Skip to main content
International Journal of Clinical and Experimental Pathology logoLink to International Journal of Clinical and Experimental Pathology
. 2010 Nov 27;4(1):22–31.

Absence of microsatellite instability in mucinous carcinomas of the breast

Magali Lacroix-Triki 1,2,*, Maryou B Lambros 1,*, Felipe C Geyer 1, Paula H Suarez 1, Jorge S Reis-Filho 1, Britta Weigelt 3
PMCID: PMC3016101  PMID: 21228925

Abstract

Microsatellite instability (MSI) is a form of genetic instability that results from defects in DNA mismatch repair. MSI is reported to be rare in unselected breast cancers, however it is a common feature in subsets of colorectal, ovarian and endometrial cancers. In these anatomical sites, MSI-high carcinomas often display a mucinous histology. The aim of this study was to determine whether mucinous carcinomas of the breast would more frequently display MSI-high than invasive ductal carcinomas of no special type (IDC-NSTs). The expression of four MSI markers (i.e. MSH2, MSH6, MLH1 and PMS2) was immunohistochemically assessed in 35 mucinous breast carcinomas and 35 histological grade- and oestrogen receptor (ER) status-matched IDC-NSTs, and in a series of 245 invasive breast cancers. Cases were considered as potentially MSI-high if tumour cells lacked expression of at least two MSI markers and internal controls displayed nuclear staining. Nine mucinous carcinomas were microdissected and subjected to MSI analysis by PCR using the MSI markers BAT26 and BAT40. No immunohistochemical evidence of MSI-high was found in the 35 mucinous carcinomas and 35 grade- and ER-matched IDC-NSTs, and in the cohort of 245 invasive breast cancers. In addition, no evidence of MSI-high was observed by PCR analysis using the BAT26 and BAT40 markers in the nine mucinous carcinomas tested. Our results demonstrate that MSI-high phenotype is remarkably rare in invasive breast cancer, and that, in contrast to mucinous carcinomas of other anatomical sites, MSI is not a common event in mucinous carcinomas of the breast.

Keywords: Mucinous carcinoma, breast cancer, mismatch repair, microsatellite instability, immunohistochemistry, genetics

Introduction

Mucinous carcinoma is a rare histological special type of invasive breast cancer, accounting for up to 2% of all breast carcinomas. Mucinous carcinoma of the breast occurs preferentially in older women and is associated with good clinical outcome [1-4]. In a way akin to mucinous cancers of other anatomical sites, mucinous carcinomas of the breast are characterised by abundant production of extracellular and/or intracellular mucin. Pure mucinous carcinomas of the breast are defined as tumours with >90% of mucinous component and display bland cytological features, with uniform neoplastic cells showing mild nuclear atypia and low mitotic activity, arranged in clusters floating in large amount of mucin [5]. Therefore, by definition, the vast majority of mucinous breast carcinomas is classified as of low histological grade [1-4,6] when graded using the Nottingham grading system [7].

We and others have shown that mucinous carcinomas of the breast are typically oestrogen receptor (ER)-positive [1,4,6] and classified as of ‘luminal’ subtype according to the molecular breast cancer taxonomy based on microarray gene expression profiling [8-14]. Furthermore, we previously demonstrated that mucinous carcinomas of the breast are (i) distinct from grade- and ER-matched invasive ductal carcinomas of no special type (IDC-NSTs) at the genomic and transcriptomic levels [6,8], (ii) display a relatively low level of genetic instability [6], and (iii) lack the concurrent loss of 16q and gain of 1q [6], a hallmark feature of grade I IDC-NSTs and classic lobular carcinomas [15-19]. These data suggest that mucinous carcinomas may evolve through genetic pathways distinct from those altered in tumours from the low-grade breast neoplasia family [20].

Microsatellite instability (MSI) is a form of genetic instability characterised by frequent errors occurring during the replication of short nucleo-tide repeats, often due to a defective DNA mismatch repair gene such as hMSH2, hMLH1, hPMS2 and hMSH6 [21-25]. In fact, two distinct MSI phenotypes have been described in cancer: MSI-high (MSI-H) cancers, which result from defective mismatch repair, and MSI-low (MSI-L) tumours, which display lower levels of MSI and are not associated with defective mismatch repair [26-28]. However, the definition of the MSI-L phenotype remains controversial [28]. In addition, several independent groups have shown that MSI-H cancers are diploid and harbour fewer chromosomal copy number aberrations than tumours lacking MSI or that are MSI-L [29,30].

Subsets of colorectal [24], gastric [31], pancreatic [31], ovarian [32] and endometrial tumours [22,31,33], and particularly those occurring in the hereditary nonpolyposis colorectal cancer (HNPCC) or Lynch syndrome [31], are characterised by microsatellite instability. Interestingly, however, MSI-H appears to be vanishingly rare in breast cancer [21,34]. Likewise, breast cancers displaying an MSI-L status are remarkably rare, whereas in tumours from other anatomical sites, such as colorectal, endometrial or ovarian cancers [27], this phenomenon is not as uncommon. Of note, in some anatomical sites (e.g. colorectal and ovarian), tumours displaying microsatellite instability often display a mucinous histology [32,35,36]. However, the prevalence of MSI in mucinous carcinomas of the breast has not yet been systematically addressed.

Immunohistochemical analysis of DNA mismatch repair proteins, in particular the combination of MLH1/PMS2 and MSH2/MSH6 immunostainings, has been used as a surrogate for the detection of MLH1 or MSH2 mutations, the most frequently mutated mismatch repair genes in MSI-H tumours [23,25,26]. This four-marker immunohistochemistry (IHC) panel has a high sensitivity (>90%) in predicting MLH1/MSH2 gene mutations and has been suggested to be an optimal first-line screening tool [23,37]. Of note, however, mismatch repair protein deficiency as explored by immunohistochemistry may also be underpinned by alternative mechanisms such as methylation. Indeed, a recent multicentric study on colorectal cancers showed that specific MLH1 promoter polymorphism predisposed to MLH1 methylation and MLH1 silencing, leading to loss of MLH1 protein as assessed by IHC and MSI [38]. This observation supports the fact that, after excluding germline mutation, mismatch repair gene methylation may be the underlying cause of at least a subset of MSI-H cancers [39].

The aims of this study were to determine whether mucinous carcinomas of the breast would display MSI, and whether mucinous carcinomas would more frequently display an MSI-H phenotype than IDC-NSTs. To address these questions, we analysed a series of pure mucinous carcinomas of the breast and grade- and ER-matched IDC-NSTs for MSI by IHC for MSH2, MLH1, MSH6 and PMS2 expression, a subset of which was subjected to polymerase chain reaction (PCR) amplification of the mononucleotide repeat markers BAT26 and BAT40. As a secondary aim, we investigated the prevalence of immunohistochemical profiles suggestive of MSI-H in a series of 245 invasive breast cancers treated with adjuvant anthracycline-based chemotherapy.

Materials and methods

Cases

Mucinous carcinomas: Consecutive cases of pure mucinous carcinomas of the breast (n=35) were retrieved from the histopathological files of the authors' institutions. Cases were centrally reviewed by at least two pathologists (MLT, FCG and/ or JRF) and detailed histopathological features are described elsewhere [6]. Briefly, nine cases were of grade I, 24 of grade II, and three of grade III as assessed using the Nottingham grading system [7]. All cases were ER-positive [6]. A tissue microarray (TMA) was constructed with triplicate 0.6mm tumour cores.

Control group: A control group of consecutive 245 invasive breast cancers was included in a separate TMA containing three replicate 0.6mm tumour cores. Tumours were graded according to the Nottingham grading system [7] and detailed histopathological features are described elsewhere [40-42]. Out of this cohort, 35 consecutive IDC-NSTs were matched according to histological grade and ER-status with the muci-nous carcinomas studied. This study was approved by local research ethics committee.

Immunohistochemistry

Immunohistochemistry (IHC) was performed on 3um TMA sections as previously described [43], using antibodies against the mismatch repair proteins MSH2, MLH1, MSH6 and PMS2. Combination of the four makers MLH1, MSH2, MSH6 and PMS2 for MSI screening by IHC stems from observations showing that mutations in MLH1 or MSH2 result in concurrent loss of MLH1/PMS2 or MSH2/MSH6 respectively, whereas mutations of PMS2 or MSH6 will result in isolated loss of PMS2 or MSH6 only [23,25,26]. Details of the antibodies and IHC protocols used are provided in Table 1. Positive (ovarian carcinomas) and negative controls (omission of the primary antibody) were included in each experiment. Slides were interpreted by two pathologists (MLT & FCG), who evaluated the expression of each marker semi-quantitatively, and resolved discordant cases on a multi-headed microscope. Only nuclear expression was considered as specific and the presence of internal positive controls (nuclear staining in either normal epithelial cells, lymphocytes or stromal cells) was assessed for each case. Cases without optimal positive internal controls were deemed inconclusive. Following the IHC interpretation criteria applied in colorectal cancers [25], tumours were recorded as MSH2, MLH1, MSH6 or PMS2 negative when there was a complete absence of nuclear staining in neoplastic cells. If a case was negative for at least two markers or was inconclusive for more than one marker, IHC was repeated on full-face sections.

Table 1.

Immunohistochemistry protocols for MSH2, MLH1, MSH6 and PMS2 antibodies

Marker Clone Dilution Antigen retrieval Detection system Company
MSH2 FE11 1:20 2 min, PC citrate pH 6.0 EnVision™ system, Dako, Glostrup, Denmark Calbiochem, EMD Chemicals, Gibbstone, NJ, USA
MLH1 G168-728 1:20 2 min, PC citrate pH 6.0 EnVision™ system, Dako, Glostrup, Denmark Pharmingen, BD Biosciences, Oxford, UK
MSH6 44/MSH6 1:15 2 min, PC citrate pH 6.0 EnVision™ system, Dako, Glostrup, Denmark Transduction Labs, BD Biosciences, Oxford, UK
PMS2 A16-4 1:10 2 min PC, EDTA pH8.0 EnVision™ system, Dako, Glostrup, Denmark Pharmingen, BD Biosciences, Oxford, UK

MSH: mutS homolog; MLH: mutL homolog; PC: pressure cooker; PMS2: postmeiotic segregation increased 2.

All breast cancers included in this study were previously characterised for their expression of ER, progesterone receptor (PR), HER2, Ki67, p53, Bcl2, cyclin D1 and basal markers [6,40].

We compared the results of the MSI IHC analysis performed in 35 pure mucinous carcinomas with those from 35 IDC-NSTs matched for histological grade and ER expression, selected from the cohort of 245 invasive breast cases, as previously described [6]. The rationale for matching case and control samples according to grade and ER status stems from several lines of evidence suggesting that grade and ER are strongly associated with the pattern of genomic changes in breast cancer [16,44,45].

Microdissection and DNA extraction

Ten representative 10um thick sections of the mucinous tumours were stained with nuclear fast red and were microdissected with a sterile needle under a stereomicroscope (Olympus SZ61, Tokyo, Japan) to ensure >70% of purity of cancer cells as previously described [43,46]. DNA was extracted as previously described using an FFPE DNA extraction kit (FFPE DNeasy Kit, Qiagen, Hamburg, Germany) [43], and DNA concentration was determined using the PicoGreen® assay according to the manufacturer's protocol (Invitrogen, Paisley, UK). Sufficient amount of DNA to perform MSI PCR analysis was obtained in nine mucinous carcinomas.

Microsatellite instability analysis

The presence of MSI was evaluated by amplification of the mononucleotide repeat markers BAT26 and BAT40. DNA was subjected to standard PCR and gel electrophoresis as previously described [25]. Briefly, forward primers with a fluorescent tag at the 5′ end to allow microsatellite detection by the ABI Prism were purchased from Applied Biosystems (Carlsbad, CA, USA). PCR conditions were as follows: 5μl of 10× PCR reaction mix buffer (AmpliTaq, Applied Biosystems); 3μl of MgCl2 (AmpliTaq); 5μl of 2.5mM dNTPs; 0.5μl of Taq polymerase (AmpliTaq); 0.5μl of each primer and 50ng of DNA in a final volume of 50μl. After initial denaturation at 94°C for 4min, 35 cycles of PCR steps (94°C for 1min, annealing at 56°C for 1min and 72°C for 3min) were run, followed by a final extension at 72°C for 7min. After amplification, PCR products were mixed with 18μl of Hi-Di formamide (Applied Biosystems) and 0.3μl of size standards (GeneScan™ 500 ROX™, Applied Biosystems), followed by denaturation at 95°C for 3min, before analysis on ABI prism using GeneScan Analysis software (Applied Biosystems). Tumours were considered MSI-H when both BAT26 and BAT40 displayed MSI, defined by multiple extra bands above background [25,27]. As control, a blood DNA sample from a healthy female donor was run in parallel in each experiment. The technique was validated for both markers on two endometrial cancer cell lines, AN3CA and EFE-184, which were MSI-H and MSI-stable, respectively. The microsatellite marker BAT26 has been shown to be particularly useful in determining the MSI-H phenotype without the requirement for matching normal DNA given that it is ‘quasi-monomorphic’ in DNA from normal individuals or microsatellite stable tumours compared to MSI-H cancers [25,26,47]. Therefore, no normal tissue matching was performed in this study.

Results

All 35 pure mucinous carcinomas of the breast analysed were positive for MLH1 and MSH6 as determined by IHC, and 33 out of 35 (94.2%) and 32 out of 34 cases (94.1%) showed expression of MSH2 and PMS2, respectively (Table 2 and Figure 1). Out of the three mucinous tumours lacking expression of MSH2 and/or PMS2, two cases were considered inconclusive and no case displayed complete lack of expression of two MSI markers. Following the guidelines for the interpretation of MSI IHC markers in colorectal carcinomas [23,25], no case in this cohort of mucinous carcinomas would be considered as potentially MSI-H. Immunohistochemical analysis of full-face sections of the negative and inconclusive cases revealed one case lacking expression of one marker (i.e. MSH2) and no case lacking expression of two or more MSI markers (Table 2). Of the 35 grade-and ER-matched IDC-NSTs, 97.1%, 97.1%, 88.6% and 100% showed nuclear expression of MLH1, MSH2, MSH6 and PMS2, respectively. One case lacked MLH1 expression, one case MSH2 expression and three cases MSH6 expression, the remaining case displaying lack of expression of MSH6 was considered inconclusive. No IDC-NST lacked expression of two or more MSI markers (Table 2). When the pattern of expression of the four MSI markers in the 35 mucinous carcinomas was compared with that of 35 grade- and ER-matched IDC-NSTs, no significant differences were observed (Table 2).

Table 2.

Comparison of MSI marker expression in 35 pure mucinous carcinomas and 35 grade- and ER-matched IDC-NSTs determined by immunohistochemistry on tissue microarrays

N Pure mucinous (n=35) IDC-NSTs (n=35) p value*
MLH1 70 0.999
 Positive 35 (100%) 34 (97.1%)
 Negative 0 1 (2.9%)
 Inconclusive 0 0
MSH2 70 0.999
 Positive 33 (94.2%) 34 (97.1%)
 Negative 1 (2.9%) 1 (2.9%)
 Inconclusive 1 (2.9%) 0
MSH6 70 0.114
 Positive 35 (100%) 31 (88.6%)
 Negative 0 3 (8.5%)
 Inconclusive 0 1 (2.9%)
PMS2 69 0.239
 Positive 32 (94.1%) 35 (100%)
 Negative 0 0
 Inconclusive 2 (5.9%) 0
Combination analysis of MSI markers# 67
 1 marker negative 1 (3%) 5 (14.7%)
 2 markers negative 0 0
 3 markers negative 0 0
 4 markers negative 0 0
*

3×2 Fisher's test

#

inconclusive cases excluded

ER: oestrogen receptor; IDC-NSTs: invasive ductal carcinomas of no special type; MLH1: mutL homolog 1; MSH: mutS homolog; MSI: microsatellite instability; PMS2: postmeiotic segregation increased 2.

Figure 1.

Figure 1

Immunohistochemical pattern of MSI marker expression in mucinous carcinoma of the breast. Representative micrographs of a pure mucinous carcinoma of the breast displaying nuclear expression of (A) MLH1, (B) MSH2, (C) MSH6 and (D) PMS2. Note the nuclear expression in isolated stromal cells in the mucin surrounding neoplastic cells, serving as internal positive control. MLH1: mutL homolog 1; MSH: mutS homolog; MSI: microsatellite instability; PMS2: postmeiotic segregation increased 2.

Albeit limited to a small number of cases given the restricted tumour tissue availability and generally low DNA yield of mucinous tumours owing to their histology, PCR amplification of the microsatellite markers BAT26 and BAT40 was successfully performed and informative in nine mucinous carcinomas of the breast. Tumours would have been considered MSI-H if both BAT26 and BAT40 displayed MSI, however none of the nine mucinous cases displayed MSI for any of these two markers (Figure 2).

Figure 2.

Figure 2

PCR-based MSI analysis of mucinous carcinoma of the breast using the mononucleotide repeat marker BAT26. Electropherograms of (A) mucinous carcinoma 1 (MSI-stable), (B) mucinous carcinoma 2 (MSI-stable), (C) blood DNA from healthy individual (control; MSI-stable), (D) endometrial cell line AN3CA (control; MSI-high), and (E) endometrial cell line EFE-184 (control; MSI-stable). MSI: microsatellite instability; PCR: polymerase chain reaction.

Analysis of the entire cohort of primary breast cancers from patients treated with adjuvant anthracycline-based chemotherapy (n=245) revealed that the vast majority of cases were positive for MLH1 (97.8%), MSH2 (91.7%), MSH6 (93.7%) and PMS2 (98.2%) (Table 3 and Supplementary Tables 1-4). Similar frequencies of positivity were observed for the subset of IDC-NSTs (n=180) (Table 3). When combining the IHC conclusive results of all four MSI markers tested in the 245 invasive breast cancers, no case showed complete lack of expression of two MSI markers on TMA cores (Table 3). Reanalysis of negative and inconclusive cases on full face tissue sections revealed focal expression of the MSI markers (Figure 3), and, therefore, these cases were considered MSI stable. Two cases that lacked expression of two MSI markers also lacked internal controls and were deemed inconclusive [25]. No significant correlations between the lack of expression of MLH1, MSH2, MSH6 or PMS2 and clinico-pathological features and immunohistochemical markers in this cohort of 245 invasive breast cancers were identified (Supplementary Tables 1-4).

Table 3.

MSI marker expression in invasive breast carcinomas determined using immunohistochemistry on tissue microarrays

N Whole cohort (n=245) N IDC-NSTs only (n=180)
MLH1 178 133
 Positive 174 (97.8%) 129 (97%)
 Negative 4 (2.2%) 4 (3%)
 Inconclusive 0 0
MSH2 180 134
 Positive 165 (91.7%) 125 (93.3%)
 Negative 14 (7.7%) 8 (6%)
 Inconclusive 1 (0.6%) 1 (0.7%)
MSH6 225 164
 Positive 211 (93.7%) 151 (92.1%)
 Negative 11 (4.9%) 10 (6.1%)
 Inconclusive 3 (1.4%) 3 (1.8%)
PMS2 172 128
 Positive 169 (98.2%) 125 (97.6%)
 Negative 2 (1.1%) 2 (1.5%)
 Inconclusive 1 (0.7%) 1 (0.9%)
Combination analysis of MSI markers# 171 127
 1 marker negative 26 (15.1%) 18 (14%)
 2 markers negative 0 0
 3 markers negative 0 0
 4 markers negative 0 0
#

inconclusive cases excluded

IDC-NSTs: invasive ductal carcinomas of no special type; MLH1: mutL homolog 1; MSH: mutS homolog; MSI: microsatellite instability; PMS2: postmeiotic segregation increased 2.

Figure 3.

Figure 3

Immunohistochemical pattern of MSI marker expression in invasive ductal carcinoma of the breast. Representative micrograph of an invasive ductal carcinoma of no special type displaying nuclear expression of (A) MLH1, (B) MSH2, (C) MSH6, and (D) PMS2. This case was negative for MLH1 (A, insert) and MSH2 (B, insert) on immunostains performed on TMA sections but displayed focal positivity on full sections. MLH1: mutL homolog 1, MSH: mutS homolog; MSI: microsatellite instability; PMS2: postmeiotic segregation increased 2; TMA: tissue microarray.

Discussion

Our findings corroborate those of previous studies, which demonstrated the lack or low prevalence (i.e. 2-3%) of MSI in invasive breast cancers [21,27,31,34,48] and breast cancer cell lines [21]. Importantly, we provide direct evidence that unlike mucinous carcinomas of the colon, ovary and endometrium, which display an MSI-H phenotype in about 30% of cases [22,24,49-51], MSI is a remarkably rare phenomenon in mucinous carcinomas of the breast. This finding is entirely consistent with the observation that unlike colorectal or endometrial cancers of mucinous subtype, mucinous carcinoma of the breast is uncommon in HNPCC/ Lynch syndrome, which is characterised by germline mutations in DNA mismatch repair genes [52].

Our results provide evidence to suggest that the assessment of mismatch repair proteins by IHC should preferably be performed on full tissue sections, as their expression may be focal in some tumours resulting in false negative results in TMA analyses. This observation supports previous reports addressing the issue of reliability of MSI IHC on small biopsy specimens and focal expression of MSI markers, in particular with MSH6 immunostaining [23]. Furthermore, there are several lines of evidence to demonstrate that immunohistochemical stainings of MSI markers should be interpreted with caution when no nuclear staining of internal controls (i.e. normal epithelial, lymphocytes or stromal cells) is observed on repeated experiments [23,25].

It should be noted that the presence of an MSI-L phenotype in the mucinous carcinomas and control invasive breast cancers has not been addressed in the present study. MSI-L tumours, which have been described in 26%, 32% and 35% of ovarian, endometrial and colorectal cancers, respectively [27], are believed to harbour distinct molecular aberrations, including MSH6 germline mutations [26]. However, these tumours did not seem to be associated with distinct clinicopathological features [27,28]. Moreover, it has been reported previously that MSI-L cancers do not display loss of MLH1, MSH2, MSH6 and PMS2 protein expression [25].

Therefore, we cannot rule out that a subset of invasive breast cancers, including mucinous carcinomas, may display a MSI-L phenotype. Despite the controversy regarding the definition of MSI-L, further studies using specific additional markers, such as those proposed by Halford et al [27,28], for the identification of the MSI-L phenotype in different histological subtypes of invasive breast cancer are warranted.

Taken together, here we demonstrate that in contrast to mucinous carcinomas of other anatomical sites, mucinous carcinomas of the breast do not display an MSI-H phenotype as determined by IHC and a PCR-based method. Moreover, no concurrent lack of MLH1/PMS2 or MSH2/MSH6 expression, the IHC surrogate markers of the most frequent mutations (i.e. mutations of MLH1 or MSH2) observed in MSI-H cancers of other anatomical sites, were found in a consecutive series of 245 invasive breast carcinomas (including IDC-NSTs, invasive lobular and mixed carcinomas).

Acknowledgments

This study was funded by Breakthrough Breast Cancer. M.L.-T. is funded by a partenariat grant from the Fédération Nationale des Centres de Lutte Contre le Cancer (FNCLCC, Paris) and the Fondation Médicale de France (Paris), B.W. by a Cancer Research UK postdoctoral fellowship.

Supplemental material

Supplementary Table 1.

Analysis of correlation between MLH1 expression, clinico-pathological features and markers in 245 invasive breast carcinomas

N MLH1 negative (n=4) MLH1 positive (n=174) p value
Tumour size 176 0.550*
 T1 3 83
 T2 1 77
 T3 0 12
Histological grade 174 0.589*
 I 0 19
 II 2 50
 III 2 101
Lympho-vascular invasion 176 0.587**
 present 2 52
 absent 2 120
Lymph node metastasis 172 0.307**
 present 4 113
 absent 0 55
ER 178 0.499**
 positive 3 147
 negative 1 27
PR 178 0.304**
 positive 2 127
 negative 2 47
HER2 178 0.095**
 positive 2 23
 negative 2 151
Ki67 166 0.674*
 low (<10%) 1 73
 intermediate 2 67
 high (>30%) 1 22
Topoisomerase IIα 163 0.625**
 positive 3 85
 negative 1 74
Cyclin D1 169 0.623*
 low (Allred score 0-3) 1 18
 intermediate (Allred score 4-5) 1 33
 high (Allred score 6-8) 2 114
Bcl2 145 0.545**
 positive 1 97
 negative 1 46
FOXA1 146 1**
 positive 3 108
 negative 0 35
p53 166 1**
 positive 1 47
 negative 2 116
Cytokeratin 5/6 170 0.311**
 positive 1 14
 negative 3 152
Cytokeratin 14 176 1**
 positive 0 13
 negative 4 159
Cytokeratin 17 175 1**
 positive 0 19
 negative 4 152
Basal cytokeratins 176 0.432**
 positive 1 22
 negative 3 150
EGFR 178 0.281**
 positive 1 13
 negative 3 161
Caveolin 1 178 1**
 positive 0 14
 negative 4 160
Caveolin 2 165 1**
 positive 0 10
 negative 4 151
Nestin 146 0.296**
 positive 1 15
 negative 2 128
Any basal marker 176 0.476**
 positive 1 25
 negative 3 147
Molecular phenotype*** 173 0.513*
 luminal 2 127
 HER2+ 1 23
 basal-like 1 29
E-cadherin 165 0.330*
 normal 2 101
 reduced 1 10
 negative 1 50
HER2 CISH 168 0.433**
 amplified 1 21
 not amplified 3 143
TOP2A CISH 169 0.295**
 amplified 1 13
 not amplified 3 152
CCND1 CISH 176 1**
 amplified 0 24
 not amplified 4 148
*

Chi-squared test

**

Fisher's exact test

***

according to criteria by Nielsen et al [53].

CCND1: cyclin D1 gene; CISH: chromogenic in situ hybridisation; MLH1: mutL homolog 1; TOP2A: topoisomerase IIα gene.

Supplementary Table 2.

Analysis of correlation between MSH2 expression, clinico-pathological features and markers in 245 invasive breast carcinomas

N MSH2 negative (n=14) MSH2 positive (n=165) MSH2 inconclusive (n=1) p value*
Tumour size 178 0.528
 T1 6 81 0
 T2 8 69 1
 T3 0 13 0
Histological grade 176 0.882
 I 2 17 0
 II 5 49 0
 III 7 95 1
Lympho-vascular invasion 178 0.781
 present 10 112 1
 absent 4 51 0
Lymph node metastasis 174 0.758
 present 9 108 1
 absent 5 51 0
ER 180 0.545
 positive 13 136 1
 negative 1 29 0
PR 180 0.385
 positive 12 116 1
 negative 2 49 0
HER2 180 0.267
 positive 0 25 0
 negative 14 140 1
Ki67 169 0.560
 low (<10%) 8 68 0
 intermediate 4 65 1
 high (>30%) 1 22 0
Topoisomerase IIα 166 0.094
 positive 3 87 0
 negative 8 67 1
Cyclin D1 172 0.101
 low (Allred score 0-3) 3 16 0
 intermediate (Allred score 4-5) 3 32 1
 high (Allred score 6-8) 5 112 0
Bcl2 148 0.760
 positive 7 91 1
 negative 4 45 0
FOXA1 148 0.149
 positive 6 107 0
 negative 3 31 1
p53 169 0.577
 positive 2 47 0
 negative 9 110 1
Cytokeratin 5/6 172 0.909
 positive 1 15 0
 negative 13 142 1
Cytokeratin 14 178 0.961
 positive 1 12 0
 negative 13 151 1
Cytokeratin 17 177 0.879
 positive 2 18 0
 negative 12 144 1
Basal cytokeratins 178 0.921
 positive 2 22 0
 negative 12 141 1
EGFR 180 0.941
 positive 1 14 0
 negative 13 151 1
Caveolin 1 180 0.941
 positive 1 14 0
 negative 13 151 1
Caveolin 2 167 0.634
 positive 0 11 0
 negative 11 144 1
Nestin 148 0.750
 positive 2 14 0
 negative 10 121 1
Any basal marker 178 0.760
 positive 3 25 0
 negative 11 138 1
Molecular phenotype** 175 0.475
 luminal 13 116 1
 HER2+ 0 25 0
 basal-like 1 19 0
E-cadherin 168 0.230
 normal 4 100 0
 reduced 1 10 0
 negative 6 46 1
HER2 CISH 171 0.261
 amplified 0 24 0
 not amplified 14 132 1
TOP2A CISH 172 0.511
 amplified 0 15 0
 not amplified 12 144 1
CCND1 CISH 178 0.306
 amplified 0 24 0
 not amplified 13 140 1
*

Chi-squared test

**

according to criteria by Nielsen et al [53].

CCND1: cyclin D1 gene; CISH: chromogenic in situ hybridisation; MSH2: mutS homolog 2; TOP2A: topoisomerase IIα gene.

Supplementary Table 3.

Analysis of correlation between PMS2 expression, clinico-pathological features and markers in 245 invasive breast carcinomas

N PMS2 negative (n=2) PMS2 positive (n=169) PMS2 inconclusive (n=1) p value*
Tumour size 170 0.853
 T1 1 82 0
 T2 1 75 1
 T3 0 10 0
Histological grade 168 0.444
 I 1 19 0
 II 0 47 0
 III 1 99 1
Lympho-vascular invasion 170 0.676
 present 1 115 1
 absent 1 52 0
Lymph node metastasis 166 0.042
 present 0 112 0
 absent 2 51 1
ER 172 0.352
 positive 1 144 1
 negative 1 25 0
PR 172 0.592
 positive 2 125 1
 negative 0 44 0
HER2 172 0.771
 positive 0 25 0
 negative 2 144 1
Ki67 160 0.771
 low (<10%) 1 72 0
 intermediate 1 63 1
 high (>30%) 0 22 0
Topoisomerase IIα 158 0.657
 positive 1 85 1
 negative 1 70 0
Cyclin D1 163 0.003
 low (Allred score 0-3) 2 17 0
 intermediate (Allred score 4-5) 0 34 0
 high (Allred score 6-8) 0 109 1
Bcl2 139 0.615
 positive 1 92 1
 negative 0 45 0
FOXA1 140 0.713
 positive 1 103 1
 negative 0 35 0
P53 160 0.188
 positive 0 44 1
 negative 2 113 0
Cytokeratin 5/6 164 0.148
 positive 1 15 0
 negative 1 146 1
Cytokeratin 14 170 0.093
 positive 1 13 0
 negative 1 154 1
Cytokeratin 17 168 0.208
 positive 1 18 0
 negative 1 147 1
Basal cytokeratins 170 0.294
 positive 1 22 0
 negative 1 145 1
EGFR 172 0.110
 positive 1 14 0
 negative 1 155 1
Caveolin 1 172 0.110
 positive 1 14 0
 negative 1 155 1
Caveolin 2 159 0.052
 positive 1 10 0
 negative 1 146 1
Nestin 140 0.885
 positive 0 15 0
 negative 1 123 1
Any basal marker 170 0.358
 positive 1 25 0
 negative 1 142 1
Molecular phenotype** 169 0.512
 luminal 1 123 1
 HER2+ 0 24 0
 basal-like 1 19 0
E-cadherin 159 0.781
 normal 2 98 1
 reduced 0 10 0
 negative 0 48 0
HER2 CISH 162 0.777
 amplified 0 23 0
 not amplified 2 136 1
TOP2A CISH 163 0.857
 amplified 0 15 0
 not amplified 2 145 1
CCND1 CISH 169 0.030
 amplified 0 21 1
 not amplified 2 145 0
*

Chi-squared test

**

according to criteria by Nielsen et al [53].

CCND1: cyclin D1 gene; CISH: chromogenic in situ hybridisation; PMS2: postmeiotic segregation increased 2; TOP2A: topoisomerase II. gene.

Supplementary Table 4.

Analysis of correlation between MSH6 expression, clinico-pathological features and markers in 245 invasive breast carcinomas

N MSH6 negative (n=11) MSH6 positive (n=211) MSH6 inconclusive (n=3) p value*
Tumour size 223 0.284
 T1 6 108 0
 T2 5 86 3
 T3 0 15 0
Histological grade 220
 I 0 22 0 0.765
 II 3 61 1
 III 8 123 2
Lympho-vascular invasion 223 0.074
 present 4 145 2
 absent 7 64 1
Lymph node metastasis 218 0.129
 present 5 140 1
 absent 6 64 2
ER 225 0.726
 positive 9 174 3
 negative 2 37 0
PR 225 0.568
 positive 8 153 3
 negative 3 58 0
HER2 225 0.679
 positive 1 31 0
 negative 10 180 3
Ki67 208 0.828
 low (<10%) 4 83 1
 intermediate 6 83 2
 high (>30%) 1 28 0
Topoisomerase IIα 198 0.319
 positive 4 104 1
 negative 7 80 2
Cyclin D1 207 0.908
 low (Allred score 0-3) 1 23 0
 intermediate (Allred score 4-5) 3 38 1
 high (Allred score 6-8) 7 132 2
Bcl2 169 0.424
 positive 6 102 3
 negative 4 54 0
FOXA1 173 0.584
 positive 6 123 1
 negative 3 39 1
P53 207 0.979
 positive 3 55 1
 negative 8 138 2
Cytokeratin 5/6 216 0.849
 positive 1 20 0
 negative 9 183 3
Cytokeratin 14 223 0.519
 positive 0 18 0
 negative 11 191 3
Cytokeratin 17 221 0.656
 positive 2 24 0
 negative 9 183 3
Basal cytokeratins 223 0.693
 positive 1 30 0
 negative 10 179 3
EGFR 225 0.870
 positive 1 17 0
 negative 10 194 3
Caveolin 1 225 0.867
 positive 1 18 0
 negative 10 193 3
Caveolin 2 196 0.164
 positive 2 9 0
 negative 9 173 3
Nestin 166 0.051
 positive 3 16 0
 negative 5 139 3
Any basal marker 223 0.449
 positive 3 33 0
 negative 8 176 3
Molecular phenotype** 218 0.766
 luminal 8 149 3
 HER2+ 1 32 0
 basal-like 2 23 0
E-cadherin 201 0.861
 normal 7 120 2
 reduced 0 14 0
 negative 4 53 1
HER2 CISH 212 0.765
 amplified 1 30 0
 not amplified 9 170 2
TOP2A CISH 213 0.862
 amplified 1 18 0
 not amplified 10 181 3
CCND1 CISH 222 0.551
 amplified 1 28 1
 not amplified 10 180 2
*

Chi-squared test

**

according to criteria by Nielsen et al [53].

CCND1: cyclin D1 gene; CISH: chromogenic in situ hybridisation; MSH6: mutS homolog 6; TOP2A: topoisomerase IIα gene.

References

  • 1.Barkley CR, Ligibel JA, Wong JS, Lipsitz S, Smith BL, Golshan M. Mucinous breast carcinoma: a large contemporary series. Am J Surg. 2008;196:549–551. doi: 10.1016/j.amjsurg.2008.06.013. [DOI] [PubMed] [Google Scholar]
  • 2.Di Saverio S, Gutierrez J, Avisar E. A retrospective review with long term follow up of 11,400 cases of pure mucinous breast carcinoma. Breast Cancer Res Treat. 2008;111:541–547. doi: 10.1007/s10549-007-9809-z. [DOI] [PubMed] [Google Scholar]
  • 3.Li CI, Uribe DJ, Daling JR. Clinical characteristics of different histologic types of breast cancer. Br J Cancer. 2005;93:1046–1052. doi: 10.1038/sj.bjc.6602787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Vo T, Xing Y, Meric-Bernstam F, Mirza N, Vlastos G, Symmans WF, Perkins GH, Buchholz TA, Babiera GV, Kuerer HM, Bedrosian I, Akins JS, Hunt KK. Long-term outcomes in patients with mucinous, medullary, tubular, and invasive ductal carcinomas after lumpectomy. Am J Surg. 2007;194:527–531. doi: 10.1016/j.amjsurg.2007.06.012. [DOI] [PubMed] [Google Scholar]
  • 5.Tavassoli FA, Devilee P, editors. Tumours fo the Breast and Female Genital Organs. Lyon: International Agency for Research of Cancer (IARC); 2003. Tumours of the breast. Pathology and Genetics. [Google Scholar]
  • 6.Lacroix-Triki M, Suarez PH, Mackay A, Lambros MB, Natrajan R, Savage K, Geyer FC, Weigelt B, Ashworth A, Reis-Filho JS. Mucinous carcinoma of the breast is genomically distinct from invasive ductal carcinomas of no special type. J Pathol. 2010;222:282–298. doi: 10.1002/path.2763. [DOI] [PubMed] [Google Scholar]
  • 7.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;19:403–410. doi: 10.1111/j.1365-2559.1991.tb00229.x. [DOI] [PubMed] [Google Scholar]
  • 8.Weigelt B, Geyer FC, Horlings HM, Kreike B, Halfwerk H, Reis-Filho JS. Mucinous and neuroendo-crine breast carcinomas are transcriptionally distinct from invasive ductal carcinomas of no special type. Mod Pathol. 2009;22:1401–1414. doi: 10.1038/modpathol.2009.112. [DOI] [PubMed] [Google Scholar]
  • 9.Weigelt B, Horlings HM, Kreike B, Hayes MM, Hauptmann M, Wessels LF, de Jong D, Van de Vijver MJ, Van't Veer LJ, Peterse JL. Refinement of breast cancer classification by molecular characterization of histological special types. J Pathol. 2008;216:141–150. doi: 10.1002/path.2407. [DOI] [PubMed] [Google Scholar]
  • 10.Weigelt B, Reis-Filho JS. Histological and molecular types of breast cancer: is there a unifying taxonomy? Nat Rev Clin Oncol. 2009;6:718–730. doi: 10.1038/nrclinonc.2009.166. [DOI] [PubMed] [Google Scholar]
  • 11.Sotiriou C, Pusztai L. Gene-expression signatures in breast cancer. N Engl J Med. 2009;360:790–800. doi: 10.1056/NEJMra0801289. [DOI] [PubMed] [Google Scholar]
  • 12.Weigelt B, Mackay A, A'Hern R, Natrajan R, Tan DS, Dowsett M, Ashworth A, Reis-Filho JS. Breast cancer molecular profiling with single sample predictors: a retrospective analysis. Lancet Oncol. 2010;11:339–349. doi: 10.1016/S1470-2045(10)70008-5. [DOI] [PubMed] [Google Scholar]
  • 13.Reis-Filho JS, Weigelt B, Fumagalli D, Sotiriou C. Molecular profiling: moving away from tumor philately. Sci Transl Med. 2010;2 doi: 10.1126/scitranslmed.3001329. 47ps43. [DOI] [PubMed] [Google Scholar]
  • 14.Weigelt B, Baehner FL, Reis-Filho JS. The contribution of gene expression profiling to breast cancer classification, prognostication and prediction: a retrospective of the last decade. J Pathol. 2010;220:263–280. doi: 10.1002/path.2648. [DOI] [PubMed] [Google Scholar]
  • 15.Bertucci F, Orsetti B, Negre V, Finetti P, Rouge C, Ahomadegbe JC, Bibeau F, Mathieu MC, Treilleux I, Jacquemier J, Ursule L, Martinec A, Wang Q, Bénard J, Puisieux A, Birnbaum D, Theillet C. Lobular and ductal carcinomas of the breast have distinct genomic and expression profiles. Oncogene. 2008;27:5359–5372. doi: 10.1038/onc.2008.158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Loo LW, Grove DI, Williams EM, Neal CL, Cousens LA, Schubert EL, Holcomb IN, Massa HF, Glogovac J, Li CI, Malone KE, Daling JR, Delrow JJ, Trask BJ, Hsu L, Porter PL. Array comparative genomic hybridization analysis of genomic alterations in breast cancer subtypes. Cancer Res. 2004;64:8541–8549. doi: 10.1158/0008-5472.CAN-04-1992. [DOI] [PubMed] [Google Scholar]
  • 17.Simpson PT, Reis-Filho JS, Gale T, Lakhani SR. Molecular evolution of breast cancer. J Pathol. 2005;205:248–254. doi: 10.1002/path.1691. [DOI] [PubMed] [Google Scholar]
  • 18.Abdel-Fatah TM, Powe DG, Hodi Z, Lee AH, Reis-Filho JS, Ellis IO. 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;31:417–426. doi: 10.1097/01.pas.0000213368.41251.b9. [DOI] [PubMed] [Google Scholar]
  • 19.Abdel-Fatah TM, Powe DG, Hodi Z, Reis-Filho JS, Lee AH, Ellis IO. Morphologic and molecular evolutionary pathways of low nuclear grade invasive breast cancers and their putative precursor lesions: further evidence to support the concept of low nuclear grade breast neoplasia family. Am J Surg Pathol. 2008;32:513–523. doi: 10.1097/PAS.0b013e318161d1a5. [DOI] [PubMed] [Google Scholar]
  • 20.Lopez-Garcia MA, Geyer FC, Lacroix-Triki M, Marchio C, Reis-Filho JS. Breast cancer precursors revisited: molecular features and progression pathways. Histopathology. 2010;57:171–192. doi: 10.1111/j.1365-2559.2010.03568.x. [DOI] [PubMed] [Google Scholar]
  • 21.Anbazhagan R, Fujii H, Gabrielson E. Microsatellite instability is uncommon in breast cancer. Clin Cancer Res. 1999;5:839–844. [PubMed] [Google Scholar]
  • 22.Helland A, Borresen-Dale AL, Peltomaki P, Hektoen M, Kristensen GB, Nesland JM, de la Chapelle A, Lothe RA. Microsatellite instability in cervical and endometrial carcinomas. Int J Cancer. 1997;70:499–501. doi: 10.1002/(sici)1097-0215(19970304)70:5<499::aid-ijc1>3.0.co;2-t. [DOI] [PubMed] [Google Scholar]
  • 23.Shia J. Immunohistochemistry versus microsatellite instability testing for screening colorectal cancer patients at risk for hereditary nonpolyposis colorectal cancer syndrome. Part I. The utility of immunohistochemistry. J Mol Diagn. 2008;10:293–300. doi: 10.2353/jmoldx.2008.080031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Thibodeau SN, Bren G, Schaid D. Microsatellite instability in cancer of the proximal colon. Science. 1993;260:816–819. doi: 10.1126/science.8484122. [DOI] [PubMed] [Google Scholar]
  • 25.Xicola RM, Llor X, Pons E, Castells A, Alenda C, Pinol V, Andreu M, Castellvi-Bel S, Paya A, Jover R, Bessa X, Giros A, Duque JM, Nicolas-Perez D, Garcia AM, Rigau J, Gassull MA. Gastrointestinal Oncology Group of the Spanish Gastroenterological Association. Performance of different microsatellite marker panels for detection of mismatch repair-deficient colorectal tumors. J Natl Cancer Inst. 2007;99:244–252. doi: 10.1093/jnci/djk033. [DOI] [PubMed] [Google Scholar]
  • 26.Boland CR, Thibodeau SN, Hamilton SR, Sidransky D, Eshleman JR, Burt RW, Meltzer SJ, Rodriguez-Bigas MA, Fodde R, Ranzani GN, Srivastava S. A National Cancer Institute Workshop on Microsatellite Instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res. 1998;58:5248–5257. [PubMed] [Google Scholar]
  • 27.Halford SE, Sawyer EJ, Lambros MB, Gorman P, Macdonald ND, Talbot IC, Foulkes WD, Gillett CE, Barnes DM, Akslen LA, Lee K, Jacobs IJ, Hanby AM, Ganesan TS, Salvesen HB, Bodmer WF, Tomlinson IP, Roylance RR. MSI-low, a real phenomenon which varies in frequency among cancer types. J Pathol. 2003;201:389–394. doi: 10.1002/path.1453. [DOI] [PubMed] [Google Scholar]
  • 28.Tomlinson I, Halford S, Aaltonen L, Hawkins N, Ward R. Does MSI-low exist? J Pathol. 2002;197:6–13. doi: 10.1002/path.1071. [DOI] [PubMed] [Google Scholar]
  • 29.Boardman LA, Johnson RA, Petersen GM, Oberg AL, Kabat BF, Slusser JP, Wang L, Morlan BW, French AJ, Smyrk TC, Lindor NM, Thibodeau SN. Higher frequency of diploidy in young-onset microsatellite-stable colorectal cancer. Clin Cancer Res. 2007;13:2323–2328. doi: 10.1158/1078-0432.CCR-06-2739. [DOI] [PubMed] [Google Scholar]
  • 30.Derks S, Postma C, Carvalho B, van den Bosch SM, Moerkerk PT, Herman JG, Weijenberg MP, de Bruine AP, Meijer GA, van Engeland M. Integrated analysis of chromosomal, microsatellite and epigenetic instability in colorectal cancer identifies specific associations between promoter methylation of pivotal tumour suppressor and DNA repair genes and specific chromosomal alterations. Carcinogenesis. 2008;29:434–439. doi: 10.1093/carcin/bgm270. [DOI] [PubMed] [Google Scholar]
  • 31.Peltomaki P, Lothe RA, Aaltonen LA, Pylkkanen L, Nystrom-Lahti M, Seruca R, David L, Holm R, Ryberg D, Haugen A, Brogger A, Borresen AL, de la Chapelle A. Microsatellite instability is associated with tumors that characterize the hereditary non-polyposis colorectal carcinoma syndrome. Cancer Res. 1993;53:5853–5855. [PubMed] [Google Scholar]
  • 32.Pal T, Permuth-Wey J, Kumar A, Sellers TA. Systematic review and meta-analysis of ovarian cancers: estimation of microsatellite-high frequency and characterization of mismatch repair deficient tumor histology. Clin Cancer Res. 2008;14:6847–6854. doi: 10.1158/1078-0432.CCR-08-1387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Shia J, Black D, Hummer AJ, Boyd J, Soslow RA. Routinely assessed morphological features correlate with microsatellite instability status in endometrial cancer. Hum Pathol. 2008;39:116–125. doi: 10.1016/j.humpath.2007.05.022. [DOI] [PubMed] [Google Scholar]
  • 34.Adem C, Soderberg CL, Cunningham JM, Reynolds C, Sebo TJ, Thibodeau SN, Hartmann LC, Jenkins RB. Microsatellite instability in hereditary and sporadic breast cancers. Int J Cancer. 2003;107:580–582. doi: 10.1002/ijc.11442. [DOI] [PubMed] [Google Scholar]
  • 35.Boland CR, Goel A. Microsatellite instability in colorectal cancer. Gastroenterology. 2010;138:2073–2087e3. doi: 10.1053/j.gastro.2009.12.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Raut CP, Pawlik TM, Rodriguez-Bigas MA. Clinico-pathologic features in colorectal cancer patients with microsatellite instability. Mutat Res. 2004;568:275–282. doi: 10.1016/j.mrfmmm.2004.05.025. [DOI] [PubMed] [Google Scholar]
  • 37.Hampel H, Frankel WL, Martin E, Arnold M, Khanduja K, Kuebler P, Clendenning M, Sotamaa K, Prior T, Westman JA, Panescu J, Fix D, Lockman J, LaJeunesse J, Comeras I, de la Chapelle A. Feasibility of screening for Lynch syndrome among patients with colorectal cancer. J Clin Oncol. 2008;26:5783–5788. doi: 10.1200/JCO.2008.17.5950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Mrkonjic M, Roslin NM, Greenwood CM, Raptis S, Pollett A, Laird PW, Pethe VV, Chiang T, Daftary D, Dicks E, Thibodeau SN, Gallinger S, Parfrey PS, Younghusband HB, Potter JD, Hudson TJ, McLaughlin JR, Green RC, Zanke BW, Newcomb PA, Paterson AD, Bapat B. Specific variants in the MLH1 gene region may drive DNA methylation, loss of protein expression, and MSI-H colorectal cancer. PLoS One. 2010;5 doi: 10.1371/journal.pone.0013314. e13314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Poynter JN, Siegmund KD, Weisenberger DJ, Long TI, Thibodeau SN, Lindor N, Young J, Jenkins MA, Hopper JL, Baron JA, Buchanan D, Casey G, Levine AJ, Le Marchand L, Gallinger S, Bapat B, Potter JD, Newcomb PA, Haile RW, Laird PW. Colon Cancer Family Registry Investigators. Molecular characterization of MSI-H colorectal cancer by MLHI promoter methylation, immunohistochemistry, and mismatch repair germline mutation screening. Cancer Epidemiol Biomarkers Prev. 2008;17:3208–3215. doi: 10.1158/1055-9965.EPI-08-0512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Dedes KJ, Lopez-Garcia MA, Geyer FC, Lambros MB, Savage K, Vatcheva R, Wilkerson P, Wetterskog D, Lacroix-Triki M, Natrajan R, Reis-Filho JS. Cortactin gene amplification and expression in breast cancer: a chromogenic in situ hybridisation and immunohistochemical study. Breast Cancer Res Treat. 2010;124:653–666. doi: 10.1007/s10549-010-0816-0. Breast Cancer Res Treat. 2010. [DOI] [PubMed] [Google Scholar]
  • 41.Lambros MB, Natrajan R, Geyer FC, Lopez-Garcia MA, Dedes KJ, Savage K, Lacroix-Triki M, Jones RL, Lord CJ, Linardopoulos S, Ashworth A, Reis-Filho JS. PPM1D gene amplification and overexpression in breast cancer: a qRT-PCR and chromogenic in situ hybridization study. Mod Pathol. 2010;23:1334–45. doi: 10.1038/modpathol.2010.121. [DOI] [PubMed] [Google Scholar]
  • 42.Dedes KJ, Natrajan R, Lambros MB, Geyer FC, Lopez-Garcia MA, Savage K, Jones RL, Reis-Filho JS. Down-regulation of the miRNA master regulators Drosha and Dicer is associated with specific subgroups of breast cancer. Eur J Cancer. 2010 doi: 10.1016/j.ejca.2010.08.007. epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 43.Marchio C, Iravani M, Natrajan R, Lambros MB, Savage K, Tamber N, Fenwick K, Mackay A, Senetta R, Di Palma S, Schmitt FC, Bussolati G, Ellis LO, Ashworth A, Sapino A, Reis-Filho JS. Genomic and immunophenotypical characterization of pure micropapillary carcinomas of the breast. J Pathol. 2008;215:398–410. doi: 10.1002/path.2368. [DOI] [PubMed] [Google Scholar]
  • 44.Natrajan R, Lambros MB, Rodriguez-Pinilla SM, Moreno-Bueno G, Tan DS, Marchio C, Vatcheva R, Rayter S, Mahler-Araujo B, Fulford LG, Hungermann D, Mackay A, Grigoriadis A, Fenwick K, Tamber N, Hardisson D, Tutt A, Palacios J, Lord CJ, Buerger H, Ashworth A, Reis-Filho JS. Tiling path genomic profiling of grade 3 invasive ductal breast cancers. Clin Cancer Res. 2009;15:2711–2722. doi: 10.1158/1078-0432.CCR-08-1878. [DOI] [PubMed] [Google Scholar]
  • 45.Natrajan R, Lambros MB, Geyer FC, Marchio C, Tan DS, Vatcheva R, Shiu KK, Hungermann D, Rodriguez-Pinilla SM, Palacios J, Ashworth A, Buerger H, Reis-Filho JS. Loss of 16q in high grade breast cancer is associated with estrogen receptor status: Evidence for progression in tumors with a luminal phenotype? Genes Chromosomes Cancer. 2009;48:351–365. doi: 10.1002/gcc.20646. [DOI] [PubMed] [Google Scholar]
  • 46.Marchio C, Iravani M, Natrajan R, Lambros MB, Geyer FC, Savage K, Parry S, Tamber N, Fenwick K, Mackay A, Schmitt FC, Bussolati G, Ellis I, Ashworth A, Sapino A, Reis-Filho JS. Mixed micropapillary-ductal carcinomas of the breast: a genomic and immunohistochemical analysis of morphologically distinct components. J Pathol. 2009;218:301–315. doi: 10.1002/path.2572. [DOI] [PubMed] [Google Scholar]
  • 47.Zhou XP, Hoang JM, Li YJ, Seruca R, Carneiro F, Sobrinho-Simoes M, Lothe RA, Gleeson CM, Russell SE, Muzeau F, Fléjou JF, Hoang-Xuan K, Lidereau R, Thomas G, Hamelin R. Determination of the replication error phenotype in human tumors without the requirement for matching normal DNA by analysis of mononucleotide repeat microsatellites. Genes Chromosomes Cancer. 1998;21:101–107. doi: 10.1002/(sici)1098-2264(199802)21:2<101::aid-gcc4>3.0.co;2-4. [DOI] [PubMed] [Google Scholar]
  • 48.Toyama T, Iwase H, Yamashita H, Iwata H, Yamashita T, Ito K, Hara Y, Suchi M, Kato T, Nakamura T, Kobayashi S. Microsatellite instability in sporadic human breast cancers. Int J Cancer. 1996;68:447–451. doi: 10.1002/(SICI)1097-0215(19961115)68:4<447::AID-IJC8>3.0.CO;2-0. [DOI] [PubMed] [Google Scholar]
  • 49.Kakar S, Aksoy S, Burgart LJ, Smyrk TC. Mucinous carcinoma of the colon: correlation of loss of mismatch repair enzymes with clinicopathologic features and survival. Mod Pathol. 2004;17:696–700. doi: 10.1038/modpathol.3800093. [DOI] [PubMed] [Google Scholar]
  • 50.Messerini L, Vitelli F, De Vitis LR, Mori S, Calzolari A, Palmirotta R, Calabro A, Papi L. Microsatellite instability in sporadic mucinous colorectal carcinomas: relationship to clinico-pathological variables. J Pathol. 1997;182:380–384. doi: 10.1002/(SICI)1096-9896(199708)182:4<380::AID-PATH871>3.0.CO;2-U. [DOI] [PubMed] [Google Scholar]
  • 51.Ohwada M, Suzuki M, Saga Y, Sato I. DNA replication errors are frequent in mucinous cystade-nocarcinoma of the ovary. Cancer Genet Cytogenet. 2000;117:61–65. doi: 10.1016/s0165-4608(99)00145-4. [DOI] [PubMed] [Google Scholar]
  • 52.Lynch HT, Lynch PM, Lanspa SJ, Snyder CL, Lynch JF, Boland CR. Review of the Lynch syndrome: history, molecular genetics, screening, differential diagnosis, and medicolegal ramifications. Clin Genet. 2009;76:1–18. doi: 10.1111/j.1399-0004.2009.01230.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Nielsen TO, Hsu FD, Jensen K, Cheang M, Karaca G, Hu Z, Hernandez-Boussard T, Livasy C, Cowan D, Dressler L, Akslen LA, Ragaz J, Gown AM, Gilks CB, van de Rijn M, Perou CM. Clin Cancer Res. 2004;10:5367–5374. doi: 10.1158/1078-0432.CCR-04-0220. [DOI] [PubMed] [Google Scholar]

Articles from International Journal of Clinical and Experimental Pathology are provided here courtesy of e-Century Publishing Corporation

RESOURCES