throbber
Vol. 9, 5813–5824, December 1, 2003
`
`Clinical Cancer Research
`
`5813
`
`Review
`Targeting the Epidermal Growth Factor Receptor in Non-Small Cell
`Lung Cancer
`
`Roy S. Herbst1 and Paul A. Bunn, Jr.2
`1Department of Thoracic/Head and Neck Medical Oncology, The
`University of Texas, M. D. Anderson Cancer Center, Houston, Texas,
`and 2Tobacco Related Malignancies Program, University of Colorado
`Cancer Center and Division of Medical Oncology, University of
`Colorado Health Sciences Center, Denver, Colorado
`
`Abstract
`Fifteen % or fewer of patients with non-small cell lung
`cancer (NSCLC) survive 5 years. The current standard of care
`for patients with locally advanced or metastatic NSCLC is
`systemic chemotherapy with a two-drug combination regimen
`that includes a platinum agent. Although systemic chemother-
`apy reduces the rate of death attributable to lung cancer,
`disease progression is inevitable and dose-limiting toxicities
`restrict their use. New molecularly targeted therapies aim to
`inhibit specific pathways and key molecules implicated in tu-
`mor growth and progression while sparing normal cells. Sev-
`eral therapies, which target signal transduction pathways in-
`volved in angiogenesis, metastasis, and apoptosis, are in clinical
`development to treat lung cancer. Among these targeted ther-
`apies are the oral, small-molecule epidermal growth factor
`receptor-tyrosine kinase (EGFR-TK) inhibitors gefitinib and
`erlotinib. Both therapies have been validated preclinically as
`new treatment approaches for NSCLC and have shown single-
`agent activity against advanced, chemorefractory NSCLC in
`clinical trials. This article focuses on the biology of the
`EGFR-TK signal transduction pathway, its role in the prolif-
`eration of solid tumors, and the rationale for the clinical de-
`velopment of EGFR-TK inhibitors. We also review clinical
`trials with EGFR-TK inhibitors in NSCLC and future direc-
`tions of investigation with these targeted agents.
`
`Introduction
`The TK3 activity of the EGFR has received considerable
`attention as a target for cancer therapy (1, 2). In recent clinical
`
`Received 5/28/03; revised 8/5/03; accepted 8/6/03.
`Grant support: Supported in part by an M.D. Anderson Cancer Center
`Physician Scientist Program Award and an American Society of Clinical
`Oncology Career Development Award (R. S. H.).
`The costs of publication of this article were defrayed in part by the
`payment of page charges. This article must therefore be hereby marked
`advertisement in accordance with 18 U.S.C. Section 1734 solely to
`indicate this fact.
`Requests for reprints: Roy S. Herbst, Department of Thoracic/Head
`and Neck Medical Oncology, M. D. Anderson Cancer Center, 1515
`Holcombe Boulevard, Unit 432, Houston, TX 77030. Phone: (713) 792-
`6363; Fax: (713) 796-8655; E-mail: rherbst@mdanderson.org.
`3 The abbreviations used are: TK, tyrosine kinase; EGFR, epidermal
`growth factor receptor; NSCLC, non-small cell lung cancer; SWOG,
`Southwest Oncology Group; ECOG, Eastern Cooperative Oncology
`Group; HER, human EGF-like receptor; TGF, transforming growth
`factor; PI3K, phosphatidylinositol 3⬘-kinase; MAPK, mitogen-activated
`
`trials, selective and orally active EGFR-TK inhibitors gefitinib
`[IRESSA (ZD1839); AstraZeneca] and erlotinib [Tarceva (OSI-
`774); OSI and Genentech] produced objective tumor responses
`and symptom improvement in some patients with NSCLC who
`had previously received chemotherapy (3–5). This was the first
`class of oral targeted therapies to produce such responses in
`advanced NSCLC. Although chemotherapy can result in life-
`threatening toxicities, the EGFR-TK inhibitors have far better
`safety profiles in patients with advanced NSCLC.
`Lung cancer is the leading cause of cancer death in both
`men and women in the United States and throughout the world
`(6, 7). The 5-year survival rate for lung cancer patients remains
`very poor with 15% or less surviving 5 years (6). Nonetheless,
`this is improved compared with the 5% 5-year survival rate in
`the United States in the 1960s and the 5% rate still seen in many
`parts of the world. The major reasons for the poor survival rate
`for lung cancer are the lack of effective screening and early
`diagnosis procedures, the propensity for early metastasis, and
`the inability of systemic therapies to cure patients with widely
`metastatic disease. This is not to conclude that there have been
`no advances in lung cancer therapy. Systemic chemotherapy
`produces a 26–32% reduction in the hazard rate of death for
`patients with advanced stage III/IV NSCLC that includes ade-
`nocarcinomas, squamous carcinomas, and large cell carcinomas
`(8–10). Chemotherapy also reduces lung cancer-related symp-
`toms and improves quality of life in patients with advanced
`NSCLC (8–11).
`The current standard of care for patients with locally ad-
`vanced or metastatic NSCLC is systemic chemotherapy with a
`two-drug combination regimen that includes a platinum agent
`(8). Such two-drug combinations, developed in the 1990s, were
`shown to be more effective than the best supportive care or
`treatment with a single chemotherapy agent. These two-drug
`combinations were also shown to be as effective as, but less
`toxic than, combinations of three or more chemotherapy drugs.
`The efficacy is similar for several of these two-drug combina-
`tions. Trials of the SWOG and the ECOG compared five dif-
`ferent two-drug combinations in previously untreated patients
`with advanced NSCLC and found that they had similar efficacy
`in terms of tumor response rates and overall survival (Table 1;
`Refs. 12 and 13). Other large randomized trials from the United
`States and Europe have also shown the equivalence of a number
`of two-drug combination regimens, and superiority compared
`with single-agent chemotherapy regardless of whether the single
`agent is an older agent such as cisplatin or a newer agent such
`as paclitaxel, docetaxel, or gemcitabine (14–18).
`The development of NSCLC disease progression on chem-
`
`protein kinase; mAb, monoclonal antibody; IDEAL, IRESSA dose
`evaluation in advanced lung cancer; LCS, Lung Cancer Subscale;
`FACT-L, Functional Assessment of Cancer Therapy—Lung; INTACT,
`(cid:50)(cid:54)(cid:44)(cid:3)(cid:21)(cid:19)(cid:22)(cid:25)
`IRESSA NSCLC trial assessing combination therapy.
`(cid:36)(cid:51)(cid:50)(cid:55)(cid:40)(cid:59)(cid:3)(cid:57)(cid:17)(cid:3)(cid:50)(cid:54)(cid:44)
`(cid:44)(cid:51)(cid:53)(cid:21)(cid:19)(cid:20)(cid:25)(cid:16)(cid:19)(cid:20)(cid:21)(cid:27)(cid:23)
`
`

`

`5814 Targeting EGFR in NSCLC
`
`Table 1 Overview of efficacy results from large comparative trials
`of first-line chemotherapy regimens for advanced NSCLCa
`Trial end point
`SWOG 9509
`ECOG 1594
`Response, %
`25–28
`17–22
`Median survival, mo
`8.1–8.6
`7.4–8.1
`Time to tumor progression, mo
`4
`3.1–4.2
`1-year survival, %
`36–38
`31–36
`a See Refs. 12, 13.
`
`otherapy is inevitable, because these regimens do not result in
`cure. Until recently, there were no Food and Drug Administra-
`tion approved agents for use in the second-line setting. Do-
`cetaxel was approved on the basis of randomized trials in
`patients whose disease had progressed on platinum-based chem-
`otherapy (19, 20). The objective response rates of docetaxel
`were only 5–10% associated with a modest survival improve-
`ment. No agent produced tumor response in more than 5% of
`patients in the third-line treatment setting.
`The cytotoxic mechanism of action of chemotherapy
`agents imposes inherent limitations on their use. These agents
`nonspecifically kill normal proliferating cells and, as a result,
`are frequently associated with dose-limiting toxicities. Many of
`these effects, such as nausea, vomiting, and hair loss, are trou-
`bling to the patient but are not life threatening. Perhaps the most
`troubling effect is fatigue. Other frequent toxicities may be
`disabling even if not life threatening. Among these would be the
`neuropathy associated with paclitaxel and the severe fluid re-
`tention or effusion associated with docetaxel. All of the cyto-
`toxic chemotherapy agents produce hematological toxicities that
`are often life threatening and occasionally fatal. The careful
`observation of sequential blood counts and the i.v. infusions of
`chemotherapeutic agents and their supportive agents are expen-
`sive and inconvenient for the patient. Such toxicities often result
`in treatment adjustments such as dose reduction, delayed ad-
`ministration or, in some cases, discontinuation. Furthermore,
`with increasing rounds of chemotherapy, there is an increased
`risk that tumors will develop multidrug resistance, thus limiting
`future therapeutic options.
`Toxicities associated with chemotherapy may interfere
`with the ability of some patients with advanced NSCLC to
`receive the standard two-drug combination chemotherapy regi-
`mens. Such patients include the elderly (ⱖ70 years of age),
`patients with poor performance status, and patients with comor-
`bidities. Several studies in elderly patients show that less than
`one-third receive therapy although it may prolong survival (21).
`In addition, patients with a poor performance status of 2 expe-
`rienced a high rate of serious adverse events in the ECOG 1594
`study of combination chemotherapy regimens (13). The study
`design was subsequently amended to include only patients with
`an ECOG performance status of 0 or 1, because patients with
`poorer performance status are, in general, more likely to expe-
`rience adverse events.
`There is a need for new therapies with novel mechanisms
`of action that are well tolerated, effective, and convenient. The
`molecularly targeted agents that are in clinical development aim
`at inhibiting specific pathways and key molecules in tumor
`growth and progression, sparing normal cells. Examples of such
`
`agents that were recently approved by the Food and Drug
`Administration include trastuzumab, a mAb targeting the HER2/
`neu receptor protein in breast cancer;
`imatinib [Gleevec
`(STI571); Novartis], a small molecule receptor TK inhibitor
`targeting Bcr/abl in chronic myelogenous leukemia and c-Kit in
`gastrointestinal stromal tumors, (22, 23); and gefitinib, an orally
`active EGFR-TK inhibitor used as monotherapy in the treatment
`of patients with advanced or metastatic NSCLC who have failed
`to respond to platinum-based and docetaxel chemotherapies.
`Depending on the specific molecule targeted and the mechanism
`of inhibition, these agents may offer novel clinical benefits
`compared with outcomes with cytotoxic chemotherapy, or at
`least the minimum comparable benefits with reduced general
`toxicity and improved convenience.
`A variety of new approaches to treat lung cancer that
`target signal transduction pathways involved in angiogenesis,
`metastasis, and apoptosis (24 –26) are in clinical develop-
`ment. These agents inhibit a wide variety of tumor-associated
`molecules including matrix metalloproteinase, farnesyltrans-
`ferase, and a number of protein kinases. The various thera-
`peutic approaches to inhibiting these molecules include
`mAbs, small-molecule inhibitors, antisense oligonucleotides,
`biological response modifiers, and vaccines (24 –26). Among
`these various approaches are small-molecule inhibitors of
`tumor cell TKs. Gefitinib and imatinib have been validated
`clinically as new treatment approaches for malignancies
`(3–5, 23). Furthermore, the EGFR-TK inhibitors gefitinib
`and erlotinib have shown single-agent activity against ad-
`vanced, chemorefractory NSCLC in clinical trials described
`below (3–5, 27).
`
`EGFR-TK: A Molecular Target in NSCLC
`EGFR-TK Biology and Signaling in Solid Tumors.
`The EGFR is a cell surface receptor encoded by the HER1 (HER
`type 1) or ErbB1 gene (1). EGFR belongs to a family of receptor
`TKs that
`includes HER2/neu (ErbB2), HER3 (ErbB3), and
`HER4 (ErbB4). EGF and TGF-␣ are the two predominant
`ligands for EGFR (28, 29). The binding of these ligands to the
`extracellular domain of EGFR results in dimerization of the
`receptor monomer with another EGFR molecule or another
`member of the HER family (Fig. 1). Dimerization produces
`structural changes in the intracellular portion of the receptor that
`activate the TK domain. The enzymatic activity of EGFR-TK
`transfers phosphate moieties from ATP to specific tyrosine
`residues in the cytoplasmic tail of the EGFR protein. These
`phosphotyrosine residues then act as docking sites for various
`downstream effectors (Fig. 2). Some of these effectors are
`adapter molecules, such as growth factor receptor-bound protein
`2 (Grb2) and Src homology collagen protein (Shc), which serve
`as platforms to assemble the downstream signaling elements
`necessary for activating cellular proliferation (30). Other mole-
`cules are enzymes that are activated on EGFR-TK-dependent
`phosphorylation, including son of sevenless (SOS), PI3K, and
`Grb 2-associated binder-1 (Gab-1). Multiple major signal trans-
`duction pathways are initiated by EGFR autophosphorylation,
`including the Ras-MAPK signaling cascade, Src, and the signal
`transducers and activators of transcription (STAT) pathways,
`which are widely used by growth signals to induce gene tran-
`
`

`

`Clinical Cancer Research
`
`5815
`
`EGFR-TK (34). In addition, EGFR-TK interacts with com-
`ponents of the integrin pathway involved in cell– cell adhe-
`sion, which is crucial for tumor cell invasion of adjacent
`tissues (29, 35, 36). EGFR-TK also promotes invasiveness
`through the up-regulation or activation of matrix metallopro-
`teinases and stimulates tumor cell motility that further con-
`tributes to metastasis (37–39).
`EGFR-TK activation indirectly inhibits apoptosis in tu-
`mor cells, promoting tumor cell survival and resistance to
`cytotoxic therapies. This activity is mediated by PI3K, which
`activates Akt, an important signaling molecule in antiapo-
`ptotic pathways involving the transcription factor nuclear
`factor ␬B. Akt also regulates activity of the Ras-MAPK
`pathway, which is important for cellular proliferation (29).
`Interaction with signals from heterologous pathways, includ-
`ing those activated by stress inducers, neurotransmitters,
`hormones, and lymphokines, adds additional complexity to
`the EGFR-TK signaling network (31). These pathways in-
`volve G-protein-coupled receptors, which can transactivate
`EGFR. Cross-talk between EGFR and other receptors allows
`for EGFR-TK signals to activate other pathways.
`Increased expression of EGFR and its signaling pathways
`has been associated with a high percentage of tumors in the
`lung, breast, head and neck, colon, prostate, esophagus, and
`cervix (1, 2). These elevated levels of EGFR may be the result
`of transcriptional or posttranscriptional alterations or genomic
`mutation (34). Differences in the methodologies used and in the
`criteria for determining EGFR expression levels make it diffi-
`cult to compare study results (2, 40). Various methods of meas-
`
`Fig. 2 Key signal transduction pathways of activated EGFR-TK and
`the various pathways affected. Grb2, growth factor receptor-bound
`protein 2; SOS, son of sevenless; STAT, signal transducer(s) and acti-
`vator(s) of transcription; MEK, MAP/Erk (extracellular regulated ki-
`nase) kinase.
`
`Fig. 1 Homodimers and heterodimers within the human EGFR (HER)/
`ErbB family, and subsequent phosphorylation.
`
`scription and promote diverse cell responses. The particular
`dimer combinations that form at the cell surface after ligand
`binding determine which signaling molecules will be recruited
`to the surface (29–33).
`Ligand binding is the most extensively studied mecha-
`nism of EGFR-TK activation, but a variety of other cellular
`mechanisms are now known to influence EGFR-TK activity
`in tumor cells. For example, some mutations in the EGFR
`gene result in expression of EGFR proteins with constitu-
`tively activated TK activity, the most well-known being the
`EGFRvIII mutation. Defective inactivation mechanisms (e.g.,
`phosphatases, and receptor endocytosis and degradation) may
`also result in sustained signaling. Heterologous receptors and
`signal
`transduction pathways,
`including interactions or
`dimerization with other ErbB receptor types, have been
`shown to cross-activate EGFR-TK.
`Cellular proliferation as a result of EGFR-TK activation
`may occur via several signal transduction pathways; however,
`proliferation signals are strongly mediated by the MAPK path-
`way. After recruitment of adapter molecules on the activated
`EGFR complex, stepwise activation of Ras, Raf, MAP/Erk
`kinase (MEK1), and extracellular regulated kinase (Erk) pro-
`teins leads to increased activity of transcription factors, such as
`Elk1 and c-fos, key molecules that prime the cell for prolifera-
`tion and activate cell cycle progression (29). Activated EGFR
`has been shown to induce the expression of cyclin D, which is
`crucial in cell cycle progression and is commonly increased in
`solid tumors.
`Activated EGFR-TK also influences the malignant pro-
`gression of solid tumors. TGF-␣ and EGF induce angiogen-
`esis by up-regulating the expression of vascular endothelial
`growth factor (VEGF) in tumor cells. Increased microvessel
`density has been found in tumors that express activated
`
`

`

`5816 Targeting EGFR in NSCLC
`
`Fig. 3 Expression patterns of EGFR in NSCLC. Scoring on immunohistochemistry scale, staining intensities: 0, none (A); 1⫹, mild (B); 2⫹,
`moderate (C); 3⫹, strong (D). All of the panels are stained with an anti-EGFR antibody.
`
`uring EGFR levels in tumor tissues include immunohistochem-
`istry (Fig. 3), immunoassays, and assessment of RNA levels.
`Although some studies have shown a correlation between high
`expression of EGFR and decreased survival times, most studies
`of NSCLC patients have failed to show that EGFR expression is
`independently prognostic of survival (2, 34). The prognostic
`value of EGFR expression is increased when analyzed in con-
`junction with its dimeric partners, such as HER2/neu/ErbB2, or
`with ligands, such as TGF-␣ or EGF (41–43). High levels of
`EGFR in tumors result in an increase in EGFR ligand-binding
`sites and higher levels of the TK enzyme, as well as an increase
`in initiation sites for signal transduction inside the tumor cell.
`These findings indicate that there is an important role for aber-
`rant EGFR signaling in the development and progression of
`various human tumors. In addition, they provide a strong ration-
`ale for EGFR-TK as a target molecule for the development of
`new cancer therapies.
`EGFR-TK Inhibitors. Different approaches to inhibit-
`ing EGFR have resulted in a number of EGFR-targeted agents in
`clinical development including small-molecule EGFR-TK in-
`hibitors, mAbs, vaccines, immunotoxins, and recombinant li-
`gand–toxin fusion proteins (1, 44).
`Small-molecule EGFR-TK inhibitors act by blocking the
`ATP binding site of the EGFR-TK enzyme inside tumor cells
`(Fig. 4). On the basis of this mechanism of action, EGFR-TK
`
`inhibitors have the potential to inhibit all mechanisms of
`EGFR-TK activation, including constitutively activating mu-
`tations and receptor cross-talk. EGFR-TK inhibitors were
`designed to selectively inhibit EGFR-TK relative to other
`kinase enzymes present
`in normal
`tissues (28). Gefitinib
`erlotinib and CI-1033 (Pfizer) are among the EGFR-TK
`inhibitors in clinical development (Table 2; Refs. 1 and
`45– 48). Both gefitinib and erlotinib selectively and revers-
`ibly inhibit EGFR-TK, whereas CI-1033 is an irreversible
`pan-ErbB family inhibitor. The small-molecule EGFR-TK
`inhibitors also inhibit signals induced by EGFR heterodimer-
`ization with other members of the ErbB family. Compared
`with anti-EGFR mAbs such as cetuximab [Erbitux (C225);
`ImClone], EGFR-TK inhibitors offer the advantages of oral
`bioavailability and once-daily treatment.
`The targeted agent cetuximab, [Erbitux (C225); ImClone],
`is a chimeric mAb directed against the extracellular, ligand-
`binding domain of EGFR that competes with ligand for receptor
`binding (1, 49, 50). Cetuximab was not studied as a single agent
`in NSCLC but is currently being evaluated in combination with
`carboplatin/paclitaxel and cisplatin/gemcitabine in untreated pa-
`tients with stage IV NSCLC, and with docetaxel in patients with
`chemotherapy-refractory tumors. ABX-EGF (Abgenix) is an-
`other anti-EGFR mAb in Phase I clinical trials. mAbs can also
`be coupled with various toxic agents such as bacterial toxins or
`
`

`

`Clinical Cancer Research
`
`5817
`
`Pharmacodynamic studies indicate that EGFR-TK inhibi-
`tors and anti-EGFR antibodies block cell cycle progression in
`the G1 phase by up-regulating p27Kip1, a cell cycle inhibitor, and
`down-regulating c-fos, a transcriptional activator that is promi-
`nent in EGFR-mediated signaling (45, 52–55). Elevated levels
`of p27Kip1 block cell cycle progression in the G1 phase of
`growth. This sustains the hypophosphorylated state of the reti-
`noblastoma (RB) gene product, which is necessary to keep cells
`from progressing in the cell cycle (37, 56).
`The inhibition of tumor growth seen with EGFR-TK inhi-
`bition is also accompanied by decreases in vascular endothelial
`growth factor (VEGF), basic fibroblast growth factor (bFGF),
`and TGF-␣, all potent inducers of tumor angiogenesis (57).
`Thus, inhibitors of EGFR/EGFR-TK may also inhibit tumor
`growth by interfering with angiogenesis (58, 59). These obser-
`vations suggest that by inhibiting EGFR-TK, gefitinib and er-
`lotinib treatment alters expression levels of key molecules in
`tumor cells that are important for stimulating proliferation, cell
`cycle progression, tumor angiogenesis, metastasis, and inhibi-
`tion of apoptosis.
`
`Clinical Trials of EGFR-TK Inhibitors in NSCLC
`Phase I Trials. Several anti-EGFR agents have been tested
`alone or in combination with other agents in Phase I trials that
`included patients with NSCLC. Phase I trials of gefitinib followed
`two escalating dose schedules: (a) once-daily gefitinib given con-
`tinuously for 28 days; or (b) intermittent gefitinib, with 14 days on
`and 14 days off treatment (60–62). In the intermittent-dosing trials,
`doses ranged from 50 to 925 mg/day. In the continuous-dosing
`trials, doses ranged from 150 to 1000 mg/day. Tumor EGFR status
`
`Agent
`ZD1839
`gefitinib)
`
`OSI-774
`(erlotinib)
`
`CI-1033 (none)
`
`Clinical status
`Phase III
`(Approved)
`
`Phase III
`
`Phase I
`
`Table 2 EGFR-targeted agents in clinical developmenta
`Mechanism of action in vitro
`IC50 HER1 (KBb,c; CGd)
`HER2 (KBc; CGd)
`EGFR-TK inhibitor
`HER1 (23–79 nM; ⱕ80 nM)
`HER2 (3.7–10 ␮M; N/A)
`EGFR-TK inhibitor
`HER1 (2–20 nM; ⱕ100 nM)
`HER2 (0.2 ␮M; ⬍3 ␮M)
`EGFR-TK/HER2 inhibitor
`HER1 (1.7 nM; 7.4 nM)
`HER2 (5 nM; N/A)
`Anti-EGFR antibody
`
`Phase II/III
`
`Anti-EGFR antibody
`
`Phase I/II
`
`Vaccine
`
`Immunotoxin
`
`Phase II/III
`
`Phase II
`
`C225
`(cetuximab)
`ABX-EGF
`(none)
`EGF-P64K
`(none)
`DAB389-EGF
`(none)
`a See Refs. 45–48.
`b KB, kinase/binding inhibition; CG, cell growth inhibition; N/A,
`not available.
`c KB varies based on the source of purified HER1/2.
`d CG varies based on cell line tested.
`
`Fig. 4 The mechanism of action of EGFR-TK inhibitors in blocking
`signal transduction through EGFR-TK.
`
`radioactive particles, which may be used as delivery devices.
`The binding of the mAbs to the extracellular domain of EGFR
`triggers endocytosis of the receptor-immunotoxin complex to
`the cytoplasm, in which the various toxins act to inhibit protein
`synthesis and induce apoptosis (51). In another approach for
`targeting toxins to EGFR-expressing tumor cells, chimeric mol-
`ecules are created by fusing portions of the genes for ligands
`(EGF, TGF-␣) with a toxin gene. One example of such a
`toxin-fusion protein, DAB389-EGF, is in Phase II clinical trials
`for NSCLC (24).
`Both EGFR-TK inhibitors and anti-EGFR antibodies are
`effective in preclinical models for inhibiting the growth of a
`variety of human tumor cell lines, including lung, colorectal,
`breast, and prostate, suggesting their potential for broad appli-
`cability for solid tumor types (1). Preclinical studies also
`showed that EGFR inhibition results in synergy with chemo-
`therapy agents or radiation therapy in cell lines that are sensitive
`to EGFR inhibitors (52). For example, in cell viability assays,
`gefitinib treatment was synergistic with the cytotoxic chemo-
`therapy agents, vinorelbine and paclitaxel, and had additive
`effects with cisplatin (52). Similarly, gefitinib in combination
`with radiation has shown growth-inhibitory effects ranging from
`synergistic to additive in gefitinib-sensitive cell lines (52). Lung
`tumor xenografts have also been inhibited by gefitinib alone or
`in combination with chemotherapy agents (53). Gefitinib, erlo-
`tinib, and cetuximab have all been shown to potentiate the
`antitumor effects of most cytotoxic agents, including platinum-
`based chemotherapy agents in preclinical models with cell lines
`sensitive to EGFR inhibition. Gefitinib also showed activity
`against NSCLC xenografts in combination with taxanes, doxo-
`rubicin, and antifolates (45, 52–54).
`
`

`

`5818 Targeting EGFR in NSCLC
`
`was not an eligibility requirement in these trials. Patients were
`selected based on having tumor types that were all known to
`express EGFR at a high rate, including NSCLC and colorectal,
`prostate, head and neck, ovarian, breast, renal, and pancreatic
`cancers.
`In total, 252 patients were recruited for Phase I trials of
`gefitinib (60–62). Almost all of the patients had received prior
`treatment with radiotherapy and/or chemotherapy, and many
`had received multiple prior chemotherapy regimens. Of the
`patients enrolled, 100 had advanced, previously treated NSCLC.
`The most common adverse events were diarrhea, acneiform
`rash, nausea, asthenia, and vomiting. The majority of adverse
`events were grade 1 or 2 and transient; grades 3 and 4 events
`were rare (62). Dose-limiting toxicities including reversible
`diarrhea and rash occurred at daily doses of 700 to 800 mg.
`Pharmacokinetics were consistent with once-daily dosing
`(60, 62).
`Histopathological studies of pretreatment and posttreat-
`ment skin biopsy specimens from Phase I trials showed that
`treatment with gefitinib suppressed EGFR phosphorylation, in-
`hibited MAPK activity, reduced the proliferation index as
`judged by staining for Ki67 (a nuclear proliferation-associated
`antigen), and increased both the apoptotic index and the expres-
`sion of p27Kip1 (55). The stratum corneum of the epidermis was
`significantly thinner in posttreatment skin samples (55). In a
`separate study, tumor biopsy samples obtained after 28 days of
`treatment with gefitinib showed decreased levels of activated
`signal
`transduction molecules compared with biopsies from
`baseline samples (63).
`In the Phase I trials of gefitinib in 100 NSCLC patients,
`partial responses were observed in 10% of patients, and disease
`stabilization was seen in 13% of patients (61). There were no
`obvious differences between the continuous- and intermittent-
`dose schedules with respect to response or toxicity. Antitumor
`activity occurred at all dose levels, with no clear dose-response
`relationship. Stable disease was observed at daily doses as low
`as 50 mg (n ⫽ 1), and partial responses were achieved at 150 mg
`(n ⫽ 2; Ref. 61). In many cases, there was anecdotal evidence
`of symptom amelioration with gefitinib treatment in the absence
`of an objective tumor response. Of the 100 patients with
`NSCLC, 28% remained on gefitinib for at least 3 months and
`20% for at least 6 months.
`Erlotinib was investigated in a Phase I trial of 40 patients
`with previously treated advanced solid tumors,
`including 4
`patients with NSCLC (64). Dose levels included 25, 50, 100,
`and 200 mg/day. Expression of EGFR in the tumor was not an
`eligibility requirement in this trial. However, EGFR expression
`was assessed immunohistochemically to determine whether pa-
`tients with EGFR-positive tumors were more likely to benefit
`from treatment with erlotinib (64). Tumor response did not show
`a correlation with EGFR expression (64). The most common
`adverse events were diarrhea and skin toxicities. These adverse
`events established the maximum tolerated dose at 150 mg/day.
`Most adverse events were grade 1 or 2 and were reversible. In
`this trial, there was one complete response (renal cell carci-
`noma), one partial response (colorectal), and several patients
`with stable disease (one of four with NSCLC).
`The mAb cetuximab, which is administered i.v., was in-
`vestigated in Phase I trials involving weekly administration of
`
`cetuximab, alone or in combination with cisplatin (65). Patients
`recruited for these studies had advanced solid tumors that over-
`expressed EGFR as documented through immunohistochemistry
`on tumor biopsies. The multiple-dose monotherapy study was
`conducted in 17 patients who had previously received chemo-
`therapy according to the standard of care for their particular
`tumor type. Another trial evaluated cetuximab in combination
`with chemotherapy in 22 patients with head and neck cancer or
`patients with NSCLC who had not been previously treated with
`a platinum agent. The dose levels of cetuximab investigated
`were 5, 20, 50, and 100 mg/m2 (65).
`The most common adverse events in the Phase I trials of
`cetuximab were fever and chills; asthenia; transaminase eleva-
`tion; nausea; and skin toxicities, including flushing, seborrhea,
`and acneiform rashes. The majority of adverse events were
`grade 1 or 2. The maximum tolerated dose was not reached in
`these trials. Because anti-EGFR mAbs such as cetuximab are
`administered i.v., gastrointestinal
`toxicities are not as pro-
`nounced as with the orally administered small-molecule
`EGFR-TK inhibitors. However, the mAb-based treatments oc-
`casionally induce immunological responses. Of the 189 patients
`treated with cetuximab in early-phase trials, 2% experienced
`grade 3 allergic reactions and 2% experienced grade 4 reactions.
`Allergic reactions to cetuximab were managed with standard
`interventions (66). Several patients in these trials experienced
`disease stabilization, particularly those with head and neck
`cancer (65).
`Randomized Phase II Trials in Advanced Refractory
`NSCLC. There were two large randomized Phase II trials that
`evaluated two doses of gefitinib in advanced, chemotherapy-
`refractory NSCLC patients. These trials were termed IDEAL-1
`and IDEAL-2 (3, 4). IDEAL-1 was a global
`trial with an
`enrollment of 210 patients with NSCLC who had failed one or
`more previous chemotherapy regimens (3). IDEAL-2 was a
`United States trial with an enrollment of 216 patients with
`NSCLC who failed two or more previous chemotherapy regi-
`mens that included a platinum agent and docetaxel (4). In both
`trials, patients were randomized to receive treatment with ge-
`fitinib at 250 mg/day or 500 mg/day. Unlike cytotoxic agents,
`for which the dose is dictated by toxicities, these once-daily oral
`doses of gefitinib were selected for study based on optimal
`biological doses that are well below the maximum tolerated
`dose (62).
`Objective tumor response (ⱖ50% inhibition of tumor
`mass), which was evaluated every 4 weeks by radiographic
`assessment, was a primary end point of both IDEAL trials. A
`co-primary end point in IDEAL-1 was safety, whereas symptom
`improvement was a co-primary end point in IDEAL-2. The LCS
`of the FACT-L questionnaire was used to assess symptom
`improvement. Because these novel targeted therapies are largely
`devoid of systemic toxicities, improvement in disease-related
`symptoms is an important basis for assessing their utility.
`The results of IDEAL-1 and IDEAL-2 are summarized in
`Table 3 (3, 4). In IDEAL-1, the following data were obtained for
`the 250 mg/day and 500 mg/day groups, respectively: objective
`tumor response rate, 18.4% versus 19%; disease control rate,
`54.4% versus 51.4%; progression-free survival, 2.7 months versus
`2.8 months; median survival, 7.6 months versus 7.9 months. Ob-
`jective tumor response rates were similar for patients who received
`
`

`

`Clinical Cancer Research
`
`5819
`
`Table 3 Results from Phase II studies of patients with NSCLC treated with gefitiniba
`IDEAL-1
`IDEAL-2
`
`Trial end point
`Objective tumor response, %
`Symptom improvement, %
`Disease control, %
`Overall survival, mo
`a See Refs. 3, 4.
`
`250 mg/day
`(n ⫽ 103)
`18.4
`40.3
`54.4
`7.6
`
`500 mg/day
`(n ⫽ 105)
`19.0
`37
`51.4
`7.9
`
`250 mg/day
`(n ⫽ 102)
`11.8
`43.1
`43.0
`6.1
`
`500 mg/day
`(n ⫽ 114)
`8.8
`35.1
`35.0
`6.0
`
`gefitinib as second-line versus third-line treatment (17.9% versus
`19.8%; Ref. 3). Adenocarcinoma, which characteristically ex-
`presses less EGFR than does squamous cell carcinoma, was iden-
`tified as one of the prognostic factors associated with objective
`response (3.5 times more likely to respond to treatment than other
`tumor histologies; Ref. 3). The overall symptom improvement rate,
`as measured by the LCS, was 40 and 37%, respectively, and
`improvement occurred rapidly with a median time to improvement
`of 8 days (3). Among responders, 78% exhibited symptom im-
`provement based on the LCS and 53% reported a quality-of-life
`improvement as measured by FACT-L (67).
`The most frequent adverse events were generally

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket