Purpose: This study aimed to investigate the expression of the ErbB family of receptor tyrosine kinases in pulmonary typical carcinoid and atypical carcinoid tumors and to understand the role of epidermal growth factor receptor (EGFR) signaling in pulmonary carcinoid tumor proliferation.

Experimental Design: Surgically resected typical carcinoid (n = 24) and atypical carcinoid (n = 7) tumor tissues were analyzed by immunohistochemical staining for EGFR, ErbB2, ErbB3, and ErbB4. Sequencing of tumor DNA of exons 18 to 21 of the EGFR gene and the KRAS gene was carried out. Biochemical analysis of lung carcinoid cell lines was used to investigate EGFR signal transduction and response to erlotinib inhibition.

Results: The analysis showed that 45.8% of typical carcinoid and 28.6% of atypical carcinoid tumors express EGFR, 100% of the tumors lack expression of ErbB2, and 100% have moderate to intense staining for ErbB3 and ErbB4. Sequencing of tumor DNA of exons 18 to 21 of the EGFR gene revealed the absence of tyrosine kinase domain mutations in these tumors. Instead, 80.6% tumors harbored a synonymous single nucleotide polymorphism in exon 20. Because EGFR and KRAS mutations tend not to be present at the same time, we sequenced the KRAS gene from pulmonary carcinoid tumor DNA and found that 100% were wild-type. Using a lung carcinoid cell line that expresses EGFR, we found that erlotinib reduced proliferation by inhibiting EGFR signal transduction.

Conclusions: Our findings suggest clinical potential for the use of EGFR inhibitors in the treatment of patients with pulmonary carcinoid tumors, particularly for patients with EGFR-positive pulmonary carcinoid tumors not amenable to surgical resection.

Translational Relevance

Little is known about the mechanisms important for the growth and proliferation of pulmonary carcinoid tumors. Because chemotherapy and radiation have limited efficacy in treating these tumors, and surgical excision is not always feasible, new and more effective strategies are needed for these tumors. Our findings of the overexpression of epidermal growth factor receptor (EGFR), ErbB3, and ErbB4 suggests that signaling by the ErbB family of receptor tyrosine kinases is important for the transmission of growth signals during pulmonary carcinoid tumor growth. The results of this study suggest the clinical potential for the use of EGFR inhibitors in the treatment of patients with pulmonary carcinoid tumors, particularly for patients with EGFR-positive pulmonary carcinoid tumors not amenable to surgical resection.

Lung cancer is the leading cause of cancer-related death in the United States (1). Pulmonary carcinoid tumors are malignant neoplasms comprising neuroendocrine cells that account for 2% to 5% of all lung cancers (2). Pulmonary typical carcinoid, which is considered a low-grade tumor, has scarce mitotic figures (<2 per 10 high-powered fields) and absence of necrosis in histologic specimens (3). Pulmonary atypical carcinoid, a higher-grade tumor, has 2 to 10 mitotic figures per 10 high-powered fields, and/or areas of focal necrosis (3). Typical carcinoid tumors usually present at an average age of 45 years, whereas atypical carcinoid tumors present a decade later. Pulmonary carcinoid tumors can develop without any known risk factors for lung cancer, and in particular less than half of patients are cigarette smokers. Epidemiologic data show that pulmonary typical carcinoid tumors are about four times more frequent than atypical carcinoid tumors, and that women are at higher risk than men of developing these tumors.

The 5-year survival for pulmonary typical carcinoid tumors averages 88% and ranges from 25% to 56% for atypical carcinoid tumors (46). Surgical resection of the tumor in the affected lung is the standard treatment, but complete surgical excision can be difficult or unattainable depending upon the location of the tumor in the chest and whether it has metastasized. Chemotherapy and radiation therapy have limited success in treating pulmonary carcinoid tumors (79). Widely metastatic pulmonary carcinoid tumors can result in carcinoid heart disease or the carcinoid syndrome (10). At the time of diagnosis, up to 30% of patients will have the carcinoid syndrome with symptoms due to excessive production of serotonin from the tumor burden (11). The outcome for patients with widely metastatic disease is poor. A better understanding of the biology of these tumors could lead to the development of novel therapeutic agents to improve patient outcomes.

Altered growth factor signaling through cell surface receptors is thought to contribute to tumorigenesis through abnormal cell proliferation. The ErbB family of receptor tyrosine kinases consists of the epidermal growth factor receptor (EGFR/ErbB1/Her1), ErbB2 (Her2/neu), ErbB3 (Her3) and ErbB4 (Her4; ref. 12). These receptor tyrosine kinases mediate cellular responses to growth factors through their intracellular domain and interact with downstream pathways important for development, differentiation, and proliferation. The EGFR gene is overexpressed and amplified in non–small cell lung cancer (NSCLC), and the presence of somatic mutations in the tyrosine kinase domain of EGFR confers sensitivity to the agents erlotinib and gefitinib (13, 14). ErbB2 (Her2/neu) overexpression in breast and ovarian cancer is associated with poor clinical outcome (1518). Treatment with trastuzumab (Herceptin) to specifically target ErbB2 (Her2/neu) in breast cancer has resulted in improved patient outcome for this cancer type (19). ErbB3 expression has been found in varying degrees in cancers of the lung, breast, ovary, prostate, and elsewhere (20). ErbB3 is the only member of this receptor tyrosine kinase family that does not have intrinsic kinase activity (21, 22). ErbB3 therefore must heterodimerize with another ErbB receptor for signal transduction. ErbB4 is required for a number of cellular responses, such as development, differentiation, and proliferation. ErbB4 is expressed mainly in developing neural tissues and myocardium (23, 24). The role of ErbB4 in tumorigenesis is complex and not completely understood. In breast cancer ErbB4 expression is low to moderate and its expression correlates with a favorable prognosis (25). ErbB4 expression in ependymoma is high and correlated with worse patient survival (26). In a study of 80 patients with NSCLC who underwent surgery, tumors that expressed ErbB4 had decreased survival compared with tumors that did not express ErbB4 (27). A positive correlation was also found with lymph node metastasis and ErbB4 expression in the tumors in this series. Overexpression of ErbB4 in a NSCLC cell line resulted in increased cell proliferation in comparison with ErbB4-negative cells (27).

In the present study, we define the expression pattern for the ErbB family of receptor tyrosine kinases from archival tumor specimens of 31 patients with pulmonary carcinoid tumors, 24 with typical carcinoid, and 7 with atypical carcinoid, for the first time. Additionally we sequenced exons 18 to 21 of the EGFR gene and the KRAS to determine whether activating mutations are present in these rare tumors. Finally using H727 lung carcinoid cells that express EGFR, we found that erlotinib reduced proliferation of this tumor cell line by inhibiting EGFR signal transduction. Our findings might have clinical application by inhibiting EGFR signaling for patients with EGFR-positive pulmonary carcinoid tumors.

Immunohistochemistry. These studies were approved by the Mayo Clinic Institutional Review Board. The Mayo Clinic Lung Cancer Specimen Registry was searched for tissue of carcinoid tumors from January 1, 2001 to January 31, 2006. Sufficient-quality frozen tissue and the corresponding archived paraffin-embedded tissue were available in 31 patients. All tumors were reviewed and confirmed as typical and atypical carcinoid tumors based on the 2004 WHO classification by an expert lung pathologist experienced in neuroendocrine lung cancers (3). Immunohistochemical stains were done on representative 4-μm formalin-fixed, paraffin-embedded tissue sections from the lung tumors using antibodies to chromogranin (Millipore; clone LK2H10; 1:500 dilution), EGFR (Dako; EGFR pharmDx), phospho-EGFR (Dako), ErbB2/Her2Neu (Dako; HercepTest), ErbB3/Her3 (Santa Cruz Biotechnology; polyclonal, 1:100), and ErbB4/Her4 (Santa Cruz Biotechnology; polyclonal, 1:100). For chromogranin, ErbB3/Her3, and ErbB4/Her4, epitope retrieval was done in a heated 1 mmol/L EDTA pH 8.0 solution for 30 min. Antigen-antibody reactions were visualized using a polymer-based detection system (Dako) using diaminobenzidine as the chromogen. For antibodies to EGFR and ErbB2/Her2Neu, the procedure was done according to the manufacturer's instructions for EGFR pharmDx and HercepTest for the Dako Autostainer. Appropriate positive and negative controls were employed for all conditions. Stains were scored 0 to 4+ based on the percentage of positive tumor cells as follows: 0, <5%; 1+, 5% to 10%; 2+, 11% to 50%; 3+, 50% to 80%; 4+, >80%.

Cell culture. H727, H720, UMC-11, and HCC827 were purchased from the American Type Culture Collection and were grown at 37°C in 5% CO2 in RPMI-1640 media with 1% antibiotics (penicillin/streptomycin) and 10% fetal bovine serum.

EGFR and KRAS gene sequencing. DNA was extracted from all of the frozen carcinoid tumor specimens using standard procedures. PCR amplification and sequencing of EGFR exon 18 to 21 was done using the identical conditions and PCR primers described by Lynch and colleagues (13). The PCR primers used were (a) exon 18: 5′-CAA-ATG-AGC-TGG-CAA-GTG-CCG-TGTC-3′, and 5′-GAG-TTT-CCC-AAA-CAC-TCA-GTG-AAAC-3′; (b) exon 19: 5′-GCA-ATA-TCA-GCC-TTA-GGT-GCG-GCTC-3′, and 5′-CAT-AGA-AAG-TGA-ACA-TTT-AGG-ATG-TG-3′; (c) exon 20: 5′-CCA-TGA-GTA-CGT-ATT-TTG-AAA-CTC-3′, and 5′-CAT-ATC-CCC-ATG-GCA-AAC-TCT-TGC-3′; and (d) exon 21: 5′-CTA-ACG-TTC-GCC-AGC-CAT-AAG-TCC-3′, and 5′-GCT-GCG-AGC-TCA-CCC-AGA-ATG-TCT-GG-3′. For KRAS the following PCR primers were used: 5′- GTA-CTG-GTG-GAG-TAT-TTG-AT-3′ and 5′-TGA-AAA-TGG-TCA-GAG-AAA-CC-3′. Amplification was done using 0.2 μmol/L primers and 100 ng DNA with the following conditions: 94°C for 5 min, then 30 cycles of 94°C for 1 min, 58°C for 1 min (55°C for KRAS), 72°C for 1 min. PCR amplicons were resolved by electrophoresis on 2% agarose gels stained with ethidium bromide. The PCR amplicons were cut from gel, gene-cleaned, and sequenced with the forward and reverse PCR primers.

Cell proliferation. Cell proliferation was determined using CyQUANT NF Cell Proliferation Assay (Invitrogen) according to the manufacturer's instructions. The assay is designed to produce a linear analytical response from 100 to 20,000 cells per well. In brief, cells were plated at density of 2,000 cells per well in a 96-well plate. After 48 h the cells were stained with the CyQUANT dye for 30 min at 37°C. The fluorescence intensity of each sample was measured using a fluorescence microplate reader (excitation of 485 nm and emission of 530 nm). The data represent averages of triplicate samples with error bars showing SD.

Cell cycle analysis. Cell cycle determination was analyzed by flow cytometry using ModFitLT software (Verity Software). H727 cells were trypsinized, washed with sterile PBS, and resuspended at 100,000 cells/mL. The cells were fixed in 70% ethanol and stained with propidium iodide (mixture of 2.5 mg/mL propidium iodide with 0.1 mg/mL RNase A and 0.05% Triton X-100) for 40 min at 37°C. The samples were analyzed by flow cytometry (Becton Dickinson FACScan; BD Bioscience).

Immunoblotting. Cellular lysates of H727 and HCC827 cells were prepared by gentle sonication on ice in lysis buffer [50 mmol/L Tris-HCl pH 7.5, 100 mmol/L NaCl, 50 mmol/L NaF, 1% (v/v) NP-40, 5 mmol/L EDTA, 1 mmol/L EGTA, 200 μmol/L sodium orthovanadate, 50 μmol/L β-glycerolpyrophosphate, 100 μmol/L phenylmethylsulfonyl fluoride, 200 μmol/L sodium fluoride, 1 μg/mL of each of leupeptin, aprotinin and pepstatin]. The protein concentration was determined spectrophotometrically using the BCA method (Pierce Biotechnology, Inc.), and equal protein was resolved by 8% SDS-PAGE, and transferred to polyvinylidene fluoride membranes. Nonspecific binding sites were blocked with TBS containing 5% milk prior to addition of the primary antibodies. Bound antibodies were detected with enhanced chemiluminescence (Amersham Biosciences).

Statistical analysis. Experiments were carried out in triplicate for all experimental conditions. Data are shown as mean ± SD. Student's t test was done on the means of two sets of sample data and considered significant if P = 0.05.

Patient characteristics. We examined the clinical characteristics of 31 patients with pulmonary carcinoid tumors diagnosed from January 1, 2001 to January 31, 2006 for which there was available fresh-frozen and paraffin-embedded tissue for study (Table 1). There were 24 patients with pulmonary typical carcinoid tumors and 7 with atypical carcinoid tumors. The entire cohort of subjects included 11 men and 20 women with a mean age of 58 years (range, 31-77 years). Overall follow-up for the entire cohort was a median time of 50 months (range, 1-84 months). Eleven patients (35%) were former cigarette smokers with an average consumption of 30 pack-years. Twenty tumors (64.5%) were located in the right chest. The most common location was the right middle lobe (nine tumors), followed by the right lower lobe (eight tumors), then the left lower lobe (five tumors). One patient had bilateral right and left upper lobe tumors. Evidence of hypercalcemia was present in 5 (16%) patients. Two patients with ectopic production of adrenocorticotropic hormone were diagnosed with Cushing's syndrome 1 month and 11 months prior to the diagnosis of pulmonary carcinoid tumor. There were a total of 16 additional malignancies in 13 (42%) patients, some of which preceded the diagnosis of pulmonary carcinoid tumor, and others that developed afterwards. There were three skin cancers (two melanoma, one squamous cell), two gastrointestinal stromal tumors, two prior pulmonary typical carcinoid tumors, two breast cancers, and prostate cancer, colon cancer, ovarian cancer, papillary thyroid cancer, parathyroid adenoma, polycythemia vera, and multiple uterine leiomyomas. This finding of the association with other malignancies in 42% of the patients has been identified in other cohorts of patients with pulmonary carcinoid tumors and highlights the need for continual cancer surveillance for these patients (28, 29). No patients had MEN 1.

Table 1.

Overall patient characteristics

HistologyAge/GenderTobacco useTumor locationSurgeryT/N/MStageOther cancerSyndromesRecurrenceOutcomeFollow-up (mo)
59/F 40 RUL Lobectomy 1/0/0 Ia    A-NED 50 
58/F RML Lobectomy 1/0/0 Ia Melanoma   A-NED 60 
70/F LLL Lobectomy 1/0/0 Ia Polycythemia vera   A-NED 60 
63/F RML Lobectomy 1/0/0 Ia    A-NED 30 
76/F RUL Lobectomy 1/0/0 Ia Papillary thyroid cancer, breast   A-NED 83 
43/M 56 RML Lobectomy 1/0/0 Ia    A-NED 34 
74/M LUL Segmentectomy 1/0/0 Ia GIST    A-NED 82 
61/F 30 LLL Lobectomy 1/0/0 Ia Breast   A-NED 80 
69/F 40 RUL Lobectomy 1/0/0 Ia  Hypercalcemia  A-NED 78 
66/M RLL Lobectomy 1/0/0 Ia    A-NED 47 
55/F Lingula Segmentectomy 1/0/0 Ia    A-NED 
40/F 20 LLL Sleeve Lobectomy 2/0/0 Ib Multiple uterine leiomyomas   A-NED 50 
56/F RML Lobectomy 2/0/0 Ib    A-NED 12 
31/M LLL Lobectomy 2/0/0 Ib    A-NED 16 
60/F RLL Lobectomy 2/0/0 Ib    A-NED 52 
67/M RML Segmentectomy 2/0/0 Ib Prostate cancer   A-NED 67 
38/M RLL Bi-Lobectomy 2/0/0 Ib  Cushings  A-NED 
53/M 70 LUL Lobectomy 1/1/0 IIa    A-NED 71 
68/F RML Lobectomy 1/1/0 IIa GIST Hypercalcemia  A-NED 13 
65/F RLL Lobectomy 2/1/0 IIb    A-NED 27 
38/M LLL Lobectomy 2/1/0 IIb    A-NED 28 
51/F RML Lobectomy 1/2/0 IIIa    A-NED 79 
59/F RML Lobectomy 4/0/0 IIIb Squamous skin   A-NED 76 
77/F 10 Lingula Wedge 4/0/0 IIIb TC 1995 RLL T1N0M0, ovary  Lingula A-NED 23 
65/F 15 RLL Lobectomy 1/0/0 Ia Parathyroid adenoma Hypercalcemia Liver A-PD 48 
57/F RML Lobectomy 1/0/0 Ia  Hypercalcemia  A-NED 51 
65/F 42 RLL Lobectomy 1/0/0 Ia    A-NED 53 
60/M RUL & LUL Lobectomy 1/0/0 Ia Bilateral carcinoids, colon   A-NED 
66/F RLL Lobectomy 2/0/0 Ib Melanoma Hypercalcemia  A-NED 41 
33/M RLL Lobectomy 1/2/0 IIIa  Cushings  A-NED 13 
41/M LUL Lobectomy 1/2/1 IV   Liver A-PD 84 
HistologyAge/GenderTobacco useTumor locationSurgeryT/N/MStageOther cancerSyndromesRecurrenceOutcomeFollow-up (mo)
59/F 40 RUL Lobectomy 1/0/0 Ia    A-NED 50 
58/F RML Lobectomy 1/0/0 Ia Melanoma   A-NED 60 
70/F LLL Lobectomy 1/0/0 Ia Polycythemia vera   A-NED 60 
63/F RML Lobectomy 1/0/0 Ia    A-NED 30 
76/F RUL Lobectomy 1/0/0 Ia Papillary thyroid cancer, breast   A-NED 83 
43/M 56 RML Lobectomy 1/0/0 Ia    A-NED 34 
74/M LUL Segmentectomy 1/0/0 Ia GIST    A-NED 82 
61/F 30 LLL Lobectomy 1/0/0 Ia Breast   A-NED 80 
69/F 40 RUL Lobectomy 1/0/0 Ia  Hypercalcemia  A-NED 78 
66/M RLL Lobectomy 1/0/0 Ia    A-NED 47 
55/F Lingula Segmentectomy 1/0/0 Ia    A-NED 
40/F 20 LLL Sleeve Lobectomy 2/0/0 Ib Multiple uterine leiomyomas   A-NED 50 
56/F RML Lobectomy 2/0/0 Ib    A-NED 12 
31/M LLL Lobectomy 2/0/0 Ib    A-NED 16 
60/F RLL Lobectomy 2/0/0 Ib    A-NED 52 
67/M RML Segmentectomy 2/0/0 Ib Prostate cancer   A-NED 67 
38/M RLL Bi-Lobectomy 2/0/0 Ib  Cushings  A-NED 
53/M 70 LUL Lobectomy 1/1/0 IIa    A-NED 71 
68/F RML Lobectomy 1/1/0 IIa GIST Hypercalcemia  A-NED 13 
65/F RLL Lobectomy 2/1/0 IIb    A-NED 27 
38/M LLL Lobectomy 2/1/0 IIb    A-NED 28 
51/F RML Lobectomy 1/2/0 IIIa    A-NED 79 
59/F RML Lobectomy 4/0/0 IIIb Squamous skin   A-NED 76 
77/F 10 Lingula Wedge 4/0/0 IIIb TC 1995 RLL T1N0M0, ovary  Lingula A-NED 23 
65/F 15 RLL Lobectomy 1/0/0 Ia Parathyroid adenoma Hypercalcemia Liver A-PD 48 
57/F RML Lobectomy 1/0/0 Ia  Hypercalcemia  A-NED 51 
65/F 42 RLL Lobectomy 1/0/0 Ia    A-NED 53 
60/M RUL & LUL Lobectomy 1/0/0 Ia Bilateral carcinoids, colon   A-NED 
66/F RLL Lobectomy 2/0/0 Ib Melanoma Hypercalcemia  A-NED 41 
33/M RLL Lobectomy 1/2/0 IIIa  Cushings  A-NED 13 
41/M LUL Lobectomy 1/2/1 IV   Liver A-PD 84 

NOTE: Listed are the characteristics of the 24 patients with typical carcinoid and the 7 patients with atypical carcinoid tumors. Tobacco use is indicated in pack-years.

Abbreviations: T, typical; A, atypical; RLL, right lower lobe; RML, right middle lobe; RUL, right upper lobe; LLL, left lower lobe; LUL, left upper lobe; A-NED, alive with no evidence of disease activity at time of last follow-up; A-PD, alive with persistent disease activity at time of last follow-up.

Among the 24 patients with pulmonary typical carcinoid tumors, 11 had stage IA disease (T1N0M0), 6 had stage IB disease (T1N1M0), 2 had stage IIA disease (T2N0M0), 2 had stage IIB disease (T2N1M0), 1 had stage IIIA disease (T1N2M0), and 2 had stage IIIB disease (both T4N0M0). All 24 patients are alive and have no evidence of disease at follow-up with a median time of 50 months.

Among the seven patients with atypical carcinoid tumors, four patients had stage IA disease (T1N0M0), one had stage IB disease (T1N1M0), one had stage IIIA disease (T1N2M0), and one had stage IV disease (T1N2M1) with metastasis to the liver. All seven patients are alive at follow-up with a median time of 48 months. There was no evidence of disease at the time of follow-up in five (71 %) patients. One patient developed liver metastasis 35 months after resection of the primary lung tumor.

The impressive survival in our cohort of patients is likely due to several factors, including the early stage of the majority of the tumors, the expertise of the team of physicians and surgeons caring for these patients, and selection bias. Indeed our study only encompasses tumors resected from 2001 to 2006 where we had matched frozen and paraffin-embedded tissue available to study.

ErbB receptor expression in pulmonary carcinoid tumors. Positive staining for EGFR was observed in 11 of 24 (45.8 %) typical carcinoid tumors and in 2 of 7 (28.6 %) atypical carcinoid tumors (Figs. 1A to D; Table 2). Five typical carcinoid tumors had 0 to 1+ EGFR immunoreactivity, whereas six had 2 to 4+ immunoreactivity. Both of the atypical carcinoid tumors had 2 to 4+ immunoreactivity. The EGFR immunoreactivity was present exclusively in the epithelial cell membranes except in one typical carcinoid tumor which had low-level membranous and cytoplasmic staining for EGFR. Additional slides were prepared for phospho-EGFR staining from the subset of tumors with EGFR immunoreactivity, but we did not observe staining with phospho-EGFR from any of these samples. Immunoreactivity for the ErbB2 receptor was absent in all samples of typical carcinoid and atypical carcinoid tumors. Receptor expression for the ErbB3 and ErbB4 receptors was intensely positive in all samples of typical carcinoid and atypical carcinoid tumors. In contrast to EGFR expression, which was present in the epithelial cell membranes, the immunoreactivity for ErbB3 and c- ErbB4 receptors was entirely cytoplasmic in all of the carcinoid tumor sections.

Fig. 1.

A, typical carcinoid (A) showing strong and diffuse expression of chromogranin (B) and focal strong expression of EGFR (C). ErbB2/Her2neu was negative (D). ErbB3 (E) and ErbB4 (F) had diffuse expression but intensity was low to moderate for ErbB3 (×400). B, atypical carcinoid (A) with diffuse immunoreactivity for chromogranin (B) also showing strong diffuse expression of EGFR (C). No expression of ErbB2/Her2neu present (D). ErbB3 (E) and ErbB4 (F) show diffuse moderate to strong expression (×400). C, typical carcinoid (A) showing no expression of EGFR (C). The expression of ErbB3 was variable within the tumor (E). [B. chromogranin, D. ErbB2/Her2neu, F. ErbB4]. (×400). D, atypical carcinoid with focal necrosis (A). No expression of EGFR (C) or ErbB2/Her2neu (D). ErbB3 (E) and ErbB4 (F) expression is moderate. (×400).

Fig. 1.

A, typical carcinoid (A) showing strong and diffuse expression of chromogranin (B) and focal strong expression of EGFR (C). ErbB2/Her2neu was negative (D). ErbB3 (E) and ErbB4 (F) had diffuse expression but intensity was low to moderate for ErbB3 (×400). B, atypical carcinoid (A) with diffuse immunoreactivity for chromogranin (B) also showing strong diffuse expression of EGFR (C). No expression of ErbB2/Her2neu present (D). ErbB3 (E) and ErbB4 (F) show diffuse moderate to strong expression (×400). C, typical carcinoid (A) showing no expression of EGFR (C). The expression of ErbB3 was variable within the tumor (E). [B. chromogranin, D. ErbB2/Her2neu, F. ErbB4]. (×400). D, atypical carcinoid with focal necrosis (A). No expression of EGFR (C) or ErbB2/Her2neu (D). ErbB3 (E) and ErbB4 (F) expression is moderate. (×400).

Close modal
Table 2.

Pulmonary carcinoid tumor ErbB receptor expression by immunohistochemistry

HistologyStageChromgraninEGFR SEQ EXON 18-21EGFR
ErbB2
ErbB3
ErbB4
Membranous (%)Cytoplasmic (%)Membranous (%)Cytoplasmic (%)Membranous (%)Cytoplasmic (%)Membranous (%)Cytoplasmic (%)
Ia SD E20 SNP G->A 30 100 100 
Ia SD E20 SNP G->A 35 100 100 
Ia SD E20 SNP G->A 100 100 
Ia MD E20 SNP G->A 100 100 
Ia SD E20 SNP G->A 100 100 
Ia SD WT 60 100 100 
Ia Mod-strong diffuse E20 SNP G->A 100 100 
Ia SD E20 SNP G->A 25 100 100 
Ia SD E20 SNP G->A 100 100 
Ia SD E20 SNP G->A 10 100 100 
Ia SD WT 100 100 
Ib SD E20 SNP G->A 100 100 
Ib MD WT 100 100 
Ib SD E20 SNP G->A 100 100 
Ib Mild-mod diffuse E20 SNP G->A 100 100 
Ib SD WT 100 100 
Ib SD E20 SNP G->A 100 100 
IIa SD E20 SNP G->A 10 100 100 
IIa SD E20 SNP G->A 40 100 100 
IIb SD E20 SNP G->A 100 100 
IIb SD E20 SNP G->A 100 100 
IIIa SD E20 SNP G->A 100 100 
IIIb SD WT 10 10 100 100 
IIIb SD E20 SNP G->A 15 100 100 
Ia SD E20 SNP G->A 30 100 100 
Ia SD WT 80 100 100 
Ia SD E20 SNP G->A 100 100 
Ia SD E20 SNP G->A 100 100 
Ib SD E20 SNP G->A 100 100 
IIIa SD E20 SNP G->A 100 100 
IV SD E20 SNP G->A 100 100 
HistologyStageChromgraninEGFR SEQ EXON 18-21EGFR
ErbB2
ErbB3
ErbB4
Membranous (%)Cytoplasmic (%)Membranous (%)Cytoplasmic (%)Membranous (%)Cytoplasmic (%)Membranous (%)Cytoplasmic (%)
Ia SD E20 SNP G->A 30 100 100 
Ia SD E20 SNP G->A 35 100 100 
Ia SD E20 SNP G->A 100 100 
Ia MD E20 SNP G->A 100 100 
Ia SD E20 SNP G->A 100 100 
Ia SD WT 60 100 100 
Ia Mod-strong diffuse E20 SNP G->A 100 100 
Ia SD E20 SNP G->A 25 100 100 
Ia SD E20 SNP G->A 100 100 
Ia SD E20 SNP G->A 10 100 100 
Ia SD WT 100 100 
Ib SD E20 SNP G->A 100 100 
Ib MD WT 100 100 
Ib SD E20 SNP G->A 100 100 
Ib Mild-mod diffuse E20 SNP G->A 100 100 
Ib SD WT 100 100 
Ib SD E20 SNP G->A 100 100 
IIa SD E20 SNP G->A 10 100 100 
IIa SD E20 SNP G->A 40 100 100 
IIb SD E20 SNP G->A 100 100 
IIb SD E20 SNP G->A 100 100 
IIIa SD E20 SNP G->A 100 100 
IIIb SD WT 10 10 100 100 
IIIb SD E20 SNP G->A 15 100 100 
Ia SD E20 SNP G->A 30 100 100 
Ia SD WT 80 100 100 
Ia SD E20 SNP G->A 100 100 
Ia SD E20 SNP G->A 100 100 
Ib SD E20 SNP G->A 100 100 
IIIa SD E20 SNP G->A 100 100 
IV SD E20 SNP G->A 100 100 

NOTE: The percent positive membranous and cytoplasmic staining for the 24 typical carcinoid and the 7 atypical carcinoid tumors is listed.Abbreviations: SD, strong diffuse; MD, moderate diffuse.

Pulmonary carcinoid tumors lack activating EGFR tyrosine kinase mutations. In a subset of patients with NSCLC, the presence of somatic mutations in the tyrosine kinase domain of the EGFR gene is associated with sensitivity to the small-molecule EGFR inhibitors gefitinib and erlotinib (13, 14). We examined whether pulmonary carcinoid tumors have mutations in exons 18 to 21 of the EGFR gene by PCR amplification and sequencing of tumor specimen DNA. We found no activating mutations in the EGFR kinase domain from the 24 typical carcinoid and 7 atypical carcinoid tumors. Interestingly the sequences were not all wild-type; 19 (79.2 %) typical carcinoid and 6 (85.7 %) atypical carcinoid tumors had a unique single nucleotide polymorphism (SNP) in exon 20 (Fig. 2). This synonymous SNP, a G to A substitution, does not change the protein coding sequence of EGFR.

Fig. 2.

Sequence of the EGFR exon 20 SNP. Shown is the sequence of exon 20 for the EGFR wild-type (WT) and the G to A substitution present in the SNP.

Fig. 2.

Sequence of the EGFR exon 20 SNP. Shown is the sequence of exon 20 for the EGFR wild-type (WT) and the G to A substitution present in the SNP.

Close modal

We compared the characteristics of the pulmonary carcinoid tumors that express EGFR with those that do not express EGFR. No statistically important correlations were observed, likely due to the small sample size. Specifically there was no difference in age, gender, tobacco use, histology, stage, recurrence, presence of the exon 20 SNP, or outcome.

Pulmonary carcinoid tumors lack KRAS mutations. Activating KRAS mutations are present in 15% to 30% of NSCLC, especially lung adenocarcinomas, and are associated with poor outcome (30). The most frequent mutations are found in codons 12 and 13 in exon 2 and result in constitutively active KRAS. EGFR mutations are common in tumors from patients who have never smoked or have minimal cigarette consumption, whereas KRAS mutations are more frequent in patient tumors with substantial cigarette consumption (31). Further, KRAS mutations have been associated with resistance to anti-EGFR treatment (32). EGFR and KRAS mutations are rarely found in the same tumors, and as discussed above none of the 31 pulmonary carcinoid tumors had EGFR tyrosine kinase domain mutations. We therefore examined whether these tumors have KRAS gene mutations by PCR amplification and sequencing of tumor specimen DNA. We found no mutations present in the KRAS gene from the 24 typical carcinoid and 7 atypical carcinoid tumors.

The reagents available for studying pulmonary carcinoid tumor biology in the laboratory are limited to a few cell lines. We obtained two “typical” lung carcinoid cell lines, H727 and UMC-11, and H720, an “atypical” lung carcinoid cell line. Although these cell lines express neuroendocrine markers, it is uncertain whether these cell lines are actually typical or atypical in histology because they were created prior to the modern WHO classification for pulmonary carcinoid tumors (3). We examined the ErbB receptor expression in H720, H727, and UMC-11 cell lines to determine whether these cell lines could be used as a model of EGFR-positive pulmonary carcinoid tumors. We used HCC827 NSCLC cells as a control, because these cells contain the L858R-activating EGFR tyrosine kinase domain mutation, have amplification of EGFR, and have been shown to be highly sensitive to EGFR inhibition (33). We found that H727 lung carcinoid cells, but not H720 or UMC-11 cells, express EGFR (Fig. 3A). The EGFR gene from H727 cells had previously been sequenced by Lynch and colleagues and shown to be without any tyrosine kinase domain mutations, a finding similar to the 31 tumors in our series (13). In a genetic screen of human cancers and cell lines for mutations in RAS and RAF genes, the H727 cancer cell line was found to have the activating KRAS2 G12V mutation and a normal B-RAF gene (34). In the subsequent experiments we used the H727 cell line as a model for EGFR-positive pulmonary carcinoid tumors.

Fig. 3.

EGFR expression and activation in pulmonary carcinoid cell lines. We first examined the expression of EGFR in three pulmonary carcinoid cell lines, H720, H727, and UMC-11, and found expression only in H727 by Western blotting (A). Next we show that we can capture EGFR from H727 cell by immunoprecipitation using both an EGFR antibody and a phosphotyrosine (pY) antibody followed by Western blotting for EGFR (B). Using a phosphospecific EGFR antibody to tyrosine 1068, we show that EGF rapidly stimulates EGFR phosphorylation in H727 cells, whereas heregulin as a control growth factor does not result in EGFR phosphorylation (C). We examined the time course of EGF-stimulated H727 cells compared with control HCC827 NSCLC cells to gain insights into EGFR signal transduction to downstream ERK and AKT pathways (D). For H727, EGF stimulation of these cells led to minimal EGFR phosphorylation at 5 min, which increased from 10 to 30 min and remained sustained for 24 h. ERK was transiently phosphorylated at 5 min, whereas AKT became phosphorylated from 30 min to 3 h. For comparison, HCC827 which has a constitutively active EGFR, showed baseline phosphorylation which increased significantly at 30 min and remained that way until 24 h when it returned to baseline. Baseline ERK phosphorylation increased at 30 min, whereas AKT phosphorylation increased from 10 min to 1 h before returning to baseline.

Fig. 3.

EGFR expression and activation in pulmonary carcinoid cell lines. We first examined the expression of EGFR in three pulmonary carcinoid cell lines, H720, H727, and UMC-11, and found expression only in H727 by Western blotting (A). Next we show that we can capture EGFR from H727 cell by immunoprecipitation using both an EGFR antibody and a phosphotyrosine (pY) antibody followed by Western blotting for EGFR (B). Using a phosphospecific EGFR antibody to tyrosine 1068, we show that EGF rapidly stimulates EGFR phosphorylation in H727 cells, whereas heregulin as a control growth factor does not result in EGFR phosphorylation (C). We examined the time course of EGF-stimulated H727 cells compared with control HCC827 NSCLC cells to gain insights into EGFR signal transduction to downstream ERK and AKT pathways (D). For H727, EGF stimulation of these cells led to minimal EGFR phosphorylation at 5 min, which increased from 10 to 30 min and remained sustained for 24 h. ERK was transiently phosphorylated at 5 min, whereas AKT became phosphorylated from 30 min to 3 h. For comparison, HCC827 which has a constitutively active EGFR, showed baseline phosphorylation which increased significantly at 30 min and remained that way until 24 h when it returned to baseline. Baseline ERK phosphorylation increased at 30 min, whereas AKT phosphorylation increased from 10 min to 1 h before returning to baseline.

Close modal

Phosphorylation status of EGFR and effect of EGFR activation on downstream signaling pathways in H727 cells. H727 cells growing in the absence of exogenous EGF were lysed, immunoprecipitated with an EGFR antibody, and blotted with an EGFR antibody or a phosphotyrosine (pY) antibody. As shown in Fig. 3B, H727 cells express EGFR that has minimal baseline phosphorylation. Because activated EGFR contains multiple tyrosine phosphorylation sites, we used a phospho-specific antibody to Tyr1068, an established EGFR autophosphorylation site, to show that the basal level of EGFR phosphorylation increases significantly with EGF stimulation (100 ng/mL for 10 minutes) of the cells (Fig. 3C). There was no apparent increased EGFR phosphorylation when the H727 cells were treated for 10 minutes with 100 ng/mL heregulin, a ligand for ErbB2-ErbB3 heterodimers and ErbB4, but is not recognized by EGFR.

Next we examined the effect of EGF stimulation of H727 cells in the context of EGFR phosphorylation and downstream signaling pathways (Fig. 3D). EGFR phosphorylation increased from baseline in H727 cells at 10 minutes and persisted for 24 hours, whereas in the control HCC827 cells the activated level of EGFR phosphorylation at baseline increased significantly by 30 minutes and then decreased after 6 hours. In the H727 cells we observed initial rapid extracellular signal-regulated kinase (ERK) phosphorylation at 5 minutes followed by absent ERK activity at all other time points. ERK phosphorylation in the HCC827 cells paralleled the phosphorylation of EGFR, increasing from 30 minutes to 6 hours, then decreasing to low levels. AKT phosphorylation in both the H727 and HCC827 cells showed increased phosphorylation from 30 minutes to 3 hours in the H727 cells and 10 minutes to 1 hour in the HCC827 cells, followed by loss of phosphorylation.

Erlotinib inhibition of EGFR activity in H727 cells results in decreased proliferation. We examined the effect of erlotinib inhibition of EGFR activity and its effect on downstream signaling pathways in H727 cells that were treated with various concentrations of erlotinib for 3 hours followed by stimulation with EGF for 60 minutes (Fig. 4A). We observed a dose- dependent decrease in EGFR phosphorylation in cells treated with increasing concentrations of erlotinib. There seemed to be a decrease in AKT phosphorylation in both the H727 and HCC827 cells. Erlotinib treatment of the H727 cells resulted in increased ERK phosphorylation, whereas in HCC827 cells the level of ERK phosphorylation decreased substantially. Next we examined the effect of erlotinib on the proliferation of these cells following treament for 48 hours. We observed a statistically significant reduction in H727 cell proliferation in a dose-dependent manner with erlotinib (Fig. 4B). H727 proliferation decreased by 32% with 0.1 μmol/L of erlotinib, 48% with 1 μmol/L of erlotinib, and 52% with 10 μmol/L of erlotinib. For any given dose of erlotinib, the HCC827 cells were more sensitive and had a greater reduction in proliferation compared with H727 cells. To investigate the mechanism of reduced cell proliferation in H727 cells due to EGFR inhibition, we examined their cell cycle transition and found that H727 cells treated with erlotinib developed a G2/M cell cycle arrest (Fig. 4C).

Fig. 4.

Erlotinib treatment of H727 cells reduces proliferation by causing a G2/M cell cycle arrest. H727 cells treated with various concentrations of erlotinib for 3 h followed by stimulation with EGF for 60 min (A) had a dose-dependent decrease in EGFR phosphorylation. AKT phosphorylation levels in both the H727 and HCC827 cells decreased. Erlotinib treatment of the H727 cells resulted in increased ERK phosphorylation, whereas in HCC827 cells the level of ERK phosphorylation decreased substantially. H727 cells treated with erlotinib for 48 h developed a dose-dependent reduction in cellular proliferation (B). Bar graphs, mean from three independent experiments ± SD. We observed a statistically significant reduction in proliferation with all dose of erlotinib. HCC827 cells are shown for comparison. Proliferating H727 cells were analyzed by flow cytometry for cell cycle phase (C) and show accumulation of cells at the G2/M phase.

Fig. 4.

Erlotinib treatment of H727 cells reduces proliferation by causing a G2/M cell cycle arrest. H727 cells treated with various concentrations of erlotinib for 3 h followed by stimulation with EGF for 60 min (A) had a dose-dependent decrease in EGFR phosphorylation. AKT phosphorylation levels in both the H727 and HCC827 cells decreased. Erlotinib treatment of the H727 cells resulted in increased ERK phosphorylation, whereas in HCC827 cells the level of ERK phosphorylation decreased substantially. H727 cells treated with erlotinib for 48 h developed a dose-dependent reduction in cellular proliferation (B). Bar graphs, mean from three independent experiments ± SD. We observed a statistically significant reduction in proliferation with all dose of erlotinib. HCC827 cells are shown for comparison. Proliferating H727 cells were analyzed by flow cytometry for cell cycle phase (C) and show accumulation of cells at the G2/M phase.

Close modal

Prior to our study there were two reports evaluating either the expression of EGFR or ErbB2 in pulmonary carcinoid tumors. Rusch and colleagues carried out immunohistochemistry for EGFR and found moderate to high staining (2+ to 4+ representing 20% to >80% positive cells) in 7 of 25 (28%) pulmonary typical carcinoid tumors and in 4 of 7 (57%) atypical carcinoid tumors (35). Wilkinson and colleagues determined there was no expression of ErbB2 in 26 pulmonary typical carcinoid and 23 atypical carcinoid tumors (36). Using the same scoring criteria as Rusch, we found moderate to high staining in 5 of 24 (21%) pulmonary typical carcinoid tumors and in 2 of 7 (29%) atypical carcinoid tumors. Six additional typical carcinoid tumors in our series had low-level staining ranging from 1% to 15% for EGFR. None of the tumors with EGFR staining had phospho-EGFR staining, suggesting that the EGFR pathway is not constitutively active in these tumors. Similarly we observed absent phospho-EGFR in the H727 carcinoid cell line, but erlotinib inhibition of EGFR activity in these cells resulted in decreased proliferation despite the baseline phospho-EGFR activity. Our study results are also concordant with the findings reported by Wilkinson showing no expression of ErbB2. Our report is the first to include staining for ErbB3 and ErbB4, which revealed 100% immunoreactivity in all of the typical and atypical pulmonary carcinoid tumors. The function of ErbB3 and ErbB4 in these tumors is unknown and provides an area for future investigation.

Other novel findings include the absence of EGFR tyrosine kinase domain mutations and the absence of KRAS mutations in all of our patient samples. The exon 20 G to A synonymous SNP was present in a high proportion of these tumors. Most synonymous SNPs create silent mutations without any functional significance, but there have been reports of synonymous SNPs affecting the stability of mRNA (37, 38). The significance of this exon 20 SNP is unknown, and did not correlate with the expression of EGFR or other patient characteristics.

We found that the kinetics of phosphorylation of EGFR in H727 lung carcinoid cells was similar to the control HCC827 cells in that sustained phosphorylation lasted for hours after EGF stimulation. This finding could be the result of altered phosphatase activity in the cells, or other factors involved in receptor recycling. Signaling to downstream kinases after EGF stimulation showed transient ERK phosphorylation in H727 cells, whereas HCC827 cells developed sustained ERK phosphorylation which is an effect of the L858R-activating EGFR tyrosine kinase domain mutation in these cells. In both H727 and HCC827 cells, AKT seemed to be constitutively phosphorylated at baseline with little increase with EGF stimulation.

Erlotinib, an oral reversible inhibitor of EGFR tyrosine kinase activity, showed improved survival compared with placebo in the BR.21 trial as monotherapy in previously treated advanced NSCLC patients (39). Gefitinib, however, failed to show a survival benefit as monotherapy in previously treated advanced NSCLC in the ISEL trial (40). In our study erlotinib treatment revealed important differences in the cellular response of the H727 lung carcinoid cells compared with the HCC827 NSCLC cells. Amann and colleagues previously reported that the mechanism of reduced proliferation in HCC827 cells treated with erlotinib occurred through reduced ERK and AKT phosphorylation, which led to a G1 cell cycle arrest (33). In H727 cells we observed that treatment with erlotinib reduced EGFR and AKT phosphorylation but resulted in ERK activation and the development of a G2/M cell cycle arrest. A similar finding of ERK activation resulting in cell cycle arrest has been reported in breast cancer and leukemia (41, 42). H727 lung carcinoid cells do not have EGFR tyrosine kinase domain mutations, yet they have a KRAS G12V mutation. Under normal unstimulated growth conditions in H727 cells, EGFR has minimal constitutive phosphorylation, and although this cell line has a KRAS G12V mutation, we did not observe constitutive ERK phosphorylation. Interestingly we noted that ERK became phosphorylated only when EGFR signaling was inhibited with erlotinib. In the absence of EGFR tyrosine kinase domain mutations, the absolute level of EGFR has been found to be an important independent factor for the response to treatment with EGFR tyrosine kinase inhibitors (43, 44). We found moderate to high staining for EGFR in 21% of the pulmonary typical carcinoid tumors and in 29% of the atypical carcinoid tumors from the 31 patient samples, and high expression of EGFR in the H727 cell line by Western blotting. Although the patient samples and H727 cells lack EGFR tyrosine kinase domain mutations, the high expression of EGFR may be responsible for the sensitivity to erlotinib.

Surgical resection of the tumor mass remains the optimal treatment for pulmonary carcinoid tumors. Complete surgical excision, including dissection of regional lung and mediastinal lymph node metastases, should be attempted to remove all of the tumor tissue for a cure. Depending upon the location of the tumor, however, this may not be possible. Lung preservation surgery, utilizing bronchial sleeve procedures, requires specialized surgical expertise (4547). The role of adjuvant therapy following the complete surgical resection of a pulmonary carcinoid tumor is undefined due to the small series and retrospective nature of the reports. There are no prospective trials that directly address the benefit of chemotherapy or radiation therapy for patients with pulmonary typical carcinoid or atypical carcinoid tumors. Our findings might have clinical application in the treatment of patients with pulmonary carcinoid tumors. The inhibition of EGFR signaling may represent a potential therapeutic strategy in the treatment of patients with EGFR-positive pulmonary carcinoid tumors, especially for those patients whose cancer cannot be surgically resected, or in patients presenting with advanced disease.

No potential conflicts of interest were disclosed.

Grant support: Mayo Foundation.

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.

1
Jemal A, Siegel R, Ward E, et al. Cancer statistics, 2008.
CA Cancer J Clin
2008
;
58
:
71
–96.
2
Thomas CF Jr, Jett JR. Bronchial carcinoid tumors. In: Rose BD (ed.), UpToDate. Wellesley, MA: UpToDate.com, 2008.
3
Gosney J, Travis WD. World Health Organization Classification of Tumors: Pathology and Genetics of Tumours of the Lung, Pleura, Thymus and Heart 2004. Lyon: IARC.
4
Thomas CF Jr, Tazelaar HD, Jett JR. Typical and atypical pulmonary carcinoids: outcome in patients presenting with regional lymph node involvement.
Chest
2001
;
119
:
1143
–50.
5
Cardillo G, Sera F, Di Martino M, et al. Bronchial carcinoid tumors: nodal status and long-term survival after resection.
Ann Thorac Surg
2004
;
77
:
1781
–5.
6
Rugge M, Fassan M, Clemente R, et al. Bronchopulmonary carcinoid: phenotype and long-term outcome in a single-institution series of Italian patients.
Clin Cancer Res
2008
;
14
:
149
–54.
7
Chakravarthy A, Abrams RA. Radiation therapy in the management of patients with malignant carcinoid tumors.
Cancer
1995
;
75
:
1386
–90.
8
Saltz L, Lauwers G, Wiseberg J, Kelsen D. A phase II trial of carboplatin in patients with advanced APUD tumors.
Cancer
1993
;
72
:
619
–22.
9
Jodrell DI, Smith IE. Carboplatin in the treatment of metastatic carcinoid tumours and paraganglioma: a phase II study.
Cancer Chemother Pharmacol
1990
;
26
:
62
–4.
10
Connolly HM, Rubin J, Seward JB, et al. Sandostatin LAR (long-acting octreotide acetate) for malignant carcinoid syndrome: a 3-year experience.
N Engl J Med
2003
;
348
:
1005
–15.
11
Modlin IM, Sandor A. An analysis of 8305 cases of carcinoid tumors.
Cancer
1997
;
79
:
813
–29.
12
Kelly K, Huang C. Biological agents in non-small cell lung cancer: a review of recent advances and clinical results with a focus on epidermal growth factor receptor and vascular endothelial growth factor.
J Thorac Oncol
2008
;
3
:
664
–73.
13
Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib.
N Engl J Med
2004
;
350
:
2129
–39.
14
Paez JG, Janne PA, Lee JC, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy.
Science
2004
;
304
:
1497
–500.
15
Rastelli F, Crispino S. Factors predictive of response to hormone therapy in breast cancer.
Tumori
2008
;
94
:
370
–83.
16
Perez EA, Baweja M. HER2-positive breast cancer: current treatment strategies.
Cancer Invest
2008
;
26
:
545
–52.
17
Cirisano FD, Karlan BY. The role of the HER-2/neu oncogene in gynecologic cancers.
J Soc Gynecol Investig
1996
;
3
:
99
–105.
18
Crijns AP, Duiker EW, de Jong S, Willemse PH, Van der Zee AG, de Vries EG. Molecular prognostic markers in ovarian cancer: toward patient-tailored therapy.
Int J Gynecol Cancer
2006
;
16 Suppl 1
:
152
–65.
19
Romond EH, Perez EA, Bryant J, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer.
N Engl J Med
2005
;
353
:
1673
–84.
20
Sithanandam G, Anderson LM. The ERBB3 receptor in cancer and cancer gene therapy.
Cancer Gene Ther
2008
;
15
:
413
–48.
21
Carraway KL, III, Sliwkowski MX, Akita R, et al. The erbB3 gene product is a receptor for heregulin.
J Biol Chem
1994
;
269
:
14303
–6.
22
Kraus MH, Issing W, Miki T, Popescu NC, Aaronson SA. Isolation and characterization of ERBB3, a third member of the ERBB/epidermal growth factor receptor family: evidence for overexpression in a subset of human mammary tumors.
Proc Natl Acad Sci U S A
1989
;
86
:
9193
–7.
23
Gassmann M, Casagranda F, Orioli D, et al. Aberrant neural and cardiac development in mice lacking the ErbB4 neuregulin receptor.
Nature
1995
;
378
:
390
–4.
24
Jones FE, Golding JP, Gassmann M. ErbB4 signaling during breast and neural development: novel genetic models reveal unique ErbB4 activities.
Cell Cycle
2003
;
2
:
555
–9.
25
Sundvall M, Iljin K, Kilpinen S, Sara H, Kallioniemi OP, Elenius K. Role of ErbB4 in breast cancer.
J Mammary Gland Biol Neoplasia
2008
;
13
:
259
–68.
26
Gilbertson RJ, Bentley L, Hernan R, et al. ERBB receptor signaling promotes ependymoma cell proliferation and represents a potential novel therapeutic target for this disease.
Clin Cancer Res
2002
;
8
:
3054
–64.
27
Starr A, Greif J, Vexler A, et al. ErbB4 increases the proliferation potential of human lung cancer cells and its blockage can be used as a target for anti-cancer therapy.
Int J Cancer
2006
;
119
:
269
–74.
28
Li AF, Hsu CY, Li A, et al. A 35-year retrospective study of carcinoid tumors in Taiwan: differences in distribution with a high probability of associated second primary malignancies.
Cancer
2008
;
112
:
274
–83.
29
Cote ML, Wenzlaff AS, Philip PA, Schwartz AG. Secondary cancers after a lung carcinoid primary: a population-based analysis.
Lung Cancer
2006
;
52
:
273
–9.
30
Kim YT, Kim TY, Lee DS, et al. Molecular changes of epidermal growth factor receptor (EGFR) and KRAS and their impact on the clinical outcomes in surgically resected adenocarcinoma of the lung.
Lung Cancer
2008
;
59
:
111
–8.
31
Tam IY, Chung LP, Suen WS, et al. Distinct epidermal growth factor receptor and KRAS mutation patterns in non-small cell lung cancer patients with different tobacco exposure and clinicopathologic features.
Clin Cancer Res
2006
;
12
:
1647
–53.
32
Massarelli E, Varella-Garcia M, Tang X, et al. KRAS mutation is an important predictor of resistance to therapy with epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer.
Clin Cancer Res
2007
;
13
:
2890
–6.
33
Amann J, Kalyankrishna S, Massion PP, et al. Aberrant epidermal growth factor receptor signaling and enhanced sensitivity to EGFR inhibitors in lung cancer.
Cancer Res
2005
;
65
:
226
–35.
34
Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer.
Nature
2002
;
417
:
949
–54.
35
Rusch VW, Klimstra DS, Venkatraman ES. Molecular markers help characterize neuroendocrine lung tumors.
Ann Thorac Surg
1996
;
62
:
798
–809, discussion 09-10.
36
Wilkinson N, Hasleton PS, Wilkes S, Quigley A. Lack of C-erbB-2 protein expression in pulmonary carcinoid tumours.
J Clin Pathol
1991
;
44
:
343
.
37
Nielsen KB, Sorensen S, Cartegni L, et al. Seemingly neutral polymorphic variants may confer immunity to splicing-inactivating mutations: a synonymous SNP in exon 5 of MCAD protects from deleterious mutations in a flanking exonic splicing enhancer.
Am J Hum Genet
2007
;
80
:
416
–32.
38
Capon F, Allen MH, Ameen M, et al. A synonymous SNP of the corneodesmosin gene leads to increased mRNA stability and demonstrates association with psoriasis across diverse ethnic groups.
Hum Mol Genet
2004
;
13
:
2361
–8.
39
Shepherd FA, Rodrigues Pereira J, Ciuleanu T, et al. Erlotinib in previously treated non-small-cell lung cancer.
N Engl J Med
2005
;
353
:
123
–32.
40
Thatcher N, Chang A, Parikh P, et al. Gefitinib plus best supportive care in previously treated patients with refractory advanced non-small-cell lung cancer: results from a randomised, placebo-controlled, multicentre study (Iressa Survival Evaluation in Lung Cancer).
Lancet
2005
;
366
:
1527
–37.
41
Moon DO, Kim MO, Kang SH, et al. Induction of G2/M arrest, endoreduplication, and apoptosis by actin depolymerization agent pextenotoxin-2 in human leukemia cells, involving activation of ERK and JNK.
Biochem Pharmacol
2008
;
76
:
312
–21.
42
Yan Y, Spieker RS, Kim M, Stoeger SM, Cowan KH. BRCA1-mediated G2/M cell cycle arrest requires ERK1/2 kinase activation.
Oncogene
2005
;
24
:
3285
–96.
43
Zhu CQ, da Cunha Santos G, Ding K, et al. Role of KRAS and EGFR as biomarkers of response to erlotinib in National Cancer Institute of Canada Clinical Trials Group Study BR.21.
J Clin Oncol
2008
;
26
:
4268
–75.
44
Tsao MS, Sakurada A, Cutz JC, et al. Erlotinib in lung cancer - molecular and clinical predictors of outcome.
N Engl J Med
2005
;
353
:
133
–44.
45
Cerfolio RJ, Deschamps C, Allen MS, Trastek VF, Pairolero PC. Mainstem bronchial sleeve resection with pulmonary preservation.
Ann Thorac Surg
1996
;
61
:
1458
–62, discussion 62-3.
46
Rizzardi G, Marulli G, Bortolotti L, Calabrese F, Sartori F, Rea F. Sleeve resections and bronchoplastic procedures in typical central carcinoid tumours.
Thorac Cardiovasc Surg
2008
;
56
:
42
–5.
47
Lucchi M, Melfi F, Ribechini A, et al. Sleeve and wedge parenchyma-sparing bronchial resections in low-grade neoplasms of the bronchial airway.
J Thorac Cardiovasc Surg
2007
;
134
:
373
–7.