Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Apr 6.
Published in final edited form as: Oncogene. 2007 May 7;26(46):6577–6592. doi: 10.1038/sj.onc.1210478

Targeting the function of the HER2 oncogene in human cancer therapeutics

Mark M Moasser 1
PMCID: PMC3071580  NIHMSID: NIHMS263195  PMID: 17486079

Abstract

The year 2007 marks exactly two decades since HER2 was functionally implicated in the pathogenesis of human breast cancer (Slamon et al. 1987). This finding established the HER2 oncogene hypothesis for the development of some human cancers. An abundance of experimental evidence compiled over the past two decades now solidly supports the HER2 oncogene hypothesis. A direct consequence of this hypothesis was the promise that inhibitors of oncogenic HER2 would be highly effective treatments for HER2-driven cancers. This treatment hypothesis has led to the development and widespread use of anti-HER2 antibodies (trastuzumab) in clinical management resulting in significantly improved clinical anti-tumor efficacies that have transformed the clinical practice of oncology. In the shadows of this irrefutable clinical success, scientific studies have not yet been able to mechanistically validate that trastuzumab inhibits oncogenic HER2 function and it remains possible that the current clinical advances are a consequence of the oncogene hypothesis but not a translation of it. These looming scientific uncertainties suggest that the full promise of the treatment hypothesis may not yet have been realized. The coming decade will see a second generation of HER2 targeting agents brought into clinical testing and a renewed attempt to treat HER2-driven cancers through the inactivation HER2. Here I review the development of treatments that target HER2 in the context of the HER2 oncogene hypothesis, and where we stand with regards to the clinical translation of the HER2 oncogene hypothesis.

INTRODUCTION

HER2 belongs to the Human Epidermal Growth Factor Receptor (HER) family of tyrosine kinases conisting of EGFR (HER1, erbB1), HER2 (erbB2, HER2/neu), HER3 (erbB3), and HER4 (erbB4). The importance of HER2 in cancer was realized in the early 1980s when a mutationaly activated form of its rodent homologue neu was identified in a search for oncogenes in a carcinogen induced rat tumorigenesis model (Shih et al. 1981). Its human homologue HER2 was simultaneously cloned and found to be amplified in a breast cancer cell line (King et al. 1985). The relevance of HER2 to human cancer was established when it was discovered that approximately 25–30% of breast cancers have amplification and overexpression of HER2 and these cancers have worse biologic behavior and prognosis (Slamon et al. 1989). This finding established the HER2 oncogene hypothesis that overexpression of HER2 is etiologically linked with tumorigenesis in some human cancers. A substantial body of experimental evidence over the past two decades has come to solidly support this hypothesis. In numerous in vitro and transgenic models, HER2 overexpression by itself is potently transforming. In addition, analysis of human breast cancers has shown that amplification of the HER2 locus is an early event in human carcinogenesis and along with the experimental evidence confirming its potently transforming functions, this makes a highly compelling case implicating HER2 overexpression in the genesis of these human cancers. The signaling functions of HER2, the body of evidence confirming the transforming functions of HER2, the numerous proposed mechanisms mediating its transforming functions, and the data establishing the relevance of these findings to human cancer pathogenesis were discussed in depth previously (Moasser 2007). A direct consequence of the HER2 oncogene hypothesis of human cancer was that inhibitors of oncogenic HER2 would be highly effective treatment for HER2 driven cancers. Here I will review where we stand with regards to testing this treatment hypothesis and where we currently stand with regards to the therapeutic implications of the HER2 oncogene hypothesis.

TUMOR DEPENDENCE ON HER2

The tumorigenic potential of HER2 is solidly supported by experimental models (Moasser 2007). This by itself proposes HER2 as a possible target for anti-cancer drugs. However its suitability as a drug target is substantially strengthened by experiments demonstrating that HER2-driven tumors are dependent on HER2 function. This dependency, recently labelled oncogene addiction, identifies oncogenes that are high value targets for drug development (Hynes and Lane 2001).

HER2-dependency of HER2 amplified human cancers

Experimental models of HER2 overexpressing cancer cells using antisense, ribozyme, or siRNA methodologies consistently show that HER2 knockdown induces apoptosis in cell culture, or tumor regression in vivo, in the absence of HER2 expression, while tumor types that do not overexpress HER2 are not sensitive to HER2 knockdown (Colomer et al. 1994; Juhl et al. 1997; Roh et al. 2000; Faltus et al. 2004; Choudhury et al. 2004). Similar results are seen with kinase-dead HER2 and intracellular single chain anti-HER2 antibodies (Messerle et al. 1994; Beerli et al. 1994; Deshane et al. 1996).

HER2-dependency in experimental models

Engineered models of HER2-driven transformation using tetracycline inducible systems confirm that HER2 induced tumors require HER2 for continued tumorigenic growth and survival. This has been demonstrated in an HER2-transformed NIH3T3 tumor model wherein tumors regress upon withdrawal of the HER2 oncogene (Baasner et al. 1996; Schiffer et al. 2003). This has also been corroborated in a tet-inducible transgenic models. Tetracycline induced expression of activated HER2 in squamous epithelia of mice results in severe hyperplastic abnormalities of squamous epithelial tissues, which reverse upon withdrawal of the HER2 transgene expression (Xie et al. 1999). Tumors in MMTV-neuT mice are also dependent on continued oncogene expression. In the MMTV-rtTA/TetO-NeuNT bitransgenic variant of this model regulated by doxycycline, when expression of the neuT oncogene is induced in the mammary tissue of adult mice, this leads to the formation of multiple mammary tumors and lung metastases, and the entire primary tumor and metastatic disease fully regresses when neuT expression is withdrawn (Moody et al. 2002).

Although each of these models is subject to criticisms relating to their simplicity, when taken in aggregate, they are highly consistent and collectively make a highly compelling case that HER2 induced tumors are addicted to HER2. This has made HER2 one of the most sought after targets in cancer drug development.

Potential to escape from HER2 addiction

The inducible models do suggest the possibility of HER2-independent tumor recurrence that occurs after a period of complete regression. Tumors that are induced in the MMTV-rtTA/TetO-NeuNT model and which completely regress upon oncogene withdrawal, eventually recur after prolonged dormancy without induction of the neuT oncogene (Moody et al. 2002). This is associated with induction of the transcriptional repressor snail, and suggests that second hits may provide escape routes for residual tumor cells leading to tumor recurrence and progression driven by alternative pathways (Moody et al. 2005). Tetracycline regulated NIH3T3-HER2 tumors cells that regress following withdrawal of oncogene expression similarly recur following a period of remission despite absence of oncogene expression, although the molecular features associated with HER2 independent recurrence in this model are not yet described (Schiffer et al. 2003). The direct relevance of these models of recurrence to human tumors is not yet known and awaits analysis of human tumors that have recurred following a complete remission induced by HER2 targeted therapies.

INHIBITION OF HER2 FOR CANCER THERAPY

The evidence to support the HER2 oncogene hypothesis that HER2 initiates and drives the progression of HER2 overexpressing cancers is almost unimpeachable at this point. The direct consequence of this hypothesis is the treatment hypothesis that inactivation of HER2 could be highly effective therapy for patients with HER2 overexpressing cancers. Due to the large number of patients with this type of cancer, testing of the HER2 treatment hypothesis has been one of the most actively pursued programs in the cancer therapeutic arena. Testing in human subjects requires the development of safe and effective therapies that inactivate HER2 in patients tumors and in the best scenario are predicted to produce complete remissions and recapitulate results from preclinical models. Correlative scientific studies of these therapeutic agents in preclinical models and in patients are essential in order to determine the validity of the treatment hypothesis. These efforts have led to the development of the anti-HER2 mAb trastuzumab which has made significant clinical impact including a reduction in mortality from HER2 overexpressing disease. But mechanistic studies have been conflicting and suggest that the treatment hypothesis may not yet have been effectively tested. If the treatment hypothesis is correct and inhibiting oncogenic HER2 function would result in complete tumor regression, the clinical impact is predicted to be much larger than currently realized and we may have merely seen the tip of the iceberg. The available data with regards to several anti-HER2 targeted therapies are reviewed below. The two modalities for which there is considerable data are antibody therapies and small molecule kinase inhibitors. These are discussed separately below.

Biologic effects of panels of anti-HER2 antibodies

Almost immediately following the discovery that the oncogene HER2 was amplified in many breast and ovarian cancers and linked with disease biology, efforts began to develop inhibitors of this oncogene. The technology to develop mouse monoclonal antibodies (mAbs) had become available at this time and since HER2 functions as a growth factor receptor, it was a highly rationale hypothesis at the time that a mAb that binds the extracellular domain of HER2 and interferes with ligand activation would inhibit tumorigenic HER2 function. The proof-of-principle experiment was initially conducted in the neuT transformation model. In this model it was found that anti-Neu mAbs downregulate NeuT expression, supress cell growth, inhibit transformation and tumor growth in mice (Drebin et al. 1985; Drebin et al. 1988). This suggested that HER2 overexpressing human cancers could also potentially be treated with mAbs. Over one hundred mAbs were developed by numerous groups against the extracellular domain of human HER2. The effects of these mAbs on HER2 overexpressing human cancers turned out to be much more complicated than predicted from the more simplistic neuT model. The activities of some of these panels of mAbs against HER2 overexpressing tumor cell lines have been characterized and published and are summarized in Table 1. The results from these studies reveal that anti-HER2 mAbs can produce highly diverse outcomes. These include both growth inhibitory or growth stimulatory effects, differentiating effects, and pro-apoptotic effects. Some mAbs induce HER2 phosphorylation and others do not, some induce HER2 downregulation and others do not, some inhibit tumorigenic growth in vivo and others do not. The results of all these studies taken together do not formulate a clear picture of the mechanism by which an anti-HER2 mAb can inhibit tumor growth. Specifically, cell growth inhibition or tumor growth inhibition does not correlate with the mAb ability to downregulate HER2. In addition, anti-HER2 mAbs downregulate mutationally activated HER2 much more effectively than wildtype HER2, reproducing the effects seen with anti-Neu mAbs in the NeuT model (van et al. 1990). Adding complexity to the picture is that even growth inhibition in vitro does not correlate with tumor growth inhibition in vivo such that some mAbs are growth stimulatory in cell culture models yet inhibit tumor growth in mice (Stancovski et al. 1991; Harwerth et al. 1993). The mechanistic principles underlying the diversity of findings from anti-HER2 mAbs remain unclear to this day. But the compelling data regarding the role of HER2 in human tumorigenesis and evidence of anti-tumor efficacy of some anti-HER2 mAbs in preclinical models drove the clinical development of at least one such agent.

TABLE 1.

Published monoclonal anti-HER2 antibodies

Reference N Biochemical findings (in Her2++ tumors) Biological findings (in Her2++ tumors) Notes
(Stancovski et al. 1991; Bacus et al. 1992; Hurwitz et al. 1995; Klapper et al. 1997; Yarden 1990) panel of 17 most induce phosphorylation, some induce endocytic degradation some are growth inhibitory and differentiating, some are growth stimulatory growth inhibition correlates with homodimerization and endocytic degradation
(van et al. 1990) panel of 4 downregulation of ErbB2 is poor, downregulation of mutant ErbB2 is strong only mutant HER2 was growth inhibited (soft agar) engineered TM domain mutation
(Harwerth et al. 1993; Harwerth et al. 1992) panel of 4 2 of 4 induce phosphorylation, 3 of 4 induce downregulation 1 of 4 inhibits cell growth 2 of 4 inhibit tumor growth no correlation between HER2 turnover and growth inhibition
(Xu et al. 1993) panel of 11 some internalize and downregulate HER2 2 inhibit monolayer growth of SkBr3, 7 inhibit growth in agar of SkBr3 no correlation between HER2 downregulation and growth inhibition, Fab fragment also inhibits growth
(Kita et al. 1996) panel of 12 most induce phosphorylation growth data not reported, one mAb induces apoptosis
(McKenzie et al. 1989) panel of 10 not reported not reported
(Tagliabue et al. 1991; Srinivas et al. 1993) panel of 2 they induce phosphorylation and internalize cause growth inhibition (Calu-3) internalization requires bivalency and Fab is inactive
(Neve et al. 2001) panel of 2 they internalize (both as scFv or IgG) no growth inhibition internalization does not require bivalency
(De et al. 2005) one mAb induces ErbB2 phosphorylation and downregulation growth inhibition in vitro and in vivo
(Belimezi et al. 2006) one mAb internalizes inhibits growth
(Hudziak et al. 1989; Sarup et al. 1991) panel of 11 4D5 downregulates HER2 some inhibit growth in vitro. 4D5 inhibits growth in vivo

Development of trastuzumab (Herceptin®)

Of the over one hundred anti-HER2 mAbs generated in the 80s and 90s, one was developed for clinical testing. The mAb 4D5 was selected among a panel of mouse anti-HER2 antibodies at Genentech, Inc. (South San Francisco, CA) for development because of its anti-tumor effects in vitro and in mouse models (Hudziak et al. 1989; Shepard et al. 1991). Mouse mAb 4D5 was humanized for clinical use yielding several humanized variants. Some of these engineered variants lost in vitro anti-proliferative efficacy despite high affinity binding to HER2, but others retained anti-proliferative efficacy and one such clone was selected for further clinical development (named trastuzumab, Herceptin®) (Carter et al. 1992). The constant regions of these humanized mAbs were engineered for optimum participation in antibody dependent cellular cytotoxicity (ADCC) or complement dependent cytotoxicity (CDC) and indeed trastuzumab is much more efficient than its murine counterpart in mediating ADCC (Carter et al. 1992; Lewis et al. 1993; Tokuda et al. 1996). Trastuzumab has reduced cell culture anti-proliferative activity compared with the murine 4D5 but equally potent anti-tumor efficacy in mouse xenograft models (Tokuda et al. 1996; Baselga et al. 1998; Clynes et al. 2000).

Clinical anti-tumor activity of trastuzumab

The clinical anti-tumor activity of trastuzumab has now been extensively characterized in numerous clinical studies spanning the past decade and a half. Initial difficulties in identifying the subset of patients with HER2 overexpressing tumors by clinically available immunohistochemical methods were finally overcome by clinical implementation of a fluorescence in situ hybridization (FISH) assay to detect HER2 gene amplification and it is now evident that trastuzumab induces tumor regression in approximately 30–35% of patients with HER2 amplified metastatic breast cancer if used as upfront therapy (Vogel et al. 2002; Mass et al. 2005) and has much less activity if used after other chemotherapies (Baselga et al. 1996). In patients with metastatic disease, trastuzumab is not curative and disease progression resumes after a median duration of approximately 5 months despite continuous trastuzumab therapy (Vogel et al. 2002). The most beneficial clinical use of trastuzumab has been in combination with various cytotoxic chemotherapies. The addition of trastuzumab to multiple chemotherapy regimens significantly increases their anti-tumor efficacy (Slamon et al. 2001; Marty et al. 2005; Burstein et al. 2003). The biggest impact of trastuzumab has been in the treatment of patients with potentially curable early stage breast cancer. In early stage HER2 amplified breast cancer patients who receive chemotherapy after surgical resection, the addition of trastuzumab to their chemotherapy regimens significantly prolongs disease-free survival and reduces the chances of disease recurrence (Figure 1) (Piccart-Gebhart et al. 2005; Romond et al. 2005). Although these adjuvant therapy studies are still in their early years of follow-up, the powerful effects seen in the early follow-up period is widely believed to translate to a significant reduction in mortality from HER2 amplified breast cancer and the use of trastuzumab has rapidly become the standard management of early stage breast cancer patients. The clinical antitumor activity of trastuzumab is limited to tumors with HER2 overexpression and trastuzumab has no significant clinical activity against breast cancers without HER2 overexpression (Vogel et al. 2002; Mass et al. 2005). At this time its single agent activity appears to be limited to breast cancers and it has much less clinical anti-tumor activity against ovarian or endometrial cancers with HER2 overexpression (Fleming et al. 2003; Bookman et al. 2003) and continues to be investigated in other types of cancer.

Figure 1.

Figure 1

Outcome results with a median follow-up time of 2.0 years in 3351 patients with early stage breast cancer treated with chemotherapy and randomized to the addition of trastuzumab or control. Panel A shows the proportion of patients in each arm that remain cancer-free at the indicated years of follow-up. Many more patients from the trastuzumab-treated arm (solid line) are cancer-free compared with the control arm (broken line). Panel B shows the proportion of patients in each arm that are alive at the indicated years. More patients from the trastuzumab-treated arm are alive compared with the control arm. The effect of trastuzumab on disease-free and overall survival is highly statistically significant. (Romond et al. 2005) Copyright 2005 Massachusetts Medical Society, All rights reserved.

Implications of trastuzumab to the HER2 oncogene hypothesis

These improvements in the clinical management of patients with HER2 amplified breast cancer afforded by trastuzumab are a direct consequence of the HER2 oncogene hypothesis of breast cancer initially proposed two decades ago and are a testament to the potential of scientific research to impact human health and disease mortality. But while the success of trastuzumab is a consequence of the HER2 oncogene hypothesis, it is not sufficient to validate it. Validation of the oncogene hypothesis requires mechanistic evidence that trastuzumab treats patients through inactivation of tumor HER2. This evidence is currently lacking and more than a decades work in trying to determine the mechanism of action of trastuzumab has produced largely conflicting and inconclusive results and a compelling mechanistic model of how and whether trastuzumab inhibits oncogenic HER2 function has not emerged.

Mechanism of action of trastuzumab – HER2 downregulation

Extensive studies over the past decade have attempted to determine the molecular mechanism underlying the clinical anti-tumor activity of trastuzumab. The simplest hypothesis derives from the previously established anti-NeuT mAb and anti-HER2 4D5 mAb data showing that these mAbs induce the degradation of the targeted surface receptors NeuT or HER2 (discussed above). Although this seems like a fairly simple hypothesis to test conclusively, extensive analyses by numerous investigators studying the effect of trastuzumab on tumor cell HER2 expression have produced conflicting results, even in similar cell types and assays. While some studies show that trastuzumab downregulates HER2 in HER2 overexpressing tumor cells (Cuello et al. 2001; Marches and Uhr 2004; Citri et al. 2002; Lee et al. 2002), other studies clearly show that it does not (Longva et al. 2005; Austin et al. 2004). Part of the complexity in this field was resolved when it was determined that trastuzumab binds and internalizes with surface HER2 but re-emerges with HER2 at the surface, merely accompanying HER2 passively along its normal endocytic recycling route (Austin et al. 2004). The most compelling evidence at this point seems to support the position that trastuzumab does not induce the downregulation of HER2 in tumor cells. Consistent with this, three clinical studies show no reduction in tumor HER2 expression in patients undergoing treatment with trastuzumab (Arnould et al. 2006; Mohsin et al. 2005; Gennari et al. 2004). Therefore it appears unlikely that the clinical antitumor activity of trastuzumab is mediated through downregulation of tumor HER2.

Mechanism of action of trastuzumab – HER2 signaling

The principal prevailing hypothesis that rationalized the development of trastuzumab and other anti-HER2 mAbs for most of the nineties was that it inhibits the activation of HER2 by yet undiscovered ligands. However the hypothesized HER2 ligand was never discovered, and biochemical screens, post-genome computional studies, and crystal structure revelations have made it clear that HER2 has no physiologic ligand, and that its ligand responsive functions are mediated through heterodimerization with its ligand-activated HER family partners (Sliwkowski 2003). In fact the extracellular domain of HER2 constitutively exists in an active conformation resembling the ligand-bound state of the other HER family proteins, precluding any potential activating role for ligands (Garrett et al. 2003; Cho et al. 2003). Therefore the hypothesis that trastuzumab inhibits direct ligand binding and activation of HER2 is all but dismissed at this point. An alternative hypothesis that has been proposed is that trastuzumab inhibits the interaction of HER2 with its HER family partners or possibly other interacting proteins. But convincing evidence in support of this hypothesis has not yet emerged. In pulldown assays trastuzumab does not inhibit HER2–HER3 interaction (Agus et al. 2002), and in fluoresence resonance energy transfer (FRET) based assays trastuzumab also does not inhibit HER2 interaction with EGFR or HER3 (Diermeier et al. 2005). In a different model using truncated HER proteins fusing them to β-galactosidase fragments in an enzyme complementation assay, trastuzumab was reported to inhibit EGFR-HER2 interaction but not HER2–HER3 interactions (Wehrman et al. 2006). The artificial nature of the truncated receptors used in the latter study makes it less reliable, specially in light of FRET evidence to the contrary.

Mechanism of action of trastuzumab – Inhibition of HER2 cleavage

Trastuzumab binding inhibits the proteolytic cleavage and shedding of HER2 by ADAM proteases (Molina et al. 2001; Liu et al. 2006). This may in part inhibit the invasive properties of HER2 transformed cells since truncated HER2 induces a more invasive morphology and is associated with increased kinase activity, increased transforming efficiency, and is increased in patients with metastatic disease (Egeblad et al. 2001; Segatto et al. 1988; Molina et al. 2002; Christianson et al. 1998). Therefore trastuzumab may inhibit this aspect of HER2 function although the transforming function of HER2 is not known to require truncation and many HER2 overexpressing breast cancers do not have significant truncation of HER2.

Mechanism of action of trastuzumab – other findings

While the therapeutically important effect of trastuzumab on the function of its direct target HER2 remains to be defined, numerous reports have emerged describing the effects of trastuzumab on downstream signaling pathways. The anti-proliferative effect of mAb 4D5 or trastuzumab in cell culture models is associated with the induction of p27 and G1 block (Lane et al. 2000; Le et al. 2005; Marches and Uhr 2004). Trastuzumab affects the expression of tumor angiogenic factors and exhibits certain anti-angiogenic properties in mouse models (Izumi et al. 2002). Trastuzumab suppresses Akt signaling in some tumor cell types but not others (Longva et al. 2005; Yakes et al. 2002; Normanno et al. 2002), increases plasma PTEN localization and activity in cells (Nagata et al. 2004; Longva et al. 2005), and its anti-proliferative and anti-tumor effects are attenuated by PTEN knockdown (Nagata et al. 2004; Fujita et al. 2006). Consistent with a functional role for PTEN in clinical anti-tumor efficacy, tumors with reduced or absent PTEN are relatively resistant to trastuzumab-containing chemotherapy regimens (Nagata et al. 2004; Fujita et al. 2006). Although these data sets are complicated by the concomitant use of cytotoxic chemotherapy regimens, they are the only currently existing evidence linking intracellular signaling with the clinical antitumor activity of trastuzumab. An association between trastuzumab resistance and PTEN loss by itself does not necessarily imply that trastuzumab inhibits tumors through direct effects on tumor signaling, since PTEN loss has also been shown to mediate immuno-resistance (Parsa et al. 2006).

Mechanism of action of trastuzumab – Immunological targeting

An increasing body of evidence suggests that the in vivo anti-tumor effects of the anti-HER2 mAb 4D5 and the humanized trastuzumab may be mediated, at least in part, if not entirely, through immunological targeting mechanisms. mAb 4D5 activates ADCC in vitro (Carter et al. 1992; Clynes et al. 2000). This activity was greatly enhanced during the process to engineer the humanized version and trastuzumab is indeed highly efficient at activating ADCC in vitro (Carter et al. 1992; Cooley et al. 1999; Carson et al. 2001; Repka et al. 2003; Kubo et al. 2003). Mouse genetic models that experimentally manipulate Fc receptor function positively or negatively clearly demonstrate the role of host immunologic mechanisms in the anti-tumor efficacy of these agents. The antitumor activity of both mAb 4D5 and trastuzumab are almost entirely abolished in the loss-of-function FcRγ−/− mice, while the antitumor activity of subtherapeutic doses of mAb 4D5 and trastuzumab are greatly enhanced in the gain-of-function FcRII−/− mouse model (Clynes et al. 2000). Furthermore, when the Fc region of the mAb 4D5 is mutated at a single position to eliminate engagement of host Fcγ receptors, the mutant mAb 4D5 retains all its in vitro anti-proliferative activity but loses its in vitro ADCC activity and loses its in vivo anti-tumor efficacy (Clynes et al. 2000). This model has provided highly compelling evidence that the antitumor activity of trastuzumab is mediated, in large part, through immunological targeting of tumor cells.

Investigators have also recently begun to look for clinical evidence of immunological targeting by trastuzumab. In a clinical study of a trastuzumab-containing chemotherapy regimen compared with case-matched controls, trastuzumab treatment was associated with significantly increased numbers of activated cytolytic natural killer (NK) cells within tumors (Arnould et al. 2006). In a second study of trastuzumab monotherapy, the treatment induced varying degress of tumor infiltration with lymphoid cells and the patients that responded to trastuzumab had the highest degree of tumor lymphocyte infiltration and higher ADCC activity measured ex-vivo (Gennari et al. 2004). The evidence that trastuzumab works through an ADCC mediated mechanism has led some investigators to determine whether this activity can be further enhanced by additional immunostimulatory approaches. Consistent with this hypothesis, IL-2 enhances the in vitro ADCC activity of trastuzumab (Carson et al. 2001; Kubo et al. 2003). However a phase I trial of this combination modality found no evidence of increased clinical activity associated with IL-2 induced NK cell expansion (Repka et al. 2003). In another approach, a bi-specific antibody that targets HER2 as well as CD3 antigen on T-cells was developed in order to recruit T-cells to tumor cells. This agent showed promising evidence of activity in its phase I study and further studies are underway to determine its anti-tumor efficacy (Kiewe et al. 2006). In another approach, the in vitro ADCC activity of trastuzumab appears to be augmented by defucosylation, and the fucosyl-negative version of trastuzumab has been proposed for clinical testing (Suzuki et al. 2007).

These preclinical experimental models and clinical observational studies have provided compelling evidence that much of the anti-tumor activity of trastuzumab is mediated through immunological targeting mechanisms. Although these data sets are still relatively few, the failure of the signaling field to confirm that the clinical activities of trastuzumab are mediated through inhibition of HER2 signaling has led to more and more attention being focused on the pursuit of the immunological targeting hypothesis and additional studies to further explore this hypothesis are underway.

Targeting HER2 with pertuzumab

A second anti-HER2 mAb is currently undergoing clinical testing. From the initial panel of anti-HER2 mAbs developed at Genentech, Inc., the mAb 2C4 was also selected for further characterization and development and has properties distinct from mAb 4D5. mAb 2C4 has been modified for human clinical use by recombinant engineering to generate the humanized version pertuzumab (Adams et al. 2006). Pertuzumab is currently undergoing clinical trials and its clinical development thus far has been oriented towards diseases other than HER2 overexpressing cancers. It’s antitumor activity in HER2 overexpressing breast cancer is not yet known, but it has little activity in breast cancers without HER2 overexpression and in unselected ovarian cancers (Cortes et al. 2005; Fleming et al. 2005).

The activities of 2C4 or pertuzumab on cell signaling pathways have only been reported by selected investigators since these agents are not yet available for study to the wider scientific community. Compared with mAb 4D5 or trastuzumab, mAb 2C4 has much less anti-proliferative activity in vitro (Hudziak et al. 1989; Takai et al. 2005) but has in vivo antitumor activity in a number of tumor types including tumors without HER2 overexpression (Agus et al. 2002; Takai et al. 2005). 2C4 is reported to inhibit heregulin mediated HER2 receptor complex formation, phosphorylation, and MAPK and Akt activation in breast cancer cells without HER2 overexpression (Agus et al. 2002). Determination of the effects of pertuzumab on HER2 overexpressing cancers awaits much more preclinical and clinical studies.

Targeting HER2 with HER kinase inhibitors

The technology to develop selective tyrosine kinase inhibitors (TKIs) for human use succeeded antibody therapeutics by almost a decade. These agents, at least in theory, have certain advantages over antibody therapies for the treatment of HER2 amplified cancers. Antibody therapies are cell impermeable agents that bind the extracellular domain of HER2 and to this day it remains unclear whether or how this binding activity can suppress oncogenic HER2 function, although they may induce the clinically important immunological targeting of HER2 overexpressing cancer cells. TKIs are cell permeable agents and can potentially inhibit the ligand dependent and independent kinase activity of HER2 residing within the intracellular domain. This strategy has a solid rational basis since kinase activity is essential for the oncogenic function of neu or HER2 (Weiner et al. 1989; Qian et al. 1994). At least in theory, these agents offer the opportunity to inactivate HER2 kinase function in patients with HER2 overexpressing cancer and for the first time directly test the HER2 oncogene hypothesis in patients. However TKIs don’t have the singular target specificity of antibodies and their off-target effects potentially limits their therapeutic index compared with antibodies.

The development of HER-selective TKIs

Synthetic and natural product inhibitors of HER kinases from diverse structures were initially studied in the early 1990s, but their limited potencies and specificities precluded their usefullness as anti-tumor agents. The field was revolutionized by the discovery of modified quinazoline compounds as highly specific and potent inhibitors of the epidermal growth factor receptor (Ward et al. 1994; Rewcastle et al. 1995). Extensive structure-activity relationships were determined and numerous improved quinazoline compounds have subsequently been developed with varying selectivity characteristics for individual HER family members. In addition to quinazolines, several other structures have now been found to potently and selectively inhibit HER kinases. Table 2 lists a number of HER TKIs that have been publicly disclosed, and for which preclinical data has been presented. In addition to these, numerous other agents are in development which are not yet disclosed at the time of this review. Almost all of these agents are ATP analogs and inhibit kinase activity by binding within the ATP pocket of the catalytic domain (Stamos et al. 2002; Wood et al. 2004). Some of these compounds bind reversibly within the ATP pocket and are competitive with ATP while others bind irreversibly and are non-competitive with ATP.

TABLE 2.

List of disclosed HER family TKIs in preclinical and clinical development.

Developer Compound Class Type Selectivity Activity demonstrated against these HER2+++ tumor models References

in vitro IC50 (nM)
EGFR HER
2
HER
4
in vitro in vivo
AstraZeneca ZD1839 gefitinib quinazoline reversible 27 370
0
BT474, SKBr3, SKOv3, MCF-7HER2 BT474, MCF-7HER2, GLM-1, GLM-4 (Wakeling et al. 2002; Moasser et al. 2001; Moulder et al. 2001; Ghossein and Bhattacharya 2001; Warburton et al. 2004; Yokoyama et al. 2006)
OSI-Genentech-Roche OSI-774 erlotinib quinazoline reversible 2 350 KPL-4, Calu-3 (Moyer et al. 1997; Akita and Sliwkowski 2003; Friess et al. 2005)
GlaxoSmithKline GW572016 lapatinib thio-quinazoline reversible 11 9 367 BT474, Calu3, N87, HB4aHER2, SUM225, H16N2HER2, SUM190, SKBr3, UACC893, UACC812 BT474 (Rusnak et al. 2001; Konecny et al. 2006)
ParkeDavis-Pfizer CI-1033 PD183805 canertinib quinazoline irreversible 0.8 19 7 (Rabindran 2005; Slichenmyer et al. 2001)
OSI-Pfizer CP-724714 quinazoline reversible 4300 8 SKBr3, another panel of 9 FREV664E-HER2 (Jani et al. 2004b; Jani et al. 2004a; Finn et al. 2004)
OSI-Pfizer CP-654577 quinazoline reversible 670 11 BT474, SKBr3 FREV664E-HER2 (Rabindran 2005; Barbacci et al. 2003)
Wyeth-Ayerst CL-387785 EKI-785 quinazoline irreversible 0.4 SKBr3, 3T3HER2 (Discafani et al. 1999; Torrance et al. 2000)
Academic AG1478 quinazoline reversible 3 200
0
SKBr3, BT474 MMTV-neu (Levitzki and Gazit 1995; Kurokawa et al. 2000; Lenferink et al. 2001; Lenferink et al. 2000)
Arry Biopharma Arry-334543 quinazoline reversible 7 2 10 BT474 BT474, Calu3, MDA-453 (Miknis et al. 2005; Pheneger et al. 2005)
Boehringer Ingelheim BIBW-2992 quinazoline irreversible 0.5 14 BT474, N87 MDA-453, N87, SKOv3 (Solca et al. 2006)
AVEO Pharm.-Misubishi AV-412 MP-412 quinazoline irreversible 1 18 BT474, KPL-4, BH248 (Robinson and Lin 2006; Fujii et al. 2005)
Novartis AEE-788 pyrrolopyrimidine reversible 6 6 160 BT474, SKBr3 HC11neuT (Traxler et al. 2004)
Novartis CGP-59326A pyrrolopyrimidine reversible 70 >10 uM SKBr3, ZR75-30 (Lydon et al. 1998)
Novartis PKI-166 CGP-75166 pyrrolopyrimidine reversible 1 11 430 BT474, SKBr3 HC11neuT (Traxler et al. 2001; Brandt et al. 2001)
Wyeth-Ayerst EKB-569 pelitinib cyanoquinoline irreversible 39 120
0
SKBr3 (Wissner et al. 2003; Torrance et al. 2000)
Wyeth-Ayerst HKI-272 cyanoquinoline irreversible 92 59 3T3neu, SKBr3, BT474 3T3neu,BT474, SKOv3 (Rabindran et al. 2004; Tsou et al. 2005)
Wyeth-Ayerst HKI-357 cyanoquinoline irreversible 33 34 SKBr3 3T3neu,BT474, SKOv3 (Kwak et al. 2005; Tsou et al. 2005)
Bristol Myers Squibb BMS-599626 pyrrolotriazine reversible 22 32 190 BT474,N87,KPL4, HCC1419,HCC1954, Sal2 KPL4, Sal2 (Wong et al. 2006)
Takeda TAK-165 mubritinib triazole >25u M 6 BT474 BT474 (Naito et al. 2002; Yoshida et al. 2002; 2006)

Although HER family kinases are highly homologous, many TKIs show selectivity among the individual family members when assayed by in vitro kinase assays using purified EGFR,HER2, or HER4 kinases, and these are listed in Table 2. However the biological relevance of these in vitro characteristics is not evident in cell based assays. The EGFR selective agent gefitinib inhibits the phosphorylation of all of the HER proteins in cells, as does the HER2-selective agent CP-654577 (Moasser et al. 2001; Normanno et al. 2002; Shepherd et al. 2001). In cell proliferation assays, HER2 overexpressing tumors are particularly sensitive to the highly EGFR-selective TKIs gefitinib, AG1478, CGP-59326A, and EKB-659 (see Table 2). In fact in an engineered HER2 overexpressing tumor model, the level of HER2 overexpression directly correlates with sensitivity to the EGFR-selective TKI AG1478 (Emlet et al. 2006). The mechanisms underlying the activities of EGFR-selective TKIs against HER2 signaling and HER2-driven growth are not currently understood, and may be due to the direct inhibition of HER2 kinase by their weaker anti-HER2 activity, high intracellular concentrations of these agents precluding their target selectivities, or a necessary role for EGFR in HER2-driven growth. The latter appears unlikely since at least in fibroblast models, HER2 is transforming even in the absence of EGFR expression (Chazin et al. 1992). These uncertainties with respect to the biologic importance of target-specificity are somewhat academic and virtually all HER TKIs show activity against HER2-driven tumors in mouse models regardless of their in vitro target selectivities (see Table 2) and they are all potentially candidate agents to test the validity of the HER2 treatment hypothesis in patients with HER2 overexpressing cancers.

Clinical anti-tumor activity of HER TKIs – Current data

Numerous TKIs are currently in various stages of preclinical and clinical development. The clinical development of each of these is prioritized towards specific cancer subtypes by their sponsors, therefore the activities of some of them against HER2 driven cancer may not be tested early in their course of development. But a limited amount of clinical data is now available to give a preliminary impression of the anti-tumor efficacy of this class of TKIs in patients with HER2 overexpressing cancers. The currently reported phase II efficacy data is summarized in Table 3. In addition to the drugs listed here, numerous multi-targeting TKIs that target other kinase families in addition to HER family kinases are in preclinical and clinical testing and although such drugs may be found to be active in the treatment of HER2-driven cancers, their multi-targeting characteristics make them less suited to test the HER2 oncogene hypothesis. Completed phase II studies of gefitinib and erlotinib have been reported in breast cancer patients. Although these were not conducted specifically in patients with HER2 overexpressing cancer, the cohorts include patients with HER2 overexpressing disease. Overall response rates of 0–10% were seen in these studies. The most informative data currently comes from clinical studies of lapatinib. Lapatinib has specifically been developed for the treatment of HER2 overexpressing breast cancer and numerous clinical efficacy studies as well as correlative scientific studies have been conducted and are ongoing to determine the activity of this agent in patients with HER2 overexpressing breast cancer. The phase II efficacy studies completed and confirmed to date show a response rate of 4–8% in patients with HER2 overexpressing breast cancer. Two additional ongoing but unconfirmed studies have reported higher response rates in the 24–30% range. A number of additional phase II studies are underway to test the efficacy of other investigational HER TKIs in patients with HER2 overrexpressing breast cancer and there will be much more data emerging in the next few years . The data so far shows evidence of weak clinical anti-tumor activity in this disease. In addition to the studies reviewed here, numerous clinical studies are underway to determine whether the addition of TKIs to cytotoxic chemotherapeutics, to trastuzumab, or to hormonal therapies produces new combinations with increased clinical activity and prolongation of survival in patients. These studies may lead to better treatment options for patients, but they are not a direct test of the HER2 oncogene hypothesis and a detailed discussion of these studies is beyond the scope of this review.

TABLE 3.

Results of currently presented phase II efficacy studies of HER TKIs in HER2 overexpressing cancers

Reference Drug phase selection criteria N Response
Rate
comments
(Baselga et al. 2005) gefitinib/ZD1839 phase II advanced breast ca 31 3%
(Robertson et al. 2003) gefitinib/ZD1839 phase II advanced breast ca - TAM-resistant 19 10%
(Albain et al. 2002) gefitinib/ZD1839 phase II advanced breast ca 63 2% 28 HER2+++ patients
(Winer et al. 2002) erlotinib/OSI-774 phase II advanced breast ca 69 3% many HER2+++ patients
(Tan et al. 2004) erlotinib/OSI-774 phase II advanced breast ca 18 0% 2 HER2+++ patients
(Blackwell et al. 2006; Blackwell et al. 2004) lapatinib/GW572016 phase II HER2+++ breast ca - after trastuzumab 81 8%
(Burstein et al. 2004) lapatinib/GW572016 phase II HER2+++ breast ca - after chemo 140 4%
(Iwata et al. 2006) lapatinib/GW572016 phase II HER2+++ breast ca - after chemo 45 24% (preliminary)
(Gomez et al. 2005) lapatinib/GW572016 phase II HER2+++ breast ca - first line 40 30% (preliminary)
(Campos et al. 2005) canertinib/CI-1033 phase II ovarian ca - after chemo 105 0% 2 HER2+++ patients

Evidence that HER TKIs inhibit HER2 function in patients

The HER2 oncogene hypothesis would predict that the vast majority of HER2-driven tumors would initially respond to treatments that suppress HER2 kinase function. Correlative studies of tumor material from patients on treatment are essential for understanding whether HER2 function and signaling has effectively been suppressed by these treatments. Such correlative studies require purely research interventions in consenting patients by performing biopsies of their tumors just before and while on therapy and these studies are extremely difficult to accomplish for a myriad of practical and ethical reasons. At least two groups thus far have succeeded in generating clinical scientific data in patients on HER TKIs. In a phase I clinical study of lapatinib, tumor biopsies were obtained before and while on therapy to determine the suppression of tumor EGFR/HER2 signaling by immunohistochemical staining (Spector et al. 2005). This study showed mixed results with variable degrees of suppression of targets, partly because this was a phase I dose escalation study involving patients with different types of cancers, including cancers not known to be HER2-dependent, and starting dose levels that are likely less effective at target suppression. The data does however show a reduction in EGFR and HER2 phosphorylation in most patients, and a reduction in MAP kinase signaling. A reduction in Akt signaling was less evident in this data set. In a phase II study of gefitinib in breast cancer patients, skin biopsies and tumor biopsies were obtained in many patients before and while on therapy for immunohistochemical analysis of target suppression (Baselga et al. 2005). This study showed effective suppression of EGFR phosphorylation and MAPK signaling in skin and tumor from drug therapy, but no suppression of Akt signaling. HER2 phosphorylation was not assayed on this study and the 3 patients with HER2 overexpressing tumors that were enrolled on this study did not have on-treatment tumor biopsies for analysis. It should be noted that the use of immunohistochemical techniques on paraffin embedded tissues using phospho-specific antibodies is fraught with technical problems that limits their reliability, and until newer techniques emerge, such studies should be interpreted with caution (Baker et al. 2005). Despite the technical problems associated with phosphoprotein immunostaining and the fact that these two studies were not designed to specifically determine target inactivation in HER2 overexpressing tumors at maximal drug dosage, they do appear to suggest that the drugs do reach their tumor targets and at least partially inactivate them. Tumor biodistribution does not seem to be a limiting step since at least with regards to gefitinib, tumor and tissue concentrations have been measured and are much higher than serum concentrations and well above concentrations that fully suppress EGFR and HER2 signaling in cell culture models (McKillop et al. 2005).

Evidence that TKIs do not fully inhibit oncogenic HER2 functions

Significant mechanistic insight into the effective suppression of oncogenic HER2 signaling by TKIs was recently afforded by the analysis of steady state HER3 and downstream Akt signaling. While TKI treatment effectively suppresses EGFR and HER2 autophosphorylation and downstream MAP kinase signaling in HER2 amplified tumors, HER3 appears to escape TKI therapy at conventional doses and concentrations (Sergina et al. 2007). This is due to Akt-driven negative feebdack signaling which restores HER3 signaling activity despite significant suppression of HER2 kinase function thereby maintaining downstream Akt signaling and numerous Akt-driven pathways important for tumor survival (Sergina et al. 2007). This feedback loop essentially buffers HER3 signaling against the incomplete loss of HER2 kinase function and underscores the tumor cells critical need to maintain Akt signaling and numerous Akt-driven pathways important for tumor survival. The fact that HER3 signaling is buffered against an incomplete inactivation of HER2 kinase raises the bar for drug development because it suggests that proper testing of the HER2 oncogene hypothesis will require drugs that can completely inactivate HER2 kinase function. Testing this principle in cell culture models with much higher concentrations of TKI or with the addition of anti-HER3 siRNA approaches shows that HER2-overexpressing tumor cells will undergo apoptosis if HER2 function, including its transactivation of HER3 and Akt signaling, is interrupted for 48 hours or longer (Sergina et al. 2007). This reaffirms the oncogene-addicted nature of HER2-driven tumors and suggests that if HER2 can be effectively inactivated in patients tumors, this could produce highly significant and rapid anti-tumor responses. But the doses required to completely inactivate HER2 and effectively suppress HER3 signaling will likely produce significant toxicities in patients due to their off-target effects and may not be safely achievable. The effective suppression of oncogenic HER2 function in patients may require drugs that are much more potent than the current generation of compounds and at the same time are highly selective with a wide therapeutic index.

Other approaches to treat HER2 overexpressing cancers

Numerous other pharmacologic approaches are being pursued to develop effective therapies for patients with HER2 overexpressing cancers. These include anti-HER2 antibodies conjugated to a number of cellular toxins or anti-HER2 antibodies placed on chemotherapy containing immunoliposomes that can deliver cytotoxics more effectively to HER2 overexpressing cancer cells. A number of agents can interfere with cellular mechanisms that regulate gene expression or protein expression and these can reduce HER2 expression in tumor cells. These include histone deacetylase inhibitors, HSP90 inhibitors, and COX-2 inhibitors. While these promising treatment approaches are a consequence of the HER2 oncogene hypothesis, their non-specific mechanisms of action limits their usefullness in validating the hypothesis and are reviewed here. Readers are referred to several excellent recent reviews for a comprehensive coverage of these HER2 targeting approaches (Kumar and Pegram 2006; Menard et al. 2003; Chen et al. 2003; Brand et al. 2006). Numerous immunological modalities are also being pursued to enable host defense mechanisms to target HER2 overexpressing cancer cells, and these are also beyond the scope of this review. In addition, numerous approaches to specifically suppress HER2 expression have been postulated over the years and continue to be developed. These include antisense, ribozyme, and siRNA approaches to inhibit HER2 expression. These modalities have not yet translated to clinically effective products that can test the validity of the HER2 oncogene hypothesis in patients. Future development of these technologies, in particular siRNA approaches, may lead to a new class of agents that can specifically and effectively inactivate oncogenic HER2 function.

Resistance to inhibitors of HER2

Numerous mutational events downstream of growth factor receptors have now been described in cancers. These include activating mutations in BRAF, Ras, PIK3CA, and inactivating mutations or deletions of PTEN. Since these genes function downstream of HER2 and since each of these mutations induces constitutive signaling activity, at least in theory these mutations can uncouple downstream pathways from HER2 rendering tumor growth independent of HER2 and resistant to inhibitors of HER2. In breast cancers Ras and BRAF mutations are rare, but HER2 overexpression occurs frequently with PIK3CA mutations, but rarely with PTEN mutation (Saal et al. 2005). There is currently no data to determine whether the co-existence of PIK3CA mutation confers resistance to TKIs in HER2 overexpressing breast cancers. Of note, the commonly used BT474 cell line commonly used as a model of HER2 amplified breast cancer, and which is sensitive to TKIs and trastuzumab, harbors an unsual mutation in exon I of PIK3CA ((Saal et al. 2005) and cosmic database). PTEN mutations are rare in breast cancers, but reduced PTEN expression may have biological significance and is often seen in breast cancers (Bose et al. 2002). PTEN loss has been shown to be induce TKI resistance in an EGFR amplified breast cancer cell line (She et al. 2003; Bianco et al. 2003), but the analogous effect has not been shown in a HER2 amplified model. Future studies will establish whether PIK3CA and PTEN are important determinants of TKI sensitivity in HER2 overexpressing tumors. However the analysis of clinical resistance will only be meaningful if drugs have been developed that fully inactivate HER2 function. As discussed above in this review, trastuzumab does not seem to inactivate HER2 and its mechanism of action remains unclear, and TKIs studied to date seem to be partial inhibitors of oncogenic HER2 signaling in vivo.

Current status of the HER2 oncogene hypothesis

The preliminary data with lapatinib is a promising hint that at least a minority of HER2 overexpressing tumors may be dependent on HER2 kinase function. But why the majority of patients fail to respond to this therapy remains to be determined. It is possible that the HER2 oncogene hypothesis is wrong, and that despite the abundant and highly compelling evidence from experimental models that HER2 overexpressing breast cancers are HER2-driven and HER2-dependent, that this hypothesis does not hold true for patients with breast cancer and the experimental models are too simplistic to predict the behavior of naturally occuring cancers. However the HER2 oncogene hypothesis can only be discounted if clinical studies show lack of anti-tumor efficacy in spite of effective inactivation of tumor HER2 function and this has certainly not been shown. The alternative possibility is that current therapies do not effectively suppress oncogenic HER2 function in tumors. The recent revelation that current TKIs are not effectively suppressing HER3/PI3K/Akt signaling strongly supports this position, and opens the way for a future generation of TKIs.

Therefore it appears almost surely the case that the HER2 oncogene hypothesis has not yet been effectively tested in patients. Much more data will be forthcoming in the coming years with regards to numerous other HER TKIs with distinct structural and biochemical properties, including their anti-tumor efficacies and their biochemical target effects in patients tumors. If these agents are unable to completely inactivate HER2, then we may have to await a newer generation of more potent agents. Alternatively, HER2 oncogenic function can be inactivated by the combination of a HER TKI and an inhibitor of the HER3/PI3k/Akt signaling pathway. Inhibitors of this pathway are also in preclinical development and the combination treatment hypothesis will likely also be tested in the coming years as these agents enter clinical testing phases.

The scientific evidence implicates a role for HER2 in breast cancer similar to bcr-abl in chronic myelogenous leukemia (CML). Since effective inhibition of bcr-abl produces complete remissions in nearly all patients in chronic phase CML, it is imperative that the analogous treatment hypothesis regarding HER2 be effectively tested in breast cancer. The potential inherent in this hypothesis is enormous and includes the scenario of the first instance of an epithelial cancer being eradicated even in advanced stages. Such an outcome would have historic significance.

References

  1. Adams CW, Allison DE, Flagella K, Presta L, Clarke J, Dybdal N, et al. Humanization of a recombinant monoclonal antibody to produce a therapeutic HER dimerization inhibitor, pertuzumab. Cancer Immunol Immunother. 2006;55:717–727. doi: 10.1007/s00262-005-0058-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Agus DB, Akita RW, Fox WD, Lewis GD, Higgins B, Pisacane PI, et al. Targeting ligand-activated ErbB2 signaling inhibits breast and prostate tumor growth. Cancer Cell. 2002;2:127–137. doi: 10.1016/s1535-6108(02)00097-1. [DOI] [PubMed] [Google Scholar]
  3. Akita RW, Sliwkowski MX. Preclinical studies with Erlotinib (Tarceva) Seminars in Oncology. 2003;30(3) suppl 7:15–24. [PubMed] [Google Scholar]
  4. Albain K, Elledge R, Gradishar WJ, Hayes DF, Rowinsky EK, Hudis C, et al. Open-label phase II multicenter trial of ZD1839 ('Iressa') in patients with advanced breast cancer. Breast Cancer Res Treat. 2002;76 S33 #20. [Google Scholar]
  5. Arnould L, Gelly M, Penault-Llorca F, Benoit L, Bonnetain F, Migeon C, et al. Trastuzumab-based treatment of HER2-positive breast cancer: an antibody-dependent cellular cytotoxicity mechanism? Br J Cancer. 2006;94:259–267. doi: 10.1038/sj.bjc.6602930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Austin CD, De Maziere AM, Pisacane PI, van Dijk SM, Eigenbrot C, Sliwkowski MX, et al. Endocytosis and sorting of ErbB2 and the site of action of cancer therapeutics trastuzumab and geldanamycin. Mol Biol Cell. 2004;15:5268–5282. doi: 10.1091/mbc.E04-07-0591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Baasner S, von MH, Klenner T, Hilgard P, Beckers T. Reversible tumorigenesis in mice by conditional expression of the HER2/c-erbB2 receptor tyrosine kinase. Oncogene. 1996;13:901–911. [PubMed] [Google Scholar]
  8. Bacus SS, Stancovski I, Huberman E, Chin D, Hurwitz E, Mills GB, et al. Tumor-inhibitory monoclonal antibodies to the HER-2/Neu receptor induce differentiation of human breast cancer cells. Cancer Res. 1992;52:2580–2589. [PubMed] [Google Scholar]
  9. Baker AF, Dragovich T, Ihle NT, Williams R, Fenoglio-Preiser C, Powis G. Stability of phosphoprotein as a biological marker of tumor signaling. Clin Cancer Res. 2005;11:4338–4340. doi: 10.1158/1078-0432.CCR-05-0422. [DOI] [PubMed] [Google Scholar]
  10. Barbacci EG, Pustilnik LR, Rossi AM, Emerson E, Miller PE, Boscoe BP, et al. The biological and biochemical effects of CP-654577, a selective erbB2 kinase inhibitor, on human breast cancer cells. Cancer Research. 2003;63:4450–4459. [PubMed] [Google Scholar]
  11. Baselga J, Albanell J, Ruiz A, Lluch A, Gascon P, Guillem V, et al. 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]
  12. Baselga J, Norton L, Albanell J, Kim YM, Mendelsohn J. Recombinant humanized anti-HER2 antibody (Herceptin) enhances the antitumor activity of paclitaxel and doxorubicin against HER2/neu overexpressing human breast cancer xenografts. Cancer Res. 1998;58:2825–2831. [PubMed] [Google Scholar]
  13. Baselga J, Tripathy D, Mendelsohn J, Baughman S, Benz CC, Dantis L, et al. Phase II study of weekly intravenous recombinant humanized anti-p185HER2 monoclonal antibody in patients with HER2/neu-overexpressing metastatic breast cancer. J Clin Onc. 1996;14:737–744. doi: 10.1200/JCO.1996.14.3.737. [DOI] [PubMed] [Google Scholar]
  14. Beerli RR, Wels W, Hynes NE. Intracellular expression of single chain antibodies reverts ErbB-2 transformation. J Biol Chem. 1994;269:23931–23936. [PubMed] [Google Scholar]
  15. Belimezi MM, Papanastassiou D, Merkouri E, Baxevanis CN, Mamalaki A. Growth inhibition of breast cancer cell lines overexpressing Her2/neu by a novel internalized fully human Fab antibody fragment. Cancer Immunol Immunother. 2006;55:1091–1099. doi: 10.1007/s00262-005-0100-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Bianco R, Shin I, Ritter CA, Yakes FM, Basso A, Rosen N, et al. 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]
  17. Blackwell K, Kaplan EH, Franco SX, Marcom PK, Maleski MJ, Sorenson MS, Berger MS. A phase II, open-label, multicenter study of GW572016 in patients with trastuzumab-refractory metastatic breast cancer. Proc Amer Soc Clin Onc. 2004;22 # 3006. [Google Scholar]
  18. Blackwell KL, Burstein H, Pegram M, Storniolo A, Salazar VM, Maleski JE, Lin X, Spector N, Stein SH, Berger MS. Determining Relevant Biomarkers from Tissue and Serum that May Predict Response to Single Agent Lapatinib in Trastuzumab Refractory Metastatic Breast Cancer. Proc Amer Soc Clin Onc. 2006;23 #3004. [Google Scholar]
  19. Bookman MA, Darcy KM, Clarke-Pearson D, Boothby RA, Horowitz IR. Evaluation of monoclonal humanized anti-HER2 antibody, trastuzumab, in patients with recurrent or refractory ovarian or primary peritoneal carcinoma with overexpression of HER2: a phase II trial of the Gynecologic Oncology Group. J Clin Oncol. 2003;21:283–290. doi: 10.1200/JCO.2003.10.104. [DOI] [PubMed] [Google Scholar]
  20. Bose S, Crane A, Hibshoosh H, Mansukhani M, Sandweis L, Parsons R. Reduced expression of PTEN correlates with breast cancer progression. Hum Pathol. 2002;33:405–409. doi: 10.1053/hupa.2002.124721. [DOI] [PubMed] [Google Scholar]
  21. Brand FX, Ravanel N, Gauchez AS, Pasquier D, Payan R, Fagret D, et al. Prospect for anti-HER2 receptor therapy in breast cancer. Anticancer Res. 2006;26:463–470. [PubMed] [Google Scholar]
  22. Brandt R, Wong AM, Hynes NE. Mammary glands reconstituted with Neu/ErbB2 transformed HC11 cells provide a novel orthotopic tumor model for testing anti-cancer agents. Oncogene. 2001;20:5459–5465. doi: 10.1038/sj.onc.1204709. [DOI] [PubMed] [Google Scholar]
  23. Burstein H, Storniolo A, Franco S, Salazar VM, Sorenson MS, Stein SH. A phase II, open-label, multicenter study of lapatinib in two cohorts of patients with advanced or metastatic breast cancer who have progressed while receiving trastuzumab-containing regimens. Annals of Oncology. 2004;15 suppl 3:27. (abstract 1040) [Google Scholar]
  24. Burstein HJ, Harris LN, Marcom PK, Lambert-Falls R, Havlin K, Overmoyer B, et al. Trastuzumab and vinorelbine as first-line therapy for HER2-overexpressing metastatic breast cancer: multicenter phase II trial with clinical outcomes, analysis of serum tumor markers as predictive factors, and cardiac surveillance algorithm. J Clin Oncol. 2003;21:2889–2895. doi: 10.1200/JCO.2003.02.018. [DOI] [PubMed] [Google Scholar]
  25. Campos S, Hamid O, Seiden MV, Oza A, Plante M, Potkul RK, et al. Multicenter, randomized phase II trial of oral CI-1033 for previously treated advanced ovarian cancer. J Clin Oncol. 2005;23:5597–5604. doi: 10.1200/JCO.2005.08.091. [DOI] [PubMed] [Google Scholar]
  26. Carson WE, Parihar R, Lindemann MJ, Personeni N, Dierksheide J, Meropol NJ, et al. Interleukin-2 enhances the natural killer cell response to Herceptin-coated Her2/neu-positive breast cancer cells. Eur J Immunol. 2001;31:3016–3025. doi: 10.1002/1521-4141(2001010)31:10<3016::aid-immu3016>3.0.co;2-j. [DOI] [PubMed] [Google Scholar]
  27. Carter P, Presta L, Gorman CM, Ridgway JB, Henner D, Wong WL, et al. Humanization of an anti-p185HER2 antibody for human cancer therapy. Proc Natl Acad Sci U S A. 1992;89:4285–4289. doi: 10.1073/pnas.89.10.4285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Chazin VR, Kaleko M, Miller AD, Slamon DJ. Transformation mediated by the human HER-2 gene independent of the epidermal growth factor receptor. Oncogene. 1992;7:1859–1866. [PubMed] [Google Scholar]
  29. Chen JS, Lan K, Hung MC. Strategies to target HER2/neu overexpression for cancer therapy. Drug Resist Updat. 2003;6:129–136. doi: 10.1016/s1368-7646(03)00040-2. [DOI] [PubMed] [Google Scholar]
  30. Cho HS, Mason K, Ramyar KX, Stanley AM, Gabelli SB, Denney DW, Jr, et al. Structure of the extracellular region of HER2 alone and in complex with the Herceptin Fab. Nature. 2003;421:756–760. doi: 10.1038/nature01392. [DOI] [PubMed] [Google Scholar]
  31. Choudhury A, Charo J, Parapuram SK, Hunt RC, Hunt DM, Seliger B, et al. Small interfering RNA (siRNA) inhibits the expression of the Her2/neu gene, upregulates HLA class I and induces apoptosis of Her2/neu positive tumor cell lines. Int J Cancer. 2004;108:71–77. doi: 10.1002/ijc.11497. [DOI] [PubMed] [Google Scholar]
  32. Christianson TA, Doherty JK, Lin YJ, Ramsey EE, Holmes R, Keenan EJ, et al. NH2-terminally truncated HER-2/neu protein: relationship with shedding of the extracellular domain and with prognostic factors in breast cancer. Cancer Res. 1998;58:5123–5129. [PubMed] [Google Scholar]
  33. Citri A, Alroy I, Lavi S, Rubin C, Xu W, Grammatikakis N, et al. Drug-induced ubiquitylation and degradation of ErbB receptor tyrosine kinases: implications for cancer therapy. EMBO J. 2002;21:2407–2417. doi: 10.1093/emboj/21.10.2407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Clynes RA, Towers TL, Presta LG, Ravetch JV. Inhibitory Fc receptors modulate in vivo cytoxicity against tumor targets. [see comment] Nature Medicine. 2000;6:443–446. doi: 10.1038/74704. [DOI] [PubMed] [Google Scholar]
  35. Colomer R, Lupu R, Bacus SS, Gelmann EP. erbB-2 antisense oligonucleotides inhibit the proliferation of breast carcinoma cells with erbB-2 oncogene amplification. Br J Cancer. 1994;70:819–825. doi: 10.1038/bjc.1994.405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Cooley S, Burns LJ, Repka T, Miller JS. Natural killer cell cytotoxicity of breast cancer targets is enhanced by two distinct mechanisms of antibody-dependent cellular cytotoxicity against LFA-3 and HER2/neu. Exp Hematol. 1999;27:1533–1541. doi: 10.1016/s0301-472x(99)00089-2. [DOI] [PubMed] [Google Scholar]
  37. Cortes JE, Baselga J, Kellokumpu-Lehtinen, Pirkko Liisa, Bianchi G, Cameron D, Miles D, Salvagni S, Wardley A, Goeminne J, Gianni L. Open label, randomized, phase II study of pertuzumab (P) in patients (pts) with metastatic breast cancer (MBC) with low expression of HER2. Proc Amer Soc Clin Onc. 2005;23 #3068. [Google Scholar]
  38. Cuello M, Ettenberg SA, Clark AS, Keane MM, Posner RH, Nau MM, et al. Down-regulation of the erbB-2 receptor by trastuzumab (herceptin) enhances tumor necrosis factor-related apoptosis-inducing ligand-mediated apoptosis in breast and ovarian cancer cell lines that overexpress erbB-2. Cancer Res. 2001;61:4892–4900. [PubMed] [Google Scholar]
  39. De LC, Cozzolino R, Carpentieri A, Pucci P, Laccetti P, D'Alessio G. Biological properties of a human compact anti-ErbB2 antibody. Carcinogenesis. 2005;26:1890–1895. doi: 10.1093/carcin/bgi146. [DOI] [PubMed] [Google Scholar]
  40. Deshane J, Grim J, Loechel S, Siegal GP, Alvarez RD, Curiel DT. Intracellular antibody against erbB-2 mediates targeted tumor cell eradication by apoptosis. Cancer Gene Ther. 1996;3:89–98. [PubMed] [Google Scholar]
  41. Diermeier S, Horvath G, Knuechel-Clarke R, Hofstaedter F, Szollosi J, Brockhoff G. Epidermal growth factor receptor coexpression modulates susceptibility to Herceptin in HER2/neu overexpressing breast cancer cells via specific erbB-receptor interaction and activation. Exp Cell Res. 2005;304:604–619. doi: 10.1016/j.yexcr.2004.12.008. [DOI] [PubMed] [Google Scholar]
  42. Discafani CM, Carroll ML, Floyd MB, Jr, Hollander IJ, Husain Z, Johnson BD, et al. Irreversible inhibition of epidermal growth factor receptor tyrosine kinase with in vivo activity by N-[4-[(3-bromophenyl)amino]-6-quinazolinyl]-2-butynamide (CL-387,785) Biochem Pharmacol. 1999;57:917–925. doi: 10.1016/s0006-2952(98)00356-6. [DOI] [PubMed] [Google Scholar]
  43. Drebin JA, Link VC, Greene MI. Monoclonal antibodies reactive with distinct domains of the neu oncogene-encoded p185 molecule exert synergistic anti-tumor effects in vivo. Oncogene. 1988;2:273–277. [PubMed] [Google Scholar]
  44. Drebin JA, Link VC, Stern DF, Weinberg RA, Greene MI. Down-modulation of an oncogene protein product and reversion of the transformed phenotype by monoclonal antibodies. Cell. 1985;41:697–706. doi: 10.1016/s0092-8674(85)80050-7. [DOI] [PubMed] [Google Scholar]
  45. Egeblad M, Mortensen OH, Jaattela M. Truncated ErbB2 receptor enhances ErbB1 signaling and induces reversible, ERK-independent loss of epithelial morphology. Int J Cancer. 2001;94:185–191. doi: 10.1002/ijc.1459. [DOI] [PubMed] [Google Scholar]
  46. Emlet DR, Schwartz R, Brown KA, Pollice AA, Smith CA, Shackney SE. HER2 expression as a potential marker for response to therapy targeted to the EGFR. Br J Cancer. 2006;94:1144–1153. doi: 10.1038/sj.bjc.6603078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Faltus T, Yuan J, Zimmer B, Kramer A, Loibl S, Kaufmann M, et al. Silencing of the HER2/neu gene by siRNA inhibits proliferation and induces apoptosis in HER2/neu-overexpressing breast cancer cells. Neoplasia. 2004;6:786–795. doi: 10.1593/neo.04313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Finn RS, Wilson CA, Chen J, Glaspy P, Dering J, Cook A, Yang E, Sanders J, Britten C, Slamon DJ. Biologic effects of CP-724,714, a selective HER-2/neu kinase inhibitor, on human breast cancer cells with variable expression of EGFR and HER-2. Proc Am Assoc Can Res. 2004 Jan 3;45 #4556. [Google Scholar]
  49. Fleming G, Gordon MS, Matei D, Aghajanian C, Matulonis UA, Brewer MA, Fleming, Hainsworth JD, Garcia AA, Pegram M, Karlan BY. Clinical activity of pertuzumab (rhuMab 2C4) in advanced, refractory or recurrent ovarian cancer (OC), and the role of HER2 activation status. Proc Amer Soc Clin Onc. 2005;23 doi: 10.1200/JCO.2005.05.4221. #5051. [DOI] [PubMed] [Google Scholar]
  50. Fleming GF, Sill MA, Thigpen JT, Adler LM, Berek JS, DiSilvestro PA, Horowitz IR. Phase II evaluation of trastuzumab in patients with advanced or recurrent endometrial carcinoma: A report on GOG 181B. Proc Amer Soc Clin Onc. 2003;22 #1821. [Google Scholar]
  51. Friess T, Scheuer W, Hasmann M. Combination treatment with erlotinib and pertuzumab against human tumor xenografts is superior to monotherapy. Clin Cancer Res. 2005;11:5300–5309. doi: 10.1158/1078-0432.CCR-04-2642. [DOI] [PubMed] [Google Scholar]
  52. Fujii A, Suzuki T, Ohya JI, Nakamura H, Fujita F, Koike M, et al. MP-412, a dual EGFR/HER2 tyrosine kinase inhibitor: 2. In vivo antitumor effects. Proc Am Assoc Can Res. 2005;46 #3394. [Google Scholar]
  53. Fujita T, Doihara H, Kawasaki K, Takabatake D, Takahashi H, Washio K, et al. PTEN activity could be a predictive marker of trastuzumab efficacy in the treatment of ErbB2-overexpressing breast cancer. Br J Cancer. 2006 doi: 10.1038/sj.bjc.6602926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Garrett TP, McKern NM, Lou M, Elleman TC, Adams TE, Lovrecz GO, et al. The crystal structure of a truncated ErbB2 ectodomain reveals an active conformation, poised to interact with other ErbB receptors. Mol Cell. 2003;11:495–505. doi: 10.1016/s1097-2765(03)00048-0. [DOI] [PubMed] [Google Scholar]
  55. Gennari R, Menard S, Fagnoni F, Ponchio L, Scelsi M, Tagliabue E, et al. Pilot study of the mechanism of action of preoperative trastuzumab in patients with primary operable breast tumors overexpressing HER2. Clin Cancer Res. 2004;10:5650–5655. doi: 10.1158/1078-0432.CCR-04-0225. [DOI] [PubMed] [Google Scholar]
  56. Ghossein RA, Bhattacharya S. Molecular detection and characterization of circulating tumor cells and micrometastases in prostatic, urothelial, and renal cell carcinomas. [Review] [51 refs] Seminars in Surgical Oncology. 2001;20:304–311. doi: 10.1002/ssu.1048. [DOI] [PubMed] [Google Scholar]
  57. Gomez HL, Chavez MA, Doval DC, Nag S, Chow LW, Ang PC, Ahmad NM, Berger M, Newstat B, Stein S, Sledge GW. Biomarker results from a phase II randomized study of lapatinib ( GW572016) as first-line treatment for patients with ErbB2 FISH-amplified advanced or metastatic breast cancer. Breast Cancer Res Treat. 2005;94 suppl 1:S63. (abstract 1071) [Google Scholar]
  58. Harwerth IM, Wels W, Marte BM, Hynes NE. Monoclonal antibodies against the extracellular domain of the erbB-2 receptor function as partial ligand agonists. J Biol Chem. 1992;267:15160–15167. [PubMed] [Google Scholar]
  59. Harwerth IM, Wels W, Schlegel J, Muller M, Hynes NE. Monoclonal antibodies directed to the erbB-2 receptor inhibit in vivo tumour cell growth. Br J Cancer. 1993;68:1140–1145. doi: 10.1038/bjc.1993.494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Hudziak RM, Lewis GD, Winget M, Fendly BM, Shepard HM, Ullrich A. p185HER2 monoclonal antibody has antiproliferative effects in vitro and sensitizes human breast tumor cells to tumor necrosis factor. Mol Cell Biol. 1989;9:1165–1172. doi: 10.1128/mcb.9.3.1165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Hurwitz E, Stancovski I, Sela M, Yarden Y. Suppression and promotion of tumor growth by monoclonal antibodies to ErbB-2 differentially correlate with cellular uptake. Proc Natl Acad Sci U S A. 1995;92:3353–3357. doi: 10.1073/pnas.92.8.3353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Hynes NE, Lane HA. Myc and mammary cancer: Myc is a downstream effector of the ErbB2 receptor tyrosine kinase. J Mammary Gland Biol Neoplasia. 2001;6:141–150. doi: 10.1023/a:1009528918064. [DOI] [PubMed] [Google Scholar]
  63. Iwata H, Toi M, Fujiwara Y, Ito Y, Fujii H, Nakamura S, Aogi K, Zaks T, Sasaki Y, Takashima S. Phase II clinical study of lapatinib ( GW572016) in patients with advanced or metastatic breast cancer. San Antonio Br Can Symp. 2006 #1091. [Google Scholar]
  64. Izumi Y, Xu L, di TE, Fukumura D, Jain RK. Tumour biology: herceptin acts as an anti-angiogenic cocktail. Nature. 2002;416:279–280. doi: 10.1038/416279b. [DOI] [PubMed] [Google Scholar]
  65. Jani JP, Barbacci EG, Bhattacharya S, Boos C, Campbell M, Clark T, Coleman K, Connell R, Cosker T. Discovery and development of CP-724714, a selective HER2 receptor tyrosine kinase inhibitor. Proc Amer Soc Clin Onc. 2004a;22 #3122. [Google Scholar]
  66. Jani JP, Barbacci EG, Bhattacharya S, Moyer JD. CP-724714, a novel erbB2 receptor tyrosine kinase inhibitor for cancer therapy. Proc Am Assoc Can Res. 2004b;45 #4637. [Google Scholar]
  67. Juhl H, Downing SG, Wellstein A, Czubayko F. HER-2/neu is rate-limiting for ovarian cancer growth. Conditional depletion of HER-2/neu by ribozyme targeting. J Biol Chem. 1997;272:29482–29486. doi: 10.1074/jbc.272.47.29482. [DOI] [PubMed] [Google Scholar]
  68. Kiewe P, Hasmuller S, Kahlert S, Heinrigs M, Rack B, Marme A, et al. Phase I trial of the trifunctional anti-HER2 x anti-CD3 antibody ertumaxomab in metastatic breast cancer. Clin Cancer Res. 2006;12:3085–3091. doi: 10.1158/1078-0432.CCR-05-2436. [DOI] [PubMed] [Google Scholar]
  69. King CR, Kraus MH, Aaronson SA. Amplification of a novel v-erbB-related gene in a human mammary carcinoma. Science. 1985;229:974–976. doi: 10.1126/science.2992089. [DOI] [PubMed] [Google Scholar]
  70. Kita Y, Tseng J, Horan T, Wen J, Philo J, Chang D, et al. ErbB receptor activation, cell morphology changes, and apoptosis induced by anti-Her2 monoclonal antibodies. Biochem Biophys Res Commun. 1996;226:59–69. doi: 10.1006/bbrc.1996.1311. [DOI] [PubMed] [Google Scholar]
  71. Klapper LN, Vaisman N, Hurwitz E, Pinkas-Kramarski R, Yarden Y, Sela M. A subclass of tumor-inhibitory monoclonal antibodies to ErbB-2/HER2 blocks crosstalk with growth factor receptors. Oncogene. 1997;14:2099–2109. doi: 10.1038/sj.onc.1201029. [DOI] [PubMed] [Google Scholar]
  72. 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;66:1630–1639. doi: 10.1158/0008-5472.CAN-05-1182. [DOI] [PubMed] [Google Scholar]
  73. Kubo M, Morisaki T, Kuroki H, Tasaki A, Yamanaka N, Matsumoto K, et al. Combination of adoptive immunotherapy with Herceptin for patients with HER2-expressing breast cancer. Anticancer Res. 2003;23:4443–4449. [PubMed] [Google Scholar]
  74. Kumar PS, Pegram M. Targeting HER2 Epitopes. Semin Oncol. 2006;33:386–391. doi: 10.1053/j.seminoncol.2006.04.004. [DOI] [PubMed] [Google Scholar]
  75. Kurokawa H, Lenferink AE, Simpson JF, Pisacane PI, Sliwkowski MX, Forbes JT, et al. Inhibition of HER2/neu (erbB-2) and mitogen-activated protein kinases enhances tamoxifen action against HER2-overexpressing, tamoxifen-resistant breast cancer cells. Cancer Res. 2000;60:5887–5894. [PubMed] [Google Scholar]
  76. Kwak EL, Sordella R, Bell DW, Godin-Heymann N, Okimoto RA, Brannigan BW, et al. Irreversible inhibitors of the EGF receptor may circumvent acquired resistance to gefitinib. Proc Natl Acad Sci USA. 2005;102:7665–7670. doi: 10.1073/pnas.0502860102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Lane HA, Beuvink I, Motoyama AB, Daly JM, Neve RM, Hynes NE. ErbB2 potentiates breast tumor proliferation through modulation of p27(Kip1)-Cdk2 complex formation: receptor overexpression does not determine growth dependency. Mol Cell Biol. 2000;20:3210–3223. doi: 10.1128/mcb.20.9.3210-3223.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Le XF, Pruefer F, Bast RC., Jr HER2-targeting antibodies modulate the cyclin-dependent kinase inhibitor p27Kip1 via multiple signaling pathways. Cell Cycle. 2005;4:87–95. doi: 10.4161/cc.4.1.1360. [DOI] [PubMed] [Google Scholar]
  79. Lee S, Yang W, Lan KH, Sellappan S, Klos K, Hortobagyi G, et al. Enhanced sensitization to taxol-induced apoptosis by herceptin pretreatment in ErbB2-overexpressing breast cancer cells. Cancer Res. 2002;62:5703–5710. [PubMed] [Google Scholar]
  80. Lenferink AE, Busse D, Flanagan WM, Yakes FM, Arteaga CL. ErbB2/neu kinase modulates cellular p27(Kip1) and cyclin D1 through multiple signaling pathways. Cancer Res. 2001;61:6583–6591. [PubMed] [Google Scholar]
  81. Lenferink AE, Simpson JF, Shawver LK, Coffey RJ, Forbes JT, Arteaga CL. 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;97:9609–9614. doi: 10.1073/pnas.160564197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Levitzki A, Gazit A. Tyrosine kinase inhibition: an approach to drug development. Science. 1995;267:1782–1788. doi: 10.1126/science.7892601. [DOI] [PubMed] [Google Scholar]
  83. Lewis GD, Figari I, Fendly B, Wong WL, Carter P, Gorman C, et al. Differential responses of human tumor cell lines to anti-p185HER2 monoclonal antibodies. Cancer Immunol Immunother. 1993;37:255–263. doi: 10.1007/BF01518520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Liu PC, Liu X, Li Y, Covington M, Wynn R, Huber R, et al. Identification of ADAM10 as a major source of HER2 ectodomain sheddase activity in HER2 overexpressing breast cancer cells. Cancer Biol Ther. 2006;5:657–664. doi: 10.4161/cbt.5.6.2708. [DOI] [PubMed] [Google Scholar]
  85. Longva KE, Pedersen NM, Haslekas C, Stang E, Madshus IH. Herceptin-induced inhibition of ErbB2 signaling involves reduced phosphorylation of Akt but not endocytic down-regulation of ErbB2. Int J Cancer. 2005;116:359–367. doi: 10.1002/ijc.21015. [DOI] [PubMed] [Google Scholar]
  86. Lydon NB, Mett H, Mueller M, Becker M, Cozens RM, Stover D, et al. A potent protein-tyrosine kinase inhibitor which selectively blocks proliferation of epidermal growth factor receptor-expressing tumor cells in vitro and in vivo. Int J Cancer. 1998;76:154–163. doi: 10.1002/(sici)1097-0215(19980330)76:1<154::aid-ijc24>3.0.co;2-b. [DOI] [PubMed] [Google Scholar]
  87. Marches R, Uhr JW. Enhancement of the p27Kip1-mediated antiproliferative effect of trastuzumab (Herceptin) on HER2-overexpressing tumor cells. Int J Cancer. 2004;112:492–501. doi: 10.1002/ijc.20378. [DOI] [PubMed] [Google Scholar]
  88. Marty M, Cognetti F, Maraninchi D, Snyder R, Mauriac L, Tubiana-Hulin M, et al. Randomized phase II trial of the efficacy and safety of trastuzumab combined with docetaxel in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer administered as first-line treatment: the M77001 study group. J Clin Onc. 2005;23:4265–4274. doi: 10.1200/JCO.2005.04.173. [DOI] [PubMed] [Google Scholar]
  89. Mass RD, Press MF, Anderson S, Cobleigh MA, Vogel CL, Dybdal N, 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;6:240–246. doi: 10.3816/CBC.2005.n.026. [DOI] [PubMed] [Google Scholar]
  90. McKenzie SJ, Marks PJ, Lam T, Morgan J, Panicali DL, Trimpe KL, et al. Generation and characterization of monoclonal antibodies specific for the human neu oncogene product, p185. Oncogene. 1989;4:543–548. [PubMed] [Google Scholar]
  91. McKillop D, Partridge EA, Kemp JV, Spence MP, Kendrew J, Barnett S, et al. Tumor penetration of gefitinib (Iressa), an epidermal growth factor receptor tyrosine kinase inhibitor. Mol Cancer Ther. 2005;4:641–649. doi: 10.1158/1535-7163.MCT-04-0329. [DOI] [PubMed] [Google Scholar]
  92. Menard S, Pupa SM, Campiglio M, Tagliabue E. Biologic and therapeutic role of HER2 in cancer. Oncogene. 2003;22:6570–6578. doi: 10.1038/sj.onc.1206779. [DOI] [PubMed] [Google Scholar]
  93. Messerle K, Schlegel J, Hynes NE, Groner B. NIH/3T3 cells transformed with the activated erbB-2 oncogene can be phenotypically reverted by a kinase deficient, dominant negative erbB-2 variant. Mol Cell Endocrinol. 1994;105:1–10. doi: 10.1016/0303-7207(94)90029-9. [DOI] [PubMed] [Google Scholar]
  94. Miknis G, Wallace E, Lyssikatos J, Lee P, Zhao Q, Hans J, et al. ARRY-334543, A potent, orally active small molecule inhibitor of EGFR and ErbB-2. Proc Am Assoc Can Res. 2005;24 #3399. [Google Scholar]
  95. Moasser MM. The oncogene HER2; Its signaling and transforming functions and its role in human cancer pathogenesis. Oncogene. 2007;26:6469–6487. doi: 10.1038/sj.onc.1210477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. 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 Research. 2001;61:7184–7188. [PubMed] [Google Scholar]
  97. Mohsin SK, Weiss HL, Gutierrez MC, Chamness GC, Schiff R, DiGiovanna MP, et al. Neoadjuvant trastuzumab induces apoptosis in primary breast cancers. J Clin Oncol. 2005;23:2460–2468. doi: 10.1200/JCO.2005.00.661. [DOI] [PubMed] [Google Scholar]
  98. Molina MA, Codony-Servat J, Albanell J, Rojo F, Arribas J, Baselga J. Trastuzumab (herceptin), a humanized anti-Her2 receptor monoclonal antibody, inhibits basal and activated Her2 ectodomain cleavage in breast cancer cells. Cancer Res. 2001;61:4744–4749. [PubMed] [Google Scholar]
  99. Molina MA, Saez R, Ramsey EE, Garcia-Barchino MJ, Rojo F, Evans AJ, et al. NH(2)-terminal truncated HER-2 protein but not full-length receptor is associated with nodal metastasis in human breast cancer. Clin Cancer Res. 2002;8:347–353. [PubMed] [Google Scholar]
  100. Moody SE, Perez D, Pan TC, Sarkisian CJ, Portocarrero CP, Sterner CJ, et al. The transcriptional repressor Snail promotes mammary tumor recurrence. Cancer Cell. 2005;8:197–209. doi: 10.1016/j.ccr.2005.07.009. [DOI] [PubMed] [Google Scholar]
  101. Moody SE, Sarkisian CJ, Hahn KT, Gunther EJ, Pickup S, Dugan KD, et al. Conditional activation of Neu in the mammary epithelium of transgenic mice results in reversible pulmonary metastasis. Cancer Cell. 2002;2:451–461. doi: 10.1016/s1535-6108(02)00212-x. [DOI] [PubMed] [Google Scholar]
  102. 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 Research. 2001;61:8887–8895. [PubMed] [Google Scholar]
  103. Moyer JD, Barbacci EG, Iwata KK, Arnold L, Boman B, Cunningham A, et al. Induction of apoptosis and cell cycle arrest by CP-358,774, an inhibitor of epidermal growth factor receptor tyrosine kinase. Cancer Res. 1997;57:4838–4848. [PubMed] [Google Scholar]
  104. Nagata Y, Lan KH, Zhou X, Tan M, Esteva FJ, Sahin AA, et al. PTEN activation contributes to tumor inhibition by trastuzumab, and loss of PTEN predicts trastuzumab resistance in patients. Cancer Cell. 2004;6:117–127. doi: 10.1016/j.ccr.2004.06.022. [DOI] [PubMed] [Google Scholar]
  105. Naito K, Matsutani E, Tamura T, Miwa K, Takakura N, Asada M, Tasaka A, Miyake A, Terashita Z. TAK-165, a selective inhibitor of HER2 tyrosine kinase: 1. Nature of tyrosine kinase inhibition and selective antitumor activity in vivo and in vitro. Proc Am Assoc Can Res. 2002;43 # 3897. [Google Scholar]
  106. Neve RM, Nielsen UB, Kirpotin DB, Poul MA, Marks JD, Benz CC. Biological effects of anti-ErbB2 single chain antibodies selected for internalizing function. Biochem Biophys Res Commun. 2001;280:274–279. doi: 10.1006/bbrc.2000.4104. [DOI] [PubMed] [Google Scholar]
  107. Normanno N, Campiglio M, De LA, Somenzi G, Maiello M, Ciardiello F, et al. Cooperative inhibitory effect of ZD1839 (Iressa) in combination with trastuzumab (Herceptin) on human breast cancer cell growth. [comment] Annals of Oncology. 2002;13:65–72. doi: 10.1093/annonc/mdf020. [DOI] [PubMed] [Google Scholar]
  108. Parsa AT, Waldron JS, Panner A, Crane CA, Parney IF, Barry JJ, et al. Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat Med. 2006 doi: 10.1038/nm1517. [DOI] [PubMed] [Google Scholar]
  109. Pheneger T, Woessner R, Lyssikatos J, Miknis G, Anderson D, Winski S, Lee PA. In vivo anti-tumor activity of arry-334543, a small molecule inhibitor of EGFR and ErbB-2; Proc AACR-NCI-EORTC Conference; 2005. # A247. [Google Scholar]
  110. 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;353:1659–1672. doi: 10.1056/NEJMoa052306. [DOI] [PubMed] [Google Scholar]
  111. Qian X, Dougall WC, Hellman ME, Greene MI. Kinase-deficient neu proteins suppress epidermal growth factor receptor function and abolish cell transformation. Oncogene. 1994;9:1507–1514. [PubMed] [Google Scholar]
  112. Rabindran SK. Antitumor activity of HER-2 inhibitors. Cancer Lett. 2005;227:9–23. doi: 10.1016/j.canlet.2004.11.015. [DOI] [PubMed] [Google Scholar]
  113. Rabindran SK, Discafani CM, Rosfjord EC, Baxter M, Floyd MB, Golas J, et al. 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]
  114. Repka T, Chiorean EG, Gay J, Herwig KE, Kohl VK, Yee D, et al. Trastuzumab and interleukin-2 in HER2-positive metastatic breast cancer: a pilot study. Clin Cancer Res. 2003;9:2440–2446. [PubMed] [Google Scholar]
  115. Rewcastle GW, Denny WA, Bridges AJ, Zhou H, Cody DR, McMichael A, et al. Tyrosine kinase inhibitors. 5. Synthesis and structure-activity relationships for 4-[(phenylmethyl)amino]- and 4-(phenylamino)quinazolines as potent adenosine 5'-triphosphate binding site inhibitors of the tyrosine kinase domain of the epidermal growth factor receptor. J Med Chem. 1995;38:3482–3487. doi: 10.1021/jm00018a008. [DOI] [PubMed] [Google Scholar]
  116. Robertson JFR, Gutteridge E, Cheung KL, Owers R, Koehler M, Hamilton L, Gee J, Nicholson RI. Gefitinib (ZD1839) is active in acquired tamoxifen (TAM)-resistant oestrogen receptor (ER)-positive and ER-negative breast cancer: Results from a phase II study. Proc Amer Soc Clin Onc. 2003;22 #23. [Google Scholar]
  117. Robinson MO, Lin J. AV-412, a potent EGFRsHER2 TK inhibitor causes tumor regression in novel genetically engineered EGFRL858R and EGFRL858R&T790M lung tumor models. European Journal of Cancer Supplements. 2006;4:169. [Google Scholar]
  118. Roh H, Pippin J, Drebin JA. Down-regulation of HER2/neu expression induces apoptosis in human cancer cells that overexpress HER2/neu. Cancer Res. 2000;60:560–565. [PubMed] [Google Scholar]
  119. Romond EH, Perez EA, Bryant J, Suman VJ, Geyer CE, Jr, Davidson NE, et al. Trastuzumab plus Adjuvant Chemotherapy for Operable HER2-Positive Breast Cancer. N Engl J Med. 2005;353:1673–1684. doi: 10.1056/NEJMoa052122. [DOI] [PubMed] [Google Scholar]
  120. Rusnak DW, Lackey K, KAffleck K, Wood ER, Alligood KJ, Rhodes N, et al. 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. Molecular Cancer Therapeutics. 2001;1:85–94. [PubMed] [Google Scholar]
  121. Saal LH, Holm K, Maurer M, Memeo L, Su T, Wang X, et al. 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]
  122. Sarup JC, Johnson RM, King KL, Fendly BM, Lipari MT, Napier MA, et al. Characterization of an anti-p185HER2 monoclonal antibody that stimulates receptor function and inhibits tumor cell growth. Growth Regul. 1991;1:72–82. [PubMed] [Google Scholar]
  123. Schiffer IB, Gebhard S, Heimerdinger CK, Heling A, Hast J, Wollscheid U, et al. Switching off HER-2/neu in a tetracycline-controlled mouse tumor model leads to apoptosis and tumor-size-dependent remission. Cancer Res. 2003;63:7221–7231. [PubMed] [Google Scholar]
  124. Segatto O, King CR, Pierce JH, Di Fiore PP, Aaronson SA. Different structural alterations upregulate in vitro tyrosine kinase activity and transforming potency of the erbB-2 gene. Mol Cell Biol. 1988;8:5570–5574. doi: 10.1128/mcb.8.12.5570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Sergina NV, Rausch M, Wang D, Blair J, Hann B, Shokat KM, et al. 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]
  126. She Q, Solit D, Basso A, Moasser MM. Resistance to gefitinib (ZD1839, Iressa) in PTEN null HER overexpressing tumor cells can be overcome through restoration of PTEN function or pharmacologic modulation of constitutive PI3K/Akt pathway signaling. Clinical Cancer Research. 2003;9:4340–4346. [PubMed] [Google Scholar]
  127. Shepard HM, Lewis GD, Sarup JC, Fendly BM, Maneval D, Mordenti J, et al. Monoclonal antibody therapy of human cancer: taking the HER2 protooncogene to the clinic. J Clin Immunol. 1991;11:117–127. doi: 10.1007/BF00918679. [DOI] [PubMed] [Google Scholar]
  128. Shepherd TG, Kockeritz L, Szrajber MR, Muller WJ, Hassell JA. The pea3 subfamily ets genes are required for HER2/Neu-mediated mammary oncogenesis. Curr Biol. 2001;11:1739–1748. doi: 10.1016/s0960-9822(01)00536-x. [DOI] [PubMed] [Google Scholar]
  129. Shih C, Padhy LC, Murray M, Weinberg RA. Transforming genes of carcinomas and neuroblastomas introduced into mouse fibroblasts. Nature. 1981;290:261–264. doi: 10.1038/290261a0. [DOI] [PubMed] [Google Scholar]
  130. 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]
  131. Slamon DJ, Godolphin W, Jones LA, Holt JA, Wong SG, Keith DE, et al. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science. 1989;244:707–712. doi: 10.1126/science.2470152. [DOI] [PubMed] [Google Scholar]
  132. Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. [see comment] N Engl J Med. 2001;344:783–792. doi: 10.1056/NEJM200103153441101. [DOI] [PubMed] [Google Scholar]
  133. Slichenmyer WJ, Elliott WL, Fry DW. CI-1033, a pan-erbB tyrosine kinase inhibitor. [Review] [19 refs] Seminars in Oncology. 2001;28:80–85. doi: 10.1016/s0093-7754(01)90285-4. [DOI] [PubMed] [Google Scholar]
  134. Sliwkowski MX. Ready to partner. Nat Struct Biol. 2003;10:158–159. doi: 10.1038/nsb0303-158. [DOI] [PubMed] [Google Scholar]
  135. Solca F, Baum A, Guth B, Colbatzky F, Blech S, Amelsberg A, Himmelsbach F. BIBW 2992, an irreversible dual EGFR/HER2 receptor tyrosine kinase inhibitor for cancer therapy; Proc AACR-NCI-EORTC Conference; 2006. # A244. [Google Scholar]
  136. Spector NL, Xia W, Burris H, III, Hurwitz H, Dees EC, Dowlati A, et al. Study of the biologic effects of lapatinib, a reversible inhibitor of ErbB1 and ErbB2 tyrosine kinases, on tumor growth and survival pathways in patients with advanced malignancies. J Clin Oncol. 2005;23:2502–2512. doi: 10.1200/JCO.2005.12.157. [DOI] [PubMed] [Google Scholar]
  137. Srinivas U, Tagliabue E, Campiglio M, Menard S, Colnaghi MI. Antibody-induced activation of p185HER2 in the human lung adenocarcinoma cell line Calu-3 requires bivalency. Cancer Immunol Immunother. 1993;36:397–402. doi: 10.1007/BF01742256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. 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]
  139. Stancovski I, Hurwitz E, Leitner O, Ullrich A, Yarden Y, Sela M. Mechanistic aspects of the opposing effects of monoclonal antibodies to the ERBB2 receptor on tumor growth. Proc Natl Acad Sci U S A. 1991;88:8691–8695. doi: 10.1073/pnas.88.19.8691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Suzuki E, Niwa R, Saji S, Muta M, Hirose M, Iida S, et al. A Nonfucosylated Anti-HER2 Antibody Augments Antibody-Dependent Cellular Cytotoxicity in Breast Cancer Patients. Clinical Cancer Research. 2007;13:1875–1882. doi: 10.1158/1078-0432.CCR-06-1335. [DOI] [PubMed] [Google Scholar]
  141. Tagliabue E, Centis F, Campiglio M, Mastroianni A, Martignone S, Pellegrini R, et al. Selection of monoclonal antibodies which induce internalization and phosphorylation of p185HER2 and growth inhibition of cells with HER2/NEU gene amplification. Int J Cancer. 1991;47:933–937. doi: 10.1002/ijc.2910470625. [DOI] [PubMed] [Google Scholar]
  142. Takai N, Jain A, Kawamata N, Popoviciu LM, Said JW, Whittaker S, et al. 2C4, a monoclonal antibody against HER2, disrupts the HER kinase signaling pathway and inhibits ovarian carcinoma cell growth. Cancer. 2005;104:2701–2708. doi: 10.1002/cncr.21533. [DOI] [PubMed] [Google Scholar]
  143. Tan AR, Yang X, Hewitt SM, Berman A, Lepper ER, Sparreboom A, et al. Evaluation of biologic end points and pharmacokinetics in patients with metastatic breast cancer after treatment with erlotinib, an epidermal growth factor receptor tyrosine kinase inhibitor. J Clin Oncol. 2004;22:3080–3090. doi: 10.1200/JCO.2004.08.189. [DOI] [PubMed] [Google Scholar]
  144. Tokuda Y, Ohnishi Y, Shimamura K, Iwasawa M, Yoshimura M, Ueyama Y, et al. In vitro and in vivo anti-tumour effects of a humanised monoclonal antibody against c-erbB-2 product. Br J Cancer. 1996;73:1362–1365. doi: 10.1038/bjc.1996.259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Torrance CJ, Jackson PE, Montgomery E, Kinzler KW, Vogelstein B, Wissner A, et al. Combinatorial chemoprevention of intestinal neoplasia. Nat Med. 2000;6:1024–1028. doi: 10.1038/79534. [DOI] [PubMed] [Google Scholar]
  146. Traxler P, Allegrini PR, Brandt R, Brueggen J, Cozens R, Fabbro D, et al. AEE788: a dual family epidermal growth factor receptor/ErbB2 and vascular endothelial growth factor receptor tyrosine kinase inhibitor with antitumor and antiangiogenic activity. Cancer Res. 2004;64:4931–4941. doi: 10.1158/0008-5472.CAN-03-3681. [DOI] [PubMed] [Google Scholar]
  147. Traxler P, Bold G, Buchdunger E, Caravatti G, Furet P, Manley P, et al. Tyrosine kinase inhibitors: from rational design to clinical trials. Med Res Rev. 2001;21:499–512. doi: 10.1002/med.1022. [DOI] [PubMed] [Google Scholar]
  148. Tsou HR, Overbeek-Klumpers EG, Hallett WA, Reich MF, Floyd MB, Johnson BD, et al. 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]
  149. van LF, van dV, Lomans J, van DL, Jenster G, Akiyama T, et al. Mutation of the human neu protein facilitates down-modulation by monoclonal antibodies. Oncogene. 1990;5:497–503. [PubMed] [Google Scholar]
  150. 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 Onc. 2002;20:719–726. doi: 10.1200/JCO.2002.20.3.719. [DOI] [PubMed] [Google Scholar]
  151. Wakeling AE, Guy SP, Woodburn JR, Ashton SE, Curry BJ, Barker AJ, et al. ZD1839 (Iressa): an orally active inhibitor of epidermal growth factor signaling with potential for cancer therapy. Cancer Research. 2002;62:5749–5754. [PubMed] [Google Scholar]
  152. Warburton C, Dragowska WH, Gelmon K, Chia S, Yan H, Masin D, et al. Treatment of HER-2/neu overexpressing breast cancer xenograft models with trastuzumab (Herceptin) and gefitinib (ZD1839): drug combination effects on tumor growth, HER-2/neu and epidermal growth factor receptor expression, and viable hypoxic cell fraction. Clin Cancer Res. 2004;10:2512–2524. doi: 10.1158/1078-0432.ccr-03-0244. [DOI] [PubMed] [Google Scholar]
  153. Ward WH, Cook PN, Slater AM, Davies DH, Holdgate GA, Green LR. Epidermal growth factor receptor tyrosine kinase. Investigation of catalytic mechanism, structure-based searching and discovery of a potent inhibitor. Biochem Pharmacol. 1994;48:659–666. doi: 10.1016/0006-2952(94)90042-6. [DOI] [PubMed] [Google Scholar]
  154. Wehrman TS, Raab WJ, Casipit CL, Doyonnas R, Pomerantz JH, Blau HM. A system for quantifying dynamic protein interactions defines a role for Herceptin in modulating ErbB2 interactions. Proc Natl Acad Sci U S A. 2006;103:19063–19068. doi: 10.1073/pnas.0605218103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Weiner DB, Kokai Y, Wada T, Cohen JA, Williams WV, Greene MI. Linkage of tyrosine kinase activity with transforming ability of the p185neu oncoprotein. Oncogene. 1989;4:1175–1183. [PubMed] [Google Scholar]
  156. Winer EP, Cobleigh M, Dickler M, Miller K, Fehrehbacher L, Jones CM, Anderson S, Eberhard D, Jones C. Phase II multicenter study to evaluate the efficacy and safety of Tarceva (Erlotinib HCl, OSI-774) in women with previously treated, locally advanced or metastatic breast cancer. San Antonio Br Can Symp. 2002:445. [Google Scholar]
  157. Wissner A, Overbeek E, Reich MF, Floyd MB, Johnson BD, Mamuya N, et al. 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]
  158. Wong TW, Lee FY, Yu C, Luo FR, Oppenheimer S, Zhang H, et al. Preclinical antitumor activity of BMS-599626, a pan-HER kinase inhibitor that inhibits HER1/HER2 homodimer and heterodimer signaling. Clin Cancer Res. 2006;12:6186–6193. doi: 10.1158/1078-0432.CCR-06-0642. [DOI] [PubMed] [Google Scholar]
  159. Wood ER, Truesdale AT, McDonald OB, Yuan D, Hassell A, Dickerson SH, et al. 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]
  160. Xie W, Chow LT, Paterson AJ, Chin E, Kudlow JE. Conditional expression of the ErbB2 oncogene elicits reversible hyperplasia in stratified epithelia and up-regulation of TGFalpha expression in transgenic mice. Oncogene. 1999;18:3593–3607. doi: 10.1038/sj.onc.1202673. [DOI] [PubMed] [Google Scholar]
  161. Xu F, Lupu R, Rodriguez GC, Whitaker RS, Boente MP, Berchuck A, et al. Antibody-induced growth inhibition is mediated through immunochemically and functionally distinct epitopes on the extracellular domain of the c-erbB-2 (HER-2/neu) gene product p185. Int J Cancer. 1993;53:401–408. doi: 10.1002/ijc.2910530310. [DOI] [PubMed] [Google Scholar]
  162. 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;62:4132–4141. [PubMed] [Google Scholar]
  163. Yarden Y. Agonistic antibodies stimulate the kinase encoded by the neu protooncogene in living cells but the oncogenic mutant is constitutively active. Proc Natl Acad Sci U S A. 1990;87:2569–2573. doi: 10.1073/pnas.87.7.2569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Yokoyama H, Ikehara Y, Kodera Y, Ikehara S, Yatabe Y, Mochizuki Y, et al. Molecular basis for sensitivity and acquired resistance to gefitinib in HER2-overexpressing human gastric cancer cell lines derived from liver metastasis. Br J Cancer. 2006;95:1504–1513. doi: 10.1038/sj.bjc.6603459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Yoshida S, Naito K, Hori A, Teratani M, Koyama M, Tasaka A, Terashita Z. Tak-165, a selective inhibitor of HER2 tyrosine kinase: 2. Mechanism of antitumor activity on HER2 signal transduction pathway. Proc Am Assoc Can Res. 2002;43 # 3898. [Google Scholar]

RESOURCES