Skip to main content
Annals of Saudi Medicine logoLink to Annals of Saudi Medicine
. 2005 Jan-Feb;25(1):1–12. doi: 10.5144/0256-4947.2005.1

Lung Cancer

Irfan Maghfoor *,, Michael C Perry
PMCID: PMC6150570  PMID: 15822487

Summary

Lung cancer is the leading cause of cancer-related mortality. Since tobacco smoking is the cause in vast majority of cases, the incidence of lung cancer is expected to rise in those countries with high or rising incidence of tobacco smoking. Even though populations at risk of developing lung cancer are easily identified, mass screening for lung cancer is not supported by currently available evidence. In case of non-small cell lung cancer, a cure may be possible with surgical resection followed by post-operative chemotherapy in those diagnosed at an early stage. A small minority of patients who present with locally advanced disease may also benefit from pre-operative chemotherapy and/or radiation therapy to down stage the tumor to render is potentially operable. In a vast majority of patients, however, lung cancer presents at an advanced stage and a cure is not possible with currently available therapeutic strategies. Similarly, small cell lung cancer confined to one hemi-thorax may be curable with a combination of chemotherapy and thoracic irradiation followed by prophylactic cranial irradiation, if complete remission is achieved at the primary site. Small cell lung cancer that is spread beyond the confines of one hemi-thorax is, however, considered incurable. In this era of molecular targeted therapies, new agents are constantly undergoing pre-clinical and clinical testing with the aim of targeting the molecular pathways thought be involved in etiology and pathogenesis of lung cancer.


The World Health Organization reported 6 million cancers worldwide in Year 2000. Lung cancer accounted for one million, one sixth of these, representing the most common cancer globally.1 In the United States, one of the countries where accurate and consistent statistics are available, there will be an estimated 186 550 new lung cancers in Year 2004 with estimated 165 130 deaths.2 Lung cancer accounts for 13% of all cancers in males and 12% of all cancers in females in the US. However, it is responsible for 32% and 25% of all cancer-related deaths in males and females, respectively, representing the largest single cause of smoking related mortality.2 Despite major financial and research efforts of the past two decades, the five-year survival for lung cancer has remained constant at a dismal 14% in the Western world.3 Only those presenting at an early stage appear to have a chance at cure.

The age-specific incidence rate for lung cancer in Saudi Arabia is 25 per 100 000 population (Figure 1). Lung cancer is the 4th most common cancer among males and 7th among females.4 Considering the incidence of smoking in males, this number appears small and may represent a lag time between the increased incidence of smoking and the rise in the incidence of lung cancer. It is thus expected that in coming years lung cancer will represent a major public health problem in the Kingdom of Saudi Arabia. Concerted efforts are thus needed not only to educate the general public regarding the hazards of smoking and early symptoms and signs of the disease, but also health care providers to not only educate the public but also to diligently diagnose lung cancer at its early and potentially curable stage. This article will thus attempt to briefly analyze etiology, prevention, and screening strategies as well as new advances in management.

Figure 1.

Figure 1

Average age-specific incidence rate (AIR) for lung cancer, Saudi Arabia, 1994–2000.

Etiology, Pathogenesis & Pathology

Smoking is the major etiologic factor for lung cancer, with approximately 90% to 95% of new lung cancers resulting from active smoking.5,6 Among non-smokers a quarter of lung cancers are caused by second hand smoke (passive smoking).79 Since only 10% of the smokers develop lung cancer, other factors seem to play a part.10 These might include the genetic make-up of an individual as well as other environmental insults such as asbestos exposure. Other etiologic factors include exposure to asbestos11 or other environmental agents like silica,12 beryllium,13 nickel,14 diesel exhaust,15 or radon,11 although the data regarding these, except for asbestos exposure are conflicting. Lung cancer starts with premalignant changes in the epithelium and develops from squamous metaplasia through carcinoma in situ to frank invasive cancer.16 Environmental factors are thought to be critical in pathogenesis, although they may not be the sole cause.

The World Health Organization divides lung malignancies into several different sub-types but for clinicians it may be divided into two broad categories, i.e. small cell and non-small cell lung cancers, the latter encompassing adenocarcinoma, squamous cell, large cell, and bronchoalveolar carcinomas.

Prevention and Screening

Lung cancer is one of the few cancers with a well-defined etiology—inhalation of tobacco smoke. In addition to first hand smoke, the United States Environmental Protection Agency has also identified passive (second hand) tobacco inhalation to be a carcinogen. At least 24 studies have identified second hand smoke as a risk factor for development of lung cancer, with a relative risk as high as 2-fold in some studies.79 Clearly, cigarette smoking is the most prevalent environmental carcinogen and concerted efforts are required by governments as well as health care providers to support smoking cessation and prevention programs.

Only 10% of all smokers develop lung cancer. Therefore, other factors play a part in the development of lung cancer in addition to tobacco smoke. An individual’s genetic susceptibility to the carcinogens present in tobacco smoke may be one such factor.17 Investigators have identified several enzymatic pathways that may be involved in activation, degradation of carcinogens, and subsequent DNA repair.18 Members of cytochrome P450,1922 glutathione S-transferase,2325 epoxide hydrolase,26 and N-acetyltransferase25,27,28 are involved in activation and degradation of certain carcinogens found in tobacco smoke. Genes encoding these enzymes may display deletions, mutations, or polymorphisms, thereby affecting the enzyme’s activity.

Certain dietary components have also been linked to an increased risk of lung cancer.29,30 The carotenoids were the first such group identified to have an inverse correlation with lung cancer risk.31 Several epidemiologic studies have suggested that individuals with diets low in beta-carotene have a higher risk of developing lung cancer.30,32,33 Other dietary micronutrients like flavonoids34 and isothiocyanates35,36 have also been suggested to have inverse correlation with lung cancer risk.

Chemoprevention is an area of intense investigation. Research to date has focused on two separate classes of agents, i.e. dietary micronutrients or their synthetic analogues, and synthetic agents such as NSAIDs.37 Retinoids have been extensively studied in this regard. One of the first studies to report positive results showed a statistically significant decrease in the incidence of second tobacco-related malignancies (head and neck, lung, esophagus) in those diagnosed with squamous cell carcinoma of head and neck and treated with cis-retinoic acid.38 A US intergroup trial studied 13-cis retinoic acid in patients with resected stage I lung cancer and found an increase in the death rate among those on supplementation.39 It also found an increased death rate among those who continued to smoke.39 13-cis retinoic acid has also been found not to influence squamous metaplasia in another study.40

Beta-carotene as a lung cancer prevention agent has been reported in two large randomized trials–ATBC (alpha tocopherol beta carotene trial)41 and CARET (carotene and retinol efficacy) trials.42 Both of these trials found a detrimental effect of beta-carotene in individuals on supplemental beta-carotene, showing a higher risk of lung cancer. This harmful effect was increased in heavy smokers41,42 and clearly established that beta-carotene is harmful in smokers. Other dietary supplements have been reported to have a beneficial effect on lung cancer risk and are currently being tested in randomized studies.

Diagnosis

Lung cancer should be suspected in chronic smokers with protracted or new onset respiratory symptoms. The work-up usually starts with a chest X-Ray and if abnormal should lead to a computer tomographic scan. Further diagnostic work-up depends upon the location of the tumor–central tumors are best approached with a bronchoscopy, which allows for direct visualization of the airways as well as tumor, and permits biopsy material to be obtained for histologic diagnosis at the same time. Peripheral tumors can be biopsied via a transthoracic approach under CT or ultrasound guidance. Further invasive techniques including thoracoscopy, mediastinoscopy, and thoracotomy are reserved for those in whom the initial diagnostic techniques mentioned above fail to yield a diagnosis. Even though sputum cytology is a non-invasive method of establishing a diagnosis, negative sputum cytology does not rule out lung cancer. In experienced hands sensitivity and specificity of sputum cytology in diagnosis of lung cancer may approach 0.66 and 0.99 respectively.43

Staging and Prognosis

The prognosis and management of lung cancer is critically dependent on appropriate staging. The work-up should include a detailed history and physical examination, chest X-ray, computed tomographic scan of chest and upper abdomen including liver and adrenals, serum chemistries including renal and liver function tests, serum calcium, and alkaline phosphatase. If history, physical examination or blood tests (calcium, alkaline phosphatase) suggest bone or central nervous system spread, a bone scan and CT scan of the brain should be obtained.44 Pulmonary function testing and invasive mediastinal staging are reserved for those being considered for curative surgical resection or radical radiation therapy. Staging of non-small cell lung cancer (NSCLC) utilizes the Tumor, Node Metastases (TNM) system as revised in 1997 (Table 1).45

Table 1.

TNM (Tumor, Node, Metastases) staging of non-small cell lung cancer.

Stage Group TNM Description 5-year survival
0 Carcinoma in situ
IA T1N0M0
  • -Tumor <3 cm surrounded by lung or visceral

  • -Pleura, not more proximal than labor bronchus (T1)

  • -No lymph nodes or distant metastases

67%
IB T2N0M0
  • -Tumor >3 cm or involves mainstem bronchus

  • ->2 cm from carina

  • -Invades visceral pleura

  • -Lobar atalectasis (not entire lung atalectasis) (T2)

57%
IIA T1N1M0
  • -T1 with involvement of ipsilateral hilar or peribronchial lymph

  • -Nodes or direct extension to intrapulmonary nodes (N1)

  • -No metastases

55%
IIB TN1M0
  • -T2 & N1 as defined

  • -No metastases

39%
T3N0M0
  • -Direct invasion by tumor of chest wall, diaphragm, mediastinal pleura, parietal pericardium

  • -In mainstem bronchus <2 cm from carina

  • -Atalectasis of entire lung (T3)

  • -No lymph nodes or metastases

38%
IIIA T3N1M0
  • -T3 & N1 as defined

  • -No metastases

25%
T1-3N2M0
  • -T1, T2, T3 as defined

  • -Involvement of ipsilateral mediastinal or subcarinal nodes (N2)

  • -No metastases

23%
IIIB T4anyNM0
  • -Involvement of heart, mediastinum, great vessels, trachea, esophagus, vertebral body, carina

  • -Malignant pleural or pericardial effusion

  • -Satellite tumor nodule in ipsilateral tumor bearing lobe of the lung (T4)

7%
AnyTN3M0 Metastases to contralateral mediastinal, or hilar nodes, ipsilateral or contralateral scalene nodes or supraclavicuar nodes 3%
IV AnyT
AnyN
M1
Presence of distant metastases 1%

Adapted from Mountain, CF45 and Mountain, CF & Dressler CM132

The prognosis of NSCLC depends upon extent of disease (stage), performance status, and weight loss. As alluded to before, despite extensive research the 5-year overall survival for NSCLC has remained remarkably constant at 14% in past the 20 years. Even in stage I patients undergoing curative surgical resection, the 5-year survival is only 60%.

Management of NSCLC

Surgical resection is the only potentially curative treatment of NSCLC. Unfortunately, it can only be offered to a minority of patients since approximately two-thirds to three-fourths of patients with NSCLC will present with advanced and potentially unresectable disease. Management depends upon the stage of disease, and surgery can be offered to those patients presenting with stage IA to stage IIIA disease who can tolerate surgery. Those with more advanced stage disease are offered chemotherapy, radiation therapy, or chemoradiotherapy. Some patients presenting with stage III disease, particularly those with stage IIIA, may be offered pre-operative chemotherapy or chemoradiotherapy to downstage the disease in an effort to render the disease surgically resectable.

Surgery

Even though surgery is curative, only one-third to one-fourth of patients present with an early enough stage to be offered surgical resection. In addition, comorbid conditions, especially those caused by tobacco smoking, namely coronary artery disease and chronic obstructive pulmonary disease, may make surgical resection technically difficult or impossible. Patients being considered for surgical resection undergo a more rigorous pre-operative work-up, which may include computed tomographic or magnetic resonance imaging of the brain as well as positron emission tomography, in addition to pulmonary function testing.

Several retrospective and prospective studies have revealed a sensitivity and specificity of 85% and 88%, respectively, for mediastinal lymph node involvement with positron emission tomography (PET).46 The sensitivity and specificity are even higher with use of both computer tomography and PET.47

Pulmonary function testing with a predicted postoperative residual forced expiratory volume (FEV1) of at least one liter is mandatory before considering surgical resection. Adequate surgical resections include lobectomy and/or pneumonectomy.

The role of post-operative radiation therapy remains controversial, and while some studies suggest a decrease in local recurrence rate without any impact on overall survival,48 others have reported a detrimental effect on overall survival from post-operative radiation therapy with a 21% increase in the relative risk of death particularly in those with resected stage I disease.49 The decision to offer post-operative radiation therapy should be individualized but should be strongly considered for those with positive or very close surgical resection margins or those with N2 disease.

Until recently post-operative chemotherapy did not have a defined role after surgical resection, but recently 4 large randomized controlled trials and a meta-analysis have demonstrated a clear benefit with post-operative chemotherapy.5053

Radiation Therapy

Radiation therapy in the management of NSCLC may be offered with a curative intent to those with early stage disease deemed medically unfit to undergo anesthesia and/or surgical resection.54,55 Patients with stages I–II treated with radiation therapy alone at a dose of at least 6000 cGy may achieve 5-year survivals of 10% to 27%.55,56

For patients with stage III disease, those with unresectable stage IIIA or stage IIIB disease excluding malignant pleural effusion, radiation therapy along with chemotherapy constitutes the standard of care (Table 3).57,58 Until recently a sequential approach with two cycles of cisplatin-based chemotherapy followed by radiation was considered standard of care based on data published by the Cancer and Leukemia Group B (CALGB) and the Radiation Therapy Oncology Group (RTOG).59,60 However, the RTOG has reported the results of a randomized trial comparing this sequential approach to concurrent chemo-radiotherapy using the same chemotherapy with an improvement in median survival from 14 months with a sequential approach to 17 months with a concurrent approach.61 A Japanese randomized trial using mitomycin, vindesine, and cisplatin has also shown improvements in 2-, 3-, 4-, and 5-year survival when radiatiotherapy was delivered concurrently with chemotherapy compared to a sequential approach.62 The toxicity is understandably higher but concurrent chemoradiotherapy should be considered for patients with unresectable stage IIIA and IIIB patients with good performance status.

Table 3.

Selected studies of chemotherapy and biologic therapy in advanced disease.

Study Treatment Remarks
Chemotherapy and radiotherapy in inoperable disease
Dillman57 Cisplatin, vinblastine and radiotherapy vs. radiotherapy alone Improved 1-,2-,3-,7-year survival with chemotherapy
Sause58 Cisplatin, vinblastine and radiotherapy vs. radiotherapy Improved survival with chemotherapy
Curran61 Cisplatin, vinblastine given concurrently with radiotherapy vs. sequentially Improved median survival with concurrent approach
Chemotherapy for advanced disease
Schiller81 Carboplatin and paclitaxel vs. cisplatin and paclitaxel, cisplatin and gemcitaine, and cisplatin and docetaxel Overall survival equal in all groups, longer progression survival but more toxicity in gemcitabine arm
Biological therapy in advanced disease
IDEAL 1103,104 Gefitinib as second line vs. observation 20% response rate, symptom improvement
IDEAL2138 Gefitinib as 3rd and 4th line vs. observation 10% response rate symptom improvement
INTACT 1 and 2106,107 Chemotherapy vs. chemotherapy and gefinitib No benefit with addition of gefinitib
TRIBUTE108 Chemotherapy vs. chemotherapy and erlotinib (OSI-774) No benefit with addition of erlotinib in first line setting
TALENT109 Chemotherapy vs. chemotherapy and erlotinib (OSI-774) No benefit with addition of erlotinib in first line setting

Radiation therapy is an excellent palliative option for control of pain, hemoptysis, and bronchial obstruction with post-obstructive pneumonia.

Chemotherapy

As mentioned, chemotherapy is not only offered in the post-operative adjuvant setting and as a neo-adjuvant approach to down-stage the disease but, a large majority of those who present with advanced stage (stage IIIB and IV) as well as those who relapse after resection, are offered this modality. As such, as many as 80% to 90% of all NSCLC patients may become candidates for chemotherapy during the course of their disease.

Pre-operative Chemotherapy

Pre-operative (neo-adjuvant, induction) chemotherapy may be offered to a selected group of patients with marginally respectable tumors, who have good performance status. Four randomized trials have addressed this issue (Table 2). Two of these were too small to draw useful conclusions63,64 but studies by Rosell65 and Roth66 demonstrated an improvement in 2- and 5- year survival in those receiving pre-operative chemotherapy. Updated results from both these studies have been reported and show a benefit with neo-adjuvant chemotherapy on long-term follow-up.67,68 Despite limitations of these studies, it may be appropriate to offer 2 to 3 cycles of pre-operative chemotherapy to this patient group if surgical resection is deemed feasible with down-staging. Several ongoing randomized trials are addressing the issue of neo-adjuvant chemotherapy for earlier stage disease since a French trial by Depierre et.al. suggested a benefit for those patients with earlier stages treated with this approach.69

Table 2.

Selected studies of neo-adjuvant and adjuvant strategies in non-small cell lung cancer

Study Treatment Comments
Neo-adjuvant Chemotherapy
Roth66 Surgery and radiotherapy vs. cisplatin and etoposide before and after surgery Increased 3-year survival with chemotherapy (56% vs. 15%)
Rosell133 Surgery and radiotherapy vs. mitomycin, ifosfamide and cisplatin before surgery and radiotherapy Increased median survival with chemotherapy (26 mo vs. 8 mo). Lower then expected survival in surgery arm
Depierre69 Mitomycin, ifosfamide, cisplatin before surgery and radiotherapy vs. surgery and radiotherapy Survival benefit at 1 and 4 years in early stage (N0 and N1). No benefit in N2
Adjuvant chemotherapy
Holmes134 Cyclophosphamide, adriamycin, cisplatin vs. immunotherapy Improved survival with adjuvant chemotherapy
Lung Cancer Study Group135 Cyclophosphamide, adriamycin cisplatin and radiotherapy vs. radiotherapy Improved 1-year survival with adjuvant chemotherapy
Keller136 Cisplatin, etoposide and radiotherapy vs radiotherapy No benefit with chemotherapy
Tonato137 Mitomycin, vindesine, cisplatin vs. observation Stage I, II IIIA. At median follow up of 63 month no benefit in overall or event free survival
International Adjuvant Lung Cancer Trial50 Cisplatin based chemotherapy vs. observation 4% absolute increase in survival at 5 years
Strauss70 Paclitaxel and carboplatin vs. observation Stage IB only. Overall survival at 4 years 71% vs. 59% in favor of adjuvant chemotherapy
Winton53 Cisplatin and vinorelbine vs. observation Stage IB and II. Overall survival 94 mo vs. 73 mo in favor of adjuvant chemotherapy
Hamada51 UFT vs. observation 5- and 7-year survival rates improved with UFT

Post-operative chemotherapy

Recently, several randomized trials have conclusively provided conclusive evidence for a survival benefit with adjuvant chemotherapy after surgical resection (Table 2). The largest of these is the International Adjuvant Lung Trial (IALT) that included 1867 patients. 50 The accrual was stopped early with slowing of accrual as well as evidence of benefit on interim analysis. Adjuvant cisplatin based chemotherapy resulted in a 4.1% increase in overall survival at 5 years. There was also an improvement in disease-free survival from 34.3% to 39.4% at five years. Other trials demonstrating survival benefit with adjuvant chemotherapy include a Japanese randomized trial as well as a meta-analysis in stages IB and II adenocarcinoma in which post-operative fluorouracil-tegafur (UFT) was given orally for 2 years post-operatively.51 The results are, however, not applicable at present outside Japan. Other more relevant studies include the CALGB and National Cancer Institute of Canada (NCIC) trials. The NCIC trial reported by Winton et al at the 40th annual meeting of American Society of Clinical Oncology in June, 2004 randomized patients with stage IB (T2N0) and stage II (excluding T3N0), after resection, to fourcycles of vinorelbine and cisplatin versus observation. In this trial overall survival was significantly prolonged in the adjuvant chemotherapy group (94 months vs. 73 months; HR=0.69, P=0.011). Relapse-free survival was also prolonged (not yet reached for adjuvant chemotherapy arm versus 46.7 months; HR=0.6, P=0.0003).53 In the same meeting Strauss et al reported a CALGB trial in which patients with resected stage IB (T2N0) NSCLC were randomized to four cycles of paclitaxel and carboplatin versus observation. The hazard ratio for death from any cause was 0.62 (P=0.028) in favor of adjuvant chemotherapy. Overall survival at 4 years was 71% versus 59% in the chemotherapy and observation arms, respectively. Failure-free survival and lung cancer mortality were similarly improved with adjuvant chemotherapy. The hazard ratio for failure-free survival was 0.69 (P=0.035) in favor of the chemotherapy group and 4-year lung cancer mortality was 16% in the chemotherapy group versus 26% in the observation group.70 In summary, platinum based adjuvant chemotherapy for three to four cycles may be considered standard of care after resection of stages IB to IIIA NSCLC.

Chemotherapy for Advanced Disease

Non small cell lung cancer presenting with stage IIIB and IV disease and relapsed NSCLC, with few exceptions, is incurable. In the past, few patients were offered chemotherapy for advanced NSCLC due to low response rates and toxicity. However, several studies, including meta-analyses, have demonstrated a modest gain of 2 to 4 months in median survival and a 1-year survival ranging from 10% to 20%.7173 Other studies have reported improvement of quality of life with chemotherapy which is highly significant in this patient population with limited survival.7476 This benefit is often restricted, however, to those who maintain a good performance status.

Several agents have shown single agent activity in NSCLC, but platinum has been used almost exclusively in single-agent chemotherapy for NSCLC. Phase III trials carried out in early 1990s, however, showed that cisplatin or carboplatin, when combined with second- and third-generation chemotherapeutic agents (paclitaxel, docetaxel, vinorelbine, gemcitabine) have a higher response rate, time-to-disease progression and, overall survival.7779 The Eastern Co-operative Oncology Group (ECOG) was the first to show the superiority of a newer generation platinum combination (paclitaxel and carboplatinum) over cisplatin and etoposide.80 In 2001, Schiller et al reported the results of a large phase III randomized trial showing equal efficacy of four third-generation platinum combinations (Table 3).81 A recent meta-analysis has, however, reported a slight superiority of gemcitabine-containing regimens over other platinum-based combinations.82 It is appropriate to offer any of these two-drug combinations as first-line chemotherapy in this setting.83 At present, no data exists to support a three-drug combination, which may offer a higher response rate with higher toxicity and without a survival benefit.84,85

The optimum duration of chemotherapy is another controversial issue and it is common practice to offer six cycles of chemotherapy to responding patients. At least three randomized trials have addressed this issue, and it appears that prolonging chemotherapy beyond three to four cycles adds toxicity without additional clinical benefit.8688

Fit elderly patients seem to derive similar benefit from chemotherapy without increased toxicity and therefore should be offered similar therapeutic options as the younger patients.8991

Patients who have responded to first-line platinum-based chemotherapy and maintain a good performance status can be offered second line docetaxel.92,93 A recently reported phase III randomized trial has shown similar efficacy with pemetrexed in this setting.94 Recently reported data also suggests that gemcitabine may have activity in second-line setting.95,96

Biologic Therapy

Non-small cell lung cancer cells over-express epidermal growth factor receptor (EGFR),97,98 which provides these cancer cells a selective growth advantage with increased ability to grow, induce angiogenesis, and metastasize.99101 This results from activation of EGFR after binding to its ligand, which leads to phosporylation of the intra-cellular domain of the receptor. This leads to an increase in EGFR tyrosine kinase activity, resulting in an intra-cellular cascade of events leading to an increase in cell proliferation, a decrease in apoptotic potential, increased angiogenesis, and metastatic potential. Strategies to block this EGFR dependent tyrosine kinase pathway have included monoclonal antibodies, anti-sense oligonucleotides, and small molecules that block phosphorylation of the intra-cellular domain of EGFR receptor. Gefitinib (ZD1839, IRESSA) is the first such compound102 and has shown a 10% to 20% activity in heavily pre-treated patients.103,104 Most importantly, the responses are seen regardless of the level of EGFR expression.105 Two large phase II trials in North America and Europe led to Food and Drug Administration (FDA) approval of gefitinib as a third-line agent (Table 3). Gefitinib has shown improvement in symptoms in up to 50% of patients.104 Orally administered, the major toxicities are skin rash and diarrhea and an approximately 1% risk of developing potentially fatal interstitial lung disease (mostly in Japanese patients). Interestingly, development of some of these side effects, especially rash, may predict response to the agent, but this is not conclusive.

The addition of gefitinib to first-line chemotherapy, on the other hand, has failed to show any benefit.106,107 Similarly, the addition of erlotinib (EGFR inhibitor) to carboplatin and paclitaxel or cisplatin and gemcitabine did not confer a survival advantage over the same chemotherapy given alone in two recently reported randomized phase III trials. 108,109 There are several other biologic agents in various stages of development with promising data already reported on erlotinib110 (EGFR inhibitor) and Avastin (vascular endothelial growth factor inhibitor). In a phase III trial of erlotinib versus placebo in a second- or third-line setting in patients with advanced NSCLC, Shepherd et al reported a prolongation of overall survival from 4.7 month with placebo to 6.7 months with erlotinib (P=0.001). There were statistically significant improvements in progression-free survival as well symptoms of cough, dyspnea, and pain.110

Patients with solitary brain metastases may achieve a 10% to 20% 5-year survival after resection of metastasis followed by whole brain radiotherapy. 111,112 Less convincing data also exist for resection of solitary adrenal metastases.113,114 In both instances control of the primary tumor by surgery must be assumed.

Small Cell Lung Cancer

Small cell lung cancer (SCLC) is biologically distinct from NSCLC in its propensity to an early spread and have a rapid tumor doubling time. The staging schema is simpler, dividing the disease into limited and extensive stages. Limited stage is defined as disease confined to one hemithorax including ipsilateral mediastinal and/or supraclavicular disease, excluding malignant pleural effusion (operationally disease that can be confined within one tolerable radiation port). All other tumors are characterized as extensive. The staging work-up should include history and physical examination, laboratory evaluation, CT scans of chest and upper abdomen, bone and brain scans.115 After a complete staging work-up only one quarter to a third of the patients will be identified as having limited disease.

Limited Stage Disease

Surgery is not considered a part of standard management of SCLC due to its propensity for early spread. On occasion, patients may undergo excision of a solitary pulmonary nodule that is subsequently identified as SCLC on pathologic examination. These patients should undergo mediastinal node dissection followed by combination chemotherapy.16 Post-operative radiotherapy should be added if mediastinal nodes are involved. All other patients should undergo combined modality therapy with chemotherapy and radiation therapy without surgery.

Randomized trials have established that combination chemotherapy is clearly superior to single-agent chemotherapy in small cell lung cancer.116,117 Cisplatin and etoposide is the most commonly used chemotherapy regimen, although randomized trials in extensive stage disease have not demonstrated a survival benefit over the regimen of cyclophosphamide, vincrisitine and adriamycin.118,119 Perry et al first reported a survival advantage with addition of thoracic radiotherapy to chemotherapy in limited stage disease (Table 4).120 Meta-analysis have later confirmed a 5% reduction in the risk of death with the addition of thoracic radiotherapy. 121,122

Table 4.

Selected studies in management of small cell lung cancer.

Study Treatment Remarks
Limited Stage disease
Perry120 Cyclophosphamide, etoposide/adriamycin, vincristine alone vs. chemotherapy and early vs. delayed radiotherapy Failure free survival overall survival improved with addition of radiotherapy
Takada123 Cisplatin and etoposide with concurrent vs. sequential radiotherapy Improved 2- and 5-year survival with concurrent approach. More myelosuppression
Turrisi124 Chemotherapy with once daily vs. twice daily radiotherapy Improved 5-year survival with twice daily radiotherapy (26% vs. 16%)
Extensive Stage Disease
Fukuoka118 Cisplatin, etoposide vs. cyclophosphamide adriamycin, vincristine vs. both regimens alternating with each other Response rate superior in etoposide containing regimens. Complete responses similar
Roth119 Cisplatin, etoposide vs. cyclophosphamide adriamycin, vincristine vs. both regimens alternating with each other No difference among treatment groups
Noda129 Cisplatin and etoposide vs. cisplatin and Irinotecan Median survival (12.8 vs. 9.4 months) and 2-year survival (19.5% vs. 5.2%) higher with irinotecan arm with increased incidence of diarrhea

The timing of radiotherapy, early or late, is another area of intensive investigation and there is no consensus. It appears that concurrent chemotherapy and radiation offers an improved 5-year survival compared to a sequential approach.123 Turrisi et al reported improved 5-year survival with hyperfractionated radiotherapy (twice daily fractionation) delivered concurrently with chemotherapy.124 There are obvious problems of increased toxicity and the logistics of twice daily delivery of radiation. This approach is, therefore, not widely practiced as a standard of care.

Patients with limited SCLC who achieve complete remission may develop brain metastases as the initial site of relapse. Studies have reported up to a 50% incidence of brain metastases in this patient population. 125,126 Several studies failed to show a survival benefit with prophylactic cranial radiation (PCI) despite decreasing the risk of brain metastases. Even though there have been concerns about long-term neurologic sequalae, studies suggest that PCI does not result in clinically significant neuropsychologic sequalae,127 especially if the radiation dose is limited to less than 3600 cGy in those without any neurologic deficit.125 A meta-analysis has shown a clinically significant survival benefit (5.4% at 3 years) with the addition of PCI in those who achieve complete remission after chemotherapy and thoracic radiation.128

Extensive Stage Disease

Extensive stage disease is managed with combination chemotherapy. The combination of cisplatin and etoposide, or cyclophosphamide, adriamycin and vincristine appear equivalent in survival benefit, although a cisplatin-based regimen may offer higher response rates and improved toxicity profile (Table 4).118,119 A recently reported randomized trial from Japan has reported improved response rates, median, and two year survivals with cisplatin and irinotecan compared to cisplatin and etoposide. The median and two year survivals were 12.8 months vs. 9.4 months and 19.5% vs. 5.2%, respectively.129 There was less severe hematologic toxicity but more severe diarrhea with the irinotecan combination. Even though confirmatory trials of cisplatin and irinotecan are ongoing, at present it is appropriate to offer etoposide or irinotecan in combination with cisplatin as first-line therapy for extensive stage SCLC. Other agents with activity in SCLC include paclitaxel and topotecan. The addition of paclitaxel to cisplatin and etoposide has been studied and showed increased toxicity without survival benefit.130 Topotecan has shown useful activity in the second-line setting and is currently being studied for use in first-line therapy.131

Acknowledgement

The authors acknowledge the help given by National Cancer Registry of Saudi Arabia particularly Mrs. Soad Arteh in providing cancer incidence in the Kingdom.

References

  • 1.GLOBOCAN 2000. Cancer Incidence, Mortality and Prevalence Worldwide. Version 1.0. Lyon: [IARCPress]. 2001. [Google Scholar]
  • 2.Jemal A, Tiwari R, Murray T, et al. Cancer statistics, 2004. CA Cancer J Clin. 2004;54:8–29. doi: 10.3322/canjclin.54.1.8. [DOI] [PubMed] [Google Scholar]
  • 3.American Cancer Society. Cancer Facts & Figures 2004. Atlanta, Georgia: American Cancer Society, Inc; 2004. [Google Scholar]
  • 4.Cancer Incidence Report. Kingdom of Saudi Arabia: National Cancer Registry; 2004. [Google Scholar]
  • 5.Mitchell G, Mitchell C. Lung cancer. Aust Fam Physician. 2004;33:321–325. [PubMed] [Google Scholar]
  • 6.Proctor R. The global smoking epidemic: a history and status report. Clin Lung Cancer. 2004;5:371–376. doi: 10.3816/CLC.2004.n.016. [DOI] [PubMed] [Google Scholar]
  • 7.Cardenas V, Thun M, Austin H, et al. Environmental tobacco smoke and lung cancer mortality in the American Cancer Society’s Cancer Prevention Study. II. Cancer Causes Control. 1997;8:57–64. doi: 10.1023/a:1018483121625. [DOI] [PubMed] [Google Scholar]
  • 8.Spitzer W, Lawrence V, Dales R, et al. Links between passive smoking and disease: a best-evidence synthesis. A report of the Working Group on Passive Smoking. Clin Invest Med. 1990;13:17–42. discussion 43–46. [PubMed] [Google Scholar]
  • 9.Wald N, Nanchahal K, Thompson S, et al. Does breathing other people’s tobacco smoke cause lung cancer? Br Med J (Clin Res Ed) 1986;293:1217–1222. doi: 10.1136/bmj.293.6556.1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Sethi T. Lung cancer. Introduction. Thorax. 2002;57:992–993. doi: 10.1136/thorax.57.11.992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Neuberger J, Field R. Occupation and lung cancer in nonsmokers. Rev Environ Health. 2003;18:251–267. doi: 10.1515/reveh.2003.18.4.251. [DOI] [PubMed] [Google Scholar]
  • 12.Attfield M, Costello J. Quantitative exposure-response for silica dust and lung cancer in Vermont granite workers. Am J Ind Med. 2004;45:129–138. doi: 10.1002/ajim.10348. [DOI] [PubMed] [Google Scholar]
  • 13.Ward E, Okun A, Ruder A, et al. A mortality study of workers at seven beryllium processing plants. Am J Ind Med. 1992;22:885–904. doi: 10.1002/ajim.4700220610. [DOI] [PubMed] [Google Scholar]
  • 14.Grimsrud T, Berge S, Martinsen J, et al. Lung cancer incidence among Norwegian nickel-refinery workers 1953–2000. J Environ Monit. 2003;5:190–197. doi: 10.1039/b211722n. [DOI] [PubMed] [Google Scholar]
  • 15.Haus B, Razavi H, Kuschner W. Occupational and environmental causes of bronchogenic carcinoma. Curr Opin Pulm Med. 2001;7:220–225. doi: 10.1097/00063198-200107000-00010. [DOI] [PubMed] [Google Scholar]
  • 16.Spira A, Ettinger D. Multidisciplinary management of lung cancer. N Engl J Med. 2004;350:379–392. doi: 10.1056/NEJMra035536. [DOI] [PubMed] [Google Scholar]
  • 17.Spitz M, Bondy M. Genetic susceptibility to cancer. Cancer. 1993;72:991–995. doi: 10.1002/1097-0142(19930801)72:3+<991::aid-cncr2820721307>3.0.co;2-5. [DOI] [PubMed] [Google Scholar]
  • 18.Perera F. Molecular epidemiology: insights into cancer susceptibility, risk assessment, and prevention. J Natl Cancer Inst. 1996;88:496–509. doi: 10.1093/jnci/88.8.496. [DOI] [PubMed] [Google Scholar]
  • 19.Li D, Firozi P, Wang L, et al. Sensitivity to DNA damage induced by benzo(a)pyrene diol epoxide and risk of lung cancer: a case-control analysis. Cancer Res. 2001;61:1445–1450. [PubMed] [Google Scholar]
  • 20.Wu X, Shi H, Jiang H, et al. Associations between cytochrome P4502E1 genotype, mutagen sensitivity, cigarette smoking and susceptibility to lung cancer. Carcinogenesis. 1997;18:967–973. doi: 10.1093/carcin/18.5.967. [DOI] [PubMed] [Google Scholar]
  • 21.Kato S, Shields P, Caporaso N, et al. Analysis of cytochrome P450 2E1 genetic polymorphisms in relation to human lung cancer. Cancer Epidemiol Biomarkers Prev. 1994;3:515–518. [PubMed] [Google Scholar]
  • 22.Nakachi K, Imai K, Hayashi S, et al. Polymorphisms of the CYP1A1 and glutathione S-transferase genes associated with susceptibility to lung cancer in relation to cigarette dose in a Japanese population. Cancer Res. 1993;53:2994–2999. [PubMed] [Google Scholar]
  • 23.Nazar-Stewart V, Motulsky A, Eaton D, et al. The glutathione S-transferase mu polymorphism as a marker for susceptibility to lung carcinoma. Cancer Res. 1993;53:2313–2318. [PubMed] [Google Scholar]
  • 24.Heckbert S, Weiss N, Hornung S, et al. Glutathione S-transferase and epoxide hydrolase activity in human leukocytes in relation to risk of lung cancer and other smoking-related cancers. J Natl Cancer Inst. 1992;84:414–422. doi: 10.1093/jnci/84.6.414. [DOI] [PubMed] [Google Scholar]
  • 25.Hou S, Felt S, Yang K, et al. Differential interactions between GSTM1 and NAT2 genotypes on aromatic DNA adduct level and HPRT mutant frequency in lung cancer patients and population controls. Cancer Epidemiol Biomarkers Prev. 2001;10:133–140. [PubMed] [Google Scholar]
  • 26.Zhou W, Liu G, Thurston S, et al. Genetic polymorphisms in N-acetyltransferase-2 and microsomal epoxide hydrolase, cumulative cigarette smoking, and lung cancer. Cancer Epidemiol Biomarkers Prev. 2002;11:15–21. [PubMed] [Google Scholar]
  • 27.Frazier M, O’Donnell F, Kong S, et al. Age-associated risk of cancer among individuals with N-acetyltransferase 2 (NAT2) mutations and mutations in DNA mismatch repair genes. Cancer Res. 2002;61:1269–1271. [PubMed] [Google Scholar]
  • 28.Zhou W, Thurston S, Liu G, et al. The interaction between microsomal epoxide hydrolase polymorphisms and cumulative cigarette smoking in different histological subtypes of lung cancer. Cancer Epidemiol Biomarkers Prev. 2001;10:461–466. [PubMed] [Google Scholar]
  • 29.Nomura A, Stemmermann G, Heilbrun L, et al. Serum vitamin levels and the risk of cancer of specific sites in men of Japanese ancestry in Hawaii. Cancer Res. 1985;45:2369–2372. [PubMed] [Google Scholar]
  • 30.Willett W, Polk B, Underwood B, et al. Relation of serum vitamins A and E and carotenoids to the risk of cancer. N Engl J Med. 1984;310:430–434. doi: 10.1056/NEJM198402163100705. [DOI] [PubMed] [Google Scholar]
  • 31.Ziegler R, Colavito E, Hartge P, et al. Importance of alpha-carotene, beta-carotene, and other phytochemicals in the etiology of lung cancer. J Natl Cancer Inst. 1996;88:612–615. doi: 10.1093/jnci/88.9.612. [DOI] [PubMed] [Google Scholar]
  • 32.Colditz G, Stampfer M, Willett W. Diet and lung cancer. A review of the epidemiologic evidence in humans. Arch Intern Med. 1987;147:157–160. doi: 10.1001/archinte.147.1.157. [DOI] [PubMed] [Google Scholar]
  • 33.Greenwald P. NCI cancer prevention and control research. Prev Med. 1993;22:642–660. doi: 10.1006/pmed.1993.1058. [DOI] [PubMed] [Google Scholar]
  • 34.Knekt P, Jorvinen R, Sepponen R, et al. Dietary flavonoids and the risk of lung cancer and other malignant neoplasms. Am J Epidemiol. 1997;146:223–230. doi: 10.1093/oxfordjournals.aje.a009257. [DOI] [PubMed] [Google Scholar]
  • 35.Hecht S. Chemoprevention of lung cancer by isothiocyanates. Adv Exp Med Biol. 1996;401:1–11. doi: 10.1007/978-1-4613-0399-2_1. [DOI] [PubMed] [Google Scholar]
  • 36.Stoner G, Morse M. Isothiocyanates and plant polyphenols as inhibitors of lung and esophageal cancer. Cancer Lett. 1997;114:113–119. doi: 10.1016/s0304-3835(97)04639-9. [DOI] [PubMed] [Google Scholar]
  • 37.Kelloff G, Hawk E, Crowell J, et al. Strategies for identification and clinical evaluation of promising chemopreventive agents. Oncology (Huntingt) 1996;10:1471–1484. discussion 1484–1488. [PubMed] [Google Scholar]
  • 38.Hong W, Lippman S, Itri L, et al. Prevention of second primary tumors with isotretinoin in squamous-ell carcinoma of the head and neck. N Engl J Med. 1990;323:795–801. doi: 10.1056/NEJM199009203231205. [DOI] [PubMed] [Google Scholar]
  • 39.Lippman S, Lee J, Karp D, et al. Randomized phase III intergroup trial of isotretinoin to prevent second primary tumors in stage I non-small-cell lung cancer. J Natl Cancer Inst. 2001;93:605–618. doi: 10.1093/jnci/93.8.605. [DOI] [PubMed] [Google Scholar]
  • 40.Lee J, Lippman S, Benner S, et al. Randomized placebo-controlled trial of isotretinoin in chemoprevention of bronchial squamous metaplasia. J Clin Oncol. 1994;12:937–945. doi: 10.1200/JCO.1994.12.5.937. [DOI] [PubMed] [Google Scholar]
  • 41.The Alpha-Tocopherol, Beta Carotene Cancer Prevention Study Group. The effect of vitamin E and beta carotene on the incidence of lung cancer and other cancers in male smokers. N Engl J Med. 1994;330:1029–1035. doi: 10.1056/NEJM199404143301501. [DOI] [PubMed] [Google Scholar]
  • 42.Omenn G, Goodman G, Thornquist M, et al. The beta-carotene and retinol efficacy trial (CARET) for chemoprevention of lung cancer in high risk populations: smokers and asbestos-exposed workers. Cancer Res. 1994;54:2038s–2043s. [PubMed] [Google Scholar]
  • 43.Rivera M, Detterbeck F, Mehta A, et al. Diagnosis of lung cancer: the guidelines. Chest. 2003;123:129S–136S. doi: 10.1378/chest.123.1_suppl.129s. [DOI] [PubMed] [Google Scholar]
  • 44.Michel F, Soler M, Imhof E, et al. Initial staging of non-small cell lung cancer: value of routine radioisotope bone scanning. Thorax. 1991;46:469–473. doi: 10.1136/thx.46.7.469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Mountain C. Revisions in the International System for Staging Lung Cancer. Chest. 1997;111:1710–1717. doi: 10.1378/chest.111.6.1710. [DOI] [PubMed] [Google Scholar]
  • 46.Pieterman RM, van Putten JW, Meuzelaar JJ, et al. Preoperative staging of non-small-cell lung cancer with positron-emission tomography. N Engl J Med. 2000;343:254–261. doi: 10.1056/NEJM200007273430404. [DOI] [PubMed] [Google Scholar]
  • 47.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. doi: 10.1097/00000421-200002000-00014. [DOI] [PubMed] [Google Scholar]
  • 48.Effects of postoperative mediastinal radiation on completely resected stage II and stage III epidermoid cancer of the lung. The Lung Cancer Study Group. N Engl J Med. 1986;315:1377–1381. doi: 10.1056/NEJM198611273152202. [DOI] [PubMed] [Google Scholar]
  • 49.Postoperative radiotherapy in non-small-cell lung cancer: systematic review and meta-analysis of individual patient data from nine randomised controlled trials. PORT Meta-analysis Trialists Group. Lancet. 1998;352:257–263. [PubMed] [Google Scholar]
  • 50.Arriagada R, Bergman B, Dunant A, et al. Cisplatin-based adjuvant chemotherapy in patients with completely resected non-small-cell lung cancer. N Engl J Med. 2004;350:351–360. doi: 10.1056/NEJMoa031644. [DOI] [PubMed] [Google Scholar]
  • 51.Hamada C, Ohta M, Wada H, et al. Survival benefit of oral UFT for adjuvant chemotherapy after completely resected non-small-cell lung cancer. Proc Am Soc Clin Oncol. 2004;22:617S. Abstract 7002. [Google Scholar]
  • 52.Strauss G, Herndon J, Maddaus M, et al. Randomized clinical trial of adjuvant chemotherapy with paclitaxel and carboplatin following resection in stage IB Non-Small Cell Lung Cancer (NSCLC): Report of Cancer and Leukemia Group B (CALGB) Protocol 9633. Proc Am Soc Clin Oncol. 2004;22:621S. Abstract 7019. [Google Scholar]
  • 53.Winton T, Livingston R, Johnson D, et al. A prospective randomized trial of adjuvant vinorelbine (VIN) and cisplatin (CIS) in completely resected stage IB and II non small cell lung cancer (NSCLC). Intergroup JBR10. Proc Am Soc Clin Oncol. 2004;22:621S. Abstract 7018. [Google Scholar]
  • 54.Noordijk EM, vd Poest Clement E, Hermans J, et al. Radiotherapy as an alternative to surgery in elderly patients with resectable lung cancer. Radiother Oncol. 1988;13:83–89. doi: 10.1016/0167-8140(88)90029-1. [DOI] [PubMed] [Google Scholar]
  • 55.Dosoretz DE, Katin MJ, Blitzer PH, et al. Radiation therapy in the management of medically inoperable carcinoma of the lung: results and implications for future treatment strategies. Int J Radiat Oncol Biol Phys. 1992;24:3–9. doi: 10.1016/0360-3016(92)91013-d. [DOI] [PubMed] [Google Scholar]
  • 56.Gauden S, Ramsay J, Tripcony L. The curative treatment by radiotherapy alone of stage I nonsmall cell carcinoma of the lung. Chest. 1995;108:1278–1282. doi: 10.1378/chest.108.5.1278. [DOI] [PubMed] [Google Scholar]
  • 57.Dillman R, Seagren S, Propert K, 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. doi: 10.1056/NEJM199010043231403. [DOI] [PubMed] [Google Scholar]
  • 58.Sause W, Scott C, Taylor S, et al. Radiation Therapy Oncology Group (RTOG) 88–08 and Eastern Cooperative Oncology Group (ECOG) 4588: preliminary results of a phase III trial in regionally advanced, unresectable non-small-cell lung cancer. J Natl Cancer Inst. 1995;87:198–205. doi: 10.1093/jnci/87.3.198. [DOI] [PubMed] [Google Scholar]
  • 59.Sause W, Kolesar P, Taylor SI, et al. Final results of phase III trial in regionally advanced unresectable non-small cell lung cancer: Radiation Therapy Oncology Group, Eastern Cooperative Oncology Group, and Southwest Oncology Group. Chest. 2000;117:358–364. doi: 10.1378/chest.117.2.358. [DOI] [PubMed] [Google Scholar]
  • 60.Dillman R, Herndon J, Seagren S, et al. 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. doi: 10.1093/jnci/88.17.1210. [DOI] [PubMed] [Google Scholar]
  • 61.Curran WJ., Jr Evolving chemoradiation treatment strategies for locally advanced non-smallcell lung cancer. Oncology (Huntingt) 2003;17:7–14. [PubMed] [Google Scholar]
  • 62.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. doi: 10.1200/JCO.1999.17.9.2692. [DOI] [PubMed] [Google Scholar]
  • 63.Pass H, Pogrebniak H, Steinberg S, et al. Randomized trial of neoadjuvant therapy for lung cancer: interim analysis. Ann Thorac Surg. 1992;53:992–998. doi: 10.1016/0003-4975(92)90373-c. [DOI] [PubMed] [Google Scholar]
  • 64.Elias A, Herndon J, Kumar P, et al. A phase III comparison of “best local regional therapy” with or without chemotherapy (CT) for stage IIIA T1-3N2 non-small cell lung cancer: Preliminary results. Proc Am Soc Clin Oncol. 1997;16:1611A. [Google Scholar]
  • 65.Rosell R, Gomez-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. doi: 10.1056/NEJM199401203300301. [DOI] [PubMed] [Google Scholar]
  • 66.Roth J, 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. doi: 10.1093/jnci/86.9.673. [DOI] [PubMed] [Google Scholar]
  • 67.Rosell R, Gomez-Codina J, Camps C, et al. Preresectional chemotherapy in stage IIIA nonsmall-cell lung cancer: a 7-year assessment of a randomized controlled trial. Lung Cancer. 1999;26:7–14. doi: 10.1016/s0169-5002(99)00045-8. [DOI] [PubMed] [Google Scholar]
  • 68.Roth J, Atkinson E, 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. doi: 10.1016/s0169-5002(98)00046-4. [DOI] [PubMed] [Google Scholar]
  • 69.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. doi: 10.1200/JCO.2002.20.1.247. [DOI] [PubMed] [Google Scholar]
  • 70.Strauss G, Herndon J, Maddaus M, et al. Randomized clinical trial of adjuvatn chemotherapy with paclitaxel and carboplatin following resection in stage IB Non-Small Cell Lung Cancer (NSCLC): Report of Cancer and Leukemia Group B (CALGB) Protocol 9633. Proc Am Soc Clin Oncol. 2004;22:621S. Abstract 7019. [Google Scholar]
  • 71.Chemotherapy in non-small cell lung cancer: a meta-analysis using updated data on individual patients from 52 randomised clinical trials. Nonsmall Cell Lung Cancer Collaborative Group. BMJ. 1995;311:899–909. [PMC free article] [PubMed] [Google Scholar]
  • 72.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. doi: 10.1002/1097-0142(19950815)76:4<593::aid-cncr2820760409>3.0.co;2-n. [DOI] [PubMed] [Google Scholar]
  • 73.Grilli R, Oxman A, Julian J. Chemotherapy for advanced non-small-cell lung cancer:how much benefit is enough? J Clin Oncol. 1993;11:1866–1872. doi: 10.1200/JCO.1993.11.10.1866. [DOI] [PubMed] [Google Scholar]
  • 74.Helsing M, Bergman B, Thaning L, et al. 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. Joint Lung Cancer Study Group. Eur J Cancer. 1998;34:1036–1044. doi: 10.1016/s0959-8049(97)10122-8. [DOI] [PubMed] [Google Scholar]
  • 75.Cullen M, Billingham L, Woodroffe C, 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. doi: 10.1200/JCO.1999.17.10.3188. [DOI] [PubMed] [Google Scholar]
  • 76.Ellis P, Smith I, Hardy J, 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. doi: 10.1038/bjc.1995.74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.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 Oct 1;18:3390–9. doi: 10.1200/JCO.2000.18.19.3390. [DOI] [PubMed] [Google Scholar]
  • 78.Sandler A, 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. doi: 10.1200/JCO.2000.18.1.122. [DOI] [PubMed] [Google Scholar]
  • 79.Wozniak A, Crowley J, Balcerzak S, 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. doi: 10.1200/JCO.1998.16.7.2459. [DOI] [PubMed] [Google Scholar]
  • 80.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. doi: 10.1200/JCO.2000.18.3.623. [DOI] [PubMed] [Google Scholar]
  • 81.Schiller J, Harrington D, Belani C, et al. Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. N Engl J Med. 2002;346:92–98. doi: 10.1056/NEJMoa011954. [DOI] [PubMed] [Google Scholar]
  • 82.Le Chevalier T, Brown A, Natale R, et al. Gemcitabine in the treatment of Non-Small Cell Lung Cancer (NSCLC): A meta-analysis of survival and progression free survival data. Lung Cancer. 2003;41:S70. [Google Scholar]
  • 83.Ettinger D. Is there a preferred combination chemotherapy regimen for metastastic non-small cell lung cancer? Oncologist. 2002;7:226–233. doi: 10.1634/theoncologist.7-3-226. [DOI] [PubMed] [Google Scholar]
  • 84.Kelly K, Mikhaeel-Kamel N, Pan Z, et al. 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. [PubMed] [Google Scholar]
  • 85.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. Southern Italy Cooperative Oncology Group. J Clin Oncol. 1999;17:2316–2325. doi: 10.1200/JCO.1999.17.8.2316. [DOI] [PubMed] [Google Scholar]
  • 86.Smith I, O’Brien M, Talbot D, 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. doi: 10.1200/JCO.2001.19.5.1336. [DOI] [PubMed] [Google Scholar]
  • 87.Socinski M, Schell M, 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. doi: 10.1200/JCO.2002.20.5.1335. [DOI] [PubMed] [Google Scholar]
  • 88.Andresen O, Sorensen S, Bergman B, et al. Duration of chemotherapy and survival in advanced non-small cell lung cancer (NSCLC). A multicenter, prospective randomised study. Lung Cancer. 2003;41:O28. [Google Scholar]
  • 89.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. doi: 10.1093/jnci/95.5.362. [DOI] [PubMed] [Google Scholar]
  • 90.Langer C, Vangel M, Schiller J, et al. Age-specific subanalysis of ECOG1594: Fit elderly patients (70–80 yrs) with NSCLC do as well as younger patients (70) Lung Cancer. 2003;41:S17. [Google Scholar]
  • 91.Belani C, Fossella F. Phase III study (Tax 326) of Docetaxel-Cisplatin (DC) and Docetaxel-Carboplatin (DCB) versus Vinorelbine-Cisplatin (VC) for the first line treatment of advanced/metastatic non-small cell lung cancer (NSCLC): analysis in elderly patients. Lung Cancer. 2003;41:S18. doi: 10.1002/cncr.21495. [DOI] [PubMed] [Google Scholar]
  • 92.Shepherd F, 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. doi: 10.1200/JCO.2000.18.10.2095. [DOI] [PubMed] [Google Scholar]
  • 93.Fossella F, DeVore R, Kerr R, et al. Randomized phase III trial of docetaxel versus vinorelbine or ifosfamide in patients with advanced non-smallcell lung cancer previously treated with platinum-containing chemotherapy regimens. The TAX 320 Non-Small Cell Lung Cancer Study Group. J Clin Oncol. 2000;18:2354–2362. doi: 10.1200/JCO.2000.18.12.2354. [DOI] [PubMed] [Google Scholar]
  • 94.De Marinis F, Pereira J, Park K, et al. Does second line therapy for non-small cell lung cancer (NSCLC) result in symptom palliation? Analysis of 484 patients from a randomized trial of pemetrexed vs. docetaxel. Proc Am Soc Clin Oncol. 2004;22:625S. Abstract 7035. [Google Scholar]
  • 95.Gillenwater HH, Tynan M, Natoli S, et al. Second-line gemcitabine in refractory stage IV non small-cell lung cancer: a phase II trial. Clin Lung Cancer. 2000;2:133–138. doi: 10.3816/clc.2000.n.026. [DOI] [PubMed] [Google Scholar]
  • 96.Manegold C. Gemcitabine (Gemzar) in nonsmall cell lung cancer. Expert Rev Anticancer Ther. 2004;4:345–360. doi: 10.1586/14737140.4.3.345. [DOI] [PubMed] [Google Scholar]
  • 97.Salomon DS, Brandt R, Ciardiello F, et al. Epidermal growth factor-related peptides and their receptors in human malignancies. Crit Rev Oncol Hematol. 1995;19:183–232. doi: 10.1016/1040-8428(94)00144-i. [DOI] [PubMed] [Google Scholar]
  • 98.Rusch V, Baselga J, Cordon-Cardo C, et al. Differential expression of the epidermal growth factor receptor and its ligands in primary nonsmall cell lung cancers and adjacent benign lung. Cancer Res. 1993;53:2379–2385. [PubMed] [Google Scholar]
  • 99.Volm M, Rittgen W, Drings P. Prognostic value of ERBB-1, VEGF, cyclin A, FOS, JUN and MYC in patients with squamous cell lung carcinomas. Br J Cancer. 1998;77:663–669. doi: 10.1038/bjc.1998.106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Pavelic K, Banjac Z, Pavelic J, et al. Evidence for a role of EGF receptor in the progression of human lung carcinoma. Anticancer Res. 1993;13:1133–1137. [PubMed] [Google Scholar]
  • 101.Fujino S, Enokibori T, Tezuka N, et al. A comparison of epidermal growth factor receptor levels and other prognostic parameters in non-small cell lung cancer. Eur J Cancer. 1996;32A:2070–2074. doi: 10.1016/s0959-8049(96)00243-2. [DOI] [PubMed] [Google Scholar]
  • 102.Ranson M, Hammond LA, Ferry D, et al. ZD1839, a selective oral epidermal growth factor receptor-tyrosine kinase inhibitor, is well tolerated and active in patients with solid, malignant tumors: results of a phase I trial. J Clin Oncol. 2002;20:2240–2250. doi: 10.1200/JCO.2002.10.112. [DOI] [PubMed] [Google Scholar]
  • 103.Fukuoka M, Yano S, Giaccone G, et al. Multi-institutional randomized phase II trial of gefitinib for previously treated patients with advanced non-small-cell lung cancer. J Clin Oncol. 2003;21:2237–2246. doi: 10.1200/JCO.2003.10.038. [DOI] [PubMed] [Google Scholar]
  • 104.Fukuoka M, Yano S, Giaccone G, et al. Final result from a phase II trial of ZD1839 (‘Iressa’) for patients with advanced non-small-cell lung cancer (IDEAL I) Proc Am Soc Clin Oncol. 2002;21:298a. [Google Scholar]
  • 105.Bailey R, Kris M, Wolf M, et al. Gefitinib (‘Iressa’, ZD1839) monotherapy for pretreated advanced non-small-cell lung cancer in IDEAL 1 and 2: tumor response is not clinically relevantly predicted from tumor EGFR membrane staining alone. Lung Cancer. 2003;41:S71. [Google Scholar]
  • 106.Giaccone G, Herbst R, Manegold C, et al. Gefitinib in combination with gemcitabine and cisplatin in advanced non-small-cell lung cancer: a phase III trial--INTACT 1. J Clin Oncol. 2004;22:777–784. doi: 10.1200/JCO.2004.08.001. [DOI] [PubMed] [Google Scholar]
  • 107.Herbst R, Giaccone G, Schiller J, et al. Gefitinib in combination with paclitaxel and carboplatin in advanced non-small-cell lung cancer: a phase III trial—INTACT 2. J Clin Oncol. 2004;22:785–794. doi: 10.1200/JCO.2004.07.215. [DOI] [PubMed] [Google Scholar]
  • 108.Herbst R, Prager D, Hermann R, et al. TRIBUTE-A phase III trial of erlotinib HCL (OSI-774) combined with carboplatin and paclitaxel (CP) chemotherapy in advanced non-small cell lung cancer. Proc Am Soc Clin Oncol. 2004;22 doi: 10.1200/JCO.2005.02.840. Abstract 7011. [DOI] [PubMed] [Google Scholar]
  • 109.Gatzemeier U, Pluzanska A, Szczesna A, et al. Results of a phase III trial of erlotinib (OSI-774) combined with cisplatin and gemcitabine (GC) chemotherapy in advanced non-small cell lung cancer (NSCLC) Proc Am Soc Clin Oncol. 2004;22:619S. Abstract 7010. [Google Scholar]
  • 110.Shepherd F, Pereira J, Ciuleanu T, et al. A randomized placebo-controlled trial of erlotinib in patients with advanced non-small cell lung cancer (NSCLC) following failure of 1st line or 2nd line chemotherapy. A National Cancer Institute of Canada Clinial Trials Group (NCIC CTG) Trial. Proc Am Soc Clin Oncol. 2004;22:7022. [Google Scholar]
  • 111.Patchell R, Tibbs P, Walsh J, et al. A randomized trial of surgery in the treatment of single metastases to the brain. N Engl J Med. 1990;322:494–500. doi: 10.1056/NEJM199002223220802. [DOI] [PubMed] [Google Scholar]
  • 112.Magilligan D, Duvernoy C, Malik G, et al. Surgical approach to lung cancer with solitary cerebral metastasis: twenty-five years’ experience. Ann Thorac Surg. 1986;42:360–364. doi: 10.1016/s0003-4975(10)60536-x. [DOI] [PubMed] [Google Scholar]
  • 113.Raviv G, Klein E, Yellin A, et al. Surgical treatment of solitary adrenal metastases from lung carcinoma. J Surg Oncol. 1990;43:123–124. doi: 10.1002/jso.2930430212. [DOI] [PubMed] [Google Scholar]
  • 114.Reyes L, Parvez Z, Nemoto T, et al. Adrenalectomy for adrenal metastasis from lung carcinoma. J Surg Oncol. 1990;44:32–34. doi: 10.1002/jso.2930440108. [DOI] [PubMed] [Google Scholar]
  • 115.Argiris A, Murren J. Staging and clinical prognostic factors for small-cell lung cancer. Cancer J. 2001;7:437–447. [PubMed] [Google Scholar]
  • 116.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. [PubMed] [Google Scholar]
  • 117.Seifter E, Ihde D. Therapy of small cell lung cancer: a perspective on two decades of clinical research. Semin Oncol. 1988;15:278–299. [PubMed] [Google Scholar]
  • 118.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. doi: 10.1093/jnci/83.12.855. [DOI] [PubMed] [Google Scholar]
  • 119.Roth B, Johnson D, Einhorn L, 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 SoutheasternCancer Study Group. J Clin Oncol. 1992;10:282–291. doi: 10.1200/JCO.1992.10.2.282. [DOI] [PubMed] [Google Scholar]
  • 120.Perry M, Eaton W, Propert K, et al. Chemotherapy with or without radiation therapy in limited small-cell carcinoma of the lung. N Engl J Med. 1987;316:912–918. doi: 10.1056/NEJM198704093161504. [DOI] [PubMed] [Google Scholar]
  • 121.Pignon J, Arriagada R, Ihde D, et al. A meta-analysis of thoracic radiotherapy for small-cell lung cancer. N Engl J Med. 1992;327:1618–1624. doi: 10.1056/NEJM199212033272302. [DOI] [PubMed] [Google Scholar]
  • 122.Arriagada R, Pignon J, Ihde D, 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. [PubMed] [Google Scholar]
  • 123.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;20:3054–360. doi: 10.1200/JCO.2002.12.071. [DOI] [PubMed] [Google Scholar]
  • 124.Turrisi A, 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. doi: 10.1056/NEJM199901283400403. [DOI] [PubMed] [Google Scholar]
  • 125.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. doi: 10.1093/jnci/87.3.183. [DOI] [PubMed] [Google Scholar]
  • 126.Komaki R, Cox J, 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. [PubMed] [Google Scholar]
  • 127.Le Pechoux C, Laplanche A, Borie F, et al. Long term results in terms of neurotoxicity among patients with limited small cell lung cancer included in a trial evaluating prophylactic cranial radiation. Lung Cancer. 2003;41:S21. [Google Scholar]
  • 128.Aupérin A, Arriagada R, Pignon J, et al. Prophylactic cranial irradiation for patients with small-cell lung cancer in complete remission. Prophylactic Cranial Irradiation Overview Collaborative Group. N Engl J Med. 1999;341:476–484. doi: 10.1056/NEJM199908123410703. [DOI] [PubMed] [Google Scholar]
  • 129.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. doi: 10.1056/NEJMoa003034. [DOI] [PubMed] [Google Scholar]
  • 130.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. doi: 10.1023/a:1011131303391. [DOI] [PubMed] [Google Scholar]
  • 131.Ardizzoni A, Hansen H, Dombernowsky P, et al. Topotecan, a new active drug in the secondline treatment of small-cell lung cancer: a phase II study in patients with refractory and sensitive disease. The European Organization for Research and Treatment of Cancer Early Clinical Studies Group and New Drug Development Office, and the Lung Cancer Cooperative Group. J Clin Oncol. 1997;15:2090–2096. doi: 10.1200/JCO.1997.15.5.2090. [DOI] [PubMed] [Google Scholar]
  • 132.Mountain C, Dresler C. Regional lymph node classification for lung cancer staging. Chest. 1997;111:1718–1723. doi: 10.1378/chest.111.6.1718. [DOI] [PubMed] [Google Scholar]
  • 133.Rosell R, Maestre J, Font A, et al. A randomized trial of mitomycin/ifosfamide/cisplatin preoperative chemotherapy plus surgery versus surgery alone in stage IIIA non-small cell lung cancer. Semin Oncol. 1994;21:28–33. [PubMed] [Google Scholar]
  • 134.Holmes E, Gail M. Surgical adjuvant therapy for stage II and stage III adenocarcinoma and large-cell undifferentiated carcinoma. J Clin Oncol. 1986;4:710–715. doi: 10.1200/JCO.1986.4.5.710. [DOI] [PubMed] [Google Scholar]
  • 135.The benefit of adjuvant treatment for resected locally advanced non-small-cell lung cancer. The Lung Cancer Study Group. J Clin Oncol. 1988;6:9–17. doi: 10.1200/JCO.1988.6.1.9. [DOI] [PubMed] [Google Scholar]
  • 136.Keller S, 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. Eastern Cooperative Oncology Group. N Engl J Med. 2000;343:1217–1222. doi: 10.1056/NEJM200010263431703. [DOI] [PubMed] [Google Scholar]
  • 137.Tonato M. Final report of the Adjuvant Lung Project Italy (ALPI): an Italian/EORTC-LCG randomised trial of adjuvant chemotherapy in completely resected non-small cell lung cancer (NSCLC) Proc Am Soc Clin Oncol. 2002;21 Abst 1157. [Google Scholar]
  • 138.Kris M, Natale R, Herbst R, et al. A phase II trial of ZD1839 (‘Iressa’) in advanced non-small cell lung cancer (NSCLC) patients who had failed platinum-and docetaxel-based regimens (IDEAL 2) Proc Am Soc Clin Oncol. 2002 Abstract 1166. [Google Scholar]

Articles from Annals of Saudi Medicine are provided here courtesy of King Faisal Specialist Hospital and Research Centre

RESOURCES