Abstract
An increasing number of tumors, including breast cancer, overexpress proteins of the epidermal growth factor receptor (EGFR) family. The interaction between family members activates signaling pathways that promote tumor progression and resistance to treatment. Human epidermal growth factor receptor type II (HER2) positive breast cancer represents a clinical challenge for current therapy. It has motivated the development of novel and more effective therapeutic EGFR family target drugs, such as tyrosine kinase inhibitors (TKIs). This review focuses on the effects of three TKIs mostly studied in HER2- positive breast cancer, lapatinib, gefitinib and neratinib. Herein, we discuss the mechanism of action, therapeutic advantages and clinical applications of these TKIs. To date, TKIs seem to be promising therapeutic agents for the treatment of HER2-overexpressing breast tumors, either as monotherapy or combined with other pharmacological agents.
Keywords: HER2-positive breast cancer, tyrosine kinase inhibitors, lapatinib, gefitinib, neratinib, epidermal growth factor receptor family
Introduction
Breast cancer is the most common malignancy in women and is a major public health issue [1,2]. In the clinic, breast cancer is mainly classified into four molecular subtypes, luminal A/B, human epidermal growth factor receptor type II (HER2) and basal-like [3,4]. The subtype HER2 represents 25-30% of all breast cancer cases and HER2 overexpression is strongly associated with aggressive phenotype and poor outcomes [5,6]. The current and approved therapy for this type of cancer is trastuzumab, a humanized monoclonal antibody that binds to the extracellular domain of HER2 [7,8]. However, de novo or acquired resistance to therapy occurs in some patients [9]. Consequently, new targeted therapies are in development, such as tyrosine kinase inhibitors (TKIs) [10]. This paper aims to integrate knowledge of the signaling pathways associated to the major TKIs proved on HER2-positive breast cancer, lapatinib, geftinib and neratinib. Moreover, we discuss molecular mechanisms, resistance and clinical trials for each drug, as well as their beneficial therapeutic effects and undesirable side effects.
EGFR family
The EGFR family comprises four distinct membrane tyrosine kinase receptors; EGFR/ErbB-1, HER2/ErbB-2, HER3/ErbB-3 and HER4/ErbB-4 which are activated upon ligand binding to the extracellular domain of these receptors. Afterwards, the formation of receptor homo- or hetero-dimers is induced resulting in phosphorylation of tyrosine kinases residues and cross-phosphorylation, that triggers numerous signaling pathways such as phosphatidylinositol-3 kinase (PI3K), mitogen-activated protein kinase/extracellular signal-regulated kinases (MAPK/ERK1/2), signal transducer and activator of transcription (STAT), phospholipase C (PLCγ), and/or the modulation of calcium channels [11], This sequence of events induces cellular responses which include proliferation, differentiation and inhibition of apoptosis, giving rise to diseases such as cancer [12].
In a wide range of epithelial cells, including breast, colon, head, neck, kidney, lung, pancreas, and prostate, the overexpression and constitutive activation of the EGFR family members, particularly EGFR and HER2, may trigger cancer initiation, metastasis, and tumor progression [13-15].
In particular, HER2 is overexpressed/amplified in 20-30% of patients with metastatic breast cancer [16]. Moreover, there is a growing evidence that heterodimer formation between receptors of EGFR members resulted in adverse response to therapy [17]. In order to block EGFRs intracellular signaling pathways in breast cancer, the development of novel therapies which include the use of TKIs is currently underway.
Tyrosine kinase inhibitors
The TKIs are oral non-peptide anilinoquinazolone compounds homologous of the adenosine triphosphate (ATP). This similarity allows them to compete for the ATP-binding domain of protein kinases preventing phosphorylation and subsequent activation of the signal transduction pathways, leading to apoptosis and decreasing cellular proliferation [18]. Moreover, TKIs target other kinase receptors due to the homology that they share with the EGFR family in the catalytic domain [19] which is highly conserved across the kinome [20]. Whereby, the actions of TKIs in several kinases cause different effects in the therapeutic use [21,22].
The main characteristics, mechanisms of action, causes of resistance and clinical evidences of the major TKIs proved on HER2-positive breast cancer, lapatinib, geftinib and neratinib, are described below.
Lapatinib
Lapatinib is a reversible dual TKI that selectively targets and inhibits HER2 and EGFR with proven effectiveness in clinical trials. This inhibitor has been approved by the US Food and Drug Administration (FDA) since 2007 for metastatic HER2-positive breast cancer treatment. It is commonly used in combination with the chemotherapeutic agent capecitabine on the treatment of breast cancer patients previously treated with trastuzumab, anthracyclines and taxane [23-25]. Moreover, the compound combined with letrozole, an aromatase inhibitor, has been accepted as first-line therapy for metastatic breast cancer that coexpresses hormone receptors and HER2 [26].
Mechanism of action of lapatinib: preclinical evidence
Lapatinib inhibits proliferation of several human cancer cell lines from vulva, breast, lung, and esophagus [27-29]. In vitro studies showed that lapatinib inhibited the activation of the three main EGFR and HER2 downstream signaling pathways, MAPK, PI3K-AKT and PLCγ, through decreased phosphorylation of target receptors and the Raf, ERK, AKT, and PLCγ1 proteins. Additionally, this TKI increased p38 expression, a stress-induced member of the MAPK pathway that is involved in apoptosis, the subG1 phase of the cell cycle (a hallmark of apoptosis), and the cyclin-dependent kinase inhibitors p21 and p27 [30-32]. Lapatinib inhibited cell proliferation and migration of breast cancer cell lines expressing different levels of EGFR and HER2; however, cells overexpressing HER2 were more sensitive to this TKI [30]. Also, lapatinib increased the pro-apoptotic protein BIM at the transcriptional level and reduced protein expression of survivin, an apoptosis inhibitor protein, which is express in 90% of all breast cancer cases and is cause of poor outcome for this pathology [33-35]. Although lapatinib is a dual TKI that targets both HER2 and EGFR, it has been demonstrated that it also inhibited phosphorylation of HER3 [36]. A resume of lapatinib mechanisms is found in Figure 1.
Figure 1.

Schematic representation of the action of three TKIs and their interaction with receptors of the EGFR family. As TKIs are homologous to ATP, they compete for the ATP-binding domain of protein kinases preventing their phosphorylation and subsequent activation of the signal transduction pathways, leading to apoptosis, decreased cellular proliferation and eventually cell cycle arrest. Inhibition of phosphorylation of the receptors by TKIs (X); disrupted heterodimer formation by gefitinib, avoid the interaction between receptors (Φ); upregulated (↑); downregulated (↓).
There is a high incidence of brain metastases in patients with HER2-overexpressing breast cancer even if they were treated with trastuzumab [37,38]. Interestingly, in a preclinical mouse model, lapatinib could prevent the metastatic outgrowth of HER2-overexpressing breast cancer cells in the brain. In this in vivo metastasis model, lapatinib reduced the phosphorylation of HER2 but it did not affect EGFR, contrary to in vitro studies [30]. Moreover, EGFR small-interfering RNA (siRNA) knockdown in HER2-positive breast cancer cells did not affect the antiproliferative activity of lapatinib, whereas depletion of HER2 causes lapatinib resistance, indicating that lapatinib effects are mediated mainly through HER2 pathway [32]. The stated above suggests a direct correlation between lapatinib sensitivity and HER2 expression only.
A subgroup of HER2-overexpressing tumors also express p95HER2, an amino terminally truncated receptor, that has kinase activity but lacks the epitope recognized by trastuzumab; hence, expression of this form confers resistance to trastuzumab [39]. In addition, p95HER2 has been considered as a biomarker of an aggressive subtype of HER2-positive breast cancer [40]. Lapatinib inhibited p95HER2, AKT, MAPK phosphorylation and the growth of cells that express the truncate receptors. Moreover, lapatinib showed antitumor activity in p95HER2 tumor xenografts [41].
Other study demonstrated that lapatinib inhibited insulin-like growth factor I (IGF-I) signaling in both trastuzumab -sensitive and -resistant HER2 overexpressing cells [42]. Cross-talk between the IGF-I receptor and HER2 in trastuzumab-resistant cells increased HER2 receptor phosphorylation [43]. Significantly, lapatinib blocked HER2 and IGF-1R crosstalk [42]. In addition, this compound also increased fragmentation of poly ADP-ribose polymerase (PARP), a protein involved in programmed cell death, and downregulated survivin expression in trastuzumab sensitive and resistant HER2 overexpressing cells [42].
In addition, lapatinib inhibited activation of nuclear factor κB (NF-κB) in HER2-overexpressing breast cancer cells [44]. The TKI inactivates NF-κB through reducing phosphorylation of its inhibitor IkB-α via blocking the PI3K/AKT cascade [44]. This fact is relevant due to co-operation between HER2 and NF-κB signaling which causes tumor resistance to radiotherapy [45].
Overexpression of EGFR and HER2 contributes to clinical radiation resistance [46] and several EGFR inhibitors sensitize tumor cells to ionizing radiation [46-48]. In this regard, lapatinib treatment enhanced the radiosensitization of EGFR- and HER2-overexpressing breast cancer cells through inhibition of MEK/ERK signaling pathway [49,50].
In the SK-BR-3 HER2-amplified breast cancer cell line prolonged exposure to lapatinib reduced the expression and activity of the enzyme topoisomerase IIα, which renders cells resistant to the cytotoxic effects of doxorubicin, etoposide, and m-AMSA [51].
Lapatinib regulates several microRNAs (miR) that play an important role in the anti-tumor action in the HER2-postive breast carcinoma cells [52]. In this regard, lapatinib treatment upregulated miR575 and miR-1225-5 expression, contributing in this manner to downregulation of the oncogenic protein phospholipase C PLCXD1 (phosphatidylinositolspecific phospholipase-C-X-domain-containing-1), a target transcript of miR-575 and miR1225-5p [52].
Lapatinib: clinical evidence and side effects
Several clinical studies in HER2 breast cancer have evaluated safety, dosing schedules and efficacy of lapatinib as monotherapy or in combination with other therapies. The outcomes depend on population features, tumor type, stage and prior cancer treatments.
In all the studies listed in Table 1, lapatinib and its combinations provided clinical benefit in different grades in HER2 breast cancer patients. In contrast, HER2 negative and EGFR positive cancer patients did not experience benefit with this TKI [53-55].
Table 1.
Clinical evidence with lapatinib in HER2-positive breast cancer
| Therapy [Ref.] | Study type | Patient population (n) | Principal findings |
|---|---|---|---|
| Lapatinib [53] | Phase II, open-label, two-stage, two-cohorts multicenter study. | Advanced or metastatic IBC, HER2 and/or EGFR positive, refractory or recurrent after treatment with an anthracycline. | Lapatinib (1500 mg/day) was well tolerated in both cohorts. It showed clinical activity (CR=7%, PR=43%, OR=16.9%, PFS=14 weeks) in heavily pretreated HER2-positive but not in EGFR-positive/HER2-negative patients (PR=6.6%, PFS=4 weeks). The most common AEs in both cohorts included grade 1/2 diarrhea, musculoskeletal pain, and rash. Serious AEs included musculoskeletal pain, dyspnea, and diarrhea. PTEN status does not preclude response to lapatinib. Coexpression of phospho HER2 and phospho HER3 in tumors seems to predict for a favorable response to lapatinib. |
| Cohort A: HER2-positive (30). | |||
| Cohort B: EGFR-positive and HER2 negative (15). | |||
| Lapatinib [163] | Phase II, open-label, single-group, multicenter study. | Advanced or MBC with HER2 overexpression, with disease progression during TZ therapy (78). | Lapatinib demonstrated clinical activity (CBR=11.5%, TTP and PFS=15.3 weeks). Doses of 1250 and 1500 mg/day were well tolerated. AEs were rash, diarrhea, nausea, and fatigue with no grade 4 events. |
| Lapatinib [54] | Phase II, open-label, single-arm, two-cohorts multicenter study. | HER2-positive (cohort A=140) and negative (cohort B=89) advanced MBC prior anthracycline and/or taxane treatment. | Lapatinib dose of 1500 mg/day provided modest clinical benefit (CBR=5.7%) only in HER2-positive patients. PFS after 4 months (34 vs 18%) and OS (29.4 vs 18.6 weeks) were higher in HER2-positive than in HER2-negative patients. The most common AEs were diarrhea, nausea and rash most grade 1/2 with maximum severity of grade 3 in both cohorts. Serious AEs were diarrhea, dehydration, nausea, and vomiting. There were 4 fatal AEs. |
| Lapatinib [164] | Phase II, open-label, multicenter. Study was originally designed with two cohorts but cohort B was closed. | Relapsed or refractory invasive, IBC with HER2 overexpression previously treated with anthracycline and taxane plus TZ. (Cohort A=126). | Lapatinib dose of 1500 mg/day was considered potentially clinically effective (ORR=40-15% depending on criteria, PFS=14.6 weeks, OS=8.4 months) in heavily pretreated patients. Likelihood of response to lapatinib was not affected by previous treatment with TZ. The most common serious AEs were dyspnea and pleural effusion. Fatal AEs were possibly treatment related. |
| Lapatinib [55] | Phase II, open-label, two-cohorts multicenter studies. | Japanese patients with advanced or MBC with HER2 positive (cohort A=122) and negative (cohort B=22), with disease progression during previously therapy with anthracycline and taxane plus TZ. | Lapatinib dose of 1500 /day was well tolerated in both cohorts and effective only in HER2-positive patients. CBR (25 vs 4.5%) and OS (58.3 vs 40 weeks) were higher in cohort A than in cohort B. The most common AE was diarrhea grade 1/2. Patients with tumours harbouring an H1047R PIK3CA mutation or low expression of PTEN derived clinical benefit from lapatinib. |
| Lapatinib [165] | Phase II, randomized open-label, parallel-group, multicenter study. | HER2-positive advanced or MBC. Could be previously treated. (69/69). | Lapatinib doses of 1500 mg once daily or 500 mg twice daily were safe and effective without significant differences in clinical activity or the AEs profile between them (ORR=22 and 26%, CBR=29 and 33%, PFS at 4 months=60 and 67%). The most common AEs were diarrhea, rash, pruritus, and nausea, grade 1/2. |
| Lapatinib [56] | Phase II | HER2-positive breast cancer with brain metastases prior TZ treatment. (39). | Lapatinib doses of 750 mg twice daily provided clinical benefit measured as absence of progression in brain metastases (ORR=2.6%, SD≥16 weeks=15.4%). The most common AEs were diarrhea and fatigue grade 2/3. |
| Lapatinib [166] | Phase I single institution study. | HER2-overexpressing breast cancer patients without any restrictions on prior therapies including TZ or lapatinib. (34). | This study used a 3+3 dose escalation design with a starting dose cohort of 1750 mg twice daily, ending with 7000 mg twice daily. The protocol had an amendment consisted in the use of exposure enhancement strategies such as take the medication with food, inhibition of CYP3A4 with ketoconazole and dose fractionation (four times a day) using a dose of 3000 mg. The majority of AEs were grade 1/2 and diarrhea was the most common. Lapatinib dose was escalated to 7000 mg per day with no dose limiting toxicity; however, plasma lapatinib concentrations plateaued in this dose range. 6 patients achieved a response and dramatic responses were seen in 3 patients with lapatinib concentrations approaching 10000 ng/mL. |
| Lapatinib [167] | Phase III, randomized double blind, placebo-controlled multicenter trial. | Patients in early stages of HER2-positive invasive breast prior adjuvant chemotherapy but not TZ. (1571/1576). | Lapatinib (1500 mg) and placebo were administrated daily for 12 months. A review of HER2 status showed that only 79% of the women were HER2-positive. In this group, with a median follow-up of 47.4 months in the lapatinib subgroup and 48.3 in the placebo group disease-free survival events occurred in 13% in the lapatinib group and 17% in the placebo group. OS=6 and 7% in lapatinib and placebo groups respectively. 6% serious AEs occurred in patients taking lapatinib and 5% in patients taking placebo with higher incidences of grade 3/4 diarrhea (6 vs 1%), rash (5 vs <1%), and hepatobiliary disorders (2 vs <1%). |
| Lapatinib+letrozole [26] | Phase III, randomized double-blind, controlled, parallel-group, multicenter study. | Postmenopausal women with hormone receptor-positive, HER2-positive MBC. (642/644). | Combination therapy (lapatinib 1500 mg/day plus letrozole 2.5 mg/day) was superior to endocrine therapy alone (letrozole 2.5 mg/day). Respectively: PFS=8.2 vs 3.0 months, CBR=48 vs 29%. Diarrhea and rash grade 3/4 were more common in the combination arm versus monotherapy arm. 3 fatal AEs events were possibly treatment related, 1 in combination therapy, 2 in monotherapy. |
| Lapatinib+vinorelbine [168] | Phase I dose-escalation multicenter study. | HER2-positive, advanced or MBC, prior TZ treatment. (30). | The maximal tolerated dose recommended for combination was 1000 mg/day of lapatinib and 22.5 mg/m2 of vinorelbine. AEs grade 1/2 were diarrhea, rash and liver enzymes elevation. AEs grade 3/4 were neutropenia, anemia, diarrhea and asthenia. |
| Lapatinib+capecitabine [23] | Phase III randomized two-arms, open label study. | HER2-positive locally advanced or MBC progressed after treatment with, but were not limited to, anthracycline, taxane, and TZ. (163/161). | Combination therapy (lapatinib 1250 mg/day plus capecitabine 2000 mg/m2/day) was superior to capecitabine monotherapy (2500 mg/m2/day). Respectively: TTP=8.4 vs 4.4 months, PFS=8.4 vs 4.1 months, ORR=22 vs 14%, CBR=44-29%. This combination was not associated with an increase in serious toxic effects. The most common AEs were diarrhea, PPE, nausea, vomiting, fatigue and rash. Grade 4 diarrhea occurred in combination therapy arm. One death related to drug toxicity in monotherapy arm. |
| Lapatinib+capecitabine [169,170] | Phase III randomized two-arms, open label study. (Updates from Geyer, 2006). | HER2-positive locally advanced or MBC progressed after treatment with, but were not limited to, anthracycline, taxane, and TZ. (198/201). | Combination therapy (lapatinib 1250 mg/day plus capecitabine 2000 mg/m2/day) showed to be superior versus capecitabine monotherapy (2500 mg/m2/day). Respectively: OS=75.0 vs 64.7%, TTP=27.1 vs 18.6 months, CBR=29.3 vs 17.4%. The most common AE was diarrhea in both arms. |
| Lapatinib+capecitabine [171] | Phase III, randomized two-arms, open label study. (Update from Geyer 2006). | HER2-positive locally advanced or MBC progressed after treatment with, but were not limited to, anthracycline, taxane, and TZ. (198/201). | Values favored combination therapy vs monotherapy. Respectively: ORR=24 vs 14% and TTP=33.3 vs 22.9 months. |
| Lapatinib+capecitabine [172] | Phase II non-randomized open-label, single-center study. | HER2-positive MBC progressed with TZ. (22). | This combination regimen (capecitabine 2000 mg/twice day for 7 days followed by a 7-day rest and lapatinib 1250 mg/day) provided clinical benefit (CBR=50%, PFS=9.4 months), was well tolerated and reduced gastrointestinal toxicity compared with standard regimen. |
| Lapatinib+capecitabine [57] | Phase II open-label, multicenter study. | HER2-positive breast cancer with brain metastases. Previous treatment was allowed except with lapatinib or capecitabine and WBRT. (44). | Combination therapy (lapatinib 1250 mg/day plus capecitabine 2000 mg/m2/day) showed to be active in terms of objective CNS response (OR=65.9%). The most common AEs were diarrhea, PPE, nausea, fatigue, rash and bilirubin increase. AEs grade 3/4 were diarrhea, PPE and fatigue. The combination therapy could change the management of selected patients with brain metastases, allowing delay to WBRT and its AEs. |
| Lapatinib In extension phase: lapatinib+capecitabine [59] | Phase II open-label, two-cohorts, multicenter study. | HER2-positive breast cancer with brain metastases prior TZ and WBRT treatment. | Lapatinib alone (750 mg/twice day) showed a discrete CNS clinical activity (OR=6%, PFS=2.4 months). In extension phase, combination therapy (lapatinib 1250 mg/day plus capecitabine 1000 mg/m2/day) showed to have clinical activity (OR=20%, PFS=3.7 months). Volumetric reduction in CNS lesions was similar in both therapies. The most common AEs were PPE, diarrhea and nausea. AEs grade 3 were PPE, nausea and diarrhea. |
| Cohort A (95): PS=0 to 1 and 1 or 2 prior TZ regimens. | |||
| Cohort B (147): PS=2 and/or >2 prior TZ regimens (95/147). | |||
| Extension phase: patients progressed with lapatinib. (50). | |||
| Lapatinib+capecitabine [173] | Single-arm open-label trial. | HER2 locally advanced or MBC that had progressed following treatment with an anthracycline, a taxane, and TZ alone or in combination. (4283). | Combination therapy (lapatinib 1250 mg/day plus capecitabine 1000 mg/m2/twice day) showed to be safe and to offer clinical benefit to this population. Patients treated with the combination and no prior capecitabine had higher OS and PFS than patients with prior capecitabine. (41.7 and 23.9 weeks vs 36 and 18.4 weeks respectively). The most common AEs were diarrhea, vomiting, and nausea and were mainly grade 3 or higher. |
| Lapatinib+capecitabine [174] | Open-label multicenter study. | Japanese patients with HER2-positive invasive and MBC previously treated with anthracyclines, taxanes and TZ. (51). | Combination therapy (lapatinib 1250 mg/day plus capecitabine 1000 mg/m2/twice daily) was well tolerated and showed clinical activity (CBR=59%, TTP=36 weeks). The most common AEs were PPE, diarrhea and stomatitis grade 1/2. |
| Lapatinib+capecitabine [175] | Single arm open-label trial. | Chinese patients with HER2-positive invasive and MBC prior therapy with a taxane and/or an anthracycline and could have received prior TZ. (52). | Combination therapy (lapatinib, 1250 mg/day plus capecitabine 2000 mg/m2/day) showed to be well tolerated and to offer clinical benefit (CBR=57.7%, PFS=6.34 months) to heavily pretreated patients. The most common AEs were PPE, diarrhea, rash, hyperbilirubinemia, and fatigue, all grade 1/2. Grade 3/4 AEs were rash, hyperbilirubinemia, fatigue and neutropenia. PIK3CA mutation status was not associated with CBR nor PFS. |
| Lapatinib+capecitabine [176] | Phase I pos hoc analysis. | HER2-positive advanced or MBC patients. (38). | Treatment schedule consisted of lapatinib 1250 mg daily, 1 hour before or after breakfast, administered as single agent for the first 10 days, then continuously, in combination with capecitabine 2000 mg/m2, starting on day 11 (for the first cycle), and then from day 8, for 14 days out of a 21-day cycle. Cholestyramine was administered twice a day on a continuous basis, long after capecitabine and lapatinib intake. Diarrhea was the main AE: 13.2% grade 1, 10.5% grade 2, 2.6% grade 3 and no grade 4 events. ORR=34.2%, CB=55.3%, PFS=10 months. OS (1 year)=71.2%. The results are comparable with previous reports of conventional administration of the lapatinib-capecitabine regimen and led to a significant reduction in the incidence and severity of diarrhea. |
| Lapatinib+Pegylated liposomal doxorubicin [177] | Phase II, open-label, single-arm multicenter study. | HER2-positive with locally advanced, inoperable or MBC with disease progression after TZ therapy. (24). | Treatment consisted in 1250 mg lapatinib daily until progression plus 40 mg/m2 of pegylated liposomal doxorubicin at every 4 weeks for a maximum of 6 cycles. This combination showed to be active and safe in HER2-positive MBC, especially suitable for patients with cardiological risk or brain metastases. ORR was 54%. PFS and OS were 5.8 and 23.3 months respectively. The one-year PFS rate was 27% and 1-year OS rate 76%. The most commonly observed AEs were diarrhea, rash and infection. No grade 4 events reported. |
| Lapatinib+capecitabine vs lapatinib+topotecan [58] | Phase II, open-label, two-arms randomized study. | HER2 breast cancer with brain metastases despite prior standard treatment with WBRT and/or stereotactic radiosurgery and TZ. (13/9). | No CNS-OR was observed with lapatinib (1250 mg/day) plus topotecan (3.2 mg/m2) therapy and showed excessive toxicity (one death possible related to it). ORR=38% in the lapatinib plus capecitabine arm. |
| Lapatinib+capecitabine vs trastuzumab+capecitabine [178] | Prospective non-randomized two-arms controlled study. | Chinese patients with HER2 invasive and MBC resistant to TZ and previously received taxane therapy. (60/60). | Lapatinib (1250 mg/day) plus capecitabine (2000 mg/m2/day) therapy was superior than TZ therapy (6 mg/kg every 21 days [after the initial 8 mg/kg loading dose]) plus capecitabine (2000 mg/m2/day) for PFS and CBR (6.0 vs 4.5 months, 48 vs 63% respectively). |
| Lapatinib+vinorelbine vs lapatinib+capecitabine [179] | A phase II randomized multicenter study. | HER2-positive MBC patients who had received no more than one chemotherapeutic regimen. | Both therapies showed similar outcomes. Combination therapies doses were: Arm A, 1250 mg/day lapatinib plus 20 mg/m2/day vinorelbine. Arm B, 1250 mg/day lapatinib plus 2000 mg/m2/day capecitabine. PFS for both therapies=6.2 months. OS=24.3 for arm A vs 19.4 months for arm B. 42 patients opted to cross over and second evaluation was make: PFS=3.25 months for lapatinib plus vinorelbine vs 4.0 months lapatinib plus capecitabine. The most commonly observed AEs were diarrhea, neutropenia, PPE, rash, nausea, and fatigue. There were more serious AEs in arm A than in arm B. |
| Arm A: lapatinib plus vinorelbine. | |||
| Arm B: lapatinib plus capecitabine. (70/35). | |||
| Lapatinib+pazopanib [180] | Phase II two-cohorts multicenter study. | HER2 relapsed or refractory IBC. | |
| Cohort 1: previous history of IBC and documented recurrence in the skin and/or other disease sites by radiologic assessments (76). | Cohort 1: combination therapy (lapatinib 1500 mg/day plus pazopanib 800 mg/day) had higher clinical activity than monotherapy (lapatinib 1500 mg/day) for ORR (29 vs 45%) respectively, but was not well tolerated. | ||
| Cohort 2: cutaneous disease documented with photographs (88). | Cohort 2: combination therapy (lapatinib 1000 mg/day plus pazopanib 400 mg/day) was well tolerated and was superior to monotherapy (lapatinib 1500 mg/day) for CBR (58 vs 47%) but it was not for PFS. Combination showed an increase in toxicity. | ||
| Lapatinib+pazopanib [181] | Phase II randomized two-cohorts, multicenter study. | HER2 invasive and MBC. No prior anticancer therapy (except hormonal therapy) was permitted in the randomized. | |
| Cohort 1: randomized (150). | Combination therapy in cohort 1 (lapatinib 1000 mg/day plus pazopanib 400 mg/day) was not superior to monotherapy (lapatinib 1500 mg/day) for PDR (36.2 vs 38.9%), but it was for ORR (36.2 vs 22.2%). | ||
| Cohort 2: nonrandomized (40). | In cohort 2: (lapatinib 1500 mg/day plus pazopanib 800 mg/day) ORR=33.3%. AEs grade 3/4 including diarrhea, hypertension and liver enzymes elevation were higher in combination in cohort 2 than in combination in cohort 1. | ||
| Lapatinib+bevacizumab [182] | Phase II open-label, multicenter study. | HER2 invasive and MBC. (52). | Lapatinib (1500 mg/day) plus bevacizumab (10 mg/kg every 2 weeks) therapy was active in terms of PFS (24.7 weeks), CBR (30.8%) and ORR (13.3%). The most common AEs were diarrhea, rash, fatigue, nausea, headache, and epistaxis, grade 1/2. AEs grade 3/4 were rash, hypertension, diarrhea, hyperbirulinemia, hydronephrosis, gastrointestinal hemorrhage and liver enzyme alteration. |
| Lapatinib+paclitaxel [183] | Phase III randomized two-arms, double-blind, multicenter study. | Newly diagnosed HER2 MBC. (215/215). | Lapatinib (1500 mg/day) combined with paclitaxel (80 mg/m2 weekly) was superior to paclitaxel alone (80 mg/m2 weekly) for OS, PFS and ORR (27.8 vs 20.5 months, 9.7 vs 6.5 months and 69 vs 50% respectively). Incidence of grades 3/4 diarrhea and neutropenia was higher in the lapatinib plus paclitaxel arm. |
| Lapatinib+paclitaxel [184] | Phase III randomized two-arms, double-blind, multicenter study. | HER2-negative or initially untested, locally advanced or MBC previously untreated. (291/288). | Combination therapy (paclitaxel , 175 mg/m2 every 3 weeks plus lapatinib 1500 mg/day) was superior to monotherapy (paclitaxel, 175 mg/m2 every 3 weeks) cómo había un subset HER2 positive si en las especificaciones dice HER2 negative or untested? in terms of ORR, CBR, TTP (63.3 vs 37.8%, 69.4 vs 40.5%, 36.4 vs 25.1 weeks, respectively). Rash and diarrhea grade 3 were higher in the combination arm. |
| Lapatinib+nab-paclitaxel [185] | Phase II open-label, single-arm, multicenter study. | HER2 MBC with no more than one prior chemotherapeutic regimen. (60). | The recommended doses for this combination therapy were lapatinib: 1000 mg and nab-paclitaxel 100 mg/m2. This combination showed to offer clinical benefit (ORR=53%, PFS=39.7 weeks, TTP=41 weeks). Data were consistent with those reported for lapatinib in combination with paclitaxel. |
| Lapatinib+paclitaxel+gemcitabine [186] | Phase I open-label study. | HER2 early breast cancer, no previously treated. (13). | The recommended doses for this combination were lapatinib 1000 mg/day, paclitaxel 80 mg/m2 on days 1 and 8 and gemcitabine 1000 mg/m2 on days 1 and 8, every 3 weeks. The most frequent AE was neutropenia grade 3/4. Other AEs of more than grade 2 were rare: liver enzyme alteration, anorexia, gastritis, abdominal pain, diarrhea and paronychia. Combination therapy was well tolerated and showed clinical benefit (PR=61.5%, CR=30.7%). |
| Lapatinib+TZ [187] | Phase I dose-escalation study. | HER2 advanced or MBC. (54). | The optimally tolerated regimen was lapatinib 1000 mg/day plus TZ 2 mg/kg weekly (after the initial 4 mg/kg loading dose). Combination therapy showed to offer clinical benefit (ORR=15%). The responders had received prior TZ therapy in combination with cytotoxic chemotherapy. The most frequent AEs were diarrhea, rash, fatigue and nausea. Most common grade 3 events included diarrhea, fatigue, and rash. |
| Lapatinib+TZ [24] | Phase III randomized two-arms open-label multicenter study. | HER2 MBC progressed in the last TZ treatment. Must have received prior anthracycline- and taxane-based regimens. (148/148). | Combination therapy (lapatinib 1000 mg/day plus TZ 2 mg/kg weekly [after the initial 4 mg/kg loading dose]) was superior to lapatinib alone (1500 mg/day) for PFS and CBR (12.0 vs 8.1 weeks and 24.7 vs 12.4% respectively). Incidence of diarrhea was higher with the combination arm. |
| Lapatinib+TZ [188] | Phase II single-arm, multicenter study. | HER2 invasive breast cancer. (66). | Combination therapy (TZ [4 mg/kg loading, then 2 mg/kg/week] and lapatinib 1000 mg/day) showed to be clinically active (CR=27%, pRR=22%). Only 3% of the patients experienced disease progression. The most common AEs were diarrhea, rash, fatigue and nausea, all grade 1/2. AEs grade 3/4 were liver enzymes alterations, diarrhea, rash and hypertension. |
| Lapatinib vs TZ [189] | Phase II randomized open-label study. | HER2 positive MBC not previously treated with chemotherapy and/or anti-HER2 agents for metastatic disease. (19). | No significant difference in terms of response at 8 weeks was observed according to treatment arm (75 and 67% MR or more with lapatinib [1500 mg daily for 8 weeks] and TZ [8 mg/kg on loading dose, followed by weekly trastuzumab at the dose of 2 mg/kg] respectively). In the overall population, persistence in protocol, PSF and OS were 3.8, 7.3 and 43 months, respectively. MBC patients bearing tumors with markers such as “HER2-enriched” subtype and/or having high HER2/p95HER2 protein expression ratio are exquisitely sensitive to anti-HER2 agents and could be candidates for studies aimed at establishing chemotherapy-free regimens. |
| Lapatinib+TZ+docetaxel [190] | Phase I open-label, dose-escalation, multicenter study. | HER2 MBC. (53). | Two regimens were recommended: lapatinib 1250 mg plus docetaxel 75 mg/m² every three weeks and lapatinib 1000 mg plus docetaxel 100 mg/m² every three weeks, both with weekly TZ (loading dose of 4 mg/kg followed by a fixed dose of 2 mg/kg) and including use of prophylactic granulocyte colony stimulating factor. Combination therapy showed to offer clinical benefit (ORR=64%). The most common AEs were diarrhea and nausea. Grades 3/4 were neutropenia, diarrhea, leukopenia, peripheral neuropathy, and rash. |
| Lapatinib+TZ+paclitaxel [191,192] | Phase III randomized open-label, multicenter study. | HER2 invasive early breast cancer. (152/154). | Combination therapy (lapatinib 1000 mg/ day plus TZ 2 mg/kg weekly [after the initial 4 mg/kg loading dose] plus paclitaxel 80 mg/m2/week) was superior to TZ alone (2 mg/kg weekly [after the initial 4 mg/kg loading dose) and to lapatinib alone (1500 mg/day) for CR (51.3 vs 29.5 vs 24.7% respectively). Grade 3 diarrhea and liver enzyme alteration were higher in the combination arm. 3 years EFS was 78% for lapatinib, 76% for TZ and 84% for combination. 3 years OS was 93% for lapatinib, 90% for TZ and 95% for combination. EFS and OS did not differ between treatment groups but patients who achieve CR after neoadjuvant anti-HER2 therapy have longer EFS and OS than do patients without CR. |
| Lapatinib+TZ+paclitaxel [193] | Phase II single-arm, multicenter study. | HER2 surgically resected invasive breast cancer. (122). | Lapatinib given concurrently with paclitaxel and TZ is feasible and did not add cardiac toxicity. The dose of lapatinib in this regimen should not exceed 750 mg daily. Diarrhea grade 3/4 was the most common serious AE. An early aggressive management of this AE is recommended. |
| TZ-emtansine vs capecitabine+lapatinib [194] | Phase III randomized two-arms study. | HER2-positive, unresectable locally advanced or MBC previously treated with TZ and taxane. (495/496). | TZ-emtansine (3.6 mg/kg intravenously every 3 weeks) delayed the time to symptom worsening compared with capecitabine (1000 mg/m2 orally twice a day, on days 1 through 14 of a every 3 week cycle) plus lapatinib (1250 mg orally daily), 7.1 vs 4.6 months, respectively. In the TZ-emtansine arm, 55.3% of patients developed clinically significant improvement in symptoms vs 49.4% in the capecitabine plus lapatinib arm. Diarrhea cases were higher in the capecitabine plus lapatinib arm than in the TZ-emtansine arm. |
| Lapatinib+TZ+chemotherapy [195] | Phase II randomized three-arms multicenter study. | HER2 infiltrating breast cancer. (36/39/46). | Combination of chemotherapy (fluorouracil 600 mg/m2, epirubicin 75 mg/m2, and cyclophosphamide 600 mg/m2 administered every 3 weeks) with lapatinib (1000 mg/day) and TZ (2 mg/kg weekly after the initial 4 mg/kg loading dose) was superior to chemotherapy plus lapatinib and chemotherapy plus TZ for CR (46.7 vs 25.0 vs 26.3% respectively). The majority of the patients required lapatinib dose reduction because high grade 3 diarrhea incidence. |
| Chemotherapy+TZ vs chemotherapy+lapatinib [196] | Phase II randomized two arms, open label, multicenter trial. | Stages I, II, III or inflammatory breast cancer HER2 positive. (50/52). | Chemotherapy consisted in epirubicin 90 mg/m2 plus cyclophosphamide 600 mg/m2 both administered on day 1 every 21 days for four cycles followed by docetaxel 100 mg/m2 administered on day 1 every 3 weeks for four cycles. The anti-HER2 therapy was added to docetaxel as follows: patients in arm A received chemotherapy plus TZ 6 mg/kg (after a loading dose of 8 mg kg/1) on day 1 every 21 days (EC-DT). Patients in arm B received chemotherapy plus a daily dose of lapatinib 1250 mg orally (EC-DL). EC-DT exhibited higher efficacy; than EC-DL in terms of CR (52.1 vs 25.5% in breast, 47.9 vs 23.5% in breast and axila). Grade 3/4 toxicity rates were similar across arms except for diarrhea, which was more frequent in the EC-DL arm. |
| Chemotherapy+TZ vs chemotherapy+lapatinib [197] | Phase III randomized two arms, open label, multicenter trial. | Untreated HER2-positive operable or locally advanced breast cancer. (309/311). | Chemotherapy consisted in epirubicin 90 mg/m2 plus cyclophosphamide 600 mg/m2 both administered every 3 weeks for four cycles followed by docetaxel 100 mg/m2 administered on day 1 every 3 weeks for four cycles. The anti-HER2 therapy was added to docetaxel as follows: patients in arm A received chemotherapy plus TZ 6 mg/kg (after a loading dose of 8 mg kg/1) on day 1 every 21 days (ECH-TH). Patients in arm B received chemotherapy plus a daily dose of lapatinib 1000-1250 mg orally (ECH-TL). ECH-TH exhibited higher efficacy in terms of CR (30.3 vs 22.7% respectively) than ECH-TL. Chemotherapy with trastuzumab was associated with more edema (39·1% vs 28·7%) and dyspnea (29·6 vs 21·4%), and ECL-TL with more diarrhea (75·0 vs 47·4%) and skin rash (54·9 vs 31·9%). Less AE’s were reported in the ECH-TH group than in the ECL-TL group. |
MBC, metastatic breast cancer; IBC, inflammatory breast cancer; CR, complete response; PR, partial response; OR, objective response; PFS, progression free survival; PTEN, tensin homologue deleted in chromosome 10; AE, adverse event; CBR, clinical benefit rate; TZ, trastuzumab; ORR, objective response rate; OS, overall survival; SD, stable disease; TTP, time to progression; CNS, central nervous system; PPE, palmar-plantar erythrodysesthesia; WBRT, whole brain radiotherapy; PS, performance status; PDR, progressive disease rate; pPR, pathological response rate; EFS, event free survival; MR, minimal response.
In general, the addition of lapatinib to another therapy improved the efficacy compared with the drug alone and in some cases without increasing the adverse events profile or its severity.
Some studies have assessed the efficacy of lapatinib alone and in combination with capecitabine in HER2 breast cancer patients with progressive brain metastases. Only modest efficacy has been found with monotherapy while combination therapy has shown to be more effective suggesting that lapatinib plus capecitabine could be an alternative treatment for specific patients to delay whole brain radiotherapy and its adverse effects (Table 1) [56-59].
Lapatinib resistance and toxicity
Several cancers generate resistance to TKIs through modifications in different proteins implicated in downstream signaling of their target receptors [60]. Herein, we will revise the proteins and cellular processes known to be involved in acquiring resistance to lapatinib.
Dysregulation of the PI3K pathway is frequent in breast cancer leading to its constitutive activation [61,62]. Mutations in the catalytic subunit of PI3K (PIK3CA) have been reported in 20-30% of breast cancer patients with HER2 amplification [63,64]. In HER2 overexpressing cells carrying PIK3CA, the antiproliferative and proapoptotic effects of lapatinib as well as its ability to inhibit survivin and AKT phosphorylation was less pronounced than in those cells without PIK3 mutations [33,65]. Interestingly, PIK3CA has been considered a biomarker of resistance to the EGFR family targeted therapy [66]. Another abnormal activation of the PI3K pathway is the loss of phosphatase and tensin homologue deleted in chromosome 10 (PTEN) function that has been observed in 20% to 25% of primary breast cancers [63,67]. In this regard, it has been demonstrated that loss of PTEN expression conferred resistance to lapatinib in both PTEN knockdown cells and tumor xenografts with HER2 overexpression, through maintaining the activation of the AKT signaling pathway [65,68]. However, another study did not find association between PTEN protein levels and resistance to lapatinib in a panel of 17 HER2-amplified cell lines [69]. This was consistent with two studies that did not show association between PTEN and lapatinib response using cell lines and tissues of inflammatory breast cancer patients with PTEN deficiency [53,70].
Hypoxia promotes lapatinib resistance in HER2-positive breast cancer cells by inducing high levels of hypoxia inducible factor (HIF)-1α and reducing the dual specificity phosphatase2 (DUSP2) expression that leads to activation of the ERK pathway [71].
Autophagy plays an important role in the development of drug resistance in breast cancer cells [72]. Indeed, increase of autophagosome formation was observed in lapatinib resistant breast cancer cell lines. Accordingly, treatment with autophagy inhibitors restored lapatinib sensitivity [73].
Acquired resistance to lapatinib in HER2-positive breast cancer cells can also be mediated by induction of heregulin (HRG) and subsequent formation of HER3-EGFR heterodimers that promote an HRG-HER3-EGFR-PI3K signaling axis [74].
In breast cancer, the overexpression of CD24, a glycosylphosphatidylinositol-anchored membrane protein, is associated with a poor patient prognosis [75]. Noteworthy, CD24 contributes to resistance to lapatinib in HER2-positive breast cancer cells. In fact, knockdown of CD24 downregulated HER2 expression, AKT phosphorylation and increased the sensitivity of cells to lapatinib treatment [76].
Similary, the overexpression of Neuromedin U (NmU), a neuropeptide that has been associated with poor patient outcome in HER2-overexpressing tumors, is also associated with resistance to lapatinib. NmU knockdown significantly decreased migration, motility, invasion and resistance to anoikis in HER2 overexpressing breast cancer cells [77].
In a HER2-overexpressing/estrogen receptor (ER) positive breast cancer cell line treated continuously with lapatinib, increased ER signaling was observed. This was due to activation of the transcription factor forkhead box protein O3a (FOXO3a) in response to ErbB2-PI3K-AKT signaling inhibition, resulting in autoresistance to this inhibitor [78]. Consequently, ER signaling assumed a more prominent role in cell survival during the development of acquired resistance to the TKI. Accordingly, the combination of lapatinib with antagonists of the ER such as tamoxifen or fulvestrant prevented the development of lapatinib resistance [78]. In addition, in tumor biopsies of patients with early-stage breast cancer, after 14 days of lapatinib therapy, increased FOXO3a, progesterone receptor and Bcl-2 expression was observed [78].
In human breast cancer lines and xenograft models it was demonstrated that FAM83A overexpression, an oncogenic protein, is involved in lapatinib resistance [79]. Interestingly, FAM83A downregulation decreased proliferation and rendered cells sensitive to lapatinib. The mechanism by which this protein conferred resistance to lapatinib may include the interaction of FAM38A with cRAF and PI3K, maintaining the activation of MAPK signaling. Indeed, breast cancer patients with high FAM83A expression have poor prognosis [79].
Some receptors with tyrosine kinase activity have been involved in lapatinib resistance. In this sense, the aberrant expression of AXL, recepteur d’origine nantais (RON) and fibroblast growth factor receptor 2 (FGFR2) has been reported to play an important role in lapatinib resistance through eliciting PI3K/AKT and ERK signaling activation. In fact, inhibitors that target AXL, RON and FGFR2 restored lapatinib sensitivity in HER2 breast cancer cell lines [36,80,81].
Protein tyrosine kinase 6 (PTK6), Src (both non-receptor tyrosine kinase proteins), the chemokine CXCL12 and its receptor (CXCR4) are involved in breast cancer progression, regulating several cellular processes involved in the malignant phenotype, such as cell proliferation, survival, invasion and migration. Current researches have demonstrated that increased levels of PTK6 and Src expression and phosphorylation respectively, are involved in lapatinib resistance through EGFR-dependent signaling. Further, Src family kinases mRNA expression was upregulated in HER2-positive tumors treated with lapatinib. In fact, treatment with Src inhibitors in combination with lapatinib reduces AKT and ERK1/2 phosphorylation restoring the sensitivity of resistant cells to lapatinib [82-85]. Moreover, lapatinib resistant cells also show high levels of expression of CXCR4. Blockade of this chemokine receptor with a specific antibody reduced the invasive ability of lapatinib resistant cells. Notably, the combination of CXCR4 antibody and a Src inhibitor saracatinib, resulted in a greater inhibition of the invasiveness compared with either agent alone in lapatinib resistant cells. These findings suggest that blockade of Src and CXCR4 could be utilized as a therapeutic option in patients with acquired resistance to lapatinib [84].
A key feature for acquired lapatinib resistance is the fact that lapatinib treatment in HER2-positive breast cells induces the activation of several networks of kinases that contribute to induce lapatinib resistance. Efforts for dealing with this tumor response include the pharmacological inhibition of the adaptive kinome response. In this regard, inhibitors of epigenetic enzymes of BET family bromodomains suppressed the kinome reprogramming induced by lapatinib [86].
Aberrant miR-630 expression is related with lapatinib resistance in HER2-positive breast cancer cells. Overexpression of this miR reduced the metastatic phenotype and restored the drug sensitivity through decreased IGF1R expression and subsequent inhibition of the phosphorylation of HER2 and EGFR in lapatinib resistant cells [87].
In HER2-positive breast cancer cells, elevated expression of the alpha catalytic subunit of cyclic AMP-activated protein kinase A (PRKACA) confers resistance to lapatinib via increased phosphorylation of the pro-apoptotic molecule BAD [88].
The Kruppel-like transcription factors (KLFs) 4 and 5 (KLF4/5) play an import role in cell proliferation, differentiation, and transformation and have been considered as possible prognostic factors in breast cancer [89,90]. In fact, a positive correlation between HER-2/neu and KLF5 has been described [90]. In HER2-positive breast cancer cells, lapatinib treatment upregulated KLF4/5 protein expression. In these cells, KLF4 and/or KLF5 depletion restored lapatinib sensibility and reduced basal mRNA and protein levels of the anti-apoptotic factors myeloid cell leukemia 1 (MCL1) and B-cell lymphoma-extra-large (BCL-XL) [91]. The activation of another transcription factor, the aryl hydrocarbon receptor (AhR), promotes several signaling pathways as EGFR, PI3K/AKT and ERK. In fact, the induction of apoptosis by lapatinib is inhibited by this receptor activated by its external ligand (2,3,7,8-tetrachlorodibenzo-p-dioxin), indicating its participation in lapatinib resistance [92].
In patients with residual breast cancers following administration of taxane/anthracycline-based chemotherapy used concurrently with trastuzumab, loss of HER2 gene amplification has been reported [93]. Interestingly, HER2 breast cancer cells also showed loss of HER2 expression after chronic exposure to lapatinib, conferring resistance to this inhibitor [81]. Another mechanism that limits the antitumor activity of lapatinib is the upregulation of HER3 induced by this TKI, maintaining the activation of AKT and MAPK pathways. These evidences advice the use of HER2/HER3 inhibitors combined in the treatment of HER2-positive breast cancer [94,95].
Gefitinib
Gefitinib is a reversible EGFR TKI that has been approved by the FDA for the treatment of advanced non-small cell lung carcinoma with activating EGFR mutations [96,97].
EGFR is overexpressed in breast cancer tissue with a positivity range of 20-70% [98,99]. Overexpression of this receptor is associated with aggressive metastatic breast tumors. In addition, breast tumors that cooverexpress EGFR and HER2 exhibited a worse outcome than tumors that overexpressed either receptor alone [100]. Interruption of EGFR function with specific TKIs may disrupt EGFR-HER2 cross-talk, resulting in HER2 inactivation [101].
Mechanism of action of gefitinib: preclinical evidence
Gefitinib inhibits the growth of both breast cancer cell lines and human tumor xenografts expressing different levels of EGFR or HER2 [101-103]. Gefitinib effects on HER2 and EGFR coexpressing breast cancer cells are mediated by reducing basal EGFR, HER2 and HER3 phosphorylation, resulting in the blockage of downstream signaling of the AKT, MAPK and STAT3 pathways [101-105]. Also, this TKI increased p27 levels and the subG1/G1 phases of the cell cycle; reduced cyclin D1 and Cdk4. In addition, gefitinib reduced the phosphorylation of glycogen synthase kinase 3 beta (GSK-3β, a target of the AKT kinase) [101,106]. In EGFR-HER2 breast cancer cells, gefitinib induced cytostatic and apoptotic effects [103]. This action of gefitinib is in part mediated by increased p38 MAPK levels [102], dephosphorylation of FOXO3a with a subsequent increased of p27Kip1, caspase 3 and BIM protein expression [105,106]. Gefitinib has also been shown to downregulate the mTOR signaling pathway in human breast cancer cells [107].
In a similar manner as described above in the cell lines, gefitinib inhibited EGFR and MAPK phosphorylation in tumor biopsies. However, gefitinib had not effect on AKT phosphorylation or Ki67 levels. Moreover, the TKI did not increase p27 levels [108].
Gefitinib treatment disrupted the formation of the HER3/HER2 heterodimer and further association of HER3 with p85α the regulator subunit of PI3K [101]. In addition, the TKI inhibited the activation of the EGFR/HER2 and EGFR/HER3 heterodimers mediated by EGF and heregulin, respectively [102].
EGFR overexpression did not determine gefitinib sensitivity in the particular case of HER2 overexpressing breast cancer [103,104]. In this regard, gefitinib was more potent to inhibit the proliferation of breast cancer cells with high levels of HER2 and low levels EGFR compared to those cells with high levels of EGFR without HER2 expression [101,102,104]. In contrast, gefitinib effects on AKT, MAPK, and p27 were not seen in EGFR-negative breast cancer cells [101]. Interestingly, inhibition of MAPK phosphorylation was observed in EGFR-negative tumor biopsies [108], suggesting that gefitinib may be inhibiting other EGFR family members [108].
In the same way as observed with lapatinib, prolonged exposure to gefitinib induced resistance to doxorubicin, etoposide, and m-AMSA through downregulation of topoisomerase IIα [51]. A graphic summary of gefitinib mechanisms of action is found in Figure 1.
Gefitinib: clinical evidence and side effects
There are only two phase I/II clinical studies that assess the effects of different gefitinib-combination therapies specifically in HER2-positive metastatic breast cancer patients. The first one was undertaken in 35 patients divided according to prior systemic therapy. It demonstrated that the combination of 250 mg/day of gefitinib with 2 mg/kg weekly (after the initial 4 mg/kg loading dose) of trastuzumab was safe. However, there was no greater clinical benefit compared to trastuzumab monotherapy. A possible explanation of the lack of clinical activity in this study was that coupling between HER2 and HER3 and activation of the last one would not be completely inhibited by the combination of gefitinib and trastuzumab [109]. The other study included 31 patients treated with 250 mg/day of gefitinib, 6 mg/kg every 3 weeks (after the initial 8 mg/kg loading dose) of trastuzumab and 60 mg/m2 every 3 weeks of docetaxel. The values obtained were considered similar to those in other three drug combination regimens including trastuzumab and chemotherapy, with the advantage of less toxicity because the avoidance of a second chemotherapeutic agent. Additionally, no relationship between assessed biomarkers expression and response to therapy was found. In regard to EGFR, 95% of the biopsies showed little or no signal when evaluated by immunohistochemistry, although 47% of this subpopulation responded to the therapy [110].
Gefitinib alone or in combination with other drugs has been assessed in several clinical studies with HER2-negative breast cancer patients. The results are inconsistent but in some cases there is a correlation between the outcome and prior therapies administrated. We discuss these studies below.
In a clinical trial conducted in women with stage IIIb/IV advanced breast cancer resistant to chemotherapy, gefitinib monotherapy showed only a stabilization of disease for at least 3 months, neither partial nor complete response were achieved. Interestingly, a good correlation between the degree of inhibition of EGFR phosphorylation in skin and in breast tumors after gefitinib treatment was reported. Therefore, the inefficacy of this TKI could be explained by the lack of EGFR tumor dependence [108]. In other study, gefitinib was not effective in patients with recurrent or refractory breast cancer previously treated with taxane and anthracycline [111].
Moreover, studies in patients with ER and/or progesterone receptor positive breast adenocarcinoma [112,113], primary untreated breast cancer [114] and metastatic breast cancer [115], it was found that the addition of gefitinib to standard treatments (chemotherapy or hormonal therapy) did not further increase the clinical benefit. However, in some cases it caused higher toxicity and serious adverse events such as dehydration, diarrhea, fatigue, stomatitis, hypokalemia and neutropenia.
In contrast, Polychronis et al., reported that gefitinib alone and in combination with anastrozol was effective for reducing the tumor size in ER-EGFR positive breast cancer naive patients. The most frequently adverse effects presented with both therapies were rash and gastrointestinal disorders such as diarrhea, dry mouth, nausea and loose stools [116]. Ciardiaello et al., also reported good response in patients with metastatic breast cancer treated with gefitinib plus docetaxel, who had not previously received any kind of therapy for metastatic disease. The most common adverse effects in this study were neutropenia, diarrhea and nausea [117]. Interestingly, Osborne et al, found that the gefitinib plus tamoxifen combination was effective only in patients with newly diagnosed metastases or those who had recurred one year or more after stopping adjuvant therapy with tamoxifen but not in patients with recurrent disease during or after adjuvant treatment with aromatase inhibitors or those progressing after first-line aromatase inhibitor treatment for metastatic disease [118].
Additionally, Kalykaki et al., demonstrated that gefitinib was effective to reduce the number of EGFR positive and negative circulating tumor cells in patients with metastatic breast cancer [119].
The most common adverse effects of gefitinib as monotherapy in breast cancer patients are diarrhea, rush, dry skin, pruritus, dry mouth, nausea, vomiting and fatigue [108,111,116].
Gefinib resistance and toxicity
The constitutive activation of signaling pathways downstream of EGFR contributes to gefitinib resistance [104,120,121]. Normanno et al., demonstrated that hiperactivation of the MAPK pathway is involved in both the intrinsic and acquired gefitinib resistance in breast cancer cells. Interestingly, blockade of the MAPK activity resulted in growth inhibition and induction of apoptosis. In addition, constitutive activation of p42-MAPK was related with gefitinib resistance [120,121].
Other study related with intrinsic gefitinib resistance demonstrated that this TKI increased the expression of genes codifying for HER specific ligands and induced active import and nuclear accumulation of the HER ligand neuregulin, suggesting its possible transcriptional role [122]. Moreover, HER3 and consequently PI3K/AKT signaling evade inhibition by gefitinib in vitro and in vivo [123].
Regarding autophagy, gefitinib induced this cellular process in various gefitinib-sensitive and -insensitive breast cancer cell lines. Treatment with gefitinib in combination with agents that inhibit late-stage autophagy such as hydroxychloroquine or bafilomycin A1 increased efficacy of gefitinib in vitro and in vivo [107].
Alternatively, other studies suggest that the upregulation of the PI3K pathway caused by loss of PTEN activity is involved in gefitinib resistance in breast cancer [104,124,125]. In fact, in an EGFR overexpressing breast cancer cell line which lacks PTEN function, gefitinib abolished the phosphorylation of both EGFR and MAPK but not AKT [124]. Moreover, gefitinib treatment in cells that PTEN function was restored resulted in the inhibition of AKT, GSK-3β and the translation repressor protein 4EBP1 phosphorylation [124], restoration of gefitinib sensitivity, arrest in the G1 phase [125], and relocalization of FOXO3a into the nucleus. This transcriptional factor is implicated in cell cycle arrest and apoptosis [126].
Other mechanisms implicated in gefitinib resistance in breast cancer involved the interplay between EGFR and different receptors. In this regard, hepatocyte growth factor receptor (MET), a tyrosine kinase receptor implicated in breast cancer progression, confers gefitinib resistance through increased EGFR phosphorylation induced by the activation of the kinase c-Src. In fact, an inhibitor of MET kinase activity decreased both EGFR phosphorylation and cell proliferation in the presence of gefitinib in HER2-positive and EGFR overexpressing breast cancer cells [127].
As with lapatinib, FAM83A dysregulation has been show to promote gefitinib insensitivity in breast cancer cells [79].
The crosstalk between G-protein coupled receptors (GPCR) and EGFR [128]; K-RAS activation (a downstream effector of EGFR signaling) [129] and EGFR mutations [130] have all been involved with gefitinib resistance in lung cancer. In this regard, GPCRs-EGFR interaction has also been reported in breast cancer [131], Moreover, K-RAS gene mutations were found in several adenocarcinomas [132-134]. Interestingly, EGFR mutations have not been reported neither in primary breast carcinomas nor in several breast cancer cell lines [135]. These issues have not yet been addressed in gefitinib resistant breast cancer cells and will have to be further analyzed.
Neratinib
Neratinib is another oral, but irreversible TKI, known as a pan-inhibitor because interacts with the catalytic domain of several EGFR family members (EGFR, HER2 and HER4) and blocks their downstream signaling pathways [136]. Neratinib covalently binds a specific and shared cysteine residue in the ATP-binding pocket of the receptors in the EGFR family [137]. In particular, neratinib binds to cysteine residues Cys-773 and Cys-805 in HER1 and HER2, respectively [138].
Neratinib derives from structural modifications of EKB-569, another potent and irreversible EGFR inhibitor [136,139]. Neratinib has significant activity in naïve and previously exposed to trastuzumab patients [140], making it an alternative treatment for HER2-positive breast cancer. Currently, this TKI is in clinical trials and has been used to treat solid tumors and metastatic HER2 breast cancer [141-144].
Mechanism of action of neratinib: preclinical evidence
There are some reports that describe the mechanism of action of neratinib in breast cancer. A pioneering work from Rabindran et al., showed that neratinib inhibited proliferation and EGFR, HER2, HER4, AKT and MEK phosphorylation in HER2 overexpressing breast cancer cell lines [136,145]. The regulation of downstream signal transduction by neratinib leads to arrest at the G1-S phase transition resulting in increased p27 levels and decreased phosphorylated retinoblastoma protein (pRb) and cyclin D1 expression. Interestingly, neratinib showed less antiproliferative activity in cell lines that express neither HER2 nor EGFR [136]. Moreover, HER2-positive breast cancer cell lines are more likely to respond to neratinib than EGFR-positive cells or HER2 non-amplified cell lines [145]. Another antineoplastic mechanism for neratinib in cancer cell lines is that it can reverse membrane-bound ATP transporters-mediated multidrug resistance [146]. The inhibition of multidrug resistance via ATP transporters by neratinib may be an alternative mechanism that could improve the response to chemotherapy agents used in HER2-positive breast cancer.
Similarly, neratinib enhanced the therapeutic response and counteracted trastuzumab resistance by decreased trastuzumab-induced HER4 nuclear translocation in HER2-positive breast cancer [145,147]. A resume of neratinib mechanisms is found in Figure 1.
Neratinib: clinical evidence and side effects
Neratinib has proved to be a promising therapy for HER2 metastatic breast cancer in five recent phase I/II clinical studies listed in Table 2. Nevertheless, in a phase II trial, Martin et al. compared two treatments: neratinib versus lapatinib plus capecitabine with inconclusive results, since neither inferiority nor superiority of the monotherapy versus combination therapy could be demonstrated. However, they confirmed relevant single agent clinical activity and acceptable overall tolerability of neratinib [148]. These studies have warranted further studies of this TKI and its combination with other therapies. In this regard, several clinical trials phase I/II (ClinicalTrials.gov identifiers: NCT01008150, NCT01111825, NCT01670877, NCT00398567, NCT00741260, NCT00915018, NCT01494662, NCT00146172) and two phase III (NCT00878709 and NCT01808573) in HER2-positive breast cancer population are in progress.
Table 2.
Clinical evidence with neratinib in HER2 breast cancer
| Therapy [Ref.] | Study type | Patient population (n) | Principal findings |
|---|---|---|---|
| Neratinib [143] | Phase II open-label, two-cohorts, multicenter study. | HER2 advanced and MBC. | Neratinib (240 mg daily) was well tolerated and had clinical activity (PFS at 16 weeks=59 and 78%, OR=24 and 56% for cohort A and cohort B respectively). The most common AEs were diarrhea, nausea, vomiting, and fatigue. Diarrhea was the most frequent grade 3/4 AE. |
| Cohort A: Prior TZ treatment (66). | |||
| Cohort B: No prior TZ treatment (70). | |||
| Neratinib+paclitaxel [142] | Phase I/II open-label, two-part study. | HER2-positive MBC and prior TZ therapy. | Combination therapy (neratinib 240 mg daily plus paclitaxel 80 mg/m2) was well tolerated with no unexpected toxicity and had clinical activity in both groups (OR=71 and 77%, PFS=63.1 and 52.1 weeks for group A and B respectively) with a CB=82% for all evaluable patients. The most common AEs were diarrhea, neuropathy, alopecia, nausea, neutropenia, leukopenia and anemia. The most common grade 3/4 AEs were diarrhea, neutropenia, leukopenia and anemia. |
| Group A: no more than one prior chemotherapy regimen and no prior lapatinib exposure (71). | |||
| Group B: no more than three prior cytotoxic chemotherapy regimens with prior lapatinib exposure permitted (31). | |||
| Neratinib+paclitaxel+TZ [198] | Phase I open label, dose escalation, multicenter study. | HER2-positive MBC previously treated with anti-HER agent(s) and a taxane. (21). | Combination therapy consisted in neratinib (120 up to 240 mg/day) with TZ (4 mg/kg loading dose, then 2 mg/kg weekly) and paclitaxel (80 mg/m2 days 1, 8, and 15 of a 28-day cycle). The recommended dose of neratinib for this combination therapy was 200 mg/kg. It showed to have clinical activity (OR=38%, CB=52% and median TTP=3.7 months). Common grade 3/4 AEs were diarrhea (38%), dehydration (14%), electrolyte imbalance (19%), and fatigue (19%). |
| Neratinib+vinorelbine [199] | Phase I/II multicenter, open-label study. | HER2-positive MBC, divided according to prior or no prior lapatinib treatment. (12/56). | Combination therapy (neratinib 240 mg plus vinorelbine 25 mg/m2) was well tolerated with no unexpected toxicity. It showed to have clinical activity (OR=8 vs 41%, PFS=24.0 vs 47.7 weeks, CB=5 vs 39%, prior lapatinib vs no prior lapatinib respectively). The most common AEs were diarrhea, neutropenia, vomiting, nausea and fatigue. The most common grade 3/4 AEs were neutropenia (46%), diarrhea (28%), and leukopenia (17%). |
| Neratinib+capecitabine [200] | Phase I/II multinational open-label study. | HER2-positive MBC or locally advanced breast cancer, previously treated with TZ and taxane. There is a sub-group previously treated with lapatinib. (61/7). | Combination therapy (neratinib 240 mg plus capecitabine 1500 mg/m2 daily) showed to have clinical activity (OR=64 vs 57%, PFS=40.3 vs 35.9 weeks, patients with no prior lapatinib exposure vs patients previously treated with lapatinib respectively). The overall most common grade 3/4 AEs were diarrhea (26%), PPE (14%), asthenia (4%) and vomiting (4%). |
MBC, metastatic breast cancer; TZ, trastuzumab; PFS, progression free survival; OR, objective response; AEs, adverse events; CB, clinical benefit; TTP, time to progression; PPE, palmar-plantar erythrodysesthesia.
Neratinib resistance and toxicity
Seyhan et al., using a pool-based lentiviral genome-wide functional RNAi screen identified the following genes involved in neratinib resistance: oncogenesis (RAB33A, RAB6A and BCL2L14), transcription factors (FOXP4, TFEC, ZNF), cellular ion transporters (CLIC3, TRAPPC2P1, P2RX2), protein ubiquitination (UBL5), cell cycle (CCNF9) and genes known to interact with breast cancer-associated genes (CCNF, FOXP4, TFEC, several ZNF factors, GNA13, IGFBP1, PMEPA1, SOX5, RAB33A, RAB6A, FXR1, DDO, TFEC, OLFM2) [149]. This analysis helps to the identification of new biomarkers related to neratinib treatment that could eventually make more effective the use of combination targeted HER2 therapy.
In a similar manner as other TKIs, neratinib also may generate resistance. In this regard, NmU overexpression and low miR-630 levels are associated with innate or acquired-resistance to neratinib in HER2-positive breast cancer cells [77,87].
In HER2-posive breast cancer cell lines, despite the initial inhibition of phosphorylation of all members of the EGFR family by neratinib, its continued exposure resulted in reactivation of HER3 and AKT signaling. This process is thought to be involved in the developing of resistance to neratinib. Interestingly, the combination of trastuzumab and neratinib blocked the reactivation of HER3 and AKT compared to neratinib alone [145].
Conclusion
Overexpression of EGFR family members and activation of their signaling pathways have been associated with the development of breast cancer and poor prognostic [150]. Interaction between these receptors is related with resistance to current therapy becoming a challenge for HER2-positive breast cancer treatment [100,151]. Targeted therapy against cell signaling pathways of EGFR family members has been introduced in the clinic in recent years since it generates fewer side effects than conventional therapy [152]. In this regard, TKIs have been developed such as lapatinb, gefitinib and neratinb. These inhibitors have a similar mechanism of action; specifically, they compete for the ATP binding pocket of the EGFR family. These agents can inhibit the same signaling pathways such as PI3K/AKT, MAPK, PLCγ, and STAT [104,153-155]. However, the differences in their responses, and their selectivity in HER2-positive breast cancer, could depend on their dissociation constant (kd) to EGFR family members, and their molecular interactions with these proteins. The biological consequences of the binding of TKIs with EGFR family members are poorly understood, and several researchers are currently examining the different TKIs interactions within the human kinome [156]. In addition, it has been described that TKIs can interact with different dimerization complexes with active or inactive forms of EGFRs [157-159]. Also, TKIs can modify the amount of EGFR family ligands [122].
However, if the three compounds have similar mechanisms of action, what is the difference between them that could explain the different responses in the clinic? A current work of Sánchez-Martín et al, in 2012 gives us an explanation about the chemical mechanism that led to increased efficacy of lapatinib and neratinib action in breast cancer versus gefitinib action. The reversible and irreversible inhibitors, lapatinib and neratinib; respectively, have in their structure an aromatic group that binds into the ATP pocket of the EGFR family while maintain it in a closed conformation, reducing the flexibility of the kinase receptor, which ultimately limits the dimerization with other receptors. Unlike lapatinib and neratinib, gefitinib lacks an aromatic group in its structure, which maintains EGFR family ATP pocket in an open conformation, favoring the dimerization with other kinase receptors [160]. Moreover, a recent work of O’Neill et al, in 2013 studied a panel of genes implicated in cell cycle progression, tumorigenesis and invasiveness, which were upregulated after treatment with neratinib in HER2 overexpressing breast cancer cells. This work demonstrated that the genic regulation induced by neratinib was similar to that seen with trastuzumab and the combination of lapatinib and capecitabine in HER2-positive breast cancer cells. In contrast, gefitinib treatment resulted in a completely different expression pattern [161,162]. This is important because the expression of genes could be predictors of the response in some patients [161]. In summary, a different gene expression profile induced by each one of the TKIs could explain the difference in the treatment response.
The TKIs have shown sustainable features to block the signaling of the EGFR family members, inhibit cell proliferation and induce apoptosis in HER2-positive breast cancer subtype. However, the search for novel therapeutic strategies comprising the use of these or other TKIs, in combination with different antineoplastic agents could further improve the prognostic in cancer patients. In order to enhance the clinical response to the TKIs therapy, we consider that tumor molecular analyses, which include the characterization of the EGFR family members, and the search for mutated or overexpressed proteins related with TKI resistance, must be performed.
Acknowledgements
This work was supported by Instituto Científico Pfizer, México, and by the Consejo Nacional de Ciencia y Tecnología (CONACyT), México grant number 154215 to DBH. MSM is a Ph.D, student from the Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, México, and recipient of a fellowship from CONACyT (324486) and PAEP, UNAM, México.
Disclosure of conflict of interest
None.
References
- 1.Beiki O, Hall P, Ekbom A, Moradi T. Breast cancer incidence and case fatality among 4.7 million women in relation to social and ethnic background: a population-based cohort study. Breast Cancer Res. 2012;14:R5. doi: 10.1186/bcr3086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Globocan. Cancer Incidence and Mortality Worldwide. IARC CancerBase (http://globocan.iarc.fr) 2008; No. 10.
- 3.Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, Fluge O, Pergamenschikov A, Williams C, Zhu SX, Lonning PE, Borresen-Dale AL, Brown PO, Botstein D. Molecular portraits of human breast tumours. Nature. 2000;406:747–752. doi: 10.1038/35021093. [DOI] [PubMed] [Google Scholar]
- 4.Sotiriou C, Pusztai L. Gene-expression signatures in breast cancer. N Engl J Med. 2009;360:790–800. doi: 10.1056/NEJMra0801289. [DOI] [PubMed] [Google Scholar]
- 5.Slamon DJ, Godolphin W, Jones LA, Holt JA, Wong SG, Keith DE, Levin WJ, Stuart SG, Udove J, Ullrich A. Studies of the HER-2/neu protooncogene in human breast and ovarian cancer. Science. 1989;244:707–712. doi: 10.1126/science.2470152. [DOI] [PubMed] [Google Scholar]
- 6.Dawood S, Broglio K, Buzdar AU, Hortobagyi GN, Giordano SH. Prognosis of women with metastatic breast cancer by HER2 status and trastuzumab treatment: an institutional-based review. J. Clin. Oncol. 2010;28:92–98. doi: 10.1200/JCO.2008.19.9844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Brower V. Cardiotoxicity Debated for Anthracyclines and Trastuzumab in Breast Cancer. J Natl Cancer Inst. 2013;105:835–6. doi: 10.1093/jnci/djt161. [DOI] [PubMed] [Google Scholar]
- 8.Dieras V, Vincent-Salomon A, Degeorges A, Beuzeboc P, Mignot L, de Cremoux P. [Trastuzumab (Herceptin) and breast cancer: mechanisms of resistance] . Bull Cancer. 2007;94:259–266. [PubMed] [Google Scholar]
- 9.Nahta R, Esteva FJ. HER2 therapy: molecular mechanisms of trastuzumab resistance. Breast Cancer Res. 2006;8:215. doi: 10.1186/bcr1612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Agrawal A, Gutteridge E, Gee JM, Nicholson RI, Robertson JF. Overview of tyrosine kinase inhibitors in clinical breast cancer. Endocr Relat Cancer. 2005;12(Suppl 1):S135–144. doi: 10.1677/erc.1.01059. [DOI] [PubMed] [Google Scholar]
- 11.Wang YN, Hung MC. Nuclear functions and subcellular trafficking mechanisms of the epidermal growth factor receptor family. Cell Biosci. 2012;2:13. doi: 10.1186/2045-3701-2-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Yarden Y. The EGFR family and its ligands in human cancer. signalling mechanisms and therapeutic opportunities. Eur J Cancer. 2001;37(Suppl 4):S3–8. doi: 10.1016/s0959-8049(01)00230-1. [DOI] [PubMed] [Google Scholar]
- 13.Asgeirsson KS, Agrawal A, Allen C, Hitch A, Ellis IO, Chapman C, Cheung KL, Robertson JF. Serum epidermal growth factor receptor and HER2 expression in primary and metastatic breast cancer patients. Breast Cancer Res. 2007;9:R75. doi: 10.1186/bcr1788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Wells A. EGF receptor. Int J Biochem Cell Biol. 1999;31:637–643. doi: 10.1016/s1357-2725(99)00015-1. [DOI] [PubMed] [Google Scholar]
- 15.Gullick WJ. Prevalence of aberrant expression of the epidermal growth factor receptor in human cancers. Br Med Bull. 1991;47:87–98. doi: 10.1093/oxfordjournals.bmb.a072464. [DOI] [PubMed] [Google Scholar]
- 16.Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science. 1987;235:177–182. doi: 10.1126/science.3798106. [DOI] [PubMed] [Google Scholar]
- 17.Zaczek A, Brandt B, Bielawski KP. The diverse signaling network of EGFR, HER2, HER3 and HER4 tyrosine kinase receptors and the consequences for therapeutic approaches. Histol Histopathol. 2005;20:1005–1015. doi: 10.14670/HH-20.1005. [DOI] [PubMed] [Google Scholar]
- 18.Traxler P. Tyrosine kinases as targets in cancer therapy - successes and failures. Expert Opin Ther Targets. 2003;7:215–234. doi: 10.1517/14728222.7.2.215. [DOI] [PubMed] [Google Scholar]
- 19.Fabian MA, Biggs WH 3rd, Treiber DK, Atteridge CE, Azimioara MD, Benedetti MG, Carter TA, Ciceri P, Edeen PT, Floyd M, Ford JM, Galvin M, Gerlach JL, Grotzfeld RM, Herrgard S, Insko DE, Insko MA, Lai AG, Lelias JM, Mehta SA, Milanov ZV, Velasco AM, Wodicka LM, Patel HK, Zarrinkar PP, Lockhart DJ. A small molecule-kinase interaction map for clinical kinase inhibitors. Nat Biotechnol. 2005;23:329–336. doi: 10.1038/nbt1068. [DOI] [PubMed] [Google Scholar]
- 20.Hanks SK, Quinn AM, Hunter T. The protein kinase family: conserved features and deduced phylogeny of the catalytic domains. Science. 1988;241:42–52. doi: 10.1126/science.3291115. [DOI] [PubMed] [Google Scholar]
- 21.Adnane L, Trail PA, Taylor I, Wilhelm SM. Sorafenib (BAY 43-9006, Nexavar), a dual-action inhibitor that targets RAF/MEK/ERK pathway in tumor cells and tyrosine kinases VEGFR/PDGFR in tumor vasculature. Methods Enzymol. 2006;407:597–612. doi: 10.1016/S0076-6879(05)07047-3. [DOI] [PubMed] [Google Scholar]
- 22.Martinelli G, Soverini S, Rosti G, Baccarani M. Dual tyrosine kinase inhibitors in chronic myeloid leukemia. Leukemia. 2005;19:1872–1879. doi: 10.1038/sj.leu.2403950. [DOI] [PubMed] [Google Scholar]
- 23.Geyer CE, Forster J, Lindquist D, Chan S, Romieu CG, Pienkowski T, Jagiello-Gruszfeld A, Crown J, Chan A, Kaufman B, Skarlos D, Campone M, Davidson N, Berger M, Oliva C, Rubin SD, Stein S, Cameron D. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med. 2006;355:2733–2743. doi: 10.1056/NEJMoa064320. [DOI] [PubMed] [Google Scholar]
- 24.Blackwell KL, Burstein HJ, Storniolo AM, Rugo H, Sledge G, Koehler M, Ellis C, Casey M, Vukelja S, Bischoff J, Baselga J, O’Shaughnessy J. Randomized study of Lapatinib alone or in combination with trastuzumab in women with ErbB2-positive, trastuzumab-refractory metastatic breast cancer. J. Clin. Oncol. 2010;28:1124–1130. doi: 10.1200/JCO.2008.21.4437. [DOI] [PubMed] [Google Scholar]
- 25.Tevaarwerk AJ, Kolesar JM. Lapatinib: a small-molecule inhibitor of epidermal growth factor receptor and human epidermal growth factor receptor-2 tyrosine kinases used in the treatment of breast cancer. Clin Ther. 2009;31 Pt 2:2332–2348. doi: 10.1016/j.clinthera.2009.11.029. [DOI] [PubMed] [Google Scholar]
- 26.Johnston S, Pippen J Jr, Pivot X, Lichinitser M, Sadeghi S, Dieras V, Gomez HL, Romieu G, Manikhas A, Kennedy MJ, Press MF, Maltzman J, Florance A, O’Rourke L, Oliva C, Stein S, Pegram M. Lapatinib combined with letrozole versus letrozole and placebo as first-line therapy for postmenopausal hormone receptor-positive metastatic breast cancer. J. Clin. Oncol. 2009;27:5538–5546. doi: 10.1200/JCO.2009.23.3734. [DOI] [PubMed] [Google Scholar]
- 27.Rusnak DW, Lackey K, Affleck K, Wood ER, Alligood KJ, Rhodes N, Keith BR, Murray DM, Knight WB, Mullin RJ, Gilmer TM. The effects of the novel, reversible epidermal growth factor receptor/ErbB-2 tyrosine kinase inhibitor, GW2016, on the growth of human normal and tumor-derived cell lines in vitro and in vivo. Mol Cancer Ther. 2001;1:85–94. [PubMed] [Google Scholar]
- 28.Xia W, Mullin RJ, Keith BR, Liu LH, Ma H, Rusnak DW, Owens G, Alligood KJ, Spector NL. Anti-tumor activity of GW572016: a dual tyrosine kinase inhibitor blocks EGF activation of EGFR/erbB2 and downstream Erk1/2 and AKT pathways. Oncogene. 2002;21:6255–6263. doi: 10.1038/sj.onc.1205794. [DOI] [PubMed] [Google Scholar]
- 29.Wainberg ZA, Anghel A, Desai AJ, Ayala R, Luo T, Safran B, Fejzo MS, Hecht JR, Slamon DJ, Finn RS. Lapatinib, a dual EGFR and HER2 kinase inhibitor, selectively inhibits HER2-amplified human gastric cancer cells and is synergistic with trastuzumab in vitro and in vivo. Clin Cancer Res. 2010;16:1509–1519. doi: 10.1158/1078-0432.CCR-09-1112. [DOI] [PubMed] [Google Scholar]
- 30.Gril B, Palmieri D, Bronder JL, Herring JM, Vega-Valle E, Feigenbaum L, Liewehr DJ, Steinberg SM, Merino MJ, Rubin SD, Steeg PS. Effect of lapatinib on the outgrowth of metastatic breast cancer cells to the brain. J Natl Cancer Inst. 2008;100:1092–1103. doi: 10.1093/jnci/djn216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Konecny GE, Pegram MD, Venkatesan N, Finn R, Yang G, Rahmeh M, Untch M, Rusnak DW, Spehar G, Mullin RJ, Keith BR, Gilmer TM, Berger M, Podratz KC, Slamon DJ. Activity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells. Cancer Res. 2006;66:1630–1639. doi: 10.1158/0008-5472.CAN-05-1182. [DOI] [PubMed] [Google Scholar]
- 32.Zhang D, Pal A, Bornmann WG, Yamasaki F, Esteva FJ, Hortobagyi GN, Bartholomeusz C, Ueno NT. Activity of lapatinib is independent of EGFR expression level in HER2-overexpressing breast cancer cells. Mol Cancer Ther. 2008;7:1846–1850. doi: 10.1158/1535-7163.MCT-08-0168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Tanizaki J, Okamoto I, Fumita S, Okamoto W, Nishio K, Nakagawa K. Roles of BIM induction and survivin downregulation in lapatinibinduced apoptosis in breast cancer cells with HER2 amplification. Oncogene. 2011;30:4097–4106. doi: 10.1038/onc.2011.111. [DOI] [PubMed] [Google Scholar]
- 34.Ryan BM, Konecny GE, Kahlert S, Wang HJ, Untch M, Meng G, Pegram MD, Podratz KC, Crown J, Slamon DJ, Duffy MJ. Survivin expression in breast cancer predicts clinical outcome and is associated with HER2, VEGF, urokinase plasminogen activator and PAI-1. Ann Oncol. 2006;17:597–604. doi: 10.1093/annonc/mdj121. [DOI] [PubMed] [Google Scholar]
- 35.Jha K, Shukla M, Pandey M. Survivin expression and targeting in breast cancer. Surg Oncol. 2012;21:125–131. doi: 10.1016/j.suronc.2011.01.001. [DOI] [PubMed] [Google Scholar]
- 36.Liu L, Greger J, Shi H, Liu Y, Greshock J, Annan R, Halsey W, Sathe GM, Martin AM, Gilmer TM. Novel mechanism of lapatinib resistance in HER2-positive breast tumor cells: activation of AXL. Cancer Res. 2009;69:6871–6878. doi: 10.1158/0008-5472.CAN-08-4490. [DOI] [PubMed] [Google Scholar]
- 37.Bendell JC, Domchek SM, Burstein HJ, Harris L, Younger J, Kuter I, Bunnell C, Rue M, Gelman R, Winer E. Central nervous system metastases in women who receive trastuzumabbased therapy for metastatic breast carcinoma. Cancer. 2003;97:2972–2977. doi: 10.1002/cncr.11436. [DOI] [PubMed] [Google Scholar]
- 38.Ono M, Ando M, Yunokawa M, Nakano E, Yonemori K, Matsumoto K, Kouno T, Shimizu C, Tamura K, Katsumata N, Fujiwara Y. Brain metastases in patients who receive trastuzumab-containing chemotherapy for HER2-overexpressing metastatic breast cancer. Int J Clin Oncol. 2009;14:48–52. doi: 10.1007/s10147-008-0797-8. [DOI] [PubMed] [Google Scholar]
- 39.Arribas J, Baselga J, Pedersen K, Parra-Palau JL. p95HER2 and breast cancer. Cancer Res. 2011;71:1515–1519. doi: 10.1158/0008-5472.CAN-10-3795. [DOI] [PubMed] [Google Scholar]
- 40.Saez R, Molina MA, Ramsey EE, Rojo F, Keenan EJ, Albanell J, Lluch A, Garcia-Conde J, Baselga J, Clinton GM. p95HER-2 predicts worse outcome in patients with HER-2-positive breast cancer. Clin Cancer Res. 2006;12:424–431. doi: 10.1158/1078-0432.CCR-05-1807. [DOI] [PubMed] [Google Scholar]
- 41.Scaltriti M, Rojo F, Ocana A, Anido J, Guzman M, Cortes J, Di Cosimo S, Matias-Guiu X, Ramon y Cajal S, Arribas J, Baselga J. Expression of p95HER2, a truncated form of the HER2 receptor, and response to anti-HER2 therapies in breast cancer. J Natl Cancer Inst. 2007;99:628–638. doi: 10.1093/jnci/djk134. [DOI] [PubMed] [Google Scholar]
- 42.Nahta R, Yuan LX, Du Y, Esteva FJ. Lapatinib induces apoptosis in trastuzumab-resistant breast cancer cells: effects on insulin-like growth factor I signaling. Mol Cancer Ther. 2007;6:667–674. doi: 10.1158/1535-7163.MCT-06-0423. [DOI] [PubMed] [Google Scholar]
- 43.Wakeling AE, Barker AJ, Davies DH, Brown DS, Green LR, Cartlidge SA, Woodburn JR. Specific inhibition of epidermal growth factor receptor tyrosine kinase by 4-anilinoquinazolines. Breast Cancer Res Treat. 1996;38:67–73. doi: 10.1007/BF01803785. [DOI] [PubMed] [Google Scholar]
- 44.Ma C, Zuo W, Wang X, Wei L, Guo Q, Song X. Lapatinib inhibits the activation of NFkappaB through reducing phosphorylation of IkappaB-alpha in breast cancer cells. Oncol Rep. 2013;29:812–818. doi: 10.3892/or.2012.2159. [DOI] [PubMed] [Google Scholar]
- 45.Ahmed KM, Cao N, Li JJ. HER-2 and NFkappaB as the targets for therapy-resistant breast cancer. Anticancer Res. 2006;26:4235–4243. [PMC free article] [PubMed] [Google Scholar]
- 46.Sartor CI. Epidermal growth factor family receptors and inhibitors: radiation response modulators. Semin Radiat Oncol. 2003;13:22–30. doi: 10.1053/srao.2003.50003. [DOI] [PubMed] [Google Scholar]
- 47.Tanaka T, Munshi A, Brooks C, Liu J, Hobbs ML, Meyn RE. Gefitinib radiosensitizes nonsmall cell lung cancer cells by suppressing cellular DNA repair capacity. Clin Cancer Res. 2008;14:1266–1273. doi: 10.1158/1078-0432.CCR-07-1606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Ochs JS. Rationale and clinical basis for combining gefitinib (IRESSA, ZD1839) with radiation therapy for solid tumors. Int J Radiat Oncol Biol Phys. 2004;58:941–949. doi: 10.1016/j.ijrobp.2003.09.094. [DOI] [PubMed] [Google Scholar]
- 49.Zhou H, Kim YS, Peletier A, McCall W, Earp HS, Sartor CI. Effects of the EGFR/HER2 kinase inhibitor GW572016 on EGFR- and HER2-overexpressing breast cancer cell line proliferation, radiosensitization, and resistance. Int J Radiat Oncol Biol Phys. 2004;58:344–352. doi: 10.1016/j.ijrobp.2003.09.046. [DOI] [PubMed] [Google Scholar]
- 50.Sambade MJ, Camp JT, Kimple RJ, Sartor CI, Shields JM. Mechanism of lapatinib-mediated radiosensitization of breast cancer cells is primarily by inhibition of the Raf>MEK>ERK mitogen-activated protein kinase cascade and radiosensitization of lapatinib-resistant cells restored by direct inhibition of MEK. Radiother Oncol. 2009;93:639–644. doi: 10.1016/j.radonc.2009.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Bhosle J, Kiakos K, Porter AC, Wu J, Makris A, Hartley JA, Hochhauser D. Treatment with gefitinib or lapatinib induces drug resistance through downregulation of topoisomerase IIalpha expression. Mol Cancer Ther. 2013;12:2897–2908. doi: 10.1158/1535-7163.MCT-12-1049. [DOI] [PubMed] [Google Scholar]
- 52.Fisher JN, Terao M, Fratelli M, Kurosaki M, Paroni G, Zanetti A, Gianni M, Bolis M, Lupi M, Tsykin A, Goodall GJ, Garattini E. MicroRNA networks regulated by all-trans retinoic acid and Lapatinib control the growth, survival and motility of breast cancer cells. Oncotarget. 2015;6:13176–200. doi: 10.18632/oncotarget.3759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Johnston S, Trudeau M, Kaufman B, Boussen H, Blackwell K, LoRusso P, Lombardi DP, Ben Ahmed S, Citrin DL, DeSilvio ML, Harris J, Westlund RE, Salazar V, Zaks TZ, Spector NL. Phase II study of predictive biomarker profiles for response targeting human epidermal growth factor receptor 2 (HER-2) in advanced inflammatory breast cancer with lapatinib monotherapy. J. Clin. Oncol. 2008;26:1066–1072. doi: 10.1200/JCO.2007.13.9949. [DOI] [PubMed] [Google Scholar]
- 54.Burstein HJ, Storniolo AM, Franco S, Forster J, Stein S, Rubin S, Salazar VM, Blackwell KL. A phase II study of lapatinib monotherapy in chemotherapy-refractory HER2-positive and HER2-negative advanced or metastatic breast cancer. Ann Oncol. 2008;19:1068–1074. doi: 10.1093/annonc/mdm601. [DOI] [PubMed] [Google Scholar]
- 55.Toi M, Iwata H, Fujiwara Y, Ito Y, Nakamura S, Tokuda Y, Taguchi T, Rai Y, Aogi K, Arai T, Watanabe J, Wakamatsu T, Katsura K, Ellis CE, Gagnon RC, Allen KE, Sasaki Y, Takashima S. Lapatinib monotherapy in patients with relapsed, advanced, or metastatic breast cancer: efficacy, safety, and biomarker results from Japanese patients phase II studies. Br J Cancer. 2009;101:1676–1682. doi: 10.1038/sj.bjc.6605343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Lin NU, Carey LA, Liu MC, Younger J, Come SE, Ewend M, Harris GJ, Bullitt E, Van den Abbeele AD, Henson JW, Li X, Gelman R, Burstein HJ, Kasparian E, Kirsch DG, Crawford A, Hochberg F, Winer EP. Phase II trial of lapatinib for brain metastases in patients with human epidermal growth factor receptor 2-positive breast cancer. J. Clin. Oncol. 2008;26:1993–1999. doi: 10.1200/JCO.2007.12.3588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Bachelot T, Romieu G, Campone M, Dieras V, Cropet C, Dalenc F, Jimenez M, Le Rhun E, Pierga JY, Goncalves A, Leheurteur M, Domont J, Gutierrez M, Cure H, Ferrero JM, Labbe-Devilliers C. Lapatinib plus capecitabine in patients with previously untreated brain metastases from HER2-positive metastatic breast cancer (LANDSCAPE): a single-group phase 2 study. Lancet Oncol. 2013;14:64–71. doi: 10.1016/S1470-2045(12)70432-1. [DOI] [PubMed] [Google Scholar]
- 58.Lin NU, Eierman W, Greil R, Campone M, Kaufman B, Steplewski K, Lane SR, Zembryki D, Rubin SD, Winer EP. Randomized phase II study of lapatinib plus capecitabine or lapatinib plus topotecan for patients with HER2-positive breast cancer brain metastases. J Neurooncol. 2011;105:613–620. doi: 10.1007/s11060-011-0629-y. [DOI] [PubMed] [Google Scholar]
- 59.Lin NU, Dieras V, Paul D, Lossignol D, Christodoulou C, Stemmler HJ, Roche H, Liu MC, Greil R, Ciruelos E, Loibl S, Gori S, Wardley A, Yardley D, Brufsky A, Blum JL, Rubin SD, Dharan B, Steplewski K, Zembryki D, Oliva C, Roychowdhury D, Paoletti P, Winer EP. Multicenter phase II study of lapatinib in patients with brain metastases from HER2-positive breast cancer. Clin Cancer Res. 2009;15:1452–1459. doi: 10.1158/1078-0432.CCR-08-1080. [DOI] [PubMed] [Google Scholar]
- 60.Sierra JR, Cepero V, Giordano S. Molecular mechanisms of acquired resistance to tyrosine kinase targeted therapy. Mol Cancer. 2010;9:75. doi: 10.1186/1476-4598-9-75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Brugge J, Hung MC, Mills GB. A new mutational AKTivation in the PI3K pathway. Cancer Cell. 2007;12:104–107. doi: 10.1016/j.ccr.2007.07.014. [DOI] [PubMed] [Google Scholar]
- 62.Coughlin CM, Johnston DS, Strahs A, Burczynski ME, Bacus S, Hill J, Feingold JM, Zacharchuk C, Berkenblit A. Approaches and limitations of phosphatidylinositol-3-kinase pathway activation status as a predictive biomarker in the clinical development of targeted therapy. Breast Cancer Res Treat. 2010;124:1–11. doi: 10.1007/s10549-010-1108-4. [DOI] [PubMed] [Google Scholar]
- 63.Saal LH, Holm K, Maurer M, Memeo L, Su T, Wang X, Yu JS, Malmstrom PO, Mansukhani M, Enoksson J, Hibshoosh H, Borg A, Parsons R. PIK3CA mutations correlate with hormone receptors, node metastasis, and ERBB2, and are mutually exclusive with PTEN loss in human breast carcinoma. Cancer Res. 2005;65:2554–2559. doi: 10.1158/0008-5472-CAN-04-3913. [DOI] [PubMed] [Google Scholar]
- 64.Stemke-Hale K, Gonzalez-Angulo AM, Lluch A, Neve RM, Kuo WL, Davies M, Carey M, Hu Z, Guan Y, Sahin A, Symmans WF, Pusztai L, Nolden LK, Horlings H, Berns K, Hung MC, van de Vijver MJ, Valero V, Gray JW, Bernards R, Mills GB, Hennessy BT. An integrative genomic and proteomic analysis of PIK3CA, PTEN, and AKT mutations in breast cancer. Cancer Res. 2008;68:6084–6091. doi: 10.1158/0008-5472.CAN-07-6854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Eichhorn PJ, Gili M, Scaltriti M, Serra V, Guzman M, Nijkamp W, Beijersbergen RL, Valero V, Seoane J, Bernards R, Baselga J. Phosphatidylinositol 3-kinase hyperactivation results in lapatinib resistance that is reversed by the mTOR/phosphatidylinositol 3-kinase inhibitor NVP-BEZ235. Cancer Res. 2008;68:9221–9230. doi: 10.1158/0008-5472.CAN-08-1740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Barton S, Starling N, Swanton C. Predictive molecular markers of response to epidermal growth factor receptor(EGFR) family-targeted therapies. Curr Cancer Drug Targets. 2010;10:799–812. doi: 10.2174/156800910793357925. [DOI] [PubMed] [Google Scholar]
- 67.Isakoff SJ, Engelman JA, Irie HY, Luo J, Brachmann SM, Pearline RV, Cantley LC, Brugge JS. Breast cancer-associated PIK3CA mutations are oncogenic in mammary epithelial cells. Cancer Res. 2005;65:10992–11000. doi: 10.1158/0008-5472.CAN-05-2612. [DOI] [PubMed] [Google Scholar]
- 68.Wang L, Zhang Q, Zhang J, Sun S, Guo H, Jia Z, Wang B, Shao Z, Wang Z, Hu X. PI3K pathway activation results in low efficacy of both trastuzumab and lapatinib. BMC Cancer. 2011;11:248. doi: 10.1186/1471-2407-11-248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.O’Brien NA, Browne BC, Chow L, Wang Y, Ginther C, Arboleda J, Duffy MJ, Crown J, O’Donovan N, Slamon DJ. Activated phosphoinositide 3-kinase/AKT signaling confers resistance to trastuzumab but not lapatinib. Mol Cancer Ther. 2010;9:1489–1502. doi: 10.1158/1535-7163.MCT-09-1171. [DOI] [PubMed] [Google Scholar]
- 70.Xia W, Husain I, Liu L, Bacus S, Saini S, Spohn J, Pry K, Westlund R, Stein SH, Spector NL. Lapatinib antitumor activity is not dependent upon phosphatase and tensin homologue deleted on chromosome 10 in ErbB2-overexpressing breast cancers. Cancer Res. 2007;67:1170–1175. doi: 10.1158/0008-5472.CAN-06-2101. [DOI] [PubMed] [Google Scholar]
- 71.Karakashev SV, Reginato MJ. Hypoxia/HIF1alpha induces lapatinib resistance in ERBB2-positive breast cancer cells via regulation of DUSP2. Oncotarget. 2015;6:1967–1980. doi: 10.18632/oncotarget.2806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Vazquez-Martin A, Oliveras-Ferraros C, Menendez JA. Autophagy facilitates the development of breast cancer resistance to the anti-HER2 monoclonal antibody trastuzumab. PLoS One. 2009;4:e6251. doi: 10.1371/journal.pone.0006251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Chen S, Li X, Feng J, Chang Y, Wang Z, Wen A. Autophagy facilitates the Lapatinib resistance of HER2 positive breast cancer cells. Med Hypotheses. 2011;77:206–208. doi: 10.1016/j.mehy.2011.04.013. [DOI] [PubMed] [Google Scholar]
- 74.Xia W, Petricoin EF 3rd, Zhao S, Liu L, Osada T, Cheng Q, Wulfkuhle JD, Gwin WR, Yang X, Gallagher RI, Bacus S, Lyerly HK, Spector NL. An heregulin-EGFR-HER3 autocrine signaling axis can mediate acquired lapatinib resistance in HER2+ breast cancer models. Breast Cancer Res. 2013;15:R85. doi: 10.1186/bcr3480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kristiansen G, Winzer KJ, Mayordomo E, Bellach J, Schluns K, Denkert C, Dahl E, Pilarsky C, Altevogt P, Guski H, Dietel M. CD24 expression is a new prognostic marker in breast cancer. Clin Cancer Res. 2003;9:4906–4913. [PubMed] [Google Scholar]
- 76.Hosonaga M, Arima Y, Sugihara E, Kohno N, Saya H. Expression of CD24 is associated with HER2 expression and supports HER2-Akt signaling in HER2-positive breast cancer cells. Cancer Sci. 2014;105:779–787. doi: 10.1111/cas.12427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Rani S, Corcoran C, Shiels L, Germano S, Breslin S, Madden S, McDermott MS, Browne BC, O’Donovan N, Crown J, Gogarty M, Byrne AT, O’Driscoll L. Neuromedin U: a candidate biomarker and therapeutic target to predict and overcome resistance to HER-tyrosine kinase inhibitors. Cancer Res. 2014;74:3821–3833. doi: 10.1158/0008-5472.CAN-13-2053. [DOI] [PubMed] [Google Scholar]
- 78.Xia W, Bacus S, Hegde P, Husain I, Strum J, Liu L, Paulazzo G, Lyass L, Trusk P, Hill J, Harris J, Spector NL. A model of acquired autoresistance to a potent ErbB2 tyrosine kinase inhibitor and a therapeutic strategy to prevent its onset in breast cancer. Proc Natl Acad Sci U S A. 2006;103:7795–7800. doi: 10.1073/pnas.0602468103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Lee SY, Meier R, Furuta S, Lenburg ME, Kenny PA, Xu R, Bissell MJ. FAM83A confers EGFR-TKI resistance in breast cancer cells and in mice. J Clin Invest. 2012;122:3211–3220. doi: 10.1172/JCI60498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Wang Q, Quan H, Zhao J, Xie C, Wang L, Lou L. RON confers lapatinib resistance in HER2-positive breast cancer cells. Cancer Lett. 2013;340:43–50. doi: 10.1016/j.canlet.2013.06.022. [DOI] [PubMed] [Google Scholar]
- 81.Azuma K, Tsurutani J, Sakai K, Kaneda H, Fujisaka Y, Takeda M, Watatani M, Arao T, Satoh T, Okamoto I, Kurata T, Nishio K, Nakagawa K. Switching addictions between HER2 and FGFR2 in HER2-positive breast tumor cells: FGFR2 as a potential target for salvage after lapatinib failure. Biochem Biophys Res Commun. 2011;407:219–224. doi: 10.1016/j.bbrc.2011.03.002. [DOI] [PubMed] [Google Scholar]
- 82.Park SH, Ito K, Olcott W, Katsyv I, Halstead-Nussloch G, Irie HY. PTK6 inhibition promotes apoptosis of Lapatinib-resistant Her2 breast cancer cells by inducing Bim. Breast Cancer Res. 2015;17:86. doi: 10.1186/s13058-015-0594-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Formisano L, Nappi L, Rosa R, Marciano R, D’Amato C, D’Amato V, Damiano V, Raimondo L, Iommelli F, Scorziello A, Troncone G, Veneziani B, Parsons SJ, De Placido S, Bianco R. Epidermal growth factor-receptor activation modulates Src-dependent resistance to lapatinib in breast cancer models. Breast Cancer Res. 2014;16:R45. doi: 10.1186/bcr3650. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 84.De Luca A, D’Alessio A, Gallo M, Maiello MR, Bode AM, Normanno N. Src and CXCR4 are involved in the invasiveness of breast cancer cells with acquired resistance to lapatinib. Cell Cycle. 2014;13:148–156. doi: 10.4161/cc.26899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Rexer BN, Ham AJ, Rinehart C, Hill S, Granja-Ingram Nde M, Gonzalez-Angulo AM, Mills GB, Dave B, Chang JC, Liebler DC, Arteaga CL. Phosphoproteomic mass spectrometry profiling links Src family kinases to escape from HER2 tyrosine kinase inhibition. Oncogene. 2011;30:4163–4174. doi: 10.1038/onc.2011.130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Stuhlmiller TJ, Miller SM, Zawistowski JS, Nakamura K, Beltran AS, Duncan JS, Angus SP, Collins KA, Granger DA, Reuther RA, Graves LM, Gomez SM, Kuan PF, Parker JS, Chen X, Sciaky N, Carey LA, Earp HS, Jin J, Johnson GL. Inhibition of Lapatinib-Induced Kinome Reprogramming in ERBB2-Positive Breast Cancer by Targeting BET Family Bromodomains. Cell Rep. 2015;11:390–404. doi: 10.1016/j.celrep.2015.03.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Corcoran C, Rani S, Breslin S, Gogarty M, Ghobrial IM, Crown J, O’Driscoll L. miR-630 targets IGF1R to regulate response to HERtargeting drugs and overall cancer cell progression in HER2 over-expressing breast cancer. Mol Cancer. 2014;13:71. doi: 10.1186/1476-4598-13-71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Moody SE, Schinzel AC, Singh S, Izzo F, Strickland MR, Luo L, Thomas SR, Boehm JS, Kim SY, Wang ZC, Hahn WC. PRKACA mediates resistance to HER2-targeted therapy in breast cancer cells and restores anti-apoptotic signaling. Oncogene. 2015;34:2061–71. doi: 10.1038/onc.2014.153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Chen CJ, Lin SE, Lin YM, Lin SH, Chen DR, Chen CL. Association of expression of kruppellike factor 4 and kruppel-like factor 5 with the clinical manifestations of breast cancer. Pathol Oncol Res. 2012;18:161–168. doi: 10.1007/s12253-011-9422-7. [DOI] [PubMed] [Google Scholar]
- 90.Tong D, Czerwenka K, Heinze G, Ryffel M, Schuster E, Witt A, Leodolter S, Zeillinger R. Expression of KLF5 is a prognostic factor for disease-free survival and overall survival in patients with breast cancer. Clin Cancer Res. 2006;12:2442–2448. doi: 10.1158/1078-0432.CCR-05-0964. [DOI] [PubMed] [Google Scholar]
- 91.Farrugia MK, Sharma SB, Lin CC, McLaughlin SL, Vanderbilt DB, Ammer AG, Salkeni MA, Stoilov P, Agazie YM, Creighton CJ, Ruppert JM. Regulation of anti-apoptotic signaling by Kruppel-like factors 4 and 5 mediates lapatinib resistance in breast cancer. Cell Death Dis. 2015;6:e1699. doi: 10.1038/cddis.2015.65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Bekki K, Vogel H, Li W, Ito T, Sweeney C, Haarmann-Stemmann T, Matsumura F, Vogel CF. The aryl hydrocarbon receptor (AhR) mediates resistance to apoptosis induced in breast cancer cells. Pestic Biochem Physiol. 2015;120:5–13. doi: 10.1016/j.pestbp.2014.12.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Mittendorf EA, Wu Y, Scaltriti M, Meric-Bernstam F, Hunt KK, Dawood S, Esteva FJ, Buzdar AU, Chen H, Eksambi S, Hortobagyi GN, Baselga J, Gonzalez-Angulo AM. Loss of HER2 amplification following trastuzumabbased neoadjuvant systemic therapy and survival outcomes. Clin Cancer Res. 2009;15:7381–7388. doi: 10.1158/1078-0432.CCR-09-1735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Garrett JT, Olivares MG, Rinehart C, Granja-Ingram ND, Sanchez V, Chakrabarty A, Dave B, Cook RS, Pao W, McKinely E, Manning HC, Chang J, Arteaga CL. Transcriptional and posttranslational up-regulation of HER3 (ErbB3) compensates for inhibition of the HER2 tyrosine kinase. Proc Natl Acad Sci U S A. 2011;108:5021–5026. doi: 10.1073/pnas.1016140108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Amin DN, Sergina N, Ahuja D, McMahon M, Blair JA, Wang D, Hann B, Koch KM, Shokat KM, Moasser MM. Resiliency and vulnerability in the HER2-HER3 tumorigenic driver. Sci Transl Med. 2010;2:16ra7. doi: 10.1126/scitranslmed.3000389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Berardi R, Santoni M, Morgese F, Ballatore Z, Savini A, Onofri A, Mazzanti P, Pistelli M, Pierantoni C, De Lisa M, Caramanti M, Pagliaretta S, Pellei C, Cascinu S. Novel small molecule EGFR inhibitors as candidate drugs in non-small cell lung cancer. Onco Targets Ther. 2013;6:563–576. doi: 10.2147/OTT.S28155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Esteva FJ. Monoclonal antibodies, small molecules, and vaccines in the treatment of breast cancer. Oncologist. 2004;9(Suppl 3):4–9. doi: 10.1634/theoncologist.9-suppl_3-4. [DOI] [PubMed] [Google Scholar]
- 98.Sainsbury JR, Farndon JR, Harris AL, Sherbet GV. Epidermal growth factor receptors on human breast cancers. Br J Surg. 1985;72:186–188. doi: 10.1002/bjs.1800720309. [DOI] [PubMed] [Google Scholar]
- 99.Ferrero JM, Ramaioli A, Largillier R, Formento JL, Francoual M, Ettore F, Namer M, Milano G. Epidermal growth factor receptor expression in 780 breast cancer patients: a reappraisal of the prognostic value based on an eight-year median follow-up. Ann Oncol. 2001;12:841–846. doi: 10.1023/a:1011183421477. [DOI] [PubMed] [Google Scholar]
- 100.Harris AL, Nicholson S, Sainsbury JR, Farndon J, Wright C. Epidermal growth factor receptors in breast cancer: association with early relapse and death, poor response to hormones and interactions with neu. J Steroid Biochem. 1989;34:123–131. doi: 10.1016/0022-4731(89)90072-1. [DOI] [PubMed] [Google Scholar]
- 101.Moulder SL, Yakes FM, Muthuswamy SK, Bianco R, Simpson JF, Arteaga CL. Epidermal growth factor receptor (HER1) tyrosine kinase inhibitor ZD1839 (Iressa) inhibits HER2/neu (erbB2)-overexpressing breast cancer cells in vitro and in vivo. Cancer Res. 2001;61:8887–8895. [PubMed] [Google Scholar]
- 102.Anido J, Matar P, Albanell J, Guzman M, Rojo F, Arribas J, Averbuch S, Baselga J. ZD1839, a specific epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, induces the formation of inactive EGFR/HER2 and EGFR/HER3 heterodimers and prevents heregulin signaling in HER2-overexpressing breast cancer cells. Clin Cancer Res. 2003;9:1274–1283. [PubMed] [Google Scholar]
- 103.Campiglio M, Locatelli A, Olgiati C, Normanno N, Somenzi G, Vigano L, Fumagalli M, Menard S, Gianni L. Inhibition of proliferation and induction of apoptosis in breast cancer cells by the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor ZD1839 (‘Iressa’) is independent of EGFR expression level. J Cell Physiol. 2004;198:259–268. doi: 10.1002/jcp.10411. [DOI] [PubMed] [Google Scholar]
- 104.Moasser MM, Basso A, Averbuch SD, Rosen N. The tyrosine kinase inhibitor ZD1839 (“Iressa”) inhibits HER2-driven signaling and suppresses the growth of HER2-overexpressing tumor cells. Cancer Res. 2001;61:7184–7188. [PubMed] [Google Scholar]
- 105.Krol J, Francis RE, Albergaria A, Sunters A, Polychronis A, Coombes RC, Lam EW. The transcription factor FOXO3a is a crucial cellular target of gefitinib (Iressa) in breast cancer cells. Mol Cancer Ther. 2007;6:3169–3179. doi: 10.1158/1535-7163.MCT-07-0507. [DOI] [PubMed] [Google Scholar]
- 106.Segovia-Mendoza M, Diaz L, Gonzalez-Gonzalez ME, Martinez-Reza I, Garcia-Quiroz J, Prado-Garcia H, Ibarra-Sanchez MJ, Esparza-Lopez J, Larrea F, Garcia-Becerra R. Calcitriol and its analogues enhance the antiproliferative activity of gefitinib in breast cancer cells. J Steroid Biochem Mol Biol. 2015;148:122–131. doi: 10.1016/j.jsbmb.2014.12.006. [DOI] [PubMed] [Google Scholar]
- 107.Dragowska WH, Weppler SA, Wang JC, Wong LY, Kapanen AI, Rawji JS, Warburton C, Qadir MA, Donohue E, Roberge M, Gorski SM, Gelmon KA, Bally MB. Induction of autophagy is an early response to gefitinib and a potential therapeutic target in breast cancer. PLoS One. 2013;8:e76503. doi: 10.1371/journal.pone.0076503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Baselga J, Albanell J, Ruiz A, Lluch A, Gascon P, Guillem V, Gonzalez S, Sauleda S, Marimon I, Tabernero JM, Koehler MT, Rojo F. Phase II and tumor pharmacodynamic study of gefitinib in patients with advanced breast cancer. J. Clin. Oncol. 2005;23:5323–5333. doi: 10.1200/JCO.2005.08.326. [DOI] [PubMed] [Google Scholar]
- 109.Arteaga CL, O’Neill A, Moulder SL, Pins M, Sparano JA, Sledge GW, Davidson NE. A phase I-II study of combined blockade of the ErbB receptor network with trastuzumab and gefitinib in patients with HER2 (ErbB2)-overexpressing metastatic breast cancer. Clin Cancer Res. 2008;14:6277–6283. doi: 10.1158/1078-0432.CCR-08-0482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Somlo G, Martel CL, Lau SK, Frankel P, Ruel C, Gu L, Hurria A, Chung C, Luu T, Morgan R Jr, Leong L, Koczywas M, McNamara M, Russell CA, Kane SE. A phase I/II prospective, single arm trial of gefitinib, trastuzumab, and docetaxel in patients with stage IV HER-2 positive metastatic breast cancer. Breast Cancer Res Treat. 2012;131:899–906. doi: 10.1007/s10549-011-1850-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.von Minckwitz G, Jonat W, Fasching P, du Bois A, Kleeberg U, Luck HJ, Kettner E, Hilfrich J, Eiermann W, Torode J, Schneeweiss A. A multicentre phase II study on gefitinib in taxane- and anthracycline-pretreated metastatic breast cancer. Breast Cancer Res Treat. 2005;89:165–172. doi: 10.1007/s10549-004-1720-2. [DOI] [PubMed] [Google Scholar]
- 112.Smith IE, Walsh G, Skene A, Llombart A, Mayordomo JI, Detre S, Salter J, Clark E, Magill P, Dowsett M. A phase II placebo-controlled trial of neoadjuvant anastrozole alone or with gefitinib in early breast cancer. J. Clin. Oncol. 2007;25:3816–3822. doi: 10.1200/JCO.2006.09.6578. [DOI] [PubMed] [Google Scholar]
- 113.Massarweh S, Tham YL, Huang J, Sexton K, Weiss H, Tsimelzon A, Beyer A, Rimawi M, Cai WY, Hilsenbeck S, Fuqua S, Elledge R. A phase II neoadjuvant trial of anastrozole, fulvestrant, and gefitinib in patients with newly diagnosed estrogen receptor positive breast cancer. Breast Cancer Res Treat. 2011;129:819–827. doi: 10.1007/s10549-011-1679-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Guarneri V, Frassoldati A, Ficarra G, Puglisi F, Andreetta C, Michelotti A, Cresti N, Boni C, Bisagni G, Berardi R, Battelli N, Santoro A, Banna G, Bottini A, Di Blasio B, Maiorana A, Piacentini F, Giovannelli S, Jovic G, Conte P. Phase II, randomized trial of preoperative epirubicin-paclitaxel +/- gefitinib with biomarker evaluation in operable breast cancer. Breast Cancer Res Treat. 2008;110:127–134. doi: 10.1007/s10549-007-9688-3. [DOI] [PubMed] [Google Scholar]
- 115.Engebraaten O, Edvardsen H, Lokkevik E, Naume B, Kristensen V, Ottestad L, Natarajan V. Gefitinib in Combination with Weekly Docetaxel in Patients with Metastatic Breast Cancer Caused Unexpected Toxicity: Results from a Randomized Phase II Clinical Trial. ISRN Oncol. 2012;2012:176789. doi: 10.5402/2012/176789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Polychronis A, Sinnett HD, Hadjiminas D, Singhal H, Mansi JL, Shivapatham D, Shousha S, Jiang J, Peston D, Barrett N, Vigushin D, Morrison K, Beresford E, Ali S, Slade MJ, Coombes RC. Preoperative gefitinib versus gefitinib and anastrozole in postmenopausal patients with oestrogen-receptor positive and epidermal-growth-factor-receptor-positive primary breast cancer: a double-blind placebocontrolled phase II randomised trial. Lancet Oncol. 2005;6:383–391. doi: 10.1016/S1470-2045(05)70176-5. [DOI] [PubMed] [Google Scholar]
- 117.Ciardiello F, Troiani T, Caputo F, De Laurentiis M, Tortora G, Palmieri G, De Vita F, Diadema MR, Orditura M, Colantuoni G, Gridelli C, Catalano G, De Placido S, Bianco AR. Phase II study of gefitinib in combination with docetaxel as first-line therapy in metastatic breast cancer. Br J Cancer. 2006;94:1604–1609. doi: 10.1038/sj.bjc.6603141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Osborne CK, Neven P, Dirix LY, Mackey JR, Robert J, Underhill C, Schiff R, Gutierrez C, Migliaccio I, Anagnostou VK, Rimm DL, Magill P, Sellers M. Gefitinib or placebo in combination with tamoxifen in patients with hormone receptor-positive metastatic breast cancer: a randomized phase II study. Clin Cancer Res. 2011;17:1147–1159. doi: 10.1158/1078-0432.CCR-10-1869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Kalykaki A, Agelaki S, Kallergi G, Xyrafas A, Mavroudis D, Georgoulias V. Elimination of EGFR-expressing circulating tumor cells in patients with metastatic breast cancer treated with gefitinib. Cancer Chemother Pharmacol. 2014;73:685–693. doi: 10.1007/s00280-014-2387-y. [DOI] [PubMed] [Google Scholar]
- 120.Normanno N, De Luca A, Maiello MR, Campiglio M, Napolitano M, Mancino M, Carotenuto A, Viglietto G, Menard S. The MEK/MAPK pathway is involved in the resistance of breast cancer cells to the EGFR tyrosine kinase inhibitor gefitinib. J Cell Physiol. 2006;207:420–427. doi: 10.1002/jcp.20588. [DOI] [PubMed] [Google Scholar]
- 121.Normanno N, Campiglio M, Maiello MR, De Luca A, Mancino M, Gallo M, D’Alessio A, Menard S. Breast cancer cells with acquired resistance to the EGFR tyrosine kinase inhibitor gefitinib show persistent activation of MAPK signaling. Breast Cancer Res Treat. 2008;112:25–33. doi: 10.1007/s10549-007-9830-2. [DOI] [PubMed] [Google Scholar]
- 122.Ferrer-Soler L, Vazquez-Martin A, Brunet J, Menendez JA, De Llorens R, Colomer R. An update of the mechanisms of resistance to EGFR-tyrosine kinase inhibitors in breast cancer: Gefitinib (Iressa) -induced changes in the expression and nucleo-cytoplasmic trafficking of HER-ligands (Review) Int J Mol Med. 2007;20:3–10. [PubMed] [Google Scholar]
- 123.Sergina NV, Rausch M, Wang D, Blair J, Hann B, Shokat KM, Moasser MM. Escape from HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3. Nature. 2007;445:437–441. doi: 10.1038/nature05474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.She QB, Solit D, Basso A, Moasser MM. Resistance to gefitinib in PTEN-null HERoverexpressing tumor cells can be overcome through restoration of PTEN function or pharmacologic modulation of constitutive phosphatidylinositol 3’-kinase/Akt pathway signaling. Clin Cancer Res. 2003;9:4340–4346. [PubMed] [Google Scholar]
- 125.Bianco R, Shin I, Ritter CA, Yakes FM, Basso A, Rosen N, Tsurutani J, Dennis PA, Mills GB, Arteaga CL. Loss of PTEN/MMAC1/TEP in EGF receptor-expressing tumor cells counteracts the antitumor action of EGFR tyrosine kinase inhibitors. Oncogene. 2003;22:2812–2822. doi: 10.1038/sj.onc.1206388. [DOI] [PubMed] [Google Scholar]
- 126.Dijkers PF, Medema RH, Pals C, Banerji L, Thomas NS, Lam EW, Burgering BM, Raaijmakers JA, Lammers JW, Koenderman L, Coffer PJ. Forkhead transcription factor FKHR-L1 modulates cytokine-dependent transcriptional regulation of p27(KIP1) Mol Cell Biol. 2000;20:9138–9148. doi: 10.1128/mcb.20.24.9138-9148.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Mueller KL, Hunter LA, Ethier SP, Boerner JL. Met and c-Src cooperate to compensate for loss of epidermal growth factor receptor kinase activity in breast cancer cells. Cancer Res. 2008;68:3314–3322. doi: 10.1158/0008-5472.CAN-08-0132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Kuzumaki N, Suzuki A, Narita M, Hosoya T, Nagasawa A, Imai S, Yamamizu K, Morita H, Suzuki T, Okada Y, Okano HJ, Yamashita JK, Okano H. Multiple analyses of G-protein coupled receptor (GPCR) expression in the development of gefitinib-resistance in transforming non-small-cell lung cancer. PLoS One. 2012;7:e44368. doi: 10.1371/journal.pone.0044368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Pao W, Wang TY, Riely GJ, Miller VA, Pan Q, Ladanyi M, Zakowski MF, Heelan RT, Kris MG, Varmus HE. KRAS mutations and primary resistance of lung adenocarcinomas to gefitinib or erlotinib. PLoS Med. 2005;2:e17. doi: 10.1371/journal.pmed.0020017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Pao W, Miller V, Zakowski M, Doherty J, Politi K, Sarkaria I, Singh B, Heelan R, Rusch V, Fulton L, Mardis E, Kupfer D, Wilson R, Kris M, Varmus H. EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc Natl Acad Sci U S A. 2004;101:13306–13311. doi: 10.1073/pnas.0405220101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Hart S, Fischer OM, Prenzel N, Zwick-Wallasch E, Schneider M, Hennighausen L, Ullrich A. GPCR-induced migration of breast carcinoma cells depends on both EGFR signal transactivation and EGFR-independent pathways. Biol Chem. 2005;386:845–855. doi: 10.1515/BC.2005.099. [DOI] [PubMed] [Google Scholar]
- 132.Han C, Zou H, Ma J, Zhou Y, Zhao J. [Comparison of EGFR and KRAS status between primary non-small cell lung cancer and corresponding metastases: a systematic review and meta-analysis] . Zhongguo Fei Ai Za Zhi. 2010;13:882–891. doi: 10.3779/j.issn.1009-3419.2010.09.09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Han CB, Li F, Ma JT, Zou HW. Concordant KRAS mutations in primary and metastatic colorectal cancer tissue specimens: a metaanalysis and systematic review. Cancer Invest. 2012;30:741–747. doi: 10.3109/07357907.2012.732159. [DOI] [PubMed] [Google Scholar]
- 134.Santarpia L, Qi Y, Stemke-Hale K, Wang B, Young EJ, Booser DJ, Holmes FA, O’Shaughnessy J, Hellerstedt B, Pippen J, Vidaurre T, Gomez H, Valero V, Hortobagyi GN, Symmans WF, Bottai G, Di Leo A, Gonzalez-Angulo AM, Pusztai L. Mutation profiling identifies numerous rare drug targets and distinct mutation patterns in different clinical subtypes of breast cancers. Breast Cancer Res Treat. 2012;134:333–343. doi: 10.1007/s10549-012-2035-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Bhargava R, Gerald WL, Li AR, Pan Q, Lal P, Ladanyi M, Chen B. EGFR gene amplification in breast cancer: correlation with epidermal growth factor receptor mRNA and protein expression and HER-2 status and absence of EGFR-activating mutations. Mod Pathol. 2005;18:1027–1033. doi: 10.1038/modpathol.3800438. [DOI] [PubMed] [Google Scholar]
- 136.Rabindran SK, Discafani CM, Rosfjord EC, Baxter M, Floyd MB, Golas J, Hallett WA, Johnson BD, Nilakantan R, Overbeek E, Reich MF, Shen R, Shi X, Tsou HR, Wang YF, Wissner A. Antitumor activity of HKI-272, an orally active, irreversible inhibitor of the HER-2 tyrosine kinase. Cancer Res. 2004;64:3958–3965. doi: 10.1158/0008-5472.CAN-03-2868. [DOI] [PubMed] [Google Scholar]
- 137.Wissner A, Overbeek E, Reich MF, Floyd MB, Johnson BD, Mamuya N, Rosfjord EC, Discafani C, Davis R, Shi X, Rabindran SK, Gruber BC, Ye F, Hallett WA, Nilakantan R, Shen R, Wang YF, Greenberger LM, Tsou HR. Synthesis and structure-activity relationships of 6,7-disubstituted 4-anilinoquinoline-3-carbonitriles. The design of an orally active, irreversible inhibitor of the tyrosine kinase activity of the epidermal growth factor receptor (EGFR) and the human epidermal growth factor receptor-2 (HER-2) J Med Chem. 2003;46:49–63. doi: 10.1021/jm020241c. [DOI] [PubMed] [Google Scholar]
- 138.Wissner A, Mansour TS. The development of HKI-272 and related compounds for the treatment of cancer. Arch Pharm (Weinheim) 2008;341:465–477. doi: 10.1002/ardp.200800009. [DOI] [PubMed] [Google Scholar]
- 139.Tsou HR, Overbeek-Klumpers EG, Hallett WA, Reich MF, Floyd MB, Johnson BD, Michalak RS, Nilakantan R, Discafani C, Golas J, Rabindran SK, Shen R, Shi X, Wang YF, Upeslacis J, Wissner A. Optimization of 6,7-disubstituted-4-(arylamino)quinoline-3-carbonitriles as orally active, irreversible inhibitors of human epidermal growth factor receptor-2 kinase activity. J Med Chem. 2005;48:1107–1131. doi: 10.1021/jm040159c. [DOI] [PubMed] [Google Scholar]
- 140.Swaby R, Blackwell K, Jiang Z, Sun Y, Dieras V, Zaman K, Zacharchuk C, Powell C, Abbas R, Thakuria M. Neratinib in combination with trastuzumab for the treatment of advanced breast cancer: A phase I/II study. J. Clin. Oncol. 2009;27:15s. [Google Scholar]
- 141.Ito Y, Suenaga M, Hatake K, Takahashi S, Yokoyama M, Onozawa Y, Yamazaki K, Hironaka S, Hashigami K, Hasegawa H, Takenaka N, Boku N. Safety, efficacy and pharmacokinetics of neratinib (HKI-272) in Japanese patients with advanced solid tumors: a Phase 1 doseescalation study. Jpn J Clin Oncol. 2012;42:278–286. doi: 10.1093/jjco/hys012. [DOI] [PubMed] [Google Scholar]
- 142.Chow LW, Xu B, Gupta S, Freyman A, Zhao Y, Abbas R, Vo Van ML, Bondarenko I. Combination neratinib (HKI-272) and paclitaxel therapy in patients with HER2-positive metastatic breast cancer. Br J Cancer. 2013;108:1985–1993. doi: 10.1038/bjc.2013.178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Burstein HJ, Sun Y, Dirix LY, Jiang Z, Paridaens R, Tan AR, Awada A, Ranade A, Jiao S, Schwartz G, Abbas R, Powell C, Turnbull K, Vermette J, Zacharchuk C, Badwe R. Neratinib, an irreversible ErbB receptor tyrosine kinase inhibitor, in patients with advanced ErbB2-positive breast cancer. J. Clin. Oncol. 2010;28:1301–1307. doi: 10.1200/JCO.2009.25.8707. [DOI] [PubMed] [Google Scholar]
- 144.Wong KK, Fracasso PM, Bukowski RM, Lynch TJ, Munster PN, Shapiro GI, Janne PA, Eder JP, Naughton MJ, Ellis MJ, Jones SF, Mekhail T, Zacharchuk C, Vermette J, Abbas R, Quinn S, Powell C, Burris HA. A phase I study with neratinib (HKI-272), an irreversible pan ErbB receptor tyrosine kinase inhibitor, in patients with solid tumors. Clin Cancer Res. 2009;15:2552–2558. doi: 10.1158/1078-0432.CCR-08-1978. [DOI] [PubMed] [Google Scholar]
- 145.Canonici A, Gijsen M, Mullooly M, Bennett R, Bouguern N, Pedersen K, O’Brien NA, Roxanis I, Li JL, Bridge E, Finn R, Siamon D, McGowan P, Duffy MJ, O’Donovan N, Crown J, Kong A. Neratinib overcomes trastuzumab resistance in HER2 amplified breast cancer. Oncotarget. 2013;4:1592–1605. doi: 10.18632/oncotarget.1148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Zhao XQ, Xie JD, Chen XG, Sim HM, Zhang X, Liang YJ, Singh S, Talele TT, Sun Y, Ambudkar SV, Chen ZS, Fu LW. Neratinib reverses ATP-binding cassette B1-mediated chemotherapeutic drug resistance in vitro, in vivo, and ex vivo. Mol Pharmacol. 2012;82:47–58. doi: 10.1124/mol.111.076299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Nafi S, Generali D, Kramer-Marek G, Gijsen M, Strina C, Cappelletti M, Andreis D, Haider S, Li JL, Bridges E, Capala J, Ioannis R, Harris AL, Kong A. Nuclear HER4 mediates acquired resistance to trastuzumab and is associated with poor outcome in HER2 positive breast cancer. Oncotarget. 2014;5:5934–49. doi: 10.18632/oncotarget.1904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Martin M, Bonneterre J, Geyer CE Jr, Ito Y, Ro J, Lang I, Kim SB, Germa C, Vermette J, Wang K, Awada A. A phase two randomised trial of neratinib monotherapy versus lapatinib plus capecitabine combination therapy in patients with HER2+ advanced breast cancer. Eur J Cancer. 2013;49:3763–3772. doi: 10.1016/j.ejca.2013.07.142. [DOI] [PubMed] [Google Scholar]
- 149.Seyhan AA, Varadarajan U, Choe S, Liu W, Ryan TE. A genome-wide RNAi screen identifies novel targets of neratinib resistance leading to identification of potential drug resistant genetic markers. Mol Biosyst. 2012;8:1553–1570. doi: 10.1039/c2mb05512k. [DOI] [PubMed] [Google Scholar]
- 150.Navolanic PM, Steelman LS, McCubrey JA. EGFR family signaling and its association with breast cancer development and resistance to chemotherapy (Review) Int J Oncol. 2003;22:237–252. [PubMed] [Google Scholar]
- 151.Dua R, Zhang J, Nhonthachit P, Penuel E, Petropoulos C, Parry G. EGFR over-expression and activation in high HER2, ER negative breast cancer cell line induces trastuzumab resistance. Breast Cancer Res Treat. 2010;122:685–697. doi: 10.1007/s10549-009-0592-x. [DOI] [PubMed] [Google Scholar]
- 152.Arora A, Scholar EM. Role of tyrosine kinase inhibitors in cancer therapy. J Pharmacol Exp Ther. 2005;315:971–979. doi: 10.1124/jpet.105.084145. [DOI] [PubMed] [Google Scholar]
- 153.Bunn PA Jr, Franklin W. Epidermal growth factor receptor expression, signal pathway, and inhibitors in non-small cell lung cancer. Semin Oncol. 2002;29:38–44. doi: 10.1053/sonc.2002.35646. [DOI] [PubMed] [Google Scholar]
- 154.Bachawal SV, Wali VB, Sylvester PW. Enhanced antiproliferative and apoptotic response to combined treatment of gamma-tocotrienol with erlotinib or gefitinib in mammary tumor cells. BMC Cancer. 2010;10:84. doi: 10.1186/1471-2407-10-84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Bachawal SV, Wali VB, Sylvester PW. Combined gamma-tocotrienol and erlotinib/gefitinib treatment suppresses Stat and Akt signaling in murine mammary tumor cells. Anticancer Res. 2010;30:429–437. [PubMed] [Google Scholar]
- 156.Karaman MW, Herrgard S, Treiber DK, Gallant P, Atteridge CE, Campbell BT, Chan KW, Ciceri P, Davis MI, Edeen PT, Faraoni R, Floyd M, Hunt JP, Lockhart DJ, Milanov ZV, Morrison MJ, Pallares G, Patel HK, Pritchard S, Wodicka LM, Zarrinkar PP. A quantitative analysis of kinase inhibitor selectivity. Nat Biotechnol. 2008;26:127–132. doi: 10.1038/nbt1358. [DOI] [PubMed] [Google Scholar]
- 157.Stamos J, Sliwkowski MX, Eigenbrot C. Structure of the epidermal growth factor receptor kinase domain alone and in complex with a 4-anilinoquinazoline inhibitor. J Biol Chem. 2002;277:46265–46272. doi: 10.1074/jbc.M207135200. [DOI] [PubMed] [Google Scholar]
- 158.Wood ER, Truesdale AT, McDonald OB, Yuan D, Hassell A, Dickerson SH, Ellis B, Pennisi C, Horne E, Lackey K, Alligood KJ, Rusnak DW, Gilmer TM, Shewchuk L. A unique structure for epidermal growth factor receptor bound to GW572016 (Lapatinib): relationships among protein conformation, inhibitor off-rate, and receptor activity in tumor cells. Cancer Res. 2004;64:6652–6659. doi: 10.1158/0008-5472.CAN-04-1168. [DOI] [PubMed] [Google Scholar]
- 159.Bublil EM, Pines G, Patel G, Fruhwirth G, Ng T, Yarden Y. Kinase-mediated quasi-dimers of EGFR. FASEB J. 2010;24:4744–4755. doi: 10.1096/fj.10-166199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Sanchez-Martin M, Pandiella A. Differential action of small molecule HER kinase inhibitors on receptor heterodimerization: therapeutic implications. Int J Cancer. 2012;131:244–252. doi: 10.1002/ijc.26358. [DOI] [PubMed] [Google Scholar]
- 161.O’Neill F, Madden SF, Clynes M, Crown J, Doolan P, Aherne ST, O’Connor R. A gene expression profile indicative of early stage HER2 targeted therapy response. Mol Cancer. 2013;12:69. doi: 10.1186/1476-4598-12-69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.O’Neill F, Madden SF, Aherne ST, Clynes M, Crown J, Doolan P, O’Connor R. Gene expression changes as markers of early lapatinib response in a panel of breast cancer cell lines. Mol Cancer. 2012;11:41. doi: 10.1186/1476-4598-11-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Blackwell KL, Pegram MD, Tan-Chiu E, Schwartzberg LS, Arbushites MC, Maltzman JD, Forster JK, Rubin SD, Stein SH, Burstein HJ. Single-agent lapatinib for HER2-overexpressing advanced or metastatic breast cancer that progressed on first- or second-line trastuzumab-containing regimens. Ann Oncol. 2009;20:1026–1031. doi: 10.1093/annonc/mdn759. [DOI] [PubMed] [Google Scholar]
- 164.Kaufman B, Trudeau M, Awada A, Blackwell K, Bachelot T, Salazar V, DeSilvio M, Westlund R, Zaks T, Spector N, Johnston S. Lapatinib monotherapy in patients with HER2-overexpressing relapsed or refractory inflammatory breast cancer: final results and survival of the expanded HER2+ cohort in EGF103009, a phase II study. Lancet Oncol. 2009;10:581–588. doi: 10.1016/S1470-2045(09)70087-7. [DOI] [PubMed] [Google Scholar]
- 165.Gomez HL, Doval DC, Chavez MA, Ang PC, Aziz Z, Nag S, Ng C, Franco SX, Chow LW, Arbushites MC, Casey MA, Berger MS, Stein SH, Sledge GW. Efficacy and safety of lapatinib as first-line therapy for ErbB2-amplified locally advanced or metastatic breast cancer. J. Clin. Oncol. 2008;26:2999–3005. doi: 10.1200/JCO.2007.14.0590. [DOI] [PubMed] [Google Scholar]
- 166.Chien AJ, Munster PN, Melisko ME, Rugo HS, Park JW, Goga A, Auerback G, Khanafshar E, Ordovas K, Koch KM, Moasser MM. Phase I dose-escalation study of 5-day intermittent oral lapatinib therapy in patients with human epidermal growth factor receptor 2-overexpressing breast cancer. J. Clin. Oncol. 2014;32:1472–1479. doi: 10.1200/JCO.2013.52.1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Goss PE, Smith IE, O’Shaughnessy J, Ejlertsen B, Kaufmann M, Boyle F, Buzdar AU, Fumoleau P, Gradishar W, Martin M, Moy B, Piccart-Gebhart M, Pritchard KI, Lindquist D, Chavarri-Guerra Y, Aktan G, Rappold E, Williams LS, Finkelstein DM TEACH investigators. Adjuvant lapatinib for women with early-stage HER2-positive breast cancer: a randomised, controlled, phase 3 trial. Lancet Oncol. 2013;14:88–96. doi: 10.1016/S1470-2045(12)70508-9. [DOI] [PubMed] [Google Scholar]
- 168.Brain E, Isambert N, Dalenc F, Dieras V, Bonneterre J, Rezai K, Jimenez M, Mefti-Lacheraf F, Cottura E, Tresca P, Vanlemmens L, Mahier-Ait Oukhatar C, Lokiec F, Fumoleau P. Phase I study of lapatinib plus vinorelbine in patients with locally advanced or metastatic breast cancer overexpressing HER2. Br J Cancer. 2012;106:673–677. doi: 10.1038/bjc.2011.591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Cameron D, Casey M, Oliva C, Newstat B, Imwalle B, Geyer CE. Lapatinib plus capecitabine in women with HER-2-positive advanced breast cancer: final survival analysis of a phase III randomized trial. Oncologist. 2010;15:924–934. doi: 10.1634/theoncologist.2009-0181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Cameron D, Casey M, Press M, Lindquist D, Pienkowski T, Romieu CG, Chan S, Jagiello-Gruszfeld A, Kaufman B, Crown J, Chan A, Campone M, Viens P, Davidson N, Gorbounova V, Raats JI, Skarlos D, Newstat B, Roychowdhury D, Paoletti P, Oliva C, Rubin S, Stein S, Geyer CE. A phase III randomized comparison of lapatinib plus capecitabine versus capecitabine alone in women with advanced breast cancer that has progressed on trastuzumab: updated efficacy and biomarker analyses. Breast Cancer Res Treat. 2008;112:533–543. doi: 10.1007/s10549-007-9885-0. [DOI] [PubMed] [Google Scholar]
- 171.Kaufman B, Stein S, Casey MA, Newstat BO. Lapatinib in combination with capecitabine in the management of ErbB2-positive (HER2-positive) advanced breast cancer. Biologics. 2008;2:61–65. doi: 10.2147/btt.s1713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Gajria D, Gonzalez J, Feigin K, Patil S, Chen C, Theodoulou M, Drullinsky P, D’Andrea G, Lake D, Norton L, Hudis CA, Traina TA. Phase II trial of a novel capecitabine dosing schedule in combination with lapatinib for the treatment of patients with HER2-positive metastatic breast cancer. Breast Cancer Res Treat. 2012;131:111–116. doi: 10.1007/s10549-011-1749-y. [DOI] [PubMed] [Google Scholar]
- 173.Capri G, Chang J, Chen SC, Conte P, Cwiertka K, Jerusalem G, Jiang Z, Johnston S, Kaufman B, Link J, Ro J, Schutte J, Oliva C, Parikh R, Preston A, Rosenlund J, Selzer M, Zembryki D, De Placido S. An open-label expanded access study of lapatinib and capecitabine in patients with HER2-overexpressing locally advanced or metastatic breast cancer. Ann Oncol. 2010;21:474–480. doi: 10.1093/annonc/mdp373. [DOI] [PubMed] [Google Scholar]
- 174.Iwata H, Fujii H, Masuda N, Mukai H, Nishimura Y, Katsura K, Ellis CE, Gagnon RC, Nakamura S. Efficacy, safety, pharmacokinetics and biomarker findings in patients with HER2-positive advanced or metastatic breast cancer treated with lapatinib in combination with capecitabine: results from 51 Japanese patients treated in a clinical study. Breast Cancer. 2015;22:192–200. doi: 10.1007/s12282-013-0475-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Xu BH, Jiang ZF, Chua D, Shao ZM, Luo RC, Wang XJ, Liu DG, Yeo W, Yu SY, Newstat B, Preston A, Martin AM, Chi HD, Wang L. Lapatinib plus capecitabine in treating HER2-positive advanced breast cancer: efficacy, safety, and biomarker results from Chinese patients. Chin J Cancer. 2011;30:327–335. doi: 10.5732/cjc.010.10507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Gamucci T, Moscetti L, Mentuccia L, Pizzuti L, Mauri M, Zampa G, Pavese I, Sperduti I, Vaccaro A, Vici P. Optimal tolerability and high efficacy of a modified schedule of lapatinib-capecitabine in advanced breast cancer patients. J Cancer Res Clin Oncol. 2014;140:221–226. doi: 10.1007/s00432-013-1556-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Pircher M, Mlineritsch B, Fridrik MA, Dittrich C, Lang A, Petru E, Weltermann A, Thaler J, Hufnagl C, Gampenrieder SP, Rinnerthaler G, Ressler S, Ulmer H, Greil R. Lapatinib-pluspegylated liposomal doxorubicin in advanced HER2-positive breast cancer following trastuzumab: a phase II trial. Anticancer Res. 2015;35:517–521. [PubMed] [Google Scholar]
- 178.Bian L, Wang T, Zhang S, Jiang Z. Trastuzumab plus capecitabine vs. lapatinib plus capecitabine in patients with trastuzumab resistance and taxane-pretreated metastatic breast cancer. Tumour Biol. 2013;34:3153–3158. doi: 10.1007/s13277-013-0884-y. [DOI] [PubMed] [Google Scholar]
- 179.Janni W, Sarosiek T, Karaszewska B, Pikiel J, Staroslawska E, Potemski P, Salat C, Brain E, Caglevic C, Briggs K, Desilvio M, Marini L, Papadimitriou C. A phase II, randomized, multicenter study evaluating the combination of lapatinib and vinorelbine in women with ErbB2 overexpressing metastatic breast cancer. Breast Cancer Res Treat. 2014;143:493–505. doi: 10.1007/s10549-013-2828-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Cristofanilli M, Johnston SR, Manikhas A, Gomez HL, Gladkov O, Shao Z, Safina S, Blackwell KL, Alvarez RH, Rubin SD, Ranganathan S, Redhu S, Trudeau ME. A randomized phase II study of lapatinib + pazopanib versus lapatinib in patients with HER2+ inflammatory breast cancer. Breast Cancer Res Treat. 2013;137:471–482. doi: 10.1007/s10549-012-2369-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Johnston SR, Gomez H, Stemmer SM, Richie M, Durante M, Pandite L, Goodman V, Slamon D. A randomized and open-label trial evaluating the addition of pazopanib to lapatinib as first-line therapy in patients with HER2-positive advanced breast cancer. Breast Cancer Res Treat. 2013;137:755–766. doi: 10.1007/s10549-012-2399-4. [DOI] [PubMed] [Google Scholar]
- 182.Rugo HS, Chien AJ, Franco SX, Stopeck AT, Glencer A, Lahiri S, Arbushites MC, Scott J, Park JW, Hudis C, Nulsen B, Dickler MN. A phase II study of lapatinib and bevacizumab as treatment for HER2-overexpressing metastatic breast cancer. Breast Cancer Res Treat. 2012;134:13–20. doi: 10.1007/s10549-011-1918-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Guan Z, Xu B, DeSilvio ML, Shen Z, Arpornwirat W, Tong Z, Lorvidhaya V, Jiang Z, Yang J, Makhson A, Leung WL, Russo MW, Newstat B, Wang L, Chen G, Oliva C, Gomez H. Randomized trial of lapatinib versus placebo added to paclitaxel in the treatment of human epidermal growth factor receptor 2-overexpressing metastatic breast cancer. J. Clin. Oncol. 2013;31:1947–1953. doi: 10.1200/JCO.2011.40.5241. [DOI] [PubMed] [Google Scholar]
- 184.Di Leo A, Gomez HL, Aziz Z, Zvirbule Z, Bines J, Arbushites MC, Guerrera SF, Koehler M, Oliva C, Stein SH, Williams LS, Dering J, Finn RS, Press MF. Phase III, double-blind, randomized study comparing lapatinib plus paclitaxel with placebo plus paclitaxel as first-line treatment for metastatic breast cancer. J. Clin. Oncol. 2008;26:5544–5552. doi: 10.1200/JCO.2008.16.2578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Yardley DA, Hart L, Bosserman L, Salleh MN, Waterhouse DM, Hagan MK, Richards P, DeSilvio ML, Mahoney JM, Nagarwala Y. Phase II study evaluating lapatinib in combination with nab-paclitaxel in HER2-overexpressing metastatic breast cancer patients who have received no more than one prior chemotherapeutic regimen. Breast Cancer Res Treat. 2013;137:457–464. doi: 10.1007/s10549-012-2341-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Park IH, Lee KS, Kang HS, Kim SW, Lee S, Jung SY, Kwon Y, Shin KH, Ko K, Nam BH, Ro J. A phase Ib study of preoperative lapatinib, paclitaxel, and gemcitabine combination therapy in women with HER2 positive early breast cancer. Invest New Drugs. 2012;30:1972–1977. doi: 10.1007/s10637-011-9759-5. [DOI] [PubMed] [Google Scholar]
- 187.Storniolo AM, Pegram MD, Overmoyer B, Silverman P, Peacock NW, Jones SF, Loftiss J, Arya N, Koch KM, Paul E, Pandite L, Fleming RA, Lebowitz PF, Ho PT, Burris HA 3rd. Phase I dose escalation and pharmacokinetic study of lapatinib in combination with trastuzumab in patients with advanced ErbB2-positive breast cancer. J. Clin. Oncol. 2008;26:3317–3323. doi: 10.1200/JCO.2007.13.5202. [DOI] [PubMed] [Google Scholar]
- 188.Rimawi MF, Mayer IA, Forero A, Nanda R, Goetz MP, Rodriguez AA, Pavlick AC, Wang T, Hilsenbeck SG, Gutierrez C, Schiff R, Osborne CK, Chang JC. Multicenter phase II study of neoadjuvant lapatinib and trastuzumab with hormonal therapy and without chemotherapy in patients with human epidermal growth factor receptor 2-overexpressing breast cancer: TBCRC 006. J. Clin. Oncol. 2013;31:1726–1731. doi: 10.1200/JCO.2012.44.8027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Montemurro F, Prat A, Rossi V, Valabrega G, Sperinde J, Peraldo-Neia C, Donadio M, Galvan P, Sapino A, Aglietta M, Baselga J, Scaltriti M. Potential biomarkers of long-term benefit from single-agent trastuzumab or lapatinib in HER2-positive metastatic breast cancer. Mol Oncol. 2014;8:20–26. doi: 10.1016/j.molonc.2013.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Crown J, Kennedy MJ, Tresca P, Marty M, Espie M, Burris HA, Desilvio M, Lau MR, Kothari D, Koch KM, Dieras V. Optimally tolerated dose of lapatinib in combination with docetaxel plus trastuzumab in first-line treatment of HER2-positive metastatic breast cancer. Ann Oncol. 2013;24:2005–2011. doi: 10.1093/annonc/mdt222. [DOI] [PubMed] [Google Scholar]
- 191.Baselga J, Bradbury I, Eidtmann H, Di Cosimo S, de Azambuja E, Aura C, Gomez H, Dinh P, Fauria K, Van Dooren V, Aktan G, Goldhirsch A, Chang TW, Horvath Z, Coccia-Portugal M, Domont J, Tseng LM, Kunz G, Sohn JH, Semiglazov V, Lerzo G, Palacova M, Probachai V, Pusztai L, Untch M, Gelber RD, Piccart-Gebhart M, Neo AST. Lapatinib with trastuzumab for HER2-positive early breast cancer (NeoALTTO): a randomised, open-label, multicentre, phase 3 trial. Lancet. 2012;379:633–640. doi: 10.1016/S0140-6736(11)61847-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.de Azambuja E, Holmes AP, Piccart-Gebhart M, Holmes E, Di Cosimo S, Swaby RF, Untch M, Jackisch C, Lang I, Smith I, Boyle F, Xu B, Barrios CH, Perez EA, Azim HA Jr, Kim SB, Kuemmel S, Huang CS, Vuylsteke P, Hsieh RK, Gorbunova V, Eniu A, Dreosti L, Tavartkiladze N, Gelber RD, Eidtmann H, Baselga J. Lapatinib with trastuzumab for HER2-positive early breast cancer (NeoALTTO): survival outcomes of a randomised, open-label, multicentre, phase 3 trial and their association with pathological complete response. Lancet Oncol. 2014;15:1137–1146. doi: 10.1016/S1470-2045(14)70320-1. [DOI] [PubMed] [Google Scholar]
- 193.Moreno-Aspitia A, Dueck AC, Ghanem-Canete I, Patel T, Dakhil S, Johnson D, Franco S, Kahanic S, Colon-Otero G, Tenner KS, Rodeheffer R, McCullough AE, Jenkins RB, Palmieri FM, Northfelt D, Perez EA. RC0639: phase II study of paclitaxel, trastuzumab, and lapatinib as adjuvant therapy for early stage HER2-positive breast cancer. Breast Cancer Res Treat. 2013;138:427–435. doi: 10.1007/s10549-013-2469-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Welslau M, Dieras V, Sohn JH, Hurvitz SA, Lalla D, Fang L, Althaus B, Guardino E, Miles D. Patient-reported outcomes from EMILIA, a randomized phase 3 study of trastuzumab emtansine (T-DM1) versus capecitabine and lapatinib in human epidermal growth factor receptor 2-positive locally advanced or metastatic breast cancer. Cancer. 2014;120:642–651. doi: 10.1002/cncr.28465. [DOI] [PubMed] [Google Scholar]
- 195.Guarneri V, Frassoldati A, Bottini A, Cagossi K, Bisagni G, Sarti S, Ravaioli A, Cavanna L, Giardina G, Musolino A, Untch M, Orlando L, Artioli F, Boni C, Generali DG, Serra P, Bagnalasta M, Marini L, Piacentini F, D’Amico R, Conte P. Preoperative chemotherapy plus trastuzumab, lapatinib, or both in human epidermal growth factor receptor 2-positive operable breast cancer: results of the randomized phase II CHER-LOB study. J. Clin. Oncol. 2012;30:1989–1995. doi: 10.1200/JCO.2011.39.0823. [DOI] [PubMed] [Google Scholar]
- 196.Alba E, Albanell J, de la Haba J, Barnadas A, Calvo L, Sanchez-Rovira P, Ramos M, Rojo F, Burgues O, Carrasco E, Caballero R, Porras I, Tibau A, Camara MC, Lluch A. Trastuzumab or lapatinib with standard chemotherapy for HER2-positive breast cancer: results from the GEICAM/2006-14 trial. Br J Cancer. 2014;110:1139–1147. doi: 10.1038/bjc.2013.831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Untch M, Loibl S, Bischoff J, Eidtmann H, Kaufmann M, Blohmer JU, Hilfrich J, Strumberg D, Fasching PA, Kreienberg R, Tesch H, Hanusch C, Gerber B, Rezai M, Jackisch C, Huober J, Kuhn T, Nekljudova V, von Minckwitz G German Breast Group (GBG); Arbeitsgemeinschaft Gynäkologische Onkologie-Breast (AGO-B) Study Group. Lapatinib versus trastuzumab in combination with neoadjuvant anthracycline-taxane-based chemotherapy (GeparQuinto, GBG 44): a randomised phase 3 trial. Lancet Oncol. 2012;13:135–144. doi: 10.1016/S1470-2045(11)70397-7. [DOI] [PubMed] [Google Scholar]
- 198.Jankowitz RC, Abraham J, Tan AR, Limentani SA, Tierno MB, Adamson LM, Buyse M, Wolmark N, Jacobs SA. Safety and efficacy of neratinib in combination with weekly paclitaxel and trastuzumab in women with metastatic HER2positive breast cancer: an NSABP Foundation Research Program phase I study. Cancer Chemother Pharmacol. 2013;72:1205–1212. doi: 10.1007/s00280-013-2262-2. [DOI] [PubMed] [Google Scholar]
- 199.Awada A, Dirix L, Manso Sanchez L, Xu B, Luu T, Dieras V, Hershman DL, Agrapart V, Ananthakrishnan R, Staroslawska E. Safety and efficacy of neratinib (HKI-272) plus vinorelbine in the treatment of patients with ErbB2-positive metastatic breast cancer pretreated with anti-HER2 therapy. Ann Oncol. 2013;24:109–116. doi: 10.1093/annonc/mds284. [DOI] [PubMed] [Google Scholar]
- 200.Saura C, Garcia-Saenz JA, Xu B, Harb W, Moroose R, Pluard T, Cortes J, Kiger C, Germa C, Wang K, Martin M, Baselga J, Kim SB. Safety and efficacy of neratinib in combination with capecitabine in patients with metastatic human epidermal growth factor receptor 2-positive breast cancer. J. Clin. Oncol. 2014;32:3626–3633. doi: 10.1200/JCO.2014.56.3809. [DOI] [PubMed] [Google Scholar]
