studies in 32,825 patients. Breast. 2008;17(4):323–334. doi: 10.1016/j.breast.2008.02.002. [DOI] [PubMed] [Google Scholar]
  • 70.Jonat W, Arnold N. Is the Ki-67 labelling index ready for clinical use? Ann Oncol. 2011;22(3):500–502. doi: 10.1093/annonc/mdq732. [DOI] [PubMed] [Google Scholar]
  • 71.DeCensi A, Guerrieri-Gonzaga A, Gandini S, et al. Prognostic significance of Ki-67 labeling index after short-term presurgical tamoxifen in women with ER-positive breast cancer. Ann Oncol. 2011;22(3):582–587. doi: 10.1093/annonc/mdq427. [DOI] [PubMed] [Google Scholar]
  • 72.Keshgegian AA, Cnaan A. Proliferation markers in breast carcinoma. Mitotic figure count, S-phase fraction, proliferating cell nuclear antigen, Ki-67 and MIB-1. Am J Clin Pathol. 1995;104(1):42–49. doi: 10.1093/ajcp/104.1.42. [DOI] [PubMed] [Google Scholar]
  • 73.Clahsen PC, van de Velde CJ, Duval C, et al. The utility of mitotic index, oestrogen receptor and Ki-67 measurements in the creation of novel prognostic indices for node-negative breast cancer. Eur J Surg Oncol. 1999;25(4):356–363. doi: 10.1053/ejso.1999.0657. [DOI] [PubMed] [Google Scholar]
  • 74.Cheang MCU, Chia SK, Voduc D, et al. Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J Natl Cancer Inst. 2009;101(10):736–750. doi: 10.1093/jnci/djp082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Inwald EC, Klinkhammer-Schalke M, Hofstädter F, et al. Ki-67 is a prognostic parameter in breast cancer patients: results of a large population-based cohort of a cancer registry. Breast Cancer Res Treat. 2013;139(2):539–552. doi: 10.1007/s10549-013-2560-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Dowsett M, Smith IE, Ebbs SR, et al. Prognostic Value of Ki67 Expression After Short-Term Presurgical Endocrine Therapy for Primary Breast Cancer. JNCI J Natl Cancer Inst. 2007;99(2):167–170. doi: 10.1093/jnci/djk020. [DOI] [PubMed] [Google Scholar]
  • 77.Nishimura R, Osako T, Okumura Y, Hayashi M, Arima N. Clinical significance of Ki-67 in neoadjuvant chemotherapy for primary breast cancer as a predictor for chemosensitivity and for prognosis. Breast Cancer. 2010;17(4):269–275. doi: 10.1007/s12282-009-0161-5. [DOI] [PubMed] [Google Scholar]
  • 78.Vuong D, Simpson PT, Green B, Cummings MC, Lakhani SR. Molecular classification of breast cancer. Virchows Arch. 2014;465(1):1–14. doi: 10.1007/s00428-014-1593-7. [DOI] [PubMed] [Google Scholar]
  • 79.Carey LA, Perou CM, Livasy CA, et al. Race, breast cancer subtypes, and survival in the Carolina Breast Cancer Study. JAMA. 2006;295(21):2492–2502. doi: 10.1001/jama.295.21.2492. [DOI] [PubMed] [Google Scholar]
  • 80.Subik K, Lee JF, Baxter L, et al. The expression patterns of ER, PR, HER2, CK5/6, EGFR, KI-67 and AR by immunohistochemical analysis in breast cancer cell lines. Breast Cancer Basic Clin Res. 2010;4(1):35–41. [PMC free article] [PubMed] [Google Scholar]
  • 81.Prat A, Cheang MCU, Martín M, et al. Prognostic significance of progesterone receptor-positive tumor cells within immunohistochemically defined luminal A breast cancer. J Clin Oncol. 2013;31(2):203–209. doi: 10.1200/JCO.2012.43.4134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Sorlie T, Tibshirani R, Parker J, et al. Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci U S A. 2003;100(14):8418–8423. doi: 10.1073/pnas.0932692100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Koboldt DC, Fulton RS, McLellan MD, et al. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490(7418):61–70. doi: 10.1038/nature11412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Prat A, Carey LA, Adamo B, et al. Molecular Features and Survival Outcomes of the Intrinsic Subtypes Within HER2-Positive Breast Cancer. JNCI J Natl Cancer Inst. 2014;106(8):dju152–dju152. doi: 10.1093/jnci/dju152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Staaf J, Ringnér M, Vallon-Christersson J, et al. Identification of subtypes in human epidermal growth factor receptor 2–positive breast cancer reveals a gene signature prognostic of outcome. J Clin Oncol. 2010;28(11):1813–1820. doi: 10.1200/JCO.2009.22.8775. [DOI] [PubMed] [Google Scholar]
  • 86.Fountzilas G, Dafni U, Bobos M, et al. Differential Response of Immunohistochemically Defined Breast Cancer Subtypes to Anthracycline-Based Adjuvant Chemotherapy with or without Paclitaxel. In: Aziz SA, editor. PLoS One. 6. Vol. 7. 2012. p. e37946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Kneubil MC, Brollo J, Botteri E, et al. Breast cancer subtype approximations and loco-regional recurrence after immediate breast reconstruction. Eur J Surg Oncol. 2013;39(3):260–265. doi: 10.1016/j.ejso.2012.12.004. [DOI] [PubMed] [Google Scholar]
  • 88.Vici P, Pizzuti L, Natoli C, et al. Triple positive breast cancer: A distinct subtype? Cancer Treat Rev. 2015;41(2):69–76. doi: 10.1016/j.ctrv.2014.12.005. [DOI] [PubMed] [Google Scholar]
  • 89.De Laurentiis M, Arpino G, Massarelli E, et al. A meta-analysis on the interaction between HER-2 expression and response to endocrine treatment in advanced breast cancer. Clin Cancer Res. 2005;11(13):4741–4748. doi: 10.1158/1078-0432.CCR-04-2569. [DOI] [PubMed] [Google Scholar]
  • 90.Dowsett M, Allred C, Knox J, et al. Relationship Between Quantitative Estrogen and Progesterone Receptor Expression and Human Epidermal Growth Factor Receptor 2 (HER-2) Status With Recurrence in the Arimidex, Tamoxifen, Alone or in Combination Trial. J Clin Oncol. 2008;26(7):1059–1065. doi: 10.1200/JCO.2007.12.9437. [DOI] [PubMed] [Google Scholar]
  • 91.Osborne CK, Schiff R. Mechanisms of Endocrine Resistance in Breast Cancer. Annu Rev Med. 2011;62(1):233–247. doi: 10.1146/annurev-med-070909-182917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Shou J, Massarweh S, Osborne CK, et al. Mechanisms of tamoxifen resistance: increased estrogen receptor-HER2/neu cross-talk in ER/HER2-positive breast cancer. J Natl Cancer Inst. 2004;96(12):926–935. doi: 10.1093/jnci/djh166. [DOI] [PubMed] [Google Scholar]
  • 93.Alqaisi A, Chen L, Romond E, et al. Impact of estrogen receptor (ER) and human epidermal growth factor receptor-2 (HER2) co-expression on breast cancer disease characteristics: implications for tumor biology and research. Breast Cancer Res Treat. 2014;148(2):437–444. doi: 10.1007/s10549-014-3145-x. [DOI] [PubMed] [Google Scholar]
  • 94.Lehmann BD, Bauer JA, Chen X, et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest. 2011;121(7):2750–2767. doi: 10.1172/JCI45014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Reis-Filho JS, Weigelt B, Fumagalli D, Sotiriou C. Molecular profiling: moving away from tumor philately. Sci Transl Med. 2010;2(47):47ps43. doi: 10.1126/scitranslmed.3001329. [DOI] [PubMed] [Google Scholar]
  • 96.Weigelt B, Baehner FL, Reis-Filho JS. The contribution of gene expression profiling to breast cancer classification, prognostication and prediction: a retrospective of the last decade. J Pathol. 2010;220(2):263–280. doi: 10.1002/path.2648. [DOI] [PubMed] [Google Scholar]
  • 97.Desmedt C, Sotiriou C. Proliferation: the most prominent predictor of clinical outcome in breast cancer. Cell Cycle. 2006;5(19):2198–2202. doi: 10.4161/cc.5.19.3254. [DOI] [PubMed] [Google Scholar]
  • 98.Pusztai L, Broglio K, Andre F, Symmans WF, Hess KR, Hortobagyi GN. Effect of molecular disease subsets on disease-free survival in randomized adjuvant chemotherapy trials for estrogen receptor-positive breast cancer. J Clin Oncol. 2008;26(28):4679–4683. doi: 10.1200/JCO.2008.17.2544. [DOI] [PubMed] [Google Scholar]
  • 99.Wirapati P, Sotiriou C, Kunkel S, et al. Meta-analysis of gene expression profiles in breast cancer: toward a unified understanding of breast cancer subtyping and prognosis signatures. Breast Cancer Res. 2008;10(4):R65. doi: 10.1186/bcr2124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Iwamoto T, Bianchini G, Booser D, et al. Gene Pathways Associated With Prognosis and Chemotherapy Sensitivity in Molecular Subtypes of Breast Cancer. JNCI J Natl Cancer Inst. 2011;103(3):264–272. doi: 10.1093/jnci/djq524. [DOI] [PubMed] [Google Scholar]
  • 101.Buyse M, Loi S, van’t Veer L, et al. Validation and Clinical Utility of a 70-Gene Prognostic Signature for Women With Node-Negative Breast Cancer. JNCI J Natl Cancer Inst. 2006;98(17):1183–1192. doi: 10.1093/jnci/djj329. [DOI] [PubMed] [Google Scholar]
  • 102.Desmedt C, Piette F, Loi S, et al. Strong time dependence of the 76-gene prognostic signature for node-negative breast cancer patients in the TRANSBIG multicenter independent validation series. Clin Cancer Res. 2007;13(11):3207–3214. doi: 10.1158/1078-0432.CCR-06-2765. [DOI] [PubMed] [Google Scholar]
  • 103.Drukker CA, Bueno-de-Mesquita JM, Retèl VP, et al. A prospective evaluation of a breast cancer prognosis signature in the observational RASTER study. Int J cancer. 2013;133(4):929–936. doi: 10.1002/ijc.28082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Cardoso F, van’t Veer LJ, Bogaerts J, et al. 70-Gene Signature as an Aid to Treatment Decisions in Early-Stage Breast Cancer. N Engl J Med. 2016;375(8):717–729. doi: 10.1056/NEJMoa1602253. [DOI] [PubMed] [Google Scholar]
  • 105.Paik S, Shak S, Tang G, et al. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med. 2004;351(27):2817–2826. doi: 10.1056/NEJMoa041588. [DOI] [PubMed] [Google Scholar]
  • 106.Dowsett M, Cuzick J, Wale C, et al. Prediction of Risk of Distant Recurrence Using the 21-Gene Recurrence Score in Node-Negative and Node-Positive Postmenopausal Patients With Breast Cancer Treated With Anastrozole or Tamoxifen: A TransATAC Study. J Clin Oncol. 2010;28(11):1829–1834. doi: 10.1200/JCO.2009.24.4798. [DOI] [PubMed] [Google Scholar]
  • 107.Albain KS, Barlow WE, Shak S, et al. Prognostic and predictive value of the 21-gene recurrence score assay in postmenopausal women with node-positive, oestrogen-receptor-positive breast cancer on chemotherapy: a retrospective analysis of a randomised trial. Lancet Oncol. 2010;11(1):55–65. doi: 10.1016/S1470-2045(09)70314-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Harris LN, Ismaila N, McShane LM, et al. Use of Biomarkers to Guide Decisions on Adjuvant Systemic Therapy for Women With Early-Stage Invasive Breast Cancer: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol. 2016;34(10):1134–1150. doi: 10.1200/JCO.2015.65.2289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Tang G, Shak S, Paik S, et al. Comparison of the prognostic and predictive utilities of the 21-gene Recurrence Score assay and Adjuvant! for women with node-negative, ER-positive breast cancer: results from NSABP B-14 and NSABP B-20. Breast Cancer Res Treat. 2011;127(1):133–142. doi: 10.1007/s10549-010-1331-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Mamounas EP, Tang G, Fisher B, et al. Association between the 21-gene recurrence score assay and risk of locoregional recurrence in node-negative, estrogen receptor-positive breast cancer: results from NSABP B-14 and NSABP B-20. J Clin Oncol. 2010;28(10):1677–1683. doi: 10.1200/JCO.2009.23.7610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Habel LA, Shak S, Jacobs MK, et al. A population-based study of tumor gene expression and risk of breast cancer death among lymph node-negative patients. Breast Cancer Res. 2006;8(3):R25. doi: 10.1186/bcr1412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Elkhuizen MD PH, van de Vijver MD PhD MJ, Hermans PhD J, Zonderland MD HM, van de Velde MD PhD CJ, Leer MD PhD J-WH. Local Recurrence After Breast-Conserving Therapy for Invasive Breast Cancer: High Incidence in Young Patients and Association with Poor Survival. Int J Radiat Oncol. 1998;40(4):859–867. doi: 10.1016/s0360-3016(97)00917-6. [DOI] [PubMed] [Google Scholar]
  • 113.Fowble BL, Schultz DJ, Overmoyer B, et al. The influence of young age on outcome in early stage breast cancer. Int J Radiat Oncol Biol Phys. 1994;30(1):23–33. doi: 10.1016/0360-3016(94)90515-0. [DOI] [PubMed] [Google Scholar]
  • 114.van Werkhoven E, Hart G, van Tinteren H, et al. Nomogram to predict ipsilateral breast relapse based on pathology review from the EORTC 22881-10882 boost versus no boost trial. Radiother Oncol. 2011;100(1):101–107. doi: 10.1016/j.radonc.2011.07.004. [DOI] [PubMed] [Google Scholar]
  • 115.Moran MS, Schnitt SJ, Giuliano AE, et al. Society of Surgical Oncology–American Society for Radiation Oncology Consensus Guideline on Margins for Breast-Conserving Surgery With Whole-Breast Irradiation in Stages I and II Invasive Breast Cancer. Int J Radiat Oncol. 2014;88(3):553–564. doi: 10.1016/j.ijrobp.2013.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Jones HA, Antonini N, Hart AAM, et al. Impact of pathological characteristics on local relapse after breast-conserving therapy: a subgroup analysis of the EORTC boost versus no boost trial. J Clin Oncol. 2009;27(30):4939–4947. doi: 10.1200/JCO.2008.21.5764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Gage I, Schnitt SJ, Nixon AJ, et al. Pathologic margin involvement and the risk of recurrence in patients treated with breast-conserving therapy. Cancer. 1996;78(9):1921–1928. doi: 10.1002/(sici)1097-0142(19961101)78:9<1921::aid-cncr12>3.0.co;2-#. [DOI] [PubMed] [Google Scholar]
  • 118.Jagsi R, Raad RA, Goldberg S, et al. Locoregional recurrence rates and prognostic factors for failure in node-negative patients treated with mastectomy: implications for postmastectomy radiation. Int J Radiat Oncol Biol Phys. 2005;62(4):1035–1039. doi: 10.1016/j.ijrobp.2004.12.014. [DOI] [PubMed] [Google Scholar]
  • 119.Nielsen HM, Overgaard M, Grau C, Jensen AR, Overgaard J. Loco-regional recurrence after mastectomy in high-risk breast cancer–risk and prognosis. An analysis of patients from the DBCG 82 b&c randomization trials. Radiother Oncol. 2006;79(2):147–155. doi: 10.1016/j.radonc.2006.04.006. [DOI] [PubMed] [Google Scholar]
  • 120.Recht A, Comen EA, Fine RE, et al. Postmastectomy Radiotherapy: An American Society of Clinical Oncology, American Society for Radiation Oncology, and Society of Surgical Oncology Focused Guideline Update. Pract Radiat Oncol. 2016;6(6):e219–e234. doi: 10.1016/j.prro.2016.08.009. [DOI] [PubMed] [Google Scholar]
  • 121.Abdulkarim BS, Cuartero J, Hanson J, Deschênes J, Lesniak D, Sabri S. Increased risk of locoregional recurrence for women with T1-2N0 triple-negative breast cancer treated with modified radical mastectomy without adjuvant radiation therapy compared with breast-conserving therapy. J Clin Oncol. 2011;29(21):2852–2858. doi: 10.1200/JCO.2010.33.4714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Clarke M, Collins R, Darby S, et al. Effects of radiotherapy and of differences in the extent of surgery for early breast cancer on local recurrence and 15-year survival: an overview of the randomised trials. Lancet (London, England) 2005;366(9503):2087–2106. doi: 10.1016/S0140-6736(05)67887-7. [DOI] [PubMed] [Google Scholar]
  • 123.Su Y-L, Li S-H, Chen Y-Y, et al. Post-mastectomy radiotherapy benefits subgroups of breast cancer patients with T1-2 tumor and 1–3 axillary lymph node(s) metastasis. Radiol Oncol. 2014;48(3):314–322. doi: 10.2478/raon-2013-0085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Metcalfe K, Lynch HT, Ghadirian P, et al. Contralateral Breast Cancer in BRCA1 and BRCA2 Mutation Carriers. J Clin Oncol. 2004;22(12):2328–2335. doi: 10.1200/JCO.2004.04.033. [DOI] [PubMed] [Google Scholar]
  • 125.Garcia-Etienne CA, Barile M, Gentilini OD, et al. Breast-conserving surgery in BRCA1/2 mutation carriers: are we approaching an answer? Ann Surg Oncol. 2009;16(12):3380–3387. doi: 10.1245/s10434-009-0638-7. [DOI] [PubMed] [Google Scholar]
  • 126.Tsoutsou PG, Vozenin M-C, Durham A-D, Bourhis J. How could breast cancer molecular features contribute to locoregional treatment decision making? Crit Rev Oncol Hematol. 2017;110:43–48. doi: 10.1016/j.critrevonc.2016.12.006. [DOI] [PubMed] [Google Scholar]
  • 127.Sanpaolo P, Barbieri V, Genovesi D. Prognostic value of breast cancer subtypes on breast cancer specific survival, distant metastases and local relapse rates in conservatively managed early stage breast cancer: A retrospective clinical study. Eur J Surg Oncol. 2011;37(10):876–882. doi: 10.1016/j.ejso.2011.07.001. [DOI] [PubMed] [Google Scholar]
  • 128.García Fernández A, Chabrera C, García Font M, et al. Mortality and recurrence patterns of breast cancer patients diagnosed under a screening programme versus comparable non-screened breast cancer patients from the same population: analytical survey from 2002 to 2012. Tumor Biol. 2014;35(3):1945–1953. doi: 10.1007/s13277-013-1260-7. [DOI] [PubMed] [Google Scholar]
  • 129.Jatoi I, Anderson WF, Jeong J-H, Redmond CK. Breast Cancer Adjuvant Therapy: Time to Consider Its Time-Dependent Effects. J Clin Oncol. 2011;29(17):2301–2304. doi: 10.1200/JCO.2010.32.3550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.van der Leij F, Elkhuizen PHM, Bartelink H, van de Vijver MJ. Predictive Factors for Local Recurrence in Breast Cancer. Semin Radiat Oncol. 2012;22(2):100–107. doi: 10.1016/j.semradonc.2011.12.001. [DOI] [PubMed] [Google Scholar]
  • 131.Haffty BG. Molecular and genetic markers in the local-regional management of breast cancer. Semin Radiat Oncol. 2002;12(4):329–340. doi: 10.1053/srao.2002.35252. [DOI] [PubMed] [Google Scholar]
  • 132.Albert JM, Gonzalez-Angulo AM, Guray M, et al. Estrogen/Progesterone Receptor Negativity and HER2 Positivity Predict Locoregional Recurrence in Patients With T1a,bN0 Breast Cancer. Int J Radiat Oncol. 2010;77(5):1296–1302. doi: 10.1016/j.ijrobp.2009.12.011. [DOI] [PubMed] [Google Scholar]
  • 133.Nguyen PL, Taghian AG, Katz MS, et al. Breast Cancer Subtype Approximated by Estrogen Receptor, Progesterone Receptor, and HER-2 Is Associated With Local and Distant Recurrence After Breast-Conserving Therapy. J Clin Oncol. 2008;26(14):2373–2378. doi: 10.1200/JCO.2007.14.4287. [DOI] [PubMed] [Google Scholar]
  • 134.Lowery AJ, Kell MR, Glynn RW, Kerin MJ, Sweeney KJ. Locoregional recurrence after breast cancer surgery: A systematic review by receptor phenotype. Breast Cancer Res Treat. 2012;133(3):831–841. doi: 10.1007/s10549-011-1891-6. [DOI] [PubMed] [Google Scholar]
  • 135.Romond EH, Perez EA, Bryant J, et al. Trastuzumab plus Adjuvant Chemotherapy for Operable HER2-Positive Breast Cancer. N Engl J Med. 2005;353(16):1673–1684. doi: 10.1056/NEJMoa052122. [DOI] [PubMed] [Google Scholar]
  • 136.Haffty BG, Brown F, Carter D, Flynn S. Evaluation of HER-2 neu oncoprotein expression as a prognostic indicator of local recurrence in conservatively treated breast cancer: a case-control study. Int J Radiat Oncol Biol Phys. 1996;35(4):751–757. doi: 10.1016/0360-3016(96)00150-2. [DOI] [PubMed] [Google Scholar]
  • 137.Elkhuizen PH, Voogd AC, van den Broek LC, et al. Risk factors for local recurrence after breast-conserving therapy for invasive carcinomas: a case-control study of histological factors and alterations in oncogene expression. Int J Radiat Oncol Biol Phys. 1999;45(1):73–83. doi: 10.1016/s0360-3016(99)00158-3. [DOI] [PubMed] [Google Scholar]
  • 138.Pierce LJ, Merino MJ, D’Angelo T, et al. Is c-erb B-2 a predictor for recurrent disease in early stage breast cancer? Int J Radiat Oncol Biol Phys. 1994;28(2):395–403. doi: 10.1016/0360-3016(94)90063-9. [DOI] [PubMed] [Google Scholar]
  • 139.Panoff JE, Hurley J, Takita C, et al. Risk of locoregional recurrence by receptor status in breast cancer patients receiving modern systemic therapy and post-mastectomy radiation. Breast Cancer Res Treat. 2011;128(3):899–906. doi: 10.1007/s10549-011-1495-1. [DOI] [PubMed] [Google Scholar]
  • 140.Yin W, Jiang Y, Shen Z, Shao Z, Lu J. Trastuzumab in the adjuvant treatment of HER2-positive early breast cancer patients: A meta-analysis of published randomized controlled trials. PLoS One. 2011;6(6) doi: 10.1371/journal.pone.0021030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Pietras RJ, Poen JC, Gallardo D, Wongvipat PN, Lee HJ, Slamon DJ. Monoclonal Antibody to HER-2/neuReceptor Modulates Repair of Radiation- induced DNA Damage and Enhances Radiosensitivity of Human Breast Cancer Cells Overexpressing This Oncogene. CANCER Res. 1999;59:1347–1355. [PubMed] [Google Scholar]
  • 142.Fehrenbacher L, Capra AM, Quesenberry CP, Fulton R, Shiraz P, Habel LA. Distant invasive breast cancer recurrence risk in human epidermal growth factor receptor 2-positive T1a and T1b node-negative localized breast cancer diagnosed from 2000 to 2006: a cohort from an integrated health care delivery system. J Clin Oncol. 2014;32(20):2151–2158. doi: 10.1200/JCO.2013.52.0858. [DOI] [PubMed] [Google Scholar]
  • 143.Malone KE, Begg CB, Haile RW, et al. Population-Based Study of the Risk of Second Primary Contralateral Breast Cancer Associated With Carrying a Mutation in BRCA1 or BRCA2. J Clin Oncol. 2010;28(14):2404–2410. doi: 10.1200/JCO.2009.24.2495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Tun N, Villani G, Ong K, Yoe L, Bo Z. Risk of having BRCA1 mutation in high-risk women with triple-negative breast cancer: a meta-analysis. Clin Genet. 2014;85(1):43–48. doi: 10.1111/cge.12270. [DOI] [PubMed] [Google Scholar]
  • 145.Bessonova L, Taylor TH, Mehta RS, Zell JA, Anton-Culver H. Risk of a Second Breast Cancer Associated with Hormone-Receptor and HER2/neu Status of the First Breast Cancer. Cancer Epidemiol Biomarkers Prev. 2011;20(2):389–396. doi: 10.1158/1055-9965.EPI-10-1016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Kumar P, Aggarwal R. An overview of triple-negative breast cancer. Arch Gynecol Obstet. 2016;293(2):247–269. doi: 10.1007/s00404-015-3859-y. [DOI] [PubMed] [Google Scholar]
  • 147.Wang J, Xie X, Wang X, et al. Locoregional and distant recurrences after breast conserving therapy in patients with triple-negative breast cancer: A meta-analysis. Surg Oncol. 2013;22(4):247–255. doi: 10.1016/j.suronc.2013.10.001. [DOI] [PubMed] [Google Scholar]
  • 148.Moran MS. Radiation therapy in the locoregional treatment of triple-negative breast cancer. Lancet Oncol. 2015;16(3):e113–e122. doi: 10.1016/S1470-2045(14)71104-0. [DOI] [PubMed] [Google Scholar]
  • 149.Huang EH, Tucker SL, Strom EA, et al. Predictors of locoregional recurrence in patients with locally advanced breast cancer treated with neoadjuvant chemotherapy, mastectomy, and radiotherapy. Int J Radiat Oncol. 2005;62(2):351–357. doi: 10.1016/j.ijrobp.2004.09.056. [DOI] [PubMed] [Google Scholar]
  • 150.Levy A, Borget I, Bahri M, et al. Loco-regional control after neo-adjuvant chemotherapy and conservative treatment for locally advanced breast cancer patients. Breast J. 2014;20(4):381–387. doi: 10.1111/tbj.12277. [DOI] [PubMed] [Google Scholar]
  • 151.Mamounas EP, Anderson SJ, Dignam JJ, et al. Predictors of locoregional recurrence after neoadjuvant chemotherapy: results from combined analysis of National Surgical Adjuvant Breast and Bowel Project B-18 and B-27. J Clin Oncol. 2012;30(32):3960–3966. doi: 10.1200/JCO.2011.40.8369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.von Minckwitz G, Untch M, Blohmer J-U, et al. Definition and Impact of Pathologic Complete Response on Prognosis After Neoadjuvant Chemotherapy in Various Intrinsic Breast Cancer Subtypes. J Clin Oncol. 30:1796–1804. doi: 10.1200/JCO.2011.38.8595. [DOI] [PubMed] [Google Scholar]
  • 153.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(8):1275–1281. doi: 10.1200/JCO.2007.14.4147. [DOI] [PubMed] [Google Scholar]
  • 154.Swisher SK, Vila J, Tucker SL, et al. Locoregional Control According to Breast Cancer Subtype and Response to Neoadjuvant Chemotherapy in Breast Cancer Patients Undergoing Breast-conserving Therapy. Ann Surg Oncol. 2016;23(3):749–756. doi: 10.1245/s10434-015-4921-5. [DOI] [PubMed] [Google Scholar]
  • 155.Peterson DJ, Truong PT, Sadek BT, et al. Locoregional recurrence and survival outcomes by type of local therapy and trastuzumab use among women with node-negative, HER2-positive breast cancer. Ann Surg Oncol. 2014;21(11):3490–3496. doi: 10.1245/s10434-014-3767-6. [DOI] [PubMed] [Google Scholar]
  • 156.Kiess AP, McArthur HL, Mahoney K, et al. Adjuvant trastuzumab reduces locoregional recurrence in women who receive breast-conservation therapy for lymph node-negative, human epidermal growth factor receptor 2-positive breast cancer. Cancer. 2012;118(8):1982–1988. doi: 10.1002/cncr.26484. [DOI] [PubMed] [Google Scholar]
  • 157.Donker M, van Tienhoven G, Straver ME, et al. Radiotherapy or surgery of the axilla after a positive sentinel node in breast cancer (EORTC 10981–22023 AMAROS): a randomised, multicentre, open-label, phase 3 non-inferiority trial. Lancet Oncol. 2014;15(12):1303–1310. doi: 10.1016/S1470-2045(14)70460-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Giuliano AE, McCall L, Beitsch P, et al. Locoregional Recurrence After Sentinel Lymph Node Dissection With or Without Axillary Dissection in Patients With Sentinel Lymph Node Metastases. Ann Surg. 2010;252(3):423–426. doi: 10.1097/SLA.0b013e3181f08f32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Giuliano AE, Ballman K, McCall L, et al. Locoregional Recurrence After Sentinel Lymph Node Dissection With or Without Axillary Dissection in Patients With Sentinel Lymph Node Metastases. Ann Surg. 2016;264(3):413–420. doi: 10.1097/SLA.0000000000001863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Morrow M. Rethinking the Local Therapy of Breast Cancer: Integration of Biology and Anatomy. Ann Surg Oncol. 2015;22(10):3168–3173. doi: 10.1245/s10434-015-4750-6. [DOI] [PubMed] [Google Scholar]
  • 161.Olivotto IA, Truong PT, Chua B. Postmastectomy Radiation Therapy: Who Needs It? J Clin Oncol. 2004;22(21):4237–4239. doi: 10.1200/JCO.2004.06.974. [DOI] [PubMed] [Google Scholar]
  • 162.Goldhirsch A, Glick JH, Gelber RD, Senn HJ. Meeting highlights: International Consensus Panel on the Treatment of Primary Breast Cancer. J Natl Cancer Inst. 1998;90(21):1601–1608. doi: 10.1093/jnci/90.21.1601. [DOI] [PubMed] [Google Scholar]
  • 163.Jwa E, Shin KH, Lim HW, et al. Identification of Risk Factors for Locoregional Recurrence in Breast Cancer Patients with Nodal Stage N0 and N1: Who Could Benefit from Post-Mastectomy Radiotherapy? In: St-Pierre Y, editor. PLoS One. 12. Vol. 10. 2015. p. e0145463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.The IDEA Study (Individualized Decisions for Endocrine Therapy Alone) https://clinicaltrials.gov/show/NCT02400190.
  • 165.The PRECISION Trial (Profiling Early Breast Cancer for Radiotherapy Omission): A Phase II Study of Breast-Conserving Surgery Without Adjuvant Radiotherapy for Favorable-Risk Breast Cancer. https://clinicaltrials.gov/ct2/show/NCT02653755.
  • 166.A Prospective Cohort Study Evaluating Risk of Local Recurrence Following Breast Conserving Surgery and Endocrine Therapy in Low Risk Luminal A Breast Cancer. Full Text View - ClinicalTrials.gov; https://clinicaltrials.gov/ct2/show/NCT01791829.
  • 167.Radiation Therapy or Standard Therapy in Treating Women With Stage II Breast Cancer Who Have Undergone Mastectomy. https://clinicaltrials.gov/ct2/show/NCT00966888.

RESOURCES