Preventing breast cancer is possible with selective estrogen receptor (ER) modulators and aromatase inhibitors, which reduce the risk of invasive disease by up to 65% (up to 73% for ER-positive and no effect for ER-negative cancer) and the risk of preinvasive disease [ductal carcinoma in situ (DCIS)] by up to 50%. Clearly, approaches for preventing ER-negative, and increased prevention of ER-positive breast cancers would benefit public health. A growing body of work (including recent preclinical and clinical data) support targeting the HER family [epidermal growth factor receptor (EGFR), or human epidermal growth factor receptor (HER) 1 or ErbB1) and HER2, HER3, and HER4] for preventing ER-negative and possibly ER-positive breast cancer. Preclinical studies of HER family–targeting drugs in mammary neoplasia show suppression of (i) ER-negative tumors in HER2-overexpressing mouse strains, (ii) ER-negative tumors in mutant Brca1/p53+/ mice, and (iii) ER-positive tumors in the methylnitrosourea (MNU) rat model; tumors arising in both the MNU and mutant Brca1/p53+/ models lack HER2 overexpression. Clinical trials include a recent placebo-controlled phase IIb presurgical trial of the dual EGFR HER2 inhibitor lapatinib that suppressed growth of breast premalignancy [including atypical ductal hyperplasia (ADH) and DCIS] and invasive cancer in patients with early-stage, HER2-overexpressing or -amplified breast cancer. These results suggest that lapatinib can clinically suppress the progression of ADH and DCIS to invasive breast cancer, an effect previously observed in a mouse model of HER2-overexpressing, ER-negative mammary cancer. The preclinical and clinical signals provide a compelling rationale for testing HER-targeting drugs for breast cancer prevention in women at moderate-to-high risk, leading perhaps to combinations that prevent ER-negative and ER-positive breast cancer. Cancer Prev Res; 4(8); 1149–57. ©2011 AACR.

Notwithstanding major advances in breast cancer prevention since the late 1990s, the incidence of breast cancer remains high and treating metastatic breast cancer remains challenging. Clearly, effective new preventive strategies for this disease are needed. Large phase III clinical trials have identified several estrogen receptor (ER)-targeting drugs that prevent breast cancer in moderate-to-high–risk women. Selective ER modulators (SERMs; e.g., tamoxifen, raloxifene, and lasofoxifene; refs. 1, 2) and the aromatase inhibitor (AI) exemestane (3) reduce the risk of breast cancer in women without prior breast cancer. Predictably, these drugs only prevent ER-positive tumors. Therefore, there is an urgent need to identify targetable pathways for preventing ER-negative breast cancer, which accounts for approximately one third of breast cancers in the United States and for a substantially higher proportion in Asian countries such as India and China. Increasing evidence, including 2 articles appearing elsewhere in this issue of the journal (4, 5), supports the HER family as potentially useful targets for preventing ER-negative breast cancer.

The HER family [also called the ErbB or epidermal growth factor receptor (EGFR) family] comprises 4 transmembrane receptor tyrosine kinases, EGFR itself (also called HER1) and HER2, HER3, and HER4 (also called ErbB2, ErbB3, and ErbB4). Signaling occurs through both homo- and heterodimeric HER complexes (Fig. 1) and can induce cell proliferation, motility, and invasion. Dysregulated expression and activity of HER family members is prevalent in human neoplasia (6). Strikingly, up to 30% of breast carcinomas overexpress HER2, frequently as a consequence of genomic amplification of a region of the long arm of chromosome 17 (17q21) that includes the HER2 locus. HER2 overexpression may be more frequent in ER-negative than in ER-positive cancers, drives aggressive disease, and thus represents an important therapeutic target. The humanized monoclonal antibody trastuzumab (Herceptin) was the first agent developed for HER2 targeting and has dramatically improved outcomes among women with HER2-positive (defined by HER2 overexpression and/or amplification) breast cancer.

Figure 1.

HER signaling and targeted breast cancer prevention.MAPKs, mitogen-activated protein kinases; JNK, c-jun N-terminal kinase; S6K, p70 S6 ribosomal kinase; 4EBP1, 4E-binding protein 1; TF, transcription factor; BC, breast cancer.

Figure 1.

HER signaling and targeted breast cancer prevention.MAPKs, mitogen-activated protein kinases; JNK, c-jun N-terminal kinase; S6K, p70 S6 ribosomal kinase; 4EBP1, 4E-binding protein 1; TF, transcription factor; BC, breast cancer.

Close modal

Effective small-molecule inhibitors of EGFR/ErbB tyrosine kinases, including the EGFR inhibitors gefitinib (Iressa) and erlotinib (Tarceva) and the dual EGFR/HER2 inhibitor lapatinib (Tykerb), have also been developed. Lapatinib is U.S. Food and Drug Administration (FDA)-approved for treating advanced or metastatic HER2-overexpressing breast cancer. The initial approval was for use in combination with capecitabine in patients with metastatic breast cancer who had received prior therapy including an anthracycline, a taxane, and trastuzumab (7). Lapatinib is also approved for use in combination with the AI letrozole to treat postmenopausal women with hormone receptor [ER or progesterone receptor (PR)]-positive, HER2-positive advanced breast cancer (http://www.cancer.gov/cancertopics/druginfo/fda-lapatinib). Subgroup analyses of clinical breast cancer therapy trials identified HER2 overexpression as a key determinant of sensitivity to lapatinib (8–10), whereas EGFR expression does not appear to be predictive (11).

Agents targeting the ER and HER axes have a spectrum of preclinical and FDA-approved clinical activity in the prevention and treatment of breast cancer (Fig. 2). Lapatinib has more complete activity in blocking HER signaling (vs. the other major HER family–targeting agents), indicating its potential for preventing ER-positive and -negative breast cancer (Fig. 1). Given that HER2 is overexpressed in a large proportion of preinvasive ductal carcinomas in situ (DCIS; refs. 12–14), lapatinib and other HER family–targeting drugs have a strong potential for breast cancer prevention.

Figure 2.

HER family targeting for breast cancer prevention. Potential clinical preventive applications of lapatinib (along with known applications of trastuzumab, SERMs, and AIs). The major circle segments reflect HER2-positive cancer (pink; ∼20% of total disease) and HER2-negative cancer (sky blue; ∼80% of total disease). Divisions within segments reflect the rough proportions of HR (ER, PR)-positive and -negative disease, which, respectively, are 50%/50% in HER2-positive cancer and 75%/25% in HER2-negative cancer (the rough proportions in all cancers are 70% HR-positive and 30% HR-negative). Doubled blocking lines indicate settings of greater HER-targeting drug effect.

Figure 2.

HER family targeting for breast cancer prevention. Potential clinical preventive applications of lapatinib (along with known applications of trastuzumab, SERMs, and AIs). The major circle segments reflect HER2-positive cancer (pink; ∼20% of total disease) and HER2-negative cancer (sky blue; ∼80% of total disease). Divisions within segments reflect the rough proportions of HR (ER, PR)-positive and -negative disease, which, respectively, are 50%/50% in HER2-positive cancer and 75%/25% in HER2-negative cancer (the rough proportions in all cancers are 70% HR-positive and 30% HR-negative). Doubled blocking lines indicate settings of greater HER-targeting drug effect.

Close modal

Preclinical studies

Several studies have explored ErbB inhibitors for chemoprevention in animal breast cancer models (Table 1). The Arteaga group provided early proof of principle for the preventive efficacy of EGFR inhibition in work using bigenic mice expressing neu (the rat HER2 homologue) and TGFα expressed from the mouse mammary tumor virus (MMTV) promoter (15). Simultaneous expression of mammary-targeted transgenes encoding neu and TGFα (an EGFR ligand) provides a model for human breast cancers coexpressing HER2 and EGFR. Administration of the EGFR tyrosine kinase inhibitor tyrphostin (AG1478) from 8 weeks onward significantly delayed tumor onset in virgin female mice. Short-term tyrphostin administration suppressed DNA synthesis in tumor cells as well as in uninvolved epithelium, but induction of apoptosis was not observed, suggesting that antiproliferative effects were the primary antitumor mechanism. Consistent with the observed reduction in proliferation, tumor lysates from tyrphostin-treated animals exhibited decreases in cyclin D1 levels and cyclin-dependent kinase 2 (cdk2) activity and increased levels of the cell-cycle inhibitor p27kip1.

Table 1.

Preclinical studies of ErbB inhibitors for mammary tumor prevention

DrugTargetAnimal modelER statusResultsRefs.
Tyrphostin EGFR Bigenic MMTV/neu, MMTV/TGFα mice Unknown Daily intraperitoneal injections (50 mg/kg, from 8 wk old) substantially delayed tumor onset; tumor incidence = 90% (controls) vs. 20% (tyrphostin) at 250 d (P = 0.0007) (15) 
Gefitinib EGFR MMTV/neu mice ER-negative tumors Daily gavage (6 d/wk; 100 mg/kg, from 3 mo old) delayed median time-to-tumor from 230 to >310 d (P < 0.001), when 75% of gefitinib-treated vs. 0% of control mice were tumor free (16) 
Gefitinib EGFR BALB-neuT mice ER-negative tumors Oral gavage (5 d/wk; 75 mg/kg, increasing to 135 mg/kg, between 5 and 14 wk old) reduced mean number of tumor-bearing glands/mouse from 9.6 to 0.6 (P < 0.0001) (17) 
Gefitinib EGFR MNU-treated rats ER-positive tumors 10 mg/kg daily reduced cancer multiplicity by 91% (P < 0.01) and reduced size of established tumors (19) 
Lapatinib EGFR/HER2 MMTV/neu mice ER-negative tumors Twice daily gavage (6 d/wk; 75 mg/kg, from 3 mo old) reduced tumor incidence from 100% to 31% at 418 d old (P < 0.001) (18) 
Lapatinib EGFR/HER2 MNU-treated rats ER-positive tumors Daily gavage (7 d/wk, from 55 d old) reduced tumor multiplicity by 31% (25 mg/kg) and 81% (75 mg/kg) at 198 d old (5) 
Erlotinib EGFR MMTV/Cre, Brca1flox/flox/p53+/− mice ER-positive, -negative tumors Daily gavage (7 d/wk; 100 mg/kg, from 3 mo old) increased tumor latency from median 256 days to 365 d (P = 0.0003) with selective suppression of ER-negative tumors (24) 
DrugTargetAnimal modelER statusResultsRefs.
Tyrphostin EGFR Bigenic MMTV/neu, MMTV/TGFα mice Unknown Daily intraperitoneal injections (50 mg/kg, from 8 wk old) substantially delayed tumor onset; tumor incidence = 90% (controls) vs. 20% (tyrphostin) at 250 d (P = 0.0007) (15) 
Gefitinib EGFR MMTV/neu mice ER-negative tumors Daily gavage (6 d/wk; 100 mg/kg, from 3 mo old) delayed median time-to-tumor from 230 to >310 d (P < 0.001), when 75% of gefitinib-treated vs. 0% of control mice were tumor free (16) 
Gefitinib EGFR BALB-neuT mice ER-negative tumors Oral gavage (5 d/wk; 75 mg/kg, increasing to 135 mg/kg, between 5 and 14 wk old) reduced mean number of tumor-bearing glands/mouse from 9.6 to 0.6 (P < 0.0001) (17) 
Gefitinib EGFR MNU-treated rats ER-positive tumors 10 mg/kg daily reduced cancer multiplicity by 91% (P < 0.01) and reduced size of established tumors (19) 
Lapatinib EGFR/HER2 MMTV/neu mice ER-negative tumors Twice daily gavage (6 d/wk; 75 mg/kg, from 3 mo old) reduced tumor incidence from 100% to 31% at 418 d old (P < 0.001) (18) 
Lapatinib EGFR/HER2 MNU-treated rats ER-positive tumors Daily gavage (7 d/wk, from 55 d old) reduced tumor multiplicity by 31% (25 mg/kg) and 81% (75 mg/kg) at 198 d old (5) 
Erlotinib EGFR MMTV/Cre, Brca1flox/flox/p53+/− mice ER-positive, -negative tumors Daily gavage (7 d/wk; 100 mg/kg, from 3 mo old) increased tumor latency from median 256 days to 365 d (P = 0.0003) with selective suppression of ER-negative tumors (24) 

Two subsequent studies examined the EGFR inhibitor gefitinib in the FVB MMTV/neu (16) and BALB/c MMTV/neuT (17) mouse models of breast cancer, in which tumor formation is driven by expression of the wild-type (in the FVB MMTV/neu model) or mutationally activated (in the BALB/c MMTV/neuT model) rat HER2 homologue. Tumors in HER2/neu transgenic mice generally lack ERα expression, and thus, these strains simultaneously provide a model of HER2-overexpressing and ER-negative disease. Both studies showed preventive efficacy of gefitinib. Lu and colleagues showed a substantial delay in tumor onset (median tumor latency increased from 230 to >310 days) in virgin FVB MMTV/neu females administered gefitinib from 3 months of age (16), and Piechocki and colleagues observed a striking reduction in tumor multiplicity in BALB/c MMTV/neuT mice treated with gefitinib for 9 weeks (age 5–14 weeks; ref. 17). Mechanistic studies indicated that gefitinib suppressed mammary tumor development predominantly through antiproliferative effects.

More recently, the dual EGFR/HER2 inhibitor lapatinib was also evaluated for cancer prevention (5, 18). As does gefitinib, lapatinib suppresses ER-negative mammary tumor development in FVB MMTV/neu mice in a dose-dependent manner, again apparently through an antiproliferative effect (18). Of note, limited duration (5 months) treatment suppressed the formation of premalignant lesions and microscopic invasive tumors. As observed for both gefitinib and tyrphostin, lapatinib suppressed proliferation in mammary epithelium, with no effect on apoptosis. Mechanistic analyses indicated commensurate molecular alterations including decreased cyclin D1 expression and increased p27 transcripts.

All of the studies discussed above utilized HER2-overexpressing mouse strains, a rational choice based on a predicted model in which ErbB inhibitors suppress HER2 signaling via modulation of EGFR–HER2 heterodimers. Intriguingly, however, 2 recent studies, one of gefitinib (19) and the other of lapatinib (5), showed that HER targeting can also be effective in a non–HER2-overexpressing model (although a physiologic level of HER2 is present). Both gefitinib and lapatinib suppressed tumor multiplicity and caused regression of established tumors in methylnitrosourea (MNU)-treated rats, a widely used model of ER-positive disease. Phospho-EGFR and phospho-HER2 levels were reduced in tumor tissues from the lapatinib-treated rats (5), suggesting that lapatinib effects are mediated by HER1 dimers and possibly also work through suppressing physiologic levels of HER2 (Fig. 1). No reduction in phosphorylated extracellular signal–regulated kinase (phospho-ERK) levels was detected in MNU-induced tumors from lapatinib-treated rats, which contrasts with previously reported effects of gefitinib and tyrphostin on phospho-ERK in HER2/neu transgenic strains (15–17) and of lapatinib on breast cancer cell lines (20, 21). Instead, phosphorylation of Src family kinase members Lyn and Lck was reduced, as were levels of phospho-AKT and IGF-1R. Molecular markers of increased apoptosis were also identified, although apoptosis was not directly assayed. The observed reduction of IGF-1R in lapatinib-treated, MNU-induced mammary tumors (5) is intriguing and potentially indicates cross-talk between the HER family and IGF-1R (22). Lapatinib is known to suppress the migration and invasion of cultured breast cancer cell lines induced by leptin and IGF-1 (23). Because these factors are elevated in association with obesity, lapatinib could potentially modify the increased breast cancer risk that has been identified for obese postmenopausal women. Therapeutic trials of lapatinib clearly identify HER2 overexpression as the key determinant of lapatinib sensitivity (8–10), whereas Li and colleagues found that lapatinib was active preclinically in a model where HER2 was present but not overexpressed (5). This apparent discrepancy suggests that ER-driven breast neoplasia could be differentially sensitive to signaling input from the HER family at preinvasive stages compared with later stages of tumor progression.

A recent study from Gerburg Wulf's group suggests that EGFR inhibition may be clinically significant in the absence of HER2 amplification, specifically in the context of BRCA1 deficiency (24). This group found that EGFR expression increased in cultured mammary epithelial cells in response to siRNA-mediated BRCA1 depletion, particularly in the subset expressing the putative stem cell marker aldehyde dehydrogenase 1 (ALDH1). Corresponding EGFR upregulation occurred in the acini of Brca1-deficient mammary glands from MMTV/Cre, Brca1flox/flox/p53+/ mice. Mammary tumor latency in these mice was significantly increased by erlotinib treatment initiated at 3 months of age. Tumors that did develop tended to lack EGFR expression and to be ER-positive, showing a selective suppression of ER-negative tumor formation. Furthermore, tumors from erlotinib-treated animals lacked ALDH1 expression, potentially suggesting a reduced stem cell component. This study suggests the exciting possibility that EGFR inhibition may be a viable strategy for reducing ER-negative breast cancer risk in carriers of mutant BRCA1 alleles (25).

Clinical trials

Anti-HER2 drugs have been used for years for treating HER2-positive breast cancer. The humanized monoclonal antibody trastuzumab was the first anti-HER2 drug shown to be active in breast cancer (Table 2). Its effectiveness was initially shown in treating metastatic HER2-positive breast cancer (26) and subsequently in treating early-stage, HER2-positive breast cancer (27) and in neoadjuvant settings involving HER2-positive disease (before surgical resection; ref. 28). Two phase III adjuvant trials of trastuzumab recently showed a dramatic prolongation of median disease-free survival in women with early-stage, HER2-positive breast cancer (29). This dramatic efficacy revolutionized the treatment of this form of breast cancer, thus converting the outcome of HER2-positive patients from relatively poor to relatively good (compared to HER2-negative, hormone receptor–negative patients).

Table 2.

Clinical studies of ErbB inhibitors in breast cancer

Drug/trial designTargetSettingResultsRefs.
Tras (I-line, randomized phase II) HER2 Metastatic HER2-positive I-line Tras produced a 26% objective response rate; response rate in 3+ HER2 overexpression was 35%, with a CBR of 48% (26) 
Tras (phase III HERA trial; initial results) HER2 Early-stage, HER2-positive Tras for 1 y after adjuvant chemotherapy improved DFS; Tras HR (vs. observation) = 0.54 (95% CI: 0.43–0.67; P < 0.0001) (49) 
Tras (longer-term HERA results) HER2 Early-stage, HER2-positive Tras for 1 y after adjuvant chemotherapy improved overall survival—HR for risk of death (vs. observation) was 0.66 (95% CI: 0.47–0.91; P = 0.0115) (27) 
Tras (phase III neo-adjuvant) HER2 HER2-positive Tras + chemotherapy (paclitaxel followed by FEC) improved pCR (65.2%) vs. chemotherapy alone (26%; P = 0.016) (50)  
Tras + chemotherapy (two phase III adjuvant trials) HER2 Early-stage, HER2-positive Dramatically prolonged median DFS–HR = 0.48 (P < 0.0001), and produced 33% reduced risk of death (P = 0.015) (29) 
Pertuzumab (phase I) HER2 Metastatic cancer PR in 2 patients (nonbreast cancer); stable disease lasting >2.5 m in 6 patients (31) 
Tras-DM1 (phase II) HER2 Metastatic HER2-positive, resistant to HER2 therapy Produced 26% objective response rate (33) 
Gefitinib (phase II) EGFR Advanced/metastatic 0% CR + PR (34) 
Erlotinib (phase II) EGFR Advanced/metastatic 3% PR (35) 
Lapat (phase III) EGFR/HER2 Metastatic HER2-positive Lapat + capecitabine prolonged TTP (vs. capecitabine; HR = 0.49; 95% CI: 0.34–0.71; P < 0.001) (37) 
Lapat (phase II) EGFR/HER2 Refractory, metastatic Modest activity (6% clinical benefit) (51) 
Lapat (1st-line phase III) EGFR/HER2 Metastatic Lapat + paclitaxel improved TTP (HR = 0.53; 95% CI: 0.31–0.89; P = 0.005) and event-free survival (HR = 0.52; 95% CI: 0.31–0.86; P = 0.004)–all vs. placebo + paclitaxel–in HER2-positive disease (9) 
Lapat + letrozole (1st-line phase III) EGFR/HER2 Metastatic, hormone- receptor–positive Letrozole + lapat reduced risk of disease progression vs. letrozole-alone in HER2-positive disease (HR = 0.71; 95% CI: 0.53–0.96; P = 0.019); no improved outcome in HER2-negative cancer (10) 
Lapat (phase II neoadjuvant) EGFR/HER2 Inflammatory breast cancer Lapat monotherapy followed by Lapat + paclitaxel produced a 78% clinical response rate in HER2-positive disease (38) 
Lapat, Tras (phase III) EGFR/HER2 Tras-resistant metastatic HER2-positive Lapat + Tras delayed progression, was superior to Lapat-alone for PFS (HR = 0.73; 95% CI: 0.57–0.93; P = 0.008) and CBR (24.7% vs. 12.4%) (40) 
Lapat or Tras (phase II neoadjuvant) HER2/EGFR Early-stage Lapat suppressed Ki67 proliferation index in breast tissue approximately 20% after 4 wk; Tras did not suppress Ki67 index (45) 
Lapat vs. Tras (phase III neoadjuvant GeparQuinto trial) EGFR/HER2 Early-stage pCR to Lapat (21.7%) and Tras (31.3%; P < 0.03) (37) 
Lapat, Tras, Lapat + Tras (phase III neoadjuvant NeoALTTO trial) EGFR/HER2 Early-stage pCR was 51.3% for Lapat + Tras (+chemotherapy) vs. 24.7% for Lapat-alone (+chemotherapy) and 29.5% for Tras-alone (+chemotherapy; P ≤ 0.0001 vs. lapatinib + Tras combination) (42) 
Tras + docetaxel vs. Tras + pertuzumab + docetaxel vs. Tras + pertuzumab vs. pertuzumab + docetaxel (4-arm phase II neoadjuvant NeoSPHERE trial) EGFR/HER2 Early-stage, HER2-positive Higher pCR with Tras/pertuzumab/docetaxel (46%) vs. Tras/docetaxel (29%; P = 0.014), and higher than in the Tras/pertuzumab arm (18%; P = 0.019) (32) 
Tras (single-dose, presurgery) HER2 DCIS No pathologic response or change in Ki67 staining (vs. pretreatment) (44) 
Lapat + Tras (phase II neoadjuvant) HER2/EGFR Early-stage, HER2-positive Lapat + Tras (without chemotherapy) produced a high pCR rate (overall 28%), especially in ER-negative disease (40%) (43) 
Lapat (phase II presurgery) HER2/EGFR Early-stage, HER2-positive Reduced proliferation (Ki67) in cancer (P = 0.008), DH (P = 0.006), and DIN (P = 0.067) (4) 
Drug/trial designTargetSettingResultsRefs.
Tras (I-line, randomized phase II) HER2 Metastatic HER2-positive I-line Tras produced a 26% objective response rate; response rate in 3+ HER2 overexpression was 35%, with a CBR of 48% (26) 
Tras (phase III HERA trial; initial results) HER2 Early-stage, HER2-positive Tras for 1 y after adjuvant chemotherapy improved DFS; Tras HR (vs. observation) = 0.54 (95% CI: 0.43–0.67; P < 0.0001) (49) 
Tras (longer-term HERA results) HER2 Early-stage, HER2-positive Tras for 1 y after adjuvant chemotherapy improved overall survival—HR for risk of death (vs. observation) was 0.66 (95% CI: 0.47–0.91; P = 0.0115) (27) 
Tras (phase III neo-adjuvant) HER2 HER2-positive Tras + chemotherapy (paclitaxel followed by FEC) improved pCR (65.2%) vs. chemotherapy alone (26%; P = 0.016) (50)  
Tras + chemotherapy (two phase III adjuvant trials) HER2 Early-stage, HER2-positive Dramatically prolonged median DFS–HR = 0.48 (P < 0.0001), and produced 33% reduced risk of death (P = 0.015) (29) 
Pertuzumab (phase I) HER2 Metastatic cancer PR in 2 patients (nonbreast cancer); stable disease lasting >2.5 m in 6 patients (31) 
Tras-DM1 (phase II) HER2 Metastatic HER2-positive, resistant to HER2 therapy Produced 26% objective response rate (33) 
Gefitinib (phase II) EGFR Advanced/metastatic 0% CR + PR (34) 
Erlotinib (phase II) EGFR Advanced/metastatic 3% PR (35) 
Lapat (phase III) EGFR/HER2 Metastatic HER2-positive Lapat + capecitabine prolonged TTP (vs. capecitabine; HR = 0.49; 95% CI: 0.34–0.71; P < 0.001) (37) 
Lapat (phase II) EGFR/HER2 Refractory, metastatic Modest activity (6% clinical benefit) (51) 
Lapat (1st-line phase III) EGFR/HER2 Metastatic Lapat + paclitaxel improved TTP (HR = 0.53; 95% CI: 0.31–0.89; P = 0.005) and event-free survival (HR = 0.52; 95% CI: 0.31–0.86; P = 0.004)–all vs. placebo + paclitaxel–in HER2-positive disease (9) 
Lapat + letrozole (1st-line phase III) EGFR/HER2 Metastatic, hormone- receptor–positive Letrozole + lapat reduced risk of disease progression vs. letrozole-alone in HER2-positive disease (HR = 0.71; 95% CI: 0.53–0.96; P = 0.019); no improved outcome in HER2-negative cancer (10) 
Lapat (phase II neoadjuvant) EGFR/HER2 Inflammatory breast cancer Lapat monotherapy followed by Lapat + paclitaxel produced a 78% clinical response rate in HER2-positive disease (38) 
Lapat, Tras (phase III) EGFR/HER2 Tras-resistant metastatic HER2-positive Lapat + Tras delayed progression, was superior to Lapat-alone for PFS (HR = 0.73; 95% CI: 0.57–0.93; P = 0.008) and CBR (24.7% vs. 12.4%) (40) 
Lapat or Tras (phase II neoadjuvant) HER2/EGFR Early-stage Lapat suppressed Ki67 proliferation index in breast tissue approximately 20% after 4 wk; Tras did not suppress Ki67 index (45) 
Lapat vs. Tras (phase III neoadjuvant GeparQuinto trial) EGFR/HER2 Early-stage pCR to Lapat (21.7%) and Tras (31.3%; P < 0.03) (37) 
Lapat, Tras, Lapat + Tras (phase III neoadjuvant NeoALTTO trial) EGFR/HER2 Early-stage pCR was 51.3% for Lapat + Tras (+chemotherapy) vs. 24.7% for Lapat-alone (+chemotherapy) and 29.5% for Tras-alone (+chemotherapy; P ≤ 0.0001 vs. lapatinib + Tras combination) (42) 
Tras + docetaxel vs. Tras + pertuzumab + docetaxel vs. Tras + pertuzumab vs. pertuzumab + docetaxel (4-arm phase II neoadjuvant NeoSPHERE trial) EGFR/HER2 Early-stage, HER2-positive Higher pCR with Tras/pertuzumab/docetaxel (46%) vs. Tras/docetaxel (29%; P = 0.014), and higher than in the Tras/pertuzumab arm (18%; P = 0.019) (32) 
Tras (single-dose, presurgery) HER2 DCIS No pathologic response or change in Ki67 staining (vs. pretreatment) (44) 
Lapat + Tras (phase II neoadjuvant) HER2/EGFR Early-stage, HER2-positive Lapat + Tras (without chemotherapy) produced a high pCR rate (overall 28%), especially in ER-negative disease (40%) (43) 
Lapat (phase II presurgery) HER2/EGFR Early-stage, HER2-positive Reduced proliferation (Ki67) in cancer (P = 0.008), DH (P = 0.006), and DIN (P = 0.067) (4) 

NOTE: Generally defined as CR or PR or as stable disease lasting 6 months.

Abbreviations: Tras, trastuzumab; CBR, clinical benefit rate; HERA, Herceptin adjuvant; DFS, disease-free survival; FEC, fluorouracil, epirubicin, cyclophosphamide; pCR, pathologic complete remission; PR, partial response; Lapat, lapatinib; TTP, time to progression; MTD, maximum tolerated dose; PFS, progression-free survival.

Two new anti-HER2 antibodies have been tested clinically with promising results. Pertuzumab functions by inhibiting heterodimerization of HER2 with HER3. This antibody suppresses the growth of breast tumors, including tumors that do not have HER2 overexpression, in mouse xenograft experiments (30), was active in early-phase clinical trials (31) and is now being tested clinically in combination with trastuzumab with encouraging results in the Neoadjuvant Study of Pertuzumab and Herceptin in an Early Regimen Evaluation (NeoSPHERE) trial (32). Trastuzumab–DM1 is a HER2 antibody drug conjugate in which trastuzumab is fused to the antimicrotubule agent DM1. Trastuzumab–DM1 is effective in treating HER2-positive metastatic breast cancer that is resistant to trastuzumab or lapatinib (33).

In addition to these intravenous monoclonal antibodies, oral anti-EGFR therapies have also been tested for the treatment of breast cancer. Erlotinib and gefitinib have been tested in metastatic breast cancer, where they have had limited activity (34, 35). The dual tyrosine kinase inhibitor lapatinib was highly active in combination with chemotherapy for treating HER2-positive metastatic breast cancer (36, 37) and in inflammatory breast cancer, which is an aggressive, biologically distinct form with a higher frequency of HER2 overexpression versus other breast cancer (38, 39). Recent therapeutic trials show that lapatinib plus trastuzumab delayed tumor progression in patients who had previously failed on trastuzumab alone (40). This effect may be due to the ability of lapatinib to inhibit EGFR (ErbB1) dimers (Fig. 1B). This clinical activity of lapatinib in the metastatic setting led to FDA approval of lapatinib in combination with capecitabine for the treatment of HER2-positive breast cancer and also provided the rationale for testing lapatinib in the clinical setting of early-stage, HER2-positive breast cancer, which is currently ongoing in the Tykerb Evaluation after Chemotherapy (TEACH) trial.

Several trials have tested anti-HER2 therapies in the neoadjuvant setting. Results from the phase III neoadjuvant GeparQuinto and Neoadjuvant Lapatinib and/or Trastuzumab Treatment Optimisation (NeoALTTO) trials, both comparing lapatinib and trastuzumab, were presented at the San Antonio Breast Cancer Symposium in December 2010. The GeparQuinto trial compared lapatinib plus standard chemotherapy with trastuzumab plus standard chemotherapy given prior to definitive surgery. The NeoALTTO trial compared standard chemotherapy plus lapatinib and trastuzumab with standard chemotherapy plus either lapatinib or trastuzumab. The primary endpoint for both trials was pathologic complete response at the time of surgery.

The GeparQuinto trial showed that both lapatinib and trastuzumab induced pathologic complete responses, although trastuzumab was significantly more active (31.3%) than with lapatinib (21.7%; P < 0.03; ref. 41). The NeoALTTO trial showed that the pathologic complete response rate was 51.3% in the lapatinib plus trastuzumab (plus standard chemotherapy) arm versus 24.7% in the lapatinib (plus standard chemotherapy) arm and 29.5% in the trastuzumab (plus standard chemotherapy) arm, with the first (lapatinib plus trastuzumab) being superior to the last (trastuzumab; P ≤ 0.0001). The complete response rates of chemotherapy with lapatinib alone (24.7%) and with trastuzumab alone (29.5%) were not statistically significantly different (P = 0.34; ref. 42). Assessments of safety, overall survival, and disease-free survival are ongoing in both trials. Future reports from these 2 important studies will help clarify the use of anti-HER2 therapy in treating breast cancer in the neoadjuvant setting.

Trastuzumab plus lapatinib without chemotherapy has also been tested clinically. The preliminary report of a multicenter phase II trial showed that trastuzumab plus lapatinib produced a high pathologic response rate in locally advanced HER2-positive breast cancer—28% overall (21% in ER-positive and 40% in ER-negative disease; ref. 43).

All of the previously discussed clinical studies were for treating invasive breast cancer. Recent clinical studies have also tested anti-HER2 drugs for breast cancer prevention. Kuerer and colleagues studied the effect of a single dose of trastuzumab given 14 to 28 days prior to surgical resection in women with DCIS lesions (44). There was no pathologic response or change in Ki67 staining (as compared with pretreatment), but there was evidence of significantly augmented antibody-dependent cell-mediated cytotoxicity in 100% of the trastuzumab-treated women. Of note, the DCIS showed HER2 overexpression in only 24 of 69 enrolled patients (35%). The substantial proportion of subjects with HER2-negative DCIS may have accounted for the lack of reduction in Ki67 staining.

DeCensi and colleagues tested a short (3 weeks) course of lapatinib for activity in ductal intraepithelial neoplasia [DIN; comprising atypical ductal hyperplasia (ADH) and DCIS], ductal hyperplasia (DH) without atypia, and invasive breast tissue in a randomized, placebo-controlled phase II presurgical trial in early-stage, HER2-positive breast cancer patients (4). Lapatinib reduced Ki67 in breast cancer tissue by 9.3% (compared with pre–lapatinib treatment), whereas placebo treatment was associated with a 15% increase in proliferation (vs. pretreatment; P = 0.008); these findings are consistent with other recent short-term lapatinib data (45). The antiproliferative effect of lapatinib (4) was statistically significant in ER-negative/PR-negative tumors but was only a trend in ER-positive/PR-positive tumors. These results suggest that HER2-positive, ER-negative tumors are more sensitive to lapatinib (vs. HER2-positive, ER-positive tumors) which is supported by some evidence from the therapeutic setting (43, 46). In addition this study suggested that phosphatase and tensin homolog (PTEN) overexpression was a potential predictive marker, consistent with other preclinical and clinical data on phosphoinositide 3-kinase (PI3K) pathway activation (47). Src pathway activation in preclinical and clinical studies is also a promising predictive marker for lapatinib (48).

Regarding effects in preinvasive tissue surrounding breast cancer in the DeCensi and colleagues trial, lapatinib treatment did not affect the prevalence of DIN lesions but did produce a trend toward reduced proliferation in DIN cells (Ki67 labeling index of 17.0%) versus placebo (23.4%; P = 0.067) and significantly reduced proliferation in DH cells (1.8%) versus placebo (2.5%; P = 0.006). These results support the hypothesis that lapatinib will suppress the progression of DH and DIN (ADH and DCIS) lesions to invasive breast cancer in humans, also supported by relevant preclinical results in MMTV/neu mice (18). An intriguing, unanswered question is whether cells of these DH and DIN lesions overexpressed HER2 or EGFR, an issue relevant to the development of predictive markers for prevention about which DeCensi and colleagues do not comment. Such results would be particularly useful for future testing of lapatinib in women with DIN or DH lesions. Despite this caveat, the results of DeCensi and colleagues provide strong support for testing lapatinib and other oral receptor tyrosine kinase inhibitors for the prevention of HER2-overexpressing breast cancer.

Including very recent preclinical and clinical data (4, 5), the growing body of work on HER family targeting certainly advances the field of treatment and prevention of EGFR- and HER2-positive breast cancer, including both ER-negative and -positive disease. Given the strong antiproliferative and anticancer activity of lapatinib and its generally acceptable toxicity profile, it is time to test lapatinib and other HER2-targeting drugs for breast cancer prevention in women at a high risk of this disease. The most appropriate population for prevention with anti-HER2 drugs would be women with HER2-positive DCIS lesions. Indeed, the NSABP is currently testing trastuzumab in an ongoing phase III trial in women with HER2-positive DCIS (NSABP B-43, NCT00769379), and investigators at MD Anderson Cancer Center are conducting a phase II multicenter presurgical trial of lapatinib in patients with EGFR- or HER2-positive DCIS (NCT00555152). These studies should provide further evidence bearing on the utility of anti-HER2 therapy for preventing invasive breast cancer. Another unanswered question is whether lapatinib will prevent the development of cancers that do not overexpress HER2. Li and colleagues provide provocative data, suggesting that lapatinib may prevent tumors that do not overexpress HER2 (presumably through its effect on EGFR and/or lower levels of HER2; ref. 5). To address this issue, it will be necessary in the future to conduct clinical trials testing lapatinib's effect on non–HER2-overexpressing DCIS.

Predictive markers (as discussed above) and risk markers remain a very important issue for future clinical prevention trials of lapatinib and other HER family–targeting drugs. Risk models integrating family history, breast density, target-tissue markers, and germ line changes (e.g., BRCA mutations and single-nucleotide polymorphisms) are helping to identify high breast cancer risk. The overall data on breast cancer prevention with estrogen-targeting and HER family–targeting agents suggest the possibility of developing combinations that may prevent ER-negative breast cancer and increase prevention of DCIS and ER-positive invasive disease.

P.H. Brown is a consultant/advisory Board for Susan G. Komen for the Cure organization.

P.H. Brown acknowledges the assistance of Ms. Kimberly Young-Jenkins and Michelle Savage in the preparation of this manuscript.

This work was supported in part by NIH grants R01 CA078480 (to P.H. Brown) and R01 CA131219 (to L.R. Howe) and a grant from the Breast Cancer Research Foundation (to P.H. Brown).

1.
Cuzick
J
,
DeCensi
A
,
Arun
B
,
Brown
PH
,
Castiglione
M
,
Dunn
B
, et al
Preventive therapy for breast cancer: a consensus statement
.
Lancet Oncol
2011
;
12
:
496
503
.
2.
Vogel
VG
,
Costantino
JP
,
Wickerham
DL
,
Cronin
WM
,
Cecchini
RS
,
Atkins
JN
, et al
Update on the national surgical adjuvant breast and bowel project study of tamoxifen and raloxifene (STAR) P-2 trial: preventing breast cancer
.
Cancer Prev Res
2010
;
3
:
696
706
.
3.
Goss
PE
,
Ingle
JN
,
Alés-Martínez
JE
,
Cheung
AM
,
Chlebowski
RT
,
Wactawski-Wende
J
, et al
Exemestane for breast-cancer prevention in postmenopausal women
.
N Engl J Med
2011
;
25
:
2381
91
.
4.
DeCensi
A
,
Puntoni
M
,
Pruneri
G
,
Guerrieri-Gonzaga
A
,
Lazzeroni
M
,
Serrano
D
, et al
Lapatinib activity in premalignant lesions and HER2-positive cancer of the breast in a randomized, placebo-controlled presurgical trial
.
Cancer Prev Res
2011
;
4
:
1181
9
.
5.
Li
J
,
Cho
Y-Y
,
Langfald
A
,
Carper
A
,
Lubet
RA
,
Grubbs
CJ
, et al
Lapatinib, a preventive/therapeutic agent against mammary cancer, suppresses RTK-mediated signaling through multiple signaling pathways
.
Cancer Prev Res
2011
;
4
:
1190
7
.
6.
Arteaga
CL
,
Moulder
SL
,
Yakes
FM
. 
HER (erbB) tyrosine kinase inhibitors in the treatment of breast cancer
.
Semin Oncol
2002
;
29
Suppl 11
:
4
10
.
7.
Ryan
Q
,
Ibrahim
A
,
Cohen
MH
,
Johnson
J
,
Ko
CW
,
Sridhara
R
, et al
FDA drug approval summary: lapatinib in combination with capecitabine for previously treated metastatic breast cancer that overexpresses HER-2
.
Oncologist
2008
;
13
:
1114
9
.
8.
Burstein
HJ
,
Storniolo
AM
,
Franco
S
,
Forster
J
,
Stein
S
,
Rubin
S
, et al
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
74
.
9.
Di Leo
A
,
Gomez
HL
,
Aziz
Z
,
Zvirbule
Z
,
Bines
J
,
Arbushites
MC
, et al
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
52
.
10.
Johnston
S
,
Pippen
J
,
Pivot
X
,
Lichinitser
M
,
Sadeghi
S
,
Dieras
V
, et al
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
46
.
11.
Press
MF
,
Finn
RS
,
Cameron
D
,
Di Leo
A
,
Geyer
CE
,
Villalobos
IE
, et al
HER-2 gene amplification, HER-2 and epidermal growth factor receptor mRNA and protein expression, and lapatinib efficacy in women with metastatic breast cancer
.
Clin Cancer Res
2008
;
14
:
7861
70
.
12.
Bartkova
J
,
Barnes
DM
,
Millis
RR
,
Gullick
WJ
. 
Immunohistochemical demonstration of c-erbB-2 protein in mammary ductal carcinoma in situ
.
Hum Pathol
1990
;
21
:
1164
7
.
13.
Park
K
,
Han
S
,
Kim
HJ
,
Kim
J
,
Shin
E
. 
HER2 status in pure ductal carcinoma in situ and in the intraductal and invasive components of invasive ductal carcinoma determined by fluorescence in situ hybridization and immunohistochemistry
.
Histopathology
2006
;
48
:
702
7
.
14.
Ramachandra
S
,
Machin
L
,
Ashley
S
,
Monaghan
P
,
Gusterson
BA
. 
Immunohistochemical distribution of c-erbB-2 in in situ breast carcinoma–a detailed morphological analysis
.
J Pathol
1990
;
161
:
7
14
.
15.
Lenferink
AE
,
Simpson
JF
,
Shawver
LK
,
Coffey
RJ
,
Forbes
JT
,
Arteaga
CL
, et al
Blockade of the epidermal growth factor receptor tyrosine kinase suppresses tumorigenesis in MMTV/Neu + MMTV/TGF-alpha bigenic mice
.
Proc Natl Acad Sci U S A
2000
;
97
:
9609
14
.
16.
Lu
C
,
Speers
C
,
Zhang
Y
,
Xu
X
,
Hill
J
,
Steinbis
E
, et al
Effect of epidermal growth factor receptor inhibitor on development of estrogen receptor-negative mammary tumors
.
J Natl Cancer Inst
2003
;
95
:
1825
33
.
17.
Piechocki
MP
,
Dibbley
SK
,
Lonardo
F
,
Yoo
GH
. 
Gefitinib prevents cancer progression in mice expressing the activated rat HER2/neu
.
Int J Cancer
2008
;
122
:
1722
9
.
18.
Strecker
TE
,
Shen
Q
,
Zhang
Y
,
Hill
JL
,
Li
Y
,
Wang
C
, et al
Effect of lapatinib on the development of estrogen receptor-negative mammary tumors in mice
.
J Natl Cancer Inst
2009
;
101
:
107
13
.
19.
Lubet
RA
,
Szabo
E
,
Christov
K
,
Bode
AM
,
Ericson
ME
,
Steele
VE
, et al
Effects of gefitinib (Iressa) on mammary cancers: preventive studies with varied dosages, combinations with vorozole or targretin, and biomarker changes
.
Mol Cancer Ther
2008
;
7
:
972
9
.
20.
Konecny
GE
,
Pegram
MD
,
Venkatesan
N
,
Finn
R
,
Yang
G
,
Rahmeh
M
, et al
Activity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells
.
Cancer Res
2006
;
66
:
1630
9
.
21.
Xia
W
,
Liu
LH
,
Ho
P
,
Spector
NL
. 
Truncated ErbB2 receptor (p95ErbB2) is regulated by heregulin through heterodimer formation with ErbB3 yet remains sensitive to the dual EGFR/ErbB2 kinase inhibitor GW572016
.
Oncogene
2004
;
23
:
646
53
.
22.
Jin
Q
,
Esteva
FJ
. 
Cross-talk between the ErbB/HER family and the type I insulin-like growth factor receptor signaling pathway in breast cancer
.
J Mammary Gland Biol Neoplasia
2008
;
13
:
485
98
.
23.
Saxena
NK
,
Taliaferro-Smith
L
,
Knight
BB
,
Merlin
D
,
Anania
FA
,
O'Regan
RM
, et al
Bidirectional crosstalk between leptin and insulin-like growth factor-I signaling promotes invasion and migration of breast cancer cells via transactivation of epidermal growth factor receptor
.
Cancer Res
2008
;
68
:
9712
22
.
24.
Burga
LN
,
Hu
H
,
Juvekar
A
,
Tung
NM
,
Troyan
SL
,
Hofstatter
EW
, et al
Loss of BRCA1 leads to an increase in epidermal growth factor receptor expression in mammary epithelial cells, and epidermal growth factor receptor inhibition prevents estrogen receptor-negative cancers in BRCA1-mutant mice
.
Breast Cancer Res
2011
;
13
:
R30
.
25.
Antoniou
AC
,
Wang
X
,
Fredericksen
ZS
,
McGuffog
L
,
Tarrell
R
,
Sinilnikova
OM
, et al
A locus on 19p13 modifies risk of breast cancer in BRCA1 mutation carriers and is associated with hormone receptor-negative breast cancer in the general population
.
Nat Genet
2010
;
42
:
885
92
.
26.
Vogel
CL
,
Cobleigh
MA
,
Tripathy
D
,
Gutheil
JC
,
Harris
LN
,
Fehrenbacher
L
, et al
Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer
.
J Clin Oncol
2002
;
20
:
719
26
.
27.
Smith
I
,
Procter
M
,
Gelber
RD
,
Guillaume
S
,
Feyereislova
A
,
Dowsett
M
, et al
2-year follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer: a randomised controlled trial
.
Lancet
2007
;
369
:
29
36
.
28.
Buzdar
AU
,
Valero
V
,
Ibrahim
NK
,
Francis
D
,
Broglio
KR
,
Theriault
RL
, et al
Neoadjuvant therapy with paclitaxel followed by 5-fluorouracil, epirubicin, and cyclophosphamide chemotherapy and concurrent trastuzumab in human epidermal growth factor receptor 2-positive operable breast cancer: an update of the initial randomized study population and data of additional patients treated with the same regimen
.
Clin Cancer Res
2007
;
13
:
228
33
.
29.
Romond
EH
,
Perez
EA
,
Bryant
J
,
Suman
VJ
,
Geyer
CE
,
Davidson
NE
, et al
Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer
.
N Engl J Med
2005
;
353
:
1673
84
.
30.
Friess
T
,
Scheuer
W
,
Hasmann
M
. 
Combination treatment with erlotinib and pertuzumab against human tumor xenografts is superior to monotherapy
.
Clin Cancer Res
2005
;
11
:
5300
9
.
31.
Agus
DB
,
Gordon
MS
,
Taylor
C
,
Natale
RB
,
Karlan
B
,
Mendelson
DS
, et al
Phase I clinical study of pertuzumab, a novel HER dimerization inhibitor, in patients with advanced cancer
.
J Clin Oncol
2005
;
23
:
2534
43
.
32.
Spector
NL
. 
Drug development—targeting HER2 beyond trastuzumab [abstract]
.
In
:
Proceedings of 33rd Annual San Antonio Breast Cancer Symposium
;
2010 Dec 8–12
;
San Antonio, TX. Philadelphia (PA)
:
AACR
; 
2010
PL2-1
.
33.
Burris
HA
 III
,
Rugo
HS
,
Vukelja
SJ
,
Vogel
CL
,
Borson
RA
,
Limentani
S
, et al
Phase II study of the antibody drug conjugate trastuzumab-DM1 for the treatment of human epidermal growth factor receptor 2 (HER2)-positive breast cancer after prior HER2-directed therapy
.
J Clin Oncol
2011
;
29
:
398
405
.
34.
Baselga
J
,
Albanell
J
,
Ruiz
A
,
Lluch
A
,
Gascón
P
,
Guillém
V
, et al
Phase II and tumor pharmacodynamic study of gefitinib in patients with advanced breast cancer
.
J Clin Oncol
2005
;
23
:
5323
33
.
35.
Dickler
MN
,
Cobleigh
MA
,
Miller
KD
,
Klein
PM
,
Winer
EP
. 
Efficacy and safety of erlotinib in patients with locally advanced or metastatic breast cancer
.
Breast Cancer Res Treat
2009
;
115
:
115
21
.
36.
Burris
HA
 III
,
Hurwitz
HI
,
Dees
EC
,
Dowlati
A
,
Blackwell
KL
,
O'Neil
B
, et al
Phase I safety, pharmacokinetics, and clinical activity study of lapatinib (GW572016), a reversible dual inhibitor of epidermal growth factor receptor tyrosine kinases, in heavily pretreated patients with metastatic carcinomas
.
J Clin Oncol
2005
;
23
:
5305
13
.
37.
Geyer
CE
,
Forster
J
,
Lindquist
D
,
Chan
S
,
Romieu
CG
,
Pienkowski
T
, et al
Lapatinib plus capecitabine for HER2-positive advanced breast cancer
.
N Engl J Med
2006
;
355
:
2733
43
.
38.
Boussen
H
,
Cristofanilli
M
,
Zaks
T
,
DeSilvio
M
,
Salazar
V
,
Spector
N
, et al
Phase II study to evaluate the efficacy and safety of neoadjuvant lapatinib plus paclitaxel in patients with inflammatory breast cancer
.
J Clin Oncol
2010
;
28
:
3248
55
.
39.
Kaufman
B
,
Trudeau
M
,
Awada
A
,
Blackwell
K
,
Bachelot
T
,
Salazar
V
, et al
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
8
.
40.
Blackwell
KL
,
Burstein
HJ
,
Storniolo
AM
,
Rugo
H
,
Sledge
G
,
Koehler
M
, et al
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
30
.
41.
Untch
M
,
Loibl
S
,
Bischoff
J
,
Eidtmann
H
,
Kaufmann
M
,
Blohmer
JU
, et al
Lapatinib vs trastuzumab in combination with neoadjuvant anthracycline-taxane-based chemotherapy: primary efficacy endpoint analysis of the GEPARQUINTO STUDY (GBG 44). [abstract]
.
In
:
Proceedings of 33rd Annual San Antonio Breast Cancer Symposium; 2010 Dec 8–12;
San Antonio, TX
.
Philadelphia (PA): AACR
; 
2010
S3
1
.
42.
Baselga
J
,
Bradbury
I
,
Eidtmann
H
,
Di Cosimo
S
,
Aura
C
,
De Azambuja
E
, et al
First results of the NeoALTTO trial (BIG 01-06/EGF 106903): a phase III, randomized, open label, neoadjuvant study of lapatinib, trastuzumab, and their combination plus paclitaxel in women with HER2-positive primary breast cancer. [abstract]
.
In
:
Proceedings of 33rd Annual San Antonio Breast Cancer Symposium
; 
2010
Dec 8–12; San Antonio, TX. Philadelphia (PA): AACR; 2010 S3–3
.
43.
Chang
JC
,
Mayer
IA
,
Forero-Torres
A
,
Nanda
R
,
Goetz
MP
,
Rodriguez
AA
, et al
TBCRC 006: a multicenter phase II study of neoadjuvant lapatinib and trastuzumab in patients with HER2-overexpressing breast cancer
.
J Clin Oncol
2011
;
29
Suppl. Abstract 505
.
44.
Kuerer
HM
,
Buzdar
AU
,
Mittendorf
EA
,
Esteva
FJ
,
Lucci
A
,
Vence
LM
, et al
Biologic and immunologic effects of preoperative trastuzumab for ductal carcinoma in situ of the breast
.
Cancer
2011
;
117
:
39
47
.
45.
Dave
B
,
Migliaccio
I
,
Gutierrez
MC
,
Wu
MF
,
Chamness
GC
,
Wong
H
, et al
Loss of phosphatase and tensin homolog or phosphoinositol-3 kinase activation and response to trastuzumab or lapatinib in human epidermal growth factor receptor 2-overexpressing locally advanced breast cancers
.
J Clin Oncol
2011
;
29
:
166
73
.
46.
Finn
RS
,
Press
MF
,
Dering
J
,
Arbushites
M
,
Koehler
M
,
Oliva
C
, et al
Estrogen receptor, progesterone receptor, human epidermal growth factor receptor 2 (HER2), and epidermal growth factor receptor expression and benefit from lapatinib in a randomized trial of paclitaxel with lapatinib or placebo as first-line treatment in HER2-negative or unknown metastatic breast cancer
.
J Clin Oncol
2009
;
27
:
3908
15
.
47.
Wang
L
,
Zhang
Q
,
Sun
S
,
Guo
H
,
Jia
Z
,
Wang
B
, et al
PI3K pathway activation results in low efficacy of both trastuzumab and lapatinib
.
BMC Cancer
2011
;
11
:
248
.
48.
Rexer
BN
,
Ham
AJ
,
Rinehart
C
,
Hill
S
,
de Matos Granja-Ingram
N
,
González-Angulo
AM
, et al
Phosphoproteomic mass spectrometry profiling links Src family kinases to escape from HER2 tyrosine kinase inhibition
.
Oncogene
2011
Apr 18
.
[Epub ahead of print]
.
49.
Piccart-Gebhart
MJ
,
Procter
M
,
Leyland-Jones
B
,
Goldhirsch
A
,
Untch
M
,
Smith
I
, et al
Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer
.
N Engl J Med
2005
;
353
:
1659
72
.
50.
Buzdar
AU
,
Ibrahim
NK
,
Francis
D
,
Booser
DJ
,
Thomas
ES
,
Theriault
RL
, et al
Significantly higher pathologic complete remission rate after neoadjuvant therapy with trastuzumab, paclitaxel, and epirubicin chemotherapy: results of a randomized trial in human epidermal growth factor receptor 2-positive operable breast cancer
.
J Clin Oncol
2005
;
23
:
3676
85
.
51.
Burstein
HJ
,
Storniolo
AM
,
Franco
S
,
Forster
J
,
Stein
S
,
Rubin
S
, et al
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
74
.