Critical Reviews in Oncology/Hematology 85 (2013) 95–111
Issues to be considered when studying cancer in vitro ∗ ˇ Beata Cunderlíková International Laser Centre, Ilkoviˇcova 3, 841 04 Bratislava, Slovakia Accepted 27 June 2012
Contents 1. 2.
3.
4.
5.
Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Cell modifications during in vitro cultivation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.1. Representativeness of tumour cell lines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.2. Cell modifications due to culturing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.3. Cultured vs. uncultured cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Lack of relevant interactions in vitro . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.1. Extracellular matrix . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.2. Stromal cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.3. Evidence from animal studies supporting importance of tumour cell environment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.4. Species specificity. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Lack of structural context in vitro . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.1. Spatial organization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.2. ECM stiffness . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Reviewers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Biography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
95 96 96 97 98 99 99 100 101 102 103 103 103 104 104 104 104 111
Abstract Various cancer treatment approaches have shown promising results when tested preclinically. The results of clinical trials, however, are often disappointing. While searching for the reasons responsible for their failures, the relevance of experimental and preclinical models has to be taken into account. Possible factors that should be considered, including cell modifications during in vitro cultivation, lack of both the relevant interactions and the structural context in vitro have been summarized in the present review. © 2012 Elsevier Ireland Ltd. All rights reserved. Keywords: In vitro; Cell culturing; Extracellular matrix; Tumour microenvironment; Tissue stiffness
1. Introduction Intensive efforts within cancer research resulted in a number of novel anticancer agents. Positive results of treatments with some of them have been documented in patients. The most promising examples include anti-epidermal ∗
Tel.: +421 2 654 21 575; fax: +421 2 654 23 244. E-mail addresses:
[email protected],
[email protected]
1040-8428/$ – see front matter © 2012 Elsevier Ireland Ltd. All rights reserved. http://dx.doi.org/10.1016/j.critrevonc.2012.06.007
growth factor receptor (EGFR) monoclonal antibodies in some patients with metastatic colorectal cancer [1] and metastatic breast cancer [2] or tyrosine kinase inhibitors in some patients with chronic myelogenous leukemia [3] or non-small cell lung cancer [4]. However, the efficacy of many of the newer biological and targeted therapies is confirmed in some individuals only. For example, only patients with colon tumours expressing wild-type KRAS or with HER2/neu-positive breast cancer are likely to benefit from
96
ˇ B. Cunderlíková / Critical Reviews in Oncology/Hematology 85 (2013) 95–111
the treatments with the anti-EGFR monoclonal antibodies [1]. Treatment with tyrosine kinase inhibitor (imatinib) is efficient in patients with BCR-ABL translocations [5]. Except of the encouraging achievements, however, negative results of clinical trials are also reported. To mention some of them, treatment with matrix metalloproteinase (MMP) inhibitors showed no survival advantage for the patients compared to conventional treatment modalities or even lead to poorer survival and had to be terminated [6–8]. Clinical trials with Fe-chelator Triapine® as anticancer drug, either alone in patients with metastatic renal cell carcinoma [9] or in combination with gemcitabine in patients with advanced pancreatic adenocarcinoma [10], were terminated as they did not give any benefit for the patients at the used doses and schedules. Clinical trials with antiEGFR agent-combining regimes in advanced non-small cell lung cancer patients showed no survival advantages over chemotherapy alone [11]. Based on in vitro observations of a dose–response effect, high-dose chemotherapy with stemcell transplantation has been evaluated for the treatment of various solid cancers and compared with conventional modalities. Although event-free survival was often found to be advantageous in favour of high-dose chemotherapy, no significant benefit in overall survival, apart from few exceptions, or even negative results were reported [12,13]. Current gene therapy has not proven very successful in clinical trials [14,15]. Understanding the reasons for the failures of clinical trials, in spite of promising preclinical results, is of high importance from scientific point of view. While searching for them, the relevance of experimental and preclinical models has to be considered. Predictions of treatment efficacies in clinical trials based on in vitro studies [16] and even preclinical mouse models [8] were reported to lead to disappointments in a number of cases. For example, Fe-chelator Dp44mT did not lead to Fe depletion within the tumour, despite its high activity at inhibiting Fe uptake from transferrin and inducing Fe mobilisation from cells in cultures [17]. Many of tumours that relapsed after treatment by radiotherapy appeared within irradiated regions, despite in vitro and preclinical data indicating that the doses used would kill >99% of both the tumour and endothelial cells [18]. Moreover, in a randomized trial of a regime combining application of a monoclonal antibody against EGFR, there was no correlation between EGFR expression and tumour response and/or symptom improvement in patients with non-small cell lung cancer [11]. In a phase III trial with a humanized monoclonal antibody against vascular endothelial growth factor (VEGF), VEGF expression, assessed by in situ hybridization in primary tumour samples, did not correlate with objective response in advanced breast cancer patients [11]. Apart from inconsistent findings between in vitro studies and clinical trials, there appear also reports that document discrepancies between results obtained under in vitro and in vivo (e.g. [19–26]) or ex vivo (e.g. [25,27]) conditions. In fact, many researchers realized limitations of usually used
in vitro and in vivo experimental systems already long time ago. It has been recognized that cell culture systems, and even animal carcinogenesis models, may not accurately represent the complexity and the true physiologic state of the diseased human tissue [28] and simplification inherent to vitro approaches might be achieved at the cost of physiological relevance [29]. In order to be able to correctly interpret findings of in vitro and preclinical experiments, it is crucial to understand the physiological and pathological relevance of the experimental conditions. Therefore, factors governing cell behaviour need to be precisely identified and the alterations of cells in cultures that are artifacts of culturing (e.g. cell modifications due to long-term culturing or cell processing) need to be distinguished. Unfortunately, inconsistency in experimental protocols and conditions used makes comparisons of the results among laboratories not easy. Issues that should be considered in experiments utilizing in vitro cell cultures and in interpreting their findings, especially when studying cancer, have been summarized in the present review.
2. Cell modifications during in vitro cultivation Use of cell cultures offers considerable simplification for biologically oriented research. Nevertheless, evidence suggests that cells undergo changes when placed in cultures [30–35] (Tables 1 and 2), which gives rise to doubts whether cell lines are representatives of their original tissues [36–38]. 2.1. Representativeness of tumour cell lines Many tumour cell lines have been derived from metastases, pleural or ascitic effusions (references in [36,39]). Such cells constitute a population of cells in a late stage of tumour evolution (references in [36]), which may differ from their corresponding primary tumours with respect to ploidy [36], antigenicity and immunogenicity and therefore the response to therapies [40,41]. Cell lines developed from primary tumours were claimed to be representatives of the tumour specimen from which they were derived [36,42,43]. Nevertheless, difficulties to establish even primary cultures from primary tumours [36,39,44–46] with majority of outgrowths arising from normal cells within the specimen [45,46] have often been reported. Success rates for cell line establishments from primary and metastatic breast cancers were 10% and 25%, respectively [47]. Low success rates in the range of 1–10% were reported for the establishment of leukaemia–lymphoma cell lines. It was suggested that the original primary cells might need to possess special features that make them prone to immortalization in vitro and only samples containing such cells may evolve into cell lines [48]. It was even mentioned that only rare specimens of breast cancer develop into cell lines [33,46]. Furthermore, the growth of primary carcinoma cells was found to be considerably slower than that of carcinoma cell lines (references in [45]). This is
ˇ B. Cunderlíková / Critical Reviews in Oncology/Hematology 85 (2013) 95–111
97
Table 1 Cell modifications observed during conventional cell culturing and processing. Influencing factor
Consequence
Reference
Substrate (culturing on plastic)
Cell dedifferentiation (loss of tissue specific gene expression, differentiated functions and structural organization) Modification of the expression of cell type markers Decrease in cell doubling time Chromosomal changes Development of features characteristic of transformation Reversion of properties of transformed phenotype to normal one Loss of chemoresistance-mediating factors Ploidy changes
[20,22,34,51,52,72,77–79]
Reduction of receptor binding sites Cell stability (possibly)
[98] [35]
Duration (extended time in culture/passaging)
Processing (extended storage at −70 ◦ C) (repeated treatment with EDTA-trypsin)
clearly opposite to the situation that we face within in vitro studies, where it is easy to culture tumour cell lines, while “normal” ones require more demanding conditions. 2.2. Cell modifications due to culturing Dedifferentiation on plastic. The changes in cell behaviour may be induced by environmental conditions during culturing, namely by extracellular matrix (ECM, [22,49–69]), cell culture substrate [70–75] as well as by composition of the cultivation medium [35,39,50,52,76]. Importantly, the most remarkable modifications compared to in vivo conditions were reported to occur in cells cultured on the most frequently used substrate-plastic. Dedifferentiation following cell culturing on plastic [20,22,51,52,72,77,78]
[39,76,84,85,99] [33,35] [33,35] [33] [75,104,105] [101] [36]
is accompanied by the loss of expression of tissue specific genes, differentiated functions as well as capability to form organized structures [34,51,77–79]. Re-appearance of various differentiated properties could be induced by providing the cells with microenvironment more closely mimicking in vivo conditions (references in [19,51,80], Fig. 1). Although these conclusions come mostly from the experiments with epithelial cells, the process of dedifferentiation has been rather extensively studied also in isolated adult rat cardiomyocytes [81–83]. The cells isolated from normal hearts acquired a number of properties of hypertrophic rather than normal myocytes in “long-term” cultures [82]. Vimentin appearance. Another property induced by cell culturing on plastic (Table 1), which is not consistent with the situation in vivo, is appearance of vimentin filaments
Table 2 Factors that should be considered when working with in vitro cell cultures. Representativeness of tissue Stability (permanent phenotypic changes due to processing, passaging, storage, medium composition, etc./genotypic drift) Context-dependent phenotype/dedifferentiation on plastic Cell-type specific behaviour
[36–46,248,249]a [33,35,36,76,98,99,102,103]
“Correct” ECM composition Matrigel (basement membrane) Type I collagen (tumour stroma) Fibroblast-derived matrices (normal, primed, activated stroma) Tissue-extracted ECM Artificial-material based matrices Co-cultures (fibroblasts, myoepithelial cells, astrocytes, macrophages, etc.) Source Type Abundance
[58,69,161,179] [150,151] [69,232,237,257] [152–154]
Structural context
3 Dimensionality ECM fibre architecture Tissue-relevant ECM stiffness
[69,100,108,161,202,227–237] [57,120,152–154,238,239] [157,237–243,247]
Others
Species dependency Hematopoietic/immune system Vascular/lymphatic system Interstitial fluid flow, local gradients, etc.
[219–223] [18,117,180,181] [161] [159]
Cell properties
Interactions cell–ECM (biochemical)
cell–cell
a
Examples of literature, including review articles with references to relevant seminal work, are included in the table.
[20,22,49–83] [58,59,63,139,161,250–256]
[155] [156–161] [66,67,130–137,144–147,199–207] [87,88,189–194] [152,182,185,186,188,194] [86,194]
ˇ B. Cunderlíková / Critical Reviews in Oncology/Hematology 85 (2013) 95–111
98
Components: conventional 2D culture
tumor epithelial cells non-tumor epithelial cells tumor-associated fibroblasts normal fibroblasts tumor-associated myoepithelial cells
D A
normal myoepithelial cells tumor-associated macrophages/other stromal cells normal macrophages/other stromal cells ECM fiber
3D culture mimicking in vivo microenvironment
basement membrane
Interactions: through direct contact through released substances
C normal ECM
activated ECM B
Fig. 1. Culturing of cells under conventional and in-vivo mimicking conditions. Components of an in vivo-mimicking culture as well as interactions important for cell behaviour and function well documented in the literature are shown. Additional factors that need to be considered are listed in Table 2. Depending on cell type and ECM used, the cells in 3D cultures may form different 3D structures, such as polarized acini with central lumens (A), polar reverted spheroids (B), disorganized colonies (C) or tubules (D).
in majority of cultured cells of epithelial and muscle origin [84,85]. The vimentin filaments could be sometimes detected even together with another type of intermediate filaments [84]. Vimentin is a major cytoskeletal component of mesenchymal cells and it is therefore frequently used as a marker of mesenchymal cells as well as cells undergoing epithelial– mesenchymal transition, whereas cytokeratins are used as markers of epithelial cells. Applications include identification and characterization of cell types [39,86–88] including cells with stem cell properties [89–92], evidence of epithelialmesenchymal transition [93] or detection of tumour micrometastases [94]. Expression of vimentin induced by cell culturing on plastic should be considered in these cases, but also with regard to the role of cytoskeleton in intracellular signalling and mechanotransduction [54,95–97]. Alterations due to cell processing. Alterations of the cultured cells could be caused by cell processing, as a consequence of extended storage at −70 ◦ C [98], repeated treatment with EDTA–trypsin [35], extended time in the culture and increased number of cultivation passages (Table 1). The latter two factors were reported to lead to decrease in doubling time and chromosomal changes [33,35], changes in adhesion complexes and increased potential to form tumours [33] or altered keratin expression [39,76,99] in epithelial cell lines, increase in proliferation of lipocytes [100] or to changes in phenotype of transformed fibroblasts [75]. Progressive loss of factors mediating chemoresistance on passaging of cells from metastases but not from primary tumours in monolayers was reported [101] and loss of tumourigenicity with cell passaging is also observed. Nevertheless, it was concluded that cell lines are relatively stable if they are kept under the same experimental conditions, however, when they are exposed to
different environments, different variants could be selected [102,103]. The appearance of cell variants with reversion of the properties of transformed cells to the parental or more normal phenotype [75,104,105] indicates rather poor control over the cell cultures. The reversion was found to be influenced by time of in vitro cell cultivation, cell density affecting cell spreading and cell shape [105], intercolony contact or by addition of substances such as cyclic AMP, DMSO and others to cell cultures [75]. Ploidy. A controversy has been noticed with regard to ploidy of the cultured cells. Selection in vitro for diploid cell populations mostly due to their better attachment to glass substrates has been reported [36]. On the other hand, majority of breast tumour cell lines was reported to be aneuploid [36]. Although these cell lines were usually established from metastasis and pleural effusions, and even after mutagenizing treatment procedures [36], suggesting that the cells may be originally aneuploid, the possibility of the evolution to aneuploidy could not be excluded [35]. Thus, it was mentioned that even cells from normal breast may become aneuploid in culture (references in [36]). In a study comparing cell lines with corresponding tumour tissues, poor correlation was noted in ploidy indices [42]. 2.3. Cultured vs. uncultured cells Distinct cell behaviour was documented when comparing cultured with uncultured cells [19,22,24,44,57] as well as primary cell cultures with cell lines [106–108]. Follicular lymphoma cells purified from lymph nodes of patients had little tendency to die in vivo, but they underwent apoptosis in vitro and expression of death suppressor bcl-xL declined
ˇ B. Cunderlíková / Critical Reviews in Oncology/Hematology 85 (2013) 95–111
rapidly within one day in culture [109]. Downregulation of bcl-xL and mcl-1 occurred also in germinal center B cells freshly purified from tonsils after several hours in culture [109]. Several attempts have been made to compare cultured tumour cells or tumour cell lines with cells from biopsies of various tumour tissues [30,37,38,110–112] by genomic or proteomic analyses. The comparisons have been hampered by cell heterogeneity of tissue biopsies containing non-tumour cells [113], still, modifications of cells occurring in vitro when cultures were established from primary tumours were suggested as an alternative explanations [30,37,38]. For example, comparison of protein profiles assayed by two-dimensional (2D) electrophoresis of non-invasive lowgrade superficial bladder transition cell carcinomas and their primary cultures has shown that short-term culturing of the lesions resulted in the altered expression of several proteins [30]. Differences in the expression of high-molecular-weight tropomyosins were found by the same method also when comparing tumour cells extracted from breast carcinoma tissues, non-cancerous human breast lesions and a number of breast carcinoma cell lines [37]. Different expression of several genes was found when serum-starved cells from marrow samples of patients with acute myeloid leukemia (AML) were compared with AML cell lines [112]. Further study compared tumour cells directly harvested from primary breast tumours by differential trypsinization, early culture passages of such cells and immortal cell lines frequently used in cancer research. Gene expression profiles of tumour cells harvested from primary breast tumour tissue were more consistent with those of the corresponding tumour cell cultures than with immortal cell lines [114]. In a study comparing broad panel of cell lines with primary tumour samples enriched for at least 50% malignant cells it was demonstrated that around 30% genes were differentially expressed in cell lines [115]. All these data indicate possibility of cell modification due to culturing that needs to be taken into account when interpreting experimental results. In case that differences in protein and gene expressions between cultured and uncultured cells are due to modifications of the cultured cells and not due to presence of contaminating cells or heterogeneity of cell population, then the results of experiments performed with cultured cells may not be applicable for in vivo situation if altered genes or proteins are involved in the studied pathway or process1 . 3. Lack of relevant interactions in vitro In the majority cases, cell culture experiments utilize monocultures, i.e. monolayers or suspensions of cells of one type (Fig. 1). In sharp contrast, solid tumours are not 1
For an example from author’s experience, when results obtained from experiments using cell line cultures were not consistent with those from studies utilizing non-cultured ex vivo samples and in vivo models, please see [25]. Modifications of the cells during cell culturing and/or lack of components of an in vivo system may be responsible.
99
clones of cancer cells only. They are composed of multiple other cell types and ECM that constitute tumour stroma and together interact with the rest of the organism [116]. As a result, tumour behaviour is not dependent exclusively on the properties of the tumour comprising cancer cells, but it is affected by what happens outside them in the tumour microenvironment [117,118]. Several lines of evidence suggest that even though the initiating mutation in the cancers of epithelial origin usually occurs in the epithelium, it is the stroma that is involved in promotion, and in some cases even in initiation, of tumour progression [119]. The importance of the stroma of solid tumours has been recognized as summarized in several review papers [116,118,120]. However, behaviour of tumour cells is dependent on the stroma not only in solid tumours. The bone marrow microenvironment was suggested to contribute to drug resistance in variety of hematopoietic malignancies (for review see [121–123]). ECM, namely fibronectin [124–129], as well as stromal cells [130–135] are shown to be involved. 3.1. Extracellular matrix ECM produced in vitro. The ECM consists of multiple components, type and abundance of which depend on the stroma and epithelium within the specific tissue and interactions between them [108,136,137]. Therefore, although cells in vitro are able to produce their own ECM, this is likely to differ from the ECM that the cells sense in vivo, especially in the case of monocultures on plastic (e.g. [138]). Fibroblasts are believed to be the main source of ECM, however, other cell types are also capable of producing ECM components in vitro [67,71,136,139–143]. The ECM deposited by cell cultures may depend on cell type [67,139]. Importantly, while fibroblastic clone of spontaneously immortalized, nontumourigenic mouse mammary cell line in pure culture lacked deposited laminin on the cell surface, it was sporadically seen on the surface of the epithelial clone cultured alone. Co-culture of cells of the epithelial clone on a monolayer of cells of fibroblastic clone gave rise to a large increase in laminin deposition in contact areas between epithelial cell clusters and fibroblastic layer [144]. Likewise, deposition of ECM components in monocultures either of Sertoli or peritubular cells, was minimal in the absence of serum. It became appreciable in co-cultures of the two cell types [145]. In cultures of mammary epithelial cells alone, little or no extracellular fibrillar material could be detected, with some of it seen after two week incubation. However, it could be seen when mammary epithelial cells were grown on fibroblasts or adipocytes [146]. Moreover, co-culturing with breast adenocarcinoma cells was reported to enhance collagen synthesis by fibroblasts [147]. Thus, co-culturing of epithelial with mesenchymal cells considerably affects ECM production and deposition in cell culture. The presence of serum [145], duration of a culture [145], most probably due to increase in cell density and cellular contacts [63,136,148,149] and culture substrate [139] are other factors
100
ˇ B. Cunderlíková / Critical Reviews in Oncology/Hematology 85 (2013) 95–111
involved. For example, in vitro experiments showed that both the ultrastructure and ECM composition could be considerably altered when cells are plated on the matrix derived from the Engelbreth–Holm–Swarm mouse sarcoma as opposed to plastic, exemplified by production of collagen III and not of collagen I and remarkable changes in proteoglycan levels [100]. Deposition of collagen IV was found to be increased when mammary epithelial cells were cultured in collagen I matrices as compared to plastic [74,136]. In vivo ECM-mimicking cell cultures. With the intention to provide cultured cells with in vivo-like environment, several culture systems have been introduced (Fig. 1). They include matrices made of Matrigel, which consists of basement membrane components (procedures and modifications of this assay have been summarized in [150,151]), collagen I gels, ECM produced in vitro by fibroblasts [152–154] or extracted from tissues [155] as well as artificial materialbased matrices ([156–158], references in [159–161]). Use of these culture systems gave clear evidence for distinct cell behaviour and signalling, gene expression as well as response to therapies when compared to plastic (e.g. [162], reviewed in [161,163–167]). Due to distinct capabilities of individual integrins, mediating interactions between cells and the ECM, to interact with particular ECM ligands [168,169], the ECM composition ensures specific ECM-cell interaction and therefore cell behaviour [73,169–171]. These specific interactions are transmitted via the cytoskeleton to the nuclear matrix, where they affect gene expression ([49,95,97,172–174], reviewed in [175,176]), suggesting that the substrate used for cell culturing belongs to the factors responsible for gene expression in cultured cells. Consistent with this, dependence of cell behaviour on the type of ECM used for culture has been recognized (e.g. [59,61,65–67,155,177,178]) implying that if cell behaviour analyzed in vitro should aid in understanding of in vivo processes, correct type of ECM needs to be used [58,69,161,179], (Table 2). 3.2. Stromal cells Among stromal cells, fibroblasts, macrophages, neutrophils, mast cells, myeloid-derived suppressor cells, lymphocytes, endothelial cells, pericytes and adipocytes were reported to influence the fate of tumour cells [116–118]. Accordingly, they should be present in or available to culture model systems of tumour cells. There are obvious limitations to achieve this. Several cell types contributing to tumour stroma, such as bone marrow-derived cells, do not originally reside in the tumours. Instead, they are actively recruited to the tumour stroma by cancer cells [18,117,180,181]. Examples include tumour-associated macrophages, which promote tumour growth [18,116] or bone marrow-derived myeloid cells and endothelial progenitor cells, proposed in restoring tumour growth after treatment [18]. The other issue concerns choice of co-cultured cells with regard to their type, source and abundance. The properties of the
tumour-associated stromal cells differ from those of the same cell types within healthy body and even from their normal counterparts in adjacent normal tissue [116,182,183]. Normal and tumour-associated fibroblasts. Fibroblasts in particular seem to play a role in the initiation and progression of carcinomas [116,182,184], and accordingly most of the studies aimed to analyze their contribution to tumourigenesis. While fibroblasts from normal breast generally inhibit epithelial growth, fibroblasts from breast carcinomas can promote it [185,186]. Indeed, gene expression profiling identified many genes differently expressed in early passage cultures of normal breast fibroblasts as compared to fibroblasts from breast carcinomas [186]. Differences not only in mRNA but also in protein levels were reported [187]. Analysis of breast carcinoma-associated fibroblasts, their adjacent counterparts isolated from histologically noncancerous region of the same affected breast and fibroblasts isolated from normal breast tissue were shown to differ in various properties among each other, even fibroblasts isolated from noncancerous region of the affected breast were different from normal fibroblasts [182]. Not only tumour-derived fibroblasts seem to differ from normal ones. Differences in expression of fibroblast growth factor-2 and ability to induce increased epithelial cell proliferation and progesterone expression between carcinoma-associated fibroblasts derived from hormone independent as compared to hormonedependent mouse mammary tumours were reported [188]. In general, population of fibroblasts seems to be very heterogenous. Considerable variability in properties among fibroblasts derived from individual patients was noticed [86,185,186] and fibroblasts vary even among tissues and sites within the body [87,189–194]. Tissue source of fibroblasts was found to be critical in promoting the extent of tumour invasion, migration and the degree of tumour differentiation in the 3D culture model of esophageal squamous cell carcinoma [88]. While fetal skin fibroblasts supported well differentiated phenotype of EGFR, hTERT and p53transduced human esophageal epithelial cells, a tendency toward poorly differentiated phenotype and more aggressive invasion were observed in the presence of fetal esophageal fibroblasts. In contrast, human adult esophageal fibroblasts did not promote invasion of the epithelial cells [88]. It was also shown that different types of fibroblasts produced matrices of clearly distinct properties, with consequences for behaviour of cells cultured within them [57,59,152,195]. Presence of fetal esophageal fibroblasts in matrices composed of a collagen I: Matrigel mixtures resulted in greater increase in stiffness than in the case of fetal skin fibroblasts [88]. Experiments with 3D in vitro co-cultures documented importance of the ratio between epithelial cells to fibroblasts [86]. Even ratio of different types of fibroblasts influenced tumourigenesis. While pure TGF--responsive and -unresponsive stromal fibroblasts supported growth of normal prostate and precancerous lesions, respectively, only their 1:1 mixtures resulted in development of prostatic adenocarcinomas [194].
ˇ B. Cunderlíková / Critical Reviews in Oncology/Hematology 85 (2013) 95–111
Other stromal cells modified in tumours. Myoepithelial cells [183] and tumour-associated macrophages (references in [116]) may be other examples of stromal cells with modified properties within tumours. Furthermore, tumour-specific autologous cytotoxic T cells were proposed to be modified by factors within tumour microenvironment, leading to suppression of their proliferative capability [196]. Substances released by stromal cells. The finding of T cell modifications by factors within tumour microenvironment emphasizes importance of production of substances by stromal cells within tumour microenvironment. Importantly, expression of some of these substances may be lost in cultured cells [57]. Analysis of MMP expression, mostly by in situ hybridization, revealed that stromal cell expression of MMPs, as a response to the tumour, is in general more common than their expression by tumour cells [6,7,197]. Tumour cells, tumour-associated fibroblasts, endothelial cells and various cells of the myeloid and lymphoid classes have been shown to supply proteolytic enzymes within a tumour microenvironment (references in [8]). Substances released by stromal cells within tumour microenvironment mediate communication among cells, serve as signals to modify properties and behaviour of the cells and ECM, activate cells within tumour or induce recruitment of distally located cells to the tumour site (reviewed in [198]). MMPs may trigger the cleavage of cytokines, their receptors (references in [196]) and ECM proteins and consequently affect cellular signalling and functions. The cleavage of ECM proteins may even result in generating fragments with new functions [6,108]. Cell–cell as well as cell–ECM interactions may be mediated by direct contact or by released substances acting in a dynamic and reciprocal manner [19,95,199,200]. Cell–cell interactions. Trials to mimic cell–cell interactions by co-culturing different cell types proved that interactions between different cell types may influence cell behaviour with regard to morphological and functional differentiation, protein secretion, ECM deposition and basement membrane formation, activation of factors, cell growth and development, gene expression, etc. ([201] and references in Table 3), often in cell-type specific manner [137,200,203]. Co-culturing of various hematopoietic tumour primary cells and cell lines with bone marrow stromal cells provided with evidence that bone marrow stromal cells through soluble factors and direct cell contact were responsible for drug resistance and protection from apoptosis of myeloma [130,131], lymphoma [132,133] and leukaemia cells [134]. Upregulation of survival genes in cancer cells could be induced by their co-culturing with astrocytes, leading to resistance to number of chemotherapeutic agents [205]. The above summarized data document complexity of the conditions in vivo, which, in the case of cancer, have been replaced by tumour cell monocultures in majority cases. There are many factors involved that operate in a specific, dynamic and reciprocal way and neglecting any of them may shift the whole process to direction different from the one which is relevant in vivo*.
101
3.3. Evidence from animal studies supporting importance of tumour cell environment The importance of microenvironment for tumour cell behaviour has been proved by animal studies. When human mammary cancer cells premixed with either Matrigel, nulliparous or involution mammary matrices, were injected into nude mice, the ability of small tumours to metastasize out the mammary fat pad was found to be dependent on the source of the matrix [155]. Lung, liver and kidney metastases were increased when involution matrix was used. Furthermore, biological behaviour of tumours may be altered by the environment of the tumour cell growth or implantation site [210–213]. In general, the growth of tumour cells in the intraperitoneal cavity, the lymph node or the spleen was documented to lead to progression towards cell populations with a higher metastatic potential in different animal tumour systems [212]. Mammary cancer cell study. Injections of human mammary epithelial cell lines of increasing tumourigenic properties into mammary fat pad of nude mice resulted in increased tumour formation frequency as compared to subcutaneous (s.c.) injections [213]. Prostate cancer cell study. Prostate cancer cell line variants with different tumour growth and metastatic potential could be isolated based on cell ability to grow within the prostate or to produce lymph node metastases [211]. Leukemia-lymphoma study. Lymphomas, characterized by their localized involvement, could be transformed into leukaemia with systemic dissemination by the tissue microenvironment in which the neoplastic lymphocytes proliferated. Neoplastic lymphocytes capable of inducing leukaemia, proliferating as transplants in spleens and lymph nodes of allogeneic animals, retained the capacity to induce leukaemia with systemic manifestations when inoculated subcutaneously, but they lose this capacity when they proliferated at subcutaneous sites and regained it again when they proliferated intrasplenically [210,212]. Glioblastoma study. Data from animal study have revealed the potential of glioblastoma multiforme (GBM) to adapt to its changing microenvironment [214]. GBM tumours normally invade adjacent brain by infiltration of single cells into the brain parenchyma. In microenvironment lacking VEGF or MMP-9, this mode of invasion was found to be blocked and deep perivascular tumour invasion occurred instead [180,214]. Isograft vs. xenograft study. Furthermore, response to some treatments may depend on whether the tumour is induced in its natural anatomical site [215]. When isografts of rat mammary carcinoma in the Fischer rats were compared with xenografts of the same carcinoma and human mesothelioma in nude mice, the response to photodynamic therapy was different in each of them, for the same tumour in different hosts as well as for different tumours in the same host [23]. Some experimental evidence demonstrated that the organ microenvironment could regulate the level of gene
102
ˇ B. Cunderlíková / Critical Reviews in Oncology/Hematology 85 (2013) 95–111
Table 3 Cell–cell interactions studied in cell co-cultures. Studied cells
Co-cultured cells
Influenced cell behaviour/property
Reference
Primary luminal mammary epithelial cells, human Mammary epithelial cell clone, mouse Cultured mammary epithelial cells, mouse
Myoepithelial cells, human
Morphological differentiation, cell polarity
[66,67]
Fibroblastic cell clone, mouse
[144]
Cultured mammary epithelial cells, mouse Cultured endometrial epithelial cells, human
Adipocyte cell line, mouse
Morphological and functional differentiation, protein secretion, ECM protein deposition Morphological and functional differentiation, protein secretion, ECM deposition, basement membrane formation Functional differentiation, cell growth
[199]
Colorectal carcinoma cell lines, human Epithelial mammary tumour cell line, human Breast cancer cell line, human Lung cancer cell line, human Melanoma cell line, human Prostate cancer cell line, human Sertoli cells, rat
Mesenchymal cell strain, human
Morphological differentiation, ECM deposition, basement membrane formation, cell growth Morphological and functional differentiation
Normal skin fibroblasts, human
Tumour forming capacity
[184]
Astrocytes, mouse Astrocytes, mouse Astrocytes, mouse Macrophages, mouse Peritubular myoid cells, rat
[205] [205] [206] [207] [137] and references therein
Sertoli cells, rat Sertoli cells, rat Aortic endothelial cells, bovine
Testicular peritubular cells, rat Germ cells, rat Pericytes, smooth muscle cells, bovine Pericytes, smooth muscle cells, bovine Fibroblasts, human and mouse; retinal pigment epithelial cells, bovine; kidney epithelial cells, Madin–Darby canine Primary hepatocytes, rat (but not human) Mammary adenocarcinoma cell lines, human Stromal cell line, mouse
Protection from apoptosis, gene expression Protection from apoptosis, gene expression Protection from apoptosis IL-6 production increase Morphological and functional differentiation, protein secretion, basement membrane formation ECM deposition Functional differentiation TGF- activation Inhibition of cell proliferation
[208]
Capillary endothelial cells, bovine
Lung microvessel endothelial cells, rat Fibroblasts, human B cell progenitors, LSK cells, mouse Myeloma cell lines, human Primary lymphoma cells, lymphoma cell lines, human Primary leukemia cells, leukemic cell lines, human
Adipocyte cell line, preadipocyte cell line, mouse
Cultured endometrial stromal cells, human
Bone marrow stromal cells, human Bone marrow stromal cell line, human Bone marrow stromal cell line, human
expression. For example, highly metastatic human prostate cancer cells implanted in nude mice at an ectopic site s.c. expressed lower levels of metastasis-related genes than those implanted in an orthotopic site into the prostate [211]. The activity of collagenolytic enzymes was shown to be influenced by tumour implantation site also in the rabbit carcinoma model [216]. Not only the nature of the implantation site, but a number of other factors need to be considered when comparing different tumour models, including hematopoietic and immune host response. Consistently, significant variation in utilization of bone-marrow derived cells and bioavailability of MMP-9 between different tumour models was reported [214].
[146]
[204]
[202]
[145] [203] References in [70]
Cell growth stimulation
Capillary morphogenesis
[209]
Stimulation of ECM protein synthesis
[147]
Cell development, gene expression
[200]
Drug resistance, protection from apoptosis
[130,131]
Drug resistance, protection from apoptosis
[132,133]
Drug resistance, protection from apoptosis
[134]
3.4. Species specificity Even animal models, which, contrary to in vitro models, represent intact systems, may not fully overcome limitations recognized particularly in the fields of immunotherapy and gene therapy [217]. Due to the complexity and the unique nature of the host–tumour immunorelationship and the fact that the immune systems in humans and animals are different [218], animal models do not seem to fully mimic the biology of human patients with cancer. That not only immune systems differ between animals and humans has been documented in several review papers [219–223]. This aspect should be also considered in in vitro models.
ˇ B. Cunderlíková / Critical Reviews in Oncology/Hematology 85 (2013) 95–111
4. Lack of structural context in vitro Although deficient composition of cell’s microenvironment is the first factor that one considers with regard to in vitro conditions, the lack of the correct structural context is another limitation of the most usually used in vitro conditions. 4.1. Spatial organization Physiologically crucial molecules are often immobilized on insoluble structural elements within the cell and nucleus [96,172]. Likewise, ECMs serve as cellular scaffolds that provide structure to tissues, bind and sequester signalling molecules, affecting their local concentration and biological activity [19,79,172]. Moreover, the macromolecules of the ECM act also as ligands for adhesion receptors of the cells that transduce signals to the cell interior. It has been detected long time ago that the biochemical signalling resulting from the specific interactions between integrins and ECM components is coupled with morphological changes of the cells [73,174]. The impact of cell spreading and cell shape has been documented in cell growth, differentiation, nuclear morphology and gene expression [97,224–226]. The importance of structural context for cell behaviour is documented by the fact that providing the cells with correct biochemical microenvironment in the form of ECM protein-forming coatings does not necessarily lead to in vivo-like cell behaviour. Cell behaviour under such conditions, and even on attached as opposed to relaxed ECM matrices, is usually similar to that seen on plastic [22,54,55,69,171]. Consistently, not only the ECM composition determines the functional behaviour of the cells in the tissue (references in [95]), but the organization and biophysical properties of the ECM also play an important role (Table 2). 2D vs. 3D cultures. Numerous studies with primary cultures of epithelial cells of different types, fibroblasts as well as various cell lines document distinct behaviour of the cells in 3D as compared to 2D environments [100,108,202,227–237]. Importantly, phenotype dependence on spatial organization appears in transformed and tumour cells as well [69,202,227,232–234]. The modifications concern cell morphology, gene expression, protein production, deposition of ECM components, proliferation and functional differentiation. Critical importance of tissue architecture has been shown also in resistance of mammary epithelial cells to apoptosis. Formation of polarized 3D structures, occurring in 3D basement membrane cultures, conferred protection to apoptosis in both non-malignant and malignant mammary epithelial cells, while both cell types exhibited apoptotic responsiveness in 2D monolayer cultures [65]. Improved clonogenic survival after ␥-irradiation in 3D collagen gel as compared to 2D plastic conditions was shown for human non-immortalized fibroblast strain, while opposite result was found for hamster transformed cell line (references in [225]). Architecture of ECM fibres. The synthesis, deposition, posttranslation modification, but also the architecture of
103
the fibrillar collagen and ECM are altered in tumours [57,120,238,239] and various effects of ECM proteins were reported to be mediated by its architecture rather than solely by protein level [57,120]. The changed collagen organization may modify cell invasion by enabling cells to migrate along the collagen fibres or influence therapeutic efficacy by hindering diffusion of drugs through dense collagen networks (references in [116]). 4.2. ECM stiffness Evidence further suggests that even biophysical properties of the ECM, namely forces exerted through matrix stiffness provide information to the cells. Force can alter the structural properties of the cytoskeleton, which in turn can activate signalling events or induce binding of other structural proteins (references in [240]). Consistently, the changes in ECM stiffness affect cell morphology, integrin signalling, and the actin cytoskeleton to control the cell cycle [241] and physiological tissue stiffness acts as a cell-cycle inhibitor in mammary epithelial and vascular smooth muscle cells [157]. In general, ECM stiffness enhances cell growth and survival, influences differentiation and promotes migration [237,238,242] and could even direct lineage specification of stem cells (references in [243]). Increased ECM stiffness in tumours thus could influence treatment response (references in [237]) by eliciting diverse effects on cellular differentiation, gene expression, proliferation, survival and migration [120]. In conventional in vitro experiments, the cells are cultured on nondeformable substrates, which are far from to the elastic ECM that cells encounter in vivo [157], (Fig. 2). Milliprobe indentation and atomic force microscopy indicated elastic moduli of normal murine tissues such as isolated mammary glands, thoracic aortae, and femoral arteries ranging from 600 to 4300 Pa [157], skeletal muscles ∼12 kPa [244] or cartilage up to 1.3 MPa [245]. Stiffness scale for normal human solid tissues was reported to range from several hundreds of Pa for soft brain [246] to microstiffness in the order of MPa for cartilage and precalcified bone [242,245]. Unconfined compression analysis of normal and malignant breast tissues from MMTV-Her2/neu, Myc, and Ras transgenic mice revealed that normal mammary tissue was rather soft with elastic moduli as low as ∼167 Pa, whereas the stromal matrix adjacent to the transformed cells and the oncogene-induced tumour itself were stiffer [238], with average elastic moduli reaching values of ∼900 and ∼4000 Pa, respectively [237]. Different elastic moduli were reported for different tumour types. Murine carcinoma xenografts were shown to be the softest tumours with the lowest collagen contents compared to rigid murine glioblastomas and sarcomas [247]. Still, elastic moduli of plastic dishes and glass coverslips are several orders of magnitude higher, ∼109 to 1012 Pa [237,241]. Such a difference should be remembered as various cells, at least normal, show distinct responses over the substrate stiffness range that is relevant to that of in vivo tissues [243].
ˇ B. Cunderlíková / Critical Reviews in Oncology/Hematology 85 (2013) 95–111
104
Reviewers Andrea Sadlonova, Ph.D., Emory University, 1462 Clifton Road NE, DSB 405, Atlanta, GA 30322, United States. Elizabeth G.E., De Vries, M.D., Ph.D., University Medical Center Groningen, Department of Internal Medicine, Hanzeplein 1, NL-9713 RB Groningen, Netherlands. Galia Staneva, Ph.D., Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, BG-1113 Sofia, Bulgaria. Antoinette Wozniak, M.D., FACP, Wayne State University, Karmanos Cancer Institute, Department of HematologyOncology, 4100 John R, Detroit, MI 48201, United States.
Acknowledgements Due to space limitations, reviews have been cited throughout the article. I, therefore, apologize for the omission of many significant references. Helpful comments from reviewers are highly acknowledged. Many thanks to K.R.
References
Fig. 2. Stiffness of solid tissues and conventional culture supports.
5. Conclusion In order to interpret the results of in vitro experiments correctly, it is crucial to understand the physiological and pathological relevance of the experimental conditions. That monocultures in plastic dishes may not represent relevant in vitro models in cancer research has been acknowledged long time ago. Despite that, they still constitute a considerable part of current research. Complexity of cancer, however, necessitates work with more sophisticated culture systems that provide with in vivo-mimicking interactions and structural context (Table 2) as well as accompanying analyses of patient material and systems biology approaches. Apart from complex physiological aspects of tissues, such as vasculature, interstitial flows or local gradients, specificity of cell behaviour with regard to cell type and type of ECM needs to be considered when developing in vitro cancer cell models. This may require use of culture systems specifically generated for the studied condition/disease based on analyses of in vivo samples and choice of proper cellular and non-cellular components with in vivo corresponding properties in addition to standardized culture systems.
[1] Enrique AA, Gema PC, Jeronimo JC, Auxiliadora GE. Role of antiEGFR target therapy in colorectal carcinoma. Frontiers in Bioscience (Elite Edition) 2012;4:12–22. [2] Tsang RY, Finn RS. Beyond trastuzumab: novel therapeutic strategies in HER2-positive metastatic breast cancer. British Journal of Cancer 2012;106:6–13. [3] Suttorp M, Eckardt L, Tauer JT, Millot F. Management of chronic myeloid leukemia in childhood. Current Hematologic Malignancy Reports 2012;(March). [4] Laack E, Sauter G, Bokemeyer C. Lessons learnt from gefitinib and erlotinib: key insights into small-molecule EGFR-targeted kinase inhibitors in non-small cell lung cancer. Lung Cancer 2010;69:259–64. [5] Harris T. Gene and drug matrix for personalized cancer therapy. Nature Reviews Drug Discovery 2010;9:660–1. [6] Egeblad M, Werb Z. New functions for the matrix metalloproteinases in cancer progression. Nature Reviews Cancer 2002;2:161–74. [7] Nelson AR, Fingleton B, Rothenberg ML, Matrisian LM. Matrix metalloproteinases: biologic activity and clinical implications. Journal of Clinical Oncology 2000;18:1135–49. [8] Martin MD, Carter KJ, Jean-Philippe SR, et al. Effect of ablation or inhibition of stromal matrix metalloproteinase-9 on lung metastasis in a breast cancer model is dependent on genetic background. Cancer Research 2008;68:6251–9. [9] Knox JJ, Hotte SJ, Kollmannsberger C, Winquist E, Fisher B, Eisenhauer EA. Phase II study of triapine(R) in patients with metastatic renal cell carcinoma: a trial of the National Cancer Institute of Canada Clinical Trials Group (NCIC IND.161). Investigational New Drugs 2007;25:471–7. [10] Mackenzie MJ, Saltman D, Hirte H, et al. A phase II study of 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP) and gemcitabine in advanced pancreatic carcinoma. A trial of the Princess Margaret hospital phase II consortium. Investigational New Drugs 2007;25:553–8. [11] Gasparini G, Longo R, Torino F, et al. Is tailored therapy feasible in oncology? Critical Reviews in Oncology/Hematology 2006;57:70–101.
ˇ B. Cunderlíková / Critical Reviews in Oncology/Hematology 85 (2013) 95–111 [12] Nieto Y, Jones RB, Shpall EJ. Stem-cell transplantation for the treatment of advanced solid tumors. Springer Seminars in Immunopathology 2004;26:31–56. [13] Nieto Y, Shpall EJ. High dose chemotherapy for high-risk primary and metastatic breast cancer: is another look warranted? Current Opinion in Oncology 2009;21:150–7. [14] http://www.ornl.gov/sci/techresources/Human Genome/medicine/ genetherapy.shtml, modified June 11, 2009. [15] Kroeger KM, Muhammad AK, Baker GJ, et al. Gene therapy and virotherapy: novel therapeutic approaches for brain tumors. Discovery Medicine 2010;10:293–304. [16] Seufferlein T, Ahn J, Krndija D, et al. Tumor biology and cancer therapy—an evolving relationship. Cell Communication and Signalling 2009;7:19. [17] Richardson DR, Kalinowski DS, Lau S, et al. Cancer cell iron metabolism and the development of potent iron chelators. Biochimica et Biophysica Acta 2009;1790:702–17. [18] Ahn G-O, Brown JM. Influence of bone marrow-derived hematopoietic cells on the tumor response to radiotherapy: experimental models and clinical perspectives. Cell Cycle 2009;8:970–6. [19] Stoker AW, Streuli CH, Martins-Green M, Bissell MJ. Designer microenvironments for the analysis of cell and tissue function. Current Opinion in Cell Biology 1990;2:864–74. [20] Walling JM, Blackmore M, Hickman JA, Townsend KMS. Role of the extracellular matrix on the growth and differentiated phenotype of murine colonic adenocarcinoma cells in vitro. International Journal of Cancer 1991;47:776–83. [21] Mani SK, Julian J, Lampelo S, Glasser SR. Initiation and maintenance of in vitro decidualization are independent of hormonal sensitization in vivo. Biology of Reproduction 1992;47:785–99. [22] Streuli CH, Schmidhauser C, Kobrin M, Bissell MJ, Derynck R. Extracellular matrix regulates expression of the TGF-1 gene. Journal of Cell Biology 1993;120:253–60. [23] Gibson SL, Foster TH, Feins RH, et al. Effects of photodynamic therapy on xenografts of human mesothelioma and rat mammary carcinoma in nude mice. British Journal of Cancer 1994;69:473–81. [24] Vollmer G, Schneider MR. The rat endometrial adenocarcinoma cell line RUCA-I: a novel hormone-responsive in vivo/in vitro tumor model. The Journal of Steroid Biochemistry and Molecular Biology 1996;58:103–15. ˇ [25] Cunderlíková B, Vasoviˇc V, Sieber F, et al. Hexaminolevulinatemediated photodynamic purging of leukemia cells from bone marrow. Bone Marrow Transplantation 2010;45:1553–61. ˇ [26] Cunderlíková B, Vasoviˇc V, Sieber F, et al. Hexaminolevulinatemediated photodynamic purging of marrow grafts with murine breast carcinoma. Bone Marrow Transplantation 2011;46:1118–27. [27] Flinterman AE, Knol EF, van Ieperen-van Dijk AG, et al. Probiotics have a different immunomodulatory potential in vitro versus ex vivo upon oral administration in children with food allergy. International Archives of Allergy and Immunology 2007;143:237–44. [28] Paweletz CP, Charboneau L, Bichsel VE, et al. Reverse phase protein microarrays which capture disease progression show activation of pro-survival pathways at the cancer invasion front. Oncogene 2001;20:1981–9. [29] Watt FM. Cell culture models of differentiation. FASEB Journal 1991;5:287–94. [30] Celis A, Rasmussen HH, Celis P, et al. Short-term culturing of lowgrade superficial bladder transitional cell carcinomas leads to changes in the expression levels of several proteins involved in key cellular activities. Electrophoresis 1999;20:355–61. [31] Bayly A, French NJ, Dive C, Roberts RA. Non-genotoxic hepatocarcinogenesis in vitro: the FaO hepatoma line responds to peroxisome proliferators and retains the ability to undergo apoptosis. Journal of Cell Science 1993;104:307–15. [32] Birgersdotter A, Sandberg R, Ernberg I. Gene expression perturbation in vitro–a growing case for three-dimensional (3D) culture systems. Seminars in Cancer Biology 2005;15:405–12.
105
[33] Gaffney EV. A cell line (HBL-100) established from human breast milk. Cell and Tissue Research 1982;227:563–8. [34] Watkin H, Streuli CH. Adenoviral-mediated gene transfer in twodimensional and three-dimensional cultures of mammary epithelial cells. Methods in Cell Biology 2002;69:403–23. [35] Soule HD, Maloney TM, Wolman SR, et al. Isolation and characterization of a spontaneously immortalized human breast epithelial cell line, MCF-10. Cancer Research 1990;50:6075–86. [36] Wolman SR, Smith HS, Stampfer M, Hackett AJ. Growth of diploid cells from breast cancers. Cancer Genetics and Cytogenetics 1985;16:49–64. [37] Franzén B, Linder S, Uryu K, et al. Expression of tropomyosin isoforms in benign and malignant human breast lesions. British Journal of Cancer 1996;73:909–13. [38] Li H-R, Wang-Rodriguez J, Nair TM, et al. Two-dimensional transcriptome profiling: identification of messenger RNA isoform signatures in prostate cancer from archived paraffin-embedded cancer specimens. Cancer Research 2006;66:4079–88. [39] Taylor-Papadimitriou J, Stampfer M, Bartek J, et al. Keratin expression in human mammary epithelial cells cultured from normal and malignant tissue: relation to in vivo phenotypes and influence of medium. Journal of Cell Science 1989;94:403–13. [40] Fidler IJ, Kripke ML. Metastasis results from preexisting variant cells within malignant tumor. Science 1977;197:893–5. [41] Sugarbaker EV, Cohen AM. Altered antigenicity in spontaneous pulmonary metastases from an antigenic murine sarcoma. Surgery 1972;72:155–61. [42] Wistuba II, Behrens C, Milchgrub S, et al. Comparison of features of human breast cancer cell lines and their corresponding tumors. Clinical Cancer Research 1998;4:2931–8. [43] Wistuba II, Bryant D, Behrens C, et al. Comparison of features of human lung cancer cell lines and their corresponding tumors. Clinical Cancer Research 1999;5:991–1000. [44] Watanabe TK, Hansen LJ, Reddy NK, Kanwar YS, Reddy JK. Differentiation of pancreatic acinar carcinoma cells cultured on rat testicular seminiferous tubular basement membranes. Cancer Research 1984;44:5361–8. [45] Blaschke RJ, Howlett AR, Desprez P-Y, Petersen OW, Bissell MJ. Cell differentiation by extracellular matrix components. Methods in Enzymology 1994;245:535–56. [46] 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. Proceedings of the National Academy of Sciences of the United States of America 1992;89:9064–8. [47] Gazdar AF, Kurvari V, Virmani A, et al. Characterization of paired tumor and non-tumor cell lines established from patients with breast cancer. International Journal of Cancer 1998;78:766–74. [48] Drexler HG. Review of alterations of the cyclin-dependent kinase inhibitor INK4 family genes p15, p16, p18 and p19 in human leukemia–lymphoma cells. Leukemia 1998;12:845–59. [49] Boyd JA, Rinehart Jr CA, Walton LA, Siegal GP, Kaufman DG. Ultrastructural characterization of two new human endometrial carcinoma cell lines and normal human endometrial epithelial cells cultured on extracellular matrix. In Vitro Cellular and Developmental Biology 1990;26:701–8. [50] Vollmer G, Ellerbrake N, Hopert A-C, et al. Extracellular matrix induces hormone responsiveness and differentiation in RUCA-I rat endometrial adenocarcinoma cells. The Journal of Steroid Biochemistry and Molecular Biology 1995;52:259–69. [51] Schatz F, Gordon RE, Laufer N, Gurpid E. Culture of human endometrial cells under polarizing conditions. Differentiation 1990;42:184–90. [52] Boudreau N, Werb Z, Bissell MJ. Suppression of apoptosis by basement membrane requires three-dimensional tissue organization and withdrawal from the cell cycle. Proceedings of the National Academy of Sciences of the United States of America 1996;93:3509–13.
106
ˇ B. Cunderlíková / Critical Reviews in Oncology/Hematology 85 (2013) 95–111
[53] Kirkland SC. Type I collagen inhibits differentiation and promotes a stem cell-like phenotype in human colorectal carcinoma cells. British Journal of Cancer 2009;101:320–6. [54] Lelièvre S, Weaver VM, Bissell MJ. Extracellular matrix signaling from the cellular membrane skeleton to the nuclear skeleton: a model of gene regulation. Recent Progress in Hormone Research 1996;51:417–32. [55] Lin CQ, Bissell MJ. Multi-faceted regulation of cell differentiation by extracellular matrix. FASEB Journal 1993;7:737–43. [56] Lin CQ, Dempsey PJ, Coffey RJ, Bissell MJ. Extracellular matrix regulates whey acidic protein gene expression by suppression of TGF-␣ in mouse mammary epithelial cells: studies in culture and in transgenic mice. Journal of Cell Biology 1995;129: 1115–26. [57] Lee H-O, Mullis SR, Franco-Barraza J, et al. FAP-overexpressing fibroblasts produce an extracellular matrix that enhances invasive velocity and directionality of pancreatic cancer cells. BMC Cancer 2011;11:245. [58] Serebriiskii I, Castelló-Cros R, Lamb A, Golemis EA, Cukierman E. Fibroblast-derived 3D matrix differentially regulates the growth and drug-responsiveness of human cancer cells. Matrix Biology 2008;27:573–85. [59] Castelló-Cros R, Khan DR, Simons J, Valianou M, Cukierman E. Staged stromal extracellular 3D matrices differentially regulate breast cancer cell responses through PI3K and beta 1-integrins. BMC Cancer 2009;9:94. [60] 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. Molecular Oncology 2007;1:84–96. [61] LaBarge MA, Nelson CM, Villadsen R, et al. Human mammary progenitor cell fate decisions are products of interactions with combinatorial microenvironments. Integrative Biology (Cambridge) 2009;1:70–9. [62] Nam J-M, Onodera Y, Bissell MJ, Park CC. Breast cancer cells in three-dimensional culture display an enhanced radioresponse after coordinate targeting of integrin ␣51 and fibronectin. Cancer Research 2010;70:5238–48. [63] Novaro V, Roskelley CD, Bissell MJ. Collagen-IV and laminin1 regulate estrogen receptor ␣ expression and function in mouse mammary epithelial cells. Journal of Cell Science 2003;116: 2975–86. [64] Weigelt B, Lo AT, Park CC, Gray JW, Bissell MJ. HER2 signaling pathway activation and response of breast cancer cells to HER2targeting agents is dependent strongly on the 3D microenvironment. Breast Cancer Research and Treatment 2010;122:35–43. [65] Weaver VM, Lelièvre S, Lakins JN, et al. 4 integrin-dependent formation of polarized three-dimensional architecture confers resistance to apoptosis in normal and malignant mammary epithelium. Cancer Cell 2002;2:205–16. [66] Gudjonsson T, Rønnov-Jessen L, Villadsen R, Bissell MJ, Petersen OW. To create the correct microenvironment: three-dimensional heterotypic collagen assays for human breast epithelial morphogenesis and neoplasia. Methods 2003;30:247–55. [67] Gudjonsson T, Rønnov-Jessen L, Villadsen R, et al. Normal and tumor-derived myoepithelial cells differ in their ability to interact with luminal breast epithelial cells for polarity and basement membrane deposition. Journal of Cell Science 2002;115:39–50. [68] Spancake KM, Anderson CB, Weaver VM, et al. E7-transduced human breast epithelial cells show partial differentiation in threedimensional culture. Cancer Research 1999;59:6042–5. [69] Wozniak MA, Keely PJ. Use of three-dimensional collagen gels to study mechanotransduction in T47D breast epithelial cells. Biological Procedures Online 2005;7:144–61. [70] Arrick BA, Lopez AR, Elfman F, et al. Altered metabolic and adhesive properties and increased tumorigenesis associated with increased expression of transforming growth factor 1. Journal of Cell Biology 1992;118:715–26.
[71] Adams JC, Clelland JD, Collett GDM, et al. Cell-matrix adhesions differentially regulate fascin phosphorylation. Molecular Biology of the Cell 1999;10:4177–90. [72] Glasser SR, Julian J, Decker GL, Tang J-P, Carson DD. Development of morphological and functional polarity in primary cutlures of immature rat uterine epithelial cells. Journal of Cell Biology 1988;107:2409–23. [73] Streuli CH, Gilmore AP. Adhesion-mediated signaling in the regulation of mammary epithelial cell survival. Journal of Mammary Gland Biology and Neoplasia 1999;4:183–91. [74] David G, Nusgens B, van der Schueren B, et al. Collagen metabolism and basement membrane formation in cultures of mouse mammary epithelial cells. Induction of ‘assembly’ on fibrillar type I collagen substrata. Experimental Cell Research 1987;170:402–16. [75] Brouty-Boyé D, Gresser I, Baldwin C. Reversion of the transformed phenotype to the parental phenotype by subcultivation of X-ray-transformed C3H/10T1/2 cells at low cell density. International Journal of Cancer 1979;24:253–60. [76] Trask DK, Band V, Zajchowski DA, et al. Keratins as markers that distinguish normal and tumor-derived mammary epithelial cells. Proceedings of the National Academy of Sciences of the United States of America 1990;87:2319–23. [77] Treeck O, Strunck E, Vollmer G. A novel basement membraneinduced gene identified in the human endometrial adenocarcinoma cell line HEC1B. FEBS Letters 1998;425:426–30. [78] Strunck E, Hopert A-C, Vollmer G. Basement membrane regulates gene expression in HEC1B(L) endometrial adenocarcinoma cells. Biochemical and Biophysical Research Communications 1996;221:346–50. [79] Streuli CH. Extracellular matrix and gene expression in mammary epithelium. Seminars in Cell Biology 1993;4:203–12. [80] Satyaswaroop PG, Tabibzadeh SS. Extracellular matrix and the patterns of differentiation of human endometrial carcinomas in vitro and in vivo. Cancer Research 1991;51:5661–6. [81] Eppenberger ME, Hauser I, Baechi T, et al. Immunocytochemical analysis of the regeneration of myofibrils in long-term cultures of adult cardiomyocytes of the rat. Developmental Biology 1988;130: 1–15. [82] Eppenberger HM, Hertig C, Eppenberger- Eberhardt M. Adult rat cardiomyocytes in culture. A model system to study the plasticity of the differentiated cardiac phenotype at the molecular and cellular levels. Trends in Cardiovascular Medicine 1994;4: 187–93. [83] Mitcheson JS, Hancox JC, Levi AJ. Cultured adult cardiac myocytes: future applications, culture methods, morphological and electrophysiological properties. Cardiovascular Research 1998;39:280–300. [84] Franke WW, Schmid E, Winter S, Osborn M, Weber K. Widespread occurrence of intermediate-sized filaments of the vimentin-type in cultured cells from diverse vertebrates. Experimental Cell Research 1979;123:25–46. [85] Sommers CL, Walker-Jones D, Heckford SE, et al. Vimentin rather than keratin expression in some hormone-independent breast cancer cell lines and in oncogene-transformed mammary epithelial cells. Cancer Research 1989;49:4258–63. [86] Sadlonova A, Novak Z, Johnson MR, et al. Breast fibroblasts modulate epithelial cell proliferation in three-dimensional in vitro co-culture. Breast Cancer Research 2005;7:R46–59. [87] Jackson II RS, Franco OE, Bhowmick NA. Gene targeting to the stroma of the prostate and bone. Differentiation 2008;76:606–23. [88] Okawa T, Michaylira CZ, Kalabis J, et al. The functional interplay between EGFR overexpression, hTERT activation, and p53 mutation in esophageal epithelial cells with activation of stromal fibroblasts induces tumor development, invasion, and differentiation. Genes and Development 2007;21:2788–803. [89] Villadsen R, Fridriksdottir AJ, Rønnov-Jessen L, et al. Evidence for a stem cell hierarchy in the adult human breast. Journal of Cell Biology 2007;177:87–101.
ˇ B. Cunderlíková / Critical Reviews in Oncology/Hematology 85 (2013) 95–111 [90] Gudjonsson T, Villadsen R, Nielsen HL, et al. Isolation, immortalization, and characterization of a human breast epithelial cell line with stem cell properties. Genes and Development 2002;16:693–706. [91] Stingl J, Eaves CJ, Zandieh I, Emerman JT. Characterization of bipotent mammary epithelial progenitor cells in normal adult human breast tissue. Breast Cancer Research and Treatment 2001;67:93–109. [92] Péchoux C, Gudjonsson T, Ronnov-Jessen L, Bissell MJ, Petersen OW. Human mammary luminal cells contain progenitors to myoepithelial cells. Developmental Biology 1999;206:88–99. [93] Ao M, Williams K, Bhowmick NA, Hayward SW. Transforming growth factor- promotes invasion in tumorigenic but not in nontumorigenic human prostatic epithelial cells. Cancer Research 2006;66:8007–16. [94] Borgen EF. Detection of disseminated tumour cells in the bone marrow of breast carvinoma patients. Thesis. Faculty of Medicine, University of Oslo; 2007. [95] Weaver VM, Roskelley CD. Extracellular matrix: the central regulator of cell and tissue homeostasis. Trends in Cell Biology 1997;7: 40–2. [96] Ingber DE. The riddle of morphogenesis: a question of solution chemistry or molecular cell engineering? Minireview. Cell 1993;75:1249–52. [97] Lelièvre SA. Contributions of extracellular matrix signaling and tissue architecture to nuclear mechanisms and spatial organization of gene expression control. Biochimica et Biophysica Acta 2009;1790:925–35. [98] Brooks SC, Locke ER, Soule HD. Estrogen receptor in a human cell line (MCF-7) from breast carcinoma. Journal of Biological Chemistry 1973;248:6251–3. [99] Paine TM, Soule HD, Pauley RJ, Dawson PJ. Characterization of epithelial phenotypes in mortal and immortal human breast cells. International Journal of Cancer 1992;50:463–73. [100] Friedman SL, Roll FJ, Boyles J, Arenson DM, Bissell DM. Maintenance of differentiated phenotype of cultured rat hepatic lipocytes by basement membrane matrix. Journal of Biological Chemistry 1989;264:10756–62. [101] Song SH, Wientjes MG, Gan Y, Au JL-S. Fibroblast growth factors: an epigenetic mechanism of broad spectrum resistance to anticancer drugs. Proceedings of the National Academy of Sciences 2000;97:8658–63. [102] Masters JRW. Human cancer cell lines: fact and fantasy. Nature Reviews Molecular Cell Biology 2000;1:233–6. [103] Roschke AV, Tonon G, Gehlhaus KS, et al. Karyotypic complexity of the NCI-60 drug screenin panel. Cancer Research 2003;63:8634–47. [104] Rabinowitz Z, Sachs L. The formation of variants with a reversion of properties of transformed cells. VI. Stability of the reversed state. International Journal of Cancer 1972;9:334–43. [105] Brouty-Boyé D, Tucker RW, Folkman J. Transformed and neoplastic phenotype: reversibility during culture by cell density and cell shape. International Journal of Cancer 1980;26:501–7. [106] Streuli CH, Bailey N, Bissell MJ. Control of mammary epithelial differentiation: basement membrane induces tissue-specific gene expression in the absence of cell-cell interaction and morphological polarity. Journal of Cell Biology 1991;115:1383–95. [107] Franza Jr BR, Maruyama K, Garrels JI, Ruley HE. In vitro establishment is not a sufficient prerequisite for transformation by activated Ras oncogenes. Cell 1986;44:409–16. [108] Katz E, Streuli CH. The extracellular matrix as an adhesion checkpoint for mammary epithelial function. International Journal of Biochemistry and Cell Biology 2007;39:715–26. [109] Ghia P, Boussiotis VA, Schultze JL, et al. Unbalanced expression of bcl-2 family proteins in follicular lymphoma: contribution of CD40 signaling in promoting survival. Blood 1998;91:244–51. [110] Perou CM, Jeffrey SS, van de Rijn M, et al. Distinctive gene expression patterns in human mammary epithelial cells and breast cancers. Proceedings of the National Academy of Sciences of the United States of America 1999;96:9212–7.
107
[111] Alon U, Barkai N, Notterman DA, et al. Broad patterns of gene expression revealed by clustering analysis of tumor and normal colon tissues probed by oligonucleotide arrays. Proceedings of the National Academy of Sciences of the United States of America 1999;96:6745–50. [112] Court EL, Smith MA, Avent ND, et al. DNA microarray screening of differential gene expression in bone marrow samples from AML, non-AML patients and AML cell lines. Leukemia Research 2004;28:743–53. [113] Celis JE, Kruhøffer M, Gromova I, et al. Gene expression profiling: monitoring transcription and translation products using DNA microarrays and proteomics. FEBS Letters 2000;480:2–16. [114] Dairkee SH, Ji Y, Ben Y, et al. A molecular ˇısignatureˇıof primary breast cancer cultures; patterns resembling tumor tissue. BMC Genomics 2004;5:47. [115] Sandberg R, Ernberg I. The molecular portrait of in vitro growth by meta-analysis of gene-expression profiles. Genome Biology 2005;6:R65. [116] Egeblad M, Nakasone ES, Werb Z. Tumors as organs: complex tissues that interface with the entire organism. Developmental Cell 2010;18:884–901. [117] Jodele S, Chantrain CF, Blavier L, et al. The contribution of bone marrow-derived cells to the tumor vasculature in neuroblastoma is matrix metalloproteinase-9 dependent. Cancer Research 2005;65:3200–8. [118] Littlepage LE, Egeblad M, Werb Z. Coevolution of cancer and stromal cellular responses. Cancer Cell 2005;7:499–500. [119] Sternlicht MD, Werb Z. How matrix metalloproteinases regulate cell behaviour. Annual Review of Cell and Developmental Biology 2001;17:463–516. [120] Egeblad M, Rasch MG, Weaver VM. Dynamic interplay between the collagen scaffold and tumor evolution. Current Opinion in Cell Biology 2010;22:697–706. [121] Shain KH, Dalton WS. Cell adhesion is a key determinant in de novo multidrug resistance (MDR): new targets for the prevention of acquired MDR. Molecular Cancer Therapeutics 2001;1:69–78. [122] Hazlehurst LA, Landowski TH, Dalton WS. Role of the tumor microenvironment in mediating de novo resistance to drugs and physiological mediators of cell death. Oncogene 2003;22:7396–402. [123] Meads MB, Hazlehurst LA, Dalton WS. The bone marrow microenvironment as a tumor sanctuary and contributor to drug resistance. Clinical Cancer Research 2008;14:2519–26. [124] Damiano JS, Hazlehurst LA, Dalton WS. Cell adhesion-mediated drug resistance (CAM-DR) protects the K562 chronic myelogenous leukemia cell line from apoptosis induced by BCR/ABL inhibition, cytotoxic drugs, and ␥-irradiation. Leukemia 2001;15:1232–9. [125] Hazlehurst LA, Damiano JS, Buyuksal I, Pledger WJ, Dalton WS. Adhesion to fibronectin via 1 integrins regulates p27kip1 levels and contributes to cell adhesion mediated drug resistance (CAM-DR). Oncogene 2000;19:4319–27. [126] Hazlehurst LA, Argilagos RF, Emmons M, et al. Cell adhesion to fibronectin (CAM-DR) influences acquired mitoxantrone resistance in U937 cells. Cancer Research 2006;66:2338–45. [127] Hazlehurst LA, Argilagos RF, Dalton WS. 1 integrin mediated adhesion increases Bim protein degradation and contributes to drug resistance in leukaemia cells. British Journal of Haematology 2006;136:269–75. [128] Shain KH, Yarde DN, Meads MB, et al. 1 integrin adhesion enhances IL-6-mediated STAT3 signaling in myeloma cells: implications for microenvironment influence on tumor survival and proliferation. Cancer Research 2009;69:1009–15. [129] Damiano JS, Cress AE, Hazlehurst LA, Shtil AA, Dalton WS. Cell adhesion mediated drug resistance (CAM-DR): role of integrins and resistance to apoptosis in human myeloma cell lines. Blood 1999;93:1658–67. [130] Nefedova Y, Cheng P, Alsina M, Dalton WS, Gabrilovich DI. Involvement of notch-1 signaling in bone marrow stroma-mediated de novo
108
[131]
[132]
[133]
[134]
[135]
[136]
[137]
[138]
[139]
[140]
[141]
[142]
[143]
[144]
[145]
[146]
[147]
[148]
ˇ B. Cunderlíková / Critical Reviews in Oncology/Hematology 85 (2013) 95–111 drug resistance of myeloma and other malignant lymphoid cell lines. Blood 2004;103:3503–10. Nefedova Y, Landowski TH, Dalton WS. Bone marrow stromalderived soluble factors and direct cell contact contribute to de novo drug resistance of myeloma cells by distinct mechanisms. Leukemia 2003;17:1175–82. Lwin T, Hazlehurst LA, Li Z, et al. Bone marrow stromal cells prevent apoptosis of lymphoma cells by upregulation of anti-apoptotic proteins associated with activation of NF-jB (RelB/p52) in nonHodgkin’s lymphoma cells. Leukemia 2007;21:1521–31. Lwin T, Crespo LA, Wu A, et al. Lymphoma cell adhesion-induced expression of B cell-activating factor of the TNF family in bone marrow stromal cells protects non-Hodgkin’s B lymphoma cells from apoptosis. Leukemia 2009;23:170–7. Garrido SM, Appelbaum FR, Willman CL, Banker DE. Acute myeloid leukemia cells are protected from spontaneous and drug-induced apoptosis by direct contact with a human bone marrow stromal cell line (HS-5). Experimental Hematology 2001;29:448–57. Bewry NN, Nair RR, Emmons MF, Boulware D, Pinilla-Ibarz J, Hazlehurst LA. Stat3 contributes to resistance toward BCR-ABL inhibitors in a bone marrow microenvironment model of drug resistance. Molecular Cancer Therapeutics 2008;7:3169–75. Streuli CH, Bissell MJ. Expression of extracellular matrix components is regulated by substratum. Journal of Cell Biology 1990;110:1405–15. Janecki A, Steinberger A. Vectorial secretion of transferrin and androgen binding protein in sertoli cell cultures: effect of extracellular matrix, peritubular myoid cells and medium composition. Molecular and Cellular Endocrinology 1987;52:125–35. Stamatoglou SC, Colin Hughes R, Lindahl U. Rat hepatocytes in serum-free primary culture elaborate an extensive extracellular matrix containing fibrin and fibronectin. Journal of Cell Biology 1987;105:2417–25. Gospodarowicz D, Greenburg G, Birdwell CR. Determination of cellular shape by the extracellular matrix and its correlation with the control of cellular growth. Cancer Research 1978;38:4155–71. Kolachala VL, Bajaj R, Wang L, et al. Epithelial-derived fibronectin expression, signalling, and function in intestinal inflammation. Journal of Biological Chemistry 2007;282:32965–73. Powers MJ, Griffith LG. Adhesion-guided in vitro morphogenesis in pure and mixed cell cultures. Microscopy Res Tech 1998;43: 379–84. Matsui R, Goldstein RH, Mihal K, Brody JS, Steele MP, Fine A. Type I collagen formation in rat type II alveolar cells immortalized by viral gene products. Thorax 1994;49:201–6. Xu Q, Norman JT, Shrivastav S, Lucio-Cazana J, Kopp JB. In vitro models of TGF--induced fibrosis suitable for high-throughput screening of antifibrotic agents. American Journal of Physiology Renal Physiology 2007;293:F631–40. Reichmann E, Ball R, Groner B, Friis RR. New mammary epithelial and fibroblastic cell clones in coculture form structures competent to differentiate functionally. Journal of Cell Biology 1989;108: 1127–38. Skinner MK, Tung PS, Fritz IB. Cooperativity between sertoli cells and testicular peritubular cells in the production and deposition of extracellular matrix components. Journal of Cell Biology 1985;100:1941–7. Wiens D, Park CS, Stockdale FE. Milk protein expression and ductal morphogenesis in the mammary gland in vitro: hormone-dependent and -independent phases of adipocyte-mammary epithelial cell interaction. Developmental Biology 1987;120:245–58. Noël A, Munaut C, Boulvain A, et al. Modulation of collagen and fibronectin synthesis in fibroblasts by normal and malignant cells. Journal of Cellular Biochemistry 1992;48:150–61. Dhawan J, Farmer SR. Regulation of ␣1(I)-collagen gene expression in response to cell adhesion in Swiss 3T3 fibroblasts. Journal of Biological Chemistry 1990;265:9015–21.
[149] Dhawan J, Lichtler AC, Rowe DW, Farmer SR. Cell adhesion regulates pro-␣1(I) collagen mRNA stability and transcription in mouse fibroblasts. Journal of Biological Chemistry 1991;266:8470–5. [150] Lee GY, Kenny PA, Lee EH, Bissell MJ. Three-dimensional culture models of normal and malignant breast epithelial cells. Nature Methods 2007;4:359–65. [151] Debnath J, Muthuswamy SK, Brugge JS. Morphogenesis and oncogenesis of MCF-10A mammary epithelial acini grown in three-dimensional basement membrane cultures. Methods 2003;30: 256–68. [152] Amantangelo MD, Bassi DE, Klein-Szanto AJP, Cukierman E. Stroma-derived three-dimensional matrices are necessary and sufficient to promote desmoplastic differentiation of normal fibroblasts. American Journal of Pathology 2005;167:475–88. [153] Beacham DA, Amantangelo MD, Cukierman E. Preparation of extracellular matrices produced by cultured and primary fibroblasts. Current Protocols in Cell Biology 2006 [Chapter 10:10.9.1-21]. [154] Castelló-Cros R, Cukierman E. Stromagenesis during tumorigenesis: characterization of tumor-associated fibroblasts and stroma-derived 3D matrices. Methods in Molecular Biology 2009;522:275–305. [155] McDaniel SM, Rumer KK, Biroc SL, et al. Remodeling of the mammary microenvironment after lactation promotes breast tumor cell metastasis. American Journal of Pathology 2006;168:608–20. [156] Williams CM, Mehta G, Peyton SR, Zeiger AS, Van Vliet KJ, Griffith LG. Autocrine-controlled formation and function of tissue-like aggregates by primary hepatocytes in micropatterned hydrogel arrays. Tissue Engineering A 2011;17:1055–68. [157] Klein EA, Yin L, Kothapalli D, et al. Cell-cycle control by physiological matrix elasticity and in vivo tissue stiffening. Current Biology 2009;19:1511–8. [158] Fischbach C, Chen R, Matsumoto T, et al. Engineering tumors with 3D scaffolds. Nature Methods 2007;4:855–60. [159] Griffith LG, Swartz MA. Capturing complex 3D tissue physiology in vitro. Nature Reviews Molecular Cell Biology 2006;7:211–24. [160] Verbridge SS, Chandler EM, Fischbach C. Tissue-engineered threedimensional tumor models to study tumor angiogenesis. Tissue Engineering A 2010;6:2147–52. [161] Yamada KM, Cukierman E. Modeling tissue morphogenesis and cancer in 3D. Cell 2007;130:601–10. [162] Birgersdotter A, Baumforth KRN, Porwit A, et al. Three-dimensional culturing of the Hodgkin lymphoma cell-line L1236 induces a HL tissue-like gene expression pattern. Leukemia and Lymphoma 2007;48:2042–53. [163] Cukierman E, Pankov R, Stevens DR, Yamada KM. Taking cellmatrix adhesions to the third dimension. Science 2001;294:1708–12. [164] Cukierman E, Pankov R, Yamada KM. Cell interactions with three-dimensional matrices. Current Opinion in Cell Biology 2002;14:633–9. [165] Cukierman E, Bassi DE. Physico-mechanical aspects of extracellular matrix influences on tumorigenic behaviours. Seminars in Cancer Biology 2010;20:139–45. [166] Bissell MJ, Radisky DC, Rizki A. The organizing principle: microenvironmental influences in the normal and malignant breast. Differentiation 2002;70:537–46. [167] Bissell MJ. Modelling molecular mechanisms of breast cancer and invasion: lessons from the normal gland. Biochemical Society Transactions 2007;35:18–22. [168] Klein CE, Dressel D, Steinmayer T, et al. Integrin ␣21 is upregulated in fibroblasts and highly aggressive melanoma cells in three-dimensional collagen lattices and mediates the reorganization of collagen I fibrils. Journal of Cell Biology 1991;115:1427–36. [169] Juliano RL, Haskill S. Signal transduction from the extracellular matrix. Journal of Cell Biology 1993;120:577–85. [170] Howlett AR, Bailey N, Damsky C, Petersen OW, Bissell MJ. Cellular growth and survival are mediated by 1 integrins in normal human breast epithelium but not in breast carcinoma. Journal of Cell Science 1995;108:1945–57.
ˇ B. Cunderlíková / Critical Reviews in Oncology/Hematology 85 (2013) 95–111 [171] Wozniak MA, Modzelewska K, Kwong L, Keely PJ. Focal adhesion regulation of cell behavior. Biochimica et Biophysica Acta 2004;1692:103–19. [172] Adams JC, Watt FM. Regulation of development and differentiation by the extracellular matrix. Development 1993;117:1183–98. [173] Aggeler J, Ward J, Blackie LM, et al. Cytodifferentiation of mouse mammary epithelial cells cultured on a reconstituted basement membrane reveals striking similarities to development in vivo. Journal of Cell Science 1991;99:407–17. [174] Boudreau N, Myers C, Bissell MJ. From laminin to lamin: regulation of tissue-specific gene expression by the ECM. Trends in Cell Biology 1995;5:1–4. [175] Spencer V, Xu R, Bissell MJ. Extracellular matrix, nuclear and chromatin structure, and gene expression in normal tissues and malignant tumors: a work in progress. Advances in Cancer Research 2007;97:275–94. [176] Spencer V, Xu R, Bissell MJ. Gene expression in the third dimension: the ECM-nucleus connection. Journal of Mammary Gland Biology and Neoplasia 2010;15:65–71. [177] Maniotis AJ, Valyi-Nagy K, Karavitis J, et al. Chromatin organization measured by AluI restriction enzyme changes with malignancy and is regulated by the extracellular matrix and the cytoskeleton. American Journal of Pathology 2005;166:1187–203. [178] Provenzano PP, Inman DR, Eliceiri KW, Keely PJ. Matrix densityinduced mechanoregulation of breast cell phenotype, signaling and gene expression through a FAK–ERK linkage. Oncogene 2009;28:4326–43. [179] Anders M, Hansen R, Ding R-X, et al. Disruption of 3D tissue integrity facilitates adenovirus infection by deregulating the coxsackievirus and adenovirus receptor. Proceedings of the National Academy of Sciences 2003;100:1943–8. [180] Du R, Lu KV, Petritsch C, et al. HIF1␣ induces the recruitment of bone marrow-derived vascular modulatory cells to regulate tumor angiogenesis and invasion. Cancer Cell 2008;13:206–20. [181] Ahn G-O, Brown JM. Matrix metalloproteinase-9 is required for tumor vasculogenesis but not for angiogenesis: role of bone marrowderived myelomonocytic cells. Cancer Cell 2008;13:193–205. [182] Hawsawi NM, Ghebeh H, Hendrayani S-F, et al. Breast carcinoma-associated fibroblasts and their counterparts display neoplastic-specific changes. Cancer Research 2008;68:2717–25. [183] Allinen M, Beroukhim R, Cai L, et al. Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell 2004;6:17–32. [184] Noël A, Hajitou A, LˇıHoir C, et al. Inhibition of stromal matrix metalloproteinases: effects on breast-tumor promotion by fibroblasts. International Journal of Cancer 1998;76:267–73. [185] Sadlonova A, Mukherjee S, Bowe DB, et al. Human breast fibroblasts inhibit growth of the MCF10AT xenograft model of proliferative breast disease. American Journal of Pathology 2007;170: 1064–76. [186] Sadlonova A, Bowe DB, Novak Z, et al. Identification of molecular distinctions between normal breast-associated fibroblasts and breast cancer-associated fibroblasts. Cancer Microenvironment 2009;2:9–21. [187] Mukherjee S, Frolova N, Sadlonova A, et al. Hedgehog signalling and response to cyclopamine differ in epithelial and stromal cells in benign breast and breast cancer. Cancer Biology & Therapy 2006;5: 674–83. [188] Giulianelli S, Cerliani JP, Lamb CA, et al. Carcinoma-associated fibroblasts activate progesterone receptors and induce hormone independent mammary tumor growth: a role for the FGF-2/FGFR-2 axis. International Journal of Cancer 2008;123:2518–31. [189] Schor SL, Schor AM, Rushton G. Fibroblasts from cancer patients display a mixture of both foetal and adult-like phenotypic characteristics. Journal of Cell Science 1988;90:401–7. [190] Bhowmick NA, Neilson EG, Moses HL. Stromal fibroblasts in cancer initiation and progression. Nature 2004;432:332–7.
109
[191] Bhowmick NA, Moses HL. Tumor-stroma interactions. Current Opinion in Genetics & Development 2005;15:97–101. [192] Cheng N, Bhowmick NA, Chytil A, et al. Loss of TGF-  type II receptor in fibroblasts promotes mammary carcinoma growth and invasion through upregulation of TGF-␣-, MSP- and HGF-mediated signaling networks. Oncogene 2005;24:5053–68. [193] Franco OE, Jiang M, Strand DW, et al. Altered TGF- signaling in a subpopulation of human stromal cells promotes prostatic carcinogenesis. Cancer Research 2011;71:1272–81. [194] Kiskowski MA, Jackson 2nd RS, Banerjee J, et al. Role for stromal heterogeneity in prostate tumirgtenesis. Cancer Research 2011;71:3459–70. [195] Quiros RM, Valianou M, Kwon Y, et al. Ovarian normal and tumorassociated fibroblasts retain in vivo stromal characteristics in a 3-D matrix-dependent manner. Gynecologic Oncology 2008;110: 99–109. [196] Sheu B-C, Hsu S-M, Ho H-N, et al. A novel role of metalloproteinase in cancer-mediated immunosuppression. Cancer Research 2001;61:237–42. [197] Littlepage LE, Sternlicht MD, Rougier N, et al. Matrix metalloproteinases contribute distinct roles in neuroendocrine prostate carcinogenesis, metastasis, and angiogenesis progression. Cancer Research 2010;70:2224–34. [198] Liotta LA, Kohn EC. The microenvironment of the tumour–host interface. Nature 2001;411:375–9. [199] Hopfer H, Rinehart Jr CA, Vollmer G, Kaufman DG. In vitro interactions of endometrial stromal and epithelial cells in Matrigel: reorganization of the extracellular matrix. Pathobiology 1994;62:104–8. [200] Zetterblad J, Qian H, Zandi S, et al. Genomics based analysis of interactions between developing B-lymphocytes and stromal cells reveal complex interactions and two-way communication. BMC Genomics 2010;11:108. [201] Schmeichel KL, Bissell MJ. Modeling tissue-specific signalling and organ function in three dimensions. Journal of Cell Science 2003;116:2377–88. [202] Richman PI, Bodmer WF. Control of differentiation in human colorectal carcinoma cell lines: epithelial–mesenchymal interactions. Journal of Pathology 1988;156:197–211. [203] Janecki A, Jakubowiak A, Steinberger A. Effect of germ cells on vectorial secretion of androgen binging protein and transferrin by immature rat Sertoli cells in vitro. Journal of Andrology 1988;9:126–32. [204] Levine JF, Stockdale FE. Cell–cell interactions promote mammary epithelial cell differentiation. Journal of Cell Biology 1985;100:1415–22. [205] Kim SJ, Kim JS, Park ES, et al. Astrocytes upregulate survival genes in tumor cells and induce protection from chemotherapy. Neoplasia 2011;13:286–98. [206] Lin Q, Balasubramanian K, Fan D, et al. Reactive astrocytes protect melanoma cells from chemotherapy by sequestering intracellular calcium through gap junction communication channels. Neoplasia 2010;12:748–54. [207] Kim SW, Kim JS, Papadopoulos J, et al. Consistent interactions between tumor cell IL-6 and macrophage TNF-␣ enhance the growth of human prostate cancer cells in the bone of nude mouse. International Immunopharmacology 2011;11:862–72. [208] Orlidge A, D’Amore PA. Inhibition of capillary endothelial cell growth by pericytes and smooth muscle cells. The Journal of Cell Biology 1987;105:1455–62. [209] Sudo R, Chung S, Zervantonakis IK, Vickerman V, Toshimitsu Y, Griffith L, Kamm RD. Transport-mediated angiogenesis in 3D epithelial coculture. FASEB Journal 2009;23:2155–64. [210] Diamandopoulos GT. Microenvironmental influences on the in vivo behavior of neoplastic lymphocytes. Proceedings of the National Academy of Sciences of the United States of America 1978;76:6456–60.
110
ˇ B. Cunderlíková / Critical Reviews in Oncology/Hematology 85 (2013) 95–111
[211] Greene GF, Kitadai Y, Pettaway CA, et al. Correlation of metastasisrelated gene expression with metastatic potential in human prostate carcinoma cells implanted in nude mice using an in situ messenger RNA hybridization technique. American Journal of Pathology 1997;150:1571–82. [212] Meyvisch C. Influence of implantation site on formation of metastases. Cancer Metastasis Reviews 1983;2:295–306. [213] Rizki A, Weaver VM, Lee S-Y, et al. A human breast cell model of preinvasive to invasive transition. Cancer Research 2008;68: 1378–87. [214] Seandel M, Butler J, Lyden D, Rafii S. A catalytic role for proangiogenic marrow-derived cells in tumor neovascularization. Cancer Cell 2008;13:181–3. [215] Iinuma S, Schomacker KT, Wagnieres G, et al. In vivo fluence rate and fractionation effects on tumor response and photobleaching: photodynamic therapy with two photosensitizers in an orthotopic rat tumor model. Cancer Research 1999;59:6164–70. [216] Biswas C, Moran WP, Bloch KJ, Gross J. Collagenolytic activity of rabbit V2 -carcinoma growing at multiple sites. Biochemical and Biophysical Research Communication 1978;80:33–8. [217] Melcher A, Gough M, Todryk S, Vile R. Apoptosis or necrosis for tumor immunotherapy: what’s in a name? Journal of Molecular Medicine 1999;77:824–33. [218] Osband ME, Ross S. Problems in the investigational study and clinical use of immunotherapy. Immunology Today 1990;11:193–5. [219] Anisimov VN, Ukraintseva SV, Yashin AI. Cancer in rodents: Does it tell us about cancer in humans? Nature Reviews Cancer 2005;5:807–19. [220] Ginis I, Luo Y, Miura T, et al. Differences between human and mouse embryonic stem cells. Developmental Biology 2004;269:360–80. [221] Rangarajan A, Weinberg RA. Comparative biology of mouse versus human cells: modelling human cancer in mice. Nature Reviews Cancer 2003;3:952–9. [222] Rangarajan A, Hong SJ, Gifford A, Weinberg RA. Species- and cell type-specific requirements for cellular transformation. Cancer Cell 2004;6:171–83. [223] Hovey RC, McFaddenTB, Akers RM. Regulation of mammary gland growth and morphogenesis by the mammary fat pad: a species comparison. Journal of Mammary Gland Biology and Neoplasia 1999;4:53–68. [224] Ingber DE. Integrins as mechanochemical transducers. Current Opinion in Cell Biology 1991;3:841–8. [225] Lukashev ME, Werb Z. ECM signalling: orchestrating cell behaviour and misbehaviour. Trends in Cell Biology 1998;8:437–41. [226] Butcher DT, Alliston T, Weaver VM. A tense situation: forcing tumour progression. Nature Reviews Cancer 2009;9:108–22. [227] Pignatelli M, Bodmer WF. Genetics and biochemistry of collagen binding-triggered glandular differentiation in a human colon carcinoma cell line. Proceedings of the National Academy of Sciences of the United States of America 1988;85:5561–5. [228] Haas T, Davis SJ, Madri JA. Three-dimensional type I collagen lattices induce coordinate expression of matrix metalloproteinases MT1-MMP and MMP-2 in microvascular endothelial cells. Journal of Biological Chemistry 1998;273:3604–10. [229] Hay ED. Extracellular matrix alters epithelial differentiation. Current Opinion in Cell Biology 1993;5:1029–35. [230] Tomasek JJ, Halliday NL, Updike DL, et al. Gelatinase A activation is regulated by the organization of the polymerized actin cytoskeleton. Journal of Biological Chemistry 1997;272:7482–7. [231] Lelièvre S, Weaver VM, Nickerson JA, et al. Tissue phenotype depends on reciprocal interactions between the extracellular matrix and the structural organization of the nucleus. Proceedings of the National Academy of Sciences of the United States of America 1998;95:14711–6. [232] Maaser K, Wolf K, Klein CE, et al. Functional hierarchy of simultaneously expressed adhesion receptors: integrin ␣21 but not CD44 mediates MV3 melanoma cell migration and matrix reorganization
[233]
[234]
[235]
[236]
[237] [238]
[239]
[240]
[241]
[242]
[243] [244]
[245]
[246]
[247]
[248]
[249]
[250]
[251]
[252]
within three-dimensional hyaluronan-containing collagen matrices. Molecular Biology of the Cell 1999;10:3067–79. Kurschat P, Zigrino P, Nischt R, et al. Tissue inhibitor of matrix metalloproteinase-2 regulates matrix metalloproteinase-2 activation by modulation of membrane-type 1 matrix metalloproteinase activity in high and low invasive melanoma cell lines. Journal of Biological Chemistry 1999;274:21056–62. Wozniak MA, Desai R, Solski PA, Der CJ, Keely PJ. ROCK-generated contractility regulates breast epithelial cell differentiation in response to the physical properties of a three-dimensional collagen matrix. Journal of Cell Biology 2003;163:583–95. Azzam HS, Thompson EW. Collagen-induced activation of the Mr 72,000 type IV collagenase in normal and malignant human fibroblastoid cells. Cancer Research 1992;52:4540–4. Azzam HS, Arand G, Lippman ME, Thompson EW. Association of MMP-2 activation potential with metastatic progression in human breast cancer cell lines independent of MMP-2 production. Journal of the National Cancer Institute 1993;85: 1758–63. Paszek MJ, Zahir N, Johnson KR, et al. Tensional homeostasis and the malignant phenotype. Cancer Cell 2005;8:241–54. Levental KR, Yu H, Kass L, et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell 2009;139: 891–906. Provenzano PP, Inman DR, Eliceiri KW, et al. Collagen density promotes mammary tumor initiation and progression. BMC Medicine 2008;6:11. von Wichert G, Krndija D, Schmid H, et al. Focal adhesion kinase mediates defects in the force-dependent reinforcement of initial integrin-cytoskeleton linkages in metastatic colon cancer cell lines. European Journal of Cell Biology 2008;87:1–16. Assoian RK, Klein EA. Growth control by intracellular tension and extracellular stiffness. Trends in Cell Biology 2008;18: 347–52. Buxboim A, Ivanovska IL, Discher DE. Matrix elasticity, cytoskeletal forces and physics of the nucleus: How deeply do cells ˇıfeelˇı outside and in? Journal of Cell Science 2010;123:297–308. Buxboim A, Discher DE. Stem cells feel the difference. Nature Methods 2010;7:695–6. Engler AJ, Griffin MA, Sen S, et al. Myotubes differentiate optimally on substrates with tissue-like stiffness: pathological implications for soft or stiff microenvironments. Journal of Cell Biology 2004;166:877–87. Stolz M, Gottardi R, Raiteri R, et al. Early detection of aging cartilage and osteoarthritis in mice and patient samples using atomic force microscopy. Nature Nanotechnology 2009;4:186–92. Flanagan LA, Ju YE, Marg B, Osterfield M, Janmey PA. Neurite branching on deformable substrates. NeuroReport 2002;13: 2411–5. Netti PA, Berk DA, Swartz MA, Grodzinsky AJ, Jain RK. Role of extracellular matrix assembly in interstitial transport in solid tumors. Cancer Research 2000;60:2497–503. Jones C, Mackay A, Grigoriadis A, et al. Expression profiling of purified normal human luminal and myoepithelial breast cells: identification of novel prognostic markers for breast cancer. Cancer Research 2004;64:3037–45. Ross DT, Perou CM. A comparison of gene expression signatures from breast tumors and breast tissue derived cell lines. Disease Markers 2001;17:99–109. Pankov R, Endo Y, Even-Ram S, et al. A Rac switch regulates random versus directionally persistent cell migration. Journal of Cell Biology 2005;170:793–802. Sahai E, Marshall CJ. Differing modes of tumour cell invasion have distinct requirements for Rho/ROCK signalling and extracellular proteolysis. Nature Cell Biology 2003;5:711–9. Sahai E, Garcia-Medina R, Pouysségur J, Vial E. Smurf1 regulates tumor cell plasticity and motility through degradation of RhoA
ˇ B. Cunderlíková / Critical Reviews in Oncology/Hematology 85 (2013) 95–111
[253] [254] [255]
[256]
[257]
leading to localized inhibition of contractility. Journal of Cell Biology 2007;176:35–42. Muthuswamy SK. 3D culture reveals a signaling network. Breast Cancer Research 2011;13:103–4. Fringer J, Grinnell F. Fibroblast quiescence in floating or released collagen matrices. Journal of Biological Chemistry 2001;276:31047–52. Novaro V, Radisky DC, Ramos Castro NE, Weisz A, Bissell MJ. Malignant mammary cells acquire independence from extracellular context for regulation of estrogen receptor ␣. Clinical Cancer Research 2004;10:402S–S409. Petersen OW, Gudjonsson T, Villadsen R, Bissell MJ, Rønnov-Jessen L. Epithelial progenitor cell lines as models of normal breast morphogenesis and neoplasia. Cell Proliferation 2003;36:33–44. Kirkland SC, Henderson K, Liu D, Pignatelli M. Organisation and gel contraction by human colonic carcinoma (HCA-7) subline grown in 3-dimensional collagen gel. International Journal of Cancer 1995;60:877–82.
111
Biography ˇ Beata Cunderlíková, Ph.D. graduated from the Comenius University in Bratislava, Slovakia. She took her post-graduate studies at the Comenius University in Bratislava and at the Institute for Cancer Research in Oslo, Norway and defended her doctoral theses at the Comenius University, Slovakia and at the University of Oslo, Norway. She was a fellow at the Department of Pathology, The Norwegian Radiumhospital/The Oslo University Hospital in Oslo, Norway. Since undergraduate studies she was mainly engaged in the field of photodynamic therapy of cancer. Currently, she is working as a researcher at the International Laser Centre in Bratislava, Slovakia.