Biologically Targeted Treatment of Non–Small-Cell Lung Cancer: Focus on Epidermal Growth Factor Receptor Ronald B. Natale
Abstract The epidermal growth factor receptor (EGFR) has emerged in recent years as a key target of molecular therapy for solid tumors. The postembryonic role of EGFR is normally limited. In cancer, however, abnormal EGFR–tyrosine kinase (TK) activity plays a central role in many of the processes involved in tumor progression, such as proliferation, angiogenesis, invasiveness, decreased apoptosis, and loss of differentiation. Several different approaches have been taken to inhibit EGFR-mediated activity in tumor cells, including monoclonal antibodies directed at the ligand-binding portion of the EGFR and small-molecule agents that directly inhibit the intracellular TK domain of EGFR. Two of these TK inhibitors, gefitinib and erlotinib (OSI-774, Tarceva™), have shown antitumor activity and good tolerability across several tumor types in early dose-finding clinical trials, particularly for non–small-cell lung cancer (NSCLC). In heavily pretreated patients with advanced NSCLC, gefitinib showed clinically significant tumor responses and symptom relief with good tolerability. Based on these results, gefitinib has now been approved for the third-line treatment of advanced NSCLC. The use of gefitinib in standard treatment programs or combined with other molecular targeted agents may substantially improve the outlook for patients with NSCLC or other types of solid tumors. Clinical Lung Cancer, Vol. 5, Suppl. 1, S11-S17, 2003
Introduction The fundamental concepts underlying cancer biology and treatment have changed a great deal during the past decade. In the past, tumors were seen to be composed simply of “cancer cells” and cancer treatment consisted of attacking the tumor from the outside using radiation, chemotherapy, or surgery. The more recent, and more sophisticated, view of cancer is that a tumor represents a microenvironment consisting of genetically heterogeneous tumor cells, stroma cells, immune cells, and blood vessels. This new understanding has changed therapeutic strategies. New therapies are now being designed either to destroy the microenvironment that supports tumor cell growth, or to destroy the way that tumor cells respond to the microenvironment. With respect to the latter, it is now known that tumor response to the microenvironment is mediated by cell-surface receptors and receptor-activated kinases, many of which are abnormally active in cancer. Cedars-Sinai Comprehensive Cancer Center, Los Angeles, CA Submitted: May 1, 2003; Revised: Aug 26, 2003; Accepted: Aug 27, 2003 Address for correspondence: Ronald B. Natale, MD, Cedars-Sinai Comprehensive Cancer Center, 8700 Beverly Blvd, Ste C2000, Los Angeles, CA 90048-1804 Fax: 310-652-8759; e-mail:
[email protected]
Key words: Gefitinib, Targeted therapy, Tyrosine kinase inhibitor
Figure 1
Signal Transduction Pathways Activated by EGFR-TK Ligands = EGF, TGF-α EGFR
P
SOS
P
Ras
Raf
MEK/ERK PI3K
Nucleus MAPK
c-Jun c-Fos Akt
Proliferation Survival Angiogenesis (VEGF, FGF)
Effect on Cell Cycle: S phase (most radiation resistant) G2/M (most radiation sensitive)
Abbreviations: EGF = epidermal growth factor; EGFR = epidermal growth factor receptor; ERK = extracellular signal–regulated kinase; FGF = fibroblast growth factor; MAPK = mitogenactivated protein kinase; MEK = mitogen-activated protein kinase kinase; PI3K = phosphatidylinositol-3 kinase; SOS = son of sevenless; TGF-α = transforming growth factor–α; TK = tyrosine kinase; VEGF = vascular endothelial growth factor
Clinical Lung Cancer Vol 5 • Suppl 1 September 2003
S11
Targeted Treatment of NSCLC: Focus on EGFR Table 1
Expression of EGFR and Its Ligands in NSCLC Rusch,9 1993
Rusch,10 1997
Fontanini,11 1998
EGFR Expression
93%
NA
81%
TGF-α Expression
86%
NA
99%
0
NA
NA
All patients
45%
70%
49%
SCC
68%
92%
57%
Non-SCC
36%
58%
38%
EGF Expression EGFR Overexpression
Abbreviations: EGF = epidermal growth factor; EGFR = epidermal growth factor receptor; NA = not applicable; NSCLC = non–small-cell lung cancer; SCC = squamous cell carcinoma; TGF-α = transforming growth factor−α
Among the most important molecules that drive tumor cell growth are the cell-surface receptors. These signal-transduction molecules have in common a membrane-spanning segment and an intracellular tyrosine kinase (TK) domain, whereas their extracellular domains vary with respect to their ligand-binding specificities. The ErbB family of receptors consists of 4 members: epidermal growth factor receptor (EGFR, ie, ErbB-1), ErbB-2 (ie, HER2/neu), ErbB-3, and ErbB-4.1-3 Of these, EGFR has the greatest number of known ligands: epidermal growth factor (EGF), transforming growth factor–α (TGF-α), amphiregulin, betacellulin, heparin-binding EGF, and epiregulin. ErbB-3 and ErbB-4 bind the heregulins, whereas ErbB-4 also binds neuregulin (NRG) 2, NRG3, and betacellulin. Although ErbB-2 has no known ligand, it is a primary partner for the activation of ErbB-1 and ErbB-3.2,3 Signal transduction through the EGFR is initiated when ligand binds to the extracellular domain (Figure 1). The receptor then dimerizes with various ErbB family members (ErbB-2 primarily), leading to activation of the TK catalytic domain and autophosphorylation of tyrosine residues in the intracellular domain. These phosphorylated tyrosine residues provide docking sites for signal transduction molecules with Src homology or
protein tyrosine binding domains.4 Such signal-transduction molecules include phosphatidylinositol-3 kinases, phospholipase Cγ, the adaptor molecules Shc and growth factor receptor–bound protein 2, and components of the p21 ras signaling cascade. The results of EGFR-TK signaling are pleiotropic, including stimulation of proliferation, suppression of apoptosis, inhibition of hormone secretion, and influences on differentiation and angiogenesis.5 In normal cells, EGFR-TK activity is tightly regulated. In cancer cells, a variety of mechanisms can lead to increased EGFR-TK activity, which results in inappropriate activation of many of these cellular processes.6,7 Increased expression or activation of EGFR is a hallmark of many types of solid human tumors and has been associated with late-stage disease, invasion, metastasis, resistance to chemotherapy and hormone therapy, and poor outcome. In non–small-cell lung cancer (NSCLC), as in other common solid tumors, EGFR and TGF-α are thought to form an autocrine loop that plays an important role in tumor progression.8 In studies of NSCLC, EGFR expression was detected in 81%-93% of tumors and TGF-α expression was seen in 86%-99% (Table 1).9-11 These and other studies found increased levels of EGFR in 45%-70% of resected tumors from patients with NSCLC. Expression levels were variously defined by the intensity of staining or the percentage of cells labeled in tumor samples in comparison to surrounding tissue. Normal, uninvolved tissue was generally found to express little or no EGFR. Thus, any expression in cancerous tissue was considered overexpression.
Novel Approaches to Inhibiting Epidermal Growth Factor Receptor-Tyrosine Kinase
The properties of EGFR described herein have made it an attractive therapeutic target for a broad range of common solid tumors, including NSCLC. The main approaches tested thus far for inhibition of EGFR include monoclonal antibodies (MoAbs) that interact with the EGFR extracellular domain and small-molecule TK inhibitors (TKIs) that are directed at the intracellular catalytic domain of the kinase enzyme. Proof of the concept of EGFR inhibition for cancer therapy will include tumor responses, clinical benefits, and good tolerability in randomized clinical trials.7 The idea of targeting tumor cells with antibodies to HER family of receptors is not new. Humanized MoAbs, such as trastuzumab, with specificity Table 2 Clinical Trials of Epidermal Growth Factor Receptor−Targeted Agents14-21 for HER2 (ErbB-2) have already been developed for advanced breast cancer.12 Chimeric Monotherapy Drug-Related MoAbs, such as the EGFR-specific cetuximab, Maximum EGFRPhase II/III Agent Tolerated Adverse Events* Positive are in advanced clinical trials in combination Trial Dose Dose Screening with cytotoxic therapies.13 The rationale for Rash Diarrhea these studies comes from xenograft tumor 700-1000 mg/day 250-500 mg/day No 37%-50% ≈50% Gefitinib models in which combination treatment with anti-EGFR MoAbs and cytotoxic drugs result150 mg/day 150 mg/day Erlotinib Yes and no 73%-79% 54% ed in enhanced antitumor activity.12 400 mg/m2 A second approach to targeting EGFR was > 400 mg/m2 then Yes 5% 9% Cetuximab 200 mg/m2/week based on the observation that mutations in the adenosine triphosphate (ATP)–binding *Mainly grade 1/2. Abbreviation: EGFR = epidermal growth factor receptor pocket of the receptor block its TK activity.
S12
Clinical Lung Cancer Vol 5 • Suppl 1 September 2003
Ronald B. Natale Pharmacophore modeling was used to design selective EGFRTKIs such as gefitinib and erlotinib (OSI-774, Tarceva™), which competitively inhibit the binding of ATP to the catalytic TK domain.8 In contrast to the extracellular EGFR MoAbs, EGFRTKIs act intracellularly to directly inhibit autophosphorylation of EGFR, thus blocking downstream signaling. Other TKIs in development include EKB-569, which is irreversible; GW572016, which targets both EGFR and HER2; and the irreversible inhibitor CI1033, which can inhibit all 4 ErbB family members.7 The different TKIs are in various stages of clinical testing as monotherapy and in combination with cytotoxic chemotherapy or radiation. EGFR-TKIs are being tested in clinical trials of various solid tumors, with initial development focusing on NSCLC and cancers of the head and neck.8,13 The status of clinical trials for the 3 anti-EGFR agents that are furthest along in development is shown in Table 2.14-21 Gefitinib, erlotinib, and cetuximab have all undergone phase I trials in which the maximum tolerated dose (MTD) was determined. The specificity that these and other molecular agents exhibit for their respective targets means that the clinically effective dose may be considerably lower than the MTD. On the basis of the phase I trials, doses were selected for phase II/III studies that were at the MTD (erlotinib) or below the MTD (gefitinib, cetuximab) for these agents. Randomized phase II clinical trials of gefitinib and erlotinib as monotherapy have resulted in tumor response and clinical benefit. Exploratory trials have shown that gefitinib, erlotinib, and cetuximab can be combined safely with several standard chemotherapy regimens.18,22-28 Gefitinib has recently been approved for the treatment of patients with advanced NSCLC after previous chemotherapy. Despite the different mechanisms of EGFR inhibition by these agents, all 3 had similar tolerability profiles in clinical trials. With respect to the most commonly seen adverse events, skin changes (especially rash) and diarrhea, the profiles suggest that these adverse events represent a class effect of EGFR-TK inhibition (Table 2). The skin rash is a maculopapular or acneiform rash on an erythematous base that is likely caused by the role of EGFR in skin biology.29-32 The rash frequency, and to some extent the severity, are dose-related and generally manageable. Differences in frequency and severity may reflect the pharmacology of the specific agent and the treatment dose. Gefitinib has been associated with a low incidence of rash, which is likely a result of treatment with doses well below the MTD. In trials of gefitinib, the rash rarely led to discontinuation of treatment.
Erlotinib (OSI-774, Tarceva™) in the Treatment of Advanced Non–Small-Cell Lung Cancer In phase I trials of erlotinib alone or in combination with chemotherapy, tumor response or stable disease was observed in patients with a variety of solid tumors.14,24-26 The most common adverse events were diarrhea and skin toxicities. The majority of adverse events were mild and reversible. As monotherapy, the MTD was 150 mg/day. Dose-limiting toxicities were observed with erlotinib at 100 mg/day in combination with cisplatin/gemcitabine and carboplatin/paclitaxel (neutropenia and diarrhea), and at 75 mg/day in combination with docetaxel (neutropenia).24-26
Figure 2 Schematic of Trial Design for IDEAL 1 and IDEAL 251-54 IDEAL 1 Patients (N = 210) ≥ 1 previous platinum chemotherapy regimens IDEAL 2 Patients (N = 216) ≥ 2 prior chemotherapy regimens including platinum and docetaxel
R A N D O M I Z E
250 mg Gefitinib once daily
500 mg Gefitinib once daily
Primary endpoints: • Objective tumor response • Safety profile (IDEAL 1 only) • Symptom improvement (IDEAL 2 only)
Continue gefitinib until disease progression or intolerable toxicity Abbreviation: IDEAL = Iressa Dose Evaluation in Advanced Lung Cancer
In a small phase II trial (N = 56) in patients with advanced, chemotherapy-refractory NSCLC, the objective response rate was 12% and median survival was 37 weeks.33 Stable disease was observed in 26% of patients. Rash was the most common adverse event, occurring in 78% of patients. Although both the phase I and II trials of erlotinib included an evaluation of EGFR expression, tumor response was not correlated with EGFR expression levels.14,33 Erlotinib is currently undergoing phase III evaluation in NSCLC as second- and third-line monotherapy and as firstline therapy in combination with carboplatin/paclitaxel or cisplatin/gemcitabine. In addition, erlotinib is being investigated in a phase III trial as first-line therapy for pancreatic cancer.
The Role of Gefitinib in the Treatment of Advanced Non–Small-Cell Lung Cancer Preclinical Data The EGFR-TKI gefitinib is an orally bioavailable quinazoline that selectively inhibits EGFR-TK activity, with an IC50 (inhibitory concentration 50%) in preclinical studies that is approximately 50 times lower than that for the related ErbB-2 receptor (EGFR IC50 = 0.023-0.079 μmol/L, ErbB-2 IC50 = 1.2-3.7 μmol/L). In a xenograft tumor model, the growth of human A549 NSCLC cells was almost completely inhibited by gefitinib. When treatment was stopped, tumor growth continued. Gefitinib has also shown antitumor activity against xenograft models of prostate, breast, colorectal, gastric, and ovarian tumors.34 Furthermore, gefitinib has shown additive and synergistic antitumor activity in some preclinical studies when combined with cytotoxic chemotherapy or radiation.35-41 For example, combination treatment with gefitinib and the chemotherapy agent paclitaxel achieved an apparent complete tumor regression in an LX-1 lung cancer xenograft model.35 This combination inhibited tumor growth more potently than did either drug alone. Moreover, overexpression of EGFR did not seem to be required for the inhibitory effect, as LX-1 cells express only low levels of EGFR.35
Phase I Monotherapy Trials Four phase I monotherapy trials of gefitinib enrolled a total of 252 patients with a median age of 59 years.42-47 Patients had
Clinical Lung Cancer Vol 5 • Suppl 1 September 2003
S13
Targeted Treatment of NSCLC: Focus on EGFR Table 3
Patient Demographics in IDEAL 1 and IDEAL 251-54 IDEAL 1
IDEAL 2
Gefitinib Gefitinib Gefitinib Gefitinib 250 mg/day 500 mg/day 250 mg/day 500 mg/day Patients Randomized
104
106
102
114
Men/Women
75%/25%
66%/34%
59%/41%
55%/45%
61
60
61
62
0
17%
19%
18%
13%
1
70%
68%
63%
66%
2
13%
13%
19%
20%
3
0
0
0
1%
Median Age (Years) Performance Status
The pharmacodynamics of treatment with gefitinib were also studied in phase I trials. Paired skin biopsies were taken from 65 patients with cancer before therapy and 28 days after therapy with gefitinib was initiated.30 Immunohistochemistry was used to assess the activation (phosphorylation) state of EGFR-TK and its downstream effectors, including mitogen-activated protein kinase, signal transducer and activator of transcription–3, and the Cdk inhibitor p27KIP1 (the latter 2 molecules are linked to keratinocyte maturation). An oral once-daily dose of gefitinib was found to inhibit EGFR-TK activation and affect downstream receptor-dependent processes in patients. The effects of gefitinib were profound at doses well below the MTD.
Phase II Monotherapy Trials
Gefitinib as monotherapy (250 mg/day vs. 500 mg/day) for the treatment of advanced NSCLC was investigated in 2 large randomized phase II trials: 67% 64% 64% 69% Adenocarcinoma* Iressa Dose Evaluation in Advanced Lung Cancer 17% 16% 24% 14% Squamous (IDEAL) 1 and IDEAL 2 (Figure 2).51-54 Both trials enrolled patients with locally advanced or 9% 3% 9% 2% Large cell metastatic NSCLC. In the international trial 8% 17% 15% Other (IDEAL 1), patients had received 1 or 2 previous 3% chemotherapy regimens, of which ≥ 1 was platPrior inum-based.53 In the US trial (IDEAL 2), patients Chemotherapy Regimens had received ≥ 2 previous chemotherapy regimens containing platinum and docetaxel, given either 0 0 1 57% 56% concurrently or separately, and all patients had ≥ 1 42% 40% 2 43% 44% disease-related symptom.51,52 The patients in IDEAL 2, therefore, represented a more extensively 36% 30% 3 0 0 pretreated population with more advanced disease ≥4 compared with those in IDEAL 1. Patient demo22% 28% 0 0 graphics for both trials are shown in Table 3.51-54 Disease Stage The primary endpoints in IDEAL 1 were tumor response rate and safety profile associat† Locally advanced 19.5% 8% ed with gefitinib.53 In IDEAL 2, the primary 80.5%† 92% Metastatic endpoints were tumor response rate and disease-related symptom improvement rate.51 Pa*Includes patients with adenosquamous histology. †Data combined for both doses. tients were therefore required to be Abbreviation: IDEAL = Iressa Dose Evaluation in Advanced Lung Cancer symptomatic at baseline (score of ≤ 24 on the Lung Cancer Subscale [LCS]) in IDEAL 2.52 a variety of solid tumors, including NSCLC (n = 100), colorectal In IDEAL 1, the radiographic tumor response rates were simi(n = 40), ovarian (n = 33), head and neck (n = 30), prostate (n = lar in the 250-mg/day group and the 500-mg/day group, with 19), breast (n = 10), and other types of solid cancer (n = 20). Over overall response rates of 18.4% and 19.0%, respectively (Table the average time of 2.4 months of monotherapy with gefitinib, ad4).53,54 The disease control rates (tumor response plus stable disverse events included diarrhea (grade 3 diarrhea was a dose-limitease) were also comparable for the 2 dose groups. Median overall ing toxicity of gefitinib 700 mg/day),48 grade 1/2 gastrointestinal survival times for patients in these groups were 7.6 months and effects, and skin rash within the first 2 months. At doses of gefi8.0 months, respectively.53,55 In summary, the 250-mg/day and tinib > 600 mg/day, adverse events required an increasing percent500-mg/day dosages provided similar efficacy in this population. age of patients to either reduce their dose level or withdraw from In IDEAL 2, the radiographic tumor response rate was 11.8% the study. No cardiac, marrow, or renal toxicity was documented for patients receiving gefitinib at 250 mg/day. Similar response in this study, nor was toxicity cumulative.16,42-44,46,47,49,50 rates were achieved with gefitinib treatment at 500 mg/day Tumor Histology
S14
Clinical Lung Cancer Vol 5 • Suppl 1 September 2003
Ronald B. Natale Table 4
IDEAL 1 Efficacy Results53,54
Table 5
IDEAL 2 Efficacy Results51,52 Gefitinib 250 mg/day
Gefitinib 500 mg/day
102
114
Gefitinib 250 mg/day
Gefitinib 500 mg/day
103
105
Overall Response Rate (95% CI)
18.4% (11.5%-27.3%)
19.0% (12.1%-27.9%)
Stable Disease Rate
31%
27%
Disease Control Rate (95% CI)*
54.4% (44.3%-64.2%)
51.4% (41.5%-61.3%)
Symptom Response Rate
43%
35%
Median Progression-Free Survival (Months)
2.7
2.8
Median Overall Survival (Months; 95% CI)
6.1 (4.8-7.7)
6.0 (4.3-7.2)
Median Overall Survival (Months)
7.6
8.0
40.3% (28.5%-53.0%)
37.0% (26.0%-49.1%)
Symptom Improvement Rate (95% CI)
*Disease control rate = tumor response + stable disease. Abbreviations: CI = confidence interval; IDEAL = Iressa Dose Evaluation in Advanced Lung Cancer
(Table 5).51,52 Overall, the rate of symptom improvement (an increase of ≥ 2 points on the LCS) was 39%. Symptom improvement was positively correlated with tumor response: in the 250-mg/day group, 100% of patients with an objective tumor response improved by ≥ 2 points on the LCS, as did 90% of patients in the 500-mg/day group. In contrast, among patients with progressive disease in the 250-mg/day and 500-mg/day groups, only 12% and 20%, respectively, showed improvement of ≥ 2 points on the LCS (Figure 3).51,52 Among patients with stable disease, 81% in the 250-mg/day group and 61% of those in the 500-mg/day group showed symptom improvement, indicating that a tumor response is not required for relief of NSCLC-associated symptoms during treatment with gefitinib. Furthermore, symptom improvement was associated with longer progression-free and overall survival times. In the 250mg/day group, median overall survival for patients without symptom improvement was 3.7 months, whereas the median survival for patients with symptom improvement had not yet been reached after a median follow-up of 9.2 months (range, 2.7-13.6 months). In the 500-mg/day group, median overall survival was 3.8 months in patients without symptom improvement and 8.1 months for patients without or with symptom improvement. Similarly, patients with symptom improvement had longer progression-free survival times than did those who did not show symptom improvement: 4.8 months and 1.0 month, respectively, for those in the 250mg/day group, and 3.7 months and 1.1 months, respectively, for those in the 500-mg/day group.52 In both trials, the majority of adverse events were mild. The most common adverse events were grade 1/2 diarrhea and skin rash. Grade 3/4 adverse events were infrequent and occurred less often at the lower dose of 250 mg/day (Table 6).51,53 The percentage of patients who withdrew from treatment because of drugrelated adverse events was low (1.9% and 9.4%, in the 250-mg/day and 500-mg/day groups, respectively, in IDEAL 1, and 1.0% and 4.4%, respectively, in IDEAL 2).55 The results from
Evaluable Patients
Overall Response Rate (95% CI) 11.8% (6.2%-19.7%) 8.8% (4.3%-15.5%)
Abbreviations: CI = confidence interval; IDEAL = Iressa Dose Evaluation in Advanced Lung Cancer
these trials show that oral treatment with gefitinib at 250 mg/day and 500 mg/day in patients with NSCLC provided clinically significant tumor responses and stable disease with good tolerability. With comparable efficacy and better tolerability, a daily dose of 250 mg is recommended in this clinical setting.
Phase III Combination Trials Based on promising additive results in combination with chemotherapy in phase I trials, gefitinib has also been investigated in phase III trials in combination with chemotherapy for the treatment of NSCLC.56,57 These trials were known as the Iressa NSCLC Trials Assessing Combination Therapy (INTACT) 1 (cisplatin/gemcitabine) and INTACT 2 (carboplatin/paclitaxel). Each trial enrolled > 1000 patients with advanced NSCLC who were previously untreated. Patients were randomized to receive either chemotherapy (maximum, 6 cycles) or chemotherapy plus 250-mg/day or 500-mg/day gefitinib. In these trials, there were no significant differences in response rates, time to progression, or survival between the gefitinib and placebo groups. Treatment with gefitinib was not associated with a worsening of adverse events compared with chemotherapy, except for an increased incidence of gastrointestiFigure 3
Symptom Improvement Rate (%)
Evaluable Patients
100
Rates of Symptom Improvement ≥ 2 Points on the Lung Cancer Subscale in IDEAL 251,52 100% 90% 81%
80
Gefitinib 250 mg/day Gefitinib 500 mg/day
61%
60 40
20%
20 0
12% (n = 12) (n = 10) Partial Response
(n = 31) (n = 31) Stable Disease
(n = 59) (n = 59) Progressive Disease
Abbreviation: IDEAL = Iressa Dose Evaluation in Advanced Lung Cancer
Clinical Lung Cancer Vol 5 • Suppl 1 September 2003
S15
Targeted Treatment of NSCLC: Focus on EGFR Table 6
Serious AEs and AEs Leading to Withdrawal in IDEAL 1 and IDEAL 2 Trials51,53 IDEAL 1
IDEAL 2
Gefitinib Gefitinib Gefitinib Gefitinib 500 250 500 250 mg/day mg/day mg/day mg/day
ing of gefitinib with radiation and other novel agents is ongoing. Gefitinib is being evaluated in common solid tumors, including colorectal, head and neck, prostate, breast, gastric, ovarian, and endometrial cancers, as well as less-common tumors such as renal cell carcinoma, glioblastoma, mesothelioma, and transitional cell carcinoma of the urothelium.
References Evaluable Patients
103
106
102
114
Grade 3/4 Drug-Related AEs
3.9%*
27.3%
6.9%
17.5%
Withdrawals Due to DrugRelated AEs
1.9%
9.4%
1.0%
4.4%
*Grade 3 only. Abbreviations: AEs = adverse events; IDEAL = Iressa Dose Evaluation in Advanced Lung Cancer
nal and skin toxicities.56,57 The lack of benefit in these trials may have resulted from issues of timing or sequencing of anti-EGFR therapy in combination with chemotherapy. Alternately, it may be that responsiveness to gefitinib is restricted to an as-yetunidentified subgroup of patients. Some tumors, especially in advanced disease, may have redundancies in deregulated growth pathways that bypass a block of EGFR-TK.58,59 It is hoped that these issues will be resolved through further investigation.
Gefitinib Approved for Treatment of Non–Small-Cell Lung Cancer Based on the results of IDEAL 1 and IDEAL 2, gefitinib has now been approved in the United States, Australia, Japan, Singapore, Korea, and Argentina for second- or third-line treatment of advanced NSCLC after the use of a platinum agent and docetaxel.60 Gefitinib is the first agent to be approved in the class of EGFR-targeted agents, and offers patients with advanced lung cancer a therapeutic option where none existed before. In postmarketing use in Japan, treatment with gefitinib has been associated with a low incidence (1.7%) of interstitial lung disease (ILD).61 Worldwide, the incidence of ILD associated with the use of gefitinib is < 1%. The relationship of gefitinib to these occurrences of ILD is unclear. Interstitial lung disease is a known complication of lung cancer and occurs to varying degrees in association with different chemotherapies and with radiation therapy.62-65 Physicians prescribing gefitinib should be aware of the symptoms of ILD, which include nonproductive cough and shortness of breath, abnormal breath sounds, fever, weight loss, fatigue, muscle and joint pain, and clubbing of the fingernails.66,67 The skin toxicities that occur with the use of gefitinib and other EGFR-targeted agents are generally mild and typically subside with time. If necessary, these side effects can be managed with clindamycin, antibiotics, or corticosteroids.
Conclusion Gefitinib is also being investigated for treatment of early-stage NSCLC and for lung cancer prevention. In addition, clinical test-
S16
Clinical Lung Cancer Vol 5 • Suppl 1 September 2003
1. Yarden Y. The EGFR family and its ligands in human cancer: signalling mechanisms and therapeutic opportunities. Eur J Cancer 2001; 37(suppl 4):S3-S8. 2. Alroy I, Yarden Y. The ErbB signaling network in embryogenesis and oncogenesis: signal diversification through combinatorial ligand-receptor interactions. FEBS Lett 1997; 410:83-86. 3. Hynes NE, Horsch K, Olayioye MA, et al. The ErbB receptor tyrosine family as signal integrators. Endocr Relat Cancer 2001; 8:151-159. 4. Prenzel N, Fischer OM, Streit S, et al. The epidermal growth factor receptor family as a central element for cellular signal transduction and diversification. Endocr Relat Cancer 2001; 8:11-31. 5. Slichenmyer WJ, Fry DW. Anticancer therapy targeting the erbB family of receptor tyrosine kinases. Semin Oncol 2001; 28 (suppl 16):67-79. 6. Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol 2001; 2:127-137. 7. Arteaga CL. The epidermal growth factor receptor: from mutant oncogene in nonhuman cancers to therapeutic target in human neoplasia. J Clin Oncol 2001; 19 (suppl):32s-40s. 8. Ciardiello F. Epidermal growth factor receptor tyrosine kinase inhibitors as anticancer agents. Drugs 2000; 60 (suppl 1):25-32. 9. Rusch V, Baselga J, Cordon-Cardo C, et al. Differential expression of the epidermal growth factor receptor and its ligands in primary non-small cell lung cancers and adjacent benign lung. Cancer Res 1993; 53:2379-2385. 10. Rusch V, Klimstra D, Venkatraman E, et al. Overexpression of the epidermal growth factor receptor and its ligand transforming growth factor a is frequent in resectable non-small cell lung cancer but does not predict tumor progression. Clin Cancer Res 1997; 3:515-522. 11. Fontanini G, De Laurentiis M, Vignati S, et al. Evaluation of epidermal growth factor-related growth factors and receptors and of neoangiogenesis in completely resected stage I-IIIA non-small-cell lung cancer: amphiregulin and microvessel count are independent prognostic indicators of survival. Clin Cancer Res 1998; 4:241-249. 12. Herbst RS. Targeted therapy in non-small-cell lung cancer. Oncology 2002; 16:19-24. 13. Dancey JE, Schoenfeldt M. Clinical trials referral resource: epidermal growth factor receptor inhibitors in clinical trials. Oncology (Huntingt) 2001; 15:748-758. 14. Hidalgo M, Siu LL, Nemunaitis J, et al. Phase I and pharmacologic study of OSI-774, an epidermal growth factor receptor tyrosine kinase inhibitor, in patients with advanced solid malignancies. J Clin Oncol 2001; 19:3267-3279. 15. Kusaba H, Tamura T, Nakagawa K, et al. A phase I intermittent dose-escalation trial of ZD1839 (Iressa™) in Japanese patients with solid malignant tumors. Clin Cancer Res 2000; 6 (suppl):4543s (Abstract #381). 16. Baselga J, Herbst R, LoRusso P, et al. Continuous administration of ZD1839 (Iressa), a novel oral epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI), in patients with five selected tumor types: evidence of activity and good tolerability. Proc Am Soc Clin Oncol 2000; 19:177a (Abstract #686). 17. Bonomi P, Perez-Soler R, Chachoua A, et al. A phase II trial of the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI), CP-358,774, following platinum-based chemotherapy in patients (pts) with advanced non-small cell lung cancer (NSCLC). Clin Cancer Res 2000; 6 (suppl):4544s (Abstract #386). 18. Laurie SA, Miller VA, Johnson D, et al. Pilot trial of ZD1839 (Iressa™), an oral inhibitor of epidermal growth factor receptor (EGFR) tyrosine kinase, in combination with carboplatin (C) and paclitaxel (P) in previously untreated advanced non-small cell lung cancer (NSCLC). Lung Cancer 2000; 29 (suppl 1):71 (Abstract #230). 19. Bonner JA, Ezekiel MP, Robert F, et al. Continued response following treatment with IMC-C225, an EGFr MoAb combined with RT in advanced head and neck malignancies. Proc Am Soc Clin Oncol 2000; 19:4a (Abstract #5F). 20. Cohen RB, Falcey JW, Paulter VJ, et al. Safety profile of the monoclonal antibody (MoAb) IMC-C225, an anti-epidermal growth factor receptor (EGFr) used in the treatment of EGFr-positive tumors. Proc Am Soc Clin Oncol 2000; 19:474a (Abstract #1862). 21. Siu LL, Hidalgo M, Nemunaitis J, et al. 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 1999; 18:388a (Abstract #1498). 22. Miller VA, Johnson D, Heelan RT, et al. 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 2001; 20:326a (Abstract #1301). 23. Gonzalez-Larriba JL, Giaccone G, van Oosterom AT, et al. ZD1839 (‘Iressa’) in combination with gemcitabine and cisplatin in chemonaive patients with advanced solid tumours: final results of a phase I trial. Proc Am Soc Clin Oncol 2002; 21:95a (Abstract #376).
Ronald B. Natale 24. Forouzesh B, Hidalgo M, Takimoto C, et al. Phase I, pharmacokinetic (PK), and biological studies of the epidermal growth factor-tyrosine kinase (EGFR-TK) inhibitor OSI-774 in combination with docetaxel. Proc Am Soc Clin Oncol 2002; 21:21a (Abstract #81). 25. Forero L, Patnaik A, Hammond LA, et al. Phase I, pharmacokinetic (PK) and biologic study of OSI-774, a selective epidermal growth factor receptor (EGFR) tyrosine kinase (TK) inhibitor in combination with paclitaxel and carboplatin. Proc Am Soc Clin Oncol 2002; 21:25b (Abstract #1908). 26. Ratain MJ, George CM, Janisch L, et al. Phase I trial of erlotinib (OSI-774) in combination with gemcitabine (G) and cisplatin (P) in patients with advanced solid tumors. Proc Am Soc Clin Oncol 2002; 21:76b (Abstract #2115). 27. Abbruzzese JL, Rosenberg A, Xiong Q, et al. Phase II study of anti-epidermal growth factor receptor (EGFR) antibody cetuximab (IMC-C225) in combination with gemcitabine in patients with advanced pancreatic cancer. Proc Am Soc Clin Oncol 2001; 20:130a (Abstract #518). 28. Hong WK, Arquette M, Nabell L, et al. Efficacy and safety of the anti-epidermal growth factor antibody (EGFR) IMC-C225, in combination with cisplatin in patients with recurrent squamous cell carcinoma of the head and neck (SCCHN) refractory to cisplatin containing chemotherapy. Proc Am Soc Clin Oncol 2001; 20:224a (Abstract #895). 29. Jost M, Kari C, Rodeck U. The EGF receptor—an essential regulator of multiple epidermal functions. Eur J Dermatol 2000; 10:505-510. 30. Albanell J, Rojo F, Averbuch S, et al. 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 2002; 20:110-124. 31. Van Doorn R, Kirtschig G, Scheffer E, et al. Follicular and epidermal alterations in patients treated with ZD1839 (Iressa), an inhibitor of the epidermal growth factor receptor. Br J Dermatol 2002; 147:598-601. 32. Busam KJ, Capodieci P, Motzer R, et al. Cutaneous side-effects in cancer patients treated with the antiepidermal growth factor receptor antibody C225. Br J Dermatol 2001; 144:1169-1176. 33. Perez-Soler R, Chachoua A, Huberman M, et al. 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, nonsmall cell lung cancer (NSCLC). Proc Am Soc Clin Oncol 2001; 20:310a (Abstract #1235). 34. Wakeling AE, Guy SP, Woodburn JR, et al. ZD1839 (Iressa): an orally active inhibitor of epidermal growth factor signaling with potential for cancer therapy. Cancer Res 2002; 62:5749-5754. 35. Sirotnak FM, Zakowski MF, Miller VA, et al. 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 2000; 6:4885-4892. 36. Ciardiello F, Caputo R, Bianco R, et al. 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 2000; 6:2053-2063. 37. Williams KJ, Telfer BA, Stratford IJ, et al. Combination of ZD1839 (“Iressa”), an EGFR-TKI, and radiotherapy increases antitumour efficacy in a human colon cancer xenograft model. Proc Am Assoc Cancer Res 2001; 42:715 (Abstract #3840). 38. She Y, Lee F, Haimovitz-Friedman A, et al. The selective epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) ZD1839 (‘Iressa’) enhances radiation sensitivity of human tumor xenografts in nude mice. Proc Am Assoc Cancer Res 2002; 43:786 (Abstract #3894). 39. Ciardiello F, Damiano V, Bianco R, et al. Inhibition of EGFR-tyrosine kinase by ZD1839 (‘Iressa’) leads to antiproliferative and antiangiogenic effects in human GEO colon cancer xenografts, in combination with paclitaxel. Proc Am Assoc Cancer Res 2001; 42:854 (Abstract #4580). 40. Solomon B, Hagekyriakou J, Trivett M, et al. Potentiation of the antitumor effect of ionizing radiation by ZD1839 (“Iressa”) in vitro and in vivo in A431 cells. Proc Am Assoc Cancer Res 2002; 43:1002 (Abstract #4966). 41. Bianco R, Bianco C, Caputo R, et al. Selective inhibition of the epidermal growth factor receptor tyrosine kinase (EGFR-TK) by ZD1839 (‘Iressa’) potentiates the antitumor effects of ionizing radiation. Proc Am Assoc Cancer Res 2002; 43:1003 (Abstract #4972). 42. Ranson M, Hammond LA, Ferry D, et al. 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 2002; 20:2240-2250. 43. Goss GD, Hirte H, Lorimer I, et al. Final results of the dose escalation phase of a phase I pharmacokinetics (PK), pharmacodynamic (PD), and biological activity study of ZD1839: NCIC CTG Ind. 122. Proc Am Soc Clin Oncol 2001; 20:85a (Abstract #335). 44. Negoro S, Nakagawa K, Fukuoka M, et al. Final results of a phase I intermittent doseescalation trial of ZD1839 (‘Iressa’) in Japanese patients with various solid tumours. Proc Am Soc Clin Oncol 2001; 20:324a (Abstract #1292). 45. Baselga J, LoRusso P, Herbst R, et al. A pharmacokinetic/pharmacodynamic trial of ZD1839 (‘Iressa’), a novel oral epidermal growth factor receptor tyrosine kinase (EGFR-TK) inhibitor, in patients with five selected tumor types (a phase I/II trial of continuous once-daily treatment). Presented at: American Association for Cancer Research-National Cancer Institute-European Organization for Research and Treatment
of Cancer; November 16-19, 1999; Washington, DC. 46. Herbst RS, Maddox AM, Rothenberg ML, et al. 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 2002; 20:3815-3825. 47. Baselga J, Rischin D, Ranson M, et al. 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 2002; 20:4292-4302. 48. Kris M, Ranson M, Ferry D, et al. Phase I study of oral ZD1839 (‘Iressa’), a novel inhibitor of epidermal growth factor receptor tyrosine kinase (EGFR-TK): evidence of good tolerability and activity. Presented at: American Association for Cancer ResearchNational Cancer Institute-European Organization for Research and Treatment of Cancer; November 16-19, 1999; Washington, DC. 49. Ferry D, Hammond L, Ranson M, et al. Intermittent oral Zd1839 (Iressa), a novel epidermal growth factor receptor tyrosine kinase inhibitor (Egfr-Tki) shows evidence of good tolerability and activity: final results from a phase I study. Proc Am Soc Clin Oncol 2000; 19:3a (Abstract #5E). 50. Kris M, Ranson M, Ferry D, et al. Phase I study of oral ZD1839 (Iressa™), a novel inhibitor of epidermal growth factor receptor tyrosine kinase (EGFR-TK): evidence of good tolerability and activity. Clin Cancer Res 1999; 5 (suppl):3749s-3750s (Abstract #99). 51. Kris MG, Natale RB, Herbst RS, et al. A phase II trial of ZD1839 (‘Iressa’) in advanced non-small cell lung cancer (NSCLC) patients who had failed platinum- and docetaxel-based regimens (IDEAL 2). Proc Am Soc Clin Oncol 2002; 21:292a (Abstract #1166). 52. Natale RB, Skarin AT, Maddox AM, et al. Improvement in symptoms and quality of life for advanced non-small-cell lung cancer patients receiving ZD1839 (‘Iressa’) in IDEAL 2. Proc Am Soc Clin Oncol 2002; 21:292a (Abstract #1167). 53. Fukuoka M, Yano S, Giaccone G, et al. Final results from a phase II trial of ZD1839 (‘Iressa’) for patients with previously treated advanced non-small-cell lung cancer (IDEAL 1). Proc Am Soc Clin Oncol 2002; 21:298a (Abstract #1188). 54. Douillard JY, Giaccone G, Horai T, et al. Improvement in disease-related symptoms and quality of life in patients with advanced non-small-cell lung cancer (NSCLC) treated with ZD1839 (‘Iressa’) (IDEAL 1). Proc Am Soc Clin Oncol 2002; 21:299a (Abstract #1195). 55. Fukuoka M, Yano S, Giaccone G, et al. Multi-institutional randomized phase II trial of gefitinib for previously treated patients with advanced non–small-cell lung cancer. J Clin Oncol 2002; 21:2237-2246. 56. Giaccone G, Johnson DH, Manegold C, et al. 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 2002; 13 (suppl 5):2-3 (Abstract #4O). 57. Johnson DH, Herbst R, Giaccone G, et al. 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 2002; 13 (suppl 5):127-128 (Abstract #468O). 58. Chakravarti A, Loeffler JS, Dyson NJ. Insulin-like growth factor receptor I mediates resistance to anti-epidermal growth factor receptor therapy in primary human glioblastoma cells through continued activation of phosphoinositide 3-kinase signaling. Cancer Res 2002; 62:200-207. 59. Bianco R, Yakes FM, Mills GB, et al. EGF receptor-overexpressing tumor cells that lack PTEN/MMAC1 and RB are resistant to small molecule EGFR kinase inhibitors. Proc Am Assoc Cancer Res 2001; 42:851 (Abstract #4567). 60. Iressa® (gefitinib) tablets prescribing information. Wilmington, DE: AstraZeneca Pharmaceuticals LP; May 2003 . 61. Inoue A, Saijo Y, Maemondo M, et al. Severe acute interstitial pneumonia and gefitinib. Lancet 2003; 361:137-139. 62. Reckzeh B, Merte H, Pflüger KH, et al. Severe lymphocytopenia and interstitial pneumonia in patients treated with paclitaxel and simultaneous radiotherapy for non–small-cell lung cancer. J Clin Oncol 1996; 14:1071-1076. 63. Chen YM, Perng RP, Lin WC, et al. Phase II study of docetaxel and gemcitabine combination chemotherapy in non–small-cell lung cancer patients failing previous chemotherapy. Am J Clin Oncol 2002; 25:509-512. 64. Abid SH, Malhotra V, Perry MC. Radiation-induced and chemotherapy-induced pulmonary injury. Curr Opin Oncol 2001; 13:242-248. 65. Thomas AL, Cox G, Sharma RA, et al. Gemcitabine and paclitaxel associated pneumonitis in non-small cell lung cancer: report of a phase I/II dose-escalating study. Eur J Cancer 2000; 36:2329-2334. 66. Collard HR, King TE Jr. Demystifying idiopathic interstitial pneumonia. Arch Intern Med 2003; 163:17-29. 67. American Thoracic Society/European Respiratory Society International Multidisciplinary Consensus Classification of the Idiopathic Interstitial Pneumonias. This joint statement of the American Thoracic Society (ATS), and the European Respiratory Society (ERS) was adopted by the ATS board of directors, June 2001 and by the ERS Executive Committee, June 2001. Am J Respir Crit Care Med 2002; 165:277-304.
Clinical Lung Cancer Vol 5 • Suppl 1 September 2003
S17