Sunday 26 June 2016

NEOPLASMS OF LUNG

The Magnitude of the Problem

In 2007, primary carcinoma of the lung affected 114,760 males and 98,620 females in the United States; 86% die within 5 years of diagnosis, making it the leading cause of cancer death in both men and women. The incidence of lung cancer peaks between ages 55 and 65 years. Lung cancer accounts for 29% of all cancer deaths (31% in men, 26% in women). Lung cancer is responsible for more deaths in the United States each year than breast cancer, colon cancer, and prostate cancer combined; more women die each year of lung cancer than of breast cancer. The age-adjusted lung cancer death rate in males is decreasing, but in females it is stable or still increasing. These death rates are related to smoking; smoking cessation efforts begun 40 years ago in men are largely responsible for the change in incidence and death rates. However, women started smoking in substantial numbers about 10–15 years later than men; smoking cessation efforts need to increase for women. The 5-year overall lung cancer survival rate (15%) has nearly doubled in the past 30 years. The improvement is due to advances in combined-modality treatment with surgery, radiotherapy, and chemotherapy. The International Agency for Research on Cancer estimates that there will be over 1.18 million deaths from lung cancer worldwide in 2007, which will rise to 10 million deaths per year by 2030. This represents one lung cancer case for every 3 million cigarettes smoked. Thus, primary carcinoma of the lung is a major health problem with a generally grim prognosis.

Pathology

The term lung cancer is used for tumors arising from the respiratory epithelium (bronchi, bronchioles, and alveoli). Mesotheliomas, lymphomas, and stromal tumors (sarcomas) are distinct from epithelial lung cancer. Four major cell types make up 88% of all primary lung neoplasms according to the World Health Organization classification (Table 85-1). These are squamous or epidermoid carcinoma, small cell (also called oat cell) carcinoma, adenocarcinoma (including bronchioloalveolar), and large cell carcinoma. The remainder include undifferentiated carcinomas, carcinoids, bronchial gland tumors (including adenoid cystic carcinomas and mucoepidermoid tumors), and rarer tumor types. The various cell types have different natural histories and responses to therapy, and thus a correct histologic diagnosis by an experienced pathologist is the first step to correct treatment. In the past 25 years, adenocarcinoma has replaced squamous cell carcinoma as the most frequent histologic subtype, and the incidence of small cell carcinoma is on the decline.

 Frequency, Age-Adjusted Incidence, and Survival Rates for Different Histologic Types of Lung Cancera
Histologic Type of Thoracic Malignancy Frequency, % Age-Adjusted Rate 5-Year Survival Rate (All Stages)
Adenocarcinoma (and all subtypes) 32 7 17
Bronchioloalveolar carcinoma 3 1.4 42
Squamous cell (epidermoid) carcinoma 29 15 15
Small cell carcinoma 18 9 5
Large cell carcinoma 9 5 11
Carcinoid 1.0 0.5 83
Mucoepidermoid carcinoma 0.1 <0.1 39
Adenoid cystic carcinoma <0.1 <0.1 48
Sarcoma and other soft tissue tumors 0.1 0.1 30
All others and unspecified carcinomas 11.0 6 NA
Total  100 52 14

aData on histology frequency and age-adjusted incidence rates per 100,000 U.S. population are from 60,514 cases of invasive lung cancer involving all races and both sexes obtained from the data for 1983–1987 of the Surveillance, Epidemiology, and End Results (SEER) Program of the National Cancer Institute; 5-year relative survival rates for all stages, all races, and both sexes are from the SEER data on 87,128 carcinomas, 1978–1986. NA, not available.

Source: Summarized from Travis et al: Cancer 75:191, 1995.

Major treatment decisions are made on the basis of whether a tumor is classified as a small cell lung carcinoma (SCLC) or as one of the non-small cell lung cancer (NSCLC) varieties (squamous, adenocarcinoma, large cell carcinoma, bronchioloalveolar carcinoma, and mixed versions of these). The histologic distinctions between SCLC and NSCLC include the following: SCLC has scant cytoplasm, small hyperchromatic nuclei with fine chromatin pattern and indistinct nucleoli with diffuse sheets of cells, while NSCLC has abundant cytoplasm, pleomorphic nuclei with coarse chromatin pattern, prominent nucleoli, and glandular or squamous architecture. Among the molecular distinctions, SCLC displays neuroendocrine properties absent in NSCLCs, production of specific peptide hormones [such as adrenocorticotropic hormone (ACTH), arginine vasopressin (AVP), atrial natriuretic factor (ANF), gastrin-releasing peptide (GRP)] and differences in oncogene and tumor-suppressor gene changes (SCLCs have RB mutations in 90% and p16 abnormalities in 10% but never have KRAS or EGFR mutations, while NSCLCs have RB mutations in only 20%, p16 changes in 50%, KRAS mutations in 30%, and EGFR mutations in ~10%). Both types have frequent p53 mutations (>70% in SCLC and >50% in NSCLC), 3p allele loss (>90% in both), telomerase expression (>90% in both), and tumor-acquired promoter methylation in multiple genes (>80% in both, often involving the same genes, including RASSF1A). SCLCs are initially very responsive to combination chemotherapy (>70% responses, with 30% complete responses) and to radiotherapy (>90% responses); however, most SCLCs ultimately relapse. By contrast, NSCLCs have objective tumor shrinkage following radiotherapy in 30–50% of cases and response to combination chemotherapy in 20–35% of cases. At presentation, SCLCs usually have already spread such that surgery is unlikely to be curative and, given their responsiveness to chemotherapy, are managed primarily by chemotherapy with or without radiotherapy. Chemotherapy clearly provides symptom relief and survival advantage. By contrast, NSCLCs that are clinically localized at the time of presentation may be cured with either surgery or radiotherapy. The beneficial role of chemotherapy in NSCLC is in palliation of symptoms and improving survival modestly.
Although it is important to differentiate whether a tumor is SCLC or NSCLC for both prognostic and therapeutic reasons, it is less important to identify the histologic subtypes of NSCLC. Stage for stage, the histology of NSCLC is not an important prognostic factor, and in the past the different subtypes of NSCLC were rarely treated differently. However, lung adenocarcinomas (often with bronchioloalveolar features) may be responsive to therapy aimed at the epidermal growth factor receptor (EGFR) (see below). In addition, patients with squamous cell carcinoma may not be appropriate candidates for antiangiogenic therapy due to an increased risk of bleeding (see below).
Eighty-five percent of patients with lung cancer of all histologic types are current or former cigarette smokers. Of the annual 213,380 new cases of lung cancer, ~50% develop in former smokers. With increased success in smoking cessation efforts, the number of former smokers will grow, and these individuals will be important candidates for early detection and chemoprevention efforts.
All histologic types of lung cancer are due to smoking. However, lung cancer can also occur in individuals who have never smoked. By far the most common form of lung cancer arising in lifetime nonsmokers, in women, and in young patients (<45 years) is adenocarcinoma. However, in nonsmokers with adenocarcinoma involving the lung, the possibility of other primary sites should be considered. Squamous and small cell cancers usually present as central masses with endobronchial growth, while adenocarcinomas and large cell cancers tend to present as peripheral nodules or masses, frequently with pleural involvement. Squamous and large cell cancers cavitate in 10–20% of cases. Bronchioloalveolar carcinoma (BAC) is a subtype of adenocarcinoma that grows along the alveoli without invasion and can present radiographically as a single mass; as a diffuse, multinodular lesion; as a fluffy infiltrate; and on screening CT scans as a "ground glass" opacity. The male to female ratio is 1:1, and while BAC can be associated with smoking, it is often found in nonsmokers. Histologically pure BAC is relatively rare. More common is adenocarcinoma with BAC features. BAC may present in a mucinous form, which tends to be multicentric, and a nonmucinous form, which tends to be solitary. Many of the EGFR mutations found in nonsmoking lung cancers occur in adenocarcinomas with BAC histologic features.
Etiology
Most lung cancers are caused by carcinogens and tumor promoters inhaled via cigarette smoking. The prevalence of smoking in the United States is 28% for males and 25% for females, age 18 years or older; 38% of high school seniors smoke. The relative risk of developing lung cancer is increased about thirteenfold by active smoking and about 1.5-fold by long-term passive exposure to cigarette smoke. Chronic obstructive pulmonary disease, which is also smoking-related, further increases the risk of developing lung cancer. The lung cancer death rate is related to the total amount (often expressed in "cigarette pack-years") of cigarettes smoked, such that the risk is increased 60- to 70-fold for a man smoking two packs a day for 20 years as compared with a nonsmoker. Conversely, the chance of developing lung cancer decreases with cessation of smoking but may never return to the nonsmoker level. The increase in lung cancer rate in women is also associated with a rise in cigarette smoking. Women have a higher relative risk per given exposure than men (~1.5-fold higher). This sex difference may be due to a greater susceptibility to tobacco carcinogens in women, although the data are controversial.
About 15% of lung cancers occur in individuals who have never smoked. The majority of these are found in women. The reason for this sex difference is not known but may be related to hormonal factors.
Efforts to get people to stop smoking are mandatory. However, smoking cessation is extremely difficult, because the smoking habit represents a powerful addiction to nicotine (Chap. 390). Smoking addiction is both biologic and psychosocial. Different methods are available to help motivated smokers give up the habit, including counseling, behavioral therapy, nicotine replacement (gum, patch, sublingual spray, inhaler), and antidepressants (such as bupropion). However, one year after starting such smoking cessation aids, the methods are successful in only 20–25% of individuals. Preventing people from starting to smoke is thus very important, and this primary prevention effort needs to be targeted to children since most cigarette smoking addiction occurs during the teenage years.
Radiation is another environmental cause of lung cancer. People exposed to high levels of radon or receiving thoracic radiation therapy have a higher than normal incidence of lung cancer, particularly if they smoke.
Biology and Molecular Pathogenesis
Molecular genetic studies have shown the acquisition by lung cancer cells of a number of genetic lesions, including activation of dominant oncogenes and inactivation of tumor-suppressor or recessive oncogenes (Chaps. 79 and 80). In fact, lung cancer cells may have to accumulate a large number (perhaps 20) of such lesions. A small subpopulation (perhaps <1%) of cells within a tumor are responsible for the full malignant behavior of the tumor;—these are referred to as cancer stem cells. As part of this concept, the large bulk of the cells in a cancer are "offspring" of these cancer stem cells and, while clonally related to the cancer stem cell subpopulation, by themselves cannot regenerate the full malignant phenotype such as metastatic disease and unlimited replicative potential. These cancer stem cells are very important to identify since successful treatment of the tumor will require eradication of this stem cell component. These cancer stem cells may be more resistant to chemotherapy than the bulk of the tumor. Features that distinguish cancer stem cells from the remaining tumor cells have not been defined and validated.
Activation of Dominant Oncogenes
Changes in dominant oncogenes include point mutations in the coding regions of the RAS family of oncogenes (particularly in the KRAS gene in adenocarcinoma of the lung); mutations in the tyrosine kinase domain of the EGFR found in adenocarcinomas from nonsmokers (~10% in the United States with rates >50% in nonsmoking East Asian patients); occasional mutations in BRAF and PIK3CA or activation of the PIK3CA/AKT/mTor pathway; amplification, rearrangement, and/or loss of transcriptional control of myc family oncogenes (c-, N-, and L-myc; changes in c-myc are found in non-small cell cancers, while changes in all myc family members are found in SCLC); overexpression of bcl-2 and other antiapoptotic proteins; overexpression of other EGFR family members such as Her-2/neu, and ERBB3; and activated expression of the telomerase gene in >90% of lung cancers. Genome-wide approaches are identifying other amplified or mutated dominant oncogenes that could be important new therapeutic targets.
Inactivation of Tumor-Suppressor Genes
A large number of tumor-suppressor genes (recessive oncogenes) have been identified that are inactivated during the pathogenesis of lung cancer. This usually occurs by a tumor-acquired inactivating mutation of one allele [seen, for example, in the p53 and retinoblastoma (RB) tumor-suppressor gene] or tumor-acquired inactivation of expression by tumor-acquired promoter DNA methylation (seen, for example, in the case of the p16 and RASSF1A tumor-suppressor genes), which is then coupled with physical loss of the other parental allele ("loss of heterozygosity"). This leaves the tumor cell with only the functionally inactive allele and thus loss of function of the growth-regulatory tumor-suppressor gene. Genome-wide approaches have identified many such genes involved in lung cancer pathogenesis, including p53, RB, RASSF1A, SEMA3B, SEMA3F, FUS1, p16, LKB1, RAR, and FHIT. Several tumor-suppressor genes on chromosome 3p appear to be involved in nearly all lung cancers. Allelic loss for this region occurs very early in lung cancer pathogenesis, including in histologically normal smoking-damaged lung epithelium.
Autocrine Growth Factors
The large number of genetic and epigenetic lesions shows that lung cancer, like other common epithelial malignancies, arises as a multistep process that is likely to involve both carcinogens causing mutation ("initiation") and tumor promoters. Prevention can be directed at both processes. Lung cancer cells produce many peptide hormones and express receptors for these hormones. They can promote tumor cell growth in an "autocrine" fashion.
Highly carcinogenic derivatives of nicotine are formed in cigarette smoke. Lung cancer cells of all histologic types (and the cells from which they are derived) express nicotinic acetylcholine receptors. Nicotine activates signaling pathways in tumor and normal cells that block apoptosis. Thus, nicotine itself could be directly involved in lung cancer pathogenesis both as a mutagen and tumor promoter.
Inherited Predisposition to Lung Cancer
While an inherited predisposition to develop lung cancer is not common, several features suggest a potential for familial association. People with inherited mutations in RB (patients with retinoblastomas living to adulthood) and p53 (Li-Fraumeni syndrome) genes may develop lung cancer. First-degree relatives of lung cancer probands have a two- to threefold excess risk of lung cancer or other cancers, many of which are not smoking-related. An as yet unidentified gene in chromosome region 6q23 was found to segregate in families at high risk of developing lung cancer of all histologic types. Finally, certain polymorphisms of the P450 enzyme system (which metabolizes carcinogens) or chromosome fragility (mutagen sensitivity) genotypes are associated with the development of lung cancer. The use of any of these inherited differences to identify persons at very high risk of developing lung cancer would be useful in early detection and prevention efforts.
Therapy Targeted at Molecular Abnormalities
A detailed understanding of the molecular pathogenesis should be applicable to new methods of early diagnosis, prevention, and treatment of lung cancer. Two examples of this translation involve EGFR and vascular endothelial growth factor (VEGF). EGFR belongs to the ERBB (HER) family of protooncogenes, including EGFR (ERBB1), Her2/neu (ERBB2), HER3 (ERBB3), and HER4 (ERBB4), cell-surface receptors consisting of an extracellular ligand-binding domain, a transmembrane structure, and an intracellular tyrosine kinase (TK) domain. The binding of ligand to receptor activates receptor dimerization and TK autophosphorylation, initiating a cascade of intracellular events, leading to increased cell proliferation, angiogenesis, metastasis, and a decrease in apoptosis (Chap. 80). Overexpression of EGFR protein or amplification of the EGFR gene has been found in as many as 70% of NSCLCs.
Activating/oncogenic mutations (usually a missense or a small deletion mutation) in the TK domain of EGFR have been identified. These are found most commonly in women, East Asians, patients who have never smoked, and those with adenocarcinoma and BAC histology. This is also the group of patients who are most likely to have dramatic responses to drugs that inhibit TK activation [tyrosine kinase inhibitors (TKIs)]. EGFR mutations are almost never found in cancers other than lung cancer, nor in lung cancers that have KRAS mutations. These EGFR mutations, often associated with amplification of the EGFR gene, usually confer sensitivity of these lung cancers to EGFR TKIs (such as gefitinib or erlotinib), resulting in clinically beneficial tumor responses that unfortunately are still not permanent. In many cases the development of EGFR TKI resistance is associated with the development of another mutation in the EGFR gene (T790M mutation), or amplification of the c-met oncogene. However, other drugs with EGFR TKI activity are in development to which the lung cancers with these resistance mutations will respond as are drugs targeting c-met or its pathways.
The discovery of EGFR mutation/amplification driving lung cancer growth and the dramatic response of these tumors to oral EGFR TKI therapy has prompted a widespread search for other drugs "targeted" against oncogenic changes in lung cancer. An important example of another such target is VEGF, which, while not mutated, is inappropriately produced by lung cancers and stimulates tumor angiogenesis (Chap. 80). VEGF is often overexpressed in lung cancer, and the resulting increase in tumor microvessel density correlates with poor prognosis. A monoclonal antibody to the VEGF ligand, bevacizumab, has significant antitumor effects when used with chemotherapy in lung cancer (see below).
Molecular Profiles Predict Survival and Response
Just as the presence of EGFR TK domain mutations and amplification is an excellent predictor of response to EGFR TKIs, molecular predictors of response to standard chemotherapy and other new targeted agents are being sought. Lung cancers can be molecularly typed at the time of diagnosis to yield information that predicts survival and defines agents to which the tumor is most likely to respond. One example is the identification of alterations in lung cancer DNA repair pathways that may predict resistance to chemotherapy. Patients whose tumors exhibit low activity of the excision-repair-cross complementation group 1 (ERCC1) proteins typically have a worse prognosis as they are unable to repair DNA adducts in the tumor. However, retrospective analysis shows that when treated with cisplatin, patients with tumors expressing low levels of ERCC1 activity appear to do better, as they are unable to repair DNA adducts caused by cisplatin, while patients with high ERCC1 activity actually do worse with cisplatin-based chemotherapy. Although these protein or gene expression "signatures" have yet to be validated in large prospective studies, it is possible that such information will allow future therapy to be tailored to the characteristics of each patient's tumor. Mass spectroscopy-based proteomic studies have identified unique protein patterns in the serum of patients, one of which allows for early diagnosis, while another can predict sensitivity or resistance to drugs. However, such methods have not been validated and may be difficult to implement in a patient care setting.

Clinical Manifestations

Lung cancer gives rise to signs and symptoms caused by local tumor growth, invasion or obstruction of adjacent structures, growth in regional nodes through lymphatic spread, growth in distant metastatic sites after hematogenous dissemination, and remote effects of tumor products (paraneoplastic syndromes) 

Although 5–15% of patients with lung cancer are identified while they are asymptomatic, usually as a result of a routine chest radiograph or through the use of screening CT scans, most patients present with some sign or symptom. Central or endobronchial growth of the primary tumor may cause cough, hemoptysis, wheeze and stridor, dyspnea, and postobstructive pneumonitis (fever and productive cough). Peripheral growth of the primary tumor may cause pain from pleural or chest wall involvement, dyspnea on a restrictive basis, and symptoms of lung abscess resulting from tumor cavitation. Regional spread of tumor in the thorax (by contiguous growth or by metastasis to regional lymph nodes) may cause tracheal obstruction, esophageal compression with dysphagia, recurrent laryngeal nerve paralysis with hoarseness, phrenic nerve paralysis with elevation of the hemidiaphragm and dyspnea, and sympathetic nerve paralysis with Horner's syndrome (enophthalmos, ptosis, miosis, and ipsilateral loss of sweating). Malignant pleural effusion often leads to dyspnea. Pancoast's (or superior sulcus tumor) syndrome results from local extension of a tumor growing in the apex of the lung with involvement of the eighth cervical and first and second thoracic nerves, with shoulder pain that characteristically radiates in the ulnar distribution of the arm, often with radiologic destruction of the first and second ribs. Often Horner's syndrome and Pancoast's syndrome coexist. Other problems of regional spread include superior vena cava syndrome from vascular obstruction; pericardial and cardiac extension with resultant tamponade, arrhythmia, or cardiac failure; lymphatic obstruction with resultant pleural effusion; and lymphangitic spread through the lungs with hypoxemia and dyspnea. In addition, BAC can spread transbronchially, producing tumor growing along multiple alveolar surfaces with impairment of gas exchange, respiratory insufficiency, dyspnea, hypoxemia, and sputum production.
Extrathoracic metastatic disease is found at autopsy in >50% of patients with squamous carcinoma, 80% of patients with adenocarcinoma and large cell carcinoma, and >95% of patients with small cell cancer. Lung cancer metastases may occur in virtually every organ system. Common clinical problems related to metastatic lung cancer include brain metastases with headache, nausea, and neurologic deficits; bone metastases with pain and pathologic fractures; bone marrow invasion with cytopenias or leukoerythroblastosis; liver metastases causing liver dysfunction, biliary obstruction, anorexia, and pain; lymph node metastases in the supraclavicular region and occasionally in the axilla and groin; and spinal cord compression syndromes from epidural or bone metastases. Adrenal metastases are common but rarely cause adrenal insufficiency.
Paraneoplastic syndromes are common in patients with lung cancer and may be the presenting finding or first sign of recurrence. In addition, paraneoplastic syndromes may mimic metastatic disease and, unless detected, lead to inappropriate palliative rather than curative treatment. Often the paraneoplastic syndrome may be relieved with successful treatment of the tumor. In some cases, the pathophysiology of the paraneoplastic syndrome is known, particularly when a hormone with biologic activity is secreted by a tumor (Chap. 96). However, in many cases the pathophysiology is unknown. Systemic symptoms of anorexia, cachexia, weight loss (seen in 30% of patients), fever, and suppressed immunity are paraneoplastic syndromes of unknown etiology. Endocrine syndromes are seen in 12% of patients: hypercalcemia and hypophosphatemia resulting from the ectopic production by squamous tumors of parathyroid hormone (PTH) or, more commonly, PTH-related peptide; hyponatremia with the syndrome of inappropriate secretion of antidiuretic hormone or possibly atrial natriuretic factor by small cell cancer; and ectopic secretion of ACTH by small cell cancer. ACTH secretion usually results in additional electrolyte disturbances, especially hypokalemia, rather than the changes in body habitus that occur in Cushing's syndrome from a pituitary adenoma.
Skeletal–connective tissue syndromes include clubbing in 30% of cases (usually non-small cell carcinomas) and hypertrophic pulmonary osteoarthropathy in 1–10% of cases (usually adenocarcinomas), with periostitis and clubbing causing pain, tenderness, and swelling over the affected bones and a positive bone scan. Neurologic-myopathic syndromes are seen in only 1% of patients but are dramatic and include the myasthenic Eaton-Lambert syndrome and retinal blindness with small cell cancer, while peripheral neuropathies, subacute cerebellar degeneration, cortical degeneration, and polymyositis are seen with all lung cancer types. Many of these are caused by autoimmune responses such as the development of anti-voltage-gated calcium channel antibodies in the Eaton-Lambert syndrome (Chap. 97). Coagulation, thrombotic, or other hematologic manifestations occur in 1–8% of patients and include migratory venous thrombophlebitis (Trousseau's syndrome), nonbacterial thrombotic (marantic) endocarditis with arterial emboli, disseminated intravascular coagulation with hemorrhage, anemia, granulocytosis, and leukoerythroblastosis. Thrombotic disease complicating cancer is usually a poor prognostic sign. Cutaneous manifestations such as dermatomyositis and acanthosis nigricans are uncommon (1%), as are the renal manifestations of nephrotic syndrome or glomerulonephritis (1%).

Diagnosis and Staging

Screening

Most patients with lung cancer present with advanced disease, raising the question of whether screening would detect these tumors at an earlier stage when they are theoretically more curable. The role of screening high-risk patients (for example current or former smokers >50 years of age) for early stage lung cancers is debated. Results from five randomized screening studies in the 1980s of chest x-rays with or without cytologic analysis of sputum did not show any impact on lung cancer–specific mortality from screening high-risk patients, although earlier-stage cancers were detected in the screened groups. These studies have been criticized for their design and statistical analyses, but they led to current recommendations not to use these tools to screen for lung cancer. However, low-dose, noncontrast, thin-slice, helical, or spiral CT has emerged as a possible new tool for lung cancer screening. Spiral CT is a scan in which only the pulmonary parenchyma is examined, thus negating the use of intravenous contrast and the necessity of a physician being present at the exam. The scan can usually be done quickly (within one breath) and involves low doses of radiation. In a nonrandomized study of current and former smokers from the Early Lung Cancer Action Project (ELCAP), low-dose CT was shown to be more sensitive than chest x-ray for detecting lung nodules and lung cancer in early stages. Survival from date of diagnosis is also long (10-year survival predicted to be 92% in screening-detected stage I NSCLC patients). Other nonrandomized CT screening studies of asymptomatic current or former smokers also found that early lung cancer cases were diagnosed more often with CT screening than predicted by standard incidence data. However, no decline in the number of advanced lung cancer cases or deaths from lung cancer was noted in the screened group. Thus, spiral CT appears to diagnose more lung cancer without improving lung cancer mortality. Concerns include the influence of lead-time bias, length-time bias, and over-diagnosis (cancers so slow-growing that they are unlikely to cause the death of the patient). Over-diagnosis is a well-established problem in prostate cancer screening, but it is surprising that some lung cancers are not fatal. However, many of the small adenocarcinomas found as "ground glass" opacities on screening CT appear to have such long doubling times (>400 days) that they may never harm the patient. While CT screening will detect lung cancer in 1–4% of the patients screened over a 5-year period, it also detects a substantial number of false-positive lung lesions (ranging from 25 to 75% in different series) that need follow-up and evaluation. The appropriate management of these small lesions is undefined. Unnecessary treatment of these patients may include thoracotomy and lung resection, thus adding to the cost, mortality, and morbidity of treatment. A large, randomized trial of CT screening for lung cancer (National Lung Cancer Screening Trial) involving ~55,000 individuals has completed accrual and will provide definitive data in the next several years on whether screening reduces lung cancer mortality. Until these results become available, routine CT screening for lung cancer cannot be recommended for any risk group. For those patients who want to be screened, physicians need to discuss the possible benefits and risks of such screening, including the risk of false-positive scans that could result in multiple follow-up CTs and possible biopsies for a malignancy that may not be life-threatening.

Establishing a Diagnosis of Lung Cancer

Once signs, symptoms, or screening studies suggest lung cancer, a tissue diagnosis must be established. Tumor tissue can be obtained by a bronchial or transbronchial biopsy during fiberoptic bronchoscopy; by node biopsy during mediastinoscopy; from the operative specimen at the time of definitive surgical resection; by percutaneous biopsy of an enlarged lymph node, soft tissue mass, lytic bone lesion, bone marrow, or pleural lesion; by fine-needle aspiration of thoracic or extrathoracic tumor masses using CT guidance; or from an adequate cell block obtained from a malignant pleural effusion. In most cases, the pathologist should be able to make a definite diagnosis of epithelial malignancy and distinguish small cell from non-small cell lung cancer.
Staging Patients with Lung Cancer
Lung cancer staging consists of two parts: first, a determination of the location of tumor (anatomic staging) and, second, an assessment of a patient's ability to withstand various antitumor treatments (physiologic staging). In a patient with NSCLC, resectability (whether the tumor can be entirely removed by a standard surgical procedure such as a lobectomy or pneumonectomy), which depends on the anatomic stage of the tumor, and operability (whether the patient can tolerate such a surgical procedure), which depends on the cardiopulmonary function of the patient, are determined.
Non-Small Cell Lung Cancer
The TNM International Staging System should be used for cases of NSCLC, particularly in preparing patients for curative attempts with surgery or radiotherapy (Table 85-2). The various T (tumor size), N (regional node involvement), and M (presence or absence of distant metastasis) factors are combined to form different stage groups. At presentation, approximately one-third of patients have disease localized enough for a curative attempt with surgery or radiotherapy (patients with stage I or II disease and some with stage IIIA disease), one-third have distant metastatic disease (stage IV disease), and one-third have local or regional disease that may or may not be amenable to a curative attempt (some patients with stage IIIA disease and others with stage IIIB disease) (see below). This staging system provides useful prognostic information.
Tumor, Node, Metastasis International Staging System for Lung Cancer
    5-Year Survival Rate, %
Stage TNM Descriptors Clinical Stage Surgical-Pathologic Stage
IA T1 N0 M0 61 67
IB T2 N0 M0 38 57
IIA T1 N1 M0 34 55
IIB T2 N1 M0 24 39
IIB T3 N0 M0 22 38
IIIA T3 N1 M0 9 25
  T1–2–3 N2 M0 13 23
IIIB T4 N0–1–2 M0 7 <5
  T1–2–3–4 N3 M0 3 <3
IV Any T any N M1 1 <1
Tumor (T) Status Descriptor 
T0 No evidence of a primary tumor
TX Primary tumor cannot be assessed, or tumor proven by the presence of malignant cells in sputum or bronchial washings but not visualized by imaging or bronchoscopy
TIS Carcinoma in situ
T1 Tumor <3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than lobar bronchus (i.e., not in main bronchus)
T2 Tumor with any of following: >3 cm in greatest dimension; involves main bronchus, 2 cm distal to the carina; invades visceral pleura; associated with atelectasis or obstructive pneumonitis extending to hilum but does not involve entire lung
T3 Tumor of any size that directly invades any of the following: chest wall (including superior sulcus tumors), diaphragm, mediastinal pleura, parietal pericardium; or tumor in main bronchus <2 cm distal to carina but without involvement of carina; or associated atelectasis or obstructive pneumonitis of entire lung
T4 Tumor of any size that invades any of the following: mediastinum, heart, great vessels, trachea, esophagus, vertebral body, carina; or tumor with a malignant pleural or pericardial effusion,a or with satellite tumor nodule(s) within the ipsilateral primary-tumor lobe of the lung.
 
Lymph Node (N) Involvement Descriptor 
NX Regional lymph nodes cannot be assessed
N0 No regional lymph node metastasis
N1 Metastasis to ipsilateral peribronchial and/or ipsilateral hilar lymph nodes, and intrapulmonary nodes involved by direct extension of the primary tumor
N2 Metastasis to ipsilateral mediastinal and/or subcarinal lymph node(s)
N3 Metastasis to contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s)
Distant Metastasis (M) Descriptor 
MX Presence of distant metastasis cannot be assessed
M0 No distant metastasis
M1 Distant metastasis presentb 

aMost pleural effusions associated with lung cancer are due to tumor. However, in a few patients with multiple negative cytopathologic exams of a non-bloody, non-exudative pleural or pericardial effusion that clinical judgment dictates is not related to the tumor, the effusion should be excluded as a staging element and the patient's disease staged as T1, T2, or T3.
bSeparate metastatic pulmonary tumor nodule(s) in the ipsilateral nonprimary tumor lobe(s) of the lung are classified as M1.

Source: Adapted from CF Mountain. Revisions in the International System for Staging of Lung Cancer. Chest 111:1710, 1997; with permission.

Small Cell Lung Cancer
A simple two-stage system is used. In this system, limited-stage disease (seen in about 30% of all patients with SCLC) is defined as disease confined to one hemithorax and regional lymph nodes (including mediastinal, contralateral hilar, and usually ipsilateral supraclavicular nodes), while extensive-stage disease (seen in about 70% of patients) is defined as disease exceeding those boundaries. Clinical studies such as physical examination, x-rays, CT and bone scans, and bone marrow examination are used in staging. In part, the definition of limited-stage disease relates to whether the known tumor can be encompassed within a tolerable radiation therapy port. Thus, contralateral supraclavicular nodes, recurrent laryngeal nerve involvement, and superior vena caval obstruction can all be part of limited-stage disease. However, cardiac tamponade, malignant pleural effusion, and bilateral pulmonary parenchymal involvement generally qualify disease as extensive-stage because the organs within a curative radiation therapy port cannot safely tolerate curative radiation doses.
Lung Cancer Staging Procedures
(Table 85-3) All patients with lung cancer should have a complete history and physical examination, with evaluation of all other medical problems, determination of performance status and history of weight loss, and a CT scan of the chest and abdomen with contrast. Positron emission tomography (PET) scans are sensitive in detecting both intrathoracic and metastatic disease. PET is useful in assessing the mediastinum and solitary pulmonary nodules. A standardized uptake value (SUV) of >2.5 is highly suspicious for malignancy. False negatives can be seen in diabetes, in slow-growing tumors such as BAC, in concurrent infection such as tuberculosis, and in lesions <8 mm. False positives can also be seen in infections and granulomatous disease. Thus, PET should never be used alone to diagnose lung cancer, mediastinal involvement, or metastases. Instead, its primary function is to help guide a mediastinal biopsy for staging purposes and to help identify sites of metastatic disease. Fiberoptic bronchoscopy obtains material for pathologic examination and information on tumor size, location, degree of bronchial obstruction (i.e., assesses resectability), and recurrence.
Pretreatment Staging Procedures for Patients with Lung Cancer
All Patients 
Complete history and physical examination
Determination of performance status and weight loss
Complete blood count with platelet determination
Measurement of serum electrolytes, glucose, and calcium; renal and liver function tests
Electrocardiogram
Skin test for tuberculosis
Chest x-ray
CT scan of chest and abdomen
CT or MRI scan of brain and radionuclide scan of bone if any finding suggests the presence of tumor metastasis in these organs
Fiberoptic bronchoscopy with washings, brushings, and biopsy of suspicious lesions unless medically contraindicated or if it would not alter therapy (e.g., very late stage patient)
X-rays of suspicious bony lesions detected by scan or symptom
Barium swallow radiographic examination if esophageal symptoms exist
Pulmonary function studies and arterial blood gas measurements if signs or symptoms of respiratory insufficiency are present
Biopsy of accessible lesions suspicious for cancer if a histologic diagnosis is not yet made or if treatment or staging decisions would be based on whether or not a lesion contained cancer
Patients with Non-small Cell Lung Cancer Who Have No Contraindicationa to Curative Surgery or Radiotherapy with or without Chemotherapy 
All the above procedures, plus the following:
PET scan to evaluate mediastinum and detect metastatic disease
Pulmonary function tests and arterial blood gas measurements
Coagulation tests
CT or MRI scan of brain if symptoms suggestive
Cardiopulmonary exercise testing if performance status or pulmonary function tests are borderline
If surgical resection is planned: surgical evaluation of the mediastinum at mediastinoscopy or at thoracotomy
If the patient is a poor surgical risk or a candidate for curative radiotherapy: transthoracic fine-needle aspiration biopsy or transbronchial forceps biopsy of peripheral lesions if material from routine fiberoptic bronchoscopy is negative
Patients Presenting with Small Cell or Advanced Non-small Cell Lung Cancer 
For proven small cell lung cancer, all the procedures under "All Patients," plus the following:
CT or MRI scan of brain
Bone marrow aspiration and biopsy (if peripheral blood counts abnormal)
For non-small cell lung cancer or cancer of unknown histology, all the procedures under "All Patients," plus the following:
Fiberoptic bronchoscopy if indicated by hemoptysis, obstruction, pneumonitis, or no histologic diagnosis of cancer
Biopsy of accessible lesions suspicious for tumor to obtain a histologic diagnosis or if therapy would be altered by finding of tumor
Transthoracic fine-needle aspiration biopsy or transbronchial forceps biopsy of peripheral lesions if fiberoptic bronchoscopy is negative and no other material exists for a histologic diagnosis
Diagnostic and therapeutic thoracentesis if a pleural effusion is present

aPatients with non-small cell lung cancer and extrathoracic metastatic disease, malignant pleural effusion, or intrathoracic disease beyond the bounds of a tolerable radiotherapy port.
Note: CT, computed tomography; PET, positron emission tomography.
Chest radiographs and CT scans are needed to evaluate tumor size and nodal involvement; old radiographs are useful for comparison. CT scans of the thorax and upper abdomen are of use in the preoperative staging of NSCLC to detect mediastinal nodes and pleural extension and occult abdominal disease (e.g., liver, adrenal), and in planning curative radiation therapy. However, mediastinal nodal involvement should be documented histologically if the findings will influence therapeutic decisions. Thus, sampling of lymph nodes via mediastinoscopy or thoracotomy to establish the presence or absence of N2 or N3 nodal involvement is crucial in considering a curative surgical approach for patients with NSCLC with clinical stage I, II, or III disease, regardless of whether the PET is positive or negative. A preoperative mediastinoscopy may not need to be done in patients with normal-size nodes (by CT) that are PET-negative, as the discovery of micrometastases is unlikely to change the preoperative management of the disease, although lymph node sampling should be done intraoperatively. A standard nomenclature for referring to the location of lymph nodes involved with cancer has evolved (Fig. 85-1). Unless the CT-detected abnormalities are unequivocal, histology of suspicious extrathoracic lesions should be confirmed by procedures such as fine-needle aspiration if the patient would otherwise be considered for curative treatment. In SCLC, CT scans are used in the planning of chest radiation treatment and in the assessment of the response to chemotherapy and radiation therapy. Surgery or radiotherapy can make interpretation of conventional chest x-rays difficult; after treatment, CT scans can provide good evidence of tumor recurrence.
If signs or symptoms suggest involvement by tumor, brain CT or bone scans are performed, as well as radiography of any suspicious bony lesions. Any accessible lesions suspicious for cancer should be biopsied if involvement would influence treatment.
In patients presenting with a mass lesion on chest x-ray or CT scan and no obvious contraindications to a curative approach after the initial evaluation, the mediastinum must be investigated. Approaches vary among centers and include performing chest CT scan and mediastinoscopy (for right-sided tumors) or mediastinotomy (for left-sided lesions) on all patients and proceeding directly to thoracotomy for staging of the mediastinum. Patients who present with disease that is confined to the chest but not resectable, and who thus are candidates for neoadjuvant chemotherapy plus surgery or for curative radiotherapy with or without chemotherapy, should have additional tests done as indicated to evaluate specific symptoms. In patients presenting with NSCLC that is not curable, all the general staging procedures are done, plus fiberoptic bronchoscopy as indicated to evaluate hemoptysis, obstruction, or pneumonitis, as well as thoracentesis with cytologic examination (and chest tube drainage as indicated) if fluid is present. As a rule, a radiographic finding of an isolated lesion (such as an enlarged adrenal gland) should be confirmed as cancer by fine-needle aspiration before a curative attempt is rejected.
Staging of Small Cell Lung Cancer
Pretreatment staging for patients with SCLC includes the initial general lung cancer evaluation with chest and abdominal CT scans (because of the high frequency of hepatic and adrenal involvement) as well as fiberoptic bronchoscopy with washings and biopsies to determine the tumor extent before therapy; brain CT scan (10% of patients have metastases); and radionuclide scans (bone) if symptoms or other findings suggest disease involvement in these areas. Bone marrow biopsies and aspirations are rarely performed given the low incidence of isolated bone marrow metastases. Chest and abdominal CT scans are very useful to evaluate and follow tumor response to therapy, and chest CT scans are helpful in planning chest radiotherapy ports.
If signs or symptoms of spinal cord compression or leptomeningitis develop at any time in lung cancer patients with disease of any histologic type, a spinal CT scan or MRI scan and examination of the cerebrospinal fluid cytology are performed. If malignant cells are detected, radiotherapy to the site of compression and intrathecal chemotherapy (usually with methotrexate) are given. In addition, a brain CT or MRI scan is performed to search for brain metastases, which often are associated with spinal cord or leptomeningeal metastases.
Resectability and Operability
In patients with NSCLC, the following are major contraindications to curative surgery or radiotherapy alone: extrathoracic metastases; superior vena cava syndrome; vocal cord and, in most cases, phrenic nerve paralysis; malignant pleural effusion; cardiac tamponade; tumor within 2 cm of the carina (not curable by surgery but potentially curable by radiotherapy); metastasis to the contralateral lung; bilateral endobronchial tumor (potentially curable by radiotherapy); metastasis to the supraclavicular lymph nodes; contralateral mediastinal node metastases (potentially curable by radiotherapy); and involvement of the main pulmonary artery. Pleural effusions are generally considered malignant regardless of whether they are cytology positive, particularly if they are exudative, bloody, and have no other probable etiology. Most patients with SCLC have unresectable disease; however, if clinical findings suggest the potential for resection (most common with peripheral lesions), that option should be considered.
Physiologic Staging
Patients with lung cancer often have cardiopulmonary and other problems related to chronic obstructive pulmonary disease as well as other medical problems. To improve their preoperative condition, correctable problems (e.g., anemia, electrolyte and fluid disorders, infections, and arrhythmias) should be addressed, smoking stopped, and appropriate chest physical therapy instituted. Since it is not always possible to predict whether a lobectomy or pneumonectomy will be required until the time of operation, a conservative approach is to restrict resectional surgery to patients who could potentially tolerate a pneumonectomy. In addition to nonambulatory performance status, a myocardial infarction within the past 3 months is a contraindication to thoracic surgery because 20% of patients will die of reinfarction. An infarction in the past 6 months is a relative contraindication. Other major contraindications include uncontrolled major arrhythmias, an FEV1 (forced expiratory volume in 1 s) <1 L, CO2 retention (resting PCO2 >45 mmHg), DLCO <40%, and severe pulmonary hypertension. Recommending surgery when the FEV1 is 1.1–2.0 L or <80% predicted requires careful judgment, while an FEV1 >2.5 L or >80% predicted usually permits a pneumonectomy. In patients with borderline lung function but a resectable tumor, cardiopulmonary exercise testing could be performed as part of the physiologic evaluation. This test allows an estimate of the maximal oxygen consumption (O2max). A O2max <15 mL/kg per min predicts for high risk of postoperative complications.
Lung Cancer: Treatment
The overall treatment approach to patients with lung cancer is shown in Table 85-4. Patients should be encouraged to stop smoking, particularly if they will be undergoing surgery or radiation therapy. Those who do fare better than those who continue to smoke.
 Summary of Treatment Approach to Patients with Lung Cancer
Non-Small Cell Lung Cancer 
Stages IA, IB, IIA, IIB, and some IIIA:
Surgical resection for stages IA, IB, IIA, and IIB
Surgical resection with complete-mediastinal lymph node dissection and consideration of neoadjuvant CRx for stage IIIA disease with "minimal N2 involvement" (discovered at thoracotomy or mediastinoscopy)
Consider postoperative RT for patients found to have N2 disease
Stage IB: discussion of risk/benefits of adjuvant CRx; not routinely given
Stage II: Adjuvant CRx
Curative potential RT for "nonoperable" patients
Stage IIIA with selected types of stage T3 tumors:
Tumors with chest wall invasion (T3): en bloc resection of tumor with involved chest wall and consideration of postoperative RT
Superior sulcus (Pancoast's) (T3) tumors: preoperative RT (30–45 Gy) and CRx followed by en bloc resection of involved lung and chest wall with postoperative RT
Proximal airway involvement (<2 cm from carina) without mediastinal nodes: sleeve resection if possible preserving distal normal lung or pneumonectomy
Stages IIIA "advanced, bulky, clinically evident N2 disease" (discovered preoperatively) and IIIB disease that can be included in a tolerable RT port:
Curative potential concurrent RT + CRx if performance status and general medical condition are reasonable; otherwise, sequential CRx followed by RT, or RT alone
Stage IIIB disease with carinal invasion (T4) but without N2 involvement:
Consider pneumonectomy with tracheal sleeve resection with direct reanastomosis to contralateral mainstem bronchus
Stage IV and more advanced IIIB disease:
RT to symptomatic local sites
CRx for ambulatory patients; consider CRx and bevacizumab for selected patients
Chest tube drainage of large malignant pleural effusions
Consider resection of primary tumor and metastasis for isolated brain or adrenal metastases
Small Cell Lung Cancer 
Limited stage (good performance status): combination CRx + concurrent chest RT
Extensive stage (good performance status): combination CRx
Complete tumor responders (all stages): consider prophylactic cranial RT
Poor-performance-status patients (all stages):
Modified-dose combination CRx
Palliative RT
All Patients 
RT for brain metastases, spinal cord compression, weight-bearing lytic bony lesions, symptomatic local lesions (nerve paralyses, obstructed airway, hemoptysis, intrathoracic large venous obstruction, in non-small cell lung cancer and in small cell cancer not responding to CRx)
Appropriate diagnosis and treatment of other medical problems and supportive care during CRx
Encouragement to stop smoking
Entrance into clinical trial, if eligible

Abbreviations: CRx, chemotherapy; RT, radiotherapy.
Management of Occult and Stage 0 Carcinomas
In the uncommon situation where malignant cells are identified in a sputum or bronchial washing specimen but the chest radiograph appears normal (TX tumor stage), the lesion must be localized. More than 90% can be localized by meticulous examination of the bronchial tree with a fiberoptic bronchoscope under general anesthesia and collection of a series of differential brushings and biopsies. Often, carcinoma in situ or multicentric lesions are found in these patients. Current recommendations are for the most conservative surgical resection, allowing removal of the cancer and conservation of lung parenchyma, even if the bronchial margins are positive for carcinoma in situ. The 5-year overall survival rate for these occult cancers is ~60%. Close follow-up of these patients is indicated because of the high incidence of second primary lung cancers (5% per patient per year). One approach to in situ or multicentric lesions uses systemically administered hematoporphyrin (which localizes to tumors and sensitizes them to light) followed by bronchoscopic phototherapy.
Solitary Pulmonary Nodule and "Ground Glass" Opacity
Occasionally, when an x-ray or CT scan is done for another purpose, a patient will present with an incidental finding of an asymptomatic, solitary pulmonary nodule (SPN, defined as an x-ray density completely surrounded by normal aerated lung, with circumscribed margins, of any shape, usually 1–6 cm in greatest diameter). A decision to resect or follow the nodule must be made. Nodules of this size discovered in CT screening for lung cancer would also be of the size requiring a biopsy for tissue. Approximately 35% of all such lesions in adults are malignant, most being primary lung cancer, while <1% are malignant in nonsmokers <35 years of age. A complete history, including a smoking history, physical examination, routine laboratory tests, chest CT scan, fiberoptic bronchoscopy, and old chest x-rays or CT scans are obtained if available.
PET scans are useful in detecting lung cancers >7–8 mm in diameter. If no diagnosis is immediately apparent, the following risk factors would all argue strongly in favor of proceeding with resection to establish a histologic diagnosis: a history of cigarette smoking; age 35 years; a relatively large lesion; lack of calcification; chest symptoms; associated atelectasis, pneumonitis, or adenopathy; growth of the lesion revealed by comparison with old x-rays/CT scans; or a positive PET scan. At present, only two radiographic criteria are reliable predictors of the benign nature of an SPN: lack of growth over a period >2 years and certain characteristic patterns of calcification. Calcification alone does not exclude malignancy. However, a dense central nidus, multiple punctate foci, and "bull's eye" (granuloma) and "popcorn ball" (hamartoma) calcifications are all highly suggestive of a benign lesion. An algorithm for evaluating an SPN is shown in Fig. 85-2.
When old x-rays are not available, the PET scan is negative, and the characteristic calcification patterns are absent, the following approach is reasonable. Nonsmoking patients <35 years can be followed with serial CT every 3 months for 1 year and then yearly; if any significant growth is found, a histologic diagnosis is needed. For patients >35 years and all patients with a smoking history, a histologic diagnosis must be made, regardless of whether the lesion is PET positive or negative, since slow-growing cancers such as BAC can be PET negative. The sample for histologic diagnosis can be obtained either at the time of nodule resection or, if the patient is a poor operative risk, via video-assisted thoracic surgery (VATS) or transthoracic fine-needle biopsy. Some institutions use preoperative fine-needle aspiration on all such lesions; however, all positive lesions have to be resected, and negative cytologic findings in most cases have to be confirmed by histology on a resected specimen. Much has been made of sparing patients an operation; however, the high probability of finding a malignancy (particularly in smokers >35 years) and the excellent chance for surgical cure when the tumor is small both suggest an aggressive approach to these lesions.
Since the advent of screening CTs, small "ground-glass" opacities ("GGOs") have often been observed, particularly as the increased sensitivity of CTs enables detection of smaller lesions. Many of these GGOs, when biopsied, are found to be BAC. Some of the GGOs are semiopaque and referred to as "partial" GGOs, which are often more slowly growing, with atypical adenomatous hyperplasia histology, a lesion of unclear prognostic significance. By contrast, "solid" GGOs have a faster growth rate and usually are typical adenocarcinomas histologically.
Non-Small Cell Lung Cancer
NSCLC Stages I and II
Surgery
In patients with NSCLC stages IA, IB, IIA and IIB (Table 85-2) who can tolerate operation, the treatment of choice is pulmonary resection. If a complete resection is possible, the 5-year survival rate for N0 disease is about 60–80%, depending on the size of the tumor. The 5-year survival drops to about 50% when N1 (hilar node involvement) disease is present.
The extent of resection is a matter of surgical judgment based on findings at exploration. Clinical trials have shown that lobectomy is superior to wedge resection in reducing the rate of local recurrence. Pneumonectomy is reserved for patients with tumors involving multiple lobes or very central tumors and should only be performed in patients with excellent pulmonary reserve. In addition, patients undergoing a right-sided pneumonectomy after induction chemotherapy and radiation therapy (see below) have a high mortality rate and should be carefully selected before surgery. Wedge resection and segmentectomy (potentially by VATS) are reserved for patients with poor pulmonary reserve and small peripheral lesions.
Radiotherapy with Curative Intent
Patients with stage I or II disease who refuse surgery or are not candidates for pulmonary resection should be considered for radiation therapy with curative intent. The decision to administer high-dose radiotherapy is based on the extent of disease and the volume of the chest that requires irradiation. Patients with distant metastases, malignant pleural effusion, or cardiac involvement are not considered candidates for curative radiation treatment. The long-term survival for patients with all stages of lung cancer who receive radiation with curative intent is about 20%. In addition to being potentially curative, radiotherapy may increase the quality and length of life by controlling the primary tumor and preventing symptoms related to local recurrence in the lung.
Treatment with curative intent usually involves midplane doses of 60–64 Gy, while palliative thoracic radiation (see below) involves delivery of 30–45 Gy. The major dose-limiting concern is the amount of lung parenchyma and other organs in the thorax that are included in the treatment plan, including the spinal cord, heart, and esophagus. In patients with a major degree of underlying pulmonary disease, the treatment plan may have to be compromised because of the deleterious effects of radiation on pulmonary function.
The most common side effect of curative thoracic radiation is esophagitis. Other side effects include fatigue, radiation myelitis (rare), and radiation pneumonitis, which can sometimes progress to pulmonary fibrosis. The risk of radiation pneumonitis is proportional to the radiation dose and the volume of lung in the field. The full clinical syndrome (dyspnea, fever, and radiographic infiltrate corresponding to the treatment port) occurs in 5% of cases and is treated with glucocorticoids. Acute radiation esophagitis occurs during treatment but is usually self-limited, unlike spinal cord injury, which may be permanent and should be avoided by careful treatment planning. Brachytherapy (local radiotherapy delivered by placing radioactive "seeds" in a catheter in the tumor bed) provides a way to give a high local dose while sparing surrounding normal tissue.
NSCLC Stage IA
Patients with resected stage IA NSCLC receive no other therapy but are at a high risk of recurrence (~2–3% annually) or developing a second primary lung cancer. Thus, it is reasonable to follow these patients with CT scans for the first 5 years and consider entering them onto early detection and chemoprevention studies.
Adjuvant Chemotherapy for NSCLC Stages IB and II
A meta-analysis of more than 4300 patients showed a trend toward improved survival of ~5% at 5 years with cisplatin-based adjuvant therapy (p = .08). Subsequently, three randomized studies demonstrated no significant survival advantage despite the addition of more "modern" postoperative adjuvant chemotherapy regimens. However, since then at least three additional randomized trials and two meta-analyses showed a survival benefit in response to postoperative adjuvant-based therapy (Table 85-5). Consequently, adjuvant chemotherapy is now routinely recommended in NSCLC patients with a good performance status and stage IIA or IIB disease, though the beneficial effects are modest.
 Randomized Studies of Adjuvant Chemotherapy in NSCLC
Study Treatment Number of Patients 5-Year Survival (%) Median Survival Hazard Ratio (95% CI) p Value 
ECOG 3590 (II–IIIA) Surgery RT vs. Surgery + post-op concurrent RT + cis/etoposide
242
246
39%
33%
39 months vs. 38 months 0.93 (0.74–1.18) 0.56
ALPI (I–IIIA) Surgery alone vs. Surgery + post-op mitomycin/vindesine/cisplatin
603
606
51%
43%
NR 0.96 (0.8–1.1) 0.59
Big Lung Trial (I–IIIB) Surgery alone vs. Surgery + post-op chemotherapya 
189
192
 
33 months
34 months
1.02 (0.77–1.35) 0.90
IALTIB–IIIA Surgery alone vs. Surgery + post-op Cis + VP16/vinca
405
361
40%
44.5%
NR 0.86 (0.76–0.98) <0.03
UFTIA–IB Surgery alone vs. Surgery + post-op UFT
488
469
85%
88%
0.71 (0.52–0.98) 0.04
CALGB IB (ASCO 06) Surgery alone vs. Surgery + post-op carbo/paclitaxel
172
172
57%
59%
78 months
95 months
0.80 (0.60–1.07) 0.10
NCI-CIB–II Surgery alone vs. Surgery + post-op Cis/vinorelbine
241
241
54%
69%
73 months
94 months
0.69 (0.52–0.91) 0.04
ANITA IB, II, IIIA Surgery alone vs. Surgery + post-op Cis/vinorelbine
433
407
43%
51%
44 months
66 months
0.79 (50–88.5) 0.017

aChemotherapy allowed: mitomycin, cisplatin, ifosfamide; mitomycin, vinblastine, cisplatin; cisplatin, vindesine; cisplatin, vinorelbine.
Note: RT, radiation therapy; NR, not reported; UFT, tegafur and uracil.
The role of adjuvant chemotherapy for stage IB disease is undefined. Subset analysis of all the randomized studies showed no benefit in patients with stage IB. In addition, one clinical trial focusing solely on IB disease and using carboplatin and paclitaxel (one of the most commonly used regimens for advanced disease) found a hazard ratio of 0.80 (20% reduction in death with adjuvant chemotherapy) that was not statistically significant. Thus, patients with stage IB NSCLC are not routinely given adjuvant therapy.
Adjuvant Radiotherapy for NSCLC Stages I–II
After apparent complete resection, postoperative adjuvant radiation therapy does not improve survival and may actually be detrimental to survival in N0 and N1 disease.
Superior Sulcus or Pancoast Tumors
Non-small cell carcinomas of the superior pulmonary sulcus producing Pancoast's syndrome appear to behave differently than lung cancers at other sites and are usually treated with combined radiotherapy and surgery. Patients with these carcinomas should have the usual preoperative staging procedures, including mediastinoscopy and CT and PET scans, to determine tumor extent and a neurologic examination (and sometimes nerve conduction studies) to document involvement or impingement of nerves in the region. If mediastinoscopy is negative, curative approaches may be used in treating Pancoast's syndrome despite its apparent locally invasive nature. The best results reported thus employed concurrent preoperative irradiation [30 Gy in 10 treatments] and cisplatin and etoposide, followed by an en bloc resection of the tumor and involved chest wall 3–6 weeks later; 65% of thoracotomy specimens showed either a complete response or minimal residual microscopic disease on pathologic evaluation. The 2-year survival rate was 55% for all eligible patients and 70% for patients who had a complete resection.
NSCLC with T3, N0 Disease (Stage IIB)
The subset of T3, N0 disease (which does not present as Pancoast tumor) was initially considered stage III disease. However, it has a different natural history and treatment strategy than stage III N2 disease and is now considered as stage IIB. Patients with peripheral chest wall invasion should have resection of the involved ribs and underlying lung. Chest wall defects are then repaired with chest wall musculature or Marlex mesh and methylmethacrylate. Five-year survival rates as high as 35–50% have been found, and adjuvant chemotherapy is usually recommended.
NSCLC Stage III
Treatment of locally advanced NSCLC is one of the most controversial issues in the management of lung cancer. Treatment options include a local therapy (surgery or radiation therapy) combined with systemic chemotherapy to control micrometastases. Interpretation of the results of clinical trials involving patients with locally advanced disease has been clouded by a number of issues, including changing diagnostic techniques, different staging systems, and heterogeneous patient populations with tumors that range from nonbulky stage IIIA (clinical N1 nodes with N2 nodes discovered only at the time of surgery, despite a negative mediastinoscopy) to bulky N2 nodes (enlarged adenopathy clearly visible on chest x-rays or multiple nodal level involvement) to clearly inoperable stage IIIB disease. Thus, a team approach involving pulmonary medicine, thoracic surgery, and medical and radiation oncology is essential for the management of these patients.
NSCLC Stage IIIA
Nonbulky IIIASurgery for N2 disease is a controversial area in the management of lung cancer. Patients with N2 disease can be divided into "minimal" disease (involvement of only one node with microscopic foci, usually discovered at thoracotomy or mediastinoscopy) and the more common "advanced" bulky disease, clinically obvious on CT scans and discovered preoperatively. Patients who have an incidental finding of N2 disease at the time of resection should receive adjuvant chemotherapy.
Bulky IIIA
No evidence suggests that patients with "bulky," multilevel ipsilateral mediastinal nodes (N2) have improved survival with surgery and either pre- or postoperative chemotherapy compared to treatment with chemotherapy plus radiation therapy. This important issue was addressed in the multicenter randomized Intergroup 0139 Trial involving patients with pathologically staged N2 disease who received 45 Gy of induction radiation therapy plus two cycles of cisplatin and etoposide to "debulk" tumors. The patients were then randomly assigned to surgical resection of any residual tumor or to boost radiation therapy plus an additional two cycles of chemotherapy. Although a significant improvement in progression-free survival was observed at 5 years for those patients randomized to surgical resection (22% vs. 11%; p = .017), the difference in 5-year overall survival while favoring surgery (22% vs. 11%; p = .10) was not significant. This is important since treatment-related mortality was greater in the surgery arm (8% vs. 2%), with the majority of deaths occurring in patients undergoing pneumonectomy. Patients who had persistent N2 disease following neoadjuvant chemotherapy did particularly poorly, leading some oncologists to conclude that surgery for bulky IIIA disease should only be conducted in patients who have clearing of their mediastinal nodes following neoadjuvant therapy. The main role of neoadjuvant chemotherapy is to control micrometastatic disease, and if this macroscopically evident disease is not sensitive to chemotherapy, it is unlikely that the microscopic disease will be controlled. Thus, surgical removal of the primary tumor after such chemotherapy is probably fruitless. Likewise, neoadjuvant chemotherapy generally should not be used to render inoperable disease operable. One exception to this approach is T4, N0 or T4, N1 (stage IIIB, see below) disease for which preoperative chemotherapy may provide enough tumor debulking to allow otherwise unresectable disease to be resected. Chemotherapy may allow chest wall resection for direct extension of tumor, tracheal sleeve pneumonectomy, and sleeve lobectomy for lesions near the carina.
Bulky NSCLC Stage IIIA and Dry IIIB (IIIB Without a Pleural Effusion)
The presence of pathologically involved N2 nodes should be confirmed histologically because enlarged nodes detected by CT will be negative for cancer in ~30% of patients. Chemotherapy plus radiation therapy is the treatment of choice for patients with bulky stage IIIA or IIIB disease without pleural effusion (referred to as "dry IIIB"). Randomized studies demonstrate an improvement in median and long-term survival with chemotherapy followed by radiation therapy, compared with radiation therapy alone. Subsequent randomized trials have shown that administering chemotherapy and radiation therapy concurrently results in improved survival compared to sequential chemotherapy and radiation therapy, albeit with more side effects, such as fatigue, esophagitis, and neutropenia. Frequently, an additional two to three cycles of chemotherapy are also given. However, it is not clear whether these additional cycles should be administered before or after the chemoradiation, what the optimal drugs are, or whether doses should be attenuated during the radiation but given more frequently. (Lower doses of drugs may "sensitize" the tumor to radiation therapy but may not by themselves remove other microscopic disease.)
Disseminated Non-Small Cell Lung Cancer
Symptomatic Management of Metastatic Disease
Patients who present with or progress to metastatic NSCLC have a poor prognosis, as do patients with pleural effusions. Untreated, the median survival of both of these patient groups is roughly 4–6 months. They are often treated in the same way. Standard medical management, the judicious use of pain medications, the appropriate use of radiotherapy, and outpatient chemotherapy form the cornerstone of this management.
Palliative Radiation Therapy
Patients whose primary tumor is causing urgent severe symptoms such as bronchial obstruction with pneumonitis, hemoptysis, upper airway or superior vena cava obstruction, brain or spinal cord compression, or painful bony metastases should have radiotherapy to the primary tumor to relieve these symptoms. Usually, radiation therapy is given as a course of 30–40 Gy over 2–4 weeks for palliative purposes. Radiation therapy provides relief of intrathoracic symptoms: hemoptysis, 84%; superior vena cava syndrome, 80%; dyspnea, 60%; cough, 60%; atelectasis, 23%; and vocal cord paralysis, 6%. Cardiac tamponade (treated with pericardiocentesis and radiation therapy to the heart), painful bony metastases (with relief in 66%), brain or spinal cord compression, and brachial plexus involvement may also be palliated with radiotherapy.
Brain metastases are often isolated sites of relapse in patients with adenocarcinoma of the lung otherwise controlled by surgery or radiotherapy. These are usually treated with radiation therapy and, in highly selected cases, with surgical resection. Usually, in addition to radiotherapy for brain metastases and cord compression, dexamethasone (25–100 mg/d in four divided doses) is also given and then rapidly tapered to the lowest dosage that relieves symptoms. Because of the high frequency of brain metastases, the use of prophylactic cranial irradiation (PCI, given to the whole brain before metastatic disease becomes manifest) has been considered. However, PCI is of no proven value. Screening asymptomatic patients with head CT scans to find such lesions before such metastases become clinically evident is also not proven beneficial.
Pleural effusions are common and are usually treated with thoracentesis. If they recur and are symptomatic, a pleurex catheter or chest tube drainage followed by pleurodesis with a sclerosing agent such as intrapleural talc, bleomycin, or tetracycline can be used. These sclerosing agents may be administered through the chest tube, or, in the case of talc, via thorascopic insufflation. In the former case, the chest cavity is completely drained. Xylocaine 1% is instilled (15 mL), followed by 50 mL normal saline. Then the sclerosing agent is dissolved in 100 mL normal saline, and this solution is injected through the chest tube. The chest tube is clamped for 4 h if tolerated, and the patient is rotated onto different sides to distribute the sclerosing agent. The chest tube is removed 24–48 h later, after drainage has become slight (usually <100 mL/24 h). While sclerosing agents have been widely used, an indwelling pleurex catheter is equivalent to chest tube drainage and better tolerated by patients. In this situation, the pleurex catheter is tunneled under the skin and can remain in place for weeks. The patient periodically drains the catheter into a specially designed bag, as needed.
Symptomatic endobronchial lesions that recur after surgery or radiotherapy or develop in patients with severely compromised pulmonary function are difficult to treat with conventional therapy. Neodymium-YAG (yttrium-aluminum-garnet) laser therapy administered through a flexible fiberoptic bronchoscope (usually under general anesthesia) can provide palliation in 80–90% of such patients even when the tumor has relapsed after radiotherapy. Local radiotherapy delivered by brachytherapy, photodynamic therapy using a photosensitizing agent, and endobronchial stents are other measures that can relieve airway obstruction from recurrent tumor.
Chemotherapy
Chemotherapy palliates symptoms, improves the quality of life, and improves survival in newly diagnosed patients with stage IV NSCLC, particularly in patients with good performance status. Whereas the median survival for untreated patients is roughly 4–6 months, and 1-year survival is 5–10%, with combination chemotherapy the median survival is 8–10 months, 1-year survival is 30–35%, and 2-year survival 10–15%. Combination chemotherapy produces an objective tumor response in 20–30% of patients, although the response is complete in <5%. In addition, economic analysis has found chemotherapy to be cost-effective palliation for stage IV NSCLC. However, the use of chemotherapy for NSCLC requires clinical experience and careful judgment to balance potential benefits and toxicities for these patients.
Chemotherapy for previously untreated, good-performance-status patients typically consists of two drugs ("doublets"). Traditionally, one of the two drugs has been either cisplatin or carboplatin, and the other drug is a taxane (paclitaxel or docetaxel), gemcitabine, or a vinca alkaloid such as vinorelbine. No major difference in outcome has been observed between the standard chemotherapy doublets, although they differ in terms of schedule, side effects, and cost. Cytotoxic chemotherapy for first-line chemotherapy is typically administered for four to six cycles; no benefit has been shown for continuing the same chemotherapy beyond that point. After four to six cycles, chemotherapy is usually stopped and the patient observed closely for tumor progression, at which point second-line chemotherapy may be started if the patient's performance status remains good. Nausea with typical first-line regimens is usually mild, particularly when 5-HT3 serotonin antagonists are used as antiemetics. Hair loss depends on the choice of regimen and should be discussed with the patient. All regimens cause myelosuppression, but the incidence of neutropenic fevers, bleeding episodes, or anemia requiring transfusions is low. Growth-factor support is rarely needed. Elderly patients without significant comorbid conditions benefit from and tolerate chemotherapy much the same as their younger counterparts. However, patients with a poorer performance status seem to obtain less benefit.
Docetaxel and pemetrexed are second-line agents for patients who have progressive disease on first-line chemotherapy and still have a good performance status. Docetaxel improves progression-free survival and overall survival compared to best supportive care, and pemetrexed has roughly the same efficacy as docetaxel, but with fewer side effects.
VEGF Targeted Therapy
Bevacizumab, a monoclonal antibody to VEGF, improves response rate, progression-free survival, and overall survival of patients with advanced disease when combined with chemotherapy (paclitaxel/carboplatin). Median, 1-year, and 2-year survival in response to chemotherapy plus bevacizumab was 12.3 months, 51%, and 23%, compared, respectively, to 10.3 months, 44%, and 15% with chemotherapy alone (hazard ratio 0.79, p = 0.003). A 1-year survival of >50% and a 2-year survival of >20% represents a significant improvement in long-term prognosis. The dose of bevacizumab administered on this trial was 15 mg/kg IV every 3 weeks. Bevacizumab side effects include bleeding, hypertension, and proteinuria, and the hemorrhagic side effects make this agent risky to use. Patients with squamous cancer cannot receive bevacizumab because of their tendency toward serious hemorrhagic side effects. Patients with brain metastases, hemoptysis, and bleeding disorders or who need anticoagulation are also not eligible to receive the agent. Despite these restrictions and careful patient selection, significant bleeding is noted in about 4% of patients.
EGFR Targeted Therapy
Erlotinib is an oral inhibitor of the EGFR kinase that is used in second- and third-line therapy of NSCLC. Clinical responses have been seen in a large fraction of the small subset of patients with tumors bearing mutations in the EGFR. Prolonged survival with EGFR TKI treatment has also been observed in some patients whose tumors have amplification of the EGFR gene or overexpression of the receptor. Side effects of erlotinib differ from chemotherapy side effects of hair loss, nausea, and neutropenia, but they include acneiform skin rash and diarrhea. For patients whose tumors respond to EGFR TKI therapy, substantial clinical benefit is seen.
Small Cell Lung Cancer
SCLC is a chemotherapy-sensitive disease. Patients with limited stage disease have high response rates (60–80%) and a 10–30% complete response rate. The response rates in patients with extensive disease are somewhat lower (50%) and almost always partial responses. Tumor regressions usually occur quickly, within the first two cycles of treatment, and provide rapid palliation of tumor-related symptoms.
Chemotherapy significantly prolongs survival. Untreated, patients with limited-stage SCLC have a median survival of 12 weeks; the median survival with chemotherapy is 18 months, and long-term (>3 year) survival is 30–40%. The median survival of extensive-stage patients is 9 months; <5% of patients survive 2 years. Thus, although initially responsive, most patients with SCLC relapse, presumably due to the emergence of chemotherapy resistance.

Chemotherapy

The chemotherapy combination most widely used for SCLC is etoposide plus cisplatin or carboplatin, given every 3 weeks on an outpatient basis for four to six cycles. Increased dose intensity of chemotherapy adds toxicity without clear survival benefit. Appropriate supportive care (antiemetics, fluid support with cisplatin, monitoring of blood counts and blood chemistries, monitoring for signs of bleeding or infection, and, as required, use of hematopoietins) and adjustment of chemotherapy doses on the basis of nadir granulocyte counts are essential.
The prognosis of patients who relapse is poor. Patients who relapse >3 months since the completion of their initial chemotherapy (so-called chemosensitive disease) have a median survival of 4–5 months; patients who do not respond to initial chemotherapy or relapse within 3 months (chemorefractory disease) have a median survival of only 2–3 months. Patients with chemosensitive disease may be retreated with their initial regimen. Topotecan has modest activity as second-line therapy, or patients can be entered onto clinical trials testing new agents.
Considerations for Therapy of SCLC Limited-Stage Disease
Combined-Modality Chemoradiotherapy
Radiation therapy to the thorax is associated with a small but significant improvement in long-term survival for patients with limited-stage SCLC (5% at 3 years). Chemotherapy given concurrently with thoracic radiation is more effective than sequential chemoradiation but is associated with significantly more esophagitis and hematologic toxicity. In one randomized study, twice-daily hyperfractionated radiation was compared with a once-daily schedule; both were administered concurrently with four cycles of cisplatin and etoposide. Survival was significantly higher with the twice-daily regimen (median survival 23 months compared with 19 months; 5-year survival 26% compared with 16%), but the twice-daily regimen gave more grade 3 esophagitis and pulmonary toxicity. Patients should be carefully selected for concurrent chemoradiation therapy based on good performance status and pulmonary reserve.
PCI significantly decreases the development of brain metastases (which occur in about two-thirds of patients who do not receive PCI) and results in a small survival benefit (~5%) in patients who have obtained a complete response to induction chemotherapy. Deficits in cognitive ability following PCI are uncommon and often difficult to sort out from effects of chemotherapy or normal aging.
Radiation Therapy for Palliation
Palliative radiation therapy is an important component of the management of SCLC patients. Cranial radiation often decreases the signs and symptoms of brain metastases. In the case of symptomatic, progressive lesions in the chest or at other critical sites, if radiotherapy has not yet been given to these areas, it may be administered in full doses (e.g., 40 Gy to the chest tumor mass).
Surgery
Although surgical resection is not routinely recommended for SCLC, occasional patients meet the usual requirements for resectability (stage I or II disease with negative mediastinal nodes). Often this histologic diagnosis is made in some patients only on review of the resected surgical specimen. However, when such SCLC patients are discovered, they should receive standard SCLC chemotherapy. Retrospective series have reported high cure rates if postoperative chemotherapy is used, although it is unclear what the outcome would be with chemoradiation therapy alone, given the relatively low bulk disease of these patients.
Lung Cancer Prevention
Deterring children from taking up smoking and helping young adults stop is likely to be the most effective lung cancer prevention. Smoking cessation programs are successful in 5–20% of volunteers; the poor efficacy is due to the addictive nature of nicotine use, which is as strong as addiction to heroin.
Chemoprevention is an experimental approach to reduce lung cancer risk; no benefit has yet been shown for chemoprevention. Two putative chemoprevention agents, vitamin E and -carotene, actually increased the risk of lung cancer in heavy smokers.
Benign Lung Neoplasms
The benign neoplasms of the lung, representing <5% of all primary tumors, include bronchial adenomas and hamartomas (90% of such lesions) and a group of very uncommon benign neoplasms (epithelial tumors such as bronchial papillomas, fibroepithelial polyps; mesenchymal tumors such as chondromas, fibromas, lipomas, hemangiomas, leiomyomas, pseudolymphomas; tumors of mixed origin such as teratomas; and other diseases such as endometriosis). The diagnostic and primary-treatment approach (surgery) is basically the same for all these neoplasms. They can present as central masses causing airway obstruction, cough, hemoptysis, and pneumonitis. The masses may or may not be visible on radiographs but are usually accessible to fiberoptic bronchoscopy. Alternatively, they can present without symptoms as SPNs and are evaluated accordingly. In all cases, the extent of surgery must be determined at operation, and a conservative procedure with appropriate reconstructions is usually performed.
Bronchial Adenomas
Bronchial adenomas (80% are central) are slow-growing endobronchial lesions; they represent 50% of all benign pulmonary neoplasms. About 80–90% are carcinoids, 10–15% are adenocystic tumors (or cylindromas), and 2–3% are mucoepidermoid tumors. Adenomas present in patients 15–60 years old (average age 45) as endobronchial lesions and are often symptomatic for several years. Patients may have a chronic cough, recurrent hemoptysis, or obstruction with atelectasis, lobar collapse, or pneumonitis and abscess formation.
Bronchial adenomas of all types, because of their endobronchial and often central location, are usually visible by fiberoptic bronchoscopy. Because they are hypervascular, they can bleed profusely after bronchoscopic biopsy, and this problem should be anticipated. Bronchial adenomas must be considered as potentially malignant, thus requiring removal for symptom relief and because they can be locally invasive or recurrent, potentially can metastasize, and may produce paraneoplastic syndromes. Surgical excision is the primary treatment for all types of bronchial adenomas. The extent of surgery is determined at operation and should be as conservative as possible. Often bronchotomy with local excision, sleeve resection, segmental resection, or lobectomy is sufficient. Five-year survival rate after surgical resection is 95%, decreasing to 70% if regional nodes are involved. The treatment of metastatic pulmonary carcinoids is unclear because they can either be indolent or behave more like SCLC (Chap. 344). Assessment of the tempo and histology of the disease in the individual patient is necessary to determine if and when chemotherapy or radiotherapy is indicated.
Carcinoid and Other Neuroendocrine Lung Tumors
Neuroendocrine lung tumors represent a spectrum of pathologic entities, including typical carcinoid, atypical carcinoid, and large cell neuroendocrine cancer, as well as SCLC. SCLC and large cell neuroendocrine cancer are high-grade neuroendocrine tumors and in general should be treated as described for SCLC. By contrast, typical carcinoid and atypical carcinoids are low- and intermediate-grade tumors with different treatment approaches and in general are resistant to chemotherapy (Chap. 44). Carcinoids, like SCLCs, may secrete other hormones, such as ACTH or AVP, and can cause paraneoplastic syndromes that resolve on resection. Uncommonly, bronchial carcinoid metastases (usually to the liver) may produce the carcinoid syndrome, with cutaneous flush, bronchoconstriction, diarrhea, and cardiac valvular lesions, which SCLC does not do. Carcinoid tumors that have an unusually aggressive histologic appearance (referred to as atypical carcinoids) metastasize in 70% of cases to regional nodes, liver, or bone, compared with only a 5% rate of metastasis for carcinoids with typical histology. Large cell neuroendocrine cancer is a high-grade NSCLC with neuroendocrine features. These tumors are characterized by histologic features similar to small cell cancer, but they are formed by larger cells. The prognosis for patients with large cell neuroendocrine cancer is significantly worse than that for patients with atypical carcinoid and classic large cell cancer. Five-year survival is 21% for patients with large cell neuroendocrine cancer, 65% for atypical carcinoid, and 90% for typical carcinoid.
Thymomas
Hamartomas
Pulmonary hamartomas have a peak incidence at age 60 and are more frequent in men than in women. Histologically, they contain normal pulmonary tissue components (smooth muscle and collagen) in a disorganized fashion. They are usually peripheral, clinically silent, and benign in their behavior. Unless the radiographic findings are pathognomonic for hamartoma, with "popcorn" calcification, the lesions usually have to be resected for diagnosis, particularly if the patient is a smoker. VATS may minimize the surgical complications.
Metastatic Pulmonary Tumors
The lung is a frequent site of metastases from primary cancers outside the lung. Usually such metastatic disease is incurable. However, two special situations should be borne in mind. The first is the development of an SPN or a mass on chest x-ray in a patient known to have an extrathoracic neoplasm. This nodule may represent a metastasis or a new primary lung cancer. Because the natural history of lung cancer is often worse than that of other primary tumors, a single pulmonary nodule in a patient with a known extrathoracic tumor is approached as though the nodule is a primary lung cancer, particularly if the patient is >35 years and a smoker. If a vigorous search for other sites of active cancer proves negative, the nodule is surgically resected. Second, in some cases multiple metastatic pulmonary nodules can be resected with curative intent. This tactic is usually recommended if, after careful staging, it is found that (1) the patient can tolerate the contemplated pulmonary resection, (2) the primary tumor has been definitively and successfully treated (disease-free for >1 year), and (3) all known metastatic disease can be encompassed by the projected pulmonary resection. Patients with uncontrolled primary tumors and other extrapulmonary metastases are not considered. Primary tumors whose pulmonary metastases have been successfully resected for cure include osteogenic and soft tissue sarcomas; colon, rectal, uterine, cervix, and corpus tumors; head and neck, breast, testis, and salivary gland cancer; melanoma; and bladder and kidney tumors. Five-year survival rates of 20–30% have been found in selected series, and dramatic results have been achieved in patients with osteogenic sarcomas, where resection of pulmonary metastases (sometimes requiring several thoracotomies) is a standard curative treatment approach.

No comments:

Post a Comment