Purpose: Antiestrogens are used to treat estrogen receptor (ER)-α-positive breast cancers and cause a p27-dependent G1 arrest. Estrogen-bound ER recruits Src to mediate proteolysis of p27 and drive cell proliferation. Here, we tested the antitumor efficacy of combined Src and aromatase inhibition for ER-positive breast cancer.

Experimental Design: Antiproliferative effects of the aromatase inhibitor, anastrozole, and Src inhibitor, AZD0530, alone or in combination were tested in vitro and in vivo on aromatase-transfected MCF-7Arom5 xenografts. Xenografts were analyzed by immunohistochemistry and proteomic analysis to identify potential biomarkers of drug response and resistance.

Results: AZD0530 and anastrozole together increased p27 and caused greater G1 cell cycle arrest than either drug alone. AZD0530 monotherapy initially retarded xenograft growth in vivo, but drug resistance rapidly emerged. Combined anastrozole/AZD0530 reduced drug resistance and showed greater antitumor efficacy in vivo with greater Src and epidermal growth factor receptor inhibition and a greater increase in p27 and reduction of Ki-67 than either drug alone, supporting further evaluation of these putative predictors of response to combined Src/aromatase inhibition in vivo. Anastrozole alone stimulated Src activity both in vitro and in vivo. AZD0530-resistant tumors showed activation of bypass pathways including MEK and phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin, raising the possibility that MEK, mammalian target of rapamycin (mTOR), or PI3K inhibitors may augment Src inhibitor efficacy.

Conclusions: These data support clinical investigation of anastrozole-AZD0530 therapy for postmenopausal ER-positive breast cancer. Loss of p27 and increased Ki-67 may predict response and further clinical studies should evaluate for activation of bypass pathways including MEK and PI3K pathways during Src inhibitor therapy.

Translational Relevance

Although aromatase inhibitors are used to treat postmenopausal estrogen receptor-positive breast cancer, resistance limits treatment efficacy. The cell cycle inhibitor p27 is a key mediator of the antiproliferative effects of antiestrogens. The Src kinase phosphorylates p27 to trigger p27 proteolysis. Because estrogens activate Src to drive p27 proteolysis, we reasoned that combined Src and aromatase inhibitor therapy might have greater efficacy than either drug alone. Xenografts rapidly developed resistance to AZD0530 alone, with strong activation of both MEK and phosphatidylinositol 3-kinase pathways in drug-resistant tumors. MEK or phosphatidylinositol 3-kinase inhibitors together with AZD0530 may have therapeutic potential in breast cancer. AZD0530/anastrozole showed greater antitumor efficacy, raising intratumoral p27 and lowering Ki-67 and pSrc more than either drug alone. Combined aromatase and Src inhibition may delay or prevent emergence of resistance to either drug alone and our preclinical data support initiation of clinical trials to test this in human breast cancer.

Estrogen is a potent mitogen and a survival factor for many estrogen receptor (ER)-α-positive human breast cancer lines and appears to play a role in the genesis and progression of a majority of breast cancers. ER blockade and drug-mediated estrogen deprivation are used in ER-positive breast cancer both as adjuvant treatments to prevent recurrence after initial breast surgery and for metastatic disease (1). Aromatase inhibitors such as anastrozole and letrozole have significantly improved the efficacy of endocrine therapy (2). Adrenal androstenedione and testosterone undergo peripheral conversion to estrogen by aromatase. Aromatase inhibitors block the last biosynthetic step in estrogen production (2) and cause >90% reduction in circulating estrogens in postmenopausal women (1). Anastrozole was the first aromatase inhibitor tested in large clinical trials in both metastatic and adjuvant settings and showed superior antitumor activity compared with tamoxifen (1, 2).

Oncogenic activation of the Src tyrosine kinase regulates proliferation, motility, and survival in many human cancers (3). We observed that >70% of primary human breast cancers show Src activation, with 40% at high levels (4). In addition, a gene expression signature of Src activation is seen in up to 40% of human breast cancers and is predictive of response to Src inhibitor drugs in vitro (5).

Src is activated in breast cancers via the ErbB family and insulin-like growth factor-I receptor pathways and other cell surface receptors (3, 6). In addition, estrogen binding to the ER leads to rapid, transient Src activation, formation of Src/Shc/ER and Src/p85/ER complexes, and signaling via phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) 1/2 (79). In addition to ER-α signaling to Src, Src can also phosphorylate ER-α on Tyr537 (10). Because of the potential for two-way crosstalk between ER-α and Src, aberrant activation of either protein initiates feed-forward loops to promote breast cancer growth.

Estrogens recruit quiescent ER-positive breast cancer cells into cell cycle and shorten the G1 phase. In quiescent, estrogen-deprived breast cancer cells, the cyclin-dependent kinase (Cdk) inhibitor p27 is elevated and p27 binds and inhibits cyclin E-Cdk2 (1114). In normal and malignant mammary epithelial cells, estrogens and growth factors stimulate p27 phosphorylation and loss of cyclin E-Cdk2, with resulting cyclin E-Cdk2 activation and p27 proteolysis promoting S-phase entry and cell cycle progression. We and others showed that the Cdk inhibitors p21 and p27 are both essential for G1 arrest induced by either estrogen withdrawal or ER blockade by tamoxifen (12, 15). Estrogens and growth factors activate Src. Src, in turn, promotes p27 proteolysis through phosphorylation of p27 at Tyr74 and Tyr88 (4).

Given the frequent activation of Src in breast cancer (4) through both estrogen-bound ER (79) and receptor tyrosine kinase action (6, 16), we postulated that blocking both Src activation and estrogen synthesis might improve antitumor efficacy. This study investigated effects of the aromatase inhibitor, anastrozole, and a novel Src inhibitor, AZD0530, alone or in combination. Both drugs together led to a greater increase in p27 levels and proliferative arrest than did either drug alone in both cell culture and in vivo in a breast cancer xenograft model. Proteomic analysis of xenograft tumors yielded potential predictive markers for drug response and revealed bypass pathway activation in tumors resistant to AZD0530 monotherapy.

Generation of MCF-7Arom5. Full-length human placental aromatase cDNA (17) was introduced into pcDNA3.0 and transfected into MCF-7. Clones were selected and screened for aromatase expression. The MCF-7Arom5 line was used for all studies.

Chemicals. The c-Src inhibitor, AZD0530, and the aromatase inhibitor, anastrozole, were provided by Astra Zeneca, dissolved in DMSO and ethanol, respectively, and diluted in tissue culture medium. Vehicle controls had no effect on analyses done.

Cell culture. MCF-7Arom5 and MCF-Ca were cultured in improved modified Eagle's medium (option Zn2+) with 5% fetal bovine serum, 1% penicillin/streptomycin, and 400 μg/mL G418.

Cell cycle effects of anastrozole and AZD0530. MCF-7Arom5 and MCF-Ca (18) were cultured in estrogen-deprived medium (with 5% charcoal-stripped fetal bovine serum) with or without 25 nmol/L 4- androstenedione and treated with different concentrations of anastrozole, 1 μmol/L AZD0530, or both. AZD0530 did not exceed 1 μmol/L to avoid off target effects.

Anastrozole is rapidly metabolized in tissue culture and concentrations required for growth inhibition are considerably higher than the 137 nmol/L trough therapeutic plasma concentration in humans. Growth-inhibitory anastrozole concentrations vary with aromatase levels, which differ by up to 20-fold between breast cancer lines (19). Prior work showed that whereas 0.1 μmol/L letrozole arrests aromatase-transfected MCF-Ca, 0.1 μmol/L anastrozole had no cell cycle effect (20). Cell cycle inhibition has been observed with 25 to 3,500 μmol/L anastrozole in other lines and 50 μmol/L caused only 50% inhibition of aromatase activity in other studies (19, 21).

Aromatase levels were 5-fold higher in MCF-7Arom5 than MCF-Ca. Anastrozole concentrations required for growth arrest of MCF-7Arom5 were titrated. Anastrozole 12.5 to 1,000 μmol/L caused increasing degrees of reversible G1 cell cycle arrest with no apparent toxicity. Subsequent investigations used 100 μmol/L anastrozole because it caused partial G1 arrest and permitted detection of further cell cycle inhibition when combined with AZD0530. Asynchronously proliferating untreated and drug-treated cells were collected 48 h later for flow cytometry and Western blotting.

Flow cytometric analysis. Cells were pulse labeled with 10 μmol/L bromodeoxyuridine and stained with anti-bromodeoxyuridine-conjugated FITC (Becton Dickinson) and propidium iodide as described (12).

Growth inhibition of MCF-7Arom5 tumor xenografts. Female ovariectomized athymic BALB/c nude mice, 4 to 6 weeks old (National Cancer Institute), were housed in a pathogen-free environment under controlled conditions and received food and water ad libitum. Animals were acclimatized 3 days after shipment before tumor inoculation (18, 22). Subconfluent MCF-7Arom5 cells were resuspended in Matrigel (10 mg/mL; Becton Dickinson) at 2.5 × 107 cells/mL. Each animal was injected with 0.1 mL cell suspension into one mammary fat pad. Median tumor size at initiation of drug treatment was 70 mm3. Mice received either no treatment (without added androstenedione with vehicle alone) or daily treatment with 4-androstenedione (100 μg/day) subcutaneously alone or with either anastrozole (0.25 mg/kg or ∼5 μg/animal) subcutaneously, AZD0530 (50 mg/kg) by oral gavage, or both drugs together. This anastrozole dose delayed xenograft growth without causing regression in prior studies (23). Oral AZD0530 50 mg/kg inhibits Src in xenograft target tissues (AstraZeneca AZD0530 Investigators Brochure). Tumors were measured weekly with calipers, and volumes calculated with the formula: 1/2 × a × b2 (a > b).

Immunoblotting and immunoprecipitation. Cells were lysed in ice-cold 1% NP-40 lysis buffer as described (12). Western analysis of cyclin E, p27, Cdk2, Cdk4, cyclin D1, MAPK, pMAPK (MAPKpT202/Y204), Akt, pAkt (AktpT473), Src, and pSrc (Src-pY416) used the following: antibodies to MAPK, pMAPK, Akt, pAkt, Src, and pSrc were obtained from Cell Signaling; antibodies to p27 were from Transduction Laboratories; antibodies to cyclin E1 (monoclonal antibodies E172 and E12) were from E. Harlow (MGH); and antibodies to β-actin were from Sigma. Other antibodies were from Santa Cruz Biotechnology. Protein levels were assayed for three different enhanced chemiluminescence exposures and quantitated by densitometry using Image Quant software from at least three different biological experiments.

Cyclin E was immunoprecipitated from 200 μg protein lysate with cyclin E monoclonal antibody E172 and associated proteins detected by immunoblotting or cyclin E-Cdk2 kinase assayed using [γ-32P]ATP (Amersham) and histone H1 (Roche) as described (12). Products were resolved by SDS-PAGE. Radioactivity in histone H1 substrate was quantitated by phosphorimager from four different biological assays and graphed as % maximum activity.

Immunohistochemistry. Xenograft tumor tissues were formalin-fixed and embedded and deparaffinized samples were incubated with p27Kip1 antibody (Transduction Laboratories; 1:1,000), Ki-67 (DAKO; 1:100), and pSrc (Src-pY416; Stressgen; 1:100), respectively, as in refs. 4, 24 and processed in a DAKO autostainer using DAKO LSAB+ peroxidase detection kit. p27 and Ki-67 were scored as % tumor nuclei positive (4, 24). The predominant activated Src staining intensity was scored as follows: 0, no staining; 1, weak staining; 2, moderate staining; and 3, intense staining as in ref. 4. Each antigen was scored independently by two pathologists (M.J. and N.G.) in at least 20 high-power fields and the mean score for each was used in statistical analysis.

Reverse-phase protein lysate array. Reverse-phase protein lysate array (RPPA) as described previously (25, 26) was used to quantify expression of 86 proteins, including MEK, extracellular signal-regulated kinase 2, c-Jun NH2-terminal kinase, Akt, glycogen synthase kinase 3, mammalian target of rapamycin (mTOR), S6, p110α, β-catenin, ER-α, and cyclin D1, and phosphorylation of MEK at Ser217/Ser221, MAPK at Thr202 and Tyr204, c-Jun NH2-terminal kinase at Thr183 and Tyr185, AKT at Thr308 and Ser473, glycogen synthase kinase-3 at Ser21, mTOR at Ser2448, and S6 at Ser235/Ser236 using antibodies from Cell Signaling, Epitomics (total MEK and p110α antibodies), Lab Vision (total ER-α antibody), and Santa Cruz Biotechnology (total extracellular signal-regulated kinase 2, c-Jun NH2-terminal kinase, glycogen synthase kinase-3, and cyclin D1 antibodies). For a complete list of proteins assayed, see Supplementary Table S1.

Statistical analysis. Mean cyclin E-Cdk2 assays and immunohistochemistry scores for tumors in each treatment group were graphed as histograms and error bars show SE. Differences were considered significant when error bars do not overlap and when the two tailed Student's t test showed differences at P < 0.05.

Analysis of potential synergy between anastrozole and AZD0530 on tumor xenograft growth used the combination ratio as in ref. 27. Individual relative tumor volume (RTV) was calculated as (tumor volume on day of tumor measurement after treatment) / (tumor volume at initial treatment). The inhibition rate was calculated as (1.0 - final RTV after treatment / final RTV with androstenedione alone) × 100% (27).

The calculated combination ratio (27) evaluated combined effects. Fractional tumor volume (FTV) was defined as the ratio of mean final tumor volume of experimental drug-treated animals divided by the mean final tumor volume of the untreated control animals that received only androstenedione. The combination ratio compared the FTV expected if there were no synergy with the observed FTV. The combination ratio was calculated as (FTV of anastrozole × FTV of AZD0530) / observed FTV of combination. Observed and expected FTV are described as follows:

  • Expected FTV = (mean FTV of anastrozole) × (mean FTV of AZD0530)

  • Observed FTV = final tumor volume combined therapy / final tumor volume androstenedione alone

  • Combination ratio = expected FTV / observed FTV.

A combination ratio of >1 indicates a synergistic effect, whereas a ratio of <1 indicates a less than additive effect.

For statistical evaluation of RPPA data, serial dilution curves were constructed for each protein in each sample and relative quantification values were subsequently assigned to each protein in each sample as described (28). Each protein and phosphoprotein measurement was subsequently corrected for loading using the average expression of 86 measured proteins (Supplementary Table S1) in each sample. NCSS software was used to generate means plots for analyzed antibodies and ANOVA was used to calculate P values for each means plot. Xcluster and Treeview software were used to generate dendrograms after log transformation and mean centering of the protein quantification data as described (28).

AZD0530 increases G1 arrest by anastrozole in MCF-7Arom5 cells. To provide a model in which to test antitumor efficacy of aromatase inhibitors, MCF-7 cells were transfected with the human aromatase cDNA (17). MCF-7Arom5 expresses high aromatase levels (Fig. 1A). Estrogen deprivation impaired MCF-7Arom5 proliferation. Incubation of MCF-7Arom5 for 48 h in estrogen-depleted medium caused partial G0-G1 arrest, with a decrease in the % S phase from 43% to 14% (Fig. 1B). Addition of 25 nmol/L androstenedione stimulated proliferation, increasing the % S-phase cells to 52% by 48 h (Fig. 1B). Treatment with 100 μmol/L anastrozole inhibited the cell cycle progression, reducing the % S-phase cells from 52% to 22% and increasing the % G0-G1. The Src inhibitor, AZD0530 1 μmol/L alone, had little effect on cell cycle, and the % S-phase cells fell only modestly from 52% to 45%. Both drugs together caused G1 arrest within 48 h, with % S-phase cells falling to 12%, significantly lower than with either drug alone (see error bars in Fig. 1B).

Fig. 1.

Effects of anastrozole and AZD0530 on cell cycle. A, Western blot for aromatase in MCF-7, MCF-7 vector control, and MCF-7Arom5. B, asynchronous (Asyn) MCF-7Arom5 cells were estrogen deprived for 48 h and then treated with androstenedione alone or with AZD0530 1 μmol/L (SI), anastrozole 100 μmol/L (Ana), or both and then recovered for flow cytometry. Bars, SE. C, asynchronous MCF-7Arom5 cells were incubated for 48 h with androstenedione and treated with increasing concentrations of anastrozole without or with 1 μmol/L AZD0530 and then recovered for flow cytometry 48 h later. Mean % S-phase cells graphed for three repeat assays. Bars, SE.

Fig. 1.

Effects of anastrozole and AZD0530 on cell cycle. A, Western blot for aromatase in MCF-7, MCF-7 vector control, and MCF-7Arom5. B, asynchronous (Asyn) MCF-7Arom5 cells were estrogen deprived for 48 h and then treated with androstenedione alone or with AZD0530 1 μmol/L (SI), anastrozole 100 μmol/L (Ana), or both and then recovered for flow cytometry. Bars, SE. C, asynchronous MCF-7Arom5 cells were incubated for 48 h with androstenedione and treated with increasing concentrations of anastrozole without or with 1 μmol/L AZD0530 and then recovered for flow cytometry 48 h later. Mean % S-phase cells graphed for three repeat assays. Bars, SE.

Close modal

Cell cycle effects of increasing anastrozole concentrations were assayed with and without 1 μmol/L AZD0530, an AZD0530 dose that on its own had little cell cycle effect (Fig. 1B). In cells grown with androstenedione as the only estrogen source, anastrozole caused a dose-dependent G1 arrest over a 100-fold concentration range (Fig. 1C). Anastrozole 12.5 μmol/L for 48 h decreased the % S phase from 52% to 43%, and considerably higher doses were required to further inhibit cell cycle progression. With 1000 μmol/L anastrozole alone, the % S phase fell to 14% (Fig. 1C). Although 1 μmol/L AZD0530 had little effect on cell proliferation (Fig. 1B), this dose of AZD0530 increased the cell cycle inhibitory effect of anastrozole by 1 log. At each anastrozole concentration used between 12.5 and 1,000 μmol/L, addition of a noninhibitory dose of AZD0530 caused a more profound increase in % G1 and decrease in % S-phase cells than observed with either drug alone (see Fig. 1B and C). Thus, although anastrozole alone was a relatively weak cell cycle inhibitor, requiring high doses to cause G0-G1 arrest, it took 10-fold less anastrozole to arrest these cells when anastrozole was used together with the 1 μmol/L dose of AZD0530, which on its own had little effect on the cell cycle. Similar results were observed in another aromatase transfected cell line, MCF-7Ca (data not shown).

Effects of anastrozole and AZD0530 on signaling and cell cycle regulators. Effects of AZD0530 and anastrozole on mitogenic signaling were assayed. These assays used 100 μmol/L anastrozole because this caused a partial G1 arrest that was increased by addition of 1 μmol/L AZD0530 (Fig. 1B and C). When added to cells grown with androstenedione as the only estrogen source, anastrozole 100 μmol/L alone and AZD0530 1 μmol/L alone each modestly reduced pAkt. pAkt in dual-treated cells was 3-fold lower than in untreated cells and 2-fold lower than with either drug alone. Anastrozole alone had little effect on MAPK phosphorylation, whereas AZD0530 caused an unexpected increase in pMAPK that was inhibited by coadministration with anastrozole. AZD0530 decreased pSrc as expected, but anastrozole paradoxically increased pSrc within 48 h, whereas AZD0530 and anastrozole together caused Src inhibition. Densitometric analysis of band intensities reveals that combined treatment with anastrozole and AZD0530 significantly inhibited Src, Akt, and MAPK activities (Fig. 2A; Supplementary Fig. S1) without affecting total kinase levels.

Fig. 2.

Effects of anastrozole and Src inhibitor AZD0530 on signaling and cell cycle regulators. Cells were grown without estrogen in 5% charcoal-stripped fetal bovine serum supplemented with androstendione (no drug) and treated with no additional drug, AZD0530 (SI), anastrozole (Ana), or both (Ana + SI) for 48 h and then assayed for (A) pSrc, Src, pAkt, Akt, pMAPK, and MAPK levels and (B) p27, cyclin E, cyclin D1, Cdk2, Cdk4, and β-actin levels. C, cyclin E was immunoprecipitated and complexes were resolved and immunoblotted for associated proteins. D, cyclin E immunoprecipitates were assayed for associated kinase activity as per Materials and Methods. Radioactivity in histone H1 substrate was quantitated. Mean ± SE % maximum kinase activity from four repeat assays.

Fig. 2.

Effects of anastrozole and Src inhibitor AZD0530 on signaling and cell cycle regulators. Cells were grown without estrogen in 5% charcoal-stripped fetal bovine serum supplemented with androstendione (no drug) and treated with no additional drug, AZD0530 (SI), anastrozole (Ana), or both (Ana + SI) for 48 h and then assayed for (A) pSrc, Src, pAkt, Akt, pMAPK, and MAPK levels and (B) p27, cyclin E, cyclin D1, Cdk2, Cdk4, and β-actin levels. C, cyclin E was immunoprecipitated and complexes were resolved and immunoblotted for associated proteins. D, cyclin E immunoprecipitates were assayed for associated kinase activity as per Materials and Methods. Radioactivity in histone H1 substrate was quantitated. Mean ± SE % maximum kinase activity from four repeat assays.

Close modal

Assays of G1 cell cycle regulators showed p27 was increased by 1.1-fold with AZD0530 and by 1.5-fold with anastrozole alone, whereas both drugs together increased p27 by 3.5-fold over levels in asynchronous androstenedione-treated cells. Densitometric analysis showed that the increase in p27 was statistically significant in all treatment groups but most marked with both drugs together (see Supplementary Fig. S1). Cyclin D1, cyclin E, and Cdk2 levels were modestly decreased, whereas Cdk4 was unchanged by anastrozole plus AZD0530 (Fig. 2B).

Anastrozole and AZD0530 inhibit cyclin E-Cdk2 activity. After 48 h, p27 bound to cyclin E-Cdk2 increased by 1.2-fold following AZD0530 alone and by 2.4-fold following anastrozole alone (Fig. 2C) and by 4.8-fold with AZD0530 and anastrozole together. Although AZD0530 alone had little effect on cyclin E-Cdk2 activity, both drugs together caused a statistically significant kinase inhibition compared with that observed with either alone (Fig. 2D). Data show the mean radioactivity incorporated into the substrate of four repeat assays, graphed as a histogram showing % maximal activity ± SE (Fig. 2D).

Effects of anastrozole and AZD0530 on breast cancer xenograft growth. To test the antitumor efficacy of aromatase inhibitors with or without AZD0530 in vivo, MCF-7Arom5 xenografts were grown in ovariectomized mice. Nude mice are deficient in adrenal androgen precursor production and animals were supplemented with androstenedione. In these xenografts, intratumoral aromatase-mediated conversion of exogenous androstenedione to estrogen provides the only estrogen source. Thus, the tumor serves as an autocrine source of estrogen that stimulates tumor growth and is responsive to aromatase inhibitors (18). Androstenedione on its own, in the absence of transfected aromatase, does not affect MCF-7 xenograft growth (22).

MCF-7Arom5 xenografts were estrogen dependent for proliferation. In the absence of androstenedione, tumors grew very little over the 14-week experiment. In contrast, xenografts grew rapidly in androstenedione-treated animals. AZD0530 administration initially showed an antitumor effect, but within 12 to 14 weeks, resistance developed and final tumor volumes were similar to the androstenedione-alone controls (Fig. 3A). The experiment could not be extended beyond 14 weeks because tumors reached maximum size permitted. Anastrozole alone suppressed tumor growth significantly compared with androstenedione alone, with 0.25 mg/kg anastrozole yielding an inhibition rate [1 - (final RTV after treatment / final RTV with androstenedione alone) × 100] of 37.9% (see Table 1A and Fig. 3A). Combined anastrozole and AZD0530 therapy more potently inhibited tumor growth, yielding an inhibition rate of 53.3% (Fig. 3A).

Fig. 3.

Anastrozole and AZD0530 inhibit breast cancer growth in vivo. A, tumor growth curve graphs RTV over time in each treatment group. Mean ± SE. B, mean ± SE weight of animals in each treatment group graphed over time.

Fig. 3.

Anastrozole and AZD0530 inhibit breast cancer growth in vivo. A, tumor growth curve graphs RTV over time in each treatment group. Mean ± SE. B, mean ± SE weight of animals in each treatment group graphed over time.

Close modal
Table 1.
(A) Synergy between anastrozole and AZD0530 therapy on human breast cancer MCF-7Arom5 xenograft growth in nude mice
GroupDosageNo. mice
Tumor volume (mm3), mean ± SE
RTV (mean ± SD)Inhibition rate (%)*
InitialEndInitialEnd
Without androstenedione 74 ± 15 179 ± 39 2.7 ± 1.8  
Androstenedione 5 mg/kg/d, s.c. 71 ± 8 1,169 ± 138 18.2 ± 8.7  
AZD0530 50 mg/kg/d, intragastrically 74 ± 11 1,242 ± 219 16.8 ± 3.3 7.7 
Anastrozole 0.25 mg/kg/d, s.c. 65 ± 7 758 ± 168 11.3 ± 5.2 37.9 
Anastrozole + AZD0530 0.25 mg/kg/d, s.c. + 50 mg/kg/d, intragastrically 68 ± 6 565 ± 122 8.5 ± 3.3§ 53.3 
     
(B) Anastrozole and AZD0530 FTV relative to untreated control*
 
    
FTVanastrozole
 
FTVAZD0530
 
Combination
 

 

 

 

 
Expected FTV Observed FTV Combination ratio 
0.66 1.07 0.71 0.45 1.57 
(A) Synergy between anastrozole and AZD0530 therapy on human breast cancer MCF-7Arom5 xenograft growth in nude mice
GroupDosageNo. mice
Tumor volume (mm3), mean ± SE
RTV (mean ± SD)Inhibition rate (%)*
InitialEndInitialEnd
Without androstenedione 74 ± 15 179 ± 39 2.7 ± 1.8  
Androstenedione 5 mg/kg/d, s.c. 71 ± 8 1,169 ± 138 18.2 ± 8.7  
AZD0530 50 mg/kg/d, intragastrically 74 ± 11 1,242 ± 219 16.8 ± 3.3 7.7 
Anastrozole 0.25 mg/kg/d, s.c. 65 ± 7 758 ± 168 11.3 ± 5.2 37.9 
Anastrozole + AZD0530 0.25 mg/kg/d, s.c. + 50 mg/kg/d, intragastrically 68 ± 6 565 ± 122 8.5 ± 3.3§ 53.3 
     
(B) Anastrozole and AZD0530 FTV relative to untreated control*
 
    
FTVanastrozole
 
FTVAZD0530
 
Combination
 

 

 

 

 
Expected FTV Observed FTV Combination ratio 
0.66 1.07 0.71 0.45 1.57 

NOTE: Student's t test compared with androstenedione group.

*

Inhibition rate = (1.0 - final RTV after treatment / final RTV with androstenedione alone) × 100.

P < 0.001.

P < 0.05.

§

P < 0.01.

To test for interaction between anastrozole and AZD0530, the effect of dual therapy was evaluated by the combination ratio of expected over observed FTV (27). FTV is defined by the ratio of mean final tumor volume of experimental drug-treated animals divided by the mean final tumor volume of the untreated control animals (27). The combination ratio = (FTV of anastrozole × FTV of AZD0530) / (observed FTV of combination). A combination ratio of >1 indicates a synergistic effect between the two drugs. In the initial few weeks of the experiment, before the emergence of resistance to AZD0530, the effect of the two drugs was not synergistic. However, as resistance to AZD0530 monotherapy emerged, by 14 weeks, the ratio was 1.57 (see Table 1B), indicative of synergy between anastrozole and AZD0530. In all treatment groups, the animals showed no signs of toxicity or weight loss throughout the experiment (Fig. 3B). Coadministration of anastrozole with AZD0530 prevented the development of resistance to AZD0530 monotherapy and augmented the effect of anastrozole alone.

Effects of anastrozole and AZD0530 on proliferation and pSrc in vivo. In an effort to identify predictors of antitumor response, p27, pY416 phosphorylated activated Src (Src-pY416), and Ki-67 were assayed by immunohistochemistry in xenografts recovered at the end of the experiment. Scores were reached by consensus of two different pathologists. As observed in cell culture, xenograft p27 levels increased more in tumors treated with anastrozole and AZD0530 together than with each drug alone. Ki-67, a proliferation indicator (29), was not reduced by AZD0530, was significantly decreased by anastrozole alone, and was lowest after combination therapy compared with controls.

pSrc was highest in androstenedione-alone control tumors. Src was not significantly inhibited in the resistant tumors recovered at the end of AZD0530 therapy. As observed in tissue culture, anastrozole alone modestly increased Src phosphorylation. Anastrozole and AZD0530 combination therapy significantly decreased Src-pY416 (Fig. 4).

Fig. 4.

p27, Ki-67, and Src-pY416 levels in MCF-7Arom5 xenograft tumors. A to C, representative photomicrographs of p27, Ki-67, and Src-pY416 immunohistochemistry from tumors treated with either androstenedione alone (no drug) or androstenedione together with either AZD0530 (SI), anastrozole (Ana), or both drugs (SI + Ana) together (×40). p27, Ki-67, and Src-pY416 immunohistochemistry scores are graphed for each treatment (mean ± SE). *, P < 0.05; **, P < 0.01, statistically different from androstenedione alone (Student's t test).

Fig. 4.

p27, Ki-67, and Src-pY416 levels in MCF-7Arom5 xenograft tumors. A to C, representative photomicrographs of p27, Ki-67, and Src-pY416 immunohistochemistry from tumors treated with either androstenedione alone (no drug) or androstenedione together with either AZD0530 (SI), anastrozole (Ana), or both drugs (SI + Ana) together (×40). p27, Ki-67, and Src-pY416 immunohistochemistry scores are graphed for each treatment (mean ± SE). *, P < 0.05; **, P < 0.01, statistically different from androstenedione alone (Student's t test).

Close modal

MEK and PI3K pathways are activated in AZD0530-treated xenografts. Proteomic analysis of 86 proteins using RPPA including apoptotic markers, X-linked inhibitor of apoptosis, cleaved caspase, and cleaved poly (ADP-ribose) polymerase showed no evidence for apoptosis in any treatment groups. Consistent with results in tissue culture (Fig. 2B) and effects on p27 and Ki-67 by immunohistochemistry (Fig. 4), xenograft cyclin D1 levels were reduced by each drug alone and most notably by AZD0530/anastrozole treatment compared with androstenedione-alone controls (P = 0.01). Dual therapy also reduced levels of phospho-activated epidermal growth factor receptor p922 (P = 0.07).

As shown in the heat maps and means plots in Fig. 5, RPPA analysis showed tumors treated with Src inhibitor alone (condition C) or in combination with anastrozole (condition D) had increased phosphorylation of components of the MEK and PI3K signaling pathways compared with untreated controls. AZD0530-resistant xenografts showed increased pMEK, pMAPK, and p110, activation of Akt, mTOR, and S6 kinase, and inhibitory phosphorylation of glycogen synthase kinase-3β compared with androstendione- or anastrozole-alone groups. β-Catenin levels were increased, in keeping with the known effect of glycogen synthase kinase-3β inhibition to stabilize β-catenin (30). This may reflect activation of these pathways after long-term Src inhibitor therapy and could contribute to resistance to the antitumor effects of AZD0530.

Fig. 5.

RPPA was used to quantify expression of the indicated proteins in the MEK (A) and PI3K (B) pathways in MCF-7Arom5 xenografts treated with androstenedione alone (A) or together with anastrozole (B), AZD0530 (C), or both anastrozole and AZD0530 (D). In the heat map, green, reduced expression levels; red, higher expression levels.

Fig. 5.

RPPA was used to quantify expression of the indicated proteins in the MEK (A) and PI3K (B) pathways in MCF-7Arom5 xenografts treated with androstenedione alone (A) or together with anastrozole (B), AZD0530 (C), or both anastrozole and AZD0530 (D). In the heat map, green, reduced expression levels; red, higher expression levels.

Close modal

Aromatase inhibitors have had a major effect on breast cancer treatment and are more effective than tamoxifen as first-line therapy for metastatic ER-positive breast cancers and as adjuvant therapy for hormone-responsive postmenopausal disease (1, 2). However, de novo and acquired resistance limit therapeutic efficacy of aromatase inhibitors (31). Oncogenic activation of various growth factor pathways can lead to antiestrogen resistance (31, 32). Given the complexity of mitogenic signaling in ER-positive breast cancer, monotherapy with signal transduction inhibitors may have only modest antitumor efficacy and prove less effective than endocrine therapy (31). Combined endocrine/signal transduction inhibitor therapy may cause greater growth inhibition and delay emergence of resistance. Several preclinical and clinical trials are now examining signal transduction inhibitors combined with aromatase inhibitors in an effort to block crosstalk between activated ER and signaling pathways and oppose resistance (3134).

Src is frequently activated in human breast cancers and high pSrc levels were observed in 37% of ER-positive breast cancers (4). Estrogen binding to the ER recruits and activates Src leading to MEK/MAPK and PI3K/Akt activation (79). Moreover, oncogenic activation of insulin-like growth factor-I receptor or the ErbB family, through ErbB1/3 overexpression or ErbB2 gene amplification, leads to constitutive Src activation and Src, in turn, further activates these receptor tyrosine kinases (6, 35). Activation of these signaling kinases modulates cell cycle regulators to stimulate cell cycle progression (12), and activated kinases phosphorylate ER-α to positively regulate its transcriptional activity (36). Up-regulation of Src mediates antiestrogen resistance in culture models (37).

The cell cycle inhibitor p27 is ubiquitously expressed and regulates cell proliferation (38). Although normal quiescent mammary epithelial tissues express high p27 (24, 39), p27 protein is reduced in up to 60% of primary human breast and this is associated with poor patient outcome (24, 3840). Estrogen stimulation of estrogen-deprived cells mediates loss of p27, leading to cyclin E-Cdk2 activation and cell cycle entry (11, 12, 14, 41). p27 and the related Cdk2 inhibitor, p21, are both required for G1 arrest by antiestrogens in human breast cancer lines (12, 15). Tyrosine phosphorylation of p27 by Abl and Src family kinases impairs the Cdk2 inhibitory action of p27 in vitro and promotes p27 proteolysis (4, 42). Src-mediated p27 phosphorylation at Y74, Y88, and Y89 led to reduced binding to cyclin E-Cdk2, loss of inhibition of cyclin E-Cdk2 by p27, and facilitated cyclin E-Cdk2-mediated T187 phosphorylation of p27 to drive p27 proteolysis. Src induction reduced the p27 t1/2, whereas Src inhibition by AZD0530 increased p27 stability (4). These findings provided the molecular rationale for the current preclinical study. Because p27 is required for the therapeutic growth-inhibitory effects of estrogen deprivation or ER blockade (12), and because Src activates p27 proteolysis (4), we reasoned that Src inhibitors may oppose antiestrogen resistance and could potentially cooperate with antiestrogens to augment antitumor efficacy.

AZD0530 is a highly selective, orally available Abl and Src family kinase inhibitor (43). It showed potent antiproliferative activity (below micromolar IC50) in over 17 human cancer lines and inhibited growth of 6 of 13 xenograft tumor models tested. It is currently in phase II clinical trials for cancer. AZD0530 can cooperate with tamoxifen to arrest proliferation of resistant ER-positive breast cancer lines (4, 44). AZD0530 and tamoxifen synergize to inhibit anchorage-independent growth in vitro (45). The growth-inhibitory effect of AZD0530 is opposed by estrogen, and Src inhibitors may require ER-α blockade for efficacy in estrogen-dependent breast cancer (45).

The present data indicate that, as for ER-blocking drugs (12), the aromatase inhibitor anastrozole up-regulates p27 protein and p27 inhibits cyclin E-Cdk2 to arrest MCF-7Arom5 cells in G1. AZD0530-anastrozole more effectively increased p27 levels and binding and inhibition of cyclin E-Cdk2 by p27 than either drug alone, causing a greater cell cycle arrest. Although the increase in p27 was modest (3.5-fold), this is sufficient for stoichiometric binding and inhibition of cyclin E-Cdk2 (46). The reduction in cyclin D1 levels observed with AZD0530-anastrozole likely reflects a loss of protein stability as cells enter G1 arrest (47) and reduced CCND1 transcription (30).

The present data suggest potential synergy between anastrozole and AZD0530 both in cell culture and in xenograft tumors. In cell culture, addition of 1 μmol/L AZD0530, which on its own had little antiproliferative effect, decreased by 10-fold the anastrozole concentration required for proliferation arrest. Although AZD0530 alone had little antiproliferative effect, this Src inhibitor appears to cooperate with estrogen deprivation by anastrozole to block cell cycle. This may, in part, result from abrogation by AZD0530 of the stimulation of Src seen with anastrozole alone in vitro and in xenografts. In xenografts treated with dual therapy, p27 levels were significantly increased and Src-pY416, cyclin D1, and the proliferation marker Ki-67 were reduced when compared with androstenedione-alone controls, supporting further study of these markers as indicators of treatment response in clinical trials. Ki-67 has been widely used to assess cancer prognosis. A change in the expression of Ki-67 after exposure of patients to therapeutic agents may represent a pharmacodynamic marker of efficacy. Recent neoadjuvant studies suggest that on-treatment measurement of Ki-67 may be a more effective predictor of treatment efficacy for both endocrine treatment and chemotherapy (48). It will be of interest to further evaluate whether changes between pretreatment and post-treatment p27 and Ki-67 levels are predictive of patient response in the context of clinical trials.

Although AZD0530 initially delayed xenograft growth compared with androstenedione-only controls, tumors rapidly acquired AZD0530 resistance. Resistance to AZD0530 was also observed in breast cancer lines after prolonged (>10 weeks) treatment in vitro (44). In contrast to AZD0530 resistance seen in cell culture, in which Src remained inhibited (44), resistant xenografts showed little Src inhibition after 14 weeks of AZD0530. Although increased AZD0530 metabolism or accelerated drug expulsion from xenografts could contribute to resistance, our proteomic analysis showed that prolonged AZD0530 therapy was associated with activation of bypass pathways not seen in untreated (androstendione-alone) controls. AZD0530 treatment alone increased pMAPK within 48 h in tissue culture and AZD0530-resistant xenografts recovered after prolonged treatment showed both MEK/MAPK and PI3K/Akt/mTOR pathway activation. RPPA analysis of xenograft tumors showed that HER-2 and epidermal growth factor receptor activities (which would activate MEK and PI3K) were also increased by AZD0530 alone but were reduced by the drug combination. Both PI3K/Akt/mTOR and MEK/MAPK pathways could mediate constitutive Src activation, competing with the effect of AZD0530 on its target. Further investigation is warranted to test if MEK or PI3K/AKT/mTOR pathway inhibitors could abort development of resistance to AZD0530 monotherapy.

In human cancers, both MEK/MAPK and PI3K/Akt alter p27 phosphorylation and oppose the Cdk2 inhibitory effects of p27 (30). Transfection of activated MEK reduces the cyclin E-Cdk2 inhibitory activity of p27 (49, 50) and, in some cell types, activates p27 proteolysis (49). Moreover, constitutive Akt activation shifts p27 into the cytoplasm away from nuclear Cdk targets, leading to loss of p27 binding to cyclin E-Cdk2 (38, 51). Thus, both MEK and PI3K bypass pathway activation could abort p27 function causing resistance to coadministered anastrozole during prolonged AI-AZD0530 therapy in humans.

Because estrogen/ER stimulates Src and MAPK, the estrogen deprivation resulting from anastrozole treatment was expected to block MAPK and Src activation in our studies. Used alone, anastrozole had little inhibitory effect on pMAPK in culture and but caused a more important MAPK inhibition in xenografts. Although anastrozole monotherapy caused a partial G1 arrest in cell culture and had an antiproliferative effect with reduced Ki-67 and increased p27 in treated tumors, paradoxically, anastrozole appears to activate Src in MCF-7Arom5 cells both in cell culture and after prolonged monotherapy in xenografts. AZD0530 combined with anastrozole effectively inhibited Src, MEK/MAPK, and Akt activation in cell culture and in xenografts and caused a sustained inhibition of xenograft growth. Src activation by anastrozole may constitute one mechanism leading to anastrozole resistance in vivo and supports further investigation of the therapeutic potential of combined aromatase and Src inhibitor therapy. The combination of the two drugs together appears to overcome the activation of the bypass pathways observed with either treatment alone in these models.

In summary, the combination of AZD0530 and anastrozole more effectively inhibits proliferation of ER-positive MCF-7Arom5 breast cancer cells than either drug alone both in vitro and in vivo. These data support further clinical investigation of the therapeutic potential of AZD0530 in combination with anastrozole for treatment of postmenopausal ER-positive breast cancer. Both PI3K and MEK pathways were activated following prolonged AZD0530 treatment but not by androstendione alone in xenograft tumors. Proteomic analysis of xenografts may permit the preclinical identification of putative predictive markers and reveal potential resistance mechanisms that can be further evaluated in the context of clinical trials.

No potential conflicts of interest were disclosed.

Grant support: Breast Cancer Research Foundation and Flight Attendants Medical Research Institute (J.M. Slingerland) and Avon Foundation-AACR International Scholar Awards in Breast Cancer (Y. Chen).

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

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

We thank the laboratory members and clinical colleagues, Stefan Gluck and Mark Pegram, for helpful discussions during this work as well as Sarath Dhananjayan for technical expertise in formatting the figures.

1
Ellis MJ. Neoadjuvant endocrine therapy for breast cancer: medical perspectives.
Clin Cancer Res
2001
;
7
:
4388
–91s.
2
Smith IE, Dowsett M. Aromatase inhibitors in breast cancer.
N Engl J Med
2003
;
348
:
2431
–42.
3
Ishizawar R, Parsons SJ. c-Src and cooperating partners in human cancer.
Cancer Cell
2004
;
6
:
209
–14.
4
Chu I, Sun J, Arnaout A, et al. p27 phosphorylation by Src regulates inhibition of cyclin E-Cdk2.
Cell
2007
;
128
:
281
–94.
5
Bild AH, Yao G, Chang JT, et al. Oncogenic pathway signatures in human cancers as a guide to targeted therapies.
Nature
2006
;
439
:
353
–7.
6
Belsches-Jablonski AP, Biscardi JS, Peavy DR, Tice DA, Romney DA, Parsons SJ. Src family kinases and HER2 interactions in human breast cancer cell growth and survival.
Oncogene
2001
;
20
:
1465
–75.
7
Castoria G, Migliaccio A, Bilancio A, et al. PI3-kinase in concert with Src promotes the S-phase entry of oestradiol-stimulated MCF-7 cells.
EMBO J
2001
;
20
:
6050
–9.
8
Migliaccio A, DiDomenico M, Castona C, et al. Tyrosine kinas/p21ras/MAP-kinase pathway activation by estradiol receptor complex in MCF-7 cells.
EMBO J
1996
;
15
:
1292
–300.
9
Coleman KM, Smith CL. Intracellular signaling pathways: nongenomic actions of estrogens and ligand-independent activation of estrogen receptors.
Front Biosci
2001
;
6
:
D1379
–91.
10
Arnold SF, Vorojeikina DP, Notides AC. Phosphorylation of tyrosine 537 on the human estrogen receptor is required for binding to an estrogen response element.
J Biol Chem
1995
;
270
:
30205
–12.
11
Planas-Silva MD, Weinberg RA. Estrogen-dependent cyclin E-cdk2 activation through p21 redistribution.
Mol Cell Biol
1997
;
17
:
4059
–69.
12
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 U S A
2000
;
97
:
9042
–6.
13
Prall OW, Rogan EM, Sutherland RL. Estrogen regulation of cell cycle progression in breast cancer cells.
J Steroid Biochem Mol Biol
1998
;
65
:
169
–74.
14
Foster J, Wimalasen J. Estrogen regulates activity of cyclin-dependent kinases and retinoblastoma protein phosphorylation in breast cancer cells.
Mol End
1996
;
10
:
488
–96.
15
Carroll JS, Prall OW, Musgrove EA, Sutherland RL. A pure estrogen antagonist inhibits cyclin E-Cdk2 activity in MCF-7 breast cancer cells and induces accumulation of p130-2F4 complexes characteristic of quiescence.
J Biol Chem
2000
;
275
:
38221
–9.
16
Song RX, Barnes CJ, Zhang ZG, Bao YD, Kumar R, Santen RJ. The role of Shc and insulin-like growth factor 1 receptor in mediating the translocation of estrogen receptor α to the plasma membrane.
Proc Natl Acad Sci U S A
2004
;
101
:
2076
–81.
17
Evans CT, Ledesma DB, Schulz TZ, Simpson ER, Mendelson CR. Isolation and characterization of a complementary DNA specific for human aromatase-system cytochrome P-450 mRNA.
Proc Natl Acad Sci U S A
1986
;
83
:
6387
–91.
18
Jelovac D, Macedo L, Handratta V, et al. Effects of exemestane and tamoxifen in a postmenopausal breast cancer model.
Clin Cancer Res
2004
;
10
:
7375
–81.
19
Weinberg OK, Marquez-Garban DC, Fishbein MC, et al. Aromatase inhibitors in human lung cancer therapy.
Cancer Res
2005
;
65
:
11287
–91.
20
Thiantanawat A, Long BJ, Brodie AM. Signaling pathways of apoptosis activated by aromatase inhibitors and antiestrogens.
Cancer Res
2003
;
63
:
8037
–50.
21
Xanthopoulos JM, Romano AE, Majumdar SK. Response of mouse breast cancer cells to anastrozole, tamoxifen, and the combination.
J Biomed Biotechnol
2005
;
2005
:
10
–9.
22
Yue W, Zhou DJ, Chen SA, Brodie A. A new nude-mouse model for postmenopausal breast-cancer using Mcf-7 cells transfected with the human aromatase gene.
Cancer Res
1994
;
54
:
5092
–5.
23
Lu Q, Yue W, Wang J, Liu Y, Long B, Brodie A. The effects of aromatase inhibitors and antiestrogens in the nude mouse model.
Breast Cancer Res Treat
1998
;
50
:
63
–71.
24
Catzavelos C, Bhattacharya N, Ung YC, et al. Decreased levels of the cell-cycle inhibitor p27Kip1 protein: prognostic implications in primary breast cancer.
Nat Med
1997
;
3
:
227
–30.
25
Tibes R, Qiu Y, Lu Y, et al. Reverse phase protein array: validation of a novel proteomic technology and utility for analysis of primary leukemia specimens and hematopoietic stem cells.
Mol Cancer Ther
2006
;
5
:
2512
–21.
26
Hennessy BT, Lu YL, Poradosu E, et al. Pharmacodynamic markers of perifosine efficacy.
Clin Cancer Res
2007
;
13
:
7421
–31.
27
Dings RP, Yokoyama Y, Ramakrishnan S, Griffioen AW, Mayo KH. The designed angiostatic peptide anginex synergistically improves chemotherapy and antiangiogenesis therapy with angiostatin.
Cancer Res
2003
;
63
:
382
–5.
28
Hu J, He X, Baggerly KA, Coombes KR, Hennessy BTJ, Mills GB. Non-parametric quantification of protein lysate arrays.
Bioinformatics
2007
;
23
:
1986
–94.
29
Scholzen T, Gerdes J. The Ki-67 protein: from the known and the unknown.
J Cell Physiol
2000
;
182
:
311
–22.
30
Liang J, Slingerland JM. Multiple roles of the PI3K/PKB (Akt) pathway in cell cycle progression.
Cell Cycle
2003
;
2
:
339
–45.
31
Johnston SR, Dowsett M. Aromatase inhibitors for breast cancer: lessons from the laboratory.
Nat Rev Cancer
2003
;
3
:
821
–31.
32
Chu I, Blackwell K, Chen S, Slingerland J. The dual ErbB1/ErbB2 inhibitor, lapatinib (GW572016), cooperates with tamoxifen to inhibit both cell proliferation- and estrogen-dependent gene expression in antiestrogen-resistant breast cancer.
Cancer Res
2005
;
65
:
18
–25.
33
Johnston SR, Martin LA, Leary A, Head J, Dowsett M. Clinical strategies for rationale combinations of aromatase inhibitors with novel therapies for breast cancer.
J Steroid Biochem Mol Biol
2007
;
106
:
180
–6.
34
Johnston SR, Martin LA, Head J, Smith I, Dowsett M. Aromatase inhibitors: combinations with fulvestrant or signal transduction inhibitors as a strategy to overcome endocrine resistance.
J Steroid Biochem Mol Biol
2005
;
95
:
173
–81.
35
Tan M, Li P, Klos KS, et al. ErbB2 promotes Src synthesis and stability: novel mechanisms of Src activation that confer breast cancer metastasis.
Cancer Res
2005
;
65
:
1858
–67.
36
Kato S, Endoh H, Masuhiro Y, et al. Activation of the estrogen receptor through phosphorylation by mitogen- activated protein kinase.
Science
1995
;
270
:
1491
–4.
37
Hiscox S, Morgan L, Green TP, Barrow D, Gee J, Nicholson RI. Elevated Src activity promotes cellular invasion and motility in tamoxifen resistant breast cancer cells. Breast Cancer Res Treat 2005;1–12.
38
Chu IM, Hengst L, Slingerland JM. The Cdk inhibitor p27 in human cancer: prognostic potential and relevance to anticancer therapy.
Nat Rev Cancer
2008
;
8
:
253
–67.
39
Porter PL, Malone KE, Heagerty PJ, et al. Expression of cell cycle regulators p27kip1 and cyclin E, alone and in combination, correlate with survival in young breast cancer patients.
Nat Med
1997
;
3
:
222
–5.
40
Tan P, Cady B, Wanner M, et al. The cell cycle inhibitor p27 is an independent prognostic marker in small (T1a,b) invasive breast carcinomas.
Cancer Res
1997
;
57
:
1259
–63.
41
Prall OWJ, Sarcevic B, Musgrove EA, Watts CKW, Sutherland RL. Estrogen induced activation of Cdk4 and Cdk2 during G1-S phase progression is accompanied by increased cyclin D1 expression and decreased cyclin dependent inhibitor association with cyclin E- Cdk2.
J Biol Chem
1997
;
272
:
10882
–94.
42
Grimmler M, Wang Y, Mund T, et al. Cdk-inhibitory activity and stability of p27Kip1 are directly regulated by oncogenic tyrosine kinases.
Cell
2007
;
128
:
269
–80.
43
Ple PA, Green TP, Hennequin LF, et al. Discovery of a new class of anilinoquinazoline inhibitors with high affinity and specificity for the tyrosine kinase domain of c-Src.
J Med Chem
2004
;
47
:
871
–87.
44
Hiscox S, Jordan NJ, Smith C, et al. Dual targeting of Src and ER prevents acquired antihormone resistance in breast cancer cells. Breast Cancer Res Treat 2008. Epub 2008 May 21. doi:10.1007/s10549-008-0058-6.
45
Herynk MH, Beyer AR, Cui Y, et al. Cooperative action of tamoxifen and c-Src inhibition in preventing the growth of estrogen receptor-positive human breast cancer cells.
Mol Cancer Ther
2006
;
5
:
3023
–31.
46
Reynisdottir I, Polyak K, Iavarone A, Massague J. Kip/Cip and Ink4 Cdk inhibitors cooperate to induce cell cycle arrest in response to TGF-β.
Genes Dev
1995
;
9
:
1831
–45.
47
Bates S, Parry D, Bonetta L, Vousden K, Dickson C, Peters G. Absence of cyclin D/cdk complexes in cells lacking functional retinoblastoma protein.
Oncogene
1994
;
9
:
1633
–40.
48
Dowsett M, Dunbier AK. Emerging biomarkers and new understanding of traditional markers in personalized therapy for breast cancer.
Clin Cancer Res
2008
;
14
:
8019
–26.
49
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
–95.
50
Cheng M, Sexl V, Sherr CJ, Roussel MF. Assembly of cyclin D-dependent kinase and titration of p27Kip1 regulated by mitogen-activated protein kinase kinase (MEK1).
Proc Natl Acad Sci U S A
1998
;
95
:
1091
–6.
51
Liang J, Zubovitz J, Petrocelli T, et al. PKB/Akt phosphorylates p27, impairs nuclear import of p27 and opposes p27-mediated G1 arrest.
Nat Med
2002
;
8
:
1153
–60.

Supplementary data