321
Views
20
CrossRef citations to date
0
Altmetric
Review

Clinical potential of novel therapeutic targets in breast cancer: CDK4/6, Src, JAK/STAT, PARP, HDAC, and PI3K/AKT/mTOR pathways

&
Pages 203-215 | Published online: 06 Aug 2014

Abstract

Breast cancers expressing estrogen receptor α, progesterone receptor, or the human epidermal growth factor receptor 2 (HER2) proto-oncogene account for approximately 90% of cases, and treatment with antiestrogens and HER2-targeted agents has resulted in drastically improved survival in many of these patients. However, de novo or acquired resistance to antiestrogen and HER2-targeted therapies is common, and many tumors will recur or progress despite these treatments. Additionally, the remaining 10% of breast tumors are negative for estrogen receptor α, progesterone receptor, and HER2 (“triple-negative”), and a clinically proven tumor-specific drug target for this group has not yet been identified. Therefore, the identification of new therapeutic targets in breast cancer is of vital clinical importance. Preclinical studies elucidating the mechanisms driving resistance to standard therapies have identified promising targets including cyclin-dependent kinase 4/6, phosphoinositide 3-kinase, poly adenosine diphosphate–ribose polymerase, Src, and histone deacetylase. Herein, we discuss the clinical potential and status of new therapeutic targets in breast cancer.

Introduction

Breast cancer is the most commonly diagnosed female cancer, and the second leading cause of cancer-related death in women in the United States with >232,000 new cases and 40,000 deaths projected in the year 2014.Citation1 Clinical breast cancer subtypes are based on the expression of the hormone receptors (HRs) for estrogen (ER) and progesterone (PR), and overexpression/amplification of the human epidermal growth factor receptor 2 (HER2) (ERBB2) proto-oncogenic receptor, tyrosine kinase. Subtype drives the selection of appropriate therapies; HR+ tumors constitute ∼75% of cases and are treated with antiestrogen therapies that block ER transcriptional activity (for example, aromatase inhibitors, tamoxifen, fulvestrant). HER2+ tumors comprise ∼20% of cases, half of which are also HR-positive, and are managed with HER2-targeted agents (for example, trastuzumab, pertuzumab, lapatinib, trastuzumab emtansine). Tumors that lack the expression of ER, PR, and HER2 are termed “triple-negative breast cancers (TNBC)”; they comprise 5%–10% of cases, and are managed only with genotoxic chemotherapy.Citation2 Breast tumors are also molecularly subtyped based on gene expression profiling into luminal A, luminal B, HER2-enriched, basal-like, claudin-low, and normal-like.Citation3Citation5 Luminal A and B tumors tend to express ER, and basal and claudin-low tumors are often triple-negative.Citation3,Citation6

While tumor-targeted agents have been extremely effective in treating HR+ and HER2+ breast cancers, de novo or acquired drug resistance is common and many cancers will recur or progress.Citation7,Citation8 Alternatively, TNBC does not yet have an obvious tumor-specific receptor or pathway to target. Therefore, identifying new drug targets in breast cancer is a high clinical priority, and combining therapeutics to simultaneously target multiple oncogenic signaling pathways may be a key to overcoming/preventing resistance.

Targeting CDK4 and CDK6 in breast cancer

The role of CDK4/6 in the cell cycle

Phases of the cell cycle are tightly controlled by oscillating levels of cyclin proteins. In the G1 phase, cells grow and prepare to replicate deoxyribonucleic acid (DNA) in the S phase. In order for a cell to proliferate, it must first satisfy the requirements of restriction checkpoints in G1 before progressing into the S phase. If these checkpoints are not satisfied, the cell exits the cell cycle and enters a quiescent state (G0) and, under some circumstances, senescence (permanent cell cycle arrest). Alternatively, sufficient mitogenic signals will drive the production of cyclin D proteins that can associate with and activate their catalytic partners, cyclin-dependent kinase (CDK)4 or CDK6. CDK4 and CDK6 have overlapping and distinct functions,Citation9 the most characterized of which is phosphorylation of the Rb (RB1) tumor suppressor. Once associated with cyclin D, CDK4/6 phosphorylates Rb at Ser780 and Ser795, resulting in Rb inactivation, thereby releasing E2F transcription factors to initiate the transcription of genes required for DNA replication. Progression through G1 is also limited by the abundance of cyclin-dependent kinase inhibitors (CKIs) such as p16INK4 proteins, which block the binding site of cyclin D on CDK4/6, and p27KIP1, which has broad specificity for the cyclin/CDK complexes.

Deregulation of the cell cycle in cancer

Aberrant cell cycle control is a hallmark of cancer,Citation10 and multiple mechanisms contribute to deregulation of the G1-to-S checkpoint. Mitogen-activated pathways that drive the production of cyclin D1 may be stimulated by aberrant expression of growth factors or growth factor receptors, which can activate cells to produce cyclin D1 in an autocrine manner.Citation11 These pathways may also be constitutively active due to the overexpression of downstream signaling molecules that drive the expression of cyclin D1, such as Ras proteins, which are frequently overexpressed in breast cancer and are associated with poor prognosis.Citation12 Tumor cells have also been shown to frequently manipulate the expression of the cyclin and CDK genes (). Many tumors alter the messenger ribonucleic acid (mRNA) and protein levels of key cell cycle regulatory components. For example, cyclin D1 mRNA is overexpressed in over 50% of breast cancers.Citation13 Similarly, overexpression of CDK4/6 or the loss of CKIs is commonly seen in tumors.Citation14 The gene encoding p16INK4A (CDKN2A) has more homozygous deletions than any other recessive cancer-related gene.Citation15 It is important to note that deregulation of cell cycle proteins has been shown to differ according to breast cancer subtype. For example, amplification of cyclin D1 (CCND1) occurs in a much greater percentage of luminal B breast cancers than in luminal A (58% versus 29%, respectively).Citation16 Similarly, the luminal B subtype is more often associated with a gain of CDK4 (25% of luminal B versus 14% of luminal A), and a loss of negative regulators including p16INK4A.Citation16 In contrast, basal-like breast cancers do not typically display alterations in cyclin D1 or CDK4/6, but 20% of cases harbor mutations or lead to the homozygous loss of RB1, indicating that this breast cancer subtype may respond less frequently to treatment with CDK4/6 inhibitors.Citation16 The relationship between cyclin D, CDK4/6, CKIs, and Rb allows cancer cells to gain a proliferative advantage by altering members of this pathway to enhance the inactivation of Rb. Therefore, it is not surprising that deregulation of at least one of these proteins is seen in nearly all human cancers.Citation14,Citation17

Figure 1 Frequencies of genetic lesions identified in primary breast tumors.

Notes: The percentages of tumors exhibiting mutations, amplifications, or homozygous deletions of genes in the PI3K/AKT/mTOR and CDK4/6 pathways are indicated. Data from the TCGA dataset containing molecular subtyping data of 515 breast tumorsCitation16 were extracted and used to generate an oncoprint plot using the cBio Cancer Genomics Portal.Citation155,Citation156 The results published here are in whole or part based upon data generated by the TCGA Research Network (http://cancergenome.nih.gov/).
Abbreviations: HER2, human epidermal growth factor receptor 2; C-L, claudin-low; PI3K, phosphoinositide 3-kinase; EGFR, epidermal growth factor receptor; IGF-1R, insulin-like growth factor-1 receptor; InsR, insulin receptor; CDK, cyclin-dependent kinase; CCND, cyclin D; mTOR, mammalian target of rapamycin; TCGA, The Cancer Genome Atlas; PTEN, phosphatase and tensin homolog.
Figure 1 Frequencies of genetic lesions identified in primary breast tumors.

CDK4/6 inhibitors in clinical development

Until recently, nonspecific CDKIs have been the primary mechanism of therapeutically targeting kinases in the cell cycle.Citation18 However, these therapeutics often elicit adverse effects in patients.Citation18 The broad-spectrum CKI, flavopiridol, had promising preclinical results in multiple tumor cell types,Citation19Citation21 but it exhibited adverse effects and high toxicity in early-phase clinical trials; furthermore, it did not meet expectations with regard to efficacy against most tumor types with the exception of leukemia.Citation22Citation26 To limit toxicity and increase antitumor efficacy, there has been interest in the development of more specific CKIs. CDK4/6 are attractive targets due to their central role in the inactivation of Rb and their frequent deregulation in tumor cells. Palbociclib (PD-0332991) is the first highly selective inhibitor of CDK4/6 to be tested in humans. This drug exhibits an in vitro half maximal inhibitory concentration (IC50) of 10–15 nM for CDK4/6, compared to >5 μM for CDK2.Citation27 Initial studies in cultured cancer cells showed a reduction in Rb phosphorylation within 4 hours of treatment with palbociclib, reaching a maximum at 16 hours posttreatment; Rb inhibition was completely reversed within 2 hours following drug removal.Citation28 When tested against a panel of breast cancer cell lines, palbociclib preferentially inhibited growth of the ER+ cell lines, particularly among those with higher Rb and cyclin D levels, and lower p16INK4A expression.Citation29 Palbociclib treatment also effectively suppressed the growth of ER+ human breast cancer xenografts in mice.Citation30 The only known mechanism of resistance to CDK4/6 inhibition is the loss of Rb function.Citation28

Despite immense progress in the treatment of ER+ breast cancer with the development of antiestrogens, approximately 50% of breast cancers will develop endocrine resistance and progress on antiestrogen therapies.Citation31 One target gene of ER is cyclin D1 (CCND1), and antiestrogens have been shown to inhibit the cell cycle via downregulation of CDK activity and the subsequent phosphorylation of Rb to cause G0/G1 arrest.Citation32Citation34 Therefore, it is not surprising that deregulation of cyclin D1 and Rb phosphorylation have been associated with antiestrogen resistance in in vitro and in vivo models.Citation35Citation37 Given that tumors progressing on endocrine therapy are likely to have an intact cell cycle program, there is reason to investigate the therapies targeting CDK4/6 in endocrine-resistant tumors. Treatment of luminal ER+ breast cancer cells with palbociclib and tamoxifen elicits a synergistic effect.Citation29 Furthermore, palbociclib treatment was able to partially restore sensitivity to tamoxifen in a tamoxifen-resistant cell line, and it abrogated growth of ER+ cells with acquired resistance to estrogen deprivation.Citation29,Citation30,Citation38 Similarly, another CDK4/6 inhibitor now in clinical development (LEE011) has shown potent antitumor activity in preclinical models, including those resistant to the phosphoinositide 3-kinase (PI3K) inhibitor, BYL719 (described within the PI3K pathway inhibitors as single agents section).Citation39 These promising preclinical data have led to numerous clinical trials of CDK4/6 inhibitors in various types of cancer.

Phase I trials of palbociclib tested different treatment schedules and doses in patients with Rb+ advanced solid tumors or non-Hodgkin’s lymphoma that were refractory to standard treatment. For both a 2-week-on, 1-week-off schedule (2:1), and a 3-week-on, 1-week-off schedule (3:1), neutropenia and thrombocytopenia were defined as the dose-limiting toxicities with maximum tolerated doses (MTDs) of 200 mg/day and 125 mg/day, respectively.Citation40,Citation41 With the 2:1 treatment schedule, three of 31 of patients exhibited stable disease for >10 cycles of treatment.Citation40 Similarly, in the 3:1 treatment group, six of 37 patients maintained disease stabilization for >10 cycles of treatment; one patient who benefitted from 50 mg/day palbociclib had breast cancer with strong Rb positivity.Citation41 Based on these results, a dose of 125 mg/day and a 3:1 schedule has been further pursued in Phase II trials of palbociclib (NCT01684215, NCT01536743, NCT01709370) ().

Table 1 Clinical development status of targeted therapeutics in breast cancer

Based on the indications that palbociclib may be an effective treatment in combination with antiestrogens, and as a treatment for endocrine-resistant breast cancer, several clinical trials are ongoing to explore these findings in patients. A randomized, placebo-controlled, Phase II study to determine the benefit of adding palbociclib to the aromatase inhibitor, letrozole, as first-line therapy for advanced ER+/HER2− breast cancer in postmenopausal women (NCT00721409) has shown promising results thus far. The median progression-free survival (PFS) of the combination treatment was 26.1 months compared to 7.5 months for letrozole/placebo (hazard ratio=0.32; 95% confidence interval [CI]: 0.19–0.56; P<0.001). This study also established clinical benefit rates of 70% and 44% for treatment with letrozole/palbociclib and letrozole/placebo, respectively.Citation42 In attempts to define biomarkers predictive of a response to palbociclib, this study analyzed the amplification of CCND1 and/or the loss of CDKN2A (p16INK4A) by fluorescence in situ hybridization. However, these alterations were not predictive of benefit from adding palbociclib, highlighting the difficulty in finding predictive biomarkers in the heterogeneous background of human tumors. The preliminary results of this trial led to palbociclib receiving “breakthrough therapy designation” from the United States Food and Drug Administration in April 2013, which allowed for the accelerated approval of drugs that treat a life-threatening disease and display significant improvement over current therapy. A Phase III trial of the same design (NCT01740427; PALOMA-2), a Phase III study of palbociclib/fulvestrant versus placebo/fulvestrant in patients with ER+/HER2− advanced disease that progressed on an antiestrogen (NCT01942135; PALOMA-3), and a Phase III study of palbociclib/antiestrogen versus placebo/antiestrogen as an adjuvant therapy in patients with early-stage ER+/HER2− disease (NCT01864746; PENELOPE-B) are underway.

Two other CDK4/6 inhibitors (LEE011 and LY2835219) have been tested in dose-finding Phase I studies.Citation43 LEE011 is now being tested in a randomized Phase Ib/II study with the PI3K inhibitor BYL719 and letrozole (NCT01872260), and in a randomized Phase Ib/II study with the mammalian target of rapamycin (mTOR) complex 1 (mTORC1) inhibitor everolimus and the aromatase inhibitor exemestane (NCT01857193), both in patients with advanced ER+/HER2− breast cancer that progressed on an antiestrogen. LEE011 is also being tested with letrozole in a randomized presurgical study in patients with early-stage ER+/HER2− breast cancer to identify molecular changes in tumors and in a Phase III study to determine the efficacy of this combination (NCT01919229; MONALEESA-1, NCT01958021; MONALEESA-2). While palbociclib and LEE011 are being administered in a 3:1 schedule to ameliorate side effects such as neutropenia, LY2835219 is being administered on a continuous schedule with a lower incidence of neutropeniaCitation44 in a Phase Ib study in combination with several antiestrogens in patients with advanced ER+/HER2− antiestrogen-resistant disease (NCT02057133). The reason that these agents induce different adverse event profiles is unclear.

Potential of CDK4/6 inhibitors in combination with genotoxic agents

Traditional chemotherapeutic agents are thought to rely on the cell cycle to induce DNA damage that promotes apoptosis; therefore, the combination of these therapies with cell cycle inhibitors like palbociclib may not be a viable treatment strategy. In vitro data suggest that a concomitant treatment of chemotherapeutics and palbociclib may result in decreased efficacy as compared to single-agent treatments, and in some cases, combination treatment antagonizes chemotherapy-induced cell death.Citation45Citation47 Palbociclib also antagonized mitotic catastrophe and subsequent cell death associated with paclitaxel, a microtubule-stabilizing agent.Citation46

Although palbociclib does not appear to enhance tumor killing in combination with chemotherapeutics, there is potential in using CDK4/6 inhibitors transiently to synchronize cells before treating them with genotoxic agents to enhance the cytotoxic effect. Short-term treatment with palbociclib before or concurrent with paclitaxel suppressed cell growth more effectively than single agents or continuous treatment with both agents.Citation46 A Phase I clinical study is ongoing to determine the MTD of palbociclib in combination with paclitaxel in patients with Rb+ advanced breast cancer (NCT01320592).

Targeting the PI3K/AKT/mTOR pathway in breast cancer

Pathway deregulation in breast cancer

PI3K/AKT/mTOR signaling regulates multiple cellular processes to promote cancer cell growth, survival, and metastasis.Citation48Citation50 This is the most frequently aberrantly activated pathway in human breast cancer, with alterations in genes encoding the pathway components occurring in >80% of cases ().Citation51 The deregulation of class 1A PI3K signaling has also been associated with the development of resistance to a variety of cancer therapies, including antiestrogens, trastuzumab, radiation, and chemotherapy.Citation52Citation54 However, mechanisms of PI3K pathway activation differ between breast cancer subtypes. Activating mutations in PIK3CA occur more frequently in luminal A, luminal B, and HER2+ breast cancers (45%, 29%, and 39%, respectively) as compared to basal-like breast cancers (9%).Citation16 In contrast, phosphatase and tensin homolog (PTEN) expression is decreased in 67% of TNBCs, compared with 29%–44% in ER+ tumors and 22% in HER2+ tumors.Citation55Citation58 The high frequency of PI3K pathway alteration in human breast tumors makes this pathway a promising target for therapeutics, and inhibitors of PI3K, AKT, and/or mTOR are in clinical development.

PI3K pathway inhibitors as single agents

PI3K inhibitors have exhibited clinical activity against advanced solid tumors and metastatic breast cancers.Citation59,Citation60 A Phase I study of the pan-PI3K inhibitor, BKM120, in patients with metastatic breast cancer reported partial responses and stable disease in 11% and 50% of patients, respectively.Citation61 A Phase II trial is ongoing to investigate BKM120 for the treatment of metastatic TNBC (NCT01629615). Preliminary results of a first-in-human Phase I trial testing the PI3K/p110α isoform-specific inhibitor, BYL719, in advanced solid tumors that harbor activating mutations in p110α (PIK3CA) (NCT01219699) suggest a favorable safety profile and an MTD of 400 mg/day. Partial responses were observed in seven of 39 patients, including two with ER+ breast cancer. At a dose of 270 mg/day, the median PFS for 15 patients with ER+/HER2− metastatic breast cancer was 5.5 months.Citation62 While the presence of PIK3CA mutations or PTEN loss is predictive of a response to PI3K and AKT inhibitors in cell lines,Citation63Citation67 it remains to be determined whether these biomarkers remain predictive in human breast cancers.Citation59,Citation68,Citation69 Phase I trials are ongoing to investigate BYL719 in combination with endocrine therapies including letrozole and exemestane in HR+ metastatic breast cancer (NCT01870505, NCT01791478).

The allosteric mTORC1 inhibitor, everolimus, also showed efficacy as a single agent in a Phase II placebo-controlled study in breast cancer patients with bone metastases. This study reported improved time-to-progression from 12.6 weeks in the placebo arm to 37 weeks in the everolimus arm (HR =0.464; 95% CI: 0.226–0.954; P=0.037, adjusted for endocrine therapy).Citation70 Another Phase II trial is ongoing to test the benefit of everolimus after preoperative chemotherapy in patients with invasive breast cancer (NCT01088893).

PI3K pathway inhibitors for the treatment of ER+ breast cancer

There is ample evidence that HR+ tumors resistant to endocrine therapy exhibit increased PI3K/AKT/mTOR signaling.Citation51,Citation53,Citation63 Accordingly, treatment of endocrine-resistant cell lines and xenografts with inhibitors of PI3K, AKT, and/or mTOR abrogates endocrine resistance.Citation63,Citation71Citation74 Therefore, treatment with inhibitors of the PI3K/ATK/mTOR pathway in combination with endocrine therapies may prevent an escape from endocrine dependence in these tumors, and restore sensitivity to endocrine agents in breast cancer.

The first PI3K/AKT/mTOR pathway-targeted therapeutic to enter routine clinical use for cancer is everolimus. A randomized Phase II trial investigating the clinical benefit of everolimus in combination with tamoxifen versus tamoxifen/placebo in patients with ER+/HER2− metastatic disease that had prior exposure to aromatase inhibitors reported an improved 6-month clinical benefit rate (61% versus 42%, exploratory P=0.045) and time-to-progression (8.6 months versus 4.5 months, exploratory P=0.002) in the combination arm compared to tamoxifen/placebo, corresponding to a reduction in the risk of progression (46% decrease) and the risk of death (55% decrease) with combination treatment.Citation75 Similarly, the randomized Phase III Breast Cancer Trials of Oral Everolimus-2 (BOLERO-2) trialCitation76 led to the approval of everolimus in combination with exemestane for the treatment of ER+/HER2− postmenopausal breast cancer patients with the disease that progressed during treatment with a nonsteroidal aromatase inhibitor. BOLERO-2 reported a median PFS of 11.0 months and 4.1 months for the combination treatment and exemestane/placebo, respectively (HR =0.38; 95% CI: 0.31–0.48; log-ranked P<0.0001).Citation76 Sequencing of 182 genes to identify a lesion or lesions predictive of benefit from exemestane/everolimus did not reveal an obvious single biomarker, but patients with tumors harboring minimal combined lesions in PIK3CA, PTEN, CCND1, FGFR1, and FGFR2 derived greater benefit from the combination (HR =0.27).Citation77 The benefit of continued endocrine therapy (with exemestane) is being tested in an ongoing randomized Phase III trial of everolimus alone versus everolimus/exemestane versus capecitabine in the same patient population (NCT01783444).

Preclinical and clinical data indicate that mTORC1 inhibition (for example, with everolimus) derepresses negative feedback that results in the upregulation of growth factor receptor, PI3K/AKT, and MEK/ERK signaling.Citation78Citation80 Thus, targeting signaling nodes upstream of mTORC1 may be more effective. A Phase Ib/II trial testing the PI3K inhibitor, BKM120, with letrozole in patients with ER+/HER2− metastatic breast cancer is ongoing.Citation81 Early results suggest that this drug combination is active with a tolerable safety profile. Unexpectedly, the mutation status of PIK3CA did not correlate with clinical benefit.Citation81 BKM120 is being tested in Phase II and III trials in combination with fulvestrant in postmenopausal women with ER+/HER2− advanced disease with cancer that progressed on an aromatase inhibitor with or without an mTORC1 inhibitor (NCT01339442, NCT01610284 [BELLE-2], NCT01633060 [BELLE-3]). Similarly, the pan-PI3K inhibitor GDC-0941 and the PI3K/mTOR dual inhibitor GDC-0980 are being tested with fulvestrant in a randomized Phase II placebo-controlled study in patients with advanced disease resistant to aromatase inhibitors (NCT01437566). In addition, a Phase I trial is testing the efficacy of the AKT inhibitor, MK2206, in combination with anastrozole or fulvestrant, or with anastrozole plus fulvestrant in patients with metastatic ER+ disease (NCT01344031).

Targeting the PI3K pathway in HER2+ breast cancer

Activation of PI3K/AKT/mTOR signaling via a PIK3CA mutation in HER2+ breast cancer has been associated with resistance to trastuzumab and lapatinib, while the effects of PTEN loss remain controversial.Citation82Citation86 In preclinical models, everolimus plus trastuzumab had greater antitumor effects than either treatment alone.Citation78 Similarly, BKM120 was shown to synergize with trastuzumab in HER2+ breast cancer cell lines, and treatment with BKM120 partially restored sensitivity to trastuzumab in trastuzumab-resistant HER2+ xenografts.Citation87 Recently, a Phase I/II trialCitation88 was completed for patients with advanced HER2+ trastuzumab-resistant disease, showing that combined treatment with BKM120 and trastuzumab was well tolerated and elicited clinical benefit in 25% of patients. These results were comparable to those of a prior studyCitation89 examining the combination of everolimus and trastuzumab in a similar patient population, which reported a 35% rate of clinical benefit. Based on these encouraging results, a Phase II placebo-controlled trial is ongoing to evaluate the efficacy of BKM120 in combination with paclitaxel and trastuzumab in the neoadjuvant setting in patients with early-stage HER2+ breast cancer (NCT01816594, NeoPHOEBE). BKM120 is also being tested in combination with lapatinib in patients with advanced trastuzumab-resistant HER2+ breast cancer harboring lesions in PIK3CA or PTEN (NCT01589861, PIKHER2).

PI3K inhibitors in combination with genotoxic agents

Activation of PI3K/AKT signaling has been associated with resistance to DNA-damaging chemotherapeutics in vitro and in vivo,Citation90 and several clinical trials are ongoing to explore the potential of combining PI3K inhibitors and genotoxic agents to prevent resistance. The benefit of BKM120 and paclitaxel as a first-line treatment for HER2-negative advanced breast cancer is being investigated in an ongoing placebo-controlled Phase II trial (NCT01572727). Similarly, GDC-0941 is being tested in combination with paclitaxel in a Phase II randomized study in patients with advanced disease (NCT01740336).

Inhibiting IGF-1R/InsR signaling in breast cancer

Treatment with antiestrogens and HER2-blocking agents has not only been shown to result in compensatory upregulation of PI3K/AKT/mTOR signaling; upstream receptor tyrosine kinases including insulin-like growth factor (IGF)-1 receptor (IGF-1R) are congruently upregulated.Citation63,Citation91,Citation92 Furthermore, IGF-1R was found to be expressed in ∼90% of breast cancers and associated with worse prognosis in ER+ breast cancer patients.Citation93Citation95 As IGF-1R is a potent driver of PI3K signaling, preclinical and clinical studies explored the efficacy of IGF-1R-targeted agents in ER+ breast cancer. IGF-1R is highly homologous to the insulin receptor (InsR), and these proteins form homo- and heterodimers that confer ligand preferences for IGF-1 or insulin. Adenosine triphosphate (ATP)-competitive inhibitors target both receptors, while IGF-1R and IGF-1 (ligand-specific) antibodies are protein-specific. In ER+ models, treatment with an ATP-competitive IGF-1R/InsR inhibitor abrogated endocrine-resistant cell and tumor growth, while an IGF-1R antibody was less effective.Citation96 Such effects are likely due, in part, to compensatory upregulation of InsR when IGF-1R is targeted.

A recent Phase I trial of everolimus combined with the monoclonal IGF-1R antibody, figitumumab, showed stable disease in 15 of 18 patients with advanced solid tumors, and partial response in one patient.Citation97 Inhibition of PI3K signaling with BYL719 was shown to activate IGF-1R signaling in PI3KCA-mutant breast cancer cells and a xenograft model, while cotreatment of BYL719 and the IGF-1R antibody, ganitumab, induced tumor regression.Citation98 A Phase Ib/II trial is ongoing to study the same drug combination in patients with PI3KCA-mutant breast cancers (NCT01708161).

There is extensive crosstalk between ER and IGF-1R signaling, and preclinical studies show that targeting both pathways may be beneficial.Citation99,Citation100 However, a Phase II placebo-controlled studyCitation101 testing the IGF-1R monoclonal antibody ganitumab (AMG-479) with exemestane or fulvestrant in postmenopausal women with HR+ metastatic disease did not result in improved PFS and negatively impacted overall survival. Likewise, the ATP-competitive IGF-1R/InsR inhibitor, OSI-906, demonstrated preclinical efficacy in tumor modelsCitation96 and was being tested in a Phase II trial with letrozole ± the epidermal growth factor receptor inhibitor, erlotinib, in patients with ER+ metastatic breast cancer; however, this trial was terminated due to unacceptable toxicity and a lack of efficacy (NCT01205685). While initial trials of IGF-1R and IGF-1R/InsR inhibitors in ER+ disease have been disappointing, further study is necessary to determine the molecular markers that may predict benefit.

In vitro studies have demonstrated that IGF-1 plays a role in the development of resistance to chemotherapy in breast cancer.Citation102,Citation103 A Phase I trial of figitumumab in patients with advanced solid tumors showed clinical benefit, with ten of 15 patients experiencing stable disease at the maximum feasible dose of 20 mg/kg.Citation104 A Phase II trial in small cell lung cancer showed that figitumumab significantly improved response to carboplatin and paclitaxel.Citation105 While these results are promising, further study in breast cancer is needed to determine the best sequencing of IGF-1R inhibitors with chemotherapy to result in maximal tumor response.

IGF-1R signaling has been associated with resistance to HER2-targeted agents in preclinical studies.Citation92,Citation100,Citation102,Citation106 In vitro data have shown that the combination of trastuzumab and an anti-IGF1R antibody increases cytotoxicity compared to trastuzumab alone in breast cancer.Citation106 A Phase II trial investigating the addition of the IGF-1R antibody, cixutumumab, to lapatinib and capecitabine in HER2+ breast cancer patients is ongoing (NCT00684983).

An alternative approach to inhibiting IGF-1R is the depletion of a ligand. BI 836845 is an antibody that binds and sequesters IGF-1 and IGF-2.Citation107 This antibody is undergoing Phase I testing (NCT01403974, NCT02123823).

Anticancer effects of bone resorption inhibitors

Bone is a frequent site of metastasis in breast cancer patients. The bone microenvironment is enriched with growth factors to support cancer cell growth. Osteoclast-induced bone resorption leads to the increased release of growth factors, further enhancing proliferation and survival of cancer cells. Furthermore, breast cancer cells secrete growth factors and cytokines that contribute to osteoclast activity and inhibit osteoblast function, thereby maintaining a feed-forward cycle of bone resorption and cancer growth.Citation108 Bisphosphonates potently inhibit osteoclast-mediated bone resorption, and these compounds have been used clinically to prevent complications related to bone metastasis. More recently, these compounds have been adopted as treatments to prevent bone loss associated with chemotherapy or endocrine therapy in breast cancer patients.Citation109Citation111

Preclinical studies revealed that bisphosphonates have antitumor activity and prevent metastasis by inhibiting tumor cell invasion and promoting apoptosis.Citation112Citation116 These observations prompted clinical investigations of bone resorption inhibitors as anticancer agents, which have produced mixed results. Clinical trials examining the bisphosphonate, zoledronic acid, showed improvements in disease-free and overall survival compared to standard therapy alone, both in chemotherapy and endocrine therapy combinations. However, the observed survival benefit was restricted to postmenopausal women, or premenopausal women who had suppressed estrogen levels due to treatment with aromatase inhibitors or ovarian suppression.Citation117 Clinical trials investigating denosumab, an antibody against the osteoclast activator RANK ligand (RANKL), in the prevention of metastasis are ongoing (NCT00556374, NCT01077154).

Inhibiting PARP in triple-negative breast cancer

TNBCs are more aggressive and result in a worse prognosis when compared to other breast cancer subtypes.Citation118,Citation119 TNBCs are a heterogeneous subtype, and a single oncogenic driver has not been identified; therefore, tumor-targeted therapeutics are lacking. Poly(adenosine diphosphate–ribose) polymerase (PARP) is involved in the recognition and repair of DNA breaks, and PARP works in concert with other proteins including BRCA1 and BRCA2 to repair DNA damage. If DNA is irreparably damaged (for example, with genotoxic drugs), a cell will be forced to undergo apoptosis rather than replicate damaged DNA.Citation120 Preclinical data demonstrate that deficiency in BRCA1/2 or other homologous recombination DNA repair proteins sensitizes cells to PARP inhibition.Citation121 Therefore, as single agents, PARP inhibitors are likely to be more effective against tumors carrying mutations in genes encoding proteins involved in compensatory DNA repair mechanisms, such as BRCA1/2, as these mutations essentially prime the cells for DNA damage-induced apoptosis.Citation122,Citation123

There are two primary mechanisms of action of PARP inhibitors: catalytic PARP inhibition; and PARP–DNA trapping, where drugs trap PARP1 and PARP2 on single-stranded DNA breaks. Veliparib acts as a strong catalytic PARP inhibitor with a weak PARP–DNA trapping effect, while niraparib, olaparib, and rucaparib act through both mechanisms.Citation124,Citation125

A Phase I trialCitation126 in breast cancer patients with germline BRCA1/2 mutations showed that the PARP inhibitor, olaparib, was effective as a monotherapy, achieving objective responses in 41% of patients. TNBCs have a gene expression signature similar to that of BRCA-deficient tumors.Citation127 Furthermore, TNBCs frequently arise in carriers of BRCA1 mutations.Citation127 It is therefore reasonable to suggest treating TNBC with PARP inhibitors in combination with genotoxic therapy to activate a synthetic lethal interaction of both PARP and BRCA deficiency in cancer cells.

In a recent Phase I trialCitation128 of olaparib in combination with paclitaxel for metastatic TNBC, 37% of patients exhibited a partial response; however, this study was relatively small (n=19) and did not characterize the BRCA1/2 mutation status of patients. Rucaparib was well tolerated in a Phase I dose-escalation studyCitation129 in patients with advanced solid tumors; among 17 breast cancer patients, one BRCA1/2-mutant patient achieved a partial response, and four patients achieved stable disease for greater than 12 weeks. An exploratory analysis of BRCA1/2 mutation status in a Phase II study of veliparib in combination with temozolomide in metastatic TNBC reported a median PFS of 5.5 months in patients with known deleterious BRCA1/2 mutations versus 1.8 months in patients without these mutations.Citation130 Thus, stratification based on BRCA1/2 status may be a useful biomarker to predict the response to PARP inhibitors. Clinical trials are investigating the use of PARP inhibitors in combination with genotoxic therapies in patients with BRCA1/2 mutations (NCT00494234, NCT01989546, NCT01074970, NCT01945775).

Histone deacetylase inhibitors for breast cancer

Preclinical studies have demonstrated that histone deacetylases (HDACs) modulate ER activity, and HDAC inhibitors reverse resistance to antiestrogen therapies in vitro.Citation131Citation133 A Phase II trialCitation134 in patients with advanced ER+ breast cancer resistant to aromatase inhibition reported that the HDAC inhibitor, entinostat, combined with exemestane improved PFS from 2.3 months to 4.3 months (HR =0.73; 95% CI: 0.50–1.07; P=0.055) and the median overall survival from 19.8 months to 28.1 months compared with exemestane/placebo (HR =0.59; 95% CI: 0.36–0.97; P=0.036). Similarly, Munster et alCitation135 found in a Phase II trial in patients with ER+ endocrine-resistant metastatic disease that the combination of the HDAC inhibitor vorinostat and tamoxifen induced an objective response rate of 19%, a clinical benefit rate of 40%, and a median response duration of 10.3 months. A Phase II trial is ongoing in patients with chemotherapy-resistant advanced TNBC or ER+ disease to investigate combination treatment with entinostat and the DNA methyltransferase inhibitor, azacitidine (NCT01349959). Preliminary results of a Phase I trialCitation136 in patients with trastuzumab-resistant HER2+ metastatic disease indicate that the HDAC inhibitor, panobinostat, with trastuzumab is well tolerated and shows promising antitumor activity, with two of 13 patients experiencing tumor reduction thus far (NCT00567879).

Targeting Src kinases in breast cancer

The Src family of nonreceptor tyrosine kinases is involved in a variety of functions (for example, proliferation, motility, and invasion) through its interaction with mediators including steroid HRs, integrins, G protein-coupled receptors, signal transducer and activator of transcription (STAT) family members, and several other receptors and intracellular proteins that may contributed to the malignant phenotype observed in cancer cells (as reviewed by FrameCitation137). Gene expression profiling of primary and metastatic breast tumors indicates that a gene expression signature of Src pathway activation in the primary tumor is strongly predictive of bone metastasis,Citation138 and high Src mRNA expression is correlated with decreased survival, making Src a potential target for therapeutic interventions.Citation139Citation141 As a single agent, the antitumor activity of Src inhibitors has been disappointing in clinical trials for advanced breast cancer.Citation142,Citation143 However, in vitro studies have shown that Src inhibitors in combination with chemotherapy or endocrine therapy may enhance tumor cell death.Citation144 In a Phase I trial,Citation145 combination of the Src inhibitor dasatinib with paclitaxel in patients with metastatic breast cancer resulted in partial responses in four of 13 patients. Another Phase I trialCitation146 reported that six of 25 advanced breast cancer patients exhibited partial responses, and 32% had stable disease following combination treatment with dasatinib plus capecitabine. The results of a randomized Phase II trialCitation147 of dasatinib plus letrozole in HR+, HER2− postmenopausal metastatic breast cancer patients showed that the combination group had doubled PFS compared to the letrozole/placebo group (20.1 months versus 9.9 months, respectively; P=0.05). A Phase II trialCitation148 investigating dasatinib plus exemestane compared with exemestane/placebo in advanced ER+ breast cancer patients that had progressed on an aromatase inhibitor reported a modest improvement in median PFS in the combination group (18.1 weeks with dasatinib/exemestane versus 16.1 weeks with exemestane/placebo; P=0.148), and the proportion of patients with clinical benefit was 30.6% in the combination group compared to 12.24% with exemestane/placebo. Overall, these findings support the use of Src inhibitors with chemotherapeutics and endocrine therapies to more effectively treat advanced breast cancers.

Targeting the JAK/STAT pathway in breast cancer

Janus kinases (JAKs) are mediators of cytokine and growth hormone signaling. Activated JAKs phosphorylate STAT proteins, leading to their nuclear translocation and the transcriptional regulation of genes that regulate cell proliferation, differentiation, and apoptosis.Citation149,Citation150 Mutations in JAK and STAT proteins have been extensively characterized in myeloproliferative disorders, and these mutations have been linked to hyperactivation of the JAK/STAT pathway and, consequently, unchecked cell proliferation.Citation151 Comparable mutations have not been well studied in breast cancer. However STAT1, STAT3, and STAT5 are often constitutively phosphorylated, seemingly due to elevated levels of cytokines and receptors.Citation152 Preclinical studies showed that the interleukin-6/JAK2/STAT3 pathway is preferentially activated in basal-like breast cancer cells, and inhibition of JAK2 with NBP-BSK805 hinders the growth of patient-derived breast tumor xenografts.Citation153 There is also evidence supporting JAK2/STAT5 inhibitors as a means to overcome resistance to PI3K/mTOR inhibition in TNBC.Citation154

The JAK1/JAK2 inhibitor, ruxolitinib, is approved for the treatment of myelofibrosis, and it is currently being tested as a single agent and in combination with paclitaxel in Phase II trials for inflammatory breast cancer and TNBC (NCT01562873, NCT02041429), and in advanced ER+ breast cancer in combination with exemestane (NCT01594216). A randomized, double-blind, placebo-controlled Phase II trial is also underway to investigate ruxolitinib in combination with capecitabine in metastatic breast cancer (NCT02120417). In patients with metastatic HER2+ breast cancer, an ongoing Phase II trial is testing ruxolitinib with trastuzumab (NCT02066532).

Conclusion

The high incidence of recurrence and progression on standard therapies highlights the importance of developing new therapeutics for breast cancer. Deregulation of the cyclin D–CDK4/6–Rb axis is frequently seen in antiestrogen-resistant tumors. Therefore, inhibitors of this pathway, including palbociclib, LEE011, and LY2835219, are promising as effective treatments for endocrine-resistant breast cancer. Activation of the PI3K/AKT/mTOR pathway has also been shown to promote drug resistance. The PI3K and mTOR inhibitors in clinical development have shown efficacy as single agents and in combination with antiestrogens or HER2-targeted agents. The search for a targeted therapeutic to treat TNBC has led to the development of PARP inhibitors, which appear to be considerably effective in a subgroup of TNBC patients harboring mutations in BRCA1/2. However, the TNBC subtype is heterogeneous, so further identification of targeted therapies for this group is needed. Immense progress has been made in identifying new targets in breast cancer, and future advances in therapy will likely include simultaneous or sequential targeting of multiple pathways to maximally inhibit tumor growth while thwarting the development of drug resistance.

Disclosure

TWM has received commercial research support from Piramal Life Sciences, Ltd. SRH reports no conflicts of interest in this work.

References

  • American Cancer SocietyBreast Cancer: Facts and Figures 2013–2014Atlanta, GAAmerican Cancer Society, Inc2013
  • CurtisCShahSPChinSFThe genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroupsNature2012486740334635222522925
  • SørlieTPerouCMTibshiraniRGene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implicationsProc Natl Acad Sci U S A20019819108691087411553815
  • PratAParkerJSKarginovaOPhenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancerBreast Cancer Res2010125R6820813035
  • PerouCMSørlieTEisenMBMolecular portraits of human breast tumoursNature2000406679774775210963602
  • BertucciFFinettiPCerveraNHow basal are triple-negative breast cancers?Int J Cancer2008123123624018398844
  • RexerBNArteagaCLIntrinsic and acquired resistance to HER2-targeted therapies in HER2 gene-amplified breast cancer: mechanisms and clinical implicationsCrit Rev Oncog201217111622471661
  • GiulianoMSchifpROsborneCKTrivediMVBiological mechanisms and clinical implications of endocrine resistance in breast cancerBreast201120Suppl 34249
  • AndersLKeNHydbringPA systematic screen for CDK4/6 substrates links FOXM1 phosphorylation to senescence suppression in cancer cellsCancer Cell201120562063422094256
  • HanahanDWeinbergRAHallmarks of cancer: the next generationCell2011144564667421376230
  • FlemingTPMatsuiTMolloyCJRobbinsKCAaronsonSAAutocrine mechanism for v-sis transformation requires cell surface localization of internally activated growth factor receptorsProc Natl Acad Sci U S A19898620806380672813378
  • ClarkGJDerCJAberrant function of the Ras signal transduction pathway in human breast cancerBreast Cancer Res Treat19953511331447612899
  • MusgroveEACaldonCEBarracloughJStoneASutherlandRLCyclin D as a therapeutic target in cancerNat Rev Cancer201111855857221734724
  • MalumbresMBarbacidMTo cycle or not to cycle: a critical decision in cancerNat Rev Cancer20011322223111902577
  • BignellGRGreenmanCDDaviesHSignatures of mutation and selection in the cancer genomeNature2010463728389389820164919
  • Cancer Genome Atlas NetworkComprehensive molecular portraits of human breast tumoursNature20124907418617023000897
  • HallMPetersGGenetic alterations of cyclins, cyclin-dependent kinases, and Cdk inhibitors in human cancerAdv Cancer Res199668671088712071
  • DicksonMASchwartzGKDevelopment of cell-cycle inhibitors for cancer therapyCurr Oncol2009162364319370178
  • BlagosklonnyMVFlavopiridol, an inhibitor of transcription: implications, problems and solutionsCell Cycle20043121537154215539947
  • ByrdJCShinnCWaselenkoJKFlavopiridol induces apoptosis in chronic lymphocytic leukemia cells via activation of caspase-3 without evidence of bcl-2 modulation or dependence on functional p53Blood19989210380438169808574
  • KitadaSZapataJMAndreeffMReedJCProtein kinase inhibitors flavopiridol and 7-hydroxy-staurosporine down-regulate antiapoptosis proteins in B-cell chronic lymphocytic leukemiaBlood200096239339710887097
  • ShapiroGISupkoJGPattersonAA phase II trial of the cyclin-dependent kinase inhibitor flavopiridol in patients with previously untreated stage IV non-small cell lung cancerClin Cancer Res2001761590159911410495
  • SchwartzGKIlsonDSaltzLPhase II study of the cyclin-dependent kinase inhibitor flavopiridol administered to patients with advanced gastric carcinomaJ Clin Oncol20011971985199211283131
  • AkliluMKindlerHLDonehowerRCManiSVokesEEPhase II study of flavopiridol in patients with advanced colorectal cancerAnn Oncol20031481270127312881391
  • LiuGGandaraDRLaraPNA Phase II trial of flavopiridol (NSC #649890) in patients with previously untreated metastatic androgen-independent prostate cancerClin Cancer Res200410392492814871968
  • CanaveseMSantoLRajeNCyclin dependent kinases in cancer: potential for therapeutic interventionCancer Biol Ther201213745145722361734
  • ToogoodPLHarveyPJRepineJTDiscovery of a potent and selective inhibitor of cyclin-dependent kinase 4/6J Med Chem20054872388240615801831
  • FryDWHarveyPJKellerPRSpecific inhibition of cyclin-dependent kinase 4/6 by PD 0332991 and associated antitumor activity in human tumor xenograftsMol Cancer Ther20043111427143815542782
  • FinnRSDeringJConklinDPD 0332991, a selective cyclin D kinase 4/6 inhibitor, preferentially inhibits proliferation of luminal estrogen receptor-positive human breast cancer cell lines in vitroBreast Cancer Res2009115R7719874578
  • MillerTWBalkoJMFoxEMERα-dependent E2F transcription can mediate resistance to estrogen deprivation in human breast cancerCancer Discov20111433835122049316
  • ClarkeRLeonessaFWelchJNSkaarTCCellular and molecular pharmacology of antiestrogen action and resistancePharmacol Rev2001531257111171938
  • WattsCKBradyASarcevicBdeFazioAMusgroveEASutherlandRLAntiestrogen inhibition of cell cycle progression in breast cancer cells in associated with inhibition of cyclin-dependent kinase activity and decreased retinoblastoma protein phosphorylationMol Endocrinol1995912180418138614416
  • WattsCKSweeneyKJWarltersAMusgroveEASutherlandRLAntiestrogen regulation of cell cycle progression and cyclin D1 gene expression in MCF-7 human breast cancer cellsBreast Cancer Res Treat1994311951057981461
  • CarrollJSPrallOWMusgroveEASutherlandRLA pure estrogen antagonist inhibits cyclin E-Cdk2 activity in MCF-7 breast cancer cells and induces accumulation of p130-E2F4 complexes characteristic of quiescenceJ Biol Chem200027549382213822910991938
  • WilckenNRPrallOWMusgroveEASutherlandRLInducible overexpression of cyclin D1 in breast cancer cells reverses the growth-inhibitory effects of antiestrogensClin Cancer Res1997368498549815758
  • RudasMLehnertMHuynhAAustrian Breast and Colorectal Cancer Study GroupCyclin D1 expression in breast cancer patients receiving adjuvant tamoxifen-based therapyClin Cancer Res20081461767177418347178
  • BoscoEEWangYXuHThe retinoblastoma tumor suppressor modifies the therapeutic response of breast cancerJ Clin Invest2007117121822817160137
  • ThangavelCDeanJLErtelATherapeutically activating RB: reestablishing cell cycle control in endocrine therapy-resistant breast cancerEndocr Relat Cancer201118333334521367843
  • KimSLooAChopraRAbstract PR02: LEE011: an orally bioavailable, selective small molecule inhibitor of CDK4/6 – reactivating Rb in cancerMol Cancer Ther201312PR02
  • SchwartzGKLoRussoPMDicksonMAPhase I study of PD 0332991, a cyclin-dependent kinase inhibitor, administered in 3-week cycles (Schedule 2/1)Br J Cancer2011104121862186821610706
  • FlahertyKTLorussoPMDemicheleAPhase I, dose-escalation trial of the oral cyclin-dependent kinase 4/6 inhibitor PD 0332991, administered using a 21-day schedule in patients with advanced cancerClin Cancer Res201218256857622090362
  • FinnRSCrownJPBoerKResults of a randomized phase 2 study of PD 0332991, a cyclin-dependent kinase (CDK) 4/6 inhibitor, in combination with letrozole vs letrozole alone for first-line treatment of ER+/HER2− advanced breast cancer(BC)2012 Cancer Res201272Suppl 24S1S6
  • InfanteJRPetronellaWGerecitanoJA phase I study of the single-agent CDK4/6 inhibitor LEE011 in pts with advanced solid tumors and lymphomasJ Clin Oncol2014325s Suppl Abstract 2528
  • ShapiroGLeeSTolcherAWA first-in-human phase I study of the CDK4/6 inhibitor, LY2835219, for patients with advanced cancerJ Clin Oncol201331Suppl Abstract 2500
  • McClendonAKDeanJLRivadeneiraDBCDK4/6 inhibition antagonizes the cytotoxic response to anthracycline therapyCell Cycle201211142747275522751436
  • DeanJLMcClendonAKKnudsenESModification of the DNA damage response by therapeutic CDK4/6 inhibitionJ Biol Chem201228734290752908722733811
  • RobertsPJBisiJEStrumJCMultiple roles of cyclin-dependent kinase 4/6 inhibitors in cancer therapyJ Natl Cancer Inst2012104647648722302033
  • VivancoISawyersCLThe phosphatidylinositol 3-Kinase AKT pathway in human cancerNat Rev Cancer20022748950112094235
  • KatsoROkkenhaugKAhmadiKWhiteSTimmsJWaterfieldMDCellular function of phosphoinositide 3-kinases: implications for development, homeostasis, and cancerAnnu Rev Cell Dev Biol20011761567511687500
  • OsakiMOshimuraMItoHPI3K-Akt pathway: its functions and alterations in human cancerApoptosis20049666767615505410
  • MillerTWRexerBNGarrettJTArteagaCLMutations in the phosphatidylinositol 3-kinase pathway: role in tumor progression and therapeutic implications in breast cancerBreast Cancer Res201113622422114931
  • BurrisHA3rdOvercoming acquired resistance to anticancer therapy: focus on the PI3K/AKT/mTOR pathwayCancer Chemother Pharmacol201371482984223377372
  • MillerTWBalkoJMArteagaCLPhosphatidylinositol 3-kinase and antiestrogen resistance in breast cancerJ Clin Oncol201129334452446122010023
  • RazisEBobosMKotoulaVEvaluation of the association of PIK3CA mutations and PTEN loss with efficacy of trastuzumab therapy in metastatic breast cancerBreast Cancer Res Treat2011128244745621594665
  • ShomanNKlassenSMcFaddenABickisMGTorlakovicEChibbarRReduced PTEN expression predicts relapse in patients with breast carcinoma treated by tamoxifenMod Pathol200518225025915475931
  • SaalLHJohanssonPHolmKPoor prognosis in carcinoma is associated with a gene expression signature of aberrant PTEN tumor suppressor pathway activityProc Natl Acad Sci U S A2007104187564756917452630
  • Pérez-TenorioGAlkhoriLOlssonBPIK3CA mutations and PTEN loss correlate with similar prognostic factors and are not mutually exclusive in breast cancerClin Cancer Res200713123577358417575221
  • Gonzalez-AnguloAmFerrer-LozanoJStemke-HaleKPI3K pathway mutations and PTEN levels in primary and metastatic breast cancerMol Cancer Ther20111061093110121490305
  • BendellJCRodonJBurrisHAPhase I, dose-escalation study of BKM120, an oral pan-Class I PI3K inhibitor, in patients with advanced solid tumorsJ Clin Oncol201230328229022162589
  • DoiTAndoYBandoHPhase I dose-escalation study of BKM120, an oral pan-class I PI3K inhibitor, in Japanese patients with advanced solid tumorsMol Cancer Ther20111011 Supplement 1 Abstract B159
  • RodonJBendellJCAbdulRARSafety profile and clinical activity of single-agent BKM120, a pan-class I PIK inhibitor, for the treatment of patients with metastatic breast carcinomaCancer Res20117124 Suppl 3 Abstract P3-16-01
  • Gonzalez-AnguloAMJuricDArgilésGSafety, pharmacokinetics, and preliminary activity of the α-specific PI3K inhibitor BYL719: results from the first-in-human studyJ Clin Oncol201331Suppl Abstract 2531
  • MillerTWHennessyBTGonzález-AnguloAMHyperactivation of phosphatidylinositol-3 kinase promotes escape from hormone dependence in estrogen receptor-positive human breast cancerJ Clin Invest201012072406241320530877
  • O’BrienCWallinJJSampathDPredictive biomarkers of sensitivity to the phosphatidylinositol 3′ kinase inhibitor GDC-0941 in breast cancer preclinical modelsClin Cancer Res201016143670368320453058
  • SheQBChandarlapatySYeQBreast tumor cells with PI3K mutation or HER2 amplification are selectively addicted to Akt signalingPLoS One200838e306518725974
  • IhleNTLemosRJrWipfPMutations in the phosphatidylinositol-3-kinase pathway predict for antitumor activity of the inhibitor PX-866 whereas oncogenic Ras is a dominant predictor for resistanceCancer Res200969114315019117997
  • WallinJJEdgarKAGuanJGDC-0980 is a novel class I PI3K/mTOR kinase inhibitor with robust activity in cancer models driven by the PI3K pathwayMol Cancer Ther201110122426243621998291
  • MayerIABalkoJMKubaMGSU2C phase Ib study of pan-PI3K inhibitor BKM120 plus aromatase inhibitor letrozole in ER+/HER2− metastatic breast cancer (MBC)Cancer Res20117124 Suppl 3 Abstract PD09-05
  • JankuFWhelerJJWestinSNPI3K/AKT/mTOR inhibitors in patients with breast and gynecologic malignancies harboring PIK3CA mutationsJ Clin Oncol201230877778222271473
  • MaassNHarbeckNMundhenkeCGerman Breast GroupEverolimus as treatment for breast cancer patients with bone metastases only: results of the phase II RADAR studyJ Cancer Res Clin Oncol2013139122047205624072232
  • CavazzoniABonelliMAFumarolaCOvercoming acquired resistance to letrozole by targeting the PI3K/AKT/mTOR pathway in breast cancer cell clonesCancer Lett20123231778722484466
  • GhayadSECohenPAInhibitors of the PI3K/Akt/mTOR pathway: new hope for breast cancer patientsRecent Pat Anticancer Drug Discov201051295719751211
  • GhayadSEBiecheIVendrellJAmTOR inhibition reverses acquired endocrine therapy resistance of breast cancer cells at the cell proliferation and gene-expression levelsCancer Sci200899101992200319016759
  • VilquinPVilledieuMGrisardEMolecular characterization of anastrozole resistance in breast cancer: pivotal role of the Akt/mTOR pathway in the emergence of de novo or acquired resistance and importance of combining the allosteric Akt inhibitor MK-2206 with an aromatase inhibitorInt J Cancer201313371589160223553037
  • 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
  • YardleyDANoguchiSPritchardKIEverolimus plus exemestane in postmenopausal patients with HR(+) breast cancer: BOLERO-2 final progression-free survival analysisAdv Ther2013301087088424158787
  • HortobagyiGNPiccart-GebhartMJRugoHSCorrelation of molecular alterations with efficacy of everolimus in hormone receptor-positive, HER2-negative advanced breast cancer: results from BOLERO-2J Clin Oncol201331Suppl Abstract LBA509
  • MillerTWForbesJTShahCInhibition of mammalian target of rapamycin is required for optimal antitumor effect of HER2 inhibitors against HER2-overexpressing cancer cellsClin Cancer Res200915237266727619934303
  • O’ReillyKERojoFSheQBmTOR inhibition induces upstream receptor tyrosine kinase signaling and activates AktCancer Res20066631500150816452206
  • CarracedoAMaLTeruya-FeldsteinJInhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancerJ Clin Invest200811893065307418725988
  • MayerIAAbramsonVGBalkoJMSU2C phase Ib study of pan-PI3K inhibitor BKM120 with letrozole in ER+/HER2− metastatic breast cancer (MBC)J Clin Oncol201230Suppl Abstract 510
  • PerezEADueckACMcCulloughAEImpact of PTEN protein expression on benefit from adjuvant trastuzumab in early-stage human epidermal growth factor receptor 2-positive breast cancer in the North Central Cancer Treatment Group N9831 trialJ Clin Oncol201331172115212223650412
  • BernsKHorlingsHMHennessyBTA functional genetic approach identifies the PI3K pathway as a major determinant of trastuzumab resistance in breast cancerCancer Cell200712439540217936563
  • O’BrienNABrowneBCChowLActivated phosphoinositide 3-kinase/AKT signaling confers resistance to trastuzumab but not lapatinibMol Cancer Ther2010961489150220501798
  • WangLZhangQZhangJPI3K pathway activation results in low efficacy of both trastuzumab and lapatinibBMC Cancer20111124821676217
  • EstevaFJGuoHZhangSPTEN, PIK3CA, p-AKT, and p-p70S6K status: association with trastuzumab response and survival in patients with HER2-positive metastatic breast cancerAm J Pathol201017741647165620813970
  • O’BrienNAMcDonaldKTongLPI3K/mTOR inhibition overcomes in vitro and in vivo trastuzumab resistance independent of feedback activation of pAKTCancer Res20127224 Suppl 3 Abstract P4-08-01
  • SauraCBendellJJerusalemGPhase Ib study of buparlisib plus trastuzumab in patients with HER2-positive advanced or metastatic breast cancer that has progressed on trastuzumab-based therapyClin Cancer Res20142071935194524470511
  • 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
  • WestKACastilloSSDennisPAActivation of the PI3K/Akt pathway and chemotherapeutic resistanceDrug Resist Updat20025623424812531180
  • MassarwehSOsborneCKCreightonCJTamoxifen resistance in breast tumors is driven by growth factor receptor signaling with repression of classic estrogen receptor genomic functionCancer Res200868382683318245484
  • LuYZiXZhaoYMascarenhasDPollakMInsulin-like growth factor-I receptor signaling and resistance to trastuzumab (Herceptin)J Natl Cancer Inst200193241852185711752009
  • VadgamaJVWuYDattaGKhanHChillarRPlasma insulin-like growth factor-I and serum IGF-binding protein 3 can be associated with the progression of breast cancer, and predict the risk of recurrence and the probability of survival in African-American and Hispanic womenOncology199957433034010575321
  • CreightonCJCasaALazardZInsulin-like growth factor-I activates gene transcription programs strongly associated with poor breast cancer prognosisJ Clin Oncol200826254078408518757322
  • BonneterreJPeyratJPBeuscartRDemailleAPrognostic significance of insulin-like growth factor 1 receptors in human breast cancerCancer Res19905021693169352170011
  • FoxEMMillerTWBalkoJMA kinome-wide screen identifies the insulin/IGF-I receptor pathway as a mechanism of escape from hormone dependence in breast cancerCancer Res201171216773678421908557
  • QuekRWangQMorganJACombination mTOR and IGF-1R inhibition: phase I trial of everolimus and figitumumab in patients with advanced sarcomas and other solid tumorsClin Cancer Res201117487187921177764
  • CaoAZPinzon-OrtizMChenYTargeting PIK3CA mutant breast cancer with the combination of PIK3CA-specific inhibitor, BYL719, and IGF1-R antibody, ganitumabJ Clin Oncol201331Suppl Abstract e13525
  • FaganDHYeeDCrosstalk between IGF1R and estrogen receptor signaling in breast cancerJ Mammary Gland Biol Neoplasia200813442342919003523
  • CohenBDBakerDASoderstromCCombination therapy enhances the inhibition of tumor growth with the fully human anti-type 1 insulin-like growth factor receptor monoclonal antibody CP-751,871Clin Cancer Res20051152063207315756033
  • RobertsonJFFerreroJMBourgeoisHGanitumab with either exemestane or fulvestrant for postmenopausal women with advanced, hormone-receptor-positive breast cancer: a randomised, controlled, double-blind, phase 2 trialLancet Oncol201314322823523414585
  • DunnSEHardmanRAKariFWBarrettJCInsulin-like growth factor 1 (IGF-1) alters drug sensitivity of HBL100 human breast cancer cells by inhibition of apoptosis induced by diverse anticancer drugsCancer Res19975713268726939205078
  • GoochJLVan Den BergCLYeeDInsulin-like growth factor (IGF)-I rescues breast cancer cells from chemotherapy-induced cell death – proliferative and anti-apoptotic effectsBreast Cancer Res Treat199956111010517338
  • HaluskaPShawHBatzelGNPhase I dose escalation study of the anti-IGF-1R monoclonal antibody CP-751,871 in patients with refractory solid tumorsJ Clin Oncol200725Suppl 18 Abstract 3586
  • KarpDDPaz-AresLGNovelloSPhase II study of the anti-insulin-like growth factor type 1 receptor antibody CP-751,871 in combination with paclitaxel and carboplatin in previously untreated, locally advanced, or metastatic non-small-cell lung cancerJ Clin Oncol200927152516252219380445
  • NahtaRYuanLXZhangBKobayashiREstevaFJInsulin-like growth factor-I receptor/human epidermal growth factor receptor 2 heterodimerization contributes to trastuzumab resistance of breast cancer cellsCancer Res20056523111181112816322262
  • FriedbichlerKHofmannMHKroezMPharmacodynamic and antineoplastic activity of BI 836845, a fully human IGF ligand-neutralizing antibody, and mechanistic rationale for combination with rapamycinMol Cancer Ther201413239940924296829
  • ChenYCSosnoskiDMMastroAMBreast cancer metastasis to the bone: mechanisms of bone lossBreast Cancer Res201012621521176175
  • HershmanDLMcMahonDJCrewKDZoledronic acid prevents bone loss in premenopausal women undergoing adjuvant chemotherapy for early-stage breast cancerJ Clin Oncol200826294739474518711172
  • GnantMMlineritschBLuschin-EbengreuthGAustrian Breast and Colorectal Cancer Study Group (ABCSG)Adjuvant endocrine therapy plus zoledronic acid in premenopausal women with early-stage breast cancer: 5-year follow-up of the ABCSG-12 bone-mineral density substudyLancet Oncol20089984084918718815
  • ShapiroCLHalabiSHarsVZoledronic acid preserves bone mineral density in premenopausal women who develop ovarian failure due to adjuvant chemotherapy: final results from CALGB trial 79809Eur J Cancer201147568368921324674
  • GreenJRBisphosphonates: preclinical reviewOncologist20049Suppl 431315459425
  • WinterMCHolenIColemanREExploring the anti-tumour activity of bisphosphonates in early breast cancerCancer Treat Rev200834545347518423992
  • DaubinéFLe GallCGasserJGreenJClézardinPAntitumor effects of clinical dosing regimens of bisphosphonates in experimental breast cancer bone metastasisJ Natl Cancer Inst200799432233017312309
  • OttewellPDLefleyDVCrossSSEvansCAColemanREHolenISustained inhibition of tumor growth and prolonged survival following sequential administration of doxorubicin and zoledronic acid in a breast cancer modelInt J Cancer2010126252253219621384
  • HiragaTWilliamsPJUedaATamuraDYonedaTZoledronic acid inhibits visceral metastases in the 4T1/luc mouse breast cancer modelClin Cancer Res200410134559456715240548
  • GnantMZoledronic acid in the treatment of early-stage breast cancer: is there a final verdict?Curr Oncol Rep2012141354322113793
  • DentRTrudeauMPritchardKITriple-negative breast cancer: clinical features and patterns of recurrenceClin Cancer Res20071315 Pt 14429443417671126
  • KenneckeHYerushalmiRWoodsRMetastatic behavior of breast cancer subtypesJ Clin Oncol201028203271327720498394
  • RoosWPKainaBDNA damage-induced cell death by apoptosisTrends Mol Med200612944045016899408
  • McCabeNTurnerNCLordCJDeficiency in the repair of DNA damage by homologous recombination and sensitivity to poly(ADP-ribose) polymerase inhibitionCancer Res200666168109811516912188
  • BryantHESchultzNThomasHDSpecific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymeraseNature2005434703591391715829966
  • FarmerHMcCabeNLordCJTargeting the DNA repair defect in BRCA mutant cells as a therapeutic strategyNature2005434703591792115829967
  • MuraiJHuangSYDasBBTrapping of PARP1 and PARP2 by clinical PARP inhibitorsCancer Res201272215588559923118055
  • MuraiJHuangSYRenaudAStereospecific PARP trapping by BMN 673 and comparison with olaparib and rucaparibMol Cancer Ther201413243344324356813
  • TuttARobsonMGarberJEOral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and advanced breast cancer: a proof-of-concept trialLancet2010376973723524420609467
  • TurnerNTuttAAshworthAHallmarks of ‘BRCAness’ in sporadic cancersNat Rev Cancer200441081481915510162
  • DentRALindemanGJClemonsMPhase I trial of the oral PARP inhibitor olaparib in combination with paclitaxel for first- or second-line treatment of patients with metastatic triple-negative breast cancerBreast Cancer Res2013155R8824063698
  • KristeleitRSShapiroGLoRussoPA phase I dose-escalation and PK study of continuous oral rucaparib in patients with advanced solid tumorsJ Clin Oncol201331Suppl Abstract 2585
  • IsakoffSJOvermoyerBTungNMA phase II trial expansion cohort of the PARP inhibitor veliparib (ABT888) and temozolomide in BRCA1/2 associated metastatic breast cancerCancer Res20117124 Suppl 3P3-16-05
  • FanJYinWJLuJSER alpha negative breast cancer cells restore response to endocrine therapy by combination treatment with both HDAC inhibitor and DNMT inhibitorJ Cancer Res Clin Oncol2008134888389018264725
  • JangERLimSJLeeESThe histone deacetylase inhibitor trichostatin A sensitizes estrogen receptor alpha-negative breast cancer cells to tamoxifenOncogene20042391724173614676837
  • ThomasSThurnKTBiçakuEMarchionDCMünsterPNAddition of a histone deacetylase inhibitor redirects tamoxifen-treated breast cancer cells into apoptosis, which is opposed by the induction of autophagyBreast Cancer Res Treat2011130243744721298336
  • YardleyDAIsmail-KhanRRMelicharBRandomized phase II, double-blind, placebo-controlled study of exemestane with or without entinostat in postmenopausal women with locally recurrent or metastatic estrogen receptor-positive breast cancer progressing on treatment with a nonsteroidal aromatase inhibitorJ Clin Oncol201331172128213523650416
  • MunsterPNThurnKTThomasSA phase II study of the histone deacetylase inhibitor vorinostat combined with tamoxifen for the treatment of patients with hormone therapy-resistant breast cancerBr J Cancer2011104121828183521559012
  • ContePCamponeMPronzatoPPhase I trial of panobinostat (LBH589) in combination with trastuzumab in pretreated HER2-positive metastatic breast cancer (mBC): preliminary safety and tolerability resultsJ Clin Oncol20092715s Suppl Abstract 1081
  • FrameMCSrc in cancer: deregulation and consequences for cell behaviorBiochim Biophys Acta20021602211413012020799
  • ZhangXHWangQGeraldWLatent bone metastasis in breast cancer tied to Src-dependent survival signalsCancer Cell2009161677819573813
  • ElsbergerBTanBAMitchellTJIs expression or activation of Src kinase associated with cancer-specific survival in ER-, PR- and HER2-negative breast cancer patients?Am J Pathol200917541389139719762712
  • ElsbergerBTanBAMallonEABruntonVGEdwardsJIs there an association with phosphorylation and dephosphorylation of Src kinase at tyrosine 530 and breast cancer patient disease-specific survivalBr J Cancer2010103121831183421063412
  • MorganLGeeJPumfordSElevated Src kinase activity attenuates Tamoxifen response in vitro and is associated with poor prognosis clinicallyCancer Biol Ther20098161550155819830888
  • HeroldCIChadaramVPetersonBLPhase II trial of dasatinib in patients with metastatic breast cancer using real-time pharmacodynamic tissue biomarkers of Src inhibition to escalate dosingClin Cancer Res201117186061607021810917
  • FinnRSBengalaCIbrahimNDasatinib as a single agent in triple-negative breast cancer: results of an open-label phase 2 studyClin Cancer Res201117216905691322028489
  • FedelePCalvaniNMarinoATargeted agents to reverse resistance to endocrine therapy in metastatic breast cancer: where are we now and where are we going?Crit Rev Oncol Hematol201284224325122494933
  • FornierMNMorrisPGAbbruzziAA phase I study of dasatinib and weekly paclitaxel for metastatic breast cancerAnn Oncol201122122575258121406471
  • SomloGAtzoriFStraussLCDasatinib plus capecitabine for advanced breast cancer: safety and efficacy in phase I study CA180004Clin Cancer Res20131971884189323403636
  • PaulDVukeljaSJHolmesFALetrozole plus dasatinib improves progression-free survival (PFS) in hormone receptor-positive, HER2-negative postmenopausal metastatic breast cancer (MBC) patients receiving first-line aromatase inhibitor (AI) therapyCancer Res201373 Abstract S3-07
  • Bristol-Myers SquibbSafety and efficacy of exemestane plus dasatinib versus placebo for advanced ER+ breast cancer Available from: http://clinicaltrials.gov/show/NCT00767520. NLM identifier: NCT00767520Accessed March 14, 2014
  • ParganasEWangDStravopodisDJak2 is essential for signaling through a variety of cytokine receptorsCell19989333853959590173
  • LevineRLPardananiATefferiAGillilandDGRole of JAK2 in the pathogenesis and therapy of myeloproliferative disordersNat Rev Cancer20077967368317721432
  • LevineRLWadleighMCoolsJActivating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosisCancer Cell20057438739715837627
  • SansonePBrombergJTargeting the interleukin-6/Jak/stat pathway in human malignanciesJ Clin Oncol20123091005101422355058
  • MarottaLLAlmendroVMarusykAThe JAK2/STAT3 signaling pathway is required for growth of CD44+CD24− stem cell-like breast cancer cells in human tumorsJ Clin Invest201112172723273521633165
  • BritschgiAAndraosRBrinkhausHJAK2/STAT5 inhibition circumvents resistance to PI3K/mTOR blockade: a rationale for cotargeting these pathways in metastatic breast cancerCancer Cell201222679681123238015
  • CeramiEGaoJDogrusozUThe cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics dataCancer Discov20122540140422588877
  • GaoJAksoyBADogrusozUIntegrative analysis of complex cancer genomics and clinical profiles using the cBioPortalSci Signal20136269pl123550210