当前位置: 首页 > 期刊 > 《肿瘤与内分泌》 > 2005年第2期 > 正文
编号:11330208
Clinical trials of intracellular signal transductions inhibitors for breast cancer — a strategy to overcome endocrine resistance
http://www.100md.com 《肿瘤与内分泌》
     Department of Medicine, Royal Marsden Hospital, London SW3 6JJ, UK

    This paper was presented at the 1st Tenovus/AstraZeneca Workshop, Cardiff (2005). AstraZeneca has supported the publication of these proceedings.

    Abstract

    Acquired resistance to endocrine therapy in breast cancer is associated with an increase in peptide growth factor signaling that results in cross-talk activation of the estrogen receptor (ER). Small molecule signal transduction inhibitors (STIs) can target components of these intracellular pathways, and may prove effective in anticancer therapy. However, early phase II clinical trials with various STIs as monotherapy in advanced breast cancer have shown only a modest level of efficacy for these intracellular inhibitors. Preclinical data suggest that combinations of tamoxifen with STIs may provide significantly greater growth inhibition than either therapy alone, and, furthermore, may delay the emergence of endocrine resistance. There are now several trials assessing the efficacy of combinations of small molecule tyrosine kinase inhibitors (TKIs), such as gefitinib and lapatinib, with either tamoxifen or aromatase inhibitors both in the second-line, endocrine-resistant and first-line, hormone-sensitive setting. Similar trials continue with both farnesyltransferase inhibitors (FTIs) and mTOR antagonists, where there are strong preclinical data to suggest additive or synergistic effects for either of these agents in combination with endocrine therapies. Biomarker studies in the presurgical setting are also being utilized as an alternative approach to investigate whether combined endocrine/STI therapy is an effective clinical strategy. This article reviews some of the preclinical evidence supporting this strategy, together with the current status of clinical trials in this area.

    Introduction

    Research into the mechanisms of endocrine resistance in breast cancer has revealed that various growth factor pathways and oncogenes involved in the signal transduction cascade become activated and utilized by breast cancer cells to bypass normal endocrine responsiveness (Nicholson et al. 1999). Significant clinical attention in recent years has focused on the monoclonal antibody trastuzumab (Herceptin), which targets the extracellular domain of the type 1 growth factor receptor, human epidermal growth factor receptor 2 (HER-2), an oncogene that is amplified in 20% of human breast carcinomas (Slamon et al. 2002). However, HER-2 signaling involves the activation of various downstream intracellular kinases, many of which are activated or oncogeneic in their own right and may override control by upstream membrane receptors such as HER-2. As such, these intracellular pathways represent attractive targets for pharmacologic intervention with small molecule signal transduction inhibitors (STIs) that target aberrantly or excessively expressed oncogene products. Many such drugs are in active development for breast cancer, including type 1 growth factor tyrosine kinase inhibitors, farnesyltransferase inhibitors (FTIs), MEK inhibitors and mTOR antagonists (Table 1).

    Preclinical efficacy of intracellular STIs in breast cancer

    Enhanced expression of type I growth factor receptors such as epidermal growth factor receptor (EGFR) and HER-2, together with subsequent downstream activation of signaling pathways regulated by the mitogen-activated protein kinase/extracellular signal-related kinase (MAPK/ERK), has been found in breast cancer cells that become resistant over time to endocrine therapy either with tamoxifen (Knowlden et al. 2003) or by long-term estrogen deprivation (LTED) (Jeng et al. 2000, Martin et al. 2003). Treatment with various STIs has been used in preclinical models in an attempt to overcome this resistance by blocking upregulated signaling pathways (Nicholson et al. 2004). For example, in MCF-7 cells that developed resistance to tamoxifen, both gefitinib, which targets the internal tyrosine kinase domain of EGFR, and trastuzumab, which blocks the external domain of HER-2, were effective at reducing downstream ERK1/2 MAPK signaling and inhibiting cell growth (Knowlden et al. 2003). EGFR and HER-2 heterodimerize in the resistant cells, such that targeting either one of the receptors can be an effective therapy. Of note, hormone-sensitive cells (in which neither receptors are overexpressed) were unaffected by either gefitinib or trastuzumab therapy. Similar data have been reported by other groups in tamoxifen-resistant, HER-2-transfected MCF-7 cells with AG1478, a HER-2 tyrosine kinase inhibitor, and with trastuzumab (Kurokawa et al. 2000, Witters et al. 2002). Likewise, in cells resistant to LTED, both growth and estrogen receptor (ER)-mediated gene transcription was abrogated by a number of different intracellular approaches to interrupt signaling, including the tyrosine kinase inhibitor gefitinib, the MEK inhibitor UO126 and the ER downregulator fulvestrant, which degrades residual ER (Martin et al. 2003). Several groups have shown that these different STIs may also inhibit the growth of breast cancer xenograft tumors in vivo (Kurokawa et al. 2000, Shou et al. 2004).

    Other intracellular signaling and cell survival pathways are also activated in hormone-resistant breast cancer, particularly the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Akt (or PKB) is a serine/threonine kinase that promotes cell survival and is activated in response to many different growth factors, including insulin, insulin-like growth factor 1 (IGF-1), basic fibroblast growth factor (bFGF), EGF, heregulin and vascular endothelial growth factor (VEGF). Once activated, Akt exerts antiapoptotic effects through phosphorylation of substrates that directly regulate the apoptotic machinery (i.e., Bad, caspase 9). In addition, the mammalian target of rapamycin (mTOR) is a down-stream effector of the PI3K/Akt signaling pathway that activates p70S6 kinase and 4E-binding protein-1, which, in turn, regulate transition through the G1/S phase of the cell cycle. Approaches to targeting these cell survival pathways have included either specific PI3K inhibitors, such as LY294002, or rapamycin analogues, such as temsirolimus (CCI-779) or everolimus (RAD-001), that target mTOR. Breast cancer cell lines with activated Akt (as by loss of the regulatory PTEN tumor suppressor gene) are especially sensitive to mTOR antagonism (Yu et al. 2001).

    Enhanced anticancer effects for STIs in combination with endocrine therapy

    Several preclinical reports have implied that, in hormone-sensitive, ER-positive breast cancer, STIs as monotherapy may have only a minimal effect on tumor growth, especially if cells lack the activation and dependence of the various signal transduction pathways described above. Emerging evidence suggests that adaptive changes occur during prolonged endocrine therapy, particularly upregulation of growth factor signaling. Thus, strategies to combine endocrine with STI therapies have been used as a means to prevent development of resistance and improve therapeutic efficacy. In vitro, combined tamoxifen and gefitinib provided nearly complete inhibition of phosphorylated ERK1/2 MAPK and Akt, together with greater G0/G1 cell-cyle arrest and suppression of the cell-survival protein bcl-2 than that observed with just tamoxifen (Gee et al. 2003). In particular, combined therapy prevented the acquired expression of EGFR/MAPK signaling and the subsequent resistance that occurred after 5 weeks in tamoxifen-alone-treated cells.

    For established hormone-resistant, HER-2-positive breast cancer, the strategy of combined STIs and endocrine therapy may also be more effective than using STIs alone (Kurokawa et al. 2000). In vivo, gefitinib and tamoxifen provided maximal growth inhibition and significantly delayed the growth of HER-2-positive MCF-7 xenografts compared with gefitinib alone (Shou et al. 2004). Moreover, similar effects were seen with gefitinib combined with estrogen deprivation, which provided greater inhibition of growth and substantially delayed acquired resistance compared with estrogen deprivation alone (Massaraweh et al. 2002). A synergistic effect has also been reported for trastuzumab combined with tamoxifen in ER-positive, HER-2-positive BT-474 breast cancer cells, with enhanced accumulation of cells in G0/G1 and reduction in S phase of the cell cycle compared with either therapy alone (Argiris et al. 2004). Of interest, there was no evidence for any induction of apoptosis. Recently, the dual EGFR/HER-2 inhibitor lapatinib has been shown to cooperate with tamoxifen to provide more rapid and profound cell-cycle arrest than either therapy alone in hormone-resistant cells (Chu et al. 2005). The two drugs together caused a greater reduction in cyclin D1, together with a greater increase in the kinase inhibitor p27 and cyclin E-cdk2 inhibition, in various tamoxifen-resistant breast cancer cell lines. Lapatinib was able to restore tamoxifen sensitivity in EGFR- or HER-2-expressing cells, while, in vivo, combined laptaninib and tamoxifen caused maximal regression of HER-2-overexpressing, tamoxifen-resistant MCF-7 xenografts (Chu et al. 2005).

    Other STIs that have only a minimal effect on hormone-sensitive breast cancer may also be more effective when combined with endocrine therapy. The FTI tipifarnib inhibits the growth of a number of human breast cancer cells lines in vitro, most of which contain normal wild-type ras genes (End et al. 2001). However, in vivo, tipifarnib produced only a modest cytostatic effect on hormone-sensitive MCF-7 xenograft growth, with evidence of induction of apoptosis and enhanced expression of the cell-cycle inhibitory protein p21 (Kelland et al. 2001). In contrast, when tipifarnib was combined with tamoxifen or estrogen deprivation therapy, combined treatment induced significantly greater tumor regression than either endocrine therapy alone (Johnston et al. 2002). Three other groups have since reported a similar interaction for FTIs with tamoxifen or aromatase inhibitors, and have suggested either a synergistic (Ellis et al. 2003) or an additive antitumor effect (Long et al. 2004). One recent study implied an additive effect on G0/G1 cell-cycle arrest, and that the FTI-277, when combined with tamoxifen, maintained higher levels of the cdk inhibitor p21waf/cip1, resulting in an additive effect on inactivation of cyclin E/Cdk2 complexes and decreased phosphorylation of pRb (Doisneau-Sixou et al. 2003).

    More recently, a similar rationale has emerged to support the combination of mTOR antagonists with either tamoxifen or an aromatase inhibitor in preclinical models of ER-positive, hormone-sensitive and resistant breast cancer (DeGraffenried et al. 2004, Rudolf et al. 2004). The estrogen-dependent growth of both wild-type MCF7 and aromatase-expressing (MCF7/Aro) breast cancer cells was inhibited in a dose-dependent manner by the mTOR antagonist everolimus (RAD-001), suggesting that mTOR signaling is required for the estrogen-dependent proliferation of these cells. In subsequent experiments with the MCF7/Aro cells, the combination of letrozole and everolimus produced maximal growth inhibition with clear evidence for additive/synergistic effects (Rudolf et al. 2004). Evidence has emerged that activation of Akt/PKB and the downstream mTOR pathway can cause resistance to tamoxifen (Clark et al. 2002, Pancholi et al. 2004). MCF-7 cells expressing a constitutively active Akt were able to proliferate under reduced estrogen conditions and were resistant to the growth inhibitory effects of tamoxifen, both in vitro and in vivo, in xenograft models (DeGraffenried et al. 2004). However, cotreatment with the mTOR inhibitor temsirolimus (CCI-779) inhibited mTOR activity and restored sensitivity to tamoxifen, primarily through induction of apoptosis, thus suggesting that Akt-induced tamoxifen resistance may in part be mediated by signaling through the mTOR pathway.

    Clinical trials with intracellular STIs in breast cancer

    Taken together, these experimental data indicate that various intracellular signaling pathways may be activated or overexpressed in breast cancer, especially in endocrine-resistant cells, and suggest that targeting such pathways may be an effective therapy. Proof of principle for therapies that target growth factor pathways in breast cancer has already been provided with trastuzumab (Herceptin), the monoclonal antibody to the external domain of HER-2. Clinical use of trastuzumab has been exclusively in HER-2-positive breast cancer, and significant activity has been seen as first-line monotherapy, with up to a 48% clinical benefit rate in HER-2-positive tumors, including a 34% objective response rate in tumors reported positive by fluorescence in situ hybridization (FISH) (Vogel et al. 2002). Thus, it was hoped that various small molecule STIs may also prove effective anticancer strategies in this setting. Early phase II clinical studies were initiated with EGFR/HER-2 tyrosine kinase inhibitors, FTIs and more recently mTOR antagonists, each as monotherapy, in patients with advanced (often heavily pretreated) disease. At the same time, the encouraging preclinical data suggesting that a more effective strategy may be to combine STIs with endocrine therapy has led to several randomized clinical trials designed to test this hypothesis in the clinic.

    Trials with EGFR tyrosine kinase inhibitors (TKIs)

    The most extensively studied TKI in breast cancer is the EGFR TKI gefitinib (Iressa), an orally active, low-molecular- weight, synthetic anilinoquinazoline and a potent selective inhibitor of EGFR-TK. In various breast cancer cell lines that express EGFR and/or HER-2, gefitinib given as a single agent induced a dose-dependent antiproliferative effect which delayed growth (Moulder et al. 2001). Experiments have demonstrated that gefitinib may inhibit the growth of acquired endocrine-resistant MCF-7 breast cancer cells in vitro (Hutcheson et al. 2003, Knowlden et al. 2003, Martin et al. 2003) and in vivo (Shou et al. 2004). There have been three phase II monotherapy studies of gefitinib in patients with advanced breast cancer (Albain et al. 2002, Baselga et al. 2003, Robertson et al. 2003). Overall, the data are relatively disappointing with low clinical response rates and short times to disease progression (Table 2). The only trial to report a significant number of responses included patients with ER-positive, tamoxifen-resistant breast cancer (Robertson et al. 2003), the setting in which preclinical models had shown the best evidence of activity for gefitinib. Pharmacodynamic studies have been performed in one of these trials, confirming that EGFR tyrosine kinase signaling is inhibited in both skin and tumor biopsies by doses of gefitinib delivered orally (Baselga et al. 2003). However, there was discordance in the effect of gefitinib on downstream intracellular signaling in treated tumor biopsies, with lack of inhibition of Ki-67 (a marker of cell proliferation) in tumor, but not in matched skin biopsies. This suggested that activation of other intracellular pathways downstream of EGFR (especially in breast cancer as opposed to normal skin cells) may determine the clinical response to gefitinib. More research is required to establish tumor phenotypes in responding versus nonresponding patients (Dancey et al. 2003). Fewer clinical data exist regarding the other EGFR TKI in breast cancer, although a phase II monotherapy trial of the selective EGFR TKI erlotinib (OSI-774) in breast cancer was relatively disappointing (Winer et al. 2002).

    Based on the preclinical evidence for added benefit outlined above, a number of small phase I/II trials have been initiated with TKIs in combination with tamoxifen, fulvestrant or an aromatase inhibitor. Some of these trials are in the second-line setting, with one trial enrolling patients whose tumor was progressing on tamoxifen, and then adding lapatinib to tamoxifen to see whether clinical responses could be observed and resistance reversed. Another trial will compare gefitinib alone to the combination of gefitinib plus tamoxifen after progression on tamoxifen. However, the ultimate clinical test for the hypothesis that TKIs enhance the efficacy of endocrine therapy is the randomized, controlled clinical trial of endocrine therapy alone versus combined endocrine/TKI therapy. Many of these trials are in the first-line setting, where early clinical (and indeed experimental) data have shown that gefitinib alone may have limited activity. Therefore, the primary endpoint for these trials is to investigate whether time to disease progression (TTP) can be significantly prolonged by the addition of a TKI to endocrine therapy, thus delaying the emergence of resistance as demonstrated in various preclinical models described above. Table 3 lists the current randomized, controlled clinical trials of endocrine therapy with or without gefitinib as first-line therapy in advanced breast cancer. The majority are randomized phase II studies with only 100–200 patients, and in some studies the primary ef.- cacy endpoint is objective response rate (ORR) rather than TTP. Such studies are asking the combination to provide greater initial antitumor activity than endocrine therapy alone, expecting to enhance the response in tumors with de novo endocrine resistance. Given the mechanism of action of these drugs in combination (that is, enhanced G0/G1 arrest without enhanced apoptosis), an enhanced clinical benefit rate (CBR) with more stable disease may be a better endpoint for these trials, which ultimately will contribute to prolonged TTP.

    It will be important in all these trials to stratify for prior endocrine therapy (usually tamoxifen) given in the adjuvant setting, and in particular the interval since completion of such therapy. This is important, as it may have implications for the presence or absence of activated growth factor signaling in the relapsed tumor, which could determine the efficacy of added gefitinib. In addition, biologic studies are required to help predict those patients more likely to benefit from combined endocrine/gefitinib therapy. For example, the gefitinib/tamoxifen trial will undertake studies to look at downstream intracellular signaling componenets of the erbB family, in addition to assessment of ER and coactivator (AIB1) expression (Fig. 1).

    Trials with dual EGFR/HER-2 tyrsoine kinase inhibitors

    Cooperative activation of different type I growth factor receptors (EGFR, HER-2, HER-3 or HER-4) may limit the efficacy of targeting just one single receptor. Lapatinib (GW572016) is a potent dual inhibitor of both EGFR and HER-2 and inhibits autophosphorylation of the receptors (Rusnack et al. 2001). In the phase I study, diarrhea and skin rash were the main toxicities (Xia et al. 2002), and clinical activity was reported in trastuzumab-resistant breast cancer patients (Spector et al. 2003). Several biologic and pharmacologic reasons may account for the efficacy of a small molecule inhibitor of HER-2 in patients resistant to monoclonal therapy with trastuzumab. A phase II trial of lapatinib has been completed in heavily pretreated patients with advanced breast cancer that progressed on prior trastuzumab-containing regimens. A recent interim analysis in the first 41 patients confirmed clinical activity for lapatinib in breast cancer, with partial responses in 7% of patients and/or stable disease in 24% of patients after 16 weeks of therapy (Blackwell et al. 2004).

    One of the pivotal trials for lapatinib will be the large, randomized, phase III trial of letrozole with or without lapatinib in ER-positive advanced metasatic breast cancer (Table 3; Fig. 2). This study in over 760 patients is powered to detect a 30% improvement in median time to disease progression from 10 to 13 months (hazard ratio of 0.769). Recruitment has started, and exporatory biologic studies will accompany the main clinical protocol to try to identifiy who benefits most from the combination. Again, the recently published preclinical data showing the ability of lapatinib to reverse hormone resistance and to cooperate with tamoxifen in providing maximal growth arrest strongly underpin the design of this study (Chu et al. 2005). Secondary endpoints will include objective response rate and clinical benefit rate, a feature which should allow the impact of the combination on delaying disease progression to be detected.

    Trials with FTIs

    Monotherapy activity has been reported for the FTI tipifarnib (Zarnestra) in advanced breast cancer. A total of 76 patients were treated with tipifarnib, either as a continuous dose of 300 or 400mg b.i.d. (n=41) or an intermittent dose of 300mg b.i.d. for 21 days followed by 7 days off therapy (n=35) (Johnston et al. 2003). In the continuous treatment arm, there were four partial responses (10%) lasting 4–12 months and six patients with stable disease (15%) for at least 6 months. In the intermittent treatment arm, there were five partial responses (14%) and three patients with stable disease (9%). Objective responses were seen in both soft-tissue and viscereal sites of disease (Fig. 3). The main toxicities were neutropenia, thrombocytopenia, neurotoxicity and fatigue. These results were independent of ras status, estrogen/progesterone receptor or HER-2/EGFR receptor status, and 40% of patients had received only prior adjuvant and/or metastatic endocrine therapy at entry into the trial. Thus, while FTIs may have modest antitumor activity as a single-agent therapy, stabilization of disease in those resistant to endocrine therapy may represent a meaningful response.

    Based on the encouraging preclinical data discussed above from several groups for additive or synergistsic effects for combining FTIs with endocrine therapy, several nonrandomized and randomized phase II studies of FTIs in combination with either tamoxifen, fulvestrant or aromatase inhibitors have been undertaken (Table 4). It is unlikely that any overlapping toxicities will be seen for combinations of FTIs with endocrine therapy, but at least two trials are ensuring that no pharmacokinetic interactions exist where by tamoxifen- or aromatase inhibitor-induced hepatic enzymes could enhance clearance of FTIs and lower serum concentrations. For example, pharmacokinetic and pharmacodynamic endpoints have been assessed by a sequential design in 11 patients treated initially with the FTI tipifarnib (either 200 or 300 mg b.i.d. for 21/28 days), and after 1 week tamoxifen was added (Lebowitz et al. 2004). There was no significant change in the pharmacokinetic profile for tipifarnib; moreover, the pharmacodynamic endpoint (inhibition of farnesyltransferase in peripheral blood mononuclear cells) was enhanced from 30% enzyme suppression to 41% by the combination. In addition, such early phase II trials may help determine the optimal schedule for the combination. There are no published efficacy data from any of the randomized endocrine/FTI trials to date.

    Trials with mTOR antagonists

    Single-agent activity has been reported for the mTOR antagonist temsirolimus (CCI-779) given by weekly intravenous administration to 109 patients with heavily pretreated, locally advanced or metastatic breast cancer whose disease relapsed or was refractory to previous anthracyclines and/or taxanes (Chan et al. 2003). Based on 94 evaluable patients, partial responses were seen in 11% patients with stable disease in an additional 33% of patients. The main toxicities included alteration in transaminases, mucositis, rash and mild nausea.

    Again based on encouraging preclinical data for possible synergistic effects when mTOR antagonists were combined with endocrine therapy (DeGraffenried et al. 2004, Rudolf et al. 2004), clinical trials looking at combined therapy have been initiated – these are not as far advanced as those described above for TKIs and FTIs. In these phase II studies, clinical response is the primary endpoint, with stable disease for at least 6 months being an additional endpoint in some studies. Preliminary data from a small, three-arm randomized study has compared two different schedules of oral temsirolimus (10mg continuous or 30 mg for 5/14 days) combined with letrozole. Initial higher doses of temsirolimus were used but were poorly tolerated (grade 2/3 stomatitis) when combined with long-term endocrine therapy (Baselga et al. 2004). Efficacy results have suggested similar response rates for the combination compared with letrozole alone. However, clinical endpoints using objective tumor response rates in small, nonrandomized phase II studies must be viewed cautiously, and the primary role of such studies should be to provide safety and supportive biologic data for the combination in advance of definitive randomized trials. A large-scale phase III clinical trial with an orally active formulation of temsirolimus (30mg for 5 out of 14 days) in combination with letrozole has started, and aims to recruit over 1200 postmenopausal patients with ER-positive, locally advanced/metasatic breast cancer that are suitable for first-line hormonal therapy with an aromatase inhibitor. The primary endpoint of this study will be progression-free survival. A similar, randomized phase III study assessing letrozole with or without everolimus is also underway.

    Presurgical biomarker studies of endocrine/STI therapy

    Randomized clinical trials of endocrine/STI therapy in metastatic breast cancer, as outlined above, are large and expensive, and always run the risk of a very heterogeneous patient population, making results potentially difficult to interpret. If any benefit for the combined therapy exists, its use in the adjuvant setting needs ultimately to be tested to see whether survival from breast cancer can be enhanced further beyond the improvement already seen with aromatase inhibitors by such an approach (ATAC Trialists 2002). However, large, randomized, adjuvant clinical trials to establish whether any benefit exists for combined endocrine/STI therapy versus endocrine therapy alone will involve many thousands of patients with follow-up extending over many years before first results emerge. Reliable intermediate endpoints that allow a more rapid evaluation of the potential benefit of combined endocrine/STI therapy in early breast cancer would be of enormous value. The neoadjuvant (preoperative) setting in which medical therapies are given and evaluated prior to surgery is being increasingly exploited with these issues in mind (Ayers et al. 2004). Clinical response to neoadjuvant chemotherapy has been found to predict disease-free survival and overall survival (Fisher et al. 1998), but there are no direct data to confirm this for endocrine therapy.

    Changes in the biologic determinants of tumor regression or progression, namely, proliferation and apoptosis, might be able to provide an earlier – and possibly more reliable — prediction of long-term outcome. Data from relatively small studies supported the possibility that biomarkers may predict tumor response to chemotherapy (Chang et al. 2000). In a neoadjuvant endocrine therapy trial, the aromatase inhibitor vorozole was compared with tamoxifen, and after 2 weeks it showed a greater fall in Ki-67 (58% vs 43%, respectively) that was weakly associated with clinical response at 12 weeks (Harper-Wynne et al. 2002). In the recently reported IMPACT trial, the neoadjuvant use of tamoxifen versus the third-generation aromatase inhibitor anastrozole versus the combination was compared in 330 postmenopausal patients with primary ER-positive breast cancer (Dowsett et al. 2005). Suppression of the proliferation marker Ki-67 after 2 and 12 weeks was significantly greater with anastrozole than with tamoxifen (P=0.004 and P<0.001), but similar between tamoxifen and the combination. There was no significant correlation overall between a fall in Ki-67 and clinical tumor response. However, the 2-week change in Ki-67 in the neoadjuvant IMPACT trial closely paralleled the improved recurrence-free survival seen after 5 years in the adjuvant ATAC trial that compared these same agents in the adjuvant setting (ATAC Trialists 2002). For the first time, therefore, change in an intermediate biomarker in an endocrine therapy trial may predict for the subsequent long-term outcome, and provide a quicker and more reliable outcome measure than clinical response criteria alone.

    This approach is now being tested for combined endocrine/STI therapy in presurgical neoadjuvant studies. Figure 4 shows the design of a multicenter, double-blind, randomized, controlled trial that will assess whether the addition of the TKI gefitinib to anastrozole will improve its efficacy. A novel aspect is that the primary endpoint will be change in cell proliferation as assessed by Ki-67. In addition, the design will look at whether the change in Ki-67 after just 2 weeks of therapy can predict long-term benefit of therapy for the combination (arm A) versus endocrine therapy alone (arm C), and in arm B whether the addition of gefitinib after an initial 2 weeks of therapy with anastrozole can further lower Ki-67 in those tumors that do not response to initial aromatase inhibitor therapy. Secondary endpoints will include clinical response, tolerability and pharmacokinetics, as well as proteomics and gene expression profiles that may predict response to therapy. Recruitment to the trial is well underway. A similar design is being considered for the combination of the mTOR antagonist everolimus and letrozole in the neoadjuvant setting.

    Conclusion — challenges for development of STIs in breast cancer

    There is much enthusiasm surrounding these novel STIs and their potential role in the treatment of breast cancer. However, considerable thought is needed in order to maximize their potential. Central to their development will be a clear understanding of the molecular biology of these pathways, in particular the differences between hormone-sensitive and hormone-resistant disease. Preclinical models both in vitro and in vivo are important to clarify the benefit and utility of combined endocrine/STI therapy. For clinical trials, appropriate patient selection will be important, and parallel biologic studies are now a requirement for development of these drugs. Randomized studies remain central to determine any added benefit of combined endocrine/STI therapy, and must be appropriately powered for relevant endpoints in order to encourage further clinical development. Presurgical studies may be an additional means of gaining biologic and clinical information about such combined therapies. Many trials have now started, and we eagerly await the results to see whether intracellular STIs can provide a significant therapeutic benefit.

    Acknowledgements

    The author declares that there is no conflict of interest that would prejudice the impartiality of this scientific work.

    References

    Albain K Elledge R, Gradishar WJ, Hayes DF, Rowinsky EK, Hudis C, Pusztai L, Tripathy D, Modi S & Rubi S 2002 Open-label phase II multicenter trial of ZD1839 (Iressa) in patients with advanced breast cancer. Breast Cancer Research and Treatment 76 A20.

    Argiris A, Wang CX, Whalen SG & DiGiovanna MP 2004 Synergistic interactions between tamoxifen and trastuzumab (Herceptin). Clinical Cancer Research 10 1409–1420.

    ATAC Trialists Group 2002 Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early breast cancer: first results of the ATAC randomised trial. Lancet 359 2131–2139.

    Ayers M, Symmans WF, Stec J, Damokosh AI, Clark E, Hess K, Lecocke M, Metivier J, Booser D, Ibrahim N, Valero V, Royce M, Arun B, Whitman G, Ross J, Sneige N, Hortobagyi GN & Pusztai L 2004 Gene expression profiles predict complete pathologic response to neoadjuvant paclitaxel and fluorouracil, doxorubicin, and cyclophosphamide chemotherapy in breast cancer. Journal of Clinical Oncology 22 2284–2293.

    Baselga J, Albanelli J, Ruiz A, Lluch A, Gascon P, Gonzalez S, Guillan V, Sauleda S, Averbuch SD & Rojo F 2003 Phase II and tumor pharmacodynamic study of gefitinib in patients with advanced breast cancer. Proceedings of the American Society of Clinical Oncology 22 A24.

    Baselga J, Fumoleau P, Gil M, Colomer R, Roche H, Cortes-Funes H, Burstein H, Kaufman P, Kong S & Moore L 2004 Phase II, 3-arm study of CCI-779 in combination with letrozole in postmenopausal women with locally advanced or metastatic breast cancer: Preliminary results. Proceedings of the American Society of Clinical Oncology 22 A544.

    Blackwell KL, Kaplan EH, Franco SX, Marcom PK, Maleski JE, Sorensen MJ & Berger MS 2004 A phase II, open-label, multicenter study of GW572016 in patients with trastuzumab-refractory metastatic breast cancer. Proceedings of the American Society of Clinical Oncology 22 3006.

    Chan S, Scheulen ME, Johnston SRD, Mross K, Piccart M, Hess D, Bouxin N & Azli N 2003 A phase II study of two dose levels of CCI-779 in locally advanced or metasatic breast cancer failing prior anthracyclines and/or taxanes regimens. Proceedings of the American Society of Clinical Oncology 22 A774.

    Chang J, Ormerod M, Powles TJ, Allred DC, Ashley SE & Dowsett M 2000 Apoptosis and proliferation as predictors of chemotherapy response in patients with breast carcinoma. Cancer 89 2145–2152.

    Chu I, Blackwell K, Chen S & Slingerland J 2005 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 Research 65 18–25.

    Clark AS, West K, Streicher S & Dennis PA 2002 Constitutive and inducible Akt activity promotes resistance to chemotherapy, trastuzumab, or tamoxifen in breast cancer cells. Molecular Cancer Therapeutics 1 707–717.

    Dancey JE & Freidlen B 2003 Targeting epidermal growth factor receptor – are we missing the mark Lancet 362 62–64.

    DeGraffenried LA, Friedrichs WE, Russel DH, Donzis EJ, Middleton AK, Silva JM, Roth RA & Hidalgo M 2004 Inhibition of mTOR activity restores tamoxifen response in breast cancer cells with aberrant Akt activity. Clinical Cancer Research 10 8059–8067.

    Doisneau-Sixou SF, Cestac P, Faye JC, Favre G & Sutherland RL 2003 Additive effects of tamoxifen and the farnesyl transferase inhibitor FTI-277 on inhibition of MCF-7 breast cancer cell-cycle progression. International Journal of Cancer 106 789–798.

    Dowsett M, Smith IE, Ebbs SR, Dixon JM, Skene A, Griffith C, Boeddinghaus I, Salter J, Detre S, Hills M, Ashley S, Francis S & Walsh G 2005 Short-term changes in Ki-67 during neoadjuvant treatment of primary breast cancer with anastrozole or tamoxifen alone or combined correlate with recurrence-free survival. Clinical Cancer Research 11 951s–958s.

    Ellis CA, Vos MD, Wickline M, Riley C, Vallecorsa T, Telford WG, Zujewskil J & Clark GJ 2003 Tamoxifen and the farnesyl transferase inhibitor FTI-277 synergize to inhibit growth in estrogen receptor-positive breast tumor cell lines. Breast Cancer Research and Treatment 78 59–67.

    End DW, Smets G, Todd AV, Applegate TL, Fuery CJ, Angibaud P, Venet M, Sanz G, Poignet H, Skrzat S, Devine A, WoutersW& Bowden C 2001 Characterization of the antitumor effects of the selective farnesyl protein transferase inhibitor R115777 in vivo and in vitro. Cancer Research 61 131–137.

    Fisher B, Bryant J, Wolmark N, Mamounas E, Brown A, Fisher ER, Wickerham DL, Begovic M, DeCillis A, Robidoux A, Margolese RG, Cruz AB Jr, Hoehn JL, Lees AW, Dimitrov NV & Bear HD 1998 Effect of preoperative chemotherapy on the outcome of women with operable breast cancer. Journal of Clinical Oncology 16 2672–2685.

    Gee JM, Harper ME, Hutcheson IR, Madden TA, Barrow D, Knowlden JM, McClelland RA, Jordan N, Wakeling AE & Nicholson RI 2003 The antiepidermal growth factor receptor agent gefitinib (ZD1839/Iressa) improves antihormone response and prevents development of resistance in breast cancer in vitro. Endocrinology 144 5105–5117.

    Harper-Wynne CL, Sacks NPM, Shenton K, MacNeill FA, Sauven P, Laidlaw IJ, Rayter Z, Miall S, Howes, A, Salter J, Hills MJ, Lowe FM, A’Hern R, Nasiri N, Doody D, Iqbal J & Dowsett M 2002 Comparison of the Systemic and Intratumoral Effects of Tamoxifen and the Aromatase Inhibitor Vorozole in Postmenopausal Patients With Primary Breast Cancer. Journal of Clinical Oncology 20 1026–1035.

    Hutcheson IR, Knowlden JM, Madden TA, Barrow D, Gee JM, Wakeling AE & Nicholson RI 2003 Oestrogen receptor-mediated modulation of the EGFR/MAPK pathway in tamoxifen-resistant MCF-7 cells. Breast Cancer Research and Treatment 81 81–93.

    Jeng MH, Yue W, Eischeid A, Wang JP & Santen RJ 2000 Role of MAP kinase in the enhanced cell proliferation of long term estrogen deprived human breast cancer cells. Breast Cancer Research and Treatment 62 167–175.

    Johnston SR, Hickish T, Ellis P, Houston S, Kelland L, Dowsett M, Salter J, Michiels B, Perez-Ruixo JJ, Palmer P & Howes A 2003 Phase II study of the efficacy and tolerability of two dosing regimens of the farnesyl transferase inhibitor, R115777, in advanced breast cancer. Journal of Clinical Oncology 21 2492–2499.

    Johnston SRD, Head J, Valenti M, Detre S & Dowsett M 2002 Endocrine therapy combined with the farnesyltransferase inhibitor R115777 produces enhanced tumour growth inhibition in hormone-sensitive MCF-7 human breast cancer xenografts in-vivo. Breast Cancer Research and Treatment 76 A245.

    Kelland LR, Smith V, Valenti M, Patterson L, Clarke PA, Detre S, End D, Howes AJ, Dowsett M, Workman P & Johnston SR 2001 Preclinical antitumor activity and pharmacodynamic studies with the farnesyl protein transferase inhibitor R115777 in human breast cancer. Clinical Cancer Research 7 3544–3550.

    Knowlden JM, Hutcheson IR, Jones HE, Madden T, Gee, JM, Harper ME, Barrow D, Wakeling AE & Nicholson RI 2003 Elevated levels of epidermal growth factor receptor/c-erbB2 heterodimers mediate an autocrine growth regulatory pathway in tamoxifenresistant MCF-7 cells. Endocrinology 144 1032–1044.

    Kurokawa H, Lenferink AEG, Simpson JF, Pisacane PI, Sliwkowski MX, Forbes JT & Arteaga CL 2000 Inhibition of HER2/neu (erbB-2) and Mitogen-activated Protein Kinases Enhances Tamoxifen Action against HER2- overexpressing, Tamoxifen-resistant Breast Cancer Cells. Cancer Research 60 5887–5894.

    Lebowitz PF, Eng-Wong J, Balis F, Widemann B, Jayaprakash N, Gantz S, Chow C, Merino M & Zujewski J 2004 A phase I trial of tipifarnib, a farnesyltransferase inhibitor, and tamoxifen in hormonereceptor positive metastatic breast cancer. Proceedings of the American Society of Clinical Oncology 22 644.

    Long BJ, Liu G, Marrinan CH, Black S, Gheyas F, Nomeir A, Liu M, Kirschmeier P & Bishop R 2004 Combining the farnesyl transferase inhibitor (FTI) lonafarnib (SCH66336) with antiestrogens and aromatase inhibitors results in enhanced growth inhibition of hormonedependent human breast cancer cells and tumor xenografts. Proceedings of the American Association of Cancer Research 45 A3868.

    Martin LA, Farmer I, Johnston SR, Ali S, Marshall C & Dowsett M 2003 Enhanced estrogen receptor (ER) alpha, ERBB2, and MAPK signal transduction pathways operate during the adaptation of MCF-7 cells to long term estrogen deprivation. Journal of Biological Chemistry 278 30458–30468.

    Massarweh S, Shou J, Mohsin SK, Ge M, Wakeling AE, Osborne CK & Schiff R 2002 Inhibition of epidermal growth factor/HER2 receptor signaling using ZD1839 (Iressa) restores tamoxifen sensitivity and delays resistance to estrogen deprivation in HER2-overexpressing breast tumors. Proceedings of the American Society of Clinical Oncology 21 33a (A130).

    Moulder SL, Yakes FM, Muthuswamy SK, Bianco R, Simpson JF & Arteaga CL 2001 Epidermal growth factor receptor (HER1) tyrosine kinase inhibitor ZD1839 (Iressa) inhibits HER2/neu (erbB2)-overexpressing breast cancer cells in vitro and in vivo. Cancer Research 61 8887–8895.

    Nicholson RI, Hutcheson IR, Knowlden JM, Jones HE, Harper ME, Jordan N, Hiscox SE, Barrow D & Gee, JMW 2004 Nonendocrine Pathways and Endocrine Resistance: Observations with Antiestrogens and Signal Transduction Inhibitors in Combination. Clinical Cancer Research 10 346S–354S.

    Nicholson RI, McClelland RA, Robertson JRF & Gee JMW 1999 Involvement of steroid hormone and growth factor cross-talk in endocrine response in breast cancer. Endocrine-Related Cancer 6 373–387.

    Pancholi S, Lykkesfeldt A, Dowsett M, Johnston SRD & Martin L-A 2004 Elevated levels of Akt and p90rsk provide tamoxifen-resistant MCF-7 cells with a survival advantage involving Bad and Bcl-2. Proceedings of the American Association of Cancer Research 45 A5168.

    Robertson JFR, Gutteridge E, Cheung KL, Owers R, Koehler M, Hamilton L, Gee JMW & Nicholson RI 2003 Gefitinib (ZD1839) is active in acquired tamoxifenresistant oestrogen receptor positive and ER-negative breast cancer: results from a phase II study. Proceedings of the American Society of Clinical Oncology 22 A23.

    Rudolf J, Boulay A, Zumstein-Mecker S, Evans DB, O’Reilly T & Lane HA 2004 The mTOR pathway in estrogen response: a potential for combining the rapamycin derivative FAD001 with the aromatase inhibitor letrozole in breast carcinomas. Proceedings of the American Association of Cancer Research 45 A5619.

    Rusnak DW, Lackey K, Affleck K, Wood ER, Alligood KJ, Rhodes N, Keith BR, Murray DM, Glennon K, Knight WB, Mullin RJ & Gilmer TM 2001 The effects of the novel, reversible epidermal growth factor receptor/erbB2 tyrosine kinase inhibitor, GW2016, on the growth of human normal and tumor-derived cell lines in-vitro and in-vivo. Molecular Cancer Therapeutics 1 85–94.

    Shou J, Massarweh S, Osborne CK, Wakeling AE, Ali S, Weiss H & Schiff R 2004 Mechanisms of tamoxifen resistance: increased estrogen receptor-HER2/neu crosstalk in ER/HER2-positive breast cancer. Journal of the National Cancer Institute 96 926–935.

    Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M, Baselga J & Norton L 2001 Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. New England Journal of Medicine 344 783–792.

    Spector N, Raefsky E, Hurwitz H, Hensing T, Dowlati A, Dees C, O’Neill B, Smith DA, Mangum S & Burris HA 2003 Safety, clinical efficacy, and biological assessments from EGF10004; a randomised phase IB study of GW572016 for patients with metastatic carcinomas overexpressing EGFR or erbB2. Proceedings of the American Society of Clinical Oncology 22 193 (A772).

    Vogel CL, Cobleigh MA, Tripathy D, Gutheil JC, Harris, LN, Fehrenbacher L, Slamon DJ, Murphy M, Novotny WF, Burchmore M, Shak S, Stewart SJ & Press M 2002 Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer. Journal of Clinical Oncology 20 719–726.

    Winer E, Cobleigh M, Dickler M, Miller K, Fehrenbacher L, Jones C & Justice R 2002 Phase II multicenter study to evaluate the efficacy and safety of Tarceva (erlotinib, OSI-774) in women with previously treated locally advanced or metastatic breast cancer. Breast Cancer Research and Treatment 76 A445.

    Witters L, Engle L & Lipton A 2002 Restoration of estrogen responsiveness by blocking the HER-2/neu pathway. Oncology Reports 9 1163–1166.

    Xia W, Mullin R, Keith B, Liu L-H, Ma H, Rusnack D, Owens G, Alligood KJ & Spector NL 2002 Antitumor activity of GW572016; a dual tyrosine kinase inhibitor blocks EGFR activation of EGFR/erbB2 and downstream Erk1/2 and AKT pathways. Oncogene 21 6255–6263.

    Yu K, Toral-Barza L, Discafani C, Zhang WG, Skotnicki, J, Frost P & Gibbons JJ 2001 mTOR, a novel target in breast cancer: the effect of CCI-779, an mTOR inhibitor, in preclinical models of breast cancer. Endocrine-Related Cancer 8 249–258.(S R D Johnston)