The majority of patients with head and neck squamous cell carcinoma (HNSCC) present with advanced-stage disease. The current standard of care is surgery followed by adjuvant radiotherapy with or without chemotherapy or chemoradiation alone. The addition of cetuximab for the treatment of patients with locally advanced or recurrent/metastatic HNSCC has improved overall survival and locoregional control; however, responses are often modest, and treatment resistance is common. A variety of therapeutic strategies are being explored to overcome cetuximab resistance by blocking candidate proteins implicated in resistance mechanisms such as HER2. Several HER2 inhibitors are in clinical development for HNSCC, and HER2-targeted therapy has been approved for several cancers. This review focuses on the biology of HER2, its role in cancer development, and the rationale for clinical investigation of HER2 targeting in HNSCC. Clin Cancer Res; 21(3); 526–33. ©2014 AACR.

Head and neck squamous cell carcinoma (HNSCC) is the seventh most common cancer worldwide by incidence, with an estimated 600,000 new cases reported each year worldwide, two thirds of which occur in industrialized nations (1). The 5-year survival rate of patients with HNSCC is approximately 40% to 60% (2), with a high rate of tumor recurrence likely due to the advanced stage (stages III and IV) at diagnosis in many cases. Locoregional disease recurrence is common, and distant metastatic disease arises in 20% to 30% of patients (3).

The standard of care for advanced HNSCC is surgical resection followed by adjuvant radiotherapy or chemoradiation as a primary treatment approach. However, treatment strategies that target the biologic mechanisms of HNSCC tumorigenesis have been, and continue to be, investigated. Currently, cetuximab, an mAb to EGFR, is the only FDA-approved molecular targeting agent for the treatment of primary or recurrent/metastatic HNSCC. Overexpression of EGFR has been found in approximately 90% of cases of HNSCC and is a predictor of poor prognosis (4–6). However, responses to cetuximab as a single agent do not exceed 13% for patients with recurrent/metastatic disease, are typically short lived, and are not correlated with EGFR expression levels in the primary tumor (7). Many studies have proposed several mechanisms for resistance, the most common of which involves overactivation of other ErbB family receptors, including HER2 (8).

HER2, commonly referred to as ErbB2, c-erbB2, or HER2/neu, is a 185-kDa receptor tyrosine kinase and a member of the ErbB family of proteins (9). The ErbB family consists of four closely related receptors: EGFR (ErbB1/HER1), ErbB2 (HER2/neu), ErbB3 (HER3), and ErbB4 (HER4; ref. 10). There is some homology among the family members; each is a membrane-spanning tyrosine kinase that exists as an inactive monomer. Upon ligand binding, the receptors homodimerize or heterodimerize with other members of the ErbB protein family, which triggers autophosphorylation of their intracellular tyrosine kinase domains and initiates a signaling cascade (11). The ErbB proteins are expressed in most epithelial cell layers and play a key role in cell differentiation during development (12).

ErbB2 was originally identified as the oncogene neu in mouse embryonic fibroblasts (NIH3T3 cells; ref. 13). HER2 has since been found to be amplified and overexpressed in a number of human cancers (12), contributing to tumor development, cell-cycle progression, and cellular motility and growth. Consequently, HER2 is an active focus of drug development and cancer research. To date, 20 articles have been reported in the peer-reviewed literature assessing HER2 expression in HNSCC tumors (14–33). Six articles have evaluated HER2 targeting in HNSCC preclinical models, and two trials have been completed and reported using HER2 inhibitors for this malignancy (18, 34–40). In this review, we evaluate the preclinical and clinical data implicating HER2 as a therapeutic target in HNSCC.

HER2 signaling pathway

Unlike the other family members, HER2 lacks a ligand-binding domain, characterizing it as an orphan receptor. The absence of a ligand likely contributes to its role as a powerful signal amplifier for the other ErbB family receptors (41). Evidence suggests that HER2 is the preferred dimerization partner among all members of the protein family, perhaps due to frequent recycling of the HER2 receptor heterodimers to the cell surface as well as the ability of HER2 to decrease the rate of ligand dissociation (42–44).

The downstream signaling effects of the HER2 receptor are complex due to the differential effects of the various HER2-containing heterodimers. For example, EGFR/HER2 heterodimers preferentially stimulate the MAPK pathway, whereas the HER2/HER3 heterodimers activate both the MAPK and the PI3K/v-akt murine thymoma viral oncogene homolog (AKT) pathway (ref. 45; Fig. 1). There are at least 10 known EGF-related peptides with varying degrees of affinity for the different heterodimers (11). These peptides do not directly bind to HER2 but instead promote the receptor's heterodimerization and cross-phosphorylation (11). EGFR/HER2 heterodimers are most commonly formed in response to stimulation with EGF, whereas formation of HER2/HER3 is driven by neuregulins (44). Even when overexpressed, HER2 maintains its dependence on other members of the HER family, namely HER3, for HER2-mediated tumorigenesis (46).

Figure 1.

HER2 Signaling. Human epidermal growth factor receptors, EGFR (HER1), HER2, HER3, and HER4, are receptor tyrosine kinases that play a role in cell growth, survival, and differentiation. Each receptor is composed of an extracellular ligand-binding domain, transmembrane domain, and tyrosine kinase domain. Upon ligand binding, the receptors are activated by auto- and cross-phosphorylation. There are no known ligands for HER2, which does not need to be bound to a ligand to be activated. HER2 is the preferential dimerization partner among all family members. Dimerization and subsequent phosphorylation lead to activation of downstream targets, including the PI3K/AKT and RAF/MEK/MAPK pathways. Activation of these pathways enhances survival, proliferation, and cell-cycle progression. AKT, v-akt murine thymomaviral oncogene homolog 1; P, phosphorylation; RAF, v-raf-1 murine leukemia viral oncogene homolog; RAS, rat sarcoma viral oncogene homolog.

Figure 1.

HER2 Signaling. Human epidermal growth factor receptors, EGFR (HER1), HER2, HER3, and HER4, are receptor tyrosine kinases that play a role in cell growth, survival, and differentiation. Each receptor is composed of an extracellular ligand-binding domain, transmembrane domain, and tyrosine kinase domain. Upon ligand binding, the receptors are activated by auto- and cross-phosphorylation. There are no known ligands for HER2, which does not need to be bound to a ligand to be activated. HER2 is the preferential dimerization partner among all family members. Dimerization and subsequent phosphorylation lead to activation of downstream targets, including the PI3K/AKT and RAF/MEK/MAPK pathways. Activation of these pathways enhances survival, proliferation, and cell-cycle progression. AKT, v-akt murine thymomaviral oncogene homolog 1; P, phosphorylation; RAF, v-raf-1 murine leukemia viral oncogene homolog; RAS, rat sarcoma viral oncogene homolog.

Close modal

Variations in detection techniques and interpretation methods of HER2 overexpression have contributed to inconsistent reporting of overexpression of HER2 in many types of cancer, including HNSCC. The American Society of Clinical Oncology (ASCO) publishes guidelines for assessing HER2 status in breast carcinomas via IHC and FISH (47). Although distinct HER2 testing protocols have also been established in gastric cancer, none exist for HNSCC (48). Consequently, pathologists apply the IHC/FISH scoring techniques for breast cancers to HNSCC, a biologically different carcinoma. Reports of HER2 overexpression range from 0% to 47% in HNSCC (15, 20, 25). In laryngeal cancer, positive HER2 staining has been reported in 68% of cases (24). Azemar and colleagues found significantly elevated levels of HER2 in 26 of 45 primary HNSCC samples (49). We assessed 426 HNSCC tumors in The Cancer Genome Atlas (TCGA) database and found 18 cases (4%) with mRNA upregulation; however, only 8 of those 18 also harbored HER2 gene amplification. In a separate analysis of 213 HNSCC tumors in The Cancer Proteome Atlas (TCPA), low levels of activated (phosphorylated HER2) or total HER2 expression were detected (Fig. 2A). Although HER2 mRNA levels correlated with HER2 protein expression, no correlation was observed with levels of phosphorylated HER2 (Fig. 2B). The protein levels of TCGA tumors were determined by reverse phase protein array (RPPA), a high throughput technique that reports HER2 scoring intensity on a continuum. Assadi and colleagues compared RPPA results with IHC and found a correlation of 0.86 (50). Comparing protein detection techniques with those for DNA or RNA is not as simple. Levels of RNA in TCGA tumors were determined by RNAseq processing, a profiling method that is subject to sample bias. Moreover, there remains a gap between mRNA and protein levels because of various degrees of regulation.

Figure 2.

Landscape of HER2 expression in HNSCC tumors (TCGA database, provisional). A, pHER2 and HER2 expression z-scores were obtained from the TCPA database for HNSCC tumors (n = 213). pHER2 expression levels were plotted as a function of frequency in R version 3.0.2. B, pHER2 levels do not correlate with HER2 mRNA expression. HER2 levels do correlate with HER2 mRNA expression. RPPA data were obtained from the TCPA database for HNSCC tumors. Protein levels and mRNA expression were compared using a Pearson correlation in R version 3.0.2. C and D, RPPA expression for (C) HER2 and (D) pHER2 correlates with RPPA expression for EGFR. E, RPPA expression for pHER2 correlates with RPPA expression for HER2. RPPA data for pHER2, HER2, and EGFR were obtained from the TCPA database for HNSCC tumors. Protein levels were compared using a Pearson correlation in R version 3.0.2.

Figure 2.

Landscape of HER2 expression in HNSCC tumors (TCGA database, provisional). A, pHER2 and HER2 expression z-scores were obtained from the TCPA database for HNSCC tumors (n = 213). pHER2 expression levels were plotted as a function of frequency in R version 3.0.2. B, pHER2 levels do not correlate with HER2 mRNA expression. HER2 levels do correlate with HER2 mRNA expression. RPPA data were obtained from the TCPA database for HNSCC tumors. Protein levels and mRNA expression were compared using a Pearson correlation in R version 3.0.2. C and D, RPPA expression for (C) HER2 and (D) pHER2 correlates with RPPA expression for EGFR. E, RPPA expression for pHER2 correlates with RPPA expression for HER2. RPPA data for pHER2, HER2, and EGFR were obtained from the TCPA database for HNSCC tumors. Protein levels were compared using a Pearson correlation in R version 3.0.2.

Close modal

Elevated HER2 overexpression has been reportedly associated with worse prognosis, increased recurrence, and decreased overall survival (OS) in HNSCC (26, 51). Cavalot and colleagues found that frequency of HER2 overexpression was significantly higher in patients with more aggressive disease occurring in conjunction with metastatic lymph nodes (15). In addition, the 5-year OS and the 5-year disease-free survival (DFS) probability were significantly lower for HER2-positive patients compared with HER2-negative individuals (15). Wei and colleagues found that in two laryngeal tumors, HER2 staining was higher in the metastases (2+ and 3+), whereas staining was lower in the corresponding primary tumors (17). However, other studies have shown that HER2 is not an independent prognostic factor (22, 29). Our analysis found that among the 213 tumors in the TCGA cohort, neither mRNA upregulation of HER2 nor increased expression of total or phosphorylated HER2 protein was an independent predictor of OS (data not shown). However, the small sample size limits the conclusiveness of this analysis.

HER2 has been found to be coexpressed with EGFR in HNSCC tumors (33). Among the 213 HNSCC tumors in the TCPA, expression of both HER2 and activated HER2 (pHER2) correlated with EGFR expression (Fig. 2C and D). As expected, expression of pHER2 correlated with total HER2 expression (Fig. 2E). These findings suggest that HER2 coexpression may contribute to the negative prognostic impact of EGFR because coexpression/activation has been reported to be associated with resistance to therapeutic agents (35, 52). Wheeler and colleagues generated cetuximab-resistant non–small cell lung cancer (NSCLC) and HNSCC cell lines and found that the cetuximab-resistant cell lines expressed higher levels of phosphorylated receptor tyrosine kinases, including HER2, than the parental cell lines (52). In addition, among all of the cetuximab-resistant cell lines, EGFR had increased coimmunoprecipitation with HER2 and HER3 when compared with the parental cell lines. Treating these cells with increasing concentrations of cetuximab had no effect on the autophosphorylation of EGFR or the activity of HER2. However, this resistance was overcome with several tyrosine kinase inhibitors (TKI), including erlotinib, gefitinib, and the pan-HER irreversible inhibitor canertinib (CI-1033). We previously reported that treatment with a dual EGFR-HER2 kinase inhibitor overcame acquired resistance of HNSCC tumors to cetuximab in xenograft tumor models (35). Together, these findings validate the active investigation of HER2 as a molecular target for HNSCC therapy.

Monoclonal antibodies: preclinical and clinical studies

A number of strategies are used to inhibit HER2, including mAbs. Trastuzumab is a humanized mAb that binds domain IV of the extracellular domain of the HER2 receptor, preventing activation of its intracellular tyrosine kinase (53, 54). Trastuzumab has several possible mechanisms of action, including prevention of HER2 receptor dimerization, increased endocytosis of the receptor, and inhibition of the generation of a constitutively active truncated intracellular HER2 molecule (53, 55). A phase II trial evaluated trastuzumab in combination with paclitaxel and cisplatin (56). The addition of trastuzumab did not significantly improve patient response rate to the chemotherapy treatments.

Pertuzumab is a humanized mAb to the HER2 receptor that binds domain II of the extracellular domain and inhibits heterodimerization of HER2 with other members of the HER family (57). Because blocking the formation of heterodimers, particularly HER2/HER3, has had promising effects in the treatment of breast cancer (58, 59), pertuzumab was studied in combination with gefitinib, an EGFR inhibitor, in HNSCC cell lines (60). The cell lines that harbored high levels of phosphorylated HER2 and HER3 were more resistant to gefitinib, and a similar trend was observed for cell lines with amplified EGFR. However, combination treatments of gefitinib and pertuzumab overcame resistance (60). Currently, there are no reports of pertuzumab in HNSCC preclinical models or patients.

Tyrosine kinase inhibitors: preclinical studies

Small-molecule TKIs that bind to the ATP binding site of the HER molecule are also effective therapeutic agents. Lapatinib, a reversible dual TKI of both EGFR and HER2, is FDA approved for the treatment of HER2-positive breast cancer (61). By preventing phosphorylation and subsequent activation of the PI3K/AKT and MAPK/ERK pathways, lapatinib results in an increase in apoptosis and decrease in cellular proliferation and growth. Just like other small-molecule TKIs, lapatinib is administered orally and well tolerated by patients (61). An ex vivo study evaluating the combined effect of lapatinib and cisplatin in 72 different patient-derived HNSCC samples found that lapatinib provided an additive effect by limiting colony formation (38). However, there were significant differences in the response of individual tumors to lapatinib treatment, evident by the wide range of IC50 concentrations (38). Another study evaluating the combined effect of lapatinib with either cisplatin or paclitaxel found additive effects both in vivo and in vitro (40). Lapatinib reduced proliferation of HNSCC cells, but no correlation was found between expression levels of EGFR or HER2 and the antiproliferative effects (40). In vivo xenograft studies in mice bearing YCU-H891 HNSCC cells demonstrated that lapatinib alone induced apoptosis and displayed antitumor activity (40). Although lapatinib alone did not inhibit angiogenesis, inhibition of angiogenesis was observed when lapatinib was combined with paclitaxel. In addition, the combination of cisplatin or paclitaxel with lapatinib resulted in enhanced antitumor activity primarily by increasing apoptosis (40).

The irreversible ErbB family blocker afatinib (Gilotrif) binds to the tyrosine kinase domains of EGFR, HER2, and HER4 (62). The FDA recently approved afatinib for the treatment of patients with NSCLC harboring EGFR mutations. Owing to its irreversible activity and multireceptor binding, afatinib is also being investigated for treatment of patients with other ErbB-driven cancers, including HNSCC (63). To date, a few studies have reported on the use of afatinib in preclinical HNSCC models. In one study, mice inoculated with FaDu cells, which are cells derived from a hypopharyngeal cancer, received varying doses of afatinib; at every dose, afatinib significantly decreased tumor volume size when compared with the control (63).

Dacomitinib, another irreversible pan-HER kinase inhibitor, is an experimental drug under investigation in NSCLC, gastric cancer, glioma, and HNSCC. It has shown preclinical efficacy alone and in combination with ionizing radiotherapy in HNSCC cell lines and mice models (64). HNSCC cell lines, UT-SCC-8 and UT-SCC-42a, showed sensitivity to dacomitinib, with IC50s of 25 nmol/L, with combination treatment of dacomitinib and ionizing radiation further reducing cell viability. Although dacomitinib was effective in inhibiting EGFR phosphorylation, reduction of HER2 phosphorylation was not evaluated (64).

Clinical trials of HER2 TKIs in HNSCC

Ongoing trials of anti-HER agents in clinical development in HNSCC are summarized in Tables 1 and 2. IC50 values for these agents are listed in Table 3. Because of the current palliative nature of treatment associated with recurrent and/or metastatic HNSCC, Tables 1 and 2 delineate the trials as either curative or palliative, with “curative” meaning therapy for nonrecurrent/nonmetastatic HNSCC.

Table 1.

Trials of anti-HER family agents for nonrecurrent/nonmetastatic HNSCC (curative therapy)

Clinical trialPhaseStudy regimensEstimated accrual; study populationPrimary endpointStatus
NCT01783587 Afatinib plus CRT or afatinib plus RT N = 38; intermediate- and high-risk HNSCC DLT Recruiting 
NCT01737008 Dacomitinib plus CRT or RT N = 34; LA HNSCC MTD Recruiting 
NCT01732640 I/II Afatinib plus carboplatin and paclitaxel N = 71; primary unresected patients with LA, HPV-negative, stage III or IV a/b HNSCC MTD, objective tumor response Recruiting 
NCT01824823 II Afatinib or placebo N = 108; patients with LA, stage III or IV HNSCC DFS Recruiting 
NCT01538381 II Afatinib or observation N = 30; newly diagnosed HNSCC Reduction of tumor Recruiting 
NCT01415674 II Afatinib N = 60; untreated nonmetastatic HNSCC Potential predictive biomarkers of afatinib activity Recruiting 
NCT00490061 II Lapatinib plus RT N = 60; stage III/IV HNSCC who cannot tolerate concurrent CRT TTP Active, not recruiting 
NCT00387127 II Lapatinib plus CRT or placebo plus CRT N = 67; stage III, IV a/b HNSCC Complete response rate Active, not recruiting 
NCT01427478 III Afatinib or placebo N = 315; resected, CRT-treated HNSCC with macroscopically complete resection of disease DFS Recruiting 
NCT02131155 (LUX-Head & Neck 4) III Afatinib or placebo N = 150; primary unresected, stage III, IV a/b LA HNSCC without evidence of disease after CRT DFS Recruiting 
NCT01345669 (LUX-Head & Neck 2) III Afatinib or placebo N = 669; primary unresected, stage III, IV a/b LA HNSCC without evidence of disease after CRT DFS Recruiting 
Clinical trialPhaseStudy regimensEstimated accrual; study populationPrimary endpointStatus
NCT01783587 Afatinib plus CRT or afatinib plus RT N = 38; intermediate- and high-risk HNSCC DLT Recruiting 
NCT01737008 Dacomitinib plus CRT or RT N = 34; LA HNSCC MTD Recruiting 
NCT01732640 I/II Afatinib plus carboplatin and paclitaxel N = 71; primary unresected patients with LA, HPV-negative, stage III or IV a/b HNSCC MTD, objective tumor response Recruiting 
NCT01824823 II Afatinib or placebo N = 108; patients with LA, stage III or IV HNSCC DFS Recruiting 
NCT01538381 II Afatinib or observation N = 30; newly diagnosed HNSCC Reduction of tumor Recruiting 
NCT01415674 II Afatinib N = 60; untreated nonmetastatic HNSCC Potential predictive biomarkers of afatinib activity Recruiting 
NCT00490061 II Lapatinib plus RT N = 60; stage III/IV HNSCC who cannot tolerate concurrent CRT TTP Active, not recruiting 
NCT00387127 II Lapatinib plus CRT or placebo plus CRT N = 67; stage III, IV a/b HNSCC Complete response rate Active, not recruiting 
NCT01427478 III Afatinib or placebo N = 315; resected, CRT-treated HNSCC with macroscopically complete resection of disease DFS Recruiting 
NCT02131155 (LUX-Head & Neck 4) III Afatinib or placebo N = 150; primary unresected, stage III, IV a/b LA HNSCC without evidence of disease after CRT DFS Recruiting 
NCT01345669 (LUX-Head & Neck 2) III Afatinib or placebo N = 669; primary unresected, stage III, IV a/b LA HNSCC without evidence of disease after CRT DFS Recruiting 

NOTE: Descriptions for ongoing (recruiting; active, not recruiting; or not yet recruiting) phase I/II clinical trials of anti-HER2 family agents for HNSCC as of September 2014 (as per listings on ClinicalTrials.gov).

Abbreviations: CRT, chemoradiation; DLT, dose-limiting toxicity; HPV, human papillomavirus; LA, locally advanced; RT, radiotherapy; TTP, time to progression.

Table 2.

Trials of anti-HER family agents for recurrent/metastatic HNSCC (palliative therapy)

Clinical trialPhaseStudy regimensEstimated accrual; study populationPrimary endpointStatus
NCT01345682 (LUX-Head & Neck 1) III Afatinib or methotrexate N = 474; R/M HNSCC with progression after at least 2 cycles platinum-based therapy PFS Active, not recruiting 
NCT01856478 (LUX-Head & Neck 3) III Afatinib or methotrexate N = 300; R/M HNSCC with progression after a cisplatin and/or carboplatin therapy PFS Recruiting 
Clinical trialPhaseStudy regimensEstimated accrual; study populationPrimary endpointStatus
NCT01345682 (LUX-Head & Neck 1) III Afatinib or methotrexate N = 474; R/M HNSCC with progression after at least 2 cycles platinum-based therapy PFS Active, not recruiting 
NCT01856478 (LUX-Head & Neck 3) III Afatinib or methotrexate N = 300; R/M HNSCC with progression after a cisplatin and/or carboplatin therapy PFS Recruiting 

NOTE: Description of ongoing clinical trials of anti-HER2 family agents for recurrent/metastatic HNSCC as of September 2014 (as per listings on Clinicaltrials.gov).

Abbreviation: R/M, recurrent/metastatic.

Table 3.

IC50 values of anti-HER family agents in HNSCC

AgentsEGFRHER2
Lapatinb 10.8 9.2 
Afatinib 0.5 14 
Dacomitinib 45.7 
AgentsEGFRHER2
Lapatinb 10.8 9.2 
Afatinib 0.5 14 
Dacomitinib 45.7 

NOTE: Values are expressed in nanomolar units.

Nonrecurrent/nonmetastatic HNSCC

Completed clinical trials for patients with nonrecurrent/nonmetastatic HNSCC include investigations of lapatinib therapy. In a randomized phase II placebo-controlled trial, patients with previously untreated locally advanced HNSCC received either lapatinib or placebo for 2 to 6 weeks, followed by chemoradiation (34). The response rate before chemoradiation was 17% (n = 4, 7% with HER2 overexpression and amplification in 4%) among 24 patients receiving lapatinib and 0% among all 16 patients receiving the placebo. Despite a clinical response, the patients who received lapatinib monotherapy did not have a significant increase in apoptosis compared with those who received placebo. All of the lapatinib responders had EGFR overexpression, and 50% had HER2 expression. Although no other biologic characteristics predicted response to lapatinib therapy, the small sample size warrants future investigation (34).

Another phase II trial did not show the same success of lapatinib as a monotherapy (36). Patients were separated based on their treatment history with an EGFR inhibitor. Patients with no prior treatment with an EGFR inhibitor (EGFR-naive) had a 6-month progression-free survival (PFS) rate of 7%, and the median PFS was 52 days. The EGFR-naive group's median OS and 6-month OS rate were 288 days and 52%, respectively. At the time of analysis, all patients in this group progressed. Similarly, all patients in the previously EGFR inhibitor–treated group had died. The 6-month median PFS was 52 days, and the PFS rate was 6%. The median OS was 155 days, and the 6-month OS rate was 39%. Most HNSCC tumors expressed both EGFR and HER2, and posttreatment biopsies showed a significant decrease in pHER2 levels. Samples were analyzed for the presence of HER2-activating mutations, but only one was found to contain a mutation, which did not produce a phenotype. Although lapatinib was well tolerated by all patients, it appeared to be largely inactive as a monotherapy in both EGFR-naïve and EGFR-refractory patients. A phase III (NCT00424255) trial did not show improvement when lapatinib was added to radiotherapy or cisplatin therapy in the postoperative setting in patients with HNSCC at high risk for recurrence. Patients with resected stage II to stage IVA HNSCC were randomized to receive chemotherapy/radiotherapy with either placebo or lapatinib before, during, and following concurrent chemoradiotherapy. There was no significant difference in DFS between treatment arms.

Although lapatinib has not demonstrated clear efficacy in HNSCC, it still remains in active clinical development for patients with nonrecurrent/nonmetastatic HNSCC. Two actively accruing trials are designed to evaluate lapatinib plus radiotherapy or chemoradiation (Table 1). In a recently completed phase III trial, examining lapatinib with concurrent chemoradiation followed by maintenance monotherapy for high-risk patients with resected HNSCC (NCT00424255), addition of lapatinib to chemoradiation failed to prolong DFS (65). HER2 mutations are rare in HNSCC (2% of TCGA) and have not been evaluated as a predictive biomarker to select patients with HNSCC likely to benefit from anti-HER2 therapy. Attempts to correlate levels of EGFR, phosphorylated EGFR (pEGFR), and HER2 with clinical benefit from lapatinib as a monotherapeutic agent have not been informative to date.

Although there are no completed clinical trials, afatinib and dacomitinib are undergoing active investigation for therapy in nonrecurrent/nonmetastatic HNSCC. Table 1 describes the several ongoing trials investigating these agents alone or in combination with radiotherapy or chemoradiation.

Recurrent/metastatic HNSCC

Among completed clinical trials, afatinib was compared with cetuximab in a recently published phase II study of patients with recurrent/metastatic HNSCC (NCT00514943; ref. 66). The disease control rate by investigator review was 50% for the afatinib-receiving cohort and 56.5% for the cetuximab-receiving cohort (P = 0.48), with similar rates observed by independent central review. Centrally reviewed median PFS was 13.0 weeks for afatinib and 15.0 weeks for the cetuximab group (P = 0.71). Preliminary report of another completed phase III trial (NCT01345682 LUX-Head & Neck 1) suggested that afatinib improved PFS as well as patient-reported outcomes. Patients with recurrent/metastatic HNSCC who progressed on or after platinum-based therapy were randomized to receive afatinib or methotrexate (MTX). Afatinib significantly improved PFS (median, 2.6 months), disease control rate (49.1%), and objective response rate (10.2%) compared with methotrexate (median, 1.7 months, 38.5%, 5.6%). Afatinib did not significantly improve OS (median, 6.8 months) compared with methotrexate (median, 6.2 months). Table 2 shows results from the phase III trials in which afatinib is being compared with methotrexate for patients with recurrent/metastatic HNSCC (NCT01345682/LUX-Head & Neck 1; NCT01856478/LUX-Head & Neck 3; Table 2).

The presence of activating HER2 mutations or HER2 overexpression, primarily due to gene amplification, has been associated with reproducible and robust responses to HER2 targeting therapy in most malignancies, including breast, lung, and gastric cancer. HER2 mutations and HER2 gene amplifications are rare in HNSCC. EGFR is a validated prognostic and therapeutic target for treatment of HNSCC, and the mAb cetuximab is the only FDA-approved molecular targeting agent in HNSCC. EGFR is frequently overexpressed in HNSCC tumors, resulting in constitutive downstream signaling activity. EGFR can heterodimerize with HER2, and preclinical evidence suggests that cotargeting of EGFR and HER2 may augment cetuximab responses and mitigate therapeutic resistance. The majority of agents under active clinical investigation in HNSCC dually target EGFR and HER2. Because a specific molecular inhibitor strategy is unlikely to be effective in all cases, the identification of biomarkers that will predict clinical responses to ErbB family blockers in HNSCC is of paramount importance.

No potential conflicts of interest were disclosed.

Editorial and formatting assistance was provided by Melissa Brunckhorst, PhD, of MedErgy, which was contracted and funded by Boehringer Ingelheim Pharmaceuticals, Inc. (BIPI). BIPI was given the opportunity to review the article for medical and scientific accuracy as well as intellectual property considerations.

J.R. Grandis was supported by the NIH under award numbers R01CA77308 and P50CA097190 and the American Cancer Society.

1.
Ferlay
J
,
Shin
HR
,
Bray
F
,
Forman
D
,
Mathers
C
,
Parkin
DM
. 
GLOBOCAN 2008: cancer incidence and mortality worldwide: IARC CancerBase No. 10 [monograph on the Internet]
.
Lyon, France
:
International Agency for Research on Cancer
; 
2010
[cited 2014 Sep 3]
.
2.
Siegel
R
,
Naishadham
D
,
Jemal
A
. 
Cancer statistics, 2013
.
CA Cancer J Clin
2013
;
63
:
11
30
.
3.
Vermorken
JB
,
Specenier
P
. 
Optimal treatment for recurrent/metastatic head and neck cancer
.
Ann Oncol
2010
;
21
Suppl 7
:
vii
252
61
.
4.
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
:
3579
84
.
5.
Ang
KK
,
Berkey
BA
,
Tu
X
,
Zhang
HZ
,
Katz
R
,
Hammond
EH
, 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
:
7350
6
.
6.
Rubin Grandis
J
,
Melhem
MF
,
Gooding
WE
,
Day
R
,
Holst
VA
,
Wagener
MM
, 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
:
824
32
.
7.
Vermorken
JB
,
Trigo
J
,
Hitt
R
,
Koralewski
P
,
az-Rubio
E
,
Rolland
F
, et al
Open-label, uncontrolled, multicenter phase II study to evaluate the efficacy and toxicity of cetuximab as a single agent in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck who failed to respond to platinum-based therapy
.
J Clin Oncol
2007
;
25
:
2171
7
.
8.
Yonesaka
K
,
Zejnullahu
K
,
Okamoto
I
,
Satoh
T
,
Cappuzzo
F
,
Souglakos
J
, et al
Activation of ERBB2 signaling causes resistance to the EGFR-directed therapeutic antibody cetuximab
.
Sci Transl Med
2011
;
3
:
99ra86
.
9.
Coussens
L
,
Yang-Feng
TL
,
Liao
YC
,
Chen
E
,
Gray
A
,
McGrath
J
, et al
Tyrosine kinase receptor with extensive homology to EGF receptor shares chromosomal location with neu oncogene
.
Science
1985
;
230
:
1132
9
.
10.
Anderson
NG
,
Ahmad
T
. 
ErbB receptor tyrosine kinase inhibitors as therapeutic agents
.
Front Biosci
2002
;
7
:
d1926
40
.
11.
Roskoski
R
 Jr
. 
The ErbB/HER receptor protein-tyrosine kinases and cancer
.
Biochem Biophys Res Commun
2004
;
319
:
1
11
.
12.
Yarden
Y
,
Sliwkowski
MX
. 
Untangling the ErbB signalling network
.
Nat Rev Mol Cell Biol
2001
;
2
:
127
37
.
13.
Shih
C
,
Padhy
LC
,
Murray
M
,
Weinberg
RA
. 
Transforming genes of carcinomas and neuroblastomas introduced into mouse fibroblasts
.
Nature
1981
;
290
:
261
4
.
14.
Xia
W
,
Lau
YK
,
Zhang
HZ
,
Xiao
FY
,
Johnston
DA
,
Liu
AR
, et al
Combination of EGFR, HER-2/neu, and HER-3 is a stronger predictor for the outcome of oral squamous cell carcinoma than any individual family members
.
Clin Cancer Res
1999
;
5
:
4164
74
.
15.
Cavalot
A
,
Martone
T
,
Roggero
N
,
Brondino
G
,
Pagano
M
,
Cortesina
G
. 
Prognostic impact of HER-2/neu expression on squamous head and neck carcinomas
.
Head Neck
2007
;
29
:
655
64
.
16.
Sheikh Ali
MA
,
Gunduz
M
,
Nagatsuka
H
,
Gunduz
E
,
Cengiz
B
,
Fukushima
K
, et al
Expression and mutation analysis of epidermal growth factor receptor in head and neck squamous cell carcinoma
.
Cancer Sci
2008
;
99
:
1589
94
.
17.
Wei
Q
,
Sheng
L
,
Shui
Y
,
Hu
Q
,
Nordgren
H
,
Carlsson
J
. 
EGFR, HER2, and HER3 expression in laryngeal primary tumors and corresponding metastases
.
Ann Surg Oncol
2008
;
15
:
1193
201
.
18.
Zhang
M
,
Taylor
CE
,
Piao
L
,
Datta
J
,
Bruno
PA
,
Bhave
S
, et al
Genetic and chemical targeting of epithelial-restricted with serine box reduces EGF receptor and potentiates the efficacy of afatinib
.
Mol Cancer Ther
2013
;
12
:
1515
25
.
19.
Pardis
S
,
Sardari
Y
,
Ashraf
MJ
,
Andisheh
TA
,
Ebrahimi
H
,
Purshahidi
S
, et al
Evaluation of tissue expression and salivary levels of HER2/neu in patients with head and neck squamous cell carcinoma
.
Iran J Otorhinolaryngol
2012
;
24
:
161
70
.
20.
Sardari
Y
,
Pardis
S
,
Tadbir
AA
,
Ashraf
MJ
,
Fattahi
MJ
,
Ebrahimi
H
, et al
HER2/neu expression in head and neck squamous cell carcinoma patients is not significantly elevated
.
Asian Pac J Cancer Prev
2012
;
13
:
2891
6
.
21.
Hama
T
,
Yuza
Y
,
Suda
T
,
Saito
Y
,
Norizoe
C
,
Kato
T
, et al
Functional mutation analysis of EGFR family genes and corresponding lymph node metastases in head and neck squamous cell carcinoma
.
Clin Exp Metastasis
2012
;
29
:
19
25
.
22.
Ali
MA
,
Gunduz
M
,
Gunduz
E
,
Tamamura
R
,
Beder
LB
,
Katase
N
, et al
Expression and mutation analysis of her2 in head and neck squamous cell carcinoma
.
Cancer Invest
2010
;
28
:
495
500
.
23.
Beckhardt
RN
,
Kiyokawa
N
,
Xi
L
,
Liu
TJ
,
Hung
MC
,
El-Naggar
AK
, et al
HER-2/neu oncogene characterization in head and neck squamous cell carcinoma
.
Arch Otolaryngol Head Neck Surg
1995
;
121
:
1265
70
.
24.
Khademi
B
,
Shirazi
FM
,
Vasei
M
,
Doroudchi
M
,
Gandomi
B
,
Modjtahedi
H
, et al
The expression of p53, c-erbB-1 and c-erbB-2 molecules and their correlation with prognostic markers in patients with head and neck tumors
.
Cancer Lett
2002
;
184
:
223
30
.
25.
Angiero
F
,
Sordo
RD
,
Dessy
E
,
Rossi
E
,
Berenzi
A
,
Stefani
M
, et al
Comparative analysis of c-erbB-2 (HER-2/neu) in squamous cell carcinoma of the tongue: does over-expression exist? And what is its correlation with traditional diagnostic parameters?
J Oral Pathol Med
2008
;
37
:
145
50
.
26.
Xia
W
,
Lau
YK
,
Zhang
HZ
,
Liu
AR
,
Li
L
,
Kiyokawa
N
, et al
Strong correlation between c-erbB-2 overexpression and overall survival of patients with oral squamous cell carcinoma
.
Clin Cancer Res
1997
;
3
:
3
9
.
27.
Ma
BB
,
Poon
TC
,
To
KF
,
Zee
B
,
Mo
FK
,
Chan
CM
, et al
Prognostic significance of tumor angiogenesis, Ki 67, p53 oncoprotein, epidermal growth factor receptor and HER2 receptor protein expression in undifferentiated nasopharyngeal carcinoma–a prospective study
.
Head Neck
2003
;
25
:
864
72
.
28.
Shiga
H
,
Rasmussen
AA
,
Johnston
PG
,
Langmacher
M
,
Baylor
A
,
Lee
M
, et al
Prognostic value of c-erbB2 and other markers in patients treated with chemotherapy for recurrent head and neck cancer
.
Head Neck
2000
;
22
:
599
608
.
29.
Freier
K
,
Joos
S
,
Flechtenmacher
C
,
Devens
F
,
Benner
A
,
Bosch
FX
, et al
Tissue microarray analysis reveals site-specific prevalence of oncogene amplifications in head and neck squamous cell carcinoma
.
Cancer Res
2003
;
63
:
1179
82
.
30.
Koynova
DK
,
Tsenova
VS
,
Jankova
RS
,
Gurov
PB
,
Toncheva
DI
. 
Tissue microarray analysis of EGFR and HER2 oncogene copy number alterations in squamous cell carcinoma of the larynx
.
J Cancer Res Clin Oncol
2005
;
131
:
199
203
.
31.
Tse
GM
,
Yu
KH
,
Chan
AW
,
King
AD
,
Chen
GG
,
Wong
KT
, et al
HER2 expression predicts improved survival in patients with cervical node-positive head and neck squamous cell carcinoma
.
Otolaryngol Head Neck Surg
2009
;
141
:
467
73
.
32.
Khan
AJ
,
King
BL
,
Smith
BD
,
Smith
GL
,
Digiovanna
MP
,
Carter
D
, et al
Characterization of the HER-2/neu oncogene by immunohistochemical and fluorescence in situ hybridization analysis in oral and oropharyngeal squamous cell carcinoma
.
Clin Cancer Res
2002
;
8
:
540
8
.
33.
Ibrahim
SO
,
Lillehaug
JR
,
Johannessen
AC
,
Liavaag
PG
,
Nilsen
R
,
Vasstrand
EN
. 
Expression of biomarkers (p53, transforming growth factor alpha, epidermal growth factor receptor, c-erbB-2/neu and the proliferative cell nuclear antigen) in oropharyngeal squamous cell carcinomas
.
Oral Oncol
1999
;
35
:
302
13
.
34.
Del Campo
JM
,
Hitt
R
,
Sebastian
P
,
Carracedo
C
,
Lokanatha
D
,
Bourhis
J
, et al
Effects of lapatinib monotherapy: results of a randomised phase II study in therapy-naive patients with locally advanced squamous cell carcinoma of the head and neck
.
Br J Cancer
2011
;
105
:
618
27
.
35.
Quesnelle
KM
,
Grandis
JR
. 
Dual kinase inhibition of EGFR and HER2 overcomes resistance to cetuximab in a novel in vivo model of acquired cetuximab resistance
.
Clin Cancer Res
2011
;
17
:
5935
44
.
36.
de Souza
JA
,
Davis
DW
,
Zhang
Y
,
Khattri
A
,
Seiwert
TY
,
Aktolga
S
, et al
A phase II study of lapatinib in recurrent/metastatic squamous cell carcinoma of the head and neck
.
Clin Cancer Res
2012
;
18
:
2336
43
.
37.
Uno
M
,
Otsuki
T
,
Kurebayashi
J
,
Sakaguchi
H
,
Isozaki
Y
,
Ueki
A
, et al
Anti-HER2-antibody enhances irradiation-induced growth inhibition in head and neck carcinoma
.
Int J Cancer
2001
;
94
:
474
9
.
38.
Schrader
C
,
Boehm
A
,
Reiche
A
,
Diet
A
,
Mozet
C
,
Wichmann
G
. 
Combined effects of lapatinib and cisplatin on colony formation of head and neck squamous cell carcinoma
.
Anticancer Res
2012
;
32
:
3191
9
.
39.
Rusnak
DW
,
Alligood
KJ
,
Mullin
RJ
,
Spehar
GM
,
Renas-Elliott
C
,
Martin
AM
, et al
Assessment of epidermal growth factor receptor (EGFR, ErbB1) and HER2 (ErbB2) protein expression levels and response to lapatinib (Tykerb, GW572016) in an expanded panel of human normal and tumour cell lines
.
Cell Prolif
2007
;
40
:
580
94
.
40.
Kondo
N
,
Tsukuda
M
,
Ishiguro
Y
,
Kimura
M
,
Fujita
K
,
Sakakibara
A
, et al
Antitumor effects of lapatinib (GW572016), a dual inhibitor of EGFR and HER-2, in combination with cisplatin or paclitaxel on head and neck squamous cell carcinoma
.
Oncol Rep
2010
;
23
:
957
63
.
41.
Arkhipov
A
,
Shan
Y
,
Kim
ET
,
Dror
RO
,
Shaw
DE
. 
Her2 activation mechanism reflects evolutionary preservation of asymmetric ectodomain dimers in the human EGFR family
.
Elife
2013
;
2
:
e00708
.
42.
Lenferink
AE
,
Pinkas-Kramarski
R
,
van de Poll
ML
,
van Vugt
MJ
,
Klapper
LN
,
Tzahar
E
, et al
Differential endocytic routing of homo- and hetero-dimeric ErbB tyrosine kinases confers signaling superiority to receptor heterodimers
.
EMBO J
1998
;
17
:
3385
97
.
43.
Lenferink
AE
,
Busse
D
,
Flanagan
WM
,
Yakes
FM
,
Arteaga
CL
. 
ErbB2/neu kinase modulates cellular p27(Kip1) and cyclin D1 through multiple signaling pathways
.
Cancer Res
2001
;
61
:
6583
91
.
44.
Tzahar
E
,
Waterman
H
,
Chen
X
,
Levkowitz
G
,
Karunagaran
D
,
Lavi
S
, et al
A hierarchical network of interreceptor interactions determines signal transduction by Neu differentiation factor/neuregulin and epidermal growth factor
.
Mol Cell Biol
1996
;
16
:
5276
87
.
45.
Holbro
T
,
Civenni
G
,
Hynes
NE
. 
The ErbB receptors and their role in cancer progression
.
Exp Cell Res
2003
;
284
:
99
110
.
46.
Holbro
T
,
Beerli
RR
,
Maurer
F
,
Koziczak
M
,
Barbas
CF
 III
,
Hynes
NE
. 
The ErbB2/ErbB3 heterodimer functions as an oncogenic unit: ErbB2 requires ErbB3 to drive breast tumor cell proliferation
.
Proc Natl Acad Sci U S A
2003
;
100
:
8933
8
.
47.
Wolff
A
,
Hammond
E
,
Hicks
D
,
Dowsett
M
,
McShane
L
,
Allison
K
, et al
Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update
.
J Clin Oncol
2013
;
31
:
3997
4013
.
48.
Ruschoff
J
,
Hanna
W
,
Bilous
M
,
Hofmann
M
,
Osamura
R
,
Penault-Llorca
F
, et al
HER2 testing in gastric cancer: a practical approach
.
Mod Pathol
2012
;
25
:
637
50
.
49.
Azemar
M
,
Schmidt
M
,
Arlt
F
,
Kennel
P
,
Brandt
B
,
Papadimitriou
A
, et al
Recombinant antibody toxins specific for ErbB2 and EGF receptor inhibit the in vitro growth of human head and neck cancer cells and cause rapid tumor regression in vivo
.
Int J Cancer
2000
;
86
:
269
75
.
50.
Assadi
M
,
Lamerz
J
,
Jarutat
T
,
Farfsing
A
,
Paul
H
,
Gierke
B
, et al
Multiple protein analysis of formalin-fixed and paraffin-embedded tissue samples with reverse phase protein arrays
.
Mol Cell Proteomics
2013
;
12
:
2615
22
.
51.
Sato-Kuwabara
Y
,
Neves
JI
,
Fregnani
JH
,
Sallum
RA
,
Soares
FA
. 
Evaluation of gene amplification and protein expression of HER-2/neu in esophageal squamous cell carcinoma using Fluorescence in situ Hybridization (FISH) and immunohistochemistry
.
BMC Cancer
2009
;
9
:
6
.
52.
Wheeler
DL
,
Huang
S
,
Kruser
TJ
,
Nechrebecki
MM
,
Armstrong
EA
,
Benavente
S
, et al
Mechanisms of acquired resistance to cetuximab: role of HER (ErbB) family members
.
Oncogene
2008
;
27
:
3944
56
.
53.
Hudis
CA
. 
Trastuzumab–mechanism of action and use in clinical practice
.
N Engl J Med
2007
;
357
:
39
51
.
54.
Kumler
I
,
Tuxen
MK
,
Nielsen
DL
. 
A systematic review of dual targeting in HER2-positive breast cancer
.
Cancer Treat Rev
2014
;
40
:
259
70
.
55.
Tse
C
,
Gauchez
AS
,
Jacot
W
,
Lamy
PJ
. 
HER2 shedding and serum HER2 extracellular domain: Biology and clinical utility in breast cancer
.
Cancer Treat Rev
2011
;
38
:
133
42
.
56.
Gillison
ML
,
Glisson
BS
,
O'Leary
E
,
Murphy
BA
,
Levine
MA
,
Kies
MS
, et al
Phase II trial of trastuzumab (T), paclitaxel (P) and cisplatin (C) in metastatic (M) or recurrent (R) head and neck squamous cell carcinoma (HNSCC): response by tumor EGFR and HER2/neu status
.
J Clin Oncol
24
:
18s
, 
2006
(
suppl; abstr 5511
).
57.
Metzger-Filho
O
,
Winer
EP
,
Krop
I
. 
Pertuzumab: optimizing HER2 blockade
.
Clin Cancer Res
2013
;
19
:
5552
6
.
58.
Baselga
J
,
Gelmon
KA
,
Verma
S
,
Wardley
A
,
Conte
P
,
Miles
D
, et al
Phase II trial of pertuzumab and trastuzumab in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer that progressed during prior trastuzumab therapy
.
J Clin Oncol
2010
;
28
:
1138
44
.
59.
Lee-Hoeflich
ST
,
Crocker
L
,
Yao
E
,
Pham
T
,
Munroe
X
,
Hoeflich
KP
, et al
A central role for HER3 in HER2-amplified breast cancer: implications for targeted therapy
.
Cancer Res
2008
;
68
:
5878
87
.
60.
Erjala
K
,
Sundvall
M
,
Junttila
TT
,
Zhang
N
,
Savisalo
M
,
Mali
P
, 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
.
Clin Cancer Res
2006
;
12
:
4103
11
.
61.
Medina
PJ
,
Goodin
S
. 
Lapatinib: a dual inhibitor of human epidermal growth factor receptor tyrosine kinases
.
Clin Ther
2008
;
30
:
1426
47
.
62.
Li
D
,
Ambrogio
L
,
Shimamura
T
,
Kubo
S
,
Takahashi
M
,
Chirieac
LR
, et al
BIBW2992, an irreversible EGFR/HER2 inhibitor highly effective in preclinical lung cancer models
.
Oncogene
2008
;
27
:
4702
11
.
63.
Harbeck
N
,
Solca
F
,
Gauler
TC
. 
Preclinical and clinical development of afatinib: a focus on breast cancer and squamous cell carcinoma of the head and neck
.
Future Oncol
2014
;
10
:
21
40
.
64.
Williams
J
,
Kim
I
,
Ito
E
,
Shi
W
,
Yue
S
,
Siu
L
, et al
Pre-clinical characterization of dacomitinib (PF-00299804), an irreversible pan-ErbB inhibitor, combined with ionizing radiation for head and neck squamous cell carcinoma
.
PLoS ONE
2014
;
9
:
e9855
.
65.
Harrington
KJ
,
Temam
S
,
D'Cruz
A
,
Jain
MM
,
D'Onofrio
I
,
Manikhas
G
, et al
Final analysis: A randomized, blinded, placebo (P)-controlled phase III study of adjuvant postoperative lapatinib (L) with concurrent chemotherapy and radiation therapy (CH-RT) in high-risk patients with squamous cell carcinoma of the head and neck (SCCHN)
.
J Clin Oncol
32
:
5s
, 
2014
(
suppl; abstr 6005
).
66.
Seiwert
TY
,
Fayette
J
,
Cupissol
D
,
Del Campo
JM
,
Clement
PM
,
Hitt
R
, et al
A randomized, phase 2 study of afatinib versus cetuximab in metastatic or recurrent squamous cell carcinoma of the head and neck
.
Ann Oncol
2014
;
25
:
1813
20
.