当前位置: 首页 > 期刊 > 《新英格兰医药杂志》 > 2004年第4期 > 正文
编号:11307640
Multidisciplinary Management of Lung Cancer
http://www.100md.com 《新英格兰医药杂志》
     In 2004, an estimated 173,700 Americans will receive a diagnosis of lung cancer, and 164,440 of them will die of the disease. Despite years of research, the prognosis for patients with lung cancer remains dismal, with a five-year survival rate of 14 percent. Nevertheless, lung cancer may be curable in its early stages, and most patients derive some benefit from treatment such as longer survival or amelioration of symptoms. The topic was last reviewed in the Journal in 1992.1 This review will focus on the multidisciplinary management and treatment of lung cancer, with particular emphasis on phase 3 studies.

    Figure 1 shows a model of the pathogenesis of lung cancer, with its progression from normal tissue to frankly malignant tissue. Lung cancer is divided into two types: non–small cell and small cell. Non–small-cell lung cancer consists of several subtypes, predominantly adenocarcinoma, squamous-cell carcinoma, and large-cell carcinoma, which are all treated in the same manner. Small-cell lung cancer is a very aggressive neuroendocrine lung carcinoma, treated primarily with chemotherapy and, occasionally, radiotherapy. Treatments and their adverse effects are managed by multiple physicians, with different subspecialty expertise; careful coordination of the patient's care is essential.

    Figure 1. Sequential Pathogenetic Changes Involved in Lung Cancer.

    Although multiple genetic markers (indicated by the arrows) are abnormal in lung cancers, the timing of their appearance during the lengthy preneoplastic process varies. Several alterations have been described in histologically normal specimens of bronchial epithelium from smokers. Other changes (such as hyperplasia and metaplasia) have been detected in slightly abnormal epithelium and are regarded as early changes. Molecular changes detected frequently in dysplasia are regarded as intermediate in timing, whereas those usually detected in carcinoma in situ or invasive stages are regarded as late changes. Modified from Hirsch et al.2

    Principles of Radiotherapy in Lung Cancer

    Radiotherapy is usually delivered by an external beam from a linear accelerator. Standard therapy for unresectable disease consists of approximately 60 Gy, with the dose divided among 30 sessions over a period of six weeks, although higher doses have been used.3 In normal organs, the toxic effects of radiation include pneumonitis, esophagitis, skin desquamation, myelopathies, and cardiac abnormalities.4 These effects may be minimized by using three-dimensional computed tomography (CT) to guide therapy or by modulating the intensity of radiotherapy in order to spare normal tissue. Concurrent chemotherapy may increase the effectiveness of radiation by sensitizing the tumor to radiation, but it can also increase the adverse effects (particularly esophagitis).

    General Approach to Chemotherapy

    Many chemotherapeutic agents are effective against both small-cell and non–small-cell lung cancer (Table 1). Among the most active are those of the platinum family: cisplatin, which cross-links DNA, and carboplatin, a cisplatin analogue. Most studies suggest that carboplatin is as efficacious as cisplatin but less toxic. However, published data do not support the frank substitution of carboplatin for cisplatin in patients with curable disease.5,6 The adverse effects of chemotherapy can be severe but are generally manageable and reversible. Major effects include nausea, vomiting, alopecia, myelosuppression, nephrotoxicity, neuropathies, high-pitch hearing loss, and electrolyte depletion. Etoposide, docetaxel, paclitaxel, gemcitabine, vinorelbine, and irinotecan are often used in combination with a platinum agent.

    Non–Small-Cell Lung Cancer

    Staging and Evaluation

    The tumor, node, and metastasis (TNM) staging system for lung cancer, developed by the American Joint Committee on Cancer, is shown in Table 2.7,8 Before treatment recommendations can be made for a given patient, the size of the tumor, lymph-node status, and possible presence of metastases must be ascertained. Lung cancer often spreads to the draining nodes in the hilum and mediastinum. Mediastinal involvement (N2) places the patient in a higher stage and may render the tumor unresectable, if it is close to vital mediastinal structures. Accurate noninvasive assessment of hilar and mediastinal nodes is difficult. Clinical staging is based on the size of the lymph node as determined by CT. Until fairly recently, the use of a cutoff for normal nodes of 10 to 15 mm provided sensitivities and specificities for detecting nodal metastases of only 40 to 70 percent, but this approach was the only noninvasive option.9,10 Even small, peripheral, T1 lesions (Table 2), often considered to be associated with a low risk of mediastinal spread, frequently involve these nodes.11

    Positron-emission tomography (PET) has emerged as an important noninvasive test for mediastinal assessment. This approach operates on the principle that tumors cause increased uptake of radiolabeled glucose that can be imaged. Retrospective data from several trials suggest a sensitivity and specificity of 85 percent and 88 percent, respectively, for the mediastinal staging of non–small-cell lung cancer with the use of PET, and the results of a prospective study support these findings.12 The combination of PET and CT appears to have an even greater sensitivity and specificity than the use of either method alone,13 and the use of both should be strongly considered as part of the preoperative evaluation. The gold standard for mediastinal evaluation is lymph-node biopsy by means of bronchoscopy or, if needed, the more invasive mediastinoscopy. To distinguish potentially curable from incurable disease, radiologic studies that indicate the presence of mediastinal disease should be followed by biopsy of identified nodes before a tumor is deemed to be unresectable. Brain magnetic resonance imaging14 and bone scanning15 should be performed in all patients who have N2 disease or other clinical indications before aggressive local therapy is considered.

    Surgery

    Surgery is the mainstay of treatment for patients with non–small-cell lung cancer, particularly for those with early disease. Surgery should include resection (pneumonectomy or lobectomy) and mediastinal-node mapping. Complete lymph-node dissection should be performed if the tumor is resectable and mediastinal nodes are involved.

    Adjuvant Therapy

    Adjuvant therapy refers to the use of radiation or chemotherapy to improve survival after a tumor has been treated surgically.

    Radiotherapy

    Adjuvant radiotherapy has been considered a means to eliminate small deposits of tumor cells adjacent to or draining from the primary tumor site. Unfortunately, the results of adjuvant radiotherapy have been quite variable; some trials show a benefit,16 whereas most show none (Table 3; additional information has been deposited with the National Auxiliary Publications Service ).21,32,33 A large meta-analysis published in 1998 suggested that postoperative radiotherapy was detrimental, with a 21 percent increase in the relative risk of death, and was particularly harmful for patients with stage I disease.34 However, this analysis included data that are very dated, given the availability of modern radiotherapy and staging techniques. One important exception must be mentioned: in 1986, a trial from the Lung Cancer Study Group demonstrated that adjuvant radiotherapy prevented local recurrence in patients with N2 disease but did not improve overall survival.16 These limited data have become the rationale for the use of postoperative radiation in otherwise healthy patients with N2 disease. Postoperative radiotherapy should not be used outside of a clinical trial for any other type of patients, unless the surgical margins are positive and repeated resection is not feasible.

    Chemotherapy

    Given the poor prognosis for patients with early non–small-cell lung cancer, even with adequate surgical resection, many patients probably have undetectable microscopic metastasis at diagnosis. In theory, chemotherapy with a cytotoxic agent may eliminate micrometastases, improving survival. Although this concept is appealing, the results of trials are mixed (Table 3 and * document 05612). Most trials evaluating chemotherapy have combined patients with different stages of disease (usually stage II and III). Modern trials usually use platinum-based regimens, since this class is the most active against non–small-cell lung cancer. Most individual trials have not shown a statistically significant benefit of adjuvant chemotherapy,19,35,36,37 and the few that did18,38 suggested that there was only a small (10 to 15 percent) survival advantage several years after diagnosis in patients with incompletely resected tumors17 or stage III tumors.39

    A large meta-analysis reported in 199540 evaluated data on adjuvant chemotherapy from all trials that took place between 1965 and 1991. The use of adjuvant therapy based on alkylating agents (mainly cyclophosphamide and nitrosourea) proved detrimental. Treatment with cisplatin-based therapy resulted in a moderate (13 percent) reduction in the risk of death that did not reach statistical significance (P=0.08). Trials that used a combination of chemotherapy and radiation had similar results.40 A subsequent study from the Eastern Cooperative Oncology Group19 similarly did not show a benefit of multimodal postoperative adjuvant therapy (cisplatin, etoposide, and radiotherapy). The results of the International Adjuvant Lung Cancer Trial are reported elsewhere in this issue of the Journal.20 In this study of 1867 patients randomly assigned to either cisplatin-based adjuvant chemotherapy or no adjuvant therapy, adjuvant chemotherapy provided an absolute advantage of 5 percent for disease-free survival at five years and of 4 percent for overall survival.20 Adjuvant chemotherapy, as currently administered, may provide a small benefit in certain patients, but this particular subgroup is very difficult to define with the use of current techniques. On the basis of the results of the International Adjuvant Lung Cancer Trial20 and the aforementioned meta-analysis,40 one should strongly consider the use of adjuvant platinum-based chemotherapy in patients with stage I, II, or IIIA non–small-cell lung cancer.

    Neoadjuvant Therapy

    Neoadjuvant therapy refers to the use of nonsurgical therapy as initial treatment (chemotherapy or radiotherapy) for cases in which surgery is a suboptimal initial approach. Ideally, neoadjuvant radiotherapy results in shrinkage of the tumor, allowing for a complete surgical resection. Neoadjuvant chemotherapy might result both in tumor shrinkage and early eradication of systemic micrometastases.

    Nonresectable Tumors

    Stage III tumors or those invading vital structures are often described as either nonresectable or marginally resectable. For many years, the mainstay of treatment for these tumors was radiotherapy (total dose, 60 Gy). The risk of local recurrence was diminished, but the rate of long-term survival was still poor (5 percent).41 The results of several phase 2 studies provided preliminary support for the addition of chemotherapy to radiotherapy, and a landmark trial of this combined approach, reported in 1990 by Dillman et al.22 (Table 3 and * document 05612), demonstrated increased rates of three-year survival (23 percent as compared with 11 percent) and long-term survival.23 Subsequent randomized trials (* document 05612) reported a variable benefit for combined therapy — some findings were positive,42,43 and others were not.44,45,46 Two large meta-analyses (Table 3 and * document 05612) have provided support for the benefits of combined chemotherapy and radiotherapy. A meta-analysis by Pritchard and Anthony showed that combined therapy for unresectable disease resulted in a significant decrease in the relative risk of death at both one and three years.47 Similarly, Marino et al. reported a 24 percent reduction in the risk of death at one year and a 30 percent reduction at two years for combined cisplatin-based chemotherapy and radiotherapy.48

    The optimal sequence of combined therapy has yet to be determined, although concurrent therapy appears to be superior to sequential (segregated) therapy.24,49 Furuse et al. demonstrated that the use of concurrent chemotherapy and radiotherapy rather than sequential therapy improved survival.49 Curran and colleagues obtained similar results in a large study: concurrent therapy resulted in a survival rate of 25 percent, as compared with 4 percent for sequential therapy (P=0.046).24 The concurrent approach appears to increase the rate of adverse events, mainly esophagitis (21 percent, as compared with 4 percent, in the study by Curran et al.).24 Given its apparent superiority, we believe that concurrent chemotherapy and radiotherapy should be used in all patients, if possible.

    Additional approaches are being explored. For example, in multicenter,50 single-center,51 and phase 2 trials,52 the combination of carboplatin, paclitaxel, and concurrent radiotherapy improved survival (median, 20.5 months), with acceptable rates of adverse effects. This apparently promising approach must be examined in phase 3 trials.

    Resectable Tumors

    In patients with non–small-cell lung cancer, resectable tumors can range from stage I to stage IIIA. Although surgery is the mainstay of therapy for such tumors, survival after surgery alone remains suboptimal. For tumors involving the chest wall, diaphragm, or pleura (T3) without visible mediastinal involvement, en bloc resection of the entire tumor should be performed. T3 tumors involving the superior sulcus of the lung (Pancoast's tumor) have a propensity to invade surrounding thoracic inlet structures and are associated with a high incidence of local recurrence, because tumor-free margins cannot be achieved. Several retrospective, single-institution studies have shown improved survival and decreased rates of local recurrence with combined neoadjuvant chemotherapy and radiotherapy.53,54,55 Studies using combined neoadjuvant chemotherapy and radiotherapy followed by surgical resection56,57 have demonstrated two-year survival rates in the range of 50 to 70 percent, which is higher than the historical rate of approximately 20 percent among patients receiving postoperative radiotherapy alone.53 Even patients with vertebral invasion may have a significant survival advantage with aggressive multimodality therapy.58 Neoadjuvant chemoradiotherapy followed by complete surgical excision is thus the preferred approach to these tumors.

    Tumors with ipsilateral mediastinal spread (N2) may be resectable but fall into the category of locally advanced tumors (stage IIIA), which are associated with poor survival8 (Table 2). Because of its success in patients with nonresectable (N2) tumors, combined neoadjuvant chemotherapy and radiotherapy have been used in patients with resectable N2 tumors. In theory, neoadjuvant therapy facilitates early systemic therapy for micrometastases, as well as tumor shrinkage, which can lead to a more complete resection. In 1989, Skarin et al. reported the results of neoadjuvant cisplatin-based chemotherapy followed by surgery and radiotherapy in patients with resectable stage III disease.59 Median survival was 32 months, and the 1-year survival rate was 75 percent, both of which were higher than previously reported rates.59

    Two randomized, controlled trials evaluating the efficacy of combined neoadjuvant therapy for resectable non–small-cell lung cancer were reported in 1994 (Table 3 and * document 05612).25,27 Roth et al. studied 60 patients who were randomly assigned to receive either six cycles of preoperative cisplatin-based therapy or surgery alone.25 Patients receiving neoadjuvant chemotherapy had a median survival of 64 months, as compared with 11 months for those undergoing surgery alone; the 3-year survival rates were 56 percent and 15 percent, respectively. Rosell et al. studied 60 patients who were randomly assigned to either surgery alone or induction cisplatin-based chemotherapy followed by surgery and radiotherapy.27 Median survival was 26 months in the combined-treatment group, as compared with 8 months in the surgery-only group. Long-term follow-up in both these studies supported the findings that this combined-treatment approach was beneficial.26,28 A third, smaller study had similar findings.60

    The studies by both Roth et al.25 and Rosell et al.27 have been criticized for several reasons, including their small size (60 patients in each), imbalances between groups, and poorer-than-expected outcomes in the control groups. Depierre and colleagues performed a much larger study that explored the uses of neoadjuvant chemotherapy in 355 patients with early non–small-cell lung cancer who were randomly assigned to receive either preoperative chemotherapy with two cycles of chemotherapy followed by surgery or surgery alone.29 Patients with chemotherapy-responsive disease underwent two additional cycles of postoperative chemotherapy, and radiotherapy was used for patients who had T3N2 disease or an incomplete resection of tumor. There was a nonsignificant trend toward a survival advantage for those who received combined therapy (P=0.15). Subgroup analysis showed that combined therapy did not benefit patients with N2 disease (relative risk of death, 1.04). Further analysis demonstrated that the risk of distant recurrence was lower in the chemotherapy group, but there was no significant difference in the risk of locoregional relapse, raising the possibility that chemotherapy eradicated microscopic metastases. Thus, the results of this larger study contrast with those of Roth et al.25 and Rosell et al.27

    The use of a neoadjuvant approach to all stages of non–small-cell lung cancer is currently being assessed. Pisters and colleagues reported the results of a phase 2 trial, the Bimodality Lung Oncology Team (BLOT) study, in which neoadjuvant carboplatin and paclitaxel followed by surgery were used in patients with early disease and appeared to be highly successful (a survival rate of 85 percent at one year).61 A second phase 2 study, which used a combination of gemcitabine and cisplatin, had a similar outcome.62 In our opinion, otherwise healthy patients with locally advanced (N2) disease should receive neoadjuvant chemotherapy, although this approach is controversial. For patients with earlier-stage disease, data concerning neoadjuvant therapy are too premature to recommend such treatment outside of a clinical trial. Future studies of operable tumors will probably address the potential benefit of additional postoperative (adjuvant) chemotherapy in patients whose tumors have responded to neoadjuvant chemotherapy.

    Advanced Disease

    Currently, virtually no patient with disease as advanced as stage IIIB or IV will be cured. Although chemotherapy is the backbone of treatment for metastatic disease, response rates are low, and survival times poor. In the past, many patients with advanced non–small-cell lung cancer received no therapy, since the toxicity of therapy was thought to outweigh the benefits. It is now clear, however, that treatment can be beneficial. Several meta-analyses have reported moderate gains in survival when chemotherapy is used, as compared with the best supportive care. Increases in median survival appear to be in the range of two to four months, and increases in the one-year survival rate appear to range from 10 to 20 percent.40,48,63 Studies have also suggested important gains in other therapeutic end points such as the time to disease progression and the quality of life.64,65,66 The benefits of therapy are usually restricted to otherwise healthy patients with lung cancer — that is, those who maintain a good functional status.

    Although many agents are active against non–small-cell lung cancer, single-agent platinum therapy remained the backbone of treatment until the 1990s. Phase 3 studies in the 1980s did not show that adding a second or third agent was beneficial.67,68 With the development of additional cytotoxic drugs, such as gemcitabine, vinorelbine, paclitaxel, and docetaxel, combination therapy was reevaluated. Several randomized trials evaluating these newer agents in combination with cisplatin, as compared with cisplatin alone, showed response rates favoring combination therapy, with minimal additional toxicity (Table 3 and * document 05612).69,70,71,72,73,74,75

    In 2000, Bonomi et al. were the first to report a better response rate with the use of a modern combination — paclitaxel and cisplatin — than with an older regimen, etoposide and cisplatin.76 In the 1990s, numerous two-drug regimens were in common use, although there were few data from direct comparisons of their efficacies. In 2002, Schiller et al. reported on a comparison of four commonly used two-drug regimens for advanced lung cancer30 (Table 3 and * document 05612). All four treatment groups had virtually identical rates of survival and adverse effects, and all response rates were higher than historical response rates with the use of a single agent. Other trials have had similar results.77,78,79 Unless specific reasons dictate otherwise, patients with advanced lung cancer should, in our opinion, receive a two-drug regimen of chemotherapy.79 Although some practitioners withhold chemotherapy solely on the basis of a patient's age (with the cutoff often arbitrarily set as 70 years), several trials have demonstrated that, as compared with younger patients, the elderly have similar rates of tolerance and receive similar benefits from chemotherapy and should therefore be treated similarly.80,81 Several trials have examined three-drug combinations. Two have shown that this approach increases toxic effects without improving survival,82,83 and such regimens should therefore not be used.

    The optimal duration of therapy has long been debated. A randomized trial compared three cycles of cisplatin-based therapy with six cycles in patients with advanced disease and found only increased toxicity with prolonged administration of chemotherapy,84 as have other trials.85,86 Patients with advanced disease should therefore initially be limited to three or four cycles of two-agent chemotherapy.

    Since in virtually all patients with advanced disease, initial therapy will ultimately fail, second-line therapy will often be necessary.87 Most patients will have received first-line platinum-based therapy, and because of presumed tumor resistance and drug toxicity, second-line platinum therapy is not usually used. Several randomized studies suggest that docetaxel may offer some survival benefit in such a second-line setting, as compared with both the best supportive care88 and other agents.89 Other agents (e.g., gemcitabine) have activity in this setting as well and may be considered for otherwise healthy patients who can maintain good, independent function.

    Patients with a single, solitary metastasis may benefit from resection of the metastatic lesion. The five-year survival rate among patients who undergo resection of a solitary brain metastasis90 followed by whole-brain radiotherapy91 can reach 10 to 20 percent; the use of subsequent chemotherapy should be considered but has not been well studied. Resection of a solitary adrenal metastasis can also increase long-term survival, although the data are less definitive.92,93

    Small-Cell Lung Cancer

    In contrast to non–small-cell lung cancer, small-cell lung cancer is characterized by its propensity for early metastases and a rapid doubling time.94 Rather than TNM staging, a more practical scheme divides small-cell lung cancer into limited and extensive disease. Limited disease is defined as a tumor that can be encompassed within a single, tolerable radiation port; all other tumors are characterized as extensive. Because of the tumor's propensity for early metastasis, all patients should undergo a staging workup consisting of a history taking and a physical examination, a basic laboratory evaluation, chest CT, bone scanning, and imaging of the brain. With the use of this evaluation, only one third of patients who present with small-cell lung cancer will be classified as having limited disease. Bone marrow evaluation, although used in the past, adds little information and is not required.95

    Limited Disease

    Because small-cell lung cancer has a propensity for early spread yet is responsive to chemotherapy, surgical resection is usually not considered part of the treatment algorithm. Patients with a solitary lung nodule and no evidence of nodal involvement on mediastinal staging should still undergo mediastinal-node dissection at the time of surgery96 and receive postoperative chemotherapy, with the addition of radiation if the mediastinum is involved on microscopical examination of lymph nodes.97 In actuality, these patients are usually first identified at the time of surgery for an undiagnosed lung nodule, because pathological examination reveals small-cell lung cancer. For patients who receive a diagnosis on the basis of a biopsy, management should consist of combined chemotherapy and radiotherapy without surgery.

    Studies in the 1970s and 1980s demonstrated that combination chemotherapy was clearly superior to single-agent therapy.98,99 Several studies compared two commonly used regimens, etoposide and cisplatin with vincristine, doxorubicin, and cyclophosphamide. The outcomes among patients treated with etoposide and cisplatin were superior in one study100 but not in the other101 (Table 4 and * document 05612). Two meta-analyses suggested a moderate benefit of combination chemotherapy and radiotherapy as compared with chemotherapy alone for patients with limited disease (on the order of a 15 percent decrease in the risk of death).105,106

    Concurrent chemotherapy and radiotherapy appear to provide better five-year survival rates than sequential therapy.103 Delivering radiotherapy earlier during chemotherapy is better than delivering it later.107 One randomized trial demonstrated improved five-year survival with minimal additional toxicity when hyperfractionated radiotherapy (i.e., given twice instead of once daily) was used, with the same total dose of 45 Gy.104 However, this approach has been slow to catch on as standard therapy, most likely because of the increased time involved on the part of both patients and physicians.

    Lad et al. did not find that the resection of a residual mass after concurrent chemotherapy and radiotherapy was beneficial in patients with limited disease.108 When combined therapy is used in patients with limited disease, the rates of thoracic recurrences are decreased, but the rates of distant recurrences, particularly in the brain, are increased. Some studies report up to a 50 percent incidence of brain metastasis two years after diagnosis.109,110 Prophylactic cranial irradiation has long been thought to decrease the incidence of brain-only metastasis. A meta-analysis showed a 5.4 percent increase in the three-year rate of overall survival among patients with small-cell lung cancer in complete remission who underwent prophylactic cranial irradiation, as well as a large reduction in the incidence of brain metastasis (relative risk as compared with those who did not undergo prophylactic cranial irradiation, 0.46).111 Once brain metastases appear, patients do not have a good response to treatment. Limited studies suggest that prophylactic cranial irradiation does not result in clinically significant neuropsychological sequelae, the most likely long-term effect,109 particularly when patients are neurologically intact before radiotherapy and the doses are limited to 24 to 36 Gy. However, over time, the radiotherapy could contribute to cognitive abnormalities. Despite this possibility, most oncologists recommend prophylactic cranial irradiation to patients with small-cell lung cancer in complete remission,112 mainly because of its potential to increase the quality of life, as well as its small survival benefit.

    Extensive Disease

    For extensive small-cell lung cancer, the treatment of choice has long been chemotherapy alone consisting of a combination of cisplatin and etoposide. Carboplatin is equivalent to cisplatin in this setting.113 A recent randomized trial compared the use of cisplatin with either irinotecan or etoposide in patients with extensive disease.102 Patients treated with irinotecan and cisplatin had an increase in both median survival (12.8 months vs. 9.4 months) and two-year survival rate (19.5 percent vs. 5.2 percent) and less severe hematologic toxic effects but a higher incidence of diarrhea.102 The addition of paclitaxel to cisplatin plus etoposide for extensive small-cell lung cancer increased the toxic effects without adding a significant survival advantage.114,115 Confirmatory trials of irinotecan are ongoing, but either etoposide or irinotecan in conjunction with cisplatin is appropriate as first-line therapy. Other agents (e.g., topotecan) are active against small-cell lung cancer, are useful for relapsed disease, and are being evaluated for first-line use.116

    Summary and Future Directions

    Table 5 summarizes the management approaches to lung cancer. Future directions for treatment are heavily weighted toward targeted therapies — namely, those aimed at molecular abnormalities involved in the pathogenesis of lung cancer117 — rather than traditional cytotoxic agents.118 A recent review describes many of these therapies, some of which have had moderate activity in a phase 2 setting.119 Cellular targets abound, with the epithelial growth factor receptor the best studied. The first compound against this receptor, gefitinib, has recently been approved for use on the basis of moderate tumor responses, as well as improvements in the quality of life.120 Unfortunately, no additional benefit was seen when gefitinib was combined with standard therapy.31 Other compounds, such as those that target protein kinase C, vascular endothelial growth factor, cyclooxygenase-2, and farnesyl transferase, are being tested. Many of the available data are suboptimal; meta-analyses are used frequently instead of adequately powered, randomized, controlled trials. Since little further progress is expected with the use of traditional cytotoxic agents,121 new agents and approaches must be evaluated if we are to advance therapy for lung cancer.

    * See NAPS document no. 05612 for 16 pages of supplementary material. To order, contact NAPS, c/o Microfiche Publications, 248 Hempstead Tpke., West Hempstead, NY 11552.

    Source Information

    From the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore.

    Address reprint requests to Dr. Ettinger at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Bunting Blaustein Cancer Research Bldg., Rm. G88, 1650 Orleans St., Baltimore, MD 21231-1000, or at ettinda@jhmi.edu.

    References

    Ihde DC. Chemotherapy of lung cancer. N Engl J Med 1992;327:1434-1441.

    Hirsch FR, Franklin WA, Gazdar AF, Bunn PA Jr. Early detection of lung cancer: clinical perspectives of recent advances in biology and radiology. Clin Cancer Res 2001;7:5-22.

    Perez CA, Bauer M, Edelstein S, Gillespie BW, Birch R. Impact of tumor control on survival in carcinoma of the lung treated with irradiation. Int J Radiat Oncol Biol Phys 1986;12:539-547.

    Bloomer WD, Hellman S. Normal tissue responses to radiation therapy. N Engl J Med 1975;293:80-83.

    Klastersky J, Sculier JP, Lacroix H, et al. A randomized study comparing cisplatin or carboplatin with etoposide in patients with advanced non-small-cell lung cancer: European Organization for Research and Treatment of Cancer Protocol 07861. J Clin Oncol 1990;8:1556-1562.

    Rosell R, Gatzemeier U, Betticher DC, et al. Phase III randomised trial comparing paclitaxel/carboplatin with paclitaxel/cisplatin in patients with advanced non-small-cell lung cancer: a cooperative multinational trial. Ann Oncol 2002;13:1539-1549.

    Mountain CF, Dresler CM. Regional lymph node classification for lung cancer staging. Chest 1997;111:1718-1723.

    Mountain CF. Revisions in the International System for Staging Lung Cancer. Chest 1997;111:1710-1717.

    McLoud TC, Bourgouin PM, Greenberg RW, et al. Bronchogenic carcinoma: analysis of staging in the mediastinum with CT by correlative lymph node mapping and sampling. Radiology 1992;182:319-323.

    Arita T, Kuramitsu T, Kawamura M, et al. Bronchogenic carcinoma: incidence of metastases to normal sized lymph nodes. Thorax 1995;50:1267-1269.

    Tahara RW, Lackner RP, Graver LM. Is there a role for routine mediastinoscopy in patients with peripheral T1 lung cancers? Am J Surg 2000;180:488-491.

    Pieterman RM, van Putten JWG, Meuzelaar JJ, et al. Preoperative staging of non-small-cell lung cancer with positron-emission tomography. N Engl J Med 2000;343:254-261.

    Weng E, Tran L, Rege S, et al. Accuracy and clinical impact of mediastinal lymph node staging with FDG-PET imaging in potentially resectable lung cancer. Am J Clin Oncol 2000;23:47-52.

    Mayr NA, Hussey DH, Yuh WT. Cost-effectiveness of high-contrast-dose MR screening of asymptomatic brain metastasis. AJNR Am J Neuroradiol 1995;16:215-217.

    Michel F, Soler M, Imhof E, Perruchoud AP. Initial staging of non-small cell lung cancer: value of routine radioisotope bone scanning. Thorax 1991;46:469-473.

    The Lung Cancer Study Group. Effects of postoperative mediastinal radiation on completely resected stage II and stage III epidermoid cancer of the lung. N Engl J Med 1986;315:1377-1381.

    Holmes EC, Gail M. Surgical adjuvant therapy for stage II and stage III adenocarcinoma and large-cell undifferentiated carcinoma. J Clin Oncol 1986;4:710-715.

    The benefit of adjuvant treatment for resected locally advanced non-small-cell lung cancer. J Clin Oncol 1988;6:9-17.

    Keller SM, Adak S, Wagner H, et al. A randomized trial of postoperative adjuvant therapy in patients with completely resected stage II or IIIA non-small-cell lung cancer. N Engl J Med 2000;343:1217-1222.

    The International Adjuvant Lung Cancer Trial Collaborative Group. Cisplatin-based adjuvant chemotherapy in patients with completely resected non-small-cell lung cancer. N Engl J Med 2004;350:351-360.

    Lafitte JJ, Ribet ME, Prevost BM, Gosselin BH, Copin MC, Brichet AH. Postresection irradiation for T2 N0 M0 non-small cell carcinoma: a prospective, randomized study. Ann Thorac Surg 1996;62:830-834.

    Dillman RO, Seagren SL, Propert KJ, et al. A randomized trial of induction chemotherapy plus high-dose radiation versus radiation alone in stage III non-small-cell lung cancer. N Engl J Med 1990;323:940-945.

    Dillman RO, Herndon J, Seagren SL, Eaton WL Jr, Green MR. Improved survival in stage III non-small-cell lung cancer: seven-year follow-up of Cancer and Leukemia Group B (CALGB) 8433 trial. J Natl Cancer Inst 1996;88:1210-1215.

    Curran WJ, Scott CB, Langer CJ, et al. Long-term benefit is observed in a phase III comparison of sequential vs. concurrent chemo-radiation for patients with unresected stage III NSCLC: RTOG 9410. Prog Proc Am Soc Clin Oncol 2003;22:621. abstract.

    Roth JA, Fossella F, Komaki R, et al. A randomized trial comparing perioperative chemotherapy and surgery with surgery alone in resectable stage IIIA non-small-cell lung cancer. J Natl Cancer Inst 1994;86:673-680.

    Roth JA, Atkinson EN, Fossella F, et al. Long-term follow-up of patients enrolled in a randomized trial comparing perioperative chemotherapy and surgery with surgery alone in resectable stage IIIA non-small-cell lung cancer. Lung Cancer 1998;21:1-6.

    Rosell R, Gómez-Codina J, Camps C, et al. A randomized trial comparing preoperative chemotherapy plus surgery with surgery alone in patients with non-small-cell lung cancer. N Engl J Med 1994;330:153-158.

    Rosell R, Gomez-Codina J, Camps C, et al. Preresectional chemotherapy in stage IIIA non-small-cell lung cancer: a 7-year assessment of a randomized controlled trial. Lung Cancer 1999;26:7-14.

    Depierre A, Milleron B, Moro-Sibilot D, et al. Preoperative chemotherapy followed by surgery compared with primary surgery in resectable stage I (except T1N0), II, and IIIa non-small-cell lung cancer. J Clin Oncol 2002;20:247-253.

    Schiller JH, Harrington D, Belani CP, et al. Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. N Engl J Med 2002;346:92-98.

    Johnson DH, Herbst R, Giaccone G, et al. ZD1839 ("Iressa") in combination with paclitaxel & carboplatin in chemotherapy-naive patients with advanced non-small-cell lung cancer (NSCLC): results from a phase III clinical trial (INTACT 2). Ann Oncol 2002;13:Suppl 5:127-128. abstract.

    Stephens RJ, Girling DJ, Bleehen NM, Moghissi K, Yosef HM, Machin D. The role of post-operative radiotherapy in non-small-cell lung cancer: a multicentre randomised trial in patients with pathologically staged T1-2, N1-2, M0 disease. Br J Cancer 1996;74:632-639.

    Debevec M, Bitenc M, Vidmar S, et al. Postoperative radiotherapy for radically resected N2 non-small-cell lung cancer (NSCLC): randomised clinical study 1988-1992. Lung Cancer 1996;14:99-107.

    PORT Meta-analysis Trialists Group. Postoperative radiotherapy in non-small-cell lung cancer: systematic review and meta-analysis of individual patient data from nine randomised controlled trials. Lancet 1998;352:257-263.

    Ohta M, Tsuchiya R, Shimoyama M, et al. Adjuvant chemotherapy for completely resected stage III non-small-cell lung cancer: results of a randomized prospective study. J Thorac Cardiovasc Surg 1993;106:703-708.

    Dautzenberg B, Chastang C, Arriagada R, et al. Adjuvant radiotherapy versus combined sequential chemotherapy followed by radiotherapy in the treatment of resected nonsmall cell lung carcinoma: a randomized trial of 267 patients. Cancer 1995;76:779-786.

    A randomized trial of postoperative adjuvant chemotherapy in non-small cell lung cancer (the second cooperative study). Eur J Surg Oncol 1995;21:69-77.

    Wada H, Hitomi S, Teramatsu T. Adjuvant chemotherapy after complete resection in non-small-cell lung cancer. J Clin Oncol 1996;14:1048-1054.

    Xu G, Rong T, Lin P. Adjuvant chemotherapy following radical surgery for non-small-cell lung cancer: a randomized study on 70 patients. Chin Med J (Engl) 2000;113:617-620.

    Chemotherapy in non-small cell lung cancer: a meta-analysis using updated data on individual patients from 52 randomised clinical trials. BMJ 1995;311:899-909.

    Perez CA, Pajak TF, Rubin P, et al. Long-term observations of the patterns of failure in patients with unresectable non-oat cell carcinoma of the lung treated with definitive radiotherapy: report by the Radiation Therapy Oncology Group. Cancer 1987;59:1874-1881.

    Le Chevalier T, Arriagada R, Quoix E, et al. Radiotherapy alone versus combined chemotherapy and radiotherapy in nonresectable non-small-cell lung cancer: first analysis of a randomized trial in 353 patients. J Natl Cancer Inst 1991;83:417-423.

    Schaake-Koning C, van den Bogaert W, Dalesio O, et al. Effects of concomitant cisplatin and radiotherapy on inoperable non-small-cell lung cancer. N Engl J Med 1992;326:524-530.

    Soresi E, Clerici M, Grilli R, et al. A randomized clinical trial comparing radiation therapy v radiation therapy plus cis-dichlorodiammine platinum (II) in the treatment of locally advanced non-small cell lung cancer. Semin Oncol 1988;15:Suppl 7:20-25.

    Blanke C, Ansari R, Mantravadi R, et al. Phase III trial of thoracic irradiation with or without cisplatin for locally advanced unresectable non-small-cell lung cancer: a Hoosier Oncology Group protocol. J Clin Oncol 1995;13:1425-1429.

    Morton RF, Jett JR, McGinnis WL, et al. Thoracic radiation therapy alone compared with combined chemoradiotherapy for locally unresectable non-small cell lung cancer: a randomized, phase III trial. Ann Intern Med 1991;115:681-686.

    Pritchard RS, Anthony SP. Chemotherapy plus radiotherapy compared with radiotherapy alone in the treatment of locally advanced, unresectable, non-small-cell lung cancer: a meta-analysis. Ann Intern Med 1996;125:723-729.

    Marino P, Preatoni A, Cantoni A. Randomized trials of radiotherapy alone versus combined chemotherapy and radiotherapy in stages IIIa and IIIb nonsmall cell lung cancer: a meta-analysis. Cancer 1995;76:593-601.

    Furuse K, Fukuoka M, Kawahara M, et al. Phase III study of concurrent versus sequential thoracic radiotherapy in combination with mitomycin, vindesine, and cisplatin in unresectable stage III non-small-cell lung cancer. J Clin Oncol 1999;17:2692-2699.

    Choy H, Akerley W, Safran H, et al. Multiinstitutional phase II trial of paclitaxel, carboplatin, and concurrent radiation therapy for locally advanced non-small-cell lung cancer. J Clin Oncol 1998;16:3316-3322.

    Langer CJ, Movsas B, Hudes R, et al. Induction paclitaxel and carboplatin followed by concurrent chemoradiotherapy in patients with unresectable, locally advanced non-small cell lung carcinoma: report of Fox Chase Cancer Center study 94-001. Semin Oncol 1997;24:Suppl 12:S12-89.

    Choy H, Curran WJ, Scott CB, et al. Preliminary report of locally advanced multimodality protocol (LAMP): ACR 427: a randomized phase II study of three chemo-radiation regimens with paclitaxel, carboplatin, and thoracic radiation (TRT) for patients with locally advanced non small cell lung cancer (LA-NSCLC). Prog Proc Am Soc Clin Oncol 2002;21:291a. abstract.

    Attar S, Krasna MJ, Sonett JR, et al. Superior sulcus (Pancoast) tumor: experience with 105 patients. Ann Thorac Surg 1998;66:193-198.

    Wright CD, Menard MT, Wain JC, et al. Induction chemoradiation compared with induction radiation for lung cancer involving the superior sulcus. Ann Thorac Surg 2002;73:1541-1544.

    Komaki R, Mountain CF, Holbert JM, et al. Superior sulcus tumors: treatment selection and results for 85 patients without metastasis (Mo) at presentation. Int J Radiat Oncol Biol Phys 1990;19:31-36.

    Rusch VW, Giroux DJ, Kraut MJ, et al. Induction chemoradiation and surgical resection for non-small cell lung carcinomas of the superior sulcus: initial results of Southwest Oncology Group Trial 9416 (Intergroup Trial 0160). J Thorac Cardiovasc Surg 2001;121:472-483.

    Barnes JB, Johnson SB, Dahiya RS, Temes RT, Herman TS, Thomas CR Jr. Concomitant weekly cisplatin and thoracic radiotherapy for Pancoast tumors of the lung: pilot experience of the San Antonio Cancer Institute. Am J Clin Oncol 2002;25:90-92.

    Gandhi S, Walsh GL, Komaki R, et al. A multidisciplinary surgical approach to superior sulcus tumors with vertebral invasion. Ann Thorac Surg 1999;68:1778-1784.

    Skarin A, Jochelson M, Sheldon T, et al. Neoadjuvant chemotherapy in marginally resectable stage III M0 non-small cell lung cancer: long-term follow-up in 41 patients. J Surg Oncol 1989;40:266-274.

    Pass HI, Pogrebniak HW, Steinberg SM, Mulshine J, Minna J. Randomized trial of neoadjuvant therapy for lung cancer: interim analysis. Ann Thorac Surg 1992;53:992-998.

    Pisters KM, Ginsberg RJ, Giroux DJ, et al. Induction chemotherapy before surgery for early-stage lung cancer: a novel approach. J Thorac Cardiovasc Surg 2000;119:429-439.

    Van Zandwijk N, Smit EF, Kramer GW, et al. Gemcitabine and cisplatin as induction regimen for patients with biopsy-proven stage IIIA N2 non-small-cell lung cancer: a phase II study of the European Organization for Research and Treatment of Cancer Lung Cancer Cooperative Group (EORTC 08955). J Clin Oncol 2000;18:2658-2664.

    Grilli R, Oxman AD, Julian JA. Chemotherapy for advanced non-small-cell lung cancer: how much benefit is enough? J Clin Oncol 1993;11:1866-1872.

    Helsing M, Bergman B, Thaning L, Hero U. Quality of life and survival in patients with advanced non-small cell lung cancer receiving supportive care plus chemotherapy with carboplatin and etoposide or supportive care only: a multicentre randomised phase III trial. Eur J Cancer 1998;34:1036-1044.

    Ellis PA, Smith IE, Hardy JR, et al. Symptom relief with MVP (mitomycin C, vinblastine and cisplatin) chemotherapy in advanced non-small-cell lung cancer. Br J Cancer 1995;71:366-370.

    Cullen MH, Billingham LJ, Woodroffe CM, et al. Mitomycin, ifosfamide, and cisplatin in unresectable non-small-cell lung cancer: effects on survival and quality of life. J Clin Oncol 1999;17:3188-3194.

    Ruckdeschel JC, Finkelstein DM, Ettinger DS, et al. A randomized trial of the four most active regimens for metastatic non-small-cell lung cancer. J Clin Oncol 1986;4:14-22.

    Ruckdeschel JC, Finkelstein DM, Mason BA, Creech RH. Chemotherapy for metastatic non-small-cell bronchogenic carcinoma: EST 2575, generation V -- a randomized comparison of four cisplatin-containing regimens. J Clin Oncol 1985;3:72-79.

    Sandler AB, Nemunaitis J, Denham C, et al. Phase III trial of gemcitabine plus cisplatin versus cisplatin alone in patients with locally advanced or metastatic non-small-cell lung cancer. J Clin Oncol 2000;18:122-130.

    Gatzemeier U, von Pawel J, Gottfried M, et al. Phase III comparative study of high-dose cisplatin versus a combination of paclitaxel and cisplatin in patients with advanced non-small-cell lung cancer. J Clin Oncol 2000;18:3390-3399.

    von Pawel J, von Roemeling R, Gatzemeier U, et al. Tirapazamine plus cisplatin versus cisplatin in advanced non-small-cell lung cancer: a report of the international CATAPULT I study group. J Clin Oncol 2000;18:1351-1359.

    Wozniak AJ, Crowley JJ, Balcerzak SP, et al. Randomized trial comparing cisplatin with cisplatin plus vinorelbine in the treatment of advanced non-small-cell lung cancer: a Southwest Oncology Group study. J Clin Oncol 1998;16:2459-2465.

    Sederholm C. Gemcitabine (G) compared with gemcitabine plus carboplatin (GC) in advanced non-small cell lung cancer (NSCLC): a phase III study by the Swedish Lung Cancer Study Group (SLUSG). Prog Proc Am Soc Clin Oncol 2002;21:291a. abstract.

    Lilenbaum RC, Herndon J, List M, et al. Single-agent (SA) versus combination chemotherapy (CC) in advanced non-small cell lung cancer (NSCLC): a CALGB randomized trial of efficacy, quality of life (QOL) and cost-effectiveness. Prog Proc Am Soc Clin Oncol 2002;21:1a. abstract.

    Georgoulias V, Ardavanis A, Agelidou M, et al. Preliminary analysis of a multicenter phase III trial comparing docetaxel (D) versus docetaxel/cisplatin (DC) in patients with inoperable advanced and metastatic non-small cell lung cancer (NSCLC). Prog Proc Am Soc Clin Oncol 2002;21:291a. abstract.

    Bonomi P, Kim K, Fairclough D, et al. Comparison of survival and quality of life in advanced non-small-cell lung cancer patients treated with two dose levels of paclitaxel combined with cisplatin versus etoposide with cisplatin: results of an Eastern Cooperative Oncology Group trial. J Clin Oncol 2000;18:623-631.

    Kosmidis P, Mylonakis N, Nicolaides C, et al. Paclitaxel plus carboplatin versus gemcitabine plus paclitaxel in advanced non-small-cell lung cancer: a phase III randomized trial. J Clin Oncol 2002;20:3578-3585.

    Kelly K, Crowley J, Bunn PA Jr, et al. Randomized phase III trial of paclitaxel plus carboplatin versus vinorelbine plus cisplatin in the treatment of patients with advanced non-small-cell lung cancer: a Southwest Oncology Group trial. J Clin Oncol 2001;19:3210-3218.

    Ettinger DS. Is there a preferred combination chemotherapy regimen for metastatic non-small cell lung cancer? Oncologist 2002;7:226-233.

    Effects of vinorelbine on quality of life and survival of elderly patients with advanced non-small-cell lung cancer. J Natl Cancer Inst 1999;91:66-72.

    Gridelli C, Perrone F, Gallo C, et al. Chemotherapy for elderly patients with advanced non-small-cell lung cancer: the Multicenter Italian Lung Cancer in the Elderly Study (MILES) phase III randomized trial. J Natl Cancer Inst 2003;95:362-372.

    Kelly K, Mikhaeel-Kamel N, Pan Z, Murphy J, Prindiville S, Bunn PA Jr. A phase I/II trial of paclitaxel, carboplatin, and gemcitabine in untreated patients with advanced non-small cell lung cancer. Clin Cancer Res 2000;6:3474-3479.

    Frasci G, Panza N, Comella P, et al. Cisplatin, gemcitabine, and paclitaxel in locally advanced or metastatic non-small-cell lung cancer: a phase I-II study. J Clin Oncol 1999;17:2316-2325.

    Smith IE, O'Brien ME, Talbot DC, et al. Duration of chemotherapy in advanced non-small-cell lung cancer: a randomized trial of three versus six courses of mitomycin, vinblastine, and cisplatin. J Clin Oncol 2001;19:1336-1343.

    Depierre A, Quoix E, Mercier M, et al. Maintenance chemotherapy in advanced non-small cell lung cancer (NSCLC): a randomized study of vinorelbine (V) versus observation (OB) in patients (Pts) responding to induction therapy. Prog Proc Am Soc Clin Oncol 2001;20:309a. abstract.

    Socinski MA, Schell MJ, Peterman A, et al. Phase III trial comparing a defined duration of therapy versus continuous therapy followed by second-line therapy in advanced-stage IIIB/IV non-small-cell lung cancer. J Clin Oncol 2002;20:1335-1343.

    Huisman C, Smit EF, Giaccone G, Postmus PE. Second-line chemotherapy in relapsing or refractory non-small-cell lung cancer: a review. J Clin Oncol 2000;18:3722-3730.

    Shepherd FA, Dancey J, Ramlau R, et al. Prospective randomized trial of docetaxel versus best supportive care in patients with non-small-cell lung cancer previously treated with platinum-based chemotherapy. J Clin Oncol 2000;18:2095-2103.

    Fossella FV, DeVore R, Kerr RN, et al. Randomized phase III trial of docetaxel versus vinorelbine or ifosfamide in patients with advanced non-small-cell lung cancer previously treated with platinum-containing chemotherapy regimens. J Clin Oncol 2000;18:2354-2362.

    Magilligan DJ Jr, Duvernoy C, Malik G, Lewis JW Jr, Knighton R, Ausman JI. Surgical approach to lung cancer with solitary cerebral metastasis: twenty-five years' experience. Ann Thorac Surg 1986;42:360-364.

    Patchell RA, Tibbs PA, Walsh JW, et al. A randomized trial of surgery in the treatment of single metastases to the brain. N Engl J Med 1990;322:494-500.

    Raviv G, Klein E, Yellin A, Schneebaum S, Ben-Ari G. Surgical treatment of solitary adrenal metastases from lung carcinoma. J Surg Oncol 1990;43:123-124.

    Reyes L, Parvez Z, Nemoto T, Regal AM, Takita H. Adrenalectomy for adrenal metastasis from lung carcinoma. J Surg Oncol 1990;44:32-34.

    Simon G, Ginsberg RJ, Ruckdeschel JC. Small-cell lung cancer. Chest Surg Clin N Am 2001;11:165-188.

    Argiris A, Murren JR. Staging and clinical prognostic factors for small-cell lung cancer. Cancer J 2001;7:437-447.

    Inoue M, Nakagawa K, Fujiwara K, Fukuhara K, Yasumitsu T. Results of preoperative mediastinoscopy for small cell lung cancer. Ann Thorac Surg 2000;70:1620-1623.

    Inoue M, Miyoshi S, Yasumitsu T, et al. Surgical results for small cell lung cancer based on the new TNM staging system. Ann Thorac Surg 2000;70:1615-1619.

    Aisner J, Alberto P, Bitran J, et al. Role of chemotherapy in small cell lung cancer: a consensus report of the International Association for the Study of Lung Cancer workshop. Cancer Treat Rep 1983;67:37-43.

    Seifter EJ, Ihde DC. Therapy of small cell lung cancer: a perspective on two decades of clinical research. Semin Oncol 1988;15:278-299.

    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;83:855-861.

    Roth BJ, Johnson DH, Einhorn LH, et al. Randomized study of cyclophosphamide, doxorubicin, and vincristine versus etoposide and cisplatin versus alternation of these two regimens in extensive small-cell lung cancer: a phase III trial of the Southeastern Cancer Study Group. J Clin Oncol 1992;10:282-291.

    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.

    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;20:3054-3060.

    Turrisi AT III, 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;340:265-271.

    Pignon J-P, Arriagada R, Ihde DC, et al. A meta-analysis of thoracic radiotherapy for small-cell lung cancer. N Engl J Med 1992;327:1618-1624.

    Arriagada R, Pignon JP, Ihde DC, et al. Effect of thoracic radiotherapy on mortality in limited small cell lung cancer: a meta-analysis of 13 randomized trials among 2,140 patients. Anticancer Res 1994;14:333-335.

    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;11:336-344.

    Lad T, Piantadosi S, Thomas P, Payne D, Ruckdeschel J, Giaccone G. A prospective randomized trial to determine the benefit of surgical resection of residual disease following response of small cell lung cancer to combination chemotherapy. Chest 1994;106:Suppl:320S-323S.

    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;87:183-190.

    Komaki R, Cox JD, Whitson W. Risk of brain metastasis from small cell carcinoma of the lung related to length of survival and prophylactic irradiation. Cancer Treat Rep 1981;65:811-814.

    Aupérin A, Arriagada R, Pignon J-P, et al. Prophylactic cranial irradiation for patients with small-cell lung cancer in complete remission. N Engl J Med 1999;341:476-484.

    Johnson BE. NCCN: small cell lung cancer. Cancer Control 2001;8:Suppl 2:32-43.

    Kosmidis PA, Samantas E, Fountzilas G, Pavlidis N, Apostolopoulou F, Skarlos D. Cisplatin/etoposide versus carboplatin/etoposide chemotherapy and irradiation in small cell lung cancer: a randomized phase III study: Hellenic Cooperative Oncology Group for Lung Cancer Trials. Semin Oncol 1994;21:Suppl 6:23-30.

    Niell HB, Herndon JE, Miller AA, et al. Randomized Phase III intergroup trial (CALGB 9732) of etoposide (VP-16) and cisplatin (DDP) with or without paclitaxel (TAX) and G-CSF in patients with extensive stage small cell lung cancer (ED-SCLC). Prog Proc Am Soc Clin Oncol 2002;21:293a. abstract.

    Mavroudis D, Papadakis E, Veslemes M, et al. A multicenter randomized clinical trial comparing paclitaxel-cisplatin-etoposide versus cisplatin-etoposide as first-line treatment in patients with small-cell lung cancer. Ann Oncol 2001;12:463-470.

    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;15:2090-2096.

    Salgia R, Skarin AT. Molecular abnormalities in lung cancer. J Clin Oncol 1998;16:1207-1217.

    Fong KM, Sekido Y, Minna JD. Molecular pathogenesis of lung cancer. J Thorac Cardiovasc Surg 1999;118:1136-1152.

    Dy GK, Adjei AA. Novel targets for lung cancer therapy. J Clin Oncol 2002;20:2881-2894.

    Fukuoka M, Yano S, Giaccone G, et al. Final results from a phase II trial of ZD1839 (`Iressa') for patients with advanced non-small-cell lung cancer (IDEAL 1). Prog Proc Am Soc Clin Oncol 2002;21:298a. abstract.

    Carney DN. Lung cancer -- time to move on from chemotherapy. N Engl J Med 2002;346:126-128.(Alexander Spira, M.D., Ph)