98
Views
0
CrossRef citations to date
0
Altmetric
Review

Clinical evidence of the efficacy of everolimus and its potential in the treatment of breast cancer

&
Pages 27-35 | Published online: 17 May 2013

Abstract

The PI3K/Akt/mTOR (phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin) pathway regulates several key cellular functions and its dysregulation creates an environment that promotes tumorigenesis as well as resistance to therapy. The mTOR inhibitor everolimus has emerged as a promising agent in the treatment of breast cancer and was recently approved in combination with exemestane for advanced hormone receptor–positive disease after progression on a nonsteroidal aromatase inhibitor. Everolimus may also be effective in combination with cytotoxic and human epidermal growth factor receptor-2-directed therapies for the treatment of other subtypes of breast cancer. This paper highlights preclinical and clinical data that have emerged on the role of mTOR inhibition in breast cancer. Although generally well tolerated, everolimus carries a unique side effect profile of which both patients and providers should be made aware. Recommendations related to the administration of everolimus in the clinical setting are also discussed.

Introduction

It is estimated that in 2012 there were approximately 226,870 new cases of invasive breast cancer among women in the United States. An estimated 39,920 breast cancer deaths were expected, making it the second leading cause of cancer death in women. Since 1990, death rates for breast cancer have steadily decreased, reflecting progress in earlier detection and improved treatment.Citation1

Treatment strategies for breast cancer vary depending on stage, hormone receptor status, human epidermal growth factor receptor-2 (HER2) status, and other tumor- and patient-specific characteristics. However, one of the major challenges of treatment is overcoming intrinsic and/or acquired drug resistance. Multiple mechanisms of resistance exist, including reduced accumulation of chemotherapy in cancer cells, alterations in drug targets, activation of detoxifying mechanisms, increased repair of drug-induced cellular damage, and alterations in cell signaling, cell cycle control, and apoptosis signaling.Citation2 Each one of these mechanisms represents a potential opportunity to develop therapeutic agents in an effort to circumvent resistance.

The field of oncology has witnessed a paradigm shift from a “one size fits all” approach to treatment to a more individualized approach in which a deeper understanding of target oncogenic proteins and pathways has allowed us to develop more rational therapies, resulting in increased efficacy and decreased toxicity.

This paper discusses our understanding of the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway in the pathogenesis of breast cancer as well as the successful development of an inhibitor of this pathway, everolimus, and its role in the treatment of breast cancer.

PI3K/Akt/mTOR pathway

The PI3K/Akt/mTOR pathway regulates several normal cellular functions, including cellular growth, proliferation, survival, and differentiation.Citation3 Dysregulation of this pathway creates an environment that promotes tumorigenesis, and activating mutations are frequently reported in human cancers.Citation4

Activation of this pathway can occur through activation or mutation of either PI3K or Akt, overexpression of growth factor receptors such as HER2 or insulin-like growth factor receptor, or through loss of tumor suppressor phosphatase and tensin homolog deleted from chromosome 10 (PTEN) activity.Citation5Citation7

Activated PI3K leads to downstream phosphorylation of Akt, a serine/threonine kinase, which in turn activates other key downstream effectors such as mTOR. mTOR is a serine/threonine kinase that is a key mediator of cellular proliferation, apoptosis, angiogenesis, and cellular metabolism.

The effects of mTOR are mediated by two distinct multiprotein complexes, mTORC1, which is sensitive to inhibition by rapamycin, and mTORC2, which is not.Citation8 mTORC1 exerts its effects via phosphorylation of the eukaryotic initiation factor 4E-binding protein 1 (4E-BP1) and the p70 ribosomal S6 kinase 1 (S6K1), leading to enhanced mRNA translation, cell proliferation, growth, and survival. The function of mTORC2 is incompletely understood but is thought to be related to cytoskeleton organization as well as cell proliferation, survival, and metabolism.Citation8,Citation9

Mutations in the PI3K catalytic alpha subunit (PIK3CA) gene and the loss of PTEN, which inhibits the mTOR pathway, are frequently observed in breast cancer.Citation10 Several studies have shown that alterations of the PIK3CA gene are observed in approximately 10%–40% of breast cancers.Citation11 Activation of the PI3K/Akt signaling pathway in breast cancer cells leads to the development of resistance to therapy, and high Akt and mTOR activity are especially associated with the development of resistance to endocrine therapy.Citation12Citation14 Similar associations have been made between mTOR activation and trastuzumab resistance.Citation15Citation17 These findings provide the rationale for the addition of mTOR inhibition to chemotherapy, endocrine therapy, anti-HER2 therapy, or a combination of these, in an effort to delay or reverse resistance.

mTOR inhibitors

Rapamycin is a naturally occurring fungicide produced by the bacterium Streptomyces hygroscopicus. It forms a complex with the cytosolic protein FK-binding protein 12 (FKBP12) which blocks the actions of 4E-BP1 and p70 ribosomal S6K, resulting in cell cycle arrest.Citation18 Historically, rapamycin has been used as an immunosuppressant in solid organ transplant recipients. In recent years, the use of synthetic rapamycin analogs, or “rapalogs,” with more favorable pharmacokinetic properties, has extended to the field of oncology.

Temsirolimus (Torisel®, Pfizer, Inc, New York, NY, USA) is US Food and Drug Administration (FDA)-approved for the treatment of renal cell carcinoma; everolimus (Afinitor®, Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA) is FDA-approved for the treatment of renal cell carcinoma, pancreatic neuroendocrine tumor, angiomyolipoma, advanced estrogen receptor (ER)+/HER2 breast cancer, and subependymal giant cell astrocytoma in individuals with tuberous sclerosis complex; ridaforolimus is currently in clinical development.

Preclinical data

Several preclinical studies have demonstrated the efficacy of mTOR inhibition in breast cancer. One of the drivers of the PI3K pathway is ER activation, and activation of both PI3K and Akt may be invoked by crosstalk between epidermal growth factor receptor and ER. Such crosstalk has been associated with estrogen-independent transcriptional activity and estrogen resistance.Citation19Citation21 DeGraffenried et al demonstrated that breast cancer cells with constitutively active Akt proliferate in the absence of exogenous estrogen and develop resistance to the growth inhibitory and proapoptotic effects of tamoxifen both in vitro and in vivo. Cotreatment of these cells with rapamycin derivatives restores the apoptotic responses to tamoxifen.Citation20

When everolimus is combined with letrozole or 4-hydroxytamoxifen (an active metabolite of tamoxifen), there is synergistic inhibition of the proliferation of estrogen-dependent breast cancer cells and increased apoptosis.Citation22,Citation23 In another study, coadministration of everolimus with letrozole and fulvestrant reversed Akt-mediated resistance to endocrine therapy and restored responsiveness to endocrine therapy in breast cancer cell lines.Citation24 Thus, ample preclinical data are available supporting the potential role of mTOR inhibitors in the treatment of breast cancer, particularly in endocrine-resistant tumors.

Temsirolimus

A randomized three-arm Phase II study evaluated temsirolimus, also known as CCI-779 or Torisel, in combination with letrozole in postmenopausal women with heavily pretreated ER+ metastatic breast cancer (MBC). Patients received letrozole 2.5 mg/day alone or with temsirolimus given orally on a daily or intermittent schedule. The combination showed tolerability and there was a suggestion that progression-free survival (PFS) was longer with the addition of temsirolimus.Citation25

A subsequent Phase III study evaluated first-line letrozole 2.5 mg/day plus temsirolimus 30 mg/day (5 days every 2 weeks) versus letrozole plus placebo in 1112 patients with aromatase inhibitor (AI)-naïve, hormone receptor–positive advanced breast cancer. However, the study was terminated prematurely after a planned interim analysis showed that the combination was unlikely to achieve the expected level of efficacy. There was no improvement in the primary endpoint of PFS (median, 9 months; hazard ratio (HR), 0.90; 95% CI, 0.76–1.07; P = 0.25).Citation26 Possible reasons for the failure of the study to meet its primary endpoint include patient selection and/or suboptimal dosing of the drug.

Everolimus

The rapamycin analog everolimus, also known as RAD 001 or Afinitor, is a highly specific mTOR inhibitor that also carries anti-angiogenic properties. After oral administration, everolimus is absorbed rapidly, with peak concentrations occurring at 1.3–1.8 hours after a single dose. After multiple doses, steady-state concentrations are achieved in approximately 7 days. It has a half-life of 18–35 hours. Everolimus has a predominantly hepatic clearance, and dose adjustment is not needed for renal insufficiency as only 5% of the drug is excreted in the urine.Citation27

Currently, everolimus is FDA-approved for use in advanced renal cell cancer, pancreatic neuroendocrine cancers, and subependymal giant cell astrocytomas. Its most recent FDA approval was granted in July 2012 for treatment of postmenopausal women with advanced hormone receptor–positive, HER2-negative breast cancer in combination with exemestane. This last indication was granted on the basis of study results that will be discussed below.

Everolimus in the treatment of hormone receptor–positive breast cancer

On the basis of the preclinical data discussed above, a number of studies were conducted evaluating the role of everolimus in the clinical setting.

A Phase I, dose-escalating study evaluated everolimus plus letrozole in 18 postmenopausal patients with stable MBC or progression after at least 4 months of first- or second-line therapy with letrozole alone.Citation28 Six patients received everolimus 5 mg/day, and 12 patients received 10 mg/day. Among these patients, one had a complete response (CR) lasting more than 22 months, and another experienced a 28% reduction in liver metastases. Both had received everolimus 10 mg/day. There was one dose-limiting toxicity, grade 3 thrombocytopenia, which occurred in a patient assigned to the higher dose. Based on the results of this study, everolimus at a daily dose of 10 mg/day was recommended for subsequent studies.

A Phase II study randomized 270 postmenopausal women with operable ER+ breast cancer to receive 4 months of neoadjuvant treatment with letrozole 2.5 mg/day plus either everolimus 10 mg/day or placebo. The primary endpoint was clinical response by palpation. The response rate (RR) in the everolimus arm was higher than that with letrozole alone (68.1% versus 59.1%). An antiproliferative response, defined by a reduction in Ki67 expression at day 15 occurred in 52 of 91 (57%) patients in the everolimus arm and in 25 of 82 (30%) patients in the placebo arm (P < 0.01). The authors concluded that everolimus significantly increased letrozole efficacy in the neoadjuvant treatment of ER+ breast cancer.Citation29

Sabine et al characterized the effects of preoperative everolimus in primary breast cancer patients through gene expression profiling. Twenty-seven patients with ER+ breast cancer completed 11–14 days of neoadjuvant everolimus 5 mg/day. Patients whose tumors responded with significant reductions in proliferation also had significant decreases in the expression of genes involved in cell cycle and p53 signaling pathways. Overall, everolimus was noted to decrease proliferation, increase apoptosis, and reduce Akt/mTOR signaling in tumors.Citation30

The Tamoxifen-RAD001 (TAMRAD) study was a Phase II study of 111 patients with ER+, HER2-negative MBC treated previously with AI therapy.Citation31 Patients were randomized to either tamoxifen alone or tamoxifen with everolimus 10 mg/day. The primary outcome was clinical benefit rate (CBR), defined as CR + partial response (PR) + stable disease (SD) at 6 months. Earlier AI therapy had been administered in either the adjuvant (31%) or metastatic setting (60%). Patients were stratified according to primary (49%) or secondary (51%) hormone resistance. Primary resistance was defined as disease relapse during or within 6 months of stopping adjuvant AI therapy, or disease progression within 6 months of starting AI therapy in the metastatic setting. Secondary resistance was defined as disease relapse greater than 6 months after stopping adjuvant AI therapy or progression after 6 months of AI therapy in the metastatic setting.

The CBR was 42% in the tamoxifen-alone group, compared with 61% in the everolimus plus tamoxifen group (exploratory P = 0.045). The time to progression (TTP) was significantly longer in the combination group (8.6 months versus 4.5 months, HR, 0.54; 95% CI, 0.36–0.81). At the time of the last update, there were 31 deaths in the tamoxifen-alone group, compared with 16 deaths in the combination group. Median overall survival (OS) had not been reached in the combination group; OS was 32.9 months in the tamoxifen group, yielding a HR for survival of 0.45 (95% CI, 0.24–0.81, exploratory P = 0.007), favoring the addition of everolimus.

Results from exploratory subgroup analyses suggested that the benefit from the addition of everolimus to tamoxifen may have been restricted to patients with secondary hormone resistance. The CBR among patients with secondary resistance was 74% with the combination, compared with 48% with tamoxifen alone; whereas the CBR in patients with primary resistance was only slightly better with the addition of everolimus (46%) compared with tamoxifen alone (36%). Similarly, the magnitude of improvement in TTP with the combination was greater in patients with secondary resistance (HR, 0.46; 95% CI, 0.26–0.83) than that observed in patients with primary resistance (HR, 0.70; 95% CI, 0.40–1.21). The main toxicities associated with tamoxifen plus everolimus were fatigue (72% versus 53% with tamoxifen alone), stomatitis (56% versus 7%), rash (44% versus 7%), anorexia (43% versus 18%), and diarrhea (39% versus 11%).

BOLERO-2 was a randomized Phase III trial that enrolled 724 postmenopausal women with ER+/HER2 locally advanced cancer or MBC that was refractory to letrozole or anastrozole.Citation32 Patients were randomized in a 2:1 ratio to receive exemestane 25 mg/day plus everolimus 10 mg/day versus exemestane plus placebo. The primary endpoint was PFS. At the time of interim analysis, the group assigned to everolimus plus exemestane demonstrated a significant improvement in PFS (6.9 months) compared with exemestane alone (2.8 months) (HR, 0.43; 95% CI, 0.35–0.54; P < 0.001). The benefit appeared consistent across all subgroups. Overall survival analysis is not yet mature. There were more grade 3 and 4 toxicities observed with the addition of everolimus, the most common being stomatitis (8% versus 1%), anemia (6% versus < 1%), dyspnea (4% versus 1%), hyperglycemia (4% versus < 1%), fatigue (4% versus 1%) and pneumonitis (3% versus 0%). Based on results from this study, in July 2012, the FDA approved the use of everolimus in combination with exemestane for hormone receptor–positive/HER2 advanced breast cancer in postmenopausal women after progression on a nonsteroidal AI.

Everolimus in the treatment of HER2-overexpressing breast cancer

HER2+ breast cancers, which account for 25%–30% of breast cancers, are biologically aggressive and are associated with altered response to therapy and poor clinical outcomes.Citation33 The development of trastuzumab, a humanized monoclonal antibody against HER2, represents a major advance in the treatment of both early-stage and advanced HER2+ breast cancer.Citation34Citation37 Nevertheless, many tumors do not respond to trastuzumab-based therapy, and even among those that do, resistance often develops.

HER2 stimulates cell proliferation and survival through the PI3K/Akt/mTOR pathway,Citation38 and activation of this pathway is associated with trastuzumab resistance.Citation15Citation17 Preclinical data have shown that everolimus has synergistic activity with trastuzumab and may overcome trastuzumab resistance.Citation39

The efficacy of everolimus in women with HER2+ MBC has been evaluated in several Phase I–II studies. In a Phase Ib study, 33 patients who were pretreated with trastuzumab received everolimus 5 mg daily, 10 mg daily, or 30 mg weekly in combination with weekly trastuzumab and paclitaxel on days 1, 8, and 15 of a 28-day cycle.Citation40 Among the 27 patients with measurable disease, two patients had a CR and 10 patients had a PR, for an overall response rate (ORR) of 44%. An additional 13 patients (48%) had SD. The median PFS was 34 weeks.

In a subsequent Phase II follow-up trial, everolimus 10 mg/day in combination with paclitaxel and trastuzumab in doses similar to the Phase Ib study described above was evaluated in patients who were resistant to taxanes and trastuzumab. Among 48 patients, 19% had a PR and 62% had SD. Median PFS was 26 weeks.Citation41

In another Phase Ib trial, everolimus was evaluated in patients with HER2+ MBC who had progressed after receiving trastuzumab.Citation42 Most patients had received taxanes and anthracyclines previously, and 24% had been pretreated with lapatinib. Patients received everolimus 5 mg/day, 20 mg/week, or 30 mg/week in combination with weekly trastuzumab and vinorelbine on days 1 and 8 of a 21-day cycle. Among 47 patients in all dosage groups, the ORR was 19% and the CBR (CR + PR + SD ≥ 24 weeks) was 54%. Median PFS was 30.7 weeks for the overall population. In the extension phase of the trial, patients were allowed to continue everolimus, and vinorelbine could be discontinued at the investigator’s discretion. Two additional patients achieved CR, one achieved PR, and the overall median PFS was 41 weeks.

A post hoc analysis was performed that pooled data from the two previously discussed Phase ICitation40,Citation42 and Phase IICitation41 studies to evaluate the efficacy of everolimus in patients pretreated with lapatinib.Citation43 Among 101 patients, the ORR was 21% for those patients treated with lapatinib compared with 29% for those not treated with lapatinib. The disease control rate was 88% and 81%, respectively; overall mean PFS was 29.0 and 36.1 weeks, respectively. These data suggest that everolimus, in combination with trastuzumab and chemotherapy, has antitumor efficacy in patients with HER2+ MBC, regardless of whether they were pretreated with lapatinib.

Patients with HER2+ MBC with progressive disease after previous HER2-targeted therapy were evaluated in two Phase I–II trials; data were combined for analysis.Citation44 Everolimus was administered at 5 or 10 mg/day plus a trastuzumab 8 mg/kg loading dose and then 6 mg/kg every 3 weeks. Among 47 patients, 15% had PR and 19% had SD. The median PFS was 4.1 months. Results from this study suggest that everolimus may have promising activity in the absence of cytotoxic chemotherapy.

The abovementioned trials show encouraging results for everolimus use in patients with HER2+ MBC. Two ongoing Phase III studies, BOLERO-1 and BOLERO-3, will help to further define the role of everolimus in this population.Citation45,Citation46 The BOLERO-1 trial is evaluating everolimus in combination with paclitaxel and trastuzumab as first-line therapy, and BOLERO-3 is evaluating everolimus in combination with vinorelbine and trastuzumab in patients with previous taxane therapy and trastuzumab resistance.

Everolimus in the treatment of triple-negative breast cancer (TNBC)

TNBCs lack the expression of ER, progesterone receptor and HER2 and comprise about 15% of all breast cancers.Citation47 Such tumors are associated with aggressive behavior and worse survival compared with other subtypes of breast cancer.Citation48 Treatment of TNBC remains a challenge, and cytotoxic chemotherapy remains the standard.

A Phase II study was conducted to evaluate the role of mTOR inhibition in combination with chemotherapy in this population.Citation49 Fifty patients with early-stage or locally advanced TNBC were randomized to receive neoadjuvant weekly paclitaxel 80 mg/m2 either alone or in combination with everolimus 30 mg/week for 12 weeks followed by 5-fluorouracil, epirubicin, and cyclophosphamide every 3 weeks for four cycles. The 12-week RR by ultrasonography was 48% when everolimus was administered in combination with paclitaxel compared with 30% when paclitaxel was used alone (P = 0.15). Pathologic CR rate was not significantly different between the two groups.

Another Phase II study evaluated the combination of everolimus at a dose of 5 mg daily plus carboplatin (area under the curve [AUC] = 6) given every 3 weeks in patients with metastastic TNBC.Citation50 The primary objective was CBR. At the time of reporting, 18 out of a planned total of 25 patients had been enrolled. One patient achieved a CR; four achieved a PR, and two SD lasting greater than 6 months. One of the patients who achieved SD had progressed on singleagent carboplatin at the time of study entry. Dose-limiting thrombocytopenia was an unexpected toxicity requiring amendment of carboplatin dosing to AUC = 4.

Due to the small sample sizes of the above studies, no clear conclusions can be drawn at this time. More studies are needed in this population to determine whether any subset exists that may benefit from the addition of everolimus.

Everolimus – toxicities and management

Safety data from the pivotal BOLERO-2 study showed that the most common adverse events (AEs) with an incidence of 30% or greater were stomatitis, infections, rash, fatigue, diarrhea, and decreased appetite. Grade 3/4 AEs with an incidence of 2% or greater included stomatitis, infections, hyperglycemia, fatigue, dyspnea, pneumonitis, and diarrhea. All these AEs occurred with greater frequency in the combination arm.Citation32

Noninfectious pneumonitis is a rare but serious AE known to occur with mTOR inhibitors.Citation52 It should be considered in patients presenting with cough, dyspnea, hypoxia, or pleural effusions when other causes have been excluded. On computed tomography scans, pneumonitis can present as ground glass or patchy opacities or infiltrates. It appears to be immunologically mediated, with biopsies showing organizing pneumonia, granulomatous inflammation, and lymphocytic infiltration or vasculitis. Management of symptomatic pneumonitis involves cessation of therapy and the use of corticosteroids if severe. Most cases have been reversible upon drug discontinuation. A pooled analysis of five studies evaluating everolimus in breast cancer patients reported variability in the incidence of noninfectious pneumonitis. In four of the studies, the incidence was approximately 3%; however, the incidence was 35% in the fifth study.Citation51

The immunosuppressive properties of everolimus may predispose patients to opportunistic infections and reactivation of previous infections including hepatitis B. Pneumonia, mycobacterial infections, other bacterial infections, and invasive fungal infections have been noted in those treated with everolimus.Citation52

Everolimus is also associated with metabolic derangements, notably hyperglycemia, hypercholesterolemia, and hypertriglyceridemia.Citation52 Optimal glycemic and lipid control should be achieved before starting everolimus, and serum glucose and lipid levels should be checked at baseline and periodically during treatment. These abnormalities can be treated using standard guidelines for diabetes and hyperlipidemia.

Everolimus – drug interactions

Everolimus is a substrate of CYP3A4 and P-glycoprotein. Coadministration with ketoconazole, a strong CYP3A4 inhibitor, results in increased everolimus blood concentrations, with resultant maximum concentration (Cmax) and AUC 3.9-fold and 15-fold higher, respectively. It is recommended that concurrent administration of strong CYP3A4 inhibitors be avoided. If coadministration with a moderate inhibitor is necessary, it is suggested that the dose of everolimus be reduced to avoid excess toxicity. Conversely, coadministration with the strong CYP3A4 inducer rifampin decreases Cmax and AUC of everolimus by 58% and 63%, respectively. If concurrent use of a strong inducer is necessary, it is suggested that the dose of everolimus be increased.Citation52

Predictive biomarkers

In this era of targeted therapy it is crucial to be able to identify subsets of patients who are either likely or unlikely to respond to a given drug. Such identification would maximize efficacy and minimize unnecessary toxicity. Currently, no reliable biomarkers exist to predict response to treatment with everolimus, although some molecular patterns are emerging as potential predictors of sensitivity and/or resistance.

Di Nicolantonio et al performed a series of elegant experiments to assess the sensitivity to everolimus of various cancer cell lines harboring mutations in PIK3CA or PTEN.Citation53 They found that the tumors could be divided into two groups based on response to everolimus: those resistant to treatment harbored both a mutation in the PI3K pathway as well as a mutation in either KRAS or BRAF. On the other hand, those that were sensitive to everolimus had only a mutation in the PI3K pathway. These results suggest that treatment with an mTOR inhibitor may be ineffective against KRAS or BRAF mutant tumors despite the presence of PI3K/AKT/mTOR pathway activation.

This hypothesis was tested by Janku et al in a study evaluating response in 25 patients with breast or gynecologic tumors harboring the PIK3CA mutation, 23 of whom were treated on a protocol that included a PI3K/AKT/mTOR pathway inhibitor.Citation54 Two (9%) of the 23 patients had SD for more than 6 months, and seven patients (30%) had a PR. Seven patients with PIK3CA mutations had coexisting mutations in KRAS, NRAS, or BRAF, and two of these patients (ovarian cancer) achieved a response. Although the numbers are small, these findings suggest that not all patients with simultaneous mutations in both pathways demonstrate resistance.

Chen et al found that expression of the cell cycle regulator p27 correlated with the anticancer activity of rapamycin and temsirolimus in breast cancer cells in vitro and in vivo. Cells expressing high levels of p27 were sensitive to treatment, whereas those with low expression demonstrated resistance. Moreover, they observed consistently that downregulation of p27 by silencing RNA rendered cells with normally high levels of expression resistant to treatment. They propose that p27 expression levels might serve as a predictive biomarker for patient selection for rapalog-based therapy.Citation55

Conclusion

The mTOR pathway is pivotal to the pathogenesis of many cancers, including breast cancer. Dysregulation of this pathway is associated with resistance both to endocrine and to HER2-directed therapies. The mTOR inhibitor everolimus has emerged as a promising agent in the treatment of several cancers and is now approved in combination with exemestane in postmenopausal hormone receptor–positive advanced breast cancer.

Everolimus carries a unique side-effect profile, which clinicians and patients should be aware of. With proper pretreatment planning and careful monitoring, treatment-related toxicities are generally manageable and the drug well tolerated.

Studies are ongoing to further define the role of everolimus in various subtypes and stages of breast cancer (see ). Identification of biomarkers capable of predicting sensitivity or resistance to mTOR inhibition in breast cancer remains an unmet need. As our understanding of the molecular profiles of tumors improves, we will be able to develop increasingly refined targeted agents as well as appropriate selection criteria that will lead to improved outcomes with minimal toxicities.

Table 1 Ongoing studies with everolimus

Disclosure

The authors disclose no potential conflicts of interest.

References

  • American Cancer SocietyCancer Facts and Figures 2012AtlantaAmerican Cancer Society2012
  • GermanoSO’DriscollLBreast cancer: understanding sensitivity and resistance to chemotherapy and targeted therapies to aid in personalised medicineCurr Cancer Drug Targets20099339841819442059
  • VivancoISawyersCLThe phosphatidylinositol 3-kinase AKT pathway in human cancerNat Rev Cance200227489501
  • WoodLDParsonsDWJonesSThe genomic landscapes of human breast and colorectal cancersScience200731858531108111317932254
  • CuiXZhangPDengWInsulin-like growth factor-I inhibits progesterone receptor expression in breast cancer cells via the phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin pathway: progesterone receptor as a potential indicator of growth factor activity in breast cancerMol Endocrinol200317457558812554765
  • ZhouBPHuMCMillerSAHER-2/neu blocks tumor necrosis factor-induced apoptosis via the Akt/NF-kappaB pathwayJ Biol Chem2000275118027803110713122
  • Stemke-HaleKGonzalez-AnguloAMLluchAAn integrative genomic and proteomic analysis of PIK3CA, PTEN, and AKT mutations in breast cancerCancer Res200868156084609118676830
  • WullschlegerSLoewithRHallMNTOR signaling in growth and metabolismCell2006124347148416469695
  • GuertinDASabatiniDMDefining the role of mTOR in cancerCancer Cell200712192217613433
  • Perez-TenorioGAlkhoriLOlssonBPIK3CA mutations and PTEN loss correlate with similar prognostic factors and are not mutually exclusive in breast cancerClin Cancer Res200713123577358417575221
  • KarakasBBachmanKEParkBHMutation of the PIK3CA oncogene in human cancersBr J Cancer200694445545916449998
  • deGraffenriedLAFriedrichsWERussellDHInhibition of mTOR activity restores tamoxifen response in breast cancer cells with aberrant Akt ActivityClin Cancer Res200410238059806715585641
  • MusgroveEASutherlandRLBiological determinants of endocrine resistance in breast cancerNat Rev Cancer20099963164319701242
  • CholletPAbrialCTaccaOMammalian target of rapamycin inhibitors in combination with letrozole in breast cancerClin Breast Cancer20067433633817092402
  • NagataYLanKHZhouXPTEN activation contributes to tumor inhibition by trastuzumab, and loss of PTEN predicts trastuzumab resistance in patientsCancer Cell20046211712715324695
  • BernsKHorlingsHMHennessyBTA functional genetic approach identifies the PI3K pathway as a major determinant of trastuzumab resistance in breast cancerCancer Cell200712439540217936563
  • KataokaYMukoharaTShimadaHSaijoNHiraiMMinamiHAssociation between gain-of-function mutations in PIK3CA and resistance to HER2-targeted agents in HER2-amplified breast cancer cell linesAnn Oncol201021225526219633047
  • BjornstiMAHoughtonPJThe TOR pathway: a target for cancer therapyNat Rev Cancer20044533534815122205
  • CampbellRABhat-NakshatriPPatelNMConstantinidouDAliSNakshatriHPhosphatidylinositol 3-kinase/AKT-mediated activation of estrogen receptor alpha: a new model for anti-estrogen resistanceJ Biol Chem2001276139817982411139588
  • DeGraffenriedLFriedrichsWFulcherLThe mTOR inhibitor CCI-779 restores tamoxifen response in breast cancer cells with high Akt activity. Proceedings of the 14th NCI-EORTC-AACR Symposium on Molecular Targets and Cancer TherapeuticsEur J Cancer2002S158
  • SmithCLCross-talk between peptide growth factor and estrogen receptor signaling pathwaysBiol Reprod19985836276329510949
  • BoulayARudloffJYeJDual inhibition of mTOR and estrogen receptor signaling in vitro induces cell death in models of breast cancerClin Cancer Res200511145319532816033851
  • TreeckOWackwitzBHausUOrtmannOEffects of a combined treatment with mTOR inhibitor RAD001 and tamoxifen in vitro on growth and apoptosis of human cancer cellsGynecol Oncol2006102229229916443261
  • BeeramMTanQTTekmalRRRussellDMiddletonADeGraffenriedLAAkt-induced endocrine therapy resistance is reversed by inhibition of mTOR signalingAnn Oncol20071881323132817693645
  • CarpenterJTRochéHCamponeMRandomized 3-arm, Phase 2 study of temsirolimus (CCI-779) in combination with letrozole in postmenopausal women with locally advanced or metastatic breast cancerJ Clin Oncol 2005 ASCO Annual Meeting Proceedings23Suppl 16 Part I of II (June 1 Suppl), 2005564
  • WolffACLazarAABondarenkoIRandomized Phase III placebo-controlled trial of letrozole plus oral temsirolimus as first-line endocrine therapy in postmenopausal women with locally advanced or metastatic breast cancerJ Clin Oncol201331219520223233719
  • KirchnerGIMeier-WiedenbachIMannsMPClinical pharmacokinetics of everolimusClin Pharmacokinet2004432839514748618
  • AwadaACardosoFFontaineCThe oral mTOR inhibitor RAD001 (everolimus) in combination with letrozole in patients with advanced breast cancer: results of a phase I study with pharmacokineticsEur J Cancer2008441849118039566
  • BaselgaJSemiglazovVvan DamPPhase II randomized study of neoadjuvant everolimus plus letrozole compared with placebo plus letrozole in patients with estrogen receptor-positive breast cancerJ Clin Oncol200927162630263719380449
  • SabineVSSimsAHMacaskillEJGene expression profiling of response to mTOR inhibitor everolimus in pre-operatively treated post-menopausal women with oestrogen receptor-positive breast cancerBreast Cancer Res Treat2010122241942820480226
  • BachelotTBourgierCCropetCRandomized Phase II trial of everolimus in combination with tamoxifen in patients with hormone receptor-positive, human epidermal growth factor receptor 2-negative metastatic breast cancer with prior exposure to aromatase inhibitors: a GINECO studyJ Clin Oncol201230222718272422565002
  • BaselgaJCamponeMPiccartMEverolimus in postmenopausal hormone-receptor-positive advanced breast cancerN Engl J Med2012366652052922149876
  • SlamonDJClarkGMWongSGLevinWJUllrichAMcGuireWLHuman breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogeneScience198723547851771823798106
  • SlamonDEiermannWRobertNAdjuvant trastuzumab in HER2-positive breast cancerN Engl J Med2011365141273128321991949
  • SlamonDJLeyland-JonesBShakSUse of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2N Engl J Med20013441178379211248153
  • RomondEHPerezEABryantJTrastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancerN Engl J Med2005353161673168416236738
  • Piccart-GebhartMJProcterMLeyland-JonesBTrastuzumab after adjuvant chemotherapy in HER2-positive breast cancerN Engl J Med2005353161659167216236737
  • HarariDYardenYMolecular mechanisms underlying ErbB2/HER2 action in breast cancerOncogene200019536102611411156523
  • LuCHWyszomierskiSLTsengLMPreclinical testing of clinically applicable strategies for overcoming trastuzumab resistance caused by PTEN deficiencyClin Cancer Res200713195883588817908983
  • AndreFCamponeMO’ReganRPhase I study of everolimus plus weekly paclitaxel and trastuzumab in patients with metastatic breast cancer pretreated with trastuzumabJ Clin Oncol201028345110511520975068
  • DalencFCamponeMHupperetsPEverolimus in combination with weekly paclitaxel and trastuzumab in patients (pts) with HER-2-overexpressing metastatic breast cancer (MBC) with prior resistance to trastuzumab and taxanes: a multicenter phase II clinical trialProceedings of the American Society of Clinical Oncology Annual MeetingJune 4–8, 2010Chicago, IL, USA
  • JerusalemGFasoloADierasVPhase I trial of oral mTOR inhibitor everolimus in combination with trastuzumab and vinorelbine in pre-treated patients with HER2-overexpressing metastatic breast cancerBreast Cancer Res Treat2011125244745521107682
  • CamponeMGianniLMassacesiCTrastuzumab and everolimus (RAD001) containing regimens are safe and active when reintroduced in patients with HER2-overexpressing metastatic breast cancer (MBC) pre-treated with lapatinib [abstract]Eur J Cancer Suppl20108186
  • MorrowPKWulfGMEnsorJPhase I/II study of trastuzumab in combination with everolimus (RAD001) in patients with HER2-overexpressing metastatic breast cancer who progressed on trastuzumab-based therapyJ Clin Oncol201129233126313221730275
  • Everolimus in Combination With Trastuzumab and Paclitaxel in the Treatment of HER2 Positive Locally Advanced or Metastatic Breast Cancer (BOLERO-1) Available from http://clinicaltrials.gov/ct2/show/NCT00876395Accessed March 21, 2013
  • Daily Everolimus in Combination With Trastuzumab and Vinorelbine in HER2/Neu Positive Women With Locally Advanced or Metastatic Breast Cancer (BOLERO-3) Available from http://clinicaltrials.gov/ct2/show/NCT01007942Accessed March 21, 2013
  • IrvinWJJrCareyLAWhat is triple-negative breast cancer?Eur J Cancer200844182799280519008097
  • DentRTrudeauMPritchardKITriple-negative breast cancer: clinical features and patterns of recurrenceClin Cancer Res20071315 Pt 14429443417671126
  • Gonzalez-AnguloAGreenMMurrayJOpen label randomized clinical trial of standard neoadjuvant chemotherapy with paclitaxel followed by FEC (T-FEC) vs the combination of paclitaxel and RAD001 followed by FEC (TR-FEC) in women with triple receptor-negative breast cancer (TNBC)Presented at the American Society of Clinical Oncology Annual MeetingJune 37, 2011Chicago, IL, USA
  • SinghJSteinSVolmMPhase II trial of RAD001 plus carboplatin in patients with triple-negative metastatic breast cancerJ Clin Oncol201230Suppl 15e11529
  • JerusalemGEllardSFasoloANon-infectious pneumonitis in breast cancer patients treated with Everolimus containing therapy: analysis of five studiesCancer Res200969supp 241115
  • Afinitor® (everolimus) tablets for oral administration [package insert]East Hanover, NJNovartis Pharmaceuticals Corporation
  • Di NicolantonioFArenaSTaberneroJGrossoSDeregulation of the PI3K and KRAS signaling pathways in human cancer cells determines their response to everolimusJ Clin Invest201012082858286620664172
  • JankuFWhelerJJWestinSNPI3K/AKT/mTOR inhibitors in patients with breast and gynecologic malignancies harboring PIK3CA mutationsJ Clin Oncol201230877778222271473
  • ChenGYangNWangXIdentification of p27/KIP1 expression level as a candidate biomarker of response to rapalogs therapy in human cancerJ Mol Med (Berl)201088994195220508912