and Karl Reinhard Aigner3
(1)
Department of Surgery, The University of Sydney, Mosman, NSW, Australia
(2)
The Royal Prince Alfred and Sydney Hospitals, Mosman, NSW, Australia
(3)
Department of Surgical Oncology, Medias Clinic Surgical Oncology, Burghausen, Germany
Disclosures
F. M. Delgado MD, PhD: none
R. Conforti MD, PhD: none
M.A. Gil-Delgado MD, PhD: none
D. Khayat Pr, MD, PhD:
Employment or leadership position to disclose: none
Consultant or advisory relationship to disclose: Celgene, Genomic Health, Sofibio, Laval Santé, Equatour.
Stock or other ownership interest: Agenus INC.
12.1 Introduction
12.1.1 Epidemiology
Breast cancer (BC) is a major public health problem for women throughout the world. Breast cancer represents the second most common cancer in the world and is the most frequent cancer among women, with 1.67 million new cases diagnosed in 2012 according to GLOBOCAN data. Breast cancer alone represents 25 % of all cancer cases and 15 % of all cancer deaths among women (521,900 in 2012). More developed countries account for about one-half of all BC cases and 38 % of deaths [1, 2].
In the European Union, BC remains the most frequent cancer in women and the second most frequent cause of cancer death. As a result, 456,000 new cases (29 % of female cancer and 12 % of the total) of invasive breast cancer were diagnosed, and 131,000 women died of breast cancer in 2012 (17 % of female deaths and 7 % of the total) [3, 4]. Nevertheless, since 1990, the death rate from BC has decreased in the United States by 24 %, and similar reductions have been observed in Europe and in other countries [5]. Although BC has traditionally been less common in non-industrialised nations, its incidence in these areas is increasing [1, 2].
This overview refers to BC of women because although BC does rarely occur in men, men with breast cancer generally follow a protocol similar to that of female breast cancer patients, including a combination of surgery, radiation, chemotherapy and anti-hormone therapies [6].
The aetiology of the vast majority of BC cases is unknown. However, numerous risk factors for the disease have been established. These risk factors include female gender, increasing patient age, family history of BC at a young age, early menarche, late menopause, older age at first live childbirth, prolonged hormone replacement therapy, previous exposure to therapeutic chest wall irradiation, benign proliferative breast disease and genetic mutations such as of the BRCA1/BRCA2 genes.
12.1.2 Workup
The starting point of a workup for evaluating breast abnormalities is a complete medical history followed by clinical breast examination. The clinical breast examination should involve palpation of the entire breast with the patient in the upright and supine position and include the axillary region as well as all nodal areas that involve the breasts (i.e. axillary, supraclavicular and internal mammary nodes). Symptoms or positive findings on physical exam can include a palpable lump or mass, asymmetric thickening/nodularity, nipple discharge in the absence of a palpable mass and skin changes such as “peau d’orange”, erythema, nipple excoriation and scaling/eczema [7]. Determination of the locoregional involvement is the keystone in deciding the first treatment. In the Western world, most patients present following an abnormal mammogram. However, around 15 % of women are diagnosed with breast cancer due to the presence of a breast mass not detected by routine mammography, and 30 % presents with a breast mass in the interval between mammograms. It is also important to consider that there is a group of women without access to screening mammography and younger than 45 years who are not undergoing routine screening mammograms and who may present with a breast or axillary mass with or without skin changes [8].
Another key requirement is to detect the signs and symptoms of metastatic breast cancer, which depend on the organs involved. Back or leg pain is a pointer of bone involvement, whilst cough and shortness of breath suggest lung metastases; abdominal pain, nausea and jaundice point to liver involvement.
Mammographic findings of BC include the presence of a soft tissue mass or density and clustered microcalcifications. The most specific feature is a spiculated soft tissue mass, suggesting in 90 % of cases an invasive cancer. Mammographic results should be reported according the Final Assessment Categories established by the American College of Radiology BI-RADS® in which six categories are described which establish a uniform “language” for reporting mammographic findings [9].
Breast ultrasound is often used to distinguish between a benign and a malignant lesion (speculation, hypo-echogenicity, microlobulation, internal calcifications, shadowing, a lesion taller than wide and angular margins) [10]. Subsequently the results are classified according to BI-RADS® categories. Categories 4 and 5 strongly suggest that a biopsy should be performed [11].
If a woman older than 40 years exhibits a nipple discharge but no palpable mass, an evaluation of the features of this discharge must be done first followed by mammography and possibly an ultrasound. A tissue biopsy should be performed for women with an overall BI-RADS® assessment in categories 4–5. In the event that a malignancy is present, the patient should be managed according to the standard procedures [8, 12, 13].
Magnetic resonance imaging (MRI) can also play a role in the diagnostic setting. For patients with BI-RADS® categories 1–3 with skin changes consistent with serious breast disease and negative biopsy of the skin or nipple, breast MRI should be included in the workup. Since a benign skin punch biopsy in a patient with a clinical suspicion of inflammatory BC (IBC) does not rule out malignancy, further evaluation is recommended. There is evidence that certain MRI features may facilitate diagnosis of IBC, and if MRI is used, this must be considered as a complementary examination and does not obviate the need for biopsy [13, 14].
12.1.3 Breast Biopsy
Breast biopsy is recommended if diagnostic mammogram and/or ultrasound findings are suspicious or highly suggestive of malignancy. This biopsy is performed by fine needle aspiration (FNA) which involves the use of a small-bore needle to obtain cytological samples from a breast mass. Advantages of FNA biopsy include its minimally invasive methodology. FNA of non-palpable lesions can be performed under imaging guidance (e.g. ultrasound), although there is evidence to indicate that both core needle biopsy (CNB) and excisional biopsy are more accurate than FNA in the evaluation of non-palpable breast lesions [7].
Advantages of breast CNB include increased accuracy over FNA when the procedure is performed in situations where no mass is palpable and an ability to obtain tissue samples of sufficient size to eliminate the need for a complementary biopsy to confirm malignancy. Clip placement is done at the time of core needle biopsy so that the radiologist can identify the location of the lesion in the event that it is entirely removed or disappears during neoadjuvant treatment of the BC [7].
An excisional biopsy involves removal of the entire breast mass or suspicious area of the breast. Needle or wire localisation is done by the radiologist immediately prior to an excisional biopsy of a non-palpable mammographic or sonographic finding to direct the surgeon. The wire localisation may bracket a lesion that had a clip placed in it at the time of the core needle biopsy. Excisional biopsy is an option when larger samples of tissue are required [7].
12.1.4 Histological Types and Molecular Subtypes
The most common histological type of invasive breast carcinoma is designated as invasive ductal (IDC), NOS (not otherwise specified). It comprises about 80 % of all cases. It is a proliferation of epithelial cells from galactophoral ducts; it may be preceded, or accompanied, by an in situ component characterised by a proliferation of cells within the ducts without interruption of the basal membrane. When this membrane is altered, the carcinoma is considered as invasive. Many morphologic variants of invasive ductal carcinoma exist, some of them being extremely rare as described in Table 12.1. The prognosis of these subtypes varies, some of them having a better and some a worse outcome than invasive ductal carcinoma NOS [15]. Invasive lobular carcinoma (ILC) is the second major type (5–10 %) of breast cancer. As with ductal carcinoma, it may be preceded or accompanied by an in situ component. It is histologically more homogeneous than ductal carcinoma, but some morphologic variations exist, such as pleomorphic and signet ring cell types [15]. The tumour features are generally graded based on the architecture (nuclear atypia and mitotic activity and amount of tubule formations), the sum of these features results in an important prognostic indicator as depicted in the Nottingham score in Table 12.2.
Table 12.1
Histological classification
Tumour location | Histological subtype |
---|---|
Carcinoma, NOS | |
Ductal | Intraductal (in situ) Invasive with predominant component (80 %) Invasive, NOS Comedo Inflammatory Medullary with lymphocytic infiltrate Mucinous (colloid) Scirrhous Papillary Tubular Other |
Lobular | In situ Invasive with predominant in situ component Invasive |
Nipple | Paget’s disease, NOS Paget’s disease with intraductal carcinoma Paget’s disease with invasive ductal carcinoma |
Other | Undifferentiated carcinoma Metaplastic |
NOS = not otherwise specified | |
Phyllodes tumour Angiosarcoma Primary lymphoma | These tumour subtypes occur in the breast but are not considered typical breast cancers |
Table 12.2
Histological grade according to Nottingham classification
Grade 0 (G0) | Grade cannot be assessed |
Grade 1 (G1) | Favourable: low combined histological grade |
Grade 2 (G2) | Moderate favourable: intermediate combined histological grade |
Grade 3 (G3) | Unfavourable: High combined histological grade |
A central component of the treatment of BC is full knowledge of the extent of disease and biological features. These factors contribute to the determination of the stage of disease, assist in the estimation of the risk of recurrence and provide information that predicts response to therapy (e.g. hormone receptors [HR], human epidermal growth factor receptor 2 [HER2] and proliferation index [i.e. Ki67]). These factors are determined by examination of excised tissue. Hormone receptor status, normally determined by immunohistochemistry (IHC) testing, oestrogen (ER) and progesterone (PR) should be established for all histological samples of BC including DCIS [16, 17]; HER2 status can be assessed by measuring the number of HER2 gene copies (fluorescence in situ hybridization [FISH]), or by a complementary method in which the quantity of HER2 cell surface receptors is assessed by IHC [16]. The accuracy of HER2 assays used in clinical practice is a major concern, and results from several studies have shown that false-positive as well as false-negative results may occur. Currently, there is a general consensus that either an IHC or FISH test is an acceptable method for making an initial determination of HER2 tumour status provided that the test method has been validated and shown to be at least 95 % concordant with another validated method. Breast cancer tumours are classified as HER2 positive if they demonstrate HER2 gene amplification by a FISH method or are scored as 3+ by an IHC method [13, 18]. Ki-67 expression is associated with common histopathological parameters but is an additional independent prognostic parameter for DFS and OS in breast cancer patients [19].
Tumours with a similar microscopic aspect and similar disease stage can have very different outcomes when treated with the same therapy. The two major microscopic subtypes of BC (i.e. ductal and lobular carcinomas) have so far been managed with similar approaches. However, ILC of the classic type seems to have a better long-term outcome than IDC, in spite of its lower sensitivity to primary chemotherapy [20]. Dian et al. [21] found that in a multivariate analysis of 2058 patients, histology was an independent prognostic factor for overall survival (OS) in invasive BC (p = 0.046) but not for disease-free survival (DFS) (p = 0.599). The analysis of OS curve between IDC and ILC showed a significantly better OS for patients with ILC (p = 0.0302). Disease-free survival was not statistically different (p = 0.6650) between the two histologies, whilst relapse is substantially later for ILC [21].
The hereditary breast carcinomas, especially those associated with BRCA-1 mutations, frequently show particular phenotypes. The BCRA1/BRCA2 is a tumour suppressor gene, originally identified in 1994 by positional cloning on chromosome 17q21, and it is a multifocal protein in many normal cellular functions including DNA repair, transcriptional regulation, cell cycle checkpoint control and ubiquitination [22, 23]. Ninety percent of BC in BRCA1 carriers is high grade and HR negative. They also have more p53 mutations and fewer HER2 overexpression rates than sporadic tumours. The medullary type is ER negative, PR negative and HER2 negative and is seen more often in BRCA1 carriers than in sporadic cases. Tumours seen in BRCA2 carriers seem to be more similar to sporadic tumours (HR positive and p53 negative). These particular features associated to BRCA1 mutations are thought to be the result of a different genetic profile [24].
The Human Genomic Project (HGP) achieved its objectives around the year 2003 [25]. The development of cDNA microarrays, which allow the simultaneous analysis of thousands of genes, and other new technologies, has started a new era in our understanding of BC biology and therapy. Therefore, several attempts have been made to establish a more complete classification of BC based on molecular/genetic features rather than morphology of the tumours.
Perou et al. [26, 27] identified tumours with distinct patterns of gene expression, based on clusters of genes. The expression of the proliferation cluster of genes correlated well with clinical features of high-grade proliferative lesions expressing Ki-67, and the ER cluster correlated with variations in clinical ER expression. There was also a subtype of tumours with HER2 overexpression and ER underexpression, whereas tumours that overexpress both HER2 and ER fall into a different cluster.
These observations led to the proposal of BC groups according to immunohistochemistry characteristics, as depicted in Tables 12.3 and 12.4.
Table 12.3
Molecular subtypes of breast cancer
Luminal group is characterised by the high expression of many genes expressed by breast luminal cells including ER and the cytokeratins (CK) 8/18. None of these tumours overexpress HER2 |
The luminal subtype A presented the highest expression of ER-a gene, GATA binding protein 3, X-box binding protein 1, trefoil factor 3, hepatocyte nuclear factor 3 alpha and the oestrogen- regulated LIV-1 |
The luminal subtype B showed moderate expression of the luminal specific genes including the ER cluster |
HER2 subtype (HER2/neu) is overexpressed and associated with the expression of several genes in the ERBB2 amplicon at 17q22.24 including ERBB2 and GRB7. These tumours also show low levels of expression of ER and of almost all of the other genes associated with ER expression, a feature they share with the basal-like tumours |
Normal-like: the tumours defined as normal breast subtype express genes distinctive of basal epithelial and adipose cells. They also have a low expression of the luminal cell gene cluster |
Basal–like group has been characterised by high expression of CK5/CK6 and CK17, laminin and fatty acid binding protein 7 and fails to express ER and most of the other genes that are usually co-expressed with it |
Table 12.4
Classical molecular features vs. basal epithelial cells
Luminal epithelial cell markers | Basal epithelial cell markers |
---|---|
Higher expression: CK7, CK8, CK18, CK19 MUC1 Alpha-6-integrin BCL2 ER PR GATA3 Epithelial cell adhesion molecules | Higher expression: CK5, CK14, CK17 Smooth muscle markers Calponin, caldesmon P63, beta-4 integrin Laminin, maspin CD10, P-cadherin, caveolin-1 NGFR HER1/EGFR |
Lower expression: Basal CK (CK5, CK14, CK17) HER2 P53 | Lower expression: ER, PR, HER2 Desmin |
The patients with luminal A breast cancer subtype have a significantly longer overall and disease-free survival, whereas basal-like and HER2+ showed the shortest survival times. These differences in survival could be explained by the overexpression of HER2 and the presence of p53 mutations that are associated with poor survival in BC; the luminal A subgroup contained only 13 % p53-mutated tumours, whereas HER2+ and basal-like subtypes had 71 % and 82 %, respectively. Conversely, BCs in BRCA1 germ line mutation carriers fell into the basal-like subgroup. This indicates that a mutation in this gene predisposes for the development of basal-like cancer subtype and is associated with the lack of expression of ER and poor prognosis [26, 28].
12.1.5 Triple-Negative Breast Cancer
Triple-negative breast cancers (TNBCs) have been characterised by several aggressive clinico-pathological features including onset at a younger age, higher mean tumour size, higher-grade tumours and, in some cases, a higher rate of node positivity. A histological study of basal-like tumours, all of which were HR/HER2 negative, illustrated marked increases in mitotic count, geographic necrosis, pushing borders of invasion and stromal lymphocytic response. The majority of TNBCs are ductal in origin; however, several other aggressive phenotypes appear to be overrepresented, including metaplastic, atypical or typical medullary and adenoid cystic [29, 30].
It is important to clarify the relationship between TNBC and the basal-like phenotype. Triple negative is a term based on clinical assays for ER, PR and HER2, whereas basal-like is a molecular phenotype that was initially defined using complementary DNA (cDNA) microarrays. Although most TNBCs do cluster within the basal-like subgroup, these terms are not synonymous, and there is up to 30 % discordance between the two groups. The term “basal-like” is used when microarray or more comprehensive IHC profiling methodology is employed and “triple negative” when the salient studies have relied on clinical assays for definition [30]. Classically, basal-like BCs have been characterised by low expression of ER, PR and HER2 and high expression of CK5, CK14, caveolin-1, CAIX, p63 and epidermal growth factor receptor (EGFR, HER1), which reflects the mammary gland basal/myoepithelial cell component [31].
Several molecules integrally involved in DNA repair are aberrantly expressed in TNBC, which may have implications for chemotherapy sensitivity. High p53 IHC expression or p53 gene mutations are common in basal-like BC [32]. Furthermore, in one series 82 % of basal-like BCs expressed a p53 mutation compared with only 13 % in the luminal A subtype (p < 0.001) [32]. Several additional and targetable molecular pathways involved in the pathogenesis of basal-like BC include the mitogen-activated protein (MAP) kinase pathway, the Akt pathway and the poly(ADP-ribose) polymerase 1 (PARP1) pathway. It has been observed that the majority of BRCA1-associated BCs express a high proportion of basal-like cytokeratins (CK5, CK14, CK17), as well as P-cadherin and HER1/EGFR [33–35].
Several studies have shown that breast tumours arising in women carrying germ line mutations of the BRCA1 tumour suppressor gene are TNBC [33]. Gene expression studies support this association; among patients with BRCA1 mutations, breast tumours tend to cluster within the basal-like category. As BRCA1 is in part responsible for DNA repair, exploitation of this essential pathway holds therapeutic implications in the context of the TNBC phenotype [33–36].
12.1.6 Staging
The recent revision of the Cancer Staging Manual seventh edition of The American Joint Committee on Cancer (AJCC) [37] incorporated the use of novel imaging and pathology techniques employed at diagnosis (Table 12.5). This revision provides more direction relating to the specific methods of clinical and pathological tumour measurements. For instance, all invasive tumours should be assigned a grade according the Nottingham grading system permitting clarification of the classification of isolated tumour cells in axillary lymph node staging. Stage I is subdivided into IA and IB based on the presence, or absence, of nodal micrometastases (N0 vs. N0mi+). A new category of M0 status is considered, taking into account the presence of tumour cells detectable in blood or in bone marrow or in other tissues, if this not exceeds 0.2 mm. It is strongly recommended to consider the prognostic factors including tumour grade, HR status (both ER and PR) and HER2 status, although these characteristics do not specifically influence the disease staging [23, 37].
Stage 0 | Tis | N0 | M0 |
Stage IA | T1a | N0 | M0 |
Stage IB | T0a | N1mi | M0 |
T1a | N1mi | M0 | |
Stage IIA | T0 | N1b | M0 |
T1a | N1b | M0 | |
T2 | N0 | M0 | |
Stage IIB | T2 | N1 | M0 |
T3 | N0 | M0 | |
Stage IIIA | T0 | N2 | M0 |
T1a | N2 | M0 | |
T2 | N2 | M0 | |
T3 | N1 | M0 | |
T3 | N2 | M0 | |
Stage IIIB | T4 | N0 | M0 |
T4 | N1 | M0 | |
T4 | N2 | M0 | |
Stage IIIC | Any T | N3 | M0 |
Stage IV | Any T | Any N | M1 |
12.2 Breast Cancer Treatment: Concepts
12.2.1 Lobular Carcinoma In Situ
Lobular carcinoma in situ (LCIS) has long been considered a risk factor for the future development of invasive breast carcinoma, but recent evidence suggests that LCIS may also be a non-obligate precursor to invasive lobular carcinoma. LCIS is rarely detected by physical examination, and it does not have specific mammographic features [38]. LCIS arises from the terminal duct apparatus and shows a rather diffuse distribution throughout the breast, which explains its presentation as a non-palpable mass in most cases. Currently about 0.5–3.9 % of image-guided CNB incidentally identifies LCIS. The incidence of LCIS is rising due to improvements in mammographic technology [38, 39]. Patients diagnosed with LCIS have an eight- to tenfold increased lifetime risk of developing breast cancer, compared with women without this diagnosis [40]. The likelihood of developing invasive breast cancer increases by about 1 % every year after LCIS diagnosis with a 13 % risk after 10 years and a 21–26 % risk after 20 years [41]. LCIS is believed to arise from atypical lobular hyperplasia, a pre-invasive lesion with morphological features similar to LCIS, except with smaller, less distended acini. The term lobular neoplasia (LN) has been adopted by many to encompass all preinvasive lobular disease. The most well-studied characteristic of LN is loss of E-cadherin, and this is clinically used to differentiate lobular from ductal lesions [42].
Debate exists concerning whether a surgical excision should be implemented in the region of LCIS diagnosed by core biopsy that is not associated with a structural mammographic abnormality or residual mammographic calcifications [13]. Studies have shown that 17–27 % of patients with LCIS diagnosed by core needle biopsy are upgraded to invasive cancer or DCIS after larger excisional biopsy. Thus, it is reasonable to perform surgical excision of LCIS found in a core biopsy to exclude an associated invasive cancer or DCIS. More than four foci of LCIS may also increase the risk for upstaging on surgical biopsy. The NCCN recommends that LCIS [13] of the usual type (involving <4 terminal ductal lobular units in a single core) found on core biopsy as a result of routine screening for calcifications and without imaging discordance may be managed by imaging follow-up [23, 43, 44].
The risk of an invasive BC after a diagnosis of LCIS is equal in both breasts. If mastectomy is considered as a risk reduction strategy, then a bilateral procedure is required to optimally minimise risk. There is evidence to support the existence of histologically aggressive variants of LCIS that may have a greater potential to develop into invasive lobular carcinoma, and in these cases surgeons must consider the need to obtain negative margins after excision [23, 45].
Data from the National Surgical Adjuvant Breast and Bowel Project (NSABP P-1) showed after a 7-year follow-up that tamoxifen given for 5 years, in the group of patients with a previous history of LCIS, reduced the risk of developing an invasive BC to 5.43 per 1000 with a risk ratio of 0.54 (95 % CI = 0.10–1.2) [46].
Accurate follow-up of these patients includes interval history and physical examinations every 6–12 months with annual mammography [23].
12.2.2 Ductal Carcinoma In Situ
Ductal carcinoma in situ (DCIS) is a breast neoplasm with the potential for progression to invasive cancer. The term includes a wide variety of diseases, ranging from low-grade, indolent lesions to high-grade, aggressive tumours that can be a precursor to invasive disease. Patients with DCIS can be asymptomatic at the time of appearance (radiographic findings on mammogram) or may present with symptoms such as a palpable mass or nipple discharge. The incidence of DCIS has markedly increased in the last years, mainly due to improvements in screening with new imaging techniques [47].
Ductal carcinoma in situ is histologically defined by the presence of malignant epithelial cells within the well-defined breast ducts. The malignant cells are, by definition, bound by an intact basement membrane without any basal myoepithelial layer invasion. There are several architectural subtypes of DCIS: solid, comedo, cribriform, micropapillary and papillary. Furthermore, DCIS is classified qualitatively by nuclear grade (high, intermediate and low, based on cytologic/structural features) and the presence or absence of necrosis. Patients often present with lesions that contain at least two architectural subtypes. Although pathologic criteria have been established to distinguish DCIS from normal hyperplasia and atypical ductal hyperplasia (ADH), the diagnosis can still be very challenging for pathologists, as these entities represent a continuum of cellular and architectural atypia. Distinguishing between ADH and DCIS can be particularly difficult, as demonstrated by significant differences in diagnosis on expert pathology review [48, 49].
Patients with DCIS and evidence of widespread disease (i.e. disease in two or more quadrants) on mammography or other imaging or biopsy require a total mastectomy without lymph node dissection. For patients with more limited disease where negative margins are achieved, breast-conserving (BC) therapy or total mastectomy are appropriate treatment options. Although mastectomy provides maximum local control, the long-term, cause-specific survival with mastectomy appears to be equivalent to that with excision and whole-breast irradiation (WBRT); these patients are candidates for breast reconstruction [50].
Prospective randomised trials have shown that the addition of whole-breast irradiation to a margin-free excision DCIS decreases the rate of in-breast disease recurrence but does not affect overall survival or distant metastasis-free survival. After breast-conserving surgery, WBRT reduces the relative risk of a local failure by approximately one-half [13].
A retrospective analysis suggests that selected patients have a low risk of in-breast recurrence with excision alone without breast irradiation [51]. In the Di Saverio et al. analysis, 10-year disease-free survival rates of patients treated with breast-conserving surgery alone were 94 % for patients with low risk and 83 % for patients with both intermediate- and high-risk DCIS [52].
Many factors, including patient age, tumour size, tumour grade and margin width, have an impact on recurrence risk. The definition of a negative margin has not been definitely established in DCIS. However it is accepted that margins >10 mm are adequate and margins less than 1 mm are inadequate, but no uniform consensus exists for margin status between these values [13, 53]. Radiotherapy could be beneficial in cases with margins less than 1 mm.
A meta-analysis of four large randomised trials confirms that the addition of radiotherapy to BC surgery for DCIS provides a statistically significant reduction in local recurrence (p < 0.0000) [54]. In another meta-analysis of 4,660 patients with DCIS treated with breast-conserving surgery (BCS) and radiation, a surgical margin of less than 2 mm was associated with increased ipsilateral recurrence compared with margins of 2 mm, although no significant differences were observed when margins of greater than 2–5 mm or greater than 5 mm were compared with 2-mm margins [55]. The results of this study suggest that wide margins (≥2 mm) do not provide additional benefit in the population of patients with DCIS receiving radiation therapy following breast-conserving therapy.
The choice of local treatment does not affect overall events, disease-free survival (DFS) and overall survival (OS), but the potential for an increased risk of local recurrence must be considered [13]. Post-excision mammography is a valuable technique that complements specimen mammography and histopathological margin assessment in confirming that an adequate excision of DCIS has been performed. Post-excision mammography should be performed on all patients with DCIS associated with mammographic calcifications who are treated with breast-conserving therapy [56].
Axillary dissection is not recommended for patients with DCIS because axillary nodal involvement in DCIS is rare [57]. It is important to consider that a small percentage of patients with an apparent DCIS on initial biopsy could have invasive BC at the time of the definitive surgical procedure. In this case axillary lymph node staging is recommended. On the other hand, for patients to be treated with mastectomy or with excision in a difficult anatomic location, for instance, the tail of the breast, which could compromise the performance of a future sentinel lymph node (SLN) dissection, an SLN procedure may be considered [13].
Tamoxifen may be considered as a strategy to reduce the risk of local recurrence in patients whose ER-positive DCIS has been previously treated. The NSABP Breast Cancer Prevention trial showed a 75 % reduction in the occurrence of invasive BC in patients with atypical ductal hyperplasia treated with tamoxifen. Moreover, a substantial reduction in the risk of developing benign breast disease was observed in patients receiving tamoxifen [46, 58, 59]. An overview analysis performed by the Early Breast Cancer Trialists’ Collaborative Group showed that patients, with ER-positive or unknown lesions, receiving tamoxifen for 5 years resulted in a 39 % reduction in the annual probabilities of recurrence or invasive breast cancer [60]. The updated analysis of the NSABP B-24 trial showed a benefit from tamoxifen for women with DCIS, after treatment with BC surgery and radiation therapy. In this analysis, 76 % of patients were ER positive, and those treated with tamoxifen showed a significant decrease in further breast cancer at 10 years (p < 0.001), whilst no benefit was encountered among ER-negative patients [61].
Follow-up of women with DCIS includes interval history and physical examination every 6–12 months for 5 years and then annually, as well as yearly diagnostic mammography. Usually, for patients undergoing breast-conserving therapy, the first follow-up mammogram should be performed 6–12 months after the completion of breast-conserving radiation therapy. Patients receiving tamoxifen should be monitored in order to detect complications, such as endometrial neoplasia [23, 62].
12.2.3 Invasive Breast Cancer: Stages I, IIA and IIB or T3, N1, M0
12.2.3.1 Locoregional Treatment
Surgery is considered the primary treatment for breast cancer in early-stage patients. The endpoints of breast cancer surgery include the complete resection of the primary tumour, achieving negative margins as this reduces the risk of local recurrence. Moreover, surgery permits the pathologic staging of the tumour and axillary lymph nodes (LN) to make prognostic information available.
Lumpectomy is defined as complete surgical resection of a primary tumour with the objective of achieving widely negative margins (ideally a 1-cm margin). It is applicable in most patients with stage I and stage II invasive carcinomas [63].
Relative contraindications to lumpectomy include small breast size, large tumour size (>5 cm) and collagen vascular disease. Lumpectomy is absolutely contraindicated if there is multifocal disease, a history of previous radiation therapy to the area of treatment, inability to undergo radiation therapy for invasive disease and pregnancy or persistent positive margins following attempts at conservation. Factors that are often considered, but should not be detrimental, include axillary node involvement and tumour location. Although achieving a good cosmetic result is important, it should never be more important than the clinical priority of obtaining negative surgical margins, so lesions involving Paget’s disease of the nipple should be treated with excision of the nipple–areolar complex and reconstruction. Larger lesions in patients who are concerned about the cosmetic result may be better served by standard modified radical mastectomy and concurrent reconstruction [63].
A total mastectomy is defined as complete removal of all breast tissue to the clavicle superiorly, the sternum medially, the inframammary crease inferiorly and the anterior axillary line laterally, with “en bloc” resection of the fascia of the pectoralis major. The nipple–areolar complex is resected along with a skin paddle to achieve a flat chest wall closure when performing a total mastectomy. A total mastectomy does not refer to removal of any axillary nodes but may be performed in conjunction with a sentinel or axillary node dissection [63].
A modified radical mastectomy is defined as a total mastectomy with axillary lymph node dissection. In contrast, a radical mastectomy is defined as a total mastectomy plus “en bloc” resection of the pectoralis major and axillary lymph node dissection [63].
A relative contraindication to modified radical mastectomy is locally advanced cancer requiring neoadjuvant therapy before surgical intervention. Complications after total mastectomy could include the risk of local recurrence (5–10 %), wound infection, seroma, mastectomy skin flap necrosis, haematoma, chronic pain, incisional “dog ears”, lymphoedema and fibrosis.
Many randomised trials support the contention that mastectomy with axillary lymph node dissection is equivalent to breast-conserving therapy with lumpectomy, axillary dissection and whole-breast irradiation, as primary breast treatment for the majority of women with stage I and stage II BCs [64, 65].
The NSABP conducted a landmark randomised study (NSABP-B06) that established breast-conserving surgery with radiation therapy to be equivalent to modified radical mastectomy. At 20-year follow-up, no significant difference was seen in OS, local or regional DFS or distant disease-free survival between the three treatment groups. Patients in the lumpectomy group without radiation had a significantly higher LR rate (39.2 %) than patients undergoing lumpectomy plus radiation therapy (14.3 %). Patients who underwent modified radical mastectomy had a 10.2 % risk of chest wall recurrence [65].
A typical woman with clinical stage I or stage II BC requires pathological assessment of the axillary lymph node status. Performance of SLN mapping and resection in the surgical staging of the axilla is recommended for assessment of the pathologic status of the axillary lymph nodes in patients with stage I or stage II B disease [23, 66]. This recommendation is supported by results of randomised clinical trials showing decreased arm and shoulder morbidity such as pain, lymphoedema and sensory loss, in patients undergoing SLN biopsy [13, 67]. No significant differences in the effectiveness of the SLN procedure or level I and II dissection in determining the presence or absence of metastases in axillary nodes were seen in these studies. However, not all women are candidates for SLN resection. Potential candidates for SLN mapping and excision should have clinically negative axillary lymph nodes, or a negative core or FNA biopsy of any clinically suspicious axillary lymph node(s) [13, 67]. If the SLN cannot be identified or is positive for metastasis, a formal axillary lymph node dissection should be performed or axillary irradiation administered. The optimal technique for axillary radiation is not established in studies, but the axillary nodes can be included in the breast tangential fields. If lymph node mapping identifies sentinel lymph nodes in the internal mammary chain, internal mammary node excision is considered optional.
Traditional level I and level II axillary dissection required that at least ten lymph nodes should be provided for pathologic evaluation to accurately stage the axilla [68]. Axillary dissection should be extended to include level III nodes only if gross disease is apparent in the level I or II nodes [68].
Additionally, in the absence of definitive data demonstrating superior survival with axillary lymph node dissection or SLN resection, these procedures may be considered optional in patients who have particularly favourable tumours, patients for whom the selection of adjuvant systemic therapy is unlikely to be affected by the results of the procedure, elderly patients and patients with serious co-morbid conditions. Women who do not undergo axillary dissection or axillary lymph node irradiation are at increased risk for ipsilateral lymph node recurrence [69]. Women who undergo mastectomy are appropriate candidates for breast reconstruction.
12.2.4 Pre-operative (Induction or Neoadjuvant) Chemotherapy for Large Clinical Stage IIA and IIB Tumours and T3, N1, M0 Tumours
12.2.4.1 Induction Chemotherapy
Induction chemotherapy, also known as primary, pre-operative or neoadjuvant chemotherapy, for BC is indicated in locally advanced breast cancer (LABC) and patients with stage II–III disease in order to downstage the tumour burden and ensure appropriate breast conservation. Neoadjuvant therapy also makes it possible to evaluate drug sensitivity. The achievement of tumour and nodal pathologic complete remission (pCR) or minimal residual disease (MDR <1 cm) favourably affects disease-free survival (DFS) and overall survival (OS) [28].
Several clinical trials have attempted to apply information on drug sensitivity to clinical management on the hypothesis that non-cross-resistant chemotherapy agents increase pCR and ultimately improve survival.
The NSABP B-27 [70] trial was designed to determine the impact of adding docetaxel (DXT) after four cycles of induction AC (doxorubicin [ADR] plus cyclophosphamide [CPM]) on clinical CR, pCR rates, DFS and OS. Compared to pre-operative AC alone, pre-operative AC followed by DXT increased the clinical complete response rate (40.1 % vs. 63.6 %; p < .001), the overall clinical response rate (85.5 % vs. 90.7 %; p < .001), the pathologic complete response rate (13.7 % vs. 26.1 %; p < .001) and the proportion of patients with negative nodes (50.8 % vs. 58.2 %; p < .001) [70]. Comparable results were observed by the EORTC trial 10902 (European Organization for Research and Treatment of Cancer). Patients received four cycles of fluorouracil, EPR and cyclophosphamide (FEC) administered either pre- or post-operatively. No difference was observed between the two groups in terms of OS and progression-free survival (PFS) and time to locoregional recurrence (p = 0.38, p = 0.27, p = 0.61, respectively) [71].
The German Pre-operative ADR and DXT Study (GEPARTRIO) [72] included 2090 patients, with large operable or locally advanced breast cancer (LABC), and the tumour reduction was assessed by ultrasound imaging (response guided) in order to make a decision on further treatment. No evidence of a difference in response to neoadjuvant chemotherapy was found by tumour stage when analysis was adjusted for baseline characteristics. Nevertheless, exploratory analysis suggested that response-guided neoadjuvant chemotherapy might improve survival and is most effective in hormone receptor-positive tumours. If confirmed, the response-guided approach could provide a clinically meaningful advantage for the neoadjuvant over the adjuvant approach in early breast cancer [73].
Bevacizumab is a recombinant humanised monoclonal IgG1 antibody that binds to and inhibits the biological activity of human vascular endothelial growth factor (VEGF) in in vitro and in vivo assay systems [74]. Bear et al. [75] conducted a trial involving 1206 HER2-negative patients. Patients were randomised to receive bevacizumab or not over the first six cycles of chemotherapy. The addition of bevacizumab significantly increased the pCR rates (28.2 % vs. 34.5 %, p = 0.02), particularly in the hormone receptor-positive subcategory of patients (15.1 % vs. 23.2 %, p = 0.007) [75]. The benefit of adding bevacizumab was confirmed in the GeparQuinto trial, carried out by von Minckwitz et al. [76]; patients with HER2-negative BC were randomly assigned to receive induction therapy with or without concomitant bevacizumab treatment. The addition of bevacizumab led to a significant increase of pCR rate (p = 0.04). Meanwhile, in this case, the benefit of bevacizumab treatment on pCR was observed only in the hormone receptor-negative subgroup (27.9 % vs. 39.3 %, p = 0.003).
12.2.4.2 Induction Endocrine Therapy
Induction endocrine treatment in postmenopausal, HR-positive, breast cancer patients has been limited to patients who were not suitable for chemotherapy and surgery. Earlier phase II studies with tamoxifen that focused primarily on elderly and/or frail patients were often unselected for HR status of the tumour and showed response rates ranging from 49 to 68 % [77]. Conflicting results are reported in the literature on the value of factors predictive of response in the neoadjuvant or induction therapy approach. In the subset of patients with ER-positive tumours, pCR rates range from 2 to 10 %, which suggests that other primary endpoints must be considered within this tumour subset [77].
Results from two randomised trials on induction endocrine therapy in HR-positive postmenopausal patients support the theory of a relationship between the probability of response and the degree of HR expression [78, 79]. Higher ER levels are significantly related to a greater probability of response in both studies. In addition, a positive significant correlation between the ER level and the degree of Ki-67 suppression after 2 and 12 weeks of endocrine treatment was reported by Dowsett et al. [80].
The level of expression of ER and progesterone (PgR) might be associated with the probability of response to induction chemotherapy. In a retrospective analysis involving 533 patients, no pCR was observed within the cohort of patients considered as highly endocrine responsive (ER and PgR ≥50 %). This compares with 3.3 % of those with ER or PgR expressed in 0–49 % of the cells and 17.7 % of those patients who are HR negative (p < 0.0001) [81]. Conversely, even with the higher incidence of pCR, a statistically significantly worse DFS and OS were observed for patients with ER and PgR absent tumours vs. patients with tumours expressing high ER and PgR (HR 6.4, 95 % CI 3.5–11.6, for DFS; HR 3.6, 95 % CI 2.4–5.6, for OS); this study conducted by Colleoni et al. shows that response and outcome after induction chemotherapy are correlated with the degree of expression of steroid hormone receptors [81].
Aromatase inhibitors (AIs) block the conversion of androgens to oestrogens and reduce oestrogen levels in tissue and plasma [82]. Third-generation AIs include the non-steroidal inhibitors, letrozole and anastrozole, and the steroidal inhibitor, exemestane. The results of large trials conducted in the metastatic and adjuvant setting [80, 83], indicating better outcomes among women given with AIs than those given with tamoxifen stimulated the investigation of these agents in the induction setting in postmenopausal women with HR-positive tumours.
A randomised study (P024 trial) compared 4 months of letrozole or tamoxifen as induction therapy for postmenopausal women with ER- and/or PgR-positive stage II or III breast cancer, which is considered to be unsuitable for breast-conserving surgery (BCS). Letrozole increased the clinical response rate (55 % vs. 36 %, p < 0.001) and the BCS rate (45 % vs. 35 %, p = 0.022) when compared with tamoxifen [84].
In the IMPACT study [79], postmenopausal women with ER-positive tumours received induction tamoxifen, anastrozole or a combination of tamoxifen and anastrozole for 3 months. The response rates were similar for all treatments, 37 % vs. 36 % vs. 39 %, respectively. In the PROACT trial, anastrozole and tamoxifen yielded a similar response rate [85]. Seo et al. conducted a meta-analysis on three studies to show an improved breast-conserving surgery rate in patients receiving AI [86]. In another randomised study in postmenopausal patients with HR-positive breast cancer, exemestane significantly increased the clinical response rate and the rate of BCS (36.8 % vs. 20 %, p = 0.05) [87].
In the ACOSOG Z1031trial, 374 postmenopausal women with clinical stage II or III ER-positive breast cancer were randomised to receive anastrozole, exemestane or letrozole for 16–18 weeks before surgery. The response rates observed were not statistically different [88].
The optimal duration of induction endocrine therapy has not yet been determined. However, an improved response rate has been reported with induction letrozole when the duration of therapy was prolonged beyond 3 months [89]. According to these observations, several authors recommend prolonging endocrine therapy for a minimum of 4–8 months [77].
Crosstalk between the oestrogen receptor (ER) and the phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathways is a mechanism of resistance to endocrine therapy, and the blockade of both pathways enhances antitumour activity in preclinical models [89, 90]. A new approach to restore endocrine responsiveness in breast cancer might be to use the combination of an AI with a signal transduction inhibitor as a PI3K/mTOR antagonist [91]. Baselga et al. [92] included 270 postmenopausal women with operable ER-positive breast cancer who were randomly assigned to receive 4 months of induction treatment with letrozole (2.5 mg/day) and either everolimus (10 mg/day) or placebo. Clinical response rate in the everolimus arm was higher than that of letrozole alone (i.e. placebo; 68.1 % vs. 59.1 %), which was statistically significant (p = 0.062).
Limited data are available on induction treatments for premenopausal patients. Masuda et al. [93] performed the unique double-blind, multicentre, randomised trial in 197 premenopausal patients with ER-positive, HER2-negative, operable breast cancer were to receive monthly goserelin plus either anastrozole and tamoxifen placebo (n = 98) or tamoxifen and anastrozole placebo (n = 99) over 24 weeks before surgery. More patients in the anastrozole group achieved complete or partial response compared to the tamoxifen group (anastrozole 70.4 % vs. tamoxifen 50.5 % [p = 0.004]).
12.2.4.3 Induction Therapy in Patients with HER2-Positive Tumours
About 20 % of patients with breast cancer will show overexpressed HER2 gene amplification, which is associated with an aggressive clinical course of the disease. Trastuzumab, a monoclonal antibody (mAb) targeting HER2, has established activity in both the metastatic and adjuvant settings for these patients. Several single-arm phase II trials in the induction therapy setting have been performed suggesting that the addition of trastuzumab to conventional chemotherapy shows promising antitumour activity, with pCR rates ranging between 12 and 76 % [94].
The NOAH (NeOAdjuvant Herceptin) study conducted by Gianni and colleagues [95] involved 235 patients with HER2-positive locally advanced or inflammatory disease. They randomly received either ADR/paclitaxel (TXL)/cyclophosphamide (CPM), methotrexate (MTX) and fluorouracil (5-FU) chemotherapy with or without simultaneous administration of trastuzumab for 30 weeks. This study achieved a significant increase in 3-year event-free survival (EFS) of 71 % with trastuzumab vs. 56 % without (p = 0.013) and an increase in the pCR rate of 38 % vs. 19 % (p = 0.001) [95].
In another study, 120 patients with stage II and III HER2-positive BC, ineligible for BCS, were randomly assigned to receive four cycles of epirubicin (EPR)/CPM (EC), followed by four cycles of DXT with or without trastuzumab concurrently with the DXT. The difference between the groups, in terms of pCR, was not statistically significant (26 % with trastuzumab vs. 19 % without) [96].
The Austrian Breast and Colorectal Cancer Study Group randomised 93 patients with HER2-positive BC to receive six cycles of EPR-DXT or EPR-DXT-capecitabine induction therapy with or without trastuzumab. The difference between the groups, in terms of pCR, was not statistically significant (pCR 38.6 % vs. 26.5 %, p = 0.212) [97].
Buzdar and colleagues [98] investigated the effect of the timing of trastuzumab administration with anthracycline and taxane induction chemotherapy in a randomised phase III trial (282 patients). Women with operable HER2-positive invasive breast cancer were randomly assigned into group A (140 pts) (“sequential group”) with 5-fluorouracil-EPR (75 mg m2)-CPM (FEC-75) for four cycles followed by weekly TXL plus weekly trastuzumab for 12 weeks, and group B (“concurrent treatment”) group (142 pts) with weekly TXL and trastuzumab for 12 weeks followed by four cycles of FEC-75 and weekly trastuzumab. The pCR rate was the primary endpoint. The sequential group reached 56.5 % pCR compared to 54.2 % in the concurrent group. Left ventricular ejection fraction dropped in 0.8 % of patients who received sequential treatment and in 2.9 % of patients who received concurrent treatment; by week 24, LVF had dropped to 7.1 % and 4.6 % of patients in the sequential and concurrent group, respectively. Both treatments reached a high response rate, but it should be stressed that concurrent administration of trastuzumab with anthracyclines offers no additional benefit and is not warranted [98].
Lapatinib is an epidermal growth factor receptor inhibitor for use in combination with capecitabine for the treatment of advanced or metastatic HER2-positive breast cancer in women who have received prior therapy and also for use in combination with letrozole to treat HR-positive and HER2-positive ABC in postmenopausal women for whom hormonal therapy is indicated [99]. The GeparQuinto phase III trial compared four cycles of EPR/CPM (EC) followed by DXT administered concurrently with either trastuzumab or lapatinib. This face-to-face comparison of trastuzumab and lapatinib showed that pCR rate with chemotherapy and lapatinib was significantly lower than that with chemotherapy and trastuzumab (p < 0.04) [100]. Baselga et al. [101] conducted the NeoALTTO trial comparing lapatinib plus Taxol vs. trastuzumab plus TXL vs. concomitant lapatinib and trastuzumab plus TXL (152 pts). The pCR rate obtained by the combination, trastuzumab plus lapatinib, was significantly better (p = 0.0001) [101]. An updated analysis performed by Piccart-Gebhart M et al. [102] demonstrated that patients achieving pCR had a better outcome compared with patients not achieving pCR [101].
Pertuzumab is a recombinant humanised MAB that targets the extracellular dimerisation domain (subdomain II) of HER2 and thus blocks ligand-dependent heterodimerisation of HER2 with other HER family members, including EGFR, HER3 and HER4. Pertuzumab inhibits ligand-initiated intracellular signalling through both mitogen-activated protein (MAP) kinase and phosphoinositide 3-kinase (PI3K) signalling pathways, resulting in cell growth arrest and apoptosis. Pertuzumab also mediates antibody-dependent cell-mediated cytotoxicity [103].
Two pivotal clinical studies have been performed with pertuzumab, trastuzumab and DXT in induction treatment for patients with HER2 positive with operable locally advanced or inflammatory breast cancer and a primary tumour size >2 cm. In the first trial, the combination of pertuzumab plus trastuzumab plus DXT, as compared with placebo plus trastuzumab plus DXT, significantly prolonged PFS (p < 0.001), with no increase in cardiac toxic effects [103]. The second study confirmed the previous results, and patients who received pertuzumab plus trastuzumab plus DXT had a significantly improved pCR rate (45.8 %) compared to those given trastuzumab plus DXT, without substantial differences in tolerability [104, 105].
12.2.4.4 Induction Therapy in Patients with TNBC
For patients with TNBC, Huober et al. reported a high pCR rate of 39 % in 509 patients treated with TAC (DXT/ADR/CPM) or TAC-NX (DXT/ADR/CPM – vinorelbine/capecitabine) [106]. Di Leo et al. [107] suggested a beneficial outcome of anthracyclines compared to CMF therapy in terms of DFS in 294 patients with TNBC. However, recent studies have cast doubt on the role of anthracyclines in early-stage TNBC [108]. An analysis conducted by von Minckwitz et al. concluded that for patients with TNBC in particular, treatment with higher cumulative doses of anthracyclines (≥300 mg/m2) and taxanes (≥400 mg/m2) provides higher pCR rates compared with lower cumulative dose regimens [109].
Recently, the use of platinum agents has received renewed interest in the treatment of TNBC. The CALGB 40603 study in TNBC patients of carboplatin or bevacizumab combined with chemotherapy resulted in an increased pCR breast rate: 60 % of patients who received carboplatin achieved pCR breast compared with 46 % of those who did not (p = 0.0018). Patients treated with a bevacizumab-containing regimen had a pCR breast rate of 59 % compared with 48 % of those who were not (p = 0.0089). Patients assigned to both agents had the highest pCR breast rate (67 %). A major limitation of this study was that it was not powered to demonstrate any difference on DFS and OS [110].
The addition of carboplatin to a regimen of a taxane, an anthracycline and bevacizumab significantly increases, in TNCB patients, the rate of patients achieving a pCR. In the GeparSixto study [111], including TNBC patients, 53.2 % achieved a pCR with carboplatin, compared with 36.9 % without (p = 0.005).
A meta-analysis has indicated that novel induction schedules in TNBC patients have achieved a significant improvement of pCR rate, particularly among those treated with carboplatin (p = 0.001)- or bevacizumab (p = 0.003)-containing regimens. Moreover, adding carboplatin for TNBC results in an absolute benefit of 13.8 % compared with control arms. However, such treatment had no impact on DFS and OS [112, 113].
12.2.4.5 Surgical Treatment After Induction Chemotherapy
Surgical treatment after induction chemotherapy focuses on the management of the tumour itself and the axillary spread. It is important to consider that one of the purposes of induction chemotherapy is breast conservation with accurate disease staging allowing avoidance of a broad axillary dissection. The development of sentinel node biopsy provides a remarkable advantage for breast conservation after induction chemotherapy [13].
If the tumour responds to induction therapy, lumpectomy plus axillary lymph node dissection or sentinel lymph node procedure may be considered. If induction therapy results in non-response or progressive disease or if lumpectomy is not possible, then mastectomy is considered with axillary staging. If a pre-chemotherapy sentinel lymph node procedure was performed and the sentinel lymph node was pathologically negative, then further axillary lymph node staging is not necessary. If a pre-chemotherapy sentinel lymph node procedure was performed and the sentinel lymph node was positive, then a level I/II axillary lymph node dissection should be performed [12, 13].
In the case of an inoperable tumour that fails to respond or where response is insufficient after several courses of induction therapy, an alternative induction must be taken into consideration followed by local treatment using either radiotherapy or surgery, or both [12, 13].
Surgery should be followed by individualised chemotherapy such as taxanes if the full course of planned chemotherapy was not administered pre-operatively, as well as breast and regional lymph node irradiation. There is no role for post-operative chemotherapy if a full course of standard chemotherapy has been completed pre-operatively. Endocrine therapy is carried out for 5 years in women with ER- and/or PR-positive tumours [12, 13].
12.2.5 Radiation Therapy
Post-operative radiotherapy in patients who underwent surgery having received previous induction treatment must be decided based on pre-chemotherapy tumour features, irrespective of tumour response to pre-operative systemic therapy. It is generally recommended that patients who have axillary nodal metastases receive radiotherapy to the chest wall and regional nodes after mastectomy or to breast and regional nodes after lumpectomy. Conversely, in patients with negative axillary nodes, radiotherapy is not classically recommended after mastectomy and is confined to the breast alone after lumpectomy [114].
12.2.5.1 Post-lumpectomy Radiation Therapy
The purpose of radiation therapy after breast-conserving surgery is to eradicate local subclinical residual disease so reducing local recurrence rates by approximately 75 %. Based on the results from several randomised studies, irradiation of the intact breast is considered the standard of care, even in the lowest-risk disease with the most favourable prognostic features [115].
There are two general approaches used to deliver radiation therapy: conventional external beam radiotherapy (EBRT) and partial breast irradiation (PBI). WBRT consists of EBRT delivered to the breast at a dose of 50–55 Gy over 5–6 weeks. This is often followed by a boost dose specifically directed to the area in the breast where the tumour was removed [115].
Common side effects of radiation therapy include fatigue, breast pain, swelling and skin desquamation. Late toxicity (lasting ≥6 months after treatment) may include persistent breast oedema, pain, fibrosis and skin hyperpigmentation [13].
Partial breast irradiation (PBI) is employed in early-stage breast cancer after breast-conserving surgery as a way of delivering larger fraction sizes whilst maintaining a low risk of late effects. Techniques that can deliver this therapy include interstitial brachytherapy and intracavitary brachytherapy (a balloon catheter inserted into the lumpectomy site [i.e. MammoSite]). Treatment is usually administered twice daily for 5 days. In several nonrandomised studies, these techniques have shown low local recurrence rates comparable to those of EBRT. The American Society of Breast Surgeons recommends several selection criteria when patients are being considered for treatment with accelerated PBI (2): age ≥45 years, invasive ductal carcinoma or ductal carcinoma in situ (DCIS), total tumour size (invasive and DCIS) ≤3 cm, negative microscopic surgical margins of excision and ALN or SLN negative [116].
An observational study with data from the SEER-Medicare Linked Database on 35,947 women aged >66 years-old with invasive breast cancer (79.9 %) or DCIS (20.1 %) established that standard EBRT was associated with a higher 5-year breast preservation rate than either lumpectomy alone or brachytherapy. These results are controversial because the study data did not take into account the use of the newest forms of brachytherapy which might reduce the applicability of these findings [117–120].
12.2.5.2 Postmastectomy Radiation Therapy
The guidelines developed by the American Society of Clinical Oncology (ASCO), based on several prospective, randomised clinical trials, recommend that postmastectomy radiation therapy must be performed according to some important criteria: positive postmastectomy margins, primary tumours >5 cm and involvement of ≥4 lymph nodes [124].
Patients with >4 positive lymph nodes should also undergo prophylactic nodal radiation therapy at doses of 45–50 Gy to the axillary and supraclavicular regions. For patients without ALND node involvement, axillary radiation therapy is not recommended.
Postmastectomy radiotherapy was shown in previous meta-analyses to reduce the risks of both recurrence and breast cancer mortality in all women with node-positive disease considered together. However, the benefit in women with only 1–3 positive LNs is uncertain. A meta-analysis of individual data for 8135 patients [125] randomly assigned to treatment groups during 1964–86 in 22 trials of radiotherapy to the chest wall and regional lymph nodes after mastectomy and axillary surgery vs. the same surgery but no radiotherapy has examined the question. Follow-up lasted 10 years for recurrence and for mortality. In this analysis 3786 women had axillary dissection to at least level II and had zero, 1–3 or ≥4 positive nodes. All were in trials in which radiotherapy included the chest wall, supraclavicular or axillary fossa (or both) and internal mammary chain. For 700 women with axillary dissection and no positive nodes, radiotherapy had no significant effect on locoregional recurrence (2P >0 · 1). For 1314 women with axillary dissection and 1–3 positive nodes, radiotherapy reduced locoregional recurrence (2P <0 · 00001), overall recurrence (2P = 0 · 00006) and BC mortality (2P = 0 · 01); 1133 of these women were in trials in which systemic therapy with CMF or tamoxifen was given in both trial groups, and for them, radiotherapy once more reduced locoregional recurrence (2P <0 · 00001), overall recurrence (2P = 0 · 00009) and BC mortality (2P = 0 · 01). For 1772 patients with axillary dissection and ≥4 positive nodes, radiotherapy reduced locoregional recurrence, overall recurrence and BC mortality (2P = 0 · 04, 2P = 0 · 0003 and 2P < 0 · 00001, respectively). The clear conclusion is that after mastectomy and axillary dissection, radiotherapy reduced both recurrence and BC mortality in the women with one to three positive lymph nodes in these trials, even when systemic therapy was given [125].
12.2.6 Breast Reconstruction
Breast reconstruction after mastectomy may be performed in the immediate or the delayed setting. Most patients undergoing mastectomy for prophylaxis or early-stage breast cancer are candidates for reconstruction. Nevertheless, most patients undergoing mastectomy do not undergo breast reconstruction due to different reasons, including provider biases, patient preferences and lack of available specialty services [13].
Immediate reconstruction, when feasible, generally provides superior cosmetic results, because a skin-sparing total mastectomy or nipple-sparing total mastectomy may be offered to selected patients, resulting in preservation of the native skin envelope and inframammary line. However, when postmastectomy radiation is likely or a reconstructive surgeon is unavailable, delayed reconstruction following all adjuvant therapies may be recommended [126].
Patients and physicians should have realistic expectations for breast reconstruction. Although excellent results may be achieved, often multiple operations are required for revisions, symmetry procedures and nipple reconstruction. Complications related to reconstruction include an infected prosthetic implant, implant rupture, capsular contracture, flap necrosis, flap loss, fat necrosis, asymmetry and scarring [13, 127].
12.2.7 Systemic Adjuvant Therapy
A number of prognostic factors predict for future recurrence or death from BC. The strongest prognostic factors are patient age, co-morbidity, tumour size, tumour grade, number of axillary lymph nodes involved, HR status and HER2 tumour status. Algorithms have been published estimating rates of recurrence, and a validated computer-based model, such as Adjuvant Online, is available to estimate 10-year disease-free and overall survival; this incorporates all of the above prognostic factors except for HER2 tumour status [23]. These tools aid the clinician to objectively estimate outcome with local treatment only and also assist in estimating the absolute benefits expected from systemic adjuvant endocrine therapy and chemotherapy. Determination of the HER2 status of the tumour is strongly recommended for prognostic purposes for patients with node-negative BC. HER2 tumour status also provides predictive information used in selecting optimal adjuvant/neoadjuvant therapy and in the selection of therapy for recurrent or metastatic disease. Retrospective analyses have demonstrated that anthracycline-based adjuvant therapy is superior to non-anthracycline-based adjuvant chemotherapy in patients with HER2-positive tumours and that the dose of ADR may be important in the treatment of tumours that are HER2 positive [128, 129]. However, prospective evidence of the predictive utility of HER2 status in early and metastatic BC is currently available for trastuzumab-containing therapies [23].
Use of DNA microarray technologies to typify BC has allowed the development of classification systems of BC by gene expression profile [26, 130, 131].
Multigene prognostic tests provide useful complementary information to tumour size and grade in HR-positive breast cancers. The tests primarily depend on quantification of ER- and proliferation-related genes and combine these into multivariate prediction models. Since ER-negative cancers tend to have higher proliferation rates, the prognostic value of current multigene tests in these cancers is limited [132]. Prognostic signatures such as Oncotype DX® and MammaPrint® are more precisely able to predict recurrence within the first 5 years than in later years. This could be a limitation with the convenience of effective extended adjuvant endocrine therapies. There is rising agreement that multigene prognostic gene signatures provide standardised, complimentary information to routine pathologic variables including tumour size, nodal status and histological grade [132]. Multigene prognostic assays are now recognised by the ASCO, St. Gallen, NCCN guidelines and ESMO as information that could assist therapeutic decision-making in ER-positive cancers [13, 124, 132, 133].
The Mammaprint® assay uses microarray technology to analyse a 70-gene expression profile from frozen breast tumour tissue. More recent versions of the test can use formalin-fixed and paraffin-embedded (FFPE) tissues [134]. The prognostic risk discrimination is good among ER-positive cancers, but almost all ER-negative cancers are classified as high risk, which limits the score’s clinical value in this disease subset. Retrospective analysis of a large multicentre patient cohort suggested that only the high-risk ER-positive patients benefited from adjuvant chemotherapy [135].
Oncotype DX® is an assay that utilises expression measurements from 16 cancer-related genes and five housekeeping genes (total 21 genes) to compute a recurrence score from 0 to 100, which can be categorised into low-risk (score <18), intermediate-risk (score 18–30) or high-risk (score ≥31) groups [132, 136]. Measurement is performed on FFPE samples in a central laboratory. Several studies on archived tissues from randomised adjuvant chemotherapy trials have demonstrated benefit from adjuvant chemotherapy primarily in patients with high recurrence score [50, 137]. A large, prospective, randomised clinical trial (TAILORx) is testing the utility of adjuvant chemotherapy in node-negative women with risk for recurrence by Oncotype DX® (score 11–25); the first results are expected by 2017 [138].
There are other multigene prognostic assays such as IHC4 [139], Prosigna®, Genomic Grade Index [132], Breast Cancer Indexsm [132] and EndoPredict test [140]. So far, Oncotype DX® is the most widely used and recommended prognostic assay for ER-positive BC [13].
12.2.7.1 Axillary Lymph Node-Negative Tumours
Tumours less than 0.5 cm in greatest diameter that do not involve the lymph nodes are considered as favourable, and as adjuvant systemic therapy does not add any benefit, it is not recommended as treatment of the invasive BC. Endocrine therapy may be considered to reduce the risk of a second contralateral BC, especially in those with ER-positive disease. The NSABP demonstrated a correlation between the ER status of a new contralateral breast tumour and the original primary tumour, reinforcing the notion that endocrine therapy is unlikely to be an effective strategy to reduce the risk of contralateral BC in patients diagnosed with ER-negative tumours [141].
Patients with invasive ductal or lobular tumours >5 cm in diameter and no lymph node involvement may be separated into those patients with a low risk of recurrence and those with unfavourable prognostic features that allow consideration of adjuvant therapy. Unfavourable prognostic features include intramammary angiolymphatic invasion, high nuclear grade, high histological grade, HER2-positive status or HR-negative status. The decision to undertake an endocrine therapy and chemotherapy in these relatively lower-risk subgroups of women must be based on balancing the expected absolute risk reduction with the patient’s general condition in the context of likely toxicity to achieve that incremental risk reduction [12, 13]. The incremental benefit of combination chemotherapy in patients with lymph node-negative, HR-positive BC may be relatively small [142]. Therefore, the tumour HR status must be included as one of the factors considered when making decisions about chemotherapy for patients with node-negative, HR-positive BC. Patients for whom this evaluation may be especially important are those with tumours characterised as 0.6–1.0 cm and HR positive that are grade 2 or 3 or have unfavourable features or greater than 1 cm and HR positive and HER2 negative. However, chemotherapy should not be withdrawn from these patients solely on the basis of ER-positive tumour status [13, 142, 143]. Assessment of the recurrence score is indicated in these cases taking into account the context of other elements of risk.
12.2.7.2 Axillary Lymph Node-Positive Tumours
Patients with LN-positive disease are candidates for chemotherapy and if the tumour is HR positive, for the addition of endocrine therapy [13]. In postmenopausal women, with HR-positive disease, an AI should be utilised unless a contraindication exists. In premenopausal women, adjuvant tamoxifen is recommended. If both chemotherapy and tamoxifen are administered, data from the Intergroup trial 0100 suggest that delaying initiation of tamoxifen until after completion of chemotherapy improves DFS compared with concomitant administration [144]. Consequently, chemotherapy followed by endocrine therapy should be the preferred therapy sequence.
It is also important to consider subsets of patients with early BC of the usual histologies based upon responsiveness to endocrine therapy and trastuzumab (i.e. hormone receptor status, HER2 status). Patients are then further stratified based upon risk for recurrence of disease assessed by anatomic and pathologic characteristics (i.e. tumour grade, tumour size, axillary lymph node status, angiolymphatic invasion [13, 23].
12.2.7.3 Adjuvant Endocrine Therapy
Patients with invasive BCs that are ER or PR positive should be considered for adjuvant endocrine therapy regardless of patient age, lymph node status or whether or not adjuvant chemotherapy is to be administered; HER2 amplification is a marker of relative endocrine resistance independent of type of endocrine therapy [145, 146]. However, the use of adjuvant endocrine therapy in the majority of women with HR-positive BC regardless of menopausal status, age or HER2 status of the tumour is recommended.
The most definitely recognised adjuvant endocrine therapy is tamoxifen for both premenopausal and postmenopausal patients [143]. In ER-positive BC patients, adjuvant tamoxifen decreases the annual probabilities of recurrence by 39 % and the annual probabilities of death by 31 % irrespective of the use of chemotherapy, patient age, menopausal status or axillary lymph node (LN) status. Prospective, randomised trials demonstrate that the optimal duration of tamoxifen appears to be 5 years. In patients receiving both tamoxifen and chemotherapy, chemotherapy should be given first, followed by tamoxifen [144].
The ten-year duration of tamoxifen treatment in women with stage I–III hormone receptor (HR)-positive breast cancer is based on the data collected mainly from two randomised trials, the aTTom and ATLAS in which more than 13,000 randomised patients were analysed. Adjuvant endocrine therapy with tamoxifen for 5 years should be offered to pre-/perimenopausal women. After this, if the patient remains premenopausal, they should be offered continued tamoxifen for an additional 5 years. If postmenopausal, patients should be treated with continued tamoxifen or an aromatase inhibitor (AI) for an additional 5 years, totalling 10 years of adjuvant endocrine therapy [13, 62, 115, 147].
In the ATLAS trial, in the second decade after diagnosis, women who continued on tamoxifen had a 25 % lower recurrence rate and 29 % (p = 0.002) lower breast cancer mortality rate compared with women who stopped after 5 years of tamoxifen. The risk of dying due to breast cancer at 5–14 years after diagnosis was 12.2 % for continuing tamoxifen vs. 15 % for those who only had 5 years of treatment, representing an absolute gain of 2.8 %, favouring continuing treatment with tamoxifen for 10 years (p = 0.01) [148].
In the aTTom study, women with breast cancer who had been taking tamoxifen for 5 years were randomised to continue receiving tamoxifen for another 5 years or stop taking the drug. Allocation to continue tamoxifen reduced breast cancer recurrence (p = 0.003). Longer treatment also reduced breast cancer mortality (392 vs. 443 deaths after recurrence, p = 0.05) [149].
Combining the similar results of aTTom and its international counterpart ATLAS enhances statistical significance of long-term effects of continuing adjuvant tamoxifen in terms of recurrence (p < 0.0001), breast cancer mortality (p = 0.002) and overall survival (p = 0.005) benefits [149].
The disadvantages of taking tamoxifen for 10 years may include continuing menopausal symptoms, such as night sweats and hot flashes. Longer duration of treatment increases the risk of endometrial cancer: in the aTTom trial, there were 102 cases and 37 (1.1 %) deaths attributed to endometrial cancer in the 10-year tamoxifen arm, compared with 45 cases and 20 (0.6 %) deaths in the 5-year group. However, the benefits demonstrated by a longer duration of treatment with tamoxifen outweighed the possible risks [149].
A number of studies have evaluated AIs in the treatment of postmenopausal women with early-stage BC. These studies have employed the AIs as initial adjuvant therapy, as sequential therapy following 2–3 years of tamoxifen or as extended therapy following 4.5–6 years of tamoxifen. The AIs are not active in the treatment of women with functioning ovaries. Tamoxifen should not be used in women whose ovarian function cannot be reliably assessed [13]. The results from two prospective, randomised clinical trials have provided evidence of an OS benefit for patients with early-stage BC receiving initial endocrine therapy with tamoxifen followed sequentially by anastrozole (p = 0.045) or exemestane (p = 0.05) when compared with tamoxifen as the only endocrine therapy [150, 151]. In addition, the National Cancer Institute Canada Clinical Trials Group (NCIC CTG) MA-17 study results in women who had completed 4.5–6 years of adjuvant tamoxifen demonstrated that extended therapy with letrozole provides benefit in postmenopausal women with HR-positive, early BC. The results showed fewer recurrences or new contralateral BCs with extended therapy (p < 0.001). No difference in OS was demonstrated, although there was a survival advantage in the subset of patients with axillary lymph node-positive disease (p = 0.04) [152].
However, no survival differences have been reported for patients receiving initial adjuvant therapy with an AI vs. first-line tamoxifen [153, 154].
Tamoxifen and AIs have different side effect profiles. Both contribute to hot flashes and night sweats and may cause vaginal dryness. Aromatase inhibitors are more commonly associated with musculoskeletal symptoms, osteoporosis and increased rate of bone fracture, whilst tamoxifen is associated with an increased risk of uterine cancer and deep venous thrombosis.
The ATAC study demonstrated that anastrozole is superior to tamoxifen or the combination of tamoxifen and anastrozole in the adjuvant endocrine therapy of postmenopausal women with HR-positive BC (p = 0.003) [146]. Nevertheless, no difference in OS has been observed (p = 0.2).
Breast International Group (BIG) BIG1-98 tested the use of tamoxifen alone for 5 years, vs. letrozole alone for 5 years, or tamoxifen for 2 years followed sequentially by letrozole for 3 years or letrozole for 2 years followed sequentially by tamoxifen for 3 years. DFS was superior in the letrozole-treated women (p = 0.003), but no difference in OS has been observed. The overall incidence of cardiac adverse events was similar (letrozole, 4.8 %; tamoxifen, 4.7 %). However, the incidence of grade 3–5 cardiac adverse events was significantly higher in the letrozole arm, and both the overall incidence and incidence of grade 3–5 thromboembolic events were significantly higher in the tamoxifen arm. A higher incidence of bone fracture was observed for women in the letrozole arm compared with those in the tamoxifen arm (9.5 % vs. 6.5 %) [155].
Four trials have studied the use of tamoxifen for 2–3 years followed sequentially by a third-generation AI vs. continued tamoxifen. The Italian Tamoxifen Anastrozole (ITA) [155, 156] and the Intergroup Exemestane Study (IES) trials both randomised postmenopausal women with BC who had completed a total of 2–3 years of tamoxifen to AI [150]. An improvement in terms of DFS was seen in all trials but not in OS except with the exemestane study in which a significant difference was observed only in patients with ER-positive tumours (log-rank test p = 0.05) compared to unknown HR status. The Austrian Breast and Colorectal Cancer Study Group (ABCSG) trial 8 [157] and the Arimidex–Nolvadex (ARNO 95) trial [158] demonstrated the superiority of sequential AIs in disease-free survival but gave conflicting results in terms of survival. A meta-analysis gathering updated data of the above-mentioned studies ABCSG 8, ARNO 95 and ITA showed significant improvement in overall survival (p = 0.04) with a switch to anastrozole [159].
The differences in design and patient populations among the studies of the AIs do not allow for the direct comparison of the results of these studies. Thus, it is not known whether initial, sequential or extended use of adjuvant aromatase inhibitors is the optimal strategy. The optimal duration of aromatase inhibitor treatment is also not known, nor is the optimal use vis-à-vis chemotherapy established [13]. The various studies are consistent in demonstrating that the use of a third-generation AIs in postmenopausal women with HR-positive BC lowers the risk of recurrence, including ipsilateral breast tumour relapse, contralateral BC and distant metastatic disease, compared to tamoxifen alone whether the AI is used as initial adjuvant therapy, sequential therapy or extended therapy. Thus, it is recommended that postmenopausal women with early BC should receive an aromatase inhibitor as initial adjuvant therapy, as sequential with tamoxifen or as extended therapy in those situations where endocrine therapy is to be utilised [13, 62].
It is very important to emphasise that the aromatase inhibitors do not adequately suppress ovarian oestrogen synthesis in women with functioning ovaries. Moreover, some patients with chemotherapy-induced amenorrhoea may regain ovarian function whilst receiving AI therapy and even become pregnant [160]. Premenopausal women should not be given with therapy with an AI outside the limits of a clinical trial. Women who are premenopausal at the time of diagnosis and who become amenorrheic with chemotherapy may have continued oestrogen production from the ovaries in the absence of menses. Serial assessment of circulating LH, FSH and oestradiol to assure a true postmenopausal status is mandatory if this subset of women is to be considered for therapy with an AI [83].
12.2.7.4 Adjuvant Cytotoxic Chemotherapy
The historical studies of CMF chemotherapy vs. no chemotherapy have shown disease-free and overall survival advantages with CMF chemotherapy [161]. Trials adding an anthracycline as in CAF/FAC (CPM, ADR, 5-FU) have shown that the use of ADR and the use of full-dose chemotherapy regimens are important [162, 163]. In the Early BC Trialists’ overview of polychemotherapy, comparison of anthracycline-containing regimens with CMF showed a further 12 % reduction in the annual probabilities of recurrence (p = 0.006) and a further 11 % reduction in the annual odds of death (p = 0.02) with anthracycline-containing regimens [161].
The results of two randomised trials comparing AC with or without sequential TXL therapy in women with axillary node-positive BC suggest improvement in DFS rates and OS with the addition of TXL [163, 164]. On retrospective analysis, the apparent advantage of the TXL-containing regimen appears greater in women with ER-negative BCs.
One trial conducted by Levine et al. [165] in premenopausal women with node-positive BC randomly assigned them to receive CMF therapy or CEF (CPM-EPR-5-FU) chemotherapy using high-dose EPR. Both 10-year relapse-free survival (53 % vs. 63 %; p = 0.009) and OS (70 % vs. 77 %; p = 0.03) favoured the CEF arm. A second trial carried out by the French Adjuvant Study Group compared CEF given at two dose levels of EPR (50 mg/m2 q 3 weeks vs. 100 mg/m2 q 3 weeks) in pre- and postmenopausal women with node-positive BC. Five-year DFS (55 % vs. 66 %; p = 0.03) and OS (65 % vs. 77 %; p = 0.007) both favoured the EPR 100 mg/m2 arm [166].
Another trial compared two dose chemotherapy levels of EC (EPR plus CPM) with CMF in women with node-positive BC. This study showed that higher-dose EC was equivalent to CMF and superior to moderate-dose EC in DFS and OS [167]. An additional randomised trial in patients with axillary LN-positive BC compared six cycles of FEC vs. three cycles of FEC followed by three cycles of DXT [167, 168]. Five-year disease-free survival (78.4 % vs. 73.2 %, adjusted p = 0.012) and overall survival (90.7 % vs. 86.7 %, p = 0.017) were superior with sequential FEC followed by DXT [168].
Final results from a randomised trial comparing DXT, ADR and CPM (TAC) vs. FAC in axillary LN-positive BC demonstrated that TAC is superior to FAC. Estimated 5-year DFS with TAC was 75 %, 68 % for FAC (p = 0.001) and 87 % for survival with TAC and 81 % with FAC (p = 0.008) [169].
At a median follow-up of 73 months, results from the 3-arm randomised NSABP B-30 trial comparing TAC vs. AT vs. AC followed by DXT (ACT) demonstrated that ACT had a significant advantage in DFS (p = 0.006) but not in OS (p = 0.086) when compared with TAC. In addition, both DFS (p = 0.001) and OS (p = 0.034) were significantly increased when ACT was compared with AT, demonstrating non-inferiority compared with TAC [170].