Small CEL Carcinoma

37
Small-cell lung cancer Jackman, David M ; Johnson, Bruce E . The Lancet 366.9494 (Oct 15-Oct 21, 2005): 1385-96. Turn on hit highlighting for speaking browsers by selecting the Enter button Hide highlighting Abstract (summary) Translate Abstract Small-cell lung carcinoma is an aggressive form of lung cancer that is strongly associated with cigarette smoking and has a tendency for early dissemination. Increasing evidence has implicated autocrine growth loops, proto-oncogenes, and tumour-suppressor genes in its development. At presentation, the vast majority of patients are symptomatic, and imaging typically reveals a hilar mass. Pathology, in most cases of samples obtained by bronchoscopic biopsy, should be undertaken by pathologists with pulmonary expertise, with the provision of additional tissue for immunohistochemical stains as needed. Staging should aim to identify any evidence of distant disease, by imaging of the chest, upper abdomen, head, and bones as appropriate. Limited-stage disease should be treated with etoposide and cisplatin and concurrent early chest irradiation. All patients who achieve complete remission should be considered for treatment with prophylactic cranial irradiation, owing to the high frequency of brain metastases in this disease. Extensive-stage disease should be managed by combination chemotherapy, with a regimen such as etoposide and cisplatin administered for four to six cycles. Thereafter, patients with progressive or recurrent disease should be treated with additional chemotherapy. For patients who survive long term, careful monitoring for development of a second primary tumour is necessary, with further investigation and treatment as appropriate. Full text Translate Full text Turn on search term navigation

Transcript of Small CEL Carcinoma

Page 1: Small CEL Carcinoma

Small-cell lung cancer

Jackman, David M ; Johnson, Bruce E . The Lancet 366.9494 (Oct 15-Oct 21, 2005): 1385-96. Turn on hit highlighting for speaking browsers by selecting the Enter buttonHide highlighting

Abstract (summary)

Translate Abstract

Small-cell lung carcinoma is an aggressive form of lung cancer that is strongly associated with cigarette smoking and has a tendency for early dissemination. Increasing evidence has implicated autocrine growth loops, proto-oncogenes, and tumour-suppressor genes in its development. At presentation, the vast majority of patients are symptomatic, and imaging typically reveals a hilar mass. Pathology, in most cases of samples obtained by bronchoscopic biopsy, should be undertaken by pathologists with pulmonary expertise, with the provision of additional tissue for immunohistochemical stains as needed. Staging should aim to identify any evidence of distant disease, by imaging of the chest, upper abdomen, head, and bones as appropriate. Limited-stage disease should be treated with etoposide and cisplatin and concurrent early chest irradiation. All patients who achieve complete remission should be considered for treatment with prophylactic cranial irradiation, owing to the high frequency of brain metastases in this disease. Extensive-stage disease should be managed by combination chemotherapy, with a regimen such as etoposide and cisplatin administered for four to six cycles. Thereafter, patients with progressive or recurrent disease should be treated with additional chemotherapy. For patients who survive long term, careful monitoring for development of a second primary tumour is necessary, with further investigation and treatment as appropriate.

Full text

Translate Full text Turn on search term navigation

Headnote

Small-cell lung carcinoma is an aggressive form of lung cancer that is strongly associated with cigarette smoking and has a tendency for early dissemination. Increasing evidence has implicated autocrine growth loops, protooncogenes, and tumour-suppressor genes in its development. At presentation, the vast majority of patients are symptomatic, and imaging typically reveals a hilar mass. Pathology, in most cases of samples obtained by bronchoscopic biopsy, should be undertaken by pathologists with pulmonary expertise, with the provision of additional tissue for immunohistochemical stains as needed. Staging should aim to identify any evidence of distant disease, by imaging of the chest, upper abdomen, head, and bones as appropriate. Limited-stage disease should be treated with etoposide and cisplatin and concurrent early chest irradiation. All patients who achieve complete remission should be considered for treatment with prophylactic cranial irradiation, owing to the high frequency of brain metastases in this disease. Extensive-stage disease should be managed by combination chemotherapy, with a regimen such as etoposide and cisplatin administered for four to six

Page 2: Small CEL Carcinoma

cycles. Thereafter, patients with progressive or recurrent disease should be treated with additional chemotherapy. For patients who survive long term, careful monitoring for development of a second primary tumour is necessary, with further investigation and treatment as appropriate.

Small-cell lung cancer is a distinct clinical and histological entity within the range of lung cancers. Its unique pathological features were first recognised in 1926 by Barnard.1 Watson and Berg2 later described the distinct clinical features of this disease in detail, noting the predominantly central location on chest radiography, the tendency for early dissemination, high initial response rates to chemotherapy, and the high frequency of metastases at autopsy.2 These observations remain accurate 40 years later: small-cell lung cancer is the most aggressive of lung-cancer subtypes. Although most patients' tumours respond to chemotherapy, more than 95% of patients eventually die from the cancer.3 Small-cell carcinoma accounts for 14% of new lung-cancer cases, or about 77 000 of the estimated 550 000 lung cancers in the USA and Europe in 2004.4,5 The vast majority, more than 95%, of these patients develop small-cell lung cancer because of tobacco smoking. The risk of developing the cancer increases both with the number of cigarettes smoked each day and with the duration of smoking.6 This risk begins to decrease with smoking cessation compared with that for individuals who continue to smoke.7

Scientific advances have been made in defining the biology of small-cell lung cancer and have increased our ability to manage this cancer clinically. This review focuses on the underlying molecular changes in the disorder, its presenting signs and symptoms, and its diagnosis, staging, and treatment.

Molecular biology

The specific sequence of genetic alterations leading to small-cell lung cancer is still unclear. However, several important genetic and molecular changes have been noted, including the identification of autocrine growth loops, proto-oncogene activation, and loss or inactivation of tumour-suppressor genes.

Autocrine growth loops

Gastrin-releasing peptide, the human homologue of the amphibian peptide bombesin, has a role in embryonic development and adult repair of bronchial epithelia.8 Small-cell-lung-cancer cell lines produce gastrin-releasing peptide and neuromedin B.9,10 Small-cell lung cancers also express receptors for these two peptides as well as bombesin receptor subtype 3.11 As many as 85% of small-cell-lung-cancer cell lines express at least one of these three receptors.12 The secretion of the peptides by the small-cell lung cancer and subsequent binding to their receptors leads to receptor activation and an autocrine-stimulated growth loop in tumour cells.13 A monoclonal antibody directed against gastrin-releasing peptide causes growth inhibition of small-cell lung cancers both in vitro and in the athymic nude mouse model.13 A patient treated with this monoclonal antibody had a complete response.14 There are no current trials with the monoclonal antibody for patients with small-cell lung cancer because of lack of clinically usable antibody.

The tyrosine kinase receptor, c-kit, is mutated and activated in most gastrointestinal stromal tumours and could have a role in small-cell lung cancer also.15 The ligand stem-cell factor is expressed by and secreted from small-cell lung cancers, binds to its receptor, c-kit, and

Page 3: Small CEL Carcinoma

causes receptor activation.16,17 This process stimulates the growth of the cancer cells, forming an autocrine loop. The c-kit receptor is found in up to 70% of small-cell-lung-cancer lines and tumours.16,18 Moreover, mutations have been identified in the c-kit receptor of 10% of small-cell lung cancers.19 The tyrosine kinase inhibitor of c-kit, imatinib, has been investigated in small-cell lung cancer. In-vitro and invivo models show inhibition of growth when some cell lines are treated with imatinib.20,21 However, two phase II trials of imatinib in small-cell lung cancer have shown no antitumour activity in previously treated or untreated patients.22,23 Owing to this lack of efficacy, no further phase II and III trials of imatinib for patients with small-cell lung cancer are planned.

Proto-oncogenes

The MYC oncogenes (CMYC, NMYC, and LMYC) encode nuclear phosphoproteins that directly regulate gene transcription, increase cell growth, and cause cell-cycle progression.24 The overexpression of MYC proteins in small-cell lung cancer is largely a result of gene amplification.25 Such overexpression leads to more rapid proliferation and loss of terminal differentiation. MYC overexpression occurs in 16-32% of small-cell lung cancers and in 40% of cell lines established from patients whose disease progressed after chemotherapy.26,27 Treatment of small-cell-lung-cancer cell lines with MYC antisense DNA and tretinoin caused down-regulation of CMYC expression and slowed the cell growth rate.28 Conversely, transfection of CMYC into small-cell-lung-cancer cell lines causes an increase in cell proliferation and a change in cellular morphology.29

Tumour-suppressor genes

The loss of genetic material from the short arm of chromosome 3 was documented in small-cell lung cancer more than 20 years ago. Frequent chromosomal deletions of 3p, 4p, 5q, 16q, 13q, and 17p have been found by both loss-of-heterozygosiry analysis30 and comparative genomic hybridisation." The loss of alleles from the short arm of chromosome 3 is the most common deletion of tumour DNA in small-cell lung cancer, found in more than 90% of tumours.30,32 Specific regions of homozygous deletions and loss of heterozygosity have been identified in the region of (3p[14-25]),33 which supports the hypothesis that the 3p arm carries several tumour-suppressor genes (table 1).

Four genes are of particular interest as possible tumour suppressors in small-cell lung cancer. First, the fragile histidine triad gene (FHIT) encodes the enzyme diadenosine triphosphate hydrolase,34 which is thought to have an indirect role in proapoptosis and cell-cycle control. Second, the RAS effector homologue (RASSF1), encodes a microtubule-binding protein. RASSF1 interacts with the microtubules, causing stabilisation and inducing G^sub 1^, G^sub 2^/M arrest.35 The loss of RASSF1 function allows tumour cells to grow more rapidly. The third potential tumour-suppressor gene is the retinoic acid receptor β, through which retinoids exert activity against activator protein 1(36) and induction of apoptosis.17 FUS1, a fourth potential tumour suppressor, is found at 3p21.3.38 Although its specific function has not been identified, myristoylation of the protein seems to be necessary for tumour suppression.39 All four of these genes are commonly deleted or inactivated in small-cell-lung-cancer cell lines.40-42 Moreover, transfection of wild-type FHIT,43 RASSF1,44 or FUS1(45) into tumour cells results in restoration of regulatory controls and growth inhibition. The specific gene (or genes) that functions as a tumour suppressor on chromosome 3 has yet to be conclusively characterised.

Page 4: Small CEL Carcinoma

The TP53 gene encodes a transcription factor that limits cell-cycle progression in the face of genomic damage.31 TP53 mutations are the most common genetic abnormalities in human cancers; they are found in more than 75% of small-cell lung cancers and cell lines.46 These mutations, whether deletions or point mutations,46 ultimately result in deregulation of cell growth controls and increased rates of proliferation.

The retinoblastoma gene RB1 encodes a nuclear phosphoprotein that helps to regulate cell-cycle progression. Dephosphorylation of the protein leads to progression from G1 to S phase. RB1 inactivation is very common in small-cell lung cancer, with inactivating mutations present in more than 90% of tumours.47

The tumour-suppressor gene PTEN encodes a protein with tyrosine and lipid phosphatase activity.48 Through phosphatase activity on phosphatidylinositol triphosphate, PTEN inhibits the phosphatidylinositol-3-kinase/Akt pathway.48 Mutation or deletion of PTEN can lead to increased activity of the pathway, more rapid cell growth, anchorage independence, and reduced apoptosis. PTEN loss of heterozygosity was found often of 11 small-cell-lung-cancer cell lines and tumours (91%),49 but homozygous deletions were detected in only three of 35 cell lines (8%).50

Presentation and diagnosis

Patients with small-cell lung cancer typically present with disseminated disease. Symptoms are related either to bulky, intrathoracic disease or to distant metastases; cough and dyspnoea are the most common findings (table 2).51 Small-cell lung cancer tends to be centrally located, with hilar masses and hilar and mediastinal adenopathy (figure 1). Bronchoscopy shows submucosal endobronchial lesions, which partly explain the low likelihood of finding tumour cells in cytological brushings or sputum samples.

Diagnosis is typically made by histological analysis of a bronchoscopic biopsy sample, or by cytological study of percutaneous or transbronchial fine-needle aspiration samples.52-54 In the WHO 1999 classification system,55 small-cell lung cancer is defined as "a malignant epithelial tumour consisting of small cells with scant cytoplasm, ill-defined cell borders, finely granular nuclear chromatin, and absent or inconspicuous nucleoli. The cells are round, oval, and spindle-shaped, and nuclear molding is prominent. The mitotic count is high" (figure 2). The classification of small-cell lung cancers has been simplified into two categories; more than 90% are typical small-cell lung cancers. The remainder are recognised as combined small-cell carcinoma-combined with any of the histological subtypes of non-small-cell lung cancer. Typically, this combination involves adenocarcinoma, squamous-cell carcinoma, or large-cell carcinoma, though spindle cells or giant cells are observed.55 Review of SEER registry lung-cancer cases showed 94% consistency between registry-reported histological diagnoses and independent reference pathologists.56 However, because small-cell samples can be vulnerable to crush artifact from biopsy foreceps and distortion on fine-needle aspiration, differentiation from non-small-cell lung cancer can be difficult in some cases. Though immunohistochemical analysis is not generally necessary for a diagnosis, these stains can be helpful in difficult cases. Epithelial markers such as cytokeratins are ubiquitous in these tumours and help to distinguish them from lymphomas. Furthermore, chromogranin, synaptophysin, and CD56, all neuroendocrine markers, can be useful in differentiating small-cell from non-small-cell lung cancer.

Paraneoplastic syndromes

Page 5: Small CEL Carcinoma

Small-cell lung carcinoma has long been associated with paraneoplastic syndromes. The endocrine paraneoplastic disorders are characterised by ectopic production of peptide hormones, and the neurological complications are related to antibody-mediated damage to the central nervous system (table 3).

Hyponatraemia can be found in up to 15% of patients with small-cell lung cancer at presentation.57 In many of these patients, ectopic production of antidiuretic hormone, also known as arginine vasopressin, leads to reduced renal clearance of free water with a subsequent decrease in serum sodium concentrations and the syndrome of inappropriate antidiuretic hormone.58-60 Atrial natriuretic peptide can also be produced ectopically by small-cell lung cancers and might have a role in hyponatraemia of malignancy.58,60,61 The syndrome is characterised by decreasing serum sodium concentrations, which can affect mental status and lead to seizures. Laboratory abnormalities include serum hypo-osmolality, urine osmolality exceeding 100 mmol/kg, and a normal to high urine sodium concentration (>40 mmol/L). The condition improves when the underlying tumour responds to effective treatment. However, other measures might be needed to improve symptomatic hyponatraemia. These measures include saline hydration, loop diuretics, and restriction of free water.62 If needed, demeclocycline can be used to antagonise the effect of ectopic arginine vasopressin at the renal collecting tubule.63

Cushing's syndrome is present in 2-5% of patients with small-cell lung cancer at the time of presentation.64 Common symptoms and signs of the disorder include central obesity, facial plethora, hirsutism, acne, weakness, glucose intolerance, and hypertension. The syndrome is caused by ectopic production of corticotropin by tumour cells. Although more than 50% of small-cell lung cancers produce immunoreactive corticotropin precursors, only 2-5% process the precursor into a biologically active form that elicits the symptoms, signs, and laboratory abnormalities of Cushing's syndrome.65 As well as treatment of the underlying tumour, management of ectopic corticotropin production might necessitate reduction of cortisol synthesis by an adrenal enzyme inhibitor such as ketoconazole.66

Lambert-Eaton myasthenic syndrome is caused by autoantibodies directed against P/Q-type voltage-gated calcium channels.67 Decreased acetycholine release at motor nerve terminals leads to the classic proximal muscle weakness.68 Patients can also experience autonomic symptoms such as dry mouth, constipation, and erectile failure. The diagnosis can be made by autoantibody detection on radioimmunoprecipitation assay.69 Electromyelography characteristically shows increasing amplitude of muscle action potentials with high-frequency stimulation.70

Treatment directed at the underlying small-cell lung cancer generally results in neurological improvement in patients with this syndrome.71 In addition, many of these patients are given 3,4-diaminopyridine. Small studies with this drug have shown significantly increased muscle strength in treated patients;72 as many as 85% of patients treated with 3,4-diaminopyridine experience neurological improvement.71 For patients with severe weakness, intravenous immunoglobulin or plasmapheresis can provide short-term benefit.73,74 For those who do not respond to 3,4-diaminopyridine, prednisone alone or with azathioprine or ciclosporin can help to improve muscle strength and provide long-term control.71

Staging

Page 6: Small CEL Carcinoma

Patients with small-cell lung cancer rarely present with disease that is sufficiently localised for surgical resection to be possible, so the TNM staging system is generally not used. Instead, staging typically uses the Veterans Administration Lung Study Group system, which classifies patients as having either limited-stage or extensive-stage disease.75 The definition of limited-stage disease varies but is generally designed to include patients whose disease is limited to one hemithorax, with hilar and mediastinal nodes that can be encompassed within one tolerable radiotherapy portal. Extensive-stage disease is any disease beyond those boundaries.

Most (60-70%) patients present with clinically obvious extensive-stage disease. These patients have median survival of 7-12 months and the proportion alive at 5 years is 2%; among patients with limited-stage disease, median survival is about 23 months and the proportion surviving 5 years 12-17%.3,76 Given the prognostic and therapeutic implications, the main goal of staging is to identify sites of disease outside of a potential radiotherapy portal. The ensuing assessment is prompted by the patient's presenting symptoms, as well as by the most likely sites of metastatic involvement (table 4).77

In addition to a history and physical examination, patients typically undergo routine haematological and serum chemistry tests. CT of the chest should be done. Chest imaging allows the extent of intrathoracic involvement to be assessed, including the presence of pleural effusion, lobar collapse, hilar and mediastinal adenopathy, and contralateral parenchymal disease. Fibreoptic bronchoscopy can be used to characterise the disease in the airways and allow the initial diagnosis. The findings are abnormal in more than 90% of cases.54,78

Clinical signs or symptoms should indicate appropriate additional staging. Any clinical evidence of neurological abnormality should prompt MRI or CT of the brain and MRI of the spinal cord, because metastases in the central nervous system will be identified in 80-90% of patients.79,80 A radionuclide bone scan should be done for patients with bony pain. Owing to the frequency of liver metastases (table 4), the chest CT should be extended caudally to include the liver and adrenal glands, if not done already, if results of liver-function tests are abnormal. Follow-up biopsy of hepatic or adrenal lesions should be done if these organs are the only potential site of metastatic disease. For a patient with abnormal complete blood count or peripheral blood smear that cannot be otherwise explained, bone-marrow biopsy should be considered, particularly if there is no other site of extensive-stage disease.

The search for extrathoracic disease can potentially become both exhausting and expensive. In patients for whom locoregional therapy for apparent limited-stage disease is being considered, the assessment should be planned because identification of distant disease could spare patients unnecessary chest radiotherapy or surgery. An assessment should include CT of the liver and adrenal glands, MRI or CT of the brain, and a bone scan. Except in clinical trials, which have more rigorous staging requirements, further staging studies can be omitted if a patient is found to have an unequivocal site of metastatic involvement that confirms extensive-stage disease. The cost of the staging assessment can be reduced by about 40% if it is curtailed in this way.80

The role of positron emission tomography with fluorodeoxyglucose in small-cell lung cancer is still being investigated. Small-cell lung cancers avidly take up fluorodeoxyglucose, and in a small prospective trial this imaging modality could identify metastatic lesions in patients who were thought to have limited-stage disease by standard staging methods.81 However, at

Page 7: Small CEL Carcinoma

this time, this approach for small-cell lung cancer has not yet become an approved indication for reimbursement in the USA.

Prognostic factors

Several pretreatment characteristics in patients with small-cell lung cancer have been associated with meaningful differences in survival that have also been noted in patients with non-small-cell lung cancer (panel).82-86 The identification of such factors allows physicians and investigators to compare different populations of patients and to interpret the contribution of treatment to differences in survival between different groups. For example, since women with small-cell lung cancer generally outlive male patients,82,83 and because more women are developing lung cancer4 and taking part in clinical trials,83 recent improvements in overall survival for patients with small-cell lung cancer could be due partly to the greater percentage of women in trials rather than a treatment effect, especially in limited-stage disease.

Treatment

Combination chemotherapy remains the cornerstone of treatment for both limited-stage and extensive-stage small-cell lung cancer. In general, the administration of etoposide and cisplatin plus chest radiotherapy for patients with good performance status and limited-stage disease should produce a complete-response rate of 80% or higher, median survival in excess of 17 months, and 5-year cancer-free survival of 12-25%.76,87 Patients with extensive-stage disease given combination chemotherapy should have a complete-response rate of more than 20% and median survival longer than 7 months; in addition, 2% of patients will be alive and without cancer at 5 years.3 The death rate from complications of therapy should be less than 5%.

Surgical resection

Surgery in small-cell lung cancer was largely abandoned in the 1970s because the proportion of patients alive at 5 years was less than 5%.88,89 Sequential phase II studies and case-series reports showed that among patients who underwent surgical resection before a histological diagnosis of small-cell lung cancer and who were given chemotherapy, chest radiotherapy, or both, 5-year survival was 35-40%.90-93 The potential benefits of resection in small-cell lung cancer are predominantly seen in patients with TNM stage I disease. At this point, careful radiological and mediastinal staging should be used to select candidates for surgery. For patients with disease of clinical stage I with peripheral lesions and no hilar or mediastinal nodal involvement, surgical resection is recommended if the patient can tolerate the surgical procedure. Patients who undergo resection for small-cell lung cancer should still receive adjuvant postoperative combination chemotherapy. Patients with hilar or mediastinal involvement should be assessed for combined modality therapy (chest radiotherapy and chemotherapy).

Treatment of limited-stage disease

The treatment of limited-stage disease has evolved over the past few decades. After attempts at surgical resection were abandoned in the 1970s, attention turned to other therapeutic approaches. Small-cell lung cancer had been shown to respond to treatment with alkylating agents but locoregional failure occurred subsequently in up to 80% of cases.94 Attention

Page 8: Small CEL Carcinoma

turned to chest radiotherapy for patients with disease confined to the lung and mediastinum. Chest radiotherapy as a single modality led to response rates of about 75%,95 effectively controlling disease in the chest and even achieving cure in about 5% of patients with limited-stage disease.94 Clinical efforts have been focused on improving and refining the chemotherapy regimen, as well as combining these regimens with chest radiotherapy.

The initial chemotherapy regimens were primarily based on cyclophosphamide, and the combination of this drug with doxorubicin and vincristine became widely used by the late 1970s. However, this regimen has been replaced with a combination of etoposide and cisplatin. In a large randomised trial, this cisplatin-based regimen conferred median survival of 14.5 months in limited-stage disease compared with 9.7 months for patients assigned cyclophosphamide, epirubicin, and vincristine.96 The etoposide and cisplatin regimen is also associated with less haematological toxicity, and it is more easily combined with chest radiotherapy.87,96,97

The etoposide and cisplatin regimen is typically administered every 3 weeks for four to six cycles. Etoposide is given intravenously at 80-120 mg/m^sup 2^ on days 1, 2, and 3, and cisplatin is administered intravenously at 60-90 mg/m^sup 2^ for 1-3 days starting on day 1.87,97-99 Attempts to improve survival by continuing therapy for longer than six cycles have been largely unsuccessful, with greater toxicity but no survival benefit for patients with limited-stage disease in eight of nine studies.100-103

A meta-analyses of 13 prospective, randomised trials for patients with limited-stage small-cell lung cancer that compared chemotherapy alone or with chest irradiation showed that 5% more patients were alive at 2 years and 3 years in the group assigned chemotherapy and radiotherapy.104 Twice-daily chest radiotherapy to a dose of 45 Gy plus etoposide and cisplatin is favoured over once-daily chest radiotherapy to 45 Gy on the basis of a 10% absolute survival benefit at 5 years.87 Whether higher doses of once-daily chest radiotherapy will be as effective as twice-daily chest radiotherapy and be easier to administer is currently being investigated.

Chest radiotherapy given concurrently with the first or second cycle of chemotherapy has proven superior to sequential radiotherapy.97,105 Therefore, the current recommendation for limited-stage disease at this time is administration of four to six cycles of chemotherapy with etoposide and cisplatin, combined with twice-daily radiation that starts in cycle 1 or 2.

Treatment of extensive-stage disease

Combination chemotherapy remains the focus of treatment for patients with extensive-stage small-cell lung cancer. Radiotherapy in extensive-stage disease is reserved for the prevention106 or treatment107,108 of brain metastases; for symptomatic bone metastases; for cord compression; or for palliative treatment of lobar collapse or superior vena cava syndrome in patients who have not responded to chemotherapy and have not received radiotherapy to those sites. Routine use of chest radiotherapy in extensive-stage disease does not prolong survival.

The combination of cisplatin and etoposide remains the most widely used regimen. The median survival for patients with extensive-stage disease remains at about 7-9 months, and only about 2% of patients survive for 5 years.3,84,96 Other strategies have been studied in

Page 9: Small CEL Carcinoma

attempts to improve on these results. Changes in the chemotherapy regimen, dose intensity, dose density, schedule, and duration have been investigated.

In a study in Japan, etoposide plus cisplatin was compared with irinotecan plus cisplatin; there was a significant survival advantage with irinotecan in patients with extensive-stage small-cell lung cancer (median survival 12.8 vs 9.4 months; p=0.002).109 The treatment benefit of topoisomerase inhibitors was not seen in subsequent trials in the USA and Europe. In the US trial that compared irinotecan plus cisplatin with etoposide plus cisplatin, there was no significant difference in response rate (48% vs 44%), median time to progression (4.1 vs 4.6 months), median survival (9 vs 10 months), or 1-year survival (35% vs 37%). Patients assigned irinotecan plus cisplatin had less myelosuppression but more diarrhoea than those assigned etoposide plus cisplatin.110 A trial comparing oral topotecan and cisplatin with intravenous etoposide plus cisplatin in a European population also did not show improved efficacy with the topoisomerase inhibitor. The median survival and 1-year survival were similar in both study groups, and time to progression favoured etoposide plus cisplatin.111

Two large randomised trials have investigated the addition of further agents to the standard regimen of etoposide plus cisplatin. In one study, patients were assigned etoposide plus cisplatin with or without ifosfamide.112 The second study compared etoposide plus cisplatin with etoposide, cisplatin, cyclophosphamide, and epirubicin.113 Both trials showed slight survival benefits favouring the regimen with three or four drugs, but at the cost of greater toxicity.

Two trials have specifically investigated the benefit of higher doses of chemotherapy with platinum-based regimens. One, a study of 90 patients with extensive-stage disease, showed no survival difference between the standard-dose and high-dose groups.98 In the other, 105 patients with limited-stage small-cell lung cancer were enrolled, with 2-year survival of 26% in the standard-dose group and 43% in the high-dose group.114 Although two phase II trials on high-dose therapy with autologous bone-marrow transplantation had encouraging results,115,116 the only completed phase III study of high-dose therapy with bone-marrow transplantation found no difference in median survival.117 At this time, higher doses of chemotherapy are not routinely used and remain under investigation.

Etoposide is typically administered intravenously for 3-5 days of a 21-day cycle. It has greater efficacy when given on a 3-5 day schedule than when given on a single day.118,119 Attempts to alter this schedule have largely been unsuccessful. Patients with extensive-stage disease who were treated with 21 days of etoposide did not have any survival advantage over those receiving the drug for 3-5 days.120

Changes in the schedule of the chemotherapy cycle have also been investigated. The use of weekly chemotherapy resulted in greater toxicity with no survival advantage.121,122 Three trials in which the interval between chemotherapy cycles was shortened, with or without growth-factor support, found no consistent benefit for the patients treated.123-125

To avoid the nausea, ototoxic, and nephrotoxic effects of cisplatin, regimens substituting carboplatin have been attempted in both single-group126 and randomised trials.127 To date, the substitution has not led to any significant difference in survival. However, because there is much more information and experience with cisplatin in this disease, especially in combination with chest radiotherapy, the cisplatin and etoposide regimen remains the standard of care.

Page 10: Small CEL Carcinoma

The duration of chemotherapy has been studied in both limited-stage and extensive-stage small-cell lung cancer. More than ten trials have investigated addition of further chemotherapy after the four to six cycles of induction chemotherapy. This maintenance treatment has included both cytotoxic chemotherapy and targeted agents. Maintenance therapy was associated with greater toxicity without a survival benefit.100-103,128,129

Although alternating chemotherapy regimens were previously of interest,128,130 this strategy has not been superior to a single combination of platinum-based regimens in later trials.131,132 Alternating therapy is not widely used and is not a major focus of current studies.

Various novel targeted agents have been investigated in small-cell lung cancer. These include inhibitors of c-kit,22,23 matrix metalloproteinases,133,134 farnesyl transferase,135 proteosomes,136 and mTOR,137 as well as antiangiogenic agents.138,139 The results of these trials have been largely negative, though further investigation is warranted, particularly of antiangiogenesis and mTOR inhibition.

Additional treatment issues

Prophylactic cranial irradiation

Patients whose small-cell lung cancer has been successfully treated nonetheless have a 50-67% risk of developing metastases in the central nervous system.140,141 Therefore, in patients who have had a complete response to chemotherapy, cranial irradiation is used prophylactically. The radiation is typically administered in doses of 24-46 Gy in eight to 15 fractions. A meta-analysis of randomised trials of prophylactic cranial irradiation showed that the intervention reduced the risk of brain metastases by 45%; moreover, patients assigned prophylactic cranial irradiation were 5% more likely to be alive at 3 years than those not assigned the intervention.106 Though concerns have been raised about possible intellectual impairment with prophylactic cranial irradiation, studies to date have not proven that the intervention is the sole cause. Neuropsychological testing showed most impairment was detectable before administration of prophylactic cranial irradiation. In addition, patients randomly assigned prophylactic cranial irradiation or observation showed no detectable difference in post-treatment neurological function when tested a year later.141 Patients with limited-stage small-cell lung cancer who achieve complete remission should receive prophylactic cranial irradiation. Patients with extensive-stage disease who have complete responses to chemotherapy who are given prophylactic cranial irradiation seem to have longer survival than patients who are not, but the evidence so far is less conclusive.140,141 We recommend that patients with extensive-stage disease and good prognostic factors be offered prophylactic cranial irradiation if they achieve complete remission.

Treatment in patients with poor performance status

Single-agent chemotherapy in elderly patients and those with poor performance status was initially intended to provide a more tolerable treatment option.142 However, in comparisons of single-agent oral etoposide with combination chemotherapy regimens, patients assigned combination regimens not only lived longer but also had fewer side-effects.143,144 Therefore, combination chemotherapy remains the standard treatment in this setting. When the tolerability of standard dose etoposide and cisplatin remains a concern, options include

Page 11: Small CEL Carcinoma

dose reduction, substitution of carboplatin for cisplatin,145,146 or a combination of low-dose cisplatin, doxorubicin, vincristine, and etoposide.147

Progressive or relapsing disease

Most patients with small-cell lung cancer relapse within a year of starting treatment. The likelihood of response to subsequent therapy can be predicted on the basis of the response to previous therapy and the duration of drug-free interval. Patients who did not respond to previous therapy or who relapsed within 3 months of completing therapy are judged refractory, whereas those who had responded to previous treatment and relapsed 3 months or longer after completing such treatment are deemed sensitive. For patients with sensitive disease, second-line regimens, such as single-agent topotecan or the combination of cyclophosphamide, doxorubicin, and vincristine yield response rates of about 20%, with median survival times of about 25 weeks.148 For patients with refractory disease, response rates are less good; single-agent topotecan achieves responses in about 8% in this setting.149,150 However, combination chemotherapy based on irinotecan or topotecan for refractory or relapsed small-cell lung cancer has shown more promise. A study of topotecan with cisplatin in 110 patients with relapsed or refractory disease found response rates of 29.4% in sensitive disease and 23.8% in refractory disease.151 Several smaller, single-group studies of combination chemotherapy with regimens based on irinotecan, topotecan, or paclitaxel have documented response rates of 29-80%.152,153 The results of larger randomised clinical trials with these newer regimens are still needed to identify the best treatment for relapsed disease.

Long-term survivors

With the widespread use of combination chemotherapy, a small but important number of patients will achieve long-term survival. After 2 years, the risk of death from the initial disease begins to decrease; by 3 years, the daily hazard drops by a factor often.154 Nonetheless, the hazard ratio remains seven to ten times that of the general population, owing partly to the development of second primary tumours.155-157 The risk of a second primary is 2-10% per patient per year, a risk ten times that of adult male smokers who have not developed lung cancer.156,157 Therefore, these patients are good candidates for participation in screening and chemoprevention trials. Moreover, any new lung masses should be promptly investigated and biopsy samples taken, because they might be surgically resectable second primary tumours.

Further information

Clinical guidelines for the treatment of small-cell lung cancer have been established by several national and international committees. For further reference, the recommendations of the National Comprehensive Cancer Network158 and the European Society for Medical Oncology can be reviewed online.159

Conflict of interest statement

Bruce E Johnson has received an unrestricted gift from Genentech. David M Jackman declares that he has no conflict of interest.

Sidebar

Page 12: Small CEL Carcinoma

Search strategy and selection criteria

This seminar is based mainly on papers published during the past 5 years, although some classic papers have been cited. The search strategy selected articles from MEDLINE by use of the PubMed system. The key words used were "carcinoma, small cell lung" cross-referenced with key words such as "epidemiology", "pathology", "cytogenetics", "molecular biology", "diagnosis", "antineoplastic agents", "radiotherapy", and "surgery". Only papers with an abstract in English were considered.

References

References

1 Barnard W. The nature of the 'oat-celled sarcoma' of the mediastinum. J Pathol 1926; 29: 241-44.

2 Watson WL, Berg JW. Oat cell lung cancer. Cancer 1962; 15: 759-68.

3 Chute JP, Chen T, Feigal E, Simon R, Johnson BE. Twenty years of phase III trials for patients with extensive-stage small-cell lung cancer: perceptible progress. J Clin Oncol 1999; 17: 1794-801.

4 Jemal A, Tiwari RC, Murray T, et al. Cancer statistics, 2004. CA Cancer J Clin 2004; 54: 8-29.

5 Tyczynski JE, Bray F, Parkin DM. Lung cancer in Europe in 2000: epidemiology, prevention, and early detection. Lancet Oncol 2003; 4: 45-55.

6 Brownson RC, Chang JC, Davis JR. Gender and histologic type variations in smoking-related risk of lung cancer. Epidemiology 1992; 3: 61-64.

7 Khuder SA, Mutgi AB. Effect of smoking cessation on major histologic types of lung cancer. Chest 2001; 120: 1577-83.

8 Spindel ER, Sunday ME, Hofler H, Wolfe HJ, Habener JF, Chin WW. Transient elevation of messenger RNA encoding gastrin-releasing peptide, a putative pulmonary growth factor in human fetal lung. J Clin Invest 1987; 80: 1172-79.

9 Cardona C, Rabbitts PH, Spindel ER, et al. Production of neuromedin B and neuromedin B gene expression in human lung tumor cell lines. Cancer Res 1991; 51: 5205-11.

10 Sausville EA, Lebacq-Verheyden AM, Spindel ER, Cuttitta F, Gazdar AF, Battey JF. Expression of the gastrin-releasing peptide gene in human small cell lung cancer. Evidence for alternative processing resulting in three distinct mRNAs. J Biol Chem 1986; 261: 2451-57.

11 Corjay MH, Dobrzanski DJ, Way JM, et al. Two distinct bombesin receptor subtypes are expressed and functional in human lung carcinoma cells. J Biol Chem 1991; 266: 18771-79.

Page 13: Small CEL Carcinoma

12 Toi-Scott M, Jones CL, Kane MA. Clinical correlates of bombesin-like peptide receptor subtype expression in human lung cancer cells. Lung Cancer 1996; 15: 341-54.

13 Cuttitta F, Carney DN, Mulshine J, et al. Bombesin-like peptides can function as autocrine growth factors in human small-cell lung cancer. Nature 1985; 316: 823-26.

14 Chaudhry A, Carrasquillo JA, Avis IL, et al. Phase I and imaging trial of a monoclonal antibody directed against gastrin-releasing peptide in patients with lung cancer. Clin Cancer Res 1999; 5: 3385-93.

15 Heinrich MC, Corless CL, Demetri GD, et al. Kinase mutations and imatinib response in patients with metastatic gastrointestinal stromal tumor. J Clin Oncol 2003; 21: 4342-49.

16 Hibi K, Takahashi T, Sekido Y, et al. Coexpression of the stem cell factor and the c-kit genes in small-cell lung cancer. Oncogene 1991; 6: 2291-96.

17 Sekido Y, Obata Y, Ueda R, et al. Preferential expression of c-kit protooncogene transcripts in small cell lung cancer. Cancer Res 1991; 51: 2416-19.

18 Rygaard K, Nakamura T, Spang-Thomsen M. Expression of the proto-oncogenes c-met and c-kit and their ligands, hepatocyte growth factor/scatter factor and stem cell factor, in SCLC cell lines and xenografts. Br J Cancer 1993; 67: 37-46.

19 Boldrini L, Ursino S, Gisfredi S, et al. Expression and mutational status of c-kit in small-cell lung cancer: prognostic relevance. Clin Cancer Res 2004; 10: 4101-08.

20 Wang WL, Healy ME, Sattler M, et al. Growth inhibition and modulation of kinase pathways of small cell lung cancer cell lines by the novel tyrosine kinase inhibitor STI 571. Oncogene 2000; 19: 3521-28.

21 Wolff NC, Randle DE, Egorin MJ, Minna JD, Ilaria RL Jr. Imatinib mesylate efficiently achieves therapeutic intratumor concentrations in vivo but has limited activity in a xenograft model of small cell lung cancer. Clin Cancer Res 2004; 10: 3528-34.

22 Johnson BE, Fischer T, Fischer B, et al. Phase II study of imatinib in patients with small cell lung cancer. Clin Cancer Res 2003; 9: 5880-87.

23 Dy GK, Miller AA, Mandrekar S, et al. A phase II NCCTG/CALGB trial of imatinib (STI571) in patients with c-kit-expressing relapsed small cell lung cancer (SCLC). Proc Am Clin Soc Oncol 2005: LBA 7048.

24 Schmidt EV. The role of c-myc in regulation of translation initiation. Oncogene 2004; 23: 3217-21.

25 Prins J, De Vries EG, Mulder NH. The myc family of oncogenes and their presence and importance in small-cell lung carcinoma and other tumour types. Anticancer Res 1993; 13: 1373-85.

Page 14: Small CEL Carcinoma

26 Johnson BE, Makuch RW, Simmons AD, Gazdar AF, Burch D, Cashell AW. myc family DNA amplification in small cell lung cancer patients' tumors and corresponding cell lines. Cancer Res 1988; 48: 5163-66.

27 Brennan J, O'Connor T, Makuch RW, et al. myc family DNA amplification in 107 tumors and tumor cell lines from patients with small cell lung cancer treated with different combination chemotherapy regimens. Cancer Res 1991; 51: 1708-12.

28 Akie K, Dosaka-Akita H, Murakami A, Kawakami Y. A combination treatment of c-myc antisense DNA with all-trans-retinoic acid inhibits cell proliferation by downregulating c-myc expression in small cell lung cancer. Antisense Nucleic Acid Drug Dev 2000; 10: 243-49.

29 Johnson BE, Battey J, Linnoila I, et al. Changes in the phenotype of human small cell lung cancer cell lines after transfection and expression of the c-myc proto-oncogene. J Clin Invest 1986; 78: 525-32.

30 Lindblad-Toh K, Tanenbaum DM, Daly MJ, et al. Loss-of-heterozygosity analysis of small-cell lung carcinomas using single-nucleotide polymorphism arrays. Nat Biotechnol 2000; 18: 1001-05.

31 Levine AJ, Momand J, Finlay CA. The p53 tumour suppressor gene. Nature 1991; 351: 453-56.

32 Naylor SL, Johnson BE, Minna JD, Sakaguchi AY. Loss of heterozygosity of chromosome 3p markers in small-cell lung cancer. Nature 1987; 329: 451-54.

33 Hibi K, Takahashi T, Yamakawa K, et al. Three distinct regions involved in 3p deletion in human lung cancer. Oncogene 1992; 7: 445-49.

34 Pekarsky Y, Palamarchuk A, Huebner K, Croce CM. FHIT as tumor suppressor: mechanisms and therapeutic opportunities. Cancer Biol Ther 2002; 1: 232-36.

35 Rong R, Jin W, Zhang J, Saeed Sheikh M, Huang Y. Tumor suppressor RASSF1A is a microtubule-binding protein that stabilizes microtubules and induces G/M arrest. Oncogene 2004; 23: 8216-30.

36 Lin F, Xiao D, Kolluri SK, Zhang X. Unique anti-activator protein-1 activity of retinoic acid receptor beta. Cancer Res 2000; 60: 3271-80.

37 Liu Y, Lee MO, Wang HG, et al. Retinoic acid receptor beta mediates the growth-inhibitory effect of retinoic acid by promoting apoptosis in human breast cancer cells. Mol Cell Biol 1996; 16: 1138-49.

38 Kondo M, Ji L, Kamibayashi C, et al. Overexpression of candidate tumor suppressor gene FUS1 isolated from the 3p21.3 homozygous deletion region leads to G1 arrest and growth inhibition of lung cancer cells. Oncogene 2001; 20: 6258-62.

39 Uno F, Sasaki J, Nishizaki M, et al. Myristoylation of the fus1 protein is required for tumor suppression in human lung cancer cells. Cancer Res 2004; 64: 2969-76.

Page 15: Small CEL Carcinoma

40 Sozzi G, Veronese ML, Negrini M, et al. The FHIT gene 3p14.2 is abnormal in lung cancer. Cell 1996; 85: 17-26.

41 Burbee DG, Forgacs E, Zochbauer-Muller S, et al. Epigenetic inactivation of RASSF1A in lung and breast cancers and malignant phenotype suppression. J Natl Cancer Inst 2001; 93: 691-99.

42 Virmani AK, Rathi A, Zochbauer-Muller S, et al. Promoter methylation and silencing of the retinoic acid receptor-beta gene in lung carcinomas. J Natl Cancer Inst 2000; 92: 1303-07.

43 Siprashvili Z, Sozzi G, Barnes LD, et al. Replacement of Fhit in cancer cells suppresses tumorigenicity. Proc Natl Acad Sci USA 1997; 94: 13771-76.

44 Dammann R, Li C, Yoon JH, Chin PL, Bates S, Pfeifer GP. Epigenetic inactivation of a RAS association domain family protein from the lung tumour suppressor locus 3p21.3. Nat Genet 2000; 25: 315-19.

45 Ito I, Ji L, Tanaka F, et al. Liposomal vector mediated delivery of the 3p FUS1 gene demonstrates potent antitumor activity against human lung cancer in vivo. Cancer Gene Ther 2004; 11: 733-39.

46 Olivier M, Eeles R, Hollstein M, Khan MA, Harris CC, Hainaut P. The IARC TP53 database: new online mutation analysis and recommendations to users. Hum Mutat 2002; 19: 607-14.

47 Kaye FJ. RB and cyclin dependent kinase pathways: defining a distinction between RB and p16 loss in lung cancer. Oncogene 2002; 21: 6908-14.

48 Cantley LC, Neel BG. New insights into tumor suppression: PTEN suppresses tumor formation by restraining the phosphoinositide 3-kinase/AKT pathway. Proc Natl Acad Sci USA 1999; 96: 4240-45.

49 Virmani AK, Fong KM, Kodagoda D, et al. Allelotyping demonstrates common and distinct patterns of chromosomal loss in human lung cancer types. Genes Chromosomes Cancer 1998; 21: 308-19.

50 Forgacs E, Biesterveld EJ, Sekido Y, et al. Mutation analysis of the PTEN/MMAC1 gene in lung cancer. Oncogene 1998; 17: 1557-65.

51 Chute CG, Greenberg ER, Baron J, Korson R, Baker J, Yates J. Presenting conditions of 1539 population-based lung cancer patients by cell type and stage in New Hampshire and Vermont. Cancer 1985; 56: 2107-11.

52 Chin R Jr, Cappellari JO, McCain TW, Case LD, Haponik EF. Increasing use of bronchoscopic needle aspiration to diagnose small cell lung cancer. Mayo Clin Proc 2000; 75: 796-801.

53 Delgado PI, Jorda M, Ganjei-Azar P. Small cell carcinoma versus other lung malignancies: diagnosis by fine-needle aspiration cytology. Cancer 2000; 90: 279-85.

Page 16: Small CEL Carcinoma

54 Tondini M, Rizzi A. Small-cell lung cancer: importance of fiberoptic bronchoscopy in the evaluation of complete remission. Tumori 1989; 75: 266-68.

55 Travis WD, Colby TV, Corrin B, Shimosato Y, Brambilla E. Epithelial rumours. In: Travis WD CT, Corrin B, et al. eds. Histological typing of lung and pleural tumours, 3rd edn. Washington DC: Armed Forces Institute of Pathology, 1999: 25-47.

56 Field RW, Smith BJ, Platz CE, et al. Lung cancer histologic type in the surveillance, epidemiology, and end results registry versus independent review. J Natl Cancer Inst 2004; 96: 1105-07.

57 Campling BG, Sarda IR, Baer KA, et al. Secretion of atrial natriuretic peptide and vasopressin by small cell lung cancer. Cancer 1995; 75: 2442-51.

58 Johnson BE, Chute JP, Rushin J, et al. A prospective study of patients with lung cancer and hyponatremia of malignancy. Am J Respir Crit Care Med 1997; 156: 1669-78.

59 George JM, Capen CC, Phillips AS. Biosynthesis of vasopressin in vitro and ultrastructure of a bronchogenic carcinoma: patient with the syndrome of inappropriate secretion of antidiuretic hormone. J Clin Invest 1972; 51: 141-48.

60 Gross AJ, Steinberg SM, Reilly JG, et al. Atrial natriuretic factor and arginine vasopressin production in tumor cell lines from patients with lung cancer and their relationship to serum sodium. Cancer Res 1993; 53: 67-74.

61 Kamoi K, Ebe T, Kobayashi O, et al. Atrial natriuretic peptide in patients with the syndrome of inappropriate antidiuretic hormone secretion and with diabetes insipidus. J Clin Endocrinol Metab 1990; 70: 1385-90.

62 Wong LL, Verbalis JG. Systemic diseases associated with disorders of water homeostasis. Endocrinol Metab Clin North Am 2002; 31: 121-40.

63 Forrest JN Jr, Cox M, Hong C, Morrison G, Bia M, Singer I. Superiority of demeclocycline over lithium in the treatment of chronic syndrome of inappropriate secretion of antidiuretic hormone. N Engl J Med 1978; 298: 173-77.

64 Shepherd FA, Laskey J, Evans WK, Goss PE, Johansen E, Khamsi F. Cushing's syndrome associated with ectopic corticotropin production and small-cell lung cancer. J Clin Oncol 1992; 10: 21-27.

65 Stewart PM, Gibson S, Crosby SR, et al. ACTH precursors characterize the ectopic ACTH syndrome. Clin Endocrinol (Oxf) 1994; 40: 199-204.

66 Winquist EW, Laskey J, Crump M, Khamsi F, Shepherd FA. Ketoconazole in the management of paraneoplastic Cushing's syndrome secondary to ectopic adrenocorticotropin production. J Clin Oncol 1995; 13: 157-64.

67 Sher E, Gotti C, Canal N, et al. Specificity of calcium channel autoantibodies in Lambert-Eaton myasthenic syndrome. Lancet 1989; 2: 640-43.

Page 17: Small CEL Carcinoma

68 Lambert EH, Elmqvist D. Quantal components of end-plate potentials in the myasthenic syndrome. Ann N Y Acad Sci 1971; 183: 183-99.

69 Voltz R, Carpentier AF, Rosenfeld MR, Posner JB, Dalmau J. P/Q-type voltage-gated calcium channel antibodies in paraneoplastic disorders of the central nervous system. Muscle Nerve 1999; 22: 119-22.

70 Tim RW, Massey JM, Sanders DB. Lambert-Eaton myasthenic syndrome (LEMS): clinical and electrodiagnostic features and response to therapy in 59 patients. Ann N Y Acad Sci 1998; 841: 823-26.

71 Chalk CH, Murray NM, Newsom-Davis J, O'Neill JH, Spiro SG. Response of the Lambert-Eaton myasthenic syndrome to treatment of associated small-cell lung carcinoma. Neurology 1990; 40: 1552-56.

72 McEvoy KM, Windebank AJ, Daube JR, Low PA. 3,4-Diaminopyridine in the treatment of Lambert-Eaton myasthenic syndrome. N Engl J Med 1989; 321: 1567-71.

73 Sanders DB. Lambert-eaton myasthenic syndrome: diagnosis and treatment. Ann N Y Acad Sci 2003; 998: 500-08.

74 Motomura M, Hamasaki S, Nakane S, Fukuda T, Nakao YK. Apheresis treatment in Lambert-Eaton myasthenic syndrome. Ther Apher 2000; 4: 287-90.

75 Zelen M. Keynote address on biostatistics and data retrieval. Cancer Chemother Rep 3 1973; 4: 31-42.

76 Janne PA, Freidlin B, Saxman S, et al. Twenty-five years of clinical research for patients with limited-stage small cell lung carcinoma in North America. Cancer 2002; 95: 1528-38.

77 Argiris A, Murren JR. Staging and clinical prognostic factors for small-cell lung cancer. Cancer J 2001; 7: 437-47.

78 Jones AM, Hanson IM, Armstrong GR, O'Driscoll BR. Value and accuracy of cytology in addition to histology in the diagnosis of lung cancer at flexible bronchoscopy. Respir Med 2001; 95: 374-78.

79 Hochstenbag MM, Twijnstra A, Wilmink JT, Wouters EF, ten Velde GP. Asymptomatic brain metastases (BM) in small cell lung cancer (SCLC): MR-imaging is useful at initial diagnosis. J Neurooncol 2000; 48: 243-48.

80 Richardson GE, Venzon DJ, Edison M, et al. Application of an algorithm for staging small-cell lung cancer can save one third of the initial evaluation costs. Arch Intern Med 1993; 153: 329-37.

81 Bradley JD, Dehdashti F, Mintun MA, Govindan R, Trinkaus K, Siegel BA. Positron emission tomography in limited-stage small-cell lung cancer: a prospective study. J Clin Oncol 2004; 22: 3248-54.

Page 18: Small CEL Carcinoma

82 Paesmans M, Sculier JP, Lecomte J, et al. Prognostic factors for patients with small cell lung carcinoma: analysis of a series of 763 patients included in 4 consecutive prospective trials with a minimum follow-up of 5 years. Cancer 2000; 89: 523-33.

83 Spiegelman D, Maurer LH, Ware JH, et al. Prognostic factors in small-cell carcinoma of the lung: an analysis of 1,521 patients. J Clin Oncol 1989; 7: 344-54.

84 Albain KS, Crowley JJ, LeBlanc M, Livingston RB. Determinants of improved outcome in small-cell lung cancer: an analysis of the 2,580-patient Southwest Oncology Group data base. J Clin Oncol 1990; 8: 1563-74.

85 Osterlind K, Andersen PK. Prognostic factors in small cell lung cancer: multivariate model based on 778 patients treated with chemotherapy with or without irradiation. Cancer Res 1986; 46: 4189-94.

86 Wolf M, Holle R, Hans K, Drings P, Havemann K. Analysis of prognostic factors in 766 patients with small cell lung cancer (SCLC): the role of sex as a predictor for survival. Br J Cancer 1991; 63: 986-92.

87 Turrisi AT 3rd, Kim K, Blum R, et al. Twice-daily compared with once-daily thoracic radiotherapy in limited small-cell lung cancer treated concurrently with cisplatin and etoposide. N Engl J Med 1999; 340: 265-71.

88 Fox W, Scadding JG. Medical Research Council comparative trial of surgery and radiotherapy for primary treatment of small-celled or oat-celled carcinoma of bronchus: ten-year follow-up. Lancet 1973; 2: 63-65.

89 Mountain CF. Clinical biology of small cell carcinoma: relationship to surgical therapy. Semin Oncol 1978; 5: 272-79.

90 Shepherd FA, Ginsberg RJ, Feld R, Evans WK, Johansen E. Surgical treatment for limited small-cell lung cancer: the University of Toronto Lung Oncology Group experience. J Thorac Cardiovasc Surg 1991; 101: 385-93.

91 Rea F, Callegaro D, Favaretto A, et al. Long term results of surgery and chemotherapy in small cell lung cancer. Eur J Cardiothorac Surg 1998; 14: 398-402.

92 Osterlind K, Hansen M, Hansen HH, Dombernowsky P. Influence of surgical resection prior to chemotherapy on the long-term results in small cell lung cancer: a study of 150 operable patients. Eur J Cancer Clin Oncol 1986; 22: 589-93.

93 Karrer K, Shields TW, Denck H, Hrabar B, Vogt-Moykopf I, Salzer GM. The importance of surgical and multimodality treatment for small cell bronchial carcinoma. J Thorac Cardiovasc Surg 1989; 97: 168-76.

94 Cohen MH, Ihde DC, Bunn PA Jr, et al. Cyclic alternating combination chemotherapy for small cell bronchogenic carcinoma. Cancer Treat Rep 1979; 63: 163-70.

Page 19: Small CEL Carcinoma

95 Salazar OM, Yee GJ, Slawson RG. Radiation therapy for chest recurrences following induction chemotherapy in small cell lung cancer. Int Radiat Oncol Biol Phys 1991; 21: 645-50.

96 Sundstrom S, Bremnes RM, Kaasa S, et al. Cisplatin and etoposide regimen is superior to cyclophosphamide, epirubicin, and vincristine regimen in small-cell lung cancer: results from a randomized phase III trial with 5 years' follow-up. J Clin Oncol 2002; 20: 4665-72.

97 Takada M, Fukuoka M, Kawahara M, et al. Phase III study of concurrent versus sequential thoracic radiotherapy in combination with cisplatin and etoposide for limited-stage small-cell lung cancer: results of the Japan Clinical Oncology Group Study 9104. J Clin Oncol 2002; 20: 3054-60.

98 Ihde DC, Mulshine JL, Kramer BS, et al. Prospective randomized comparison of high-dose and standard-dose etoposide and cisplatin chemotherapy in patients with extensive-stage small-cell lung cancer. J Clin Oncol 1994; 12: 2022-34.

99 Murray N. Coy P, Pater JL, et al. Importance of timing for thoracic irradiation in the combined modality treatment of limited-stage small-cell lung cancer. J Clin Oncol 1993; 11: 336-44.

100 Byrne MJ, van Hazel G, Trotter J, et al. Maintenance chemotherapy in limited small cell lung cancer: a randomised controlled clinical trial. Br J Cancer 1989; 60: 413-18.

101 Giaccone G, Dalesio O, McVie GJ, et al. Maintenance chemotherapy in small-cell lung cancer: long-term results of a randomized trial. J Clin Oncol 1993; 11: 1230-40.

102 Sculier JP, Paesmans M, Bureau G, et al. Randomized trial comparing induction chemotherapy versus induction chemotherapy followed by maintenance chemotherapy in small-cell lung cancer. J Clin Oncol 1996; 14: 2337-44.

103 Beith JM, Clarke SJ, Woods RL, Bell DR, Levi JA. Long-term follow-up of a randomised trial of combined chemoradiotherapy induction treatment, with and without maintenance chemotherapy in patients with small cell carcinoma of the lung. Eur J Cancer 1996; 32A: 438-43.

104 Pignon JP, Arriagada R, Ihde DC, et al. A meta-analysis of thoracic radiotherapy for small-cell lung cancer. N Engl J Med 1992; 327: 1618-24.

105 Jeremic B, Shibamoto Y, Acimovic L, Milisavljevic S. Initial versus delayed accelerated hyperfractionated radiation therapy and concurrent chemotherapy in limited small-cell lung cancer: a randomized study. J Clin Oncol 1997; 15: 893-900.

106 Auperin A, Arriagada R, Pignon JP, et al. Prophylactic cranial irradiation for patients with small-cell lung cancer in complete remission. N Engl J Med 1999; 341: 476-84.

107 Breneman JC, Warnick RE, Albright RE, Jr., et al. Stereotactic radiosurgery for the treatment of brain metastases: results of a single institution series. Cancer 1997; 79: 551-57.

Page 20: Small CEL Carcinoma

108 Li B, Yu J, Suntharalingam M, et al. Comparison of three treatment options for single brain metastasis from lung cancer. Int J Cancer 2000; 90: 37-45.

109 Noda K, Nishiwaki Y, Kawahara M, et al. Irinotecan plus cisplatin compared with etoposide plus cisplatin for extensive small-cell lung cancer. N Engl J Med 2002; 346: 85-91.

110 Hanna NH, Einhorn LH, Sandler A, et al. Randomized, phase III trial comparing irinotecan/cisplatin (IP) with etoposide/cisplatin (EP) in patients with previously untreated, extensive-stage (ES) small cell lung cancer (SCLC). Proc Am Soc Clin Oncol 2005: LBA 7004.

111 Eckardt JR, von Pawel J, Manikhas G, et al. Comparable activity with oral topotecan/cisplatin (TC) and IV etoposide/cisplatin (PE) as treatment for chemotherapy-naive patients with extensive disease small cell lung cancer (ED-SCLC): final results of a randomized phase III trial. Proc Am Soc Clin Oncol 2005: LBA 7003.

112 Loehrer PJ Sr, Ansari R, Gonin R. et al. Cisplatin plus etoposide with and without ifosfamide in extensive small-cell lung cancer: a Hoosier Oncology Group study. J Clin Oncol 1995; 13: 2594-99.

113 Pujol JL, Daures JP, Riviere A, et al. Etoposide plus cisplatin with or without the combination of 4'-epidoxorubicin plus cyclophosphamide in treatment of extensive small-cell lung cancer: a French Federation of Cancer Institutes multicenter phase III randomized study. J Natl Cancer Inst 2001; 93: 300-08.

114 Arriagada R, Le Chevalier T, Pignon JP, et al. Initial chemotherapeutic doses and survival in patients with limited small-cell lung cancer. N Engl J Med 1993; 329: 1848-52.

115 Fetscher S, Brugger W, Engelhardt R, et al. Dose-intense therapy with etoposide, ifosfamide, cisplatin, and epirubicin (VIP-E) in 107 consecutive patients with limited- and extensive-stage non-small-cell lung cancer. Ann Oncol 1997; 8: 57-64.

116 Elias A, Ibrahim J, Skarin AT, et al. Dose-intensive therapy for limited-stage small-cell lung cancer: long-term outcome. J Clin Oncol 1999; 17: 1175.

117 Humblet Y, Symann M, Bosly A, et al. Late intensification chemotherapy with autologous bone marrow transplantation in selected small-cell carcinoma of the lung: a randomized study. J Clin Oncol 1987; 5: 1864-73.

118 Slevin ML, Clark PI, Joel SP, et al. A randomized trial to evaluate the effect of schedule on the activity of etoposide in small-cell lung cancer. J Clin Oncol 1989; 7: 1333-40.

119 Cavalli F, Sonntag RW, Jungi F, Senn HJ, Brunner KW. VP-16-213 monotherapy for remission induction of small cell lung cancer: a randomized trial using three dosage schedules. Cancer Treat Rep 1978; 62: 473-75.

120 Miller AA, Herndon JE 2nd, Hollis DR, et al. Schedule dependency of 21-day oral versus 3-day intravenous etoposide in combination with intravenous cisplatin in extensive-stage small-cell lung cancer: a randomized phase III study of the Cancer and Leukemia Group B. J Clin Oncol 1995; 13: 1871-79.

Page 21: Small CEL Carcinoma

121 Furuse K, Fukuoka M, Nishiwaki Y, et al. Phase III study of intensive weekly chemotherapy with recombinant human granulocyte colony-stimulating factor versus standard chemotherapy in extensive-disease small-cell lung cancer. J Clin Oncol 1998; 16: 2126-32.

122 Murray N, Shah A, Osoba D, et al. Intensive weekly chemotherapy for the treatment of extensive-stage small-cell lung cancer. J Clin Oncol 1991; 9: 1632-38.

123 Sculier JP, Paesmans M, Lecomte J, et al. A three-arm phase III randomised trial assessing, in patients with extensive-disease small-cell lung cancer, accelerated chemotherapy with support of haematological growth factor or oral antibiotics. Br J Cancer 2001; 85: 1444-51.

124 Steward WP, von Pawel J, Gatzemeier U, et al. Effects of granulocyte-macrophage colony-stimulating factor and dose intensification of V-ICE chemotherapy in small-cell lung cancer: a prospective randomized study of 300 patients. J Clin Oncol 1998; 16: 642-50.

125 Thatcher N, Girling DJ, Hopwood P, Sambrook RJ, Qian W, Stephens RJ. Improving survival without reducing quality of life in small-cell lung cancer patients by increasing the dose-intensity of chemotherapy with granulocyte colony-stimulating factor support: results of a British Medical Research Council Multicenter Randomized Trial. J Clin Oncol 2000; 18: 395-404.

126 Bishop JF, Raghavan D, Stuart-Harris R, et al. Carboplatin (CBDCA, JM-8) and VP-16-213 in previously untreated patients with small-cell lung cancer. J Clin Oncol 1987; 5: 1574-78.

127 Skarlos DV, Samantas E, Kosmidis P, et al. Randomized comparison of etoposide-cisplatin vs. etoposide-carboplatin and irradiation in small-cell lung cancer. Ann Oncol 1994; 5: 601-07.

128 Ettinger DS, Finkelstein DM, Abeloff MD, Ruckdeschel JC, Aisner SC, Eggleston JC. A randomized comparison of standard chemotherapy versus alternating chemotherapy and maintenance versus no maintenance therapy for extensive-stage small-cell lung cancer: a phase III study of the Eastern Cooperative Oncology Group. J Clin Oncol 1990; 8: 230-40.

129 Schiller JH, Adak S, Cella D, DeVore RF 3rd, Johnson DH. Topotecan versus observation after cisplatin plus etoposide in extensive-stage small-cell lung cancer: E7593, a phase III trial of the Eastern Cooperative Oncology Group. J Clin Oncol 2001; 19: 2114-22.

130 Evans WK, Feld R, Murray N, et al. Superiority of alternating non-cross-resistant chemotherapy in extensive small cell lung cancer: a multicenter, randomized clinical trial by the National Cancer Institute of Canada. Ann Intern Med 1987; 107: 451-58.

131 Fukuoka M, Furuse K, Saijo N, et al. Randomized trial of cyclophosphamide, doxorubicin, and vincristine versus cisplatin and etoposide versus alternation of these regimens in small-cell lung cancer. J Natl Cancer Inst 1991; 83: 855-61.

132 Roth BJ, Johnson DH, Einhorn LH, et al. Randomized study of cyclophosphamide, doxorubicin, and vincristine versus etoposide and cisplatin versus alternation of these two

Page 22: Small CEL Carcinoma

regimens in extensive small-cell lung cancer: a phase III trial of the Southeastern Cancer Study Group. J Clin Oncol 1992; 10: 282-91.

133 Rigas JR, Denham CA, Rinaldi D, et al. Adjuvant targeted therapy in unresectable lung cancer: the results of two randomized placebo-controlled trials of BAY 12-9566, a matrix metalloproteinase inhibitor (MMPI). Lung Cancer 2003; 41 (suppl 2): S34 [abstr O107].

134 Shepherd FA, Giaccone G, Seymour L, et al. Prospective, randomized, double-blind, placebo-controlled trial of marimastat after response to first-line chemotherapy in patients with small-cell lung cancer: a trial of the National Cancer Institute of Canada-Clinical Trials Group and the European Organization for Research and Treatment of Cancer. J Clin Oncol 2002; 20: 4434-39.

135 Heymach JV, De Porre PM, DeVore RF, et al. Phase II study of the farnesyl transferase inhibitorr (FTI) R115777 (Zarnestra) in patients with relapsed small cell lung cancer (SCLC). Proc Am Soc Clin Oncol 2002; 21: 319a [abstr 1275].

136 Johl J, Chansky K, Lara PN, Davies AM, Bold R, Gandara DR. The proteosome inhibitor PS-341 (Bortezomib) in platinum (plat)-treated extensive-stage small cell lung cancer (E-SCLC): a SWOG (0327) phase II trial. Proc Am Soc Clin Oncol 2005: LBA 7047.

137 Pandya KJ, Levy DE, Hidalgo M, et al. A randomized, phase II ECOG trial of two dose levels of temsirolimus (CCI-779) in patients with extensive stage small cell lung cancer in remission after induction chemotherapy: a preliminary report. Proc Am Soc Clin Oncol 2005: LBA 7005.

138 Phase III randomized study of carboplatin and etoposide with versus without thalidomide in patients with limited or extensive stage small cell lung cancer. Protocol IDs: LLCG STUDY-12, EU-20207 NCT00061919. Available at: http://www.nci.nih.gov/clinical trials (accessed Aug 24, 2005).

139 Phase II randomized study of ZD6474 in patients with small cell lung cancer who responded to induction chemotherapy with or without radiotherapy. Protocol IDs: CAN-NCIC-BR20, ZENECA-6474IL/0005, NCT00066313. Available at: http://www.nci.nih.gov/dinicaltrials (accessed Aug 24, 2005).

140 Arriagada R, Le Chevalier T, Borie F, et al. Prophylactic cranial irradiation for patients with small-cell lung cancer in complete remission. J Natl Cancer Inst 1995; 87: 183-90.

141 Gregor A, Cull A, Stephens RJ, et al. Prophylactic cranial irradiation is indicated following complete response to induction therapy in small cell lung cancer: results of a multicentre randomised trial. Eur J Cancer 1997; 33: 1752-58.

142 Bork E, Ersboll J, Dombernowsky P, Bergman B, Hansen M, Hansen HH. Teniposide and etoposide in previously untreated small-cell lung cancer: a randomized study. J Clin Oncol 1991; 9: 1627-31.

143 Souhami RL, Spiro SG, Rudd RM. et al. Five-day oral etoposide treatment for advanced small-cell lung cancer: randomized comparison with intravenous chemotherapy. J Natl Cancer Inst 1997; 89: 577-80.

Page 23: Small CEL Carcinoma

144 Girling DJ. Comparison of oral etoposide and standard intravenous multidrug chemotherapy for small-cell lung cancer: a stopped multicentre randomised trial. Lancet 1996; 348: 563-66.

145 Evans WK, Radwi A, Tomiak E, et al. Oral etoposide and carboplatin: effective therapy for elderly patients with small cell lung cancer. Am J Clin Oncol 1995; 18: 149-55.

146 Matsui K, Masuda N, Fukuoka M, et al. Phase II trial of carboplatin plus oral etoposide for elderly patients with small-cell lung cancer. Br J Cancer 1998; 77: 1961-65.

147 Westeel V, Murray N, Gelmon K, et al. New combination of the old drugs for elderly patients with small-cell lung cancer: a phase II study of the PAVE regimen. J Clin Oncol 1998; 16: 1940-47.

148 von Pawel J, Schiller JH, Shepherd FA, et al. Topotecan versus cydophosphamide, doxorubicin, and vincristine for the treatment of recurrent small-cell lung cancer. J Clin Oncol 1999; 17: 658-67.

149 Perez-Soler R, Glisson BS, Lee JS, et al. Treatment of patients with small-cell lung cancer refractory to etoposide and cisplatin with the topoisomerase I poison topotecan. J Clin Oncol 1996; 14: 2785-90.

150 Ardizzoni A, Hansen H, Dombernowsky P, et al. Topotecan, a new active drug in the second-line treatment of small-cell lung cancer: a phase II study in patients with refractory and sensitive disease. J Clin Oncol 1997; 15: 2090-96.

151 Ardizzoni A, Manegold C, Debruyne C. et al. European organization for research and treatment of cancer (EORTC) 08957 phase II study of topotecan in combination with cisplatin as second-line treatment of refractory and sensitive small cell lung cancer. Clin Cancer Res 2003; 9: 143-50.

152 Masuda N, Matsui K, Negoro S, et al. Combination of irinotecan and etoposide for treatment of refractory or relapsed small-cell lung cancer. J Clin Oncol 1998; 16: 3329-34.

153 Groen HJ, Fokkema E, Biesma B, et al. Paclitaxel and carboplatin in the treatment of small-cell lung cancer patients resistant to cyclophosphamide, doxorubicin, and etoposide: a non-cross-resistant schedule. J Clin Oncol 1999; 17: 927-32.

154 Stephens RJ, Bailey AJ, Machin D. Long-term survival in small cell lung cancer: the case for a standard definition. Lung Cancer 1996; 15: 297-309.

155 Lassen U, Osterlind K, Hansen M, Dombernowsky P, Bergman B, Hansen HH. Long-term survival in small-cell lung cancer: posttreatment characteristics in patients surviving 5 to 18+ years-an analysis of 1,714 consecutive patients. J Clin Oncol 1995; 13: 1215-20.

156 Tucker MA, Murray N, Shaw EG, et al. Second primary cancers related to smoking and treatment of small-cell lung cancer. J Natl Cancer Inst 1997; 89: 1782-88.

Page 24: Small CEL Carcinoma

157 van der Gaast A, Postmus PE, Burghouts J, van Bolhuis C, Stam J, Splinter TA. Long term survival of small cell lung cancer patients after chemotherapy. Br J Cancer 1993; 67: 822-24.

158 Johnson BE, Argiris A, Crawford J, et al. NCCN practice guidelines in oncology: small cell lung cancer, 2005. http://www.nccn.org/professionals/physician_gls/PDF/sclc.pdf (accessed Aug 2, 2005).

159 Felip E, Stahel RA, Pavlidis N. ESMO minimum clinical recommendations for diagnosis, treatment, and follow-up of small-cell lung cancer (SCLC). Ann Oncol 2005; 16 (suppl 1): i30-31.

AuthorAffiliation

Dana Farber Cancer Institute and Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA (D M Jackman MD, B E Johnson MD)

Correspondence to: Dr David M Jackman, Dana Farber Cancer Institute, 44 Binney Street, Boston, MA 02115, USA

[email protected]

Copyright Lancet Ltd. Oct 15-Oct 21, 2005