Context Malignancies arising from the aerodigestive epithelium, including lung, head and neck, and esophageal carcinomas, are the leading causes of cancer-related mortality worldwide. Given the biological importance of epidermal growth factor receptor (EGFR) in cancer development and progression, EGFR inhibitors have emerged as promising novel therapies.
Objectives To summarize the current status of EGFR inhibitors in aerodigestive carcinomas (ADCs), highlight ongoing research designed to optimize their therapeutic effectiveness, and consider the future role of these agents.
Evidence Acquisition Systematic MEDLINE search of English-language literature (1966-April 2007) performed using the terms EGFR, EGFR inhibitors, monoclonal antibodies, tyrosine kinase inhibitors, lung cancer, head and neck cancer, esophageal cancer, and EGFR predictive factors. Quality assessment of selected studies included clinical pertinence, with an emphasis on controlled study design, publication in peer-reviewed journals, adequate number of enrolled patients, objectivity of measurements, and techniques used to minimize bias.
Evidence Synthesis The role of EGFR in ADC pathogenesis has been extensively studied, and multiple EGFR inhibition strategies are under evaluation. Erlotinib, an EGFR tyrosine kinase inhibitor used as a single agent, and cetuximab, an anti-EGFR monoclonal antibody used in combination with radiation, have conferred survival benefit in 1 trial of patients with advanced non–small cell lung cancer (median survival, 6.7 vs 4.7 months; hazard ratio, 0.70; 95% confidence interval, 0.58-0.87; P < .001) and in 1 trial of patients with locally advanced head and neck squamous cell carcinoma (median survival, 49 vs 29.3 months; hazard ratio, 0.74; 95% confidence interval, 0.57-0.97; P = .03), respectively. However, other trials have not shown these degrees of improvement. EGFR inhibitors toxicities include rash, diarrhea, and hypomagnesemia. Somatic mutations and other molecular tumoral characteristics offer opportunities for treatment individualization and optimal patient selection for anti-EGFR therapy.
Conclusions EGFR is a promising therapeutic target in ADC. Further translational research is needed to optimize ways of inhibiting EGFR using single-agent or combination regimens and to identify patients who benefit the most from these therapies.
Quiz Ref IDCancers that originate from the aerodigestive epithelium, including carcinomas of the head and neck, lung, and esophagus (referred to as aerodigestive carcinomas [ADCs]), are the leading causes of cancer-related mortality worldwide, accounting for about 2 million deaths annually.1 Their pathogenesis is causally linked with exposure to carcinogens, mainly tobacco and alcohol, and they are usually diagnosed at advanced stages, in which available treatments have limited curative potential. Even small advances made in the management of ADC might affect millions of lives. The cornerstone of the current era of therapeutics is the rational development of anticancer agents that has emerged from the in-depth understanding of cancer biology. Seminal features of cancer that relate to cell growth, replication potential, signaling pathways, cell-to-cell interactions, angiogenesis, and other factors are being elucidated and successfully exploited for therapeutic purposes, thus enabling accelerated development of novel compounds.
Quiz Ref IDEpithelial carcinogenesis is the phenotypic outcome of serial genetic and epigenetic deviations resulting in the deregulation of cellular homeostasis.2 Accumulated evidence suggests that membrane receptor signaling pathways may participate in the acquisition of malignant phenotype. Epidermal growth factor receptor (EGFR) is a type I tyrosine kinase (TK) membrane receptor that regulates key cellular functions in epithelial malignancies.3 Multiple studies that used various methods have demonstrated that EGFR is commonly overexpressed in ADCs, typically more in squamous cell carcinomas than in adenocarcinomas; EGFR protein expression is detected in about 50% or more of non–small cell lung cancers (NSCLCs),4-7 more than 90% of head and neck squamous cell carcinomas (HNSCCs),8 and 30% to 70% of esophageal carcinomas.9-11 High levels of EGFR protein expression have been correlated with worse patient survival in NSCLC, although the association has not been robust and results not consistent between studies,12-18 HNSCC,8,19 and esophageal carcinoma.10,11,20 Moreover, a high EGFR gene copy number, which variably correlates with EGFR protein expression, also has been reported as a poor prognostic marker in ADC.5,21 Therefore, EGFR represents a rational target for ADC therapeutics.
Although blockade of EGFR function can be accomplished by several methods (Table 1), 2 approaches have been most extensively studied in the laboratory and clinic: monoclonal antibodies (mAbs) directed against the extracellular receptor domain, and small-molecule TK inhibitors of the EGFR intracellular domain (EGFR-TKIs). Agents of both categories have been incorporated into the standard management of ADC. However, their clinical application has generated a number of challenges.22 Foremost, the identification of molecular predictors of outcome that may potentially optimize patient selection and therapeutic efficacy remains the subject of intense ongoing basic and clinical investigation. Here, we will summarize the current status of clinical testing with EGFR inhibitors in ADC, highlight ongoing research regarding molecular predictive factors, and consider the future perspectives of these agents.
We performed a MEDLINE search of the English-language literature (1966 to April 2007) using the terms EGFR, EGFR inhibitors, monoclonal antibodies, tyrosine kinase inhibitors, lung cancer, head and neck cancer, esophageal cancer, and EGFR predictive factors. Relevant bibliographies of literature were manually reviewed for additional material. In evaluating the benefits of EGFR inhibitors in ADC, data from randomized trials were emphasized. Quality assessment of selected studies also included publication in peer-reviewed journals, adequate number of enrolled patients, objectivity of measurements, and techniques used to minimize bias. On review of clinical trials, clinical end points of focus, in decreasing order of importance, were survival benefit, progression-free survival, response rate, disease stabilization rate, and quality-of-life improvement. Further information was obtained in oral and abstract form from the 2004-2006 American Society of Clinical Oncology and the 2004-2007 American Association for Cancer Research meetings. Published National Comprehensive Cancer Network and American Society of Clinical Oncology guidelines also were reviewed. Information on ongoing clinical trials was derived from the US National Institutes of Health Web site (http://www.clinicaltrials.gov).
EGFR (ERBB1) is a member of the ERBB growth factor receptor TK family, which also includes ERBB2, ERBB3, and ERBB4. These receptors possess an extracellular ligand-binding domain, a transmembrane-anchoring region, and an intracellular domain that carries the TK activity, with the exception of ERBB3. The binding of a ligand such as epidermal growth factor or transforming growth factor α causes the EGFR to dimerize with itself or with another member of the ERBB family.23 EGFR dimerization causes activation of the receptor-linked TK, recruitment of signaling complexes, and phosphorylation (activation) of multiple downstream cascades.24
The EGFR-activated networks include the RAS/mitogen-activated protein kinase,25 phospatidylinositol-3-kinase/AKT,26 Janus kinase/signal transducers and activators of transcription (JAK/STAT),27 and phospholipase-Cγ/protein kinase C pathways28 (Figure). These cascades are potent regulators of a variety of intracellular and intercellular processes such as proliferation, apoptosis, invasion, angiogenesis, and metastasis. EGFR activation also can be achieved through cross-talk with other receptors, such as G-protein–coupled receptors,29 platelet-derived growth factor receptor,30 insulin-like growth factor receptor 1,31 and hormone receptors32 (Figure).
An important regulatory component of the cell surface EGFR consists of a 2-step endocytosis process.33 First, a rapid ligand-dependent internalization removes activated receptors from the cell membrane and enclaves them in endosomes. Second, internalized receptors either are targeted to lysosomes, where they undergo degradation, or are recycled back to the plasma membrane. EGFR sorting for lysosomal degradation requires receptor autophosphorylation, which then recruits Cbl—a ubiquitin E3 ligase responsible for EGFR ubiquitination24 (Figure). Ligand-induced receptor endocytosis generally down-regulates EGFR signaling, even though there is some evidence that the internalized receptors may retain their capability of coupling effector proteins and activating signaling cascades.34
The pathological manifestations of EGFR in ADC are achieved by at least 4 major mechanisms: (1) overexpression of EGFR ligands and establishment of autocrine/paracrine loops, (2) mutational activation of EGFR, (3) amplification of EGFR, and (4) transactivation by other receptor and nonreceptor TKs.23,35 Two main categories of EGFR mutations have been identified: deletion mutations of the extracellular domain and somatic mutations in the TK activity of intracellular domain. Several classes of EGFR ectodomain mutations (vI-vVII) have been discovered, although EGFRvIII is the one most commonly found in ADC. It was recently shown that EGFRvIII in HNSCC leads to constitutive ligand-independent receptor activation, enhanced downstream effects, and resistance to wild-type EGFR targeting.36 Activating mutations of EGFR-TK domain are more commonly found in NSCLCs compared with other ADCs.37-40 Deletions in exon 19 and nucleotide substitutions in exon 21 are the most common EGFR mutations in NSCLCs. These mutations affect the adenosine triphosphatebinding pocket of the EGFR-TK domain and endow the receptor with unopposed TK activity. Tumors harboring EGFR mutations respond better to EGFR-TKIs, suggesting that they are dependent on mutant EGFR signaling. However, it should be noted that although EGFR-driven molecular pathway aberrations are important, they cannot be considered the dominant sole genetic alteration in the molecular pathogenesis of ADC.41
Quiz Ref IDEGFR signaling is important for a variety of normal tissues, including those of the skin, gut, and kidney, which explains the development of certain toxicities from EGFR targeting, such as rash, diarrhea, and hypomagnesemia. For example, acneiform rash and other skin toxicities are encountered in the majority of patients who receive EGFR inhibitors.42 Moreover, EGFR blockade may interfere with magnesium transport, possibly due to the strong expression of EGFR in the ascending limb of the loop of Henle, where most of the filtered magnesium is reabsorbed, which could explain the high frequency of hypomagnesemia during cetuximab treatment.43,44
Strategies of EGFR Inhibition
Various strategies have been developed to disrupt the EGFR signal transduction pathway (Table 1). Among them, anti-EGFR mAbs and EGFR-TKIs have undergone the most extensive investigation. Putative mechanisms of EGFR mAb–based anticancer activity can be classified into 2 categories. First, they prevent ligands from binding to the EGFR extracellular domain, inhibit subsequent receptor dimerization/activation, and finally induce receptor degradation.45 The second potential mechanism of EGFR mAb therapy is indirect action mediated by the immune system. There are unique immune-effector mechanisms that have been found to be triggered by therapeutic mAbs, such as antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity, and complement-dependent cell-mediated cytotoxicity.46,47 An alternative mode of targeting EGFR is by conjugating anti-EGFR mAbs with toxins to selectively attack tumor cells overexpressing EGFR.48
Receptor and nonreceptor TKs are critical mediators in EGFR signaling pathways, and many are deregulated during ADC tumorigenesis. Small-molecule inhibitors that target these TKs are directly acting in tumor cells rather than mediating immune responses. Most small-molecule TK inhibitors are adenosine triphospate mimetics, thus competing (reversibly or irreversibly) with adenosine triphosphate binding within the catalytic kinase cleft. Unlike mAbs, they cross the plasma membrane and interact with the cytoplasmic domain of cell-surface receptors or modulate intracellular signaling molecules. Tyrosine kinase inhibitors have a variable selectivity for TKs, and some are dual-selective or multiselective (Table 1). EGFR-TKIs are generally thought to be less specific than mAbs, which is potentially advantageous for antitumor activity but may be associated with increased toxicities due to the inhibition of several signaling pathways that potentially interfere with normal cell functions. A number of newer-generation TK inhibitors show a broad-spectrum activity against various receptor TKs (eg, EGFR, vascular endothelial growth factor [VEGF] receptor), nonreceptor TKs (eg, SRC kinase), and/or downstream molecules (eg, RAS/mitogen-activated protein kinase).49
The development of new EGFR inhibitors has also been attempted using high-throughput screening approaches, rational drug design, or a combination of both. High-throughput screening of compounds is a way of identifying novel EGFR inhibitors that can then be further modified by medicinal chemistry approaches to develop drugs that potentially can be used in tumors with acquired resistance to current anti-EGFR agents.50 Structure-based rational approaches have been used for the design of nucleic acid–directed gene-silencing molecules, eg, antisense oligodeoxynucleotides, RNA interference, and ribozymes, that prevent EGFR messenger RNA (mRNA) translation and are still in early phases of investigation.51 Antisense technology uses a sequence complementary to a specific mRNA target (eg, EGFR), inhibiting its expression and the transfer of the genetic information from DNA to the protein level. RNA interference has been recently recognized as a powerful mechanism for silencing gene expression by targeted degradation of mRNA. Short double-stranded RNAs, known as small interfering RNAs and designed to have homology to sections of the mRNA of the target protein, can be introduced in the cytoplasm and trigger hydrolysis of the mRNA through the RNA interference pathway. Preclinical studies using these techniques have demonstrated specific inhibition of tumor cell growth as well as synergistic inhibitory effects with conventional chemotherapeutic drugs.52 However, nucleic acid–directed therapies need to be further investigated in patients with ADC to determine their antitumor activity, optimal delivery methods, and toxicity.
Lung Cancer. Lung cancer is the leading cause of cancer death worldwide; most patients develop and die of distant metastatic disease.1 Newer cytotoxic agents developed over the past decade have only marginally affected survival. Thus, there is an immense need for the development of novel systemic therapies that could improve patient outcomes. Advances in the understanding of the molecular pathogenesis of lung cancer have allowed the identification of potential targets for selective therapeutic intervention. Agents targeting EGFR were the first to be successfully tested in patients with advanced NSCLC.53,54
The EGFR-TKIs gefitinib and erlotinib achieved responses of 10% to 20%, respectively, in phase 2 clinical trials in patients with previously treated, advanced NSCLC.55-57 Based on these observations, 2 randomized, double-blind, placebo-controlled phase 3 trials were initiated to evaluate the overall survival with EGFR-TKIs compared with best supportive care alone in patients with advanced NSCLC who had received first- or second-line chemotherapy (Table 2). In the first study, patients were randomized to receive either erlotinib (150 mg orally, once daily) or placebo. Overall survival was 6.7 months for patients in the erlotinib group compared with 4.7 months in patients in the control group (P < .001).58 Analysis of quality of life showed that patients in the erlotinib group had longer time to deterioration of tumor-related symptoms.68
In the second trial, gefitinib at a daily dose of 250 mg did not improve overall survival compared with placebo in a similar cohort of patients with NSCLC, even though subset analysis showed a survival benefit in Asians and nonsmokers.61 The reasons for the discrepancy of the results of these 2 similarly designed clinical trials are unclear. The possibility that gefitinib was given at a suboptimal biological dose has been suggested as a plausible explanation and is also supported by preclinical observations.69 Whether gefitinib dose escalation beyond 500 mg would improve its clinical efficacy remains to be demonstrated in clinical studies.70 Gefitinib also produced negative results as adjuvant monotherapy after combined-modality therapy with chemotherapy and radiotherapy in a phase 3 trial in patients with stage III NSCLC64 (Table 2). Although a phase 3 study of adjuvant gefitinib in patients with surgically resected NSCLC was prematurely terminated, the use of erlotinib as adjuvant or maintenance therapy is currently under investigation (Table 3).
Four phase 3 trials evaluated an EGFR-TKI, erlotinib or gefitinib, in combination with standard first-line chemotherapy in patients with advanced NSCLC59,60,62,63 (Table 2). Quiz Ref IDNo advantage could be elicited with the addition of an EGFR-TKI to chemotherapy in survival or any other efficacy end point in these trials, in spite of preclinical work on human tumor xenografts supporting such combinations.53 It has been postulated that EGFR-TKIs and chemotherapy would be more efficacious when administered sequentially, not simultaneously, and this concept is undergoing clinical evaluation71 (Table 3). However, it is likely that the lack of advantage found in the phase 3 trials evaluating EGFR inhibitors in combination with chemotherapy was due to the lack of patient selection, stratification based on molecular tumor characteristics, or both. For example, the design of these studies did not take into consideration the presence of EGFR-activating mutations, EGFR gene copy number, or both, which recently emerged as predictors of EGFR-TKI efficacy. Thus, small benefits may have been obscured due to the molecular heterogeneity of the study population.
The anti-EGFR antibodies, mainly cetuximab, also have been studied in NSCLC as single agents and in combination with chemotherapy. In 1 study in 66 patients with previously treated, advanced NSCLC, cetuximab monotherapy yielded a low response rate (4.5%) but a promising median overall survival of 8.9 months.72 A number of ongoing phase 3 randomized trials are evaluating the addition of cetuximab to chemotherapy in the first- and second-line treatment of advanced NSCLC (Table 3).
Head and Neck Cancer. EGFR is important in the pathogenesis of HNSCC, and its overexpression has been reported in more than 80% of cases.23 Moreover, EGFR up-regulation also has been documented in the normal-appearing epithelium adjacent to malignant tissue, thus supporting the “field cancerization” concept, which refers to the multifocal development of premalignant and malignant lesions within the entire carcinogen-exposed epithelium.73 Therefore, inhibition of EGFR signaling represents a rational new strategy in HNSCC therapeutics.
Several TK inhibitors with preclinical activity against EGFR have been tested in HNSCC. Many single-group phase 2 trials using gefitinib or erlotinib have shown modest single-agent activity (responses of 1%-11%) in patients with recurrent or metastatic HNSCCs,74-77 whereas lapatinib, a novel dual EGFR and ERBB2 inhibitor, was not active in the same setting.78 Recently, promising preliminary results of clinical trials evaluating the combination of gefitinib or erlotinib with chemotherapeutic agents or other biological agents (eg, bevacizumab) and radiotherapy have been reported.79 Moreover, 2 phase 3 studies have evaluated gefitinib in recurrent or metastatic HNSCC. The first is an international multicenter trial that compared gefitinib (at either a 250-mg or a 500-mg daily dose) with methotrexate and that recently reported results showing that gefitinib is not superior to methotrexate in this setting65 (Table 2); the second is an ongoing study that compares docetaxel with or without gefitinib as first- or second-line treatment79 (Table 3).
Cetuximab was the first novel agent to obtain regulatory approval in the United States for the treatment of patients with HNSCC. This was based on positive survival data from a phase 3 randomized clinical trial that compared radiation and cetuximab with radiation alone in patients with locally advanced HNSCC67 (Table 2). The improvement in locoregional control and survival achieved with the addition of cetuximab to radiation was comparable to that achieved with platinum-based regimens. However, no randomized trial has yet compared radiation plus cetuximab with radiation plus cisplatin. Instead, a main focus of investigation has been in further reinforcing treatment by adding cetuximab to platinum-based chemoradiotherapy. Multiple phase 2 studies have been completed or are ongoing evaluating combinations of radiation, cetuximab, and platinum compounds. A recently reported phase 2 study evaluated the efficacy and toxicity of the combination of cetuximab with cisplatin and accelerated boost radiotherapy, with promising preliminary efficacy. However, the study was prematurely closed because of significant adverse events, including 2 deaths that may have not been specifically related to cetuximab.80 A phase 3 trial is currently comparing this regimen to radiation and cisplatin alone (Table 3).
Cetuximab has also been extensively studied in the treatment of recurrent or metastatic HNSCC. The Eastern Cooperative Oncology Group conducted a randomized study that compared cisplatin with or without cetuximab for the first-line treatment of recurrent or metastatic HNSCC.66 Although this study had a small sample size of 117 analyzable patients, it showed a significant improvement in response rates and a trend toward improved median progression-free survival with the addition of cetuximab (Table 2). Cetuximab has also shown efficacy in patients with platinum-refractory HNSCC, either as a single agent,81 which formed the basis for regulatory approval in the United States for this indication as well, or in combination with platinum.82,83 A recently completed phase 3 trial investigated the combination of a platinum agent (cisplatin or carboplatin) and 5-fluorouracil with or without cetuximab in recurrent or metastatic HNSCC84 (Table 3).
Esophageal Cancer. Squamous cell carcinoma of the esophagus remains a common disease1; however, the incidence of adenocarcinoma of the esophagus is gradually increasing. These 2 entities may be distinct in their etiology, pathogenesis, and prognosis.85 Several studies have reported EGFR protein expression of 30% to 70% in esophageal carcinomas, which is more common in squamous cell carcinomas than in adenocarcinomas9-11 and has been correlated with poor patient prognosis and inferior response to conventional treatment. EGFR amplification has been documented in approximately 15% of esophageal carcinomas,9,86 whereas EGFR mutations are rarely seen (0%-11%).9,39,87,88 Phase 2 studies using EGFR-TKIs as monotherapy in the first- and second-line setting in unselected patients with advanced esophageal cancer showed modest activity,86,89 while the combination of TK inhibitors or mAbs with chemotherapy, radiotherapy, or both is currently under evaluation.90,91
Predictive Markers of Anti-EGFR Therapy
Clinical Predictive Factors. Clinical trials with EGFR-TKIs in patients with NSCLC have identified a number of characteristics that predict increased benefit with EGFR-TKIs, such as no prior smoking history, female sex, adenocarcinoma or bronchioloalveolar carcinoma (BAC) histology, and Asian ethnicity.54 The molecular basis of these associations is not always apparent. Also, the distinction between prognostic and predictor factors can be potentially made only in randomized trials that included a treatment group without an EGFR inhibitor.92 The higher probability of response to EGFR-TKIs in never smokers with NSCLC has been a robust and particular intriguing observation that may be partially related to the higher frequency of EGFR mutations in this group.93 Moreover, patients with pure BAC or with adenocarcinoma having BAC features appear to derive substantial benefit from EGFR-TKIs, as demonstrated in phase 2 trials,94,95 though confirmatory data from phase 3 trials are not yet available.
It also has been consistently shown that the onset and severity of skin rash due to EGFR-TKIs and mAbs correlates with patient outcome across many tumor types, including HNSCC and NSCLC.96 The skin toxicities associated with EGFR inhibitors are attributable to the prominent role of EGFR in skin physiology.42 An interesting hypothesis is that patients with certain polymorphisms in intron 1 of the EGFR gene may have a higher likelihood of a favorable tumor response as well as of developing skin toxicities with EGFR inhibitor treatment.97
Molecular Predictive Markers. EGFR Mutations. Activating somatic mutations in the EGFR-TK domain were first described in NSCLC.37,38 It is noteworthy that these mutations are present in a fraction of patients with NSCLC that varies with race or ethnicity (approximately 10% in unselected populations in the United States but markedly higher in East Asians), smoking history, sex, and histology.98,99 Similar EGFR-TK mutations have been detected but appear to be less frequent in HNSCCs (1%-7%)40,100 and esophageal carcinomas (0%-11%).9,39,87,88 The presence of activating EGFR mutations appears to be a prognostic factor associated with prolonged survival in patients with advanced NSCLCs treated with chemotherapy with or without an EGFR-TKI.92 Moreover, in multiple studies of NSCLCs,101-103 but not in a placebo-controlled randomized trial of advanced NSCLC treated with erlotinib,104 EGFR mutations were predictive of survival in patients treated with EGFR-TKIs. A secondary point mutation in the EGFR-TK domain, T790M, that is associated with acquired resistance has also been reported.105
Although EGFR mutations are potential important predictors of outcome, they can only partially account for the documented clinical benefit from EGFR-TKIs in patients with NSCLC, while in other ADCs, EGFR mutations appear to be of limited significance.106 Multiple clinical trials are evaluating EGFR-TKIs as upfront treatment in chemotherapy-naive patients with advanced NSCLC and EGFR-activating mutations.107-109 Both gefitinib and erlotinib have so far produced dramatic antitumor activity, with objective response rates of 75% to 82% in this setting.107-109 A potential limitation for the prospective evaluation of EGFR mutations is the difficulty in obtaining sufficient tumor tissue for analysis in patients with inoperable NSCLC. Detection of the mutations in malignant pleural effusions is under study.110
EGFR Gene Copy Number and Polymorphisms. The EGFR gene copy number has emerged as an important predictor of the efficacy of EGFR inhibitors in NSCLC. High EGFR gene copy number, amplification, or high polysomy, as evaluated by fluorescent in-situ hybridization, has been documented in 31% to 45% of cases of NSCLC and associated with increased responsiveness and longer survival after treatment with EGFR-TKIs.104,111-113 The potential role of EGFR amplification as a prognostic factor is also being studied in HNSCC21 and squamous cell cancer of the esophagus,9 whereas data on its use as a predictor of outcome in studies with EGFR inhibitors in these cancers are awaited. The significance of genomic gain for other ERBB family members (eg, ERBB2, ERBB3) in patients treated with EGFR inhibitors is also under investigation.114,115
The regulation of EGFR expression is complex and involves multiple regulatory steps. Interindividual or ethnic polymorphic variations in the EGFR gene also can alter EGFR expression, activity, or both.116,117 Such variations also might account for the observed diverse anti-EGFR treatment efficacy and should be further investigated.118 Recently, a pharmacokinetic model was developed to assess the influence of cytochrome P450 3A activity on EGFR-TKI metabolism, antitumor activity, and toxicity.119
EGFR Protein Expression and Other Molecular Markers. High EGFR protein expression, as determined by immunohistochemistry, is observed in the vast majority of ADCs, with the highest expression seen in HNSCC.23,120 However, EGFR protein expression has not been found to consistently correlate with the antitumor activity of EGFR inhibitors.66,81,104,113,120,121 For example, mixed results have been obtained in studies that evaluated EGFR protein expression and EGFR-TKI activity in advanced NSCLC.122 The discordant results regarding the predictive value of EGFR protein expression may be partially attributed to variability in population characteristics and methodology. Nevertheless, it is suggested by analysis of phase 3 trials that patients with NSCLC negative for EGFR protein expression derive no survival benefit from EGFR-TKIs.104,113
Perturbations in the RAS/mitogen-activated protein kinase, phospatidylinositol-3-kinase/AKT, and JAK/STAT pathways, which are the major downstream effectors of EGFR signaling, are being evaluated as potential predictors of outcome.123,124 For example, it was recently shown in vitro that AKT phosphorylation without EGFR ligand stimulation has an important role in sensitivity to EGFR-TKI,125 probably due to ERBB3-mediated activation.126 NSCLC arising in smokers appears to have a different spectrum of molecular abnormalities than NSCLC in nonsmokers, suggesting differences that emerge early in carcinogenesis and may possibly relate to prognosis.127 K-RAS mutations strongly correlate with smoking history and have been associated with poor prognosis.128 Activating mutations in K-RAS and EGFR may be mutually exclusive, since they almost never occur in the same tumor.129 It appears that tumor growth can depend on activation of either pathway but not both. Accumulating preclinical and clinical evidence suggests that patients with NSCLC and K-RAS mutations may represent a distinct patient population with intrinsic resistance to anti-EGFR treatment approaches.92,130,131
Although data on predictors of efficacy are rapidly accumulating, at present there is no single biomarker that can be recommended for patient selection for anti-EGFR therapies. It is possible that an algorithm that incorporates a panel of biomarkers, including EGFR mutations, EGFR gene copy number, or EGFR protein levels may provide a useful clinical tool.
The introduction of EGFR inhibitors was one of the first steps in the development of successful molecularly targeted therapies in ADC. Certainly, optimizing strategies and learning from failures in the developmental process of these agents is critical. The molecular events implicated in intrinsic or acquired resistance to anti-EGFR therapy are yet to be fully unveiled. Quiz Ref IDOne of the potential strategies for improving antitumor effects is combined treatment with distinct anti-EGFR agents that can maximize EGFR signaling inhibition.132 Thus, the combination of EGFR-TKIs and mAbs is of interest and has entered clinical testing.133 However, anti-EGFR treatment alone may not be sufficient. Aerodigestive carcinomas usually acquire aberrations in more than 1 signaling pathway during carcinogenesis, and EGFR is only one of many targets in a complex system. Thus, EGFR targeting may rather become a component of a dual-targeting or multitargeting strategy that addresses the complexity of signaling pathways and avoids the emergence of resistance. For example, EGFR cross-talk networks with the VEGF/VEGF-receptor pathway are well established. Preclinical and clinical resistance to EGFR inhibitors has been attributed to increased VEGF levels.134 Therefore, the combination of EGFR inhibitors and antiangiogenesis agents has surfaced as an appealing novel strategy. Dual inhibitors of EGFR and VEGF receptor, such as vandetanib, also have been generated.135
Molecular profiling of ADCs has considerable potential to improve the results of anti-EGFR therapies by identifying tumors that are susceptible to this approach. Conducting clinical trials in patient populations enriched with certain molecular features is more likely to yield the desired outcomes. Moreover, tumor sampling before and after treatment may elucidate the mechanisms of action and resistance to novel agents.
The biological effect of EGFR inhibitors also has been assessed in biopsy samples of the skin, an easily accessible surrogate tissue; however, results from skin and tumor biopsies may not be concordant.136 Validation of appropriate pharmacodynamic end points that could guide dosing and response assessment remains of paramount importance for the development of anti-EGFR strategies. Furthermore, because tumor stabilization, not only tumor shrinkage, is relevant to the antitumor efficacy of EGFR inhibitors, study end points in early clinical trials that include time to progression are desirable. Finally, it might be important to consider the most appropriate treatment sequence of EGFR inhibitors and chemotherapy. It has been suggested that better results are likely to be obtained with intermittent dosing of EGFR-targeted agents when combined with chemotherapeutic agents.137 Clinical trials with EGFR-TKIs in advanced NSCLC are ongoing to test these hypotheses.
Therapies against the EGFR are becoming an integral component of the anticancer armamentarium and currently are being evaluated in a variety of clinical settings, including combinations with chemotherapy and radiotherapy for potentially curable disease. At the same time, newer EGFR inhibitors and multitargeted approaches are being explored in ADCs and other malignancies. A deeper understanding of the biology of ADCs should enable further optimization of anti-EGFR and other targeted therapies.
Corresponding Author: Athanassios Argiris, MD, Division of Hematology-Oncology, University of Pittsburgh, UPMC Cancer Pavilion, Fifth Floor, Pittsburgh, PA 15232 (argirisae@upmc.edu).
Author Contributions: Dr Karamouzis had full access to all of the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.
Study concept and design: Karamouzis, Grandis, Argiris.
Acquisition of data: Karamouzis, Grandis, Argiris.
Analysis and interpretation of data: Karamouzis, Argiris.
Drafting of the manuscript: Karamouzis, Argiris.
Critical revision of the manuscript for important intellectual content: Karamouzis, Grandis, Argiris.
Administrative, technical, or material support: Karamouzis, Grandis, Argiris.
Financial Disclosures: Dr Grandis reported receiving honoraria from Bristol-Myers Squibb and AstraZeneca and research support from the National Cancer Institute and OSI Pharmaceuticals. Dr Argiris reported serving as a consultant for Amgen; receiving research support from the National Cancer Institute and from AstraZeneca, Bristol-Myers Squibb, Genentech, Lilly Oncology, Millennium Pharmaceuticals, M's Science Corporation, Novartis, and Pfizer; and receiving honoraria from AstraZeneca and Genentech. No other disclosures were reported.
Funding/Support: This work was supported in part by the National Cancer Institute, Head and Neck SPORE: P50 CA097190-01A1, and Lung SPORE: P50 CA90440-06.
Role of the Sponsors: The funding organization had no role in the design and conduct of the study; the collection, management, analysis, and interpretation of the data; or the preparation, review, or approval of the manuscript.
1.Kamangar F, Dores GM, Anderson WF. Patterns of cancer incidence, mortality, and prevalence across five continents: defining priorities to reduce cancer disparities in different geographic regions of the world.
J Clin Oncol. 2006;24(14):2137-215016682732
Google ScholarCrossref 3.Mendelsohn J, Baselga J. Epidermal growth factor receptor targeting in cancer.
Semin Oncol. 2006;33(4):369-38516890793
Google ScholarCrossref 4.Fontanini G, Vignati S, Bigini D.
et al. Epidermal growth factor receptor (EGFr) expression in non-small cell lung carcinomas correlates with metastatic involvement of hilar and mediastinal lymph nodes in the squamous subtype.
Eur J Cancer. 1995;31A(2):178-1837718322
Google ScholarCrossref 5.Hirsch FR, Varella-Garcia M, Bunn PA Jr.
et al. Epidermal growth factor receptor in non-small-cell lung carcinomas: correlation between gene copy number and protein expression and impact on prognosis.
J Clin Oncol. 2003;21(20):3798-380712953099
Google ScholarCrossref 6.Rusch V, Baselga J, Cordon-Cardo C.
et al. Differential expression of the epidermal growth factor receptor and its ligands in primary non-small cell lung cancers and adjacent benign lung.
Cancer Res. 1993;53(10):(suppl)
2379-23857683573
Google Scholar 7.Brabender J, Danenberg KD, Metzger R.
et al. Epidermal growth factor receptor and HER2-neu mRNA expression in non-small cell lung cancer is correlated with survival.
Clin Cancer Res. 2001;7(7):1850-185511448895
Google Scholar 8.Ang KK, Berkey BA, Tu X.
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):7350-735612499279
Google Scholar 9.Hanawa M, Suzuki S, Dobashi Y.
et al. EGFR protein overexpression and gene amplification in squamous cell carcinomas of the esophagus.
Int J Cancer. 2006;118(5):1173-118016161046
Google ScholarCrossref 10.Gibault L, Metges JP, Conan-Charlet V.
et al. Diffuse EGFR staining is associated with reduced overall survival in locally advanced oesophageal squamous cell cancer.
Br J Cancer. 2005;93(1):107-11515986037
Google ScholarCrossref 11.Wang KL, Wu TT, Choi IS.
et al. Expression of epidermal growth factor receptor in esophageal and esophagogastric junction adenocarcinomas: association with poor outcome.
Cancer. 2007;109(4):658-66717211865
Google ScholarCrossref 12.Scagliotti GV, Selvaggi G, Novello S, Hirsch FR. The biology of epidermal growth factor receptor in lung cancer.
Clin Cancer Res. 2004;10(12, pt 2):4227s-4232s15217963
Google ScholarCrossref 13.Meert AP, Martin B, Delmotte P.
et al. The role of EGF-R expression on patient survival in lung cancer: a systematic review with meta-analysis.
Eur Respir J. 2002;20(4):975-98112412692
Google ScholarCrossref 14.Rusch V, Klimstra D, Venkatraman E, Pisters PW, Langenfeld J, Dmitrovsky E. Overexpression of the epidermal growth factor receptor and its ligand transforming growth factor alpha is frequent in resectable non-small cell lung cancer but does not predict tumor progression.
Clin Cancer Res. 1997;3(4):515-5229815714
Google Scholar 15.Fujino S, Enokibori T, Tezuka N.
et al. A comparison of epidermal growth factor receptor levels and other prognostic parameters in non-small cell lung cancer.
Eur J Cancer. 1996;32A(12):2070-20749014747
Google ScholarCrossref 16.Pfeiffer P, Clausen PP, Andersen K, Rose C. Lack of prognostic significance of epidermal growth factor receptor and the oncoprotein p185HER-2 in patients with systemically untreated non-small-cell lung cancer: an immunohistochemical study on cryosections.
Br J Cancer. 1996;74(1):86-918679464
Google ScholarCrossref 17.Pastorino U, Andreola S, Tagliabue E.
et al. Immunocytochemical markers in stage I lung cancer: relevance to prognosis.
J Clin Oncol. 1997;15(8):2858-28659256129
Google Scholar 18.Volm M, Rittgen W, Drings P. Prognostic value of ERBB-1, VEGF, cyclin A, FOS, JUN and MYC in patients with squamous cell lung carcinomas.
Br J Cancer. 1998;77(4):663-6699484827
Google ScholarCrossref 19.Rubin Grandis J, Melhem MF, Gooding WE.
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):824-8329625170
Google ScholarCrossref 20.Ozawa S, Ueda M, Ando N, Shimizu N, Abe O. Prognostic significance of epidermal growth factor receptor in esophageal squamous cell carcinomas.
Cancer. 1989;63(11):2169-21732720567
Google ScholarCrossref 21.Chung CH, Ely K, McGavran L.
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):4170-417616943533
Google ScholarCrossref 22.Marshall J. Clinical implications of the mechanism of epidermal growth factor receptor inhibitors.
Cancer. 2006;107(6):1207-121816909423
Google ScholarCrossref 23.Kalyankrishna S, Grandis JR. Epidermal growth factor receptor biology in head and neck cancer.
J Clin Oncol. 2006;24(17):2666-267216763281
Google ScholarCrossref 24.Citri A, Yarden Y. EGF-ERBB signalling: towards the systems level.
Nat Rev Mol Cell Biol. 2006;7(7):505-51616829981
Google ScholarCrossref 25.Adjei AA. Novel combinations based on epidermal growth factor receptor inhibition.
Clin Cancer Res. 2006;12(14, pt 2):4446s-4450s16857826
Google ScholarCrossref 26.Toker A, Yoeli-Lerner M. Akt signaling and cancer: surviving but not moving on.
Cancer Res. 2006;66(8):3963-396616618711
Google ScholarCrossref 27.Leeman RJ, Lui VW, Grandis JR. STAT3 as a therapeutic target in head and neck cancer.
Expert Opin Biol Ther. 2006;6(3):231-24116503733
Google ScholarCrossref 28.Thomas SM, Coppelli FM, Wells A.
et al. Epidermal growth factor receptor-stimulated activation of phospholipase Cgamma-1 promotes invasion of head and neck squamous cell carcinoma.
Cancer Res. 2003;63(17):5629-563514500405
Google Scholar 29.Thomas SM, Bhola NE, Zhang Q.
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):11831-1183917178880
Google ScholarCrossref 30.Ingram JL, Bonner JC. EGF and PDGF receptor tyrosine kinases as therapeutic targets for chronic lung diseases.
Curr Mol Med. 2006;6(4):409-42116900664
Google ScholarCrossref 31.Karamouzis MV, Papavassiliou AG. The IGF-1 network in lung carcinoma therapeutics.
Trends Mol Med. 2006;12(12):595-60217055338
Google ScholarCrossref 32.Gururaj AE, Rayala SK, Vadlamudi RK, Kumar R. Novel mechanisms of resistance to endocrine therapy: genomic and nongenomic considerations.
Clin Cancer Res. 2006;12(3, pt 2):1001s-1007s16467116
Google ScholarCrossref 33.Frey MR, Dise RS, Edelblum KL, Polk DB. p38 kinase regulates epidermal growth factor receptor downregulation and cellular migration.
EMBO J. 2006;25(24):5683-569217139251
Google ScholarCrossref 34.Wiley HS. Trafficking of the ErbB receptors and its influence on signaling.
Exp Cell Res. 2003;284(1):78-8812648467
Google ScholarCrossref 35.Oliveira S, van Bergen en Henegouwen PM, Storm G, Schiffelers RM. Molecular biology of epidermal growth factor receptor inhibition for cancer therapy.
Expert Opin Biol Ther. 2006;6(6):605-61716706607
Google ScholarCrossref 36.Sok JC, Coppelli FM, Thomas SM.
et al. Mutant epidermal growth factor receptor (EGFRvIII) contributes to head and neck cancer growth and resistance to EGFR targeting.
Clin Cancer Res. 2006;12(17):5064-507316951222
Google ScholarCrossref 37.Lynch TJ, Bell DW, Sordella R.
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):2129-213915118073
Google ScholarCrossref 38.Paez JG, Janne PA, Lee JC.
et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy.
Science. 2004;304(5676):1497-150015118125
Google ScholarCrossref 39.Guo M, Liu S, Lu F. Gefitinib-sensitizing mutations in esophageal carcinoma.
N Engl J Med. 2006;354(20):2193-219416707764
Google ScholarCrossref 40.Willmore-Payne C, Holden JA, Layfield LJ. Detection of EGFR- and HER2-activating mutations in squamous cell carcinoma involving the head and neck.
Mod Pathol. 2006;19(5):634-64016547470
Google ScholarCrossref 41.Baselga J. Targeting tyrosine kinases in cancer: the second wave.
Science. 2006;312(5777):1175-117816728632
Google ScholarCrossref 42.Lacouture ME. Mechanisms of cutaneous toxicities to EGFR inhibitors.
Nat Rev Cancer. 2006;6(10):803-81216990857
Google ScholarCrossref 43.Tejpar S, Piessevaux H, Claes K.
et al. Magnesium wasting associated with epidermal-growth-factor receptor-targeting antibodies in colorectal cancer: a prospective study.
Lancet Oncol. 2007;8(5):387-39417466895
Google ScholarCrossref 44.Schrag D, Chung KY, Flombaum C, Saltz L. Cetuximab therapy and symptomatic hypomagnesemia.
J Natl Cancer Inst. 2005;97(16):1221-122416106027
Google ScholarCrossref 45.Li S, Schmitz KR, Jeffrey PD, Wiltzius JJ, Kussie P, Ferguson KM. Structural basis for inhibition of the epidermal growth factor receptor by cetuximab.
Cancer Cell. 2005;7(4):301-31115837620
Google ScholarCrossref 46.Imai K, Takaoka A. Comparing antibody and small-molecule therapies for cancer.
Nat Rev Cancer. 2006;6(9):714-72716929325
Google ScholarCrossref 47.Kawaguchi Y, Kono K, Mimura K, Sugai H, Akaike H, Fujii H. Cetuximab induce antibody-dependent cellular cytotoxicity against EGFR-expressing esophageal squamous cell carcinoma.
Int J Cancer. 2007;120(4):781-78717096332
Google ScholarCrossref 48.Govindan SV, Griffiths GL, Hansen HJ, Horak ID, Goldenberg DM. Cancer therapy with radiolabeled and drug/toxin-conjugated antibodies.
Technol Cancer Res Treat. 2005;4(4):375-39116029057
Google Scholar 49.Heymach JV, Nilsson M, Blumenschein G, Papadimitrakopoulou V, Herbst R. Epidermal growth factor receptor inhibitors in development for the treatment of non-small cell lung cancer.
Clin Cancer Res. 2006;12(14, pt 2):4441s-4445s16857825
Google ScholarCrossref 50.Carter TA, Wodicka LM, Shah NP.
et al. Inhibition of drug-resistant mutants of ABL, KIT, and EGF receptor kinases.
Proc Natl Acad Sci U S A. 2005;102(31):11011-1101616046538
Google ScholarCrossref 51.Verreault M, Webb MS, Ramsay EC, Bally MB. Gene silencing in the development of personalized cancer treatment: the targets, the agents and the delivery systems.
Curr Gene Ther. 2006;6(4):505-53316918337
Google ScholarCrossref 52.Coppelli FM, Grandis JR. Oligonucleotides as anticancer agents: from the benchside to the clinic and beyond.
Curr Pharm Des. 2005;11(22):2825-284016101440
Google ScholarCrossref 53.Giaccone G. Epidermal growth factor receptor inhibitors in the treatment of non-small-cell lung cancer.
J Clin Oncol. 2005;23(14):3235-324215886311
Google ScholarCrossref 54.Patel JD, Pasche B, Argiris A. Targeting non-small cell lung cancer with epidermal growth factor tyrosine kinase inhibitors: where do we stand, where do we go.
Crit Rev Oncol Hematol. 2004;50(3):175-18615182824
Google ScholarCrossref 55.Fukuoka M, Yano S, Giaccone G.
et al. Multi-institutional randomized phase II trial of gefitinib for previously treated patients with advanced non-small-cell lung cancer (The IDEAL 1 Trial) [published correction appears in
J Clin Oncol. 2004;22(23):4811].
J Clin Oncol. 2003;21(12):2237-224612748244
Google ScholarCrossref 56.Kris MG, Natale RB, Herbst RS.
et al. Efficacy of gefitinib, an inhibitor of the epidermal growth factor receptor tyrosine kinase, in symptomatic patients with non-small cell lung cancer: a randomized trial.
JAMA. 2003;290(16):2149-215814570950
Google ScholarCrossref 57.Pérez-Soler R, Chachoua A, Hammond LA.
et al. Determinants of tumor response and survival with erlotinib in patients with non–small-cell lung cancer.
J Clin Oncol. 2004;22(16):3238-324715310767
Google ScholarCrossref 58.Shepherd FA, Rodrigues Pereira J, Ciuleanu T.
et al. Erlotinib in previously treated non-small-cell lung cancer.
N Engl J Med. 2005;353(2):123-13216014882
Google ScholarCrossref 59.Herbst RS, Prager D, Hermann R.
et al. TRIBUTE: a phase III trial of erlotinib hydrochloride (OSI-774) combined with carboplatin and paclitaxel chemotherapy in advanced non-small-cell lung cancer.
J Clin Oncol. 2005;23(25):5892-589916043829
Google ScholarCrossref 60.Gatzemeier U, Pluzanska A, Szczesna A.
et al. Phase III study of erlotinib in combination with cisplatin and gemcitabine in advanced non-small-cell lung cancer: the Tarceva Lung Cancer Investigation Trial.
J Clin Oncol. 2007;25(12):1545-155217442998
Google ScholarCrossref 61.Thatcher N, Chang A, Parikh P.
et al. Gefitinib plus best supportive care in previously treated patients with refractory advanced non-small-cell lung cancer: results from a randomised, placebo-controlled, multicentre study (Iressa Survival Evaluation in Lung Cancer).
Lancet. 2005;366(9496):1527-153716257339
Google ScholarCrossref 62.Herbst RS, Giaccone G, Schiller JH.
et al. Gefitinib in combination with paclitaxel and carboplatin in advanced non-small-cell lung cancer: a phase III trial—INTACT 2.
J Clin Oncol. 2004;22(5):785-79414990633
Google ScholarCrossref 63.Giaccone G, Herbst RS, Manegold C.
et al. Gefitinib in combination with gemcitabine and cisplatin in advanced non-small-cell lung cancer: a phase III trial—INTACT 1.
J Clin Oncol. 2004;22(5):777-78414990632
Google ScholarCrossref 64.Kelly K, Gaspar L, Chansky K, Albain K, Crowley J, Gandara D. Low incidence of pneumonitis on SWOG 0023: a preliminary analysis of an ongoing phase III trial of concurrent chemoradiotherapy followed by consolidation docetaxel and Iressa/placebo maintenance in patients with inoperable stage III non-small cell lung cancer.
J Clin Oncol. 2005;23(16):(suppl)
7058
Google Scholar 65.Stewart S, Cohen E, Licitra L.
et al. A phase III randomized parallel-group study of gefitinib (Iressa) versus methotrexate (IMEX) in patients with recurrent squamous cell carcinoma of the head and neck.
Proc Am Assoc Cancer Res. 2007;A3522
Google Scholar 66.Burtness B, Goldwasser MA, Flood W, Mattar B, Forastiere AA. 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):8646-865416314626
Google ScholarCrossref 67.Bonner JA, Harari PM, Giralt J.
et al. Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck.
N Engl J Med. 2006;354(6):567-57816467544
Google ScholarCrossref 68.Bezjak A, Tu D, Seymour L.
et al. Symptom improvement in lung cancer patients treated with erlotinib: quality of life analysis of the National Cancer Institute of Canada Clinical Trials Group Study BR.21.
J Clin Oncol. 2006;24(24):3831-383716921034
Google ScholarCrossref 69.Akita RW, Sliwkowski MX. Preclinical studies with erlotinib (Tarceva).
Semin Oncol. 2003;30(3):(suppl 7)
15-2412840797
Google ScholarCrossref 70.Pedersen MW, Pedersen N, Ottesen LH, Poulsen HS. Differential response to gefitinib of cells expressing normal EGFR and the mutant EGFRvIII.
Br J Cancer. 2005;93(8):915-92316189524
Google ScholarCrossref 71.Gandara DR, Gumerlock PH. Epidermal growth factor receptor tyrosine kinase inhibitors plus chemotherapy: case closed or is the jury still out?
J Clin Oncol. 2005;23(25):5856-585816043825
Google ScholarCrossref 72.Hanna N, Lilenbaum R, Ansari R.
et al. Phase II trial of cetuximab in patients with previously treated non–small-cell lung cancer.
J Clin Oncol. 2006;24(33):5253-525817114658
Google ScholarCrossref 73.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):3579-35848339264
Google Scholar 74.Cohen EE, Kane MA, List MA.
et al. Phase II trial of gefitinib 250 mg daily in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck.
Clin Cancer Res. 2005;11(23):8418-842416322304
Google ScholarCrossref 75.Cohen EE, Rosen F, Stadler WM.
et al. Phase II trial of ZD1839 in recurrent or metastatic squamous cell carcinoma of the head and neck.
J Clin Oncol. 2003;21(10):1980-198712743152
Google ScholarCrossref 76.Soulieres D, Senzer NN, Vokes EE, Hidalgo M, Agarwala SS, Siu LL. 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):77-8514701768
Google ScholarCrossref 77.Kirby AM, A’Hern RP, D’Ambrosio C.
et al. Gefitinib (ZD1839, Iressa) as palliative treatment in recurrent or metastatic head and neck cancer.
Br J Cancer. 2006;94(5):631-63616495923
Google Scholar 78.Abidoye OO, Cohen EE, Wong SJ.
et al. A phase II study of lapatinib (GW572016) in recurrent/metastatic (R/M) squamous cell carcinoma of the head and neck (SCCHN).
J Clin Oncol. 2006;24(18):(suppl)
5568
Google Scholar 79.Cohen EE. Role of epidermal growth factor receptor pathway-targeted therapy in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck.
J Clin Oncol. 2006;24(17):2659-266516763280
Google ScholarCrossref 80.Pfister DG, Su YB, Kraus DH.
et al. Concurrent cetuximab, cisplatin, and concomitant boost radiotherapy for locoregionally advanced, squamous cell head and neck cancer: a pilot phase II study of a new combined-modality paradigm.
J Clin Oncol. 2006;24(7):1072-107816505426
Google ScholarCrossref 81.Trigo J, Hitt R, Koralewski P.
et al. Cetuximab monotherapy is active in patients (pts) with platinum-refractory recurrent/metastatic squamous cell carcinoma of the head and neck (SCCHN): results of a phase II study.
J Clin Oncol. 2004;22(14):(suppl)
5502
Google Scholar 82.Baselga J, Trigo JM, Bourhis J.
et al. Phase II multicenter study of the antiepidermal growth factor receptor monoclonal antibody cetuximab in combination with platinum-based chemotherapy in patients with platinum-refractory metastatic and/or recurrent squamous cell carcinoma of the head and neck.
J Clin Oncol. 2005;23(24):5568-557716009950
Google ScholarCrossref 83.Herbst RS, Arquette M, Shin DM.
et al. Phase II multicenter study of the epidermal growth factor receptor antibody cetuximab and cisplatin for recurrent and refractory squamous cell carcinoma of the head and neck.
J Clin Oncol. 2005;23(24):5578-558716009949
Google ScholarCrossref 84.Vermorken J, Mesia R, Vega-Villegas M.
et al. Cetuximab in combination with cisplatin or carboplatin and 5-fluorouracil (5-FU) in the first-line treatment of patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck (R&M SCCHN) (EXTREME).
J Clin Oncol. 2006;24(18):(suppl)
5537
Google Scholar 85.Holmes RS, Vaughan TL. Epidemiology and pathogenesis of esophageal cancer.
Semin Radiat Oncol. 2007;17(1):2-917185192
Google ScholarCrossref 86.Janmaat ML, Gallegos-Ruiz MI, Rodriguez JA.
et al. Predictive factors for outcome in a phase II study of gefitinib in second-line treatment of advanced esophageal cancer patients.
J Clin Oncol. 2006;24(10):1612-161916575012
Google ScholarCrossref 87.Sudo T, Mimori K, Nagahara H.
et al. Identification of EGFR mutations in esophageal cancer.
Eur J Surg Oncol. 2007;33(1):44-4817142003
Google ScholarCrossref 88.Kwak EL, Jankowski J, Thayer SP.
et al. Epidermal growth factor receptor kinase domain mutations in esophageal and pancreatic adenocarcinomas.
Clin Cancer Res. 2006;12(14, pt 1):4283-428716857803
Google ScholarCrossref 89.Dragovich T, McCoy S, Fenoglio-Preiser CM.
et al. Phase II trial of erlotinib in gastroesophageal junction and gastric adenocarcinomas: SWOG 0127.
J Clin Oncol. 2006;24(30):4922-492717050876
Google ScholarCrossref 90.Enzinger PC, Yock T, Suh W.
et al. Phase II cisplatin, irinotecan, cetuximab and concurrent radiation therapy followed by surgery for locally advanced esophageal cancer.
J Clin Oncol. 2006;24(18):(suppl)
4064
Google Scholar 91.Dobelbower MC, Russo SM, Raisch KP.
et al. Epidermal growth factor receptor tyrosine kinase inhibitor, erlotinib, and concurrent 5-fluorouracil, cisplatin and radiotherapy for patients with esophageal cancer: a phase I study.
Anticancer Drugs. 2006;17(1):95-10216317296
Google ScholarCrossref 92.Eberhard DA, Johnson BE, Amler LC.
et al. Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib.
J Clin Oncol. 2005;23(25):5900-590916043828
Google ScholarCrossref 93.Pham D, Kris MG, Riely GJ.
et al. Use of cigarette-smoking history to estimate the likelihood of mutations in epidermal growth factor receptor gene exons 19 and 21 in lung adenocarcinomas.
J Clin Oncol. 2006;24(11):1700-170416505411
Google ScholarCrossref 94.West HL, Franklin WA, McCoy J.
et al. Gefitinib therapy in advanced bronchioloalveolar carcinoma: Southwest Oncology Group Study S0126.
J Clin Oncol. 2006;24(12):1807-181316622257
Google ScholarCrossref 95.Raz DJ, He B, Rosell R, Jablons DM. Current concepts in bronchioloalveolar carcinoma biology.
Clin Cancer Res. 2006;12(12):3698-370416778095
Google ScholarCrossref 96.Pérez-Soler R, Saltz L. Cutaneous adverse effects with HER1/EGFR-targeted agents: is there a silver lining?
J Clin Oncol. 2005;23(22):5235-524616051966
Google ScholarCrossref 97.Amador ML, Oppenheimer D, Perea S.
et al. An epidermal growth factor receptor intron 1 polymorphism mediates response to epidermal growth factor receptor inhibitors.
Cancer Res. 2004;64(24):9139-914315604284
Google ScholarCrossref 98.Calvo E, Baselga J. Ethnic differences in response to epidermal growth factor receptor tyrosine kinase inhibitors.
J Clin Oncol. 2006;24(14):2158-216316682734
Google ScholarCrossref 99.Shigematsu H, Gazdar AF. Somatic mutations of epidermal growth factor receptor signaling pathway in lung cancers.
Int J Cancer. 2006;118(2):257-26216231326
Google ScholarCrossref 100.Loeffler-Ragg J, Witsch-Baumgartner M, Tzankov A.
et al. Low incidence of mutations in EGFR kinase domain in Caucasian patients with head and neck squamous cell carcinoma.
Eur J Cancer. 2006;42(1):109-11116324836
Google ScholarCrossref 101.Mitsudomi T, Kosaka T, Endoh H.
et al. Mutations of the epidermal growth factor receptor gene predict prolonged survival after gefitinib treatment in patients with non-small-cell lung cancer with postoperative recurrence.
J Clin Oncol. 2005;23(11):2513-252015738541
Google ScholarCrossref 102.Han SW, Kim TY, Hwang PG.
et al. Predictive and prognostic impact of epidermal growth factor receptor mutation in non-small-cell lung cancer patients treated with gefitinib.
J Clin Oncol. 2005;23(11):2493-250115710947
Google ScholarCrossref 103.Taron M, Ichinose Y, Rosell R.
et al. Activating mutations in the tyrosine kinase domain of the epidermal growth factor receptor are associated with improved survival in gefitinib-treated chemorefractory lung adenocarcinomas.
Clin Cancer Res. 2005;11(16):5878-588516115929
Google ScholarCrossref 104.Tsao MS, Sakurada A, Cutz JC.
et al. Erlotinib in lung cancer—molecular and clinical predictors of outcome.
N Engl J Med. 2005;353(2):133-14416014883
Google ScholarCrossref 105.Kobayashi S, Boggon TJ, Dayaram T.
et al. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib.
N Engl J Med. 2005;352(8):786-79215728811
Google ScholarCrossref 106.Wirth LJ, Haddad RI, Lindeman NI.
et al. Phase I study of gefitinib plus celecoxib in recurrent or metastatic squamous cell carcinoma of the head and neck.
J Clin Oncol. 2005;23(28):6976-698116172459
Google ScholarCrossref 107.Inoue A, Suzuki T, Fukuhara T.
et al. Prospective phase II study of gefitinib for chemotherapy-naive patients with advanced non-small-cell lung cancer with epidermal growth factor receptor gene mutations.
J Clin Oncol. 2006;24(21):3340-334616785471
Google ScholarCrossref 108.Asahina H, Yamazaki K, Kinoshita I.
et al. A phase II trial of gefitinib as first-line therapy for advanced non-small cell lung cancer with epidermal growth factor receptor mutations.
Br J Cancer. 2006;95(8):998-100417047648
Google ScholarCrossref 109.Paz-Ares L, Sanchez JM, Garcia-Velasco A.
et al. A prospective phase II trial of erlotinib in advanced non-small cell lung cancer (NSCLC) patients with mutations in the tyrosine kinase (TK) domain of the epidermal growth factor receptor (EGFR).
J Clin Oncol. 2006;24(18):(suppl)
7020
Google Scholar 110.Kimura H, Fujiwara Y, Sone T.
et al. High sensitivity detection of epidermal growth factor receptor mutations in the pleural effusion of non-small cell lung cancer patients.
Cancer Sci. 2006;97(7):642-64816827805
Google ScholarCrossref 111.Cappuzzo F, Hirsch FR, Rossi E.
et al. Epidermal growth factor receptor gene and protein and gefitinib sensitivity in non-small-cell lung cancer.
J Natl Cancer Inst. 2005;97(9):643-65515870435
Google ScholarCrossref 112.Hirsch FR, Varella-Garcia M, McCoy J.
et al. Increased epidermal growth factor receptor gene copy number detected by fluorescence in situ hybridization associates with increased sensitivity to gefitinib in patients with bronchioloalveolar carcinoma subtypes: a Southwest Oncology Group Study.
J Clin Oncol. 2005;23(28):6838-684515998906
Google ScholarCrossref 113.Hirsch FR, Varella-Garcia M, Bunn PA Jr.
et al. Molecular predictors of outcome with gefitinib in a phase III placebo-controlled study in advanced non-small-cell lung cancer.
J Clin Oncol. 2006;24(31):5034-504217075123
Google ScholarCrossref 114.Cappuzzo F, Toschi L, Domenichini I.
et al. HER3 genomic gain and sensitivity to gefitinib in advanced non-small-cell lung cancer patients.
Br J Cancer. 2005;93(12):1334-134016288303
Google ScholarCrossref 115.Cappuzzo F, Varella-Garcia M, Shigematsu H.
et al. Increased HER2 gene copy number is associated with response to gefitinib therapy in epidermal growth factor receptor-positive non-small-cell lung cancer patients.
J Clin Oncol. 2005;23(22):5007-501816051952
Google ScholarCrossref 116.Ichihara S, Toyooka S, Fujiwara Y.
et al. The impact of epidermal growth factor receptor gene status on gefitinib-treated Japanese patients with non-small-cell lung cancer.
Int J Cancer. 2007;120(6):1239-124717192902
Google ScholarCrossref 117.Liu W, Innocenti F, Chen P, Das S, Cook EH Jr, Ratain MJ. Interethnic difference in the allelic distribution of human epidermal growth factor receptor intron 1 polymorphism.
Clin Cancer Res. 2003;9(3):1009-101212631599
Google Scholar 118.Araújo A, Ribeiro R, Azevedo I.
et al. Genetic polymorphisms of the epidermal growth factor and related receptor in non-small cell lung cancer—a review of the literature.
Oncologist. 2007;12(2):201-21017296816
Google ScholarCrossref 119.Li J, Karlsson MO, Brahmer J.
et al. CYP3A phenotyping approach to predict systemic exposure to EGFR tyrosine kinase inhibitors.
J Natl Cancer Inst. 2006;98(23):1714-172317148773
Google ScholarCrossref 120.Dziadziuszko R, Hirsch FR, Varella-Garcia M, Bunn PA Jr. Selecting lung cancer patients for treatment with epidermal growth factor receptor tyrosine kinase inhibitors by immunohistochemistry and fluorescence in situ hybridization—why, when, and how?
Clin Cancer Res. 2006;12(14, pt 2):4409s-4415s16857819
Google ScholarCrossref 121.Bailey LR, Kris M, Wolf M.
et al. Tumor epidermal growth factor receptor (EGFR) expression levels does not predict for response in patients (pts) receiving gefitinib (“Iressa,” ZD1839) monotherapy for pretreated advanced non-small-cell lung cancer (NSCLC): IDEAL 1 and 2.
Proc Am Assoc Cancer Res. 2003;A1362
Google Scholar 122.Toschi L, Cappuzzo F. Understanding the new genetics of responsiveness to epidermal growth factor receptor tyrosine kinase inhibitors.
Oncologist. 2007;12(2):211-22017296817
Google ScholarCrossref 123.Cappuzzo F, Toschi L, Tallini G.
et al. Insulin-like growth factor receptor 1 (IGFR-1) is significantly associated with longer survival in non-small-cell lung cancer patients treated with gefitinib.
Ann Oncol. 2006;17(7):1120-112716600976
Google ScholarCrossref 124.Cappuzzo F, Magrini E, Ceresoli GL.
et al. Akt phosphorylation and gefitinib efficacy in patients with advanced non-small-cell lung cancer.
J Natl Cancer Inst. 2004;96(15):1133-114115292385
Google ScholarCrossref 125.Noro R, Gemma A, Kosaihira S.
et al. Gefitinib (IRESSA) sensitive lung cancer cell lines show phosphorylation of Akt without ligand stimulation.
BMC Cancer. 2006;6(1):27717150102
Google ScholarCrossref 126.Sergina NV, Rausch M, Wang D.
et al. Escape from HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3.
Nature. 2007;445(7126):437-44117206155
Google ScholarCrossref 127.Toyooka S, Tokumo M, Shigematsu H.
et al. Mutational and epigenetic evidence for independent pathways for lung adenocarcinomas arising in smokers and never smokers.
Cancer Res. 2006;66(3):1371-137516452191
Google ScholarCrossref 128.Aviel-Ronen S, Blackhall FH, Shepherd FA, Tsao MS. K-ras mutations in non-small-cell lung carcinoma: a review.
Clin Lung Cancer. 2006;8(1):30-3816870043
Google ScholarCrossref 129.Pao W, Miller VA. Epidermal growth factor receptor mutations, small-molecule kinase inhibitors, and non-small-cell lung cancer: current knowledge and future directions.
J Clin Oncol. 2005;23(11):2556-256815767641
Google ScholarCrossref 130.Miller VA, Zakowski M, Riely GJ.
et al. EGFR mutations and copy number, EGFR expression and KRAS mutation as predictors of outcome with erlotinib in bronchioalveolar cell carcinoma (BAC): results of a prospective study.
J Clin Oncol. 2006;24(18):(suppl)
364
Google Scholar 131.Tsao M, Zhu C, Sakurada A.
et al. An analysis of the prognostic and predictive importance of K-ras mutation status in the National Cancer Institute of Canada Clinical Trials Group BR.21 study of erlotinib versus placebo in the treatment of non-small cell lung cancer.
J Clin Oncol. 2006;24(18):(suppl)
365
Google Scholar 132.Huang S, Armstrong EA, Benavente S, Chinnaiyan P, Harari PM. Dual-agent molecular targeting of the epidermal growth factor receptor (EGFR): combining anti-EGFR antibody with tyrosine kinase inhibitor.
Cancer Res. 2004;64(15):5355-536215289342
Google ScholarCrossref 133.Baselga J, Schöffski P, Rojo F.
et al. A phase I pharmacokinetic (PK) and molecular pharmacodynamic (PD) study of the combination of two anti-EGFR therapies, the monoclonal antibody (MAb) cetuximab (C) and the tyrosine kinase inhibitor (TKI) gefitinib (G), in patients (pts) with advanced colorectal (CRC), head and neck (HNC) and non-small cell lung cancer (NSCLC).
J Clin Oncol. 2006;24(18):(suppl)
3006
Google Scholar 134.Viloria-Petit A, Crombet T, Jothy S.
et al. Acquired resistance to the antitumor effect of epidermal growth factor receptor-blocking antibodies in vivo: a role for altered tumor angiogenesis.
Cancer Res. 2001;61(13):5090-510111431346
Google Scholar 135.Bozec A, Fischel JL, Milano G. Epidermal growth factor receptor/angiogenesis dual targeting: preclinical experience.
Curr Opin Oncol. 2006;18(4):330-33416721126
Google ScholarCrossref 136.Calvo E, Malik S, Siu L.
et al. Assessment of erlotinib pharmacodynamics in tumors and skin of patients with head and neck cancer.
Ann Oncol. 2007;18(4):761-76717317676
Google ScholarCrossref 137.Solit DB, She Y, Lobo J.
et al. Pulsatile administration of the epidermal growth factor receptor inhibitor gefitinib is significantly more effective than continuous dosing for sensitizing tumors to paclitaxel.
Clin Cancer Res. 2005;11(5):1983-198915756024
Google ScholarCrossref