ARTICLE

Akt Phosphorylation and Gefitinib Efficacy in Patients With Advanced Non–Small-Cell Lung Cancer

Federico Cappuzzo, Elisabetta Magrini, Giovanni Luca Ceresoli, Stefania Bartolini, Elisa Rossi, Vienna Ludovini, Vanesa Gregorc, Claudia Ligorio, Alessandra Cancellieri, Stefania Damiani, Anna Spreafico, Carlo Terenzio Paties, Laura Lombardo, Cesare Calandri, Guido Bellezza, Maurizio Tonato, Lucio Crinò

Affiliations of authors: Department of Oncologic Sciences, Bellaria-Maggiore Hospital, Bologna, Italy (FC, EM, SB, CL, AC, SD, LL, CC, LC); Division of Radiochemotherapy, Scientific Institute University Hospital San Raffaele-Milano, Italy (GLC, VG, AS, CTP); CINECA—Interuniversity Consortium, Bologna, Italy (ER); Policlinico Monteluce, Division of Medical Oncology, Perugia, Italy (VL, GB, MT)

Correspondence to: Federico Cappuzzo, MD, Bellaria Hospital, Division of Medical Oncology, Via Altura 3, 40139-Bologna, Italy (e-mail: federico.cappuzzo{at}ausl.bo.it)


    ABSTRACT
 Top
 Notes
 Abstract
 Introduction
 Patients and Methods
 Results
 Discussion
 References
 
Background: Gefitinib, a specific epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, has activity against approximately 10% of unselected non–small-cell lung cancer (NSCLC) patients. Phosphatidylinositol 3'-kinase (PI3K)/Akt and Ras/Raf/mitogen-activated protein kinase (MAPK), the two main EGFR-signaling pathways, mediate EGFR effects on proliferation and survival. Because activation of these pathways is dependent on the phosphorylation status of the components, we evaluated the association between phosphorylation status of Akt (P-Akt) and MAPK (P-MAPK) and gefitinib activity in patients with advanced NSCLC. Methods: Consecutive patients (n = 106) with NSCLC who had progressed or relapsed on standard therapy received gefitinib (250 mg/day) until disease progression, unacceptable toxicity, or patient refusal. P-Akt and P-MAPK positivity was determined with immunohistochemistry using tumor tissues obtained before any anticancer treatment. Association of P-Akt and time to progression was determined by univariable and multivariable analyses. All statistical tests were two-sided. Results: Of the 103 evaluable patients, 51 (49.5%) had tumors that were positive for P-Akt, and 23 (22.3%) had tumors that were positive for P-MAPK. P-Akt–positivity status was statistically significantly associated with being female (P<.001), with never-smoking history (P = .004), and with bronchioloalveolar carcinoma histology (P = .034). Compared with patients whose tumors were negative for P-Akt, patients whose tumors were positive for P-Akt had a better response rate (26.1% versus 3.9%; P = .003), disease control rate (60.9% versus 23.5%; P<.001), and time to progression (5.5 versus 2.8 months; P = .004). Response rate, disease control rate, and time to progression did not differ according to P-MAPK status. The multivariable analysis showed that P-Akt positivity was associated with a reduced risk of disease progression (hazard ratio = 0.58, 95% confidence interval = 0.35 to 0.94). Conclusions: Patients with P-Akt–positive tumors who received gefitinib had a better response rate, disease control rate, and time to progression than patients with P-Akt–negative tumors, suggesting that gefitinib may be most effective in patients with basal Akt activation.



    INTRODUCTION
 Top
 Notes
 Abstract
 Introduction
 Patients and Methods
 Results
 Discussion
 References
 
Lung cancer is the leading cause of cancer death among men and women in Europe and North America (1). Despite aggressive surgical and chemotherapeutic intervention, the prognosis of patients with non–small-cell lung cancer (NSCLC) remains poor, with an overall cure rate of less than 15% (2). Given the rapid advances in the molecular and biological understanding of the tumorigenic process, several new strategies, including those that involve specific molecular targets, have been developed for the treatment of various cancers.

One potential family of therapeutic targets is the epidermal growth factor receptor (EGFR) superfamily. These cell membrane receptors have been implicated in the development and progression of cancer through their effects on cell cycle progression, apoptosis, angiogenesis, and metastasis (36). The EGFR superfamily includes four distinct receptors: EGFR/ErbB-1, HER2/ErbB-2, HER3/ErbB-3, and HER4/ErbB-4. Although specific soluble ligands that bind to the extracellular domains of EGFR, HER3, and HER4 have been identified, no ligand has been identified for the HER2 receptor. Several ligands can bind to EGFR, including EGF and transforming growth factor {alpha} (TGF-{alpha}). After the ligand binds the receptor, the receptor dimerizes—either as a homodimer or as a heterodimer with other members of the EGFR family, but preferentially with HER2—and undergoes autophosphorylation at specific tyrosine residues within the intracellular domain. These autophosphorylation events in turn activate downstream signaling pathways, including the Ras/Raf/mitogen-activated protein kinase (MAPK) pathway and the phosphatidylinositol 3'-kinase (PI3K)–Akt pathway. Activation of Ras initiates a multistep phosphorylation cascade that leads to the activation of MAPKs. The MAPKs ERK1 and ERK2 subsequently regulate gene transcription and have been linked to cell proliferation, survival, and transformation in laboratory studies (7). Akt also plays a critical role in controlling the balance between cell survival and apoptosis (8). Phosphorylation of Akt is required for its activation; once activated, Akt inactivates proapoptotic proteins, including the Bcl-2 family member Bad and caspase-9, and cell cycle–regulatory molecules (910). Akt function is also important for cell survival when cells are exposed to different apoptotic stimuli, such as growth factor withdrawal, ultraviolet radiation, and DNA damage (1115). Given its association with cell survival, it is not surprising that Akt has been found to be overexpressed in several cancers, including lung (15), pancreatic (16), thyroid (17), and ovarian (1819) cancers. Recently, Akt phosphorylation has been observed in more than 60% of patients with NSCLC (20).

Drugs interfering with EGFR pathways are thought to be of potential therapeutic benefit in tumors expressing or overexpressing EGFR, and during the last decade, several molecules have been synthesized to inhibit the tyrosine kinase domain of EGFR (21,22). Among the more promising new drugs is ZD-1839 (gefitinib or Iressa; AstraZeneca, London, U.K.), an orally active, selective EGFR tyrosine kinase inhibitor with antitumor activity against a variety of human cancer cell lines expressing EGFR (23). After objective responses were observed in 11% of study participants in phase I trails (2427), two large phase II trials (IDEAL 1 and 2) (28,29) were conducted. The results of the IDEAL trials confirmed that gefitinib is active in approximately 10% of patients with NSCLC in whom standard therapy failed (28,29). Although these results were considered encouraging because they were obtained from patients in whom standard therapies were largely ineffective, only a small percentage of patients responded to single-agent gefitinib. Preclinical data have suggested that EGFR expression is not the critical factor in determining whether a patient will respond to gefitinib (30), although tumors that overexpress HER2 are highly sensitive to gefitinib (31). In our previous trial (32), conducted on 63 patients with NSCLC who gave their consent for EGFR and HER2 analysis, we examined the association between efficacy and tolerability of gefitinib and EGFR and HER2 expression. In that small study (32), in which HER2 status was determined with immunohistochemistry, there was no association between HER2 expression and gefitinib activity.

Although the degree of EGFR expression does not affect response to gefitinib, clinical responses to EGFR-blocking agents suggest that, in some patients with NSCLC, the EGFR signaling pathway may be the critical pathway for tumor growth. It is possible that patients who respond to EGFR-blocking agents such as gefitinib are those patients in whom tumor cell survival is maintained by activated EGFR-dependent pathways. To test this hypothesis and to determine if it is possible to identify which patients may respond to gefitinib therapy, we evaluated the association between the phosphorylation status of MAPK and Akt in patients with advanced NSCLC before starting gefitinib therapy and evaluated their response to gefitinib therapy. This trial was designed to evaluate possible associations between the phosphorylation status of Akt and of MAPK and gefitinib activity in terms of response rate and time to progression.


    PATIENTS AND METHODS
 Top
 Notes
 Abstract
 Introduction
 Patients and Methods
 Results
 Discussion
 References
 
Patients

Study enrollment began in February 2001, and the last patients were enrolled in August 2003. In the period, February 2001–June 2002, because of the ongoing HER2 study (32), patients were asked to give their consent for inclusion in the HER2 study (N = 55), the Akt/MAPK study (N = 22) (the results of which are described in this article), or both studies (N = 8).

Patients included in the present study had histologically confirmed, measurable, locally advanced or metastatic NSCLC and had progressed or relapsed after standard therapy. Patients who had not received any previous chemotherapy were included in the study if they were considered not eligible for systemic therapy because of age, poor performance status, or any medical condition contraindicating chemotherapy. To be eligible for the study, patients had to be aged older than 18 years, have an ECOG performance status of 2 or less, have a white blood cell count of at least 3.5 x 109/L, have a platelet count of at least 100 x 109/L, have a hemoglobin level of at least 9 g/dL, have an absolute granulocyte count of more than 2.0 x 109/L, have a bilirubin level that was less than 1.5-fold the upper limit of normal, have a prothrombin time or activated partial thromboplastin time of less than 1.5 times that of control, have an alanine aminotransferase (ALT) or aspartate aminotransferase (AST) levels of less than threefold the upper limits of normal (this level was increased to fivefold for patients with known hepatic metastases), and have a calculated creatinine clearance rate of more than 45 mL/min. Patients were ineligible for the study if they had evidence of prior or concurrent malignancy, with the exception of in situ carcinoma of the cervix, adequately treated basal cell carcinoma of the skin, or no evidence of recurrence of a malignancy treated 5 or more years ago.

Before patients were included in the trial, all patients were asked about their smoking history and classified as nonsmoker (never smoker), former smoker (stopped smoking more than 6 months before enrollment onto the trial), or current smoker (stopped smoking less than 6 months before enrollment onto the trial or still an active smoker). Lung cancer histology was defined according to the World Health Organization pathology classification (33). Written informed consent was obtained from each patient entering the study. The study was approved by the appropriate ethics review boards and followed the recommendations of the Declaration of Helsinki for biomedical research involving human subjects and the guidelines for good clinical practice.

Study Design and Treatment

In this study, 106 consecutive patients with NSCLC who progressed or relapsed after standard therapy failed received gefitinib daily at a dose of 250 mg. Patients received the drug until their disease progressed (n = 77), they experienced unacceptable toxicity (n = 1), or they refused to comply further (n = 1). The drug was provided by AstraZeneca on a compassionate-use basis.

Before the time of entry on the trial, a baseline evaluation was performed for each patient that included a complete history and physical examination, a complete blood cell count and serum chemistry analysis, urinalysis, an electrocardiogram, chest x-ray, and a total-body computed tomography (CT) scan. Other imaging modalities, such as magnetic resonance imaging and bone scintigraphy, were performed according to specific clinical indications. All baseline imaging procedures were performed within 4 weeks before study entry. Biochemical screening, which was performed every 4 weeks, included assessments of serum levels of creatinine, electrolytes, alkaline phosphatase, bilirubin, AST, ALT, calcium, magnesium, and total protein. Patients were evaluated for response according to the RECIST (response evaluation criteria in solid tumors) criteria (36). Tumor response was assessed by CT scan every 2 months, with a confirmatory evaluation that was repeated in patients who had not progressed to therapy at least 4 weeks after the initial determination of response.

Immunohistochemical Staining

Tumor specimens obtained at the time of primary diagnosis or at the time of study entry were collected for P-Akt and P-MAPK immunohistochemistry. Paraffin-embedded tissue sections were stained with antibodies against phospho-Akt (P-Akt; Cell Signaling Technology, Beverly, MA) and phospho-MAPK (P-MAPK; Cell Signaling Technology), according to the manufacturer's recommended protocol. Briefly, 4-µm–thick tissue sections were placed on glass slides and deparaffinized. The tissue sections were incubated in 1x Microstain Unmasker Buffer (pH 8) (Ventana, Tucson, AZ) for 40 minutes at 98 °C to unmask the antigens. The reaction was quenched with 1% hydrogen peroxide. Nonspecific binding sites were blocked with 5% goat serum in phosphate-buffered saline (PBS) for 1 hour at room temperature. The sections were then incubated with P-Akt (Ser 473) rabbit polyclonal antibody (1 : 50 diluted in phosphate buffer) or P-p44/42 Map Kinase (Thr202/Tyr204) polyclonal antibody (1 : 100 diluted in phosphate buffer) overnight at 4 °C. The sections were developed by using a two-step method involving a large-volume biotinylated goat polyvalent antibody and a large-volume streptavidin peroxidase reagent (Lab Vision, Fremont, CA). The negative controls were incubated with nonimmune solution in place of primary antibody.

Immunohistochemically stained sections were interpreted independently by two pathologists who were blinded to all patient information. The intensity of staining seen in different areas of the same slide was analyzed according to criteria described previously for p53 (34). Immunohistochemical analyses were centralized, and all were performed at the Bellaria Hospital Pathology Department. Immunohistochemical results were blinded, and the referring physician was informed of the results only at the end of the trial or when the patient was withdrawn from the study.

At present, there are no validated scoring systems for interpreting immunohistochemical staining for P-MAPK or P-Akt. We used a system for interpreting P-MAPK staining that was based on staining intensity. If none of the tumor cells stained, the intensity was coded as 0; if more than 10% of the tumor cells stained weakly, the intensity was coded as 1+; if more than 10% of the tumor cells stained moderately, the intensity was coded as 2+; and if more than 10% of the tumor cells stained strongly, the intensity was coded as 3+. Specimens having a score of 0 or 1+ were considered negative, whereas a score of 2+ or 3+ was considered positive. We used a system for interpreting P-Akt staining that was based on subcellular staining localization. Activation of Akt by phosphorylation results in its translocation from the cytoplasm to the nucleus (35). Thus, staining was considered positive if nuclear staining was present and negative if nuclear staining was absent.

Statistical Analysis

Intent-to-treat analyses were performed on data from all patients who entered the study and received treatment. The trial was designed to detect a difference in response rate between the patients whose tumors were P-Akt positive and patients whose tumors were P-Akt negative. A sample size of 94 patients, with 47 patients in each group, was considered adequate to detect a difference of 25% (5%–30%) between the two groups, with a two-sided significance level ({alpha}) of 5% and a power of 90%. Sample size was calculated using SPSS version 11.5.1 (SPSS Italia srl, Bologna, Italy).

Statistical analyses were performed with respect to clinical characteristics and response to therapy. For both proteins, differences between the two groups (positive and negative) were compared by Fisher's exact test or chi-square test for nominal variables and the Mann–Whitney test or Student's t test for continuous variables. Normality of the distribution of continuous variables was assessed with the Kolmogorov–Smirnov test. Time to progression, overall survival, and 95% confidence intervals (CIs) were evaluated with the Kaplan–Meier method (37), and differences between the two groups were evaluated with the log-rank test. Risk factors associated with time to progression were evaluated using Cox's proportional hazards regression model with a step-down procedure (38). Proportional hazard assumptions were checked and satisfied. Only those variables with statistically significant results in univariate analysis were included in the multivariable analysis. The criterion for removing a variable was the likelihood ratio statistic, which was based on the maximum partial likelihood estimate (default P value of .10 for removal from the model). Data analysis was planned for 3 months after the enrollment of the last patient. This time was selected because standard therapies had failed in our cohort, and the patients therefore had a poor prognosis. The degree of agreement between the two pathologists involved in the study (EM and SD) was assessed by a weighted {kappa} statistic (39). All statistical analyses were performed using SPSS version 11.5.1 (SPSS Italia srl).


    RESULTS
 Top
 Notes
 Abstract
 Introduction
 Patients and Methods
 Results
 Discussion
 References
 
Patient Characteristics

From February 2001 through August 2003, 106 consecutive patients from three Italian institutions fulfilled the selection criteria and received gefitinib daily at a dose of 250 mg. The majority of patients (76 patients) were enrolled after June 2002. Tumor tissue specimens were not suitable for immunohistochemistry from four patients: sections from two patients could not be stained for P-Akt and P-MAPK, sections from one patient could not be stained for P-Akt, and sections from one patient could not be stained for P-MAPK. Thus, 103 patients contributed immunohistochemistry results for either P-Akt or P-MAPK. Patient characteristics are listed in Table 1. The majority of the patients were male (63.1%), with a median age of 63 years (range = 25–83 years) and with a good ECOG (Eastern Cooperative Oncology Group) performance status (86.4% had a performance status of 0 or 1). Among the patient cohort, 55.3% had adenocarcinoma, 12.6% had bronchioloalveolar carcinoma, 20.4% had squamous-cell carcinoma, 9.8% had undifferentiated carcinoma, and 1.9% had large-cell carcinoma. Seven patients (6.8%) received gefitinib as first-line therapy: One patient was aged older than 80 years, and six patients had comorbidities contraindicating chemotherapy. The remaining 96 patients had all received chemotherapy before gefitinib, and of these patients, 74.7% had received chemotherapy that included a platinum salt. At the time of study entry, the majority of patients were current (50.5%) or former (31.1%) smokers.


View this table:
[in this window]
[in a new window]
 
Table 1. Patient characteristics and association with the phosphorylation status of Akt and MAPK determined by immunohistochemistry in evaluable patients*

 
Immunohistochemistry was performed on primary lung tumor specimens obtained before any anticancer therapy. Tumor tissue specimens were obtained at the time of surgery for 26 patients, at the time of original diagnosis of advanced disease for 73 patients, and immediately before starting gefitinib therapy for seven patients. Among the 103 patients evaluated for P-Akt and P-MAPK status, 52 (50.5%) had tumors that were negative for P-Akt and 51 (49.5%) had tumors that were positive for P-Akt, 80 (77.7%) had tumors that were negative (0–1+) for P-MAPK, 14 (13.6%) had tumors that were moderately positive (2+) for P-MAPK, and nine (8.7%) had tumors that were strongly positive (3+) for P-MAPK. The agreement between the two pathologists who reviewed all immunohistochemical staining was very strong, with a weighted {kappa} statistic of 0.973 for P-Akt (P<.001) and 0.966 for P-MAPK (P<.001).

We next evaluated the association between P-Akt and P-MAPK status and clinical variables. P-Akt status was statistically significantly associated with being female (P<.001), never-smoking history (P = .004), and a bronchioloalveolar carcinoma histology (P = .034). No association was found between P-Akt status and other tumor histologies. P-MAPK status was not statistically significantly related to any clinical variable.

Response to Therapy

One hundred patients were evaluable for response. Six patients were considered not evaluable for response because of the absence of any measurable lesion (one patient) or because the patient was lost to follow-up after the baseline visit (five patients). Gefitinib was administered orally, with a median treatment duration of 3.6 months (range = 0.6–23.5 months). One (1%) patient had a complete response, 13 (13%) patients had a partial response, and 26 (26%) patients had stable disease, for an overall response rate of 14% (95% CI = 7.9% to 22.4%). The overall response rate, including complete responses and partial responses, was 26.1% for patients with P-Akt–positive tumors and 3.9% for patients with P-Akt–negative tumors (mean difference = 22.2%, 95% CI = 14.0 to 30.4; P = .003). The disease control rate, which includes complete responses, partial responses, and stable disease, was 60.9% for patients with P-Akt–positive tumors and 23.5% for patients with P-Akt–negative tumors (mean difference = 37.4%, 95% CI = 27.9 to 46.9; P<.001). There was no difference in terms of response rate and disease control rate among patients with P-MAPK–negative tumors and patients with P-MAPK–positive tumors (response rates = 12% versus 22.7%, respectively; P = .298; disease control rate = 38.7% versus 50%, respectively; P = .461) (Table 2). Eighteen patients had tumors that were positive for both P-Akt and P-MAPK. In this subgroup of patients, the response rate was 29.4% and disease control rate was 58.8%, values that were not statistically significantly different from those observed for patients with tumors positive for P-Akt and negative for P-MAPK (response rate = 24.1%, disease control rate = 62.1%) but statistically significantly better than those observed for patients whose tumors were negative for both P-Akt and P-MAPK (response rate = 4.4%; P = .014; and disease control rate = 24.4%; P = .011) (Table 3). No comparisons were made with six patients whose tumors were negative for P-Akt but positive for P-MAPK because of the small sample size.


View this table:
[in this window]
[in a new window]
 
Table 2. Response to gefitinib therapy in patients with advanced non–small-cell lung cancer according to the phosphorylation status of Akt and MAPK

 

View this table:
[in this window]
[in a new window]
 
Table 3. Association between response to gefitinib therapy and phosphorylation status of Akt and MAPK in patients with advanced non–small-cell lung cancer

 
Time to Progression and Survival Analysis

Final analysis was performed in November 2003, after more than 3 months since the enrollment of the last patient. With a median follow-up time of 6.6 months (range = 0.6–24.1 months), the median time to progression for the entire cohort was 3.4 months (range = 0.6–24.1 months), and median overall survival time was 9.4 months (range = 0.6–24.1 months). At the time of the final analysis, 33 patients with P-Akt–positive tumors and 44 patients with P-Akt–negative tumors had radiologically confirmed disease progression, whereas 27 patients with P-Akt–positive tumors and 41 patients with P-Akt–negative tumors had symptomatic progression (onset of new symptoms or worsening of existing disease-related symptoms). Median time to progression was statistically significantly longer for patients with P-Akt–positive tumors than for patients with P-Akt–negative tumors (5.5 versus 2.8 months, mean difference = 2.7 months, 95% CI = 1.5 to 3.9 months; P = .004) (Fig. 1). Time to progression did not differ on the basis of P-MAPK status (3.2 months for patients with P-MAPK–negative tumors versus 4.8 months for patients with P-MAPK–negative tumors; P = .29). Median time to progression was 6.2 months for patients with tumors that were positive for both P-Akt and P-MAPK, which was not statistically significantly different from the median time to progression of patients with tumors that were negative for P-Akt and P-MAPK (2.8 months; P = .05) or from that of patients with tumors that were positive for P-Akt and negative for P-MAPK (0/1+) (5.3 months; P = .63).



View larger version (19K):
[in this window]
[in a new window]
 
Fig. 1. Time to progression in patients with advanced non–small-cell lung cancer who received gefitinib therapy according to the phosphorylation status of Akt. Phosphorylation status was determined on the basis of immunohistochemistry on tumor sections stained before the patients received gefitinib therapy. Median time to progression was 5.5 months (95% CI = 3.8 to 7.2 months) for patients with P-Akt–positive tumors and 2.8 months (95% CI = 2.3 to 3.3 months) for patients with P-Akt–negative tumors. Differences between the two groups were evaluated with the log-rank test.

 
To define which variables were predictive of improved time to progression, we evaluated prespecified prognostic factors at study entry, such as age, sex (male versus female), performance status (0/1 versus 2), disease stage (III versus IV), smoking history (current and former smoker versus never smoker), histology (adenocarcinoma including bronchioloalveolar versus other histotypes), P-MAPK status (P-MAPK 0/1+ versus P-MAPK 2+/3+), P-Akt status (P-Akt positive versus P-Akt negative) in a univariate analysis, and those variables associated with statistically significant relationships (sex, performance status, smoking history, and P-Akt status) were included in the multivariable model. P-Akt status, smoking history, and performance status were statistically significantly associated with risk of progression. Patients with P-Akt–positive tumors had a statistically significantly lower risk of disease progression than patients with P-Akt–negative tumors (hazard ratio [HR] = 0.58, 95% CI = 0.35 to 0.94), whereas patients with a performance status of 2 or patients who smoked (current or former) had a statistically significantly higher risk of disease progression (HR = 2.65, 95% CI = 1.33 to 5.27; and HR = 1.75, 95% CI = 1.08 to 2.85, respectively). Median survival time was 15.0 months (95% CI = 7.4 to 22.6 months) for patients with P-Akt–positive tumors and 8.3 months (95% CI = 4.8 to 11.8 months) for patients with P-Akt–negative tumors (P = .32) (Fig. 2). Median survival times were similar among patients whose tumors differed on the basis of P-MAPK status (P = .96).



View larger version (19K):
[in this window]
[in a new window]
 
Fig. 2. Survival in patients with advanced non–small-cell lung cancer who received gefitinib therapy according to the phosphorylation status of Akt. Phosphorylation status was determined on the basis of immunohistochemistry on tumor sections stained before the patients received gefitinib therapy. Median survival was 15.0 months (95% CI = 7.4 to 22.6 months) for patients with P-Akt–positive tumors and 8.3 months (95% CI = 4.8 to 11.8 months) for patients with P-Akt–negative tumors. Differences between the two groups were evaluated with the log-rank test.

 

    DISCUSSION
 Top
 Notes
 Abstract
 Introduction
 Patients and Methods
 Results
 Discussion
 References
 
We evaluated the relationship between the efficacy of gefitinib therapy and P-Akt/P-MAPK status in patients with advanced NSCLC, and we found that patients whose tumors showed activation of the PI3K–Akt pathway (i.e., whose tumors were positive for P-Akt) had statistically significantly better response rates, disease control rates, and times to progression. Gefitinib is a synthetic anilinoquinazoline tyrosine kinase inhibitor that is selective for the EGFR. Although it could be hypothesized that EGFR expression is a prerequisite for a response to the drug, EGFR expression has not turned out to be useful in the selection of patients who respond to gefitinib. Indeed, preclinical data suggest that gefitinib sensitivity is not related to EGFR expression (30)—a finding that is supported by the fact that patients with squamous-cell carcinomas had lower response rates than patients with adenocarcinoma, despite their higher levels of EGFR expression. Because EGFR preferentially dimerizes with HER2, and because preclinical data suggested that HER2-overexpressing tumors are more sensitive to gefitinib therapy (31), we previously analyzed the association between HER2 status and gefitinib activity (32). Although in that small study no association was found between gefitinib activity and HER2 expression, the results confirmed that a small subgroup of patients are still particularly sensitive to gefitinib.

On the basis of previous findings, we hypothesized that response to gefitinib would occur only in patients whose tumors were dependent on sustained activation of the EGFR signaling pathways, and only in those in whom these pathways were suppressed by the drug. In this study, we evaluated the two major EGFR signaling pathways (PI3K–Akt and Ras/Raf/MAPK). Only the PI3K–Akt pathway was statistically significantly associated with gefitinib activity. Compared with patients whose tumors were negative for P-Akt, patients whose tumors were positive for P-Akt had a better response rate (26.1% versus 3.9%; P = .003) and disease control rate (60.9% versus 23.5%; P<.001). No difference in response and disease control rate was observed between patients with P-MAPK–negative and P-MAPK–positive tumors. Patients whose tumors were positive for both P-Akt and P-MAPK had better response and disease control rates only in comparison with patients whose tumors were negative for both markers (P = .01) and not in comparison with patients whose tumors were positive for P-Akt but negative for P-MAPK. These findings are consistent with preclinical data suggesting that the PI3K–Akt pathway plays a critical role in the antitumor effects of gefitinib and that inhibition of the MAPK pathway is not sufficient to mediate response to the drug (40).

In two large phase II studies evaluating gefitinib, only being female and having an adenocarcinoma histology were associated with a clinical response (28,29). More recently, Shah et al. (41) also reported that, in a multivariable analysis of 140 patients, those patients who had never smoked cigarettes and those with bronchioloalveolar carcinoma histology were more likely to respond to gefitinib. These findings are in agreement with those of our study, in which P-Akt status was statistically significantly associated with being female (P<.001), never smoking history (P = .004), and bronchioloalveolar carcinoma histology (P = .034). Results from analyses of the time-to-progression data support the central role of Akt activation in gefitinib activity. Compared with patients whose tumors were negative for P-Akt, patients whose tumors were positive for P-Akt had a statistically significantly longer time to progression (5.5 versus 2.8 months; P = .004). We found no difference in the time to progression among patients grouped on the basis of P-MAPK status (P = .29), although we noted slight, albeit not statistically significant, evidence of a longer time to progression among patients whose tumors were positive for both markers compared with patients whose tumors were negative for both markers (P = .05).

To further investigate the difference in time to progression observed among patients whose tumors were positive or negative for P-Akt, we performed a planned multivariable analysis, and only those variables that were statistically significant in the univariate analysis (sex, performance status, smoking history, and P-Akt status) were included in the model. To ensure that only relevant factors were retained in the multivariable model, the backward regression technique was used at the 10% significance level. In the multivariable analysis, P-Akt status was statistically significantly associated with a reduced risk of disease progression (HR = 0.58, 95% CI = 0.35 to 0.94). Importantly, after being adjusted for P-Akt status, performance status and smoking history remained statistically significantly associated with an increased risk of disease progression (HR = 2.65 [95% CI = 1.33 to 5.27] and 1.75 [95% CI = 1.08 to 2.85], respectively), and female sex was immediately removed at the first step of the backward elimination. These data indicate that P-Akt status, performance status, and smoking history are independent factors for disease progression but that being female is not when Akt status is considered. These findings do not represent a variation in respect to the IDEAL trial results because in those studies (28,29), sex was related to response rate and not to time to progression. Moreover, the impact of P-Akt was not evaluated in the multivariable model used in the IDEAL trials.

Although response rate, disease control rate, and time to progression were better for 51 patients whose tumors were positive for P-Akt than for 52 patients whose tumors were negative, two patients among the latter group had major responses and 10 patients had stable disease. This finding may be explained by the fact that the tumor specimens used for the immunohistochemical assays were generally obtained at the time of primary diagnosis and not immediately before starting the trial. Preclinical data showed that NSCLC tumors may become more dependent on the EGFR signaling pathway, and therefore more sensitive to EGFR blocking strategies, as they are exposed to different chemotherapies (42,43). This finding may also explain the negative results of the INTACT trials, in which standard chemotherapy plus gefitinib was compared with standard chemotherapy alone in previously untreated patients with NSCLC (44,45). In our study, only 6.6% of individuals received gefitinib as first-line therapy, and in five of those patients, the PI3K–Akt pathway was not activated (i.e., their tumors were P-Akt negative). Another important finding is that almost 40% of patients with P-Akt–positive tumors did not benefit from gefitinib therapy. Recent data indicate that sensitivity to gefitinib therapy requires intact EGFR-stimulated Akt signaling activity and that loss of PTEN, a phosphatase that negatively regulates Akt by dephosphorylating it, can lead to aberrant Akt activation and, finally, to gefitinib resistance (40,46,47). In this study, PTEN status was not determined, but a retrospective analysis of PTEN status in all patients included in this trial is ongoing.

No difference in survival was found among any group of patients separated on the basis of Akt or MAPK phosphorylation status, but survival was not the primary end point of the study, and at the time of this analysis, median follow-up was too short and the number of censored cases too high to detect any difference. Analysis of survival curves does show a trend that favors patients whose tumors are positive for P-Akt, and it is possible that with longer follow-up a difference in survival outcome could be detected.

Recent reports (48,49) showed that specific missense and deletion mutations in the tyrosine kinase domain of the EGFR gene are statistically significantly associated with gefitinib sensitivity. However, although objective responses were reported in up to 18%, and symptomatic improvement in 40%, of the unselected gefitinib-treated NSCLC patients (28,29), the low frequency of these mutations in unselected U.S. patients (49) suggests that other mechanisms could be involved in the response to gefitinib, such as Akt activation. Further studies should evaluate the association between Akt activation and EGFR gene status.

Finally, this trial showed that Akt is activated in approximately 50% of patients with NSCLC. Our findings are in agreement with the results of Brognard et al. (15), who showed that Akt is constitutively active in many cases of NSCLC, and our findings confirm the recent report by Lee et al. (20), who showed that tumors in patients with NSCLC frequently express phosphorylated Akt. Despite the observation that many patients with NSCLC express activated Akt, the relationship between Akt activation and lung carcinogenesis remains unclear.

In conclusion, our findings suggest that gefitinib therapy is more active in patients with tumors that are positive for P-Akt than in those with tumors that are negative for P-Akt. Further prospective studies are needed to evaluate the role of other associated markers such as PTEN and to assess the impact of previous therapies on Akt signaling pathway.


    NOTES
 Top
 Notes
 Abstract
 Introduction
 Patients and Methods
 Results
 Discussion
 References
 
We thank Katherine Brandt Tonato and Dr. Thomas Chanin for their editorial assistance and Dr. Fred Hirsch for his useful comments on the manuscript.


    REFERENCES
 Top
 Notes
 Abstract
 Introduction
 Patients and Methods
 Results
 Discussion
 References
 

1 Greenlee RT, Hill-Harmon MB, Murray T, Thun M. Cancer statistics, 2001. CA Cancer J Clin 2001;51:15–36.[Abstract/Free Full Text]

2 Jemal A, Thomas A, Murray T, Thun M. Cancer statistics, 2002. CA Cancer J Clin 2002;52:23–47.[Abstract/Free Full Text]

3 Salomon D, Gullick W. The erbB family of receptors and their ligands: multiple targets for therapy. Signal 2001;2:4–11.

4 Ciardello F, Tortora G. A novel approach in the treatment of cancer: targeting the epidermal growth factor receptor. Clin Cancer Res 2001;7:2958–70.[Abstract/Free Full Text]

5 Arteaga CL. Overview of epidermal growth factor receptor biology and its role as a therapeutic target in human neoplasia. Semin Oncol 2002;29(5 Suppl 14):3–9.

6 Bunn PA Jr, Franklin W. Epidermal growth factor receptor expression, signal pathway, and inhibition in non-small cell lung cancer. Semin Oncol 2002;29(5 Suppl 14):38–44.

7 Lewis TS, Shapiro PS, Ahn NG. Signal transduction through MAP kinase cascades. Adv Cancer Res 1998;74:49–139.[ISI][Medline]

8 Franke TF, Kaplan DR, Cantley LC. PI3K: downstream AKTion blocks apoptosis. Cell 1997;88:435–7.[ISI][Medline]

9 Datta SR, Dudek H, Tao X, Masters S, Fu H, Gotoh Y, et al. Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery. Cell 1997;91:231–41.[ISI][Medline]

10 Cardone MH, Roy N, Stennicke HR, Salvesen GS, Franke TF, Stanbridge E, et al. Regulation of cell death protease caspase-9 by phosphorylation. Science 1998;282:1318–21.[Abstract/Free Full Text]

11 Hemmings BA. AKT signaling: linking membrane events to life and death decision. Science 1997;275:628–30.[Free Full Text]

12 Kulik G, Weber MJ. Akt-dependent and -independent survival signaling pathways utilized by insulin-like growth factor I. Mol Cell Biol 1998;18:6711–8.[Abstract/Free Full Text]

13 Kulik G, Klippel A, Weber MJ. Antiapoptotic signalling by the insulin-like growth factor I receptor, phosphatidylinositol 3-kinase, and Akt. Mol Cell Biol 1997;17:1595–606.[Abstract]

14 Ng SSW, Tsao MS, Chow S, Hedley DW. Inhibition of phosphatidylinositide 3-kinase enhances gemcitabine-induced apoptosis in human pancreatic cancer cells. Cancer Res 2000;60:5451–5.[Abstract/Free Full Text]

15 Brognard J, Clark AS, Ni Y, Dennis PA. Akt/protein kinase B is constitutively active in non-small cell lung cancer cells and promotes cellular survival and resistance to chemotherapy and radiation. Cancer Res 2001;61:3986–97.[Abstract/Free Full Text]

16 Cheng JQ, Ruggeri B, Klein WM, Sonoda G, Altomare DA, Watson DK, et al. Amplification of AKT2 in human pancreatic cells and inhibition of AKT2 expression and tumorigenicity by antisense RNA. Proc Natl Acad Sci U S A 1996;93:3636–41.[Abstract/Free Full Text]

17 Vasko V, Saji M, Hardy E, Kruhlak M, Larin A, Savchenko V, et al. Akt activation and localisation correlate with tumour invasion and oncogene expression in thyroid cancer. J Med Genet 2004;41:161–70.[Abstract/Free Full Text]

18 Yuan ZQ, Sun M, Feldman RI, Wang G, Ma X, Jiang C, et al. Frequent activation of AKT2 and induction of apoptosis by inhibition of phosphoinositide-3-OH kinase/Akt pathway in human ovarian cancer. Oncogene 2000;19:2324–30.[CrossRef][ISI][Medline]

19 Cheng JQ, Godwin AK, Bellacosa A, Taguchi T, Franke TF, Hamilton TC, et al. AKT2, a putative oncogene encoding a member of a subfamily of protein-serine/threonine kinases, is amplified in human ovarian carcinomas. Proc Natl Acad Sci U S A 1992;89:9267–71.[Abstract]

20 Lee SH, Kim HS, Park WS, Kim SY, Lee KY, Kim SH, et al. Non-small cell lung cancer frequently express phosphorylated Akt; an immunohistochemical study. APMIS 2002;110:587–92.[CrossRef][ISI][Medline]

21 Levitzki A, Gazit A. Tyrosine kinase inhibition: an approach to drug development. Science 1995;267:1782–8.[ISI][Medline]

22 Levitt ML, Koty PP. Tyrosine kinase inhibitors in preclinical development. Invest New Drugs 1989;7:213–26.[ISI][Medline]

23 Ciardiello F, Caputo R, Bianco R, Damiano V, Pomatico G, De Placido S, et al. Antitumour effect and potentiation of cytotoxic drug activity in human cancer cells by ZD1839 (Iressa), an epidermal growth factor receptor-selective tyrosine kinase inhibitor. Clin Cancer Res 2000;6:2053–63.[Abstract/Free Full Text]

24 Kris MG, Herbst R, Rischin D, LoRusso P, Baselga J, Hammond L, et al. Objective regression in non-small cell lung cancer patients treated in phase I trials of oral ZD 1839 (Iressa), a selective tyrosine kinase inhibitor that blocks the epidermal growth factor receptor (EGFR) [abstract 233]. Lung Cancer 2000;29(Suppl 1):72.

25 Baselga J, Rischin D, Ranson M, Calvert H, Raymond E, Kieback DG, 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;20:4292–302.[Abstract/Free Full Text]

26 Herbst RS, Maddox AM, Rothenberg ML, Small EJ, Rubin EH, Baselga J, 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;20:3815–25.[Abstract/Free Full Text]

27 Ranson M, Hammond LA, Ferry D, Kris M, Tullo A, Murray PI, 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;20:2240–50.[Abstract/Free Full Text]

28 Fukuoka M, Yano S, Giaccone G, Tamura T, Nakagawa K, Douillard JY, et al. Multi-institutional randomized phase II trial of gefitinib for previously treated patients with advanced non-small-cell lung cancer. J Clin Oncol 2003;21:2237–46.[Abstract/Free Full Text]

29 Kris MG, Natale RB, Herbst RS, Lynch TJ Jr, Prager D, Belani CP, et al. Efficacy of gefitinib, an inhibitor of the epidermal growth factor receptor tyrosine kinase, in symptomatic patients with non-small cell lung cancer: a randomized trial. JAMA 2003;290:2149–58.[Abstract/Free Full Text]

30 Sirotnak FM, Zakowski MF, Miller VA, Scher HI, Kris MG. 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;6:4885–92.[Abstract/Free Full Text]

31 Moasser MM, Basso A, Averbuch SD, Rosen N. The tyrosine kinase inhibitor ZD1839 ("Iressa") inhibits HER2-driven signaling and suppresses the growth of HER2-overexpressing tumor cells. Cancer Res 2001;61:7184–8.[Abstract/Free Full Text]

32 Cappuzzo F, Gregorc V, Rossi E, Cancellieri A, Magrini E, Paties CT, et al. Gefitinib in pretreated non-small-cell lung cancer (NSCLC): analysis of efficacy and correlation with HER2 and epidermal growth factor receptor expression in locally advanced or metastatic NSCLC. J Clin Oncol 2003;21:2658–63.[Abstract/Free Full Text]

33 Travis WD, Colby TV, Corrin B, Shimosato Y, Brambilla E. Histological typing of lung and pleural tumors. 3rd ed. Berlin (Germany): Springer; 1999.

34 Allred DC, Clark GM, Elledge R, Fuqua SA, Brown RW, Chamness GC, et al. Association of p53 protein expression with tumor cell proliferation rate and clinical outcome in node-negative breast cancer. J Natl Cancer Inst 1993;85:200–6.[Abstract]

35 Meier R, Alessi DR, Cron P, Andjelkovic M, Hemmings BA. Mitogenic activation, phosphorylation, and nuclear translocation of protein kinase Bbeta. J Biol Chem 1997;272:30491–7.[Abstract/Free Full Text]

36 Therasse P. New guidelines to evaluate the response to treatment in solid tumors. J Natl Cancer Inst 2000;92:205–16.[Abstract/Free Full Text]

37 Kaplan EL, Meier P: Nonparametric estimation from incomplete observations. J Am Stat Assoc 1985;53:457–81.

38 Cox DR. Regression models and life tables. J R Stat Soc B 1972;34:187–220.[ISI]

39 Agresti A. Categorical data analysis. New York (NY): John Wiley & Sons; 1990.

40 She QB, Solit D, Basso A, Moasser MM. Resistance to gefitinib in PTEN-null HER-overexpressing tumor cells can be overcome through restoration of PTEN function or pharmacologic modulation of constitutive phosphatidylinositol 3'-kinase/Akt pathway signaling. Clin Cancer Res 2003;9:4340–6.[Abstract/Free Full Text]

41 Shah NT, Miller VA, Kris MG, Patel J, Venkatraman E, Benporat L, et al. Bronchioloalveolar histology and smoking history predict response to gefitinib [abstract 2524]. Proc ASCO 2003;22:628.

42 Sato JD, Kawamoto T, Le AD, Mendelsohn J, Polikoff J, Sato GH. Biological effects in vitro of monoclonal antibodies to human epidermal growth factor receptors. Mol Biol Med 1983;1:511–29.[Medline]

43 Perez-Soler R, Dai Q, Ling YH, Zou Y, Lia M, Kroog G, et al. Molecular mechanisms of sensitivity and resistance to the HER1/EGFR tyrosine Kinase inhibitor erlotinib [abstract O-247]. Lung Cancer 2003;41:s72.

44 Giaccone G, Herbst RS, Manegold C, Scagliotti G, Rosell R, Miller V, et al. Gefitinib in combination with gemcitabine and cisplatin in advanced non-small-cell lung cancer: a phase III trial—INTACT 1. J Clin Oncol 2004;22:777–84.[Abstract/Free Full Text]

45 Herbst RS, Giaccone G, Schiller JH, Natale RB, Miller V, et al. Gefitinib in combination with paclitaxel and carboplatin in advanced non-small-cell lung cancer: a phase III trial—INTACT 2. J Clin Oncol 2004;22:785–94.[Abstract/Free Full Text]

46 Bianco R, Shin I, Ritter CA, Yakes FM, Basso A, Rosen N, et al. Loss of PTEN/MMAC1/TEP in EGF receptor-expressing tumor cells counteracts the antitumor action of EGFR tyrosine kinase inhibitors. Oncogene 2003;22:2812–22.[CrossRef][ISI][Medline]

47 Janmaat ML, Kruyt FA, Rodriguez JA, Giaccone G. Response to epidermal growth factor receptor inhibitors in non-small cell lung cancer cells: limited antiproliferative effects and absence of apoptosis associated with persistent activity of extracellular signal-regulated kinase or Akt kinase pathways. Clin Cancer Res 2003;9:2316–26.[Abstract/Free Full Text]

48 Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 2004;350:2129–39.[Abstract/Free Full Text]

49 Paez JG, Janne PA, Lee JC, Tracy S, Greulich H, Gabriel S, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 2004;304:1497–500.[Abstract/Free Full Text]

Manuscript received January 8, 2004; revised May 28, 2004; accepted June 4, 2004.


This article has been cited by other articles in HighWire Press-hosted journals:


Correspondence about this Article

Editorial about this Article

Related Memo to the Media

             
Copyright © 2004 Oxford University Press (unless otherwise stated)
Oxford University Press Privacy Policy and Legal Statement