A crucial step in human breast cancer progression is the acquisition of invasiveness. There is a distinct lack of human cell culture models to study the transition from preinvasive to invasive phenotype as it may occur “spontaneously” in vivo. To delineate molecular alterations important for this transition, we isolated human breast epithelial cell lines that showed partial loss of tissue polarity in three-dimensional reconstituted basement membrane cultures. These cells remained noninvasive; however, unlike their nonmalignant counterparts, they exhibited a high propensity to acquire invasiveness through basement membrane in culture. The genomic aberrations and gene expression profiles of the cells in this model showed a high degree of similarity to primary breast tumor profiles. The xenograft tumors formed by the cell lines in three different microenvironments in nude mice displayed metaplastic phenotypes, including squamous and basal characteristics, with invasive cells exhibiting features of higher-grade tumors. To find functionally significant changes in transition from preinvasive to invasive phenotype, we performed attribute profile clustering analysis on the list of genes differentially expressed between preinvasive and invasive cells. We found integral membrane proteins, transcription factors, kinases, transport molecules, and chemokines to be highly represented. In addition, expression of matrix metalloproteinases MMP9, MMP13, MMP15, and MMP17 was up-regulated in the invasive cells. Using small interfering RNA–based approaches, we found these MMPs to be required for the invasive phenotype. This model provides a new tool for dissection of mechanisms by which preinvasive breast cells could acquire invasiveness in a metaplastic context. [Cancer Res 2008;68(5):1378–87]

Breast carcinomas are phenotypically and behaviorally heterogeneous (1). Subtypes of breast cancer with distinct behavioral characteristics have been recognized morphologically as well as by molecular analyses. For all breast cancers, conversion to the invasive phenotype is distinct from metastasis but is an obligate prerequisite for metastasis, thus representing a crucial step in cancer progression. Molecular profiling studies have begun to determine markers that are associated with progression in different breast cancer subtypes (217). Relevant models that recapitulate the morphologic and molecular aspects of progression within subtypes are needed to determine which of these molecular changes function in acquisition of the invasive phenotype.

Culture models of human breast cells could provide such an opportunity if relevance to cancer progression in vivo were established. Pathways associated with transition to malignancy have traditionally been studied using either transgenic mouse models or primary human cells engineered to express genes with oncogenic properties. To deal with the heterogeneity of human breast tumors, cell lines originating from a large number of malignant breast carcinomas have been used (18). It would also be useful to have models of “spontaneous” transition to malignancy, as opposed to transgenic, which could include intermediate noninvasive and preinvasive stages of carcinogenesis, as opposed to modeling only the invasive carcinoma stage. Such models could facilitate the discovery of steps in progression to the invasive phenotype in a context that recapitulates more the complexity of carcinogenesis.

The HMT-3522 human breast epithelial cell line series (S1, S2, and T4-2) were derived from a reduction mammoplasty specimen of a patient with fibrocystic breast disease more than 20 years ago. The epithelial component of the tissue was grown in defined medium to give rise to S1 cells that became immortalized spontaneously; these cells are epidermal growth factor (EGF) dependent for growth and are nontumorigenic (19). Continuous culturing in the absence of EGF gave rise to a cell population referred to as S2, a cell line that is heterogeneous and essentially still nonmalignant (20). Inoculation of S2 cells into mice produced a rare tumor that was cultured and passaged again through mice (20). The cell line derived from this tumor is referred to as T4-2 (20). Previously, we designed a three-dimensional laminin-rich basement membrane (3DlrBM) assay to model both the normal breast and breast cancers (ref. 21; Fig. 1A). In 3DlrBM, S1 cells produce growth-arrested acini that express differentiation markers mimicking in vivo polar patterns of localization of a number of markers (21): S2 colonies are heterogeneous (this study) and T4-2 cells form large and disorganized colonies (22).

Figure 1.

Partial loss of tissue polarity and preinvasive phenotypes of S3 cells. A, steps involved in the isolation of the HMT-3522 series of cell lines. B, 3DlrBM cultures of HMT-3522-S2 cells and small (S3-A), medium (S3-B), and large (S3-C) colonies isolated from the parent culture of S2 on day 10. C, confocal images of immunostained colonies in 3DlrBM, day 10. D, invasion assays: two experiments, triplicate samples. Black columns, untreated cells; gray columns, cells treated with T4-2 conditioned medium (CM).

Figure 1.

Partial loss of tissue polarity and preinvasive phenotypes of S3 cells. A, steps involved in the isolation of the HMT-3522 series of cell lines. B, 3DlrBM cultures of HMT-3522-S2 cells and small (S3-A), medium (S3-B), and large (S3-C) colonies isolated from the parent culture of S2 on day 10. C, confocal images of immunostained colonies in 3DlrBM, day 10. D, invasion assays: two experiments, triplicate samples. Black columns, untreated cells; gray columns, cells treated with T4-2 conditioned medium (CM).

Close modal

Here, we isolated three essentially homogeneous cell populations from S2 cells, which we refer to as S3-A, S3-B, and S3-C, using colony size in 3DlrBM as a screening tool (Fig. 1A). These cells were noninvasive but had a higher potential for acquiring invasiveness than the parent S2. We found a substantial number of similarities between the model of S3 versus T4-2 and preinvasive to invasive transition in breast carcinogenesis based on progressive loss of tissue polarity, ability to form tumors with squamous and basal metaplastic histology, 76% similarity to recurrent comparative genomic hybridization (CGH) abnormalities found in breast tumors, and gene expression changes in classes of genes previously identified as being important for malignancy and invasion. These observations establish the S3 cell lines as the preinvasive counterpart of a model of breast cancer progression within a metaplastic context. Using matrix metalloproteinases (MMP), we show the utility of this model in identifying functionally significant changes in transition from the preinvasive to the invasive phenotype in metaplastic breast cancer. The usefulness of the model is further shown in another report (23), which describes the discovery of a novel pathway involved in invasion using this progression series.

Cell Culture

S1 and T4-2 cells were grown in tissue culture monolayers two-dimensionally using Falcon tissue culture plastic or three-dimensionally in laminin-rich basement membrane (Matrigel) in defined medium as previously described (21, 22); S2 and S3 cells were grown under the same conditions as T4-2.

Indirect Immunofluorescence Analysis and Image Acquisition in 3DlrBM

Cells in 3DlrBM were perfused in sucrose and frozen on dry ice in Tissue-Tek optimum cutting temperature (Miles Laboratories), sectioned, and immunostained as previously described (22).

Comparative Genomic Hybridization

Chromosomal CGH and BAC array CGH were done essentially as previously described (24). Hierarchical clustering analysis was done with GeneSpring software (Silicon Genetics), using standard correlation with a minimum distance metric of 0.001 to determine relatedness of cell lines. This was done using both the chromosomal and the BAC array data, with only significant amplification/deletion data points, as well as with the complete data sets, producing comparable results.

Analysis of Tumor Formation In vivo

We injected the cells into female BALB/c athymic nude mice (Simonsen Laboratories), measured tumors weekly with a caliper, and graphed the tumor frequency and volumes of tumors taken out after 9 to 10 weeks of tumor growth, at which time the animals were sacrificed. Statistical analysis of the mean tumor volumes was done by pairwise comparison using one-tailed homoscedastic t test analysis. We injected 10 million cells s.c. into the rear flanks, 5 million cells in 50% Matrigel s.c. into the front flanks, or 2 million cells into the left and right fourth inguinal mammary glands. Tumors from all injections and surrounding tissues were fixed in 4% formaldehyde, paraffin embedded, sectioned into 5-μm slices, and stained with H&E (HRL Laboratories). Three hundred fifty H&E sections were independently examined by two expert breast pathologists (A.D.B. and R.A.J.).

Global mRNA Expression Analysis by cDNA Microarrays

cDNA microarrays with ∼8,000 known gene spots on poly-l-lysine–coated chips (custom arrayed at LBNL using Research Genetics 8k human clones) were used. mRNA samples were directly compared with each other by cohybridization to the same slide using dendrimer technology to label with red-Cy5 or green-Cy3 (Genisphere). Total RNA (1 μg) isolated with Qiagen RNEasy reagents was used for each sample hybridized. Cells in 3DlrBM were extracted with 5 mmol/L EDTA in cold PBS to dissolve the Matrigel. For each comparison, three independent sets of cultures were processed, and cells were examined at day 10 of culture. For each comparison, four slides were hybridized. This corresponded to three sets of RNAs from independent cultures plus a dye-swap experiment in which the red and green labels were switched for the two samples in question. Data were analyzed as described in Supplementary File 8.

Reverse Transcription-PCR

Quantitative. The RNA prepared for microarrays was used to prepare cDNA after DNase I (Invitrogen) treatment, using Superscript First-Strand Synthesis System for reverse transcription-PCR (RT-PCR; Invitrogen). Quantitative real-time PCR was done using LightCycler (Roche) and FastStart SYBR Green (Roche). Primer sequences were as follows (5′-3′): MMP13 forward, aagatgcatccaggggtcct; MMP13 reverse, gtccaggtttcatcatcatca; glyceraldehyde-3-phosphate dehydrogenase (GAPDH) forward, cccctggccaaggtcatccatgac; and GAPDH reverse, cataccaggaaatgagcttgacaaag. LightCycler reaction cycles were as recommended by Roche with the following modifications: for MMP13, 57°C for annealing, 27 seconds of extension, and signal acquisition at 84°C; for GAPDH, 56°C for annealing and 25 seconds for extension.

Semiquantitative. For MMP9, MMP13, MMP15, and MMP17, semiquantitative RT-PCR was done using the following primer sets (5′-3′): MMP9, atgcgtggagagtcgaaatc (forward) and tacacgcgagtgaaggtgag (reverse); MMP13, aagatgcatccaggggtcct (forward) and gtccaggtttcatcatcatca (reverse); MMP15, ccatatgtccaccatgcgtt (forward) and atgatggcattggggttgct (reverse); and MMP17, acgcaagaggagctgtctaag (forward) and acatggcttaacccaatggc (reverse). The conditions used (after having determined linear range) were, for MMP9, MMP15, and MMP17, 96°C 3 minutes, 34 × (96°C 30 seconds, 55°C 1 minutes, 72°C 1 minutes), 72°C 5 minutes; for MMP13, 96°C 3 minutes, 29 × (96°C 30 seconds, 55°C 30 seconds, 72°C 1 minutes), 72°C 5 minutes.

Expression Analysis of Cell-Surface Proteins by Fluorescence-Activated Cell Sorting

Live cells were immunostained in suspension before fixing with 2% paraformaldehyde. Primary antibodies were used at 1:10 dilution, and FITC-conjugated secondary antibodies were used at 1:100 dilution. Primary antibodies were MMP15, clone 162-22G5 (Oncogene); MMP17, rabbit polyclonal 475934 (Calbiochem). Fluorescence-activated cell sorting (FACS) analysis was done using EPICS XL-MCL data acquisition and display software on XL flow cytometry analyzers (University of California Berkeley, Flow Cytometry Facility). Gating of forward light scatter versus light scatter (90-degree scatter) allowed us to obtain data relevant to intact cells only; FITC fluorescence peak was evaluated for its median value and was corrected using samples that had not been treated with primary antibody.

Gelatin Zymography

Conditioned medium (for 48 hours) from 10-day cultures of S1, S2, S3, and T4-2 were used. Samples were analyzed by SDS-PAGE zymography (25) to determine the molecular weights and the relative abundance of the gelatinases present.

Invasion Assay

The ability to invade through laminin-rich basement membrane (Matrigel) was measured in Boyden chamber assays, essentially as described (26). The number of invading T4-2 or S3-C cells (of 1 × 105 seeded) was determined after 48 hours of incubation in regular growth medium, in medium containing T4-2 conditioned medium, in medium with DMSO with different concentrations of the MMP inhibitor GM6001 (AMS Scientific) or its inactive analogue (C1004), or in conditioned medium that had previously been treated with DMSO, GM6001, or C1004. For small interfering RNA (siRNA)–treated T4-2 cells, transfection of 30 to 150 nmol/L oligo with 4 μL of siPORT NeoFX (Ambion) per milliliter of media was done 1 day after plating cells at regular density. siRNAs were allowed to down-regulate protein levels for 2 days. Cells were then trypsinized and 1 × 105 cells were seeded for Boyden chamber assays. siRNAs against MMP9 (oligo 1, Ambion ID143941; oligo 2, Ambion ID 113182: oligo 3, Ambion ID 113183), MMP13 (oligo 1, Ambion ID 143556; oligo 2, Ambion ID 212725; oligo 3, Ambion ID 112915), MMP15 (oligo 1, Ambion ID 112917; oligo 2, Ambion ID 143557; oligo 3, Ambion ID 112916), MMP17 (oligo 1, Ambion ID 105396; oligo 2, Ambion ID 113514; oligo 3, Ambion ID 24016), or scrambled control siRNA (Ambion, Silencer-Cy3 labeled) were used.

Immunohistochemistry

Ki67. Formalin-fixed mouse xenograft tissue was paraffin embedded and sectioned into 5-μm-thick tissue sections (HRL Laboratories). The paraffin was removed by serial incubation in xylene, 100% ethanol, 95% ethanol, 70% ethanol, and water. Tissues were blocked with 3% hydrogen peroxide in PBS for 5 minutes. Antigen retrieval was done by incubating in 0.01% prewarmed trypsin in PBS for 15 minutes, followed by 10 minutes of microwaving in 10 mmol/L sodium citrate buffer. Tissues were blocked in 1.5% normal horse serum in PBS for 30 minutes and incubated with primary Ki67 antibody, which specifically recognizes human, but not mouse, Ki67 protein [Calbiochem, anti-Ki67 human (mouse), NA59] at 5 μg/mL overnight at 4°C. Slides were washed with PBS and incubated with biotinylated antirabbit antibody [1:200 dilution; Vector Laboratories, biotinylated antimouse IgG/antirabbit IgG (H + L), BA-1400] for 30 minutes at room temperature, followed by streptavidin-horseradish peroxidase (Vector Laboratories, Vectastain ABC kit, Elite PK-6100) for 30 minutes and complete 3,3′-diaminobenzidine tetrahydrochloride (Sigma) medium for 5 minutes. Slides were washed and counterstained with hematoxylin, followed by dehydration in 70% ethanol, 95% ethanol, 100% ethanol, and xylene; coverslips were mounted using Permount (Fisher Scientific, SP15). Strong nuclear staining was scored as positive and %Ki67 positive nuclei was determined for at least three tumor areas for each cell type injection, counting a minimum of 100 cells per area.

p63. Immunohistochemistry was done essentially as described for Ki67, except for omitting the trypsin step in antigen retrieval and increasing hydrogen peroxide blocking to 15 minutes. Primary antibody (Lab Vision, p63 Ab-1, clone 4A4) was used at 2 μg/mL.

CK5/6, estrogen receptor, and progesterone receptor. Immunohistochemistry was done as described in ref. 27, using CK5/6 primary antibodies from Zymed (1:100), estrogen receptor from DAKO (1:25), and progesterone receptor from DAKO (1:200); for HER2-neu, the Herceptest kit from DAKO was used.

S3 cells display partial loss of tissue polarity in 3DlrBM, a preinvasive phenotype. Using colony size in 3DlrBM as an initial screen, we isolated multiple small-, medium-, and large-sized colonies; expanded and propagated these clones for at least six generations; and examined their morphology in the three-dimensional assay (Fig. 1B). We chose three isolates that essentially bred true for the size of colony formed. We refer to these as S3-A, S3-B, and S3-C cell lines where colony size is A < B < C. S3-A cultures contain ∼10% very large colonies (not shown); otherwise the colony size, especially for S3-Cs, is essentially homogeneous. S3 cells displayed intermediate phenotypes compared with S1 and T4-2 colonies in 3DlrBM, as determined by markers that have been extensively used to describe tissue (acinar) polarity (22, 2831), such as basolateral expression of β-catenin, basal β4 integrin, cortical actin, and Ki67 labeling (Fig. 1C). In Boyden chamber assays, S1, S2, and S3 cell lines did not invade laminin-rich basement membrane, but T4-2 cells were invasive (Fig. 1D). When conditioned medium from T4-2 cells was added to the chambers, S1 and S2 cells did not invade, whereas S3-A, S3-B, and S3-C became invasive, with the S3-C cells displaying the highest propensity to invade: T4-2 > C > B > A.

S3 cells display an intermediate potential to form tumors. We determined tumorigenicity of the cells when injected into nude mice under three different microenvironments: in the flank with cells alone or cells plus Matrigel, and into mammary fat pads with cells alone (Fig. 2). In none of the injections did S1 cells or media control produce any measurable growth but T4-2 cells formed palpable tumors at high frequency, as shown also previously (refs. 20, 22, 32, 33; Fig. 2A,–C, top) When injected s.c., S2 cells and the S3 series formed very small tumors at low frequencies. S.c. injection in the presence of Matrigel (Fig. 2A,–C, middle) resulted in tumor formation in 50% of S2, 75% of S3-A, 73% of S3-B, 56% of S3-C (see following section on tumor histology), and 100% of T4-2 cell injection sites. Importantly, mammary fat pad injections (Fig. 2A,–C, bottom) resulted in progressively increasing tumor frequency going from S2, S3-A, S3-B, S3-C, to T4-2, mirroring the rate of invasion observed in Boyden chambers (Fig. 1D). Comparing the P values for tumor volume (Fig. 2C) showed that the mammary fat pad most clearly distinguished the nontumorigenic S1, the preinvasive S3, and the malignant T4-2.

Figure 2.

S3 cells display low tumorigenicity and squamous metaplastic phenotype in nude mice. A, percent injection sites that produced tumors sustained for the duration of the experiment. Top, s.c. injections (three experiments); 14 S1, 22 S2, 20 S3-A, 16 S3-B, 22 S3-C, 30 T4-2, and 3 media controls. Middle, s.c. + Matrigel (laminin-rich basement membrane, lrBM) injections (two experiments); 18 S1, 18 S2, 20 S3-A, 22 S3-B, 18 S3-C, and 18 T4-2 injections. Bottom, mammary fat pad injections; 15 S1, 16 S2, 16 S3-A, 16 S3-B, 15 S3-C, 24 T4-2, and 4 media controls. B, mean tumor volume (in mm3) excised from sacrificed animals. C, P values for the volume comparisons indicated; P < 0.05 in bold. Top, s.c. injections; middle, s.c. + Matrigel injections; bottom, mammary fat pad injections. D, H&E images of S1, S2, and S3-B s.c. + Matrigel tumors and of S3-A, S3-C, and T4-2 fat pad tumors. Bar, 50 μm. S1, low-grade adenosquamous carcinoma-like phenotype at the injection site; S2, squamous differentiation with whorls and bridges, transition from cuboidal cells to larger squamous cells in the center; S3-A, well-differentiated area with a small cyst, less well differentiated area with calcification; S3-B, cords of cells surrounding areas of extracellular matrix showing solid tumor areas with proliferation and squamous differentiation and displaying pleomorphic adenoma phenotype; S3-C, squamous differentiation, foamy squamous carcinoma cells, abundant stromal reaction; T4-2, squamous carcinoma invading the skeletal muscle.

Figure 2.

S3 cells display low tumorigenicity and squamous metaplastic phenotype in nude mice. A, percent injection sites that produced tumors sustained for the duration of the experiment. Top, s.c. injections (three experiments); 14 S1, 22 S2, 20 S3-A, 16 S3-B, 22 S3-C, 30 T4-2, and 3 media controls. Middle, s.c. + Matrigel (laminin-rich basement membrane, lrBM) injections (two experiments); 18 S1, 18 S2, 20 S3-A, 22 S3-B, 18 S3-C, and 18 T4-2 injections. Bottom, mammary fat pad injections; 15 S1, 16 S2, 16 S3-A, 16 S3-B, 15 S3-C, 24 T4-2, and 4 media controls. B, mean tumor volume (in mm3) excised from sacrificed animals. C, P values for the volume comparisons indicated; P < 0.05 in bold. Top, s.c. injections; middle, s.c. + Matrigel injections; bottom, mammary fat pad injections. D, H&E images of S1, S2, and S3-B s.c. + Matrigel tumors and of S3-A, S3-C, and T4-2 fat pad tumors. Bar, 50 μm. S1, low-grade adenosquamous carcinoma-like phenotype at the injection site; S2, squamous differentiation with whorls and bridges, transition from cuboidal cells to larger squamous cells in the center; S3-A, well-differentiated area with a small cyst, less well differentiated area with calcification; S3-B, cords of cells surrounding areas of extracellular matrix showing solid tumor areas with proliferation and squamous differentiation and displaying pleomorphic adenoma phenotype; S3-C, squamous differentiation, foamy squamous carcinoma cells, abundant stromal reaction; T4-2, squamous carcinoma invading the skeletal muscle.

Close modal

Tumor histology reveals similarities to cancer progression within a metaplastic context. In s.c. injections (Supplementary File 1B), the T4-2 tumors had viable, dividing cells with squamous metaplastic morphology. The S2 and S3 tumors had nondividing cells, suggesting they did not maintain growth, but they contained keratin clusters and inflammatory cells, which are characteristics of well-differentiated squamous carcinomas of the breast. An average increase of 27-fold in tumor volume was observed when half the number of cells was injected in the presence of Matrigel. In the Matrigel injections (Fig. 2A,–D, middle; Supplementary File 1A and C), 5 of 18 of the S1 injection sites showed a phenotype similar to low-grade adenosquamous carcinomas despite the lack of a palpable tumor at the time of sacrifice (Fig. 2J). Whereas 100% of the S2 and S3 tumors displayed the more benign mixed tumor histology, only 25% of the T4-2 tumors were mixed, with the remainder being pure squamous. In addition, T4-2 tumors had features of higher-grade squamous carcinoma than did the S2 and S3 tumors: 83% of the S2 and S3 tumors were well differentiated, compared with only 11% of the T4-2 tumors. Soft tissue involvement, indicating either invasion or proliferative expansion into neighboring tissues, was observed only in T4-2 tumors (Fig. 2J). Similarly, calcifications, which are an indication of necrosis usually observed in high-grade tumors, were found in almost all T4-2 tumors but only in a few of S2 or S3s. The fat pad tumor histology also showed similarities to squamous metaplasias of the breast (Fig. 2J; Supplementary File 1D), with T4-2 tumors being of higher “grade” in general based on the degree of differentiation, soft tissue involvement, and calcification phenotypes.

S3 cells display genomic aberrations that are distinct from those found in S1 and T4-2, but are relevant to some human breast cancers. We determined the genomically amplified or deleted regions by CGH (Supplementary File 2). The number of aberrations increased progressively when S1, S2-S3, and T4-2 cells were compared (Fig. 3A and B). Hierarchical clustering analysis (Fig. 3C) showed that the S2-S3 and T4-2 cell lines were more closely related to each other than they were to the S1 cells, and that the malignant T4-2 chromosomal aberration profiles were distinct from those of the S2-S3 cells. S2, S3, and T4-2 cell lines contained 76% (22 of 29) of the chromosomal gains or losses that are reported to be commonly found in primary breast tumors (Fig. 3D ; Supplementary File 2). Of the aberrations recurrently found in carcinoma in situ (preinvasive), 6 of 10 were present in S3-A, S3-B, and S3-C cells, but all 10 were found in T4-2. The aberrations that were present in T4-2, but not in S3, were those found in high-grade in situ or invasive carcinomas.

Figure 3.

CGH profile of S3 cells compared with other HMT-3522 cells and with primary tumors (for details of CGH data, see Supplementary File 2). A, chromosomal amplifications (red) and deletions (green) for each cell type examined at the indicated passage number (e.g., S1_27: S1 cells at passage 27). B, chromosomal aberrations common to all cell lines shown on the left (e.g., “S1” means common to all S1 lineages shown in A). C, hierarchical clustering of 119 significantly amplified or deleted regions for the indicated cell lines. D, chromosomal aberrations commonly found in primary tumors, as described in each study cited; shown in bold if shared with HMT-3522 cell lines; complete list of aberrations shared between the cell lines and primary tumors listed at the bottom (TOTAL).

Figure 3.

CGH profile of S3 cells compared with other HMT-3522 cells and with primary tumors (for details of CGH data, see Supplementary File 2). A, chromosomal amplifications (red) and deletions (green) for each cell type examined at the indicated passage number (e.g., S1_27: S1 cells at passage 27). B, chromosomal aberrations common to all cell lines shown on the left (e.g., “S1” means common to all S1 lineages shown in A). C, hierarchical clustering of 119 significantly amplified or deleted regions for the indicated cell lines. D, chromosomal aberrations commonly found in primary tumors, as described in each study cited; shown in bold if shared with HMT-3522 cell lines; complete list of aberrations shared between the cell lines and primary tumors listed at the bottom (TOTAL).

Close modal

Transition from S3-C to T4-2 phenotype is associated with altered expression of gene classes previously implicated in human breast cancer progression. We determined differences in global gene expression between S3-C and T4-2 when cells were grown in either two-dimensional (monolayers) or 3DlrBM cultures (Supplementary File 3). To discover groups of genes with shared attributes in an unbiased manner, we analyzed these two sets of differentially expressed genes by model-based clustering according to their Gene Ontology (GO) terms and other annotations by developing and using a method called Attribute Profile Clustering (ref. 34; Supplementary File 8). The 141 genes altered between S3-C and T4-2 in two dimensions were divided into four groups summarized as integral membrane proteins, transcription factors, kinases, and transport molecules (Supplementary Files 4, 6, and 8). The groups for the 502 genes with different expression levels between S3-C and T4-2, when grown in three-dimensional cultures were integral membrane proteins, transcription factors, kinases, and chemokines (Supplementary Files 5, 7, and 8). The best-defined group in the comparison of S3-C and T4-2 in three dimensions, but not in two dimensions, revealed a class with 100% of the genes sharing the GO term “Chemokine Activity.” Chemokines have been shown previously to play a significant role in metastasis (35) and were shown to increase dramatically during human breast cancer progression (36).

MMP9, MMP13, MMP15, and MMP17 are functionally significant in the acquisition of invasiveness. In addition to the genes that were relevant to human breast cancer progression by attribute clustering analysis, the microarray data contained another class of genes, the MMPs, which showed differential expression when S3-C and T4-2 were compared only in 3DlrBM. Microarray analysis showed that MMP13 (collagenase-3), MMP15 (MT-2 MMP), and MMP17 (MT-4 MMP) had ∼30% higher expression in T4-2 versus S3-C in three-dimensional cultures (Supplementary File 3). For MMP13, we confirmed these results by quantitative RT-PCR (Fig. 4A) and Western blots of conditioned medium (not shown). For the membrane-bound MMP15 and MMP17, FACS analysis showed that cell-surface expression was higher in T4-2 than in S3-C cells (Fig. 4B and C). MMP15 has been shown to activate proMMP2 by cleavage (37). Therefore, we determined overall gelatinolytic activity, including MMP2 and MMP9 activity, present in S1, S2, S3, and T4-2 cell lines using zymograms. S1 cells expressed proMMP2 but not MMP9; S2, S3-A, and S3-B had no detectable expression of MMP2 or MMP9; S3-C had a small amount of proMMP9 expression; and T4-2 showed high levels of proMMP9 but no MMP2 expression (Fig. 4D).

Figure 4.

Candidate MMPs from microarrays are differentially expressed between S3-C and T4-2 cells. A, quantitative RT-PCR for MMP13, normalized to GAPDH internal control, for S3-C and T4-2 in 3DlrBM (P < 0.05). B and C, cell-surface expression of MMP15 and MMP17; four independent experiments (P < 0.05). D, zymogram detecting gelatinolytic activity in cell lines, compared with p-aminophenylmercuric acetate–activated recombinant MMP2 control.

Figure 4.

Candidate MMPs from microarrays are differentially expressed between S3-C and T4-2 cells. A, quantitative RT-PCR for MMP13, normalized to GAPDH internal control, for S3-C and T4-2 in 3DlrBM (P < 0.05). B and C, cell-surface expression of MMP15 and MMP17; four independent experiments (P < 0.05). D, zymogram detecting gelatinolytic activity in cell lines, compared with p-aminophenylmercuric acetate–activated recombinant MMP2 control.

Close modal

In this model, we could directly assess the significance of MMP expression changes for the invasive phenotype. A broad-spectrum MMP inhibitor, GM6001 (38), abrogated T4-2 invasiveness whereas treatment of cells with C1004, an inactive analogue of GM6001, had no detectable effect (Fig. 5B). To confirm the inhibitor data specifically, we used siRNAs to transiently and individually knock down the levels of MMP9, MMP13, MMP15, and MMP17 (Fig. 5A) and found that siRNAs that successfully knocked down these MMPs decreased the amount of invasiveness of T4-2 cells compared with the scrambled control siRNA (Fig. 5C). MMPs were important for acquisition of invasiveness in S3-C cells as well: addition of GM6001, and not the control C1004, to either the S3-C or T4-2 media (the conditioned medium was taken from the latter) inhibited the ability of the T4-2 conditioned medium to induce invasiveness in S3-C (Fig. 5D).

Figure 5.

Candidate MMPs function in invasion through laminin-rich basement membrane. A, RT-PCR for MMP9, MMP13, MMP15, and MMP17 in cells transfected with siRNAs to the respective MMPs or a scrambled control. B, invasion assays for T4-2 cells treated with the indicated MMP inhibitor or control; two experiments, duplicate samples. C, invasion assays for T4-2 cells transfected with MMP9, MMP13, MMP15, or MMP17 siRNAs versus scrambled control; P values compared with scrambled control. D, invasion assays for S3-C cells; number of invading cells normalized to S3-C cells treated with T4-2 conditioned medium. Posttreated, conditioned medium was added to the S3-C medium after the indicated MMP inhibitor or control (40 μmol/L); Pretreated, conditioned medium was pretreated with the MMP inhibitor or control (40 μmol/L) for 48 h before being added to the S3-C medium in the invasion assays; three experiments.

Figure 5.

Candidate MMPs function in invasion through laminin-rich basement membrane. A, RT-PCR for MMP9, MMP13, MMP15, and MMP17 in cells transfected with siRNAs to the respective MMPs or a scrambled control. B, invasion assays for T4-2 cells treated with the indicated MMP inhibitor or control; two experiments, duplicate samples. C, invasion assays for T4-2 cells transfected with MMP9, MMP13, MMP15, or MMP17 siRNAs versus scrambled control; P values compared with scrambled control. D, invasion assays for S3-C cells; number of invading cells normalized to S3-C cells treated with T4-2 conditioned medium. Posttreated, conditioned medium was added to the S3-C medium after the indicated MMP inhibitor or control (40 μmol/L); Pretreated, conditioned medium was pretreated with the MMP inhibitor or control (40 μmol/L) for 48 h before being added to the S3-C medium in the invasion assays; three experiments.

Close modal

Modeling human breast cancer in culture traditionally has relied on carcinoma-derived cell lines or addition of potent oncogenic stimuli to nonmalignant cells. What we have lacked thus far are models with which we can recapitulate intermediate stages of cancer progression to simulate the complexity of different types of breast cancers (39). Here we describe one such human cell culture model. Neither the process of immortalization nor the process of selection of the preinvasive S3 or malignant T4-2 cells entailed introduction of oncogenic transgenes. The T4-2 population was generated by removal of EGF but provides a transgene-free opportunity to study the involvement of different and possibly unrelated pathways in malignant transition (Table 1; Supplementary File 8). The cell lines produced tumors with various metaplastic features, such as rare low-grade adenosquamous carcinoma, mixed benign tumors, and aggressive squamous metaplasia, suggesting progression within a metaplastic context (Supplementary File 1).

Table 1.

Attribute profile clustering of differentially expressed genes between S3-C and T4-2

GenBank no.GeneAvgPGenBank no.GeneAvgPGenBank no.GeneAvgP
2D integral membrane (20/30)    3D integral membrane (62/62)        
NM_005050 ABCD4 1.17 0.0295 NM_000927 ABCB1 0.82 0.0398 AA450336 NRXN3 0.74 0.0210 
NM_014567 BCAR1 0.74 0.0222 NM_012089 ABCB10 0.61 0.0165 AA598582 OXA1L 0.62 0.0054 
NM_004054 C3AR1 1.20 0.0350 NM_005689 ABCB6 1.22 0.0029 NM_002561 P2RX5 0.74 0.0467 
NM_000722 CACNA2D1 0.62 0.0450 NM_019624 ABCB9 1.33 0.0175 NM_005446 P2RXL1 0.68 0.0446 
NM_013230 CD24 1.29 0.0093 NM_000683 ADRA2C 1.24 0.0321 NM_000919 PAM 1.24 0.0170 
NM_000760 CSF3R 1.17 0.0488 NM_001671 ASGR1 1.50 0.0384 AA663440 PLXNC1 0.83 0.0460 
NM_001381 DOK1 0.80 0.0062 NM_001681 ATP2A2 1.43 0.0335 NM_000952 PTAFR 1.22 0.0019 
NM_000118 ENG 0.64 0.0315 NM_004335 BST2 1.42 0.0106 NM_002838 PTPRC 0.72 0.0020 
NM_014211 GABRP 0.68 0.0164 NM_006137 CD7 1.31 0.0421 NM_002855 PVRL1 1.09 0.0137 
NM_004482 GALNT3 0.88 0.0219 NM_001782 CD72 1.24 0.0439 NM_001037 SCN1B 0.69 0.0030 
H07878 GPR19 1.23 0.0295 NM_004356 CD81 0.80 0.0182 NM_005072 SLC12A4 0.54 0.0230 
NM_000856 GUCY1A3 1.16 0.0171 NM_004233 CD83 0.75 0.0092 NM_015865 SLC14A1 0.77 0.0417 
H11482 IFNGR1 0.74 0.0029 NM_005745 DXS1357E 0.84 0.0156 NM_004207 SLC16A3 0.79 0.0054 
NM_000612 IGF2 1.13 0.0280 NM_001400 EDG1 1.69 0.0181 NM_004694 SLC16A6 1.23 0.0273 
NM_000598 IGFBP3 0.62 0.0092 NM_000604 FGFR1 1.25 0.0311 NM_005495 SLC17A4 1.17 0.0471 
NM_002195 INSL4 1.14 0.0360 NM_006338 GAC1 0.89 0.0454 NM_007256 SLC21A9 0.56 0.0133 
NM_012282 KCNE1L 1.15 0.0007 NM_000160 GCGR 0.72 0.0317 NM_000441 SLC26A4 0.77 0.0389 
NM_002333 LRP3 1.19 0.0085 NM_000832 GRIN1 1.50 0.0285 NM_001860 SLC31A2 0.83 0.0102 
AA496022 MFAP4 0.60 0.0469 NM_000840 GRM3 0.60 0.0367 NM_014437 SLC39A1 1.31 0.0039 
AA598582 OXA1L 0.52 0.0009 NM_005477 HCN4 0.75 0.0172 NM_002394 SLC3A2 1.30 0.0077 
NM_002571 PAEP 1.15 0.0092 AA464246 HLA-C 0.79 0.0248 AA487543 SORL1 0.83 0.0397 
NM_024411 PDYN 1.37 0.0492 NM_033554 HLA-DPA1 1.31 0.0498 NM_000544 TAP2 1.46 0.0217 
NM_004705 PRKRIR 0.80 0.0021 H11482 IFNGR1 0.59 0.0003 NM_006335 TIMM17A 1.28 0.0042 
NM_000452 SLC10A1 1.06 0.0296 NM_000877 IL1R1 0.80 0.0169 NM_005834 TIMM17B 0.79 0.0361 
NM_006749 SLC20A2 0.85 0.0041 NM_004633 IL1R2 0.63 0.0217 NM_003265 TLR3 0.71 0.0272 
NM_003127 SPTAN1 1.17 0.0450 NM_012276 ILT7 1.28 0.0260 NM_006827 TMP21 1.17 0.0109 
NM_003235 TG 1.19 0.0297 NM_002223 ITPR2 0.75 0.0100 NM_001192 TNFRSF17 1.28 0.0190 
NM_003243 TGFBR3 1.41 0.0218 NM_002233 KCNA4 0.73 0.0097 NM_003810 TNFSF10 1.15 0.0373 
NM_012471 TRPC5 0.61 0.0346 NM_002286 LAG3 0.73 0.0064 NM_006564 TYMSTR 0.45 0.0322 
NM_000371 TTR 0.87 0.0485 NM_015364 MD-2 0.68 0.0027 NM_004738 VAPB 1.35 0.0212 
    NM_000908 NPR3 1.17 0.0156 NM_021083 XK 1.23 0.0150 
2D transcription (23/26)    3D transcription (51/51)        
NM_004301 BAF53A 1.19 0.0280 NM_005180 BMI1 0.78 0.0355 NM_006186 NR4A2 1.18 0.0166 
NM_006317 BASP1 0.92 0.0007 NM_000092 COL4A4 0.68 0.0387 NM_003884 PCAF 0.62 0.0031 
NM_012116 CBLC 0.67 0.0249 H54686 DAB2 0.64 0.0163 NM_021128 POLR2L 0.88 0.0464 
NM_003651 CSDA 0.86 0.0318 NM_000399 EGR2 1.37 0.0486 NM_007244 PROL4 1.43 0.0057 
NM_004111 FEN1 0.82 0.0081 NM_006874 ELF2 0.74 0.0044 NM_002892 RBBP1 0.63 0.0012 
NM_001465 FYB 0.96 0.0002 AA010400 ETV4 1.36 0.0371 NM_002919 RFX3 1.22 0.0469 
NM_001515 GTF2H2 0.86 0.0261 NM_001987 ETV6 1.28 0.0386 NM_002938 RNF4 1.22 0.0414 
NM_005324 H3F3B 1.10 0.0482 AA181023 EVI1 1.31 0.0024 NM_002955 RREB1 1.36 0.0482 
NM_005336 HDLBP 0.50 0.0200 AI346582 EVX1 0.69 0.0477 NM_003071 SMARCA3 1.30 0.0208 
NM_014213 HOXD9 0.81 0.0059 NM_003862 FGF18 0.55 0.0374 NM_007017 SOX30 0.82 0.0371 
NM_016270 KLF2 1.32 0.0361 NM_000801 FKBP1A 0.87 0.0237 NM_004509 SP110 0.66 0.0173 
NM_002485 NBS1 1.22 0.0159 NM_005087 FXR1 0.74 0.0038 NM_012446 SSBP2 1.35 0.0139 
NM_004555 NFATC3 1.13 0.0302 NM_002095 GTF2E2 1.32 0.0018 NM_005636 SSX4 0.85 0.0450 
NM_002582 PARN 0.78 0.0034 NM_003483 HMGA2 1.05 0.4378 NM_007375 TARDBP 1.36 0.0089 
NM_002779 PSD 0.59 0.0062 NM_014213 HOXD9 0.58 0.0021 NM_007108 TCEB2 1.19 0.0476 
NM_002938 RNF4 1.17 0.0420 NM_002200 IRF5 1.22 0.0153 NM_003201 TFAM 1.15 0.0263 
NM_007252 RPF-1 0.60 0.0308 NM_005853 IRX5 0.59 0.0014 NM_005077 TLE1 1.18 0.0242 
NM_003079 SMARCE1 1.14 0.0106 NM_002228 JUN 1.30 0.0038 NM_005120 TNRC11 0.61 0.0064 
NM_003084 SNAPC3 0.91 0.0081 NM_014368 LHX6 0.74 0.0210 NM_001068 TOP2B 1.24 0.0498 
NM_005839 SRRM1 1.05 0.0047 NM_012321 LSM4 0.78 0.0046 NM_016936 UBN1 1.27 0.0347 
NM_003152 STAT5A 1.14 0.0462 NM_002397 MEF2C 0.84 0.0118 AI925821 USP22 0.76 0.0034 
NM_006284 TAF10 1.17 0.0133 NM_005933 MLL 0.58 0.0164 NM_000378 WT1 0.67 0.0150 
NM_005644 TAF12 1.24 0.0035 NM_004641 MLLT10 1.24 0.0292 NM_003443 ZNF151 0.62 0.0241 
NM_005481 TRAP95 0.59 0.0271 NM_002479 MYOG 0.73 0.0299 NM_016423 ZNF219 1.27 0.0198 
NM_004236 TRIP15 0.65 0.0394 NM_016170 NCX 1.36 0.0116 NM_005741 ZNF263 1.49 0.0055 
NM_005082 ZNF147 0.63 0.0243 AA703115 NFAT5 1.22 0.0183     
2D kinases (4/4)    3D kinases (23/23)        
NM_001105 ACVR1 1.24 0.0206 NM_000020 ACVRL1 0.64 0.0383 NM_014002 IKKE 0.81 0.0493 
NM_001259 CDK6 0.79 0.0258 NM_004327 BCR 1.26 0.0102 NM_003010 MAP2K4 1.23 0.0109 
NM_004783 TAO1 1.24 0.0053 NM_004333 BRAF 1.28 0.0071 NM_005923 MAP3K5 0.79 0.0114 
NM_003565 ULK1 0.80 0.0293 NM_016508 CDKL3 1.17 0.0413 NM_002378 MATK 0.73 0.0410 
    NM_001274 CHEK1 0.74 0.0073 AA496964 NEK1 0.76 0.0475 
2D intracellular transport (4/4)    NM_001278 CHUK 0.71 0.0341 NM_005030 PLK 0.81 0.0046 
    NM_001328 CTBP1 1.34 0.0251 NM_005406 ROCK1 0.80 0.0333 
NM_016451 COPB 0.67 0.0461 NM_004734 DCAMKL1 1.32 0.0095 NM_003161 RPS6KB1 1.36 0.0272 
NM_007357 COG2 0.82 0.0162 NM_001982 ERBB3 1.21 0.0281 NM_005627 SGK 0.77 0.0403 
NM_006323 SEC24B 0.91 0.0056 NM_001465 FYB 0.63 0.0039 NM_004783 TAO1 1.31 0.0137 
NM_004766 COPB2 1.31 0.0074 NM_005734 HIPK3 1.31 0.0080 NM_003331 TYK2 0.77 0.0382 
    NM_000875 IGF1R 1.30 0.0233     
    3D chemokines (4/4)        
    NM_006419 SCYB13 1.38 0.0039 NM_000609 SDF1 1.37 0.0246 
    NM_001735 C5 1.23 0.0203 NM_000584 IL8 1.45 0.0157 
GenBank no.GeneAvgPGenBank no.GeneAvgPGenBank no.GeneAvgP
2D integral membrane (20/30)    3D integral membrane (62/62)        
NM_005050 ABCD4 1.17 0.0295 NM_000927 ABCB1 0.82 0.0398 AA450336 NRXN3 0.74 0.0210 
NM_014567 BCAR1 0.74 0.0222 NM_012089 ABCB10 0.61 0.0165 AA598582 OXA1L 0.62 0.0054 
NM_004054 C3AR1 1.20 0.0350 NM_005689 ABCB6 1.22 0.0029 NM_002561 P2RX5 0.74 0.0467 
NM_000722 CACNA2D1 0.62 0.0450 NM_019624 ABCB9 1.33 0.0175 NM_005446 P2RXL1 0.68 0.0446 
NM_013230 CD24 1.29 0.0093 NM_000683 ADRA2C 1.24 0.0321 NM_000919 PAM 1.24 0.0170 
NM_000760 CSF3R 1.17 0.0488 NM_001671 ASGR1 1.50 0.0384 AA663440 PLXNC1 0.83 0.0460 
NM_001381 DOK1 0.80 0.0062 NM_001681 ATP2A2 1.43 0.0335 NM_000952 PTAFR 1.22 0.0019 
NM_000118 ENG 0.64 0.0315 NM_004335 BST2 1.42 0.0106 NM_002838 PTPRC 0.72 0.0020 
NM_014211 GABRP 0.68 0.0164 NM_006137 CD7 1.31 0.0421 NM_002855 PVRL1 1.09 0.0137 
NM_004482 GALNT3 0.88 0.0219 NM_001782 CD72 1.24 0.0439 NM_001037 SCN1B 0.69 0.0030 
H07878 GPR19 1.23 0.0295 NM_004356 CD81 0.80 0.0182 NM_005072 SLC12A4 0.54 0.0230 
NM_000856 GUCY1A3 1.16 0.0171 NM_004233 CD83 0.75 0.0092 NM_015865 SLC14A1 0.77 0.0417 
H11482 IFNGR1 0.74 0.0029 NM_005745 DXS1357E 0.84 0.0156 NM_004207 SLC16A3 0.79 0.0054 
NM_000612 IGF2 1.13 0.0280 NM_001400 EDG1 1.69 0.0181 NM_004694 SLC16A6 1.23 0.0273 
NM_000598 IGFBP3 0.62 0.0092 NM_000604 FGFR1 1.25 0.0311 NM_005495 SLC17A4 1.17 0.0471 
NM_002195 INSL4 1.14 0.0360 NM_006338 GAC1 0.89 0.0454 NM_007256 SLC21A9 0.56 0.0133 
NM_012282 KCNE1L 1.15 0.0007 NM_000160 GCGR 0.72 0.0317 NM_000441 SLC26A4 0.77 0.0389 
NM_002333 LRP3 1.19 0.0085 NM_000832 GRIN1 1.50 0.0285 NM_001860 SLC31A2 0.83 0.0102 
AA496022 MFAP4 0.60 0.0469 NM_000840 GRM3 0.60 0.0367 NM_014437 SLC39A1 1.31 0.0039 
AA598582 OXA1L 0.52 0.0009 NM_005477 HCN4 0.75 0.0172 NM_002394 SLC3A2 1.30 0.0077 
NM_002571 PAEP 1.15 0.0092 AA464246 HLA-C 0.79 0.0248 AA487543 SORL1 0.83 0.0397 
NM_024411 PDYN 1.37 0.0492 NM_033554 HLA-DPA1 1.31 0.0498 NM_000544 TAP2 1.46 0.0217 
NM_004705 PRKRIR 0.80 0.0021 H11482 IFNGR1 0.59 0.0003 NM_006335 TIMM17A 1.28 0.0042 
NM_000452 SLC10A1 1.06 0.0296 NM_000877 IL1R1 0.80 0.0169 NM_005834 TIMM17B 0.79 0.0361 
NM_006749 SLC20A2 0.85 0.0041 NM_004633 IL1R2 0.63 0.0217 NM_003265 TLR3 0.71 0.0272 
NM_003127 SPTAN1 1.17 0.0450 NM_012276 ILT7 1.28 0.0260 NM_006827 TMP21 1.17 0.0109 
NM_003235 TG 1.19 0.0297 NM_002223 ITPR2 0.75 0.0100 NM_001192 TNFRSF17 1.28 0.0190 
NM_003243 TGFBR3 1.41 0.0218 NM_002233 KCNA4 0.73 0.0097 NM_003810 TNFSF10 1.15 0.0373 
NM_012471 TRPC5 0.61 0.0346 NM_002286 LAG3 0.73 0.0064 NM_006564 TYMSTR 0.45 0.0322 
NM_000371 TTR 0.87 0.0485 NM_015364 MD-2 0.68 0.0027 NM_004738 VAPB 1.35 0.0212 
    NM_000908 NPR3 1.17 0.0156 NM_021083 XK 1.23 0.0150 
2D transcription (23/26)    3D transcription (51/51)        
NM_004301 BAF53A 1.19 0.0280 NM_005180 BMI1 0.78 0.0355 NM_006186 NR4A2 1.18 0.0166 
NM_006317 BASP1 0.92 0.0007 NM_000092 COL4A4 0.68 0.0387 NM_003884 PCAF 0.62 0.0031 
NM_012116 CBLC 0.67 0.0249 H54686 DAB2 0.64 0.0163 NM_021128 POLR2L 0.88 0.0464 
NM_003651 CSDA 0.86 0.0318 NM_000399 EGR2 1.37 0.0486 NM_007244 PROL4 1.43 0.0057 
NM_004111 FEN1 0.82 0.0081 NM_006874 ELF2 0.74 0.0044 NM_002892 RBBP1 0.63 0.0012 
NM_001465 FYB 0.96 0.0002 AA010400 ETV4 1.36 0.0371 NM_002919 RFX3 1.22 0.0469 
NM_001515 GTF2H2 0.86 0.0261 NM_001987 ETV6 1.28 0.0386 NM_002938 RNF4 1.22 0.0414 
NM_005324 H3F3B 1.10 0.0482 AA181023 EVI1 1.31 0.0024 NM_002955 RREB1 1.36 0.0482 
NM_005336 HDLBP 0.50 0.0200 AI346582 EVX1 0.69 0.0477 NM_003071 SMARCA3 1.30 0.0208 
NM_014213 HOXD9 0.81 0.0059 NM_003862 FGF18 0.55 0.0374 NM_007017 SOX30 0.82 0.0371 
NM_016270 KLF2 1.32 0.0361 NM_000801 FKBP1A 0.87 0.0237 NM_004509 SP110 0.66 0.0173 
NM_002485 NBS1 1.22 0.0159 NM_005087 FXR1 0.74 0.0038 NM_012446 SSBP2 1.35 0.0139 
NM_004555 NFATC3 1.13 0.0302 NM_002095 GTF2E2 1.32 0.0018 NM_005636 SSX4 0.85 0.0450 
NM_002582 PARN 0.78 0.0034 NM_003483 HMGA2 1.05 0.4378 NM_007375 TARDBP 1.36 0.0089 
NM_002779 PSD 0.59 0.0062 NM_014213 HOXD9 0.58 0.0021 NM_007108 TCEB2 1.19 0.0476 
NM_002938 RNF4 1.17 0.0420 NM_002200 IRF5 1.22 0.0153 NM_003201 TFAM 1.15 0.0263 
NM_007252 RPF-1 0.60 0.0308 NM_005853 IRX5 0.59 0.0014 NM_005077 TLE1 1.18 0.0242 
NM_003079 SMARCE1 1.14 0.0106 NM_002228 JUN 1.30 0.0038 NM_005120 TNRC11 0.61 0.0064 
NM_003084 SNAPC3 0.91 0.0081 NM_014368 LHX6 0.74 0.0210 NM_001068 TOP2B 1.24 0.0498 
NM_005839 SRRM1 1.05 0.0047 NM_012321 LSM4 0.78 0.0046 NM_016936 UBN1 1.27 0.0347 
NM_003152 STAT5A 1.14 0.0462 NM_002397 MEF2C 0.84 0.0118 AI925821 USP22 0.76 0.0034 
NM_006284 TAF10 1.17 0.0133 NM_005933 MLL 0.58 0.0164 NM_000378 WT1 0.67 0.0150 
NM_005644 TAF12 1.24 0.0035 NM_004641 MLLT10 1.24 0.0292 NM_003443 ZNF151 0.62 0.0241 
NM_005481 TRAP95 0.59 0.0271 NM_002479 MYOG 0.73 0.0299 NM_016423 ZNF219 1.27 0.0198 
NM_004236 TRIP15 0.65 0.0394 NM_016170 NCX 1.36 0.0116 NM_005741 ZNF263 1.49 0.0055 
NM_005082 ZNF147 0.63 0.0243 AA703115 NFAT5 1.22 0.0183     
2D kinases (4/4)    3D kinases (23/23)        
NM_001105 ACVR1 1.24 0.0206 NM_000020 ACVRL1 0.64 0.0383 NM_014002 IKKE 0.81 0.0493 
NM_001259 CDK6 0.79 0.0258 NM_004327 BCR 1.26 0.0102 NM_003010 MAP2K4 1.23 0.0109 
NM_004783 TAO1 1.24 0.0053 NM_004333 BRAF 1.28 0.0071 NM_005923 MAP3K5 0.79 0.0114 
NM_003565 ULK1 0.80 0.0293 NM_016508 CDKL3 1.17 0.0413 NM_002378 MATK 0.73 0.0410 
    NM_001274 CHEK1 0.74 0.0073 AA496964 NEK1 0.76 0.0475 
2D intracellular transport (4/4)    NM_001278 CHUK 0.71 0.0341 NM_005030 PLK 0.81 0.0046 
    NM_001328 CTBP1 1.34 0.0251 NM_005406 ROCK1 0.80 0.0333 
NM_016451 COPB 0.67 0.0461 NM_004734 DCAMKL1 1.32 0.0095 NM_003161 RPS6KB1 1.36 0.0272 
NM_007357 COG2 0.82 0.0162 NM_001982 ERBB3 1.21 0.0281 NM_005627 SGK 0.77 0.0403 
NM_006323 SEC24B 0.91 0.0056 NM_001465 FYB 0.63 0.0039 NM_004783 TAO1 1.31 0.0137 
NM_004766 COPB2 1.31 0.0074 NM_005734 HIPK3 1.31 0.0080 NM_003331 TYK2 0.77 0.0382 
    NM_000875 IGF1R 1.30 0.0233     
    3D chemokines (4/4)        
    NM_006419 SCYB13 1.38 0.0039 NM_000609 SDF1 1.37 0.0246 
    NM_001735 C5 1.23 0.0203 NM_000584 IL8 1.45 0.0157 

NOTE: The titles (e.g., transport, kinase, etc.) describe the most common gene function in each class; the ratio of genes in the class that fit this functional definition is shown in parentheses. For each gene, the row of information contains the GenBank number (e.g., NM_005050), gene name (e.g., ABCD4), the mean value of the ratio of T4-2/S3-C expression over four microarray experiments (e.g., 0.15), and t-test penalized P value (P < 0.05).

Abbreviations: 2D, two-dimensional; 3D, three-dimensional; Avg, average.

Pure squamous carcinomas of the breast are rare, although mixed adenocarcinomas are diagnosed at a higher frequency (40). Some studies describe squamous metaplastic breast cancers as being extremely aggressive and they have been characterized to be at least as aggressive as grade 3 hormone receptor–negative adenocarcinomas (4043). Five-year survival for patients with pure squamous carcinomas of the breast seems to be only marginally worse than what is reported for all breast cancers, although these comparisons suffer from lack of a sufficient number of squamous carcinomas with follow up data (41). Squamous metaplasias of the breast share similarities with the aggressive basal-like subtype of carcinomas in clinical behavior as well as in the expression of a panel of markers (40, 44). HMT-3522 cells resemble the basal-like subtype of breast cancers at the molecular level by gene expression profiling (45). In addition, the HMT-3522 xenograft tumors are p63 and CK5/6 positive as well as estrogen receptor, progesterone receptor, and HER2 negative (Supplementary File 1F and G), similar to squamous metaplastic carcinomas (46). In addition to the rare pure squamous carcinomas of the breast, 39% of breast cancers in premenopausal African Americans and the majority of tumors in BRCA1 mutation carriers are estrogen receptor and progesterone receptor negative and have basal characteristics (47, 48). Understanding whether such similarities in marker expression translate to overlap in the mechanism of transition to invasiveness would require developing models that recapitulate these subtypes, as described for at least one subtype developed here.

The HMT-3522 cells display xenograft tumor histology consistent with the breast cancer type represented by their molecular makeup. This is a rare situation. Most human breast cancer cell line xenografts display an undifferentiated phenotype (observations of A.D.B.). To our knowledge, in addition to the cell line model described here, the MCF10A series of Ha-Ras transformed cell lines compose the only other model that recapitulates the histologic characteristics of human carcinomas (ductal carcinoma in situ and infiltrating ductal carcinoma) as xenograft tumors (49). To capture a more complete picture of the malignant progression in vivo, more complex microenvironments need to be created in three-dimensional cultures, and a number of laboratories including ours are engaged in this endeavor.

Comparison of the phenotypes of S3 cells in culture to their ability to form tumors in three different microenvironments in vivo show that these cells are particularly sensitive to their host microenvironment, producing complex phenotypes in vivo. For example, the S3-A cell line throws off ∼10% large colonies in the 3DlrBM, but these do not invade through laminin-rich basement membrane in Boyden chambers, possibly because the duration of the assay is too short (2 days). However, S3-A tumor frequency is higher than the other S3s in the skin injections, but interestingly not in the skin + Matrigel or fat pad injections, and the S3-A tumor volumes in the skin + Matrigel and fat pad injections are higher than the other S3s (Fig. 2). The other S3 cell lines also seem to be very sensitive to their particular host microenvironment, resulting in the breakdown of a one-to-one correlation between the progressive phenotypes observed in culture for S3-A, S3-B, S3-C, and the more complex phenotypes found in the xenografts. It is conceivable that, given the length of the 9- to 10-week time period, the host microenvironment within which the injected cells are in dynamic and reciprocal interactions with the stromal and humoral factors could allow for selection of the aggressive subpopulations, resulting in gain or loss of necessary functions to become malignant. In addition, results confirm that the cells poised to become malignant have not yet inactivated all pathways or gained all the new functions needed for a stable malignant phenotype.

What was found to be consistent across all assays done (invasion, three-dimensional polarity, CGH profile, tumorigenicity) is that all of the S3 cells displayed a phenotype distinct from both S1 and T4-2 cells in vivo as well as in the 3DlrBM assay. For tumorigenicity, this was supported by statistical analysis of tumor volume, as well as by histologic analysis showing that S3 tumors manifested more benign characteristics than the T4-2 tumors. In addition, S3 cells displayed more homogeneous phenotypes and a higher propensity to become invasive than the parent S2 cells when induced with T4-2 conditioned medium. Therefore, by these criteria, S3 cells have indeed progressed further than their parent S2 cells toward malignancy and are intermediate between the nontumorigenic S1 and malignant T4-2s.

Here we have described this model as one of transition from preinvasive to invasive phenotype to mimic the multistep carcinogenesis hypothesis of breast cancer (50, 51). However, the emerging cancer stem/progenitor cell hypothesis also needs to be considered. Sontag and Axelrod (1) have proposed four separate models to describe how atypical hyperplasia, ductal carcinoma in situ, invasive ductal carcinoma, and metastasis may relate to each other. These are the linear, nonlinear, branched, and parallel pathways. Unlike the other three hypotheses, the parallel pathway does not assume that in situ carcinoma develops into invasive carcinoma, but rather that in situ and invasive carcinomas can arise from the same progenitor. In our cell lines, S2 gave rise to both the preinvasive S3-C and the invasive T4-2 cells via different steps of manipulation in culture and in mice, respectively (Fig. 1A). Therefore, it is tempting to speculate that the model described here could be placed as supporting the parallel progression hypothesis. Regardless of which mathematical model we consider, what we have developed are human breast epithelial cells with preinvasive and invasive characteristics, which recapitulate some aspects of preinvasive and invasive carcinomas with basal and squamous histologic and molecular phenotypes.

Analysis of this malignant transition in three-dimensional cultures allowed us to identify candidate genes as potential therapeutic targets. For example, of the MMPs we identified as having a function in invasion, MMP9, MMP13, and, recently, MMP17 have been shown to be associated with, and increased in, breast cancer progression (5254). Although broad-spectrum inhibitors of MMPs have failed in clinical trials, targeting specific genes for certain subtypes of cancers may eliminate nonspecific effects, thereby producing more desirable clinical outcomes (55). In addition to the list of genes differentially expressed between S3-C and T4-2 (Table 1; Supplementary File 8), this model system is conducive to functional siRNA screens probing either gain of invasion function in S3-C or loss of invasion of T4-2 to find new genes involved in invasion. Using the same model, we recently discovered a new pathway of invasion regulated by polo-like kinase I (23). In general, the results described here provide a proof-of-principle for the development of spontaneous transition models for additional subtypes of breast cancers, with potential utility in discovering other mechanisms of invasion and other potential targets for therapy.

Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

Current address for A. Rizki: Department of Radiation Oncology, Virginia Commonwealth University, Richmond, VA 23298; Current address for C.A. Myers: Department of Pathology and Immunology, Washington University, St. Loius, MO 63110; Current address for L.R. Grate: Molecular, Cell, and Developmental Biology Department, University of California at Santa Cruz, Santa Cruz, CA 95064.

Grant support: U.S. Department of Energy, Office of Biological and Environmental Research grant DEAC0376SF00098 (M.J. Bissell); NIH grant CA64786 (M.J. Bissell and O.W. Petersen); Department of Defense (DOD) Breast Cancer Research Program (BCRP) Innovator Award DAMD170210438 (M.J. Bissell); California Breast Cancer Research Program (CBCRP) Postdoctoral Fellowship Award 8FB0184 (A. Rizki); DOD BCRP Innovative, Developmental, and Exploratory Award DAMD170110368, NIH grant CA78731, and American Cancer Society New Investigator Internal Award IRG7800223 (V.M. Weaver); DOD BCRP Predoctoral Fellowship Award DAMD170310421 (G.I. Rozenberg); NIH grant CA88858 (J.D. Mott); NIH grants CA80067 and CA098131 (R.A. Jensen); CBCRP grant 8PB0171 and NIH grants P01 AG017242 and U01 ES011044 (I.S. Mian); DOD BCRP Predoctoral Fellowship Award BC021231 (J.L. Bascom); and NIH grant CA58207 (K. Chin and J.W. Gray). NIH grant CA50519 and DOD BCRP grant DAMD170210439 (D.J. Chen). M.J. Bissell is a recipient of a distinguished fellowship from the U.S. Department of Energy, Office of Biological and Environmental Research.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

We thank Eva Lee, Hui Zhang, Genee Lee, and Shraddha Ravani (Lawrence Berkeley National Laboratory) and Travis Mullan (Virginia Commonwealth University) for technical assistance and/or advice.

1
Sontag L, Axelrod DE. Evaluation of pathways for progression of heterogeneous breast tumors.
J Theor Biol
2005
;
232
:
179
–89.
2
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
:
396
–402.
3
Etzell JE, Devries S, Chew K, et al. Loss of chromosome 16q in lobular carcinoma in situ.
Hum Pathol
2001
;
32
:
292
–6.
4
Gong G, DeVries S, Chew KL, Cha I, Ljung BM, Waldman FM. Genetic changes in paired atypical and usual ductal hyperplasia of the breast by comparative genomic hybridization.
Clin Cancer Res
2001
;
7
:
2410
–4.
5
Isola J, Chu L, DeVries S, et al. Genetic alterations in ERBB2-amplified breast carcinomas.
Clin Cancer Res
1999
;
5
:
4140
–5.
6
Isola JJ, Kallioniemi OP, Chu LW, et al. Genetic aberrations detected by comparative genomic hybridization predict outcome in node-negative breast cancer.
Am J Pathol
1995
;
147
:
905
–11.
7
Loveday RL, Greenman J, Simcox DL, et al. Genetic changes in breast cancer detected by comparative genomic hybridisation.
Int J Cancer
2000
;
86
:
494
–500.
8
Ma XJ, Salunga R, Tuggle JT, et al. Gene expression profiles of human breast cancer progression.
Proc Natl Acad Sci U S A
2003
;
100
:
5974
–9.
9
Nishizaki T, DeVries S, Chew K, et al. Genetic alterations in primary breast cancers and their metastases: direct comparison using modified comparative genomic hybridization.
Genes Chromosomes Cancer
1997
;
19
:
267
–72.
10
Nishizaki T, Chew K, Chu L, et al. Genetic alterations in lobular breast cancer by comparative genomic hybridization.
Int J Cancer
1997
;
74
:
513
–7.
11
Polyak K. Molecular alterations in ductal carcinoma in situ of the breast.
Curr Opin Oncol
2002
;
14
:
92
–6.
12
Porter DA, Krop IE, Nasser S, et al. A SAGE (serial analysis of gene expression) view of breast tumor progression.
Cancer Res
2001
;
61
:
5697
–702.
13
Sgroi DC, Teng S, Robinson G, LeVangie R, Hudson JR, Jr., Elkahloun AG. In vivo gene expression profile analysis of human breast cancer progression.
Cancer Res
1999
;
59
:
5656
–61.
14
Thor AD, Eng C, Devries S, et al. Invasive micropapillary carcinoma of the breast is associated with chromosome 8 abnormalities detected by comparative genomic hybridization.
Hum Pathol
2002
;
33
:
628
–31.
15
Tirkkonen M, Tanner M, Karhu R, Kallioniemi A, Isola J, Kallioniemi OP. Molecular cytogenetics of primary breast cancer by CGH.
Genes Chromosomes Cancer
1998
;
21
:
177
–84.
16
Waldman FM, DeVries S, Chew KL, Moore DH II, Kerlikowske K, Ljung BM. Chromosomal alterations in ductal carcinomas in situ and their in situ recurrences.
J Natl Cancer Inst
2000
;
92
:
313
–20.
17
Waldman FM, Hwang ES, Etzell J, et al. Genomic alterations in tubular breast carcinomas.
Hum Pathol
2001
;
32
:
222
–6.
18
Neve RM, Chin K, Fridlyand J, et al. A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes.
Cancer Cell
2006
;
10
:
515
–27.
19
Briand P, Petersen OW, Van Deurs B. A new diploid nontumorigenic human breast epithelial cell line isolated and propagated in chemically defined medium.
In Vitro Cell Dev Biol
1987
;
23
:
181
–8.
20
Briand P, Nielsen KV, Madsen MW, Petersen OW. Trisomy 7p and malignant transformation of human breast epithelial cells following epidermal growth factor withdrawal.
Cancer Res
1996
;
56
:
2039
–44.
21
Petersen OW, Ronnov-Jessen L, Howlett AR, Bissell MJ. Interaction with basement membrane serves to rapidly distinguish growth and differentiation pattern of normal and malignant human breast epithelial cells.
Proc Natl Acad Sci U S A
1992
;
89
:
9064
–8.
22
Weaver VM, Petersen OW, Wang F, et al. Reversion of the malignant phenotype of human breast cells in three-dimensional culture and in vivo by integrin blocking antibodies.
J Cell Biol
1997
;
137
:
231
–45.
23
Rizki A, Mott JD, Bissell MJ. Polo-like kinase I is involved in invasion through extracellular matrix.
Cancer Res
2007
;
67
:
11106
–10.
24
Pinkel D, Segraves R, Sudar D, et al. High resolution analysis of DNA copy number variation using comparative genomic hybridization to microarrays.
Nat Genet
1998
;
20
:
207
–11.
25
Herron GS, Werb Z, Dwyer K, Banda MJ. Secretion of metalloproteinases by stimulated capillary endothelial cells. I. Production of procollagenase and prostromelysin exceeds expression of proteolytic activity.
J Biol Chem
1986
;
261
:
2810
–3.
26
Lochter A, Srebrow A, Sympson CJ, Terracio N, Werb Z, Bissell MJ. Misregulation of stromelysin-1 expression in mouse mammary tumor cells accompanies acquisition of stromelysin-1-dependent invasive properties.
J Biol Chem
1997
;
272
:
5007
–15.
27
Taylor MA, Cote RJ. Immunomicroscopy: a diagnostic tool for the surgical pathologist. Philadelphia: W.B. Saunders; 1994. p. 300–78.
28
Liu H, Radisky DC, Wang F, Bissell MJ. Polarity and proliferation are controlled by distinct signaling pathways downstream of PI3-kinase in breast epithelial tumor cells.
J Cell Biol
2004
;
164
:
603
–12.
29
Muthuswamy SK, Li D, Lelievre S, Bissell MJ, Brugge JS. ErbB2, but not ErbB1, reinitiates proliferation and induces luminal repopulation in epithelial acini.
Nat Cell Biol
2001
;
3
:
785
–92.
30
Weaver VM, Lelievre S, Lakins JN, et al. β4 integrin-dependent formation of polarized three-dimensional architecture confers resistance to apoptosis in normal and malignant mammary epithelium.
Cancer Cell
2002
;
2
:
205
–16.
31
Lelievre SA, Weaver VM, Nickerson JA, et al. Tissue phenotype depends on reciprocal interactions between the extracellular matrix and the structural organization of the nucleus.
Proc Natl Acad Sci U S A
1998
;
95
:
14711
–6.
32
Muschler J, Levy D, Boudreau R, Henry M, Campbell K, Bissell MJ. A role for dystroglycan in epithelial polarization: loss of function in breast tumor cells.
Cancer Res
2002
;
62
:
7102
–9.
33
Wang F, Weaver VM, Petersen OW, et al. Reciprocal interactions between β1-integrin and epidermal growth factor receptor in three-dimensional basement membrane breast cultures: a different perspective in epithelial biology.
Proc Natl Acad Sci U S A
1998
;
95
:
14821
–6.
34
Semeiks JR, Rizki A, Bissell MJ, Mian IS. Ensemble attribute profile clustering: discovering and characterizing groups of genes with similar patterns of biological features.
BMC Bioinformatics
2006
;
7
:
147
.
35
Muller A, Homey B, Soto H, et al. Involvement of chemokine receptors in breast cancer metastasis.
Nature
2001
;
410
:
50
–6.
36
Allinen M, Beroukhim R, Cai L, et al. Molecular characterization of the tumor microenvironment in breast cancer.
Cancer Cell
2004
;
6
:
17
–32.
37
d'Ortho MP, Will H, Atkinson S, et al. Membrane-type matrix metalloproteinases 1 and 2 exhibit broad-spectrum proteolytic capacities comparable to many matrix metalloproteinases.
Eur J Biochem
1997
;
250
:
751
–7.
38
Grobelny D, Poncz L, Galardy RE. Inhibition of human skin fibroblast collagenase, thermolysin, and Pseudomonas aeruginosa elastase by peptide hydroxamic acids.
Biochemistry
1992
;
31
:
7152
–4.
39
Leaf C. Why we're losing the war on cancer (and how to win it).
Fortune
2004
;
149
:
76
–82, 4–6, 8 passim.
40
Hennessy BT, Krishnamurthy S, Giordano S, et al. Squamous cell carcinoma of the breast.
J Clin Oncol
2005
;
23
:
7827
–35.
41
Aparicio I, Martinez A, Hernandez G, Hardisson D, De Santiago J. Squamous cell carcinoma of the breast. Eur J Obstet Gynecol Reprod Biol 2007; online ahead of print (dx.doi.org/10.1016/j.ejogrb.2007.03.021).
42
Tse GM, Tan PH, Putti TC, Lui PC, Chaiwun B, Law BK. Metaplastic carcinoma of the breast: a clinico-pathological review.
J Clin Pathol
2006
;
59
:
1079
–83.
43
Behranwala KA, Nasiri N, Abdullah N, Trott PA, Gui GP. Squamous cell carcinoma of the breast: clinico-pathologic implications and outcome.
Eur J Surg Oncol
2003
;
29
:
386
–9.
44
Reis-Filho JS, Milanezi F, Steele D, et al. Metaplastic breast carcinomas are basal-like tumours.
Histopathology
2006
;
49
:
10
–21.
45
Kenny PA, Lee GY, Myers CA, et al. The morphologies of breast cancer cell lines in three-dimensional assays correlate with their profiles of gene expression.
Mol Oncol
2007
;
1
:
84
–96.
46
Tse GM, Tan PH, Chaiwun B, et al. p63 is useful in the diagnosis of mammary metaplastic carcinomas.
Pathology
2006
;
38
:
16
–20.
47
Carey LA, Perou CM, Livasy CA, et al. Race, breast cancer subtypes, and survival in the Carolina Breast Cancer Study.
JAMA
2006
;
295
:
2492
–502.
48
Honrado E, Benitez J, Palacios J. Histopathology of BRCA1- and BRCA2-associated breast cancer.
Crit Rev Oncol Hematol
2006
;
59
:
27
–39.
49
Santner SJ, Dawson PJ, Tait L, et al. Malignant MCF10CA1 cell lines derived from premalignant human breast epithelial MCF10AT cells.
Breast Cancer Res Treat
2001
;
65
:
101
–10.
50
Allred DC, Mohsin SK. Biological features of premalignant disease in the human breast.
J Mammary Gland Biol Neoplasia
2000
;
5
:
351
–64.
51
Burstein HJ, Polyak K, Wong JS, Lester SC, Kaelin CM. Ductal carcinoma in situ of the breast.
N Engl J Med
2004
;
350
:
1430
–41.
52
Nielsen BS, Rank F, Lopez JM, et al. Collagenase-3 expression in breast myofibroblasts as a molecular marker of transition of ductal carcinoma in situ lesions to invasive ductal carcinomas.
Cancer Res
2001
;
61
:
7091
–100.
53
Rha SY, Yang WI, Kim JH, et al. Different expression patterns of MMP-2 and MMP-9 in breast cancer.
Oncol Rep
1998
;
5
:
875
–9.
54
Chabottaux V, Sounni NE, Pennington CJ, et al. Membrane-type 4 matrix metalloproteinase promotes breast cancer growth and metastases.
Cancer Res
2006
;
66
:
5165
–72.
55
Fingleton B. Matrix metalloproteinases: roles in cancer and metastasis.
Front Biosci
2006
;
11
:
479
–91.