This experimental protocol describes the isolation of BCSCs from breast cancer cell and tissue samples as well as the in vitro and in vivo assays that can be used to assess BCSC phenotype and function.
Breast cancer stem cells (BCSCs) are cancer cells with inherited or acquired stem cell-like characteristics. Despite their low frequency, they are major contributors to breast cancer initiation, relapse, metastasis and therapy resistance. It is imperative to understand the biology of breast cancer stem cells in order to identify novel therapeutic targets to treat breast cancer. Breast cancer stem cells are isolated and characterized based on expression of unique cell surface markers such as CD44, CD24 and enzymatic activity of aldehyde dehydrogenase (ALDH). These ALDHhighCD44+CD24– cells constitute the BCSC population and can be isolated by fluorescence-activated cell sorting (FACS) for downstream functional studies. Depending on the scientific question, different in vitro and in vivo methods can be used to assess the functional characteristics of BCSCs. Here, we provide a detailed experimental protocol for isolation of human BCSCs from both heterogenous populations of breast cancer cells as well as primary tumor tissue obtained from breast cancer patients. In addition, we highlight downstream in vitro and in vivo functional assays including colony forming assays, mammosphere assays, 3D culture models and tumor xenograft assays that can be used to assess BCSC function.
Understanding the cellular and molecular mechanisms of human breast cancer stem cells (BCSCs) is crucial for addressing the challenges encountered in breast cancer treatment. The emergence of the BCSC concept dates back to the early 21st century, where a small population of CD44+CD24-/low breast cancer cells were found to be capable of generating heterogenous tumors in mice1,2. Subsequently, it was observed that human breast cancer cells with high enzymatic activity of aldehyde dehydrogenase (ALDHhigh) also displayed similar stem cell-like properties3. These BCSCs represent a small population of cells capable of self-renewal and differentiation, contributing to the heterogenous nature of bulk tumors1,2,3. Accumulating evidence suggest that alterations in evolutionarily conserved signaling pathways drive BCSC survival and maintenance4,5,6,7,8,9,10,11,12,13,14. In addition, the cell extrinsic microenvironment has been shown to play a pivotal role in dictating different BCSC functions15,16,17. These molecular pathways and the external factors regulating BCSC function contribute to breast cancer relapse, metastasis18 and development of resistance to therapies19,20,21, with the residual existence of BCSCs post-treatment posing a major challenge to the overall survival of breast cancer patients22,23. Pre-clinical evaluation of these factors is therefore very important for identifying BCSC-targeting therapies that could be beneficial for achieving better treatment outcomes and improved overall survival in breast cancer patients.
Several in vitro human breast cancer cell line models and in vivo human xenograft models have been used to characterize BCSCs24,25,26,27,28,29. The ability of cell lines to continuously repopulate after every successive passage makes these an ideal model system to perform omics-based and pharmacogenomic studies. However, cell lines often fail to recapitulate the heterogeneity observed in patient samples. Hence, it is important to complement cell line data with patient-derived samples. Isolation of BCSCs in their purest form is important for enabling detailed characterization of BCSCs. Achieving this purity depends on the selection of phenotypic markers that are specific to BCSCs. Currently, the ALDHhighCD44+CD24– cell phenotype is most commonly used to distinguish and isolate human BCSCs from bulk breast cancer cell populations using fluorescence activated cell sorting (FACS) for maximum purity1,3,26. Furthermore, the properties of isolated BCSCs such as self-renewal, proliferation, and differentiation can be evaluated using in vitro and in vivo techniques.
For example, in vitro colony forming assays can be used to assess the ability of a single cell to self-renew to form a colony of 50 cells or more in presence of different treatment conditions30. Mammosphere assays can also be used to assess the self-renewal potential of breast cancer cells under anchorage-independent conditions. This assay measures the ability of single cells to generate and grow as spheres (mixture of BCSCs and non-BCSCs) at each successive passage in serum-free non-adherent culture conditions31. Additionally, 3-Dimensional (3D) culture models can be used to assess BCSC function, including cell-cell and cell-matrix interactions that closely recapitulate the in vivo microenvironment and allow investigation of the activity of potential BCSC-targeted therapies32. Despite the diverse applications of in vitro models, it is difficult to model the complexity of in vivo conditions using only in vitro assays. This challenge can be overcome by use of mouse xenograft models to evaluate BCSC behavior in vivo. In particular, such models serve as an ideal system for assessing breast cancer metastasis33, investigating interactions with the microenvironment during disease progression34, in vivo imaging35, and for predicting patient-specific toxicity and efficacy of antitumor agents34.
This protocol provides a detailed description for the isolation of human ALDHhighCD44+CD24– BCSCs at maximum purity from bulk populations of heterogenous breast cancer cells. We also provide a detailed description of three in vitro techniques (colony forming assay, mammosphere assay, and 3D culture model) and an in vivo tumor xenograft assay that can be used to assess different functions of BCSCs. These methods would be appropriate for use by investigators interested in isolating and characterizing BCSCs from human breast cancer cell lines or primary-patient derived breast cancer cells and tumor tissue for the purposes of understanding BCSC biology and/or investigating novel BCSC-targeting therapies.
Collection of patient-derived surgical or biopsy samples directly from consenting breast cancer patients were carried out under approved human ethics protocol approved by the institutional ethic board. All mice used to generate patient-derived xenograft models were maintained and housed in an institution approved animal facility. The tumor tissue from patient-derived xenograft models using mice were generated as per approved ethics protocol approved by the institutional animal care committee.
1. Preparation of cell lines
2. Preparation of breast cancer tumor tissue
3. Generation of single cell suspensions of breast cancer cells
4. Generation of single cell suspension from tissue samples
5. Isolation of breast cancer stem cells (BCSCs)
Figure 1: FACS gating strategy for isolation of BCSCs from breast cancer cell lines and tissue samples. (A) Flowchart describing the procedure of BCSC isolation. (B) Representative FACS plots showing the sort strategy used to isolate viable BCSCs and non-BCSCs from a heterogenous pool of cells. MDA-MB-231 human breast cancer cells are concurrently labeled with 7-AAD, CD44-APC, CD24-PE and the ALDH substrate. Cell subsets were isolated using a four-color protocol on a FACS machine. Cells are selected based on expected light scatter, then for singlets, and viability based on 7-AAD exclusion. Cells are then analyzed for ALDH activity and the top 20% most positive are selected as the ALDHhigh population, while the bottom 20% of cells with the lowest ALDH activity were deemed to be ALDHlow. Finally, 50% of the ALDHlow cells are further selected based on a CD44low/-CD24+ phenotype, and 50% of the ALDHhigh cells are selected based on CD44+CD24– phenotype. This figure has been adapted from Chu et al.17. Please click here to view a larger version of this figure.
Figure 2: BCSCs proportions are variable in different breast cancer cell lines. Representative image showing the differential proportion of BCSCs and non-BCSCs in (A) SUM159 and (B) MDA-MD-468 triple negative breast cancer cell lines following labelling and sorting as described in Figure 1. Please click here to view a larger version of this figure.
6. Colony forming assay
7. Mammosphere assay
8. 3D culture model
Figure 3: In vitro assays to assess BCSC cell function. In vitro assays were performed as described in protocol sections 6.1 to 6.5 (A), 7.1 to 7.4 (B), or 81. to 8.4 + 8.6 (C). (A) Representative image showing the colonies generated by MDA-MB-231 human breast cancer cells; (B) Representative images showing mammosphere formation by MCF7, SUM159, or MDA-MB-468 human cell lines as well as patient-derived LRCP17 breast cancer cells. (C) Representative images showing the 3D structures formed by MCF7 and MDA-MB-231 breast cancer cells in 3D cultures models. Please click here to view a larger version of this figure.
NOTE: Perform animal experiments under an animal ethics protocol approved by the institutional animal care committee.
9. In vivo xenograft model
The described protocol allows isolation of human BCSCs from a heterogenous population of breast cancer cells, either from cell lines or from dissociated tumor tissue. For any given cell line or tissue sample, it is crucial to generate a uniform single cell suspension to isolate BCSCs at maximum purity as contaminating non-BCSC populations could result in variable cellular responses, especially if the study aim is to evaluate the efficacy of therapeutic agents targeting BCSCs. Application of a stringent sorting strategy will minimize the presence of contaminating non-BCSCs and result in the ability to collect the proportion of breast cancer cells with stem cell-like characteristics that display a cellular phenotype that distinguishes them from bulk population of cancer cells. Human breast cancer cells that exhibit enhanced ALDH enzymatic activity, express high levels of the cell surface marker CD44, and low/negative expression of CD24 have an ALDHhighCD44+CD24– phenotype and can be classified as BCSCs. The proportion of BCSCs within the bulk population can vary between cell lines or patients (Figure 2), and often depends on disease stage, with more aggressive breast cancer usually displaying a higher proportion of BCSCs26,36,37.
Isolated BCSCs can be used to perform different in vitro and in vivo assays where their behavior and function can be compared to that of the bulk and/or non-BCSC populations. For example, the ability of a single breast cancer cell to self-renew and generate colonies of 50 cells can be assessed by colony-forming assays (Figure 3A). The ability of BCSCs to self-renew under anchorage-independent experimental conditions can be assessed by mammosphere assays, where variable sphere number, size, and sphere-initiating capacity can be analyzed and correlated with the presence and function of BCSCs (Figure 3B). It is important to determine the seeding cell densities for different breast cancer cell lines or breast tumor samples to obtain optimal results. This is particularly important when performing SLDA, as higher cell densities could lead to cell aggregation resulting in misinterpretation of cellular activity.
Culturing breast cancer cells in BME allows BCSCs to form 3D structures that recapitulate in vivo conditions (Figure 3C). 3D culture of breast cancer cells in the presence of other microenvironmental cell types such as fibroblasts, endothelial cells, and/or immune cells has the added capacity for investigating the role of microenvironment in 3D growth of BCSCs38,39. The specific cell numbers required to generate 3D organoids may vary depending on the cell line or patient tumor source, and thus it is important to optimize the culture conditions and cell numbers prior to any large-scale experiments.
Finally, in vivo mouse xenograft models can be used to understand the differences in growth (Figure 4) self-renewal, differentiation and/or tumor-initiating ability of BCSCs in vivo compared to non-BCSCs or bulk cell populations. Often, the in vitro cellular responses observed in the presence of exogenous factors or therapeutic agents is not representative of in vivo setting, suggesting that in vitro observation should be complimented with in vivo studies whenever feasible. Using in vivo xenograft models, the cellular heterogeneity and tumor architecture is preserved and thus these models can serve as a system that closely mimics the microenvironment in human patients. In vivo LDA can be performed to determine the proportion of tumor-initiating cells in a given mixed population of cancer cells (BCSCs or non-BCSCs)40,41. The range of cell dilutions used should be optimized and will depend on the frequency of initiating cells in the cell population of interest. Ideally these dilutions should include doses that result in 100% tumor formation, down to cell doses with no tumor formation and a reasonable range in between. The frequency of tumor-initiating cells in primary samples can be variable, and in instances where breast tumors have very low numbers or heterogenous populations of tumor-initiating cells, performing LDA can be particularly challenging42. In these cases, injecting larger number of cells would be more appropriate for understanding breast cancer biology.
Figure 4: In vivo xenograft assays to assess BCSC function. MDA-MB-231 breast cancer cells were isolated by FACS as described in Figure 1 and injected into the right thoracic mammary fat pad of female NSG mice as described in protocol sections 9.1 to 9.8 (5 x 105 cells/mouse; 4 mice/cell population). Primary breast tumor growth kinetics are shown for ALDHhiCD44+CD24– (■) versus ALDHlowCD44low/-CD24+ (□) populations. Data represented as the mean ± S.E.M. * = significantly different tumor size than respective ALDHlowCD44low/- subsets at the same time-point (P < 0.05). This figure has been adapted from Croker et al.26. Please click here to view a larger version of this figure.
Breast cancer metastasis and resistance to therapy have become major cause of mortality in women worldwide. The existence of a sub-population of breast cancer stem cells (BCSCs) contributes to enhanced metastasis26,43,44,45,46 and therapy resistance21,47,48. Therefore, the focus of future treatments should aim at eradicating BCSCs to achieve better treatment outcomes, and this requires accurate methods for isolating and characterizing the functional characteristics of BCSCs using both in vitro and in vivo methods.
Immortalized cell lines derived from different subtypes of breast cancer have proven to be feasible models to study breast cancer biology including the isolation and characterization of BCSCs26,49,50. The high proliferative capacity and unlimited expansion ability of cell lines provides an ideal model system for performing studies that are highly reproducible and technically straightforward. However, due to the clonal origin of cell lines, they may fail to recapitulate the heterogeneity exhibited by different patients and/or by cancer cells within tumor tissue. In addition, genetic alterations can be acquired during serial passaging of cell lines and may induce genotypic or phenotypic changes that can confound experimental results51. In contrast, primary patient-derived cells, despite their limited proliferative and expansion ability, may provide a more accurate model to that observed in vivo. However, such samples may be more difficult to acquire and be more technically challenging to work with. All of these factors should be considered when choosing a starting model system with which to isolate and characterize BCSCs.
FACS is a commonly used technique to isolate cells of interest based on cell surface marker expression52,53. Based on cell surface antigens (CD44 and CD24) and ALDH enzymatic activity, human BCSCs can be isolated at high purity from both breast cancer cell lines and tumor tissues1,2. The sorting efficiency determines the purity of sorted sample, and it is recommended that users analyze a small portion of sorted sample incubated with viability dye to check the efficiency of sorting53,54. The sorting efficiency can be confounded by many factors including the presence of cell clumps, a high number of dead or dying cells, improper compensation of the fluorochromes and/or damage to cell surface antigens due to sensitivity to trypsin or collagenase during pre-sorting dissociation steps53,54,55,56. Therefore, generation of a proper single cell suspension and use of appropriate cell dissociation techniques will increase the sorting efficiency. While performing multiparameter cell sorting, it is important to choose fluorochromes that minimizes spectral overlap. In some cases, where spectral overlap cannot be avoided, a control that contains all the fluorochromes except one (fluorescence minus one, FMO) should be used to minimize the spillover of fluorescent signals into other channels54. Alternatively, the spectral overlap can reduced by immunomagnetically isolating cell populations prior to final FACS isolation of cells of interest56.
In vitro assays such as the colony-forming and mammosphere assays described in this protocol have been extensively used to study the self-renewal and proliferative ability of BCSCs57,58,59,60,61,62. Additionally, these assays can be used to assess the activity of different therapeutic drugs on BCSC function. Several evolutionarily conserved signaling pathways have been implemented in BCSC maintenance63, and both colony-forming64,65,66 and mammosphere assays64,67 have been used to assess the value of therapeutic disruption of these pathways as an intervention to block BCSC intrinsic signaling and reduce BCSC activity and disease progression. Colony forming assay using primary cells can be challenging due to low cell density, variation between samples and lack of its adaptability to isolated in vitro conditions. These challenges can be overcome by culturing BCSCs on a soft agar layer or by coculturing them with fibroblasts on a collagen-coated cell culture dish68,69,70. In addition, supplementing growth factors into the culture media (such as FGF771) could also improve the colony-forming ability of cells isolated from tissue samples. In addition, over-digestion of tissue using collagenase or trypsin during single cell suspension generation step can result in low to zero colony-forming ability and reduce mammosphere-forming efficiency31. In both assays, care should be taken to incubate the assay plates undisturbed to avoid disruption of colony or sphere structures as they are forming. It is also recommended that users extend the incubation period for primary cells (relative to cell lines) as it might take longer for these cells to form colonies or spheres.
Multiple lines of evidence have demonstrated the critical role of extracellular matrix (ECM)15,17,72 and stromal components, such as fibroblasts, immune cells, endothelial cells and adipocytes in influencing BCSC functions15. Thus, the 3D culture model we describe in this protocol can provide a useful experimental system for helping to recapitulate the in vivo tumor microenvironment in an in vitro setting. Although the 3D culture system closely resembles the tumor microenvironment in cancer patients, long term maintenance of cells as organoids can be difficult. In addition, optimization of the 3D culture conditions and the ability to accurately investigate self-renewal and differentiation ability of BCSCs is challenging73. The efficiency of organoids formed in 3D culture system depends on the growth factors supplemented in the culture media74. Absence of key components (for example, ROCK inhibitor) could lead to reduced or no organoid formation74. Media should be replenished every 3-4 days to maintain optimal cellular function and the sustainability of the culture. In order to recapitulate in vivo conditions and response, it is always important to allow the cells to form organoids prior to any kind of exogenous treatment75. Cells derived from patient samples should be giving sufficient time to form organoids, particularly if the objective is the evaluate drug response75.
While these in vitro methods are attractive and accessible experimental tools for characterizing BCSC function, tumor heterogeneity and the effect of tumor microenvironment on BCSC behavior cannot be studied with complete effectiveness. These in vitro assays should therefore be complemented with in vivo xenograft models whenever feasible in order to further validate experimental findings related to BSCS biology and/or response to novel therapeutics. Different in vivo models have been used study BCSC tumorigenicity and metastasis. Ectopic (subcutaneous engraftment) and orthotopic (MFP engraftment) mouse models have been used to generate breast tumors and assess longitudinal changes in tumor growth over time50. Although both in vivo injection approaches can be used to study BCSC biology, the native stromal and vasculature-related components of the MFP allow more accurate recapitulation of primary breast tumor progression as observed in patients, and thus MFP injection is preferred76,77,78. Finally, the use of immunocompromised mice is required for engraftment of human BCSCs and tumor growth, and this prevents incorporating the role of immune cells in tumorigenesis and metastasis studies79. More recently, this limitation has been addressed through the use of humanized mice in which a human immune system is reconstituted via bone marrow transplantation prior to the initiation of xenograft studies80,81,82. However, these models are expensive and technically challenging, and thus are still not commonly used83.
In summary, here we have provided a protocol for the isolation of human BCSCs from both breast cancer cell lines and patient-derived tumor tissue samples. We have also described in vitro and in vivo protocols for downstream assays that can be used to study BCSC function, with the ability to be optimized for different breast cancer cell sources and the flexibility to be performed under different experimental conditions. These protocols will be useful for investigators interested in cancer stem cells, breast cancer biology and therapeutic development, with the ultimate goal of improving patient outcomes in the future.
The authors have nothing to disclose.
We thank members of our laboratory for their helpful discussions and support. Our research on breast cancer stem cells and the tumor microenvironment is funded by grants from the Canadian Cancer Research Society Research Institute and the U.S. Army Department of Defense Breast Cancer Program (Grant # BC160912). V.B. is supported by a Western Postdoctoral Fellowship (Western University), and both A.L.A. and V.B. are supported by the Breast Cancer Society of Canada. C.L. is supported by a Vanier Canada Graduate Scholarship from the Government of Canada.
7-Aminoactinomycin D (7AAD) | BD | 51-68981E | suggested: 0.25 µg/1×106 cells |
Acetone | Fisher | A18-1 | |
Aldehyde dehydrogenase (ALDH) substrate | Stemcell Technologies | 1700 | Sold commerically as part of the ALDEFLOUR Assay kit; follow manufacturer's instructions for ALDH substrate preparation |
Basement membrane extract (BME) | Corning | 354234 | Sold under the commercial name Matrigel |
Cell culture plates: 6 well | Corning | 877218 | |
Cell culture plates: 60mm | Corning | 353002 | |
Cell culture plates: 96-well ultra low attachment | Corning | 3474 | |
Cell strainer: 40 micron | BD | 352340 | |
Collagen | Stemcell Technologies | 7001 | Prepare 1:30 dilution of 3 mg/mL collagen in PBS |
Collagenase | Sigma | 11088807001 | 1x |
Conical tubes: 50 mL | Fisher scientific | 05-539-7 | |
Crystal violet | Sigma | C6158 | Use 0.05% crystal violet solution in water for staining |
Dispase | Stemcell Technologies | 7913 | 5U/mL |
DMEM:F12 | Gibco | 11330-032 | 1x, With L-glutamine and 15 mM HEPES |
DNAse | Sigma | D5052 | 0.1 mg/mL final concentration |
FBS | Avantor Seradigm Lifescience | 97068-085 | |
Flow tubes: 5ml | BD | 352063 | Polypropylene round-bottom tubes |
Methanol | Fisher | 84124 | |
mouse anti-Human CD24 antibody | BD | 561646 | R-phycoerythrin and Cyanine dye conjugated Clone: ML5 |
mouse anti-Human CD44 antibody | BD | 555479 | R-phycoerythrin conjugated, Clone: G44-26 |
N,N-diethylaminobenzaldehyde (DEAB) | Stemcell Technologies | 1700 | Sold commerically as part of the ALDEFLOUR Assay kit; follow manufacturer's instructions DEAB preparation |
PBS | Wisent Inc | 311-425-CL | 1x, Without calcium and magnesium |
Trypsin-EDTA | Gibco | 25200-056 | |
Mammosphere Media Composition | |||
B27 | Gibco | 17504-44 | 1x |
bFGF | Sigma | F2006 | 10 ng/mL |
BSA | Bioshop | ALB003 | 04% |
DMEM:F12 | Gibco | 11330-032 | 1x, With L-glutamine and 15 mM HEPES |
EGF | Sigma | E9644 | 20 ng/mL |
Insulin | Sigma | 16634 | 5 µg/mL |
3D Organoid Media Composition | |||
A8301 | Tocris | 2939 | 500 nM |
B27 | Gibco | 17504-44 | 1x |
DMEM:F12 | Gibco | 11330-032 | 1x, With L-glutamine and 15 mM HEPES |
EGF | Sigma | E9644 | 5 ng/mL |
FGF10 | Peprotech | 100-26 | 20 ng/mL |
FGF7 | Peprotech | 100-19 | 5 ng/mL |
GlutaMax | Invitrogen | 35050-061 | 1x |
HEPES | Gibco | 15630-080 | 10 mM |
N-acetylcysteine | Sigma | A9165 | 1.25 mM |
Neuregulin β1 | Peprotech | 100-03 | 5 nM |
Nicotinamide | Sigma | N0636 | 5 mM |
Noggin | Peprotech | 120-10C | 100 ng/mL |
R-spondin3 | R&D | 3500 | 250 ng/mL |
SB202190 | Sigma | S7067 | 500 nM |
Y-27632 | Tocris | 1254 | 5 µM |