Вы находитесь на странице: 1из 12

Pathology (January 2009) 41(1), pp.

7788

ANCILLARY TOOLS IN BREAST PATHOLOGY

The role of molecular analysis in breast cancer


`* FELIPE C. GEYER*, CATERINA MARCHIO AND

JORGE S. REIS-FILHO

Molecular Pathology Laboratory, The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, United Kingdom; *these authors contributed equally to this review

Summary Breast cancer is a heterogeneous disease, encompassing a wide variety of histological types and clinical behaviours. Current histopathological classication systems for breast cancer are based on descriptive entities that are of prognostic signicance. Few predictive biomarkers are currently available. High throughput molecular technologies are reshaping our understanding of breast cancer and a molecular taxonomy that has stronger predictive power is slowly emerging. Novel therapeutic targets and prognostic/predictive gene signatures have been identied. This review will address the contribution of molecular methods to our understanding of breast cancer and its precursors, their use in breast cancer translational research and their impact on diagnostic breast cancer histopathology.
Key words: Microarrays, comparative genomic hybridisation, prognostic marker, predictive marker, genomic signature, histological grade, breast cancer. Received 20 July, revised 5 August, accepted 7 August 2008

Molecular pathology has already had a great impact on the diagnosis of haematological and soft tissue neoplasia. The contribution of molecular techniques to the classication of carcinomas has not yet been so profound. In the last few years, breast cancer has been the epithelial malignancy most studied by molecular techniques; therefore, it is not surprising that not only predictive markers, but also novel therapeutic targets are emerging. The focus of this review will be on the impact of molecular proling analysis and molecular genetics on our understanding of breast cancer and its precursors.

AN ARRAY OF CHANGES
The boom of high throughput technologies and the class discovery studies pioneered by the Stanford group4,5 have not only brought forward the fact that breast cancer is not a single entity but also provided a working model for a molecular taxonomy for breast cancer.48 Expression proling analyses using microarrays have demonstrated that breast cancers can be classied according to their expression patterns into at least ve groups:48 luminal A, luminal B, normal breast-like, HER2 and basallike. The most robust distinction observed by microarray analysis is between the transcriptome of ER positive and ER negative breast cancers. Luminal tumours are described as those that show expression patterns reminiscent of normal luminal epithelial cells of the breast, including consistent expression of low molecular weight cytokeratins 8/18, ER and genes associated with an active ER pathway.48 At least two subgroups of luminal tumours have been identied: luminal A, which are usually of low histological grade, have an excellent prognosis and show high levels of expression of ER-activated genes; and luminal B, which are more often of higher histological grade, have higher proliferation rates and a poorer prognosis than luminal A tumours.48 Normal breast-like cancers are rather poorly characterised tumours; one of the dening features of these lesions is that they consistently cluster together with samples of broadenomas and normal breast samples. The clinical signicance of normal breastlike tumours is yet to be determined48 and some have suggested that this subgroup may be a mere artefact of expression proling (i.e., disproportionally high content of stromal cells). HER2 tumours are usually ER negative and characterised by over-expression of HER2 and genes associated with HER2 pathway and/or HER2 amplicon on 17q12. HER2 cancers have very aggressive clinical behaviour; however, they are amenable to novel tailored

INTRODUCTION
Breast cancer is a heterogeneous disease encompassing a wide variety of pathological entities that are reported to have distinct clinical behaviours.1,2 Pathologists have acknowledged the complexity of breast cancer and endeavoured to devise classication systems to account for its diversity. However, current classication systems are descriptive, based on morphological entities that have been shown to have prognostic implications. For the success of targeted therapies and individualised medicine, a predictive, rather than purely prognostic, classication system is required. Despite the translational research eorts of the 1980s and 90s, only three biomarkers are routinely used in breast cancer patient management, namely oestrogen receptor (ER), progesterone receptor (PR) and HER2. Interestingly, all of these biomarkers have optimal negative predictive values (i.e., patients with ER negative breast cancer are highly unlikely to respond to endocrine therapy; HER2 negative breast cancers fail to respond to humanised monoclonal antibodies against HER2). However, their positive predictive value is rather limited, with a substantial proportion of patients with HER2 positive disease either harbouring de novo resistance or developing resistance to trastuzumab over time.3

Print ISSN 0031-3025/Online ISSN 1465-3931 # 2009 Royal College of Pathologists of Australasia DOI: 10.1080/00313020802563536

78

GEYER et al.

Pathology (2009), 41(1), January

therapies using either humanised monoclonal antibodies against HER2 or HER2 tyrosine kinase inhibitors.413 Although the vast majority (480%) of HER2 cancers as dened by microarrays harbour HER2 gene amplication or HER2 3 immunohistochemical expression,6,14 not all tumours that are HER2 amplied fall into the HER2 cluster by expression arrays analysis. There is also evidence to suggest that some HER2 amplied, ER positive cancers fall within the luminal subtypes rather than the HER2 microarray subtype.6 Basal-like cancers, another group of ER negative cancers, are so named because the neoplastic cells of this tumour type consistently express genes usually expressed in normal basal/myoepithelial cells of the breast, including high molecular weight cytokeratins (5/6, 14 and 17), P-cadherin, caveolins 1 and 21522 and epidermal growth factor receptor (EGFR)17 and, in a minority of cases, harbour EGFR gene amplication.7 These tumours are usually of high histological grade and characterised by high mitotic indices, the presence of central necrotic zones, pushing borders, conspicuous lymphocytic inltrate and typical/atypical medullary features.23,24 In addition, metaplastic elements are not uncommonly found.25 The morphological and immunohistochemical features of basal-like cancers are remarkably similar to those described for tumours arising in BRCA1 germline mutation carriers21 and there is a growing body of evidence to suggest that BRCA1 pathway is dysfunctional in sporadic basal-like cancers.26 In fact, engineered mouse models targeting Brca1 and Trp53 genes in luminal or basal cells of the mouse mammary gland resulted in the development of tumours displaying morphological and immunohistochemical features that recapitulate those seen in human basal-like breast carcinomas.27,28

BASAL-LIKE CARCINOMAS: BACK TO THE BASIS


Before discussing the implications of basal-like breast carcinomas, it is worth mentioning that two misconceptions have plagued the literature on this topic. First, there is a pervasive misconception that basal-like cancers were discovered by gene expression proling analysis. In fact, the existence of a subgroup of aggressive breast carcinomas showing features of myoepithelial/basal dierentiation has been known since the 1960s16,2932 and that these tumours more often express the immunohistochemical features now known to be characteristic of basallike cancers has been known since the 1990s.33 However, it would be fair to say that basal-like breast carcinomas only gained widespread interest after their rediscovery and systematic classication by microarray-based expression proling analysis. Another important misconception is that basal-like and triple negative (ER, PR and HER2 negative) cancers are synonymous.34 Although the vast majority of triple negative cancers are of basal-like phenotype3537 and the vast majority of tumours expressing basal markers are triple negative,17,38 there is a signicant number of triple negative cancers that do not express basal markers and a small, but still signicant, subgroup of basal-like cancers that express either hormone receptors or HER2.14,37,3941 Bertucci et al.39 have addressed this issue directly and conrmed that not all triple negative tumours when

analysed by gene expression proling were classied as basal-like cancers (i.e., only 71% were of basal-like phenotype) and not all basal-like breast carcinomas classied by expression arrays displayed a triple negative phenotype (i.e., 77% were of triple negative phenotype). Taken all together, these results are in accord with the concept that the triple negative phenotype is not an ideal surrogate marker for basal-like breast cancers37,39,42 and call for caution in the interpretation of ongoing therapeutic trials whose selection of patients was made on the basis of lack of ER, PR and HER2 expression.39 Furthermore, there are several lines of evidence to suggest that the group of triple negative cancers is substantially more heterogeneous than the group encompassed by basal-like breast cancers.3739 Although the gold standard for diagnosis of basal-like carcinomas remains microarray-based expression proling, this technology is unlikely to be rolled out as a diagnostic tool in most pathology departments. In fact, several groups have endeavoured to develop surrogate markers for the intrinsic gene list molecular taxonomy. Real-time PCR based methods have already been developed and validated in archival, formalin-xed, paran-embedded tissue samples.43,44 However, for routine management of breast specimens, immunohistochemistry is likely to become the method used to identify these subtypes. Although there is no internationally accepted immunohistochemical denition for basal-like breast cancer,45 Nielsen et al. have demonstrated that, using a panel of four immunohistochemical markers, basal-like carcinomas can be detected with a sensitivity of 76% and a specicity of 100%.17 Basallike carcinomas are thus dened as being ER negative, HER2 negative and positive for expression of cytokeratin 5/6 and/or EGFR. Importantly, this immunohistochemical panel has been shown to be of prognostic signicance in dierent cohorts.38,42 Further renement of this panel by the same group38 has demonstrated that expression of basal markers (i.e., cytokeratin 5/6 and/or EGFR) in ER, PR and HER2 negative cases, identies a subgroup of triple negative cancers with a signicantly worse outcome than patients with triple negative carcinomas that are negative for basal markers. Basal-like carcinomas have been shown to display a rather aggressive clinical behaviour,15,18,41,46,47 with most recurrences happening within the rst ve years after diagnosis.41,47 Late recurrences are reported not to be as common. Basal-like cancers appear to respond to neoadjuvant chemotherapy,14,48,49 however, patients with basal-like tumours, paradoxically, still have a worse outcome when compared with those with tumours pertaining to other molecular subgroups.14,15,18,35,46,48 However, there is substantial circumstantial evidence to suggest that patients with basal-like breast cancer who evolve to pathological complete response following neoadjuvant chemotherapy have an excellent prognosis, whereas those who only display a partial response have remarkably poor outcomes.48,50 This underpins the concept that basal-like cancers are a heterogeneous group in terms of their expression proles, molecular genetic patterns51 and clinical behaviour.41,47 Interestingly, basal-like and tumours arising in BRCA1 mutation carriers have recently been shown to preferentially express markers consistent with a cancer stem cell/ cancer progenitor cell phenotype.52,53 However, it should

MOLECULAR ANALYSIS IN BREAST CANCER

79

be emphasised that not all basal-like cancers display CD44/CD24 cells.52 Interestingly, although breast cancer stem cells are reported to be resistant to chemotherapy and cancer stem cells are more prevalent in basal-like breast cancers, the highest prevalence of pathological complete response following neoadjuvant chemotherapy is found in the subgroup of basal-like cancers.48,50 Although several theories have been advanced to reconcile these contrasting results, they still remain purely theoretical.

GENE-EXPRESSION PROFILING: PROGNOSTIC SIGNATURES


Pathology-derived details have long been used to tailor therapy for cancer patients. Their diligent use has contributed to the reduction of breast cancer mortality and morbidity, with a breast pathologist having a crucial role in the treatment decision making. As an example, sentinel lymph node biopsy has been responsible for great improvement in breast cancer treatment and its recognised high negative predictive value for axillary nodal status54,55 depends on adequate histopathological examination.56,57 Adjuvant!Online (www.adjuvantonline.com, accessed July 2008), Nottingham prognostic index, St Gallen guidelines, and National Institutes of Health (NIH) criteria,58,59 tools often used by oncologists to decide which patients should receive chemotherapy, are based on tumour size, histological grade, ER status, vascular invasion and presence of lymph node metastasis. However, oncologists, pathologists and surgeons would not disagree that the parameters currently available are not sucient to capture the complexity of breast cancer and to tailor therapy for individual patients. Therefore, several research groups have carried out comprehensive microarray gene-expression proling studies with the aim of improving on traditional clinico-pathological parameters. The rst breast cancer prognostic signature described60 was developed through a class prediction analysis of a series of 78 young patients with small (55 cm), lymph node negative cancers who did not receive adjuvant chemotherapy. By comparing the expression proles of tumours from patients who developed distant metastasis within 5 years with those who did not, the authors identied a multi-gene prognostic predictive score comprising 70 genes (MammaPrint; Agendia, The Netherlands). Later the signature was validated in larger cohorts with node positive and node negative patients.61,62 A dierent approach has been undertaken by Wang et al.63 who rst identied 16 and 60 genes associated with relapse in ER positive and ER negative breast carcinomas, respectively, and assembled them into a 76-gene prognostic signature (VDX2 gene chips; Veridex LLC, USA). In the studies above, statistical analyses have arguably shown that the predictive power of these signatures was signicantly superior to the standard clinico-pathological parameters. However, subsequent statistical analyses have called into question the actual contribution of these signatures for breast cancer patient management.6466 Following the attention these two prognostic signatures attracted, several other prognostic signatures have been or are currently being developed. Fan et al.67 applied multiple signatures (i.e., intrinsic subtypes, 70-gene signature,

wound response signature and 21-gene recurrence score) to a cohort of breast cancer patients and conrmed their prognostic value.67 On the other hand, others have failed to nd a superior predictive value for the 70-gene signature when compared with that of regular clinico-pathological features.14,64,65 Although all of these signatures are based on the expression of genes related to similar pathways and to some extent correlate with tumour proliferation,67,68 many were surprised by the fact that the overlap in terms of genes belonging to these signatures is negligible. As a consequence, both pathologists and clinicians face the challenge of which signature should be used. Disappointingly, combining dierent signatures does not seem to result in a signicant improvement of accuracy.67 To complicate matters even further, many doubts regarding the reliability and reproducibility of the technique have arisen.7,69,70 One of the reasons for this apparent failure of microarrays in realising their potential stems from the disparate paces of microarray technology development and the development of bioinformatics and statistics applied to microarray analysis. For instance, in the beginning of this century, data over-tting was a poorly understood concept and methods for power calculation for microarray analysis were yet to be described.71 Fortunately, this eld is maturing very rapidly and currently there are clear guidelines as to how a therapeutically signicant gene signature should be developed and validated.72,73 Unfortunately, none of the microarray-based signatures described to date fulls all criteria required. Although we believe that microarray-based technology, or most likely one of its derivatives, is likely to be incorporated in breast pathology practice, we argue that there are practical issues that need to be resolved before gene expression proling can be translated to routine clinical use. First of all, there are data to suggest that the prognostic power of the gene signatures reported to date is rather limited in ER negative, high grade disease.68 In addition, although microarrays are quite reproducible and can certainly be applied to class discovery studies and preclinical analysis, their accuracy and reproducibility are not sucient for clinical use. For instance, up to 15% of error in qualitative assessment of gene expression has been demonstrated when the optimal samples were analysed with the same platform and protocols in dierent laboratories.74 Furthermore, the applicability of microarrays to readily available, formalin-xed, paran-embedded material is limited, as this technology has been shown to perform suboptimally when RNA extracted from this type of sample is used. Although Illumina (USA; http://www.illumina. com/, accessed July 2008) has recently provided a method for microarray-based gene expression analysis of formalinxed, paran-embedded samples (DASL gene expression), which has been shown to produce reasonable results when applied to matched frozen and formalin-xed, paranembedded specimens,75 neither the 70-gene nor the 76-gene signature has been converted into this platform. As stated above, we anticipate that one of the derivatives of microarray analysis, rather than microarrays themselves, is likely to be incorporated into clinical practice. In fact, quantitative reverse transcriptase PCR (qRT-PCR)-based signatures and immunohistochemical panels have already been developed as alternative methods for expression proling analysis. The prototype of this approach is

80

GEYER et al.

Pathology (2009), 41(1), January

Oncotype DX (Genomic Health, USA),76 which is based on the mRNA expression levels of 21 genes (16 cancer related genes and ve reference genes). These 16 genes comprise components of ER pathway (ER, PGR, BCL2 and SCUBE2), proliferation (Ki67, STK15, Survivin, CCNB1 and MYBL2), HER2 amplicon (HER2 and GRB7), invasion (MMP11 and CTSL2) and GSTM1, CD68 and BAG1. The analysis of the gene expression by qRT-PCR gives rise to the recurrence score, which has been shown to be an independent prognostic factor in early stage, endocrine responsive, tamoxifen treated patients. Patients with a high recurrence score are thought to benet from chemotherapy. Intermediate and low scores are assembled under the good prognosis group and chemotherapy would be of limited value. This assay has proven suitable for the analysis of formalin-xed paran-embedded samples, however the recurrence score was not validated in two studies.77,78 Furthermore, some have argued that it only recapitulates the information provided by grade, ER, HER2 and MIB179 and it remains to be determined if this signature will be of any prognostic value for patients receiving aromatase inhibitors or anthracyline- and taxanebased therapeutic regimens.80

GRADING OF BREAST CANCER: THE END OF NOTTINGHAM GRADE?


Molecular studies have conrmed the importance of histological grade in breast carcinomas and this should not be surprising, given that morphological features visible in H&E sections such as nuclear atypia, tubule formation and mitosis represent the expression of thousands of genes and the interactions of tens of thousands of proteins.81 It has been demonstrated that grade, more than any other clinico-pathological parameter or tumour intrinsic characteristic, is associated with the type, pattern and complexity of molecular changes seen in breast cancer and its precursors.1,2 Ma et al. analysed matched normal, preinvasive and invasive breast lesions of dierent grades by means of expression proling analysis and observed that samples preferentially clustered according to histological grade rather than stage.82 Currently, histological grading of breast carcinomas is one of the cornerstones of treatment decision making. It is performed by histopathological analysis using the modied Bloom and Richardson method,83 also known as Nottingham grade. Despite its importance, concordance between pathologists has been reported to be suboptimal.84 In fact intra- and inter-observer agreement ranges from 50% to 86%. The clinical signicance of grade I and grade III are well dened, but only about half of breast carcinomas are classied as such. Often oncologists have to care for a patient with a grade II invasive ductal carcinoma of no special type, which is not helpful in clinical decision making. This has prompted several groups to investigate methods that could rene the current grading system, making it more reproducible and assigning high and low risk subgroups. Sotiriou et al. undertook a hypothesis-driven approach focusing on histological grade.85 They analysed grade I and grade III ER positive breast carcinomas using expression arrays and proposed the gene expression

grade index (GGI). GGI is based on 97 genes, most of them are associated to cell cycle progression and proliferation. Indeed, proliferation has been reported to be the most important predictor of outcome in breast cancer, especially in ER positive patients.86 GGI could identify grade I and grade III tumours in the validation set with an accuracy of *90% and was more strongly associated with relapse-free survival than histological grade. Importantly, it was shown that GGI could stratify grade II tumours into genomic low grade and genomic high grade and that these groups were of prognostic signicance: histological grade II GGI low tumours had outcomes similar to GGI low cancers, whereas histological grade II GGI high tumours had outcomes similar to GGI high cancers. GGI has been subsequently validated by the same group.87 Another genetic grade signature was proposed by Ivshina et al.88 who proled 347 breast invasive tumours with expression arrays. Using two class prediction algorithms, six grade-associated genes were identied and accurately classied grade I and grade III tumours in low and high genomic grade, respectively. Grade II tumours were not a molecularly distinct group but could be separated into grade IIa (low grade-like) or grade IIb (high grade-like). Grade IIa patients had a signicantly better outcome than grade IIb patients, whereas no dierence was observed between the grade IIa and grade I survival curves, or the grade IIb and grade III curves. These results have been further validated by the authors in datasets generated with dierent microarray platforms.88 However, further analyses have shown that there were signicant molecular and clinico-pathological dierences between the grade I and grade IIa tumours and the grade IIb and grade III tumours. More recently, using a similar approach, Ma et al. reported a ve-gene molecular grade index.89 In a way akin to the signatures described above, this molecular grade index could classify grade I and grade III tumours with 89% accuracy and grade II tumours were stratied into two clinically signicant groups. The results were validated in a qRT-PCR assay suitable for formalin-xed, paranembedded clinical samples. Therefore, the authors argued that this signature could be incorporated in the daily practice, as it removes the subjectivity and inter-/intraobserver variability associated with conventional histological grading. It has also been proposed that grading signatures could replace histological grade in currently used prognostic models such as Adjuvant!Online and Nottingham prognostic index.68,86,88 Ivshina et al. have described that incorporating their genetic grade signature into the Nottingham prognostic index, some patients classied as having worse prognosis shifted to the good prognosis group. Hence, the use of genetic grade may help improve the identication of patients who could be spared toxic adjuvant systemic therapy.88 Although there is a great enthusiasm about genomic grading, additional independent validation of the above genomic grade signatures is still required and their discriminatory power in ER negative tumours remains to be determined. Therefore, despite the issues related to reproducibility of Nottingham histological grade, histological grade still remains a useful and less expensive tool to help tailor the therapy of breast cancer patients.

MOLECULAR ANALYSIS IN BREAST CANCER

81

PREDICTIVE SIGNATURES AND BIOMARKERS


Hormone receptors and HER2 are the only mandatory biomarkers to be tested for breast cancer patient decision making, and tamoxifen and trastuzumab represent two of the few successful examples of targeted therapy in breast cancer. In recent years, a paradigm shift has occurred in the way mechanisms of response or resistance to therapy and the predictive value of a biomarker are perceived. There are data to suggest that the signicance of predictive markers may depend on the molecular subtype of the tumour.90,91 Biological pathways might be important in one particular molecular subtype, such as TP53 mutation in ER positive chemotherapy resistant tumours;92 whereas the impact of TP53 mutation in ER negative disease may dier. Therefore, studies designed to discover new predictive markers of response to systemic therapy should take into account the distinctive molecular features of the dierent subtypes, otherwise they might under- or over-estimate their performance (Fig. 1).86,90,91 Several groups have tried to identify reliable predictive factors in the context of tamoxifen treated patients.93,94 Tamoxifen is the most frequently prescribed anti-oestrogen agent in women with ER positive breast carcinomas in both

early stage and advanced disease; however, a signicant proportion of patients relapse due to intrinsic or acquired resistance. Ma et al. analysed the gene expression prole of 60 early-stage tamoxifen-treated patients and identied three genes associated with outcome: HOXB13, IL17BR and CHDH.94 High levels of HOXB13 mRNA and low levels of IL17BR mRNA are associated with recurrence. A simple two-gene ratio HOXB13:IL17BR was then proposed as a novel biomarker to predict recurrence in tamoxifen treated patients. This group developed a qRT-PCR assay and achieved comparable results. Although initial validation studies failed to demonstrate the predictive power of this two-gene signature,95 subsequent studies validated the results in two dierent cohorts and showed the ratio has prognostic power and is predictive of tamoxifen response.96,97 In both cohorts the ratio was not signicant in node-positive patients,96,97 explaining its lack of predictive value in the study by Reid et al.95 Moreover, it has also been shown that the ratio has complementary prognostic value when combined with the ve-gene molecular grade index described above.89 HER2 over-expression and HER2 gene amplication are both predictors of response to humanised monoclonal antibodies anti-HER2 or to HER2 tyrosine kinase

FIG. 1 Schematic illustration of data integration for prognostic/predictive markers and therapeutic targets identication based on high-throughput molecular methods. The term oncogene addiction has been used to describe the phenomenon in which tumour cells become reliant upon the activity of specic oncogenes for growth and progression.163 Consequently, identifying and inhibiting the activity or expression of the proteins encoded by these addictive oncogenes is a highly attractive therapeutic strategy in human cancer.164 (A) Representative genome plot of DNA copy number and chromosome position of a HER2 amplied breast carcinoma. Log2 ratios are plotted on the Y axis against each clone according to genomic location on the X axis. The centromere is represented by a vertical dotted line. BACs categorised as displaying genomic gains or amplication are plotted in green and those categorised as genomic losses in red. (B) Representative image of a scanned microarray chip. Red, relative high expression; green, relative low expression. (C) Representative micrograph of an invasive ductal carcinoma harbouring HER2 amplication by CISH. (D) Representative micrograph of an invasive ductal carcinoma displaying HER2 over-expression by IHC. ArrayCGH, microarray comparative genomic hybridisation; FISH, uorescent in situ hybridisation; CISH, chromogenic in situ hybridisation; qRT-PCR, quantitative reverse transcriptase PCR; IHC, immunohistochemistry; siRNA, small interfering RNA; shRNA, short hairpin RNA.

82

GEYER et al.

Pathology (2009), 41(1), January

inhibitors.413 However, there is strong evidence to suggest that current methods to determine which patients should receive trastuzumab or HER2 tyrosine kinase inhibitors have good negative predictive value, but suboptimal positive predictive values. In the last years, several mechanisms of de novo and acquired resistance to trastuzumab have been identied. It is currently accepted that PIK3CA activating mutations, PTEN inactivation, IGF1R overexpression or expression of p95 HER2 isoform may all play roles in both de novo or acquired resistance to therapies that target HER2.3,98 Furthermore, it has recently been suggested that HER2 amplied breast cancers that harbour a basal-like transcriptome are less sensitive to anti-HER2 therapies.99 Therefore, one can anticipate that additional molecular testing of HER2 positive cancers is likely to be incorporated as predictive markers for patients with tumours pertaining to this molecular subgroup. In addition, HER2 has been reported to predict response to anthracycline-based chemotherapy.100,101 Although HER2 is considered to be the target gene for amplication at chromosome 17q12-q21, the HER2 amplicon encompasses several other genes, such as TOPO2A. TOPO2A gene encodes for topoisomerase-IIa, the direct molecular target of anthracycline,102 it is often co-amplied with HER2 and its amplication seems to be restricted to HER2 amplied tumours.103106 In this context TOPO2A has emerged as a new putative predictor biomarker of response, as it has been shown that tumours with HER2/TOPO2A co-amplication show a signicantly better response to anthracycline-based chemotherapy.104,107,108

TUMOUR MICROENVIRONMENT AND STROMAL SIGNATURES


The major focus of cancer research has been on the malignant cell itself. Most of the analyses described above were performed using whole tissue samples and those judged to possess insucient tumour epithelial cell content were generally excluded. Thus, the specic contribution of epithelial cells, stromal cells and extra-cellular matrix to these tumour classiers and prognostic/predictive signatures remains uncertain. However, some groups have focused their studies on the tumour microenvironment in breast cancer.109116 Comprehensive gene expression prole of each cell type has shown that changes at the transcriptional level occur in epithelial, myoepithelial and stromal cells, already evident at the in situ carcinoma stage.109 However, genetic changes detected by microarray-based comparative genomic hybridisation appear to be limited to cancer epithelial cells.109 The studies of epithelial-mesenchymal interactions have brought some interesting insights about cancer progression.112,114,116 As dierent groups have failed to detect signicant dierences in genetic alterations between ductal carcinoma in situ (DCIS) and invasive tumours,82 the progression of in situ carcinoma to invasive carcinoma may not be due to the intrinsic properties of the tumour epithelial cells but determined by complex interactions between epithelial cells and all the cell types that compose the tumour microenvironment.112,116 It has been hypothesised that progression to invasion may be promoted by broblasts and inhibited by myoepithelial cells.116

Based on the above concepts, it is perhaps not surprising that prognostic signatures derived from the proles of stromal cells have been developed. Using gene signatures of two broblastic tumours, desmoid-type bromatosis (DTF) and solitary brous tumour (SFT), West et al.115 could stratify a breast carcinoma dataset in at least two subgroups with prognostic value independent of clinicopathological risk factors, suggesting that there might be distinct broblastic reaction patterns in breast cancer. The rst cluster expressed DTF genes and was associated with a good prognosis, whereas the second was less homogeneous and composed of two subgroups enriched for SFT genes. These results have been conrmed in dierent datasets and, at the protein level, breast cancers with strong expression of one single gene (SPARC) of the DTF cluster have shown a trend for increased survival.110 However, it should be noted that a previous study demonstrated that SPARC expression in a large consecutive series of breast cancers was associated with poor prognosis.117 One could hypothesise that the DTF stromal reaction pattern would be similar to that seen in the desmoplastic stromal pattern typically found in tubular carcinomas; however, no correlation between molecular and morphological features has been described as yet. Using a dierent approach, Bergamaschi et al.111 grossly dissected 28 breast cancers, with some retention of extracellular matrix, and carried out an unsupervised hierarchical clustering analysis. This analysis revealed that tumours were classied into four dierent groups according to the expression of 278 selected extra-cellular matrix-related genes. Interestingly, in the validation set, the authors found a strong association between one of the groups (Group 1) and high grade, ER negativity, TP53 mutations and poor outcome. When the ve previously dened molecular subtypes4,5 were compared with the stromal signature, it was observed that 81% of all basal-like, 4% of luminal A and 23% of luminal B tumours fell into Group 1. The luminal A and B tumours falling into this group displayed a signicantly worse outcome than other luminal tumours, indicating that the extra-cellular matrix signature can give additional information and is partly independent of the intrinsic subgroups. Furthermore, the authors identied three genes out of the 278 previously selected that were associated with prognosis. Tumours harbouring high expression levels of MARCO and low levels of PUNC and SPARC displayed a signicantly worse outcome. More recently, Finak et al.113 have isolated tumour stroma and matched normal stroma from breast tumours and derived a 26-gene signature strongly associated with outcome called stroma-derived prognosis predictor (SDPP). SDPP predicted prognosis with greater accuracy than the 70-gene signature (MammaPrint), could also stratify several published whole-tumour derived datasets and was independent of grade, age, lymph node involvement, chemotherapy, hormonal therapy and both ER and HER2 status. The poor outcome cluster showed enrichment for markers of hypoxic and angiogenic response, whereas the good outcome cluster over-expressed immune response genes. Interestingly, an immune response gene expression signature has been shown to identify a good outcome subtype of ER negative patients.118 Surprisingly, this subtype was not related either to the extent of lymphocytic

MOLECULAR ANALYSIS IN BREAST CANCER

83

inltrate, medullary carcinomas or BRCA mutation status. The immune system seems to play an important role in a subgroup of ER negative patients who may benet from treatments targeting tumour cells via the immune response, such as vaccine therapies.113 Interestingly, a small percentage of basal-like carcinomas, a group still without reliable prognostic markers or signatures that reliably identify those with good or poor prognosis, was included in this subtype.118

MOLECULAR GENETICS OF BREAST CANCER: FROM GENOMIC ARCHITECTURE TO BREAST CANCER GENETIC PATHWAYS
The concept that breast cancer encompasses a plethora of entities with distinctive biological characteristics and clinical behaviour is also underpinned at the molecular genetic level by a complex array of genetic alterations that aect the function and control of individual genes and cellular processes.1,2 Not only expression proling analysis, but also the study of cancer genetics has had a profound impact on our understanding of the evolutionary pathways and causative factors in the initiation, development and progression of breast cancer. Microarray-based comparative genomic hybridisation (aCGH) enables researchers to perform rapid and high resolution screening of genomes and a detailed analysis of the global copy number changes.119 While expression proling analysis looks at the transcriptional level, aCGH

scrutinises the genome-wide pattern of genetic alterations. Reliable results using DNA extracted from formalin-xed, paran-embedded samples can be achieved, in particular if BAC arrays are used. aCGH studies in breast cancer have shown that copy number changes are associated with dierent clinico-pathological features to the geneexpression subtypes previously described4,5 and to dierent outcomes.120,121 Three patterns of genomic changes have been recently described, probably related to dierent mechanisms of DNA repair defects found in breast cancers (Fig. 2).121 The rst pattern is called simplex and is dened by broad segments of gains and losses, usually comprising entire chromosomes or chromosome arms. Tumours with this pattern often display gains of 1q and losses of the whole 16q arm. This pattern accounts for 60% of diploid tumours and is related to low histological grade and luminal A phenotype.122 The remaining tumours fall into a category called complex. Tumours with a complex genomic architecture have been shown to have shorter survival in retrospective analysis and to comprise at least two distinct patterns: sawtooth (25%) and restorm or amplier (5%).121,122 The sawtooth pattern is characterised by many segments of gains and losses of varying sizes, often alternating; copy number changes aect almost the entire genome, however loci with high level amplication are usually not seen. The restorm pattern resembles the simplex, but shows at least one localised region of clustered, narrow peaks of amplication, with each cluster conned to

FIG. 2 Schematic illustration of the genomic patterns in breast cancer, as described by Hicks et al.,121 and their association with molecular characteristics, histological features and clinical variables. At the top representative genome plots of DNA copy number and chromosome position of the three distinct genomic proles. Log2 ratios are plotted on the Y axis against each clone according to genomic location on the X axis. The centromere is represented by a vertical dotted line. BACs categorised as displaying genomic gains or amplication are plotted in green and those categorised as genomic losses in red. *Chin et al.122 **Basal-like carcinomas do not display as many high level amplications as HER2 and luminal B carcinomas and they are predominantly of sawtooth pattern; HER2 carcinomas are typically of complex-amplier/restorm pattern.

84

GEYER et al.

Pathology (2009), 41(1), January

a single chromosome arm. Interestingly, complex patterns, in particular the restorm pattern, were associated with a worse outcome. Historically, breast cancer progression pathways were seen as a multistep model, similar to that described by Vogelstein et al.123 for colorectal cancer, where normal breast epithelial cells would transform and progress through a series of morphologically identiable precursors (i.e., normal hyperplasia of usual type atypical ductal hyperplasia low grade DCIS high grade DCIS invasive carcinoma).2 However, in the last few years it has become clear that breast cancer molecular pathways are much more complex than previously appreciated.1,2,124127 In fact, breast cancer molecular pathways encompass a network of inter-related multi-step pathways, which can be broadly classied into two groups/arms, based on histological grade.1,2 The low grade arm encompasses most of the precursors pathologists are aware of, including columnar cell changes, atypical ductal hyperplasia, lobular neoplasia (ALH/LCIS), and their invasive counterparts. These lesions are characterised by low nuclear grade, consistent positivity for hormone receptors and lack of HER2 expression, and quite simple diploid/near diploid karyotypes, with few recurrent changes: deletions of 16q and gains of 1q and 16p.1,2 On the other hand, the high grade arm encompasses high grade DCIS and invasive high grade invasive ductal carcinoma. These tumours frequently lack hormone receptors and express HER2 or basal markers. At the genetic level, tumours of the high grade arm are aneuploid, have complex karyotypes, with a plethora of unbalanced changes mapping to several chromosomal arms.1,2 Interestingly, 510% of high grade breast cancers harbour deletions of 16q and even when they do, the mechanisms leading to 16q deletions are dierent from those described in low grade breast cancer.128 Taken together, these lines of evidence support the idea that progression from low to high grade breast cancer is an exceedingly rare biological phenomenon. This is further supported by the fact that recurrences of DCIS have similar histological grade and molecular genetic features when compared with those of index cases.129 Another line of evidence stems from expression array analysis of matched atypical ductal hyperplasia/at epithelial atypia, DCIS and invasive breast carcinomas.1,2 As mentioned above, Ma et al. have demonstrated that lesions of similar histological grade rather than progression stage cluster together, suggesting that dierent histological grades are associated with distinct gene expression signatures. Last but not least, this is further corroborated by a recent observation that a mixture of grade I, grade II and grade III DCIS is seen in only 9.2% of cases.130 Results of molecular genetic studies, on the other hand, have blurred the boundaries between low grade ductal carcinomas and lobular neoplasia.1,2 Both show remarkably similar immunohistochemical and molecular genetic proles, the main dierence being the target gene of 16q losses.1,2,131 Whilst in ductal lesions the target gene remains to be identied,132134 in lobular carcinomas it has been proven to be the CDH1 gene.135,136 This gene encodes E-cadherin, an adhesion molecule that mediates homophylic-homotypic adhesions. This gene is reported to be inactivated in lobular neoplasia and invasive lobular carcinomas through a combination of genetic and epigenetic mechanisms.137,138 Moreover, identical CDH1 gene

mutations have been described in matched lobular neoplasia and invasive lobular cancer,136 providing strong evidence to suggest that at least some examples of lobular neoplasia are non-obligate precursors of invasive lobular carcinoma. Given the consistent down-regulation of Ecadherin in lobular lesions, anti-E-cadherin antibodies have proven to be useful in the diagnosis of lesions with indeterminate phenotype and in the characterisation of the pleomorphic variant of lobular carcinoma.139,140 Our group and others have demonstrated that pleomorphic LCIS and invasive pleomorphic lobular carcinoma are variants of classic lobular cancer, harbouring similar molecular genetic changes, including deletions of 16q141 144 and E-cadherin down-regulation.141144 However, pleomorphic LCIS (PLCIS) and invasive pleomorphic lobular carcinomas (iPLC) harbour additional genetic hits mapping to key oncogenes, such as MYC and HER2, which may account for the high nuclear grade and the more aggressive clinical behaviour reported.143,144 Given that PLCIS and iPLC are genetically advanced lesions, which are remarkably similar at the morphological, immunohistochemical and molecular genetic levels, PLCIS should be considered a direct non-obligate precursor of iPLC143,144 and managed less conservatively than classic lobular neoplasia. Molecular genetic analysis has also helped clarify the role of some putative breast cancer precursors. Some lesions once considered part of breast cancer progression pathways (e.g., hyperplasia of usual type) have been shown to harbour rare and fairly random chromosomal changes and their role as precursors have been called into question.1,2,145147 On the other hand, other lesions such as columnar cell changes/at epithelial atypia,148 have proven to be clonal and neoplastic, and to harbour molecular genetic changes similar to those found in low grade ductal carcinomas, including 16q deletions. In addition, apocrine lesions have also been shown to harbour recurrent unbalanced chromosomal changes;149 however, their actual role in breast cancer molecular pathways is yet to be determined. Interestingly, a recent morphological study of low grade breast lesions has given further support for the concept of a low grade breast neoplasia family, which encompasses columnar cell changes/at epithelial atypia, atypical ductal hyperplasia, low grade DCIS, lobular neoplasia and their respective invasive counterparts: tubular/tubularcribriform breast carcinomas, grade I invasive ductal carcinomas, invasive lobular carcinomas and tubulolobular carcinomas.150 These tumours seem to have a typical luminal A phenotype and, as mentioned above, are characterised by positivity for hormone receptors, deletions of 16q and have a rather indolent clinical course.2,7,15,46,148 Based on these lines of evidence it seems reasonable to consider at epithelial atypia, low grade ductal and lobular lesions as part of the same group of lesions. Furthermore, a recent expression proling analysis of special types of breast cancer has demonstrated the existence of a group of ER positive cancers encompassing classic lobular carcinomas and tubular carcinomas.131 On the other hand, there are few similarities between low grade ER positive and high grade ER negative ductal carcinomas in terms of their morphological, immunohistochemical and molecular features, suggesting that they constitute distinct entities.

MOLECULAR ANALYSIS IN BREAST CANCER

85

BREAST CANCER SPECIAL TYPES: GENOTYPIC-PHENOTYPIC CORRELATION


Although histological grade has been extensively analysed from a molecular perspective, histological type has not received the same attention. It should be noted, however, that an obvious example of genotypic-phenotypic correlation in breast cancer is the secretory carcinoma of the breast. This tumour, which has an indolent clinical behaviour and not uncommonly aects prepubertal patients, has been shown to consistently harbour a t(13;15) chromosomal translocation,151 involving the genes ETV6 and NTRK3.151154 Subsequent studies have shown that this translocation in breast cancer is specic to secretory breast carcinomas and that even tumours once thought to be variants of this special type (i.e., acinic cell carcinomas of the breast) do not harbour this translocation.155 As mentioned above, there are several lines of evidence to suggest a genotypic-phenotypic correlation between BRCA1 germline mutations and specic morphological features of breast cancer. When compared with sporadic controls, BRCA1 tumours have been shown to be enriched for medullary cancers, to be of histological grade III, have higher mitotic counts, pushing borders and brisk lymphocytic inltrate.156158 At the immunohistochemical level, BRCA1 cancers have been shown to more often lack hormone receptors and HER2 when compared with controls157159 and to preferentially be of basal-like phenotype.158,160,161 The link between BRCA1 mutations and the phenotypic characteristics listed above is further conrmed by two engineered mouse models developed by two independent groups, where Brca1 and Trp53 were inactivated in the epithelial cells of the mouse mammary gland.27,28,158 Tumours developing in these animals were characterised by the same constellation of morphological features and immunohistochemical prole as described for human BRCA1 tumours. A recent detailed expression proling analysis of special types of breast cancer demonstrated that some of the histological types of breast cancer have a distinctive transcriptome.131 One of the best examples of this phenomenon is invasive micropapillary carcinoma, which displayed a luminal phenotype, but consistently formed a distinct cluster in hierarchical clustering analysis.131 These results are in accord with our ndings that demonstrate that micropapillary carcinomas of the breast have an immunohistochemical phenotype and a pattern of genetic aberrations that are consistent with a luminal B phenotype.162 On the other hand, Weigelt et al.131 demonstrated that other special types of breast cancer, such as apocrine carcinoma, may not constitute a distinct entity. From a pathologists standpoint, the study of the molecular features of special types of breast cancer may reveal not only the basis for histological type, but also help identify novel therapeutic targets for these tumours, given that each subtype has been proven to be more homogeneous at the molecular level than invasive ductal carcinomas of no special type.131

tailored therapies, a paradigm shift from morphological and prognostic classication systems to predictive models is of paramount importance. It is anticipated that by combining morphological, immunohistochemial and molecular techniques, a more biologically and clinically meaningful classication of breast cancer and its precursors will emerge. ACKNOWLEDGMENTS Jorge S. Reis-Filho and Felipe C. Geyer are funded by Breakthrough Breast Cancer. Caterina ` is part of the PhD programme Tecniche avanzate di Marchio localizzazione dei tumori umani (University of Turin) and is funded in part by AIRC (Milam, Regional Grant 1182) and Breakthrough Breast Cancer. The authors would like to thank the Microarray Laboratory of the Breakthrough Breast Cancer Research Centre for the courtesy of the microarray gene expression image.
Address for correspondence: Dr J. S. Reis-Filho, The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, United Kingdom. E-mail: Jorge.Reis-Filho@icr.ac.uk

References
1. Reis-Filho JS, Simpson PT, Gale T, Lakhani SR. The molecular genetics of breast cancer: the contribution of comparative genomic hybridization. Pathol Res Pract 2005; 201: 71325. 2. Simpson PT, Reis-Filho JS, Gale T, Lakhani SR. Molecular evolution of breast cancer. J Pathol 2005; 205: 24854. 3. Nahta R, Esteva FJ. Trastuzumab: triumphs and tribulations. Oncogene 2007; 26: 363743. 4. Perou CM, Sorlie T, Eisen MB, et al. Molecular portraits of human breast tumours. Nature 2000; 406: 74752. 5. Sorlie T, Perou CM, Tibshirani R, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA 2001; 98: 1086974. 6. Brenton JD, Carey LA, Ahmed AA, Caldas C. Molecular classication and molecular forecasting of breast cancer: ready for clinical application? J Clin Oncol 2005; 23: 735060. 7. Reis-Filho JS, Westbury C, Pierga JY. The impact of expression proling on prognostic and predictive testing in breast cancer. J Clin Pathol 2006; 59: 22531. 8. Sorlie T, Tibshirani R, Parker J, et al. Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci USA 2003; 100: 841823. 9. Geyer CE, Forster J, Lindquist D, et al. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med 2006; 355: 273343. 10. Joensuu H, Kellokumpu-Lehtinen PL, Bono P, et al. Adjuvant docetaxel or vinorelbine with or without trastuzumab for breast cancer. N Engl J Med 2006; 354: 80920. 11. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med 2005; 353: 165972. 12. Romond EH, Perez EA, Bryant J, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med 2005; 353: 167384. 13. Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 2001; 344: 78392. 14. Rouzier R, Perou CM, Symmans WF, et al. Breast cancer molecular subtypes respond dierently to preoperative chemotherapy. Clin Cancer Res 2005; 11: 567885. 15. Abd El-Rehim DM, Pinder SE, Paish CE, et al. Expression of luminal and basal cytokeratins in human breast carcinoma. J Pathol 2004; 203: 66171. 16. Gusterson BA, Ross DT, Heath VJ, Stein T. Basal cytokeratins and their relationship to the cellular origin and functional classication of breast cancer. Breast Cancer Res 2005; 7: 1438. 17. 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: 536774. 18. Rakha EA, Putti TC, Abd El-Rehim DM, et al. Morphological and immunophenotypic analysis of breast carcinomas with basal and myoepithelial dierentiation. J Pathol 2006; 208: 495506.

CONCLUSION
In conclusion, high throughput molecular techniques are reshaping the way breast cancer is perceived. In this era of

86

GEYER et al.

Pathology (2009), 41(1), January

19. Savage K, Lambros MB, Robertson D, et al. Caveolin 1 is overexpressed and amplied in a subset of basal-like and metaplastic breast carcinomas: a morphologic, ultrastructural, immunohistochemical, and in situ hybridization analysis. Clin Cancer Res 2007; 13: 90101. 20. Savage K, Leung S, Todd SK, et al. Distribution and signicance of caveolin 2 expression in normal breast and invasive breast cancer: an immunouorescence and immunohistochemical analysis. Breast Cancer Res Treat 2008; 110: 24556. 21. Turner NC, Reis-Filho JS. Basal-like breast cancer and the BRCA1 phenotype. Oncogene 2006; 25: 584653. 22. van de Rijn M, Perou CM, Tibshirani R, et al. Expression of cytokeratins 17 and 5 identies a group of breast carcinomas with poor clinical outcome. Am J Pathol 2002; 161: 19916. 23. Fulford LG, Easton DF, Reis-Filho JS, et al. Specic morphological features predictive for the basal phenotype in grade 3 invasive ductal carcinoma of breast. Histopathology 2006; 49: 2234. 24. Livasy CA, Karaca G, Nanda R, et al. Phenotypic evaluation of the basal-like subtype of invasive breast carcinoma. Mod Pathol 2006; 19: 26471. 25. Reis-Filho JS, Milanezi F, Steele D, et al. Metaplastic breast carcinomas are basal-like tumours. Histopathology 2006; 49: 1021. 26. Turner NC, Reis-Filho JS, Russell AM, et al. BRCA1 dysfunction in sporadic basal-like breast cancer. Oncogene 2007; 26: 212632. 27. Liu X, Holstege H, van der Gulden H, et al. Somatic loss of BRCA1 and p53 in mice induces mammary tumors with features of human BRCA1-mutated basal-like breast cancer. Proc Natl Acad Sci USA 2007; 104: 121116. 28. McCarthy A, Savage K, Gabriel A, et al. A mouse model of basal-like breast carcinoma with metaplastic elements. J Pathol 2007; 211: 38998. 29. Dairkee SH, Mayall BH, Smith HS, Hackett AJ. Monoclonal marker that predicts early recurrence of breast cancer. Lancet 1987; 1: 514. 30. Gould VE, Koukoulis GK, Jansson DS, et al. Coexpression patterns of vimentin and glial lament protein with cytokeratins in the normal, hyperplastic, and neoplastic breast. Am J Pathol 1990; 137: 114355. 31. Malzahn K, Mitze M, Thoenes M, Moll R. Biological and prognostic signicance of stratied epithelial cytokeratins in inltrating ductal breast carcinomas. Virchows Arch 1998; 433: 11929. 32. Murad TM, Scharpelli DG. The ultrastructure of medullary and scirrhous mammary duct carcinoma. Am J Pathol 1967; 50: 33560. 33. Santini D, Ceccarelli C, Taurelli M, Pileri S, Marrano D. Dierentiation pathways in primary invasive breast carcinoma as suggested by intermediate lament and biopathological marker expression. J Pathol 1996; 179: 38691. 34. Kreike B, van Kouwenhove M, Horlings H, et al. Gene expression proling and histopathological characterization of triple-negative/ basal-like breast carcinomas. Breast Cancer Res 2007; 9: R65. 35. Banerjee S, Reis-Filho JS, Ashley S, et al. Basal-like breast carcinomas: clinical outcome and response to chemotherapy. J Clin Pathol 2006; 59: 72935. 36. Rakha EA, El-Sayed ME, Green AR, et al. Prognostic markers in triple-negative breast cancer. Cancer 2007; 109: 2532. 37. Rakha EA, Tan DS, Foulkes WD, et al. Are triple-negative tumours and basal-like breast cancer synonymous? Breast Cancer Res 2007; 9: 404; author reply 405. 38. Cheang MC, Voduc D, Bajdik C, et al. Basal-like breast cancer dened by ve biomarkers has superior prognostic value than triplenegative phenotype. Clin Cancer Res 2008; 14: 136876. 39. Bertucci F, Finetti P, Cervera N, et al. How basal are triple-negative breast cancers? Int J Cancer 2008; 123: 23640. 40. Calza S, Hall P, Auer G, et al. Intrinsic molecular signature of breast cancer in a population-based cohort of 412 patients. Breast Cancer Res 2006; 8: R34. 41. Jumppanen M, Gruvberger-Saal S, Kauraniemi P, et al. Basal-like phenotype is not associated with patient survival in estrogen-receptornegative breast cancers. Breast Cancer Res 2007; 9: R16. 42. Tan DS, Marchio C, Jones RL, et al. Triple negative breast cancer: molecular proling and prognostic impact in adjuvant anthracyclinetreated patients. Breast Cancer Res Treat 2008; 111: 2744. 43. Parker J, Mullins M, Cheang MC, et al. A supervised risk predictor of breast cancer based on biological subtypes. J Clin Oncol 2008; 26: (in press). 44. Perreard L, Fan C, Quackenbush JF, et al. Classication and risk stratication of invasive breast carcinomas using a real-time quantitative RT-PCR assay. Breast Cancer Res 2006; 8: R23. 45. Rakha EA, Reis-Filho JS, Ellis IO. Basal-like breast cancer: a critical review. J Clin Oncol 2008; 26: 256881. 46. Abd El-Rehim DM, Ball G, Pinder SE, et al. High-throughput protein expression analysis using tissue microarray technology of a large well-characterised series identies biologically distinct classes of breast cancer conrming recent cDNA expression analyses. Int J Cancer 2005; 116: 34050.

47. Fulford LG, Reis-Filho JS, Ryder K, et al. Basal-like grade III invasive ductal carcinoma of the breast: patterns of metastasis and long-term survival. Breast Cancer Res 2007; 9: R4. 48. Carey LA, Dees EC, Sawyer L, et al. The triple negative paradox: primary tumor chemosensitivity of breast cancer subtypes. Clin Cancer Res 2007; 13: 232934. 49. Dent R, Trudeau M, Pritchard KI, et al. Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res 2007; 13: 442934. 50. 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: 127581. 51. Jones C, Ford E, Gillett C, et al. Molecular cytogenetic identication of subgroups of grade III invasive ductal breast carcinomas with dierent clinical outcomes. Clin Cancer Res 2004; 10: 598897. 52. Honeth G, Bendahl PO, Ringner M, et al. The CD44/CD24phenotype is enriched in basal-like breast tumors. Breast Cancer Res 2008; 10: R53. 53. Wright MH, Calcagno AM, Salcido CD, et al. Brca1 breast tumors contain distinct CD44/CD24- and CD133 cells with cancer stem cell characteristics. Breast Cancer Res 2008; 10: R10. 54. Krag D, Weaver D, Ashikaga T, et al. The sentinel node in breast cancera multicenter validation study. N Engl J Med 1998; 339: 941 6. 55. Veronesi U, Paganelli G, Galimberti V, et al. Sentinel-node biopsy to avoid axillary dissection in breast cancer with clinically negative lymph-nodes. Lancet 1997; 349: 18647. 56. Rutgers EJ. Sentinel node biopsy: interpretation and management of patients with immunohistochemistry-positive sentinel nodes and those with micrometastases. J Clin Oncol 2008; 26: 698702. 57. Viale G, Maiorano E, Mazzarol G, et al. Histologic detection and clinical implications of micrometastases in axillary sentinel lymph nodes for patients with breast carcinoma. Cancer 2001; 92: 137884. 58. Eifel P, Axelson JA, Costa J, et al. National Institutes of Health Consensus Development Conference Statement: adjuvant therapy for breast cancer, November 13, 2000. J Natl Cancer Inst 2001; 93: 979 89. 59. Goldhirsch A, Wood WC, Gelber RD, et al. Meeting highlights: updated international expert consensus on the primary therapy of early breast cancer. J Clin Oncol 2003; 21: 335765. 60. vant Veer LJ, Dai H, van de Vijver MJ, et al. Gene expression proling predicts clinical outcome of breast cancer. Nature 2002; 415: 5306. 61. Buyse M, Loi S, vant Veer L, et al. Validation and clinical utility of a 70-gene prognostic signature for women with node-negative breast cancer. J Natl Cancer Inst 2006; 98: 118392. 62. van de Vijver MJ, He YD, vant Veer LJ, et al. A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med 2002; 347: 19992009. 63. Wang Y, Klijn JG, Zhang Y, et al. Gene-expression proles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet 2005; 365: 6719. 64. Dunkler D, Michiels S, Schemper M. Gene expression proling: does it add predictive accuracy to clinical characteristics in cancer prognosis? Eur J Cancer 2007; 43: 74551. 65. Eden P, Ritz C, Rose C, Ferno M, Peterson C. Good Old clinical markers have similar power in breast cancer prognosis as microarray gene expression prolers. Eur J Cancer 2004; 40: 18371841. 66. Lu X, Lu X, Wang ZC, et al. Predicting features of breast cancer with gene expression patterns. Breast Cancer Res Treat 2008; 108: 191201. 67. Fan C, Oh DS, Wessels L, et al. Concordance among gene-expressionbased predictors for breast cancer. N Engl J Med 2006; 355: 5609. 68. Sotiriou C, Piccart MJ. Taking gene-expression proling to the clinic: when will molecular signatures become relevant to patient care? Nat Rev Cancer 2007; 7: 54553. 69. Ioannidis JP. Microarrays and molecular research: noise discovery? Lancet 2005; 365: 4545. 70. Michiels S, Koscielny S, Hill C. Interpretation of microarray data in cancer. Br J Cancer 2007; 96: 11558. 71. Ein-Dor L, Zuk O, Domany E. Thousands of samples are needed to generate a robust gene list for predicting outcome in cancer. Proc Natl Acad Sci USA 2006; 103: 59238. 72. Simon R. Roadmap for developing and validating therapeutically relevant genomic classiers. J Clin Oncol 2005; 23: 733241. 73. Simon R, Radmacher MD, Dobbin K, McShane LM. Pitfalls in the use of DNA microarray data for diagnostic and prognostic classication. J Natl Cancer Inst 2003; 95: 148. 74. Shi L, Reid LH, Jones WD, et al. The MicroArray Quality Control (MAQC) project shows inter- and intraplatform reproducibility of gene expression measurements. Nat Biotechnol 2006; 24: 115161.

MOLECULAR ANALYSIS IN BREAST CANCER

87

75. Ravo M, Mutarelli M, Ferraro L, et al. Quantitative expression proling of highly degraded RNA from formalin-xed, paranembedded breast tumor biopsies by oligonucleotide microarrays. Lab Invest 2008; 88: 43040. 76. 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: 281726. 77. Esteva FJ, Sahin AA, Cristofanilli M, et al. Prognostic role of a multigene reverse transcriptase-PCR assay in patients with nodenegative breast cancer not receiving adjuvant systemic therapy. Clin Cancer Res 2005; 11: 33159. 78. Mina L, Soule SE, Badve S, et al. Predicting response to primary chemotherapy: gene expression proling of paran-embedded core biopsy tissue. Breast Cancer Res Treat 2007; 103: 197208. 79. Flanagan MB, Dabbs DJ, Brufsky AM, Beriwal S, Bhargava R. Histopathologic variables predict Oncotype DX recurrence score. Mod Pathol 2008; 21: 125561. 80. Pusztai L, Cristofanilli M, Paik S. New generation of molecular prognostic and predictive tests for breast cancer. Semin Oncol 2007; 34: S106. 81. Rosai J. Why microscopy will remain a cornerstone of surgical pathology. Lab Invest 2007; 87: 4038. 82. Ma XJ, Salunga R, Tuggle JT, et al. Gene expression proles of human breast cancer progression. Proc Natl Acad Sci USA 2003; 100: 59749. 83. Elston CW, Ellis IO. Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 1991; 19: 403 10. 84. Robbins P, Pinder S, de Klerk N, et al. Histological grading of breast carcinomas: a study of interobserver agreement. Hum Pathol 1995; 26: 8739. 85. Sotiriou C, Wirapati P, Loi S, et al. Gene expression proling in breast cancer: understanding the molecular basis of histologic grade to improve prognosis. J Natl Cancer Inst 2006; 98: 26272. 86. Desmedt C, Sotiriou C. Proliferation: the most prominent predictor of clinical outcome in breast cancer. Cell Cycle 2006; 5: 2198202. 87. Loi S, Haibe-Kains B, Desmedt C, et al. Denition of clinically distinct molecular subtypes in estrogen receptor-positive breast carcinomas through genomic grade. J Clin Oncol 2007; 25: 123946. 88. Ivshina AV, George J, Senko O, et al. Genetic reclassication of histologic grade delineates new clinical subtypes of breast cancer. Cancer Res 2006; 66: 10292301. 89. Ma XJ, Salunga R, Dahiya S, et al. A ve-gene molecular grade index and HOXB13:IL17BR are complementary prognostic factors in early stage breast cancer. Clin Cancer Res 2008; 14: 26018. 90. Andre F, Mazouni C, Hortobagyi GN, Pusztai L. DNA arrays as predictors of ecacy of adjuvant/neoadjuvant chemotherapy in breast cancer patients: current data and issues on study design. Biochim Biophys Acta 2006; 1766: 197204. 91. Andre F, Pusztai L. Molecular classication of breast cancer: implications for selection of adjuvant chemotherapy. Nat Clin Pract Oncol 2006; 3: 62132. 92. Tordai A, Wang J, Andre F, et al. Evaluation of biological pathways involved in chemotherapy response in breast cancer. Breast Cancer Res 2008; 10: R37. 93. Loi S, Haibe-Kains B, Desmedt C, et al. Predicting prognosis using molecular proling in estrogen receptor-positive breast cancer treated with tamoxifen. BMC Genomics 2008; 9: 239. 94. Ma XJ, Wang Z, Ryan PD, et al. A two-gene expression ratio predicts clinical outcome in breast cancer patients treated with tamoxifen. Cancer Cell 2004; 5: 60716. 95. Reid JF, Lusa L, De Cecco L, et al. Limits of predictive models using microarray data for breast cancer clinical treatment outcome. J Natl Cancer Inst 2005; 97: 92730. 96. Goetz MP, Suman VJ, Ingle JN, et al. A two-gene expression ratio of homeobox 13 and interleukin-17B receptor for prediction of recurrence and survival in women receiving adjuvant tamoxifen. Clin Cancer Res 2006; 12: 20807. 97. Ma XJ, Hilsenbeck SG, Wang W, et al. The HOXB13:IL17BR expression index is a prognostic factor in early-stage breast cancer. J Clin Oncol 2006; 24: 46119. 98. Hudis CA. Trastuzumabmechanism of action and use in clinical practice. N Engl J Med 2007; 357: 3951. 99. Harris LN, You F, Schnitt SJ, et al. Predictors of resistance to preoperative trastuzumab and vinorelbine for HER2-positive early breast cancer. Clin Cancer Res 2007; 13: 1198207. 100. Muss HB, Thor AD, Berry DA, et al. c-erbB-2 expression and response to adjuvant therapy in women with node-positive early breast cancer. N Engl J Med 1994; 330: 12606.

101. Paik S, Bryant J, Park C, et al. erbB-2 and response to doxorubicin in patients with axillary lymph node-positive, hormone receptornegative breast cancer. J Natl Cancer Inst 1998; 90: 136170. 102. Wang JC. DNA topoisomerases. Annu Rev Biochem 1996; 65: 63592. 103. Arriola E, Marchio C, Tan DS, et al. Genomic analysis of the HER2/ TOP2A amplicon in breast cancer and breast cancer cell lines. Lab Invest 2008; 88: 491503. 104. Arriola E, Rodriguez-Pinilla SM, Lambros MB, et al. Topoisomerase II alpha amplication may predict benet from adjuvant anthracyclines in HER2 positive early breast cancer. Breast Cancer Res Treat 2007; 106: 1819. 105. Hicks DG, Yoder BJ, Pettay J, et al. The incidence of topoisomerase II-alpha genomic alterations in adenocarcinoma of the breast and their relationship to human epidermal growth factor receptor-2 gene amplication: a uorescence in situ hybridization study. Hum Pathol 2005; 36: 34856. 106. Jarvinen TA, Tanner M, Barlund M, Borg A, Isola J. Characterization of topoisomerase II alpha gene amplication and deletion in breast cancer. Genes Chromosomes Cancer 1999; 26: 142 50. 107. Orlando L, Del Curto B, Gandini S, et al. Topoisomerase IIalpha gene status and prediction of pathological complete remission after anthracycline-based neoadjuvant chemotherapy in endocrine non-responsive Her2/neu-positive breast cancer. Breast 2008; 17: 50611. 108. Tanner M, Isola J, Wiklund T, et al. Topoisomerase IIalpha gene amplication predicts favorable treatment response to tailored and dose-escalated anthracycline-based adjuvant chemotherapy in HER2/neu-amplied breast cancer: Scandinavian Breast Group Trial 9401. J Clin Oncol 2006; 24: 242836. 109. Allinen M, Beroukhim R, Cai L, et al. Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell 2004; 6: 1732. 110. Beck AH, Espinosa I, Gilks CB, van de Rijn M, West RB. The bromatosis signature denes a robust stromal response in breast carcinoma. Lab Invest 2008; 88: 591601. 111. Bergamaschi A, Tagliabue E, Sorlie T, et al. Extracellular matrix signature identies breast cancer subgroups with dierent clinical outcome. J Pathol 2008; 214: 35767. 112. Casey T, Bond J, Tighe S, et al. Molecular signatures suggest a major role for stromal cells in development of invasive breast cancer. Breast Cancer Res Treat 2008; Mar 29 (Epub ahead of print). 113. Finak G, Bertos N, Pepin F, et al. Stromal gene expression predicts clinical outcome in breast cancer. Nat Med 2008; 14: 51827. 114. Weigelt B, Bissell MJ. Unraveling the microenvironmental inuences on the normal mammary gland and breast cancer. Semin Cancer Biol 2008; 18: 31121. 115. West RB, Nuyten DS, Subramanian S, et al. Determination of stromal signatures in breast carcinoma. PLoS Biol 2005; 3: e187. 116. Hu M, Yao J, Carroll DK, et al. Regulation of in situ to invasive breast carcinoma transition. Cancer Cell 2008; 13: 394406. 117. Jones C, Mackay A, Grigoriadis A, et al. Expression proling of puried normal human luminal and myoepithelial breast cells: identication of novel prognostic markers for breast cancer. Cancer Res 2004; 64: 303745. 118. Teschendor AE, Miremadi A, Pinder SE, Ellis IO, Caldas C. An immune response gene expression module identies a good prognosis subtype in estrogen receptor negative breast cancer. Genome Biol 2007; 8: R157. 119. Tan DS, Lambros MB, Natrajan R, Reis-Filho JS. Getting it right: designing microarray (and not microawry) comparative genomic hybridization studies for cancer research. Lab Invest 2007; 87: 73754. 120. Bergamaschi A, Kim YH, Wang P, et al. Distinct patterns of DNA copy number alteration are associated with dierent clinicopathological features and gene-expression subtypes of breast cancer. Genes Chromosomes Cancer 2006; 45: 103340. 121. Hicks J, Krasnitz A, Lakshmi B, et al. Novel patterns of genome rearrangement and their association with survival in breast cancer. Genome Res 2006; 16: 146579. 122. Chin K, DeVries S, Fridlyand J, et al. Genomic and transcriptional aberrations linked to breast cancer pathophysiologies. Cancer Cell 2006; 10: 52941. 123. Vogelstein B, Fearon ER, Hamilton SR, et al. Genetic alterations during colorectal-tumor development. N Engl J Med 1988; 319: 525 32. 124. Buerger H, Otterbach F, Simon R, et al. Comparative genomic hybridization of ductal carcinoma in situ of the breast-evidence of multiple genetic pathways. J Pathol 1999; 187: 396402. 125. Buerger H, Otterbach F, Simon R, et al. Dierent genetic pathways in the evolution of invasive breast cancer are associated with distinct morphological subtypes. J Pathol 1999; 189: 5216.

88

GEYER et al.

Pathology (2009), 41(1), January

126. Roylance R, Gorman P, Harris W, et al. Comparative genomic hybridization of breast tumors stratied by histological grade reveals new insights into the biological progression of breast cancer. Cancer Res 1999; 59: 14336. 127. Stange DE, Radlwimmer B, Schubert F, et al. High-resolution genomic proling reveals association of chromosomal aberrations on 1q and 16p with histologic and genetic subgroups of invasive breast cancer. Clin Cancer Res 2006; 12: 34552. 128. Cleton-Jansen AM, Buerger H, Haar N, et al. Dierent mechanisms of chromosome 16 loss of heterozygosity in well- versus poorly dierentiated ductal breast cancer. Genes Chromosomes Cancer 2004; 41: 10916. 129. Waldman FM, DeVries S, Chew KL, et al. Chromosomal alterations in ductal carcinomas in situ and their in situ recurrences. J Natl Cancer Inst 2000; 92: 31320. 130. Allred DC, Wu Y, Mao S, et al. Ductal carcinoma in situ and the emergence of diversity during breast cancer evolution. Clin Cancer Res 2008; 14: 3708. 131. Weigelt B, Horlings H, Kreike B, et al. Renement of breast cancer classication by molecular characterization of histological special types. J Pathol 2008; 216: 14150. 132. Kumar R, Neilsen PM, Crawford J, et al. FBXO31 is the chromosome 16q24.3 senescence gene, a candidate breast tumor suppressor, and a component of an SCF complex. Cancer Res 2005; 65: 1130413. 133. Rakha EA, Green AR, Powe DG, Roylance R, Ellis IO. Chromosome 16 tumor-suppressor genes in breast cancer. Genes Chromosomes Cancer 2006; 45: 52735. 134. van Wezel T, Lombaerts M, van Roon EH, et al. Expression analysis of candidate breast tumour suppressor genes on chromosome 16q. Breast Cancer Res 2005; 7: R9981004. 135. Cleton-Jansen AM. E-cadherin and loss of heterozygosity at chromosome 16 in breast carcinogenesis: dierent genetic pathways in ductal and lobular breast cancer? Breast Cancer Res 2002; 4: 58. 136. Vos CB, Cleton-Jansen AM, Berx G, et al. E-cadherin inactivation in lobular carcinoma in situ of the breast: an early event in tumorigenesis. Br J Cancer 1997; 76: 11313. 137. Droufakou S, Deshmane V, Roylance R, et al. Multiple ways of silencing E-cadherin gene expression in lobular carcinoma of the breast. Int J Cancer 2001; 92: 4048. 138. Sarrio D, Moreno-Bueno G, Hardisson D, et al. Epigenetic and genetic alterations of APC and CDH1 genes in lobular breast cancer: relationships with abnormal E-cadherin and catenin expression and microsatellite instability. Int J Cancer 2003; 106: 20815. 139. Jacobs TW, Pliss N, Kouria G, Schnitt SJ. Carcinomas in situ of the breast with indeterminate features: role of E-cadherin staining in categorization. Am J Surg Pathol 2001; 25: 22936. 140. Maluf HM. Dierential diagnosis of solid carcinoma in situ. Semin Diagn Pathol 2004; 21: 2531. 141. Nishizaki T, Chew K, Chu L, et al. Genetic alterations in lobular breast cancer by comparative genomic hybridization. Int J Cancer 1997; 74: 5137. 142. Palacios J, Sarrio D, Garcia-Macias MC, et al. Frequent E-cadherin gene inactivation by loss of heterozygosity in pleomorphic lobular carcinoma of the breast. Mod Pathol 2003; 16: 6748. 143. Reis-Filho JS, Simpson PT, Jones C, et al. Pleomorphic lobular carcinoma of the breast: role of comprehensive molecular pathology in characterization of an entity. J Pathol 2005; 207: 113. 144. Simpson P, Reis-Filho J, Lambros M, et al. Molecular proling pleomorphic lobular carcinomas of the breast: evidence for a common molecular genetic pathway with classic lobular carcinomas. J Pathol 2008; 215: 23144. 145. Boecker W, Buerger H, Schmitz K, et al. Ductal epithelial proliferations of the breast: a biological continuum? Comparative genomic hybridization and high-molecular-weight cytokeratin expression patterns. J Pathol 2001; 195: 41521.

146. Boecker W, Moll R, Dervan P, et al. Usual ductal hyperplasia of the breast is a committed stem (progenitor) cell lesion distinct from atypical ductal hyperplasia and ductal carcinoma in situ. J Pathol 2002; 198: 45867. 147. OConnell P. Genetic and cytogenetic analyses of breast cancer yield dierent perspectives of a complex disease. Breast Cancer Res Treat 2003; 78: 34757. 148. Simpson PT, Gale T, Reis-Filho JS, et al. Columnar cell lesions of the breast: the missing link in breast cancer progression? A morphological and molecular analysis. Am J Surg Pathol 2005; 29: 73446. 149. Jones C, Nonni AV, Fulford L, et al. CGH analysis of ductal carcinoma of the breast with basaloid/myoepithelial cell dierentiation. Br J Cancer 2001; 85: 4227. 150. Abdel-Fatah TM, Powe DG, Hodi Z, et al. High frequency of coexistence of columnar cell lesions, lobular neoplasia, and low grade ductal carcinoma in situ with invasive tubular carcinoma and invasive lobular carcinoma. Am J Surg Pathol 2007; 31: 417 26. 151. Tognon C, Knezevich SR, Huntsman D, et al. Expression of the ETV6-NTRK3 gene fusion as a primary event in human secretory breast carcinoma. Cancer Cell 2002; 2: 36776. 152. Diallo R, Schaefer KL, Bankfalvi A, et al. Secretory carcinoma of the breast: a distinct variant of invasive ductal carcinoma assessed by comparative genomic hybridization and immunohistochemistry. Hum Pathol 2003; 34: 1299305. 153. Diallo R, Tognon C, Knezevich SR, Sorensen P, Poremba C. Secretory carcinoma of the breast: a genetically dened carcinoma entity. Verh Dtsch Ges Pathol 2003; 87: 193203. 154. Makretsov N, He M, Hayes M, et al. A uorescence in situ hybridization study of ETV6-NTRK3 fusion gene in secretory breast carcinoma. Genes Chromosomes Cancer 2004; 40: 1527. 155. Reis-Filho JS, Natrajan R, Vatcheva R, et al. Is acinic cell carcinoma a variant of secretory carcinoma? A FISH study using ETV6 split apart probes. Histopathology 2008; 52: 8406. 156. Lakhani SR, Jacquemier J, Sloane JP, et al. Multifactorial analysis of dierences between sporadic breast cancers and cancers involving BRCA1 and BRCA2 mutations. J Natl Cancer Inst 1998; 90: 113845. 157. Honrado E, Benitez J, Palacios J. The molecular pathology of hereditary breast cancer: genetic testing and therapeutic implications. Mod Pathol 2005; 18: 130520. 158. Tan DS, Marchio C, Reis-Filho JS. Hereditary breast cancer: from molecular pathology to tailored therapies. J Clin Pathol 2008; 61: 107382. 159. Lakhani SR, Van De Vijver MJ, Jacquemier J, et al. The pathology of familial breast cancer: predictive value of immunohistochemical markers estrogen receptor, progesterone receptor, HER-2, and p53 in patients with mutations in BRCA1 and BRCA2. J Clin Oncol 2002; 20: 23108. 160. Lakhani SR, Reis-Filho JS, Fulford L, et al. Prediction of BRCA1 status in patients with breast cancer using estrogen receptor and basal phenotype. Clin Cancer Res 2005; 11: 517580. 161. Foulkes WD, Stefansson IM, Chappuis PO, et al. Germline BRCA1 mutations and a basal epithelial phenotype in breast cancer. J Natl Cancer Inst 2003; 95: 14825. 162. Marchio C, Iravani M, Natrajan R, et al. Genomic and immunophenotypical characterization of pure micropapillary carcinomas of the breast. J Pathol 2008; 215: 398410. 163. Weinstein IB. Cancer. Addiction to oncogenesthe Achilles heal of cancer. Science 2002; 297: 634. 164. Weinstein IB, Joe AK. Mechanisms of disease: Oncogene addiction a rationale for molecular targeting in cancer therapy. Nat Clin Pract Oncol 2006; 3: 44857.

Вам также может понравиться