Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Jun 1.
Published in final edited form as: Expert Opin Investig Drugs. 2010 Jun;19(6):709–722. doi: 10.1517/13543781003769844

Investigational EGFR-targeted therapies in HNSCC

Andre Cassell 1, Jennifer R Grandis 1,2
PMCID: PMC2947445  NIHMSID: NIHMS187844  PMID: 20415598

Abstract

Importance of the Field

The epidermal growth factor receptor (EGFR) is an established therapeutic target in head and neck squamous cell carcinoma (HNSCC). The EGFR-targeting monoclonal antibody cetuximab (™Erbitux) was FDA-approved for use in HNSCC in 2006. The molecular basis for the efficacy of an antibody approach compared with inhibition of EGFR tyrosine kinase function using small molecule inhibitors, or downregulation of protein expression via antisense strategies remains incompletely understood.

Areas covered in this review

A literature search was performed to identify studies elucidating mechanisms of action of several approaches to targeting EGFR in HNSCC (monoclonal antibodies, tyrosine kinase inhibitors, antisense approaches, and ligand toxin conjugates).

What the reader will gain

Monoclonal antibodies decrease tumor growth via receptor endocytosis and recruitment of host immune defenses. Tyrosine kinase inhibitors bind to the ATP binding pocket of the tyrosine kinase domain, inhibiting signaling. Antisense approaches decrease EGFR expression with high specificity although drug delivery remains problematic. Ligand-toxin conjugates facilitate the entry of toxin and the ADP-ribosylation of the ribosome, thereby inhibiting translation.

Take home message

Elucidation mechanisms by which these different strategies inhibit EGFR function may enhance the development of more effective treatments for HNSCC and enable prospective identification of individuals who will benefit from EGFR inhibition.

Keywords: EGFR, TKI, monoclonal antibodies, antisense oligonucleotides

1. INTRODUCTION

Head and Neck squamous cell carcinoma (HNSCC) is the sixth most prevalent cancer worldwide and is responsible for 90% of the cancers that arise in the mucosal surfaces of the head and neck [1]. There are 40,000 new HNSCC cases and 12,000 deaths per year in the United States alone [1]. In addition, there are 600,000 new HNSCC cases worldwide annually [2-3]. Risk factors for the development of HNSCC include tobacco smoking and alcohol consumption [1]. In addition, the Human Papilloma Virus (HPV) has emerged as a risk factor for HNSCC, primarily HPV-16 [4]. HPV16 seropositivity has been associated with increased risk for HSNCC [5]. Current treatment for HNSCC involves radiation or surgery in early stages of the disease [6]. Treatment for more advanced stages involves either radiation or surgery in combination with chemotherapy [7]. Despite improvements in surgical techniques, chemotherapy agents and radiation delivery, long-term survival is relatively unchanged [8]. New therapeutic approaches, including molecular targeted therapies are needed to increase long-term survival.

Epidermal Growth Factor Receptor (EGFR) has been implicated in the pathogenesis of HNSCC. EGFR is overexpressed in up to 90 % of head and neck cancers compared with levels in normal mucosa [9-10] where expression levels correlate with decreased survival, independent of therapy [11-12]. Targeting EGFR using a variety of strategies abrogates tumor growth in preclinical HNSCC models [13-15]. Despite ubiquitous EGFR expression in HNSCC, only a subset of patients will respond to EGFR inhibitors, including cetuximab. Even in the setting of upstream EGFR inhibition, alternative downstream signaling pathways are persistently activated [16]. This persistent activation has been shown to result, at least in part, from signaling through other cell-surface receptors including G-protein-coupled receptors (GPCRs), Insulin Growth Factor Receptor (IGF-R) and Met [17]. Combined targeted therapy of both EGFR and key intermediates in GPCR-EGFR crosstalk pathways showed an enhanced effect on migration and invasion compared to either therapy alone [17]. New treatments must be devised to improve the low cure rate with conventional therapies and the appearance of secondary tumors after initial intervention. Knowledge of the EGFR protein and its downstream signaling effects has advanced the field of targeted agents significantly. The promise that combined therapy holds, especially in the context of combined EGFR/GPCR targeting or radiation and targeted therapy may reveal that clinical therapy may involve many of these treatment modalities. In this review, we will describe the efficacy of individual EGFR targeting strategies in the context of HNSCC. We will also discuss how the mechanisms of anti-tumor effects may confer sensitivity or resistance to specific therapeutic approaches [18].

2. HNSCC AND EGFR BIOLOGY

EGFR (erbB1, HER1), a 170 kDa cell-surface protein, regulates the growth and differentiation of cells. EGFR is a member of the erbB family of receptor tyrosine kinases, four closely related cell membrane receptors: EGFR (HER1 or erbB1), erbB2 (HER2), erbB3 (HER3), and erbB4 (HER4), which contribute to proliferation and invasion of cancer cells, including HNSCC cells, in response to stimulation with growth factors [19]. EGFR is activated in response to specific ligands, including EGFR, TGF-alpha, HB-EGF, and amphiregulin [20].

Under normal circumstances, EGF ligand binds to the extracellular domain of the EGFR and induces a conformational change. The binding of EGF to the extracellular domain (also known as domains I and III) causes a single EGF receptor to undergo a more extended conformation, due to the severing of intramolecular bonds acting upon domains II and IV [21]. In this process, domain II, a cystine-rich domain becomes exposed and is the driving force for the formation of a “dimerization arm” between two EGF-receptors [21]. These conformational changes result in the homodimerization or heterodimerization of the EGFR with other members of the ErbB family. Once dimerization occurs, the receptor becomes activated [21]. X-ray crystallography studies have shown that disrupting domains II and IV through deletion or mutation or artificially creating an extended EGFR conformation does not create an active EGFR protein. In fact, it is hypothesized that the activated form of EGFR is induced through a combination of effects, including the unfolding of EGFR and the exposure of the wild-type form of domain II [21]. This activation sequence is in contrast to other members of the receptor tyrosine kinases, which are activated by receptor-mediated dimerization instead of undergoing a stabilization upon ligand binding before activation [22].

The mechanisms by which EGFR activation occurs has implications for potential strategies to downmodulate EGFR. Therapeutic antibodies that compete for EGFR ligand binding to the extracellular domain of the receptor, stabilize the tethered conformation, block the extended state, and directly inhibit ligand binding and receptor dimerization [23]. X-ray crystallography studies demonstrate that cetuximab competes with EGF ligand(s) by binding to an epitope on domain III of the EGFR, which significantly overlaps with the extracellular binding domain [23-24]. This binding also causes a steric blockade of the unfolding of EGFR into the extended conformation that is required for dimerization [5-6]. Direct inhibition of the dimerization domain by monoclonal antibodies to the ErbB2 form of domain II also represents a therapeutic mechanism for monoclonal antibodies. Pertuzumab blocks the heterodimerization of ErbB2 and ErbB3 with EGFR and is used for breast cancer therapy [5].

The physical juxtaposition of the receptors due to domain II has implications for EGFR activation mechanisms. Studies by Zhang and colleagues showed that the CDK/cyclin-like interface in the EGFR protein is required for receptor activation [25]. Point mutations in this interface result in abrogation of EGFR activation [25]. Studies have also focused on defining which phosphotyrosine residues are responsible for ErbB downstream signaling and what interacting partners are associated with these residues. Using a novel, high throughput screen to examine binding partners of the 89 cytosolic tyrosine residues, Matthias Mann and colleagues have determined that the binding partners of ErbB1-ErbB4 phosphotyrosine residues are conserved, such as Grb and Src [26]. Interactors of phosphotyrosine residues then function as intermediates with the Ras-Raf-Mek-MAPK, Jak-Stat, PLC-gamma, and/or the PI3K/Akt signaling pathways. These contribute to cell survival, proliferation, invasion, and metastasis [9, 19].

3.1. MECHANISMS OF RESISTANCE

EGFR transactivation by GPCRs as been implicated as a mechanism of persistent downstream EGFR signaling [27] and represents one potential mechanism of resistance (Figure 1). In cetuximab resistant clones derived by prolonged in vitro exposure, an increase in EGFR, ErbB2 and ErbB3 was detected compared to parental lines [28]. To determine the effect of EGFR phosphorylation on the activation of Her2 and Her3, Harari and colleagues used TKIs to inhibit the 1173 phosphotyrosine residue on EGFR and examined expression levels of Her2, Her3, cMet, Akt, and MAPK [28]. Levels of these proteins were decreased compared to non-treated controls, indicating that EGFR activation contributes upregulation of Her2 and Her3, increased downstream signaling, and consequent resistance to antibodies [28]. Evidence supporting the contribution of Her2 and Her3 to cetuximab resistance involved the use of 2C4, an inhibitor to Her2 dimerization. Suppression of Akt and Her3 were seen upon treatment with cetuximab and 2C4 compared to cetuximab alone, revealing the dependence of resistant cells on Her2 expression [28]. In addition, loss of Her3 resensitizes resistant cell lines to cetuximab, implicating Her3 in resistance [28].

Figure 1.

Figure 1

Signaling pathways that may contribute to resistance to EGFR inhibitors in HNSCC. Binding of ligand to EGFR induces a conformational change that trigger molecular cascades responsible for survival and proliferation. G-protein-coupled receptors (GPCRs) maintain persistent EGFR signaling in the presence of EGFR inhibitors. The loss of E-cadherin and tight-junction expression and the transition of tumor cells from an epithelial to transitional morphology also contribute to cell survival. Her2 overexpression and consequent increased heterodimerization also results in increased downstream EGFR signaling and is associated with cetuximab resistance. The EGFRvIII variant is also associated with resistance; its truncated extracellular binding domain and constitutive signaling decreases response to cetuximab. EGFR: Epidermal Growth Factor Receptor; PI3K: Phosphoinositide 3-kinase; PDK1: Phosphoinositide-dependent kinase 1; mTOR: mammalian Target of Rapamycin; Ras: Renin-angiotensin system; Raf: Relative angiostatic factor; MAPK: Mitogen-activated protein kinase; Mek: MAPK kinase; Jak: Janus kinase; STAT: Signal Transducers and Activators of Transcription.

In addition to increased transactivation of EGFR with Her2 and Her3 conferring resistance to therapy, genomic amplification can also result in resistance. EGFR copy number was assessed through the ratio of the real-time PCR level of EGFR vs. Met in ten HNSCC lines. Twenty percent of the cell lines showed relative copy numbers greater than 5 and half of the cell lines tested revealed a copy number between 2 and 5, indicating a low to moderate amount of EGFR amplification [14]. In addition, high EGFR copy numbers was statistically associated with cetuximab and gefitinib resistance [14]. High expression of ErbB2 and ErbB3 has also been implicated in gefitinib resistance where increased levels or ErbB2 and ErbB3 expression correlated with high IC50s in three HNSCC cell lines [14]. Other studies have shown that EGFR FISH (Flourescent in situ hybridization) copy number has been implicated in poor prognosis [29]. Chung and colleagues have found that in 75 HNSCC tumor samples, 58% were FISH positive and that tumor differentiation was weakly associated with FISH status [29]. FISH status was also a significant prognostic indicator of progression-free and overall survival [29].

Kinase domain mutations in of EGFR in HNSCC are extremely rare but may be associated with altered responses to EGFR inhibitors when they occur [30]. In one study, tumor samples of 100 patients with advanced primary or relapsed HNSCC were analyzed by PCR. Results revealed that one patient K745R mutation in the ATP binding site. This mutation may confer resistance to TKIs due to the stabilization of residues involved in binding to both ATP and TKIs [30]. This mechanism has been hypothesized to explain TKI resistance in NSCLC, involving a mutation in close proximity to the K745R mutation.

In addition, the induction of the epithelial to mesenchymal transition (EMT) has been shown to be a marker of resistance to EGFR-targeted therapy [31]. In high risk HNSCC, loss of tight and adherens junctions, dysregulation of E-cadherin and the conversion of cells to a more spindle-shaped morphology facilitates movement across the basement membrane and increased metastasis [32]. The resistance to tyrosine kinase inhibitors may be due to the expression of proteins like vimentin and loss of the epithelial cell adhesion molecule EpCAM [31]. In addition, the phenotype of the cell itself rather than the expression of proteins may be responsible for TKI resistance. The loss of cell to cell adhesion through decreased claudin expression and increased cell motility confer a more aggressive phenotype to the tumor and may explain resistance [31]. These changes in gene expression has been shown to predict gefinitib resistance in lung cancer and HNSCC models [31].

Furthermore, EGFR transactivation by GPCRs has been implicated as another mechanism of persistent downstream signaling and resistance [27]. When used as monotherapy, EGFR inhibitors show little success in treating HNSCC and suggest a collateral mechanism may be involved. Upon stimulation of GPCRs, the release of EFGR ligand increases, explaining continued EGFR signaling. In addition, GPCRs have also been shown to act in both EGFR-dependent and independent mechanisms. Bradykinin (BK) and lysophosphatidic acid (LPA), two GPCR ligands, have been shown to activate MAPK in both the presence and absence of inhibitors to EGFR [27]. Interestingly, the EGFRvIII variant requires no ligand binding for activation and represents another mechanism of resistance. The EGFRvIII variant results from a deletion of exons 2 through 7, which codes for the extracellular binding domain [33]. The resulting protein is 150kDa and is weakly constitutively active; the absence of a binding domain precludes binding of monoclonal antibodies to the protein. EGFRvIII transfected cells have been shown to be more resistant to cetuximab when compared to vector-transfected cells in vitro and in vivo [33]. Activated EGFR may then activate pathways that are responsible for resistance, including STAT proteins, and the PI3K/Akt pathway.

3.2 Preclinical Data and Mechanisms

Monoclonal antibodies exert their effects by binding to the extracellular domain of the EGFR and competing for ligands including TGF-alpha and amphiregulin, which act as autocrine ligands for EGFR [34-35]. Unlike endogenous ligands, which stimulate downstream EGFR effectors, monoclonal antibodies result in the abrogation of signal transduction [36]. Cetuximab abrogated EGFR-induced upregulation of EGFR, determined by decreases in Erk1/2 activation [36]. In contrast, when these cells were incubated with EGF and cisplatin alone, there was an upregulation of EGFR [36]. This suggests that abrogation of EGFR activation is mediated by competitive binding of the antibody to its receptor.

In addition to competitive binding, increased degradation and internalization of the EGFR has also been shown to be a mechanism by which monoclonal antibodies decrease EGFR signaling [37]. In an effort to investigate the cellular localization of the EGFR in the presence of monoclonal antibodies, HNSCC cells treated with EGFR monoclonal antibodies showed a decrease in the cell surface expression of EGFR compared to controls [37]. Internalization of the receptor prevents EGFR from binding to its ligands and thereby interferes with signaling.

X-ray crystallography studies performed by the Ferguson lab have shown that the humanized antibody Fab11F8 interacts exclusively with domain III, preventing EGF from binding to domain III and results in the tethered phenotype [24]. In addition, most of the interactions of Fab11F8 involve interactions between domain III and the heavy-chain complementary-determining regions (CDRs) [24]. H1 lies at the center of the interaction between VH and domain III and stabilizes the interaction between H2 and H3 [24].

The structural basis for antibody targeting is not the only mechanism that makes antibodies effective at downmodulating EGFR/ErbB expression, however. Cross-linking and surface depletion has been shown to be instrumental as well. The Yarden lab has shown that antibodies engages endocytosis of EGFR at a much slower rate than EGF, as determined by biotinylation and Western blot [38]. Combinations of antibodies directed[39] at different epitopes of EGFR caused a synergistic downregulation of EGFR [38]. This downregulation, then, seems to be dependent on the amount of cross-linking at the cell surface [38].

Monoclonal antibodies may also exert their anti-tumor properties via their effects on the immune systems of patients with HNSCC. Patients with HNSCC may exhibit upregulation of angiogenic and inflammatory factors that promote tumorigenesis, like VEGF[40]. Much work has been performed to evaluate the effects of using antibodies to neutralize the effects of VEGF in a variety of tumors. Interestingly, one side effect and challenge of therapy against VEGF is a compensatory increase in circulating VEGF levels [40-41]. Other factors include increased production of proangiogenic factors including IL8 and a dysregulation in the cytotoxic ability of effector T-cells [42-43]. Antibodies have demonstrated the ability to increase the cytotoxic capacity of natural killer cells and dendritic cells in head and neck cancer cells in vitro [42-43]. In addition, monoclonal antibodies also competitively binding to the EGFR, increase internalization and degradation of the EGFR, and induce antibody-dependent cell-mediated cytotoxicity (ADCC).

Chimeric monoclonal antibodies, by virtue of their Fc constant regions, can elicit ADCC, which may contribute to the clinical response mediated by monoclonal antibodies by recruiting effector cells [4-5]. In the process of ADCC, cells of the innate immune response bind to the constant regions of antibodies through their Fc-gamma receptors. Opsonization of a cancer cell with antibodies to EGFR with consequent binding of effector cells to these antibodies results in phagocytosis and cell lysis [39]. Both cetuximab and panitumumab have been shown to induce ADCC in the HNSCC cell lines. Cytotoxicity assays showed that in the presence of peripheral blood mononuclear cells and either cetuximab or panitumumab, levels of lactate dehydrogenase were significantly higher than compared to controls, indicating greater levels of cell lysis [39]. This finding suggests that ADCC represents a possible mechanism by which monoclonal antibodies exert their antitumor activity. The Fc gammaR constant regions on effector cells also contribute to the mechanism of in vivo cytotoxicity. Mice deficient for Fc gamma receptor IIB maintained a protective effect while mice deficient in Fc gamma receptor III showed an increase in tumor activity upon treatment with monoclonal antibodies [44]. In addition, antibody efficacy was enhanced by polymorphisms in the Fc receptor region, in the setting of rituximab treatment for lymphoma [45]. The Fc gamma RIIIa 158 valine/valine and the Fc gamma RIIa 131 histidine/histidine polymorphisms were independently associated with outcome [45]. In addition, more polymorphisms that seemed to bestow greater response in patients with metastatic colorectal cancer are the Fc gamma IIIa V/F and IIa V/R polymorphisms [27]. Other mechanisms involved in the efficacy of monoclonal antibodies comes from examining the effect of inhibiting human adenocarcinoma cell lines with C225. Work performed by the Mendelsohn lab show that G1 cell cycle arrest is induced upon administration of C225 to DiFi cells. This cell cycle arrest is also dependent on p27Kip1 [46].

4. MONOCLONAL ANTIBODIES

4.1. Clinical Data

Monoclonal antibodies represent a substantial subset of current targeted therapy toward EGFR and have exhibited clinical benefit. Cetuximab (C225, erbitux), a chimeric, mouse-human IgG1 monoclonal antibody, was approved by the FDA in 2006 in combination with radiation for the treatment of HNSCC. More recently, cetuximab plus chemotherapy was shown to be effective in recurrent/metastatic HNSCC [47]. Patients were randomized to receive either platinum plus fluorouracil or platinum, fluorouracil, and cetuximab. Median progression-free survival was 5.6 months in the treatment group vs. 3.3 months in the group that did not receive cetuximab. Based on promising results in two phase I and II trials [48-49], the clinical development of, cetuximab, was rapidly tested in three phase III trials [47, 50-51]. While phase III studies demonstrated improved clinical responses when cetuximab was added to radiation [51] or chemotherapy [47, 50], the combination of radiation and cetuximab significantly enhanced HNSCC survival. This led to rapid FDA approval of cetuximab for the treatment of newly diagnosed HNSCC [50-51]. A definitive randomized phase III clinical trial is underway to investigate cetuximab plus chemoradiation therapy (RTOG 0522).

Ongoing studies are testing the benefit of adding cetuximab to concurrent chemoradiation therapy (CRT) compared to chemoradiation alone, due to the promise of combined cetuximab and chemotherapy [51-52]. In addition, trials are evaluating the benefit of administering cetuximab prior radiation therapy as a radiosensitizing approach [53]. Panitumumab, a fully humanized IgG2 monoclonal antibody, is being evaluated in phase II clinical trials to determine its clinical benefit, as it has shown much promise in metastastic colorectal cancer (ClinicalTrials.gov NCT00547157). Zalitumumab and nimotuzumab are also being investigated in the context of combination therapies for HNSCC, as these may have enhanced ADCC properties (Zalitumumab) and unique binding sites (nimotuzumab) (Table 1).

Table 1. EGFR monoclonal antibodies in HNSCC clinical trials.

Agent Phas
e
Stage Therapy Reference Uniquenes
s of
Agent/Stu
dy
Cetuximab I Advanced solid
tumors
Cetuximab plus
bortezomib
(proteasome
inhibitor)
[97] Preclinical
data
suggests
enhanced
effect of
combining
these two
agents
Cetuximab II Recurrent/Metast
atic
Cetuximab +
BIBW2992
(ErbB2 TKI)
ClinicalTrials.gov
NCT00514943
Comparing
C225 vs.
TKI in
platinum
refractory
patient
Cetuximab III Recurrent/Metast
atic
Cisplatin vs.
Cisplatin plus
cetuximab
[50] Prior
clinical
efficacy
shown in
combined
therapy
Cetuximab II Resectable Stage
III/IV
Cetuximab plus
carboplatin,
placlitaxel
[98]
Cetuximab II Stage III/IV Cetuximab plus
cisplatin/docetaxe
l plus radiation
ClinicalTrials.gov
NCT00084318
Cetuximab II Stage III/IV Cetuximab plus
radiation and
chemotherapy
ClinicalTrials.gov
NCT00646659
Hu-Max
(Zalitumum
ab)
I/II Recurrent/Metast
atic
Monotherapy ClinicalTrials.gov
NCT00093041
Possible
antineiplast
ic and
ADCC
mechanism
s
demonstrat
ed by the
antibody
Zalitumum
ab
Nimotuzum
ab
II Localy
Advanced
Nimotuzumab
plus
cisplatin/radiothe
rapy
ClinicalTrials.gov
NCT00702481
Nimotuzu
mab
demonstrat
es unique
heavy
chain
sequences
and
exhibits a
good safety
profile
Panitumum
ab
I Locally
Advanced
HNSCC
Panitumumab
plus induction
chemotherapy
[99] Has shown
promice in
metastatic
colorectal
cancer
Cetuximab
and
Erlotinib
I/II Advanced (Phase
I)
Cetuximab plus
erlotinib
ClinicalTrials.gov
NCT00397384
Cetuximab III Stage III/IV Cetuximab plus
Radiotherapy vs
radiotherapy
alone
[100]

Despite ubiquitous EGFR expression in HNSCC tumors, only a subset of patients will respond to EGFR-directed antibody therapy. Elucidation of potential mechanisms of response and/or resistance may facilitate more effective use of these therapies. EGFR expression has not generally served as a marker of clinical response. One Phase III clinical trial found that the combination of cetuximab plus cisplatin improved response rates when compared to cisplatin alone in HNSCC. Clinical responses were greater for patients with EGFR staining in less than 80% of their tumor cells [50]. Harari and colleagues generated a cetuximab-resistant HNSCC cell line (UM-SCC1) in vitro and found that these cells expressed higher levels of Her2 and Her3 as well as increased hereodimerization of Her2 or Her3 with EGFR [28]. Downmodulating the expression of these Her family members with small molecules restored cetuximab sensitivity [28]. Trials of pan-HER inhibitors are underway in HNSCC.

The EGFRvIII variant contains a truncated ligand-binding domain, which may influence its binding to monoclonal antibodies [54]. EGFRvIII expressing tumors have shown decreased in vitro response to cetuximab [33]. EGFRvIII expression is therefore a plausible mechanism of cetuximab resistance. In an EGFRvIII model, xenografts were resistant to cetuximab treatment [33]. Additional models of EGFR antibody resistance are needed that mimic the clinical scenario to more fully elucidate mechanisms of acquired and de novo resistance in HNSCC. While the structure of EGFR may present some challenges for targeted therapy, a novel monoclonal antibody, mAb806, has been raised that is active against wild type EGFR as well as the EGFRvIII variant [55]. mAb806 recognizes an epitope that is present in cells that overexpress wild type EGFR and EGFRvIII [55]. Murine mouse models show that mAb806 results in dramatic tumor regression in lung cancer models compared with cetuximab [55]

5. TYROSINE KINASE INHIBITORS

5.1. Preclinical Data and Mechanisms

Tyrosine kinase inhbitors (TKIs) cause an abrogation of EGFR signaling through preventing the phosphorylation events necessary for downstream signaling. When ligand is bound to EGFR, the intracellular domain unfolds. Domain II is exposed and forms a bridge with the domain II of another member of the ErbB family. This dimerization results in the transphosphorylation of tyrosine residues in the activation loop (A-loop) in the C-terminal portion of EGFR [56]. There is also conformational change in the A-loop that reduces the steric hindrance associated with ligand binding. Two important interactions take place. There is the interaction between Tyr845 and Glu848, important for activation loop conformation [56]. In addition, there is a hydrogen-bond interaction between Tyr845 and Arg812, which is critical for the catalytic machinery responsible for transfer of a phosphate [56]. Furthermore, there is a three amino acid sequence, Leu-Val-Ile (LVI) sequence near the carboxy-terminus that is hypothesized to regulate transphosphorylation events [56]. After LVI sequence regulation, the catalytic domains on either side of the kinase domain (N and C lobes) interact [56]. This interaction is dependent on binding of ATP in the ATP cleft located between the N and C lobes at K745 [56-57]. After initial tyrosine phosphorylation, there is subsequent phosphorylation of docking sites [56]. Adaptor proteins that contain an SH2 domain can then associate with phosphotyrosine residues located on the carboxy-terminal portion of EGFR on these docking sites. Tyrosine kinase inhibitors like gefitinib and erlotinib are anilinoquinazolines which bind reversibly to the K745 site in the ATP binding pocket [57].

Mutations in the tyrosine kinase domain of EGFR are rare in patients with HSNCC (1% of Caucasian and 7% of Asian individuals) [33]. However, tyrosine kinase mutations are frequently seen in NSCLC and have offered much insight into the structure and function of EGFR. One common mutation seen in NSCLC patients that bestows sensitivity to TKIs is the L858R mutation [58-59]. This mutation is a gain of function mutation and also located in a group of amino acids near the ATP binding cleft known to regulate binging of TKIs [59]. It is hypothesized that the disruption of these critical residues results in the stabilization of their interaction with ATP and with gefitinib [59]. Retrospective studies have been performed to evaluate tumor response to gefitinib or erlotinib in the context of the L858R and exon 19 mutations (which also has been shown to confer sensitivity) [60]. Results show that clinical response was better and time to progression was longer for those patients that harbored the exon 19 deletion [60]. One possible explanation is that exon 19 deletions result in greater inhibition by gefitinib and erlotinib, but such conclusions are not supported by in vitro data [60]. Most of the activating mutations have been shown to be in exons 18-21 in NSCLC [61].

Other mutations, however, result in resistance to tyrosine kinase inhibitors. The T790M mutation is a common mutation seen in gefitinib and erlotinib resistant NSCLC; this mutation has been shown to change the conformation of the ATP binding site such that these drugs have an extremely low binding affinity [58]. The D761Y mutation,, is located in the alpha-C-helix of EGFR adjacent to E762 [58]. E762 forms a salt bridge with L745 that interacts with ATP and may explain why the D761Y mutation confers the resistant phenotype [58].

Using in vitro models of HNSCC, the decrease in activation of EGFR through tyrosine kinase inhibitors results in decreased growth. The proliferation of HNSCC cell lines is reduced following treatement of the preclinical EGFR-specific tyrosine kinase inhibitor PD153035 [13]. In addition to decreased growth, tyrosine kinase inhibitors abrogate the activation of downstream effectors of EGFR, including Akt and Erk, which serve as markers of cell survival and proliferation [62]. Downmodulation of EGFR kinase activity through erlotinib-treated cell lines also leads to the induction of apoptosis [1]. In tumors harvested from mouse xenograft models, gefitinib was shown to decrease markers of proliferation, such as Akt, Erk, and Ki67 [63].

Preclinical HNSCC animal models demonstrated a modest decrease in tumor volume after 40 days of EGFR TKI treatment when compared to control [64]. Combination treatment using gefitinib and IFN-alpha (an antitumor cytokine) resulted in marked decreases in tumor volume when compared to gefitinib or IFN-alpha alone or polyethylene glycol treated control groups. Other studies using the EGFR-specific tyrosine kinase inhibitor CP-358,774 reported significant reduction in tumor volume when compared to controls [65].

5.2. Clinical Data

Clinical trials to date have reported modest benefits with the use of EGFR tyrosine kinase inhibitors in HNSCC. A multicenter phase II trial to evaluate the effects of erlotinib on recurrent/metastatic oral squamous cell carcinoma showed an overall response rate of 4.3% and disease stabilization for a median of 16.1 weeks in 38.3% of patients [66]. Median overall survival was 6.0 months [66]. Other phase II clinical trials have showed similar responses using single agent therapy [67]. A phase II study examining the combination of radiotherapy and gefitinib in HNSCC reported 32% complete remission and 53% partial remission [68]. However a phase III trial demonstrated no survival advantage when gefitnib (Iressa) was combined with methotrexate for the treatment of recurrent/metastatic HNSCC [69]. While the clinical benefit of EGFR TKI in HNSCC is met with limited success, to date ongoing studies will determine the role of these agents in HNSCC. For example, clinical trials are evaluating combination studies using combination tyrosine kinase inhibitor and monoclonal antibody in HNSCC (Table 2).

Table 2. EGFR tyrosine kinase inhibitors (TKI) in HNSCC clinical trials.

Agent Phase Stage Therapy Reference
Cetuximab and
Erlotinib
I/II Advanced (Phase I) Cetuximab
plus erlotinib
ClinicalTrials.gov
NCT00397384
Erlotinib I/II Early Stage (I/II) Erlotinib plus
celecoxib
[101]
Erlotinib I/II Locally Advanced
HNSCC
Erlotinib plus
Bevacizumab
(VEGF
monoclonal
antibody)
[102]
Erlotinib II Stage III/IV Erlotinib plus
docetaxel
plus
Radiation
therapy
[103]
Gefitinib I Advanced/Recurrent Gefitinib plus
paclitaxel and
radiation
therapy
[104]
Gefitinib/ZD1839 I Locoregional,
recurrent
Radiotherapy
plus cisplatin
ClinicalTrials.gov
NCT00185835
Lapatinib I/II Locally Advanced Lapatinib
plus
combined
chemotherapy
and radiation
therapy
[105]

5.3. Monoclonal Antibodies vs. Tyrosine Kinase Inhibitors

Several mechanisms may explain the differences in clinical efficacy of EGFR monoclonal antibodies compared with tyrosine kinase inhibitors. Tyrosine kinase inhibitors do not induce ADCC or internalization of receptor. As a result, the immune system of the host is not harnessed and there is no decrease in surface expression of EGFR. In addition, monoclonal antibodies may demonstrate greater efficacy in HNSCC may be due to the lack of tyrosine kinase mutations in this cancer [33]. Tyrosine kinase inhibitors have been shown to be very effective in decreasing tumor burden in non-small cell lung carcinoma (NSCLC) in patients who harbor mutations in the tyrosine kinase domain of EGFR [70].These mutations allow for greater sensitivity and increased response to tyrosine kinase inhibitors [70]. However, tyrosine kinase mutations of EGFR in HNSCC are exceedingly rare [33]. As a result, one would not expect the increased sensitivity to tyrosine kinase inhibitors in HNSCC such as that seen in NSCLC. In addition, many HNSCC cell lines demonstrate IC50 values above the 10 micromolar range [71]. This suggests that these cell lines are relatively resistant to tyrosine kinase inhibitors. The dose required to cause downregulation of EGFR through tyrosine kinase inhibitors in patients may not be practical.

6. ANTISENSE OLIGONUCLEOTIDES AND siRNA

6.1. Preclinical studies and mechanism

Antisense oligonucleotides against EGFR are in the early stage of clinical investigation based on promising preclinical findings. Antisense oligonucleotides consist of a stretch of a single stranded DNA molecule of ~20 nucleotides in length. They exert their anti-tumor effect through binding of mRNA, sterically hindering ribosomes and preventing translation [72]. Unmodified antisense oligonucleotides are susceptible to degradation by endonucleases and exonucleases. Chemical modification of oligonucleotides with phosphorothioate moieties enhances resistance to nuclease degradation [73]. Cloning of the antisense oligonucleotide into a gene expression construct allows for the delivery of increased concentrations of the antisense sequence into the tumor. Systemic delivery however, is relatively challenging when compared to other modes of targeted agents (Table 3).

Table 3. Comparision of Potential Advantages and Disadvantages of EGFR Targeting Strategies.

Advantages Disadvantages
Monoclonal Antibodies
  • High specificity for target

  • Induces antibody-dependent cell mediated cytotoxicity (ADCC)

  • Mild side effects

  • Size of monoclonal antibodies may inhibit tissue penetration and clearance

  • IV administration

Tyrosine Kinase Inhibitors
  • Oral administration

  • Mild side effects

  • Low specificity for target, resulting in off target effects

Antisense
Oligonucleotides/Gene
Therapy
  • High specificity for target

  • Systemic administration a challenge

  • Delivery via cationic polymers can be toxic

Ligand-Toxin Conjugates
  • Sustained activity even if patient is immunocompromised

  • May be immunogenic

  • Hepatotoxicity common

The ability of antisense therapy to reduce translation is illustrated by preclinical studies downregulating EGFR in HNSCC. Xenograft studies demonstrated that tumors treated with EGFR antisense gene therapy were significantly smaller than vector treated controls [74]. Furthermore, EFGR-antisense treated tumors demonstrated decreased EGFR expression and increased apoptosis through staining for DNA fragmentation [74]. An antisense strategy has also been used to target the EGFR ligand, TGF-alpha [75]. HNSCC xenografts treated with TGF-alpha antisense DNA plus liposomes showed an inhibition of tumor growth when compared to controls [75]. Moreover, this antitumor effect was further supported by decreases in Bcl-XL, an apoptotic protein [75]. The mechanisms of antisense oligonucleotides and TGF-alpha-specific antibodies may be similar [75]. DNA synthesis decreases upon treatment of HNSCC and ovarian cancer cell lines with TGF-alpha antibodies [75].

EGFR antisense approaches may be enhanced by combinations with chemotherapy. Targeting EGFR protein production with antisense oligonucleotides and docetaxel in a HNSCC xenograft model resulted in downmodulation of EGFR, Akt, and Ki67 expression, as well as significant decreases in tumor volume when compared to those mice treated with docetaxel alone [76]. The increased response of this combined approach may result from the use of several methods to target EGFR, as docetaxel has also been shown to decrease EGFR expression [40-41]. While more conventional strategies to inhibit EGFR including antibodies and kinase inhibitors result in a response in about 5-10% of patients, their limited efficacy might stem from receptor turnover and a lack of full blockade of the receptor [76]. The addition of antisense approaches may be a way to circumvent these barriers to treatment.

siRNA strategeies are emerging as potential approaches to target gene expression. siRNA consists of double-stranded RNA, containing a sequence necessary to silence the translation of a target protein [77]. When the stand of RNA enters cells, it is fragmented by the enzyme Dicer, which creates 20 bp double stranded RNA molecules with 2-nucleotide 3′ overhangs [78]. These strands of RNA bind to the RNA-induced silencing complex (RISC), unwind, and then are guided by RISC to its complementary mRNA strand [79]. The RISC complex contains slicer, which cleaves target mRNA [80]. Preclinical studies using siRNA have shown that it can significantly decrease EGFR expression compared to scrambled siRNA-transfected controls, as evaluated by flow cytometry [15]. In addition, HNSCC xenografts treated with EGFR siRNA and cisplatin revealed a significant decrease in tumor burden when compared to controls [15].

While preclinical studies using siRNA to downregulate EGFR expression in HNSCC are limited, others have used this strategy in glioblastoma cell lines and xenograft mouse models. Glioblastoma, like HNSCC, has been shown to be associated with overexpression of EGFR [81]. EGFR siRNA-transfected glioma cells proliferated less than vector-transfected controls [82]. Furthermore, glioma xenografts treated with EGFR siRNA demonstrated a significant reduction in tumor volume (p<0.01) when compared to controls [82]. One interesting advantage that siRNA may have over conventional EGFR targeted therapies is that siRNA can be designed to specifically decrease EGFRvIII expression [82]. Several studies using siRNA engineered to the Exon1/Exon8 junction, reported decreased Akt levels and cell cycle arrest [82-83].

6.2. Clinical studies

While early phase clinical trials of antisense oligonucleotide therapy against Bcl-2 demonstrated that it was well tolerated and able to downregulate the expression of Bcl-2, no significant therapeutic benefit was demonstrated in phase III studies and this treatment failed to receive FDA approval [84]. We have shown that in a Phase I study, antisense oligonucleotides, when delivered using an expression construct, are well-tolerated and exhibit antitumor activity in patients. In a cohort of twenty patients, no grade 3 or 4 toxicities were reported. In addition, median survival was 5.4 months [85]. The disease-control group, as defined by complete remission, partial remission, or stable disease, demonstrated a median survival of 7.9 months as compared to 3.4 months for the partial disease group [85]. Another clinical trial is currently in progress investigating antisense oligonucleotide therapy in combination with radiation and cetuximab in HNSCC (ClinicalTrials.gov NCT00903461). A phase II trial will sonn open to investigate the effects of adding EGFR antisense gene therapy to cetuximab plus radiation for advanced locoregional HNSCC. (ClinicalTrials.gov NCT00903461)

Phase I studies are also being conducted using siRNA in solid tumors, directed towards the M2 subunit of ribonucleotide reductase, a well-known cancer target. SiRNA is also being investigated in a Phase I study for metastatic melanoma, targeting the immunoproteasome subunits LMP2, LMP7, and MECL1 (ClinicalTrials.gov NCT00672542. However, clinical studies are yet to be performed using siRNA to target EGFR in the context of HNSCC.

7. LIGAND AND ANTIBODY-TOXIN CONJUGATES

7.1. Preclinical Studies

Other strategies involve targeting EGFR with conjugated toxin plus anti-EGFR antibody or a toxin conjugated to an EGFR ligand. Ligand-toxin conjugates exert their anti-tumor effects by binding to their targets leading to the internalization of the toxin. Once internalized, the toxin ADP-ribosylates proteins involved in translation [86]. Toxins, such as a modified version of the Pseudomonas toxin, alpha PE-38, have been conjugated to TGF-alpha and have demonstrated efficacy in a glioblastoma xeongraft model [87]. Moreover, antibodies conjugated to Pseudomonas toxin have shown to inhibit the growth of HNSCC in vitro and in vivo. An advantage of ligand-toxin conjugates compared with monoclonal antibodies is the variable nature of the target antibodies. Antibodies can be subject to decreased internalization to antibody-target complexes, variable expression of tumor antigens, and limited cytotoxicity [87]. TGF-alpha conjugated to alpha-PE38, was used in HNSCC models, where cleaved PARP, a marker of apoptosis, was significantly higher in HNSCC cells that received treatment as opposed to controls [88]. Mice treated with this toxin-ligand conjugate intratumorally, exhibited a decrease in tumor volume and increased numbers of tumor cells when compared to controls [88]. Other preclinical studies have investigated bivatuzumab mertansine, an antibody-toxin conjugate directed against CD44v6, a HNSCC tumor-associated cell-surface protein. In this mode of therapy, bivatuzumab, a humanized IgG1 was conjugated to mertansine, a member of the vinca alkaloids and an inhibitor of microtubule assembly [89]. In HNSCC xenografts, bivatuzumab mertansine showed dose-dependent antitumor efficacy and stable regression of tumor when compared to controls [89]. The efficacy of immunotoxins have also been shown to be effective in Her2-positive breast cancer. Conjugating trastuzumab with the microtubule-depolarizing agent maytansinoid resulted in inhibition of proliferation in trastuzumab-resistant breast cancer cell lines [90]. Furthermore, tumor regression using this approach was evident in mouse models of breast cancer [90].

7.2. Clinical studies

Ligand-toxin conjugates have demonstrated some clinical efficacy and safety in phase I trials for glioblastoma multiforme [91].The adverse effects associated with therapy were not due to the toxin itself, but to pharmacokinetic considerations including infusion volume, tumor, or catheter placement for delivery of drug [91]. Two out of fifteen patients with radiographic evidence of glioblastoma showed some response to treatment Clinical trials using this approach are few in HNSCC. One Phase I study investigating the safety of bivatuzumab mertansine in HNSCC revealed that ten percent of participants experienced an adverse event and 29 out of 32 patients experienced a serious adverse event [89]. While the study had to be terminated prematurely, preliminary investigation shows partial response in 3 patients, including the restoration of the ability to swallow [89].

8. EXPERT OPINION

EGFR is overexpressed and aberrantly activated in the majority of HNSCC tumors. To date, EGFR expression and/or gene amplification in the tumor has not been a consistent predictor of response to EGFR targeted therapies [92]. Current treatment strategies using EGFR inhibitors generally combine EGFR targeting agents with standard approaches including with chemotherapy and/or radiation. Targeting EGFR in combination with with inhibitors of other cellular signaling pathways including Src family kinases, STAT3, Met, and IGF-1R, among others, in HNSCC preclinical models, has shown promise [93-96]. Monoclonal antibodies targeting EGFR have been most extensively studied and were FDA-approved for use in HNSCC in 2006. EGFR-specific tyrosine kinase efforts are under active investivation. Current research efforts are focused on prospectively identifying those individuals who are likely to benefit from EGFR targeting strategies. Additional preclinical HNSCC models of EGFR inhibitor resistance (both de novo and acquired) are needed to further elucidate mechanisms of resistance to EGFR targeting agents and facilitate the development of more effective approaches. Most importantly, tumor biopsies from patients treated with EGFR inhibitors are required to determine both the effect of these agents on the human HNSCC tumor and determine the characteristics of tumors that fail to respond to EGFR targeting agents, despite expression of this growth factor receptor.

Article Highlights.

  • -

    EGFR is upregulated in HNSCC. The ability of EGFR gene expression levels to predict response to therapy are currently being studied.

  • -

    Monoclonal antibodies, tyrosine kinase inhibitors, antisense therapy, and ligand-toxin conjugates have all shown promise in preclinical studies and are under active investigation.

  • -

    Combination therapies of targeted agents and of targeted agents with conventional chemotherapy and radiation are currently in clinical trials.

  • -

    Developing the most efficient targeted therapy modality involves increased understanding of the mechanisms involved in EGFR-mediated carcinogenesis.

Acknowledgements

This work was supported by NIH grants T32DC000066 and P50CA097190 and an American Cancer Society Clinical Research Professorship (to JRG)

Declaration of interest

The authors are funded by NIH grants T32000066 and P50CA097190 and an American Cancer Society Clinical Research Professorship to J.R. Grandis.

REFERENCES

  • 1.Zhang Q, et al. Antitumor mechanisms of combined gastrin-releasing peptide receptor and epidermal growth factor receptor targeting in head and neck cancer. Mol Cancer Ther. 2007;6:1414–1424. doi: 10.1158/1535-7163.MCT-06-0678. [DOI] [PubMed] [Google Scholar]
  • 2.Yoo GH, et al. Effect of Docetaxel on the Surgical Tumor Microenvironment of Head and Neck Cancer in Murine Models. Arch Otolaryngol Head Neck Surg. 2008;134(7):735–742. doi: 10.1001/archotol.134.7.735. [DOI] [PubMed] [Google Scholar]
  • 3.Parkin DM, et al. Global Cancer Statistics, 2002. CA Cancer J Clin. 2005;55(2):74–108. doi: 10.3322/canjclin.55.2.74. [DOI] [PubMed] [Google Scholar]
  • 4.Mork J, et al. Human Papillomavirus Infection as a Risk Factor for Squamous-Cell Carcinoma of the Head and Neck. N Engl J Med. 2001;344(15):1125–1131. doi: 10.1056/NEJM200104123441503. [DOI] [PubMed] [Google Scholar]
  • 5.Furniss CS, et al. Human papillomavirus 16 and head and neck squamous cell carcinoma. International Journal of Cancer. 2007;120(11):2386–2392. doi: 10.1002/ijc.22633. [DOI] [PubMed] [Google Scholar]
  • 6.Khuri FR, et al. The Impact of Smoking Status, Disease Stage, and Index Tumor Site on Second Primary Tumor Incidence and Tumor Recurrence in the Head and Neck Retinoid Chemoprevention Trial. Cancer Epidemiology Biomarkers & Prevention. 2001;10(8):823–829. [PubMed] [Google Scholar]
  • 7.Cohen EEW, Lingen MW, Vokes EE. The Expanding Role of Systemic Therapy in Head and Neck Cancer. J Clin Oncol. 2004;22(9):1743–1752. doi: 10.1200/JCO.2004.06.147. [DOI] [PubMed] [Google Scholar]
  • 8.Psyrri A, et al. Cisplatin, Fluorouracil, and Leucovorin Induction Chemotherapy Followed by Concurrent Cisplatin Chemoradiotherapy for Organ Preservation and Cure in Patients With Advanced Head and Neck Cancer: Long-Term Follow-Up. J Clin Oncol. 2004;22(15):3061–3069. doi: 10.1200/JCO.2004.01.108. [DOI] [PubMed] [Google Scholar]
  • 9.Kalyankrishna S, Grandis JR. Epidermal Growth Factor Receptor Biology in Head and Neck Cancer. J Clin Oncol. 2006;24(17):2666–2672. doi: 10.1200/JCO.2005.04.8306. [DOI] [PubMed] [Google Scholar]
  • 10.Grandis JR, Tweardy DJ. Elevated Levels of Transforming Growth Factor {alpha} and Epidermal Growth Factor Receptor Messenger RNA Are Early Markers of Carcinogenesis in Head and Neck Cancer. Cancer Res. 1993;53(15):3579–3584. [PubMed] [Google Scholar]
  • 11.Grandis J, et al. Levels of TGF-alpha and EGFR protein in head and neck squamous cell carcinoma and patient survival. J. Natl. Cancer Inst. 1998;90(11):824–832. doi: 10.1093/jnci/90.11.824. [DOI] [PubMed] [Google Scholar]
  • 12.Ang KK, et al. Impact of Epidermal Growth Factor Receptor Expression on Survival and Pattern of Relapse in Patients with Advanced Head and Neck Carcinoma. Cancer Res. 2002;62(24):7350–7356. [PubMed] [Google Scholar]
  • 13.Grandis JR, et al. Inhibition of epidermal growth factor receptor gene expression and function decreases proliferation of head and neck squamous carcinoma but not normal mucosal epithelial cells. Oncogene. 1997;15:409–416. doi: 10.1038/sj.onc.1201188. [DOI] [PubMed] [Google Scholar]
  • 14.Erjala K, et al. Signaling via ErbB2 and ErbB3 Associates with Resistance and Epidermal Growth Factor Receptor (EGFR) Amplification with Sensitivity to EGFR Inhibitor Gefitinib in Head and Neck Squamous Cell Carcinoma Cells. Clinical Cancer Research. 2006;12(13):4103–4111. doi: 10.1158/1078-0432.CCR-05-2404. [DOI] [PubMed] [Google Scholar]
  • 15.Nozawa H, et al. Small interfering RNA targeting epidermal growth factor receptor enhances chemosensitivity to cisplatin, 5-fluorouracil and docetaxel in head and neck squamous cell carcinoma. Cancer Science. 2006;97(10):1115–1124. doi: 10.1111/j.1349-7006.2006.00287.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Seethala RR, et al. Immunohistochemical Analysis of Phosphotyrosine Signal Transducer and Activator of Transcription 3 and Epidermal Growth Factor Receptor Autocrine Signaling Pathways in Head and Neck Cancers and Metastatic Lymph Nodes. Clinical Cancer Research. 2008;14(5):1303–1309. doi: 10.1158/1078-0432.CCR-07-1543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Thomas SM, et al. Cross-talk between G Protein-Coupled Receptor and Epidermal Growth Factor Receptor Signaling Pathways Contributes to Growth and Invasion of Head and Neck Squamous Cell Carcinoma. Cancer Res. 2006;66(24):11831–11839. doi: 10.1158/0008-5472.CAN-06-2876. [DOI] [PubMed] [Google Scholar]
  • 18.Alhasan SA, Aranha O, Sarkar FH. Genistein Elicits Pleiotropic Molecular Effects on Head and Neck Cancer Cells. Clinical Cancer Research. 2001;7(12):4174–4181. [PubMed] [Google Scholar]
  • 19.Baselga J, Swain SM. Novel anticancer targets: revisiting ERBB2 and discovering ERBB3. Nat Rev Cancer. 2009;9(7):463–475. doi: 10.1038/nrc2656. [DOI] [PubMed] [Google Scholar]
  • 20.Yotsumoto F, et al. Validation of HB-EGF and amphiregulin as targets for human cancer therapy. Biochemical and Biophysical Research Communications. 2008;365(3):555–561. doi: 10.1016/j.bbrc.2007.11.015. [DOI] [PubMed] [Google Scholar]
  • 21.Dawson JP, Bu Z, Lemmon MA. Ligand-Induced Structural Transitions in ErbB Receptor Extracellular Domains. Structure. 2007;15(8):942–954. doi: 10.1016/j.str.2007.06.013. [DOI] [PubMed] [Google Scholar]
  • 22.Ferguson KM. Structural aspects of extracellular EGFR signaling. FASEB J. 2009;23:198.3. 1_MeetingAbstracts. [Google Scholar]
  • 23.Schmitz KR, Ferguson KM. Interaction of antibodies with ErbB receptor extracellular regions. Experimental Cell Research. 2009;315(4):659–670. doi: 10.1016/j.yexcr.2008.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Li S, Kussie P, Ferguson KM. Structural Basis for EGF Receptor Inhibition by the Therapeutic Antibody IMC-11F8. Structure. 2008;16(2):216–227. doi: 10.1016/j.str.2007.11.009. [DOI] [PubMed] [Google Scholar]
  • 25.Zhang X, et al. An Allosteric Mechanism for Activation of the Kinase Domain of Epidermal Growth Factor Receptor. 2006;125(6):1137–1149. doi: 10.1016/j.cell.2006.05.013. [DOI] [PubMed] [Google Scholar]
  • 26.Schulze WX, Deng L, Mann M. Phosphotyrosine interactome of the ErbB-receptor kinase family. Mol Syst Biol. 2005;1 doi: 10.1038/msb4100012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Zhang Q, et al. Phosphorylation of TNF-α converting enzyme by gastrin-releasing peptide induces amphiregulin release and EGF receptor activation. PNAS. 2006;103(18):6901–6906. doi: 10.1073/pnas.0509719103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Wheeler DL, et al. Mechanisms of acquired resistance to cetuximab: role of HER (ErbB) family members. Oncogene. 2008;27(28):3944–3956. doi: 10.1038/onc.2008.19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Chung CH, et al. Increased Epidermal Growth Factor Receptor Gene Copy Number Is Associated With Poor Prognosis in Head and Neck Squamous Cell Carcinomas. J Clin Oncol. 2006;24(25):4170–4176. doi: 10.1200/JCO.2006.07.2587. [DOI] [PubMed] [Google Scholar]
  • 30.Loeffler-Ragg J, et al. Low incidence of mutations in EGFR kinase domain in Caucasian patients with head and neck squamous cell carcinoma. European Journal of Cancer. 2006;42(1):109–111. doi: 10.1016/j.ejca.2005.08.034. [DOI] [PubMed] [Google Scholar]
  • 31.Frederick BA, et al. Epithelial to mesenchymal transition predicts gefitinib resistance in cell lines of head and neck squamous cell carcinoma and non–small cell lung carcinoma. Molecular Cancer Therapeutics. 2007;6(6):1683–1691. doi: 10.1158/1535-7163.MCT-07-0138. [DOI] [PubMed] [Google Scholar]
  • 32.Chung CH, et al. Gene Expression Profiles Identify Epithelial-to-Mesenchymal Transition and Activation of Nuclear Factor-{kappa}B Signaling as Characteristics of a High-risk Head and Neck Squamous Cell Carcinoma. Cancer Res. 2006;66(16):8210–8218. doi: 10.1158/0008-5472.CAN-06-1213. [DOI] [PubMed] [Google Scholar]
  • 33.Sok JC, et al. Mutant Epidermal Growth Factor Receptor (EGFRvIII) Contributes to Head and Neck Cancer Growth and Resistance to EGFR Targeting. Clinical Cancer Research. 2006;12(17):5064–5073. doi: 10.1158/1078-0432.CCR-06-0913. [DOI] [PubMed] [Google Scholar]
  • 34.Matar P, et al. Combined Epidermal Growth Factor Receptor Targeting with the Tyrosine Kinase Inhibitor Gefitinib (ZD1839) and the Monoclonal Antibody Cetuximab (IMC-C225) Clinical Cancer Research. 2004;10(19):6487–6501. doi: 10.1158/1078-0432.CCR-04-0870. [DOI] [PubMed] [Google Scholar]
  • 35.Zhang Q, et al. Src Family Kinases Mediate Epidermal Growth Factor Receptor Ligand Cleavage, Proliferation, and Invasion of Head and Neck Cancer Cells. Cancer Res. 2004;64(17):6166–6173. doi: 10.1158/0008-5472.CAN-04-0504. [DOI] [PubMed] [Google Scholar]
  • 36.Yoshida T, et al. The anti-EGFR monoclonal antibody blocks cisplatin-induced activation of EGFR signaling mediated by HB-EGF. FEBS Letters. 2008;582(30):4125–4130. doi: 10.1016/j.febslet.2008.11.010. [DOI] [PubMed] [Google Scholar]
  • 37.Mandic R, et al. Treatment of HNSCC cell lines with the EGFR-specific inhibitor cetuximab (Erbitux®) results in paradox phosphorylation of tyrosine 1173 in the receptor. FEBS Letters. 2006;580(20):4973–4800. doi: 10.1016/j.febslet.2006.07.064. [DOI] [PubMed] [Google Scholar]
  • 38.Friedman LM, et al. Synergistic down-regulation of receptor tyrosine kinases by combinations of mAbs: Implications for cancer immunotherapy. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(6):1915–1920. doi: 10.1073/pnas.0409610102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lopez-Albaitero A, Ferris RL. Immune Activation by Epidermal Growth Factor Receptor Specific Monoclonal Antibody Therapy for Head and Neck Cancer. Arch Otolaryngol Head Neck Surg. 2007;133(12):1277–1281. doi: 10.1001/archotol.133.12.1277. [DOI] [PubMed] [Google Scholar]
  • 40.Bocci G, et al. Increased Plasma Vascular Endothelial Growth Factor (VEGF) as a Surrogate Marker for Optimal Therapeutic Dosing of VEGF Receptor-2 Monoclonal Antibodies. Cancer Res. 2004;64(18):6616–6625. doi: 10.1158/0008-5472.CAN-04-0401. [DOI] [PubMed] [Google Scholar]
  • 41.Ebos JML, et al. Multiple circulating proangiogenic factors induced by sunitinib malate are tumor-independent and correlate with antitumor efficacy. Proceedings of the National Academy of Sciences. 2007;104(43):17069–17074. doi: 10.1073/pnas.0708148104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Almand B, et al. Clinical Significance of Defective Dendritic Cell Differentiation in Cancer. Clinical Cancer Research. 2000;6(5):1755–1766. [PubMed] [Google Scholar]
  • 43.Mickel RA, et al. Natural Killer Cell Cytotoxicity in the Peripheral Blood, Cervical Lymph Nodes, and Tumor of Head and Neck Cancer Patients. Cancer Res. 1988;48(17):5017–5022. [PubMed] [Google Scholar]
  • 44.Clynes RA, et al. Inhibitory Fc receptors modulate in vivo cytoxicity against tumor targets. Nat Med. 2000;6(4):443–446. doi: 10.1038/74704. [DOI] [PubMed] [Google Scholar]
  • 45.Weng W-K, Levy R. Two Immunoglobulin G Fragment C Receptor Polymorphisms Independently Predict Response to Rituximab in Patients With Follicular Lymphoma. J Clin Oncol. 2003;21(21):3940–3947. doi: 10.1200/JCO.2003.05.013. [DOI] [PubMed] [Google Scholar]
  • 46.Wu X, Fan Z, DeBlasio T, Soos T, Koff A, Mendelsohn J. Involvement of p27KIP1 in G1 arrest mediated by an anti-epidermal growth factor receptor monoclonal antibody. Oncogene. 1996;12(7):1397–403. R.M. [PubMed] [Google Scholar]
  • 47.Vermorken JB, et al. Platinum-Based Chemotherapy plus Cetuximab in Head and Neck Cancer. N Engl J Med. 2008;359(11):1116–1127. doi: 10.1056/NEJMoa0802656. [DOI] [PubMed] [Google Scholar]
  • 48.Shin DM, et al. Epidermal Growth Factor Receptor-targeted Therapy with C225 and Cisplatin in Patients with Head and Neck Cancer. Clinical Cancer Research. 2001;7(5):1204–1213. [PubMed] [Google Scholar]
  • 49.Robert F, et al. Phase I Study of Anti-Epidermal Growth Factor Receptor Antibody Cetuximab in Combination With Radiation Therapy in Patients With Advanced Head and Neck Cancer. J Clin Oncol. 2001;19(13):3234–3243. doi: 10.1200/JCO.2001.19.13.3234. [DOI] [PubMed] [Google Scholar]
  • 50.Burtness B, et al. Phase III Randomized Trial of Cisplatin Plus Placebo Compared With Cisplatin Plus Cetuximab in Metastatic/Recurrent Head and Neck Cancer: An Eastern Cooperative Oncology Group Study. J Clin Oncol. 2005;23(34):8646–8654. doi: 10.1200/JCO.2005.02.4646. [DOI] [PubMed] [Google Scholar]
  • 51.Bonner JA, et al. Radiotherapy plus Cetuximab for Squamous-Cell Carcinoma of the Head and Neck. N Engl J Med. 2006;354(6):567–578. doi: 10.1056/NEJMoa053422. [DOI] [PubMed] [Google Scholar]
  • 52.Bonner JA, et al. Enhanced Apoptosis With Combination C225/Radiation Treatment Serves as the Impetus for Clinical Investigation in Head and Neck Cancers. J Clin Oncol. 2000;18(suppl_1):47s–53. [PubMed] [Google Scholar]
  • 53.Bonner JA, Harari PM, Cohen R, Jones C, Sur RK, Rabin D, Azarnia N, Needle MN, Ang KK. Journal of Clinical Oncology, 2004 ASCO Annual Meeting Proceedings (Post-Meeting Edition) 2004 July 15;22(14S):5507. J.G. Supplement. [Google Scholar]
  • 54.Huether A, et al. EGFR blockade by cetuximab alone or as combination therapy for growth control of hepatocellular cancer. Biochemical Pharmacology. 2005;70(11):1568–1578. doi: 10.1016/j.bcp.2005.09.007. [DOI] [PubMed] [Google Scholar]
  • 55.Li D, et al. Therapeutic anti-EGFR antibody 806 generates responses in murine de novo EGFR mutant–dependent lung carcinomas. The Journal of Clinical Investigation. 2007;117(2):346–352. doi: 10.1172/JCI30446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Stamos J, Sliwkowski MX, Eigenbrot C. Structure of the Epidermal Growth Factor Receptor Kinase Domain Alone and in Complex with a 4-Anilinoquinazoline Inhibitor. Journal of Biological Chemistry. 2002;277(48):46265–46272. doi: 10.1074/jbc.M207135200. [DOI] [PubMed] [Google Scholar]
  • 57.Pao W, et al. EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib. Proceedings of the National Academy of Sciences. 2004;101(36):13306–13311. doi: 10.1073/pnas.0405220101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Balak MN, et al. Novel D761Y and Common Secondary T790M Mutations in Epidermal Growth Factor Receptor–Mutant Lung Adenocarcinomas with Acquired Resistance to Kinase Inhibitors. Clinical Cancer Research. 2006;12(21):6494–6501. doi: 10.1158/1078-0432.CCR-06-1570. [DOI] [PubMed] [Google Scholar]
  • 59.Lynch TJ, et al. Activating Mutations in the Epidermal Growth Factor Receptor Underlying Responsiveness of Non-Small-Cell Lung Cancer to Gefitinib. N Engl J Med. 2004;350(21):2129–2139. doi: 10.1056/NEJMoa040938. [DOI] [PubMed] [Google Scholar]
  • 60.Jackman DM, et al. Exon 19 Deletion Mutations of Epidermal Growth Factor Receptor Are Associated with Prolonged Survival in Non–Small Cell Lung Cancer Patients Treated with Gefitinib or Erlotinib. Clinical Cancer Research. 2006;12(13):3908–3914. doi: 10.1158/1078-0432.CCR-06-0462. [DOI] [PubMed] [Google Scholar]
  • 61.Shigematsu H, Gazdar AF. Somatic mutations of epidermal growth factor receptor signaling pathway in lung cancers. International Journal of Cancer. 2006;118(2):257–262. doi: 10.1002/ijc.21496. [DOI] [PubMed] [Google Scholar]
  • 62.Patel V, et al. Profiling EGFR activity in head and neck squamous cell carcinoma by using a novel layered membrane Western blot technology. Oral Oncology. 2005;41(5):503–508. doi: 10.1016/j.oraloncology.2004.12.010. [DOI] [PubMed] [Google Scholar]
  • 63.Feng FY, et al. Effect of Epidermal Growth Factor Receptor Inhibitor Class in the Treatment of Head and Neck Cancer with Concurrent Radiochemotherapy In vivo. Clinical Cancer Research. 2007;13(8):2512–2518. doi: 10.1158/1078-0432.CCR-06-2582. [DOI] [PubMed] [Google Scholar]
  • 64.Bruzzese F, et al. Synergistic Antitumor Activity of Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor Gefitinib and IFN-α in Head and Neck Cancer Cells In vitro and In vivo. Clinical Cancer Research. 2006;12(2):617–625. doi: 10.1158/1078-0432.CCR-05-1671. [DOI] [PubMed] [Google Scholar]
  • 65.Pollack VA, et al. Inhibition of Epidermal Growth Factor Receptor-Associated Tyrosine Phosphorylation in Human Carcinomas with CP-358,774: Dynamics of Receptor Inhibition In Situ and Antitumor Effects in Athymic Mice. Journal of Pharmacology and Experimental Therapeutics. 1999;291(2):739–748. [PubMed] [Google Scholar]
  • 66.Soulieres D, et al. Multicenter Phase II Study of Erlotinib, an Oral Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor, in Patients With Recurrent or Metastatic Squamous Cell Cancer of the Head and Neck. J Clin Oncol. 2004;22(1):77–85. doi: 10.1200/JCO.2004.06.075. [DOI] [PubMed] [Google Scholar]
  • 67.Wheeler RH, et al. Clinical and molecular phase II study of gefitinib in patients (pts) with recurrent squamous cell cancer of the head and neck (H&N Ca) J Clin Oncol (Meeting Abstracts) 2005;23(16_suppl):5531. [Google Scholar]
  • 68.Doss HH, et al. Induction chemotherapy + gefitinib followed by concurrent chemotherapy/radiation therapy/gefitinib for patients (pts) with locally advanced squamous carcinoma of the head and neck: A phase I/II trial of the Minnie Pearl Cancer Research Network. J Clin Oncol (Meeting Abstracts) 2006;24(18_suppl):5543. [Google Scholar]
  • 69.Stewart JSW, et al. Phase III Study of Gefitinib Compared With Intravenous Methotrexate for Recurrent Squamous Cell Carcinoma of the Head and Neck. J Clin Oncol. 2009;27(11):1864–1871. doi: 10.1200/JCO.2008.17.0530. [DOI] [PubMed] [Google Scholar]
  • 70.Paez JG, et al. EGFR Mutations in Lung Cancer: Correlation with Clinical Response to Gefitinib Therapy. Science. 2004;304(5676):1497–1500. doi: 10.1126/science.1099314. [DOI] [PubMed] [Google Scholar]
  • 71.Kalish LH, et al. Deregulated Cyclin D1 Expression Is Associated with Decreased Efficacy of the Selective Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor Gefitinib in Head and Neck Squamous Cell Carcinoma Cell Lines. Clinical Cancer Research. 2004;10(22):7764–7774. doi: 10.1158/1078-0432.CCR-04-0012. [DOI] [PubMed] [Google Scholar]
  • 72.Gee JE, van der Laan AC, van Boom JH, Colombier C, Leng M, Raible AM, Nelson JS, Lebleu B. Assessment of high-affinity hybridization, RNase H cleavage, and covalent linkage in translation arrest by antisense oligonucleotides. Antisense Nucleic Acid Drug Dev. 1998 Apr;8(2):103–11. doi: 10.1089/oli.1.1998.8.103. R.I. [DOI] [PubMed] [Google Scholar]
  • 73.Griffey RH, et al. 2′-O-Aminopropyl Ribonucleotides: A Zwitterionic Modification That Enhances the Exonuclease Resistance and Biological Activity of Antisense Oligonucleotides†. Journal of Medicinal Chemistry. 1996;39(26):5100–5109. doi: 10.1021/jm950937o. [DOI] [PubMed] [Google Scholar]
  • 74.He Y, et al. Inhibition of human squamous cell carcinoma growth in vivo by epidermal growth factor receptor antisense RNA transcribed from the U6 promoter. J. Natl. Cancer Inst. 1998;90(14):1080–1087. doi: 10.1093/jnci/90.14.1080. [DOI] [PubMed] [Google Scholar]
  • 75.Endo S, Zeng Q, He Y, Melhem MF, Watkins SF, Lango MN, Drenning SD, Huang L, Grands JR. TGF-alpha antisense gene therapy inhibits head and neck squamous cell carcinoma growth in viv. Gene Therapy. 2000;7(22):1906–1914. doi: 10.1038/sj.gt.3301315. [DOI] [PubMed] [Google Scholar]
  • 76.Thomas SM, et al. Antitumor Mechanisms of Systemically Administered Epidermal Growth Factor Receptor Antisense Oligonucleotides in Combination with Docetaxel in Squamous Cell Carcinoma of the Head and Neck. Molecular Pharmacology. 2008;73(3):627–638. doi: 10.1124/mol.107.041160. [DOI] [PubMed] [Google Scholar]
  • 77.Ketting RF, Bernstein E, Sijen T, Hannon GJ, Plasterk RH. Dicer functions in RNA interference and in synthesis of small RNA involved in developmental timing in C. elegans. Genes & Dev. 2001 Oct 15;15(20):2654–9. doi: 10.1101/gad.927801. F.S. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Elbashir SM, Lendeckel W, Tuschl T. RNA interference is mediated by 21- and 22-nucleotide RNAs. Genes & Development. 2001;15(2):188–200. doi: 10.1101/gad.862301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Schwarz DS, et al. Asymmetry in the Assembly of the RNAi Enzyme Complex. Cell. 2003;115(2):199–208. doi: 10.1016/s0092-8674(03)00759-1. [DOI] [PubMed] [Google Scholar]
  • 80.Song J-J, et al. Crystal Structure of Argonaute and Its Implications for RISC Slicer Activity. Science. 2004;305(5689):1434–1437. doi: 10.1126/science.1102514. [DOI] [PubMed] [Google Scholar]
  • 81.Libermann TA, et al. Amplification, enhanced expression and possible rearrangement of EGF receptor gene in primary human brain tumours of glial origin. Nature. 1985;313(5998):144–147. doi: 10.1038/313144a0. [DOI] [PubMed] [Google Scholar]
  • 82.Kang CS, et al. Suppression of EGFR expression by antisense or small interference RNA inhibits U251 glioma cell growth in vitro and in vivo. Cancer Gene Ther. 2006;13(5):530–538. doi: 10.1038/sj.cgt.7700932. [DOI] [PubMed] [Google Scholar]
  • 83.Fan Q-W, Weiss WA. RNA interference against a glioma-derived allele of EGFR induces blockade at G2M. Oncogene. 2004;24(5):829–837. doi: 10.1038/sj.onc.1208227. [DOI] [PubMed] [Google Scholar]
  • 84.Morris MJ, et al. Phase I Trial of BCL-2 Antisense Oligonucleotide (G3139) Administered by Continuous Intravenous Infusion in Patients with Advanced Cancer. Clinical Cancer Research. 2002;8(3):679–683. [PubMed] [Google Scholar]
  • 85.Lai SY, et al. Intratumoral Epidermal Growth Factor Receptor Antisense DNA Therapy in Head and Neck Cancer: First Human Application and Potential Antitumor Mechanisms. J Clin Oncol. 2009;27(8):1235–1242. doi: 10.1200/JCO.2008.17.8251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Bachran C, et al. Quantification of Diphtheria Toxin Mediated ADP-Ribosylation in a Solid-Phase Assay. Clin Chem. 2007;53(9):1676–1683. doi: 10.1373/clinchem.2007.085365. [DOI] [PubMed] [Google Scholar]
  • 87.Phillips PC, et al. Transforming Growth Factor-{alpha} Pseudomonas Exotoxin Fusion Protein (TGF-{alpha}-PE38) Treatment of Subcutaneous and Intracranial Human Glioma and Medulloblastoma Xenografts in Athymic Mice. Cancer Res. 1994;54(4):1008–1015. [PubMed] [Google Scholar]
  • 88.Thomas SM, et al. Abrogation of Head and Neck Squamous Cell Carcinoma Growth by Epidermal Growth Factor Receptor Ligand Fused to Pseudomonas Exotoxin Transforming Growth Factor α-PE38. Clinical Cancer Research. 2004;10(20):7079–7087. doi: 10.1158/1078-0432.CCR-04-0587. [DOI] [PubMed] [Google Scholar]
  • 89.Riechelmann H, Sauter A, Golze W, Hanft G, Schroen C, Hoermann K, Erhardt T, Gronau S. Phase I trial with the CD44v6-targeting immunoconjugate bivatuzumab mertansine in head and neck squamous cell carcinoma. Oral Oncology. 2007;44(9):823–829. doi: 10.1016/j.oraloncology.2007.10.009. [DOI] [PubMed] [Google Scholar]
  • 90.Lewis Phillips GD, et al. Targeting HER2-Positive Breast Cancer with Trastuzumab-DM1, an Antibody-Cytotoxic Drug Conjugate. Cancer Res. 2008;68(22):9280–9290. doi: 10.1158/0008-5472.CAN-08-1776. [DOI] [PubMed] [Google Scholar]
  • 91.Sampson JH, Archer GE, Bigner DD, Berger MS, Friedman AH, Friedman HS, Herndon JE, 2nd, Kunwar S, Marcus S, McLendon RE, Paolino A, Penne K, Provenzale J, Quinn J, Reardon DA, Rich J, Stenzel T, Tourt-Uhlig S, Wikstrand C, Wong T, Williams R, Yuan F, Zalutsky MR, Pastan I. Progress report of a Phase I study of the intracerebral microinfusion of a recombinant chimeric protein composed of transforming growth factor (TGF)-alpha and a mutated form of the Pseudomonas exotoxin termed PE-38 (TP-38) for the treatment of malignant brain tumors. J Neurooncol. 2003 Oct;65(1):27–35. doi: 10.1023/a:1026290315809. A.G. [DOI] [PubMed] [Google Scholar]
  • 92.Hitt R, et al. Prognostic value of the epidermal growth factor receptor (EGRF) and p53 in advanced head and neck squamous cell carcinoma patients treated with induction chemotherapy. European Journal of Cancer. 2005;41(3):453–460. doi: 10.1016/j.ejca.2004.10.014. [DOI] [PubMed] [Google Scholar]
  • 93.Koppikar P, et al. Combined Inhibition of c-Src and Epidermal Growth Factor Receptor Abrogates Growth and Invasion of Head and Neck Squamous Cell Carcinoma. Clinical Cancer Research. 2008;14(13):4284–4291. doi: 10.1158/1078-0432.CCR-07-5226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Leeman-Neill RJ, et al. Guggulsterone enhances head and neck cancer therapies via inhibition of signal transducer and activator of transcription-3. Carcinogenesis. 2009;30(11):1848–1856. doi: 10.1093/carcin/bgp211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Buck E, et al. Feedback Mechanisms Promote Cooperativity for Small Molecule Inhibitors of Epidermal and Insulin-Like Growth Factor Receptors. Cancer Res. 2008;68(20):8322–8332. doi: 10.1158/0008-5472.CAN-07-6720. [DOI] [PubMed] [Google Scholar]
  • 96.Seiwert TY, et al. The MET Receptor Tyrosine Kinase Is a Potential Novel Therapeutic Target for Head and Neck Squamous Cell Carcinoma. Cancer Res. 2009;69(7):3021–3031. doi: 10.1158/0008-5472.CAN-08-2881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Dudek A, et al. Phase I study of bortezomib plus cetuximab in patients with tumors expressing epidermal growth factor receptor (EGFR) J Clin Oncol (Meeting Abstracts) 2007;25(18_suppl):18143. [Google Scholar]
  • 98.Wanebo HJ, et al. Phase II evaluation of cetuximab (C225) combined with induction paclitaxel and carboplatin followed by C225, paclitaxel, carboplatin, and radiation for stage III/IV operable squamous cancer of the head and neck (ECOG, E2303) J Clin Oncol (Meeting Abstracts) 2007;25(18_suppl):6015. [Google Scholar]
  • 99.Wirth LJ, et al. Phase I study of panitumumab, chemotherapy and intensity-modulated radiotherapy (IMRT) for head and neck cancer (HNC): Early results. J Clin Oncol (Meeting Abstracts) 2007;25(18_suppl):6083. [Google Scholar]
  • 100.Bonner JA, et al. Cetuximab prolongs survival in patients with locoregionally advanced squamous cell carcinoma of head and neck: A phase III study of high dose radiation therapy with or without cetuximab. J Clin Oncol (Meeting Abstracts) 2004;22(14_suppl):5507. [Google Scholar]
  • 101.Altorki NK, et al. Celecoxib, a Selective Cyclo-Oxygenase-2 Inhibitor, Enhances the Response to Preoperative Paclitaxel and Carboplatin in Early-Stage Non-Small-Cell Lung Cancer. J Clin Oncol. 2003;21(14):2645–2650. doi: 10.1200/JCO.2003.07.127. [DOI] [PubMed] [Google Scholar]
  • 102.Cohen EEW, et al. Erlotinib and bevacizumab in patients with recurrent or metastatic squamous-cell carcinoma of the head and neck: a phase I/II study. The Lancet Oncology. 2009;10(3):247–257. doi: 10.1016/S1470-2045(09)70002-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Savvides P, et al. Phase I study of the EGFR tyrosine kinase inhibitor erlotinib in combination with docetaxel and radiation in locally advanced squamous cell cancer of the head and neck (SCCHN) J Clin Oncol (Meeting Abstracts) 2006;24(18_suppl):5545. [Google Scholar]
  • 104.Sharp H, Morris JC, Van Waes C, Gius D, Cooley-Zgela T, Singh AK. High Incidence of Oral Dysesthesias on a Trial of Gefitinib, Paclitaxel, and Concurrent External Beam Radiation for Locally Advanced Head and Neck Cancers. America Journal of Clinical Oncology. 2008;31(6) doi: 10.1097/COC.0b013e318172d5de. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Specenier PM, et al. EORTC 24051: Unexpected side effects of a phase I study of TPF induction chemotherapy (IC) followed by chemoradiation (CRT) with lapatinib (LAP), a dual EGFR/ErbB2 inhibitor, in patients with locally advanced larynx and hypopharynx squamous cell carcinoma (LA-LxHxSCC) J Clin Oncol (Meeting Abstracts) 2009;27(15S):6017. doi: 10.1016/j.radonc.2012.08.006. [DOI] [PubMed] [Google Scholar]

RESOURCES