Acta Biochim Biophys Sin 2014, 46: 190 – 198 | ª The Author 2014. Published by ABBS Editorial Office in association with Oxford University Press on behalf of the Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences. DOI: 10.1093/abbs/gmt150. Advance Access Publication 20 January 2014

Review

Role of erbB3 receptors in cancer therapeutic resistance Youngseok Lee1,2†, Jian Ma3†, Hui Lyu1, Jingcao Huang1, Aeree Kim2, and Bolin Liu1 * 1

Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA Department of Pathology, Korea University Guro Hospital, Seoul 152-703, Korea 3 International Medical Centre of PLA General Hospital, Beijing 100853, China † These authors contributed equally to this work. *Correspondence address. Tel: þ1-303-724-3749; Fax: þ1-303-724-3712; E-mail: [email protected] 2

Keywords erbB3; receptor signaling; resistance; cancer Received: October 28, 2013

tyrosine

kinase;

cell

Accepted: December 10, 2013

Introduction The erbB receptor tyrosine kinase (RTK) family, including the epidermal growth factor receptor (EGFR), erbB2 (HER2/ Acta Biochim Biophys Sin (2014) | Volume 46 | Issue 3 | Page 190

neu), erbB3 (HER3), and erbB4 (HER4), is arguably the most important receptor family in the context of development and tumorigenesis [1,2]. These receptors are widely expressed in epithelial, mesenchymal, and neuronal cells [3]. Abnormal expression of erbB receptors is involved in carcinogenesis of diverse types of human cancers [4,5]. The shared molecular structure of erbB receptors consists of an extracellular ligandbinding domain, a transmembrane domain, and an intracellular domain containing a region with tyrosine kinase activity and a non-catalytic region of C-terminal tail. Unlike other family members, erbB2 has no known ligands. When a ligand binds to EGFR, erbB3, or erbB4, a dimerization arm in their extracellular domains is exposed and receptor–receptor interactions are promoted [6]. Dimerization of erbB receptors is an essential step for the activation of the cytoplasmic signaling pathways [5,7]. ErbB receptors normally exist as inactive monomers with molecular folding to prevent dimerization, with the exception of erbB2 [8,9]. ErbB2 is in a constitutively active conformation with an exposed dimerization arm even in the absence of a ligand [8], thus making it the most preferred dimerization partner for other erbB receptors [10]. As a consequence of erbB receptor dimerization, the intracellular tyrosine kinases are activated and the tyrosine residues on C-terminal tails are phosphorylated, leading to the recruitment of adaptor proteins, and activation of the downstream signaling, such as phosphoinositide-3-kinase (PI-3K)/protein kinase B (Akt) pathway, mitogen-activated protein kinase kinase (MEK)/mitogen-activated protein kinase (MAPK) pathway, and Janus kinase (Jak)/signal transducer and activator of transcription (Stat) pathways, Src kinase, etc., imparting various biological responses, including cellular proliferation, maturation, survival, apoptosis, and angiogenesis [5,11]. ErbB3 receptor has long been considered an inactive ‘pseudokinase’ [12,13], although a recent study suggested that it possesses weak kinase activity that can trans-autophosphorylate its intracellular region [14]. To fully transduce cell signaling, however, erbB3 has to be phosphorylated by its interactive partners, of those, erbB2 is the most important one [15]. Like

Downloaded from http://abbs.oxfordjournals.org/ at Kainan University on March 12, 2015

ErbB3 receptors are unique members of the erbB receptor tyrosine kinases (RTKs), which are often aberrantly expressed and/or activated in human cancers. Unlike other members in the family, erbB3 lacks or has impaired kinase activity. To transduce cell signaling, erbB3 has to interact with other RTKs and to be phosphorylated by its interactive partners, of those, erbB2 is the most important one. ErbB3 is frequently co-expressed with other RTKs in cancer cells to activate oncogenic signaling, such as phosphoinositide-3-kinase/protein kinase B (Akt) pathway, mitogen-activated protein kinase kinase (MEK)/mitogenactivated protein kinase (MAPK) pathway, Janus kinase (Jak)/signal transducer and activator of transcription (Stat) pathway, etc. and thereby promote tumorigenesis. Numerous studies have demonstrated that activation of erbB3 signaling plays an important role in the progression of a variety of tumor types, such as erbB2-overexpressing breast cancer, castration-resistant prostate cancer, platinum refractory/resistant ovarian cancer, epidermal growth factor receptor TKI-resistant non-small-cell lung cancer, and others. Basic research on the underlying mechanisms implicated the functions of erbB3 as a major cause of treatment failure in cancer therapy. Thus, concomitant inhibition of erbB3 is thought to be required to overcome the resistance and to effectively treat human cancers. This review focuses on the latest advances in our understanding of erbB3-initiated signaling in the development of resistance to cancer treatments.

ErbB3 in treatment resistance of cancer

The Unique Biology of erbB3 Receptors in Human Cancer Development ErbB3 has a unique characteristic among the erbB family members. It cannot form homodimers and lacks or possesses much lower intracellular tyrosine kinase activity [14,27]. A sequence comparison of the protein kinases reveals that certain residues, such as Cys-721, His-740, and Asn-815, have nonconservative substitutions in erbB3. These changes diminish the catalytic activity of erbB3 in its tyrosine kinase domain [28]. However, once the ligand, heregulin (HRG) or neuregulin (NRG), binds to erbB3, it recruits another receptor containing tyrosine kinase activity to form a heterodimer, leading to the activation of erbB3. Thus, to fully transduce cell signaling, erbB3 has to be phosphorylated by its interactive partners, of those, erbB2 is the most important one [15]. ErbB3 is a more potent partner than other family members for the oncogenic activity of erbB2 [18,29–31]. It is thought that erbB3 functions primarily to drive erbB2-mediated cell signaling [18,32]. We have shown that erbB3 expression is required to maintain the tyrosine kinase activity of erbB2 in both mouse and human breast cancer cells [33,34], further emphasizing the pivotal role of an erbB3 receptor in erbB2 activation. Elevated expression of erbB3 is frequently seen in various malignancies, such as cancers of breast, gastric, ovarian, prostate, and bladder, colorectal carcinoma, squamous cell carcinoma of the head and neck, melanoma [18,35,36]. It is worth noting that whereas both amplification and overexpression of erbB3 were observed in some cancers, erbB3 mutations were rarely found in human tumors until recently. Jaiswal et al. [37] were the first to report the identification of

erbB3 somatic mutations in 11% of colon and gastric cancers. Detailed studies demonstrated that the oncogenic activity of mutant erbB3 depends upon the kinase-active erbB2 and the erbB3 mutants transform colonic and breast epithelial cells in a ligand-independent manner [37]. In breast cancer, both mRNA expression and protein levels of erbB3 are up-regulated. Most metastatic breast cancers show expression for either EGFR or erbB2, and less often for both [38]. In contrast, co-expression of erbB2 and erbB3 is common in breast cancer [39] and breast cancer cell lines [40]. Overexpression of endogenous erbB3 and its association with the transgene encoded erbB2 promote mammary tumorigenesis in erbB2/neu-transgenic mice [41,42]. Studies indicated that erbB3 serves as a critical co-receptor of erbB2, and its expression is a rate-limiting factor for erbB2-induced breast cancer cell survival and proliferation [17,18]. In erbB2þ breast cancer tissues, preferential phosphorylation of erbB3, but not EGFR, has been observed [18]. It appears that erbB3 is a preferred dimerization partner for EGFR in melanoma and pancreatic cancer [43,44]. Elevated expression of erbB3 has been reported in 50%– 70% of human breast cancers [45–47], and it seems to be associated with tumor size, metastasis, risk of local recurrence, etc. [48,49]. However, the prognostic value of erbB3 expression in breast cancer has rather been poorly documented, and the available data are not conclusive [47–51]. Some studies showed that erbB3 expression, measured by immunohistochemistries (IHCs) [52], fluorescence in situ hybridization and IHC [53], or quantitative real-time PCR (qRT-PCR) [54], significantly reduced the overall and disease-free survival of the breast cancer patients. In contrast, other studies have reported a positive prognostic significance for erbB3 status. In a study that examined the expression of erbB family members with qRT-PCR, a positive main effect of erbB3 mRNA on survival was reported [51]. The expression of erbB3 was also correlated with positive estrogen receptor (ER) and progesterone receptor (PR) status and inversely associated with histological grade [55]. Recently, a study analyzed the transcriptional profiling of erbB genes and showed a correlation of erbB3 mRNA levels with ER positivity and a favorable prognostic value of overall survival [56]. While a definitive explanation for the differences among the studies with respect to the prognostic significance of erbB3 expression in breast cancer cannot be given at present, a number of hypotheses have been proposed. A naturally occurring secreted isoform of erbB3 (p85-soluble erbB3) is unique in that it can bind to the ligand HRG with high affinity, thereby blocks HRG binding to the full-length erbB3 and inhibits HRG-induced activation of erbB3 [57]. This observation could be associated with the positive prognostic value of erbB3 in some studies. Moreover, recent evidence suggests that the subcellular distribution of erbB family members affects substantially their biological activities. ErbB3 pool is mainly within intracellular compartments. It seems that the Acta Biochim Biophys Sin (2014) | Volume 46 | Issue 3 | Page 191

Downloaded from http://abbs.oxfordjournals.org/ at Kainan University on March 12, 2015

other family members, erbB3 is often aberrantly activated in a variety of human tumors [1,2]. It is frequently co-expressed with other RTKs in cancer cells to activate oncogenic signaling, with the PI-3K/Akt pathway as the most important one, and thereby promote tumor initiation and progression. Numerous studies on the underlying mechanisms implicate the functions of erbB3 as a major cause of treatment failure in cancer therapy [16]. As the erbB3/PI-3K/Akt signaling also plays a pivotal role in the development of erbB2-overexpressing (erbB2þ) breast cancer [17,18], castration-resistant prostate cancer (CRPC) [19], ovarian cancer [20,21], and EGFR tyrosine kinase inhibitor (TKI)-resistant lung cancer [22,23], concomitant inhibition of erbB3 is thought to be required to overcome resistance and effectively treat the cancer patients who have become resistant to the treatments. Several anti-erbB3 monoclonal antibodies (Abs) that prevent activation of erbB3 signaling are actively under preclinical investigations [24–26] and clinical studies (http://www.clinicaltrials. gov). Here, we discuss the latest progress in our understanding of the biology of erbB3 receptors in tumorigenesis and drug resistance of cancer.

ErbB3 in treatment resistance of cancer

ErbB3 Signaling in the Development of Cancer Therapeutic Resistance Homo- or heterodimerization of erbB receptors activates multiple downstream signaling pathways, such as PI-3K/Akt, RAS/RAF/MEK/MAPK, Phospholipase Cg (PLCg)/Protein kinase C (PKC), Jak/Stat, etc. [13]. The signaling pathways not only contribute to normal development, they also critically involve in multiple biological consequences and thereby promote tumor initiation and progression (Fig. 1). Therapeutic resistance associated with erbB signaling is frequently observed in cancer treatment. Because the expression and functions of erbB3 are regulated through a multitude of mechanisms in a number of cancer types, this highly dynamic nature facilitates erbB3 elicit refractory to many cancer therapeutics [16]. It has been shown that the activation of erbB family, mainly through MAPK, and Stat3 signaling, induces resistance to ALK inhibitors in the treatment of non-small-cell lung cancer (NSCLC) [61]. The erbB3 receptor and a non-RTK Src may form a compensatory signaling to enhance cancer cell survival under ionizing radiation [62]. Activation of both the PI-3K/Akt and the RAS/RAF/MEK/MAPK pathways via amplification of erbB2 or increased levels of the erbB3 ligand HRG results in resistance to EGFR-targeted therapy in colorectal cancers [63]. Thus, a recent study suggests that blockade of both erbB2 and erbB3 exhibits superior antitumor activity when compared with the combination of MEK and Akt inhibitors [64]. Of the four erbB receptors, erbB3 is the best suited to activate PI-3K/Akt signaling, because it has most tyrosine residues on its C-terminal tail, once being phosphorylated, capable of binding to the p85 subunit of PI-3K [15,65]. In fact, among all of erbB dimerization complexes, the erbB2/erbB3 heterodimer is the most biologically active and potent for the activation of the PI-3K/ AKT signaling pathway [66,67]. While erbB3 and erbB4 are capable of binding to the p85 regulatory subunit of PI-3K Acta Biochim Biophys Sin (2014) | Volume 46 | Issue 3 | Page 192

Figure 1. ErbB2/erbB3-initiated signal transduction pathways and the subsequent biological consequences Heterodimerization between erbB2 and erbB3 elicits tyrosine phosphorylation of both receptors. The phosphorylated tyrosine residues recruit adaptor proteins, such as PI-3K’s p85 subunit, GRB2, PLCg, or Jak, to activate the downstream signaling pathways. All these pathways will then converge to induce gene transcription and promote cell cycle progression, and subsequently enhance tumor cell survival and proliferation/maturation and increase therapeutic resistance and tumor metastasis.

directly, EGFR and erbB2 bind indirectly to p85 through adaptors or through heterodimerization with erbB3 or erbB4 [68,69]. Since the PI-3K/Akt signaling is the most important survival pathway in cell proliferation and its activation often leads to multidrug resistance in cancer treatment [70], it is not surprising that studies on the underlying mechanisms implicated the function of erbB3 as a major cause of treatment failure in cancer therapy [16].

Resistance to hormonal therapy Treatment of ER positive (ERþ) breast cancer patients with an anti-estrogen, such as tamoxifen, is commonly used and has significantly improved survival of the patients. However, both primary (de novo) and acquired resistances to tamoxifen frequently occur [71]. The mechanisms contributing to tamoxifen resistance include loss or mutation of ER, alterations in the intracellular signaling of breast cancer cells, ligandindependent ER-mediated transcription, and perturbation of the interactions of ER with some co-repressors [72]. Recently, activation of erbB family members has been linked to resistance to tamoxifen, since the increased erbB receptors facilitate

Downloaded from http://abbs.oxfordjournals.org/ at Kainan University on March 12, 2015

levels of phosphorylated erbB3 (P-erbB3) and its activity are associated with erbB3 re-localization to the plasma membrane. Furthermore, it has been demonstrated that ligands such as HRGs may affect the distribution of erbB3 and increase the membrane levels of the receptor [58]. In consequence, the impact of erbB3 on clinical outcomes of breast cancer patients may be better assessed when one considers not only its expression and interactions with other receptors like erbB2, but also its subcellular distribution as well as the expression levels of erbB3 ligands. Nonetheless, overexpression of erbB3 has been generally considered as a poor prognostic factor in breast cancer patients [59]. This was strongly supported by a recent study [60] showing that expression of erbB3 is associated with worse survival in solid tumors, including breast cancer, and the influence of erbB3 may be greater in the tumors where erbB2 is commonly overexpressed.

ErbB3 in treatment resistance of cancer

Resistance to targeted therapy Because of their extensive overexpression in tumor tissues and important function in cancer progression, attempts to target erbB receptors in cancer therapy have been the focus of research and have reached clinical applications. Currently used erbB-targeted therapy in clinic can be divided into two strategies: blocking Ab, such as cetuximab (Erbitux) against EGFR and trastuzumab (Herceptin) against erbB2; smallmolecule TKI, such as both gefitinib (Iressa) and erlotinib (Tarceva) targeting EGFR and lapatinib (Tykerb/Tyverb) dual-targeting EGFR and erbB2. These EGFR- and/or erbB2-targeted therapies have been successfully used in the treatment of various human cancers, including (cetuximab for) metastatic colorectal cancers and head and neck tumors,

(gefitinib and erlotinib for) EGFR mutant NSCLC, and (trastuzumab and lapatinib for) erbB2þ breast cancers and gastric cancers. However, almost all the tumors inevitably developed resistance to the therapeutic Abs and/or the inhibitors within 1 year. Numerous studies indicated that activation of erbB3 signaling is one of the major mechanisms contributing to the resistance to EGFR- and/or erbB2-targeted therapy [63,83]. It has been reported that a subset of colorectal cancer patients who exhibit either de novo or acquired resistance to cetuximab-based therapy has erbB2 amplification or high levels of circulating HRG, which induces activation of erbB3 signaling [84]. While multiple mechanisms exist in EGFR-TKI (gefitinib and erlotinib) resistance [63], another RTK MET amplification has been shown to lead to gefitinib resistance in lung cancer treatment by activating erbB3 signaling [22]. A recent study showing that dual targeting of EGFR and erbB3 is able to overcome acquired resistance to cetuximab and erlotinib further emphasized the importance of erbB3 signaling in resistance to EGFR-targeted therapy [23]. Trastuzumab (also known as Herceptin, a humanized monoclonal Ab against erbB2) is the first erbB2-targeted therapy approved by Food and Drug Administration (FDA) and has significantly improved the outcomes of early and metastatic erbB2þ breast cancer patients [85,86]. Unfortunately, resistance to trastuzumab is common and currently represents a huge clinical problem [87,88]. Tremendous efforts have been made to better understand the molecular mechanisms of trastuzumab resistance and to develop novel strategies/agents that can overcome the resistant phenotypes [89]. At the level of receptor complex, dimerization between erbB2 and another RTK, such as erbB3, EGFR, or the insulin-like growth factor I receptor (IGF-1R), may impair trastuzumab binding to erbB2. At the level of intracellular signaling, activation of the PI-3K/ Akt pathway and Src kinase have been identified as the major determinants of trastuzumab resistance [90,91]. It is well documented that both erbB3 and IGF-1R-initiated signaling pathways contribute to trastuzumab resistance [92–94]. Nonetheless, the relationship between erbB3 and IGF-IR in trastuzumab resistance remains unclear. We recently discovered that the erbB2 receptor simultaneously interacted with erbB3 and IGF-1R to form a trimeric complex in trastuzumabresistant breast cancer cells, and that it was the heterotrimerization of erbB3/erbB2/IGF-1R, not the heterodimer of erbB2/ erbB3, or IGF-1R/erbB2, that played a critical role in the breast cancer cells resistant to trastuzumab [95]. Further studies showed that specific knockdown of either erbB3 or IGF-1R was able to reverse trastuzumab resistance, and significantly enhances trastuzumab-mediated growth inhibitory effects on the otherwise resistant cells. For the downstream signaling, specific knockdown of erbB3 decreased the levels of both P-Akt and P-Src, whereas IGF-1R knockdown only gave rise to reduction of P-Src [95]. Our data suggested that erbB3 and IGF-1R initiate different signaling pathways contributing to Acta Biochim Biophys Sin (2014) | Volume 46 | Issue 3 | Page 193

Downloaded from http://abbs.oxfordjournals.org/ at Kainan University on March 12, 2015

breast cancer cells to bypass normal endocrine responsiveness [73,74]. It has been shown that the cross-talk between ER and erbB2/EGFR signaling promotes hormone-independent growth of breast cancer cells [75–77]. The importance of erbB3 in the development of tamoxifen-resistant phenotype is emerging. Clinical studies examining a large retrospective group of tamoxifen treated, ERþ breast cancer patients revealed that the patients with co-expression of erbB2 and erbB3 were significantly more likely to relapse on tamoxifen [78]. The tamoxifen-sensitive MCF-7 cells transfected with a HRGb-2 cDNA become estrogen-independent and resistant to tamoxifen both in vitro and in vivo [77]. Direct demonstration of the involvement of erbB3 in tamoxifen resistance comes from studies of Liu et al. who showed that down-regulation of erbB3 by a siRNA abrogated erbB2-mediated tamoxifen resistance in breast cancer cells [33]. Specific knockdown of erbB3 had no effect on ERa expression or its phosphorylation status, but significantly enhanced tamoxifen-associated growth inhibition in both MCF-7 and the erbB2-transfected MCF-7 (MCF-7/erbB2) cells via increased apoptosis [33]. The molecular mechanism responsible for the increased sensitivity to tamoxifen upon erbB3 down-regulation may be due to decreased levels of P-Akt, as it is well known that activation of PI-3K/Akt signaling is associated with tamoxifen resistance and MCF-7 cells expressing a constitutively active Akt proliferate under reduced estrogen conditions and are resistant to tamoxifen-mediated growth inhibition [79–81]. In addition, the erbB3/PI-3K/Akt signaling has also been shown to play a critical role in the development of CRPC. Although androgen withdrawal therapy (AWT) is currently the primary, first-line, an effective therapeutic intervention for recurrent prostate cancer, most patients ultimately develop resistance and progress to metastatic CRPC (mCRPC) [82]. Elevated expression of erbB3 in CRPC leads to androgen receptor stabilization and activation of PI-3K/Akt signaling [19]. Thus, erbB3 receptors may serve as useful biomarkers for modulating tamoxifen treatment and AWT in luminal B (ERþ, erbB2þ) breast cancers and CRPC, respectively.

ErbB3 in treatment resistance of cancer

the survival signaling, PI-3K/Akt pathway, it is understandable that activation of erbB3 signaling also results in chemoresistance in cancer treatment. Docetaxel-based chemotherapy has been established as the standard of care for mCRPC. However, only half of the patients benefit from docetaxel, and those who respond initially once again become resistance and eventually die of mCRPC [82,104]. Mechanistic studies suggested that activation of erbB3 mainly through PI-3K/Akt signaling plays a vital role in the progression of mCRPC into docetaxel resistance [19]. Increased secretion of HRG has been found in a subset of ovarian cancers, and thereby stimulates ovarian cancer cell proliferation via activated erbB3/HRG autocrine loop [21]. Recent studies suggested that erbB3 signaling may also contribute to chemoresistance in ovarian cancer, since the chemotherapeutic drug doxorubicin up-regulates HRG to activate the erbB3/PI-3K/Akt signaling in ovarian cancer cells [105]. It was postulated that targeting of erbB3 may significantly sensitize ovarian tumors to the toxic effects of platinum-based or other chemotherapy regimens [20]. Our early studies showed that co-expression of erbB2 and erbB3 in human breast cancer cell lines induced activation of PI-3K/Akt signaling and was associated with an increased resistance to multiple chemotherapeutic agents, such as paclitaxel, doxorubicin, 5-fluorouracil, etoposide, and camptothecin [70]. In identifying the key downstream mediator that contributes to chemoresistance, we discovered that elevated expression of erbB3 confers paclitaxel resistance in erbB2þ breast cancer cells via PI-3K/Akt-dependent up-regulation of Survivin [106]. A large body of evidence indicated that Survivin functions as a critical inhibitor of apoptosis to promote cell survival and proliferation, and regulates mitosis during cell cycle progression [107]. Survivin is selectively expressed in a variety of human malignancies and its overexpression positively correlates with poor prognosis, tumor recurrence, and therapeutic resistance [107]. Thus, novel strategies targeting Survivin, such as antisense oligonucleotide and pharmacological inhibitors may significantly enhance chemotherapeutic efficacy and are currently under clinical testing for cancer treatment [107–109]. Nonetheless, the precise mechanism by which the erbB3 signaling specifically up-regulates Survivin, but not the functionally related molecules, like Mcl-1 and Bcl-xL in erbB2þ breast cancer cells [106] remains unknown.

Perspectives Resistance to chemotherapy Chemotherapy, as an important conventional approach to treat human cancers, usually exerts antitumor activity via induction of apoptosis. The tumor cells with enhanced survival signaling and/or defects in apoptotic pathways may escape from chemotherapy. Since erbB3 (via interactions with other RTKs, particularly erbB2) is a potent activator of Acta Biochim Biophys Sin (2014) | Volume 46 | Issue 3 | Page 194

Studies on erbB RTKs have primarily focused on EGFR and erbB2 in cancer research. Recent advances dramatically improve our understanding of erbB3 as an obligate partner for other RTKs and draw our attention to the important role of erbB3 in both primary and acquired resistances to cancer therapeutics. The erbB3 signaling in cancer progression,

Downloaded from http://abbs.oxfordjournals.org/ at Kainan University on March 12, 2015

trastuzumab resistance—erbB3 activates both PI-3K/Akt signaling and Src kinase, whereas IGF-1R mainly elicits Src activation. Lapatinib is another erbB2-targeted therapy approved by FDA and has been used to treat erbB2þ breast cancer that has progressed after trastuzumab-based therapy [96]. Unfortunately, the efficacy of lapatinib is also compromised by resistance [97]. Some evidence showed that lapatinib and trastuzumab do not share common mechanisms of resistance, since lapatinib has activity in trastuzumab-resistant breast cancer [98]. However, a recent report indicated that loss of PTEN and the resulting activation of PI-3K/Akt signaling lead to lapatinib resistance [99]. Increased Src family kinase activity is also thought to be a mechanism of lapatinib resistance [100], and a recent study showed that a small-molecule Src inhibitor, saracatinib, significantly enhances lapatinibmediated antitumor activity against breast cancer brain metastasis [101]. As we have shown that erbB3 activates both PI-3K/Akt signaling and Src kinase in the trastuzumabresistant breast cancer cells [95], it is conceivable to hypothesize that activation of erbB3 signaling will induce lapatinib resistance as well. Nonetheless, whether erbB3 may function as a predictive biomarker for lapatinib sensitivity in breast cancer treatment remains to be further elucidated. In addition to causing resistance to erbB-targeted therapy, transcriptional up-regulation of erbB3 has been recently shown to involve in the resistance to RAF/MEK inhibitors in the treatment of melanoma and BRAF mutant thyroid carcinomas [102,103]. It appears that different tumor types utilize distinct mechanisms to up-regulate erbB3. The RAF inhibitor PLX4720 in melanoma enhanced erbB3 expression through a transcription factor, FOXD3 [102], whereas inhibition of RAF in thyroid cancers with vemurafenib (PLX4032) induced erbB3 transcription via decreased promoter occupancy by the transcriptional repressors C-terminal-binding proteins 1 and 2 (CtBP1/2) [103]. Once again, the increased erbB3 depended upon erbB2 to activate the downstream signaling Akt [102] or MAPK [103], and thereby resulted in resistance to the RAF inhibitors. Thus, in both the studies, therapeutic targeting of erbB2 with lapatinib was able to overcome the resistant phenotypes [102,103]. In light of the importance of enhanced erbB3 expression, we hypothesize that novel strategy inactivating the erbB3 signaling or reducing erbB3 protein levels may exhibit an even better efficacy in combination with the RAF inhibitors.

ErbB3 in treatment resistance of cancer

Funding This work was supported in part by the Front Range Ride Award (University of Colorado Cancer Center), the AEF

Bridge Funding (School of Medicine, University of Colorado Anschutz Medical Campus), and a research grant from Elsa U. Pardee Foundation (BL).

References 1. Baselga J and Swain SM. Novel anticancer targets: revisiting ERBB2 and discovering ERBB3. Nat Rev Cancer 2009, 9: 463 –475. 2. Hynes NE and MacDonald G. ErbB receptors and signaling pathways in cancer. Curr Opin Cell Biol 2009, 21: 177 – 184. 3. Hynes NE and Lane HA. ERBB receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer 2005, 5: 341– 354. 4. DeFazio A, Chiew YE, Sini RL, Janes PW and Sutherland RL. Expression of c-erbB receptors, heregulin and oestrogen receptor in human breast cell lines. Int J Cancer 2000, 87: 487 – 498. 5. Olayioye MA, Neve RM, Lane HA and Hynes NE. The ErbB signaling network: receptor heterodimerization in development and cancer. EMBO J 2000, 19: 3159. 6. Ogiso H, Ishitani R, Nureki O, Fukai S, Yamanaka M, Kim JH and Saito K, et al. Crystal structure of the complex of human epidermal growth factor and receptor extracellular domains. Cell 2002, 110: 775– 787. 7. Ferguson KM, Berger MB, Mendrola JM, Cho HS, Leahy DJ and Lemmon MA. EGF activates its receptor by removing interactions that autoinhibit ectodomain dimerization. Mol Cell 2003, 11: 507 –517. 8. Burgess AW, Cho HS, Eigenbrot C, Ferguson KM, Garrett TP, Leahy DJ and Lemmon MA, et al. An open-and-shut case? Recent insights into the activation of EGF/ErbB receptors. Mol Cell 2003, 12: 541 – 552. 9. Cho HS and Leahy DJ. Structure of the extracellular region of HER3 reveals an interdomain tether. Science 2002, 297: 1330– 1333. 10. Graus-Porta D, Beerli RR, Daly JM and Hynes NE. ErbB-2, the preferred heterodimerization partner of all ErbB receptors, is a mediator of lateral signaling. EMBO J 1997, 16: 1647– 1655. 11. Mendelsohn J and Baselga J. Status of epidermal growth factor receptor antagonists in the biology and treatment of cancer. J Clin Oncol 2003, 21: 2787 – 2799. 12. Boudeau J, Miranda-Saavedra D, Barton GJ and Alessi DR. Emerging roles of pseudokinases. Trends Cell Biol 2006, 16: 443– 452. 13. Citri A, Skaria KB and Yarden Y. The deaf and the dumb: the biology of ErbB-2 and ErbB-3. Exp Cell Res 2003, 284: 54 –65. 14. Shi F, Telesco SE, Liu Y, Radhakrishnan R and Lemmon MA. ErbB3/ HER3 intracellular domain is competent to bind ATP and catalyze autophosphorylation. Proc Natl Acad Sci USA 2010, 107: 7692– 7697. 15. Schulze WX, Deng L and Mann M. Phosphotyrosine interactome of the ErbB-receptor kinase family. Mol Syst Biol 2005, 1: 2005.0008. 16. Amin DN, Campbell MR and Moasser MM. The role of HER3, the unpretentious member of the HER family, in cancer biology and cancer therapeutics. Semin Cell Dev Biol 2010, 21: 944 –950. 17. Holbro T, Beerli RR, Maurer F, Koziczak M, Barbas CF, III and Hynes NE. The ErbB2/ErbB3 heterodimer functions as an oncogenic unit: ErbB2 requires ErbB3 to drive breast tumor cell proliferation. Proc Natl Acad Sci USA 2003, 100: 8933 – 8938. 18. Lee-Hoeflich ST, Crocker L, Yao E, Pham T, Munroe X, Hoeflich KP and Sliwkowski MX, et al. A central role for HER3 in HER2-amplified breast cancer: implications for targeted therapy. Cancer Res 2008, 68: 5878 – 5887. 19. Jathal MK, Chen L, Mudryj M and Ghosh PM. Targeting ErbB3: the new RTK(id) on the prostate cancer block. Immunol Endocr Metab Agents Med Chem 2011, 11: 131 –149. 20. Mills GB and Yarden Y. The rebirth of a phoenix: ovarian cancers are addicted to ErbB-3. Cancer Cell 2010, 17: 217 – 218.

Acta Biochim Biophys Sin (2014) | Volume 46 | Issue 3 | Page 195

Downloaded from http://abbs.oxfordjournals.org/ at Kainan University on March 12, 2015

particularly tumor recurrence following drug resistance has several implications for future investigations. ErbB3 may be considered as a valuable biomarker to predict the efficacy of EGFR- and/or erbB2-targeted therapy in the treatment of NSCLC and erbB2þ breast cancer, respectively. Therapeutic targeting of erbB3 may be an efficient way to conquer treatment failure and significantly enhance antitumor activity of hormonal therapy, EGFR/erbB2-targeted therapy, chemotherapy, and radiotherapy. Although the unique biological features of erbB3 receptor in cancer biology have been thoroughly investigated, several critical questions remain unanswered. First, activation of the erbB3 signaling, mainly through PI-3K/ Akt and MEK/MAPK pathways, not only confers drug resistance in cancer treatment, but also promotes tumor metastasis [110–114]. The key downstream mediators that are responsible for erbB3 signaling-mediated cancer progression remain unclear. We discovered that the PI-3K/Akt-dependent up-regulation of Survivin played a pivotal role in erbB3mediated paclitaxel resistance in erbB2þ breast cancer cells, and specific knockdown of Survivin abrogated the resistance [106]. However, it is not known whether the increased Survivin leads to resistance to all therapeutic agents; and the precise mechanism of erbB3/PI-3K/Akt-mediated upregulation of Survivin needs to be elucidated. Identification of additional mediators of erbB3 signaling may provide more opportunities to develop novel strategies revoking drug resistance and tumor metastasis. Secondly, both erbB2 and erbB3 have defects when they are considered individually. The two receptors rely on each other to elicit the most biologically active and potent activation of PI-3K/Akt signaling via heterodimerization [66,67]. Nonetheless, the molecular basis of erbB2/erbB3 co-expression in tumor cells remains elusive. It is unclear whether the same mechanisms are utilized by the tumor cells to simultaneously regulate erbB2 and erbB3, or tumor cells overexpress one receptor first and subsequently enhance the expression of the other. Finally, it is well-known that the ligand, HRG is critical to induce activation of erbB3 signaling. Aberrant production and/or maturation of HRG will alter tumor cell survival and proliferation. We believe that studies on the dysregulation of HRG in various human cancers may further our understanding of the ligand’s biological function in erbB3 signaling-mediated tumor initiation and progression and facilitate the development of novel approaches for cancer treatment. In conclusion, erbB3 is a focal point in erbB receptors-mediated tumorigenesis, and as such, constitutes a new potential biomarker and important target for cancer therapy.

ErbB3 in treatment resistance of cancer

Acta Biochim Biophys Sin (2014) | Volume 46 | Issue 3 | Page 196

40. deFazio A, Chiew YE, Sini RL, Janes PW and Sutherland RL. Expression of c-erbB receptors, heregulin and oestrogen receptor in human breast cell lines. Int J Cancer 2000, 87: 487 – 498. 41. Kim A, Liu B, Ordonez-Ercan D, Alvarez KM, Jones LD, McKimmey C and Edgerton SM, et al. Functional interaction between mouse erbB3 and wild-type rat c-neu in transgenic mouse mammary tumor cells. Breast Cancer Res 2005, 7: R708 – R718. 42. Siegel PM, Ryan ED, Cardiff RD and Muller WJ. Elevated expression of activated forms of Neu/ErbB-2 and ErbB-3 are involved in the induction of mammary tumors in transgenic mice: implications for human breast cancer. EMBO J 1999, 18: 2149– 2164. 43. Liles JS, Arnoletti JP, Tzeng CW, Howard JH, Kossenkov AV, Kulesza P and Heslin MJ, et al. ErbB3 expression promotes tumorigenesis in pancreatic adenocarcinoma. Cancer Biol Ther 2010, 10: 555 – 563. 44. Reschke M, Mihic-Probst D, van der Horst EH, Knyazev P, Wild PJ, Hutterer M and Meyer S, et al. HER3 is a determinant for poor prognosis in melanoma. Clin Cancer Res 2008, 14: 5188 –5197. 45. Barnes NL, Khavari S, Boland GP, Cramer A, Knox WF and Bundred NJ. Absence of HER4 expression predicts recurrence of ductal carcinoma in situ of the breast. Clin Cancer Res 2005, 11: 2163– 2168. 46. Naidu R, Yadav M, Nair S and Kutty M. Expression of c-erbB3 protein in primary breast carcinomas. Br J Cancer 1998, 78: 1385– 1390. 47. Quinn C, Ostrowski J, Lane S, Loney D, Teasdale J and Benson E. c-erbB-3 protein expression in human breast cancer: comparison with other tumour variables and survival. Histopathology 1994, 25: 247– 252. 48. Lemoine N, Barnes D, Hollywood D, Hughes C, Smith P, Dublin E and Prigent S, et al. Expression of the ERBB3 gene product in breast cancer. Br J Cancer 1992, 66: 1116 – 1121. 49. Travis A, Pinder S, Robertson J, Bell J, Wencyk P, Gullick W and Nicholson R, et al. C-erbB-3 in human breast carcinoma: expression and relation to prognosis and established prognostic indicators. Br J Cancer 1996, 74: 229 – 233. 50. Gasparini G, Pozza F, Bevilacqua P, Gullick W, Lemoine N, Maluta S and Palma P, et al. c-erbB-3 and c-erbB-2 protein expression in node-negative breast carcinoma—an immunocytochemical study. Eur J Cancer 1994, 30: 16 – 22. 51. Pawlowski V, Re´villion F, Hebbar M, Hornez L and Peyrat JP. Prognostic value of the type I growth factor receptors in a large series of human primary breast cancers quantified with a real-time reverse transcriptionpolymerase chain reaction assay. Clin Cancer Res 2000, 6: 4217– 4225. 52. Witton CJ, Reeves JR, Going JJ, Cooke TG and Bartlett J. Expression of the HER1 – 4 family of receptor tyrosine kinases in breast cancer. J Pathol 2003, 200: 290 – 297. 53. Sassen A, Rochon J, Wild P, Hartmann A, Hofstaedter F, Schwarz S and Brockhoff G. Cytogenetic analysis of HER1/EGFR, HER2, HER3 and HER4 in 278 breast cancer patients. Breast Cancer Res 2008, 10: R2. . 54. Bie`che I, Onody P, Tozlu S, Driouch K, Vidaud M and Lidereau R. Prognostic value of ERBB family mRNA expression in breast carcinomas. Int J Cancer 2003, 106: 758– 765. 55. Lee Y, Cho S, Seo JH, Shin BK, Kim HK, Kim I and Kim A. Correlated expression of erbB-3 with hormone receptor expression and favorable clinical outcome in invasive ductal carcinomas of the breast. Am J Clin Pathol 2007, 128: 1041 –1049. 56. Koutras A, Kalogeras K, Dimopoulos M, Wirtz R, Dafni U, Briasoulis E and Pectasides D, et al. Evaluation of the prognostic and predictive value of HER family mRNA expression in high-risk early breast cancer: a Hellenic Cooperative Oncology Group (HeCOG) study. Br J Cancer 2008, 99: 1775– 1785. 57. Lee H, Akita RW, Sliwkowski MX and Maihle NJ. A naturally occurring secreted human ErbB3 receptor isoform inhibits heregulinstimulated activation of ErbB2, ErbB3, and ErbB4. Cancer Res 2001, 61: 4467 – 4473.

Downloaded from http://abbs.oxfordjournals.org/ at Kainan University on March 12, 2015

21. Sheng Q, Liu X, Fleming E, Yuan K, Piao H, Chen J and Moustafa Z, et al. An activated ErbB3/NRG1 autocrine loop supports in vivo proliferation in ovarian cancer cells. Cancer Cell 2010, 17: 298 –310. 22. Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO and Lindeman N, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007, 316: 1039–1043. 23. Huang S, Li C, Armstrong EA, Peet CR, Saker J, Amler LC and Sliwkowski MX, et al. Dual targeting of EGFR and HER3 with MEHD7945A overcomes acquired resistance to EGFR inhibitors and radiation. Cancer Res 2013, 73: 824–833. 24. Aurisicchio L, Marra E, Luberto L, Carlomosti F, De Vitis C, Noto A and Gunes Z, et al. Novel anti-ErbB3 monoclonal antibodies show therapeutic efficacy in xenografted and spontaneous mouse tumors. J Cell Physiol 2012, 227: 3381 –3388. 25. Aurisicchio L, Marra E, Roscilli G, Mancini R and Ciliberto G. The promise of anti-ErbB3 monoclonals as new cancer therapeutics. Oncotarget 2012, 3: 744– 758. 26. Jiang N, Saba NF and Chen ZG. Advances in targeting HER3 as an anticancer therapy. Chemother Res Pract 2012, 2012: 817304. 27. Berger MB, Mendrola JM and Lemmon MA. ErbB3/HER3 does not homodimerize upon neuregulin binding at the cell surface. FEBS Lett 2004, 569: 332 – 336. 28. Plowman GD, Whitney GS, Neubauer MG, Green JM, McDonald VL, Todaro GJ and Shoyab M. Molecular cloning and expression of an additional epidermal growth factor receptor-related gene. Proc Natl Acad Sci USA 1990, 87: 4905– 4909. 29. Alimandi M, Romano A, Curia MC, Muraro R, Fedi P, Aaronson SA and Di Fiore PP, et al. Cooperative signaling of ErbB3 and ErbB2 in neoplastic transformation and human mammary carcinomas. Oncogene 1995, 10: 1813. 30. Hsieh A and Moasser M. Targeting HER proteins in cancer therapy and the role of the non-target HER3. Br J Cancer 2007, 97: 453– 457. 31. Wallasch C, Weiss F, Niederfellner G, Jallal B, Issing W and Ullrich A. Heregulin-dependent regulation of HER2/neu oncogenic signaling by heterodimerization with HER3. EMBO J 1995, 14: 4267 –4275. 32. Chakrabarty A, Rexer BN, Wang SE, Cook RS, Engelman JA and Arteaga C. H1047R phosphatidylinositol 3-kinase mutant enhances HER2-mediated transformation by heregulin production and activation of HER3. Oncogene 2010, 29: 5193–5203. 33. Liu B, Ordonez-Ercan D, Fan Z, Edgerton SM, Yang X and Thor AD. Downregulation of erbB3 abrogates erbB2-mediated tamoxifen resistance in breast cancer cells. Int J Cancer 2007, 120: 1874 –1882. 34. Liu B, Ordonez-Ercan D, Fan Z, Huang X, Edgerton SM, Yang X and Thor AD. Estrogenic promotion of ErbB2 tyrosine kinase activity in mammary tumor cells requires activation of ErbB3 signaling. Mol Cancer Res 2009, 7: 1882– 1892. 35. Beji A, Horst D, Engel J, Kirchner T and Ullrich A. Toward the prognostic significance and therapeutic potential of HER3 receptor tyrosine kinase in human colon cancer. Clin Cancer Res 2012, 18: 956– 968. 36. Maurer CA, Friess H, Kretschmann B, Zimmermann A, Stauffer A, Baer HU and Korc M, et al. Increased expression of erbB3 in colorectal cancer is associated with concomitant increase in the level of erbB2. Hum Pathol 1998, 29: 771 – 777. 37. Jaiswal BS, Kljavin NM, Stawiski EW, Chan E, Parikh C, Durinck S and Chaudhuri S, et al. Oncogenic ERBB3 mutations in human cancers. Cancer Cell 2013, 23: 603 –617. 38. Grupka NL, Lear-Kaul KC, Kleinschmidt-DeMasters BK and Singh M. Epidermal growth factor receptor status in breast cancer metastases to the central nervous system. Comparison with HER-2/neu status. Arch Pathol Lab Med 2004, 128: 974– 979. 39. Bieche I, Onody P, Tozlu S, Driouch K, Vidaud M and Lidereau R. Prognostic value of ERBB family mRNA expression in breast carcinomas. Int J Cancer 2003, 106: 758– 765.

ErbB3 in treatment resistance of cancer

77. Tang CK, Perez C, Grunt T, Waibel C, Cho C and Lupu R. Involvement of heregulin-b2 in the acquisition of the hormone-independent phenotype of breast cancer cells. Cancer Res 1996, 56: 3350– 3358. 78. Tovey S, Dunne B, Witton CJ, Forsyth A, Cooke TG and Bartlett JM. Can molecular markers predict when to implement treatment with aromatase inhibitors in invasive breast cancer? Clin Cancer Res 2005, 11: 4835–4842. 79. Clark AS, West K, Streicher S and Dennis PA. Constitutive and inducible Akt activity promotes resistance to chemotherapy, trastuzumab, or tamoxifen in breast cancer cells. Mol Cancer Ther 2002, 1: 707– 717. 80. Friedrichs WE, Russell DH, Donzis EJ, Middleton AK, Silva JM, Roth RA and Hidalgo M. Inhibition of mTOR activity restores tamoxifen response in breast cancer cells with aberrant Akt activity. Clin Cancer Res 2004, 10: 8059 – 8067. 81. Jordan NJ, Gee JM, Barrow D, Wakeling AE and Nicholson RI. Increased constitutive activity of PKB/Akt in tamoxifen resistant breast cancer MCF-7 cells. Breast Cancer Res Treat 2004, 87: 167– 180. 82. Seruga B and Tannock IF. Chemotherapy-based treatment for castrationresistant prostate cancer. J Clin Oncol 2011, 29: 3686 – 3694. 83. Kruser TJ and Wheeler DL. Mechanisms of resistance to HER family targeting antibodies. Exp Cell Res 2010, 316: 1083 – 1100. 84. Yonesaka K, Zejnullahu K, Okamoto I, Satoh T, Cappuzzo F, Souglakos J and Ercan D, et al. Activation of ERBB2 signaling causes resistance to the EGFR-directed therapeutic antibody cetuximab. Sci Transl Med 2011, 3: 99ra86. 85. Incorvati JA, Shah S, Mu Y and Lu J. Targeted therapy for HER2 positive breast cancer. J Hematol Oncol 2013, 6: 38. 86. Nielsen DL, Kumler I, Palshof JA and Andersson M. Efficacy of HER2-targeted therapy in metastatic breast cancer. Monoclonal antibodies and tyrosine kinase inhibitors. Breast 2013, 22: 1 –12. 87. Hurvitz SA, Hu Y, O’Brien N and Finn RS. Current approaches and future directions in the treatment of HER2-positive breast cancer. Cancer Treat Rev 2013, 39: 219– 229. 88. Rexer BN and Arteaga CL. Intrinsic and acquired resistance to HER2-targeted therapies in HER2 gene-amplified breast cancer: mechanisms and clinical implications. Crit Rev Oncog 2012, 17: 1– 16. 89. Stern HM. Improving treatment of HER2-positive cancers: opportunities and challenges. Sci Transl Med 2012, 4: 127rv122. 90. Berns K, Horlings HM, Hennessy BT, Madiredjo M, Hijmans EM, Beelen K and Linn SC, et al. A functional genetic approach identifies the PI3K pathway as a major determinant of trastuzumab resistance in breast cancer. Cancer Cell 2007, 12: 395 –402. 91. Zhang S, Huang WC, Li P, Guo H, Poh SB, Brady SW and Xiong Y, et al. Combating trastuzumab resistance by targeting SRC, a common node downstream of multiple resistance pathways. Nat Med 2011, 17: 461 – 469. 92. Agus DB, Akita RW, Fox WD, Lewis GD, Higgins B, Pisacane PI and Lofgren JA, et al. Targeting ligand-activated ErbB2 signaling inhibits breast and prostate tumor growth. Cancer Cell 2002, 2: 127– 137. 93. Lu Y, Zi X, Zhao Y, Mascarenhas D and Pollak M. Insulin-like growth factor-I receptor signaling and resistance to trastuzumab (herceptin). J Natl Cancer Inst 2001, 93: 1852– 1857. 94. Nahta R, Yuan LX, Zhang B, Kobayashi R and Esteva FJ. Insulin-like growth factor-I receptor/human epidermal growth factor receptor 2 heterodimerization contributes to trastuzumab resistance of breast cancer cells. Cancer Res 2005, 65: 11118 –11128. 95. Huang X, Gao L, Wang S, McManaman JL, Thor AD, Yang X and Esteva FJ, et al. Heterotrimerization of the growth factor receptors erbB2, erbB3, and insulin-like growth factor-i receptor in breast cancer cells resistant to herceptin. Cancer Res 2010, 70: 1204 –1214. 96. Geyer CE, Forster J, Lindquist D, Chan S, Romieu CG, Pienkowski T and Jagiello-Gruszfeld A, et al. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med 2006, 355: 2733 –2743.

Acta Biochim Biophys Sin (2014) | Volume 46 | Issue 3 | Page 197

Downloaded from http://abbs.oxfordjournals.org/ at Kainan University on March 12, 2015

58. Offterdinger M, Scho¨fer C, Weipoltshammer K and Grunt TW. c-erbB-3 a nuclear protein in mammary epithelial cells. J Cell Biol 2002, 157: 929 – 940. 59. Chiu CG, Masoudi H, Leung S, Voduc DK, Gilks B, Huntsman DG and Wiseman SM. HER-3 overexpression is prognostic of reduced breast cancer survival: a study of 4046 patients. Ann Surg 2010, 251: 1107–1116. 60. Ocana A, Vera-Badillo F, Seruga B, Templeton A, Pandiella A and Amir E. HER3 overexpression and survival in solid tumors: a meta-analysis. J Natl Cancer Inst 2013, 105: 266 –273. 61. Tanizaki J, Okamoto I, Okabe T, Sakai K, Tanaka K, Hayashi H and Kaneda H, et al. Activation of HER family signaling as a mechanism of acquired resistance to ALK inhibitors in EML4-ALK-positive non-small cell lung cancer. Clin Cancer Res 2012, 18: 6219– 6226. 62. Contessa JN, Abell A, Mikkelsen RB, Valerie K and Schmidt-Ullrich RK. Compensatory ErbB3/c-Src signaling enhances carcinoma cell survival to ionizing radiation. Breast Cancer Res Treat 2006, 95: 17– 27. 63. Vlacich G and Coffey RJ. Resistance to EGFR-targeted therapy: a family affair. Cancer Cell 2011, 20: 423– 425. 64. Kirouac DC, Du JY, Lahdenranta J, Overland R, Yarar D, Paragas V and Pace E, et al. Computational modeling of ERBB2-amplified breast cancer identifies combined ErbB2/3 blockade as superior to the combination of MEK and AKT inhibitors. Sci Signal 2013, 6: ra68. 65. Campbell MR, Amin D and Moasser MM. HER3 comes of age: new insights into its functions and role in signaling, tumor biology, and cancer therapy. Clin Cancer Res 2010, 16: 1373– 1383. 66. Mattoon D, Lamothe B, Lax I and Schlessinger J. The docking protein Gab1 is the primary mediator of EGF-stimulated activation of the PI-3K/ Akt cell survival pathway. BMC Biol 2004, 2: 24. 67. Suenaga A, Takada N, Hatakeyama M, Ichikawa M, Yu X, Tomii K and Okimoto N, et al. Novel mechanism of interaction of p85 subunit of phosphatidylinositol 3-kinase and ErbB3 receptor-derived phosphotyrosyl peptides. J Biol Chem 2005, 280: 1321 – 1326. 68. Hemi R, Paz K, Wertheim N, Karasik A, Zick Y and Kanety H. Transactivation of ErbB2 and ErbB3 by tumor necrosis factor-a and anisomycin leads to impaired insulin signaling through serine/threonine phosphorylation of IRS proteins. J Biol Chem 2002, 277: 8961 – 8969. 69. Vivanco I and Sawyers CL. The phosphatidylinositol-3-kinase—AKT pathway in human cancer. Nat Rev Cancer 2002, 2: 489 – 501. 70. Knuefermann C, Lu Y, Liu B, Jin W, Liang K, Wu L and Schmidt M, et al. HER2/PI-3K/Akt activation leads to a multidrug resistance in human breast adenocarcinoma cells. Oncogene 2003, 22: 3205– 3212. 71. Clarke R, Leonessa F, Welch JN and Skaar TC. Cellular and molecular pharmacology of antiestrogen action and resistance. Pharmacol Rev 2001, 53: 25 –72. 72. Mouridsen HT, Rose C, Brodie AH and Smith IE. Challenges in the endocrine management of breast cancer. Breast 2003, 12: S2 – S19. 73. Newby JC, Johnston S, Smith IE and Dowsett M. Expression of epidermal growth factor receptor and c-erbB2 during the development of tamoxifen resistance in human breast cancer. Clin Cancer Res 1997, 3: 1643 – 1651. 74. Shou J, Massarweh S, Osborne CK, Wakeling AE, Ali S, Weiss H and Schiff R. Mechanisms of tamoxifen resistance: increased estrogen receptor-HER2/neu cross-talk in ER/HER2 – positive breast cancer. J Natl Cancer Inst 2004, 96: 926– 935. 75. Benz CC, Scott GK, Sarup JC, Johnson RM, Tripathy D, Coronado E and Shepard HM, et al. Estrogen-dependent, tamoxifen-resistant tumorigenic growth of MCF-7 cells transfected with HER2/neu. Breast Cancer Res Treat 1992, 24: 85 – 95. 76. Pietras RJ, Arboleda J, Reese DM, Wongvipat N, Pegram MD, Ramos L and Gorman CM, et al. HER-2 tyrosine kinase pathway targets estrogen receptor and promotes hormone-independent growth in human breast cancer cells. Oncogene 1995, 10: 2435 –2446.

ErbB3 in treatment resistance of cancer

Acta Biochim Biophys Sin (2014) | Volume 46 | Issue 3 | Page 198

105. Bezler M, Hengstler JG and Ullrich A. Inhibition of doxorubicin-induced HER3-PI3K-AKT signalling enhances apoptosis of ovarian cancer cells. Mol Oncol 2012, 6: 516 – 529. 106. Wang S, Huang X, Lee CK and Liu B. Elevated expression of erbB3 confers paclitaxel resistance in erbB2-overexpressing breast cancer cells via upregulation of survivin. Oncogene 2010, 29: 4225– 4236. 107. Church DN and Talbot DC. Survivin in solid tumors: rationale for development of inhibitors. Curr Oncol Rep 2012, 14: 120 – 128. 108. Kanwar JR, Kamalapuram SK and Kanwar RK. Targeting survivin in cancer: the cell-signalling perspective. Drug Discov Today 2011, 16: 485 – 494. 109. Kelly RJ, Lopez-Chavez A, Citrin D, Janik JE and Morris JC. Impacting tumor cell-fate by targeting the inhibitor of apoptosis protein survivin. Mol Cancer 2011, 10: 35. 110. Katso R, Okkenhaug K, Ahmadi K, White S, Timms J and Waterfield MD. Cellular function of phosphoinositide 3-kinases: implications for development, homeostasis, and cancer. Annu Rev Cell Dev Biol 2001, 17: 615 – 675. 111. Reddy KB, Nabha SM and Atanaskova N. Role of MAP kinase in tumor progression and invasion. Cancer Metastasis Rev 2003, 22: 395–403. 112. Summy JM and Gallick GE. Src family kinases in tumor progression and metastasis. Cancer Metastasis Rev 2003, 22: 337– 358. 113. Webb DJ, Donais K, Whitmore LA, Thomas SM, Turner CE, Parsons JT and Horwitz AF. FAK-Src signalling through paxillin, ERK and MLCK regulates adhesion disassembly. Nat Cell Biol 2004, 6: 154 – 161. 114. Xue C, Liang F, Mahmood R, Vuolo M, Wyckoff J, Qian H and Tsai KL, et al. ErbB3-dependent motility and intravasation in breast cancer metastasis. Cancer Res 2006, 66: 1418– 1426.

Downloaded from http://abbs.oxfordjournals.org/ at Kainan University on March 12, 2015

97. Chen FL, Xia W and Spector NL. Acquired resistance to small molecule ErbB2 tyrosine kinase inhibitors. Clin Cancer Res 2008, 14: 6730– 6734. 98. McArthur H. An overview of HER-targeted therapy with lapatinib in breast cancer. Adv Ther 2009, 26: 263– 271. 99. Eichhorn PJ, Gili M, Scaltriti M, Serra V, Guzman M, Nijkamp W and Beijersbergen RL, et al. Phosphatidylinositol 3-kinase hyperactivation results in lapatinib resistance that is reversed by the mTOR/ phosphatidylinositol 3-kinase inhibitor NVP-BEZ235. Cancer Res 2008, 68: 9221– 9230. 100. Rexer BN, Ham AJ, Rinehart C, Hill S, Granja-Ingram Nde M, Gonzalez-Angulo AM and Mills GB, et al. Phosphoproteomic mass spectrometry profiling links Src family kinases to escape from HER2 tyrosine kinase inhibition. Oncogene 2011, 30: 4163– 4174. 101. Zhang S, Huang WC, Zhang L, Zhang C, Lowery FJ, Ding Z and Guo H, et al. SRC family kinases as novel therapeutic targets to treat breast cancer brain metastases. Cancer Res 2013, 73: 5764– 5774. 102. Abel EV, Basile KJ, Kugel CH, III, Witkiewicz AK, Le K, Amaravadi RK and Karakousis GC, et al. Melanoma adapts to RAF/MEK inhibitors through FOXD3-mediated upregulation of ERBB3. J Clin Invest 2013, 123: 2155– 2168. 103. Montero-Conde C, Ruiz-Llorente S, Dominguez JM, Knauf JA, Viale A, Sherman EJ and Ryder M, et al. Relief of feedback inhibition of HER3 transcription by RAF and MEK inhibitors attenuates their antitumor effects in BRAF-mutant thyroid carcinomas. Cancer Discov 2013, 3: 520 – 533. 104. Sartor AO. Progression of metastatic castrate-resistant prostate cancer: impact of therapeutic intervention in the post-docetaxel space. J Hematol Oncol 2011, 4: 18.

Role of erbB3 receptors in cancer therapeutic resistance.

ErbB3 receptors are unique members of the erbB receptor tyrosine kinases (RTKs), which are often aberrantly expressed and/or activated in human cancer...
266KB Sizes 1 Downloads 0 Views