Skip to main content
British Journal of Cancer logoLink to British Journal of Cancer
. 2004 Feb 3;90(3):566–572. doi: 10.1038/sj.bjc.6601550

Gefitinib (‘Iressa’, ZD1839) and new epidermal growth factor receptor inhibitors

G Blackledge 1,*, S Averbuch 2
PMCID: PMC2409596  PMID: 14760365

Abstract

The epidermal growth factor receptor (EGFR) is a promising target for cancer therapy and a number of EGFR-targeted agents have been developed. Those most advanced in development are the EGFR tyrosine kinase inhibitors gefitinib (‘Iressa’, ZD1839) and erlotinib (‘Tarceva’, OSI-774), and the monoclonal antibody cetuximab (‘Erbitux’, IMC-C225). This review provides a clinical overview of these agents, highlighting their antitumour activities in different tumour types. Epidermal growth factor receptor–targeted agents are generally well tolerated and are not typically associated with the severe adverse events often seen with cytotoxic chemotherapy. Gefitinib is the agent with the most extensive clinical experience, particularly in non-small-cell lung cancer (NSCLC). Recently, gefitinib became the first-approved EGFR-targeted agent, for use in patients with previously treated advanced NSCLC in Japan, the USA and other countries. Further studies are required to explore the full potential of these novel agents either as monotherapy or combination therapy.

Keywords: EGFR-targeting agents, gefitinib, erlotinib, cetuximab


Conventional cytotoxic anticancer agents have limited efficacy and a narrow therapeutic index. Identification of molecular targets important for cancer cell proliferation and survival has provided an opportunity for improved efficacy and more selective action; on tumour rather than normal tissue (Baselga, 2002; Herbst and Kies, 2002).

One such target is the epidermal growth factor receptor (EGFR), which is highly expressed in a variety of solid tumours. Expression correlates with disease progression, poor survival, poor response to therapy and resistance to cytotoxic agents (Arteaga, 2002). The EGFR is a transmembrane glycoprotein comprising an extracellular ligand-binding domain, a transmembrane hydrophobic domain and an intracellular domain with tyrosine kinase activity involved in signal transduction (Figure 1). Upon ligand binding, receptor dimerisation activates tyrosine kinase activity and tyrosine autophosphorylation. This initiates a signalling cascade that leads to cell proliferation, increased angiogenesis, invasion and metastasis and decreased apoptosis (Baselga, 2002).

Figure 1.

Figure 1

EGFR signalling and anti-EGFR approaches (reproduced with permission from: Baselga (2002); ©AlphaMed Press 1083–7159).

A variety of strategies to target EGFR signalling have been investigated, including: (1) small molecule tyrosine kinase inhibitors that prevent ATP from binding to the intracellular tyrosine kinase domain of EGFR, thereby inhibiting tyrosine kinase activity and autophosphorylation, and subsequent signal transduction; (2) monoclonal antibodies that target the extracellular ligand-binding domain or bispecific antibodies that also target epitopes on the surface of immune effector cells; (3) immunotoxin conjugates using cytotoxic single-chain fragment variable antibodies conjugated to toxins such as pseudomonas endotoxin A; (4) EGF vaccines such as EGF-P64k that contained recombinant human EGF conjugated to P64k, a highly immunogenic recombinant bacterial protein; (5) antisense oligonucleotides to block the translation of the ligand or the EGFR (Figure 1) (Baselga, 2002). The first two of these approaches have proved to be the most successful (Table 1 ).

Table 1. EGFR-targeted therapies in clinical development.

Class of agent Agent Phase of development
EGFR tyrosine kinase inhibitors    
  Gefitinib, ZD1839 III/marketed
  Erlotinib, OSI-74 III
  Canertinib, CI-1033 II
  EKB-569 II
  Lapatinib, GW572016 II
Monoclonal antibodies    
  Human–murine chimeric monoclonal antibodies  
  Cetuximab, IMC-C225 III/pre-registration
  Fully humanised monoclonal antibodies  
  ABX-EGF II
  EMD-72000 II
  Thera CIM-h-R3 II
  HuMax-EGFR I/II

This review will concentrate on the agents most advanced in clinical development: the EGFR tyrosine kinase inhibitors gefitinib (‘Iressa’, ZD1839) and erlotinib (‘Tarceva’, OSI-774), and the chimeric human–mouse monoclonal antibody cetuximab (‘Erbitux’, IMC-C225) (Table 2 ). Gefitinib has recently received approval, the first for an EGFR-targeted agent, for the treatment of patients with previously treated advanced non-small-cell lung cancer (NSCLC) in Japan, the USA and other countries. Erlotinib is currently in the follow-up stage of three large Phase III trials and cetuximab is at the pre-registration stage for colorectal cancer.

Table 2. Clinical development of gefitinib, erlotinib and cetuximab.

Agent Class of agent Phase I trial design MTD Dose selection parameters Dose selected for further study Tumour types Phase of development
Gefitinib EGFR-TKI Monotherapy up to 1000 mg day−1 ⩾700 mg day−1 Optimum biological dose: tumour response/stable disease, target activity and pharmacokinetics 250 and 500 mg day−1 NSCLC, SCCHN, colorectal, breast III/marketed (NSCLC)
Erlotinib EGFR-TKI Three monotherapy regimens (100–1600 mg) administered: once weekly every 3 out of 4 weeks; 3 consecutive days for 3 weeks; daily for 3 weeks 150 mg day−1 MTD 150 mg day−1 NSCLC, SCCHN, pancreatic III (NSCLC, pancreatic)
Cetuximab Monoclonal antibody Administered intravenously as single infusion (5–100 mg m−2); weekly for 4 weeks (5–100 mg m−2); weekly for 4 weeks combined with cisplatin (400 mg/m2) Saturates systemic clearance, 200–400 mg m−2 Optimum biological dose: pharmacokinetics, EGFR saturation Loading dose 400 mg m−2 in week 1 followed by weekly maintenance dose of 250 mg m−2 SCCHN, colorectal, NSCLC III/preregistration, (SCCHN, colorectal)

MTD=maximum tolerated dose; NSCLC=non-small-cell lung cancer; SCCHN=squamous-cell carcinoma of the head and neck.

PRECLINICAL DATA

As monotherapy, gefitinib, erlotinib and cetuximab were antiproliferative and increased apoptosis in different cancer cell lines and human tumours xenografted to immunodeficient mice (Ciardiello et al, 2000; Ciardiello and Tortora, 2001). Interestingly, although there was a spectrum of dose-dependent antitumour activity ranging from dramatic tumour regression to de novo resistance when gefitinib was tested in multiple human tumour xenograft studies, the level of EGFR expression did not predict tumour response (Wakeling et al, 2002). Many studies demonstrated additive or synergistic antitumour effects when combining EGFR-targeted agents with chemotherapy, ionising radiation or other novel agents (Milas et al, 2000; Sirotnak et al, 2000; Tortora et al, 2001; Bianco et al, 2002; Buchsbaum et al, 2002; Huang et al, 2002; Magne et al, 2002; Normanno et al, 2002; Williams et al, 2002; Prewett et al, 2003; Solomon et al, 2003; Xu et al, 2003).

CLINICAL OVERVIEW

Epidermal growth factor receptor-targeted agents have been investigated in several human cancers. Many studies have involved patients with tumours that widely express EGFR, including NSCLC, colorectal cancer, squamous-cell carcinoma of the head and neck (SCCHN) and breast cancer. Existing treatments improve survival times, but many patients with advanced disease relapse and require further therapy to palliate symptoms and improve survival. This highlights the significant unmet need for novel, targeted agents (Herbst and Kies, 2002; O’Dwyer and Benson, 2002; Herbst and Langer, 2002; Cohen, 2003; Cohen et al, 2003a).

Gefitinib

Four multicentre, open-label, Phase I trials investigated the tolerability and efficacy of oral gefitinib (up to 1000 mg day−1) in patients with a variety of solid tumours, including NSCLC (Baselga et al, 2002; Herbst et al, 2002; Ranson et al, 2002; Nakagawa et al, 2003). As gefitinib is not a traditional cytotoxic agent, dose selection for further study was based on identification of the optimum biological dose, combining maximum efficacy with minimum adverse events (AEs).

Gefitinib was generally well tolerated; the most common AEs were mild/moderate (grade 1/2) reversible rash and diarrhoea, whose incidence and severity increased with increasing dose. Gefitinib was not typically associated with the cytotoxic AEs of chemotherapy. The maximum tolerated dose (MTD) was ⩾700 mg day−1. Promising antitumour activity was observed in a number of tumour types, particularly NSCLC; 10 out of 100 NSCLC patients experienced a partial tumour response (Herbst et al, 2002; Ranson et al, 2002; Nakagawa et al, 2003). Partial responses and disease stabilisation were observed at doses ⩾150 mg day−1 with no suggestion that higher doses provided greater antitumour activity (Herbst and Kies, 2002; Ranson et al, 2002). Similarly, pre- and post-treatment skin biopsy results from cancer patients revealed that EGFR signalling was inhibited at doses ⩾150 mg day−1, with no clear dose dependence above this level (Albanell et al, 2002). Based on these results, two doses below the MTD were chosen for evaluation in Phase II studies: 250 mg day−1, at which dose responses had been seen with minimum toxicity, and 500 mg day−1, the highest dose that has been tolerated on prolonged treatment.

Gefitinib monotherapy in NSCLC

Two large-scale Phase II, multicentre, dose-randomised, double-blind, parallel-group studies, Iressa Dose Evaluation in Advanced Lung Cancer (IDEAL) 1 and 2, evaluated gefitinib monotherapy (250 and 500 mg day−1) in patients with locally advanced or metastatic NSCLC who had received platinum-based chemotherapy (Fukuoka et al, 2003; Kris et al, 2003). Doses were given daily until disease progression or withdrawal due to intolerable toxicity. Patients in IDEAL 1 were recruited from Europe, Japan, Australia and South Africa (103 patients at 250 mg day−1; 106 patients at 500 mg day−1) and must have received one or two prior chemotherapy regimens, at least one of which contained platinum. Patients in IDEAL 2 were recruited from the USA (102 patients at 250 mg day−1; 114 patients at 500 mg day−1), and must have received at least two prior regimens including platinum and docetaxel given either concurrently or separately. Disease-related symptoms were measured weekly using the independently validated Lung Cancer Subscale (LCS) of the Functional Assessment of Cancer Therapy-Lung (FACT-L) quality-of-life questionnaire (Cella et al, 2002). Using the LCS, the severities of seven disease-related symptoms are recorded on a five-point Likert scale where the maximum (best) score possible is 28. All patients in IDEAL 2 were symptomatic, with a baseline LCS score of ⩽24 points.

Both doses showed clinically meaningful antitumour activity with no greater efficacy at the higher dose (Figure 2). Adverse events were fewer and less severe at the lower dose. Accordingly, 250 mg day−1 gefitinib (approximately one-third of the MTD) is the recommended/optimum biological dose in NSCLC, causing minimum AEs without compromising efficacy. Only results for this dose will be discussed here. For IDEAL 1 and 2, respectively, response rates were 18.4 and 11.8%, disease control rates were 54.4 and 42.2%, median progression-free survival was 2.7 and 1.9 months and median overall survival was 7.6 and 6.5 months. Objective responses were achieved regardless of the number of prior chemotherapy regimens.

Figure 2.

Figure 2

Gefitinib antitumour activity in advanced NSCLC: IDEAL 1 and 2.

Many patients’ disease-related symptoms improved significantly, important because many NSCLC patients have frequent and severe symptoms. Symptom improvement rates for IDEAL 1 and 2 were 40.3 and 43.1%, with a median time to improvement of 8 and 10 days, respectively. Symptom improvement correlated with objective tumour response and was associated with increased overall and progression-free survival (Herbst and Kies, 2002). In IDEAL 2, the median overall survival for patients with symptom improvement was 13.6 months, compared with 3.7 months for patients without symptom improvement (Douillard et al, 2002).

In IDEAL 1 and 2, 24 and 34% of patients had an improved quality of life, with a median time to improvement of 29 and 30 days, respectively. Most patients with a partial/complete response or stable disease also showed improved quality of life (Natale and Zaretsky, 2002).

The most common AEs were skin rash (47 and 43% in IDEAL 1 and 2, respectively) and diarrhoea (40 and 48%, respectively). The majority of these AEs were Common Toxicity Criteria grade 1 and, across both IDEAL 1 and 2, grade 3 diarrhoea and skin rash were each observed in only one patient. The incidences of dose reductions (<1%) and withdrawals (1.9 and 1.0% in IDEAL 1 and 2, respectively) due to AEs were also low (Schiller et al, 2002).

Gefitinib combination therapy in NSCLC

Two Phase I trials using carboplatin/paclitaxel and cisplatin/gemcitabine, respectively, showed combination therapy to have acceptable tolerability and no unexpected or cumulative toxicity (Miller et al, 2001; Gonzalez-Larriba et al, 2002).

Two multinational, randomised, double-blind, placebo-controlled Phase III studies investigated gefitinib combined with standard platinum-based first-line chemotherapy regimens. The ‘Iressa’ NSCLC Trials Assessing Combination Treatment (INTACT) 1 and 2 involved more than 2100 chemonaive patients with advanced NSCLC (Giaccone et al, 2002; Johnson et al, 2002). Disappointingly, there were no improvements in overall survival or other efficacy outcomes in the first-line setting. However, these placebo-controlled studies confirmed the favourable safety profile of gefitinib observed in the IDEAL trials. The combination toxicity profile was similar to that of chemotherapy alone except for the addition of dose-dependent diarrhoea and skin rash.

Gefitinib therapy against other cancers

A Phase II study investigating gefitinib monotherapy (500 mg day−1) in patients with recurrent or metastatic SCCHN showed promising antitumour activity and acceptable toxicity. Of 47 evaluable patients, the response rate was 10.6% and the disease control rate 53%. The median time to disease progression and overall survival were 3.4 and 8.1 months, respectively. This compares with median survival times of 6–8 months from Phase III trials of other agents in this setting. Skin toxicities, never greater than grade 2, were observed in 48% of patients, and diarrhoea was observed in 50% of patients (at grade 3 in three patients) (Cohen et al, 2003a). A Phase II trial is recruiting patients (n=63) with recurrent SCCHN to investigate gefitinib monotherapy (250 mg day−1). Of the 14 patients currently evaluable for response, four had stable disease. Toxicity was assessed in 17 patients; the most common AEs were grade 1/2 rash in 50% of patients and grade 1/2 diarrhoea in 20% of patients, with no grade 3/4 AEs (Cohen et al, 2003b). Further data from this patient population will be needed to determine whether 250 or 500 mg day−1 has optimum activity in recurrent SCCHN.

A Phase II study investigating combination therapy of gefitinib (500 mg day−1) with FOLFOX-4, a triple-combination standard treatment regimen for patients with advanced colorectal cancer, showed antitumour activity. For untreated patients, the response rate was 75% and for patients who had relapsed after chemotherapy, 29%. These values compare with historical response rates of 30–55 and 9%, respectively. Combination therapy was generally well tolerated with the most common grade 3/4 AE being diarrhoea (Cho et al, 2003).

Two Phase II studies investigated gefitinib monotherapy (500 mg day−1) in patients with advanced breast cancer. In the first study (n=31), 10 patients (32%) had stable disease for ⩾3 months (Baselga et al, 2003). The second study involved nine evaluable patients with acquired tamoxifen-resistant oestrogen-receptor-positive breast cancer and 18 evaluable patients with oestrogen-receptor-negative breast cancer. Of the tamoxifen-resistant patients, one patient had a partial response and five had stable disease. Of the oestrogen-receptor-negative patients, one patient had a partial response and one patient had stable disease (Robertson et al, 2003).

Erlotinib

In Phase I trials involving pretreated patients with advanced solid tumours including NSCLC, three monotherapy regimens were administered (dose range 100–1600 mg): once weekly every 3 out of 4 weeks; on 3 consecutive days for 3 weeks; daily for 3 weeks. The MTD was not reached in the once-weekly schedule and diarrhoea was the dose-limiting toxicity in the once-daily schedule, at 200 mg day−1 (Siu et al, 1999). Common AEs were skin rashes and diarrhoea. Using conventional chemotherapy dose-selection methods, 150 mg day−1 (MTD) was selected for Phase II studies (Ciardiello and Tortora, 2001; Kim and Murren, 2002).

Erlotinib in NSCLC

In a Phase II trial, 56 patients with EGFR-positive NSCLC recurrent or progressive after platinum-based chemotherapy received erlotinib monotherapy. At 12 weeks, 10.7% had a confirmed partial response and 33.9% had stable disease. Treatment was generally well tolerated and the most common AE was a maculopapular acneiform rash in 78% of patients. No patients discontinued treatment due to toxicity and only two patients had dose reductions to 100 mg day−1 (Perez-Soler et al, 2001).

Current Phase III trials in NSCLC are investigating monotherapy in refractory patients randomised to receive erlotinib or placebo and first-line combination therapy with carboplatin/paclitaxel or gemcitabine/cisplatin (Kim and Murren, 2002). Preliminary results from the first-line combination trials have shown that combination with erlotinib did not result in an improvement in overall survival compared with chemotherapy alone.

Erlotinib and other cancers

Erlotinib monotherapy has antitumour activity in other tumour types. Out of 114 patients with SCCHN, 78 were evaluable for response: 12.8% had a partial response and 29.5% had disease stabilisation. The most common AE (n=114) was acneiform rash in 72% of patients (Senzer et al, 2001). Of 30 evaluable patients with pretreated advanced refractory ovarian cancer, 6.7% had a partial response and 10% had disease stabilisation at 5–6 months. Treatment was generally well tolerated and rash was the most common AE, in 88% of patients (Finkler et al, 2001).

A Phase III trial is investigating erlotinib (100 mg day−1) and gemcitabine combination therapy in 800 patients with pancreatic cancer (Kim and Murren, 2002).

Cetuximab

In three consecutive open-label Phase I trials, 52 patients with advanced tumours expressing high levels of EGFR were administered cetuximab intravenously as a single infusion (5–100 mg m−2), weekly for 4 weeks (5–100 mg m−2) or weekly for 4 weeks (up to 400 mg m−2) combined with cisplatin. The MTD was not reached in any study; toxicity was minimal and unrelated to dose or number of cycles administered. Common AEs were skin toxicities, fever and chills, asthenia, transient transaminase elevations and nausea. Four patients had grade 3/4 AEs. One patient receiving monotherapy had grade 3 aseptic meningitis. When cetuximab was combined with cisplatin, one patient had diarrhoea (grade 3), one patient had an anaphylactic reaction (grade 3) and one patient had both epiglottitis (grade 3) and dyspnoea (grade 4) (Baselga et al, 2000).

Cetuximab displays nonlinear pharmacokinetics and, due to its long half-life, can be administered weekly (Herbst and Hong, 2002). In Phase Ib clinical trials, a loading dose of 400 mg m−2 at week 1 followed by a weekly maintenance dose of 250 mg m−2 achieved almost complete saturation of EGFR in tumour tissue. This was the recommended dose for Phase II and III clinical trials (Shin et al, 2001).

Replacing the constant region of the original mouse monoclonal antibody with the constant region of human IgG1 reduced immunogenicity. However, 4–6% of patients experience a serious allergic event within minutes of infusion and 2% have anaphylactic reactions (O’Dwyer and Benson, 2002; Baselga, 2002).

Cetuximab combination therapy

Combining cetuximab with standard anticancer treatments in patients with SCCHN, colorectal or pancreatic cancer did not increase toxicity (Needle, 2002).

A Phase I study combining different doses of cetuximab with radiotherapy showed promising activity in therapy-naive patients with advanced SCCHN. Of 15 patients, 86.7% had a complete and 13.3% a partial response (Robert et al, 2001). A Phase II study investigated cetuximab/cisplatin combination therapy in patients with SCCHN; 12% of 78 evaluable patients with disease refractory to cisplatin responded (Kies and Harari, 2002). A Phase III study recruited 121 patients with untreated SCCHN metastatic disease to compare cisplatin and cetuximab combination therapy with cisplatin and placebo. The response rate was higher in the combination treatment group: 23 vs 9%. However, there was no significant difference in time to tumour progression (4.1 and 3.4 months, respectively) or overall median survival (9.2 and 8.0 months, respectively). Toxicity data was available for 64 patients. Grade 3/4 AEs were hypersensitivity (6%), neutropenia (17%) and rash/desquamation (11%) (Burtness et al, 2002; Kies and Harari, 2002).

Cetuximab monotherapy vs cetuximab combined with irinotecan was investigated in 329 EGFR-positive, irinotecan-refractory patients with metastatic colorectal cancer. The response rate for the 218 patients who received combination therapy was 17.9% and the median time to progression was 126 days. In contrast, for monotherapy patients, these values were 9.9% and 45 days. The 65 AEs potentially related to treatment were consistent with the safety profiles of the individual agents (Cunningham et al, 2003).

Cetuximab combination therapy with cisplatin/vinorelbine vs cisplatin/vinorelbine alone is being investigated as first-line treatment in patients with EGFR-positive advanced NSCLC. Preliminary response rates for 18 and 17 patients are 50 and 29%, respectively. Only two serious AEs have been related to treatment with cetuximab (Gatzemeier et al, 2003).

DISCUSSION

Of the EGFR-targeting agents discussed in this review, gefitinib has undergone the most extensive clinical evaluation. While the agents share many similarities, differences in their properties and approach to clinical development might influence their clinical profile.

Gefitinib and erlotinib are administered orally, once daily, so would be more suitable for an outpatient setting. In contrast, cetuximab is administered intravenously and, as it is a chimeric human–mouse monoclonal antibody, it can cause allergic reactions (Ciardiello and Tortora, 2001; Baselga, 2002). Other monoclonal antibodies in clinical development are fully humanised antibodies that do not generate human antimouse antibodies, thereby reducing the risk of inducing hypersensitivity reactions in patients and potentially prolonging their in vivo lifetime.

Different approaches to dose selection have been used. As gefitinib is not a cytotoxic agent, it does not need to be given at the MTD. In NSCLC patients, the 250 mg day−1 recommended dose (about one-third of the MTD) showed equivalent efficacy to 500 mg day−1, but was associated with fewer grade 3/4 AEs, dose reductions, and withdrawals. This supports Phase I trials that show flat dose–response curves for efficacy while AEs increase with dose. Cetuximab is also dosed below the MTD, whereas erlotinib has followed a conventional cytotoxic dose-selection process, with dosing at the MTD.

All three agents have shown monotherapy activity although there are less data for cetuximab. It is harder to assess the efficacy of combination treatment over the activity of individual agents. Some studies provide promising results, whereas others demonstrate no advantage. Further studies are required to assess and optimise combination treatment with these agents.

The most common AEs for these EGFR-targeting agents are rash and diarrhoea and are higher for erlotinib, which is dosed at the MTD. These agents are not associated with the typical cytotoxic AEs affecting patients treated with chemotherapy (Ciardiello and Tortora, 2001).

In Japan, interstitial lung disease (ILD) has been observed in gefitinib-treated patients with an incidence of 1.7% (Inoue et al, 2003). This is higher than the worldwide reported incidence of 1% in over 92 000 patients treated (up to September 2003) and 0.38% in >39 000 patients treated as part of a compassionate-use programme (Forsythe and Faulkner, 2003). The incidence might be higher in Japanese patients due to greater awareness of ILD compared with the rest of the world, differences in ILD definitions or increased genetic susceptibility. In one retrospective study of 711 Japanese patients with lung cancer who had undergone surgical resection, 7.5% had idiopathic pulmonary fibrosis, a type of ILD (Kawasaki et al, 2002). Interstitial lung disease is a known complication of chemotherapy and radiotherapy in patients with lung cancer (Abid et al, 2001) and many patients with advanced NSCLC have no further treatment options, so the benefits of gefitinib treatment outweigh the risks of ILD.

Epidermal growth factor receptor-targeted agents have also shown promise in the treatment of patients with bronchioalveolar carcinoma (BAC), which is considered to be a subtype of adenocarcinoma of the lung without pleural, stromal or vascular invasion (World Health Organization classification). In a recent presentation at the European Cancer Conference, 13 out of 52 evaluable patients (25%) with BAC had a partial response to treatment with erlotinib (Patel et al, 2003). Similar results have been shown for gefitinib, with response rates of 20% (first line) and 12% (pretreated) reported in patients with advanced BAC (West et al, 2003).

The use of targeted agents has raised the possibility of selecting the patients most likely to respond to treatment. Although some studies involving EGFR-targeted agents selected patients according to EGFR expression, there is no evidence for an association between EGFR levels and response to small-molecule EGFR-targeted agents. Hence, there are no data to support EGFR screening to select patients who would benefit from treatment (Woodburn et al, 2000; Arteaga, 2002). As with conventional chemotherapy, some clinical baseline characteristics were predictive of greater response rates, for example, response rates to gefitinib were higher for women and patients with adenocarcinoma (Schiller et al, 2003). However, responses were observed in all groups. Further studies are required to identify the molecular profiles that predict the patients most likely to benefit from treatment with these agents.

CONCLUSIONS

As monotherapy or combination therapy, EGFR-targeted agents have demonstrated promise in treatment of several tumour types. The most extensive clinical experience has been with gefitinib (>92 000 patients) and two large Phase II studies have demonstrated clinically relevant antitumour activity in patients with previously treated advanced NSCLC.

The EGFR agents discussed have favourable AE profiles and are not typically associated with the heavy toxicity burden of chemotherapy agents, allowing long-term treatment. The maximum efficacy for targeted agents occurs below toxic doses, allowing both gefitinib and cetuximab to be administered at doses lower than the MTD to maximise the benefit : risk ratio. Gefitinib is the first EGFR agent to be approved for cancer treatment in Japan, the USA and other countries.

These novel therapies offer new treatment strategies to cancer patients with limited treatment options and further studies are underway to explore their full potential.

‘Iressa’ is a trademark of the AstraZeneca group of companies, ‘Tarceva’ is a trademark of OSI Pharmaceuticals, Inc., ‘Erbitux’ is a trademark of ImClone Systems Incorporated of New York.

References

  1. Abid SH, Malhotra V, Perry MC (2001) Radiation-induced and chemotherapy-induced pulmonary injury. Curr Opin Oncol 13: 242–248 [DOI] [PubMed] [Google Scholar]
  2. Albanell J, Rojo F, Averbuch S, Feyereislova A, Mascaro JM, Herbst R, LoRusso P, Rischin D, Sauleda S, Gee J, Nicholson RI, Baselga J (2002) Pharmacodynamic studies of the epidermal growth factor receptor inhibitor ZD1839 in skin from cancer patients: histopathologic and molecular consequences of receptor inhibition. J Clin Oncol 20: 110–124 [DOI] [PubMed] [Google Scholar]
  3. Arteaga CL (2002) Epidermal growth factor receptor dependence in human tumors: more than just expression? Oncologist 7(Suppl 4): 31–39 [DOI] [PubMed] [Google Scholar]
  4. Baselga J (2002) Why the epidermal growth factor receptor? The rationale for cancer therapy. Oncologist 7(Suppl 4): 2–8 [DOI] [PubMed] [Google Scholar]
  5. Baselga J, Albanell J, Ruiz A, Lluch A, Gascon P, Gonzalez S, Guillen V, Sauleda S, Averbuch S, Rojo F (2003) Phase II and tumor pharmacodynamic study of gefitinib (ZD1839) in patients with advanced breast cancer. Oral presentation at ASCO. Proc Am Soc Clin Oncol 22: 7 (abstr 24) [DOI] [PubMed] [Google Scholar]
  6. Baselga J, Pfister D, Cooper MR, Cohen R, Burtness B, Bos M, D'Andrea G, Seidman A, Norton L, Gunnett K, Falcey J, Anderson V, Waksal H, Mendelsohn J (2000) Phase I studies of anti-epidermal growth factor receptor chimeric antibody C225 alone and in combination with cisplatin. J Clin Oncol 18: 904–914 [DOI] [PubMed] [Google Scholar]
  7. Baselga J, Rischin D, Ranson M, Calvert H, Raymond E, Kieback DG, Kaye SB, Gianni L, Harris A, Bjork T, Averbuch SD, Feyereislova A, Swaisland H, Rojo F, Albanell J (2002) Phase I safety, pharmacokinetic, and pharmacodynamic trial of ZD1839, a selective oral epidermal growth factor receptor tyrosine kinase inhibitor, in patients with five selected solid tumor types. J Clin Oncol 20: 4292–4302 [DOI] [PubMed] [Google Scholar]
  8. Bianco C, Tortora G, Bianco R, Caputo R, Veneziani BM, Caputo R, Damiano V, Troiani T, Fontanini G, Raben D, Pepe S, Bianco AR, Ciardiello F (2002) Enhancement of antitumor activity of ionizing radiation by combined treatment with the selective epidermal growth factor receptor-tyrosine kinase inhibitor ZD1839 (Iressa). Clin Cancer Res 8: 3250–3258 [PubMed] [Google Scholar]
  9. Buchsbaum DJ, Bonner JA, Grizzle WE, Stackhouse MA, Carr B, Hicklin DJ, Bohlen P, Raisch KP (2002) Treatment of pancreatic cancer xenografts with erbitux (IMC-C225) anti-EGFR antibody, gemcitabine, and radiation. Int J Radiat Oncol Biol Phys 54: 1180–1193 [DOI] [PubMed] [Google Scholar]
  10. Burtness BA, Li Y, Flood W, Mattar BI, Forastiere AA (2002) Phase III trial comparing cisplatin (C) + placebo (P) to C + anti-epidermal growth factor antibody (EGF-R) C225 in patients (pts) with metastatic/recurrent head & neck cancer (HNC). Proc Am Soc Clin Oncol 21: 226a (abstr 901) [Google Scholar]
  11. Cella D, Eton DT, Fairclough DL, Bonomi P, Heyes AE, Silberman C, Wolf MK, Johnson DH (2002) What is a clinically meaningful change on the functional assessment of cancer therapy-lung (FACT-L) questionnaire? Results from Eastern Cooperative Oncology Group (ECOG) Study 5592. J Clin Epidemiol 55: 285–295 [DOI] [PubMed] [Google Scholar]
  12. Cho CD, Fisher GA, Halsey J, Jambalos CN, Advani RH, Wakelee H, Lum BL, Sikic BI (2003) A phase II study of gefitinib in combination with FOLFOX-4 (IFOX) in patients with unresectable or metastatic colorectal cancer. Poster presented at the ASCO, Chicago, IL, May 31–June 3. Poster number 1062
  13. Ciardiello F, Caputo R, Bianco R, Damiano V, Pomatico G, De Placido S, Bianco AR, Tortora G (2000) Antitumor effect and potentiation of cytotoxic drugs activity in human cancer cells by ZD-1839 (Iressa), an epidermal growth factor receptor-selective tyrosine kinase inhibitor. Clin Cancer Res 6: 2053–2063 [PubMed] [Google Scholar]
  14. Ciardiello F, Tortora G (2001) A novel approach in the treatment of cancer: targeting the epidermal growth factor receptor. Clin Cancer Res 7: 2958–2970 [PubMed] [Google Scholar]
  15. Cohen EEW, Rosen F, Stadler WM, Recant W, Stanson K, Huo D, Vokes EE (2003a) Phase II trial of ZD1839 in recurrent or metastatic squamous cell carcinoma of the head and neck. J Clin Oncol 21: 1980–1987 [DOI] [PubMed] [Google Scholar]
  16. Cohen EEW, Stenson K, Gustin DM, Lamont E, Mauer AM, Blair E, Stadler WM, Dekker A, Mallon W, Vokes EE (2003b) A phase II study of 250-mg gefitinib (ZD1839) monotherapy in recurrent or metastatic squamous cell carcinoma of the head and neck (SCCHN). Proc Am Soc Clin Oncol 22: 502 (abstr 2021) [Google Scholar]
  17. Cohen RB (2003) Epidermal growth factor receptor as a therapeutic target in colorectal cancer. Clin Colorectal Cancer 2: 246–251 [DOI] [PubMed] [Google Scholar]
  18. Cunningham D, Humblet Y, Siena S, Khayat D, Bleiberg H, Santoro A, Bets D, Mueser M, Harstrick A, Van Cutsem E (2003) Cetuximab (C225) alone or in combination with irinotecan (CPT-11) in patients with epidermal growth factor receptor (EGFR)-positive, irinotecan-refractory metastatic colorectal cancer (MCRC). Proc Am Soc Clin Oncol 22: 252 (abstr 1012) [Google Scholar]
  19. Douillard J-Y, Natale R, Giaccone G, Lynch T, Nakagawa K, Brahmer J, Averbuch S, Kay A (2002) Gefitinib (‘Iressa’, ZD1839) provides clinically significant antitumor activity and improves disease-related symptoms in pretreated patients with advanced non-small-cell lung cancer: results of two Phase II trials (IDEAL 1 and IDEAL 2). Poster presented at the EORTC-NCI-AACR. Poster number 177
  20. Finkler N, Gordon A, Crozier M, Edwards R, Figueroa J, Garcia A, Hainsworth J, Irwin D, Silberman S, Allen L, Ferrante K, Fisher D, Nadler P (2001) Phase 2 evaluation of OSI-774, a potent oral antagonist of the EGFR-TK in patients with advanced ovarian carcinoma. Proc Am Soc Clin Oncol 20: 208a (abs 831) [Google Scholar]
  21. Forsythe B, Faulkner K (2003) Safety and tolerability of gefitinib (‘Iressa’, ZD1839) in advanced NSCLC: overview of clinical experience. Poster presented at the ERS 13th Annual Congress, Vienna, Austria, September 27–October 1. Poster number P327
  22. Fukuoka M, Yano S, Giaccone G, Tamura T, Nakagawa K, Douillard J-Y, Nishiwaki Y, Vansteenkiste J, Kudoh S, Rischin D, Eek R, Horai T, Noda K, Takata I, Smit E, Averbuch S, Macleod A, Feyereislova A, Dong R-P, Baselga J (2003) Multi-institutional randomized phase II trial of gefitinib for previously treated patients with advanced non-small-cell lung cancer. J Clin Oncol 21: 2237–2246 [DOI] [PubMed] [Google Scholar]
  23. Gatzemeier U, Rosell R, Ramlau R, Robinet G, Szczesna A, Quoix E, Font A, Jimenez E, Mueser M, Harstrick A (2003) Cetuximab (C225) in combination with cisplatin/vinorelbine vs. cisplatin/vinorelbine alone in the first-line treatment of patients (pts) with epidermal growth factor receptor (EGFR) positive advanced non-small-cell lung cancer (NSCLC). Proc Am Soc Clin Oncol 22: 642 (abstr 2582) [Google Scholar]
  24. Giaccone G, Johnson DH, Manegold C, Scagliotti GV, Rosell R, Wolf M, Rennie P, Ochs J, Averbuch S, Fandi A (2002) A phase III clinical trial of ZD1839 (‘Iressa’) in combination with gemcitabine and cisplatin in chemotherapy-naive patients with advanced non-small-cell lung cancer (INTACT 1). Ann Oncol 13(Suppl 5): 2 (abstr 40) [Google Scholar]
  25. Gonzalez-Larriba JL, Giaccone G, van Oosterom AT, Alfonso R, Smit EF, Martens M, Peters GJ, Van Der Vijgh WJF, Smith R, Fandi A, Averbuch S (2002) ZD1839 (‘Iressa’) in combination with gemcitabine and cisplatin in chemonaive patients with advanced solid tumors: final results of a phase I trial. Poster presented at the ASCO, Orlando, FL, 18–21 May. Poster number 376
  26. Herbst RS, Hong WK (2002) IMC-C225, an anti-epidermal growth factor receptor monoclonal antibody for treatment of head and neck cancer. Semin Oncol 29: 18–30 [DOI] [PubMed] [Google Scholar]
  27. Herbst RS, Kies MS (2002) ZD1839 (Iressa™) in non-small cell lung cancer. Oncologist 7(Suppl 4): 9–15 [DOI] [PubMed] [Google Scholar]
  28. Herbst RS, Langer CJ (2002) Epidermal growth factor receptors as a target for cancer treatment: the emerging role of IMC-C225 in the treatment of lung and head and neck cancers. Semin Oncol 29: 27–36 [DOI] [PubMed] [Google Scholar]
  29. Herbst RS, Maddox AM, Rothenberg ML, Small EJ, Rubin EH, Baselga J, Rojo F, Hong WK, Swaisland H, Averbuch SD, Ochs J, LoRusso PM (2002) Selective oral epidermal growth factor receptor tyrosine kinase inhibitor ZD1839 is generally well-tolerated and has activity in non-small-cell lung cancer and other solid tumors: results of a phase I trial. J Clin Oncol 20: 3815–3825 [DOI] [PubMed] [Google Scholar]
  30. Huang SM, Li J, Armstrong EA, Harari PM (2002) Modulation of radiation response and tumor-induced angiogenesis after epidermal growth factor receptor inhibition by ZD1839 (Iressa). Cancer Res 62: 4300–4306 [PubMed] [Google Scholar]
  31. Inoue A, Saijo Y, Maemondo M, Gomi K, Tokue Y, Kimura Y, Ebina M, Kikuchi T, Moriya T, Nukiwa T (2003) Severe acute interstitial pneumonia and gefitinib. Lancet 361: 137–139 [DOI] [PubMed] [Google Scholar]
  32. Johnson DH, Herbst R, Giaccone G, Schiller J, Natale RB, Miller V, Wolf M, Helton A, Averbuch S, Grous J (2002) ZD1839 (‘Iressa’) in combination with paclitaxel & carboplatin in chemotherapy-naive patients with advanced non-small-cell lung cancer (NSCLC): results from a phase III clinical trial (INTACT 2). Ann Oncol 13(Suppl 5): 127 (abstr 4680)12401678 [Google Scholar]
  33. Kawasaki H, Nagai K, Yoshida J, Nishimura M, Nishiwaki Y (2002) Postoperative morbidity, mortality, and survival in lung cancer associated with idiopathic pulmonary fibrosis. J Surg Oncol 81: 33–37 [DOI] [PubMed] [Google Scholar]
  34. Kies MS, Harari PM (2002) Cetuximab (ImClone/Merck/Bristol-Myers Squibb). Curr Opin Investig Drugs 3: 1092–1100 [PubMed] [Google Scholar]
  35. Kim TE, Murren JR (2002) Erlotinib OSI/Roche/Genentech. Curr Opin Investig Drugs 3: 1385–1395 [PubMed] [Google Scholar]
  36. Kris MG, Natale RB, Herbst RS, Lynch Jr TJ, Prager D, Belini CP, Schiller JH, Kelly K, Spiridonidis H, Sandler A, Cella D, Wolf MK, Averbuch SD, Ochs JJ, Kay AC (2003) Efficacy and safety of gefitinib (Iressa, ZD1839), an inhibitor of the epidermal growth factor receptor tyrosine kinase, in symptomatic patients with advanced non-small cell lung cancer. JAMA 290: 2149–2158 [DOI] [PubMed] [Google Scholar]
  37. Magne N, Fischel JL, Dubreuil A, Formento P, Marcie S, Lagrange J-L, Milano G (2002) Sequence-dependent effects of ZD1839 (‘Iressa’) in combination with cytotoxic treatment in human head and neck cancer. Br J Cancer 86: 819–827 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Milas L, Mason K, Hunter N, Petersen S, Yamakawa M, Ang K, Mendelsohn J, Fan Z (2000) In vivo enhancement of tumor radioresponse by C225 antiepidermal growth factor receptor antibody. Clin Cancer Res 6: 701–708 [PubMed] [Google Scholar]
  39. Miller VA, Johnson D, Heelan RT, Pizzo BA, Perez WJ, Bass A, Kris MG, Ochs J, Averbuch S (2001) A pilot trial demonstrates the safety of ZD1839 (‘Iressa’), an oral epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI), in combination with carboplatin (C) and paclitaxel (P) in previously untreated advanced non-small cell lung cancer (NSCLC). Proc Am Soc Clin Oncol 20: 326a (abstr 1301) [Google Scholar]
  40. Nakagawa K, Tamura T, Negoro S, Kudoh S, Yamamoto N, Yamamoto N, Takeda K, Swaisland H, Nakatani I, Hirose M, Dong R-P, Fukuoka M (2003) Phase I pharmacokinetic trial of the selective oral epidermal growth factor receptor tyrosine kinase inhibitor gefitinib (‘Iressa’, ZD1839) in Japanese patients with solid malignant tumours. Ann Oncol 14: 922–930 [DOI] [PubMed] [Google Scholar]
  41. Natale RB, Zaretsky SL (2002) ZD1839 (Iressa™): what's in it for the patient? Oncologist 7(Suppl 4): 25–30 [DOI] [PubMed] [Google Scholar]
  42. Needle MN (2002) Safety experience with IMC-C225, an anti-epidermal growth factor receptor antibody. Semin Oncol 29: 55–60 [DOI] [PubMed] [Google Scholar]
  43. Normanno N, Campiglio M, De Luca A, Somenzi G, Maiello M, Ciardiello F, Gianni L, Salomon DS, Menard S (2002) Cooperative inhibitory effect of ZD1839 (Iressa) in combination with trastuzumab (Herceptin) on human breast cancer cell growth. Ann Oncol 13: 65–72 [DOI] [PubMed] [Google Scholar]
  44. O’Dwyer PJ, Benson AB (2002) Epidermal growth factor receptor-targeted therapy in colorectal cancer. Semin Oncol 29: 10–17 [DOI] [PubMed] [Google Scholar]
  45. Patel JD, Miller VA, Kris MG, Shah NT, Pizzo B, Tyson L, Zakowski M, Memoli N, Heelan R, Johnson DH (2003) Encouraging activity and durable responses demonstrated by the epithelial growth factor receptor-tyrosine kinase inhibitor, erlotinib (Tarceva TM, OSI774), in patients with advanced bronchioloalveolar (BAC) cell carcinoma. Lung Cancer 41(Suppl 2): S56 (abstr O-188) [Google Scholar]
  46. Perez-Soler R, Chachoua A, Huberman M, Karp D, Rigas J, Hammond L, Rowinsky E, Preston G, Ferrante KJ, Allen LF, Nadler PI, Bonomi P (2001) A phase II trial of the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor OSI-774, following platinum-based chemotherapy, in patients (pts) with advanced, EGFR-expressing, non-small cell lung cancer (NSCLC). Proc Am Soc Clin Oncol 20: 310a (abstr 1235) [Google Scholar]
  47. Prewett MC, Hooper AT, Bassi R, Ellis LM, Waksal HW, Hicklin DJ (2003) Enhanced antitumor activity of anti-epidermal growth factor receptor monoclonal antibody IMC-C225 in combination with irinotecan (CPT-11) against human colorectal tumor xenografts. Clin Cancer Res 8: 994–1003 [PubMed] [Google Scholar]
  48. Ranson M, Hammond LA, Ferry D, Kris M, Tullo A, Murray PI, Miller V, Averbuch S, Ochs J, Morris C, Feyereislova A, Swaisland H, Rowinsky EK (2002) ZD1839, a selective oral epidermal growth factor receptor-tyrosine kinase inhibitor, is well tolerated and active in patients with solid, malignant tumors: results of a phase I trial. J Clin Oncol 20: 2240–2250 [DOI] [PubMed] [Google Scholar]
  49. Robert F, Ezekiel MP, Spencer SA, Meredith RF, Bonner JA, Khazaeli MB, Saleh MN, Carey D, LoBuglio AF, Wheeler RH, Cooper MR, Waksal HW (2001) Phase I study of anti-epidermal growth factor receptor antibody cetuximab in combination with radiation therapy in patients with advanced head and neck cancer. J Clin Oncol 19: 3234–3243 [DOI] [PubMed] [Google Scholar]
  50. Robertson JFR, Gutteridge E, Cheung KL, Owers R, Koehler M, Hamilton L, Gee J, Nicholson RI (2003) Gefitinib (ZD1839) is active in acquired tamoxifen (TAM)-resistant oestrogen receptor (ER)-positive and ER-negative breast cancer: results from a phase II study. Oral presentation at ASCO. Proc Am Soc Clin Oncol 22: 7 (abstr 23) [Google Scholar]
  51. Schiller J, Eek R, Hammond L, Horai T, Gandara D, Noda K, Averbuch S, Wolf M, Kay A, Lowe E (2003) Targeting the epidermal growth factor receptor tyrosine kinase with gefitinib (‘Iressa’, ZD1839): preliminary investigations of baseline factors associated with response in patients with advanced non-small-cell lung cancer. Poster presented at the Molecular Targets for Cancer Therapy. Poster number 314
  52. Schiller JH, Fukuoka M, Natale R, Lynch T, Averbuch S, Kay A (2002) Results from two phase II trials (IDEAL 1 and IDEAL 2) of ZD1839 (‘Iressa’) in patients with locally advanced or metastatic non-small-cell lung cancer. Poster presented at the Chest Congress. Poster number 372
  53. Senzer NN, Soulieres D, Siu L, Agarwala S, Vokes E, Hidalgo M, Silberman S, Allen L, Ferrante K, Fisher D, Marsolais C, Nadler P (2001) Phase 2 evaluation of OSI-774, a potent oral antagonist of the EGFR-TK in patients with advanced squamous cell carcinoma of the head and neck. Proc Am Soc Clin Oncol 20: 2a (abstr 6) [Google Scholar]
  54. Shin DM, Donato NJ, Perez-Soler R, Shin HJC, Wu JY, Zhang P, Lawhorn K, Khuri FR, Glisson BS, Myers J, Clayman G, Pfister D, Falcey J, Waksal H, Mendelsohn J, Hong WK (2001) Epidermal growth factor receptor-targeted therapy with C225 and cisplatin in patients with head and neck cancer. Clin Cancer Res 7: 1204–1213 [PubMed] [Google Scholar]
  55. Sirotnak FM, Zakowski MF, Miller VA, Scher HI, Kris MG (2000) Efficacy of cytotoxic agents against human tumor xenografts is markedly enhanced by coadministration of ZD1839 (Iressa), an inhibitor of EGFR tyrosine kinase. Clin Cancer Res 6: 4885–4892 [PubMed] [Google Scholar]
  56. Siu LL, Hidalgo M, Nemunaitis J, Rizzo J, Moczygemba J, Eckhardt SG, Tolcher A, Smith L, Hammond L, Blackburn A, Tensfeldt T, Silberman S, Von Hoff DD, Rowinsky EK (1999) Dose and schedule-duration escalation of the epidermal growth factor receptor (EGFR) tyrosine kinase (TK) inhibitor CP-358, 774: a phase I and pharmacokinetic (PK) study. Proc Am Soc Clin Oncol 18: 388a (abstr 1498) [Google Scholar]
  57. Solomon B, Hagekyriakou J, Trivett MK, Stacker SA, McArthur GA, Cullinane C (2003) EGFR blockade with ZD1839 (‘Iressa’) potentiates the antitumor effects of single and multiple fractions of ionizing radiation in human A431 squamous cell carcinoma. Int J Radiat Oncol Biol Phys 55: 713–723 [DOI] [PubMed] [Google Scholar]
  58. Tortora G, Caputo R, Damiano V, Fontanini G, Melisi D, Veneziani BM, Zunino F, Bianco AR, Ciardiello F (2001) Oral administration of a novel taxane, an antisense oligonucleotide targeting protein kinase A, and the epidermal growth factor receptor inhibitor Iressa causes cooperative antitumor and antiangiogenic activity. Clin Cancer Res 7: 4156–4163 [PubMed] [Google Scholar]
  59. Wakeling AE, Guy SP, Woodburn JR, Ashton SE, Curry BJ, Barker AJ, Gibson KH (2002) ZD1839 (Iressa): an orally active inhibitor of epidermal growth factor signaling with potential for cancer therapy. Cancer Res 62: 5749–5754 [PubMed] [Google Scholar]
  60. West HL, Franklin WA, Gumerlock P, Vance RB, Lau DHM, Chansky K, Crowley J, McCoy J, Gandara DR (2003) ZD1839 (Iressa) in advanced bronchioloalveolar carcinoma (BAC): a preliminary report of SWOG S0126. Lung Cancer 41(Suppl 2): S56 (abstr O-187) [Google Scholar]
  61. Williams KJ, Telfer BA, Stratford IJ, Wedge SR (2002) ZD1839 (‘Iressa’), a specific oral epidermal growth factor receptor-tyrosine kinase inhibitor, potentiates radiotherapy in a human colorectal cancer xenograft model. Br J Cancer 86: 1157–1161 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Woodburn JR, Kendrew J, Fennell M, Wakeling AE (2000) ZD1839 (‘Iressa’) a selective epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TK1): inhibition of c-fos mRNA, an intermediate marker of EGFR activation, correlates with tumor growth inhibition. Proc Am Assoc Cancer Res 41: 402 (abstr 2552) [Google Scholar]
  63. Xu JM, Azzariti A, Colucci G, Paradiso A (2003) The effect of gefitinib (Iressa, ZD1839) in combination with oxaliplatin is schedule-dependent in colon cancer cell lines. Cancer Chemother Pharmacol 52: 442–448 [DOI] [PubMed] [Google Scholar]

Articles from British Journal of Cancer are provided here courtesy of Cancer Research UK

RESOURCES