touchONCOLOGY touchONCOLOGY
Head and Neck Cancer
Read Time: 5 mins

Targeting the ErbB Family in Head and Neck Cancer

Copy Link
Published Online: Jul 20th 2011 European Oncological Disease, 2007;1(2):35-40 DOI: https://doi.org/10.17925/EOH.2007.0.2.35
Authors: Pol M Specenier, Jan B Vermorken
Quick Links:
Article
Article Information
Article:

Head and neck cancer is the sixth most common cancer, representing about 5% of all cancers. Each year, more than half a million new cases are diagnosed worldwide.1–3 Over the past decade, the important role of different growth factors and their receptors and signal transduction pathways in the genesis and progression of tumours has been well recognised, and their mechanism of action and interaction is gradually being unravelled.4–10 The ErbB family comprises four members: human epidermal growth factor receptor (EFGR; HER 1), HER2 (c-erb-B2), HER3 (cerb- B3) and HER4 (c-erb-B4).
Epidermal Growth Factor Receptor
EGFR and its ligand-transforming growth factor-alpha (TGF-α) are overexpressed in the vast majority of cases of squamous cell carcinoma of the head and neck (SCCHN). EGFR overexpression is an early event in carcinogenesis and steadily increases in parallel with histological abnormalities from hyperplasia, through carcinoma in situ to invasive carcinoma.11 EGFR overexpression is the result of an enhanced transcription or a gene amplification.12,13 There is a large body of evidence indicating that EGFR overexpression is a strong, independent and unfavourable prognostic factor for loco-regional control, disease-free survival and overall survival in SCCHN.14–21 Two of the potential EGFR-targeting strategies are currently in use. Monoclonal antibodies (MAbs) are directed at the extracellular domain of the receptor and at the small-molecule and adenosine tri-phosphate (ATP)-competitive tyrosine kinase inhibitors (TKIs) (see Table 1).22–29
Monoclonal Antibodies
MAbs targeting EGFR that are under clinical development include IMC225 (cetuximab), EMD-72000 (matuzumab), ABX-EGF (panitumumab), h-R3 (nimotuzumab), 2F8 (zalutumumab) and ICR62.22 The first MAb to enter the clinic was cetuximab (IMC225). Pre-clinically, it has antitumoral activity on its own and acts synergistically with chemotherapy and radiotherapy.25,26
Cetuximab
Cetuximab is a chimeric human/murine immunoglobulin IgG1 antibody that binds with higher affinity to the EGFR than the natural ligands EGF and tumour necrosis factor-alpha (TNF-α). The cetuximab-bound EGFR is internalised and degraded without receptor phosphorylation and activation. The availability of EGFR on the cell surface is reduced and the receptor downregulated. The most common side effects are fever, chills, asthenia, nausea, transaminase elevation and skin toxicity.25,26 A recently published large, multicentre, international phase III trial30 randomised 424 patients with stage III or IV non-metastatic squamous cell carcinoma of the oropharynx, hypopharynx or larynx to radiotherapy alone or to radiotherapy in combination with weekly cetuximab. There was a significant increase in the median duration of loco-regional control (24.4 versus 14.9 months; p=0.005) and the median duration of survival (49 versus 29.3 months; p=0.03). The only differences in grade 3/4 acute toxicity were acneiform rash and infusion reactions, which occurred in 17 and 3% of patients, respectively. In particular, the grade 3/4 radiationrelated
mucosal toxicity was no worse in the cetuximab arm (54 versus 52%). The trial was conceived before the publication of a meta-analysis that clearly established the superiority of cisplatin-based chemoradiation over radiotherapy alone as definitive treatment for loco-regionally advanced SCCHN. Thus, the control arm used in the trial can no longer be considered the current standard of care for this population.

Cetuximab was added to cisplatin and concomitant boost radiotherapy in a pilot phase II study conducted at the Memorial Sloan-Kettering Cancer Center.31 Twenty-two previously untreated patients with stage III or IV non-metastatic SCCHN were enrolled. The study was closed before it reached planned accrual because of toxicity concerns. Two patients died while still on study (one from pneumonia and one from an unknown cause). After that, three more non-fatal but serious adverse events occurred (one bacteraemia, one atrial fibrillation and one myocardial infarction), prompting study closure. However, three-year overall survival, progression-free survival (PFS) and loco-regional control rates were 76, 5 and 71%, respectively. Despite the early deaths, outcome compared favourably with those reported in the literature for patients with locally advanced SCCHN who were treated with chemoradiation alone.
Moreover, in retrospect the significant adverse events were not clearly attributable to the investigational therapy. Therefore, although this regimen could not be recommended outside a clinical trial, the results were compelling enough to launch a randomised phase III Radiation Therapy Oncology Group (RTOG) study comparing this regimen with standard cisplatin-based chemoradiation. This trial will hopefully help to elucidate the question of how cetuximab fits into the current treatment strategies for loco-regionally advanced SCCHN. Kies et al.32 added cetuximab to weekly paclitaxel 135mg/m2 and carboplatin area under the curve (AUC) 2 as induction regimen in treatment-naïve locally advanced SCCHN. After six weekly administrations of the regimen all patients had a response, with 27% of these complete responses. Severe skin toxicity occurred in 47% of the patients. Severe skin reactions were observed by others when cetuximab was added to a taxane-containing triplet (Vermorken, personal communication).
Cetuximab has been studied extensively in recurrent and metastatic disease, both in first- and second-line treatment (see Table 2). In terms of first-line treatment, Burtness et al.33 randomised 117 patients with recurrent or metastatic SCCHN to cisplatin plus placebo or cisplatin plus cetuximab. No prior chemotherapy for recurrent and/or metastatic disease had been given to these patients. Objective response rate was 26% in the cetuximab arm and 10% in the placebo arm. PFS was longer in the cetuximab arm (4.2 versus 2.7 months). However, this difference was not statistically significant. This was probably due to the longer than expected PFS in the control arm, which resulted in a lower than intended power of the study to detect a statistically significant difference. There was no difference in overall survival between the two arms for the entire study population. However, there was a survival benefit for the subset of patients who developed cetuximab-related skin toxicity. Bourhis et al.34 added cetuximab to carboplatin AUC 5 or cisplatin 100mg/m2 on day one followed by a continuous infusion of 5-fluorouracil (5-FU) at escalating doses of 600, 800 and 1,000mg/m2/day. Eligible patients had recurrent and/or metastatic SCCHN and had not received prior chemotherapy for this disease setting. Fifty-three patients were included in this phase I/II study.
The dose of 1,000mg/m2 of 5-FU was judged to be safe and tolerable and was recommended for further studies. The overall response rate was 36%. Median time to progression in this latter study was 5.1 months and median overall survival was 9.8 months. The benefit of this combined approach (cetuximab plus platinum/5-FU) was studied in a direct comparison with platinum/5-FU alone in a large phase III trial (Erbitux in First-line Treatment of Recurrent or Metastatic Head and Neck Cancer [EXTREME] study).35 The addition of cetuximab prolongs overall survival (Vermorken, personal communication). Four hundred and forty-two patients were randomised to receive either chemotherapy alone (cisplatin 100mg/m2 or carboplatin AUC 5 on day one followed by 5-fluorouracil 1,000mg/m2/day for four days) or the same regimen combined with weekly cetuximab. Cycles were repeated every three weeks for a maximum of six cycles. Thereafter, in the combined arm cetuximab was continued as a single agent until disease progression or unacceptable toxicity – whichever came first. No cross-over was permitted in this study. Patients who had received prior chemotherapy were excluded, except when this had been part of their primary treatment, provided this chemotherapy was ended at least six months before inclusion in the study.

The primary end-point was overall survival. The study was designed to detect an increase in median survival from 7.5 to 9.5 months. The addition of cetuximab increased the median overall survival by 2.7 months from 7.4 to 10.1 months (hazard ratio 0.797, p=0.036). With the addition of cetuximab, the response rate increased from 19.5 to 35.6% (p<0.0001) and the median PFS was prolonged by 2.3 months from 3.3 to 5.6 months (p<0.0001). The addition of cetuximab did not modify the characteristic adverse event profile of platinum-based chemotherapy and did not have a negative impact on quality of life.
As for second-line treatment, three phase II trials examined the role of cetuximab in platinum-resistant disease.36–38 Baselga et al.36 addedweekly cetuximab to platinum-based chemotherapy in 96 patients with platinum-refractory SCCHN. The objective response rate in this multicentre phase II study was 10% and the median time to progression and overall survival were 85 and 183 days, respectively. Herbst et al.37 studied the combination of cetuximab and chemotherapy in a heterogenous population of 130 patients with recurrent and/or metastatic SCCHN. Patients either had stable disease after two cycles or had progressed under cisplatin-based chemotherapy. After the addition of cetuximab to the same regimen, 13% of the patients achieved a partial response. Median duration of response was about four months in the patients with progressive disease at the time they entered the study and 7.4 months in those who had stable disease. Responses in these two studies could be attributed to the reversal of cisplatin resistance or to the effect of cetuximab alone. The results of the third phase II trial38 suggest that the latter may be the case. In a population of 103 patients with platinum-resistant disease, a response rate of 13% and a median time to progression and survival of 2.3 and 5.9 months, respectively, were observed. These results are not dissimilar to the results of the add-on trials. Interestingly, the survival of around six months achieved with cetuximab in platinum-refractory disease was similar to that seen with first-line therapy without cetuximab and represented an increase in survival of 2.5 months compared with platinum-refractory historical controls.39,40 Based on these results, cetuximab monotherapy seems to be a good option in patients with recurrent/metastatic SCCHN who have progressed on platinum-based chemotherapy.
Other Monoclonal Antibodies
Matuzumab (EMD72000)25,41–43 is a humanised IgG1 mouse anti-EGFR antibody that is in phase II testing in SCCHN. In phase I, the maximum tolerated dose was 1,600mg/week, with headache and fever as doselimiting toxicities and acneiform rash being the most common grade 1 or 2 toxicity. It has a prolonged half-life that allows for administration every two or three weeks. The optimal biological dose was 1,200mg every three weeks. Panitumumab (ABX-EGF) is a human IgG2 antibody.25 It blocks ligand binding and induces internalisation of the receptor with no receptor degradation. Side effects include pruritus, skin rash, dyspnoea, fatigue, abdominal pain, asthenia and diarrhoea. Panitumumab at a weekly dose of 2.5mg/kg has an acceptable tolerability and encouraging clinical activity in patients with a variety of tumour types.44 Panitumumab is being studied as a single agent, in combination with chemotherapy in recurrent or metastatic disease and in combination with induction chemotherapy and chemoradiation in locally advanced SCCHN.
Nimotuzumab (h-R3)45,46 is a humanised IgG1 mouse antibody. Preliminary data suggest that therapeutic levels of h-R3 can be achieved without eliciting skin toxicity. This is the most common side effect of the other anti- EGFR directed antibodies. In phase I studies h-R3 was added to radiation in patients with locally advanced SCCHN. The optimal dose for further phase II testing was determined to be 200mg/m2.45,46 Zalutumumab (2F8)47 is a fully human IgG1 EGFR-directed MAb that also exerts antibodydependent cell-mediated cytotoxicity. The frequency of the acneiform skin rash observed with this antibody increases with dose. In a phase I/II trial by the Danish Head and Neck Cancer Study Group (DAHANCA), no doselimiting toxicities were observed at doses of up to 8mg/kg/week.
Metabolic responses, as evaluated by fluorodeoxyglucose (FDG) positron emission tomography (PET), were observed in six of the 15 evaluable patients and response evaluation by computed tomography (CT) showed a partial response in two of the 16 evaluable patients.47 Zalutumumab is currently undergoing phase III testing in patients with cisplatin refractory disease. The rat EGFR-directed antibody ICR62 is a rat MAb to the EGFR.48

A phase I clinical trial with a single dose of ICR62 demonstrated that this antibody could be administered safely to patients with SCCHN and nonsmall- cell lung cancer (NSCLC). Four out of 20 patients developed human antirat antibodies. ICR62 could be found in metastatic tumour sites in biopsies taken 24 hours after administration. MDX-447 (Humax®)49,50 is a bispecific antibody comprising humanised Fab anti-CD64 and humanised Fab anti-EGFR. MDX-447 recognises the CD64 receptor and directs the cytotoxic effector cells to the EGFR-expressing tumour cells. The dose-limiting toxicity is hypotension.
Epidermal Growth Factor Receptor-targeted Tyrosine Kinase Inhibitors
Tyrosine kinase inhibitors (TKIs) compete with ATP for the cytoplasmatic catalytic domain of EGFR. Gefitinib and erlotinib are reversible specific EGFR TKIs and belong to the group of quinazole TKIs. This group also includes: PD153035 and GW 572016 (lapatinib), which are reversible dual EGFR/HER-2 inhibitors; EKB-569, which irreversibly inhibits the EGFR and HER-2 TK; and the irreversible pan-ErbB TKI CI 1033 (canertinib). PKI-166 (dual EGFR/ErbB-2) belongs to the pyrrolotriazine TKIs, which also include AEE788 (dual EGFR/ErbB-2) and BMS 599626. ARRY-334543 (dual EGFR/ErbB-2) and PD1578 belong to the pyridopyrimidine TKIs.25 In a phase II trial in 52 patients51 with recurrent or metastatic disease, 85% of whom had received prior chemotherapy, the overall response rate with gefitinib 500mg/day was 10.6%. The only grade 3 toxicity was diarrhoea, which occurred in 6% of the patients. Median time to progression was 3.4 months and median overall survival was 8.1 months. Forty-seven patients with locally recurrent and/or metastatic SCCHN were enrolled in an expanded access programme of gefitinib at the Royal Marsden and St George’s hospitals.52 Patients were eligible if they had received prior radiotherapy and/or chemotherapy or were medically unfit for chemotherapy. The patients received 500mg/day and the only grade 3 toxicity was folliculitis, which occurred in 8%. The observed clinical response rate was 8%. Median time to progression was 2.6 months and median overall survival was 4.3 months.
Wheeler et al.53 treated 20 patients who had received only chemotherapy as their primary management for localised disease with gefitinib 500mg/day and observed three objective responses (15%), with a median response duration of six months. There were no objective responses in 12 patients with chemotherapy-refractory disease. Grade 3 toxicities were skin rash and gastrointestinal, and were observed in eight of the 32 patients. There was no association between rash and response. Median time to progression was three months and median overall survival was six months. In a multicentre phase II trial in the US, 70 patients54 with recurrent and/or metastatic SCCHN were treated with gefitinib 250mg/day. Grade 3 toxicities included diarrhoea (2%), rash (1%), pulmonary toxicity (1%) and nausea (1%).
Median PFS was 1.8 months. The response rate of 1.4% contrasted unfavourably with the response rates observed with a daily dose of 500mg in the prior studies, suggesting that there may be a dose–response relationship for gefitinib in SCCHN. This latter aspect was tested in a large phase III trial (1839 IL/0704).55 In the trial, 486 patients with recurrent and/or metastatic SCCHN unresponsive to or unfit for platinum were randomised to receive weekly methotrexate, gefitinib 250mg/day orgefitinib 500mg/day. Overall response rates were 3.9, 2.7% and 7.6%, respectively. Median overall survival times were 6.7, 5.6 and 6 months, respectively (p=0.39). Soulieres et al.56 treated 115 patients with recurrent and/or metastatic SCCHN with erlotinib at an initial dose of 150mg/day. Ninety-seven per cent of these had previously been irradiated and 37% had received prior chemotherapy. The most common drugrelated toxicities were rash and diarrhoea, which was grade 1 or 2 in the vast majority of cases. Response rate was 4.3%. Median PFS was 9.6 weeks and median overall survival was six months. No difference was detected based on HER1/EGFR expression, but patients who developed at least grade 2 skin rash had a significantly better survival (see Table 3).

The optimal way of integrating the TKI into the current treatment strategies for head and neck cancer has yet to be determined, taking into consideration the experience with EGFR TKIs in NSCLC, where adding erlotinib or gefitinib to standard chemotherapy did not improve the outcome compared with chemotherapy alone.57–59 The radiosensitising and chemosensitising properties of gefitinib are sequence-dependent in several SCCHN lines.60–62 Synergy was observed when gefitinib was applied before radiation or before or during cisplatin and 5-FU. However, antagonism occurred when gefitinib application followed chemotherapy.60 Other investigators detected synergism when gefitinib was applied during or after docetaxel or gemcitabine, but antagonism in some cell lines when gefitinib preceded docetaxel.61,62 Induction chemotherapy followed by concomitant chemoradiation is a treatment paradigm that has recently gained much attention. Cohen et al.63 treated 69 patients with stage III or IV SCCHN with two cycles of induction chemotherapy (carboplatin and paclitaxel) followed by an alternating chemotherapy and radiation regimen and added gefitinib 250mg/day. After chemoradiation, gefitinib was continued for two years. Thirty-seven of the 42 evaluable patients (88%) had a complete response. Grade 3/4 toxicities were consistent with prior chemoradiation trials using the same regimen. After a median follow-up of 294 days, two patients had progressive disease and four patients had died.
Doss et al.64 added gefitinib 250mg/day to an induction regimen of docetaxel 60mg/m2 and carboplatin (AUC 5) given on days one and 22, 5-FU given as a continuous infusion at a dose of 200mg/m2 on days one to 43. Beginning on day 43, patients received radiotherapy (68.4 in daily fractions of 1.8Gy) concurrently with docetaxel 20mg/m2/week and gefitinib 250mg/day. Forty-five patients entered the study. Grade 3/4 myelosuppression was common and all patients experienced grade 3/4 mucositis during the combined modality programme. There was one treatment-related death. Thirty-five patients had completed the regimen and were evaluable for response. Response was evaluated by CT scan and endoscopy. There was a complete response in 32% of patients and a partial response in 53%. Erlotinib 150mg/day can be safely combined with concurrent chemoradiation and weekly docetaxel 20mg/m2,65 or cisplatin 100mg/m266 every three weeks, without jeopardising timely administration of radiotherapy. Complete response rates were 83 and 84%, respectively. Phase II trials are planned or ongoing. However, as there are data suggesting that when erlotinib is added to weekly docetaxel lowering of the dose of both drugs may be necessary,67 caution is still warranted when combining chemotherapy and TKIs.
In a so-called ‘window of opportunity’ study, Delord et al.68 treated 35 patients who had resectable SCCHN with erlotinib 150mg/day for the 14–27 days between diagnosis and surgery. As expected, rash and diarrhoea were the most frequent side effects. Twenty-five subjects were assessable for efficacy, of whom 36% responded (all partial responses). No delay in surgery was observed. Normal and tumour tissue biopsies were taken before and after treatment. Laboratory studies included EGFR TK mutation analysis and immunohistochemistry, including EGFR, phosphorylated-EGFR, phosphotyrosine (p-Tyr), phospho-Akt (p-Akt), signal transducer and activator of transcription 3 (STAT3), phosphorylated extracellular signal-regulated kinase (pERK) 1/2, p21 and p27. No mutations in the catalytic domain of the EGFR TK were found.
Interestingly, p21 expression was found to predict response to erlotinib (p=0.0002). Therefore, this trial demonstrated a useful prospective method to potentially aid patient selection and improve the design of post-operative adjuvant treatment combinations. In vitro and in vivo data suggest that combined targeting of the extracellular ligand-binding domain by an anti-EGFR antibody and the intracellular receptor kinase domain by a TKI has a complementary effect on downstream signalling, apoptosis, proliferation and tumour xenograft growth.69,70

Epidermal Growth Factor Receptor Mutations
The search for EGFR mutations is an area of active investigation. An EGFR mutation of particular importance is EGFRvIII. It is a ligand-independent, constitutively phosphorylated form of EGFR with enhanced tumourigenicity and radioresistance in pre-clinical models.71–73 EGFRvIII involves deletion of exon 2-7A and is frequently present in glioblastoma. It was detected by Sok et al.74 in 14 out of 33 SCCHN specimens by immunostaining and reverse transcription polymerase chain reaction (PCR) for EGFRvIII expression. An EGFRvIII transfected SCCHN cell line showed an increased proliferation and growth in vitro and in vivo, decreased apoptosis in response to cisplatin and decreased growth inhibition by cetuximab compared with vector transfected controls. Somatic activating mutations in the EGFR kinase domain were first described in NSCLC.75,76 In NSCLC, EGFR mutations in the TK domain are more frequent in never-smokers, females, patients of East Asian origin and patients with adenocarcinoma. Eighty-six per cent of the EGFR mutations in NSCLC occur in exon 19 or 21. The most common mutations are deletions in exon 19 and a missense mutation in exon 21 (L858R). The exon 19 deletions L858R, L861Q and G719A/C/S are consistently associated with in vitro and in vivo sensitivity to gefitinib and erlotinib, while exon 20 insertions and the T790M mutation are associated with resistance to gefitinib and erlotinib. The significance of other mutations is not yet entirely clear.77–82 In vitro NSCLC cell lines harbouring EGFR mutations were more sensitive to gefitinib, but their sensitivity to cetuximab was similar to that of EGFR wild-type cell lines.83 Gefitinib-sensitising EGFR mutations have been described in an oesophageal cancer cell line84 and in oesophageal tumours.85
The incidence of EGFR mutations in SCCHN is currently not known and varies from series to series.86–92 Lee et al.86 analysed the EGFR gene in 41 SCCHN and detected three (7.3%) deletions in exon 19 in a Korean population. Willmore-Payne87 detected two mutations in 24 tumours of the head and neck, one exon 20-activating mutation (N771VinsG) and one exon 19-activating mutation (G729E). Loeffler-Ragg88 detected an EGFR missense mutation in one out of 100 tumour samples of SCCHN patients. In a series of 82 patients, of whom eight had a gefitinib-responsive tumour, Cohen et al.90 did not find any EGFR TK-activating mutation. They did detect one ERbB2 mutation within exon 20. Also, Delord et al.68 did not find any mutation in 35 patients with newly diagnosed resectable SCCHN. Moreover, Moijca90 did not detect any mutation in exon 19 or 21 in 11 SCCHN tumour specimens at Roswell Park Cancer Insititute. The exact incidence of specific mutations and their possible predictive role in SCCHN have yet to be determined. Preliminary data suggest that EGFR mutations may be associated with a better outcome in tongue and tonsil cancer.92
Human Epidermal Growth Factor 2
HER2 (or ErbB2) is expressed in 40–60% of SCCHN and is also associated with a worse outcome.93 The ErbB2-directed antibody trastuzumab was added to paclitaxel and carboplatin in a phase II study that included patients with metastatic or recurrent SCCHN.94 The response rate (36%) was not higher than expected with this chemotherapy regimen alone. Lapatinib is a selective and potent dual competitive reversible inhibitor of the EGFR and ErbB2 TK.25 As a single agent, lapatinib is well tolerated but appears to have little, if any, activity in either EGFR inhibitor-naïve or -refractory patients with recurrent or metastatic SCCHN.95 Abidoye et al.95 did not observe any objective responses in 42 patients and found a median PFS of six weeks. The combination of lapatinib with conventional radiation (70Gy) and cisplatin 100mg/m2 every three weeks was tested in a phase I dose-escalating study.96 The optimal dose was determined to be 1,500mg/day. Seventeen of the 22 patients were evaluable and 74% achieved a complete response. The others had a partial response. Toxicities were no different to those typically observed with concurrent cisplatin and radiotherapy.

Combinations of Targeting Agents
Cyclo-oxygenase 2 (COX-2) expression was found to be an independent prognostic predictor of disease-free survival in some series of oral squamous cell carcinoma.97–105 EGFR and COX-2 signalling pathways form a positive feedback loop.106–107 The EGFR ligands EGF and TGF-α induce COX-2 expression, and COX-2 induces transactivation or increased EGFR expression. The combination of the TKI AG1478 or gefitinib and celecoxib inhibits, either additively or synergistically, the growth of five SCCHN cell lines.108,109 EGFR TKIs and COX-2 inhibitors have a non-overlapping toxicity profile. The combination of celecoxib and gefitinib was tested in a phase I dose-finding study at the Dana Farber Cancer Institute.110 All patients had previously been treated with chemotherapy or radiochemotherapy. At doses of 400 and 500mg twice-daily, no dose-limiting toxicities were encountered. The incidence of diarrhoea and rash was similar to the reported incidence of these toxicities in phase II studies of gefitinib 250 or 500mg/day in SCCHN. The response rate was 22%. The median duration of response was 4.8 months; the median progression and overall survival were three and six months, respectively. The majority of studies examining the prognostic significance of vascular endothelial growth factor (VEGF) expression observed a worse outcome in patients with SCCHN expressing VEGF and VEGF receptor-2.111 Mauer et al.112 combined erlotinib 150mg/day with the humanised VEGF-A directed antibody bevacizumab, which was administered every three weeks in doses increasing from 5 to 15mg/kg. There were no dose-limiting toxicities. After two cycles of treatment, seven of the nine evaluable patients had stable disease and a partial response was documented in one. The dose of bevacizumab could be safely raised to 15mg/m2 every three weeks. This dose was subsequently taken into a phase II study. Preliminary data on 28 evaluable patients were reported.113 Five out of 28 patients had a response (two complete responses and three partial responses). Median progression-free survival was 127 days and median overall survival was 226 days. Pre-clinical data114 suggest synergy when gefitinib and irradiation are combined with AZD2171, a highly potent, orally active, VEGF signalling inhibitor.
Conclusion
EGFR is an obvious target for therapy as it is overexpressed in virtually all SCCHN. The EGFR-directed MAb cetuximab has activity as a single agent in platinum-resistant disease and acts synergistically with radiotherapy and first-line platinum-based chemotherapy. Thus far, the results with EGFR TKIs are less promising. Areas of investigation are the integration of targeted therapies into existing treatment strategies and the combined use of agents targeting the same pathway at multiple levels or targeting multiple pathways. â– 

References

  1. Ferlay J, Bray F, Pisani P, et al., GLOBOCAN 2002: Cancer Incidence, Mortality and Prevalence Worldwide, IARC CancerBase, 2004, IARC Press.
  2. Jemal ZA, Tiwari RC, Murray T, et al., American Cancer Society, CA Cancer J Clin, 2004;54:8–29.
  3. Vermorken JB, Medical treatment in head and neck cancer, Ann Oncol, 2005;16:S258–64.
  4. Cohen S, The stimulation of epidermal proliferation by a specific protein (EGF), Dev Biol, 1965;12:394–407.
  5. Vermorken JB, Hitt R, Geoffrois L, et al., Cetuximab plus platinum-based therapy first-line in recurrent and/or metastatic squamous cell carcinoma of the head and nexk: efficacy and safety results of a randomized phase III trial (EXTREME), Eur J Cancer, 2007;43(Suppl. 4):abstract 5501.
  6. Hynes NE, Lane HA, ERBB receptors and cancer: the complexity of targeted inhibitors, Nat Rev Cancer, 2005;5:341–54.
  7. Pal SK, Pegram M, Epidermal Growth Factor Receptor and signal transduction: potential targets for anticancer therapy, Anticancer Drugs, 2005;16:483–94.
  8. Yarden Y, Sliwkowski MX, Untangling the ErbB signalling network, Nat Rev Cancer, 2001;2:127–37.
  9. Schlessinger J, Cell signalling by receptor tyrosine kinases, Cell, 2000;103:211–25.
  10. Zimmerman M, Zouhair A, Azria D, et al., The epidermal growth factor receptor (EGFR) in head and neck cancer: its role and treatment implications, Radiat Oncol, 2006;1:11.
  11. Grandis JR, Tweardy DJ, Elevated levels of transforming growth factor receptor-alpha and epidermal growth factor receptor messenger RNA are early markers of carcinogenesis in head and neck cancer, Cancer Res, 1993;53:3579–84.
  12. Chung CH, Ely K, L 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;24:4,170–76.
  13. Mrhalova M, Plzak J, Betka J, et al., Epidermal growth factor receptor – its expression and copy numbers of EGFR gene in patients with head and neck squamous cell carcinomas, Neoplasma, 2005;52:338–43.
  14. Nicholson RI, Gee JMW, Harper ME, EGFR and cancer prognosis, Eur J Cancer, 2001;37:S9–15.
  15. Demiral AN, Sarioglu S, Birlik B, et al., Prognostic significance of EGF receptor expression in early glottic cancer, Auris Nasus Larynx, 2004;31:417–24.
  16. 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;62:7350–56.
  17. Grandis JR, Melhem MF, Gooding WE, et al., Levels of TGF and EGFR protein in head and neck squamous cell carcinoma and patient survival, J Natl Cancer Inst, 1998;90:824–32.
  18. Grandis JR, Prognostic biomarkers in head and neck cancer, Clin Cancer Res, 2006;2:5005–6.
  19. Dassonville O, Formento JL, Francoul M, et al., Expression of epidermal growth factor receptor and survival in upper aerodigestive tract cancer, J Clin Oncol, 1993;11:1873–8.
  20. Maurizi M, Almadori G, Ferrandina G, et al., Prognostic significance of epidermal growth factor receptor in laryngeal squamous cell carcinoma, Br J Cancer, 1996;74:1253–7.
  21. Smid EJ, Stoter TR, Bloemena E, et al., The importance of immunohistochemical expression of EGFR in squamous cell carcinoma of the oral cavity treated with surgery and postoperative radiotherapy, Int J Radiat Oncol Biol Phys, 2006;65:1323–9.
  22. Specenier P, Vermorken JB, Targeted therapies in head and neck cancer, Targ Oncol, 2007;2:73–88.
  23. Cruz JJ, Ocana A, Del Barco E, Pandiella A, Targeting tyrosine kinases and their signal transduction routes in head and neck cancer, Ann Oncol, 2007;18:421–30.
  24. El-Rayes BF, LoRusso PM, Targeting the epidermal growth factor receptor, Br J Cancer, 2004;91:418–24.
  25. Marshall L, Clinical implications of the mechanism of Epidermal Growth Factor Receptor Inhibitors, Cancer, 2006;107:1207–18.
  26. Modjtahedi H, Molecular therapy of head and neck cancer, Cancer Metastasis Rev, 2006;24:129–45.
  27. Choong NW, Cohen EEW, Epidermal growth factor receptordirected therapy in head and neck cancer, Crit Rev Oncol Hematol, 2006;57:25–43.
  28. Baselga J, Artega CL, Critical Update and Emerging Trends in Epidermal Growth Factor Receptor Targeting in Cancer, J Clin Oncol, 2005;23:2445–59.
  29. Cohen EEW, Role of epidermal growth factor receptor pathwaytargeted therapy in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck, J Clin Oncol, 2006;24:2659–65.
  30. Bonner JA, Harrari P, Giralt J, et al., Radiotherapy plus cetuximab for squamous cell carcinoma of the head and neck, N Engl J Med, 2006;354:567–78.
  31. 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;24:1072–8.
  32. Kies MS, Garden AS, Holsinger C, et al., Induction chemotherapy with weekly paclitaxel, carboplatin, and cetuximab for squamous cell carcinoma of the head and neck, J Clin Oncol, 2006;24: abstract 520.
  33. Burtness B, Goldwasser M, Flood W, et al., Phase III randomised trial of cisplatin plus placebo compared with cisplatin plus cetuximab in metastatic/recurrent head and neck cancer: an Eastern Co-operative Oncology Group Study, J Clin Oncol, 2005;23:8646–54.
  34. Bourhis J, Rivera F, Mesia R, et al., Phase I/II study of cetuximab in combination with cisplatin or carboplatin and fluorouracil in patients with recurrent or metastatic squamous cell carcinoma of the head and neck, J Clin Oncol, 2006;24:2866–72.
  35. Vermorken JB, Mesia R, Vega-Villegas ME, 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 (SCCHN) (EXTREME), J Clin Oncol, 2006;24: abstract 5537.
  36. Baselga J, Trigo JM, Bourhis J, et al., Phase II multicentre 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;24:5568–77.
  37. Herbst RS, Arquette M, Shin DM, et al., Phase II multicentre study of the epidermal growth factor antibody cetuximab and cisplatin for recurrent and refractory squamous cell carcinoma of the head and neck, J Clin Oncol, 2005;23:5578–87.
  38. Vermorken JB, Trigo J, Hitt R, et al., Open-label, uncontrolled, multicentre 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.
  39. Leon X, Hitt R, Constenla M, et al., A retrospective analysis of the outcome of patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck refractory to a platinumbased chemotherapy, Clin Oncol, 2005;17:418–24.
  40. Vermorken J, Bourhis J, Trigo J, et al., Cetuximab (Erbitux®) in recurrent/metastatic (R&M) squamous cell carcinoma of the head and neck (SCCHN) refractory to first-line platinum-based therapies, J Clin Oncol, 2005;23: abstract 5505.
  41. Bier H, Hoffman T, Hauser U, et al., Clinical trial with escalating doses of the antiepidermal growth factor receptor humanised monoclonal antibody EMD72000 in patients with advanced squamous cell carcinoma of the larynx and hypopharynx, Cancer Chemother Pharmacol, 2001;47:519–24.
  42. Vanhoefer U, Tewes M, Rojo F, et al., Phase I study of the humanised Anti-EGFR Monoconal Antibody EMD72000 in patients with advanced solid tumours that express the Epidermal Growth Factor receptor, J Clin Oncol, 2004;22:175–84.
  43. Salazar R, Tabarnero J, Rojo F, et al., Dose-dependent inhibition of the EGFR and signalling pathways with the EGFR monoclonal antibody EMD 72000 administered every three weeks. A phase I pharmacokinetic/pharmacodynamic study to define the optimal biological dose, J Clin Oncol, 2004;22: abstract 2002.
  44. Foon KA, Yang XD, Weiner LM, et al., Pre-clinical and clinical evaluations of ABX-EGF, a fully human anti-EGF receptor monoclonal antibody, Int J Radiat Oncol Biol Phys, 2004;58: 984–90.
  45. Winquist E, Nabid A, Sicheri D, et al., A phase I dose escalation study of a humanised monoclonal antibody to EGFR (hR3) in patients with locally advanced squamous cell cancer of the head and neck (SCCHN) treated with radiotherapy (RT), J Clin Oncol, 2002;21: abstract 926.
  46. Crombet T, Osorio M, Cruz T, et al., Use of the humanised Anita- EGFR Monoclonal Antibody h-R3 in combination with radiotherapy in the treatment of locally advanced head and neck cancer patients, J Clin Oncol, 2004;22:1646–54.
  47. Bastholt L, Specht L, Jensen K, et al., A novel fully human monoclonal antibody against epidermal growth factor receptor (EGFR). First clinical and FDG-PET imaging results from a phase I/II trial conducted by the Danish Head and Neck Cancer Study Group (DAHANCA) in patients with squamous cell carcinoma of the head and neck (SCCHN), J Clin Oncol, 2004;23: abstract 5530.
  48. Modjtahedi H, Hickish T, Nicolson M, et al., Phase I trial and tumour localisation of the anti-EGFR monoclonal antibody ICR62 in head and neck or lung cancer, Br J Cancer, 1996;73:228–35.
  49. Bleeker WK, Lammerts van Bueren JJ, van Ojik HH, et al., Dual mode of action of a Human Anti-EGFR Monoclonal Antibody for cancer therapy, J Immun, 2004;173:4699–4707.
  50. Curnow RT, Clinical experience with CD-64-directed immunotherapy. An overview, Cancer Immunol Immunother, 1997;45:181–94.
  51. Cohen EEW, 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;21:1980–77.
  52. 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;94:631–6.
  53. Wheeler RH, Jones D, Sharma P, et al., Clinical and molecular phase II study of gefitinib in patients (pts) with recurrent squamous cell cancer of the head and neck, J Clin Oncol, 2005;23: abstract 5531.
  54. Cohen EEW, Kane MA, List MA, et al., Phase II trial of gefitinib 250mg daily in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck, Clin Cancer Res, 2005;11:8418–24.
  55. Stewart JSW, Cohen EEW, Licitra L, et al., A phase III randomised 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; abstract 3522.
  56. Soulieres D, Senzer NN, Vokes EE, et al., Multicentre 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;22:77–85.
  57. 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;23:5892–9.
  58. 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;22:777–84.
  59. Herbst RS, Giaccone G, Schiller JH, et al., Gefitinib in combination with paclitaxel and carboplatin in advanced non-small-cell lung cancer, J Clin Oncol, 2004;22:785–94.
  60. Magne N, Fischel JL, Dubreuil A, et al., Sequence-dependent effects of ZD1839 (Iressa®) in combination with cytotoxic treatment in human head and neck cancer, Br J Cancer, 2002;86: 819–27.
  61. Choe MS, Chen Z, Zhang X, et al., Sequence-dependent effect of epidermal growth factor receptor-tyrosine kinase inhibitor (EGFRTKI), ZD1839 or Iressa in combination with docetaxel and cisplatin in squamous cell carcinoma of the head and neck (SCCHN), Proc Am Assoc Cancer Res, 2005; abstract 1396.
  62. Mukesh K, Nyati MN, Chun PY, et al., Optimum scheduling for better therapeutic outcome is required for Gefitinib and Gemcitabine treatment for Head and Neck cancer, Proc Am Assoc Cancer Res, 2005; abstract 122.
  63. Cohen EE, Harad DJ, Stenson KM, et al., Integration of Gefitinib (G), into a Concurrent Chemoradiation (CRT) Regimen Followed by Adjuvant Therapy in Patients with Locally Advanced Head and Neck Cancer (HNC) – a Phase II Trial, J Clin Oncol, 2005;23: abstract 5506.
  64. Doss HH, Greco FA, Meluch AA, et al., Induction chemotherapy + gefitinib followed by concurrent chemotherapy/radiation therapy/gefitinib for patients with locally advanced squamous carcinoma of the head and neck: A phase I/II trial of the Minnie Pearl Cancer Research Network, J Clin Oncol, 2006;24: abstract 5543.
  65. Savvides P, Agarwala SS, Greskovich J, et al., Phase I study of the EGFR tyrosine kinase inhibitor erlotinib in combination with docetaxel and radiation in locally advanced squamous cell cancer of the head and neck (SCCHN), J Clin Oncol, 2006;24: abstract 5545.
  66. Herchenhorn D, Dias FL, Ferreira CG, et al., Phase I/II study of erlotinib combined with cisplatin and radiotherapy for locally advanced squamous cell carcinoma of the head and neck (SCCHN), J Clin Oncol, 2006;24: abstract 5575.
  67. Rhoades CA, Kraut E, Schuller D, et al., Phase I and II study of OSI-774 and docetaxel in squamous cell carcinoma of the head and neck (SCCHN), J Clin Oncol, 2006;22: abstract 5541.
  68. Delord J, Thomas F, Benlyazid A, et al., Neo-adjuvant treatment with erlotinib in squamous cell carcinoma head and neck (SCCHN) patients before surgery provides an opportunity to find predictive factors of response, J Clin Oncol, 2006;24: abstract 5513.
  69. Huang S, Armstrong EA, Benavente S, et al., Dual-agent molecular targeting of the Epidermal Growth Factor Receptor (EGFR): combining Anti-EGFR antibody with Tyrosine Kinase Inhibitor, Cancer Res, 2004;64:5355–62.
  70. Matar P, Rojo F, Cassia R, et al., Combined Epidermal Growth Factor Receptor-targeting with the tyrosine kinase inhibitor gefitinib (ZD1839) and the monoclonal antibody cetuximab (IMC- 225); superiority over single-agent receptor targeting, Clin Cancer Res, 2004;10:6487–6501.
  71. Lal A, Glazer CA, Martinson HM, et al., Mutant epidermal growth factor receptor up-regulates molecular effectors of tumour invasion, Cancer Res, 2002;62:3335–9.
  72. Li B, Yuan M, Kim I-A, et al., Mutant epidermal growth factor receptor displays increased signalling through the phophatidylinositol-3 kinase/AKT pathway and promotes radioresistance in cells of astrocytic origin, Oncogene, 2002;23: 4594–4602.
  73. Lammering G, Valerie K, Lin PS, et al., Radiation-induced activation of a common variant of EGFR confers enhanced radioresistance, Radiother Oncol, 2004;72:267–73.
  74. 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;12:5064–73.
  75. Paez JG, Jänne PA, Lee JC, et al., EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy, Science, 2004;304:1497–1500.
  76. 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: 2129–39.
  77. Jänne PA, Johnson BE, Effect of epidermal growth factor receptor tyrosine kinase domain mutations on the outcome of patients with non-small cell lung cancer treated with epidermal growth factor receptor kinase inhibitors, Clin Cancer Res, 2006;12: S4418–20.
  78. Inoue A, Suzuki T, Fukuhara T, et al., Prospective phase II study of gefitinib for chemotherapy-naïve patients with advanced nonsmall cell lung cancer with epidermal growth factor receptor gene mutations, J Clin Oncol, 2006;24:3340–46.
  79. Riely GJ, Pao W, Pham DK, et al., Clinical course of patients with non-small cell lung cancer and epidermal growth factor receptor exon 19 and exon 21 mutations treated with gefitinib or erlotinib, Clin Cancer Res, 2006;12:839–44.
  80. Uramato H, Sugio K, Oyama T, et al., Epidermal growth factor receptor mutations are associated with gefitinib sensitivity in nonsmall cell lung cancer in Japanese, Lung Cancer, 2006;51:71–7.
  81. 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: 2556–68.
  82. Eberhard DA, Johnson BE, Amler L, 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;23:5900–09.
  83. Mukohara T, Engelman JA, Hanna N, et al., Differential effects of gefitinib and cetuximab on Non-Small Cell Lung Cancer cell lines bearing Epidermal Growth Factor Receptor mutation, J Natl Cancer Inst, 2005;97:1185–94.
  84. Guo M, Liu S, Herman JG, et al., Gefitinib-sensitising mutation in oesophageal carcinoma cell line Kyse 450, Cancer Biol Ther, 2006;5:152–5.
  85. Guo M, Liu S, Lu F, Gefitinib-sensitising mutations in oesophageal carcinoma, N Engl J Med, 2006;354:2193–4.
  86. Lee JW, Soung YH, Kim SY, et al., Somatic mutations of EGFR gene in squamous cell carcinoma of the head and neck, Clin Cancer Res, 2005;11:2879–82.
  87. 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;19:634–40.
  88. 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, 2005;42:09–111.
  89. Cohen EEW, Lingen MW, Martin LE, et al., Response of some head and neck cancers to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors may be linked to Mutation of ERBB2 rather than EGFR, Clin Cancer Res, 2005;11:8105–8.
  90. Mojica WD, Khoury T, Starostik PP, et al., Frequency of gefitinib responder mutations in EGFR (+) squamous cell carcinoma of the head and neck, Proc Am Assoc Cancer Res, 2005;992–3.
  91. Murray S, Linardou H, Razis E, et al., Screening for EGFR mutations in patients with head and neck cancer treated with gefitinib on a compassionate-use program: a Hellenic Cooperative Oncology Group study, Ann Oncol, 2006;17: abstract 578.
  92. Na I, Kang H, Park Y, et al., EGFR mutations and human papillomavirus in squamous cell carcinoma of tongue and tonsil, J Clin Oncol, 2006;24: abstract 10027.
  93. Xia W, Lau YK, Zhang HZ, et al., Combination of EGFR, HER- 2/neu, and HER3 is a stronger predictor for the outcome of oral squamous cell carcinoma than any individual family members, Clin Cancer Res, 1999;5:4164–74.
  94. Gillison ML, Glisson BS, O’Leary E, et al., Phase II trial of trastuzumab (T), paclitaxel (P), and cisplatin© in metastatic (M) or recurrent(R) head and neck squamous cell carcinoma (HNSCC): response by tumour EGFR and HER2/neu status, J Clin Oncol, 2006;24:5511.
  95. 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: abstract 5568.
  96. Bourhis J, Harrington K, Rosine D, et al., A phase I, open-label study (EGF100262) of lapatinib plus chemoradiation in patients with locally advanced squamous of carcinoma of the head and neck, Ann Oncol, 2006;17: abstract 576.
  97. Thomas GR, Nadiminti H, Regalado J, Molecular predictors of clinical outcome in patients with head and neck squamous cell carcinoma, Int J Exp Pathol, 2005;86:347–63.
  98. Gallo O, Masini E, Bianchi B, et al., Prognostic significance of cyclooxygenase-2 pathway and angiogenesis in head and neck squamous cell carcinoma, Hum Pathol, 2002;33:708–14.
  99. Peng J, Su C, Chang H, Overexpression of cyclooxygenase-2 in squamous cell carcinoma of the hypopharynx, Hum Pathol, 2002;33:100–104.
  100. Lin D, Subbaramaiah K, Shah J, et al., Cyclo-oxygenase-2: a novel molecular target for the prevention and treatment of head and neck cancer, Head Neck, 2002;24:792–9.
  101. Itoh S, Matsui K, Furuta I, et al., Immunohistochemical study of overexpression of cyclooxygenase-2 in squamous cell carcinoma of the oral cavity: its importance as a prognostic predictor, Oral Oncol, 2003;39:829–35.
  102. Renkonen J, Wolff, Paavonen T, Expression of cyclooxygenase-2 in human tongue carcinoma and its precursor lesions, Virchows Arch, 2002;440:594–7.
  103. Pannone G, Bufo P, Caiaffa MF, et al., Cyclooxygenase-2 expression in oral squamous cell carcinoma, Int J Immunopathol Pharmacol, 2004;17:373–82.
  104. Chan G, Boyle JO, Yang EY, et al., Cyclooxygenase-2 expression is up-regulated in squamous cell carcinoma of the head and neck, Cancer Res, 1999;59:991–4.
  105. Grau JJ, Monzo JC, Palmero R, et al., Expression of cyclooxygenase-2 mRNA (COX2-mRNA) in peripheral blood of head and neck cancer patents and healthy controls, J Clin Oncol, 2004;22: abstract 5521.
  106. Lippman SM, Gibson N, Subbaramaiah K, et al., Combined targeting of the EGFR and cyclooxygenase-2 pathways, Clin Cancer Res, 2005;11:6097–9.
  107. Choe MS, Zhang X, JC Chin, et al., Interaction between epidermal growth factor receptor and cyclooxygenase 2-mediated pathways and its implications for the chemoprevention of head and neck cancer, Mol Cancer Ther, 2005;4:1448–55.
  108. Chen Z, Zhang X, Li M, et al., Simultaneously targeting Epidermal Growth Factor Receptor, Tyrosine Kinase and Cyclooxygenase-2, an efficient approach to inhibition of Squamous Cell Carcinoma of the Head and Neck, Clin Cancer Res, 2004;10:5930–39.
  109. Zhang X, Chen ZG, Choe MS, et al., Tumour growth inhibition by simultaneously blocking epidermal growth factor receptor and cyclooxygenase-2 in a xenograft model, Clin Cancer Res, 2005;11:6261–9.
  110. 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;23:6976–81.
  111. Kyzas PA, Cunha IW, Ioannidis JPA, Prognostic significance of Vascular Endothelial Growth Factor Immunohistochemical Expression in Head and Neck Squamous Cell Carcinoma: A Meta- Analysis, Clin Cancer Res, 2005;11:1434–40.
  112. Mauer AM, Cohen EEW, Wong SJ, et al., Phase I study of epidermal growth factor receptor (EGFR) inhibitor, erlotinib, and vascular endothelial growth factor monoclonal antibody, Bevacizumab, in recurrent and/or metastatic squamous cell carcinoma of the head and neck (SCCHN), J Clin Oncol, 2004;22: abstract 5539.
  113. Vokes EE, Cohen EE, Mauer AM, et al., A phase I study of erlotinib and bevacizumab for recurrent or metastatic squamous cell carcinoma of the head and neck, J Clin Oncol, 2005;23: abstract 5504.
  114. Bozec A, Formento P, Fischel J-L, et al., Combined effect of EGFR-targeting with antiangiogenesis and irradiation, Proc Am Assoc Cancer Res, 2006; abstract 55.

Further Resources

Share this Article
Related Content In Head and Neck Cancer
  • Copied to clipboard!
    accredited arrow-down-editablearrow-downarrow_leftarrow-right-bluearrow-right-dark-bluearrow-right-greenarrow-right-greyarrow-right-orangearrow-right-whitearrow-right-bluearrow-up-orangeavatarcalendarchevron-down consultant-pathologist-nurseconsultant-pathologistcrosscrossdownloademailexclaimationfeedbackfiltergraph-arrowinterviewslinkmdt_iconmenumore_dots nurse-consultantpadlock patient-advocate-pathologistpatient-consultantpatientperson pharmacist-nurseplay_buttonplay-colour-tmcplay-colourAsset 1podcastprinter scenerysearch share single-doctor social_facebooksocial_googleplussocial_instagramsocial_linkedin_altsocial_linkedin_altsocial_pinterestlogo-twitter-glyph-32social_youtubeshape-star (1)tick-bluetick-orangetick-red tick-whiteticktimetranscriptup-arrowwebinar Sponsored Department Location NEW TMM Corporate Services Icons-07NEW TMM Corporate Services Icons-08NEW TMM Corporate Services Icons-09NEW TMM Corporate Services Icons-10NEW TMM Corporate Services Icons-11NEW TMM Corporate Services Icons-12Salary £ TMM-Corp-Site-Icons-01TMM-Corp-Site-Icons-02TMM-Corp-Site-Icons-03TMM-Corp-Site-Icons-04TMM-Corp-Site-Icons-05TMM-Corp-Site-Icons-06TMM-Corp-Site-Icons-07TMM-Corp-Site-Icons-08TMM-Corp-Site-Icons-09TMM-Corp-Site-Icons-10TMM-Corp-Site-Icons-11TMM-Corp-Site-Icons-12TMM-Corp-Site-Icons-13TMM-Corp-Site-Icons-14TMM-Corp-Site-Icons-15TMM-Corp-Site-Icons-16TMM-Corp-Site-Icons-17TMM-Corp-Site-Icons-18TMM-Corp-Site-Icons-19TMM-Corp-Site-Icons-20TMM-Corp-Site-Icons-21TMM-Corp-Site-Icons-22TMM-Corp-Site-Icons-23TMM-Corp-Site-Icons-24TMM-Corp-Site-Icons-25TMM-Corp-Site-Icons-26TMM-Corp-Site-Icons-27TMM-Corp-Site-Icons-28TMM-Corp-Site-Icons-29TMM-Corp-Site-Icons-30TMM-Corp-Site-Icons-31TMM-Corp-Site-Icons-32TMM-Corp-Site-Icons-33TMM-Corp-Site-Icons-34TMM-Corp-Site-Icons-35TMM-Corp-Site-Icons-36TMM-Corp-Site-Icons-37TMM-Corp-Site-Icons-38TMM-Corp-Site-Icons-39TMM-Corp-Site-Icons-40TMM-Corp-Site-Icons-41TMM-Corp-Site-Icons-42TMM-Corp-Site-Icons-43TMM-Corp-Site-Icons-44TMM-Corp-Site-Icons-45TMM-Corp-Site-Icons-46TMM-Corp-Site-Icons-47TMM-Corp-Site-Icons-48TMM-Corp-Site-Icons-49TMM-Corp-Site-Icons-50TMM-Corp-Site-Icons-51TMM-Corp-Site-Icons-52TMM-Corp-Site-Icons-53TMM-Corp-Site-Icons-54TMM-Corp-Site-Icons-55TMM-Corp-Site-Icons-56TMM-Corp-Site-Icons-57TMM-Corp-Site-Icons-58TMM-Corp-Site-Icons-59TMM-Corp-Site-Icons-60TMM-Corp-Site-Icons-61TMM-Corp-Site-Icons-62TMM-Corp-Site-Icons-63TMM-Corp-Site-Icons-64TMM-Corp-Site-Icons-65TMM-Corp-Site-Icons-66TMM-Corp-Site-Icons-67TMM-Corp-Site-Icons-68TMM-Corp-Site-Icons-69TMM-Corp-Site-Icons-70TMM-Corp-Site-Icons-71TMM-Corp-Site-Icons-72