Skip to main content
  • Research article
  • Published:

Reduced expression of p27 is a novel mechanism of docetaxel resistance in breast cancer cells

Abstract

Introduction

Docetaxel is one of the most effective chemotherapeutic agents in the treatment of breast cancer. Breast cancers can have an inherent or acquired resistance to docetaxel but the causes of this resistance remain unclear. However, apoptosis and cell cycle regulation are key mechanisms by which most chemotherapeutic agents exert their cytotoxic effects.

Methods

We created two docetaxel-resistant human breast cancer cell lines (MCF-7 and MDA-MB-231) and performed cDNA microarray analysis to identify candidate genes associated with docetaxel resistance. Gene expression changes were validated at the RNA and protein levels by reverse transcription PCR and western analysis, respectively.

Results

Gene expression cDNA microarray analysis demonstrated reduced p27 expression in docetaxel-resistant breast cancer cells. Although p27 mRNA expression was found to be reduced only in MCF-7 docetaxel-resistant sublines (2.47-fold), reduced expression of p27 protein was noted in both MCF-7 and MDA-MB-231 docetaxel-resistant breast cancer cells (2.83-fold and 3.80-fold, respectively).

Conclusions

This study demonstrates that reduced expression of p27 is associated with acquired resistance to docetaxel in breast cancer cells. An understanding of the genes that are involved in resistance to chemotherapy may allow further development in modulating drug resistance, and may permit selection of those patients who are most likely to benefit from such therapies.

Introduction

Recent developments in chemotherapy have focused on the taxanes docetaxel and paclitaxel. Docetaxel is used in the treatment of breast cancer and is being evaluated in other solid tumours, including lung, gastro-oesophageal and, more recently, prostate cancers [1–3]. It is currently the most effective agent in the treatment of patients with advanced breast cancer. Up to 50% of patients who have previously been treated with or without anthracycline drugs, and have developed disease recurrence, will exhibit an objective response to docetaxel therapy [4]. Unfortunately, however, many patients do not respond to docetaxel or, having had an initial response, develop disease progression. This may occur either due to an inherent or an acquired resistance to docetaxel.

The mechanisms of docetaxel activity include binding to the β-tubulin subunits of microtubules, which prevents their depolymerization and thus blocks cell growth in the G2–M phase. This consequently results in cell death by phosphorylation of bcl-2 [5], which is integral to the apoptotic pathway. However, the mechanisms of docetaxel resistance are poorly understood, although there are some that have been identified as probably being involved in resistance. For example, mutations in the β-tubulin gene [6] and differential expression of β-tubulin isotypes have been associated with resistance to docetaxel and paclitaxel in breast, ovarian and lung cancers [7–9]. Furthermore, in lung and prostate cancers docetaxel has been shown to induce expression of the p27 protein, which is another key protein involved in apoptosis [5, 10].

The p27Kip1 gene is a member of the cyclin-dependent kinase inhibitors, which arrest progression of the cell cycle [11]. The p27 protein interacts with cdk2 and cyclin E to prevent subsequent entry into S phase of the cell cycle [12–14]. It also acts as a tumour suppressor gene and has been shown to be involved in cell adhesion, apoptosis and triggering of differentiation [13, 15–17]. Unlike many other tumour suppressor genes, however, gene mutation and loss of heterozygosity is infrequent, although p27 levels may be controlled by DNA methylation [18] and protein degradation through the ubiquitin pathway [19].

The clinical significance of this protein is supported by the fact that loss of p27 expression has been shown to be a strong predictor of reduced survival in patients with breast cancer and also correlates with increasing histological grade. Furthermore, loss of p27 is involved in tumour resistance to hormonal treatment [20–23]. In experimental animal models, mice that lack p27 protein expression are larger than wild-type mice and develop large organs and pituitary tumours, most probably resulting from uncontrolled cell proliferation [24–26]. Overexpression of p27, by recombinant adenovirus, can also induce spontaneous apoptosis and cell cycle arrest in breast and oral carcinomas in vitro [27, 28]. Other studies suggest that p27 expression can have an antiapoptotic effect and prevent drug-induced apoptosis by DNA damaging agents, such as cisplatin, leading to drug resistance [29]. The effect of p27, therefore, may be cell or tissue specific [14, 30]. The ability of p27 to initiate apoptosis may account for its possible involvement in chemotherapy-induced apoptosis [31].

Whether modulation of p27 expression plays a role in the development of resistance to docetaxel in breast cancer cells has not previously been investigated. This study focuses on the role played by p27 in docetaxel resistance, at RNA and protein levels, in an in vitro breast cancer model of acquired docetaxel resistance. In addition, our use of cDNA microarrays allowed identification of other candidate genes, which may be important for subsequent evaluation of docetaxel resistance.

Methods

Cell culture

Human breast cancer cell lines MCF-7 (oestrogen receptor positive) and MDA-MB-231 (oestrogen receptor negative) were cultured in RPMI-1640 medium, supplemented with 10% (vol:vol) foetal calf serum, 0.2% (weight:vol) sodium bicarbonate and 1% (vol:vol) penicillin–streptomycin, at 37°C in a humidified atmosphere containing 5% carbon dioxide. The cells were made resistant to docetaxel by short-term in vitro exposure to docetaxel (a gift from Aventis Pharma Ltd, West Malling, Kent, UK) for 1 hour, which was immediately followed by washing of the cells several times with culture media, trypsinization, and splitting the cells for subsequent cell growth recovery. The cells were initially exposed to 10 nmol/l docetaxel increasing to 500 nmol/l for 1 hour. After this point, the cells were exposed to 1 μmol/l docetaxel increasing to 30 μmol/l docetaxel for 24 hours.

Cytotoxic assay

Docetaxel resistance was demonstrated in cell lines by means of the 3-(4-5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) dye reduction assay [32]. The MCF-7 and MDA-MB-231 cells and their resistant sublines were plated onto 96-well plates, with a seeding density of 5 × 104 cells/well in 100 μl RPMI-1640 culture medium. Cells were exposed to varying concentrations of docetaxel and incubated at 37°C in a humidified atmosphere containing 5% carbon dioxide for 24 hours. Following exposure of cells to docetaxel, cells were treated with 50 μl 12 mmol/l MTT dye and incubated at 37°C for 4 hours. Following this incubation period, all the liquid was aspirated from each well before the addition of 200 μl dimethylsulphoxide in order to dissolve the MTT–formazan crystals. The number of viable cells was determined by measuring the absorbance at 570 nm and 630 nm for each well using a microplate spectrophotometer (DynaTech MR5000, DynaTech Laboratories Inc., Chantilly, VA, USA). All experiments were repeated three times with six replicates per experiment.

The concentration of docetaxel required to cause 50% inhibition of cell growth was calculated by direct cell counts in the presence of docetaxel. MCF-7 and MDA-MB-231 cells and their resistant sublines were plated onto petri dishes in RPMI-1640 culture medium. Cells were incubated at 37°C in a humidified atmosphere containing 5% carbon dioxide for 24 hours. After this time, the medium was removed and replaced with fresh culture medium containing varying concentrations of docetaxel and incubated for a further 24 hours. The medium was then removed and the cells resuspended in 1 ml culture medium. In order to count the cells, 100 μl of the cell solution was added to 100 μl trypan blue, and the cells were counted using a haemocytometer by light microscopy. The relative survival was expressed as a percentage inhibition of growth relative to the control. All experiments were repeated two times with two replicates per experiment.

cDNA microarray analysis

Total RNA was isolated from the cell lines using Trizol reagent (Invitrogen, Paisley, UK) in accordance with the manufacturer's instructions. RNA quality was enhanced using the RNeasy Mini Kit (Qiagen, Crawley, West Sussex, UK), and the integrity of RNA was checked by separating RNA by electrophoresis through a 1% (weight:vol) agarose gel. Following electrophoresis, the presence of two distinct bands (representing the 28S and 18S species of rRNA), with no or minimal smearing, confirmed the presence of undegraded RNA. In addition, the purity of RNA was determined by measuring the absorbance at 260 nm and 280 nm using spectrophotometry. Expression of cancer pathway genes was evaluated using Cancer Pathway Finder gene expression arrays (Super Array Inc., Frederick, MA, USA), in accordance with the manufacturer's instructions. In brief, gene-specific biotin-labelled cDNA probes were generated from 2.5 μg total RNA using gene-specific primers for reverse transcription using 200 U MMLV reverse transcriptase (Promega, Southampton, UK) and 40 U RNase inhibitor (Promega). The cDNA probe was denatured by heating at 94°C for 5 min and quickly cooled on ice before hybridization, with the cDNA membrane array, at 68°C overnight. Prior to hybridization, the membrane was prehybridized at 68°C for 30 min in the supplied hybridization buffer (Super Array Inc.). The membrane was washed accordingly and then the hybridization signals were detected using the CPD Star chemiluminescent detection kit (Super Array Inc.). Following this, the membrane was scanned with a Fluor S phosphorimager (Biorad, Hemel Hempstead, Hertfordshire, UK) and the scanned image was converted into digital data and analyzed using GEArray Analyzer software (Super Array Inc.)

Reverse-transcription polymerase chain reaction

Total RNA (2 μg) was reverse transcribed in 20 μl reverse transcription reaction buffer (50 mmol/l Tris, pH 8.3, 75 mmol/l potassium chloride, 15 mmol/l magnesium chloride, 10 mmol/l dithiothreitol), containing 250 ng random hexamers (Amersham Pharmacia, Little Chalfont, Buckinghamshire, UK), 500 μmol/l dNTP and 200 U Superscript II reverse transcriptase (Invitrogen). Reverse transcription was performed at 42°C for 1 hour, followed by heating at 70°C for 10 min. The cDNA (1 μl) was amplified in 25 μl reaction volumes, which contained the following components: 10 mmol/l Tris, pH8.3, 50 mmol/l potassium chloride, 0.1 mg/ml gelatin, 2.5 mmol/l magnesium chloride, 200 μmol/l dNTP, 10 pmol of each oligonucleotide primer, and 0.5 U Taq DNA polymerase (Roche Diagnostics, Lewes, East Sussex, UK). PCR was carried out under the following conditions: 94°C for 2 min followed by 23 cycles of 94°C for 30 s, 60°C for 30 s, 72°C for 30 s and a final extension step of 72°C for 10 min. The oligonucleotide primer sequences for p27 produced a 253 bp PCR product: sense primer 5'-TGG AGA AGC ACT GCA GAG AC-3' and antisense primer 5'-GCG TGT CCT CAG AG T TAG CC-3'. In addition, QuantumRNA™ 18S internal control standards (Ambion Inc., Huntingdon, UK), which produced a PCR product of 489 bp, were used at a ratio of 10:1 competitors to primers, in accordance with the manufacturer's instructions, to act as an internal loading control to normalize between samples during densitometry. PCR products were electrophoresed through a 1.5% (weight:vol) agarose gel. Gel images were captured by Genesnap software (Syngene, Cambridge, UK) and band densities calculated using a Fluor S phosphoimager (Biorad). The experiments were repeated in triplicate with RNA isolated from two independent extractions.

Western analysis

Cells were homogenized in a lysis buffer (20 mmol/l Tris, 0.25 mol/l sucrose, 10 mmol/l EGTA, 2 mmol/l EDTA, 1 mmol/l sodium orthovanadate, 25 mmol/l sodium β-glycerophosphate, 50 mmol/l sodium fluoride, pH 7.5). Prior to use, 0.1% (vol:vol) protease inhibitor cocktail (Sigma, Gillingham, Dorset, UK) was added. Cells were lysed by sonication and aspiration through a 25 G needle. Protein concentration was quantified using the DC Protein Assay (Biorad). Ten micrograms of protein was electrophoresed through a precast 15% polyacrylamide gel (Cambrex Bioscience, Nottingham, UK) for 2 hours at 25 V. Following electrophoresis, the separated proteins were transferred to nitrocellulose membranes (Biorad) and each membrane was blocked with 5% (weight:vol) skimmed milk in Tris-buffered saline with 0.1% (vol:vol) Tween 20 (TBST) at 4°C overnight. Each membrane was incubated with 1:250 dilution (in 5% [weight:vol] milk/TBST solution) of monoclonal mouse anti-human p27 antibody (DakoCytomation, Ely, UK) or 1:5000 β-actin (Abchem, Cambridge, UK) for 1 hour at room temperature. β-Actin was used as an internal loading control to normalize between samples during densitometry. The membrane was washed six times for 5 min in TBST solution before addition of the secondary antibody, HRP labelled goat anti-mouse IgG (Oncogene Research Products, San Diego, CA, USA), at a 1:5000 dilution in 5% (weight:vol) milk/TBST solution for 1 hour at room temperature. The membrane was further washed five times for 5 min in TBST solution and then once for 5 min in phosphate-buffered saline solution. Bands were visualized using ECL+plus™ chemiluminescent detection kit (Amersham Pharmacia) in accordance with the manufacturer's instructions, and the blots were scanned with a Fluor S phosphorimager (Biorad). The experiments were repeated in duplicate with protein isolated from two independent extractions. Cells were also checked by flow cytometry to ensure they were in the same cell cycle phase (data not shown).

Results

Docetaxel-induced cytotoxicity is reduced in docetaxel-resistant sublines

First, the in vitro effect of docetaxel on cell growth was determined using a standard cell viability assay. This demonstrated that docetaxel was more cytotoxic in MCF-7 and MDA-MB-231 cells than in their resistant sublines, which were able to withstand 24 hours of exposure of 30 μmol/l docetaxel. MCF-7 docetaxel-resistant sublines (MCF-7 TAX30) exhibited a 666-fold greater resistance to docetaxel than did MCF-7 cells. Furthermore, MDA-MB-231 docetaxel-resistant sublines (MDA-MB-231 TAX30) exhibited a 1375-fold greater resistance to MDA-MB-231 cells (Table 1).

Table 1 50% Inhibitory concentrations for docetaxel in breast cancer cell lines

Membrane-based microarray identifies p27 as a candidate gene associated with docetaxel resistance

In order to identify genes associated with docetaxel resistance, we applied a cDNA expression array to our in vitro docetaxel-resistant breast cancer model. The human cancer pathway array contained 96 genes that are involved in various pathways of cancer development, including tumour suppressor genes, oncogenes, signal transduction pathway genes, growth factors and receptors, and genes involved in angiogenesis. There was a 2.3-fold downregulation of p27 in MCF-7 docetaxel-resistant cells as compared with the parental cells (Table 2). Because p27 is a direct target for docetaxel, we selected this as a candidate for the present study. In addition, there were further changes in gene expression as illustrated in Table 2.

Table 2 Gene expression differences in docetaxel-resistant breast cancer cells

Expression of p27 mRNA is different in two docetaxel-resistant breast cancer cell lines

In order to confirm the results obtained using cDNA microarrays, the expression of p27 at the RNA level was determined by semiquantitative reverse transcription PCR analysis. In comparison with MCF-7 cells, MCF-7 docetaxel-resistant sublines exhibited a 2.47 ± 0.15 fold decrease in p27 mRNA expression (Fig. 1). In contrast, p27 mRNA expression was increased in MDA-MB-231 resistant cells (2.29 ± 0.76 fold).

Figure 1
figure 1

Modulation of p27 mRNA expression in docetaxel-resistant breast cancer cells. Total RNA extracted from each cell line was amplified using specific oligonucleotide primer sequences and separated by agarose gel electrophoresis. The p27 and 18S internal control amplified PCR products produced fragment sizes of 253 bp and 489 bp, respectively. Lane 1, DNA size standards; lane 2, MCF-7 docetaxel-resistant subline; lane 3, MCF-7 breast cancer cells; lane 4, MDA-MB-231 breast cancer cells; lane 5, MDA-MB-231 docetaxel-resistant subline.

Reduction in p27 protein in docetaxel-resistant breast cancer cells

The conflicting p27 mRNA expression difference between the two docetaxel-resistant breast cancer cell lines was validated at the protein level by western analysis. It was observed that p27 protein expression was reduced in both docetaxel-resistant cell lines (Fig. 2). There was a 2.83 ± 0.15 fold decrease in MCF-7 resistant cells and a 3.80-fold decrease in MDA-MB-231 resistant cells.

Figure 2
figure 2

Modulation of p27 protein expression in docetaxel-resistant breast cancer cells. Protein extracted from each cell line was separated by PAGE, transferred to nitrocellulose membranes and probed with anti-human p27 and β-actin antibodies. Lane 1, MCF-7 breast cancer cells; lane 2, MCF-7 docetaxel-resistant subline; lane 3, MDA-MB-231 breast cancer cells; lane 4, MDA-MB-231 docetaxel-resistant subline.

Discussion

This study is the first to demonstrate that reduced p27 expression is associated with acquired resistance to docetaxel in breast cancer cells in vitro. There may be several mechanisms that are involved in this process and that may be the result of either transcriptional or translational regulation, with modulation of gene expression and/or resultant production of the protein. In addition, there may be an increased degradation of p27 via the ubiquitin degradation pathway.

Previous studies have indicated that decreased expression of p27 is associated with drug resistance to hormonal therapy (e.g. tamoxifen) in breast cancer cells and to chemotherapy (e.g. cisplatin in malignant gliomas), mediated through mitogen-activated protein kinase activation [20, 21, 33]. It has also been reported that reduced p27 protein expression in epithelial ovarian tumours was observed, as compared with normal ovaries, from patients receiving cisplatin and paclitaxel treatment [34]. Reduced p27 expression was significantly associated with presumed chemoresistance (i.e. the patients had persistent disease after treatment) [34]. Furthermore, resistance to the anti-tumour agent rapamycin (a potent immunosuppressant that was recently reported to be effective in breast cancer treatment) has also been shown to be associated with reduced p27 expression in murine myogenic cells [35]. Another study noted that decreased p27 expression correlated with chronic Helicobacter pylori infection, which resulted in a phenotype that was resistant to apoptosis (and therefore potentially drug resistant because the majority of chemotherapeutic agents induce apoptosis) in gastric cancer patients [36]. It is possible that decreased p27 expression may prevent apoptosis or arrest cells in G1 phase, thereby reducing the cytotoxic effect of chemotherapeutic drugs that act on proliferating cells.

Increased p27 expression has also been implicated in causing drug resistance in leukaemia cell suspensions by preventing apoptosis [30]. The authors of that study reported that overexpression of p27 in the cells resulted in resistance to drug-induced apoptosis, including DNA damaging and non-DNA damaging agents. In addition, increased p27 expression is associated with cisplatin resistance in colorectal cancer cells [29]. Naumann and colleagues [33] also reported that increased p27 expression could increase resistance to vincristine, camptothecin and teniposide in malignant glioma cells, whereas reducing p27 expression (by antisense mRNA) sensitized the same cells to cisplatin, although effects appeared to be drug specific. Therefore, it may be that the effects of an individual antitumour agent are dependent on the mechanisms of action involved.

It has generally been regarded that increased p27 can initiate apoptosis by increasing the expression of the proapoptotic protein bax [31]. Docetaxel-induced cytotoxicity is mediated by increased p27 expression in lung and prostate cancers [10]. Furthermore, if the expression of p27 is increased, by using the proteosome inhibitor PS-341 (which inhibits the activity of the proteosome enzyme) [37], then this increases docetaxel-induced apoptosis in lung cancer cells [38]. Reduced p27 levels, therefore, may prevent cell death occurring by this mechanism. Indeed, p27 was demonstrated to induce apoptosis in response to low doses of docetaxel independent of bcl-2 phosphorylation, which is a recognized event for apoptosis regulation by docetaxel [5].

The precise mechanism by which decreased p27 expression may lead to docetaxel resistance is not yet fully understood. The p27 protein is primarily involved in cell cycle arrest (G1) via its ability to bind with and inactivate cyclin E–cdk2 complexes [39]. It is likely, therefore, that reduced p27 expression will prevent cell cycle arrest. In addition, downregulation of p27, with antisense molecules, leads to re-entry into the cell cycle, which leads to cell proliferation [40]. It has also been suggested that a loss of p27 may desensitize tumour cells to antimitogenic signals, thus preventing apoptosis in their evolution [41]. Continued exposure to docetaxel may therefore select for apoptosis-resistant tumour cells that express decreased levels of p27. One recent study demonstrated that a decrease in p27 expression in normal mammary tissue during pregnancy results in both an increase in cell proliferation and in apoptosis, suggesting that under normal circumstances a reduced expression of p27 would not result in a significant growth advantage [42]. If the apoptotic pathway is impaired, however, then cells could proliferate after exposure to docetaxel, and this may contribute to drug resistance.

In addition to p27, cDNA microarray analysis revealed several other genes with expression that may be altered in docetaxel-resistant breast cancer cell lines (Table 2). These include decreased expression of cell cycle (CCNE1 and CDKN1B) and proapoptotic genes (BAX and TNFRSF1A), and increased expression of antiapoptotic genes (BIRC5), which could lead to resistance to the mechanisms of action of docetaxel. Increased expression of angiogenic (IL8) and growth factors (Erbb2) could also lead to a more aggressive cell type that is perhaps able to escape the cytotoxic effects of the drug. These changes, however, remain to be validated at the mRNA and protein levels.

One of the limitations of the present study is the use of an in vitro model of drug resistance, which is, by definition, an artificial environment. In particular, our experimental approach selected cells with an acquired resistance to docetaxel rather than utilizing cells that exhibit de novo resistance to the drug. We acknowledge that it is possible that different mechanisms may be involved in de novo docetaxel resistance. However, a previous study successfully used colon cancer cell lines and their resistant sublines to identify a candidate gene, thymidylate synthase, that is involved in resistance to thymidylate synthase inhibitors [43].

Conclusion

The results from this study used cDNA microarrays based on specific molecular pathways that are believed to be involved in tumour progression and drug resistance. There are other, more comprehensive cDNA microarray methods available, which produce copious amounts of data. However, our experimental approach provides a quicker, less complicated and more focused way to identify possible candidate genes. To the best of our knowledge, this is the first description of an in vitro model of acquired docetaxel resistance in breast cancer cells. We directly compared breast cancer cells with their docetaxel-resistant sublines to identify gene expression changes associated with docetaxel resistance. In particular, the fact that p27 protein expression is reduced in both docetaxel-resistant sublines suggests that it plays a role in docetaxel resistance in breast cancer. Further studies, involving genetic profiling, may be important in allowing identification of those patients who are most likely to respond to a particular treatment.

Abbreviations

bp:

base pairs

MTT:

3-(4-5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide

PCR:

polymerase chain reaction

TBST:

Tris-buffered saline with 0.1% (vol:vol) Tween 20.

References

  1. Figgitt DP, Wiseman LR: Docetaxel: an update of its use in advanced breast cancer. Drugs. 2000, 59: 621-651.

    Article  CAS  PubMed  Google Scholar 

  2. Petrylak DP, Shepherd FA, Fossella FV, Lynch T, Armand JP, Rigas JR, Kris MG, Figgitt DP, Wiseman LR: Docetaxel (Taxotere) in hormone-refractory prostate cancer. Semin Oncol. 2000, 27: 24-29.

    CAS  PubMed  Google Scholar 

  3. Shepherd FA, Fossella FV, Lynch T, Armand JP, Rigas JR, Kris MG, Figgitt DP, Wiseman LR: Docetaxel (Taxotere) shows survival and quality-of-life benefits in the second-line treatment of non-small cell lung cancer: a review of two phase III trials. Semin Oncol. 2001, 28: 4-9. 10.1053/sonc.2001.18389.

    Article  CAS  Google Scholar 

  4. Valero V, Holmes FA, Walters RS, Theriault RL, Esparza L, Fraschini G, Fonseca GA, Bellet RE, Buzdar AU, Hortobagyi GN: Phase II trial of docetaxel: a new, highly effective antineoplastic agent in the management of patients with anthracycline-resistant metastatic breast cancer. J Clin Oncol. 1995, 13: 2886-2894.

    Article  CAS  PubMed  Google Scholar 

  5. Haldar S, Basu A, Croce CM: Bcl2 is the guardian of microtubule integrity. Cancer Res. 1997, 57: 229-233.

    CAS  PubMed  Google Scholar 

  6. Monzo M, Rosell R, Sanchez JJ, Lee JS, O'Brate A, Gonzalez-Larriba JL, Alberola V, Lorenzo JC, Nunez L, Ro JY, Martin C: Paclitaxel resistance in non-small-cell lung cancer associated with beta-tubulin gene mutations. J Clin Oncol. 1999, 17: 1786-1793.

    Article  CAS  PubMed  Google Scholar 

  7. Banerjee A: Increased levels of tyrosinated alpha-, beta(III)-, and beta(IV)-tubulin isotypes in paclitaxel-resistant MCF-7 breast cancer cells. Biochem Biophys Res Commun. 2002, 293: 598-601. 10.1016/S0006-291X(02)00269-3.

    Article  CAS  PubMed  Google Scholar 

  8. Kavallaris M, Burkhart CA, Horwitz SB: Antisense oligonucleotides to class III beta-tubulin sensitize drug-resistant cells to Taxol. Br J Cancer. 1999, 80: 1020-1025. 10.1038/sj.bjc.6690507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hasegawa S, Miyoshi Y, Egawa C, Ishitobi M, Taguchi T, Tamaki Y, Monden M, Noguchi S: Prediction of response to docetaxel by quantitative analysis of class I and III beta-tubulin isotype mRNA expression in human breast cancers. Clin Cancer Res. 2003, 9: 2992-2997.

    CAS  PubMed  Google Scholar 

  10. Gumerlock P, Mack P, Gustafsson M, Togonon M, Gandara D: p27 induction as a potential p53-independent mechanism of apoptotic response to docetaxel in non-small cell lung (NSCL) and prostate carcinomas (CaP). Clin Cancer Res. 1999, 5: s3859-

    Google Scholar 

  11. Polyak K, Lee MH, Erdjument-Bromage H, Koff A, Roberts JM, Tempst P, Massague J: Cloning of p27Kip1, a cyclin-dependent kinase inhibitor and a potential mediator of extracellular antimitogenic signals. Cell. 1994, 78: 59-66. 10.1016/0092-8674(94)90572-X.

    Article  CAS  PubMed  Google Scholar 

  12. Sherr CJ: Cancer cell cycles. Science. 1996, 274: 1672-1677. 10.1126/science.274.5293.1672.

    Article  CAS  PubMed  Google Scholar 

  13. Thomas GV, Szigeti K, Murphy M, Draetta G, Pagano M, Loda M: Down-regulation of p27 is associated with development of colorectal adenocarcinoma metastases. Am J Pathol. 1998, 153: 681-687.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. St Croix B, Florenes VA, Rak JW, Flanagan M, Bhattacharya N, Slingerland JM, Kerbel RS: Impact of the cyclin-dependent kinase inhibitor p27Kip1 on resistance of tumor cells to anticancer agents. Nat Med. 1996, 2: 1204-1210.

    Article  CAS  PubMed  Google Scholar 

  15. Levkau B, Koyama H, Raines EW, Clurman BE, Herren B, Orth K, Roberts JM, Ross R: Cleavage of p21Cip1/Waf1 and p27Kip1 mediates apoptosis in endothelial cells through activation of Cdk2: role of a caspase cascade. Mol Cell. 1998, 1: 553-563. 10.1016/S1097-2765(00)80055-6.

    Article  CAS  PubMed  Google Scholar 

  16. de Putte GV, Holm R, Lie AK, Trope CG, Kristensen GB: Expression of p27, p21, and p16 protein in early squamous cervical cancer and its relation to prognosis. Gynecol Oncol. 2003, 89: 140-147. 10.1016/S0090-8258(03)00010-6.

    Article  Google Scholar 

  17. Durand B, Gao FB, Raff M: Accumulation of the cyclin-dependent kinase inhibitor p27/Kip1 and the timing of oligodendrocyte differentiation. Embo J. 1997, 16: 306-317. 10.1093/emboj/16.2.306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Qian X, Jin L, Kulig E, Lloyd RV: DNA methylation regulates p27kip1 expression in rodent pituitary cell lines. Am J Pathol. 1998, 153: 1475-1482.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Pagano M, Tam SW, Theodoras AM, Beer-Romero P, Del Sal G, Chau V, Yew PR, Draetta GF, Rolfe M: Role of the ubiquitin-proteasome pathway in regulating abundance of the cyclin-dependent kinase inhibitor p27. Science. 1995, 269: 682-685.

    Article  CAS  PubMed  Google Scholar 

  20. Cariou S, Donovan JC, Flanagan WM, Milic A, Bhattacharya N, Slingerland JM: Down-regulation of p21WAF1/CIP1 or p27Kip1 abrogates antiestrogen-mediated cell cycle arrest in human breast cancer cells. Proc Natl Acad Sci USA. 2000, 97: 9042-9046. 10.1073/pnas.160016897.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Donovan JC, Milic A, Slingerland JM: Constitutive MEK/MAPK activation leads to p27(Kip1) deregulation and antiestrogen resistance in human breast cancer cells. J Biol Chem. 2001, 276: 40888-40895. 10.1074/jbc.M106448200.

    Article  CAS  PubMed  Google Scholar 

  22. Fredersdorf S, Burns J, Milne AM, Packham G, Fallis L, Gillett CE, Royds JA, Peston D, Hall PA, Hanby AM, Barnes DM, Shousha S, O'Hare MJ, Lu X: High level expression of p27(kip1) and cyclin D1 in some human breast cancer cells: inverse correlation between the expression of p27(kip1) and degree of malignancy in human breast and colorectal cancers. Proc Natl Acad Sci USA. 1997, 94: 6380-6385. 10.1073/pnas.94.12.6380.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Catzavelos C, Bhattacharya N, Ung YC, Wilson JA, Roncari L, Sandhu C, Shaw P, Yeger H, Morava-Protzner I, Kapusta L, Franssen E, Pritchard KI, Shingerland JM: Decreased levels of the cell-cycle inhibitor p27Kip1 protein: prognostic implications in primary breast cancer. Nat Med. 1997, 3: 227-230.

    Article  CAS  PubMed  Google Scholar 

  24. Kiyokawa H, Kineman RD, Manova-Todorova KO, Soares VC, Hoffman ES, Ono M, Khanam D, Hayday AC, Frohman LA, Koff A: Enhanced growth of mice lacking the cyclin-dependent kinase inhibitor function of p27(Kip1). Cell. 1996, 85: 721-732. 10.1016/S0092-8674(00)81238-6.

    Article  CAS  PubMed  Google Scholar 

  25. Fero ML, Rivkin M, Tasch M, Porter P, Carow CE, Firpo E, Polyak K, Tsai LH, Broudy V, Perlmutter RM, Kaushansky K, Roberts JM: A syndrome of multiorgan hyperplasia with features of gigantism, tumorigenesis, and female sterility in p27(Kip1)-deficient mice. Cell. 1996, 85: 733-744. 10.1016/S0092-8674(00)81239-8.

    Article  CAS  PubMed  Google Scholar 

  26. Nakayama K, Ishida N, Shirane M, Inomata A, Inoue T, Shishido N, Horii I, Loh DY: Mice lacking p27(Kip1) display increased body size, multiple organ hyperplasia, retinal dysplasia, and pituitary tumors. Cell. 1996, 85: 707-720. 10.1016/S0092-8674(00)81237-4.

    Article  CAS  PubMed  Google Scholar 

  27. Supriatno , Harada K, Hoque MO, Bando T, Yoshida H, Sato M: Overexpression of p27(Kip1) induces growth arrest and apoptosis in an oral cancer cell line. Oral Oncol. 2002, 38: 730-736. 10.1016/S1368-8375(02)00011-8.

    Article  CAS  PubMed  Google Scholar 

  28. Craig C, Wersto R, Kim M, Ohri E, Li Z, Katayose D, Lee SJ, Trepel J, Cowan K, Seth P: A recombinant adenovirus expressing p27Kip1 induces cell cycle arrest and loss of cyclin-Cdk activity in human breast cancer cells. Oncogene. 1997, 14: 2283-2289. 10.1038/sj.onc.1201064.

    Article  CAS  PubMed  Google Scholar 

  29. Dimanche-Boitrel MT, Micheau O, Hammann A, Haugg M, Eymin B, Chauffert B, Solary E: Contribution of the cyclin-dependent kinase inhibitor p27KIP1 to the confluence-dependent resistance of HT29 human colon carcinoma cells. Int J Cancer. 1998, 77: 796-802. 10.1002/(SICI)1097-0215(19980831)77:5<796::AID-IJC20>3.3.CO;2-N.

    Article  CAS  PubMed  Google Scholar 

  30. Eymin B, Haugg M, Droin N, Sordet O, Dimanche-Boitrel MT, Solary E: p27Kip1 induces drug resistance by preventing apoptosis upstream of cytochrome c release and procaspase-3 activation in leukemic cells. Oncogene. 1999, 18: 1411-1418. 10.1038/sj.onc.1202437.

    Article  CAS  PubMed  Google Scholar 

  31. Fujieda S, Inuzuka M, Tanaka N, Sunaga H, Fan GK, Ito T, Sugimoto C, Tsuzuki H, Saito H: Expression of p27 is associated with Bax expression and spontaneous apoptosis in oral and oropharyngeal carcinoma. Int J Cancer. 1999, 84: 315-320. 10.1002/(SICI)1097-0215(19990621)84:3<315::AID-IJC20>3.3.CO;2-L.

    Article  CAS  PubMed  Google Scholar 

  32. Mosmann T: Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods. 1983, 65: 55-63. 10.1016/0022-1759(83)90303-4.

    Article  CAS  PubMed  Google Scholar 

  33. Naumann U, Weit S, Rieger L, Meyermann R, Weller M: p27 modulates cell cycle progression and chemosensitivity in human malignant glioma. Biochem Biophys Res Commun. 1999, 261: 890-896. 10.1006/bbrc.1999.1126.

    Article  CAS  PubMed  Google Scholar 

  34. Goff BA, Paley PJ, Greer BE, Gown AM: Evaluation of chemoresistance markers in women with epithelial ovarian carcinoma. Gynecol Oncol. 2001, 81: 18-24. 10.1006/gyno.2000.6105.

    Article  CAS  PubMed  Google Scholar 

  35. Luo Y, Marx SO, Kiyokawa H, Koff A, Massague J, Marks AR: Rapamycin resistance tied to defective regulation of p27Kip1. Mol Cell Biol. 1996, 16: 6744-6751.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Shirin H, Sordillo EM, Kolevska TK, Hibshoosh H, Kawabata Y, Oh SH, Kuebler JF, Delohery T, Weghorst CM, Weinstein IB, Moss SF: Chronic Helicobacter pylori infection induces an apoptosis-resistant phenotype associated with decreased expression of p27(kip1). Infect Immun. 2000, 68: 5321-5328. 10.1128/IAI.68.9.5321-5328.2000.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Adams J, Palombella VJ, Sausville EA, Johnson J, Destree A, Lazarus DD, Maas J, Pien CS, Prakash S, Elliott PJ: Proteasome inhibitors: a novel class of potent and effective antitumor agents. Cancer Res. 1999, 59: 2615-2622.

    CAS  PubMed  Google Scholar 

  38. Mack PC, Davies AM, Lara PN, Gumerlock PH, Gandara DR: Integration of the proteasome inhibitor PS-341 (Velcade) into the therapeutic approach to lung cancer. Lung Cancer. 2003, Suppl 1: S89-S96. 10.1016/S0169-5002(03)00149-1.

    Article  Google Scholar 

  39. Coats S, Flanagan WM, Nourse J, Roberts JM: Requirement of p27Kip1 for restriction point control of the fibroblast cell cycle. Science. 1996, 272: 877-880.

    Article  CAS  PubMed  Google Scholar 

  40. Rivard N, L'Allemain G, Bartek J, Pouyssegur J: Abrogation of p27Kip1 by cDNA antisense suppresses quiescence (G0 state) in fibroblasts. J Biol Chem. 1996, 271: 18337-18341. 10.1074/jbc.271.31.18337.

    Article  CAS  PubMed  Google Scholar 

  41. Park MS, Rosai J, Nguyen HT, Capodieci P, Cordon-Cardo C, Koff A: p27 and Rb are on overlapping pathways suppressing tumorigenesis in mice. Proc Natl Acad Sci USA. 1999, 96: 6382-6387. 10.1073/pnas.96.11.6382.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Davison EA, Lee CS, Naylor MJ, Oakes SR, Sutherland RL, Hennighausen L, Ormandy CJ, Musgrove EA: The cyclin-dependent kinase inhibitor p27 (Kip1) regulates both DNA synthesis and apoptosis in mammary epithelium but is not required for its functional development during pregnancy. Mol Endocrinol. 2003, 17: 2436-2447. 10.1210/me.2003-0199.

    Article  CAS  PubMed  Google Scholar 

  43. Rooney PH, Stevenson DA, Marsh S, Johnston PG, Haites NE, Cassidy J, McLeod HL: Comparative genomic hybridization analysis of chromosomal alterations induced by the development of resistance to thymidylate synthase inhibitors. Cancer Res. 1998, 58: 5042-5045.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to acknowledge Aventis Pharma Limited for the kind gift of docetaxel. In addition, we would like to thank the Breast Cancer Campaign, Gates Trust, Aberdeen Royal Infirmary Breast Unit and NHS Grampian, for financial support.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Iain Brown.

Additional information

Competing interests

None declared.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Authors’ original file for figure 2

Rights and permissions

Reprints and permissions

About this article

Cite this article

Brown, I., Shalli, K., McDonald, S.L. et al. Reduced expression of p27 is a novel mechanism of docetaxel resistance in breast cancer cells. Breast Cancer Res 6, R601 (2004). https://doi.org/10.1186/bcr918

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/bcr918

Keywords