Cancer of the Lung : Non–Small Cell Lung Cancer and Small Cell Lung Cancer


Summary of Key Points

  • Non–Small Cell Lung Cancer

  • Non–small cell lung cancer (NSCLC) constitutes 80% to 85% of new cases of lung cancer in North America.

  • The most frequent histologic types are adenocarcinoma, squamous cell carcinoma, and large cell carcinoma.

  • NSCLC is the leading cause of cancer-related death in the United States for both men and women; it is one of the most preventable forms of cancer death.

  • Eighty percent of lung cancer deaths are attributed to tobacco use.

  • Most guidelines recommend annual low-dose computed tomography (CT) scan in high-risk individuals (based on results from the National Lung Screening Trial).

  • The staging evaluation includes a history and physical examination and routine hematologic and biochemical testing.

  • Imaging studies include:

    • CT scan (with contrast) of lungs, mediastinum, liver, and adrenals

    • Positron emission tomography (PET) scan; bone scan if PET is not available

    • Magnetic resonance imaging (MRI) of brain if patient has locally advanced or metastatic disease

  • Bronchoscopy and/or CT-guided and/or ultrasound-guided biopsy should be performed.

  • If disease is clinically more advanced, use the least invasive biopsy procedure and sample to confirm advanced disease.

  • Mediastinal node evaluation is as follows: If disease is in early stage (I, II), PET scan of mediastinum is negative, and lymph nodes are not enlarged, proceed directly to surgery. If PET scan is positive, proceed to biopsy with endobronchial ultrasound (EBUS) or endoscopic ultrasound (EUS) or mediastinoscopy depending on node location.

  • Primary therapy for stage I disease:

    • Surgery is often curative.

    • The role of preoperative or postoperative chemotherapy is unclear. Chemotherapy is often recommended in patients with stage IB tumors 4 cm or lager.

    • Stereotactic body radiation therapy (SBRT) is performed in selected patients.

  • Primary therapy for stage II disease:

    • Surgery is often curative.

    • Postoperative chemotherapy prolongs survival in patients with a good performance status.

    • SBRT is performed in selected patients.

    • Postoperative radiation therapy (PORT) is recommended in selected patients.

  • Primary therapy for stage IIIA disease:

    • Surgery followed by postoperative chemotherapy is recommended in patients with a good performance status.

    • PORT is performed in selected patients.

    • The role of preoperative chemotherapy with or without radiation therapy for stage IIIA cancer is unknown except for Pancoast tumors, in which it is recommended.

    • Concurrent chemotherapy combined with radiation therapy is recommended in patients with a good performance status and N2 disease.

  • Primary therapy for stage IIIB disease:

    • Concurrent chemotherapy combined with radiation is superior to radiation alone; increased toxicity limits this approach in frail patients.

    • Clinical trials of new approaches should be a high priority.

  • Primary therapy for stage IV disease:

    • All tumors should be sent for molecular testing and PD-L1 immunohistochemistry assessment before first-line therapy decisions are made.

    • Targeted therapy with an epidermal growth factor receptor (EGFR), ALK, or ROS tyrosine kinase inhibitor (TKI) is used as first-line therapy in patients with known EGFR, ALK, or ROS mutation or fusions.

    • Pembrolizumab is used as first-line therapy in patients with tumor positive for PD-L1 in 50% or more tumor cells.

    • Primary chemotherapy with a platinum-based doublet improves the quality and quantity of life.

    • There is no advantage to triplet chemotherapy.

  • Second- or Third-Line Therapy

    • Further treatment with any of several agents is appropriate with progression after chemotherapy, including immune checkpoint inhibitors nivolumab, pembrolizumab, or atezolizumab, docetaxel with or without ramucirumab, or pemetrexed. All of these agents have been approved by the US Food and Drug Administration (FDA).

    • Local recurrence after surgery can be treated with combined chemotherapy and radiation or stereotactic radiosurgery after complete restaging.

  • Small Cell Lung Cancer

  • Small cell lung cancer (SCLC) accounts for approximately 15% of new cases of lung cancer each year in the United States. The incidence is decreasing.

  • Subtypes include small cell carcinoma and combined small cell carcinoma (most often combined with squamous cell carcinoma, adenocarcinoma, or large cell carcinoma).

  • Staging evaluation includes history and physical examination and routine histologic and biochemical testing.

  • Imaging includes:

    • CT scan (with contrast) of lungs, mediastinum, liver, and adrenals

    • PET (useful with clinically early-stage disease)

    • Brain evaluation (MRI preferred)

    • If there are signs or symptoms of bone involvement: bone scan or PET scan recommended

    • Mediastinal node evaluation not required unless as a convenient site for biopsy

  • Therapy for limited disease:

    • Concurrent etoposide plus cisplatin is administered, with radiation to intrathoracic disease.

    • Prophylactic cranial irradiation (PCI) is administered in complete (or near-complete) responders.

  • Therapy for extensive disease:

    • Etoposide (or irinotecan) plus cisplatin or carboplatin is administered.

    • There is no evidence that a third agent or dose intensification improves outcome.

    • Research studies are a clear priority, given the lack of progress in this arena.

    • PCI may be administered in complete (or near-complete) responders.

    • Consolidative radiation therapy to thoracic disease may be considered in complete (or near-complete) responders.

  • Therapy for recurrent disease:

    • Second-line therapy with a platinum agent or etoposide may be useful mainly in patents who have experienced a longer treatment-free interval and who have a good performance status.

    • Topotecan has been approved by the FDA; however, any of several chemotherapy agents such as paclitaxel or irinotecan also offer short-term benefit.

    • Research protocols are a preferred choice for patients with disease recurrence.

Although reports of pulmonary malignancies date to antiquity, lung cancer is largely a disease of modern humans. Before 1900, lung cancers were viewed as “matters of medical curiosity not known to be in any degree influenced by medicine and too rare to be of much practical importance.” By the mid-20th century after the introduction of cheap, mass-produced cigarettes, however, lung cancer had become epidemic and firmly established as the leading cause of cancer-related death in North America and Europe. It should not be forgotten that lung cancer is potentially one of the most preventable of all of the major malignancies affecting humans. Its primary cause is tobacco smoke. King James I was among the first to chronicle the adverse health effects of tobacco smoke, but it was Raymond Pearl's landmark 1938 report that conclusively established the devastating impact of smoking on longevity. It would be another decade before tobacco smoking was firmly established as a causative agent of lung cancer and nearly 30 more years before the emergence of the US Surgeon General's initial report of the ill effects of tobacco smoking. Regrettably, it would be yet another three decades before the tobacco industry publicly acknowledged this obvious truth, but only after a long, drawn-out battle of misinformation and deception ironically helped along by the (perhaps) unwitting complicity of physicians. With the belated recognition of the etiologic role of tobacco smoke, the incidence of lung cancer has started to decline in North America and parts of Europe. For the most part the decline is seen more clearly in men. Only recently has this decline become apparent in women in the United States after a similar decline in men 15 to 20 years prior. In short, the story of lung cancer is replete with controversy, politics, pessimism, and, more recently, guarded optimism. This chapter focuses on our perceptions of modern-day management of lung cancer, with an emphasis on advances made over the past decades.

Epidemiology

Lung cancer was an uncommon disease in the early part of the 20th century but then began an epidemic rise to far surpass all other cancers in numbers and rates of death. Lung cancer is also the leading cause of cancer-related mortality worldwide. The World Health Organization (WHO) reported approximately 1.8 million new cases and more than 1.5 million deaths caused by lung cancer in 2012. Lung cancer is relatively rare below age 40, but rates rise steadily until age 80 and then taper off. In the United States, the projected lifetime probability of developing lung cancer is approximately 7% among males and approximately 6% among females. The incidence and mortality of lung cancer vary by racial and ethnic group, with the highest age-adjusted incidence rates among African American men ( Fig. 69.1 ). The excess in age-adjusted rates among African Americans occurs only among men, but examinations of age-specific rates show that below age 50, mortality from lung cancer is more than 25% higher among African American compared with white women. Incidence and mortality rates among Hispanic, Native, and Asian Americans are only 40% to 50% those of whites ( Fig. 69.2 ).

Figure 69.1, Lung and bronchus cancer incidence rates by race/ethnicity and sex, United States, 1975–2013. SEER, Surveillance, Epidemiology, and End Results Program.

Figure 69.2, Age-adjusted SEER lung cancer incidence and mortality. CDC, Centers for Disease Control and Prevention; SEER, Surveillance, Epidemiology, and End Results Program.

The trends in overall lung cancer rates mask differences in temporal patterns according to lung cancer cell type. Among men, squamous cell cancers predominated in the first two-thirds of this period. The decline in incidence of lung cancer among men was first apparent for squamous cell tumors, with decreases beginning in the early 1980s so that by the mid-1990s the rates of squamous cell carcinoma among men had dropped below those for adenocarcinoma, which did not peak until over a decade later. Among women, adenocarcinomas have been more common than squamous cell carcinomas across the three decades. The adenocarcinoma excess among women has become more pronounced over time; rates of squamous cell cancers increased steadily through the 1980s before beginning to decline, whereas rates of adenocarcinoma did not plateau until about a decade later. Although not shown, rates of small cell lung cancer (SCLC), the third most frequent cell type, tended to parallel those of squamous cell cancer in both sexes. Trends for other cell types of lung cancer, including large cell carcinomas, tend to be intermediate between those of squamous cell carcinomas and adenocarcinomas. As discussed later in the chapter, although 5-year relative survival rates for lung cancer have improved over time, the survival rates are low, currently about 15% overall. Some variation exists by sex, race, cell type, and molecular status, with slightly higher survival among whites than blacks and females than males, but for no group does the overall 5-year relative survival exceed 20%.

Risk Factors

The cause of the large majority (80%) of lung cancers is cigarette smoking. There is a massive compilation of scientific evidence from epidemiologic studies conducted around the world since the 1950s demonstrating the link between smoking and lung cancer. Epidemiologic research has also revealed that several other factors have been implicated as causes of lung cancer, although none to the extent of tobacco. Cigarette smokers have been shown to have large increases in the risk of lung cancer. Deep sequencing studies have estimated that smoking a pack of cigarettes a day for a year may cause normal lung cells to accumulate on average 150 mutations. Numerous investigations typically show 10-fold or greater increases in risk of this cancer among smokers compared with those who have never smoked. One of the largest studies is the American Cancer Society's prospective cohort study of over 1 million Americans, in which an over 20-fold excess of lung cancer has been observed among men who were current smokers at the start of the follow-up in the early 1980s. In contrast, even the most prolonged and intense exposures to asbestos, perhaps the most prominent occupational cause of lung cancer, are associated with no more than about fivefold increases in risk of lung cancer.

Risks of lung cancer are lower in persons who quit smoking than in those who continue smoking. The reductions in risk indicate that quitting smoking is beneficial (and conversely that continuing to smoke is harmful). Risk among former smokers on average is less than one-half that of those who continue to smoke. In the aforementioned American Cancer Society cohort study, former smokers had a ninefold increase in lung cancer compared with men who had never smoked versus the 20-fold excess in those who continued to smoke. Such relative reductions have been consistently seen, with the size of the reduction in risk increasing the longer the person has continued to not smoke, although in general, even long-term former smokers have higher risks of lung cancer than those who never smoked.

Cigarette smoking has been shown to increase risk of all the major lung cancer cell types. The magnitude of the increase varies by histologic type, however, with highest risks for squamous cell, small cell, and large cell carcinomas of the lung. Some early studies tended to show only small increases in risk of adenocarcinoma among smokers, but more recent studies have indicated that the excess of lung adenocarcinoma is substantial.

Cigarette smoke has also been implicated in increasing risk of lung cancer among nonsmokers. A 2006 update of the Surgeon General's report declared that there is sufficient evidence to list passive smoking as an established cause of lung cancer and called for further control of environmental tobacco smoke (ETS) exposures. The risk from ETS is far less than from active smoking, with about a 20% to 30% increase in lung cancer observed among nonsmokers married for many years to smokers, in comparison with the 2000% increase among continuing active smokers. Nevertheless, cancer control activities based on the knowledge that ETS exposure may convey an increased risk of lung cancer have helped reduce exposures in public places and have also provided additional incentive for smokers to quit the habit.

Although cigarette smoking is the dominant cause of lung cancer, several other risk factors for this cancer have been identified. These include occupational exposures to asbestos and some other workplace agents, some of which have been evaluated for nearly as long as cigarette smoking. Among the occupational agents considered to be known lung carcinogens are arsenic, bischloromethyl ether, hexavalent chromium, mustard gas, nickel (as in certain nickel refining processes), and polycyclic aromatic hydrocarbons. Several other occupational exposures have been associated with increased rates of lung cancer, but the causal nature of the association is not clear. Epidemiologic studies have attempted to assess the potentially synergistic interrelationship between certain workplace exposures and smoking. Risk of lung cancer in men exposed to asbestos who also smoked was originally thought to be exceptionally high (with early reports of 50-fold or greater excesses compared with unexposed nonsmokers), but modeling of larger data pools has suggested that asbestos and tobacco combine to enhance lung cancer risk in a less than multiplicative manner. Occupational observations have also provided clues to the mechanisms of lung cancer induction. Risk of lung cancer among asbestos-exposed workers, for example, is increased primarily in those with underlying asbestosis, raising the possibility that the scarring and inflammation produced by this fibrotic nonmalignant lung disease may in many cases (though likely not in all) be the trigger for asbestos-induced lung cancer.

Increased risks of lung cancer have also been associated with other factors. Diet and nutrition are thought to be involved, because numerous investigations have shown somewhat higher risks of this cancer among those with low fruit and vegetable intake during adulthood. The early observational studies led to hypotheses that specific nutrients, in particular retinoids and carotenoids, might have chemopreventive effects for lung cancer. Randomized clinical trials were launched but failed to validate this hypothesis when reports from interventions involving supplementation with β-carotene in trials both in Finland and in the United States found increased rather than decreased incidence of lung cancer among the supplemented. The current consensus regarding diet and lung cancer remains muddled, with a minor role for nutritional factors likely, but difficult to assess epidemiologically. Ionizing radiation has been established as a lung carcinogen, most convincingly demonstrated from studies showing modestly increased rates of this cancer among persons exposed to the atomic bombs of Hiroshima and Nagasaki and large excesses among workers exposed to alpha irradiation from radon in underground uranium mining. Extrapolations from the high exposures in mines to low-level radon exposures in homes, and direct observations from case-control studies assessing measured levels in homes, have suggested that prolonged radon exposures above the recommended remedial levels might impart a risk of lung cancer equal to or greater than that of ETS. Prior lung diseases such as asbestosis (mentioned earlier), chronic bronchitis, emphysema, and tuberculosis also have been linked to increased risks of lung cancer. Although smoking itself is a cause of chronic obstructive pulmonary disease (COPD), the link between chronic bronchitis and emphysema and lung cancer persists after adjustment for smoking, with up to about a doubled smoking-adjusted cancer risk among those with COPD.

Familial clustering of lung cancer has been observed, raising the possibility of inherited traits that may increase risk among some individuals, with risk about doubled in families with prior lung cancer. Individuals with inherited mutations in retinoblastoma (RB) and p53 (Li-Fraumeni syndrome) genes may develop lung cancer. Genome-wide association studies (GWASs) have identified four genetic loci for lung cancer risk including two on 5p15 (TERT-CLPM1L), 15q25.1 (CHRNA5-CHRNA-3 nicotinic acetylcholine receptor subunits) and 6p21 (BAT3-MSH5). A rare germline mutation involving the epidermal growth factor receptor (EGFR) within exon 20 that results in an amino acid substitution at position 790 from threonine to methionine (T790M) has been linked to lung cancer susceptibility in never-smokers. Smoking also clusters within families, so some of the familial aggregation of lung cancer may be smoking related. Nevertheless, there appear to be multiple genetic factors that help determine the way in which individuals metabolize, detoxify, repair, or otherwise respond to lung carcinogens, including the carcinogens in cigarette smoke, as discussed in more detail later in this chapter.

Smoking Cessation

Given the undeniable link between cigarette smoking and lung cancer, it is incumbent on physicians to promote tobacco abstinence and help their patients who smoke to stop smoking. Smoking cessation, even well into middle age, can minimize an individual's subsequent risk of lung cancer, and stopping before middle age avoids more than 90% of the risk attributable to tobacco. By contrast, there is little health benefit realized by simply “cutting back.” Among victims of lung cancer, smoking cessation is associated with improved survival, fewer side effects from therapy, and an overall improvement in quality of life. Often forgotten is the fact that smoking alters the metabolism of many chemotherapy drugs, potentially adversely altering the toxicities and therapeutic benefits of the agents. Therefore it is important to promote smoking cessation even after the diagnosis of lung cancer has been established. Although smoking cessation is extremely difficult, patients with lung cancer tend to be highly motivated, and success rates mirror those of other disease states. However, the individual must want to stop smoking and must be willing to work hard to achieve the goal of smoking abstinence. Nicotine replacement therapies bupropion and varenicline (an α4β2 nicotinic acetylcholine receptor partial agonist) have been approved by the US Food and Drug Administration (FDA) as first-line treatments for nicotine dependence. Varenicline has been demonstrated to be significantly more efficacious than bupropion alone for smoking cessation. Furthermore, prolonged use of varenicline beyond the initial induction phase proved useful in maintaining smoking abstinence. Clonidine and nortriptyline are recommended as second-line treatments. A systematic review of extant smoking cessation studies indicated that self-help strategies alone only marginally affect quit rates, whereas individual and combined pharmacotherapies and counseling either alone or in combination can significantly increase rates of cessation.

Pathology

In recent years, significant advances in the treatment of pulmonary adenocarcinoma have affected the pathologic diagnosis of lung cancer. The WHO classification of lung tumors from 2004 was a purely morphologic classification and geared toward resection specimens. Most pathologic diagnoses of lung cancer, however, are rendered on assessment of small biopsy or cytologic specimens. A new multidisciplinary classification of lung adenocarcinoma has been proposed jointly by the International Association for the Study of Lung Cancer (IASLC), American Thoracic Society (ATS), and European Respiratory Society (ERS) ( Table 69.1 ) and is formally recognized in the current WHO classification of lung tumors ( Table 69.2 ). Although the system is based predominantly on histologic findings, its purpose is to provide an integrated clinical, molecular, radiologic, and pathologic approach to the classification of lung adenocarcinoma. In addition, this classification addresses the diagnosis of lung cancer in small biopsy and cytologic specimens and emphasizes the use of immunohistochemistry (IHC) for the subtyping of non–small cell lung cancer (NSCLC).

Table 69.1
IASLC/ATS/ERS Classification of Lung Adenocarcinoma in Resection Specimens
  • Preinvasive lesions

    • Atypical adenomatous hyperplasia

    • Adenocarcinoma in situ (≤3 cm; formerly BAC)

      • Nonmucinous

      • Mucinous

      • Mixed mucinous and nonmucinous

  • Minimally invasive adenocarcinoma (≤3 cm lepidic predominant tumor with ≤5 mm invasion)

    • Nonmucinous

    • Mucinous

    • Mixed mucinous and nonmucinous

  • Invasive adenocarcinoma

    • Lepidic predominant (formerly nonmucinous BAC pattern, with >5 mm invasion)

    • Acinar predominant

    • Papillary predominant

    • Micropapillary predominant

    • Solid predominant with mucin production

  • Variants of invasive adenocarcinoma

    • Invasive mucinous adenocarcinoma (formerly mucinous BAC)

    • Colloid

    • Fetal (low and high grade)

    • Enteric

ATS, American Thoracic Society; BAC, bronchioloalveolar carcinoma; ERS, European Respiratory Society; IASLC, International Association for the Study of Lung Cancer.

Table 69.2
2015 World Health Organization (WHO) Classification of Malignant Epithelial Tumors
  • Adenocarcinoma

    • Lepidic adenocarcinoma

    • Acinar adenocarcinoma

    • Papillary adenocarcinoma

    • Micropapillary adenocarcinoma

    • Solid adenocarcinoma

    • Invasive mucinous adenocarcinoma

      • Mixed invasive mucinous and

      • Nonmucinous adenocarcinoma

    • Colloid adenocarcinoma

    • Fetal adenocarcinoma

    • Enteric adenocarcinoma

    • Minimally invasive adenocarcinoma

      • Nonmucinous

      • Mucinous

    • Preinvasive lesions

      • Atypical adenomatous hyperplasia

      • Adenocarcinoma in situ

      • Nonmucinous

      • Mucinous

  • Squamous cell carcinoma

    • Keratinizing squamous cell carcinoma

    • Nonkeratinizing squamous cell carcinoma

    • Basaloid squamous cell carcinoma

    • Preinvasive lesion

      • Squamous cell carcinoma in situ

  • Neuroendocrine tumors

    • Small cell carcinoma

    • Combined small cell carcinoma

    • Large cell neuroendocrine carcinoma

      • Combined large cell neuroendocrine carcinoma

    • Carcinoid tumors

      • Typical carcinoid tumor

      • Atypical carcinoid tumor

    • Preinvasive lesion

      • Diffuse idiopathic pulmonary neuroendocrine cell hyperplasia

  • Large cell carcinoma

  • Adenosquamous carcinoma

  • Pleomorphic carcinoma

  • Spindle cell carcinoma

  • Giant cell carcinoma

  • Carcinosarcoma

  • Pulmonary blastoma

  • Other and unclassified carcinomas

    • Lymphoepithelioma-like carcinoma

    • NUT carcinoma

  • Salivary gland tumors

    • Mucoepidermoid carcinoma

    • Adenoid cystic carcinoma

    • Epithelial-myoepithelial carcinoma

Tissue and Cytologic Diagnosis of Lung Cancer

The diagnosis of lung cancer can be made based on tissue specimens such as endobronchial and transbronchial biopsy specimens or resected specimens, or by assessment of cytologic specimens from procedures such as transbronchial or transthoracic fine-needle aspiration (FNA), bronchial brushings and washings, or bronchoalveolar lavage or by evaluation of sputum or pleural fluid. The diagnostic yield depends on several factors including location (accessibility) of the tumor, tumor size, tumor type, and technical aspects of the diagnostic procedure including the experience level of the bronchoscopist and pathologist. In general, central lesions such as squamous cell carcinomas and SCLC, or endobronchial lesions such as carcinoid tumors, are more readily diagnosed with bronchoscopic examination whereas peripheral lesions such as adenocarcinomas and large cell carcinomas are more amenable to transthoracic FNA or biopsy. Diagnostic accuracy for most specimens in distinguishing SCLC and NSCLC is excellent, with less accuracy for subtypes of NSCLC if immunohistochemical staining is not used.

Bronchoscopic specimens are obtained through bronchial brushings, washings, bronchoalveolar lavage, and transbronchial FNA. Of these, transbronchial FNA consistently demonstrates the highest sensitivity, surpassed only by the use of a combination of bronchoscopic specimens. Overall sensitivity for combined use of bronchoscopic methods is approximately 80%, and if performed together with tissue biopsy, the yield increases to 85% to 90%. In fact, transbronchial FNA is more often diagnostic than transbronchial biopsy when the lesion of interest is submucosal.

Like transbronchial FNA specimens, transthoracic FNA specimens are also diagnostically useful, yielding diagnostic material in 70% to 95% of cases. Sensitivity is highest for larger lesions and peripheral tumors. In general, specimens obtained with FNA, whether transbronchial, transthoracic, or endoscopic ultrasound guided, are superior to other specimen types. The superiority of FNA specimens is primarily due to the higher yield of lesional tumor cells with fewer confounding factors such as obscuring inflammation and reactive nonneoplastic cells.

Cytologic assessment of sputum is inexpensive and noninvasive but has a lower yield than other specimen types owing to poor preservation of the cells and more variability in acquiring a good-quality specimen. The yield for sputum cytology is highest for larger and centrally located tumors such as squamous cell carcinoma and SCLC, although occasionally an accurate diagnosis is possible with tumors located more peripherally within the lung. The specificity for sputum cytologic assessment averages close to 100%, although sensitivity is generally less than 70%. The accuracy of sputum cytology improves with increased numbers of specimens analyzed, and consequently analysis of at least three sputum specimens is recommended. Sputum cytologic evaluation has also been extensively studied with varying success as a screening tool for early detection of lung cancers. Sputum cytologic analysis is not currently recommended as a routine screening tool, but advances in molecular diagnostic techniques may result in a resurgence in its use.

Squamous Cell Carcinoma

Squamous cell carcinoma ( Fig. 69.3 ) is typically a centrally located tumor affecting large airways. It is strongly associated with tobacco smoking. The current WHO classification recognizes two major subtypes: keratinizing and nonkeratinizing squamous cell carcinoma. Keratinizing squamous cell carcinomas show varying light microscopic evidence of keratinization (in the form of keratin pearls, individual cell keratinization, and/or intercellular bridges). Nonkeratinizing squamous cell carcinomas are poorly differentiated tumors lacking easily recognizable features of keratinization and require immunohistochemical confirmation of their squamous nature. The most common pattern is one of infiltrating nests of malignant squamous cells ( Fig. 69.3A ) with central necrosis, often resulting in cavitation. The tumor commonly affects central airways ( Fig. 69.3B ), displaying exophytic and endobronchial growth. A basaloid variant ( Fig. 69.3C ) can mimic other basaloid or neuroendocrine tumors histologically. In evaluating cytologic specimens containing squamous cell carcinoma, intercellular bridges are not usually identified, but keratin can be clearly seen when present. In addition, the tumor tends to consist of sheets of cells rather than the three-dimensional groups of cells characteristic of adenocarcinomas ( Fig. 69.3D ).

Figure 69.3, Squamous cell carcinoma. (A) Nests of invasive squamous cell carcinoma are seen within fibrous stroma. The tumor cells show abundant eosinophilic cytoplasm. (B) Cytologically (Papanicolaou stain), there are malignant squamous cells in clusters with irregular nuclear shape and nuclear chromatin, and abundant “hard” (keratinized) cytoplasm. (C) Squamous cell carcinoma is commonly associated with central airways, and images can show papillary architecture. (D) The basaloid variant of squamous cell carcinoma is composed of tumor cell nests with peripheral nuclear palisading and keratinization toward the center of the nests.

Squamous cell carcinomas of the lung are morphologically identical to extrapulmonary squamous cell carcinomas, and IHC cannot be used to distinguish primary from metastatic tumors. Separating a primary from metastatic squamous cell carcinoma requires clinical correlation. In rare cases in which the invasive tumor is clearly associated with squamous dysplasia or a squamous cell carcinoma in situ (CIS) component, a primary tumor is highly likely. The differential diagnosis of squamous cell carcinoma of the lung includes reactive processes that may result in squamous metaplasia with reactive atypia such as that observed with infection or radiation-induced injury. In these cases, clinical correlation is also essential. Poorly differentiated squamous cell carcinomas, such as basaloid squamous cell carcinoma, commonly elicits the differential diagnosis of SCLC, and use of immunohistochemical stains including neuroendocrine markers is required to separate these tumors.

Adenocarcinoma

In North America and Japan, adenocarcinoma is the most common histologic type of lung cancer. As in other lung carcinomas, adenocarcinomas occur predominantly in smokers, although nonsmokers are more likely to develop adenocarcinoma compared with other lung cancer types. Adenocarcinomas tend to occur in the periphery of the lung but can occur centrally, can be multifocal, or can fill an entire lobe. Radiographically, they are associated with solid opacities, ground-glass opacities (GGOs), or mixed patterns, generally correlating with the amount of in situ and invasive components of the tumor. The histologic diagnosis is made based on the light microscopic features of glandular differentiation, which include the presence of glands (acini), papillary and/or micropapillary structures, lepidic (bronchioloalveolar) growth pattern, and in poorly differentiated tumors with a solid pattern of growth, the presence of intracellular mucin ( Fig. 69.4A–E ). Eighty percent of adenocarcinomas demonstrate a mixture of at least two of these patterns, and many mixed adenocarcinomas (>20%) show a focal lepidic pattern. Of these patterns, the solid and micropapillary patterns in adenocarcinomas are associated with a worse prognosis. On cytologic preparations, three-dimensional groups or glandular and papillary patterns may be identified and are diagnostic of adenocarcinoma ( Fig. 69.4F ). Variants of adenocarcinoma include colloid, enteric, and fetal adenocarcinomas.

Figure 69.4, Growth patterns of adenocarcinoma. (A) Image of the acinar (glandular) pattern shows malignant glandular structures invading a fibrous stroma. (B) The lepidic pattern can be pure, as in adenocarcinoma in situ (AIS), or mixed with other patterns of adenocarcinoma. It consists of malignant epithelial cells lining alveolar walls without invasion. (C) Image of papillary adenocarcinoma shows tumor cells lining papillary structures with fibrovascular cores and loss of the underlying alveolar architecture. (D) Image of micropapillary pattern shows tumor cells forming small simple papillary structures without fibrovascular cores. (E) Image of solid pattern shows sheets or nests of polygonal cells that may contain mucin-containing cytoplasmic vacuoles. (F) Image of adenocarcinoma by cytology (Papanicolaou stain) shows three-dimensional clusters of malignant cells with vacuolated cytoplasm.

The 2004 WHO classification introduced the concept of atypical adenomatous hyperplasia (AAH) as a precursor of some adenocarcinomas. Bronchioloalveolar carcinoma (BAC) was defined as a noninvasive tumor, if the strict criteria were followed. This definition arose out of studies showing that noninvasive tumors smaller than 3 cm resulted in a 100% 5-year patient survival ; in addition, tumors with limited fibrosis, smaller size, and limited invasion had a better prognosis. Minimally invasive tumors, although not defined as BAC in the strictest sense, also carried an excellent prognosis. The IASCL/ATS/ERS classification discourages the use of the term bronchioloalveolar carcinoma and advocates use of the diagnosis adenocarcinoma in situ (AIS) for nonmucinous and rarely mucinous lesions 3 cm or smaller with purely lepidic growth (tumor cells lining alveolar walls). The term minimally invasive adenocarcinoma (MIA) is to be applied to nonmucinous and rarely mucinous lesions 3 cm or smaller with invasion limited to 5 mm or less. Lepidic predominant adenocarcinoma is a diagnosis proposed for nonmucinous lesions with foci of invasion greater than 5 mm. Most tumors previously called mucinous BAC are felt to actually represent invasive tumors and are thus reclassified as invasive mucinous adenocarcinoma in the IASLC/ATS/ERS criteria. Of note, the diagnosis of AIS or MIA cannot be rendered on small biopsy or cytologic specimens, because the entire tumor has to be examined to exclude or measure invasion.

AAH is a small preinvasive lesion (usually <5 mm) consisting of mild to moderately atypical cells lining the alveoli. AAH is an incidental lesion usually found in lung tumor resection specimens. Because these lesions (along with AIS) preserve the underlying lung architecture and alveolar spaces, they may appear as GGOs radiographically. Although when associated with malignancy it is most often associated with adenocarcinoma of the lung, AAH has also been identified in conjunction with other lung carcinomas and metastasis.

The IASLC/ATS/ERS classification recommends use of com­prehensive histologic subtyping of adenocarcinomas to make a semiquantitative assessment of the percentages of the various histologic patterns of adenocarcinoma: acinar, papillary, micropapillary, lepidic, and solid. At the time of pathologic diagnosis, tumors should be classified according to the predominant histologic subtype. If additional subtypes are present, they also should be listed. Studies using this classification have demonstrated prognostic significance in stage I cancers based on the predominant pattern. AIS and MIA resulted in 100% disease-free survival (DFS) at 5 years. Lepidic, acinar, and papillary predominant tumors had intermediate prognosis. Solid, micropapillary, and colloid predominant and invasive mucinous and mixed mucinous and nonmucinous adenocarcinomas had the worst prognosis.

The differential diagnosis of adenocarcinoma is broad and includes not only metastatic adenocarcinomas from other sites, but also tumors such as mesothelioma, which can show histologic similarities to adenocarcinoma. Clinical data including location of the tumor or tumors and history of prior malignancy are the most helpful feature in differentiating primary from metastatic tumor, but immunohistochemical stains may help further narrow the differential diagnosis when a metastatic lesion is suspected (see later discussion of IHC).

Other Non–Small Cell Carcinomas

Less common aggressive NSCLC subtypes include large cell carcinoma, pleomorphic carcinoma, spindle cell carcinoma, and giant cell carcinoma, each representing single-digit percentages of the lung carcinoma pie. Carcinosarcoma, pulmonary blastoma, lymphoepithelioma-like carcinoma, and NUT carcinoma are even rarer. Large cell carcinoma is defined as an undifferentiated NSCLC without light microscopic and immunohistochemical evidence of squamous or glandular differentiation after examination of the entire tumor at resection ( Fig. 69.5 ). Histologically, large cell carcinoma shows sheets of large malignant epithelioid cells with abundant cytoplasm and vesicular nuclei with prominent nucleoli. There are frequent mitoses and tumor necrosis. In the search for glandular or squamous differentiation, the diagnosis of large cell carcinoma requires examination of the entire tumor mass, so it cannot be rendered on small biopsy specimens or cytologic preparations. Some tumors histologically classified as large cell carcinoma may show evidence of glandular, squamous, or neuroendocrine differentiation with electron microscopy. Furthermore, because IHC is now routinely used to highlight areas of glandular and squamous differentiation (see later discussion of IHC), more tumors are classified as either squamous cell carcinoma or adenocarcinoma and thus the diagnosis of large cell carcinoma is becoming less common. Pleomorphic carcinoma is a poorly differentiated NSCLC that shows at least 10% of spindle cell and/or giant cell carcinoma components in combination with adenocarcinoma or squamous cell carcinoma or undifferentiated NSCLC components ( Fig. 69.6 ). Spindle cell carcinomas and giant cell carcinomas are composed of only respective single cell–type components. The tumors carry a poor prognosis even in early-stage disease. Because of the aggressive nature, distant metastasis, including to unusual sites, are common at presentation. Approximately 40% to 60% of the tumors show expression of the site-specific markers, which can be helpful in separating these tumors from metastasis.

Figure 69.5, Large cell carcinoma consists of sheets of undifferentiated cells without glandular or squamous differentiation. Prominent nucleoli are shown.

Figure 69.6, Pleomorphic carcinoma. (A) In this example, the tumor is composed of adenocarcinoma and spindle cell carcinoma components. (B) Image of the spindle cell carcinoma component shows cytologically malignant tumor cells arranged in fascicles.

Neuroendocrine Tumors of the Lung

Neuroendocrine tumors of the lung include a spectrum of lesions from the indolent to the most aggressive pulmonary neoplasms. The four main neuroendocrine tumors of the lung are typical carcinoid (TC), atypical carcinoid (AC), large cell neuroendocrine carcinoma (LCNEC), and SCLC ( Fig. 69.7 ). The main distinguishing histopathologic features of these tumors include the presence of neuroendocrine morphologic features (peripheral nuclear palisading, nuclear rosettes, and trabeculation) in combination with immunohistochemical evidence of neuroendocrine differentiation (expression of neuroendocrine markers). In general, it is helpful to think of these tumors according to their clinical characteristics. Carcinoid tumors tend to occur in younger nonsmokers and are less aggressive, whereas SCLC and LCNEC occur in smokers and behave aggressively. Separation of these tumors relies on cytologic features and histologic evaluation of proliferative activity (mitotic count and presence or absence of necrosis). The WHO classification emphasizes mitotic count in defining and separating these tumors ( Table 69.3 ). Although the use of IHC for Ki-67 (MIB1)—a cell proliferation marker—is not formally recognized in the WHO classification, its usefulness in separating carcinoid tumors from SCLC and LCNEC has been documented in limited biopsy material. The rate of Ki-67 staining is usually less than 2% for TC, less than 20% (and usually approximately 10%) for AC, and greater than 25% (commonly greater than 50%) for SCLC and LCNEC. In addition to assessment for the distinct histologic appearance, IHC may also be performed to verify the neuroendocrine nature of a tumor and thus differentiate neuroendocrine tumors from other NSCLC subtypes.

Figure 69.7, Neuroendocrine tumors of the lung. (A) A typical carcinoid tumor has an organoid (nested) and trabecular pattern of growth. (B) An atypical carcinoid tumor has tumor nests with peripheral nuclear palisading and focal tumor necrosis. (C) Image of a large cell neuroendocrine carcinoma displays tumor nests with peripheral nuclear palisading and nuclear rosettes. Tumor cell nucleoli and frequent mitoses are readily identified. (D) Image of a small cell carcinoma shows hyperchromatic nuclei with inconspicuous nucleoli, scant cytoplasm, and frequent mitoses.

Table 69.3
General Histologic Features of Neuroendocrine Tumors
Diagnosis Morphologic Features Mitotic Count Per Square Millimeter
Typical carcinoid tumor Neuroendocrine morphologic features, minimal cytologic atypia <2
Atypical carcinoid tumor Neuroendocrine morphologic features with or without focal necrosis 2–10
Large cell neuroendocrine carcinoma High-grade neuroendocrine carcinoma—larger cells with prominent nucleoli >10
Small cell carcinoma High-grade neuroendocrine carcinoma—smaller cells with inconspicuous nucleoli >10

Large Cell Neuroendocrine Carcinoma

LCNEC accounts for approximately 3% of lung cancers. LCNEC is a high-grade carcinoma that shows neuroendocrine architectural features (formation of rosettes, trabeculae, organoid nests, or perilobular palisading patterns), more than 10 mitoses per 2 mm 2 , and expression of neuroendocrine immunohistochemical markers. LCNEC can be difficult to diagnose cytologically and to differentiate from SCLC. Features seen in cytologic specimens include evidence of neuroendocrine differentiation, such as sheets or groups of cells with peripheral nuclear palisading or rosette formation, as seen in other neuroendocrine tumors. Unlike SCLC, these tumors have larger cells with vesicular nuclei and prominent nucleoli. The prognosis for these tumors is intermediate between those of other SCLCs and NSCLCs.

Small Cell Carcinoma

SCLC is a poorly differentiated neuroendocrine tumor that tends to occur centrally and has strong association with cigarette smoking. Incidence rates of SCLC are higher in men than in women, but a higher percentage of lung cancers are of SCLC type in women than in men. SCLC consists of cytologically malignant epithelioid cells with scant cytoplasm, nuclei with granular (“salt and pepper”) chromatin without prominent nucleoli, and more than 10 mitoses per 2 mm 2 (usually more than 50 mitoses per 2 mm 2 ). The tumor cells are arranged in sheets, but also present are the rosettes, trabeculae, or peripheral palisading of cells along the edges of nests. In small biopsy specimens, crush artifact with smearing of nuclear material and nuclear “molding” is commonly observed. The tumor cells display scant cytoplasm and thus appear small—up to two to three times the size of a resting lymphocyte; however, variation in size can be seen, and it is the nuclear features that allow diagnosis, not strictly cell size. The characteristic features are usually easily identified in well-preserved cytologic specimens but may be more difficult to differentiate from other poorly differentiated tumors at liquid-based cytologic assessment or with less well-preserved specimens or small biopsy specimens ( Fig. 69.8 ). As with other histologic types of lung carcinoma, SCLC may occur in combination with NSCLC components. Combined SCLC and LCNEC, combined SCLC and adenocarcinoma, and combined SCLC and squamous cell carcinoma have all been well documented. The differential diagnosis of SCLC includes poorly differentiated NSCLC, especially poorly differentiated squamous cell carcinoma, and neuroendocrine carcinomas, in addition to nonepithelial malignancies such as lymphoma, and some rare sarcomas (e.g., synovial sarcoma).

Figure 69.8, Small cell carcinoma. At cytologic evaluation (hematoxylin-eosin stain), the nuclear features (“salt-and-pepper” chromatin pattern), scant cytoplasm, and crushing of cells are evident.

Typical Carcinoid Tumor

TC is a low-grade neuroendocrine tumor that is characterized by low mitotic activity (fewer than two mitoses per 2 mm 2 ) and absence of necrosis. TC is composed of bland uniform cells with minimal cytologic atypia. TC occurs in nonsmokers, most often as an endobronchial lesion, but can occur at the lung periphery. Cytologic specimens contain uniform bland cells with finely stippled chromatin. Although these tumors are known for their excellent prognosis, up to 10% to 15% of patients can have metastases at diagnosis. A common differential diagnosis includes carcinoid tumorlet, which represent a morphologic continuum between two lesions. Carcinoid tumorlet is defined as a neuroendocrine cell proliferation measuring less than 5 mm. Although tumorlets are usually an incidental finding, they can be multiple, associated with carcinoid tumors or neuroendocrine cell hyperplasia.

Atypical Carcinoid Tumor

AC is an intermediate-grade neuroendocrine tumor with morphologic features similar to those of TC. It is defined by the presence of mitoses in the range of 2 to 10 per 2 mm 2 or the presence of necrosis. It has a higher risk of metastasis (up to 50% lymph node metastasis at presentation) than TC. Cytologic preparations of TC and AC tumors show similar findings in cytologic preparations, although AC may show more atypia and nuclear enlargement.

Immunohistochemistry of Lung Tumors

With the advent of different therapies for adenocarcinoma and squamous cell carcinoma, it has become important to separate the two on small biopsy and cytologic specimens. When the light microscopic features characteristic of these tumors are lacking, their further subclassification relies on immunohistochemical studies. Expression of TTF-1 (thyroid transcription factor 1, a protein that regulates transcription of genes specific for the thyroid, lung, and diencephalon) and/or napsin A (a member of the aspartic protease family, expressed in normal lung and kidney) ( Fig. 69.9 ) in poorly differentiated NSCLC favors a diagnosis of adenocarcinoma. Expression of high-molecular-weight cytokeratins, such as CK5/6, and/or p63 (a member of the p53 family of transcription factors), and/or p40 (more specific isoform of p63) in poorly differentiated NSCLC favors a diagnosis of squamous cell carcinoma. A percentage of poorly differentiated NSCLC may not show clear separation at immunohistochemical evaluation and will still fall into the category of NSCLC, not otherwise specified.

Figure 69.9, Immunohistochemistry (IHC) for subtyping non–small cell carcinoma. (A) Poorly differentiated carcinoma, hematoxylin-eosin stain. (B) IHC for thyroid transcription factor 1 (TTF-1) stains the nuclei of tumor cells. (C) IHC for napsin A stains the cytoplasm of tumor cells. (D) IHC for p63 shows only focal weak staining of nuclei of tumor cells. This staining pattern is consistent with adenocarcinoma (TTF-1+, napsin A+, p63− [or weak]).

IHC may also be used to confirm neuroendocrine differentiation within a tumor. Neuroendocrine markers include CD56 or neural cell adhesion molecule (NCAM), synaptophysin, and chromogranin A. Neuron specific enolase (NSE) shows significant nonspecific staining of nonneuroendocrine tumors and should not be used. Most often a combination of these stains (e.g., CD56, synaptophysin, and chromogranin A) is used to establish a diagnosis. These markers support neuroendocrine differentiation, but do not distinguish between specific types of neuroendocrine tumors.

IHC is also helpful in distinguishing primary lung tumors from malignancies metastatic to the lung. This is especially true for separating primary lung from metastatic adenocarcinomas. TTF-1 is expressed in over 70% of pulmonary adenocarcinomas. When positive, TTF-1 is a reliable indicator of a lung primary, provided a thyroid origin has been excluded. A negative TTF-1 finding, however, does not exclude the possibility of a lung primary. Important to note, TTF-1 has been recognized to lose its site specificity in high-grade neuroendocrine tumors because its expression has been documented in SCLC of extrapulmonary origin. Cytokeratins 7 and 20 used in combination also assist in categorizing certain tumors. These stains are not specific for a particular site of origin but can narrow the differential diagnosis based on their pattern of expression.

Because there is significant morphologic overlap between lung carcinomas and mesothelioma, IHC is required in diagnosis of these tumors. Immunohistochemical markers for CK5/6, calretinin, and Wilms tumor gene 1 (WT1) are among most useful in supporting the diagnosis of mesothelioma; however, because none of these display 100% sensitivity, they are supplemented by so-called adenocarcinoma markers such as BerEP4, MOC31, and carcinoma embryonic antigen (CEA) and used as a panel. For a reliable diagnosis of mesothelioma, it is recommended that expression of at least two “mesothelial” markers be shown.

A summary of commonly used immunohistochemical stains is shown in Table 69.4 .

Table 69.4
Common Immunohistochemical Markers Used in the Diagnosis of Lung Tumors
Diagnosis Positive Immunohistochemical Markers
Squamous cell carcinoma Cytokeratin (CK) cocktail (e.g., AE1/AE3)
p63
p40
CK5/6
CK7 in up to 30%
Adenocarcinoma including adenocarcinoma in situ or minimally invasive adenocarcinoma, nonmucinous CK cocktail
CK7
TTF-1
NapsinA
In situ and invasive mucinous adenocarcinoma CK cocktail
CK7
CK20
cdx-2
TTF-1 rare
Large cell neuroendocrine carcinoma CK cocktail
TTF-1
CD56
Chromogranin A
Synaptophysin
Carcinoid tumor CK cocktail
TTF-1 (weaker than in high-grade neuroendocrine tumors)
CD56
Chromogranin A
Synaptophysin
Atypical carcinoid tumor CK cocktail (tends to be patchy)
TTF-1 (weaker than in high-grade neuroendocrine tumors)
CD56
Chromogranin A
Synaptophysin
Common differential diagnoses
  • Colonic adenocarcinoma

CK20+/CK7−
  • Lung, breast, pancreatobiliary, upper gastrointestinal adenocarcinoma

CK7+/CK20−
  • Urothelial carcinoma

CK7+/CK20+
  • Prostatic adenocarcinoma

CK7−/CK20−
  • Mesothelioma

Calretinin, WT1, CK5/6
  • Malignant melanoma

S-100, HMB-45, Melan-A

Biology of Lung Cancer

The specific events that trigger malignant transformation of bronchoepithelial cells are unknown in the vast majority of cases. However, it is clear that exposure to environmental carcinogens, such as those found in tobacco smoke or asbestos fibers, induce or facilitate the transformation (extrinsic component). The contribution of the extrinsic carcinogen on transformation is modulated by genetic variations in genes (intrinsic component) that affect aspects of carcinogen metabolism, such as the conversion of procarcinogens to carcinogens and their subsequent inactivation. These genetic variations occur at relatively high frequency in the population. Their contribution to an individual's lung cancer risk is generally low, but because of their population frequency, their overall impact on lung cancer risk could be high. Epidemiologic studies have further suggested that a familial predisposition to lung cancer exists that is independent of tobacco smoke exposure. One study found evidence for an autosomal dominant model linked to 6q23–25, but other studies have proposed a complex multigene model for inherited risk. As mentioned earlier, familial clustering of lung cancer cases has been reported, with an inherited T790M mutation in the EGFR gene. The identification of individuals at particularly high risk for the development of lung cancer could justify more intense screening regimens, and the identification of the responsible chromosomal loci for lung cancer susceptibility genes could allow the development of specific chemopreventive strategies.

Environmental factors, as modified by inherited modulators, likely affect specific genes by deregulating important pathways to enable the cancer phenotype. Particularly important in lung cancer are acquired abnormalities in RAS, RB, TP53, AKT, LKB1, and BRAF (reviewed by Fong and colleagues ). However, the most clinically significant acquired genetic abnormalities in lung cancer are mutation of EGFR, the anaplastic lymphoma kinase (ALK) fusion, and the ROS1 fusion. EGFR mutations occur in approximately 10% of lung cancers in the United States and 35% of cancers in Asia. They are found primarily in exons 19 (in-frame deletions) and 20 (L858R point mutants), result in constitutive signaling and AKT activation, and are associated with very high response rates (response rate; 60% to 90%) to the specific tyrosine kinase inhibitors (TKIs) gefitinib and erlotinib. ALK and ROS1 fusions have been reported in 3% to 7% and 2% of lung cancer patients, respectively, and are associated with high response rate to crizotinib. The EGFR, ALK, and ROS1 abnormalities are nonoverlapping. Interesting to note, EGFR mutations and ALK and ROS1 fusions are associated with younger age, light (<10 pack-year) smoking history and never-smokers, and adenocarcinoma histologic type. Response to specific EGFR, ALK, and ROS1 inhibitors are often dramatic and occur even in heavily pretreated patients, demonstrating that tumors with these mutations are “addicted” to the activation of this pathway. Almost all patients with these dramatic responses, however, develop progressive, resistant disease. The most common mechanism of acquired resistance to EGFR TKIs is a second site mutation within exon 20 that results in an amino acid substitution at position 790 in EGFR, from threonine to methionine (T790M). The T790M mutation is associated with approximately 1% of primary resistance but over 50% of acquired resistance to EGFR TKI treatment. To date, several second site mutations within the ALK tyrosine kinase domain have been reported as mechanisms of acquired resistance to the ALK TKI crizotinib.

In addition to mutations in EGFR, KRAS, ALK, and ROS1, other “driver” mutations have been discovered, such as mutations in BRAF, PIK3CA, NRAS, AKT1, MET, NRAS, and MEK1 (MAP2K1) and NTRK fusions ( Fig. 69.10 ). Each of these mutations occurs in less than 1% to 3% of lung adenocarcinomas. The great majority of the driver mutations are mutually exclusive, and there are ongoing clinical studies for their specific inhibitors. Most of these mutations are present in adenocarcinoma; however, mutations that may be linked to future targeted therapies in squamous cell carcinomas are emerging. These include mutations in FGFR1, DDR2, and PIK3CA. As more comprehensive data are generated in projects such as The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC), new discoveries will be made that may further advance our knowledge of lung cancer biology and improve treatment results.

Figure 69.10, Driver mutations identified in lung adenocarcinomas.

The most commonly used method to detect mutation of EGFR is gene sequencing, although ALK and ROS1 fusions are routinely tested with fluorescence in situ hybridization (FISH) or IHC. In the past years, though, some institutions moved the testing platform to multiplex techniques such as SNaPshot, Sequenom, and next-generation sequencing (NGS). NGS allows identification of mutations, copy number alterations, and rearrangements in hundreds to thousands of genes simultaneously, at decreasing cost and timeframe. Other advances in molecular biology include the availability of different techniques of liquid biopsy, which have been quickly incorporated into NSCLC care. The most frequent use is in patients whose cancer has progressed on a first-line EGFR TKI, in order to assess for a resistance mutation (T790M). Patients whose tumor harbors T790M may benefit from third-generation TKIs. Liquid biopsy is also useful for up-front molecular screening in patients whose biopsy specimen is insufficient, and in whom a repeat biopsy is undesirable.

It is clear that the vast majority of lung cancers are not driven by single aberrant genes, however, and not all aberrancies are mutations. Complex networks of genes are finely tuned in normal cells to maintain normal growth, apoptotic responses, and differentiation. These networks can be perturbed at multiple points to deregulate key pathways in lung cancer tumors. A simple example is the mutation of Rb and the loss of p16, a regulator of Rb function; NSCLCs disrupt cell cycle control by either mutation of Rb or loss of expression of p16, but not both ( Table 69.5 ).

Table 69.5
Examples of Common Molecular Alterations in Lung Tumors
Diagnosis Common Molecular Alterations
Squamous preneoplasia LOH—3p, 9p21, 8p21–23, aneuploidy, methylation
Atypical adenomatous hyperplasia LOH—3p, 9p, aneuploidy
K-ras codon 12 mutation
Adenocarcinoma p53 mutation
p16 mutation/inactivation
K-Ras (42%); smokers more common
EGFR overexpression (40%)
EGFR mutation
Her2/neu, COX-2 overexpression
Squamous cell carcinoma p53 mutation
p16 inactivation
Allellic loss 3p
EGFR overexpression (80%)
Large cell carcinoma K-Ras, p53, loss p16
Large cell neuroendocrine carcinoma p53
bcl-2 overexpression
Rb mutation
3p21, FHIT, 3p22–24, 5q21,9p21
Small cell carcinoma Rb mutation (80+%)
p53 mutation 50%–80%
BCL-2 expression
3p21, FHIT, 3p22–24, 5q21,9p21
COX-2, Cyclooxygenase 2; EGFR, epidermal growth factor receptor; LOH, loss of heterozygosity.

The histologic sequence of events that leads to the various forms of lung cancer is not well understood, and it is clearly different for the various histopathologic entities. Current knowledge suggests that squamous cell carcinoma arises in a relatively ordered progression that includes squamous metaplasia and CIS. Peripheral adenocarcinomas are thought to arise from atypical adenomatous hyperplastic lesions, but this process is much more obscure, largely because this type of lesion is much less accessible with bronchoscopy. SCLC might arise from neuroendocrine hyperplasia, but evidence in support of this hypothesis is scarce.

The molecular biology of premalignancy is of great clinical importance, not only to better understand the process of cancer development, but also to provide potential therapeutic targets for intervention in this process and intermediate biomarkers for assessment of risk and evaluation of candidate chemoprevention strategies. Premalignant lesions of the lung have been investigated for molecular alterations in NSCLC, but much less information is available for SCLC. Microdissected specimens derived from normal, hyperplastic, metaplastic, dysplastic epithelium, and CIS, as well as invasive neoplastic foci of patients with lung cancer, were studied for gene mutations, promotor hypermethylation, and allele loss. Results obtained to date suggest that allele loss on chromosome 3p is the earliest event, followed by allele loss or hypermethylation on chromosome 9p and subsequently on chromosome 8p. Loss of heterozygosity (LOH) at the p53 gene locus (17q13.1) is relatively rare (10%) and occurs predominantly at the dysplasia or CIS stage. Point mutations in the p53 gene and the EGFR gene, however, have been observed in morphologically normal bronchial epithelium obtained from the airways of patients with lung cancer. In contrast, K-ras gene mutations represent a late event, found only in CIS or invasive cancers. In addition to copy number changes and mutations, alterations in the expression of retinoic acid receptor beta (RARβ) have also been used as a molecular marker for premalignancy and as an intermediate biomarker for chemoprevention trials.

The fate of morphologically or molecularly abnormal areas is currently an intense area of research. One study showed that 54% of patients with high-grade dysplastic lesions developed lung cancer within 2 years, but 80% of those arose in a different part of the lung than the CIS. None of the patients with low-grade dysplastic lesions progressed to develop cancer in this study. Of the low-grade lesions in this study, 82% spontaneously regressed, 18% remained unchanged, and none progressed to CIS. Preliminary data also suggested that abnormal regions display reproducible molecular changes at repeat biopsies over time, and that these and additional abnormalities can be observed in tumors arising from these lesions.

Part of the problem with the studies of preneoplasia is that we may be evaluating the microscopic appearance and molecular characteristics of the wrong population of cells. The vast majority of the respiratory epithelium (and most other tissues) is thought to be terminally differentiated and incapable of sustained replication; however, one theory holds that a small subset of the cells have unlimited, but normally tightly regulated, replicative potential. These are the so-called pulmonary “stem cells,” whose biology is very poorly understood. The stem cell concept may also underlie the failure of standard medical therapies to eradicate lung cancers, even when there is a clinical complete response (CR). The theory is that therapies have been refined to induce measurable reductions in the bulk tumor mass, but true replicative potential exists only in a small subset of “cancer stem cells” that are not effectively targeted by current therapies. Specific isolation of these cells and development of stem cell–targeted therapies may therefore not cause rapid tumor regressions, but rather result in significantly increased long-term survival.

Early Detection and Screening

Early detection is a process that involves screening tests, surveillance, and diagnosis and also implies early treatment, whereas screening is defined as the systematic testing of asymptomatic individuals for preclinical disease. The purpose of screening is to prevent or delay the development of advanced disease in patients with preclinical disease through early detection and treatment. Screening presumes that a test or series of tests will identify asymptomatic persons at risk for a specific disease and that a positive result will lead to further testing to definitively establish the presence or absence of disease. The monitoring of these subjects (surveillance) intends to detect the disease early and to treat it early. Under ideal circumstances, early intervention should change the course of the disease once the diagnosis has been established, resulting in a decrease in disease-related mortality (the number of disease-specific deaths relative to the total number of persons evaluated). In addition, screening should apply to large populations that would benefit from early detection (therefore with chance of survival for longer than 5 years). Finally, screening should cause no harm and should be cost effective.

Until recently, lung cancer screening efforts using periodic chest radiographs coupled with regular cytologic assessment of sputum failed to demonstrate a decrease in lung cancer–related mortality. Details of these trials have been extensively reviewed elsewhere. Ironically, more lung cancers were diagnosed in the screening arms of these trials. The lung cancers identified in the screened population were often found at early stages, allowing more patients in the screened arms to go on to definitive surgery. Nonetheless, there was not an improvement in lung cancer–related mortality, which is considered a requirement to validate potential screening methods. Several hypotheses have been advanced to explain the findings, including flawed trial design, lead-time bias, length-time bias, and overdiagnosis bias. Briefly, these biases can be defined as follows. Lead-time bias implies that earlier detection could result in longer survival from the time of diagnosis even if death is not delayed. Length-time bias occurs when screening examination detects slow-growing cancers. In other words, the slower the growth of the neoplasm, the longer it is present without symptoms and the greater the likelihood of detection. Overdiagnosis bias refers to the phenomenon of detecting a lung cancer that would otherwise have remained subclinical before death occurred from other causes. Overdiagnosis is of some concern in lung cancer screening because newer screening modalities can reveal small nodules of unknown clinical significance. Mayo Clinic investigators found a persistence of excess lung cancer cases in the intervention arm of their original screening after an additional 16 years of follow-up, providing support for some overdiagnosis in lung cancer screening. In recent years the field has advanced dramatically. The progress is summarized in the following sections, and so are the many challenges it brings.

High-Risk Population: A Susceptible Subgroup

As noted, a challenging problem in screening for lung cancer is the definition of a “high-risk” population—the population that could best benefit from lung cancer screening. Individuals “at risk” include current and former smokers, those with specific various occupational exposures (e.g., asbestos; see section on epidemiology), the presence of airflow obstruction, a family history of lung cancer, older individuals, and those with a prior history of a cancer of the aerodigestive tract. Individuals who smoke expose their entire aerodigestive tract to multiple carcinogens, and therefore it is not surprising that this population experiences a high rate of second primary tumors, estimated at 1% to 4% per patient per year. However, mere recognition of these features is not sufficient to identify high-risk individuals. For example, although smoking is an obvious risk factor for lung cancer—the cumulative risk of dying from lung cancer for a lifelong smoker is estimated to be approximately 16% in men and approximately 10% in women —the risk of developing lung cancer varies greatly among individual current and former smokers. For example, in the Carotene and Retinol Efficacy Trial (CARET), a large, randomized trial of lung cancer prevention, the 10-year cancer risk among current and former smokers ranged from less than 1% to 15%. An example of an individual with a relatively low risk of developing lung cancer would be a 51-year-old woman who smoked one pack per day for 28 years and quit 9 years ago. By contrast, a 68-year-old man who smoked two packs per day for 50 years and continued to smoke might have a 15% risk of lung cancer. Both of these individuals would be potentially eligible for screening, and yet the payoff could be quite different. This suggests that an accurate risk prediction model may greatly facilitate the admission of subjects in screening (or chemoprevention) trials. Many attempts to improve these prediction models have been made—for example, with use of demographic and clinical and now molecular biomarkers such as DNA SEX6L polymorphism or DNA sputum cytometric analysis or imaging biomarkers such as the presence of a nodule or emphysema at baseline.

Relative to smokers with normal lung function, smokers with COPD have a fourfold to sixfold elevated risk of lung cancer, making COPD the greatest clinical risk factor in ever-smokers. Low-dose chest computed tomography (LDCT) screens provide an in vivo assessment of both biologically relevant features of indeterminate nodules and the extent of injury reflected by emphysema. The integration of biologic and imaging-based biomarkers to individualize lung cancer risk not only enables identification of individuals most likely to benefit from screening, and perhaps more intensive screening, but also could be used to identify individual smokers at great risk of developing cancer and further convince them to stop smoking, which is the first and most effective step in the prevention of lung cancer and all other smoking-related diseases.

The presence of cytologic atypia in sputum samples reflects the abnormalities that develop in the bronchial epithelium. Moreover, the presence of cytologic atypia has been shown to predict lung cancer risk. The presence of moderate dysplasia or worse cytologic atypia was associated with an increased risk of developing lung cancer in a cohort of heavy smokers with airflow obstruction (adjusted hazard ratio [HR] of 2.8). This translates into a cumulative lung cancer incidence of 10% at 3 years and 20% at 6 years. Given the limitations of cytologic evaluation of sputum samples, however, the study of molecular abnormalities in the sputum (e.g., methylation patterns of specific genes involved in lung cancer progression and cytogenetic alterations) may strengthen the assessment of risk for lung cancer in this population.

Molecular epidemiology may be used to identify patients at risk for developing lung cancer through the identification of specific genes, single-nucleotide polymorphisms, or other genetic traits associated with increased susceptibility to lung cancer. Although many genotypes show increased risk (relatively low odds ratios [ORs]) with lung cancer (e.g., CYP2A6, thymidylate synthase, GSTM1, XPA), their large number and low penetrance make targeted interventions extremely challenging. Assessing genetic susceptibility to lung cancer may allow the identification of susceptible subgroups most likely to represent ideal candidates for early detection. With the exception of the rare EGFR T790M polymorphism, autosomal dominant genes have not been found in association with family history of lung cancer; there is epidemiologic evidence demonstrating a 2.5-times increased risk in any patient with a family history of lung cancer (two affected first-degree relatives) after smoking was controlled for. A region on chromosome 6q23–25 has been identified as a locus of susceptibility with a maximum heterogeneity logarithm of the odds (LOD) score of 2.79, 3.47, and 4.26 for families with three, four, or five or more affected individuals. Further complicating the picture is the role of the environment in modifying these specific genes, the interaction among genes, and evidence to support a genetic predisposition to smoking addiction. If markers of these genetic predispositions can be firmly established, intensive intervention to alter risk factors in these selected populations may alter the clinical outcome.

Imaging Approach

The last decade has witnessed a dramatic improvement in technology allowing for faster, higher-resolution imaging of the chest with reduced radiation exposure. The CT scanner first became available in the 1970s but was impractical for screening owing to its slow speed and high radiation dose. In the mid-1990s, low-dose scanners capable of imaging the chest in less than 15 seconds with radiation doses equivalent to 10 radiographs became available, opening the door to their potential use for screening. Moreover, new bronchoscopic methods also demonstrate promise for the detection of preinvasive lesions. Other imaging modalities such as positron emission tomography (PET) are able to provide metabolic information on lesions and can be combined with CT scan to give detailed resolution.

Low-Dose Spiral Computed Tomography Scan

In recent years there has been a substantial increase in the use of spiral computed tomography (CT) scans to screen for lung cancers in former and current smokers. Spiral CT screening allows for a rapid and comprehensive evaluation of the lungs and is attractive because of its potentially increased sensitivity, low radiation exposure, and potential cost-effectiveness. Multiple pilot studies using spiral CT scans to screen for lung cancers have shown that this technique can identify a higher percentage of early-stage lung cancer compared with conventional imaging studies. These results are summarized in Table 69.6 and discussed in detail elsewhere. Lung cancer prevalence rates range from 0.4% to 2.7%, depending on the population screened. In general, these prevalence rates are significantly higher than that reported with conventional imaging studies. The mean diameter of screened detected cancers ranges from 14 to 21 mm. Incidence rates based on detection of new malignancies at annual repeat screening range from 0.07% to 1.1%. Notably, up to 85% of CT screen–detected lung cancers are clinical stage I lesions. By contrast, only 15% of lung cancers diagnosed through routine clinical care are found to be stage I. Because stage I lung cancer is the most curable form of this disease, a high frequency of detection of stage I tumors is considered a necessary (although not sufficient) indication of a favorable screening outcome.

Table 69.6
Results of Prevalence and Incidence Screens in Computed Tomography (CT) Screening Trials
Study Year Patients Screened No. of Abnormal CT Scans Cancers Detected Stage I Lesions
PREVALENCE CT
Henschke 1999 1000 233 27 (2.7%) 81%
Sone 2001 5483 676 22 (0.4%) 100%
Swensen 2002 1520 782 22 (1.4%) 59%
Sobue 2002 1611 186 14 (0.9%) 77%
Pastorino 2003 1035 199 11 (1.1%) 55%
Henschke 2006 31567 4186 410 (1.3%) 85%
Aberle 2011 26309 7191 270 (1.0%) 63%
INCIDENCE CT
Henschke 2001 1184 63 7 (0.6%) 85%
Sone 2001 8303 518 34 (0.4%) 100%
Swensen 2002 1464 191 3 (0.2%) 0%
Sobue 2002 7891 721 22 (0.3%) 82%
Pastorino 2003 996 99 11 (1.1%) 100%
Henschke 2006 27456 1460 74 (0.3%) 86%
Berg 2011 48817 10955 379 (0.8%) 63%

The Early Lung Cancer Action Program (ELCAP) was a large lung cancer screening trial started in the 1990s in which chest CT imaging was used. It showed an improved detection rate and survival of patients with early-stage lung cancers. The International Early Lung Cancer Action Program (IELCAP) investigators reported a 92% 10-year survival rate among patients with screen-detected clinical stage I lung cancer who underwent surgical resection within 1 month after diagnosis. In a separate report, the IELCAP investigators noted that the cancers identified with CT screening met standard criteria for full-fledged aggressive lung cancer. Although these data are intriguing and encouraging, long-term follow-up of these patients will be very important to exclude lead-time bias (simply diagnosing the cancer earlier, but not altering its outcome) as a confounder for improved survival and reduced disease-related mortality.

These results prompted the design of the large randomized National Lung Screening Trial (NLST). The NLST is a prospective comparison of spiral CT and standard chest radiography in 50,000 current smokers or ex-smokers between the ages of 55 and 74 years and is designed to determine the best strategy to reduce lung cancer–related mortality. A similar study is ongoing in the Netherlands and Belgium (the NELSON trial) comparing CT scanning with standard of care in 20,000 participants with a history of heavy smoking. Exciting results from the NLST showed a 20% reduction in lung cancer–specific mortality when LDCT screening was used for patients at high risk for lung cancer after a median follow-up of 6.5 years, compared with chest radiography. This is the first large randomized screening study of lung cancer with low-dose chest CT to show an improvement in overall survival (OS), thus giving new hope that broader application of this technology will result in a significant reduction in the death rate for this cancer. The results of this trial were further strengthened by the results of the PLCO study, which found that annual screening with chest radiography did not reduce lung cancer mortality compared with usual care. However, all screening effort comes at some cost. In the NLST, only 4% of the nodules detected ended up being malignant (a 96% false-positive rate). A total of 24% of the patients in whom clinical suspicion for cancer was high enough for a diagnostic surgical procedure to be performed (mediastinoscopy, thoracoscopy, or thoracotomy) had benign disease. As LDCT screening for lung cancer evolves, better predictive models that incorporate biomarkers are needed to determine which patients have lung cancer. New imaging and molecular biomarkers are considered to be frontrunners in the field of early detection. In summary, LDCT screening, although yet to be broadly adopted, was shown to reduce lung cancer mortality in a large, US-based clinical trial, and is now recommended by several major medical organizations in specific high-risk groups within the United States.

Among the unanswered questions with LDCT screening are the duration of screening, the interval between tests, the definition of the high-risk individuals, the management of indeterminate pulmonary nodules, the cost-effectiveness analysis, the benefit of computerized characterization of lung nodules and lung parenchyma over routine visual analysis, and the assessment of the radiation risk associated with LDCT screening. Although the amount of radiation exposure from a low-dose spiral CT scan is relatively modest (roughly equal to 10 chest radiographs or one-tenth that experienced with a regular chest CT scan), it is not an insignificant issue, and if applied to very low-risk populations some calculations actually show a theoretically increased risk of lung cancer.

A major challenge confronting advocates of CT screening is the high false-positive rate. On initial screening of at-risk populations, false-positive rates range from 10% to 20% of the screened population but can be as high as 50%. Positive predictive values range from 2.8% to 11.6%. False positives can have a substantial impact on patients through the expense and risk of unneeded further invasive and morbidity-associated evaluation, in addition to creating uncertainty and emotional stress. False-positive rates and positive predictive values are somewhat improved in annual follow-up CT scans, but there is still significant room for improvement. Based on extant data it appears that nodules smaller than 5 mm are unlikely to be cancerous and those 5 to 10 mm in diameter (25%–40% of noncalcified nodules detected) are of uncertain significance. The management of patients with these nodules usually consists of repeated CT scans over time to see if the nodules grow, attempted FNA, or surgical resection.

Finally, CT scanning has also shown promise in the detection of some premalignant lesions. High-resolution CT imaging has proven to be sensitive enough to detect GGOs, some of which may represent inflammatory lesions, AIS, or atypical adenomatous hyperplasia (AAH), a presumed precursor lesion to adenocarcinoma. Because adenocarcinoma is now the most common histologic type of lung cancer in the United States, there is some hope that CT scanning may be able to improve survival for these patients by revealing a precursor lesion before its transformation to invasive carcinoma. Chest CT scanning, however, is not yet sensitive enough to reveal premalignant epithelial lesions of the bronchial tree with squamous differentiation, likely precursors of squamous carcinoma of the lung.

Positron Emission Tomography

Many CT screening protocols now employ PET-CT as a method of reducing this high false-positive rate. The data regarding this application remain controversial. PET scanning attempts to identify malignancy based on glucose metabolism by measuring the uptake of fluorine-18 fluorodeoxyglucose ( 18 F-FDG). Lung cancers will preferentially take up 18 F-FDG and appear as “hot spots.” Currently, PET has been mostly used for clinical staging and detection of metastases in lung cancer and in the diagnosis of nodules larger than 15 mm in diameter. In combination with another imaging modality, PET may provide information about the metabolic state of lung nodules. Combined 18 F-FDG PET-CT scan has been shown to improve the accuracy of staging in lung cancer compared with visual correlation of PET and CT or either study alone. The added information gained from PET and combined PET-CT may help reduce the high false-positive rates seen in trials using CT alone. A trial of low-dose chest CT (LDCT), used in combination with PET scan of 1035 patients resulted in only six false positives, defined as a surgical biopsy of a benign nodule, out of 27 surgical biopsies of suspicious nodules. There are important limitations to this study, however. First, lung nodules less than or equal to 5 mm in size were followed with repeat CT in 1 year and no other intervention. Second, PET scanning was performed only on larger nodules (≥7 mm). Typically, PET has not performed well in the identification of small nodules (<15 mm), so its usefulness in intermediate lesions remains in question, in particular in areas with high prevalence of pulmonary fungal infections. There is no evidence of a survival benefit when PET scan is used a part of a screening protocol.

In fact, PET has limited usefulness in detection of lesions smaller than 1 cm and has shown to be of limited value in adenocarcinoma, particularly very well-differentiated subtypes. Each method of evaluation is costly, and some cause morbidity. The impact on patient outcome of waiting to assess nodule growth is also not clear, but can only decrease curability. Even for patients with disease that is highly suspected to be lung cancer on clinical grounds, there is a 10% to 20% incidence of “futile thoracotomies” in which the suspicious lesion is found to be benign and the patient unnecessarily incurred the morbidity and potential mortality of a thoracotomy. Eliminating unnecessary operations should be one of our priorities, although this may not be feasible without the development of other strategies for early detection.

Other Imaging Techniques

With fluorescence endoscopy, differences in the autofluorescence characteristics of normal and neoplastic epithelium are used to localize lesions. Fluorescence bronchoscopy has been shown to be more sensitive than white light bronchoscopy in the detection of preneoplastic lesions in many studies, including a randomized trial. A randomized trial demonstrated that the use of laser-induced fluorescence endoscopy (LIFE) resulted in a 46.9% absolute increase in the sensitivity of detection of moderate dysplasia or worse in high-risk patients when compared with white light bronchoscopy, although the specificity was worse. Fluorescence bronchoscopy has not been proven as a validated method of early detection of lung cancer, but it is a very important research tool.

New imaging techniques are currently being developed not only to help improve the resolution of current imaging techniques, but also to perform imaging of lesions based on biologic activity. Both magnetic resonance and gamma camera techniques are being tested as molecular imaging tools. Near-infrared Raman spectroscopy, optical coherence tomography, and confocal microscopy use optical differences within tissue to allow imaging of individual cell nuclei. This technology is being adapted for use during endoscopic examinations and may be able to provide real-time histologic evaluation of bronchial mucosa.

Biofluid-Based Biomarkers for Lung Cancer

The underlying premise of biofluid-based biomarker research is that molecular alterations of tumor cells lead to the synthesis and shedding of distinct molecular species that can be detected in biofluids. Biofluid-based detection strategies are an attractive approach for screening, because of their ease of acquisition. Biofluids, including peripheral blood and its components (circulating cells, plasma, and serum), exhaled breath condensate (EBC), urine, and sputum, offer noninvasive access to large quantities of samples for analysis. These alterations can lead to the generation of disease-specific molecular species such as altered or methylated DNA, overexpressed mRNA, microRNA, or proteins that can potentially be released into the extracellular microenvironment at a rate that is dependent on multiple variables but mainly tumor growth and shedding rates. Therefore molecular analyses of early-stage lung cancer–related biofluids represent an attractive choice for the discovery and validation of diagnostic biomarkers. The success of this approach will also depend not only on our ability to improve the specificity of our candidate biomarkers but also on the sensitivity of our detection assays. Although various serum biomarkers have been investigated in lung cancer, none has proved useful in general clinical practice, mainly because of the lack of sufficient sensitivity and specificity (reviewed by Barrera and colleagues ). For example, cytokeratin fragment antigen 21.1 (Cyfra-21.1), a marker of cytokeratin, CEA, and tissue polypeptide antigen (TPA) were found to have relatively poor sensitivity (31%–64%) when specificity limits of 95% were set. In addition, most markers reach better sensitivity in advanced disease stages as compared with stage I lung cancer. Therefore their use for early diagnosis or screening has not had an impact on patient care in the clinic. Blood-derived biomarkers used in combination with other clinical, imaging, or molecular tools might play an important role in early detection, in monitoring response to therapy, in risk assessment of recurrence, and in prognosis, especially in intermediate-risk nodules in low-risk individuals or that are PET negative.

In conclusion, LDCT in individuals at high risk now offers excellent sensitivity for detection of lung cancer at an early stage, brings a stage shift among tumors diagnosed, and saves lives. The high sensitivity of these screening tests, however, is associated with a low specificity. Better selection of individuals at highest risk of lung cancer through use of biomarkers of disease and genetic susceptibility may improve their positive predictive values, minimize the false-positive rates and associated unnecessary investigations or treatment, and reduce the cost of the early detection process.

Clinical Presentation

Presenting Signs and Symptoms

Symptoms, signs, and laboratory test abnormalities relating to lung cancer can be classified as those caused directly by the primary lesion, those related to intrathoracic spread or to distant metastasis, and those related to paraneoplastic syndromes. The prototypical lung cancer patient is a current or former smoker of either sex, usually in the seventh decade of life, with symptoms attributable to bulky intrathoracic disease (i.e., cough, dyspnea, chest pain, hoarseness, and/or hemoptysis) or distant metastases (e.g., bone pain, central nervous system [CNS] symptoms). Constitutional symptoms may include weakness, anorexia, weight loss, and, rarely, fever. Apart from the brevity of symptom duration, these parameters fail to clearly distinguish SCLC from NSCLC or even from neoplasms metastatic to the lungs. Lung cancer arising in a lifelong never-smoker is more common in women and certain ethnic groups and tends to be an adenocarcinoma. Such patients also tend to be slightly younger than their smoking counterparts at the time of diagnosis. However, the clinical presentation of lung cancer in never-smokers tends to mirror that of current and former smokers even though lung cancers arising in never-smokers appear to be biologically distinct as evidenced by the differing molecular abnormalities found in tumors derived from smokers and nonsmokers. The prognosis of lung cancer in never-smokers is generally improved compared with cancers in current or former smokers, irrespective of stage. This may be due to the better overall condition of these patients, the higher frequency of actionable genetic abnormalities in tumors from these patients (to be discussed later), or other as yet unknown reasons.

Cough, dyspnea, and chest discomfort are the most common presenting symptoms in lung cancer ( Table 69.7 ). A history of chronic cough with or without hemoptysis in a current or former smoker with COPD aged 40 years or older should prompt a thorough investigation for lung cancer even in the face of a normal chest radiograph. A persistent “pneumonia” without constitutional symptoms and unresponsive to repeated courses of antibiotics also should prompt an evaluation for an underlying cause (e.g., an occult endobronchial lesion), especially if the patient is a current or former smoker. Less frequently, patients present with hemoptysis, which rarely is massive and is usually described as streaks of fresh or old blood in sputum. The spread of disease within the chest may result in hoarseness secondary to a left recurrent laryngeal nerve paralysis and, less frequently, phrenic nerve paralysis. The latter is associated with an elevated hemidiaphragm on standard chest radiographs. Chest wall involvement is commonly accompanied by pain that in turn may serve as a more accurate indicator of chest wall invasion than radiographic studies. Chest wall pain is usually related to direct invasion of the pleura or chest wall by the primary tumor or is due to a rib metastasis. Tenderness may be elicited at the site of rib involvement and, rarely, a soft-tissue mass can be palpated. The chest pain may have a pleuritic component if there is pleural involvement. The disappearance of pleuritic chest pain may signify the development of a pleural effusion that in turn may cause shortness of breath or may worsen existing dyspnea. Venous distention of the neck and chest wall, cyanosis, facial plethora, and upper extremity edema may indicate obstruction of the superior vena cava, which today is most commonly seen with SCLC. Although the heart and other mediastinal structures are often involved with tumor at postmortem examination, only rarely does this involvement serve as the source of a presenting symptom.

Table 69.7
Presenting Sign and Symptoms of Lung Cancer
Reproduced with permission from Beckles MA, Spiro SG, Colice GL, Rudd RM. Initial evaluation of the patient with lung cancer: symptoms, signs, laboratory tests, and paraneoplastic syndromes. Chest. 2003;123:97–104.
Symptoms and Signs Range of Frequency
Cough 8%–75%
Weight loss 0%–68%
Dyspnea 3%–60%
Chest pain 20%–49%
Hemoptysis 6%–35%
Bone pain 6%–25%
Clubbing 0%–20%
Fever 0%–20%
Weakness 0%–10%
SVCO 0%–4%
Dysphagia 0%–2%
Wheezing and stridor 0%–2%
SVCO, superior vena cava obstruction.

Approximately one-third of patients have symptoms as a result of distant metastases. The most common sites of distant metastasis from lung cancer are the bones; liver, adrenal glands, and intraabdominal lymph nodes; brain and spinal cord; and lymph nodes and skin. Lung cancer can metastasize to virtually any bone, with pain being the primary presenting symptom in up to 25% of patients. Similarly, liver metastases are common at initial presentation in both SCLC and NSCLC. However, liver function test results are seldom abnormal until the metastases are numerous and large. Hepatic metastases most commonly produce symptoms of weakness and weight loss. Adrenal lesions (only rarely associated with adrenal insufficiency) and paraaortic lymph node metastases are most commonly seen with SCLC. Intracranial metastases at presentation are most common in SCLC and adenocarcinomas of the lung. Presenting symptoms may include headache, nausea and vomiting, focal neurologic symptoms or signs, seizures, confusion, and personality changes.

The stigmata of COPD in smokers may be the only findings on physical examination, or one may detect lymphadenopathy, hepatomegaly, bone tenderness, or abnormal neurologic findings. Digital clubbing and hypertrophic pulmonary osteoarthropathy (HPO) may be associated with any histologic subtype of lung cancer but are most frequently associated with squamous cell and adenocarcinoma and are least likely to occur in a patient with SCLC. Digital clubbing is more common than HPO, and the latter is characterized by painful symmetric arthropathy and periosteal new bone formation of the distal limbs. Its mechanism of development is unknown. In a published series of 111 consecutive lung cancer patients, clubbing was noted in 29%, with an incidence of 35% in NSCLC and only 4% in SCLC. Up to 20% of patients develop palpable lymphadenopathy in the supraclavicular fossa during the course of the disease. Subcutaneous metastases, although rare, portend a poor prognosis but serve as a readily accessible source of diagnostic material.

Paraneoplastic Disorders

Although many of the symptoms of NSCLC and SCLC are attributable to mass effect and direct impingement on vital organs, less commonly patients with lung cancer have symptoms related to hypercalcemia, hyponatremia, Cushing syndrome, Lambert Eaton syndrome, and other neurologic disorders. These so-called paraneoplastic phenomena can be seen in any histologic type of lung cancer, but are most frequently associated with SCLC. In general, a majority of these paraneoplastic phenomena fall into endocrine or neurologic categories.

Hypercalcemia

Hypercalcemia of malignancy (HCM) is the most common life-threatening metabolic complication of malignancy, affecting approximately 10% to 20% of patients with advanced cancer. Hypercalcemia may be associated with or due to production of a parathyroid hormone–related peptide. The incidence of HCM varies widely by cancer type, but it occurs most frequently in patients with multiple myeloma and carcinomas of the lung, breast, kidney, and head and neck. With respect to lung cancer, squamous cell carcinoma is the most common histologic subtype. Clinical symptoms of HCM include nausea, vomiting, abdominal pain, constipation, polyuria and thirst, and altered mental status. HCM may lead to renal failure. The early symptoms of nausea, vomiting, and constipation may be easily confused with the initiation of narcotics for pain control.

Hyponatremia and the Syndrome of Inappropriate Antidiuretic Hormone

The inappropriate secretion of antidiuretic hormone, or arginine vasopressin (AVP), with its resultant euvolemic, refractory, hypoosmolar hyponatremia, is observed in up to 15% of SCLC patients. However, up to one-third of patients with hyponatremia have no evidence of ectopic AVP production. In such patients hyponatremia may be caused by ectopic production of atrial natriuretic peptide (ANP). SCLC is the most common malignant cause of acute or chronic hyponatremia and the syndrome of inappropriate antidiuretic hormone (SIADH). The presence of SIADH does not correlate with clinical stage, distribution of metastatic sites, or sex, nor does SIADH influence response to chemotherapy or OS as an independent variable. As with most paraneoplastic syndromes, the best therapy for SIADH is effective treatment of the underlying SCLC. SIADH typically resolves with 1 to 4 weeks of initiation of chemotherapy in the vast majority of cases. While awaiting the effects of chemotherapy, serum sodium can usually be managed and maintained above 128 mEq/L via strict fluid restriction alone. Demeclocycline, which blocks the action of vasopressin at the level of the renal tubule, can be a useful adjunctive measure when fluid restriction alone is insufficient to restore sodium level. Tolvaptan, an oral vasopressin V2-receptor nonpeptide antagonist, also is effective in increasing serum sodium concentrations in patients with euvolemic and hypervolemia hyponatremia. Of note, patients with ectopic ANP secretion do not respond to fluid restriction. In fact, fluid restriction may actually worsen hyponatremia if sodium intake is not concomitantly increased. Accordingly, if hyponatremia fails to improve or if it worsens after 3 to 4 days of adequate fluid restriction, plasma levels of AVP and ANP should be measured to determine whether inappropriate secretion of ADH or ANP is the causative syndrome.

Ectopic Adrenocorticotropic Hormone Production

Cushing syndrome may be due to ectopic secretion of adrenocorticotropic hormone (ACTH) from a nonpituitary tumor resulting in bilateral adrenocortical hyperplasia and hypercortisolemia. Neuroendocrine lung tumors including SCLC and pulmonary carcinoids account for approximately half the cases of ectopic ACTH-producing tumors. Although hypercortisolemia has been documented in up to 50% of SCLC cases, only 2% to 5% of SCLC patients have the characteristic clinical features of Cushing syndrome. Unlike in Cushing disease, the onset of symptoms in ectopic ACTH is often abrupt because of the characteristic rapid growth of SCLC. Consequently the classic features of Cushing disease—a buffalo hump, striae, and moon face—are frequently absent. By contrast, hypokalemic alkalosis, hypertension, and hyperglycemia are common. The effect of Cushing syndrome on survival is unclear, although some investigators hold that its onset heralds a more aggressive tumor behavior. Treatment with standard medications, such as metyrapone and ketoconazole, is largely ineffective owing to extremely high cortisol levels. Some patients require bilateral adrenalectomy to control symptoms. The most effective strategy for management of the Cushing syndrome is effective treatment of the underlying SCLC.

Neurologic Paraneoplastic Syndromes

The paraneoplastic neurologic disorders are a diverse group of diseases characterized by the presence of neurologic dysfunction in the setting of a remote cancer. They are often the result of production of antibodies that react with both the SCLC cells and with normal host tissue. Well-described syndromes include Lambert-Eaton myasthenic syndrome (LEMS), paraneoplastic encephalomyelitis, sensorimotor neuropathy, and paraneoplastic cerebellar degeneration. Less frequent abnormalities include subacute sensory neuropathy, autonomic disturbances, myelopathies, progressive encephalopathy, and visual paraneoplastic syndromes.

LEMS is caused by autoantibodies directed against presynaptic voltage-gated P/Q calcium channels. The P/Q calcium channel autoantibodies decrease calcium entry into the presynaptic terminal, which prevents binding of vesicles to the presynaptic membrane and acetylcholine release. Patients with this disorder have proximal muscle weakness, usually in the lower extremities, occasional autonomic dysfunction, and, rarely, cranial nerve symptoms or involvement of the bulbar or respiratory muscles. Depressed deep tendon reflexes are frequently present. In contrast to patients with myasthenia gravis, strength improves with serial effort. The diagnosis is confirmed with electrophysiologic testing, which demonstrates small compound muscle action potentials and facilitation with exercise or 20-Hz repetitive stimulation. A serum test for voltage-gated calcium channel antibodies, estimated to occur in 5% of patients with SCLC, is commercially available. Plasma exchange and intravenous immunoglobulin can provide short-term benefit; 3,4-diaminopyridine, which enhances the release of acetylcholine from presynaptic terminals, prednisone, and azathioprine can provide limited long-term benefit. Some patients who respond to chemotherapy will have resolution of the neurologic abnormalities, and this is the initial treatment of choice.

Paraneoplastic encephalomyelitis and sensory neuropathies, cerebellar degeneration, limbic encephalitis, and brainstem encephalitis occur in SCLC in association with a variety of antineuronal antibodies such as anti-Hu, anti-CRMP5, and ANNA-3. These antibodies have been found in up to 25% of SCLC patients, although not always in association with a clinically obvious neurologic disorder. These disorders may predate the diagnosis of SCLC. Most paraneoplastic neuropathies are sensorimotor and axonal, symmetric in distribution, and frequently disabling. Limbic encephalitis is characterized by degeneration of neurons in the medial temporal lobe with clinical features that include behavioral changes, hallucinations, short-term memory loss, anosmia, ageusia, and dementia. Symptoms of brainstem encephalitis include vertigo, nystagmus, oscillopsia, ataxia, diplopia, dysarthria, and dysphagia, reflecting the predominant involvement of the floor of the fourth ventricle and inferior olives. Some patients develop respiratory insufficiency and require assisted ventilation. Cerebrospinal fluid (CSF) studies may show pleocytosis and elevated protein levels. Brain magnetic resonance imaging (MRI) scans are typically normal. As with voltage-gated calcium channel antibodies, the presence of anti-Hu antibodies does not correlate with neurologic symptoms nor with an improved prognosis.

Paraneoplastic cerebellar degeneration, manifesting with ataxia, dysarthria, and nystagmus, may be associated with anti-Hu, anti-Yo or P/Q calcium channel autoantibodies. Patients often develop loss of coordination that usually starts on one side and rapidly progresses over days to weeks to involve both sides equally. Additional presenting symptoms include limb and truncal ataxia, lack of coordination, dysarthria, and nystagmus. More rarely, patients experience opsoclonus, myoclonus, memory disturbances, pyramidal signs, sensory disturbances, or hyporeflexia. After progressing for a few weeks, the symptoms stabilize, leaving the patient in a severely disabled state. On examination, patients may be unable to stand without assistance owing to severe truncal and neck ataxia with markedly ataxic gait. Ocular findings may include horizontal or vertical nystagmus, disconjugate gaze, ocular dysmetria, and opsoclonus. Speech also can be affected severely, with impairment developing initially as mild dysarthria and progressing to incomprehensible words in severe cases. Mild deterioration of mental status may also occur, but marked changes in mental status are not compatible with this diagnosis. Treatment may include steroids, plasmapheresis, and chemotherapy. However, treatment of the tumor and/or immunomodulation does not alter the course of paraneoplastic cerebellar degeneration, but may improve LEMS symptoms. Death is frequently due to neurologic complications. Paradoxically, the tumor frequently remains localized to the chest or not detected.

Diagnostic Workup and Staging

Assessment of Intrathoracic Disease

Accurate clinical staging of lung cancer is extremely important because treatment options and prognosis are dictated by stage at presentation. The most significant dividing line is between patients who are candidates for treatment with curative intent (surgery or definitive chemoradiation) and those who are not suitable for such interventions but who will benefit from chemotherapy, radiation therapy (RT), or both. Staging with regard to a patient's potential for surgical resection is most applicable to NSCLC. The basis for staging NSCLC is the TNM system, which was updated in the eighth edition of the Union for International Cancer Control (UICC)/American Joint Committee on Cancer (AJCC)/IASLC staging system ( Tables 69.8A and 69.8B ), in effect as of January 2018. For SCLC, a more simplified staging classification is often used (described later). From a practical standpoint, metastases to the hilar or mediastinal lymph nodes are reflected in the N designator. An international lymph node map has been developed as part of the staging process. Determination of metastases to mediastinal lymph nodes constitutes a critical point in staging and treatment recommendations. Patients with mediastinal node metastases may also be selected for multidisciplinary therapy including induction therapy (either chemotherapy or chemoradiotherapy) followed by resection.

Table 69.8A
TNM Classification of Lung Cancer (Eighth Edition of the UICC/AJCC/IASLC Staging System)
T: PRIMARY TUMOR
Tx Primary tumor cannot be assessed or tumor proven by presence of malignant cells in sputum or bronchial washings but not visualized at imaging or bronchoscopy
T0 No evidence of primary tumor
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 the lobar bronchus (i.e., not in the main bronchus) a
T1a(mi) Minimally invasive adenocarcinoma b
T1a Tumor ≤1 cm in greatest dimension a
T1b Tumor >1 cm but ≤2 cm in greatest dimension a
T1c Tumor >2 cm but ≤3 cm in greatest dimension a
T2 Tumor >3 cm but ≤5 cm or tumor with any of the following features c :

  • Involves main bronchus regardless of distance from the carina but without involvement of the carina

  • Invades visceral pleura

  • Associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung

T2a Tumor >3 cm but ≤4 cm in greatest dimension
T2b Tumor >4 cm but ≤5 cm in greatest dimension
T3 Tumor >5 cm but ≤7 cm in greatest dimension or associated with separate tumor nodule(s) in the same lobe as the primary tumor or directly invades any of the following structures: chest wall (including the parietal pleura and superior sulcus tumors), phrenic nerve, parietal pericardium
T4 Tumor >7 cm in greatest dimension or associated with separate tumor nodule(s) in a different ipsilateral lobe than that of the primary tumor or invades any of the following structures: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, and carina
N: REGIONAL LYMPH NODE INVOLVEMENT
Nx Regional lymph nodes cannot be assessed
N0 No regional lymph node metastasis
N1 Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension
N2 Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s)
N3 Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s)
M: DISTANT METASTASIS
M0 No distant metastasis
M1 Distant metastasis present
M1a Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodule(s) or malignant pleural or pericardial effusion d
M1b Single extrathoracic metastasis e
M1c Multiple extrathoracic metastases in one or more organs

AJCC, American Joint Committee on Cancer; IASLC, International Association for the Study of Lung Cancer; UICC, Union for International Cancer Control.

a The uncommon superficial spreading tumor of any size with its invasive component limited to the bronchial wall, which may extend proximal to the main bronchus, is also classified as T1a.

b Solitary adenocarcinoma, 3 cm with a predominately lepidic pattern and 5 mm of invasion in any one focus.

c T2 tumors with these features are classified as T2a if 4 cm in greatest dimension or if size cannot be determined, and T2b if >4 cm but 5 cm in greatest dimension.

d Most pleural (pericardial) effusions with lung cancer are due to tumor. In a few patients, however, multiple microscopic examinations of pleural (pericardial) fluid are negative for tumor and the fluid is nonbloody and not an exudate. When these elements and clinical judgment dictate that the effusion is not related to the tumor, the effusion should be excluded as a staging descriptor.

e This includes involvement of a single distant (nonregional) lymph node.

Table 69.8B
Stage Grouping of Lung Cancer (Eighth Edition of the UICC/AJCC/IASLC Staging System)
Stage Group T Descriptor N Descriptor M Descriptor
Occult carcinoma TX N0 M0
Stage 0 Tis N0 M0
Stage IA1 T1a(mi) N0 M0
T1a N0 M0
Stage IA2 T1b N0 M0
Stage IA3 T1c N0 M0
Stage IB T2a N0 M0
Stage IIA T2b N0 M0
Stage IIB T1a-c N1 M0
T2a N1 M0
T2b N1 M0
T3 N0 M0
Stage IIIA T1a-c N2 M0
T2a-b N2 M0
T3 N1 M0
T4 N0 M0
T4 N1 M0
Stage IIIB T1a-c N3 M0
T2a-b N3 M0
T3 N2 M0
T4 N2 M0
Stage IIIC T3 N3 M0
T4 N3 M0
Stage IVA Any T Any N M1a
Any T Any N M1b
Stage IVB Any T Any N Mic
AJCC, American Joint Committee on Cancer; IASLC, International Association for the Study of Lung Cancer; M, metastasis; N, node; T, tumor; T1a(mi), minimally invasive adenocarcinoma; Tis, carcinoma in situ; UICC, Union for International Cancer Control.

Several noninvasive imaging techniques are available to aid in identifying the extent of disease both within and outside of the chest. Some lung cancers are detected with plain chest radiography ( Fig. 69.11 ). However, the chest radiograph is not sufficiently sensitive to accurately assess the mediastinum. Accordingly, a chest CT scan should be performed in all patients with known or suspected NSCLC. The chest CT scan provides anatomic detail that better identifies the location of the tumor, its proximity to local structures, and whether or not lymph nodes in the mediastinum are enlarged (see Fig. 69.11 ). Unfortunately, the accuracy of chest CT scanning in differentiating benign from malignant lymph nodes in the mediastinum based on node size is low. The most commonly used size criterion is a short axis diameter of 1 cm or greater on a transverse CT scan as an indicator of a “suspicious” lymph node. Whole-body PET scanning provides functional information on metabolic activity, is more sensitive and specific than chest CT scanning for staging lung cancer in the mediastinum ( Fig. 69.12 ), and can reveal occult metastases. Positive findings on PET scans can occur from nonmalignant conditions (e.g., infections), so tissue sampling to confirm the suspected malignancy is strongly recommended. MRI can be useful in selected circumstances such as superior sulcus tumors to rule out brachial plexus involvement but in general does not play a major role in NSCLC staging. Of course, abnormalities detected with any of the aforementioned imaging studies are not necessarily cancer. Unless overwhelming evidence of metastatic disease is present on imaging studies, curative-intent therapy should not be withheld based on imaging studies alone, but putative metastatic disease should be documented with tissue confirmation of malignancy.

Figure 69.11, Imaging findings in non–small cell lung cancer. (A) Posteroanterior and (B) lateral chest radiographs of patients with locally advanced disease. (C) Computed tomography scans using lung and (D) mediastinal windows.

Figure 69.12, Fluorine-18 fluorodeoxyglucose–positron emission tomography ( 18 F-FDG–PET) with integrated computed tomography. FDG-PET demonstrates (A) activity in the left upper lobe lesions and (B) activity in the subcarinal space. Physiologic uptake is noted in the brain, bones, liver, spleen, kidney, and musculature. Increased activity is also identified at the intravenous insertion site, renal collecting system, and bladder. Fused computed tomography–PET images confirm (C) FDG-avid lymph node in the anteroposterior window and (D) mild FDG uptake in the subcarinal lymph node.

Assessment of Extrathoracic Disease

The best initial predictor of metastatic disease remains a careful history and physical examination. If signs, symptoms, or findings from the physical examination suggest the presence of malignancy, then sequential imaging, starting with the most appropriate study based on the clues obtained during the clinical evaluation should be performed. If the findings from the clinical evaluation are negative, then additional imaging studies such as a CT scan of the head, a bone scan, or an abdominal CT scan are often unnecessary, and the search for metastatic disease is complete. Clinical findings suggestive of metastatic disease are listed in Table 69.9 . Patients with abnormal clinical evaluation findings should undergo imaging for extrathoracic metastases. Site-specific symptoms warrant directed evaluation of that site with the most appropriate study (e.g., CT scan of the head, bone scan, and abdominal CT scan). More controversial is how one should assess patients with known clinical stage III disease. Additional imaging may be required, given the high frequency of brain metastases, for example.

Table 69.9
Clinical Findings Suggesting Metastatic Disease
Reproduced with permission from Silvestri GA, Tanoue LT, Margolis ML, Barker J, Detterbeck F. The noninvasive staging of non–small cell lung cancer: the guidelines. Chest. 2003;123(1 suppl):147S–156S.
Symptoms elicited in history Constitutional: weight loss >10 lb
Musculoskeletal: focal skeletal pain
Neurologic: headaches, syncope, seizures, extremity weakness, recent change in mental status
Signs found on physical examination Lymphadenopathy (>1 cm)
Hoarseness, superior vena cava syndrome
Bone tenderness
Hepatomegaly (>13-cm span)
Focal neurologic signs, papilledema
Soft tissue mass
Routine laboratory test results Hematocrit <40% in men, <35% in women
Elevated alkaline phosphatase, GGT, SGOT, and calcium levels
GGT, γ-Glutamyltransferase; SGOT, serum glutamic-oxaloacetic transaminase.

Solitary Pulmonary Nodule

The approach to a patient with a solitary pulmonary nodule is based on an estimate of the probability of cancer, determined according to the size of the nodule, the presence or absence of a history of smoking, the patient's age, and characteristics of the nodule's margins on CT images. Mayo Clinic investigators found that reported clinical characteristics (age, cigarette-smoking status, and prior cancer diagnosis ≥5 years ago) and three radiologic characteristics (diameter, spiculation, and upper lobe location) were independent predictors of malignancy. An efficient algorithm for assessing these lesions is outlined in Fig. 69.13 .

Figure 69.13, Algorithm for evaluation of a solid solitary pulmonary nodule > 8 mm in diameter in patients with an acceptable surgical risk profile.

Non–Small Cell Lung Cancer

The four major histologic types of lung cancer—squamous cell carcinoma, adenocarcinoma, large cell carcinoma, and small cell carcinoma—together account for more than 90% of lung cancer cases in the United States. The first three are traditionally lumped together into the category of NSCLC. The incidence of NSCLC has been decreasing since the mid-1970s to 1980s among men in North America, northwestern Europe, Australia, and New Zealand but the age-adjusted rate continues to increase among women in these countries, and among both men and women in southern and Eastern Europe. These trends followed changes in smoking behavior. Over time there has been a significant shift in the incidence rates of lung cancer by histologic type. After a steadily increasing occurrence during the period 1973 to 1987, adenocarcinoma supplanted squamous cell carcinoma as the most frequent form of lung cancer. Several hypotheses have been posited as to the underlying cause of this shift, but data have suggested that changes in smoking behavior and cigarette design are major contributing factors.

Presurgical Evaluation

Physiologic Evaluation

The physiologic evaluation must be individualized for each patient, but in general emphasizes pulmonary and cardiac function and follows a stepwise progression. In some patients, history, physical examination, and routine spirometry are all that is required for physiologic assessment. For others, further physiologic assessment is indicated before pulmonary resection. The assessment of a patient's ability to tolerate lung resection from a cardiopulmonary or physiologic standpoint is fundamental to patient selection for surgery. The most oncologically sound and perfectly executed operation for lung cancer falls far short of success in the event of a major cardiopulmonary complication or severe long-term functional disability.

Smoking Cessation

Cigarette smoking is associated with up to a sixfold increase in the incidence of postoperative pulmonary complications after surgery. A clear concise statement from the physician to stop smoking completely and to never restart smoking is required. Ideally, patients are smoke free for a minimum of 2 weeks and preferably for 4 to 8 weeks before surgery, although smoking cessation at any time is valuable. Although there are few studies specific to pulmonary resection, there is evidence that preoperative smoking cessation within 8 weeks of the operation does not increase the risk of pulmonary complications. The powerful addictive properties of cigarette smoking must be recognized. Patients should be offered pharmacologic adjuncts and should be enrolled in a formal tobacco cessation program before consideration for resection. Resection in patients with NSCLC who continue to smoke should carefully balance the risks of significant morbidity of pneumonia in the postoperative period with the benefits of resection.

Nutritional Status

Weight loss and malnutrition are very common among patients with NSCLC. In a recent study, more than 40% of patients undergoing resection for NSCLC had a body mass index and skin fold thickness below the 25th percentile. Poor nutrition in surgical patients correlates with impaired wound healing and a greater propensity to develop postoperative infection.

Impact of Age

Advanced age is an independent predictor of mortality after resection for NSCLC. With recent advances in patient selection and perioperative care, however, lung resection may be performed with acceptable rates of morbidity and mortality in patients well beyond the age of 70 years. Advanced age does predict a higher incidence of perioperative cardiovascular complications, but age reflects a surrogate marker for additional comorbidities rather than an independent risk factor.

Spirometry and Pulmonary Diffusion Capacity

Spirometry is mandatory for patients under consideration for pulmonary resection for NSCLC and provides an objective assessment of pulmonary function. Advanced pulmonary disease may carry significant to prohibitive risk in more than one-third of patients with otherwise resectable NSCLC. The forced expiratory volume in 1 second (FEV 1 ) is the historical standard to determine suitability for resection; a predicted postoperative FEV 1 (ppoFEV 1 ) can be estimated based on the planned extent of resection: ppoFEV 1 = preoperative FEV 1 × (number of segments remaining/total segments).

FEV 1 is an independent predictor of mortality from surgery for lung cancer and serves as the primary determinant of the need for further physiologic assessment before surgery for NSCLC. The criteria of ppoFEV 1 of at least 0.8 L has been widely used in decisions for lung cancer resection ; however, an absolute value of FEV 1 predicts postresection pulmonary function less accurately than FEV 1 expressed as a percentage of the expected value for age and size. The use of absolute FEV 1 measurements in patient selection may bias against older patients, those of small stature, and female patients. Patients deemed unable to tolerate lobectomy from a pulmonary functional standpoint may be candidates for more limited resections, such as wedge or anatomic segmental resection, although such procedures may be associated with significantly higher rates of local recurrence and a trend toward decreased survival, or may benefit from minimally invasive video-assisted thoracic surgery (VATS) techniques and improved postoperative pain management techniques (e.g., patient-controlled analgesia, epidural anesthesia).

Pulmonary diffusing capacity for carbon monoxide (D lco ) is an adjunctive test to spirometry and lung volume measurements. D lco provides a measurement of the lung surface area available for gas exchange and is determined by measuring expired carbon monoxide levels during controlled exhalation. The D lco measures the rate at which test molecules such as carbon monoxide move from the alveolar space to combine with hemoglobin in the red blood cells. The D lco is determined by calculating the difference between inspired and expired samples of gas. D lco levels below 50% are associated with increased perioperative risk. A low D lco reflects the presence of emphysema, fibrosis, or pulmonary vascular disease. Similar to FEV 1 , preoperative D lco measurement is most useful when expressed as a percentage of predicted value and may be used to estimate predicted postoperative D lco (ppoD lco ).

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here