Skip to main content
Erschienen in: Breast Cancer Research and Treatment 1/2018

Open Access 09.09.2017 | Brief Report

Multidimensional phenotyping of breast cancer cell lines to guide preclinical research

verfasst von: Jodi M. Saunus, Chanel E. Smart, Jamie R. Kutasovic, Rebecca L. Johnston, Priyakshi Kalita-de Croft, Mariska Miranda, Esdy N. Rozali, Ana Cristina Vargas, Lynne E. Reid, Eva Lorsy, Sibylle Cocciardi, Tatjana Seidens, Amy E. McCart Reed, Andrew J. Dalley, Leesa F. Wockner, Julie Johnson, Debina Sarkar, Marjan E. Askarian-Amiri, Peter T. Simpson, Kum Kum Khanna, Georgia Chenevix-Trench, Fares Al-Ejeh, Sunil R. Lakhani

Erschienen in: Breast Cancer Research and Treatment | Ausgabe 1/2018

Abstract

Purpose

Cell lines are extremely useful tools in breast cancer research. Their key benefits include a high degree of control over experimental variables and reproducibility. However, the advantages must be balanced against the limitations of modelling such a complex disease in vitro. Informed selection of cell line(s) for a given experiment now requires essential knowledge about molecular and phenotypic context in the culture dish.

Methods

We performed multidimensional profiling of 36 widely used breast cancer cell lines that were cultured under standardised conditions. Flow cytometry and digital immunohistochemistry were used to compare the expression of 14 classical breast cancer biomarkers related to intrinsic molecular profiles and differentiation states: EpCAM, CD24, CD49f, CD44, ER, AR, HER2, EGFR, E-cadherin, p53, vimentin, and cytokeratins 5, 8/18 and 19.

Results

This cell-by-cell analysis revealed striking heterogeneity within cultures of individual lines that would be otherwise obscured by analysing cell homogenates, particularly amongst the triple-negative lines. High levels of p53 protein, but not RNA, were associated with somatic mutations (p = 0.008). We also identified new subgroups using the nanoString PanCancer Pathways panel (730 transcripts representing 13 canonical cancer pathways). Unsupervised clustering identified five groups: luminal/HER2, immortalised (‘normal’), claudin-low and two basal clusters, distinguished mostly by baseline expression of TGF-beta and PI3-kinase pathway genes.

Conclusion

These features are compared with other published genotype and phenotype information in a user-friendly reference table to help guide selection of the most appropriate models for in vitro and in vivo studies, and as a framework for classifying new patient-derived cancer cell lines and xenografts.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1007/​s10549-017-4496-x) contains supplementary material, which is available to authorized users.
Jodi M. Saunus, Chanel E. Smart, Fares Al-Ejeh, Sunil R. Lakhani have contributed equally to this work.

Introduction

Heterogeneity pervades the molecular and cellular landscapes of breast tumours at virtually every level; a consequence of transformation-associated changes coalescing with epithelial lineage commitment programs inherited from the tumour cell(s)-of-origin. Patient-derived cell lines (PDCL) and xenografts (PDX) can model this complexity fairly accurately and are becoming more prevalent in preclinical research. While PDCL/PDX are in many ways ‘better’ models of breast cancer, established cell lines remain extremely useful in earlier-phase studies because they are versatile, reliable, reproducible, amenable to genetic manipulation and conducive to hypothesis testing under controlled experimental conditions.
Over the past decade, several groups have mapped the genotypes, phenotypes, in vitro and in vivo behaviour of cell line models in order to define their relevance to human disease, provide benchmarks for assessing the integrity of cell line stocks around the world and to help contextualise experimental data. The approaches have included exome sequencing, copy-number analysis, whole-transcriptome and targeted analysis of biomarkers relevant to breast cancer or mammary gland development [19]. These studies proposed various frameworks in which each line could be categorized. For example, expression array studies identified three major intrinsic subtypes (Luminal, Basal-A and Basal-B/claudin-low) [1, 4, 7], and 4–6 triple-negative (TN) cell line subgroups [3, 10]. Expression of breast epithelial lineage commitment markers can also discriminate phenotypic and functional classes, such as CD24 and CD44 (associated with differentiation and tumourigenicity); and EpCAM and CD49f, which exhibit restricted expression in the two major functional compartments (luminal or myoepithelial) of the mammary gland [2, 11, 12].
Here, we provide an update on breast cancer cell line classification,1 incorporating single-cell protein profiling, targeted digital expression analysis and functional pathway mapping. In performing this analysis, we investigated which approaches could be applied to classify new cell lines, using a previously undescribed line derived in our lab from a metastatic bone deposit (P7731). We also applied the data to quantify the relationship between TP53 mutation status, RNA and protein expression. The data are interpreted in a context that recognises heterogeneity and the trending, anti-reductionist view of complex biological systems in cancer research. The cell lines used in this study were genetically authenticated, certified to be free of mycoplasma, and cultured under standardized conditions.

Materials and methods

Cell culture

Cell lines were sourced from the ATCC, or from the collection developed and authenticated by the Brisbane Breast Bank from 2010 to 2012 ([13] and Table 1). P7731 was derived in-house from a metastatic bone deposit from a breast cancer patient enrolled in the Victorian Tissue Bank (manuscript under review), who provided informed written consent to use donated tissue for research purposes. We acknowledge the existence of genotypic and phenotypic variants of some cell lines, and emphasise that the data presented here reflect our specific collection. Nonetheless, it broadly represents cell line collections used in standard breast cancer research laboratories around the world, and multiple quality control measures were in place to ensure its integrity. The QC process was as follows: (1) At baseline (earliest passage), the morphology of each line was checked for consistency with published information, cultures were photographed for future comparison, and expanded to generate cryopreserved ‘master stocks’. (2) Mycoplasma testing was performed using the Mycoalert® kit (Lonza) according to the manufacturer’s instructions, using media from cells cultured without antibiotic for 2 weeks. All master stocks used in this study were mycoplasma-negative. (3) DNA was extracted for STR-profiling and targeted mutation analysis (see below). (4) Thawed master stocks were cultured for at least 48 h to 70% confluence prior to each experiment, and used within five passages of the authenticated baseline (Table 1), with ongoing re-analysis of morphology and growth rates, and regular re-testing to confirm negative mycoplasma status.
Table 1
Cell lines used in this study
https://static-content.springer.com/image/art%3A10.1007%2Fs10549-017-4496-x/MediaObjects/10549_2017_4496_Figa_HTML.gif
Culture media were prepared according to ATCC recommendations ([14] and Table 1), and cells were maintained at 37 °C in a humidified atmosphere with 5% CO2. Base media and supplements were purchased from Thermofisher Scientific (TFS) or Sigma-Aldrich (SA): DMEM (TFS catalogue no. 11995065), RPMI (TFS-11875093), McCoy’s 5A modified medium (TFS-16600082), DMEM/F12 (TFS-11330032), Ham’s F12 (TFS-11765054), foetal bovine serum (FBS; TFS-16000044), horse serum (TFS-16050130), bovine pituitary extract (TFS-13028014), Insulin (SA-I5500), Hydrocortisone (SA-H0888), cholera toxin (SA-C8052), epidermal growth factor (SA-E9644), transferrin (SA-T8158). Antibiotic/antimycotic was routinely included in culture media (TFS-15240062). Master stocks were cryopreserved in a solution comprising 50% of the regular growth medium, 10% dimethyl sulfoxide (DMSO) and 40% FBS.

Short tandem repeat (STR)-profiling and targeted mutation analysis

DNA extraction from cell pellets was performed using the QIAamp DNA Mini kit (QIAGEN) and profiling was performed using the Cell ID™ System (Promega). The assay allows co-amplification and three-color detection of 9 STR loci (D21S11, TH01, TPOX, vWA, CSF1PO, D16S539, D7S820, D13S317 and D5S818) and Amelogenin for gender identification and collectively provides a genetic profile with a random match probability of 1 in 2.92 x 109. The amplified and fluorescently-tagged loci were analysed by capillary electrophoresis on an ABI Prism 3100 Genetic Analyser (Applied Biosystems) at QIMR Berghofer (Table-S1). Of the 35 lines tested, 25 matched profiles published by either the ATCC or other laboratories, seven had not been previously published, and three gave partial matches (HBL100, the subject of an authenticity debate (below); Hs578T, which exhibited loss-of-heterozygosity (LOH) at 2/10 STR loci; and MDAMB435, LOH at one locus and different number of STRs at another. The three partially matched lines are triple-negative and may exhibit heightened genomic instability [4].
Targeted mutation analysis of PIK3CA, HRAS, KRAS and BRAF was performed using the Oncocarta Assay (v1.0; Sequenom [15]). Variants were validated by High Resolution Melt analysis, iPLEX (using different PCR and extension primers), repeat OncoCarta analysis and/or Sanger sequencing. Lines were also Sanger-sequenced to identify mutations in EGFR (NC_000007.1; Ex 5–11/23–28) and TP53 (NC_000017.9; exons 4–10 as per International Agency for Research on Cancer (IARC) recommendations [16, 17]). Comparing our findings to cell line mutation data repositories [18, 19] revealed several differences: four instances where we could only detect the wild-type allele but a mutation had been reported previously, possibly indicating LOH at these loci (BT474 (PIK3CA, EGFR), MDAMB231 (BRAF), MDAMB436 (BRAF) and ZR751 (HRAS); MDAMB436, where a BRAF deletion was found by the CCLE but not COSMIC or our study; and two with discordant findings between the CCLE and COSMIC (BRAF mutations in MDAMB361 and MDAMB415). These findings highlight the existence of clonal and phenotypic drift in different cell line stocks around the world (Table-S1).
Based on these analyses we can also make the following comments about cell line authenticity:
  • Previous reports have suggested that KPL1 and MCF7 are the same cell line [20]. Indeed, our stocks had identical STR profiles, shared the E545 K PIK3CA mutation (Table-S1) and had very similar transcriptional profiles (Fig. 3/S3), though there were some phenotypic differences noted with single-cell analysis (Figs. 1,2), consistent with KPL1 being a clonal derivative of MCF7.
  • We did not detect a Y chromosome in HBL100 (Table-S1), which has been the basis for its exclusion from cell line repositories and cell line panel studies.
  • MDAMB435 has also been the subject of an historic authenticity debate, with conflicting reports that it could either be the M14 melanoma cell line or a breast-derived line that exhibits lineage infidelity, including melanocytic features [2123]. This line was an outlier in multiple experiments in our study, though others have performed more comprehensive analyses [24]. The most recent consensus is that the line was indeed contaminated with melanoma cells and should not be used as a breast cancer model [25].

Molecular classification

Transcriptome subtype (Tx-ST) was assigned based on previous gene expression array studies, summarised in Table-S2. Six lines in the cohort had not been previously classified, and so we applied the surrogate cytokeratin expression method of Hollestelle et al. [1] (Fig. 2 and Table-S2). Briefly, the criteria are: Luminal phenotype: CK8/18+ and/or CK19+, CK5-; Basal-like luminal phenotype (L+B): CK8/18+ and/or CK19+, CK5+; null (claudin-low): CK8/18 low, CK19-, CK5-; basal: CK8/18 low, CK19-, CK5+. Triple-negative (TN) cell lines were sub-classified using the TNBCtype tool [3] (Table-S2).

Multiplexed flow cytometry analysis (FC)

Cells were prepared from three separate cultures as previously described [14]. Single cell suspensions were stained with SYTOX BLUE (Molecular Probes), CD49f-PE-Cy5, EpCAM-FITC, CD44-APC H7 and CD24-PE (Becton–Dickinson (BD); Table 2), or isotype and fluorophore-matched control antibodies. Raw fluorescence data were collected on either FACSAria-I or LSRFortessa flow cytometers (BD) using FACSDiva acquisition software (v6.1.3; BD). Particles and dead cells were excluded based on low light scatter and SYTOX BLUE positivity. Doublet discrimination was also performed by gating out cells with disproportionate forward scatter height and area. Positivity thresholds were determined based on the signal from isotype controls for each marker and cell line combination (Fig-S1). At least 1x104 events representing live, single cells were collected for each sample. Fluorescence compensation was performed on each occasion, then retrospectively checked and modified if necessary using FCS Express software (v6.0; DeNovo). Population frequencies were determined for individual and combined parameters from an average of three cultures (consecutive passages). The proportions of luminal-1, luminal-2, basal and mesenchymal subpopulations were determined as described [2].
Table 2
Antibodies used in this study
Antigen
Full name/alias
Function
Rationale for use in this study
Method
Supplier (clone)
Antigen retrieval
Dilution
AR
Androgen receptor
ER-independent hormone signalling
Associated with oestrogen-independent hormone signalling
dIHC
Dako (AR441)
Citrate
1:100
CD24
Heat-stable antigen (HSA)
Cell adhesion molecule
Associated with luminal differentiation
FC
BD (ML5; PE conjugate)
na
1:100
CD44
Receptor for hyaluronic acid (& other ligands)
Cell adhesion molecule
Basal marker in normal breast; expression may be enriched in cancer cells with progenitor activity
FC
BD (C26; APC-H7 conjugate)
na
1:100
CD49f
a6 integrin
Laminin receptor subunit
Basal marker in normal breast; expressed at the myoepithelial-basement membrane interface
FC
BD (GoH3; PE-Cy5 conjugate)
na
1:100
CK19
Cytokeratin-19
Intermediate filament
Associated with luminal differentiation
dIHC
Dako (BA17)
Citrate
1:40
CK5
Cytokeratin-5
Intermediate filament
Associated with basal differentiation; expressed in luminal progenitor cells
dIHC
Novocastra (XM26)
Citrate
1:100
CK8/18
Cytokeratins 8 & 18
Intermediate filament
Associated with luminal differentiation
dIHC
Novocastra (5D3)
Citrate
1:100
E-cad
Epithelial cadherin, CD324
Cell–cell adhesion, component of the adherens junction
Marker of terminally differentiated luminal epithelia
dIHC
Novocastra (36B5)
Citrate
1:100
ER
Oestrogen receptor
Hormone signalling
Expressed by luminal epithelia; highly prognostic and predictive of hormone therapy response in BC
dIHC
Dako (1D5)
EDTA
1:100
HER2
Human epidermal growth factor receptor-2
Growth factor signalling, overexpression marks gene amplification
Amplified and overexpressed in some BCs; highly prognostic and predictive of response to targeted therapy
dIHC
Dako
HercepTest
 
p53
Tumour protein 53
Key tumour suppressor
Frequently mutated BC driver; mutation status a key determinant of genetic background in BC models
dIHC
Dako (D07)
Citrate
1:300
vim
Vimentin
Intermediate filament
Type III intermediate filament expressed in mesenchymal cells
dIHC
Dako (V9)
Citrate
1:500
dIHC digital immunohistochemistry, FC flow cytometry

Immunohistochemistry (IHC)

Cultures were washed twice in PBS and then harvested with cell scrapers after soaking in versene (EDTA) for 5 min, so as not to remove trypsin-sensitive cell surface proteins. An equal volume of growth media was added before centrifuging, then washed twice in PBS, resuspended in a minimal volume of PBS and fixed in 5 mL 10% neutral buffered formalin for 30 min at room temperature. After centrifuging (200 × g), samples were washed twice in PBS before processed for paraffin embedding. A cell line TMA was then constructed from 0.6 mm cores. 4 μm TMA sections were cut for IHC. For each of the primary antibodies used, Table 2 lists the working dilution and one of the following antigen retrieval methods: heat retrieval in a decloaking chamber (Biocare Medical) with either: (EDTA) 0.001 M Tris-ethylenediaminetetraacetic acid pH 8.8, at 105 °C for 15 min; (citrate) 0.01 M citric acid buffer pH 6.0, at 125 °C for 5 min; (Chymo) 0.1% Chymotrypsin in 0.01 M CaCl2 + 0.05 M Tris buffer, pH 7.8 at 37 °C for 10 min. The Dako EnVision + (Dakocytomation) and Vectastain® Universal ABC kit (Vector laboratories) were used for detection according to the manufacturers’ instructions. The Dako HercepTest™ kit was used for HER2 staining. Sections were reviewed and described by a qualified pathologist (ACV). We used at least two separate cultures (successive passages) for each cell line.

Digital IHC analysis

Stained TMA sections were scanned on an Aperio ScanScope T2 high-resolution slide scanner (20 x magnification). Digital images were imported into Definiens Tissue Studio 3.0 for automated analysis. Cells were segmented based on nuclear haematoxylin-defined staining and antigen specific DAB-defined chromatic staining was assessed per cell either localised to nucleus for nuclear stains (ER, p53, AR) or in the cytoplasm by dilating the area segmented by the nucleus. The intensity of staining for each antigen was divided into negative, low, moderate and strong categories was verified by visual assessment of a small panel of positive and negative controls. These thresholds were then used to calculate the frequency of cells in each category across all cores in the TMAs. An overall histological score for each antigen was calculated using an algorithm that considers the frequency of cells assigned to each intensity category. Histological score = (1 x  % weak + 2 x  % moderate + 3 x  % strong). TMA sections had to contain at least 100 cells for inclusion. Where multiple cores were analysed, an average was taken. Unsupervised hierarchical clustering was performed on the eight antigens (ER, E-cadherin, CK5, CK8/18, CK19, Vimentin, EGFR and HER2) using Euclidean distance and complete linkage method for clustering via the R Project for Statistical Computing [26].

Nanostring® targeted pancancer pathways assay

RNA was extracted from each line using the RNeasy kit (QIAGEN), then quantified using Nanodrop. We used the PanCancer Pathways Panel, a set of barcoded probes for 730 genes and 40 housekeeping genes (nanoString®). Assays and analysis were performed according to the manufacturer’s instructions using 50 ng of total RNA [9]. Data were collected using the nCounter® Dx Digital Analyzer (QIMR Berghofer) and processed using nSolver Software (v3.0 NanoString). Unsupervised clustering based on the expression of all the genes in the PanCancer Pathways panel was performed in R [26], using rank correlation as the distance metric and centroid linkage, with genes ordered by cluster tightness. Significance thresholds (p) used to assign functional enrichment were 0.01 (genes) and 0.05 (samples).

Results and discussion

Single cell biomarker profiling of key breast cancer biomarkers

Established cell lines are perpetual and (relatively) genetically and phenotypically stable, therefore a proportion of the culture population must self-renew. Keller [2] proposed that epithelial lineage differentiation hierarchies inherited from the tumour-of-origin might be maintained in cell cultures in some form, despite in vitro adherent selection and genetic drift over time. Analysis of 20 cell lines for expression of cell surface markers that discriminate luminal and myoepithelial/basal compartments in the mammary gland (EpCAM, CD49f, CD24, CD44), showed that each line exhibited a restricted version of the epithelial lineage hierarchy in normal breast, consistent with intrinsic subtype. To investigate how widespread these categories are, we performed multiplex flow cytometry analysis on 34 breast cell lines (including 26 different from the Keller study; Table 1). We assessed at least three independent cultures to encompass normal variability/plasticity (Fig. 1 and Fig-S1). Consistent with the previous findings [2], luminal lines were predominantly EpCAM+/CD49f+/CD24+, and only a few retained a ‘Luminal-1′ subpopulation (EpCAM+/CD49f/CD24+); most notably SKBr3, which exhibits pronounced epithelioid morphology in vitro. EpCAM/CD24-negativity featured prominently in lines with mesenchymal morphology. EpCAM+/CD49f+/CD24 subpopulations were most common in basal lines, though not exclusively. Basal and claudin-low lines exhibited the most heterogeneity.
We screened the panel for expression of an additional nine biomarkers using digital (d)IHC analysis of fixed cell pellets, calculating the frequencies of cells exhibiting no, low, moderate and strong staining as a readout of heterogeneity, and integrated these data to calculate the overall Histo(H)-score for each marker in each line. (Fig. 2). Unsupervised clustering of the H-scores produced three major groups that conformed with intrinsic subtype 85% of the time (Fig. 2B). The discordant cases included two immortalised ‘normal’ lines (184A1, MCF10A), P7731, PMC42ET and HCC1143 (expression subtype for the latter two was ascribed using cytokeratin surrogate markers [1]; Table-S2). Statistically, vimentin, EGFR and ER best discriminate these groups (Fig-S2A), but a minimal panel of vimentin, EGFR, CK19 and HER2 would be most useful for classifying individual lines (Fig. 2C).
Vimentin was the only marker with categorically positive or negative expression by the majority of cells in each population. The others exhibited striking variability, with co-existence of cells exhibiting a range of staining intensities. While overall expression of lineage markers like ER, CK19 and CK8/18 and CK5 was generally associated with luminal or basal subtype, this was not exclusive or uniform. For example, ER expression was highly heterogeneous amongst the luminal lines, and cytokeratins traditionally associated with basal or luminal differentiation were often expressed in subpopulations of the ‘opposite’ subtype. For example, CK19/8/18-positivity was common in CK5+ lines classified basal-like based on the transcriptomic average of the whole culture, and CK5+ subpopulations were present in cultures of ‘luminal’ cell lines. This diversity was unlikely to be introduced by technical bias, as we analysed at least two cores per line (minimum 100 cells/core), the scoring algorithm was based foremost on nucleus recognition and none of the markers bore any relationship to the regions-of-interest analysed (Fig-S2B). The data likely reflect bona fide biological diversity in the culture dish.
Taken together, these findings are consistent with the idea [2] that cultures of established cell lines comprise diverse subpopulations, even after extensive in vitro passage. The large cohort and panel of lineage markers investigated here confirm that this at least partly reflects their maintenance of restricted differentiation hierarchies. Heterogeneous expression of lineage markers is also evident on IHC-stained human tissue sections (for example, ‘basal-like’ tumours express CK19 focally or diffusely; and luminal, CK5+ cells lacking expression of basal/myoepithelial markers (vimentin, SMA and CK14) exist in the luminal layer of normal breast tissue [27]). Determining how faithfully this resembles hierarchies in the normal breast or the tumour-of-origin is outside the scope of this study, but could be investigated using single-cell transcriptomics, or time-lapse immunofluorescence to trace successive generations in vitro.

Pan-cancer pathway analysis using digital RNA counting

In the years since the original array-based taxonomy of breast tumours and spin-off classifiers for cell lines [35], we have learned much about molecular processes that distinguish the major divisions. Technology for quantifying gene expression has also evolved considerably, including digital platforms that directly count specific RNA molecules from complex mixtures, which drastically improves dynamic range and accuracy compared with amplification/hybridisation-based methods. We applied the nanoString PanCancer Pathways panel to the cohort using the nCounter® Dx system, to quantify 730 transcripts representing 13 canonical cancer pathways. We reasoned that a pathway-focused, digital analysis may identify new phenotypic groups.
Unsupervised analysis of these data produced five separate clusters, which generally conformed with classical gene-expression subtype (Fig. 3A/S3; Table-S3), with three exceptions:
(1)
Lines classified as ‘basal-like’ based on whole transcriptome profile or surrogate cytokeratin expression were divided: 4/7 were more similar to luminal/HER2 group (BT20, MDAMB468, HCC70 and HCC1954), while 3/7 exhibited more similarity to the claudin-low and normal groups (P7731, HCC1143 and PMC42ET, referred to as ‘basal-mesenchymal’). Indeed, analysing the expression of each pathway separately showed this basal stratification produced the lowest error (Fig. 3B). Basal and basal-mes groups were mostly discriminated by expression of PI3 K, TGFβ and transcriptional misregulation pathways (corrected p-values 2.1E−03, 4.6E−03 and 1.7E−03; Fig. 3C/S3B, Table-S4). Interestingly, basal-mes lines were indistinguishable from normal/immortalised lines by PanCancer pathway expression.
 
(2)
All five normal/immortalised lines co-segregated in this analysis (MCF10A and 184A1 have been classified as claudin-low, while 184B5, Bre80hTERT and SVCT are basal-like according to surrogate cytokeratin protein expression profiles [1] (Table-S2)). The luminal/HER2 group exhibited the most distinct phenotypes overall, marked by relatively low baseline expression of growth factor receptor signalling networks (Wnt/Hedgehog (HH), Ras, MAPK, JAK/STAT and PI3 K pathways).
 
(3)
There were two outliers in this analysis: MDAMB435, which exhibits lineage infidelity, including neuronal and melanocytic features [22, 23]; and MDAMB453, a luminal AR+ line (Fig. 2) [28].
 

p53 protein expression is associated with TP53 mutation status but is not a reliable surrogate in breast cancer cell lines

Functional abrogation of TP53 through germline (Li-Fraumeni syndrome) or somatic mutation is an important driver of breast cancer development, particularly in HER2+ and TN disease [29, 30]. Missense mutations can disrupt feedback signaling and/or stabilize the protein, resulting in accumulation within the nucleus (so-called “over-expression”). An optimized IHC method has recently been developed that shows p53 over-expression in serous ovarian cancer is an accurate predictor of mutation status [31], though the use of IHC as a TP53 mutation surrogate is currently not supported by the WHO/IARC [16, 17].
TP53 background is an important consideration for in vitro modelling of breast cancer, but genotype-expression relationships have not been clearly defined in cell lines. We therefore sequenced exons 4–10 of TP53 (encodes the DNA-binding domain and contains 99% of human mutations [16, 17]) in our cell line cohort, and identified mutations in 18/32 lines (56%; Table-S1). Using nanostring and dIHC data, we then analysed the relationships between TP53 mutation status, RNA and protein expression to see if expression might be a useful surrogate for loss-of-function, and whether this is impacted by protein heterogeneity. There was no association with RNA levels. p53 protein expression was higher in lines harbouring nonsynonymous mutations (p = 0.01), though there were multiple outliers in this analysis indicating that this method is not a reliable substitute for mutation analysis (Fig. 4).

Summary

Cell lines are extremely useful tools in breast cancer research. However, their advantages must be weighed against the limitations of modelling such a complex disease in vitro. Apart from the existence of clonal variants and phenotypic drift, we found that cultures of established lines exhibit striking heterogeneity (Fig. 5/Table-S5), at least partly reflecting maintenance of restricted lineage differentiation hierarchies in the culture dish. These factors are important to consider when selecting cell lines to test particular hypotheses, and interpreting data from in vitro assays. Targeted digital mRNA counting identified new subgroupings based on expression of important cancer pathways; notably, PanCan transcriptomes segregated basal lines into two subgroups that aligned with either luminal or mesenchymal phenotypes. This compendium of molecular profiling information has utility for informing selection of appropriate cell line models, and as a framework in which new PDCL and PDX can be characterised.

Acknowledgements

We would like to acknowledge the Brisbane Breast Bank for assistance with sample preparation and pathology review, and thank the following collaborators for their assistance and contributions: Tony Blick and Rik Thompson (St. Vincent’s Hospital, Melbourne and Queensland University of Technology (QUT)); Melissa Brown (UQ); Lisa Chopin (QUT); Lily Huschtscha (Westmead, The University of Sydney); Patricia Keith and Neelam Malik (UQ); Gillian Lehrbach, Chris Ormandy and Alex Swarbrick (Garvan Institute of Medical Research, Sydney); Mike McGuckin (Mater Medical Research Institute); and Sarah Roberts Thompson and Greg Monteith (UQ).

Funding

This work was supported by funding from the National Health and Medical Research Council (NHMRC program APP1017028 and APP1113867, NHMRC Project grant APP1082458), Australian Research Council (fellowship to FA, FT130101417) and the National Breast Cancer Foundation (fellowship to PTS).

Compliance with Ethical Standards

Conflict of Interest

All authors declare that they have no conflicts of interest.

Ethical approval

This article does not contain any studies with human participants or animals performed by any of the authors.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Gynäkologie

Kombi-Abonnement

Mit e.Med Gynäkologie erhalten Sie Zugang zu CME-Fortbildungen der beiden Fachgebiete, den Premium-Inhalten der Fachzeitschriften, inklusive einer gedruckten gynäkologischen oder urologischen Zeitschrift Ihrer Wahl.

Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Fußnoten
1
Cell line nomenclature has been simplified in this paper for ease of analysis, by removing dashes and spaces (e.g. MDA-MB-231 to MDAMB231).
 
Literatur
1.
Zurück zum Zitat Hollestelle A et al (2009) Distinct gene mutation profiles among luminal-type and basal-type breast cancer cell lines. Breast Cancer Res Treat 121(1):53–64CrossRefPubMed Hollestelle A et al (2009) Distinct gene mutation profiles among luminal-type and basal-type breast cancer cell lines. Breast Cancer Res Treat 121(1):53–64CrossRefPubMed
2.
Zurück zum Zitat Keller PJ et al (2010) Mapping the cellular and molecular heterogeneity of normal and malignant breast tissues and cultured cell lines. Breast cancer research: BCR 12(5):R87CrossRefPubMedPubMedCentral Keller PJ et al (2010) Mapping the cellular and molecular heterogeneity of normal and malignant breast tissues and cultured cell lines. Breast cancer research: BCR 12(5):R87CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Lehmann BD et al (2011) Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest 121(7):2750–2767CrossRefPubMedPubMedCentral Lehmann BD et al (2011) Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest 121(7):2750–2767CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Neve RM et al (2006) A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell 10(6):515–527CrossRefPubMedPubMedCentral Neve RM et al (2006) A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell 10(6):515–527CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Prat A et al (2010) Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast cancer research: BCR 12(5):R68CrossRefPubMedPubMedCentral Prat A et al (2010) Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast cancer research: BCR 12(5):R68CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Smart CE et al (2013) In vitro analysis of breast cancer cell line tumourspheres and primary human breast epithelia mammospheres demonstrates inter- and intrasphere heterogeneity. PLoS ONE 8(6):e64388CrossRefPubMedPubMedCentral Smart CE et al (2013) In vitro analysis of breast cancer cell line tumourspheres and primary human breast epithelia mammospheres demonstrates inter- and intrasphere heterogeneity. PLoS ONE 8(6):e64388CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Kao J et al (2009) Molecular Profiling of Breast Cancer Cell Lines Defines Relevant Tumor Models and Provides a Resource for Cancer Gene Discovery. PLoS ONE 4(7):e6146CrossRefPubMedPubMedCentral Kao J et al (2009) Molecular Profiling of Breast Cancer Cell Lines Defines Relevant Tumor Models and Provides a Resource for Cancer Gene Discovery. PLoS ONE 4(7):e6146CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Barretina J et al (2012) The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 483(7391):603–607CrossRefPubMedPubMedCentral Barretina J et al (2012) The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 483(7391):603–607CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Lehmann BD et al (2016) Refinement of Triple-Negative Breast Cancer Molecular Subtypes: implications for Neoadjuvant Chemotherapy Selection. PLoS ONE 11(6):e0157368CrossRefPubMedPubMedCentral Lehmann BD et al (2016) Refinement of Triple-Negative Breast Cancer Molecular Subtypes: implications for Neoadjuvant Chemotherapy Selection. PLoS ONE 11(6):e0157368CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Fillmore CM, Kuperwasser C (2008) Human breast cancer cell lines contain stem-like cells that self-renew, give rise to phenotypically diverse progeny and survive chemotherapy. Breast Cancer Res 10(2):R25CrossRefPubMedPubMedCentral Fillmore CM, Kuperwasser C (2008) Human breast cancer cell lines contain stem-like cells that self-renew, give rise to phenotypically diverse progeny and survive chemotherapy. Breast Cancer Res 10(2):R25CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Lim E et al (2010) Transcriptome analyses of mouse and human mammary cell subpopulations reveal multiple conserved genes and pathways. Breast Cancer Res 12(2):R21CrossRefPubMedPubMedCentral Lim E et al (2010) Transcriptome analyses of mouse and human mammary cell subpopulations reveal multiple conserved genes and pathways. Breast Cancer Res 12(2):R21CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat McCart Reed, A.E., et al., The Brisbane Breast Bank. Open Journal of Bioresources, 2017. In Press McCart Reed, A.E., et al., The Brisbane Breast Bank. Open Journal of Bioresources, 2017. In Press
14.
Zurück zum Zitat Smart CE et al (2013) In vitro analysis of breast cancer cell line tumourspheres and primary human breast epithelia mammospheres demonstrates inter- and intrasphere heterogeneity. PLoS ONE 8(6):e64388CrossRefPubMedPubMedCentral Smart CE et al (2013) In vitro analysis of breast cancer cell line tumourspheres and primary human breast epithelia mammospheres demonstrates inter- and intrasphere heterogeneity. PLoS ONE 8(6):e64388CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Da Silva L et al (2010) HER3 and downstream pathways are involved in colonization of brain metastases from breast cancer. Breast Cancer Res 12(4):R46CrossRefPubMedPubMedCentral Da Silva L et al (2010) HER3 and downstream pathways are involved in colonization of brain metastases from breast cancer. Breast Cancer Res 12(4):R46CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Bouaoun L et al (2016) TP53 Variations in Human Cancers: new Lessons from the IARC TP53 database and genomics data. Hum Mutat 37(9):865–876CrossRefPubMed Bouaoun L et al (2016) TP53 Variations in Human Cancers: new Lessons from the IARC TP53 database and genomics data. Hum Mutat 37(9):865–876CrossRefPubMed
20.
21.
Zurück zum Zitat Chambers AF (2009) MDA-MB-435 and M14 cell lines: identical but not M14 melanoma? Cancer Res 69(13):5292–5293CrossRefPubMed Chambers AF (2009) MDA-MB-435 and M14 cell lines: identical but not M14 melanoma? Cancer Res 69(13):5292–5293CrossRefPubMed
22.
Zurück zum Zitat Hollestelle A, Schutte M (2009) Comment Re: mDA-MB-435 and M14 cell lines: identical but not M14 Melanoma? Cancer Res 69(19):7893CrossRefPubMed Hollestelle A, Schutte M (2009) Comment Re: mDA-MB-435 and M14 cell lines: identical but not M14 Melanoma? Cancer Res 69(19):7893CrossRefPubMed
24.
Zurück zum Zitat Rae JM et al (2007) MDA-MB-435 cells are derived from M14 melanoma cells–a loss for breast cancer, but a boon for melanoma research. Breast Cancer Res Treat 104(1):13–19CrossRefPubMed Rae JM et al (2007) MDA-MB-435 cells are derived from M14 melanoma cells–a loss for breast cancer, but a boon for melanoma research. Breast Cancer Res Treat 104(1):13–19CrossRefPubMed
25.
Zurück zum Zitat Prasad VV, Gopalan RO (2015) Continued use of MDA-MB-435, a melanoma cell line, as a model for human breast cancer, even in year, 2014. NPJ Breast Cancer 1:15002CrossRefPubMedPubMedCentral Prasad VV, Gopalan RO (2015) Continued use of MDA-MB-435, a melanoma cell line, as a model for human breast cancer, even in year, 2014. NPJ Breast Cancer 1:15002CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Johnston RL et al (2016) High content screening application for cell-type specific behaviour in heterogeneous primary breast epithelial subpopulations. Breast Cancer Res 18(1):18CrossRefPubMedPubMedCentral Johnston RL et al (2016) High content screening application for cell-type specific behaviour in heterogeneous primary breast epithelial subpopulations. Breast Cancer Res 18(1):18CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Moore NL et al (2012) An androgen receptor mutation in the MDA-MB-453 cell line model of molecular apocrine breast cancer compromises receptor activity. Endocr Relat Cancer 19(4):599–613CrossRefPubMed Moore NL et al (2012) An androgen receptor mutation in the MDA-MB-453 cell line model of molecular apocrine breast cancer compromises receptor activity. Endocr Relat Cancer 19(4):599–613CrossRefPubMed
29.
Zurück zum Zitat Alsner J et al (2000) Heterogeneity in the clinical phenotype of TP53 mutations in breast cancer patients. Clin Cancer Res 6(10):3923–3931PubMed Alsner J et al (2000) Heterogeneity in the clinical phenotype of TP53 mutations in breast cancer patients. Clin Cancer Res 6(10):3923–3931PubMed
30.
Zurück zum Zitat TCGA, Cancer Genome Atlas Network: Comprehensive molecular portraits of human breast tumours. Nature, 2012. 490(7418): p. 61-70 TCGA, Cancer Genome Atlas Network: Comprehensive molecular portraits of human breast tumours. Nature, 2012. 490(7418): p. 61-70
31.
Zurück zum Zitat Kobel M et al (2016) Optimized p53 immunohistochemistry is an accurate predictor of TP53 mutation in ovarian carcinoma. J Pathol Clin Res 2(4):247–258CrossRefPubMedPubMedCentral Kobel M et al (2016) Optimized p53 immunohistochemistry is an accurate predictor of TP53 mutation in ovarian carcinoma. J Pathol Clin Res 2(4):247–258CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Forbes, SA et al (2011) COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res 39:945–950CrossRef Forbes, SA et al (2011) COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res 39:945–950CrossRef
Metadaten
Titel
Multidimensional phenotyping of breast cancer cell lines to guide preclinical research
verfasst von
Jodi M. Saunus
Chanel E. Smart
Jamie R. Kutasovic
Rebecca L. Johnston
Priyakshi Kalita-de Croft
Mariska Miranda
Esdy N. Rozali
Ana Cristina Vargas
Lynne E. Reid
Eva Lorsy
Sibylle Cocciardi
Tatjana Seidens
Amy E. McCart Reed
Andrew J. Dalley
Leesa F. Wockner
Julie Johnson
Debina Sarkar
Marjan E. Askarian-Amiri
Peter T. Simpson
Kum Kum Khanna
Georgia Chenevix-Trench
Fares Al-Ejeh
Sunil R. Lakhani
Publikationsdatum
09.09.2017
Verlag
Springer US
Erschienen in
Breast Cancer Research and Treatment / Ausgabe 1/2018
Print ISSN: 0167-6806
Elektronische ISSN: 1573-7217
DOI
https://doi.org/10.1007/s10549-017-4496-x

Weitere Artikel der Ausgabe 1/2018

Breast Cancer Research and Treatment 1/2018 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.