Abstract
The anti-HER2 drugs trastuzumab and lapatinib are increasingly changing the natural history of early and metastatic HER2-overexpressing breast cancer. Many other agents targeted against the HER2 signaling network are in clinical development. These are or will soon be combined with the currently approved anti-HER2 therapies. We review herein recent data in support of the early use of combinations of agents targeted to the HER2 network as the most rational approach against this subtype of breast cancer. We propose the optimal combination or combinations of anti-HER2 agents delivered early in the natural history of HER2+ breast cancer should close to eliminate acquired drug resistance, shorten the duration of therapy, and potentially dispense with the need of concurrent chemotherapy.
Background
The antibody trastuzumab and the tyrosine kinase inhibitor (TKI) lapatinib are approved by the FDA for the treatment of HER2-overexpressing (HER2+) breast cancer (MBC). Trastuzumab binds to an epitope in the juxtamembrane region of the HER2 receptor. This binding induces uncoupling of ligand-independent HER2-HER3 heterodimers and inhibition of downstream signaling (1) as well as antibody-dependent, cell-mediated cytotoxicity (ADCC)(2). Several randomized adjuvant trials (NCCTG N9831, NSABP B-31, BCIRG 006, and HERA), have demonstrated that the addition of trastuzumab to standard chemotherapy reduces disease recurrence and the risk of death compared to chemotherapy alone in patients with surgically-resected tumors (3–5). In N-9831, a recent interim analysis showed that the benefit of concurrent trastuzumab and chemotherapy was more pronounced than that of chemotherapy followed by trastuzumab (6). Based on these data, the addition of trastuzumab to adjuvant chemotherapy has become standard of care in women with HER2+ early breast cancer.
The trastuzumab adjuvant trials focused on high-risk, lymph node positive HER2+ tumors. Thus, there is limited to no data on small tumors (≤1 cm) with negative nodes and patient outcome. However, two recent studies found a significantly higher rate of recurrence among T1abN0 HER2+ compared to HER2-negative tumors regardless of ER status (7, 8), suggesting adjuvant trastuzumab should be considered for these patients. However, the amount and type of chemotherapy to combine with the antibody in this setting is undetermined. Most of the adjuvant trials used one year of trastuzumab. One study delivered only 9 weeks of the antibody, whereas the HERA trial included an arm where it was given for 2 years. In the first study, patients in the trastuzumab arm exhibited fewer overall recurrences and improved overall survival compared to patients treated with chemotherapy alone (9). Results in the 2-year arm in HERA are pending.
The dual EGFR/HER2 TKI lapatinib is active as first line monotherapy in patients with HER2+ MBC and in combination with capecitabine improves progression free survival compared to capecitabine alone (10, 11). In the latter registration trial, fewer brain metastases occurred in women in the combination than in the monotherapy arm, suggesting a potential difference between lapatinib and trastuzumab as it applies to recurrences in the CNS (11). In the registration study and in a second randomized trial of paclitaxel ± lapatinib in patients with MBC, the clinical benefit of lapatinib was limited to patients with HER2 overexpression by IHC and/or FISH (12).
On the Horizon
HER2 testing, discordance, and conversion
The clinical activity of anti-HER2 agents has been limited to patients with HER2+ tumors as defined by intense membrane staining with HER2 antibodies in the majority of tumor cells (3+ by immunohistochemistry – IHC) or ≥2 copies of the HER2 gene determined by fluorescent in situ hybridization (FISH). In general, HER2 IHC and FISH correlate with each other (13–15). FISH appears superior to IHC to reproducibly assess tumors for HER2 overexpression at outside/local laboratories for entry into clinical trials (16). Intrinsic limitations of IHC are the variability in fixation methods and the impact of fixation of antigenicity of the HER2 protein. Conversely, the more stable DNA, whose loci are measured by FISH, is less succeptible to tissue fixation. For these reasons, excess copies of the HER2 gene (so called HER2 positivity) defined by FISH has gained ground as the standard to define odds of tumor dependence on HER2 and, therefore, response to HER2 antagonists (17).
A reanalysis in a central laboratory of NSABP B-31 showed that 9.7% of patients enrolled on the basis of a test performed in a local laboratory had tumors that did not meet criteria for HER2 amplification by FISH or IHC (18). Notably, these patients also benefited from trastuzumab. This suggests that the local laboratory was correct and/or there is discordance in the levels of HER2 expression between micrometastases, whose clinical recurrence defines the endpoint of adjuvant trials, and the primary tumor, where the HER2 alteration was measured. This possibility is further suggested by a study where 9/24 patients with breast cancer whose primary tumor was HER2-negative (HER2−) acquired HER2 amplification in their circulating tumor cells (CTCs) during cancer progression (19). In another study, 10% of patients that recurred on adjuvant tamoxifen converted from HER2− to HER2+ in the relapsing tumor (20). Of note, however, the HER2 status of CTCs has yet to be linked to clinical outcome. Based on data like these, the NSABP is initiating a phase III trial in which patients with 1+ or 2+ HER2 by IHC and no HER2 amplification by FISH will be randomized to adjuvant chemotherapy followed by one year of trastuzumab vs. placebo.
Several studies have shown changes in HER2 status in patients treated with trastuzumab. Pectasides et al. showed that 37% of patients with HER2+ primary tumors no longer exhibited HER2 amplification in their metastatic lesions. Further, these patients exhibited a shorter TTP than the group that remained HER2+ (21). Hurley et al. reported that following treatment with neoadjuvant trastuzumab and chemotherapy, 43% of HER2+ tumors became HER2− as measured by FISH (22). Finally, Mittendorf et al. also reported that 32% of HER2+ tumors treated with neoadjuvant chemotherapy and trastuzumab ‘converted’ to HER2− by FISH. Notably, at 37 months, relapse free survival (RFS) was statistically superior in patients whose residual tumors retained HER2 amplification (23). These results have several implications. First, the change in HER2 status may reflect heterogeneity in HER2 expression in the primary tumor; the anti-oncogene therapy eliminates the HER2+ compartment and enriches for HER2− clones. Second, patients with HER2+ breast cancer who relapse after adjuvant or neoadjuvant anti-HER2 therapy should considering having their recurrent disease biopsied for reassessment of the HER2 status. Third, patients with ER−/HER2+ tumors at diagnosis that ‘convert’ to HER2− after treatment are at high risk of early recurrence. Further, there are no clear adjuvant (targeted) therapy standards for these patients who as a result, may exhibit a poor outcome.
Antibody-chemotherapy conjugates
Trastuzumab (T)-DM1 is an antibody-drug conjugate in which one molecule of trastuzumab is covalently bonded via a non-cleavable linker to three molecules of the microtubule polymerization inhibitor derivative of maytansine 1, DM1 (24). T-DM1 binds to HER2 with similar affinity as trastuzumab. It is postulated that after binding, the T-DM1/HER2 complex is internalized followed by degradation in the lysosome, release of DM1, and subsequent cell lysis. Although used at lower doses and frequency than trastuzumab, T-DM1 retains the ability to inhibit signaling and engaging immune effectors that mediate ADCC and is active against lapatinib-resistant xenografts (25). Phase I–II studies of T-DM1 demonstrated mild, reversible toxicity and a remarkable clinical response rate in excess of 25% in patients with heavily pretreated HER2+ MBC who had progressed after trastuzumab and lapatinib (26, 27). T-DM1 is being further evaluated in two large phase III randomized studies. The first trial compares T-DM1 vs. T-DM1 plus pertuzumab vs. the standard of trastuzumab plus a taxane in patients with HER2+ MBC previously untreated in the metastatic setting. The second trial compares T-DM1 vs. the standard of lapatinib and capecitabine in similar patients but who have previously received trastuzumab (Table 1).
Table 1.
Planned and ongoing high impact randomized phase III trials in patients with HER2-overexpressing breast cancer.
| Trial | Design | Population | # of patients | Treatment | Trial endpoints | Data expected |
|---|---|---|---|---|---|---|
| TEACH | Phase III, adjuvant | Stage I–IIIC, s/p neoadjuvant chemotx with an anthracycline, a taxane, and CMF | 3000 | Placebo vs. Lapatinib 1500 mg/day for 12 months | DFS, RFS, DRFS, OS, rate of CNS recurrence, QoL | 2012 |
| ALTTO, BIG2-06/N063D | Phase III, adjuvant | Stage I–IIIC, s/p neoadjuvant anthracycline based chemotx (≥4 cycles) | 8000 | Standard chemotx with a) H, b) L, c) H x 12 weeks → L after 6-week washout, d) H+L (all regimens for up to 1 year) | DFS, OS, TTDR, TTR, CNS recurrence rate | 2013 |
| ExteNET | Phase III, adjuvant | Stage II–IIIC after adjuvant H for 1 year | 120 | Placebo vs. Neratinib | DFS | 2017 |
| NEFERTT | Phase III, MBC | HER2+, no prior therapy for MBC | 1200 | P + H vs. P + Neratinib | PFS, OS | 2012 |
| Neo-ALTTO | Phase III, neoadjuvant | HER2+ >2 cm | 450 | a) L x 6 wks →L+P x 12 wks vs. b) H x 6 wks → H+P x 12 wks vs. c) L+H x 6 wks → H+L+P x 12 wks; surgery; FECx3 → a) L vs. b) H vs. c) H+L to complete 1 year | pCR rate, ORR, DFS, OS | 2011 |
| NSABP B-41 | Phase III, neoadjuvant | HER2+ >2 cm | 522 | ACx4 → a) P+H x 12 wks vs. b) P+L x 12 wks vs. c) P+H+L x 12 wks | pCR rate, ORR, RFS, OS | 2010 |
| CALGB 40601 | Phase III, neoadjuvant | HER2+ Stage II–III | H+P vs. L+P vs. H+L+P →surgery → adjuvant chemotx | pCR rate, ORR, DFS, OS | 2011 | |
| Gepar-Quinto | Phase III, neoadjuvant | HER2+ requiring neoadjuvant chemotx | 2547 (whole trial) | EC → D → H vs. EC → D → L | pCR rate | 2010 |
| BETH, NSABP B-44 | Phase III, adjuvant | HER2+, LN+ or high-risk LN− | 3500 | TCH→H (up to 1 y) vs, TCH → H + bevacizumab (up to 1 y) | DFS, OS, RFS | 2012 |
| BO22589 | Phase III, MBC | HER2+, no prior therapy for MBC | 1092 | H + taxane vs. T-DM1 vs. T-DM1 + pertuzumab | PFS | 2012 |
| EMILIA | Phase III, MBC | HER2+ LABC or MBC | 580 | T-DM1 vs. Capecitabine + Lapatinib | PFS, OS, ORR | 2013 |
| CLEOPATRA | Phase III, MBC | HER2+, no prior therapy for MBC | 800 | Docetaxel + H + placebo vs. Docetaxel + H + pertuzumab | PFS | 2011 |
| PHEREXA | Phase II, MBC | HER2+ MBC, 2nd line after Herceptin | 450 | Capecitabine + H vs. Capecitabine + H + pertuzumab | PFS, TTP, TTF, ORR, CBR | 2015 |
| BOLERO-1 | Phase III, MBC | HER2+, no prior therapy for MBC | 717 | P + H vs. P + H + everolimus | PFS, OS, ORR, CBR, TTR | 2012 |
| BOLERO3 | Phase III, MBC | HER2+, resistant to H | 572 | Vinorelbine + H vs. Vinorelbine + H + everolimus | PFS, OS, ORR, CBR | 2012 |
H=Herceptin; L=Lapatinib; P=Paclitaxel; D=docetaxel; TC= docetaxel, carboplatin; TCH=docetaxel, carboplatin, herceptin; DFS=disease-free survival’ RFS=relapse-free survival; DRFS=distant relapse-free survival; OS=overall survival; QoL=quality of life; TTR=time to recurrence; TTDR=time to distant recurrence; DFS=disease-free survival; pCR=pathological complete response; ORR=overall response rate; PFS=progression-free survival; TTP=time to progression; TTF=time to treatment failure; CBR=clinical benefit rate; ALTTO=adjuvant lapatinib & trastuzumab treatment optimization; AC=adriamycin/taxol; LABC=locally advanced breast cancer; MBC=metastatic breast cancer; T-DM1=trastuzumab-derivative of mayntansine1; CMF=cyclophosphamide, methotrexate, 5-fluoruracil
Combination of anti-HER2 therapies and abrogation of drug resistance
Dual HER2 blockade
Many HER2-amplified breast cancers do not respond to or eventually escape trastuzumab, suggesting both de novo and acquired mechanisms of resistance. A possible mechanism of de novo resistance is expression of the HER2 receptor as a kinase-active 95-kDa cytosolic fragment that lacks the trastuzumab binding epitope (28). Analysis of a cohort of patients with HER2+ MBC treated with trastuzumab and chemotherapy showed a very low response rate in tumors with cytosolic p95HER2 compared to those without (29). Lapatinib has been shown to inhibit the catalytic activity of p95HER2. Therefore, patients with p95HER2-positive breast cancers treated with lapatinib alone or in combination with capecitabline exhibited a similar PFS and overall response rate compared to p95HER2-negative tumors (30), suggesting a clinical setting where a HER2 TKI might be advantageous.
Like lapatinib, the HER2/EGFR dual TKI neratinib (31) has shown clinical activity in patients with HER2+ MBC who have progressed on trastuzumab. These data suggest that trastuzumab-resistant tumors continue to be dependent on the HER2 tyrosine kinase. However, the response to each single agent TKI tends to be short-lived (10, 11). Further, these patients may still need trastuzumab beyond progression as suggested by a recent study where the combination of lapatinib and trastuzumab was superior to lapatinib alone at improving PFS, clinical response, and overall survival of patients with HER2+ MBC who had progressed on trastuzumab (32). Taken together, these data imply that even in advanced stages, HER2+ breast cancers remain dependent on HER2 and that single-agent trastuzumab and lapatinib are not adequate to inhibit the HER2 network completely. They also imply that using combinations of HER2-targeted agents delivered early against HER2+ breast cancer is the rational approach to take. Along these lines, Adjuvant Lapatinib and/or Trastuzumab Treatment Optimization (ALTTO) is an ongoing large international adjuvant study comparing trastuzumab vs. lapapatinib vs. dual HER2 blockade using both drugs. The activity of trastuzumab beyond progression is not limited to combinations with TKIs as it has also been shown in a study where the combination of trastuzumab plus capecitabine was clearly superior to capecitabine alone (33).
Inhibition of HER3 and HER2/HER3 dimers
The HER2 co-receptor HER3, is the key adaptor that once dimerized with and phosphorylated by HER2 engages and activates the phosphatidylinositol-3 kinase (PI3K)/Akt pathway (1, 34, 35). The association of HER2/HER3 dimers with PI3K is essential for the viability of HER2-dependent cells (36, 37). Indeed, HER2+ breast cancer cells are particularly sensitive to apoptosis induced by PI3K inhibitors (38), further underscoring the importance of HER3-mediated signaling in HER2-dependent cells. It generally accepted that sustained inhibition of the output of HER2/HER3 to PI3K/Akt is required for the antitumor effect of HER2 inhibitors. Interestingly, HER2+ breast cancer cells upregulate HER3 expression upon inhibition of HER2 with lapatinib (39). This is problematic because several kinases such as MET, EGFR, Src, FGFR2 can phosphorylate HER3 (40–42), thus partially maintaining PI3K activity and limiting the anti-tumor effect of the HER2 inhibitor (Fig. 1).
Figure 1.
Upregulation of HER3 and resistance to anti-HER2 therapies. The kinase dead HER3 receptor, but not EGFR or HER2, can couple directly to p85, the regulatory subunit to PI3K. HER2/HER3 heterodimers are the most potent signaling complexes of the HER receptor network. Upon inhibition of HER2 and PI3K (as with lapatinib in figure), tumor cells upregulate expression of the HER3 protein. Other tyrosine kinases such as MET, FGFR2, Src, and EGFR present in HER2+ cancer cells can then phosphorylate tyrosines in the C-terminus of HER3 which, in turn, couples to p85 to partially maintain PI3K active and thus limiting the anti-tumor effect of HER2 inhibitors. Alternatively, the cell can use other receptor networks, i.e., IGF-IR to activate the PI3K/Akt survival pathway.
Trastuzumab has been shown to block ligand-independent association between HER2 and HER3 whereas pertuzumab, an antibody that recognizes an epitope in heterodimerization domain II of HER2, blocks ligand-induced HER2-HER3 dimerization (43). Recent data using CLIA-certified dimer assays suggest levels HER2-containing homo and heterodimers (with EGFR and HER3) can be measured in situ and are highly variable among HER2+ tumors (44, 45). We hypothesize, for example, that a tumor with high levels of HER2/HER3 heterodimers would be relatively unresponsive to trastuzumab and thus may be a candidate for lapatinib or pertuzumab, each in combination with trastuzumab. These speculations and the question whether the addition of quantitative dimer assays to FISH and IHC for HER2 will refine the selection of the type of anti-HER2 therapy remain to be investigated.
In trastuzumab-resistant xenografts and in patients with HER2+ breast cancer that have progressed on trastuzumab, only the combination of pertuzumab and trastuzumab but not each antibody alone exhibited clinical activity (46, 47). These data suggest that both HER2 antibodies might be required to completely inhibit HER2-HER3 dimerization in situ, potentially explaining their clinical activity in combination. To test this hypothesis, the phase III Cleopatra study (Table 1) is currently randomizing patients with HER2+ MBC to trastuzumab and docetaxel ± pertuzumab as first line therapy in the metastatic setting using PFS as a primary endpoint. Of note, in the recently reported NeoSphere trial in patients with HER2+ primary breast cancer (Table 1), the path CR rate was 45.8% vs. 29% (p=0.01) in patients treated with neoadjuvant docetaxel/trastuzumab/pertuzumab vs. docetaxel/trastuzumab, respectively (48). Currently, the HER3 monoclonal antibodies AMG-888 (49) and MM-121 (50) are completing phase I testing. We anticipate that like pertuzumab, they may also exert a synergistic effect in combination with trastuzumab or lapatinib in patients with HER2+ MBC.
PI3K and drug resistance
Amplification of PI3K signaling as a result co-expression of PIK3CA-activating mutations or loss of the lipid phosphatase PTEN in HER2+ breast cancer cells and primary tumors is associated with a lower response to trastuzumab and lapatinib (51–55). Several PI3K pathway antagonists are in clinical development and are the subject of recent reviews (56, 57). In preclinical studies, the addition of some of these inhibitors to trastuzumab or lapatinib has inhibited growth of HER2+ tumors resistant to anti-HER2 therapy (53, 54). Interestingly, inhibitors of mTOR, a serine-threonine kinase downstream PI3K, have shown activity after progression on trastuzumab. Dalenc et al. recently reported a multicenter phase II study of 55 women with HER2+ MBC whose tumors were resistant to trastuzumab and taxanes. Patients were treated with the TOR inhibitor everolimus, paclitaxel, and trastuzumab, exhibiting an impressive partial response rate of 19% and an overall clinical benefit rate of 81% (58).
Neoadjuvant therapy as a platform for clinical research
Trastuzumab has been administered with chemotherapy in the neoadjuvant setting with rates of pathologic complete response (path CR) as high as 65% (22, 59–62). Achievement of path CR after neoadjuvant chemotherapy has been widely associated with improved long term outcome. Although not yet clear, recent data suggest similar conclusions may eventually also apply to patients treated with neoadjuvant anti-HER2 therapy. The neoadjuvant herceptin (NOAH) trial tested the efficacy of chemotherapy ± trastuzumab in patients with HER2+ locally advanced or inflammatory breast cancer. Path CR rate was 38% vs. 19% in the trastuzumab vs. the control group. There was a 71% vs. 56% three-year event-free survival in the trastuzumab vs. control arms in all subgroups tested. Overall survival was not different between both arms but this finding is qualified by the fact that a significant proportion of patients ‘crossed-over’ to adjuvant trastuzumab (62). Nonetheless, NOAH is the first trial in patients with HER2+ tumors where path CR mirrors longer term event-free survival, suggesting use of the neoadjuvant therapy space as a platform for clinical investigation which we will discuss below.
Neo-ALTTO is a 450 patient study where HER2+ tumors >2 cm were randomized to trastuzumab, lapatinib, or the combination for 6 weeks, time at which paclitaxel is added to each of the arms for additional 12 weeks. After surgery, all 3 arms will receive adjuvant chemotherapy with FEC followed by the respective HER2 inhibitor either alone or in combination for 34 weeks. About half of the patients enrolled had ER+ tumors. There was increased but manageable toxicity in the lapatinib arms (diarrhea, transaminitis). Path CR, defined as no invasive cancer in the breast or only DCIS in the breast specimen, was significantly higher in the combination arm (51.3%) vs. 29.5% and 24.7% in the trastuzumab and lapatinib arms, respectively. In all 3 arms, the path CR rate was lower in the ER+ vs. the ER− tumors (63). Whether path CR correlates with disease-free and overall survival is pending further follow-up.
Other combinations
The data summarized above suggest that in addition to the combination of trastuzumab and lapatinib, many other rational combinations are or will soon be available for clinical testing. Examples of two-drug combinations that will inhibit the HER2 network and its output to PI3K/Akt more comprehensively are: trastuzumab plus pertuzumab, trastuzumab or lapatinib plus a HER3 neutralizing antibody, trastuzumab or lapatinib plus a PI3K or an AKT inhibitor, a PI3K pathway inhibitor plus a HER3 antibody, T-DM1 plus pertuzumab or a PI3K inhibitor, a combination of HER2 and IGF-IR pathway antagonists, among others. Some of these are shown in Table 1. We speculate many of these or all will be well tolerated and effective against HER2+ MBC but that gain will likely be only incremental. With the establishment of current anti-HER2 agents as adjuvant therapy for HER2+ early disease and the increasing limitation in patient resources, it will be difficult to move these combinations to the adjuvant setting to test their true anti-(micro)metastatic potency using survival as an endpoint.
However, the increasing use of pre-operative therapy should provide a clinical research platform where these combinations can be compared and triaged using path CR as a clinical endpoint predictive of long term outcome. Another benefit of a pre-operative platform is that residual tumor tissue is available at the time of surgery. These ‘drug resistant’ residual cancers may well reflect the molecular profile of drug-resistant micrometastases and can be interrogated with open ended molecular approaches to identify biomarkers and/or effectors of resistance to anti-HER2 therapies. It would not be too surprising if the clinical activity of these combinations turns out to be equivalent. However, toxicity and cost may turn out to be important differentiating factors. If so, just like options for endocrine therapy in ER+ breast cancer, this scenario would provide a plethora of treatment choices for patients with HER2-overexpressing breast cancer who in the end will be the winners.
Acknowledgments
Supported in part by NCI R01 grant CA80195, ACS Clinical Research Professorship Grant CRP-07-234, the Lee Jeans Translational Breast Cancer Research Program, Breast Cancer Specialized Program of Research Excellence (SPORE) P50 CA98131, and Vanderbilt-Ingram Cancer Center Support Grant P30 CA68485
References
- 1.Junttila TT, Akita RW, Parsons K, et al. Ligand-independent HER2/HER3/PI3K complex is disrupted by trastuzumab and is effectively inhibited by the PI3K inhibitor GDC-0941. Cancer Cell. 2009;15:429–40. doi: 10.1016/j.ccr.2009.03.020. [DOI] [PubMed] [Google Scholar]
- 2.Park S, Jiang Z, Mortenson ED, et al. The therapeutic effect of anti-HER2/neu antibody depends on both innate and adaptive immunity. Cancer Cell. 2010;18:160–70. doi: 10.1016/j.ccr.2010.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Romond EH, Perez EA, Bryant J, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med. 2005;353:1673–84. doi: 10.1056/NEJMoa052122. [DOI] [PubMed] [Google Scholar]
- 4.Smith I, Procter M, Gelber RD, 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. doi: 10.1016/S0140-6736(07)60028-2. [DOI] [PubMed] [Google Scholar]
- 5.Robert N, Leyland-Jones B, Asmar L, et al. Randomized phase III study of trastuzumab, paclitaxel, and carboplatin compared with trastuzumab and paclitaxel in women with HER-2-overexpressing metastatic breast cancer. J Clin Oncol. 2006;24:2786–92. doi: 10.1200/JCO.2005.04.1764. [DOI] [PubMed] [Google Scholar]
- 6.Perez EA, Suman VJ, Davidson NE, et al. Results of Chemotherapy Alone, with Sequential or Concurrent Addition of 52 Weeks of Trastuzumab in the NCCTG N9831 HER2-Positive Adjuvant Breast Cancer Trial. Cancer Res. 2009;69:Abstr 80. [Google Scholar]
- 7.Curigliano G, Viale G, Bagnardi V, et al. Clinical relevance of HER2 overexpression/amplification in patients with small tumor size and node-negative breast cancer. J Clin Oncol. 2009;27:5693–9. doi: 10.1200/JCO.2009.22.0962. [DOI] [PubMed] [Google Scholar]
- 8.Gonzalez-Angulo AM, Litton JK, Broglio KR, et al. High risk of recurrence for patients with breast cancer who have human epidermal growth factor receptor 2-positive, node-negative tumors 1 cm or smaller. J Clin Oncol. 2009;27:5700–6. doi: 10.1200/JCO.2009.23.2025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Joensuu H, Kellokumpu-Lehtinen PL, Bono P, et al. Adjuvant docetaxel or vinorelbine with or without trastuzumab for breast cancer. N Engl J Med. 2006;354:809–20. doi: 10.1056/NEJMoa053028. [DOI] [PubMed] [Google Scholar]
- 10.Gomez HL, Doval DC, Chavez MA, et al. 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]
- 11.Geyer CE, Forster J, Lindquist D, et al. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med. 2006;355:2733–43. doi: 10.1056/NEJMoa064320. [DOI] [PubMed] [Google Scholar]
- 12.Press MF, Finn RS, Cameron D, 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. doi: 10.1158/1078-0432.CCR-08-1056. [DOI] [PubMed] [Google Scholar]
- 13.Press MF, Slamon DJ, Flom KJ, Park J, Zhou JY, Bernstein L. Evaluation of HER-2/neu gene amplification and overexpression: comparison of frequently used assay methods in a molecularly characterized cohort of breast cancer specimens. J Clin Oncol. 2002;20:3095–105. doi: 10.1200/JCO.2002.09.094. [DOI] [PubMed] [Google Scholar]
- 14.Mass RD, Press MF, Anderson S, et al. Evaluation of clinical outcomes according to HER2 detection by fluorescence in situ hybridization in women with metastatic breast cancer treated with trastuzumab. Clin Breast Cancer. 2005;6:240–6. doi: 10.3816/CBC.2005.n.026. [DOI] [PubMed] [Google Scholar]
- 15.Dybdal N, Leiberman G, Anderson S, et al. Determination of HER2 gene amplification by fluorescence in situ hybridization and concordance with the clinical trials immunohistochemical assay in women with metastatic breast cancer evaluated for treatment with trastuzumab. Breast Cancer Res Treat. 2005;93:3–11. doi: 10.1007/s10549-004-6275-8. [DOI] [PubMed] [Google Scholar]
- 16.Press MF, Sauter G, Bernstein L, et al. Diagnostic evaluation of HER-2 as a molecular target: an assessment of accuracy and reproducibility of laboratory testing in large, prospective, randomized clinical trials. Clin Cancer Res. 2005;11:6598–607. doi: 10.1158/1078-0432.CCR-05-0636. [DOI] [PubMed] [Google Scholar]
- 17.Sauter G, Lee J, Bartlett JM, Slamon DJ, Press MF. Guidelines for human epidermal growth factor receptor 2 testing: biologic and methodologic considerations. J Clin Oncol. 2009;27:1323–33. doi: 10.1200/JCO.2007.14.8197. [DOI] [PubMed] [Google Scholar]
- 18.Paik S, Kim C, Wolmark N. HER2 status and benefit from adjuvant trastuzumab in breast cancer. N Engl J Med. 2008;358:1409–11. doi: 10.1056/NEJMc0801440. [DOI] [PubMed] [Google Scholar]
- 19.Meng S, Tripathy D, Shete S, et al. HER-2 gene amplification can be acquired as breast cancer progresses. Proc Natl Acad Sci U S A. 2004;101:9393–8. doi: 10.1073/pnas.0402993101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Gutierrez MC, Detre S, Johnston S, et al. Molecular changes in tamoxifen-resistant breast cancer: relationship between estrogen receptor, HER-2, and p38 mitogen-activated protein kinase. J Clin Oncol. 2005;23:2469–76. doi: 10.1200/JCO.2005.01.172. [DOI] [PubMed] [Google Scholar]
- 21.Pectasides D, Gaglia A, Arapantoni-Dadioti P, et al. HER-2/neu status of primary breast cancer and corresponding metastatic sites in patients with advanced breast cancer treated with trastuzumab-based therapy. Anticancer Res. 2006;26:647–53. [PubMed] [Google Scholar]
- 22.Hurley J, Doliny P, Reis I, et al. Docetaxel, cisplatin, and trastuzumab as primary systemic therapy for human epidermal growth factor receptor 2-positive locally advanced breast cancer. J Clin Oncol. 2006;24:1831–8. doi: 10.1200/JCO.2005.02.8886. [DOI] [PubMed] [Google Scholar]
- 23.Mittendorf EA, Wu Y, Scaltriti M, et al. Loss of HER2 amplification following trastuzumab-based neoadjuvant systemic therapy and survival outcomes. Clin Cancer Res. 2009;15:7381–8. doi: 10.1158/1078-0432.CCR-09-1735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Lewis Phillips GD, Li G, Dugger DL, et al. Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res. 2008;68:9280–90. doi: 10.1158/0008-5472.CAN-08-1776. [DOI] [PubMed] [Google Scholar]
- 25.Junttila TT, Li G, Parsons K, Phillips GL, Sliwkowski MX. Trastuzumab-DM1 (T-DM1) retains all the mechanisms of action of trastuzumab and efficiently inhibits growth of lapatinib insensitive breast cancer. Breast Cancer Res Treat. 2010 doi: 10.1007/s10549-010-1090-x. Epub Date 2010/08/24. [DOI] [PubMed] [Google Scholar]
- 26.Krop IE, Beeram M, Modi S, et al. Phase I study of trastuzumab-DM1, an HER2 antibody-drug conjugate, given every 3 weeks to patients with HER2-positive metastatic breast cancer. J Clin Oncol. 2010;28:2698–704. doi: 10.1200/JCO.2009.26.2071. [DOI] [PubMed] [Google Scholar]
- 27.Burris HA, 3rd, Rugo HS, Vukelja SJ, 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. 2010 doi: 10.1200/JCO.2010.29.5865. Epub Date 2010/12/22. [DOI] [PubMed] [Google Scholar]
- 28.Anido J, Scaltriti M, Bech Serra JJ, et al. Biosynthesis of tumorigenic HER2 C-terminal fragments by alternative initiation of translation. Embo J. 2006;25:3234–44. doi: 10.1038/sj.emboj.7601191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Scaltriti M, Rojo F, Ocana A, et al. Expression of p95HER2, a truncated form of the HER2 receptor, and response to anti-HER2 therapies in breast cancer. J Natl Cancer Inst. 2007;99:628–38. doi: 10.1093/jnci/djk134. [DOI] [PubMed] [Google Scholar]
- 30.Scaltriti M, Chandarlapaty S, Prudkin L, et al. Clinical benefit of lapatinib-based therapy in patients with human epidermal growth factor receptor 2-positive breast tumors coexpressing the truncated p95HER2 receptor. Clin Cancer Res. 2010;16:2688–95. doi: 10.1158/1078-0432.CCR-09-3407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Burstein HJ, Sun Y, Dirix LY, et al. Neratinib, an irreversible ErbB receptor tyrosine kinase inhibitor, in patients with advanced ErbB2-positive breast cancer. J Clin Oncol. 2010;28:1301–7. doi: 10.1200/JCO.2009.25.8707. [DOI] [PubMed] [Google Scholar]
- 32.Blackwell KL, Burstein HJ, Storniolo AM, 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. doi: 10.1200/JCO.2008.21.4437. [DOI] [PubMed] [Google Scholar]
- 33.von Minckwitz G, du Bois A, Schmidt M, et al. Trastuzumab beyond progression in human epidermal growth factor receptor 2-positive advanced breast cancer: a german breast group 26/breast international group 03-05 study. J Clin Oncol. 2009;27:1999–2006. doi: 10.1200/JCO.2008.19.6618. [DOI] [PubMed] [Google Scholar]
- 34.Yakes FM, Chinratanalab W, Ritter CA, King W, Seelig S, Arteaga CL. Herceptin-induced inhibition of phosphatidylinositol-3 kinase and Akt Is required for antibody-mediated effects on p27, cyclin D1, and antitumor action. Cancer Res. 2002;62:4132–41. [PubMed] [Google Scholar]
- 35.Hellyer NJ, Kim MS, Koland JG. Heregulin-dependent activation of phosphoinositide 3-kinase and Akt via the ErbB2/ErbB3 co-receptor. J Biol Chem. 2001;276:42153–61. doi: 10.1074/jbc.M102079200. [DOI] [PubMed] [Google Scholar]
- 36.Holbro T, Beerli RR, Maurer F, Koziczak M, Barbas CF, 3rd, Hynes NE. The ErbB2/ErbB3 heterodimer functions as an oncogenic unit: ErbB2 requires ErbB3 to drive breast tumor cell proliferation. Proc Natl Acad Sci U S A. 2003;100:8933–8. doi: 10.1073/pnas.1537685100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Lee-Hoeflich ST, Crocker L, Yao E, et al. A central role for HER3 in HER2-amplified breast cancer: implications for targeted therapy. Cancer Res. 2008;68:5878–87. doi: 10.1158/0008-5472.CAN-08-0380. [DOI] [PubMed] [Google Scholar]
- 38.Brachmann SM, Hofmann I, Schnell C, et al. Specific apoptosis induction by the dual PI3K/mTor inhibitor NVP-BEZ235 in HER2 amplified and PIK3CA mutant breast cancer cells. Proc Natl Acad Sci U S A. 2009;106:22299–304. doi: 10.1073/pnas.0905152106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Amin DN, Sergina N, Ahuja D, et al. 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]
- 40.Engelman JA, Zejnullahu K, Mitsudomi T, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316:1039–43. doi: 10.1126/science.1141478. [DOI] [PubMed] [Google Scholar]
- 41.Ishizawar RC, Miyake T, Parsons SJ. c-Src modulates ErbB2 and ErbB3 heterocomplex formation and function. Oncogene. 2007;26:3503–10. doi: 10.1038/sj.onc.1210138. [DOI] [PubMed] [Google Scholar]
- 42.Kunii K, Davis L, Gorenstein J, et al. FGFR2-amplified gastric cancer cell lines require FGFR2 and Erbb3 signaling for growth and survival. Cancer Res. 2008;68:2340–8. doi: 10.1158/0008-5472.CAN-07-5229. [DOI] [PubMed] [Google Scholar]
- 43.Agus DB, Akita RW, Fox WD, et al. Targeting ligand-activated ErbB2 signaling inhibits breast and prostate tumor growth. Cancer Cell. 2002;2:127–37. doi: 10.1016/s1535-6108(02)00097-1. [DOI] [PubMed] [Google Scholar]
- 44.Desmedt C, Sperinde J, Piette F, Huang W, Jin X, Tan Y, Larsimont D, Giuliani R, Chappey C, Buyse M, Winslow J, Piccart M, Sotiriou C, Petropoulos C, Bates M. Quantitation of HER2 Expression or HER2:HER2 dimers and differential survival in a cohort of metastatic breast cancer patients carefully selected for trastuzumab treatment primarily by FISH. Diagn Mol Pathol. 2009;18:22–9. doi: 10.1097/PDM.0b013e31818ebc69. [DOI] [PubMed] [Google Scholar]
- 45.Williams SJ, Mukherjee A, Badal Y, et al. Profiling PI3K pathway activation in formalin-fixed, paraffin-embedded tumor cell lines and breast and ovarian tumors using novel proximity assays. Proc Am Assoc Cancer Res. 2009:Abstr 5251. [Google Scholar]
- 46.Scheuer W, Friess T, Burtscher H, Bossenmaier B, Endl J, Hasmann M. Strongly enhanced antitumor activity of trastuzumab and pertuzumab combination treatment on HER2-positive human xenograft tumor models. Cancer Res. 2009;69:9330–6. doi: 10.1158/0008-5472.CAN-08-4597. [DOI] [PubMed] [Google Scholar]
- 47.Baselga J, Gelmon KA, Verma S, et al. Phase II trial of pertuzumab and trastuzumab in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer that progressed during prior trastuzumab therapy. J Clin Oncol. 2010;28:1138–44. doi: 10.1200/JCO.2009.24.2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Gianni L, Pienkowski T, Im Y-H, et al. Neoadjuvant pertuzumab (P) and trastuzumab (T): Antitumor and safety analysis of randomized phase II study (NeoSphere) Cancer Res. 2010;70:82s. [Google Scholar]
- 49.Freeman D, Ogbagabriel S, Rothe M, et al. Fully human anti-HER3 monoclonal antibodies (mAbs) have unique in vitro and in vivo functional and anti-tumor activities versus othe HER family inhibitors. Proc Am Assoc Cancer Res. 2008:49. [Google Scholar]
- 50.Schoeberl B, Faber AC, Li D, et al. An ErbB3 antibody, MM-121, is active in cancers with ligand-dependent activation. Cancer Res. 2010;70:2485–94. doi: 10.1158/0008-5472.CAN-09-3145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Berns K, Horlings HM, Hennessy BT, et al. A functional genetic approach identifies the PI3K pathway as a major determinant of trastuzumab resistance in breast cancer. Cancer Cell. 2007;12:395–402. doi: 10.1016/j.ccr.2007.08.030. [DOI] [PubMed] [Google Scholar]
- 52.Nagata Y, Lan KH, Zhou X, et al. PTEN activation contributes to tumor inhibition by trastuzumab, and loss of PTEN predicts trastuzumab resistance in patients. Cancer Cell. 2004;6:117–27. doi: 10.1016/j.ccr.2004.06.022. [DOI] [PubMed] [Google Scholar]
- 53.Serra V, Markman B, Scaltriti M, et al. NVP-BEZ235, a dual PI3K/mTOR inhibitor, prevents PI3K signaling and inhibits the growth of cancer cells with activating PI3K mutations. Cancer Res. 2008;68:8022–30. doi: 10.1158/0008-5472.CAN-08-1385. [DOI] [PubMed] [Google Scholar]
- 54.Eichhorn PJ, Gili M, Scaltriti M, et al. Phosphatidylinositol 3-kinase hyperactivation results in lapatinib resistance that is reversed by the mTOR/phosphatidylinositol 3-kinase inhibitor NVP-BEZ235. Cancer Res. 2008;68:9221–30. doi: 10.1158/0008-5472.CAN-08-1740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Esteva FJ, Guo H, Zhang S, et al. PTEN, PIK3CA, p-AKT, and p-p70S6K status: association with trastuzumab response and survival in patients with HER2-positive metastatic breast cancer. Am J Pathol. 2010;177:1647–56. doi: 10.2353/ajpath.2010.090885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Engelman JA. Targeting PI3K signalling in cancer: opportunities, challenges and limitations. Nat Rev Cancer. 2009;9:550–62. doi: 10.1038/nrc2664. [DOI] [PubMed] [Google Scholar]
- 57.Brachmann S, Fritsch C, Maira SM, Garcia-Echeverria C. PI3K and mTOR inhibitors: a new generation of targeted anticancer agents. Curr Opin Cell Biol. 2009;21:194–8. doi: 10.1016/j.ceb.2008.12.011. [DOI] [PubMed] [Google Scholar]
- 58.Dalenc F, Campone M, Hupperets P, et al. Everolimus in combination with weekly paclitaxel and trastuzumab in patients (pts) with HER2-overexpressing metastatic breast cancer (MBC) with prior resistance to trastuzumab and taxanes: A multicenter phase II clinical trial. J Clin Oncol. 2010;28 (Suppl):15s. [Google Scholar]
- 59.Burstein HJ, Harris LN, Gelman R, et al. Preoperative therapy with trastuzumab and paclitaxel followed by sequential adjuvant doxorubicin/cyclophosphamide for HER2 overexpressing stage II or III breast cancer: a pilot study. J Clin Oncol. 2003;21:46–53. doi: 10.1200/JCO.2003.03.124. [DOI] [PubMed] [Google Scholar]
- 60.Buzdar AU, Ibrahim NK, Francis D, 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. doi: 10.1200/JCO.2005.07.032. [DOI] [PubMed] [Google Scholar]
- 61.Harris LN, You F, Schnitt SJ, et al. Predictors of resistance to preoperative trastuzumab and vinorelbine for HER2-positive early breast cancer. Clin Cancer Res. 2007;13:1198–207. doi: 10.1158/1078-0432.CCR-06-1304. [DOI] [PubMed] [Google Scholar]
- 62.Gianni L, Eiermann W, Semiglazov V, et al. Neoadjuvant chemotherapy with trastuzumab followed by adjuvant trastuzumab versus neoadjuvant chemotherapy alone, in patients with HER2-positive locally advanced breast cancer (the NOAH trial): a randomised controlled superiority trial with a parallel HER2-negative cohort. Lancet. 2010;375:377–84. doi: 10.1016/S0140-6736(09)61964-4. [DOI] [PubMed] [Google Scholar]
- 63.Baselga J, Bradbury I, Eidtmann H, 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 plust paclitaxel in women with HER2-positive primary breast cancer. Cancer Res. 2010;70:82s. [Google Scholar]

