Non-Small Cell Lung Cancer (NSCLC)

Back

Practice Essentials

Non–small cell lung cancer (NSCLC) accounts for approximately 85% of all lung cancers. Histologically, NSCLC is divided into adenocarcinoma, squamous cell carcinoma (SCC) (see the image below), and large cell carcinoma. Patients with NSCLC require a complete staging workup to evaluate the extent of disease, because stage plays a major role in determining the choice of treatment.



View Image

Non–small cell lung cancer. A cavitating right lower lobe squamous cell carcinoma.

See the Critical Images slideshow Cutaneous Clues to Diagnosing Metastatic Cancer to help identify various skin lesions that are cause for concern. 

Go to Small Cell Lung Cancer for complete information on this topic. Go to Oncology Decision Point for expert commentary on NSCLC treatment decisions and related guidelines.

Signs and symptoms

NSCLC is often insidious, producing no symptoms until the disease is well advanced. Early recognition of symptoms may be beneficial to outcome.

At initial diagnosis, 20% of patients have localized disease, 25% of patients have regional metastasis, and 55% of patients have distant spread of disease. Symptoms depend on the location of cancer.[1]

The most common signs and symptoms of lung cancer include the following:

Metastatic signs and symptoms may include the following:

See Presentation for more detail.

Diagnosis

Testing

After physical examination and CBC, chest x-ray is often the first test performed. Chest radiographs may show the following:

There are several methods of confirming diagnosis, with the choice determined partly by lesion location. These methods include the following:

Staging

A chest CT scan is the standard for staging lung cancer. The TNM (tumor-node-metastasis) staging system from the American Joint Committee for Cancer Staging and End Results Reporting is used for all lung carcinomas except small-cell lung cancer. The TNM takes into account the following key pieces of information:

Primary tumor (T) involvement is as follows:

Lymph node (N) involvement is as follows:

Metastatic (M) involvement is as follows:

See Workup for more detail. See also Lung Cancer Staging -- Radiologic Options, a Critical Images slideshow, to help identify stages of the disease process.

Management

Surgery, chemotherapy, and radiation are the main treatment options for NSCLC. Because most lung cancers cannot be cured with currently available therapeutic modalities, the appropriate application of skilled palliative care is an important part of the treatment of patients with NSCLC.

Surgery

Surgery is the treatment of choice for stage I and stage II NSCLC. Several different types of surgery can be used, as follows:

Chemotherapy

Approximately 80% of all patients with lung cancer are considered for chemotherapy at some point during the course of their illness. Multiple randomized, controlled trials and large meta-analyses all confirm the superiority of combination chemotherapy regimens up front for advanced NSCLC.

The American Society for Clinical Oncology (ASCO) guidelines recommend that first-line treatment for NSCLC include a platinum combination. In younger patients, with a good performance status or in the adjuvant setting, cisplatin is preferred, but in older patients or those with significant comorbidities, carboplatin may be substituted.

Radiation

In the treatment of stage I and stage II NSCLC, radiation therapy alone is considered only when surgical resection is not possible.[2]  Stereotactic radiation is a reasonable option for lung cancer treatment among those who are not candidates for surgery.[3] Beta blockers have been found to improve overall survival, disease-free survival, and distant metastasis–free survival, though not locoregional progession–free survival, in patients with NSCLC undergoing radiotherapy.[4]

See Treatment for more detail.

Background

Lung cancers are generally divided into two main categories: small cell lung cancer (SCLC) and non–small cell lung cancer (NSCLC). NSCLC accounts for approximately 85% of all lung cancers. Histologically, NSCLC is divided further into adenocarcinoma, squamous cell carcinoma (SCC), and large cell carcinoma. (See Pathophysiology.)

Lung cancer was a rare entity in the early 1900s but has since become far more prevalent. The prevalence of lung cancer is second only to that of prostate cancer in men and breast cancer in women. By the end of the 1900s, lung cancer had become the leading cause of preventable death in the United States,[5] and recently, it surpassed heart disease as the leading cause of smoking-related mortality.

Lung cancer is the leading cause of cancer-related mortality in both men and women not only in the United States but also throughout the world. In 2016, the disease is expected to cause approximately 158,000 deaths in the United States—more than colorectal, breast, and prostate cancers combined.[6] The types of lung cancer in the United States, as well as in many other countries, have also changed in the past few decades: the frequency of adenocarcinoma has risen, and that of SCC has declined. (See Epidemiology.)

Most lung carcinomas are diagnosed at an advanced stage, conferring a poor prognosis. The need to diagnose lung cancer at an early and potentially curable stage is thus obvious. (See Prognosis.) In addition, most patients who develop lung cancer have been smokers and have smoking-related damage to the heart and lungs, making aggressive surgical or multimodality therapies less viable options.

Lung cancer is often insidious, producing no symptoms until the disease is well advanced. In approximately 7-10% of cases, lung cancers are diagnosed incidentally in asymptomatic patients, when a chest radiograph performed for other reasons reveals the disease. Numerous pulmonary signs may be associated with NSCLC. Systemic findings may include unexplained weight loss and low-grade fever. (See Presentation.)

Because of the importance of stage on the therapeutic decision-making process, all patients with NSCLC must be staged adequately. A complete staging workup for NSCLC should be carried out to evaluate the extent of disease. (See Workup.)

Treatment primarily involves surgery, chemotherapy, or radiation therapy. Because most lung cancers cannot be cured with currently available therapeutic modalities, the appropriate application of skilled palliative care is an important part of the treatment of patients with NSCLC. (See Treatment.)

Go to Small Cell Lung Cancer for complete information on this topic. 

Pathophysiology

Both exposure (environmental or occupational) to particular agents and an individual’s susceptibility to these agents are thought to contribute to one’s risk of developing lung cancer. In the United States, active smoking is responsible for approximately 90% of lung cancer cases. Occupational exposures to carcinogens account for approximately 9-15% of lung cancer cases.

Exposure to carcinogens

Tobacco smoke contains more than 300 harmful substances with at least 40 known potent carcinogens. Polyaromatic hydrocarbons and nicotine-derived nitrosamine ketone (NNK) are known to cause DNA damage by forming DNA adducts in animal models. Benzo-A-pyrine also appears to induce molecular signaling such as AKT, as well as inducing mutations in p53 and other tumor suppressor genes.

The most common occupational risk factor for lung cancer is exposure to asbestos. Studies have shown radon exposure to be associated with 10% of lung cancer cases, while outdoor air pollution accounts for perhaps 1-2%.[7] In addition, preexisting nonmalignant lung diseases, such as chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, and tuberculosis have all been shown to be associated with increased lung cancer rates.

The current multiple hit theory suggests that a series of toxic cellular insults disrupts orderly genetic reproduction. Symptoms ultimately develop from the uncontrolled disorganized growth that interferes with local or distant anatomy or physiologic processes.[7]

A study by Ito et al assessed the shift in histologic types of lung cancer in Japan and the United States in relation to the shift from nonfiltered to filtered cigarettes.[8] The study determined that the shift in cigarette types only altered the most frequent type of lung cancer, which shifted from SCC to adenocarcinoma.

Genetic susceptibility

Advanced molecular techniques have identified amplification of oncogenes and inactivation of tumor suppressor genes in NSCLC. The most important abnormalities detected are mutations involving the ras family of oncogenes. The ras oncogene family has 3 members: H-ras, K-ras, and N-ras. These genes encode a protein on the inner surface of the cell membrane with guanosine triphosphatase activity and may be involved in signal transduction.

Studies performed on mice suggest the involvement of ras mutations in the molecular pathogenesis of NSCLC. Studies in humans suggest that ras activation contributes to tumor progression in persons with lung cancer. The ras gene mutations occur almost exclusively in adenocarcinoma and are found in 30% of such cases. These mutations were not identified in adenocarcinomas that developed in persons who do not smoke. The K-ras mutation appears to be an independent prognostic factor.

Studies are ongoing to develop management plans according to the presence or absence of ras gene mutations.

Other molecular abnormalities found in NSCLC include mutations in the oncogenes c-myc and c-raf and in the tumor suppressor genes retinoblastoma (Rb) and p53.

Two studies have documented early and extensive mutations in lung cancers that result in pronounced intratumor heterogeneity by the time these cancers manifest clinically—thus helping to explain why these cases so often fail to respond to treatment. A study by Zhang and colleagues identified 20 of 21 known cancer gene mutations in all regions of 11 localized lung adenocarcinomas. On follow-up, patients who had postsurgical relapse had significantly larger fractions of subclonal mutations in their primary tumors.[9]

Similarly, a study by de Bruin and colleagues in seven operable NSCLCs determined that there was a long period of tumor latency between early mutations and clinical symptoms, which appeared after new mutations triggered rapid disease growth. In some former smokers, the initial mutations dated back to when they were smoking cigarettes, two decades earlier. Over time, however, those mutations became less important, with more recent mutations resulting from a new process controlled by a protein called APOBEC.[10]

Classification of lung cancer

Lung cancers are generally divided into 2 main categories: SCLC and NSCLC. NSCLC accounts for approximately 85% of all lung cancers. NSCLC is divided further into adenocarcinoma, SCC, and large cell carcinoma. All share similar treatment approaches and prognoses but have distinct histologic and clinical characteristics.

Adenocarcinoma

Adenocarcinoma, arising from the bronchial mucosal glands, is the most common NSCLC cancer in the United States, representing 35-40% of all lung cancers. It is the subtype observed most commonly in persons who do not smoke. It usually occurs in a peripheral location within the lung, in some cases at the site of pre-existing scars, wounds, or inflammation (ie, a “scar carcinoma”).

Bronchoalveolar carcinoma is a distinct subtype of adenocarcinoma with a classic manifestation as an interstitial lung disease on chest radiograph. Bronchoalveolar carcinoma arises from type II pneumocytes and grows along alveolar septa. This subtype may manifest as a solitary peripheral nodule, multifocal disease, or a rapidly progressing pneumonic form. A characteristic finding in persons with advanced disease is voluminous watery sputum.

Squamous cell carcinoma

SCC accounts for 25-30% of all lung cancers. Whereas adenocarcinoma tumors are peripheral in origin, SCC is found in the central parts of the lung (see the image below). The classic manifestation is a cavitary lesion in a proximal bronchus. This type is characterized histologically by the presence of keratin pearls and can be detected with cytologic studies because it has a tendency to exfoliate. It is the type most often associated with hypercalcemia.



View Image

Non–small cell lung cancer. A cavitating right lower lobe squamous cell carcinoma.

Large-cell carcinoma

Large-cell carcinoma accounts for 10-15% of lung cancers, typically manifesting as a large peripheral mass on chest radiograph. Histologically, this type has sheets of highly atypical cells with focal necrosis, with no evidence of keratinization (as is typical of SCC) or gland formation (as is typical of adenocarcinomas).

With improved histopathologic procedures and the use of electron microscopy, most NSCLCs that would previously have been classified as large-cell carcinomas are identified as undifferentiated adenocarcinomas or, less frequently, as SCCs.[11] Large-cell undifferentiated cancers have the same prognosis as do adenocarcinomas and are combined with them in clinical trials.

Etiology

Causes of lung cancer include the following:

Unlike many other malignancies, whose causes are largely unknown, lung cancer is known to be caused by tobacco smoking in as many as 90% of patients. However, two recent studies have reported rising NSCLC rates in persons who have never smoked: In a United States study, rates increased from 8.9% in 1990–1995 to 19.5% in 2011–2013, while a study from the United Kingdom reported an increase from 13% to 28% during a 6-year period.[12]

Because not all smokers develop lung cancer and not all lung cancer patients have a history of smoking, other factors (eg, genetic susceptibility [see Pathophysiology], arsenic exposure, radiation exposure, and other environmental carcinogens[13] ) also play a causative role, either independently or in conjunction with smoking. Genetic factors probably contribute in all populations, but the contribution of other factors is population-specific.

A study by Bagnardi et al determined that alcohol is not an independent factor in the etiology of lung cancer.[14]

Smoking

Smoking prevalence in the United States has gradually declined over last 4 decades. In 2012, there were an estimated 42.1 million active smokers in the United States. Overall smoking prevalence declined from 20.9% in 2005 to 18.1% in 2012.[15] Worldwide, the incidence of smoking in developing countries is on the rise, with almost 320 million smokers in China alone.

The development of lung cancer is directly related to number of cigarettes smoked, length of smoking history, and the tar and nicotine content of the cigarettes. Risk is highest among current smokers and lowest among nonsmokers. A large trial showed that persistent smokers had a 16-fold elevated lung cancer risk, which was further doubled in those who started smoking when younger than 16 years.[16] The age-adjusted incidence rates range from 4.8-20.8 per 100,000 among nonsmokers to 140-362 per 100,000 among active smokers.

Although tobacco smoking is the major cause of lung cancer, it is now believed that males and females may differ in their susceptibility to the carcinogenic effects of tobacco smoke. This difference may be due to differences in DNA repair mechanisms. Although still considered controversial, it is well known that women are more likely to develop adenocarcinomas and that, stage for stage, women live longer. In addition, differences in response to certain biologic therapies (eg, epidermal growth factor [EGF] inhibitors) and antiangiogenic agents have been observed between sexes.

The risk of lung cancer declines slowly after smoking cessation. Long-term follow-up studies show that the relative risk remains high in the first 10 years after cessation and gradually declines to 2-fold approximately 30 years after cessation. This long-term risk explains the development of almost 50% of United States lung cancer cases in past smokers.

Strong cardiorespiratory fitness might help reduce lung cancer risk in men who smoke or used to smoke, accordng to the findings from a study that assessed 1602 former smokers (40 pack-years) and 1377 current smokers (43 pack-years). All were men, aged 42 to 76 years, who were free from lung cancer at baseline. Over a follow-up period of 4.6 to 18.6 years, 46 former smokers and 53 current smokers developed lung cancer. Of this group, 40 former smokers and 39 current smokers died. Men who had higher fitness levels at baseline, measured with a maximal treadmill exercise test, had a lower incidence of lung cancer during follow-up and had better survival if they did get lung cancer.[17]

Secondhand smoking

Cigarette smoke containing the carcinogenic N-nitrosamines and aromatic polycyclic hydrocarbons can be inhaled passively by nonsmokers (secondhand smoke); urinary levels of these carcinogens in nonsmokers are 1-5% of those found in active smokers. As many as 25% of the lung cancers in persons who do not smoke are believed to be caused by secondhand smoke.[18]

The US Environmental Protection Agency has recognized passive smoking as a potential carcinogen. About 3000 cases of lung cancer appear to be related to passive exposure. This awareness has led to local ordinances restricting smoking in enclosed public places, including restaurants and government buildings.

Lung cancer in never-smokers

A minority of lung cancers develop in persons who have never smoked. These lung cancers are genetically distinct from smoking-related NSCLC, and this distinction may have therapeutic implications. The observed genetic differences include a lower frequency of K-ras and a higher frequency of mutations in the EGF receptor and likely are responsible for the higher efficacy of EGF receptor inhibitors in this patient population.

Asbestos exposure

The silicate type of asbestos fiber is an important carcinogen. Asbestos exposure has been shown to be strongly associated with the causation of lung cancer, malignant pleural mesothelioma, and pulmonary fibrosis. Asbestos exposure increases the risk of developing lung cancer by as much as 5 times.

Tobacco smoke and asbestos exposure act synergistically, and the risk of developing lung cancer for persons who currently smoke tobacco and have a history of asbestos exposure approaches 80-90 times that of control populations.

Radon exposure

Radon is an inert gas produced as a result of uranium decay. Radon exposure is a well-established risk factor for lung cancer in uranium miners. Approximately 2-3% of lung cancers annually are estimated to be caused by radon exposure. Household exposure to radon, however, has never been clearly shown to cause lung cancer.

The US National Research Council’s report of the Sixth Committee on Biological Effects of Ionizing Radiation has estimated that radon exposure causes 2100 new lung cancers each year, while it contributes to lung cancer causation in approximately 9100 persons who smoke.

HIV infection

Persons with HIV infection have a higher lung cancer risk than those without HIV infection, with relative risk estimates ranging from 2 to 11. In persons with HIV infection, lung cancer is the most common and most fatal non-AIDS-associated malignancy, accounting for about 16% of deaths.[19] A majority of these cases are adenocarcinomas. In most, but not all, studies the incidence and risk of lung cancer in HIV-infected persons did not change significantly with the advent of highly active antiretroviral therapy.[20]

Lung cancer in HIV-infected persons develops almost exclusively in smokers, but HIV infection appears to increase lung cancer risk independent of smoking status, by a factor of at least 2.5-fold. Compared with lung cancer patients in the general population, HIV-infected patients with lung cancer are significantly younger. Most patients with HIV infection and lung cancer present with advanced-stage disease and have significantly shorter median survival.[19]

Other environmental agents

Beryllium, nickel, copper, chromium, and cadmium have all been implicated in causing lung cancer.

Dietary fiber and vegetables have been suggested as protective from lung cancer. Although diets rich in fruits and vegetables appear to be associated with lower rates of lung cancer, trials of supplemental beta-carotene, alone or in combination with vitamin E or retinyl palmitate, in persons at high risk for lung cancer found that this supplementation actually increased the incidence of lung cancers.[21]

Epidemiology

United States statistics

In the United States, lung cancer is the second most common cancer, after prostate cancer in men and breast cancer in women, but the most common cause of cancer deaths. The American Cancer Society projects that 228,820 cancers of the lung and bronchus will be diagnosed in the United States in 2020, with 135,720 deaths.[6] Approximately 85% of those cases are expected to be NSCLC.

In US men, the incidence of lung cancer has been decreasing since the mid-1980s. In US women, however, the rate has been decreasing only since the mid-2000s. From 2004 to 2013, the incidence of lung cancer decreased by 2% per year in men and by 1% per year in women.[6]

Lung cancer death rates for US women are among the highest in the world. Although in the United States, death rates are higher in men than in women, rates for US men are still lower than rates for men in several other countries.[22] These trends in US death rates parallel trends in smoking prevalence over the past 50 years.[6]

However, lung cancer death rates in the US have been decreasing at an accelerated rate. In men, lung cancer mortality showed a 3% annual decline in 2008-2013, then a 5% decline in 2013-2017. In women, annual declines in those periods were 2% to almost 4%: from 2016 to 2017, mortality decreased by 2.2.%, the largest ever single‐year drop in overall cancer mortality.[6]

International statistics

Lung cancer is the most commonly diagnosed cancer worldwide, and its incidence continues to grow. In 2018, an estimated 2.1 million new cases of lung cancer were diagnosed globally, accounting for approximately 11.6% of the global cancer burden. An estimated 1.76 million lung cancer deaths occurred in 2018.[23] Among all cancers, lung cancer now has the highest mortality rate in most countries, with industrialized regions such as North America and Europe having the highest rates.

Several differences exist in lung cancer incidence according to geographic area. The highest incidence occurs in Polynesia (38.1 cases per 100,000 population per year). The lowest incidence rate is in western Africa (approximately 1.7 cases per 100,000 population per year).[23] With increased smoking in developing countries, the incidence is expected to increase in the next few years, notably in China and India.

Generally, global lung cancer trends have followed the trends in smoking, with a lag time of several decades. Lung cancer incidence has been declining in several countries, including the United States, Canada, the United Kingdom, and Australia, following the decreasing rate of smoking. Lung cancer incidence among women, however, continues to increase in several parts of the globe, although it has begun to plateau in the United States. Notably, despite a very low rate of smoking, Chinese females have a higher incidence of lung cancer than European females.

Age and sex distribution

Lung cancer occurs predominately in persons aged 50-70 years. The probability of developing lung cancer remains very low until age 39 years in both sexes. It then slowly starts to rise and peaks among those older than 70 years. The risk of developing lung cancer remains higher among men in all age groups after age 40 years.

Overall, lung cancer is more common in men than in women. In the United States, Northern Europe, and Western Europe, the prevalence of lung cancer has been decreasing in men. In Eastern and Southern European countries, the incidence of lung cancer has been rapidly increasing. Most Western countries have encountered a disturbing trend of increasing prevalence in women and younger patients. Women have a higher incidence of localized disease at presentation and of adenocarcinoma and typically are younger when they present with symptoms.

Over the past two decades, the incidence of lung cancer has generally decreased in both men and women 30 to 54 years of age in all races and ethnic groups. However, the incidence has declined more steeply in men. As a result, lung cancer rates in younger women have become higher than those in younger men. In non-Hispanic whites and Hispanics ages 44 to 49 years, for example, the female-to-male rate ratio for lung cancer incidence rose from 0.88 during 1995-1999 to 1.17 during 2010-2014.[24]

This reversal can be explained in part by increased rates of cigarette smoking in women born since 1965. However, while the difference in smoking rates in that age group has narrowed, rates in women have generally not exceeded the rates in men, so other factors may be playing a role. For example, women may be more susceptible to the oncogenic effects of smoking.[24]

Incidence and survival by race

Whereas lung cancer incidence rates are similar among African-American and white women, lung cancer occurrence is approximately 45% higher in African-American men than in white men.[22] This increased incidence has been attributed to differences in smoking habits; however, recent evidence suggests a slight difference in susceptibility.

From 1995-2001, the 5-year relative survival rate was 13% lower in African Americans compared with white individuals.[22] This racial gap persisted within each stage at diagnosis for both men and women.

Trends in 5-year survival rates in lung cancer from 1975-2003 revealed that while modest gains occurred in 5-year survival rates among whites, survival rates remained unchanged in the African-American population. Current 5-year survival rates are estimated to be 16% among whites and 13% among non-whites.

Prognosis

Lung cancer is highly lethal. In Europe, the 5-year overall survival rate is 12.3%. The highest recorded 5-year patient survival rates are observed in the United States. US data collected from 2009–2015 indicate that the 5-year relative survival rate for lung cancer was 19.4%, reflecting a steady but slow improvement from 12.5% in 1975.[22, 25] However, the 5-year relative survival rate varies markedly, depending on how advanced the disease is at diagnosis, as follows[25] :

Prognostic factors for NSCLC are summarized in the image below.



View Image

Non–small cell lung cancer. Prognostic factors for lung cancer.

A retrospective Surveillance, Epidemiology, and End Results (SEER) data analysis suggests that the number of nodes with cancer may be predictive of survival. Mean lung cancer-specific survival decreased from 8.8 years for patients with one positive lymph node to 3.9 years for patients with more than eight positive lymph nodes.[26]

Patients with in situ and stage I lung cancer may respond to surgery. Their prognosis is far better than that of patients with more advanced disease. In patients with radiologically occult lung neoplasms, the 5-year survival rate is 24-26%; in those with abnormal chest radiographic findings, the rate is 12%. If the cancer is nonresectable, the prognosis is poor, with a mean survival rate of 8-14 months.

Mostertz et al found that in some patient populations, the oncogenic pathway activation profile of the tumor can have prognostic significance.[27] Retrospective analysis of 787 patients with predominantly early-stage NSCLC, using gene expression profiling, showed the following:

A meta-analysis by Parsons et al suggests that smoking cessation after diagnosis of early-stage lung cancer may improve prognosis, probably by reducing cancer progression. Life table modelling on the basis of data from 9 studies gave an estimated 5-year survival rate of 33% in 65-year-old patients with early-stage NSCLC who continued to smoke compared with 70% in those who quit smoking.[28]

In an analysis of data on 4200 patients who participated in the National Comprehensive Cancer Network's NSCLC Database Project, patients who were current smokers at the time of diagnosis had worse survival compared with patients who never smoked, and among younger patients with stage IV disease, current smokers had worse survival compared with former smokers who quit smoking more than 12 months before being diagnosed.[29]

Secondary analyses of the Women’s Health Initiative (WHI) randomized, placebo-controlled trial demonstrated an association between the use of daily conjugated equine estrogen (CEE, 0.625 mg) plus medroxyprogesterone acetate (MPA, 2.5 mg) and NSCLC. Women who used CEE plus MPA for more than 5 years were at increased risk for NSCLC, and women using CEE plus MPA who were diagnosed with NSCLC had higher mortality than women with NSCLC who do not take hormone therapy.[30]

The WHI analyses included 16,608 multiethnic postmenopausal women aged 50-79 years. Confirmation of lung cancers was completed by medical record review. This area deserves more attention and study to determine the risks and benefits of hormone therapy for postmenopausal women who smoke.

In contrast, a study by Bouchardy et al found that patients who had received antiestrogen treatment for breast cancer had a lower lung cancer mortality rate. However, use of antiestrogens did not significantly lower standardized incidence ratios for lung cancer.[31]

A review of eight trials by Rothwell et al found that allocation to daily aspirin reduced death caused by a variety of cancers, including adenocarcinoma of the lung (but no other form of lung cancer). A latent period of 5 years was observed before risk of death was decreased for lung cancer, but 20-year risk of cancer death remained lower in the aspirin groups. Benefit was unrelated to aspirin dose (75 mg or higher), sex, or smoking, but increased with age, with the absolute reduction in 20-year risk of cancer death reaching 7.08% at age 65 years and older.[32]

Although tumor-node-metastasis (TNM) staging is the best prognostic factor for NSCLC, a study by Hofman et al concluded that preoperative detection of circulating tumor cells (CTCs) has prognostic significance.[33] The results showed that the presence and level of 50 or more circulating nonhematologic cells (CNHC) were associated with worse survival among patients with resectable NSCLC. Although CTCs are potentially interesting, the significance of their presence is still being debated.[34]

Risk of recurrence

In a 2012 retrospective review of 1402 consecutive stage I-III (N0-N1) NSCLC patients who underwent complete resection without adjuvant radiation therapy, significant risk factors for local recurrence included surgical procedure (single/multiple wedges + segmentectomy versus lobectomy + bilobectomy + pneumonectomy), visceral pleural invasion, and tumor size >2.7 cm. Significant risk factors for regional recurrence included pathologic N1 stage, lymphovascular space invasion, and visceral pleural invasion.[35]

In a study of 452 cases of stage I lung adenocarcinoma, thyroid transcription factor–1 (TTF-1) expression independently predicted the risk of disease recurrence. The 5-year cumulative incidence of recurrence was 40% for patients with negative TTF-1 expression, versus 15% for those with positive TTF-1 expression (P < 0.001.[36]

According to a 2013 retrospective analysis of 734 patients with stage I adenocarcinoma no larger than 2 cm, recurrence of small, early-stage adenocarcinoma after limited lung resection is three times more likely when the micropapillary component of the tumor is 5% or greater. In the 258 study patients who underwent wedge resection or segmentectomy, after adjustment for both vascular and lymphatic invasion, the presence of a micropapillary component of 5% or greater was independently associated with a 5-year cumulative incidence of recurrence (hazard ratio = 3.11). Micropapillary status was not significantly associated with recurrence in the 476 patients who underwent lobectomy.[37, 38]

Patient Education

Advise patients that smoking cessation is the most important measure for preventing lung cancer; it may also improve prognosis in patients with early-stage lung cancer.[28] Smoking cessation by others who share the patient’s home, car, or both is also important. According to published data, the use of nicotine alternatives (eg, gum, patch, spray) instead of cigarettes reduces the incidence of lung cancer, although it does not affect the incidence of ischemic heart disease.

Advise the patient to avoid asbestos exposure. Consider prophylactic administration of retinoids, such as beta-carotene.

For patient education information, see the Cancer and Tumors Center, as well as Lung Cancer, Bronchoscopy, and Understanding Lung Cancer Medications.

History

Lung cancer is often insidious, producing no symptoms until the disease is well advanced. In approximately 7-10% of cases, lung cancer is diagnosed in asymptomatic patients when a chest radiograph performed for other reasons reveals the disease. At initial diagnosis, 20% of patients have localized disease, 25% of patients have regional metastasis, and 55% of patients have distant spread of disease.

Signs and symptoms of lung cancers may be due to the primary tumor, locoregional spread, metastatic disease, or ectopic hormone production (see the image below). Cough is reported to be the most common presenting symptom of lung cancer. Other respiratory symptoms include dyspnea, chest pain, and hemoptysis. Hemoptysis has been described as the one symptom often prompting more rapid presentation.[39]



View Image

Non–small cell lung cancer. Symptoms and signs of lung cancer.

Symptoms due to primary tumor

The symptoms produced by the primary tumor depend on its location (ie, central, peripheral).[1] Central tumors are generally squamous cell carcinomas (SCCs) and produce cough, dyspnea, atelectasis, postobstructive pneumonia, wheezing, and hemoptysis.

Most peripheral tumors are adenocarcinomas or large cell carcinomas and, in addition to causing cough and dyspnea, can cause symptoms due to pleural effusion, and severe pain as a result of infiltration of parietal pleura and the chest wall. Because of their peripheral location, adenocarcinomas may not call attention to themselves until they have produced extrathoracic metastases. For example, patients may present with clinical signs of bone spread or intracranial metastatic disease.

Symptoms due to locoregional spread

Symptoms due to locoregional spread can include superior vena cava obstruction, paralysis of the recurrent laryngeal nerve, and phrenic nerve palsy, causing hoarseness and paralysis of the diaphragm; pressure on the sympathetic plexus, causing Horner syndrome; dysphagia resulting from esophageal compression; and pericardial effusion.

Superior sulcus tumors (Pancoast tumors) can cause compression of the brachial plexus roots as they exit the neural foramina, resulting in intense, radiating neuropathic pain in the ipsilateral upper extremity.

Symptoms by location of cancer

Endobronchial cancers may produce the following signs:

Mediastinal cancers may produce the following signs and symptoms:

Pleural cancers may produce the following signs and symptoms:

Neurologic signs and symptoms include the following:

Metastatic cancer may produce the following signs (8-68%):

Central nervous system (CNS) signs and symptoms include the following:

Vascular signs include the following:

Musculoskeletal manifestations include the following:

Paraneoplastic syndromes occur in 10-20% of patients. Most paraneoplastic syndromes are caused by small cell lung cancer (SCLC). However many paraneoplastic syndromes also occur in non–small cell lung cancer (NSCLC) patients. Some examples include:

Physical Examination

Numerous signs may be associated with NSCLC (see the image below). Extrapulmonary findings may include adenopathy and clubbing. Systemic findings may include unexplained weight loss and low-grade fever.



View Image

Non–small cell lung cancer. Symptoms and signs of lung cancer.

In approximately two thirds to three fourths of patients, the cancer is not diagnosed until it has reached an advanced stage; patients may have lost weight and may have obvious respiratory distress. Subtle findings on physical examination may provide clues for early detection.

About one third of patients present with symptoms as a result of distant metastases. The most common sites of distant metastasis from lung cancer are as follows:

Lung cancer can metastasize to virtually any bone, although the axial skeleton and proximal long bones are most commonly involved.

Head and neck

Commonly, no signs are found upon examination of the head and neck regions. However, when the cancer has spread to the supraclavicular lymph nodes, careful examination may reveal enlargement of involved nodes, which helps in the clinical staging process.

Superior sulcus tumors (Pancoast tumors), because of their presence at the apex of the lung, can compress the cervical sympathetic plexus, causing classic Horner syndrome. Findings involve ipsilateral ptosis, miosis, enophthalmos, and anhidrosis (ie, lack of sweating).

The superior vena cava syndrome (SVCS) results from obstruction of blood flow to the heart from the head and neck regions and upper extremities as a consequence of compression of the superior vena cava, either from direct invasion by the primary tumor into the mediastinum or from lymphatic spread with enlarged right paratracheal lymph nodes. It is commonly caused by SCLC but can result from any centrally located tumor or mediastinal spread.

Signs of SVCS include the following:

Respiratory system

Findings are variable and depend on tumor location and spread. Centrally located obstructing tumors can cause collapse of the entire lung with an absence of breath sounds on the side of the lesion. Peripheral lesions can cause individual segments or lobes to collapse, leading to findings of dullness to percussion and/or decreased breath sounds.

Pleural effusions give rise to characteristic findings of dullness and decreased breath sounds, depending on the size.

Respiratory insufficiency is signaled by dyspnea and increased work of breathing, retractions, orthopnea, and cyanosis. Upper airway obstruction is manifested by stridor and wheezing. Lower airway obstruction is manifested by asymmetric breath sounds, pleural effusion, pneumothorax, infiltrate, and post obstructive processes.

Cardiovascular system

Cardiac findings are usually noted when the tumor causes a pericardial effusion. Findings can range from simple effusion to tamponade. Direct cardiac involvement may also occur.

Gastrointestinal tract

The most common site of metastatic spread is the liver, which may manifest as tender hepatomegaly. In addition, Ogilvie intestinal pseudo-obstruction may occur, as manifested by the following:

Musculoskeletal system

Bone is another common site of spread for lung carcinomas. Patients may report bone pain, and tender spots may be found during examination.The examination should include fist percussion of the spine to look for tender spots, which may suggest vertebral column metastases.

A Pancoast tumor may give rise to any of the following:

Central nervous system

A neurologic examination should be performed to assess for focal neurological deficits caused by brain metastases and for signs of spinal cord compression. The skeletal system is a common site of spread of lung cancer, and metastatic lesions in the spine may grow and compress the spinal cord. Patients usually report back pain and neurological symptoms in the form of decreased sensation in the lower half of the body, decreased strength, loss of bowel control, and urinary incontinence or retention. A careful neurologic examination usually localizes the level of compression.

Suspected spinal cord compression is an emergency. Patients should immediately receive an adequate dose of a corticosteroid (usually intravenous dexamethasone, 10 mg followed by 4 mg q6h) and should undergo an immediate MRI scan of the vertebral column. If documented, spinal cord compression should be treated emergently with radiation therapy, and steroids should be tapered slowly.

A study by Fadel et al found that en bloc resection can achieve good long-term survival in highly selected patients with NSCLC that invades the thoracic inlet and spine. Factors that independently affected survival were incomplete resection and subclavian artery involvement.[41]

Paraneoplastic syndromes

Paraneoplastic syndromes are more likely to occur in small cell lung cancer than in NSCLC. Nevertheless, findings indicative of the following paraneoplastic syndromes may be noted[42] :

Paraneoplastic syndromes are unrelated to the size of the primary tumor. Sometimes paraneoplastic syndromes precede the diagnosis of a malignancy, sometimes they occur late in the disease, and sometimes they are the first signs of a recurrence.

Approach Considerations

In a patient with a long history of smoking or other risk factors for lung cancer, the presence of persistent respiratory symptoms should prompt a chest radiograph. Because benign conditions and metastatic malignancies can mimic lung cancer on radiographs, histologic confirmation is necessary. This can be achieved by sputum cytologic studies, bronchoscopy, or computed tomography (CT)-guided transthoracic needle biopsy of the mass, depending on the location of the tumor (see the image below).



View Image

Non–small cell lung cancer. Diagnostic approach for possible lung cancer.

Staging workup

Because of the importance of stage in the therapeutic decision-making process, all patients with non–small cell lung cancer (NSCLC) must be staged adequately. A complete staging workup for NSCLC should be carried out to evaluate the extent of disease. In the United States, the standard staging workup for NSCLC includes 7 main components (see the table below).



View Image

Staging workup for non–small cell lung cancer.

Information obtained from these tests can then be used to guide further testing (eg, imaging studies). Invasive staging procedures such as mediastinoscopy and mediastinotomy may be required to assess mediastinal lymph nodes in patients who are candidates for potentially curative surgical resection. Positron emission tomography (PET) scans may be useful in the detection of involved nodes, the presence of which may influence decisions about operability.

A study by Annema et al determined that among patients with suspected NSCLC, a combination of endosonography and surgical staging had a greater sensitivity for mediastinal nodal metastases than surgical staging alone.[44] This resulted in fewer unnecessary thoracotomies.

See Non-Small Cell Lung Cancer Staging for summary tables.

See Lung Cancer Staging -- Radiologic Options, a Critical Images slideshow, to help identify stages of the disease process.

Other procedures

An electrocardiogram (ECG) is helpful in establishing baseline findings and differentiating clinical symptoms (eg, chest pain, dyspnea). Changing lung hemodynamics often alter ECG wave patterns.

Bedside tests for peak expiratory flow provide good indicators of significant airflow obstruction. Lung cancer is more closely linked to chronic obstructive pulmonary disease with airflow compromise than to the disease without significant airway obstruction.

Laboratory Studies

For staging purposes, a complete blood count (CBC) should be obtained in every patient, especially before instituting chemotherapy. In an emergency setting, a CBC is not helpful in the initial evaluation. Obtain a CBC in patients with widely metastatic disease to aid in determining whether an infiltrate is potentially infectious. Obtain a CBC in patients with fever who have a recent history of chemotherapy to check for neutropenia (absolute neutrophil count < 1000/μL).

Lung cancers have a propensity to cause paraneoplastic syndromes (see the table below). Appropriate studies in such patients may include assays of serum electrolytes, blood urea nitrogen (BUN), creatinine, calcium, and magnesium.



View Image

Non–small cell lung cancer. Symptoms and signs of lung cancer.

The most common metabolic abnormality associated with NSCLC is hypercalcemia, which usually occurs with squamous cell carcinoma and results from secretion of parathyroid hormone–related peptide (PTH-rP) by the tumor. This can be distinguished from hyperparathyroidism by confirmation of normal serum parathyroid hormone (PTH) levels.

Other electrolyte abnormalities can include hyponatremia, in which case the syndrome of inappropriate antidiuretic hormone secretion (SIADH) should be considered. The combination of hyponatremia, serum osmolality < 280 mOsm/kg, and high urine osmolality is the hallmark of SIADH.

Liver function tests (aspirate aminotransferase [AST], alanine aminotransferase [ALT], gamma-glutamyl transferase [GGT], prothrombin time [PT]/international normalized ratio [INR]) and alkaline phosphatase level are usually not helpful initially. In patients with advanced disease, however, elevated results may be an indication of hepatic metastasis and bone metastasis, respectively.

Arterial blood gas (ABG) levels are useful in the detection of respiratory failure (eg, acidosis, hypercarbia, hypoxia) in sick patients. Obtain ABG levels in patients with active systemic diseases or abnormal labored breathing.

Chest Radiography

A chest radiograph is usually the first test ordered in patients in whom a lung malignancy is suggested. Clues from the chest radiograph may suggest the diagnosis of lung cancer, but may not be helpful in identifying a histologic subtype. If the tumor is clearly visible and measurable, chest radiography can sometimes be used to monitor response to therapy.

Chest radiographs may show the following:



View Image

Non–small cell lung cancer. Left pleural effusion and volume loss secondary to non–small cell carcinoma of the left lower lobe. The pleural effusion w....

Popcorn calcification is usually a radiologic characteristic of benign lesions.

The percentage of patients found to have lung cancer incidentally through chest radiographs has been consistently low. Randomized controlled trials have shown that the use of screening chest radiographs does not reduce lung cancer mortality.[45, 46]

Go to Imaging in Non-Small Cell Lung Cancer for complete information on this topic.

Computed Tomography

A chest CT scan (see the image below) is the standard for staging. The findings of CT scans of the chest and clinical presentation usually allow a presumptive differentiation between NSCLC and small cell lung cancer (SCLC). Massive lymphadenopathy and direct mediastinal invasion are commonly associated with small cell carcinoma. A mass in or adjacent to the hilum is a particular characteristic of SCLC and is seen in about 78% of cases.[47]



View Image

Lung cancer, small cell. Contrast-enhanced CT scan of the chest shows a large left lung and a hilar mass, with invasion of the left pulmonary artery.

Common sites of spread of NSCLC include the liver and adrenals; hence, CT scanning of the chest and upper abdomen that includes the liver and adrenals is the minimum standard for a staging workup for a person newly diagnosed with NSCLC. Lung nodules incidentally detected on abdominal CT are often benign.[48]

A CT scan or magnetic resonance imaging (MRI) scan of the brain may be required if neurologic symptoms or signs (eg, mental status change) are present. Most thoracic surgeons perform imaging of the brain before attempting definitive resection of a lung malignancy.

Go to Imaging in Non-Small Cell Lung Cancer for complete information on this topic.

Magnetic Resonance Imaging

MRI is most useful when evaluating a patient in whom spinal cord compression is suggested. In addition, brain MRI has a greater sensitivity than CT scan for detection of central nervous system (CNS) metastasis. MRI may be used when findings of superior sulcus and brachial plexus tumors are equivocal on CT scans.

Go to Imaging in Non-Small Cell Lung Cancer for complete information on this topic.

Bone Scintigraphy

The skeletal system is common site of metastases for lung cancers. If patients report bone pain or if their serum calcium and/or alkaline phosphatase levels are elevated, a bone scan should be obtained to search for bone metastases (see the image below).



View Image

Lung cancer, small cell. Whole-body nuclear medicine bone scanning with anterior and posterior images reveal multiple abnormal areas of increased radi....

Positron Emission Tomography

PET scanning (see the image below) using fluoro-18–2-deoxyglucose (FDG) has proven to be an excellent modality for evaluating solitary pulmonary nodules and has been approved by the US Food and Drug Administration (FDA) for this indication. The average sensitivity and specificity of FDG-PET scanning for detecting a malignancy was reported to be 0.97 and 0.78, respectively.[49] However, a meta-analysis by Deppen and colleagues found that FDG-PET had lower specificity for diagnosing malignancies in areas with endemic infectious lung disease compared with areas with nonendemic disease.[50]



View Image

Lung cancer, small cell. Coronal positron emission tomogram shows abnormal areas of increased metabolic activity in the left hilar and left adrenal re....

Studies also suggest that PET scanning is useful for searching for systemic spread if other diagnostic modalities cannot clarify an abnormality that may change the treatment of the patient’s condition. However, false-positive and false-negative results occur.

Additional data have emerged that underscore the importance of PET scanning in patients with NSCLC. PET scans appear to be more sensitive, specific, and accurate than CT scans for staging mediastinal disease. Whereas radiographs and CT scans show images of structures, PET scans reveal the nature of the area under study. PET scans often detect abnormalities not demonstrated on CT scans.

Published reports suggest that staging of NSCLC may be influenced by PET scan results in up to 60% of the cases and that as many as 25% of cases may be upstaged after PET scanning.

Caution is required when interpreting the results of PET scans in patients who may be denied potentially curative surgical resection based on PET results.

Go to Imaging in Non-Small Cell Lung Cancer for complete information on this topic.

Sputum Cytologic Studies

Centrally located endobronchial tumors may exfoliate malignant cells into sputum. (This location and tendency to exfoliate are most common in squamous cell carcinomas [SCCs].) Therefore, sputum cytology can be a quick and inexpensive diagnostic test if results are positive. The false-positive rate for sputum cytology is 1%, but the false-negative rate is as high as 40%.

Sputum cytology does not provide reliable distinction between different histologic subtypes. Discordant results are often observed between cytologic and histologic findings of specimens obtained from bronchoscopy or transthoracic biopsy.

The diagnostic accuracy of sputum cytology depends on rigorous specimen sampling (at least 3 specimens) and preservation techniques, as well as on the location (central vs peripheral) and size of the tumor.[51] The test detects 71% of central tumors but less than 50% of peripheral tumors; therefore, further testing must always follow a negative result.

Several large studies have not revealed that screening with sputum cytology and chest radiography is cost-effective in early detection. In one small study, a cytologic specimen was used to measure EGFR and KRAS mutations; however, this practice still needs to be validated.[52]

Sputum cytology is suggested for high-risk patients in whom semi-invasive procedures such as bronchoscopy or transthoracic needle aspiration (see below) might pose a higher risk. Currently, however, with the development of advanced x-ray imaging techniques and biopsy procedures, sputum cytology is not commonly employed in the diagnosis of NSCLC.

Bronchoscopy

When a lung cancer is suggested, bronchoscopy provides a means for direct visualization of the tumor, allows determination of the extent of airway obstruction, and allows collection of diagnostic material under direct visualization with direct biopsy of the visualized tumor, bronchial brushings and washing, and transbronchial biopsies.

The decision whether to pursue a diagnostic bronchoscopy for a lesion that is suspected of being lung cancer largely depends on the location of the lesion (central vs peripheral).[53] Bronchoscopy is the study of choice in patients with central tumors, with a combined sensitivity of 88%. The addition of transbronchial needle aspiration with endobronchial ultrasound to obtain cytology or histology samples when there is submucosal tumor spread or peribronchial tumor causing extrinsic compression further increases the sensitivity of bronchoscopy.[54]

Biopsy

Transthoracic needle biopsy, guided by CT or fluoroscopy, is preferred for tumors located in the periphery of the lungs because peripheral tumors may not be accessible through a bronchoscope. A positive finding for cancer is reliable; however, the false-negative rate is high at 26%, and, thus, transthoracic biopsy is generally not useful in ruling out cancer.

Diagnostic material can also be obtained from other abnormal sites (eg, enlarged palpable lymph nodes, liver, pleural or pericardial effusions, accessible bone lesions).

Needle Thoracentesis (Ultrasound Guided)

Needle thoracentesis is both diagnostic and therapeutic in patients presenting with respiratory distress. Thoracentesis has a sensitivity of only 80% with a specificity greater than 90%. In patients suspected of having lung cancer who have an accessible pleural effusion, if the pleural fluid cytology finding is negative (after at least 2 thoracenteses), thoracoscopy is recommended as the next step to aid in diagnosis.

Thoracoscopy and Mediastinoscopy

Thoracoscopy is usually reserved for tumors that remain undiagnosed after bronchoscopy or CT-guided biopsy. Thoracoscopy is also an important tool in the management of malignant pleural effusions.

Video-assisted thoracoscopy (VATS) is a newer modality that may be used to sample small peripheral tumors (less than 2 cm in diameter), pleural tumors, or pleural effusions for diagnostic or staging purposes.[54] It is safe and can provide a definitive diagnosis with a high degree of accuracy and minimal risk to the patient. The reported sensitivity rate ranges between 0.80 and 0.99, the specificity rate ranges between 0.93 and 1, and the negative predictive value ranges between 0.93 and 0.96.[54] Survival with assisted VATS is comparable to complete VATS and can be cost-effective.[55]

Mediastinoscopy

Mediastinoscopy may be used to obtain tissue from cancer that has infiltrated into the mediastinum.[56] It is usually performed to evaluate the status of enlarged mediastinal lymph nodes (seen on CT scan) before attempting definitive surgical resection of lung cancer.

Molecular Testing

Molecular testing forms an important part of the full pathologic evaluation of patients with metastatic non–small cell lung cancer (NSCLC). This includes testing for epidermal growth factor receptor (EGFR) mutation and anaplastic lymphoma kinase (ALK) mutation. Such testing is indicated because very effective, less toxic targeted treatments are available for patients with EGFR and ALK mutations.

According to new international evidence-based guidelines jointly published by the College of American Pathologists (CAP), the International Association for the Study of Lung Cancer (IASLC), and the Association for Molecular Pathology (AMP), all lung cancer patients with adenocarcinomas should be tested for the genetic abnormalities that indicate suitability for treatment with targeted agents, irrespective of clinical variables such as sex, ethnicity, or smoking status.[57]

Clinical trial data demonstrate that patients who are tested for these abnormalities and treated with the appropriate targeted therapy have better outcomes.[57] Targeted agents currently available include epidermal growth factor receptor (EGFR) inhibitors (eg, erlotinib, gefitinib) and anaplastic lymphoma kinase (ALK) inhibitors (ie, crizotinib, ceritinib). Various other targeted agents are in the late stages of development.

In May 2013, the US Food and Drug Administration (FDA) approved of the cobas EGFR Mutation Test, a companion diagnostic for erlotinib.[58] This is the first FDA-approved companion diagnostic that can detect EGFR gene mutations. The mutation test allows physicians to identify patients with NSCLC who are candidates for receiving erlotinib as first-line therapy.

The safety and effectiveness of the cobas EGFR Mutation Test was established with clinical data from the EURTAC study and showed progression-free survival in patients with NSCLC who had specific types of EGFR mutations (exon 19 deletions or exon 21 [L858R] substitution mutations) for 10.4 months when they received erlotinib treatment, compared with 5.4 months for those who received standard therapy.[59]

Testing for Ras mutation can also be done. Ras mutation is a marker for aggressive disease and poor prognosis. New agents and regimens to target this mutation (eg, selumetinib plus docetaxel[60] ) are currently under development, so these patients should be considered for enrollment in a clinical trial.

For more information, see Genetics of Non-Small Cell Lung Cancer.

Histologic Findings

The updated World Health Organization (WHO) classification of lung cancer is widely used. Non–small cell lung cancer (NSCLC) includes squamous cell carcinoma (SCC), adenocarcinoma, and large cell carcinoma. Some lung cancers exhibit two or more histologic patterns. SCC was previously the most common type of NSCLC, but adenocarcinoma appears to be increasing in incidence, especially in women.

SCC has a distinct dose-response relationship to tobacco smoking and usually develops in proximal airways, progressing through stages of squamous metaplasia to carcinoma in situ. Well-differentiated SCCs contain keratin pearls, while poorly differentiated SCCs may stain positive for keratin. Microscopic examination reveals cells with large, irregular nuclei and coarse nuclear chromatin with large nucleoli. Cells are arranged in sheets, and the presence of intercellular bridging is diagnostic.

Histologically, adenocarcinomas form glands and produce mucin. Mucin production can be identified with mucicarmine or periodic acid-Schiff staining. The WHO classification of lung cancer divides adenocarcinomas into (1) acinar, (2) papillary, (3) bronchoalveolar, and (4) mucus-secreting. Bronchoalveolar carcinoma is a distinct clinicopathologic entity that appears to arise from type II pneumocytes and may manifest as a solitary peripheral nodule, multifocal disease, or a pneumonic form, which can spread rapidly from one lobe to another.

Stage for stage, adenocarcinomas are associated with worse prognoses than SCCs, with the exception of T1 N0 M0 tumors.

Large cell carcinoma is the least common of all NSCLCs. It is composed of large cells with prominent nucleoli, and no mucin production or intercellular bridging is identified. Many tumors previously diagnosed as large cell carcinomas are identified as poorly differentiated adenocarcinomas or SCCs after advanced immunohistochemical staining, electron microscopy, and monoclonal antibody studies.

A variant of large cell carcinoma has been identified; it contains neuroendocrine features and is called large cell neuroendocrine carcinoma. Large cell neuroendocrine carcinomas are associated with a worse prognosis than large cell carcinomas.

WHO classification of epithelial lung tumors

WHO divides epithelial lung tumors into preinvasive lesions and invasive malignant lesions. Preinvasive lesions include the following:

Invasive malignant lesions include the following:

Staging

The most important prognostic indicator in lung cancer is the extent of disease and lymph node involvement. The American Joint Committee for Cancer Staging and End Results Reporting has developed the TNM (tumor-node-metastasis) staging system, which takes into account the degree of spread of primary tumor, the extent of regional lymph node involvement, and the presence or absence of distant metastases (see the table below).[61] The TNM system is used for all lung carcinomas except SCLCs.

AJCC TNM staging and grouping system

Primary tumor (T) involvement is as follows:

Lymph node (N) involvement is as follows:

Metastatic (M) involvement is as follows:

AJCC prognostic groups for NSCLC comprise 4 stages, with further subdivision of stages into subtypes. These stages have important therapeutic and prognostic implications, which are discussed later.

Stage grouping of the TNM system is as follows:

Go to Imaging in Lung Cancer Staging and Non-Small Cell Lung Cancer Staging for complete information on this topic.

See also Lung Cancer Staging -- Radiologic Options, a Critical Images slideshow, to help identify stages of the disease process.

Workup for Special Populations

Patients with CNS metastasis, immunosuppression, superior vena cava syndrome (SVCS), Pancoast tumor, and/or Ogilvie intestinal pseudo-obstruction may require specific workup, as described below. If no pathologic process is present, discharge the patient with a prescription for continuous analgesic use until follow-up care can be arranged with the patient’s personal physician.[62]

Patients with CNS metastasis and known cancer

Head CT scanning, with and without contrast enhancement to depict masses, may be indicated. Obtain a neurosurgical consultation. Admit patients for possible whole-brain irradiation or resection.

Headache and brain edema may respond to dexamethasone (10 mg IV). Seizures are treated with anticonvulsants, but patients with brain metastases and no history of seizures do not generally require anticonvulsant therapy.[63]

Immunosuppressed patients with cancer and infections

Obtain a CBC for evaluation of neutropenia and other blood cell derangements. Assess electrolyte levels for signs of dehydration. The chest radiograph may show only subtle infiltrate. If diarrhea is present, perform urinalysis with a culture, blood cultures with samples from peripheral sites, cultures with samples from any indwelling catheters, and stool cultures for Clostridium difficile.

Administer broad-spectrum empiric antibiotics (eg, piperacillin, gentamicin, second- or third-generation cephalosporin) and an aminoglycoside. If the patient has a penicillin allergy, replace penicillin with carbapenem (if mild penicillin allergy) or aztreonam.

Treatment with granulocyte colony-stimulating factor (G-CSF) may be appropriate for raising neutrophil levels. Consultation with an oncologist is indicated to begin G-CSF therapy.

Patients with Pancoast tumor

An MRI is superior to a CT scan in depicting superior sulcus tumors. Admit the patient for transthoracic needle aspiration. Perform bronchoscopy if endobronchial involvement is present.

Patients with SVCS

Lung cancer accounts for 60-80% of SVCS. Head elevation, cautious administration of fluids, and supplemental oxygen is indicated. Diuretics and glucocorticoids (methylprednisolone 125 mg IV) may help with symptoms, but their roles are unclear. Definitive treatment is usually by radiotherapy or chemotherapy and/or vena caval stenting.

Patients with Ogilvie intestinal pseudo-obstruction

Abdominal radiograph shows massive dilation of the colon and small intestine, with or without air-fluid levels. Check electrolyte levels and correct abnormalities. Place a nasogastric tube and rectal tube.

Admit the patient for possible colonic decompression and treatment of the underlying cause (eg, lung cancer producing autoantibodies to the myenteric neural plexus). For cancer patients with severe pain and advanced disease, administer opioid analgesics. Nasogastric tube and rectal tube placement may help with pain.

Screening

Guidelines from the American Cancer Society (ACS), American College of Chest Physicians (ACCP), National Comprehensive Cancer Network (NCCN), and U.S. Preventive Services Task Force (USPSTF) recommend offering annual screening with low-dose computed tomography (LDCT) scanning to patients aged 55 to 74 years (the USPSTF extends the recommended age range to 80 years and the NCCN extends to age 77 years ) and who have at least a 30 pack-year smoking history and either continue to smoke or have quit within the past 15 years.[64, 65, 46, 66]

The NCCN guidelines recommend beginning screening at age 50 and lowering the threshold to at least a 20 pack-year smoking history when a patient has one of the following additional risk factors[64] :

The USPSTF recommends discontinuing screening in patients who either have not smoked for 15 years or who have developed a health condition that will substantially limit their life expectancy, the feasibility of curative lung surgery, or their willingness to undergo such surgery.[66]

Using data from the National Health Interview Survey, Cheung et al estimated the number of US smokers eligible for screening on the basis of either USPSTF criteria or the Lung Cancer Risk Assessment Tool and the number of lung cancer deaths preventable with each method. They determined that with risk-based screening, more people would be screened and more deaths prevented. In 2015, risk-based screening would have prevented 5000 more lung cancer deaths than USPSTF-based screening.[67]  However, an analysis by Kumar et al concluded that risk-based screening provides only attenuated and modest benefits with respect to life-years, quality-adjusted life-years, and cost-effectiveness.[68]

A randomized trial designed to assess the value of prolonged lung cancer screening beyond 5 years found LDCT screening benefit improved beyond the 5th year of screening, with a 58% reduced risk of lung cancer mortality (HR 0.42; 95% CI 0.22–0.79), and 32% reduction of overall mortality (HR 0.68; 95% CI 0.49–0.94).[69]

Limitations of screening

In a 2013 analysis of data on 53,452 individuals at high risk for lung cancer, derived from the National Lung Screening Trial, Patz et al determined that performing lung screens with LDCT scanning carries a 22.5% probability of NSCLC overdiagnosis (ie, detection of indolent cancers), as well as an 18.5% probability of overdiagnosis for lung cancer in general. Patz et al concluded that overdiagnosis—which can lead to increases in treatment costs, anxiety, and treatment-related morbidity—should be a consideration when physicians are discussing the risks of LDCT lung cancer screening.[70]

Subsequently, in a retrospective cohort analysis of data from the National Lung Screening Trial participants, Patz et al reported that patients whose initial LDCT scan is negative have a lower incidence of lung cancer and lung cancer-specific mortality. These authors proposed that a longer interval between screens might be warranted in patients whose initial LDCT screening scan is negative.[71]

In this study, lung cancer incidence per 100,000 person-years was 371.88 in the 19,066 participants with a negative LDCT, versus 661.23 in the overall cohort of 26,231 participants. Lung cancer–related mortality rates per 100 000 person-years were 185.82 versus 277.20 for the two cohorts, respectively.[71]

In a study by Kinsinger et al of lung cancer screening in 2106 patients at Veterans Health Administration medical centers, LDCT identified nodules in 59.7% of screened patients, but just 1.5% of patients had lung cancer diagnosed within 330 days. The rate of false-positive test results was 97.5%. These authors concluded that implementing a lung cancer screening program for Veterans Health Administration patients "would potentially require substantial resources and effort by clinical staff and facilities for an uncertain benefit of reduced mortality from lung cancer."[72, 73]

Risk models for screening

A study by researchers from the National Cancer Institute (NCI) and the American Cancer Society that reviewed nine risk prediction models determined that the following four models were more accurate than the others for predicting lung cancer risk and for selecting patients who had ever-smoked for lung cancer screening:

Although the researchers concluded that that any of those models could be used to select US smokers who are at the greatest risk for lung cancer incidence or death, all the models have limitations. The Bach model does not account for race/ethnicity, family history of lung cancer, or presence of chronic obstructive pulmonary disease; the PLCO-M2012 model underestimated lung cancer risk in people of Hispanic descent by a factor of 2 to 3, and the LCRAT and LCDRAT models both underestimated risk in the "Asian/other" subgroup.[74, 75]

Biomarker screening

A collaborative study has identified and validated a panel of circulating protein biomarkers that may improve lung cancer risk assessment and may be used to define eligibility for CT screening.[76, 77] Using prediagnostic blood samples from patients at high risk for lung cancer, the Integrative Analysis of Lung Cancer Etiology and Risk (INTEGRAL) Consortium for Early Detection of Lung Cancer created a risk assessment tool consists of a panel of the following proteins:

In the validation study of 63 ever-smoking patients with lung cancer and 90 matched controls, an integrated risk prediction model that combined smoking exposure with the biomarker panel score identified 40 of the 63 lung cancer cases, corresponding to a sensitivity of 0.63. By comparison, the US Preventive Services Task Force screening criteria demonstrated a sensitivity of 0.42 for these cases.[76, 77]

Approach Considerations

Surgery is the treatment of choice for patients with non–small cell lung cancer (NSCLC) stages I through IIIA.[11] In addition, patients with resected lung cancer have a high risk of relapse and so are treated with adjuvant chemotherapy.[78] Patients with stage IIIB and IV NSCLC are usually offered chemotherapy with the option of surgery. Molecular-targeted therapy plays an increasingly important role in the treatment of advanced NSCLC.

Radiation is a reasonable option for treatment in patients who are not candidates for surgery. The role of adjuvant radiation therapy after resection of the primary tumor remains controversial.

See Non-Small Cell Lung Cancer Treatment Protocols. Go to Oncology Decision Point for expert commentary on NSCLC treatment decisions and related guidelines.

Because most NSCLC cannot be cured with currently available therapeutic modalities, the appropriate application of skilled palliative care is an important part of treatment. Increasing evidence supports offering palliative care concurrently with standard oncologic care at the initial diagnosis of advanced NSCLC.[79]

For example, a clinical trial found that patients with metastatic NSCLC randomized to early palliative care had a better quality of life and, surprisingly, longer median survival than those randomized to standard oncologic care alone. The palliative care group also had less depressive symptoms, and fewer patients in this group received aggressive end-of-life care.[80]

Emergency Treatment

All patients thought to have lung cancer should be encouraged to obtain follow-up care with their primary care physician. In almost all cases, document the possible diagnosis and discuss it with the patient. Definitive treatment of the underlying cancer is not the purview of the emergency department (ED).

Emergency treatment is based on symptoms. In cases of upper airway obstruction, admit the patient to the intensive care unit (ICU), prepare for intubation and/or cricothyrotomy, and obtain otolaryngologic and/or surgical consultation for fiberoptic laryngoscopy or intraoperative tracheostomy.

If hemoptysis is noted, administer supplemental oxygen and perform suctioning. If a threat of imminent demise exists, consider placing a double-lumen endotracheal tube. Position the patient with the bleeding hemithorax in a dependent position. Perform arterial blood gas (ABG) and complete blood count (CBC) (type and crossmatching) coagulation studies if the bleeding is more than trivial. A pulmonologist may have to perform fiberoptic bronchoscopy. Admit patients, except those with the most minor bleeding, to the ICU.

Surgical Treatment

Surgical resection remains the mainstay of treatment for all patients with stage I and II NSCLC—that is, those patients with no evidence of mediastinal disease or invasion of local organs. Lobectomy is the procedure of choice. Outcomes are better when the procedure is performed by a surgeon with specialty training, or is done in a higher-volume center or in a teaching facility.[81]

A study patients who underwent planned resection after an unexpected finding of N2 disease at the time of thoracoscopy or thoracotomy found that proceeding with lobectomy did not appear to compromise outcomes if adjuvant chemotherapy with or without radiation therapy was administered following surgery.[82]

The role of surgery for stage III disease is controversial. (See Stage-Based Management.) Patients with completely resectable primary tumors (ie, T4 N0) have a much better prognosis than those with spread to ipsilateral mediastinal or subcarinal lymph nodes (ie, N2), signifying that spread beyond the primary tumor is associated with a poor prognosis. Patients with stage IIIB or IV tumors are almost never surgical candidates.

Preoperative evaluation

Preoperative evaluation should include a careful assessment of resectability, cardiopulmonary reserve, and perioperative risk. High-resolution computed tomography (CT) and positron emission tomography (PET) scanning are helpful for preoperative planning in early-stage lung cancer.[83]

As a general guideline, most patients with a preoperative forced expiratory volume in one second (FEV1) of greater than 2.5 L are able to tolerate pneumonectomy. With an FEV1 of 1.1-2.4 L, a lobectomy is possible. Patients with an FEV1 of less than 1 L are not considered candidates for surgery. These factors are further modified by the presence of cardiac disease or other comorbid conditions.

Lobectomy and pneumonectomy

The standard surgical approach remains a lobectomy, which helps preserve pulmonary function while allowing a good resection. Hilar and other proximal tumors may require more extensive surgery, including a pneumonectomy, which carries significant operative mortality and long-term morbidity. In such patients, alternative approaches such as sleeve resection may be of value.[84]

Retrospective data from the Surveillance, Epidemiology, and End Results (SEER) database show that lobectomy and segmentectomy result in similar survival among patients with small lung cancers (< 1 cm).[85] This needs to be validated in a randomized phase III study.

Wedge resection/segmentectomy

Sublobar resections are used for patients with poor pulmonary reserve and are increasingly being used in conjunction with video-assisted thoracoscopic surgery (VATS). An older Lung Cancer Study Group trial, of stage IA cancers randomized to standard lobectomy versus sublobar resections, suggested a much higher local recurrence rate (75%), with a near-significant trend towards an increased cancer-specific mortality of 50%.[86]

However, a review of SEER data from 1988-2008 found that the survival benefit of lobectomy over sublobar resection for stage I NSCLCs ≤ 2 cm in size decreased over that time. By 2005-2008, both wedge resections and segmentectomies were equivalent to lobectomy.[87] A Cancer and Leukemia Group B (CALGB) phase III trial randomizing patients to lobectomy or limited resection for small peripheral IA lesions is ongoing, and should provide more clarity in this area.[86]

In patients older than 74 years with stage IA NSCLC, Okami et al found no significant difference in 5-year survival after sublobar resection versus standard lobectomy, although locoregional recurrence rates were higher after sublobar resection.[88] A study by Wolf et al showed that sublobar resection is a reasonable option for elderly patients with compromised cardiopulmonary status.[89]

Video-assisted thoracoscopic surgery

Video-assisted thoracoscopic surgery (VATS) is a minimally invasive surgical modality being used for both diagnostic and therapeutic lung cancer surgery. It offers low perioperative morbidity and mortality as well as decreased pain and hospitalization.

Recurrence rates and 5-year and long-term overall survival appear similar to those with traditional open thoracotomies. This approach is also better tolerated in older populations.[90] Finally, patients treated with VATS appear to have fewer delays and dose reductions in adjuvant chemotherapy. Practice guidelines suggest that VATS is feasible as long as adequate resection is possible.[91, 92]

Mediastinal lymphadenectomy

The role of routine mediastinal lymphadenectomy versus lymph node sampling remains controversial. The authors of a large randomized trial recommend that an adequate mediastinal lymphadenectomy should include exploration and removal of lymph nodes from stations 2R, 4R, 7, 8, and 9 for right-side cancers and stations 4L, 5, 6, 7, 8, and 9 for left-side cancers.[93]

For details on this procedure, see Mediastinal Lymphadenectomy.

Postoperative evaluation and complications

Residual pulmonary function after surgical resection is estimated using pulmonary function tests and radionuclide lung scans.

A study by Allen et al assessed curative-intent resections in a major US metropolitan area. The study found that most resections did not achieve good quality surgical resection (GQR) standards.[94] Although surgical sampling of mediastinal (level 2) lymph nodes was most lacking, evaluation of level 1 lymph nodes was also suboptimal. Further studies are indicated to assess surgical practices in order to achieve minimum standards for accurate staging, prognostication, and eligibility for clinical trials.

An understanding of post-surgical quality of life can help surgeons provide lung cancer patients with important information regarding postoperative outcomes. A study that measured health-related quality of life (HRQOL) in patients who underwent lung cancer surgery found that survivors exhibited clinically meaningful worse dyspnea, coughing, chest pain, and financial problems than the general population.[95]

The perioperative mortality rate is 6% for pneumonectomy, 3% for lobectomy, and 1% for segmentectomy. These rates reflect improvements in anesthesia and surgical techniques.

Radiation Therapy

In the treatment of stage I and stage II NSCLC, radiation therapy alone is considered only when surgical resection is not possible because of limited pulmonary reserve or the presence of comorbidities.[2] Radiation is a reasonable option for lung cancer treatment in patients who are not candidates for surgery.[3] Radiation therapy alone as local therapy, in patients who are not surgical candidates, has been associated with 5-year cancer specific survival rates of 13-39% in early-stage NSCLC (ie, T1 and T2 disease).[96]

This inferior survival reflects the poor functional status of these patients, as well as the likelihood of these patients actually having a higher stage, given the known limitations of clinical staging. Survival appears to be enhanced by the use of hyperfractionation schedules, such as continuous hyperfractionated accelerated radiotherapy (CHART) at 1.5 Gy 3 times a day for 12 days, as opposed to conventional radiation therapy at 60 Gy in 30 daily fractions. Overall survival at 4 years was 18% vs 12%.

A study by Jeremić et al assessed independent prognosticators of outcome for hyperfractionated radiation therapy treatment.[97] The study found that female sex, lower Karnofsky performance score (KPS), less pronounced weight loss, squamous histology, lower stage, shorter interfraction interval, and treatment independently predicted better overall survival and progression-free survival. Age did not influence overall survival or progression-free survival.

A retrospective study of 722 patients with NSCLC undergoing radiotherapy who were taking beta blockers for another condition found that these patients had better overall survival, disease-free survival, and distant metastasis–free survival than patients not taking these drugs. Data showed a 22% improvement in overall survival in the beta-blocker group. However, there was no improvement in locoregional progression-free survival, which suggests that beta blockers affect the metastatic tumors rather than the primary tumor.[4]

In a population-based study of 10,376 elderly patients with unresectable stage III NSCLC who were not candidates for chemotherapy, treatment with radiotherapy alone (specifically, complex radiotherapy) was associated with significantly improved survival. Median overall survival was found to be 9 months in the radiation-treated group (n=6468) and 7 months in the untreated patient group.[98]

However, this was benefit was seen only in patients treated with complex radiotherapy, not in those who received intermediate complexity radiotherapy. In addition, patients treated with radiation were more likely to be hospitalized with pneumonitis or esophagitis.

Stereotactic body radiotherapy (SBRT) is another technique for nonoperative treatment of early-stage lung cancers. SBRT uses precise targeting of high-dose radiation to the tumor, typically in 1-2 fractions, while minimizing toxicity to normal tissues. Patients best suited for SBRT include those with a peripheral node-negative tumor that is less than 5 cm, in whom definitive surgery is contraindicated.

In a phase II study by Timmerman et al, use of SBRT in patients with inoperable NSCLC resulted in a 3-year survival rate of 55.8% (versus the 20%-35% seen with current management), high rates of local tumor control, and moderate treatment-related morbidity.[99] A large Japanese retrospective analysis showed that patients treated with SBRT at doses higher than 100 Gy had a local recurrence rate of 8.4%, and a 5-year overall survival of 70.8%.[100] Randomized studies of SBRT are being conducted by the Radiation Therapy Oncology Group (RTOG).

SBRT was associated with shorter overall survival but similar recurrence rates and cause-specific mortality in a nonrandomized study by Grills et al. The study compared outcomes in patients with stage T1-2N0M0 NSCLC who were ineligible for lobectomy and thus underwent either wedge resection or, if deemed medically inoperable, SBRT.[101]

Radiofrequency ablation (RFA) has also been used for inoperable patients who have peripheral tumors that are less than 3 cm in size, and occasionally in a palliative setting. In a single small nonrandomized prospective study, 2-year overall survival with stage I NSCLC was 75% (45-92%). This may be an option for patients in whom both surgery as well as traditional external beam radiation therapy may be contraindicated.[102]

Planning 4-dimensional computed tomography (4DCT) scans can be used to minimize target volumes for lung cancer radiotherapy, but 4DCT results may not be fully representative of patient movement during radiotherapy. Dutch investigators have reported that megavoltage cinema images can identify patients in whom planning 4DCT scans are not representative and thus could be helpful in the treatment of small lesions.[103]

Adjuvant radiation therapy

The role of adjuvant radiation therapy after resection of the primary tumor remains controversial.[104, 105] Radiation therapy reduces local failures in completely resected (stages II and IIIA) NSCLC but has not been shown to improve overall survival rates. In one study, 5-year overall survival was actually worse (30% vs 53%). A retrospective SEER analysis also showed that survival was lower for this population.

A single phase III study using small fractions, with 3D treatment planning, showed a 5-year survival benefit in the radiation treatment arm (67% vs 58%).[106] This finding has not been replicated; hence, at this time, postoperative radiation therapy for stage I and II lung cancer is reserved for cases with positive margins, until further trials are conducted with modern radiation therapy planning and delivery.

Systemic Chemotherapy

The treatment of NSCLC has drastically changed over the past few years, with the advent of molecular-targeted agents. Nevertheless, cytotoxic chemotherapy retains a role in management..

Only 30-35% of patients with NSCLC present with sufficiently localized disease at diagnosis that curative surgical resection may be attempted (stages IA and IB, IIA and IIB, and IIIA). Furthermore, approximately 50% of patients who undergo surgical resection experience local or systemic relapse. Thus, approximately 80% of all patients with lung cancer are considered for chemotherapy at some point during the course of their illness.

At present, chemotherapy alone has no role in potentially curative therapy for NSCLC. Some trials have shown a survival benefit with adjuvant chemotherapy (ie, chemotherapy given after surgery) in resected stage IIA, IIB, and IIIA NSCLC, with a survival advantage of 5-10%.[107, 108] However, adjuvant chemotherapy in elderly patients with resected stage IIIA NSCLC is not associated with survival advantage.[109]

Two small, randomized trials have suggested that neoadjuvant chemotherapy (ie, chemotherapy given prior to surgery) prolongs survival in subjects with stage IIIA disease. Other similarly designed trials failed to confirm this.

Chemotherapy may be considered as part of multimodality therapy for locally advanced NSCLC and is used alone in the palliative treatment of stage IIIB NSCLC (owing to malignant pleural effusion) and stage IV NSCLC.[64] In advanced NSCLC, patients with good performance status (ie, 0-2 on the Zubrod or Eastern Cooperative Oncology Group [ECOG] scale) or greater than 70% on the Karnofsky scale; see the table below), and less than 10% body weight loss are good candidates for chemotherapy.



View Image

Non–small cell lung cancer. Performance status scales for patients with cancer.

Large meta-analyses from 16 randomized trials showed a significant survival advantage to patients getting chemotherapy as opposed to best supportive care. One-year survival in the chemotherapy arm was 29% as opposed to 20% in those receiving supportive care alone. This survival benefit was present irrespective of age or histology. There appears to be no detriment in quality of life in the patients treated with chemotherapy; hence, palliative chemotherapy should be offered to all patients who are willing and able to receive chemotherapy.[110, 111]

NSCLC is only moderately sensitive to chemotherapy, with single-agent response rates in the range of 15% or better. Newer agents (eg, gemcitabine, pemetrexed, docetaxel, vinorelbine) have shown promising single-agent activity, with response rates from 20-25%.

A study by Wanders et al found that patients with stage III NSCLC who are 75 years and older could have improved survival if they are provided treatment with curative intent (eg, radiotherapy only, sequential chemotherapy and radiation).[112] In an observational cohort study of patients aged 65 years or older, the benefit of adjuvant chemotherapy was similar to that seen with younger patients. However, a subset analysis of patients aged 80 years or older suggested that adjuvant chemotherapy might have more adverse effects in this population.[113]

Platinum-based regimens

Cisplatin has been the cornerstone of most combination regimens studied in advanced NSCLC.[114] A recent meta-analysis of 16 trials comparing platinum-based regimens to nonplatinum agents showed a statistically significant improved response rate as well as 1-year survival favoring cisplatin. A beneficial trend was noted with carboplatin-based combinations but this was not significant. Gastrointestinal toxicity was higher with cisplatin.[115]

American Society for Clinical Oncology (ASCO) guidelines recommend that first-line treatment for NSCLC should include a platinum combination. In younger patients, with a good performance status or in the adjuvant setting, cisplatin is preferred, but in older patients or those with significant comorbidities, carboplatin may be substituted. Some recent trials have studied nonplatinum combinations such as gemcitabine with a taxane, which have shown noninferiority, and may be an option for selected patients.

Several randomized controlled trials have failed to show a clear superiority of one platinum-containing combination over another. A landmark ECOG trial comparing cisplatin-gemcitabine, cisplatin-paclitaxel, cisplatin-docetaxel, and carboplatin-paclitaxel, suggested similar overall response rates (approximately 19%), and median survival (7.9 mo). One- and 2-year overall survival was also similar at 33% and 11%, respectively.

In October 2012, the US Food and Drug Administration (FDA) approved protein-bound paclitaxel (Abraxane) for locally advanced or metastatic NSCLC, as first-line treatment in combination with carboplatin, in patients who are not candidates for curative surgery or radiation therapy. Approval was based on a single-phase, multicenter, randomized open-label study in which patients with advanced NSCLC received either weekly protein-bound paclitaxel plus carboplatin every 3 weeks or solvent-based paclitaxel plus carboplatin every 3 weeks.[116]

Patients who received the protein-bound paclitaxel demonstrated a statistically significantly higher overall response rate (ORR) compared with those in the solvent-based paclitaxel arm (33% vs 25%; P = 0.005). In patients with squamous histology, the ORR was also statistically superior in the protein-bound paclitaxel group (41% vs 24%; P < 0.001). There was no statistically significant difference in overall survival between the 2 study arms.[116]

The cisplatin-gemcitabine did appear to have an increased progression free survival, compared with the standard treatment arm of cisplatin-paclitaxel (4.2 mo vs 3.4 mo), with increased renal toxicity. This finding was confirmed by 2 Italian and Japanese studies, which showed similar efficacy of these combinations as measured by response rates or survival.

However, a meta-analysis comparing cisplatin-gemcitabine with other platinum-containing regimens suggested an improved median survival (9 vs 8.2 mo), and an absolute improvement in 1-year overall survival of 3.9% as compared with nongemcitabine combinations. This effect was not sustained when compared against other third-generation cisplatin combinations.[117]

A study by Quoix et al found that platinum-based doublet chemotherapy was associated with survival benefits among elderly patients with NSCLC compared with vinorelbine or gemcitabine monotherapy, despite increased toxic effects.[118] A separate study by Pallis et al found that chemotherapy treatment among older patients with NSCLC is feasible, with no significant differences in response compared with younger patients, although increased toxicity is noted.[119]

Histologic factors in chemotherapy responsiveness

For some time, NSCLC histology was thought to not impact chemotherapy responsiveness. A phase III trial comparing upfront cisplatin-pemetrexed with cisplatin-gemcitabine in stage III and IV NSCLC showed similar response rates (30.6% vs 28.2%), median survival (10.3 mo each), and 2-year overall survival (18.9% vs 14%). These were statistically similar.

However, in a preplanned subset analysis, median survival in patients with nonsquamous histology was significantly better with cisplatin-pemetrexed than with cisplatin-gemcitabine: 12.6 vs 10.9 mo for patients with adenocarcinoma and 10.4 vs 6.7 mo for those with large cell histology. In contrast, the patients with squamous cell histology did better with the cisplatin-gemcitabine combination: 10.8 vs 9.4 mo. Cisplatin-pemetrexed is now the preferred combination for adenocarcinoma.[120]

Genetic factors in resistance to platinum compounds

Although platinum-based chemotherapy is currently standard of care in NSCLC, data suggest that certain individual tumors may have inherent resistance to platinum compounds. Excision repair cross-complementation group 1 (ERCC1) is one such genetic abnormality, and high levels of ERCC1 messenger RNA in tumor tissue have been associated with resistance to platinum.

Holm et al found that, in patients receiving carboplatin and gemcitabine for inoperable NSCLC, the expression of ERCC1 had different effects on survival in men and women. In a retrospective study in 163 patients, men whose tumors were ERCC1 negative survived significantly longer than men with ERCC1 -positive tumors (median survival, 11.8 mo vs 7.9 mo). Conversely, ERCC1 status had no effect on survival in women.[121]

As with ERCC1, increased expression of ribonucleotide reductase subunit 1 (RRM1) has been associated with decreased response to gemcitabine and platinum.

In the future, therapy for NSCLC may need to be tailored on the basis of the genetic characteristics of individual tumors. Gene expression profiling has been studied for accurate classification of NSCLC histology as well as prognostication, but this remains experimental.

Second-line chemotherapy

Selected patients with good responses to first-line chemotherapy, good performance status, and a long disease-free period between initial chemotherapy and relapse may be candidates for second-line chemotherapy. Docetaxel and pemetrexed have been approved by the US Food and Drug Administration (FDA) in this clinical setting, but other drugs (eg, gemcitabine, vinorelbine), if not used in the first-line regimen, may result in similar palliation and clinical benefit.

In December 2014, the FDA approved ramucirumab (Cyramza) in combination with docetaxel for metastatic NSCLC with disease progression on or after platinum-based chemotherapy. Patients with EGFR or ALK genomic tumor aberrations should have disease progression on FDA-approved therapy for these aberrations prior to receiving ramucirumab.

Ramucirumab’s approval was based on improved overall survival (OS) in a multicenter, double-blind, placebo-controlled study (n = 1253) in patients with previously treated metastatic NSCLC. Patients were randomized to receive either ramucirumab (10 mg/kg q3wk) in combination with docetaxel (75 mg/m2 q3wk) on day 1 of a 21-day cycle (n=628) or matching placebo plus docetaxel (n=625). Results showed a statistically significant prolonged OS and PFS in patients treated with ramucirumab plus docetaxel.[122]

A phase III study published in 2009 compared immediate and delayed docetaxel after front-line therapy with gemcitabine plus carboplatin in advanced NSCLC and found a statistically significant improvement in progression-free survival when docetaxel was administered immediately after front-line gemcitabine plus carboplatin, without increasing toxicity or decreasing quality of life. The increase in overall survival was not statistically significant.[123]

Another phase III trial of pemetrexed versus docetaxel showed similar efficacy for both agents in recurrent NSCLC when administered as single-agent chemotherapy in second-line settings. Response rates (9.1% vs 8.8%) and overall survival (8.3 mo vs 7.9 mo) were similar.[124]

In a phase III study comparing docetaxel and docetaxel/aflibercept, the addition of ziv-aflibercept to standard docetaxel therapy did not improve overall survival in platinum-pretreated patients with advanced or metastatic nonsquamous NSCLC.[125]

Complications

Chemotherapy can give rise to various adverse effects, as follows:

Combined Chemoradiation Therapy

The current standard of care in the management of good-risk (ie, Karnofsky performance score of 70-100, minimal weight loss) patients with locally advanced unresectable (stage IIIA) non–small cell lung cancer (NSCLC) is combined-modality therapy consisting of platinum-based chemotherapy in conjunction with radiation therapy. This combination results in statistically significant improvement in both disease-free and overall survival rates compared with either modality used alone.[126, 127]

Randomized studies show longer survival in patients with unresectable stage III disease when treated with concurrent (rather than sequential) platinum-based chemotherapy and radiation therapy.[128, 129] In a Radiation Therapy Oncology Group (RTOG) study that compared cisplatin/vinblastine either given concurrently with radiation therapy or followed by radiation therapy, the concurrent group showed better median survival as well as overall survival (17 vs 14.6 mo and 21% vs 12%, respectively).[130, 131, 132]

Chemotherapy regimens that have been studied in combination with radiation therapy include cisplatin/vinblastine and cisplatin/etoposide (5-y survival of 15%).[128] In elderly patients or those with comorbidities and contraindications to cisplatin, weekly carboplatin/paclitaxel may be used, based on a phase II study that showed a median survival of 16.7 months.[133, 134, 135]

Consolidation chemotherapy after chemoradiation had initially been shown to be beneficial in phase II studies, with docetaxel after chemoradiation with cisplatin/etoposide showing a median survival of 26 months and a 5-year survival of 29%. In phase III testing, however, this regimen did not show improved survival and it proved to be more toxic, so it is no longer recommended outside a clinical trial setting.[136]

In a phase III study of 610 patients, treatment with a combination of cisplatin, etoposide, and radiation conferred significant overall survival. These researchers concluded that this regimen should be the standard in patients with stage III lung cancer.[137]

More recently, consolidation therapy with the anti-programmed death ligand–1 antibody durvalumab following concomitant chemoradiation has shown significant benefit. In a study in 713 patients with stage III NSCLC who did not have disease progression after two or more cycles of platinum-based chemoradiotherapy, median progression-free survival was 16.8 months with durvalumab versus 5.6 months with placebo, and the 18-month progression-free survival rate was 44.2% versus 27.0%.[138]

The high locoregional failure rate with chemoradiation alone has led to study of chemoradiation followed by surgical resection.[105, 107, 139] Uncontrolled phase II studies suggested possible survival benefit from this approach, but a phase III study showed only a nonsignificant trend toward better 5-year overall survival (27% vs 20%) despite an improvement in progression-free survival. The postoperative mortality rate was higher in those patients undergoing surgical resection.

A European Organization for Research and Treatment of Cancer (EORTC) study failed to show any benefit of resection over radiation therapy after neoadjuvant chemotherapy in patients with stage IIIA (N2) disease.[140]

In a small study in patients who were node negative with T3 and T4 NSCLC, neoadjuvant chemoradiation followed by surgery led to better survival.[141]

 

Molecular-Targeted Therapy

In recent years, the increased understanding of molecular abnormalities in lung cancer, identification of molecular targets, and development of molecular-targeted therapies have produced a paradigm shift. The treatment of NSCLC has entered a new era.

The possibility of finding a mutation that is susceptible to molecular-targeted therapy is driving more frequent mutation testing in NSCLC. However, the likelihood of a mutation depends on the histologic subtype of the cancer. Consequently, histologic testing should precede mutation testing.[91]

All patients with NSCLC should have their tumor tissue tested for mutations, such as in the genes that code for endothelial growth factor receptor (EGFR), KRAS, anaplastic lymphoma kinase (ALK), ROS1, and programmed death ligand–1 (PDL-1). The results will help determine the patient's eligibility for treatment with specific molecular-targeted agents.

EGFR testing that identifies sensitivity to EGFR-directed tyrosine kinase inhibitors (TKIs) include exon 19 deletions or the L858R point mutation. The T790M and exon 20 insertion have been associated with low response or acquired resistance to TKIs. In patients with adenocarcinomas that have EGFR mutations consistent with TKI sensitivity, options for single-agent targeted therapy without chemotherapy (first-, second-, or subsequent-line) include the following:

NCCN guidelines recommend osimertinib as the preferred category 1 option for first-line therapy in patients who have EGFR mutations documented before first-line chemotherapy.[91] Other EGFR-directed TKIs are also recommended as first-line agents include erlotinib, afatinib, gefitinib, or dacomitinib. Additional options include the following:

NSCLC adenocarcinoma with ALK rearrangements more commonly occurs in light smokers or non-smokers. These patients do not benefit from treatment with EGFR-directed TKIs. Instead, first-line therapy is with one of the following:

Treatment for NSCLC with BRAF mutation is with dabrafenib (Tafinlar) in combination with trametinib (Mekinist).

For patients with RET fusion-positive NSCLC, selpercatinib (Retevmo) is indicated.

For patients with VEGF mutations, the following agents are used in combination with chemotherapy:

For patients with ROS-1 mutations, the following agents are used:

PDL-1 testing results direct therapy as follows:

In patients with NSCLC that does not demonstrate EGFR or ALK but has high PDL-1 expression, the immune checkpoint inhibitors pembrolizumab (Keytruda) and atezolizumab (Tecentriq) are approved for use as first-line therapy. They are also used in combination with chemotherapy or as a single agent in second-line and third-line therapy.

The immune checkpoint inhibitor nivolumab (Opdivo) is used in combination with ipilimumab for first-line treatment, and as monotherapy in patients with progression of metastatic NSCLC on or after platinum-based chemotherapy.

EGFR mutations

Overexpression of EGFR is common in NSCLC. Cancers overexpressing EGFR have been shown to have increased resistance to therapy, increased metastatic potential, and thus a poorer prognosis.

Stimulation of the EGFR pathway leads to increased autophosphorylation of a tyrosine kinase pathway associated with EGFR. This leads to a series of intracellular events culminating in increased mitotic and growth potential, increased ability to metastasize, and increased angiogenesis (new blood vessel formation) in the cancer cells.

Rosell et al evaluated the feasibility of large-scale screening for EGFR mutations in patients with advanced NSCLC. EGFR mutations were found in 350 of 2105 patients (16.6%). Mutations were found more frequently in women (69.7%), patients who had never smoked (66.6%), and those with adenocarcinomas (80.9%). These researchers concluded that large-scale screening of patients with lung cancer for EGFR mutations is feasible and can have a role in decisions about treatment.[144]

The NCCN guidelines add that EGFR mutations are present in adenocarcinomas in approximately 10% of Western patients and up to 50% of Asian patients, and that the EGFR mutation frequency is higher in nonsmokers, women, and patients with non-mucinous cancers. In squamous cell carcinomas, however, the observed incidence of EGFR mutations is 2.7% and the true incidence can be confidently posited as less than 3.6%—too low to justify routine testing of all tumor specimens.[91]

EGFR-directed therapies are described below, in the order of their FDA approval.

Afatinib

Afatinib (Gilotrif) is a TKI that was approved by the FDA in July 2013 for first-line treatment of patients with metastatic NSCLC whose tumors have EGFR exon 19 deletions or exon 21 (L858R) substitution mutations, as detected by the diagnostic companion test therascreen EGFR RGQ PCR kit. Approval was based on data from the LUX-Lung 3 trial, in which progression-free survival in patients receiving afatinib was 11.1 months, compared with 6.9 months for those treated with pemetrexed/cisplatin.[145] In 2018, the indication for first-line use in metastatic NSCLC was expanded to include 3 additional nonresistant EGFR mutations (ie, L861Q, G719X, S768I).[146]

Additionally, in patients with tumors expressing the two most common EGFR mutations (Del19 or L858R), progression-free survival was 13.6 months in those who received afatinib, versus 6.9 months for those in the chemotherapy arm.[145]

In April 2016, the FDA approved afatinib for metastatic squamous NSCLC that has progressed after platinum-based chemotherapy. Approval was based on the LUX-Lung 8 clinical trial. Compared with erlotinib, afatinib obtained a significant delay in progression of lung cancer, reducing the risk of cancer progression by 18%. Also observed was a significant improvement in overall survival (p=0.0077), reducing the risk of death by 19%. A significantly improved disease control rate was also shown (51% vs 40%; P=0.002).[147]

The most common serious adverse events in patients receiving afatinib included diarrhea (6.6%), vomiting (4.8%), dyspnea, fatigue, and hypokalemia (1.7%). Fatal adverse events included pulmonary toxicity/interstitial lung disease (1.3%), sepsis (0.43%), and pneumonia (43%).[145]

Gefitinib

Gefitinib represents a class of EGFR tyrosine kinase inhibitors (TKIs) that act intracellularly to block activation of the EGFR pathway.[148, 149]

In July 2015, gefitinib returned to the United States market following restricted availability. It was approved by the FDA as first-line treatment of patients with metastatic NSCLC whose tumors have EGFR exon 19 deletions or exon 21 (L858R) substitution mutations as detected by an FDA-approved test. Approval as first-line treatment for metastatic NSCLC is based on data from the IFUM (IRESSA Follow-Up Measure) clinical trial, which showed an overall response rate (ORR) of about 50% with a median duration of response of 6 months.[150]

The IFUM results were supported by the most recent analysis of the IPASS (IRESSA Pan-ASia Study) study which assessed gefitinib vs. carboplatin/paclitaxel as a first-line treatment in these patients. The subset population consisted of 186 of 1217 patients (15%) determined to be EGFR positive by the same clinical trial assay as used in IFUM and had radiographic scans available for a retrospective assessment. IPASS showed an ORR of 67% with a median duration of response of 9.6 months in gefitinib-treated patients vs. 41% ORR with a median duration of response of 5.5 months for the carboplatin/paclitaxel group. Mean Progression Free Survival (PFS) was 10.8 months in the gefitinib group vs. 5.4 for the carboplatin/paclitaxel patients.[151]

Two large phase II trials led to the expedited original approval of gefitinib in the United States as a third-line therapy.

However, the Iressa Survival Evaluation in Lung Cancer (ISEL) trial, a large phase III randomized trial that compared gefitinib with placebo in patients who had progressed following first-line chemotherapy, found no significant improvement in median survival (5.6 vs 5.1 mo) overall and also in the adenocarcinoma subset (6.3 vs 5.4 mo). Planned subset analyses in patients who had never been smokers and those of Asian ethnicity showed significantly longer survival (8.9 vs 6.1 mo and 9.5 vs 5.5 mo) than with placebo.[149]

The INTEREST trial studied gefitinib versus docetaxel in the second-line setting and found no significant difference in survival (7.6 vs 8 mo). Based on these data, gefitinib was removed from the United States market for use in new patients and was only available by a restricted access program.

Mok et al conducted a large phase III study that compared gefitinib with carboplatin-paclitaxel in the first-line setting in Asian patients who had adenocarcinoma and had never smoked or were former light smokers (none in last 15 years). Patients receiving gefitinib had a higher response rate (43% vs 32%), with similar median survival (18.6 vs 17.3 mo). Of patients who were positive for the EGFR gene mutation, those in the gefitinib group had significantly longer progression-free survival than those in the carboplatin-paclitaxel group. Conversely, patients who were negative for the mutation had significantly longer progression-free survival with carboplatin-paclitaxel.[152]

In an early IPASS study, EGFR mutation was found to be the strongest predictor of progression-free survival and response to gefitinib.[153]

Erlotinib

A second EGFR TKI, erlotinib, improved survival rates compared with placebo in the second- and third-line setting.[154, 155] Erlotinib demonstrated improved response rates (8% vs < 1%), and overall survival (6.7 vs 4.6 mo). This led to the initial FDA approval of erlotinib in the second-line setting. In late 2016, approval was extended to use for first-line treatment, maintenance treatment, or second- or subsequent-line treatment after progression following at least 1 prior chemotherapy regimen in patients with NSCLC that has EGFR exon 19 deletions or exon 21 (L858R) substitution mutations. as detected by an FDA-approved test.

In an open-label, randomized, phase III trial of Chinese patients with adenocarcinoma of the lung with an EGFR mutation, progression-free survival was longer in patients treated with erlotinib than in those receiving standard chemotherapy with gemcitabine plus carboplatin (13.1 mo vs 4.6 mo).[156]

An open-label randomized phase III trial in European patients with NSCLC also found erlotinib to be superior to standard chemotherapy for first-line treatment of NSCLC with EGFR mutations. In this study, progression-free survival in patients with either of two specific types of EGFR mutations (exon 19 deletions or exon 21 [L858R] substitution mutations) was 9.7 months with erlotinib treatment, compared with 5.2 months for those who received standard therapy.[59]

Similar to the experience with gefitinib, no benefit was seen when erlotinib was combined with chemotherapy. Earlier studies also showed better response rates and survival with females, Asian persons, nonsmokers, particularly those with adenocarcinoma histology (especially bronchioalveolar cancer), as was seen with gefitinib.

In contrast, a study in 760 unselected patients with advanced NSCLC found that first-line treatment with erlotinib followed at disease progression by cisplatin-gemcitabine was significantly inferior in terms of overall survival compared with the standard sequence of chemotherapy with cisplatin-gemcitabine followed by erlotinib.[157]

A study by Herbst et al found no benefit when erlotinib was combined with bevacizumab compared with erlotinib alone in patients who experienced a failure of standard first-line chemotherapy.[158] A separate study by Hirsch et al compared the use of erlotinib alone or intercalated with chemotherapy among chemotherapy-naïve patients with advanced NSCLC who were positive for EGFR protein expression, had a high EGFR gene copy number, or both. The study results did not support combined chemotherapy and erlotinib in this setting; patients with tumors harboring EGFR mutations had better outcomes with erlotinib alone.[159]

Osimertinib

Osimertinib (Tagrisso) is an irreversible EGFR-TKI inhibitor designed to inhibit both EGFR-sensitizing and EGFR T790M-resistance mutations, with clinical activity against CNS metastases. In March 2017, FDA fully approved osimertinib for patients with EGFR T79M-positive NSCLC whose disease progressed on or after EGFR TKI therapy. In April 2018, the FDA also approved osimertinib for the first-line treatment of patients with metastatic NSCLC whose tumors express EGFR exon 19 deletions or exon 21 L858R mutations, as detected by an FDA-approved test.[160, 161]

Approval was based on a phase III FLAURA study conducted in 556 patients with EGFR exon 19 deletion or exon 21 L858R mutation-positive, unresectable or metastatic NSCLC who had not received previous systemic treatment for advanced disease. Patients were randomized to receive osimertinib or a standard EGFR-TKI (gefitinib or erlotinib). Of those randomized to the standard EGFR-TKIs, 20% received osimertinib as the next line of antineoplastic therapy. The estimated median PFS was 18.9 months in the osimertinib arm and 10.2 months in the standard EGFR-TKI arms. The ORR was 77% for the osimertinib arm and 69% with standard EGFR-TKI.. The estimated median response durations for the osimertinib and standard therapy arms were 17.6 and 9.6 months, respectively.[162]

Dacomitinib

Dacomitinib (Vizimpro) is an irreversible kinase inhibitor of the human EGFR family (EGFR/HER1, HER2, and HER4) and certain EGFR-activating mutations (exon 19 deletion or the exon 21 L858R substitution mutation). It is indicated for first-line treatment of patients with metastatic non-small lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) exon 19 deletion or exon 21 L858R substitution mutations as detected by an FDA-approved test.

Approval of dacomitinib was based on the ARCHER 1050 trial (n=452). Dacomitinib was compared with gefitinib. The results showed a statistically significant improvement in progression-free survival (PFS) with dacomitinib: median PFS was 14.7 months with dacomitinib compared with 9.2 months with gefitinib (hazard ratio, 0.59; P < .0001).[163] Overall survival analysis showed slight improvement with dacomitinib 34.1 months compared with gefitinib 26.8 months.[164]

Cetuximab

Cetuximab, a monoclonal antibody that binds the EGFR receptor, is also used in colorectal cancer and squamous cell cancer (SCC) of the head and neck. It was studied in the first-line setting, in combination with cisplatin-vinorelbine, compared with cisplatin-vinorelbine alone, in patients with NSCLC that expressed EGFR by immunohistochemistry.[165]

The chemotherapy was given in combination with cetuximab for up to 6 cycles, and in responding patients, the cetuximab was continued until progression. Patients receiving cetuximab had an increased response rate (36% vs 29%), and improved median survival (11.3 vs 10.1 mo). Whites appeared to benefit more than Asian persons, who seemed to do worse with this regimen.

A phase 3 study found that the addition of cetuximab to first-line chemotherapy in patients with advanced NSCLC provided a survival benefit in patients with high EGFR expression (immunohistochemistry score ≥ 200), increasing overall survival from a median of 9.6 to 12.0 months. No corresponding survival benefit was seen in those with low EGFR expression.[166]

The cetuximab/cisplatin/vinorelbine chemotherapy regimen is not recommended by NCCN guidelines due to small benefit of 1 month, patient have por tolerance to this regimen and most patients have multiple comorbid conditions.

Capmatinib

Resistance to EGFR TKI therapy can result from mesenchymal-epithelial transition (MET) exon 14 skipping. In May 2020, the FDA approved capmatinib (Tabrecta) for treatment of adults with metastatic NSCLC with exon 14 skipping. Approval was based on a study in which the ORR in treatment-naïve patients (n=28) was 68% (complete response in 4%, partial response in 64%) and the ORR in previously treated patients (n=69) was 41%, with all having a partial response. Median duration of response was 12.6 months in treatment-naïve patients and 9.7 months in previously treated patients.[167]

The FDA also approved a companion diagnostic for capmatinib, the FoundationOne CDx assay (F1CDx). F1CDx is a next-generation sequencing–based, in vitro diagnostic device that detects MET exon 14 skipping mutations, as well as other mutations.[167]

Squamous Cell NSCLC

For patients with metastatic squamous cell lung cancer it is important to test for EGFR mutation. If EGFR mutation is present, chemotherapy with cisplatin and gemcitabine and necitumumab should be prescribed. Other options could include oral EGFR inhibitors.

Necitumumab

Necitumumab is another monoclonal antibody that binds to human EFGR and blocks the interaction between EGFR and its ligands. It was approved in November 2015 for first-line treatment of metastatic squamous NSCLC in combination with gemcitabine and cisplatin. Approval was based on the SQUIRE trial, conducted in 1093 patients, which showed a 1.6-month improvement in overall survival (OS) when necitumumab was added to a regimen of gemcitabine and cisplatin. The median OS was 11.5 months, compared with 9.9 months for gemcitabine and cisplatin alone.[168]

However, current NCCN guidelines recommend against use of the necitumumab/cisplatin/gemcitabine regimen for patients with metastatic squamous cell NSCLC, based on the opinion that the addition of necitumumab to the regimen is not beneficial based on toxicity, cost, and limited improvement in efficacy when compared with cisplatin/gemcitabine. Specifically, the NCCN panel notes that the addition of necitumumab resulted in only a slight improvement in overall survival and an increase in grade 3 or higher adverse events.[91]

Pembrolizumab

Pembrolizumab, a PD-1 inhibitor, gained approval in 2018 as a first-line treatment option for metastatic squamous NSCLC in combination with carboplatin and either paclitaxel or nab-paclitaxel. Approval was supported by the KEYNOTE-407 study. In the trial, the combination of pembrolizumab plus chemotherapy (carboplatin and paclitaxel or nab-paclitaxel) significantly improved overall survival (OS) compared with chemotherapy alone, at a median of 15.9 months compared with 11.3 months (hazard ratio [HR], 0.64; P = 0.0017).[169]

Vandetanib

A randomized, double-blind, phase III trial assessed the use of vandetanib, a once-daily oral inhibitor of vascular EGFR and EGFR signaling.[170] This second-line therapy did not meet the primary end point of statistically significant progression-free survival. However, the combination of vandetanib (100 mg/d) plus pemetrexed (500 mg/m2) produced a significantly higher objective response rate and a significant delay in time to worsening of symptoms compared with placebo. The combination also had an acceptable safety profile. This indication is not yet approved by the FDA.

Non-Squamous NSCLC: VEGF (vascular endothelial growth factor)

In adenocarcinoma and large cell lung cancers that lack mutations in EGFR, ROS1 or ALK, combination chemotherapy and targeted therapy may be offered.

Bevacizumab

In an Eastern Cooperative Oncology Group (ECOG) study, addition of the anti-angiogenesis agent bevacizumab to standard first-line carboplatin-paclitaxel resulted in significant prolongation of survival. Bevacizumab was continued in patients who appeared to respond to four to six cycles of chemotherapy. The median overall survival was improved (12.3-10.3 mo), as was the response rate (35% vs 15%) compared with chemotherapy alone.[171]

Patients with squamous cell histology, brain metastases, clinically significant hemoptysis, and ECOG performance status of greater than 1 were excluded.[171] Despite increased hemorrhagic complications and treatment-related deaths, bevacizumab has been approved for use in this setting in combination with chemotherapy.

Bevacizumab has also been studied in combination with cisplatin-gemcitabine as first-line therapy for nonsquamous NSCLC, with improved response rates (20.1% vs 34.1%) and modest improvement in progression-free survival (6.7 vs 6.1 mo). Overall survival was not different.[172]

A 2012 systematic review and meta-analysis of randomized phase II/III trials concluded that the addition of bevacizumab to platinum-based chemotherapy as first-line treatment in patients with advanced NSCLC significantly prolonged overall and progression-free survival. The effects of bevacizumab on overall survival were significantly greater in patients with adenocarcinoma versus other histologies. No unexpected toxicity was observed.[173]

In September 2017, FDA approved Mvasi (bevacizumab-awwb) as a biosimilar to Avastin (bevacizumab) to treat non-squamous NSCLC. It is used in combination with carboplatin and paclitaxel for first-line treatment of unresectable, locally advanced, recurrent or metastatic disease. The approval was based on evidence from animal study data, human pharmacokinetic and pharmacodynamics data, and clinical immunogenicity data.[174]

Ramucirumab

Ramucirumab is a human IgG1 monoclonal antibody that targets the extracellular domain of VEGFR-2. A& phase 3 trial in patients with stage IV NSCLC who had progressed during or after first-line platinum-based chemotherapy reported a 1-month survival benefit with ramucirumab in combination with docetaxel compared with docetaxel plus placebo.[175]

ALK-directed targeted therapies

Chromosomal rearrangement of ALK occurs in about 5% of NSCLC cases. In most of these patients, treatment is initiated with crizotinib. This agent is also indicated for NSCLC with ROS-1 mutations. Second-line agents for ALK-positive NSCLC are brigatinib, ceritinib, and alectinib.

Crizotinib

Crizotinib (Xalkori) was approved by the FDA in August 2011 for the treatment of locally advanced or metastatic NSCLC that is ALK positive, as detected by the Vysis ALK Break Apart FISH Probe test (Abbott Molecular). Approval was based on 2 multicenter trials (n=255) in which median response duration ranged from 41.9-48.1 weeks.[176, 177]

In a study of patients with previously treated advanced NSCLC who had ALK gene rearrangement, treatment with crizotinib was superior to standard chemotherapy.[178] Risk of progression was reduced by 50% with crizotinib. Study participants with locally advanced or metastatic ALK-positive lung cancer treated with one previous platinum-based regimen were randomized to oral crizotinib 250 mg twice daily or to chemotherapy with intravenous pemetrexed (500 mg/m²) or docetaxel (75 mg/m²) every 3 weeks. As part of a separate analysis, patients in the chemotherapy group with disease progression were allowed to cross over to crizotinib.[178]

Median progression-free survival was longer with crizotinib treatment (7.7 mo) than with chemotherapy (3.0 mo). The hazard ratio for progression or death with crizotinib was 0.49 (95% confidence interval [CI], 0.37-0.64; P< 0.001), and the response rate was higher in the crizotinib group (65%) than in the chemotherapy group (20%) (P< 0.001). Overall survival was similar in the two groups.[178]

In March 2016 the FDA expanded use of crizotinib to include patients with metastatic NSCLC whose tumors harbor a ROS-1 gene mutation. Study results showed crizotinib exhibited marked antitumor activity in this population with an objective response rate of 66% by an independent radiology review. There was 1 complete response and 32 partial responses. The median duration of response was 18.3 months.[179]

Brigatinib

Brigatinib (Alunbrig) was approved in April 2017 for ALK-positive metastatic NSCLC in patients who have progressed on or are intolerant to crizotinib. Approval was based on the noncomparative, open-label, multicenter ALTA clinical trial. After a median follow-up of 8 months, median duration of response was 13.8 months in patients randomized to receive brigatinib at oral doses of either 90 mg once daily (n = 112) or 180 mg once daily following a 7-day lead-in at 90 mg once daily (n = 110).[180]

Ceritinib

Ceritinib, an ALK inhibitor, received accelerated approval from the FDA in 2014 for patients with ALK-positive metastatic NSCLC whose disease had progressed or who were intolerant to crizotinib based on a blinded independent review committee (BIRC)-assessed ORR of 44% among 163 patients in a single-arm trial. A phase I study found that ceritinib at the dose of 400 mg daily provided a response rate of 58%, with a median progression-free survival of 7 months. Dose-limiting toxicity included diarrhea, vomiting, dehydration, elevated aminotransferase level, and hypophosphatemia. In this study, ceritinib was effective for both crizotinib-naive patients and those who had tumor progression with crizotinib.[181]

In May 2017, ceritinib was granted regular approval for patients with metastatic NSCLC whose tumors are ALK positive, as detected by an FDA-approved test. This approval also included use as first-line treatment. Approval was based on data from ASCEND-4, a randomized, multicenter, open-label, active-controlled trial conducted in patients with untreated ALK-positive NSCLC. All patients were required to have evidence of ALK-rearrangement identified by the VENTANA ALK (D5F3) test performed through central laboratory testing.

ASCEND-4 randomized 376 patients (1:1) to receive either ceritinib (n=189) 750 mg orally once daily until disease progression or platinum-pemetrexed doublet chemotherapy (n=187). Patients in the chemotherapy arm received pemetrexed (500 mg/m2) with either cisplatin (75 mg/m2) or carboplatin (AUC 5-6) on day 1 of every 21-day cycle for up to 4 cycles, followed by pemetrexed maintenance therapy. Results demonstrated improved PFS as assessed by BIRC, with a hazard ratio (HR) of 0.55 (P < 0.0001). The estimated median PFS was 16.6 months in the ceritinib arm and 8.1 months in the chemotherapy arm. Confirmed ORR, was 73% and 27% in the ceritinib and chemotherapy arms, respectively. Estimated median response durations were 23.9 months and 11.1 months in the ceritinib and chemotherapy arms, respectively. Overall survival data are immature.[182]

In the ASCEND-8 trial, 137 patients receiving ceritinib 450 mg or 600 mg daily with food (~100-500 calories and 1.5-15 grams of fat) or 750 mg daily under fasted conditions, there was no clinically meaningful difference in the systemic steady-state exposure of ceritinib (AUC) for the 450 mg with food arm compared to the 750 mg fasted arm. The steady-state AUC increased by 24% and the peak plasma concentration increased by 25% in the 600 mg with food arm compared to the 750 mg fasted arm.[183]

Alectinib

In November 2017, the FDA approved alectinib (Alecensa), a TKI that targets ALK and RET, for the frontline treatment of patients with ALK-positive metastatic NSCLC. In addition to granting this new indication, the FDA also converted alectinib’s indication for patients with ALK-positive NSCLC who have progressed on crizotinib from accelerated approval to full approval. Alectinib is indicated for ALK-positive, metastatic NSCLC in patients whose disease has progressed on crizotinib or who are intolerant to crizotinib.

Approval was primarily based on findings from the phase III ALEX study. In the trial, treatment-naïve patients were randomly assigned to alectinib 600 mg PO BID or crizotinib 250 mg PO BID. Median PFS, as determined by an independent review committee, was 25.7 months in the alectinib arm versus 10.4 months in the crizotinib arm. The ORR with alectinib was 79% versus 72% with crizotinib. The complete response rates were 13% versus 6%, respectively, and the partial response rate was 66% in both arms. Eighty-two percent of patients receiving alectinib had a response duration ≥6 months, with 64% and 37%, having response durations ≥12 months and ≥18 months, respectively. The corresponding rates in the crizotinib arm were 57%, 36%, and 14%.[184]

A separate phase III study, the Japanese phase III J-ALEX trial, also demonstrated the benefit of crizotinib in ALK-positive NSCLC. In J-ALEX, 207 Japanese patients with ALK-positive advanced or recurrent NSCLC who had not been previously treated with an ALK inhibitor were randomized to alectinib 300 mg PO BID or 250 mg of crizotinib 250 mg PO BID. The median PFS was 25.9 months in the alectinib arm versus 10.2 months in the crizotinib arm. Alectinib reduced the risk of progression in the CNS by 81% in patients without brain metastases at baseline, and by 49% in patients with brain metastases at baseline.[185]

Lorlatinib

Lorlatinib (Lorbrena) was approved in 2018 for patients with ALK-positive metastatic NSCLC who have progressed on crizotinib and at least 1 other ALK inhibitor, or has progressed on alectinib or ceritinib as first ALK inhibitor therapy. Accelerated approval was based on a single, open-label trial (N=215) in patients with ALK-positive metastatic NSCLC previously treated with an ALK inhibitor, in which the overall response rate with loratinib was 48% (4% complete response, 44% partial response), with a median duration of response of 12.5 months. Additionally, in a subgroup of patients with CNS metastases (N=89), the intracranial response rate was 60% (21% complete response, 38% partial response) with a median duration of response of 19.5 months.[186]

Programmed cell death–1 inhibitors

Immune checkpoint inhibitors such as programmed cell death (PD-1) inhibitors have gained increasing importance in cancer therapy. PD-1 is expressed on the surface of activated CD8+ T cells, and cancers that express PD-1 ligand (PDL-1) can inactivate these T cells and thus avoid attack by them. PD-1 inhibitors bind to PD-1, preventing the interaction with PD-1 ligand but not inactivating the T cell, so the T cells maintain their ability to target the cancer cells.

Nivolumab

Nivolumab (Opdivo) was approved for use in NSCLC in March 2015 for NSCLC with squamous pathology. Nivolumab is a monoclonal antibody inhibitor of PD-1. It is the first immunotherapy approved for NSCLC. Current NCCN guidelines recommend nivolumab as a treatment option in patients with squamous cell carcinoma that has progressed on first-line therapy.[91] In October 2015, the FDA approved an expanded indication to include nonsquamous NSCLC.

FDA approval was supported by results from a clinical trial that was stopped early after showing a survival benefit. In the open-label trial, CheckMate 017, 272 patients with advanced or metastatic squamous cell NSCLC were randomly assigned to treatment with either nivolumab 3 mg/kg IV every 2 weeks or docetaxel 75 mg/m² IV every 3 weeks. At 1 year, the overall survival rate was 42% with nivolumab versus 24% with docetaxel. The response rate was 20% with nivolumab versus 9% with docetaxel. The median progression-free survival was 3.5 months with nivolumab versus 2.8 months with docetaxel.[187]

In an early phase I trial of nivolumab in a heavily pretreated group of patients, the response rate was 17%; at a dose of 3 mg/kg, the median overall survival was 14.9 months. Among those patients who responded, the responses were durable, with 24% alive at 24 months. The dose-limiting toxicity was pneumonitis, which resulted in three deaths.[99]

In May 2020, nivolumab combined with ipilimumab was approved for metastatic NSCLC in patients whose tumors express PD-L1 (≥1%), with no EGFR or ALK tumor aberrations. The approval was based the randomized, open-label trial, Checkmate-227. Patients (n=793) with metastatic or recurrent NSCLC and no prior anticancer therapy were randomized to receive either nivolumab plus ipilimumab or platinum-doublet chemotherapy. The median OS was 17.1 months in the nivolumab plus ipilimumab arm compared with 14.9 months in the platinum chemotherapy arm.[188]

Pembrolizumab

In October 2015, a second PD-1 inhibitor, pembrolizumab (Keytruda), was approved for metastatic NSCLC. Approval of pembrolizumab was based in part on data from the KEYNOTE-001 trial, in which pembrolizumab produced an overall response rate of nearly 20% in 495 previously treated and treatment-naive patients with advanced or metastatic NSCLC. The overall response rate was much higher, at 45.2%, in a cohort of patients with NSCLC that showed high expression of PD-L1. The median duration of response exceeded 1 year (12.5 months) in all responders, regardless of the degree of PD-L1 expression. Median overall survival was 12.0 months (95% confidence interval [CI], 9.3–14.7 months) for all patients, 9.3 months (95% CI, 8.4–12.4 months) for previously treated patients, and 16.2 months (95% CI, 16.2 months–not reached) for previously untreated patients.[189]

In October 2016, pembrolizumab was approved as monotherapy for first-line treatment of patients with metastatic NSCLC whose tumors have high PD-L1 expression (Tumor Proportion Score [TPS] ≥50%) as determined by an FDA-approved test, with no EGFR or ALK genomic tumor aberrations. This expanded approval was based on results from the KEYNOTE-024 trial, which showed significantly longer PFS and OS and fewer adverse events with pembrolizumab than with platinum-based chemotherapy. Median PFS was 10.3 months (95% CI, 6.7 to not reached) in the pembrolizumab group compared with 6.0 months (95% CI, 4.2 to 6.2) in the chemotherapy group (hazard ratio for disease progression or death, 0.50; 95% CI, 0.37 to 0.68; P< 0.001).[190]

The above indication was expanded in April 2019 to include patients with stage III NSCLC who are not candidates for surgical resection or definitive chemoradiation, or metastatic NSCLC, and whose tumors express PD-L1 (TPS ≥1%) with no EGFR or ALK genomic tumor aberrations. Approval was based on the KEYNOTE-042 trial (n=1274) that compared pembrolizumab with the investigator’s choice of platinum-based chemotherapy. Results suggest that pembrolizumab monotherapy can be extended as first-line therapy to patients with locally advanced or metastatic non-small-cell lung cancer without sensitizing EGFR or ALK alterations and with low PD-L1 TPS. Overall survival was significantly longer in the pembrolizumab group than in the chemotherapy group in all three TPS populations (ie, ≥50%, ≥20%, and ≥1%).[191]

First-line combination treatment of pembrolizumab was approved in May 2017 for metastatic nonsquamous NSCLC. Approval was based on data from the KEYNOTE-021 trial (Cohort G1) in 123 previously untreated patients with metastatic nonsquamous NSCLC with no EGFR or ALK genomic tumor aberrations and irrespective of PD-L1 expression. In this trial, pembrolizumab plus pemetrexed and carboplatin demonstrated an ORR that was nearly double the ORR of pemetrexed/carboplatin (55% vs 29% respectively; all responses were partial responses). Among patients who received pembrolizumab plus pemetrexed/carboplatin, 93% had a duration of response of 6 months or more (range 1.4+ to 13.0+ months) compared to 81% who received pemetrexed/carboplatin alone (range 1.4+ to 15.2+ months). Progression free survival also improved (median 13.0 months [95% CI]) compared to 8.9 months (95% CI) with pemetrexed/carboplatin alone.[192]

In the KEYNOTE-010 trial, which compared pembrolizumab with docetaxel in 1034 patients with previously treated NSCLC who had PD-L1 expression on at least 1% of tumor cells, pembrolizumab resulted in longer overall survival and had a more favorable benefit-to-risk profile. In this randomized, open-label trial, median overall survival was 10.4 months with pembrolizumab 2 mg/kg, 12.7 months with pembrolizumab 10 mg/kg, and 8.5 months with docetaxel.[193]

In the KEYNOTE-189 trial, the addition of pembrolizumab to standard chemotherapy with pemetrexed and a platinum-based agent resulted in significantly longer overall and progression-free survival. The trial involved 616 patients with metastatic nonsquamous NSCLC without sensitizing EGFR or ALK mutations who had received no previous treatment for metastatic disease. Study patients received either 200 mg of pembrolizumab or placebo every 3 weeks for four cycles, followed by pembrolizumab or placebo for up to a total of 35 cycles plus pemetrexed maintenance therapy.[194]

In KEYNOTE-189, 12-month survival was 69% with pembrolizumab versus 49% with placebo; median progression-free survival was 8.8 vs. 4.9 months, respectively. Survival benefit was seen in all subgroups receiving pembrolizumab, including those with a PD-L1 tumor proportion score of less than 1%, although the benefit was greater in patients with a higher score.[194]

Approval for treatment of metastatic squamous NSCLC with pembrolizumab in combination with carboplatin and either paclitaxel or nab-paclitaxel was supported by the KEYNOTE-407 study. The combination of pembrolizumab plus chemotherapy (carboplatin and paclitaxel or nab-paclitaxel) significantly improved overall survival (OS) compared with chemotherapy alone, at a median of 15.9 months compared with 11.3 months (hazard ratio [HR], 0.64; P = 0.0017).[169]

Atezolizumab

In October 2016, atezolizumab (Tecentriq) was approved for patients with metastatic NSCLC who have disease progression during or following platinum-containing chemotherapy. Patients with EGFR or ALK genomic tumor aberrations should have disease progression on FDA-approved therapy for these aberrations prior to receiving atezolizumab. Atezolizumab is a monoclonal antibody to PD-L1. Approval was based on the phase III OAK and phase II POPLAR studies. In the OAK study, survival benefit of atezolizumab was compared with docetaxel chemotherapy, regardless of PD-L1 status. Patients receiving atezolizumab lived a median 4.2 months longer than those treated with docetaxel chemotherapy.[195, 196]

In May 2020, atezolizumab was FDA-approved for first-line treatment for metastatic NSCLC in patients whose tumors have high PD-L1 expression (PD-L1 stained ≥50% of tumor cells [TC ≥50%] or PD-L1 stained tumor-infiltrating immune cells [IC] covering ≥10% of the tumor area [IC ≥10%]). Approval was based on the phase III IMpower110 trial, a multicenter, international, randomized, open-label trial. Patients were randomized to receive either atezolizumab or a platinum-based chemotherapy. Median overall survival was 20.2 months in the atezolizumab arm and 13.1 months in the chemotherapy arm, which was a 7.1-month improvement in overall survival in the atezolizumab arm.[197]

Durvalumab

In February 2018, the FDA approved durvalumab for unresectable, stage III NSCLC in patients whose disease has not progressed following concurrent platinum-based chemotherapy and radiation therapy. Approval was based on a randomized trial of 713 patients whose NSCLC had not progressed after completing chemotherapy and radiation. Median progression-free survival for patients taking durvalumab was 16.8 months compared with 5.6 months for patients receiving a placebo (P < 0.001). The median time to death or distant metastasis was longer with durvalumab than with placebo (23.2 months vs. 14.6 months; P< 0.001).[138]

Figitumumab

In a phase II trial, Karp et al studied the effect of the anti–insulinlike growth factor 1 receptor antibody CP-751,871 (figitumumab) combined with paclitaxel and carboplatin in advanced treatment-naive NSCLC. Patients were randomized (2:1) to receive paclitaxel 200 mg/m2, carboplatin (area under the concentration-time curve [AUC] of 6), and CP-751,871 at a dose of 10 or 20 mg/kg (PCI[10] or PCI [20]); or paclitaxel and carboplatin alone (PC) every 3 weeks for up to 6 cycles. The PCI(20)/PC hazard ratio for progression-free survival was 0.8-0.56, indicating safety and efficacy in NSCLC. A randomized, open-label, phase III trial is currently under way.[198]

BRAF-directed therapy

Testing for BRAF mutations should be done. If the BRAF V600E mutation is found, treatment with the combination dabrafenib and trametinib is indicated.

Dabrafenib with trametinib

In June 2017, the FDA approved the combination of dabrafenib (a selective BRAF kinase inhibitor) and trametinib (an MEK1 inhibitor) for targeted treatment of metastatic NSCLC with BRAF V600E mutation.[199] Approval was based on a phase II open-label, nonrandomized study in patients who received dabrafenib 150 mg twice daily plus trametinib 2 mg once daily in continuous 21-day cycles until disease progression.

Of the 93 patients in the study, 36 had received no prior systemic therapy for metastatic NSCLC, and 57 had demonstrated disease progression despite receiving at least one platinum-based chemotherapy regimen. In the previously treated group, the overall response rate (ORR) was 63% and the median duration of treatment response was 9.0 months. The overall disease control rate was 79% when patients who had stable disease for 12 weeks or more were included.[199]

RET inhibitors

Selpercatinib is the first targeted therapy to be approved by the FDA for tumors that have rearranged during transfection (RET) mutations. It is indicated for metastatic RET-fusion–positive NSCLC. 

Accelerated approval in May 2020 for NSCLC was based on the open-label LIBRETTO-001 phase I/II clinical trial (n = 144). ORR was 64% in treatment experienced patients (n = 105) and 85% in treatment naïve patients (n = 39). The phase III confirmatory trial (LIBRETTO-431) is under way.[200]

Immunotherapy-related toxicity

Immune-mediated adverse effects are a common complication of checkpoint inhibitor therapy, with autoimmune toxicity of any grade occurring in approximately 30% of patients and grade 3-5 toxicity occurring in up to 10% of patients. Dermatologic, pulmonary, gastrointestinal, endocrine, neurologic, cardiovascular, and musculoskeletal involvement have all been reported.[201]

A study by Berner et al in 73 patients with NSCLC who received anti–PD-1 therapy with nivolumab or pembrolizumab found that autoimmune skin toxic effects were more frequent in patients with complete remission or partial remission (68.2%) than in those with progressive or stable disease (19.6%). These authors identified nine T-cell antigens that were present in both tumor tissue and skin and were able to stimulate CD8+ and CD4+ T cells in vitro. Several of the antigen-specific T cells found in blood samples were present in autoimmune skin lesions and tumors of patients who responded to anti–PD-1 therapy.[202]

Treatment of immunotherapy-related toxicity depends on its severity and the organ system involved. Corticosteroid therapy is indicated for most symptomatic toxicity. Holding immunotherapy may be indicated.[203]

National Comprehensive Cancer Network (NCCN) guidelines include the following recommendations on initiation of corticosteroid therapy for specific immunotherapy-related toxicities[203] :

Stage-Based Management

Stage IA/IB (T1aN0M0 , T1bN0M0, T2aN0M0)

Surgery is the treatment of choice for stage I non–small cell lung cancer (NSCLC). A careful assessment of residual pulmonary reserve should be carried out as part of surgical planning. Although lobectomy is generally considered to be the optimal procedure, patients with limited pulmonary reserve may be considered for more limited intervention with either a segmental or a wedge resection. The risk of local recurrence is higher with limited resection, but no adverse effect on overall survival was reported in a randomized trial by the Lung Cancer Study Group.

Video-assisted thoracoscopic surgery (VATS) may be used for surgical resection. VATS offers apparently similar resection capability and decreased postoperative morbidity.

Patients with insufficient pulmonary reserve to undergo a resection may be treated with radiation therapy alone with curative intent. Retrospective data report a 5-year survival ranging from 10-25% with radiation therapy alone in this setting. Selected patients may be candidates for either stereotactic body radiotherapy or radiofrequency ablation for isolated lesions.

The role of postoperative radiation has been explored and a meta-analysis of 9 randomized trials revealed a reduction in overall survival with postoperative radiation therapy in stage I and II NSCLC. However, it remains to be seen whether the use of modern radiation techniques with better definition of target volume and cardiac sparing could alter these outcomes.

Adjuvant chemotherapy has been extensively explored in NSCLC.[204] A meta-analysis concluded that adjuvant cisplatin-based therapy improved survival in resected stage IB, II, and III NSCLC. The absolute benefit in survival at 5 years was 6.9%, but in subset analyses, the benefit in stage IB was not statistically significant. No impact of age, sex, histology, or type of surgery was noted. The CALGB 9633 study randomized resected stage IB patients to 4 cycles of carboplatin-paclitaxel versus observation.

This study initially showed improved overall survival at 4 years (71% vs 59%), but a longer term follow-up at 74 months showed no change in overall survival, except in patients with a tumor size greater than 4 cm.[205]

This contrasted with the results of a Canadian study that showed significantly improved 5-year survival for stage IB and II patients treated with adjuvant cisplatin-vinorelbine for 4 cycles. Patients with resected stage IB NSCLC should, therefore, be counseled about risks and benefits of adjuvant chemotherapy and may be offered either 4 cycles of platinum-based doublet chemotherapy, preferably cisplatin, or observation.

Stage IIA/IIB (T2bN0M0, T1aN1M0, T1bN1M0, T1bN1M0, T2aN1M0, T2bN1M0, T3N1M0)

Surgical resection is the treatment of choice for stage II NSCLC, except for those patients who are not surgical candidates because of comorbid conditions or poor pulmonary reserve.

Long-term survival of 10-25% has been reported in patients with radiation therapy alone delivered with curative intent. In such cases, however, the dose of radiation therapy should be approximately 60 Gy, with careful planning to define tumor volume and avoid critical structures. Frequently a cone-down boost is used to enhance local control.

Patient with resected stage II disease are candidates for platinum-based adjuvant chemotherapy (see below) and should be offered four cycles of platinum-based adjuvant chemotherapy.

Stage IIIA (T1aN2M0, T1bN2M0, T2aN2M0, T3N1M0, T3N2M0, T4N1M0)

The management of stage IIIA NSCLC is quite controversial. Surgical resection, chemotherapy, radiation therapy, or a combination of any of these modalities may be the optimal choice, depending on the clinical situation. Overall 5-year survival of stage IIIA (N2) ranges from 10-15%, as resectability rates are low and very few patients (5-10%) achieve long-term benefit with radiation therapy alone. Consequently, stage IIIA has been an area of active research.

Patients who have mediastinal node involvement (N2 or N3 stage) have poor results from surgery and hence should be considered for definitive chemoradiation therapy. Cisplatin-based combinations (eg, with etoposide) are preferred, with carboplatin an acceptable alternative in patients with contraindications to cisplatin. Radiation is usually given in daily fractions for a total of 60 Gy. A cone-down boost may be useful.

Hyperfractionation schedules appear to be better, but are not widely available. Further chemotherapy or surgery does not appear to provide significant survival benefit for in patients treated upfront with chemoradiation.

A large randomized trial conducted by the European Organisation for Research and Treatment of Cancer (EORTC) compared surgery versus radiation therapy following neoadjuvant chemotherapy and found no significant difference between the two approaches in stage IIIA N2 disease. Neoadjuvant chemotherapy followed by surgery may, however, be considered for younger patients with stage IIIA disease who have good performance status.

Patients with stage III (T3-4, N1) disease of the superior sulcus are usually treated with neoadjuvant chemotherapy followed by surgical resection Two-year survival in this group is 50-70%.

Patients with stage IIIA (T3, N1) disease who are candidates for surgical resection should be offered adjuvant chemotherapy after a definitive surgical resection, based on the results of the International Adjuvant Lung Trial (IALT) and meta-analysis of adjuvant chemotherapy trials showing a hazard ratio of 0.87 with adjuvant chemotherapy. These patients should also undergo a mediastinal node dissection. In patients with positive margins, radiation therapy may be considered concurrently with chemotherapy.

Several retrospective series have suggested that postoperative radiation therapy may improve local control in patients with involved mediastinal nodes. Prospective trials have yielded conflicting results with regard to reduction in local recurrence with postoperative radiation therapy. A meta-analysis of 9 randomized trials of postoperative radiation therapy did not find a survival benefit in the entire group or in the subgroup with N2 disease.

Two small reports have shown improvement in disease-free and overall survival rates with neoadjuvant cisplatin-based chemotherapy for stage IIIA NSCLC; this approach may be considered in patients with good performance status. This approach may also be employed for patients who have tumors that are too large for a radiation port, prior to definitive chemoradiation.

A phase III trial compared prophylactic cranial irradiation (PCI) versus observation in patients with stage III NSCLC.[206] The study determined that among patients with stage III disease who did not have progression of disease after treatment with surgery or radiation therapy, PCI decreased the rate of brain metastasis but did not improve overall survival or disease-free survival. No significant differences in global cognitive function or quality of life were noted after PCI; however, a significant decline in memory was noted at 1 year.[207]

Stage IIIB (T1aN3M0, T1bN3M0, T2aN3M0, T2bN3M0, T3N3M0, T4N2M0, T4N3M0)

Patients with satellite lesions (T4 N0-1) should undergo a surgical resection if possible, followed by adjuvant chemotherapy. All other patients with stage IIIB disease are usually not candidates for surgical resection and are best managed with chemotherapy, combined chemoradiation therapy, or radiation therapy alone, depending on extent of disease, sites of involvement, and performance status of the patient. Patients who have malignant pleural effusion are not candidates for radiation therapy and are managed as stage IV (see below).

In an open-label, phase III study in chemotherapy-naive patients with stage IIIB NSCLC (n=676), cetuximab combined with first-line taxane/carboplatin chemotherapy did not reach statistically significant improvement for progression-free survival or overall survival; however, significant improvement was shown in overall response rate.[208]

The NVALT3 study found that chemotherapy in patients aged 70 years or older treated with carboplatin/paclitaxel or carboplatin/gemcitabine did not have a decline in their quality of life.[209]

A meta-analysis of 10 randomized trials of combined chemoradiation therapy revealed a 10% reduction in risk of death with combined modality therapy compared with radiation alone. It appears that in appropriate candidates (with good performance status), chemotherapy given concurrently with radiation results in superior survival compared with chemotherapy followed by radiation therapy.

Patients with stage IIIB NSCLC and poor performance status are not good candidates for chemotherapy or a combined-modality approach. These patients may benefit from radiation therapy alone to palliate the symptoms of shortness of breath, cough, and hemoptysis. Patients with invasive airway obstruction may be candidates for palliative endobronchial curettage or stenting to relieve obstructive atelectasis and dyspnea.

Stage IV (Any T, any N, M1a; Any T, any N, M1b)

Patients with advanced NSCLC should be evaluated for the presence of distant metastases. Patients with solitary brain lesions may benefit from a surgical resection, or stereotactic radiosurgery, if their primary disease is well controlled. Isolated adrenal masses should be resected, since many adrenal masses are benign and even oligometastatic adrenal disease can occasionally be well controlled.

In a small study, patients with isolated adrenal metastasis from lung cancer treated with surgical resection compared with nonoperative management have a better 5-year survival (34% vs 0%).[210]

Isolated synchronous nodules (either in the same or the opposite lung) should be treated as two separate primaries. These patients may need PET scanning to identify occult metastases or serial scans prior to definitive surgical resection that may be counterproductive.

In first-line systemic therapy for stage IV disease, cisplatin-based regimens have provided clear evidence of improved median survival and reductions in risk of death. Patients with good performance status should be offered chemotherapy with a platinum-based combination. Older patients (>70 y) or those with contraindications may be treated with a carboplatin-based regimen, such as carboplatin-paclitaxel. Younger patients (< 70 y) with nonsquamous histology may be candidates for treatment with cisplatin-pemetrexed, which appears to be somewhat better than cisplatin-gemcitabine.

Patients with nonsquamous histology, absence of cranial metastases, and no hemoptysis may be candidates for treatment with bevacizumab, which has been studied in combination with carboplatin-paclitaxel and cisplatin-gemcitabine. Antiangiogenic therapy is very expensive and potentially toxic even in carefully selected patients; hence, a detailed discussion with patients about its modest benefits versus the risks and costs is important.

Two-drug combinations have been found to be superior to single-agent treatment. However, no therapeutic advantage is obtained with the use of three drugs.

No clear survival benefit is observed from maintenance non–cross-resistant chemotherapy, though this is being studied with agents such as pemetrexed. For example, a randomized, placebo-controlled, double-blind phase 3 study of maintenance therapy with pemetrexed in 663 patients with stage IIIB or IV disease who had not progressed on four cycles of platinum-based chemotherapy found that although pemetrexed therapy was associated with toxic effects, it significantly improved progression-free survival and overall survival compared with placebo.[211]

In other randomized, double-blind, placebo-controlled studies, quality of life during maintenance therapy with pemetrexed was similar to that seen with placebo, although a small increase in loss of appetite was observed. Pemetrexed maintenance therapy significantly delayed worsening of pain and hemoptysis and was associated with improvements in overall and progression-free survival, making it a treatment option for patients with advanced nonsquamous NSCLC who have not progressed after induction therapy with platinum-based regimens.[212, 213]

Small-molecule EGFR tyrosine kinase inhibitors such as gefitinib and erlotinib may benefit nonsmokers with adenocarcinomas, particularly bronchoalveolar carcinoma; women of Asian origin are particularly likely to benefit. In such patients, it may be helpful to evaluate for EGFR mutations and use these agents first line.

Similarly, patients with EGFR expression and absence of K -ras mutations may be considered for the addition of cetuximab to first-line chemotherapy. However, the combination of anti–vascular endothelial growth factor (VEGF) agents such as bevacizumab with anti-EGFR antibodies appears to detrimental in other settings, and its use in NSCLC should be avoided.

Patients with progressive disease and good performance status may be candidates for treatment with single cytotoxic drugs such as docetaxel, pemetrexed, or gemcitabine, if they were not exposed to these drugs in the first-line setting. Selected patients can also be treated with erlotinib, though this is typically used in the third-line setting.

Patients with ECOG/Zubrod performance scores greater than 2 should be considered for palliative care, focused on symptom control. These patients should be recommended for enrollment in hospice care programs.

American Society of Clinical Oncology (ASCO) guidelines on chemotherapy for stage IV disease includes the following recommendations[214] :

First-line therapy

Maintenance therapy 

Second-line therapy 

Third-line therapy

Patients with large-cell neuroendocrine carcinoma should receive platinum plus etoposide or the same treatment as other patients with nonsquamous carcinoma.

Physical Activity

A patient's activity level, as measured by a performance status scale (eg, Zubrod, Karnofsky) is an important prognostic factor. Patients should be encouraged to remain active during and after treatment for lung cancer. A declining activity level usually signifies progressive or recurrent disease but also may be due to adverse effects of treatment.

 

Prevention of NSCLC

Smoking cessation

Cigarette smoking is the most common etiologic factor for lung cancer; thus, the primary way to decrease the prevalence of lung cancer is to decrease the prevalence of smoking. Some measures for doing so include the following:

Combining nicotine replacement, bupropion, and social or behavioral support can increase the quit rate to 35%.[215]

Although the relative risk of cancer does not decline to baseline levels for as long as 10 years after cessation, linked conditions (eg, chronic bronchitis, chronic obstructive pulmonary disease) show more rapid improvement or stabilization.[216]

Other measures

Workers exposed to asbestos or radioactive materials should always wear required safety equipment.

Some studies have shown a reduction in lung cancer incidence with daily use of aspirin. This reflects similar studies showing as association between use of nonsteroidal anti-inflammatory drugs (NSAIDs) and a reduced incidence of colorectal cancer and adenoma.[217]

Screening

Prevention is the more effective modality for decreasing the prevalence of NSCLC. However, several organizations recommend screening with low-dose computed tomography in patients at high risk for lung cancer. See the Screening subsection in Workup.

 

 

Consultations

The management of lung cancer is best achieved with a multidisciplinary approach; therefore, after diagnosis, consultations should be sought from the following specialists:

To address complications caused by spread of the disease, consultation with one or more of the following services may be needed:

Long-Term Monitoring

Recommendations from the National Comprehensive Cancer Network (NCCN) regarding cancer surveillance in survivors of non–small cell lung cancer (NSCLC) include the following[91] :

Other NCCN recommendations for long-term monitoring include the following[91] :

Guidelines Summary

Lung Cancer Screening

Guidelines on lung cancer screening have been issued by the following organizations:

The guidelines are in agreement that annual screening with low-dose, computed tomography (LDCT) scanning should be offered to patients aged 55 to 74 years and who have at least a 30 pack-year smoking history and either continue to smoke or have quit within the past 15 years.[65, 46, 64, 66, 218]

The USPSTF extends the recommended age range to 80 years, while the NCCN extends screening to 77 years.[66, 64]  

The American Association for Thoracic Surgery (AATS) extends the recommended age range to 79 years and also recommends annual screening starting at age 50 for patients who have a 20 pack-year smoking history and additional comorbid conditions that produce a cumulative risk for cancer of at least 5% over the next 5 years. Additionally, it recommends annual screening in long-term cancer survivors aged 55 to 79 years.[218]

The NCCN guidelines also recommend screening starting at age 50 in patients with at least a 20 pack-year smoking history and one or more of the following risk factors[64] :

The groups also agree that the shared decision making is required and should include a discussion of benefits and risks.

None of the guidelines recommend screening asymptomatic patients for lung cancer with chest radiograph or sputum cytology.

See Small Cell Lung Cancer: Beating the Spread, a Critical Images slideshow, to help identify the key clinical and biologic characteristics of small cell lung cancer, the staging criteria, and the common sites of spread.

ACCP Diagnosis and Management Guidelines

The American College of Chest Physicians (ACCP) updated its comprehensive set of lung cancer guidelines in 2013. The guideline set of more than 275 recommendations includes an executive summary of current recommendations for diagnosis and treatment, along with additional recommendations for screening, chemoprevention and treatment of tobacco use in patients with lung cancer.[81]

Diagnosis of Pleural Abnormalities

The updated ACCP guidelines recommendations for diagnosis of pleural abnormalities include the following[81] :

Diagnosis of Primary Tumor

The updated ACCP guidelines recommendations for diagnosis of primary tumor include the following[81] :

Treatment of Clinical Stage I and II NSCLC

The updated ACCP guidelines recommendations for treatment of clinical stage I and II NSCLC include[81] :

Treatment of Clinical Stage III NSCLC

The updated ACCP guidelines recommendations for treatment of clinical Stage III NSCLC include the following[81] :

Treatment of Clinical Stage IV NSCL

The updated ACCP guidelines recommendations for treatment of clinical stage IV NSCLC include the following[81] :

Palliative Care

The updated ACCP guidelines recommendations for palliative and end-of-life care include the following[81] :

Molecular Testing and Treatment

International evidence-based guidelines jointly published by the College of American Pathologists (CAP), the International Association for the Study of Lung Cancer (IASLC), and the Association for Molecular Pathology (AMP) in 2013 recommend all lung cancer patients with adenocarcinomas should be tested for the genetic abnormalities that indicate suitability for treatment with targeted agents, irrespective of clinical variables such as sex, ethnicity, or smoking status.[57]  These guidelines were endorsed by the American Society for Clinical Oncology (ASCO) in 2014 [219]

National Comprehensive Cancer Network recommendations

In patients with adenocarcinoma, large cell NSCLC, and NSCLC not otherwise specified, the NCCN recommends the following molecular testing, conducted as part of broad molecular profiling[91] :

In patients with squamous cell NSCLC, the NCCN recommends the following molecular testing, conducted as part of broad molecular profiling:

For patients with a sensitizing EGFR mutation, the NCCN recommends the following for first-line therapy:

For ALK rearrangement–positive patients, the NCCN recommends the following for first-line therapy:

For subsequent therapy in ALK rearrangement–positive patients, the NCCN recommends the following:

For ROS1 rearrangement–positive patients, the NCCN recommends the following for first-line therapy:

For BRAF V600E mutation–positive patients, the NCCN recommends dabrafenib/trametinib for first-line as well as subsequent therapy.

For NTRK gene fusion–positive patients, the NCCN recommends larotrectinib or entrectinib for first-line as well as subsequent therapy.

For patients with PD-L1 ≥1%, the NCCN recommends the following for first-line therapy:

For MET exon 14 skipping mutation–positive patients, the NCCN recommends capmatinib or crizotinib for first-line as well as subsequent therapy.

For MET rearrangment–positive patients, the NCCN recommends selpercatinib, cabozantinib, or vandetanib for first-line as well as subsequent therapy.

ESMO recommendations

Guidelines from the European Society for Medical Oncology (ESMO) contain the following recommendations on molecular testing in patients wqith NSCLC[220] :

In patients with metastatic NSCLC with positive molecular tests, ESMO treatment recommendations are as follows:

 

 

 

 

Video-Assisted thoracoscopy surgery (VATS)

NCCN practice guidelines recommend that VATS or minimally invasive surgery (including robotic-assisted approaches) be strongly considered for patients who have no anatomic or surgical contraindications, provided that standard oncologic and dissection principles of thoracic surgery are not compromised. The guideline notes that in high-volume centers with significant VATS experience, VATS lobectomy in selected patients results in improved early outcomes (ie, decreased pain, reduced hospital length of stay, more rapid return to function, fewer complications) without compromise of cancer outcomes.[91]

Chemotherapy for Stage IV Disease

The American Society of Clinical Oncology (ASCO) issued guidelines on chemotherapy for stage IV NSCLC in 2015[214] and updated them in 2017.[221] For patients with performance status (PS) 0 to 1 who receive chemotherapy (and for selected well-informed patients with PS 2 who desire aggressive treatment), ASCO recommends therapy with a combination of two cytotoxic drugs. Platinum combinations, unless contraindicated, are recommended over nonplatinum therapy. In addition, ASCO recommends early palliative care assistance for all patients with stage IV NSCLC, because it improves survival and well-being in patients with this incurable condition.[214]

First-line therapy

First-line therapy recommendations for patients with negative or unknown tumor EGFR-sensitizing mutation, ALK or ROS1 gene rearrangement status, and PS 0-1 (or possibly PS 2) include the following[221] :

Cisplatin-based two-drug combinations include the following as the second drug:

Carboplatin-based two-drug combinations include the following as the second drug:

For patients receiving carboplatin plus paclitaxel, the guidelines recommend adding bevacizumab, 15 mg/kg once every 3 weeks, and continuing it as tolerated until disease progression, Exceptions include patients with any of the following:

For patients with PS 2, non-SCC, and negative or unknown tumor EGFR-sensitizing mutation and ALK or ROS1 gene rearrangement status, the guidelines recommend combination therapy, single-agent therapy, or palliative therapy alone, selected on the basis of shared decision making.

For patients SCC, negative or unknown tumor EGFR-sensitizing mutation, ALK or ROS1 gene rearrangement status, and PS 0-1 (or possibly PS 2), the guidelines recommend the following:

Cisplatin-based two-drug combinations include the following as the second drug:

Carboplatin-based two-drug combinations include the following as the second drug:

For patients with a sensitizing EGFR mutation, first-line options are as follows:

Second-line therapy

Recommendations for patients with SCC and non-SCC disease and negative or unknown EGFR mutation, ALK gene rearrangement status, or ROS1 gene rearrangement status are as follows:

For patients with a sensitizing EGFR mutation who experienced disease progression with a first-line EGFR tyrosine kinase inhibitor (TKI) recommendations include the following:

For patients with ROS1 rearrangement, recommendations include the following:

For patients with BRAF mutations, recommendations include the following:

Third-line therapy

Recommendations for third-line therapy include the following"

Radiation Therapy for Locally Advanced Disease

In 2015, the American Society for Radiation Oncology (ASTRO) released evidence-based guidelines for definitive and adjuvant radiotherapy in locally advanced NSCLC.[222, 223]  The American Society for Clinical Oncology has endorsed these guidelines.[224]

The guidelines cover patients with stage II or III locally advanced NSCLC whose disease is unresectable, and patients with stage II or III disease who are eligible for surgery. Key recommendation include the following[222, 223] :

Curative-intent Treatment

Adjunctive Therapy

COVID-19

The American College of Surgeons has released a guideline on COVID-19–related triage of patients with thoracic cancer.  As a general recommendation, the guideline recommends that determination of case status (ie, risk of death time frame) be made by Division, ideally in a multi-clinician setting (case review conference).

Suggested consent language: You are being offered surgery now, because at this time we feel that your risk of being harmed by infections, including coronavirus, within the hospital is low, and that delaying surgery could reduce your chances of being cured of cancer.  It is not possible to know either the risk of delaying surgery or the chance of getting an infection with perfect accuracy, but I did consult my colleagues and it is our group’s opinion that surgery is a reasonable thing to do.

Specific guideline recommendations are divided into three phases, depending on the COVID-19 status at a given hospital.

Phase I – Semi-urgent Setting (Preparation Phase)

Features of this phase are as follows:

In phase I, surgery should be restricted to patients whose survival is likely to be compromised if surgery not performed within next 3 months. The following cases need to be done as soon as feasible (recognizing that the status of each hospital is likely to evolve over next week or two):

Cases that should be deferred include the following:

The following alternative treatment approaches can be considered (assuming resources permit):

Phase II  Urgent Setting

Features of this phase are as follows:

Surgery should be restricted to patients whose survival is likely to be compromised if surgery is not performed within the next few days. Cases that need to be done as soon as feasible (recognizing that the hospital’s status is likely to progress over next few days):

All thoracic procedures typically scheduled as routine/elective (ie, not add-ons) should be deferred.

Alternative treatment approaches that are recommended, assuming resources permit, are as follows:

Phase III

In this phase, hospital resources are all routed to COVID-19 patients, the hospital has no ventilator or ICU capacity, and OR supplies are exhausted. Surgery should be restricted to patients whose survival is likely to be compromised if surgery is not performed within next few hours.

Cases that need to be done as soon as feasible (status of hospital likely to progress in hours) are as follows:

All other cases should be deferred. Recommended alternative treatments are the same as for Phase II.

Medication Summary

Chemotherapy is used as an adjuvant to surgery in selected patients with non–small cell lung cancer (NSCLC). Unresectable NSCLC is treated with chemotherapy or a combination of chemotherapy and radiation therapy. Molecularly targeted treatments have also become standard of care.

Aggressive antiemetic support and growth-factor support, when appropriate, are other integral parts of medical treatment of these patients. Antibiotics are commonly required for treatment of infectious complications but are not discussed in this article. Aggressive antiemetic support to prevent, not treat, nausea and vomiting is essential because of the highly emetogenic potential of chemotherapy drugs and the doses used in the treatment of NSCLC. This holds especially true for platinum-based chemotherapeutic regimens. The most common and effective agents are corticosteroids and the serotonin receptor antagonists, which include ondansetron, granisetron, and dolasetron.

Carboplatin

Clinical Context:  Carboplatin has a mechanism of action similar to that of cisplatin. It is approved for ovarian cancer but is used commonly in squamous cell carcinoma (SCC) of the head, neck, cervix, and lungs. Its main advantages over cisplatin include less nephrotoxicity and ototoxicity (not requiring extensive prehydration) and reduced likelihood of inducing nausea and vomiting. It is more likely to induce myelotoxicity.

Vinorelbine (Navelbine)

Clinical Context:  Vinorelbine is a semisynthetic vinca alkaloid that inhibits tubulin polymerization during the G2 phase of cell division, thereby inhibiting mitosis. Vinorelbine alone or in combination with cisplatin is indicated as first-line treatment of ambulatory patients with unresectable, advanced NSCLC and for stage IV NSCLC. In stage III NSCLC, vinorelbine is indicated in combination with cisplatin.

Paclitaxel

Clinical Context:  Paclitaxel is a naturally occurring chemical derived from the Pacific yew tree (Taxus brevifolia). It inhibits tubulin depolymerization in the spindle during cell division. Paclitaxel is used in combination with cisplatin for the first-line treatment of NSCLC in patients who are not candidates for potentially curative surgery or radiation therapy.

Paclitaxel protein bound (Abraxane)

Clinical Context:  Protein-bound paclitaxel is a microtubular inhibitor (albumin-conjugated formulation). It is a natural taxane, and it prevents depolymerization of cellular microtubules, which results in DNA, RNA, and protein synthesis inhibition. It is indicated for locally advanced or metastatic non–small cell lung cancer (NSCLC), as first-line treatment in combination with carboplatin, in patients who are not candidates for curative surgery or radiation therapy.

Cisplatin

Clinical Context:  Cisplatin is an alkylating agent that causes intrastrand and interstrand cross-linking of DNA, leading to strand breakage. It has a very broad range of antitumor activity and is approved for use in testicular, ovarian, and transitional cell carcinomas. It forms the basis of currently available approved combination chemotherapy regimens for NSCLC. Administer it as a single-dose intravenous (IV) infusion or in divided doses over several days; this can be repeated only after complete hematologic recovery; cycles are typically separated by 3-4 wk.

Gemcitabine (Gemzar)

Clinical Context:  Gemcitabine is an antimetabolite that acts as an inhibitor of DNA synthesis. It is cell-cycle specific for the S phase. Gemcitabine is used as first-line treatment of patients with inoperable, locally advanced (stage IIIA or IIIB), or metastatic (stage IV) NSCLC in combination with cisplatin.

Docetaxel (Taxotere)

Clinical Context:  Docetaxel is a semisynthetic taxane, a class of drugs that inhibits cancer cell growth by promoting assembly and blocking disassembly of microtubules, thereby preventing cancer cell division, leading to cell death. It is indicated for the treatment of patients with locally advanced or metastatic NSCLC after failure of prior platinum-based chemotherapy when used alone. It can be used in combination with cisplatin for the treatment of patients with unresectable, locally advanced, or metastatic NSCLC who have not previously received chemotherapy for this condition.

Pemetrexed disodium (Alimta)

Clinical Context:  Pemetrexed disrupts folate-dependent metabolic processes essential for cell replication. It specifically inhibits thymidylate synthase (TS), dihydrofolate reductase (DHFR), and glycinamide ribonucleotide formyltransferase (GARFT), which are folate-dependent enzymes involved in de novo biosynthesis of thymidine and purine nucleotides. It is indicated for nonsquamous NSCLC as follows:

1) Initial treatment in combination with pembrolizumab and platinum chemotherapy for patients with metastatic disease with no EGFR or ALK genomic tumor aberrations

2) Initial treatment in combination with cisplatin for patients with locally advanced or metastatic disease

3) Single agent for maintenance in patients with locally advanced or metastatic disease that has not progressed after 4 cycles of platinum-based first-line chemotherapy

4) Single agent for treatment of patients with recurrent metastatic disease after prior therapy

Cyclophosphamide

Clinical Context:  Cyclophosphamide is chemically related to nitrogen mustards; as an alkylating agent, the mechanism of action of active metabolites may involve cross-linking of DNA, which may interfere with growth of normal and neoplastic cells.

Doxorubicin

Clinical Context:  Doxorubicin is an anthracycline that inhibits topoisomerase II and produces free radicals, which may cause destruction of DNA; the combination of these 2 events can, in turn, inhibit the growth of neoplastic cells.

Vincristine

Clinical Context:  Vincristine is a vinca alkaloid whose mechanism of action is uncertain. It may involve a decrease in reticuloendothelial cell function or an increase in platelet production. However, neither of these mechanisms fully explains the effect in thrombotic thrombocytopenic purpura and hemolytic uremic syndrome. The antineoplastic effects of vincristine are related to its binding to tubulin and inhibition of microtubule formation.

Etoposide

Clinical Context:  Etoposide inhibits topoisomerase II and causes DNA strand breakage, causing cell proliferation to arrest in the late S or early G2 portion of cell cycle; do not administer IT.

Ramucirumab (Cyramza)

Clinical Context:  Ramucirumab is a vascular endothelial growth factor receptor 2 (VEGFR2) antagonist. This action results in inhibition of ligand-induced proliferation, and migration of human endothelial cells. It is indicated for metastatic non-small cell lung cancer (NSCLC) with disease progression on or after platinum-based chemotherapy. Patients with EGFR or ALK genomic tumor aberrations should have disease progression on FDA-approved therapy for these aberrations prior to receiving ramucirumab.

Class Summary

Antineoplastic agents are used either to prolong survival or to palliate symptoms in advanced or unresectable lung cancer.

Crizotinib (Xalkori)

Clinical Context:  Inhibitor of receptor tyrosine kinases including, anaplastic lymphoma kinase (ALK), hepatocyte growth factor receptor (HGFR, c-Met), and recepteur d'origine nantais (RON). The gene's expression and signaling that contribute to increased cell proliferation and survival of the tumors becomes activated following the expression of ALK oncogenic fusion proteins. Indicated for locally advanced or metastatic non-small cell lung cancer (NSCLC) that is ALK positive as detected by an FDA-approved test. It is also indicated for NSCLC tumors positive for the ROS-1 gene mutation.

Ceritinib (Zykadia)

Clinical Context:  Ceritinib is a tyrosine kinase inhibitor that targets anaplastic lymphoma kinase (ALK), insulin-like growth factor 1 receptor (IGF-1R), insulin receptor (InsR), and ROS1. It is indicated for ALK-positive, metastatic NSCLC.

Alectinib (Alecensa)

Clinical Context:  Tyrosine kinase inhibitor that targets ALK and RET. The major active metabolite of alectinib, M4, showed similar in vitro potency and activity. In nonclinical studies, alectinib inhibited ALK phosphorylation and ALK-mediated activation of the downstream signaling proteins STAT3 and AKT, and decreased tumor cell viability in multiple cell lines harboring ALK fusions, amplifications, or activating mutations.. It is indicated for the treatment of patients with anaplastic lymphoma kinase (ALK)-positive, metastatic non-small cell lung cancer (NSCLC) as detected by an FDA-approved test.

Brigatinib (Alunbrig)

Clinical Context:  Brigatinib inhibits autophosphorylation of ALK and ALK-mediated phosphorylation of the downstream signaling proteins STAT3, AKT, ERK1/2, and S6 in in vitro and in vivo assays. It is indicated for ALK-positive metastatic NSCLC in patients who have progressed on or are intolerant to crizotinib.

Lorlatinib (Lorbrena)

Clinical Context:  ALK/ROS1 tyrosine kinase inhibitor indicated for patients with ALK-positive metastatic NSCLC who have progressed on crizotinib and at least 1 other ALK inhibitor, or, alectinib or ceritinib as first ALK inhibitor therapy.

Class Summary

Testing for ALK rearrangement is recommended in patients with metastatic NSCLC adenocarcinoma to determine if a therapy targeted toward ALK would be beneficial.

Nivolumab (Opdivo)

Clinical Context:  Monoclonal antibody to programmed cell death-1 protein (PD-1). It blocks the interaction between PD-1 and its ligands, PD-L1 and PD-L2. Restores cytokine secretion and T cell activity. It is indicated for metastatic squamous and nonsquamous (including adenocarcinoma) NSCLC with progression on or after platinum-based chemotherapy. It is also indicated in combination with ipilimumab, for metastatic NSCLC in patients whose tumors express PD-L1 (≥1%), with no EGFR or ALK tumor aberrations.

Pembrolizumab (Keytruda)

Clinical Context:  Pembrolizumab is a monoclonal antibody to programmed cell death-1 protein (PD-1). It blocks the interaction between PD-1 and its ligands, PD-L1 and PD-L2. It is indicated as a single agent for first-line treatment of patients with stage III NSCLC who are not candidates for surgical resection or definitive chemoradiation, or metastatic NSCLC whose tumors express PD-L1 (Tumor Proportion Score [TPS] ≥1%) as determined by an FDA-approved test, with no EGFR or ALK genomic tumor aberrations. Pembrolizumab is also indicated as a single agent for the treatment of patients with metastatic NSCLC whose tumors express PD-L1 (TPS ≥1%) as determined by an FDA-approved test, with disease progression on or after platinum-containing chemotherapy; patients with EGFR or ALK genomic tumor aberrations should have disease progression on FDA-approved therapy for these aberrations prior to receiving pembrolizumab.

Pembrolizumab is indicated in combination with pemetrexed and carboplatin for first-line treatment of patients with metastatic nonsquamous NSCLC irrespective of PD-L1 expression. It is also indicated first-line in combination with carboplatin and either paclitaxel or nab-paclitaxel for patients with metastatic squamous NSCLC.

Atezolizumab (Tecentriq)

Clinical Context:  Atezolizumab is a monoclonal antibody to PD-L1. It is indicated for patients with metastatic non-small cell lung cancer (NSCLC) who have disease progression during or following platinum-containing chemotherapy. Patients with EGFR or ALK genomic tumor aberrations should have disease progression on FDA-approved therapy for these aberrations prior to receiving atezolizumab. It is also indicated for first-line treatment for metastatic NSCLC in patients whose tumors have high PD-L1 expression (PD-L1 stained ≥50% of tumor cells [TC ≥50%] or PD-L1 stained tumor-infiltrating immune cells [IC] covering ≥10% of the tumor area [IC ≥10%]).

Durvalumab (Imfinzi)

Clinical Context:  Human IgG1 kappa monoclonal antibody that blocks PD-L1 binding to PD-1 and CD80, and therefore overcoming/preventing PD-L1-mediated inhibition/suppression of T-cell activation. It is indicated for unresectable, Stage III NSCLC in patients whose disease has not progressed following concurrent platinum-based chemotherapy and radiation therapy.

Class Summary

PD-1 and related target PD-ligand 1 (PD-L1) are expressed on the surface of activated T cells. Under normal conditions, PD-L1/PD-1 interaction inhibits immune activation and reduces T-cell cytotoxic activity when bound. This negative feedback loop is essential for maintaining normal immune responses and limits T-cell activity to protect normal cells during chronic inflammation. Tumor cells may circumvent T-cell–mediated cytotoxicity by expressing PD-L1 on the tumor itself or on tumor-infiltrating immune cells, resulting in the inhibition of immune-mediated killing of tumor cells; therefore, inhibiting the ligand binding will enhance T-cell mediated immune response.

Osimertinib (Tagrisso)

Clinical Context:  Kinase inhibitor of the EGFR, which binds irreversibly to certain mutant forms of EGFR (T790M, L858R, and exon 19 deletion). It is indicated for metastatic EGFR T790M mutation positive non-small cell lung cancer NSCLC, as detected by an FDA approved test, in patients who have progressed on or after EGFR TKI therapy. Also, indicated for first-line treatment of patients with metastatic NSCLC whose tumors have EGFR exon 19 deletions or exon 21 L858R mutations, as detected by an FDA-approved test. 

Gefitinib (Iressa)

Clinical Context:  Gefitinib reversibly inhibits the kinase activity of wild-type and certain activating mutations of epidermal growth factor receptor (EGFR), preventing autophosphorylation of tyrosine residues associated with the receptor, thereby inhibiting further downstream signalling and blocking EGFR-dependent proliferation. It is indicated for the treatment of patients with metastatic non-small cell lung cancer (NSCLC) whose tumors have epidermal growth factor receptor (EGFR) exon 19 deletions or exon 21 (L858R) substitution mutations as detected by an FDA-approved test.

Erlotinib (Tarceva)

Clinical Context:  Erlotinib is pharmacologically classified as a HER1/epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI). EGFR is expressed on the cell surface of normal cells and cancer cells. As of late 2016, it is approved for NSCLC in patients whose tumors have EGFR exon 19 deletions or exon 21 (L858R) substitution mutations as detected by an FDA-approved test for first-line treatment, maintenance treatment, or as second or greater line treatment after progression following at least 1 prior chemotherapy regimen.

Afatinib (Gilotrif)

Clinical Context:  Afatinib covalently binds to the kinase domains of EGFR (ErbB1), HER2 (ErbB2), and HER4 (ErbB4) and irreversibly inhibits tyrosine kinase autophosphorylation, resulting in downregulation of ErbB signaling. It demonstrates inhibition of autophosphorylation and in vitro proliferation of cell lines expressing wild-type EGFR or those expressing selected EGFR exon 19 deletion mutations or exon 21 L858R mutations, including some with a secondary T790M mutations

Afatinib is indicated for first-line treatment of patients with metastatic non-small cell lung cancer (NSCLC) who tumors have non-resistant epidermal growth factor receptor (EGFR) mutations as detected by an FDA-approved test. Additionally, afatinib is indicated for first-line use in metastatic NSCLC for 3 additional nonresistant EGFR mutations (ie, L861Q, G719X, S768I). It is also indicated for metastatic squamous NSCLC which has progressed after platinum-based chemotherapy.

Dacomitinib (Vizimpro)

Clinical Context:  Irreversible kinase inhibitor of the human EGFR family (EGFR/HER1, HER2, and HER4) and certain EGFR-activating mutations (exon 19 deletion or the exon 21 L858R substitution mutation). It is indicated for first-line treatment of patients with metastatic non-small lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) exon 19 deletion or exon 21 L858R substitution mutations as detected by an FDA-approved test.

Necitumumab (Portrazza)

Clinical Context:  Monoclonal antibody that binds to the human EFGR and blocks interaction between EGFR and its ligands. It is indicated for first-line treatment of metastatic squamous NSCLC in combination with gemcitabine and cisplatin.

Class Summary

EGFR is expressed on the cell surface of both normal and cancer cells and plays a role in the processes of cell growth and proliferation.

Ondansetron (Zofran, Zuplenz)

Clinical Context:  Ondansetron blocks serotonin 5-HT3 receptor antagonists. It is not clear whether the effect is mediated centrally, peripherally, or both. Ondansetron is indicated in the prevention of chemotherapy-induced nausea and vomiting.

Granisetron (Granisol, Sancuso, Sustol)

Clinical Context:  Granisetron blocks serotonin 5-HT3 receptor antagonists. It is not clear whether the effect is mediated centrally, peripherally, or both. Granisetron is indicated in the prevention of chemotherapy-induced nausea and vomiting.

Dolasetron (Anzemet)

Clinical Context:  Dolasetron blocks serotonin 5-HT3 receptor antagonists. It is not clear whether the effect is mediated centrally, peripherally, or both. Dolasetron is indicated in the prevention of chemotherapy-induced nausea and vomiting.

Palonosetron (Aloxi)

Clinical Context:  Palonosetron is a selective 5-HT3 receptor antagonist with a long half-life (40 h). It is indicated for the prevention of acute and delayed nausea and vomiting associated with initial and repeat courses of chemotherapy. It blocks 5-HT3 receptors peripherally and centrally in the chemoreceptor trigger zone.

Dexamethasone

Clinical Context:  Dexamethasone is a synthetic adrenocortical steroid with multiple indications. It is widely used in prevention of nausea and vomiting caused by highly emetogenic agents (eg, cisplatin) in combination with serotonin receptor antagonists.

Class Summary

Antiemetic agents are useful in the treatment of symptomatic nausea caused by chemotherapy.

Dabrafenib (Tafinlar)

Clinical Context:  Dabrafenib selectively inhibits multiple mutated BRAF kinases, BRAF V600E, BRAF V600K, and BRAF V600D. BRAF kinase inhibition primarily stunts cells growth. The combination with trametinib targets two different kinases in the RAS/ MEK pathway causing greater growth inhibition of BRAF V600 mutation.  

Class Summary

BRAF genes direct protein production which induces signal transmission along the RAS/MAPK pathway. RAS/MAPK pathway regulates cell growth, cell differentiation, cell migration, and apoptosis. BRAF mutations can cause dysregulation of cell growth resulting in tumor progression[225]

Trametinib (Mekinist)

Clinical Context:  Trametinib is a selective reversible inhibitor of mitogen-activated extracellular signal regulated kinase 1 (MEK1) and MEK2 activation and kinase activity. Trametinib inhibits activation of MEK by BRAF and inhibits MEK kinase activity BRAF V600E mutations result in constitutive activation of the BRAF pathway, which includes MEK1 and MEK2. Trametinib inhibits cell growth in BRAF V600 mutation positive tumors in vitro and in vivo. It is indicated, in combination with dabrafenib, for the treatment of patients with metastatic NSCLC with BRAF V600E mutation as detected by an FDA-approved test.

Class Summary

Mitogen extracellular signal-regulated kinase (MEK) protein acts downstream from Ras in the mitogen-activated protein kinase (MAPK) pathway. MAPK pathway regulates cell growth, cell differentiation, cell migration, and apoptosis.[225]  Blockage of MEK protein may affect tumors with mutated Ras proteins or other proteins influenced by MAPK pathway. 

Entrectinib (Rozlytrek)

Clinical Context:  Entrectinib is indicated for metastatic NSCLC in adults whose tumors are ROS1-positive.

Capmatinib (Tabrecta)

Clinical Context:  Capmatinib is a kinase inhibitor that targets mesenchymal-epithelial transition (MET) , including the exon 14 skipping mutation. MET tyrosine kinase stimulates cell scattering, invasion, protection from apoptosis, and angiogenesis. A variety of cancers (eg, lung, gastric) are associated with dysregulation of MET, owing to MET amplifications and exon 14 skipping mutations. Capmatinib is indicated for metastatic NSCLC in adults whose tumors have a mutation that leads to exon 14 skipping.

Class Summary

Entrectinib and its major metabolite inhibit tropomyosin receptor tyrosine kinases (TRKs), proto-oncogene tyrosine-protein kinase ROS1 (ROS1), and anaplastic lymphoma kinase (ALK).Approval of entrectinib for NSCLC was based on pooled analysis of 3 multicenter, single-arm, open-label trials (ALKA, STARTRK-1, STARTRK-2). Of the 51 patients with ROS1-positive confirmed NSCLC who were assessed, ORR was 78% and 55% had a DOR that lasted for at least 12 months. Favorable results were also shown in patients with CNS metastases.[142, 143]

Selpercatinib (Retevmo)

Clinical Context:  Selpercatinib is a kinase inhibitor of wild-type rearranged during transfection (RET) and multiple mutated RET isoforms, as well as vascular endothelial growth factor receptors (VEGFR1, VEGFR3). It is indicated for metastatic RET fusion-positive NSCLC. 

Class Summary

Genomic alterations in rearranged during transfection (RET) kinase, which include fusions and activating point mutations, lead to overactive RET signaling and uncontrolled cell growth.

Bevacizumab (Avastin, Mvasi)

Clinical Context:  Bevacizumab is a murine-derived monoclonal antibody that inhibits angiogenesis by targeting and inhibiting vascular endothelial growth factor (VEGF). It inhibits new blood vessel formation, denying blood, oxygen, and other nutrients needed for tumor growth. It is indicated in combination with carboplatin and paclitaxel for the first-line treatment of patients with unresectable, locally advanced, recurrent, or metastatic nonsquamous NSCLC. Mvasi has been FDA-approved as a biosimilar to Avastin but not as an interchangeable product.

Ipilimumab (Yervoy)

Clinical Context:  Recombinant, human cytotoxic T-lymphocyte antigen. Proposed mechanism of action is indirect, possibly through inhibition of CTLA-4 signaling, which can in turn reduce T-regulatory cell function and may contribute to a general increase in T cell responsiveness, including the anti-tumor immune response. It is indicated in combination with nivolumab, for metastatic NSCLC in patients whose tumors express PD-L1 (≥1%), with no EGFR or ALK tumor aberrations.

What is non–small cell lung cancer (NSCLC)?How frequently is non–small cell lung cancer (NSCLC) diagnosed with metastasis?What are the most common signs and symptoms of non–small cell lung cancer (NSCLC)?What are the metastatic signs and symptoms of non–small cell lung cancer (NSCLC)?Which chest radiography findings suggest non–small cell lung cancer (NSCLC)?How is the diagnosis of non–small cell lung cancer (NSCLC) confirmed?What is the TNM (tumor-node-metastasis) staging system for non–small cell lung cancer (NSCLC)?How is primary tumor (T) involvement in non–small cell lung cancer (NSCLC) staged?How is lymph node (N) involvement in non–small cell lung cancer (NSCLC) staged?How is metastatic (M) involvement in non–small cell lung cancer (NSCLC) staged?What are the treatment options for non–small cell lung cancer (NSCLC)?What is the role of surgery in the treatment of non–small cell lung cancer (NSCLC)?What is the role of chemotherapy in the treatment of non–small cell lung cancer (NSCLC)?What is the role of radiation therapy in the treatment of non–small cell lung cancer (NSCLC)?How is non–small cell lung cancer (NSCLC) categorized?What is the prevalence of lung cancer in the US?What is the mortality associated with non–small cell lung cancer (NSCLC)?Why is the prognosis of non–small cell lung cancer (NSCLC) generally poor?How frequently is non–small cell lung cancer (NSCLC) diagnosed in asymptomatic patients?Why should a staging workup be performed in non–small cell lung cancer (NSCLC)?What are the treatment options for non–small cell lung cancer (NSCLC)?What are the major risk factors for non–small cell lung cancer (NSCLC)?What is the role of the exposure to carcinogens in the pathophysiology of non–small cell lung cancer (NSCLC)?What is the role of genetics in the pathophysiology of non–small cell lung cancer (NSCLC)?What genetic factor may explain why non–small cell lung cancer (NSCLC) often fails to respond to treatment?What is the prevalence of non–small cell lung cancer (NSCLC) among lung cancers?How is the adenocarcinoma subtype of non–small cell lung cancer (NSCLC) characterized?How is squamous cell carcinoma (SCC) subtype of non–small cell lung cancer (NSCLC) characterized?How is large-cell carcinoma subtype of non–small cell lung cancer (NSCLC) characterized?What are the causes of non–small cell lung cancer (NSCLC)?How frequently is tobacco smoking the cause of non–small cell lung cancer (NSCLC)?Other than smoking, what are the risk factors for non–small cell lung cancer (NSCLC)?What is the global prevalence of smoking?How does the incidence of non–small cell lung cancer (NSCLC) vary between smokers and nonsmokers?What are the differences in non–small cell lung cancer (NSCLC) susceptibility between males and females?How quickly does the risk for non–small cell lung cancer (NSCLC) decline after smoking cessation?What causes non–small cell lung cancer (NSCLC) in persons who have never smoked?What is the role of asbestos exposure in the etiology of non–small cell lung cancer (NSCLC)?What is the role of radon exposure in the etiology of non–small cell lung cancer (NSCLC)?What is the role of HIV infection in the etiology of non–small cell lung cancer (NSCLC)?Which metals and chemical elements may have an etiologic role in the development of non–small cell lung cancer (NSCLC)?What is the role of diet in the etiology of non–small cell lung cancer (NSCLC)?What is the incidence of non–small cell lung cancer (NSCLC) in the US?What is the global prevalence of non–small cell lung cancer (NSCLC)?Which patient groups are at highest risk for non–small cell lung cancer (NSCLC)?What is the racial predilections of non–small cell lung cancer (NSCLC)?What are the survival rates for non–small cell lung cancer (NSCLC)?What are prognostic factors for non–small cell lung cancer (NSCLC)?What is the prognosis of in situ and stage I non–small cell lung cancer (NSCLC)?What is the prognostic significance of oncogenic pathway in non–small cell lung cancer (NSCLC)?What are the benefits of smoking cessation after the diagnosis of early-stage non–small cell lung cancer (NSCLC)?What is the effect of hormone therapy on the prognosis of non–small cell lung cancer (NSCLC) in women?What are the effect of daily aspirin on the mortality rates for non–small cell lung cancer (NSCLC)?How does detection of circulating tumor cells (CTCs) affect the prognosis of non–small cell lung cancer (NSCLC)?What is the risk of recurrence in patients treated for non–small cell lung cancer (NSCLC)?What is included in patient education about non–small cell lung cancer (NSCLC)?What are the respiratory signs and symptoms of non–small cell lung cancer (NSCLC)?How do the symptoms of non–small cell lung cancer (NSCLC) vary by subtype?What are the symptoms of non–small cell lung cancer (NSCLC) due to locoregional spread?What is the symptoms of endobronchial non–small cell lung cancer (NSCLC)?What are the symptoms of mediastinal non–small cell lung cancer (NSCLC)?What are the symptoms of pleural non–small cell lung cancer (NSCLC)?What are the neurologic symptoms of non–small cell lung cancer (NSCLC)?What are the symptoms of metastatic non–small cell lung cancer (NSCLC)?What are the CNS signs and symptoms of non–small cell lung cancer (NSCLC)?What are the vascular symptoms of non–small cell lung cancer (NSCLC)?What are the musculoskeletal symptoms of non–small cell lung cancer (NSCLC)?Which paraneoplastic syndromes may occur in non–small cell lung cancer (NSCLC)?Which physical findings are characteristic of non–small cell lung cancer (NSCLC)?What are common sites of distant metastasis in non–small cell lung cancer (NSCLC)?Which physical findings in the head and neck regions are characteristic of non–small cell lung cancer (NSCLC)?What are the signs of superior vena cava syndrome (SVCS) in non–small cell lung cancer (NSCLC)?Which respiratory system findings are characteristic of non–small cell lung cancer (NSCLC)?Which cardiovascular findings are characteristic of non–small cell lung cancer (NSCLC)?Which GI findings are characteristic of non–small cell lung cancer (NSCLC)?Which musculoskeletal findings are characteristic of non–small cell lung cancer (NSCLC)?Which neurologic findings are characteristic of non–small cell lung cancer (NSCLC)?Which physical findings are characteristic of paraneoplastic syndromes in non–small cell lung cancer (NSCLC)?Which conditions should be included in the differential diagnoses of non–small cell lung cancer (NSCLC)?What are the differential diagnoses for Non-Small Cell Lung Cancer (NSCLC)?How is a diagnosis of non–small cell lung cancer (NSCLC) confirmed?What is included in a staging workup for non–small cell lung cancer (NSCLC)?What is the role of ECG and pulmonary function testing in the workup of non–small cell lung cancer (NSCLC)?Which lab studies are indicated in the workup of non–small cell lung cancer (NSCLC)?Which electrolyte abnormalities are commonly identified during the workup for non–small cell lung cancer (NSCLC)?What is the role of liver function tests in the workup of non–small cell lung cancer (NSCLC)?What is the role of arterial blood gas (ABG) testing in the workup of non–small cell lung cancer (NSCLC)?What is the role of chest radiography in the workup of non–small cell lung cancer (NSCLC)?Which chest radiograph findings are characteristic of non–small cell lung cancer (NSCLC)?What is the role of CT scanning in the workup of non–small cell lung cancer (NSCLC)?What is the role of MRI in the workup of non–small cell lung cancer (NSCLC)?What is the role of bone scintigraphy in the workup of non–small cell lung cancer (NSCLC)?What is the role of positron emission tomography (PET) scanning in the workup of non–small cell lung cancer (NSCLC)?What is the role of sputum cytologic studies in the workup of non–small cell lung cancer (NSCLC)?What is the role of bronchoscopy in the workup of non–small cell lung cancer (NSCLC)?What is the role of biopsy in the workup of non–small cell lung cancer (NSCLC)?What is the role of needle thoracentesis in the workup of non–small cell lung cancer (NSCLC)?What is the role of thoracoscopy in the workup of non–small cell lung cancer (NSCLC)?What is the role of mediastinoscopy in the workup of non–small cell lung cancer (NSCLC)?What is the role of molecular testing in the workup of non–small cell lung cancer (NSCLC)?What are the guidelines for molecular testing in the workup of non–small cell lung cancer (NSCLC)?How are molecular testing results used to guide treatment for non–small cell lung cancer (NSCLC)?Which histologic findings indicate the squamous cell carcinoma (SCC) subtype of non–small cell lung cancer (NSCLC)?Which histologic findings indicate the adenocarcinoma subtype of non–small cell lung cancer (NSCLC)?What are preinvasive lesions of epithelial lung tumors in non–small cell lung cancer (NSCLC)?What are invasive malignant lesions of epithelial lung tumors in non–small cell lung cancer (NSCLC)?What is the most important prognostic indicator non–small cell lung cancer (NSCLC)?What is the staging for primary tumor (T) involvement in non–small cell lung cancer (NSCLC)?What is the staging for lymph node (N) involvement in non–small cell lung cancer (NSCLC)?What is the staging for metastatic (M) involvement in non–small cell lung cancer (NSCLC)?What is the stage grouping of the tumor-node-metastasis (TNM) staging system for non–small cell lung cancer (NSCLC)?Which patient groups may require additional workup for non–small cell lung cancer (NSCLC)?What is included in the workup of non–small cell lung cancer (NSCLC) in patients with CNS metastasis and known cancer?What is included in the workup of non–small cell lung cancer (NSCLC) in patients with immunosuppression?What is included in the workup of non–small cell lung cancer (NSCLC) in patients with Pancoast tumor?What is included in the workup of non–small cell lung cancer (NSCLC) in patients with superior vena cava syndrome (SVCS)?What is included in the workup of non–small cell lung cancer (NSCLC) in patients with Ogilvie intestinal pseudo-obstruction?Who should be screened for non–small cell lung cancer (NSCLC)?What is the role of biomarker screening in the workup of non–small cell lung cancer (NSCLC)?What are the guidelines for non–small cell lung cancer (NSCLC) screening?What are the limitations of non–small cell lung cancer (NSCLC) screening?Which risk models are preferred by the NCI and ACS for non–small cell lung cancer (NSCLC) screening?What are the treatment options for non–small cell lung cancer (NSCLC)?When should palliative care be considered in the treatment of non–small cell lung cancer (NSCLC)?What is included in emergency treatment for non–small cell lung cancer (NSCLC)?What is the role of surgery in the treatment of non–small cell lung cancer (NSCLC)?What is involved in preoperative evaluation of non–small cell lung cancer (NSCLC)?What is the standard surgical approach for treatment of non–small cell lung cancer (NSCLC)?What is the role of wedge resection/segmentectomy in the treatment of non–small cell lung cancer (NSCLC)?What is the role of video-assisted thoracoscopic surgery (VATS) in the treatment of non–small cell lung cancer (NSCLC)?What is the role of mediastinal lymphadenectomy in the treatment of non–small cell lung cancer (NSCLC)?What is included in the postoperative evaluation of non–small cell lung cancer (NSCLC), and what are the potential complications following surgery?What is the role of radiation therapy in the treatment of non–small cell lung cancer (NSCLC)?What are prognostic indicators of better outcome for hyperfractionated radiation therapy treatment of non–small cell lung cancer (NSCLC)?What is the efficacy of radiotherapy for the treatment of unresectable stage III non–small cell lung cancer (NSCLC)?What is stereotactic body radiotherapy (SBRT) for the treatment of non–small cell lung cancer (NSCLC)?What is the efficacy of stereotactic body radiotherapy (SBRT) for the treatment of non–small cell lung cancer (NSCLC)?What is the role of radiofrequency ablation (RFA) for the treatment of non–small cell lung cancer (NSCLC)?What is the role of 4-dimensional computed tomography (4DCT) scans in the treatment of non–small cell lung cancer (NSCLC)?What is the role of adjuvant radiation therapy in the treatment of non–small cell lung cancer (NSCLC)?What has caused a paradigm shift in the approach to treatment of non–small cell lung cancer (NSCLC)?How frequently is chemotherapy utilized in the treatment of non–small cell lung cancer (NSCLC)?What is the role of chemotherapy in the treatment of non–small cell lung cancer (NSCLC)?What is the role of neoadjuvant chemotherapy in the treatment of non–small cell lung cancer (NSCLC)?When is chemotherapy considered in the treatment of locally advanced non–small cell lung cancer (NSCLC)?What are the advantages of chemotherapy in the treatment of non–small cell lung cancer (NSCLC)?What are the response rates to chemotherapy in non–small cell lung cancer (NSCLC)?What are the benefits of adjuvant chemotherapy for non–small cell lung cancer (NSCLC)?What is the role of cisplatin in the treatment of advanced non–small cell lung cancer (NSCLC)?What is recommended by ASCO as first-line treatment for non–small cell lung cancer (NSCLC)?Which platinum-containing combination therapy is preferred in the treatment of non–small cell lung cancer (NSCLC)?What is the role of paclitaxel in the treatment of non–small cell lung cancer (NSCLC)?What is the efficacy of paclitaxel (Abraxane) in the treatment of non–small cell lung cancer (NSCLC)?What is the role of platinum-based doublet chemotherapy in the treatment of non–small cell lung cancer (NSCLC)?Which histologic factors affect chemotherapy responsiveness in patients with non–small cell lung cancer (NSCLC)?What is the role of genetics in resistance to platinum-based chemotherapy for the treatment of non–small cell lung cancer (NSCLC)?What are the indications for second-line chemotherapy for non–small cell lung cancer (NSCLC)?What is the role of ramucirumab (Cyramza) in the treatment of non–small cell lung cancer (NSCLC)?What is the efficacy of second-line chemotherapy in the treatment of non–small cell lung cancer (NSCLC)?What are possible adverse effects of chemotherapy for non–small cell lung cancer (NSCLC)?What is the role of combination chemoradiation therapy for the treatment of non–small cell lung cancer (NSCLC)?What is the efficacy of combined chemoradiation therapy for the treatment of non–small cell lung cancer (NSCLC)?Which chemotherapy regimens have been used in combination with radiation therapy for non–small cell lung cancer (NSCLC)?What is the role of consolidation chemotherapy after chemoradiation in the treatment of non–small cell lung cancer (NSCLC)?What is an effective combination therapy for the treatment of stage III non–small cell lung cancer (NSCLC)?What is the role of durvalumab in the treatment of non–small cell lung cancer (NSCLC)?What is the efficacy of chemoradiation followed by surgical resection for the treatment of non–small cell lung cancer (NSCLC)?What are the combination treatment options for non–small cell lung cancer (NSCLC) in patients with vascular endothelial growth factor (VEGF) mutations?What are the treatment options for non–small cell lung cancer (NSCLC) in patients with ROS-1 mutations?What is the role of osimertinib (Tagrisso) in the treatment of non–small cell lung cancer (NSCLC)?What is the role of ceritinib in the treatment of non–small cell lung cancer (NSCLC)?What is the role of molecular-targeted therapy in the treatment of non–small cell lung cancer (NSCLC)?What are options for targeted therapy in patients with non–small cell lung cancer (NSCLC) adenocarcinoma and endothelial growth factor receptor (EGFR) mutations?What are first-line therapy options for patients with non–small cell lung cancer (NSCLC) adenocarcinoma and anaplastic lymphoma kinase (ALK) rearrangements?How is non–small cell lung cancer (NSCLC) treated in patients with BRAF mutation?What are the indications for selpercatinib (Retevmo) in the treatment of non-small cell lung cancer (NSCLC)?What are the treatment options for non–small cell lung cancer (NSCLC) in patients with PD-1 ligand (PDL-1)?Which medications are used in the treatment of non–small cell lung cancer (NSCLC) in patients who do not demonstrate EGFR or ALK but have high PDL-1 expression?How does overexpression of endothelial growth factor receptor (EGFR) affect the prognosis of non–small cell lung cancer (NSCLC)?How common are endothelial growth factor receptor (EGFR) mutations in patients with advanced non–small cell lung cancer (NSCLC)?What is the role of afatinib (Gilotrif) in the treatment of non–small cell lung cancer (NSCLC)?What are serious adverse events of afatinib (Gilotrif) for treatment of non–small cell lung cancer (NSCLC)?What is the method of action of gefitinib in the treatment of non–small cell lung cancer (NSCLC)?When is gefitinib indicated in the treatment of non–small cell lung cancer (NSCLC)?What is the efficacy of gefitinib for the treatment of non–small cell lung cancer (NSCLC)?What is the efficacy of gefitinib versus docetaxel as second-line therapy for non–small cell lung cancer (NSCLC)?What is the efficacy of gefitinib versus carboplatin-paclitaxel as first-line therapy for non–small cell lung cancer (NSCLC)?What is the strongest predictor of progression-free survival and response to gefitinib in non–small cell lung cancer (NSCLC)?What is the role of erlotinib in the treatment of non–small cell lung cancer (NSCLC)?What are the benefits of erlotinib combined with chemotherapy for the treatment of non–small cell lung cancer (NSCLC)?What is the efficacy of first-line treatment with erlotinib followed by cisplatin-gemcitabine in non–small cell lung cancer (NSCLC)?What are the benefits of erlotinib combined with bevacizumab for the treatment of non–small cell lung cancer (NSCLC)?What is the role of dacomitinib in the treatment of non–small cell lung cancer (NSCLC)?What is the role of cetuximab in the treatment of non–small cell lung cancer (NSCLC)?What is the role of pembrolizumab in the treatment of non–small cell lung cancer (NSCLC)?What is the role of combined capmatinib (Tabrecta) in the treatment of non–small cell lung cancer (NSCLC)?What is the role of loratinib in the treatment of non–small cell lung cancer (NSCLC)?What are the treatment options for squamous cell non–small cell lung cancer (NSCLC) in patients with EGFR mutations?What is the role of necitumumab in the treatment of non–small cell lung cancer (NSCLC)?What is the role of vandetanib in the treatment of non–small cell lung cancer (NSCLC)?What are treatment options for adenocarcinoma and large cell lung cancer?What is the role of bevacizumab (Avastin) in the treatment of non–small cell lung cancer (NSCLC)?What are the response rates for bevacizumab in combination with cisplatin-gemcitabine as first-line therapy for nonsquamous non–small cell lung cancer (NSCLC)?What is the efficacy of adding bevacizumab to platinum-based chemotherapy as first-line treatment of non–small cell lung cancer (NSCLC)?What is the role of bevacizumab-awwb (Mvasi) in the treatment of non-squamous non–small cell lung cancer (NSCLC)?What is the role of ramucirumab in the treatment of non–small cell lung cancer (NSCLC)?How common are anaplastic lymphoma kinase (ALK) mutations in non–small cell lung cancer (NSCLC)?What is the role of crizotinib (Xalkori) in the treatment of non–small cell lung cancer (NSCLC)?What is the role of brigatinib (Alunbrig) in the treatment of non–small cell lung cancer (NSCLC)?What is the role of alectinib in the treatment of non–small cell lung cancer (NSCLC)?What is the role of crizotinib in the treatment of non–small cell lung cancer (NSCLC)?When are third-generation endothelial growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) indicated for the treatment of non–small cell lung cancer (NSCLC)?What is the role of osimertinib in the treatment of non–small cell lung cancer (NSCLC)?What is the role of programmed cell death (PD-1) inhibitors in the treatment of non–small cell lung cancer (NSCLC) therapy?What is the role of nivolumab (Opdivo) in the treatment of non–small cell lung cancer (NSCLC)?What is the role of pembrolizumab (Keytruda) in the treatment of metastatic non–small cell lung cancer (NSCLC)?What is the efficacy of pembrolizumab for the treatment of metastatic non–small cell lung cancer (NSCLC)?What is the role of atezolizumab (Tecentriq) in the treatment of metastatic non–small cell lung cancer (NSCLC)?What is the role of durvalumab in the treatment of stage III non–small cell lung cancer (NSCLC)?What is the role of combination figitumumab, paclitaxel and carboplatin in the treatment of non–small cell lung cancer (NSCLC)?What is BRAF-directed therapy for non–small cell lung cancer (NSCLC)?What is the role of selpercatinib (Retevmo) in the treatment of non-small cell lung cancer (NSCLC)?What are the ASCO recommendations on staged therapy for stage IV non–small cell lung cancer (NSCLC)?What is the treatment of choice for stage I non–small cell lung cancer (NSCLC)?How is non–small cell lung cancer (NSCLC) treated in patients who cannot undergo resection?What is the role of postoperative radiation therapy in stage I and II non–small cell lung cancer (NSCLC)?What is the role of adjuvant chemotherapy in non–small cell lung cancer (NSCLC)?What is the treatment of choice for stage II non–small cell lung cancer (NSCLC)?What are treatment options for stage IIIA non–small cell lung cancer (NSCLC)?Which patients with stage IIIA non–small cell lung cancer (NSCLC) should be considered for definitive chemoradiation therapy?What are the advantages of surgery versus radiation therapy following neoadjuvant chemotherapy for the treatment of stage IIIA non–small cell lung cancer (NSCLC)?What is the role of adjuvant chemotherapy in the treatment of stage IIIA non–small cell lung cancer (NSCLC)?What is the role of postoperative radiation therapy in patients with stage IIIA non–small cell lung cancer (NSCLC)?What is the role of neoadjuvant cisplatin-based chemotherapy for the treatment of stage IIIA non–small cell lung cancer (NSCLC)?What is the role of prophylactic cranial irradiation (PCI) in the treatment of non–small cell lung cancer (NSCLC)?What is the treatment options for stage IIIB non–small cell lung cancer (NSCLC)?What is the efficacy of treatments for stage IIIB non–small cell lung cancer (NSCLC)?What are the treatment options for stage IIIB non–small cell lung cancer (NSCLC) and poor performance status?What are the treatment options for advanced non–small cell lung cancer (NSCLC)?What is the efficacy of treatments for stage IV non–small cell lung cancer (NSCLC)?Which patients with stage IV non–small cell lung cancer (NSCLC) may be candidates for treatment with bevacizumab?What is the role of maintenance non–cross-resistant chemotherapy in the treatment of stage IV non–small cell lung cancer (NSCLC)?Which patients with stage IV non–small cell lung cancer (NSCLC) may benefit from treatment with gefitinib or erlotinib?Which patients with stage IV non–small cell lung cancer (NSCLC) may be candidates for treatment with cytotoxic drugs?Which patients with stage IV non–small cell lung cancer (NSCLC) should be considered for palliative care?How is stage IV non–small cell lung cancer (NSCLC) treated in patients with large-cell neuroendocrine carcinoma?How is activity level used in the prognosis of non–small cell lung cancer (NSCLC)?How is non–small cell lung cancer (NSCLC) prevented?What measures can be taken to decrease occupational exposure to carcinogens causing non–small cell lung cancer (NSCLC)?Which patients should receive non–small cell lung cancer (NSCLC) screening?Which specialist consultations are recommended for the management of non–small cell lung cancer (NSCLC)?Which specialist consultations are recommended to address complications caused by spread of non–small cell lung cancer (NSCLC)?What are the NCCN recommendations for cancer surveillance in survivors of non–small cell lung cancer (NSCLC)?What are the NCCN recommendations for long-term monitoring of non–small cell lung cancer (NSCLC)?Which organizations have issued guidelines for lung cancer screening?What are recommendations for lung cancer screening?What is included in the American College of Chest Physicians (ACCP) lung cancer guidelines?What are the ACCP diagnostic guidelines for pleural abnormalities in non–small cell lung cancer (NSCLC)?What are the ACCP recommendations for the diagnosis of primary tumor in lung cancer?What are the ACCP recommendations for treatment of clinical stage I and II non–small cell lung cancer (NSCLC)?What are ACCP recommendations for treatment of clinical stage III non–small cell lung cancer (NSCLC)?What are the ACCP recommendations for treatment of stage IV non–small cell lung cancer (NSCLC)?What are ACCP recommendations for palliative and end-of-life care for non–small cell lung cancer (NSCLC)?What are the guidelines for molecular testing and treatment in non–small cell lung cancer (NSCLC)?What are NCCN recommendations for video-assisted thoracoscopy surgery (VATS) in non–small cell lung cancer (NSCLC)?What are the ASCO recommendations for first-line therapy for negative or unknown tumor EGFR-sensitizing mutation, ALK or ROS1 gene rearrangement status in non–small cell lung cancer (NSCLC)?What are the ASCO guidelines for chemotherapy for stage IV non–small cell lung cancer (NSCLC)?What are cisplatin-based two-drug combinations for stage IV non–small cell lung cancer (NSCLC)?What are carboplatin-based two-drug combinations for stage IV non–small cell lung cancer (NSCLC)?What are ASCO recommendations for stage IV non–small cell lung cancer (NSCLC) receiving carboplatin plus paclitaxel?What are the ASCO recommendations for treatment of stage IV non–small cell lung cancer (NSCLC) in patients with PS 2, non-SCC, and negative or unknown tumor EGFR-sensitizing mutation and ALK or ROS1 gene rearrangement status?What are the ASCO recommendations for stage IV squamous cell carcinoma (SCC) non–small cell lung cancer (NSCLC)?What are cisplatin-based two-drug combinations for stage IV squamous cell carcinoma (SCC) non–small cell lung cancer (NSCLC)?What are carboplatin-based two-drug combinations for stage IV squamous cell carcinoma (SCC) non–small cell lung cancer (NSCLC)?What are first-line options for treatment of stage IV squamous cell carcinoma (SCC) non–small cell lung cancer (NSCLC) in patients with a sensitizing endothelial growth factor receptor (EGFR) mutation?What are ASCO recommendations for second-line therapy for patients with stage IV squamous cell carcinoma (SCC) and non-squamous cell carcinoma (SCC) non–small cell lung cancer (NSCLC)?What are ASCO recommendations for second-line therapy for stage IV non–small cell lung cancer (NSCLC) in patients with sensitizing endothelial growth factor receptor (EGFR) mutation?What are ASCO recommendations for second-line therapy of stage IV non–small cell lung cancer (NSCLC) in patients with ROS1 rearrangement?What are ASCO recommendations for second-line therapy of stage IV non–small cell lung cancer (NSCLC) in patients with BRAF mutations?What are ASCO recommendations for third-line therapy of stage IV non–small cell lung cancer (NSCLC)?What is included in the American Society for Radiation Oncology (ASTRO) guidelines for radiation therapy for non–small cell lung cancer (NSCLC)?What are ASTRO recommendations for curative-intent radiation therapy for locally advanced non–small cell lung cancer (NSCLC)?What are ASTRO recommendations for adjunctive radiation therapy for non–small cell lung cancer (NSCLC)?What is the role of medications in the treatment of non–small cell lung cancer (NSCLC)?Which medications in the drug class Antineoplastics, MEK Inhibitors are used in the treatment of Non-Small Cell Lung Cancer (NSCLC)?Which medications in the drug class Antineoplastics, BRAF Kinase Inhibitor are used in the treatment of Non-Small Cell Lung Cancer (NSCLC)?Which medications in the drug class Antiemetic Agents are used in the treatment of Non-Small Cell Lung Cancer (NSCLC)?Which medications in the drug class Antineoplastics, EGFR Inhibitor are used in the treatment of Non-Small Cell Lung Cancer (NSCLC)?Which medications in the drug class PD-1/PD-L1 Inhibitors are used in the treatment of Non-Small Cell Lung Cancer (NSCLC)?Which medications in the drug class Antineoplastics, Anaplastic Lymphoma Kinase Inhibitor are used in the treatment of Non-Small Cell Lung Cancer (NSCLC)?Which medications in the drug class Antineoplastic Agents are used in the treatment of Non-Small Cell Lung Cancer (NSCLC)?Which medications in the drug class Antineoplastics, Tyrosine Kinase Inhibitor are used in the treatment of Non-Small Cell Lung Cancer (NSCLC)?Which medications in the drug class RET Kinase Inhibitors are used in the treatment of Non-Small Cell Lung Cancer (NSCLC)?Which medications in the drug class Antineoplastics, Monoclonal Antibody are used in the treatment of Non-Small Cell Lung Cancer (NSCLC)?

Author

Winston W Tan, MD, FACP, Associate Professor of Medicine, Mayo Medical School; Consultant and Person-in-Charge of Genitourinary Oncology-Medical Oncology, Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic Jacksonville; Vice Chairman, Division of Hematology/Oncology Education, Chair, Cancer Survivorship Program, Associate Chair, Department of Medicine Faculty Development, Mayo Clinic Florida; Vice President, Florida Society of Clinical Oncology

Disclosure: Nothing to disclose.

Coauthor(s)

Syed Huq, MD, Fellow, Division of Hematology-Oncology, Department of Internal Medicine, University of Missouri-Columbia School of Medicine, Ellis Fischel Cancer Center

Disclosure: Nothing to disclose.

Specialty Editors

Mary L Windle, PharmD, Adjunct Associate Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Nothing to disclose.

Chief Editor

Nagla Abdel Karim, MD, PhD, Associate Professor of Medicine, Associate Director of Experimental Therapeutics, Division of Hematology/Oncology, University of Cincinnati Cancer Institute, Department of Internal Medicine, University of Cincinnati College of Medicine

Disclosure: Nothing to disclose.

Acknowledgements

Jeffrey L Arnold, MD, FACEP Chairman, Department of Emergency Medicine, Santa Clara Valley Medical Center

Jeffrey L Arnold, MD, FACEP is a member of the following medical societies: American Academy of Emergency Medicine and American College of Physicians

Disclosure: Nothing to disclose.

Barry E Brenner, MD, PhD, FACEP Professor of Emergency Medicine, Professor of Internal Medicine, Program Director for Emergency Medicine, Case Medical Center, University Hospitals, Case Western Reserve University School of Medicine

Barry E Brenner, MD, PhD, FACEP is a member of the following medical societies: Alpha Omega Alpha, American Academy of Emergency Medicine, American College of Chest Physicians, American College of Emergency Physicians, American College of Physicians, American Heart Association, American Thoracic Society, Arkansas Medical Society, New York Academy of Medicine, New York Academy of Sciences, and Society for Academic Emergency Medicine

Disclosure: Nothing to disclose.

Gino A Farina, MD, FACEP, FAAEM Associate Professor of Emergency Medicine, Hofstra North Shore LIJ School of Medicine and Albert Einstein College of Medicine; Program Director, Department of Emergency Medicine, Long Island Jewish Medical Center

Gino A Farina, MD, FACEP, FAAEM is a member of the following medical societies: American Academy of Emergency Medicine, American College of Emergency Physicians, and Society for Academic Emergency Medicine

Disclosure: Nothing to disclose.

Edmond A Hooker II, MD, DrPH, FAAEM Associate Professor, Department of Health Services Administration, Xavier University, Cincinnati, Ohio; Assistant Professor, Department of Emergency Medicine, University of Cincinnati College of Medicine

Edmond A Hooker II, MD, DrPH, FAAEM is a member of the following medical societies: American Academy of Emergency Medicine, American Public Health Association, Society for Academic Emergency Medicine, and Southern Medical Association

Disclosure: Nothing to disclose.

Irfan Maghfoor, MD Consulting Oncologist, Department of Oncology, King Faisal Specialist Hospital and Research Center, Saudi Arabia

Irfan Maghfoor, MD is a member of the following medical societies: American Society of Hematology

Disclosure: Nothing to disclose.

Tamas Peredy, MD Assistant Professor, Department of Emergency Medicine, Maine Medical Center

Disclosure: Nothing to disclose.

Michael Perry, MD, MS, MACP Nellie B Smith Chair of Oncology Emeritus, Director, Division of Hematology and Medical Oncology, Deputy Director, Ellis Fischel Cancer Center, University of Missouri-Columbia School of Medicine

Michael Perry, MD, MS, MACP is a member of the following medical societies: Alpha Omega Alpha, American Association for Cancer Research, American College of Physicians, American College of Physicians-American Society of Internal Medicine, American Medical Association, American Society of Clinical Oncology, American Society of Hematology, International Association for the Study of Lung Cancer, and Missouri State Medical Association

Disclosure: Nothing to disclose.

Peter T Porrello, MD, FACEP Clinical Instructor, Department of Emergency Medicine, Yale University School of Medicine; Chief Medical Informatics Officer, Consulting Staff, Waterbury Hospital

Disclosure: Nothing to disclose.

Mityanand Ramnarine, MD Chief Resident Physician, Department of Emergency Medicine, Albert Einstein College of Medicine at Long Island Jewish Medical Center

Mityanand Ramnarine, MD is a member of the following medical societies: Alpha Omega Alpha, American College of Emergency Physicians, Emergency Medicine Residents Association, and Society for Academic Emergency Medicine

Disclosure: Nothing to disclose.

Francisco Talavera, PharmD, PhD Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Medscape Salary Employment

References

  1. Spiro SG, Gould MK, Colice GL. Initial evaluation of the patient with lung cancer: symptoms, signs, laboratory tests, and paraneoplastic syndromes: ACCP evidenced-based clinical practice guidelines (2nd edition). Chest. 2007 Sep. 132(3 Suppl):149S-160S. [View Abstract]
  2. Rowell NP, Williams CJ. Radical radiotherapy for stage I/II non-small cell lung cancer in patients not sufficiently fit for or declining surgery (medically inoperable): a systematic review. Thorax. 2001 Aug. 56(8):628-38. [View Abstract]
  3. Strand TE, Brunsvig PF, Johannessen DC, et al. Potentially curative radiotherapy for non-small-cell lung cancer in Norway: a population-based study of survival. Int J Radiat Oncol Biol Phys. 2011 May 1. 80(1):133-41. [View Abstract]
  4. Wang HM, Liao ZX, Komaki R, et al. Improved survival outcomes with the incidental use of beta-blockers among patients with non-small-cell lung cancer treated with definitive radiation therapy. Ann Oncol. 2013 Jan 8. [View Abstract]
  5. Rosen G. A History of Public Health: Expanded Edition. Baltimore, MD.: The Johns Hopkins University Press; 1993.
  6. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020 Jan. 70 (1):7-30. [View Abstract]
  7. National Research Council, Committee on Health Risks of Exposure to Radon, Board on Radiation Effects Research, Commission on Life Sciences. Health effects of exposure to radon (BEIR VI). Washington, DC.: National Academy Press.; 1999.
  8. Ito H, Matsuo K, Tanaka H, et al. Nonfilter and filter cigarette consumption and the incidence of lung cancer by histological type in Japan and the United States: analysis of 30-year data from population-based cancer registries. Int J Cancer. 2011 Apr 15. 128(8):1918-28. [View Abstract]
  9. Zhang J, Fujimoto J, Zhang J, et al. Intratumor heterogeneity in localized lung adenocarcinomas delineated by multiregion sequencing. Science. 2014 Oct 10. 346(6206):256-9. [View Abstract]
  10. de Bruin EC, McGranahan N, Mitter R, et al. Spatial and temporal diversity in genomic instability processes defines lung cancer evolution. Science. 2014 Oct 10. 346(6206):251-6. [View Abstract]
  11. Molina JR, Yang P, Cassivi SD, Schild SE, Adjei AA. Non-small cell lung cancer: epidemiology, risk factors, treatment, and survivorship. Mayo Clin Proc. 2008 May. 83(5):584-94. [View Abstract]
  12. Nelson R. Lung Cancer Rates Surging in Never-Smokers. Medscape Medical News. Available at http://www.medscape.com/viewarticle/850708. September 9, 2015; Accessed: April 13, 2020.
  13. Beckett WS. Epidemiology and etiology of lung cancer. Clin Chest Med. 1993 Mar. 14(1):1-15. [View Abstract]
  14. Bagnardi V, Rota M, Botteri E, et al. Alcohol consumption and lung cancer risk in never smokers: a meta-analysis. Ann Oncol. 2011 Dec. 22(12):2631-9. [View Abstract]
  15. Agaku IT, King BA, Dube SR. Current cigarette smoking among adults - United States, 2005-2012. MMWR Morb Mortal Wkly Rep. 2014 Jan 17. 63(2):29-34. [View Abstract]
  16. Ginsberg RJ, Vokes EE, Raben A. Non-small cell lung cancer. In: DeVita VT Jr, Hellman S, Rosenberg SA, eds. Cancer: Principles and Practice of Oncology. 5th ed. Philadelphia, Pa: Lippincott-Raven;. 1997:858-911.
  17. Lowry F. Physical Fitness Offsets Harmful Effects of Smoking. Medscape Medical News. May 30, 2019. Available at https://www.medscape.com/viewarticle/913712
  18. Zhong L, Goldberg MS, Parent ME, Hanley JA. Exposure to environmental tobacco smoke and the risk of lung cancer: a meta-analysis. Lung Cancer. 2000 Jan. 27(1):3-18. [View Abstract]
  19. Alberg AJ, Brock MV, Ford JG, Samet JM, Spivack SD. Epidemiology of lung cancer: Diagnosis and management of lung cancer, 3rd ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2013 May. 143(5 Suppl):e1S-29S. [View Abstract]
  20. Hou W, Fu J, Ge Y, Du J, Hua S. Incidence and risk of lung cancer in HIV-infected patients. J Cancer Res Clin Oncol. 2013 Nov. 139(11):1781-94. [View Abstract]
  21. Cortes-Jofre M, Rueda JR, Corsini-Munoz G, Fonseca-Cortes C, Caraballoso M, Bonfill Cosp X. Drugs for preventing lung cancer in healthy people. Cochrane Database Syst Rev. 2012 Oct 17. 10:CD002141. [View Abstract]
  22. Ries L, Eisner M, Kosary C. Cancer statistics review, 1975-2002. National Cancer Institute.; 2005.
  23. World Health Organization, International Agency for Research on Cancer. Globocan 2018: Lung Cancer. International Agency for Research on Cancer. Available at http://gco.iarc.fr/today/data/factsheets/cancers/15-Lung-fact-sheet.pdf. Accessed: May 19, 2020.
  24. Jemal A, Miller KD, Ma J, Siegel RL, Fedewa SA, Islami F, et al. Higher Lung Cancer Incidence in Young Women Than Young Men in the United States. N Engl J Med. 2018 May 24. 378 (21):1999-2009. [View Abstract]
  25. SEER Stat Fact Sheets: Lung and Bronchus Cancer. Surveillance, Epidemiology, and End Results Program. Available at http://seer.cancer.gov/statfacts/html/lungb.html. Accessed: April 12, 2019.
  26. Jonnalagadda S, Smith C, Mhango G, Wisnivesky JP. The Number of Lymph Node Metastases as a Prognostic Factor in Patients With N1 Non-small Cell Lung Cancer. Chest. 2011 Aug. 140(2):433-40. [View Abstract]
  27. Mostertz W, Stevenson M, Acharya C, et al. Age- and sex-specific genomic profiles in non-small cell lung cancer. JAMA. 2010 Feb 10. 303(6):535-43. [View Abstract]
  28. Parsons A, Daley A, Begh R, Aveyard P. Influence of smoking cessation after diagnosis of early stage lung cancer on prognosis: systematic review of observational studies with meta-analysis. BMJ. 2010 Jan 21. 340:b5569. [View Abstract]
  29. Ferketich AK, Niland JC, Mamet R, et al. Smoking status and survival in the national comprehensive cancer network non-small cell lung cancer cohort. Cancer. 2012 Sep 28. [View Abstract]
  30. Chlebowski RT, Schwartz WH, Anderson GL, et al. Non-small cell lung cancer and estrogen plus progestin use in postmenopausal women in the Women’s Health Initiative randomized clinical trial2009 American Society of Clinical Oncology (ASCO) Annual Meeting. J Clin Oncol. 2009. 27(suppl; abstr CRA1500):18s.
  31. Bouchardy C, Benhamou S, Schaffar R, et al. Lung cancer mortality risk among breast cancer patients treated with anti-estrogens. Cancer. 2011 Mar 15. 117(6):1288-95. [View Abstract]
  32. Rothwell PM, Fowkes GR, Belch JF, Ogawa H, Warlow CP, Meade TW. Effect of daily aspirin on long-term risk of death due to cancer: analysis of individual patient data from randomized trials. Lancet. Dec 7/2010; Early online publication.
  33. Hofman V, Bonnetaud C, Ilie MI, et al. Preoperative circulating tumor cell detection using the isolation by size of epithelial tumor cell method for patients with lung cancer is a new prognostic biomarker. Clin Cancer Res. 2011 Feb 15. 17(4):827-35. [View Abstract]
  34. Wicha MS, Hayes DF. Circulating tumor cells: not all detected cells are bad and not all bad cells are detected. J Clin Oncol. 2011 Apr 20. 29(12):1508-11. [View Abstract]
  35. Lopez Guerra JL, Gomez DR, Lin SH, et al. Risk factors for local and regional recurrence in patients with resected N0-N1 non-small-cell lung cancer, with implications for patient selection for adjuvant radiation therapy. Ann Oncol. 2012 Sep 20. [View Abstract]
  36. Kadota K, Nitadori JI, Sarkaria IS, et al. Thyroid transcription factor-1 expression is an independent predictor of recurrence and correlates with the IASLC/ATS/ERS histologic classification in patients with stage I lung adenocarcinoma. Cancer. 2012 Oct 23. [View Abstract]
  37. Nitadori J, Bograd AJ, Kadota K, et al. Impact of Micropapillary Histologic Subtype in Selecting Limited Resection vs Lobectomy for Lung Adenocarcinoma of 2cm or Smaller. J Natl Cancer Inst. 2013 Aug 21. 105(16):1212-20. [View Abstract]
  38. Saunders R. Histologic Subtype Predicts Recurrence of Early Lung Adenocarcinoma. Medscape [serial online]. Available at http://www.medscape.com/viewarticle/809365. August 13, 2013; Accessed: April 12, 2019.
  39. Corner J, Hopkinson J, Fitzsimmons D, Barclay S, Muers M. Is late diagnosis of lung cancer inevitable? Interview study of patients' recollections of symptoms before diagnosis. Thorax. 2005 Apr. 60(4):314-9. [View Abstract]
  40. Ejaz S, Vassilopoulou-Sellin R, Busaidy NL, et al. Cushing syndrome secondary to ectopic adrenocorticotropic hormone secretion: the University of Texas MD Anderson Cancer Center Experience. Cancer. 2011 Oct 1. 117(19):4381-9. [View Abstract]
  41. Fadel E, Missenard G, Court C, et al. Long-term outcomes of en bloc resection of non-small cell lung cancer invading the thoracic inlet and spine. Ann Thorac Surg. 2011 Sep. 92(3):1024-30; discussion 1030. [View Abstract]
  42. Patel AM, Davila DG, Peters SG. Paraneoplastic syndromes associated with lung cancer. Mayo Clin Proc. 1993 Mar. 68(3):278-87. [View Abstract]
  43. Sher T, Dy GK, Adjei AA. Small cell lung cancer. Mayo Clin Proc. 2008 Mar. 83(3):355-67. [View Abstract]
  44. Annema JT, van Meerbeeck JP, Rintoul RC, et al. Mediastinoscopy vs endosonography for mediastinal nodal staging of lung cancer: a randomized trial. JAMA. 2010 Nov 24. 304(20):2245-52. [View Abstract]
  45. Oken MM, Hocking WG, Kvale PA, et al. Screening by chest radiograph and lung cancer mortality: the Prostate, Lung, Colorectal, and Ovarian (PLCO) randomized trial. JAMA. 2011 Nov 2. 306(17):1865-73. [View Abstract]
  46. [Guideline] Mazzone PJ, Silvestri GA, Patel S, Kanne JP, Kinsinger LS, Wiener RS, et al. Screening for Lung Cancer: CHEST Guideline and Expert Panel Report. Chest. 2018 Apr. 153 (4):954-985. [View Abstract]
  47. Foster BB, Muller NL, Miller RR, et al. Neuroendocrine carcinomas of the lung: clinical, radiologic and pathologic correlation. Radiology. 1989. 170:441-445.
  48. Alpert JB, Fantauzzi JP, Melamud K, Greenwood H, Naidich DP, Ko JP. Clinical Significance of Lung Nodules Reported on Abdominal CT. AJR Am J Roentgenol. 2012 Apr. 198(4):793-9. [View Abstract]
  49. Gould MK, Maclean CC, Kuschner WG, Rydzak CE, Owens DK. Accuracy of positron emission tomography for diagnosis of pulmonary nodules and mass lesions: a meta-analysis. JAMA. 2001 Feb 21. 285(7):914-24. [View Abstract]
  50. Deppen SA, Blume JD, Kensinger CD, et al. Accuracy of FDG-PET to diagnose lung cancer in areas with infectious lung disease: a meta-analysis. JAMA. 2014 Sep 24. 312(12):1227-36. [View Abstract]
  51. Erkilic S, Ozsarac C, Kullu S. Sputum cytology for the diagnosis of lung cancer. Comparison of smear and modified cell block methods. Acta Cytol. 2003 Nov-Dec. 47(6):1023-7. [View Abstract]
  52. Billah S, Stewart J, Staerkel G, et al. EGFR and KRAS mutations in lung carcinoma: molecular testing by using cytology specimens. Cancer Cytopathol. 2011 Apr 25. 119(2):111-7. [View Abstract]
  53. Arroliga AC, Matthay RA. The role of bronchoscopy in lung cancer. Clin Chest Med. 1993 Mar. 14(1):87-98. [View Abstract]
  54. Schreiber G, McCrory DC. Performance characteristics of different modalities for diagnosis of suspected lung cancer: summary of published evidence. Chest. 2003 Jan. 123(1 Suppl):115S-128S. [View Abstract]
  55. He J, Shao W, Cao C, et al. Long-term outcome and cost-effectiveness of complete versus assisted video-assisted thoracic surgery for non-small cell lung cancer. J Surg Oncol. 2011 Aug 1. 104(2):162-8. [View Abstract]
  56. Mentzer SJ, Swanson SJ, DeCamp MM, Bueno R, Sugarbaker DJ. Mediastinoscopy, thoracoscopy, and video-assisted thoracic surgery in the diagnosis and staging of lung cancer. Chest. 1997 Oct. 112(4 Suppl):239S-241S. [View Abstract]
  57. [Guideline] Lindeman NI, Cagle PT, Aisner DL, Arcila ME, Beasley MB, Bernicker EH, et al. Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment With Targeted Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. Arch Pathol Lab Med. 2018 Mar. 142 (3):321-346. [View Abstract]
  58. Chustecka Z. FDA approves companion genetic diagnostic test for Tarceva in NSCLC. Medscape Medical News. May 14, 2013.
  59. Rosell R, Carcereny E, Gervais R, et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2012 Mar. 13(3):239-46. [View Abstract]
  60. Janne PA, Shaw AT, Pereira JR, et al. Selumetinib plus docetaxel for KRAS-mutant advanced non-small-cell lung cancer: a randomised, multicentre, placebo-controlled, phase 2 study. Lancet Oncol. 2013 Jan. 14(1):38-47. [View Abstract]
  61. American Joint Committee on Cancer. Lung. Amin MB, Edge S, Greene F, Byrd DR, Brookland RK, et al, eds. AJCC Cancer Staging Handbook. 8th ed. Chicago, Ill: Springer; 2017. Chapter 25.
  62. Tintinalli JE. Emergency complications of malignancy. Emergency Medicine: A Comprehensive Guide. 2004. 1319-1368.
  63. Halfdanarson TR, Hogan WJ, Moynihan TJ. Oncologic emergencies: diagnosis and treatment. Mayo Clin Proc. 2006 Jun. 81(6):835-48. [View Abstract]
  64. [Guideline] National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology. Lung Cancer Screening. Available at http://www.nccn.org/professionals/physician_gls/pdf/lung_screening.pdf. Version 1.2020 — May 14, 2019; Accessed: May 19, 2020.
  65. [Guideline] Wender R, Fontham ET, Barrera E Jr, Colditz GA, Church TR, Ettinger DS, et al. American Cancer Society lung cancer screening guidelines. CA Cancer J Clin. 2013 Mar-Apr. 63 (2):107-17. [View Abstract]
  66. [Guideline] U.S. Preventive Services Task Force. Final Recommendation Statement: Lung Cancer: Screening. Available at https://www.uspreventiveservicestaskforce.org/Page/Document/RecommendationStatementFinal/lung-cancer-screening. December 20, 2016; Accessed: June 15, 2019.
  67. Cheung LC, Katki HA, Chaturvedi AK, Jemal A, Berg CD. Preventing Lung Cancer Mortality by Computed Tomography Screening: The Effect of Risk-Based Versus U.S. Preventive Services Task Force Eligibility Criteria, 2005–2015. Ann Intern Med. 2 January 2018.
  68. Kumar V, Cohen JT, van Klaveren D, Soeteman DI, Wong JB, Neumann PJ, et al. Risk-Targeted Lung Cancer Screening: A Cost-Effectiveness Analysis. Ann Intern Med. 2 January 2018.
  69. Pastorino U, Silva M, Sestini S, Sabia F, Boeri M, Cantarutti A, et al. Prolonged lung cancer screening reduced 10-year mortality in the MILD trial: new confirmation of lung cancer screening efficacy. Ann Oncol. 2019 Jun 5. [View Abstract]
  70. Patz EF Jr, Pinsky P, Gatsonis C, et al. Overdiagnosis in low-dose computed tomography screening for lung cancer. JAMA Intern Med. 2013 Dec 9. [View Abstract]
  71. Patz EF Jr, Greco E, Gatsonis C, Pinsky P, Kramer BS, Aberle DR. Lung cancer incidence and mortality in National Lung Screening Trial participants who underwent low-dose CT prevalence screening: a retrospective cohort analysis of a randomised, multicentre, diagnostic screening trial. Lancet Oncol. 2016 Mar 18. [View Abstract]
  72. Kinsinger LS, Anderson C, Kim J, Larson M, Chan SH, King HA, et al. Implementation of Lung Cancer Screening in the Veterans Health Administration. JAMA Intern Med. 2017 Jan 30. [View Abstract]
  73. Castellino M. Lung Cancer Screening -- Benefits Few, May Harm Many. Medscape Medical News. Available at http://www.medscape.com/viewarticle/875091. January 30, 2017; Accessed: June 15, 2019.
  74. Katki HA, Kovalchik SA, Petito LC, Cheung LC, Jacobs E, Jemal A, et al. Implications of Nine Risk Prediction Models for Selecting Ever-Smokers for Computed Tomography Lung Cancer Screening. Ann Intern Med. 15 May 2018.
  75. Jenkins K. Which Smokers Should Be Screened for Lung Cancer?. Medscape Medical News. Available at https://www.medscape.com/viewarticle/896582. May 14, 2018; Accessed: June 15, 2019.
  76. Jenkins K. Blood Test to Identify Individuals for Lung Cancer Screening. Medscape Medical News. Available at https://www.medscape.com/viewarticle/899380. July 16, 2018; Accessed: June 15, 2018.
  77. Integrative Analysis of Lung Cancer Etiology and Risk (INTEGRAL) Consortium for Early Detection of Lung Cancer., Guida F, Sun N, Bantis LE, et al. Assessment of Lung Cancer Risk on the Basis of a Biomarker Panel of Circulating Proteins. JAMA Oncol. 2018 Jul 12. e182078. [View Abstract]
  78. Arriagada R, Bergman B, Dunant A, Le Chevalier T, Pignon JP, Vansteenkiste J. Cisplatin-based adjuvant chemotherapy in patients with completely resected non-small-cell lung cancer. N Engl J Med. 2004 Jan 22. 350(4):351-60. [View Abstract]
  79. Greer JA, Jackson VA, Meier DE, Temel JS. Early integration of palliative care services with standard oncology care for patients with advanced cancer. CA Cancer J Clin. 2013 Sep. 63(5):349-63. [View Abstract]
  80. Temel JS, Greer JA, Muzikansky A, Gallagher ER, Admane S, Jackson VA. Early palliative care for patients with metastatic non-small-cell lung cancer. N Engl J Med. 2010 Aug 19. 363(8):733-42. [View Abstract]
  81. [Guideline] Detterbeck FC, Lewis SZ, Diekemper R, Addrizzo-Harris D, Alberts WM. Executive Summary: Diagnosis and management of lung cancer, 3rd ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2013 May. 143(5 Suppl):7S-37S. [View Abstract]
  82. Yang CF, Kumar A, Gulack BC, Mulvihill MS, Hartwig MG, Wang X, et al. Long-term outcomes after lobectomy for non-small cell lung cancer when unsuspected pN2 disease is found: A National Cancer Data Base analysis. J Thorac Cardiovasc Surg. 2016 May. 151 (5):1380-8. [View Abstract]
  83. Okada M, Nakayama H, Okumura S, et al. Multicenter analysis of high-resolution computed tomography and positron emission tomography/computed tomography findings to choose therapeutic strategies for clinical stage IA lung adenocarcinoma. J Thorac Cardiovasc Surg. 2011 Jun. 141(6):1384-91. [View Abstract]
  84. Ma Z, Dong A, Fan J, Cheng H. Does sleeve lobectomy concomitant with or without pulmonary artery reconstruction (double sleeve) have favorable results for non-small cell lung cancer compared with pneumonectomy? A meta-analysis. Eur J Cardiothorac Surg. 2007 Jul. 32(1):20-8. [View Abstract]
  85. Kates M, Swanson S, Wisnivesky JP. Survival following lobectomy and limited resection for the treatment of stage I non-small cell lung cancer<=1 cm in size: a review of SEER data. Chest. 2011 Mar. 139(3):491-6. [View Abstract]
  86. Comparison of Different Types of Surgery in Treating Patients With Stage IA Non-Small Cell Lung Cancer. National Cancer Institute. Available at https://clinicaltrials.gov/ct2/show/NCT00499330. May 28, 2018; Accessed: June 15, 2019.
  87. Yendamuri S, Sharma R, Demmy M, et al. Temporal trends in outcomes following sublobar and lobar resections for small (= 2 cm) non-small cell lung cancers--a Surveillance Epidemiology End Results database analysis. J Surg Res. 2013 Jul. 183(1):27-32. [View Abstract]
  88. Okami J, Ito Y, Higashiyama M, et al. Sublobar resection provides an equivalent survival after lobectomy in elderly patients with early lung cancer. Ann Thorac Surg. 2010 Nov. 90(5):1651-6. [View Abstract]
  89. Wolf AS, Richards WG, Jaklitsch MT, et al. Lobectomy versus sublobar resection for small (2 cm or less) non-small cell lung cancers. Ann Thorac Surg. 2011 Nov. 92(5):1819-23; discussion 1824-5. [View Abstract]
  90. Cattaneo SM, Park BJ, Wilton AS, et al. Use of video-assisted thoracic surgery for lobectomy in the elderly results in fewer complications. Ann Thorac Surg. 2008 Jan. 85(1):231-5; discussion 235-6. [View Abstract]
  91. [Guideline] National Comprehensive Cancer Network. National Comprehensive Cancer Network. Non-Small Cell Lung Cancer. NCCN. Available at https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf. Version 4.2020 — May 15, 2020; Accessed: May 19, 2020.
  92. Swanson SJ, Herndon JE 2nd, D'Amico TA, et al. Video-assisted thoracic surgery lobectomy: report of CALGB 39802--a prospective, multi-institution feasibility study. J Clin Oncol. 2007 Nov 1. 25(31):4993-7. [View Abstract]
  93. Darling GE, Allen MS, Decker PA, et al. Number of lymph nodes harvested from a mediastinal lymphadenectomy: results of the randomized, prospective American College of Surgeons Oncology Group Z0030 trial. Chest. 2011 May. 139(5):1124-9. [View Abstract]
  94. Allen JW, Farooq A, O'Brien TF, Osarogiagbon RU. Quality of surgical resection for nonsmall cell lung cancer in a US metropolitan area. Cancer. 2011 Jan 1. 117(1):134-42. [View Abstract]
  95. Yun YH, Kim YA, Min YH, et al. Health-Related Quality of Life in Disease-Free Survivors of Surgically Treated Lung Cancer Compared With the General Population. Ann Surg. 2012 Mar 30. [View Abstract]
  96. Dosoretz DE, Katin MJ, Blitzer PH, et al. Radiation therapy in the management of medically inoperable carcinoma of the lung: results and implications for future treatment strategies. Int J Radiat Oncol Biol Phys. 1992. 24(1):3-9. [View Abstract]
  97. Jeremic B, Milicic B, Milisavljevic S. Clinical prognostic factors in patients with locally advanced (stage III) nonsmall cell lung cancer treated with hyperfractionated radiation therapy with and without concurrent chemotherapy: single-Institution Experience in 600 Patients. Cancer. 2011 Jan 10. [View Abstract]
  98. Sigel K, Lurslurchachai L, Bonomi M, et al. Effectiveness of radiation therapy alone for elderly patients with unresected stage III non-small cell lung cancer. Lung Cancer. 2013 Sep 4. [View Abstract]
  99. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012 Jun 28. 366(26):2443-54. [View Abstract]
  100. Onishi H, Shirato H, Nagata Y, Hiraoka M, Fujino M, Gomi K. Hypofractionated stereotactic radiotherapy (HypoFXSRT) for stage I non-small cell lung cancer: updated results of 257 patients in a Japanese multi-institutional study. J Thorac Oncol. 2007 Jul. 2(7 Suppl 3):S94-100. [View Abstract]
  101. Grills IS, Mangona VS, Welsh R, et al. Outcomes after stereotactic lung radiotherapy or wedge resection for stage I non-small-cell lung cancer. J Clin Oncol. 2010 Feb 20. 28(6):928-35. [View Abstract]
  102. Lencioni R, Crocetti L, Cioni R, Suh R, Glenn D, Regge D. Response to radiofrequency ablation of pulmonary tumours: a prospective, intention-to-treat, multicentre clinical trial (the RAPTURE study). Lancet Oncol. 2008 Jul. 9(7):621-8. [View Abstract]
  103. Muirhead R, van der Weide L, van Sornsen de Koste JR, Cover KS, Senan S. Use of megavoltage cine-images for studying intra-thoracic motion during radiotherapy for locally advanced lung cancer. Radiother Oncol. 2011 May. 99(2):155-60. [View Abstract]
  104. PORT Meta-analysis Trialists Group. Postoperative radiotherapy in non-small-cell lung cancer: systematic review and meta-analysis of individual patient data from nine randomised controlled trials. Lancet. 1998 Jul 25. 352(9124):257-63. [View Abstract]
  105. Rusch VW, Albain KS, Crowley JJ, et al. Surgical resection of stage IIIA and stage IIIB non-small-cell lung cancer after concurrent induction chemoradiotherapy. A Southwest Oncology Group trial. J Thorac Cardiovasc Surg. 1993 Jan. 105(1):97-104; discussion 104-6. [View Abstract]
  106. Azzoli CG, Temin S, Aliff T, et al. 2011 Focused Update of 2009 American Society of Clinical Oncology Clinical Practice Guideline Update on Chemotherapy for Stage IV Non-Small-Cell Lung Cancer. J Clin Oncol. 2011 Oct 1. 29(28):3825-31. [View Abstract]
  107. Rosell R, Gomez-Codina J, Camps C, et al. A randomized trial comparing preoperative chemotherapy plus surgery with surgery alone in patients with non-small-cell lung cancer. N Engl J Med. 1994. 330:153-158. [View Abstract]
  108. Roth JA, Fossella F, Komaki R, et al. A randomized trial comparing perioperative chemotherapy and surgery with surgery alone in resectable stage IIIA non-small-cell lung cancer. J Natl Cancer Inst. 1994. 86:673-680. [View Abstract]
  109. Sigel K, Mhango G, Cohen J, et al. Outcomes After Adjuvant Platinum-based Chemotherapy in Elderly NSCLC Patients with T4 Disease. Ann Surg Oncol. 2012 Nov 1. [View Abstract]
  110. NSCLC Meta-Analyses collaborative Group. Chemotherapy in addition to supportive care improves survival in advanced non-small-cell lung cancer: a systematic review and meta-analysis of individual patient data from 16 randomized controlled trials. J Clin Oncol. 2008 Oct 1. 26(28):4617-25. [View Abstract]
  111. Marino P, Pampallona S, Preatoni A, et al. Chemotherapy vs supportive care in advanced non-small-cell lung cancer. Results of a meta-analysis of the literature. Chest. 1994 Sep. 106(3):861-5. [View Abstract]
  112. Wanders R, Steevens J, Botterweck A, et al. Treatment with curative intent of stage III non-small cell lung cancer patients of 75 years: a prospective population-based study. Eur J Cancer. 2011 Dec. 47(18):2691-7. [View Abstract]
  113. Wisnivesky JP, Smith CB, Packer S, et al. Survival and risk of adverse events in older patients receiving postoperative adjuvant chemotherapy for resected stages II-IIIA lung cancer: observational cohort study. BMJ. 2011 Jul 14. 343:d4013. [View Abstract]
  114. Weick JK, Crowley J, Natale RB, et al. A randomized trial of five cisplatin-containing treatments in patients with metastatic non-small-cell lung cancer: a Southwest Oncology Group study. J Clin Oncol. 1991 Jul. 9(7):1157-62. [View Abstract]
  115. Rajeswaran A, Trojan A, Burnand B, Giannelli M. Efficacy and side effects of cisplatin- and carboplatin-based doublet chemotherapeutic regimens versus non-platinum-based doublet chemotherapeutic regimens as first line treatment of metastatic non-small cell lung carcinoma: a systematic review of randomized controlled trials. Lung Cancer. 2008 Jan. 59(1):1-11. [View Abstract]
  116. Socinski MA, Bondarenko I, Karaseva NA, et al. Weekly nab-paclitaxel in combination with carboplatin versus solvent-based paclitaxel plus carboplatin as first-line therapy in patients with advanced non-small-cell lung cancer: final results of a phase III trial. J Clin Oncol. 2012 Jun 10. 30(17):2055-62. [View Abstract]
  117. Le Chevalier T, Scagliotti G, Natale R, et al. Efficacy of gemcitabine plus platinum chemotherapy compared with other platinum containing regimens in advanced non-small-cell lung cancer: a meta-analysis of survival outcomes. Lung Cancer. 2005 Jan. 47(1):69-80. [View Abstract]
  118. Quoix E, Zalcman G, Oster JP, et al. Carboplatin and weekly paclitaxel doublet chemotherapy compared with monotherapy in elderly patients with advanced non-small-cell lung cancer: IFCT-0501 randomised, phase 3 trial. Lancet. 2011 Sep 17. 378(9796):1079-88. [View Abstract]
  119. Pallis AG, Karampeazis A, Vamvakas L, et al. Efficacy and treatment tolerance in older patients with NSCLC: a meta-analysis of five phase III randomized trials conducted by the Hellenic Oncology Research Group. Ann Oncol. 2011 Nov. 22(11):2448-55. [View Abstract]
  120. Scagliotti GV, Parikh P, von Pawel J, et al. Phase III study comparing cisplatin plus gemcitabine with cisplatin plus pemetrexed in chemotherapy-naive patients with advanced-stage non-small-cell lung cancer. J Clin Oncol. 2008 Jul 20. 26(21):3543-51. [View Abstract]
  121. Holm B, Mellemgaard A, Skov T, Skov BG. Different impact of excision repair cross-complementation group 1 on survival in male and female patients with inoperable non-small-cell lung cancer treated with carboplatin and gemcitabine. J Clin Oncol. 2009 Sep 10. 27(26):4254-9. [View Abstract]
  122. Garon EB, Ciuleanu TE, Arrieta O, et al. Ramucirumab plus docetaxel versus placebo plus docetaxel for second-line treatment of stage IV non-small-cell lung cancer after disease progression on platinum-based therapy (REVEL): a multicentre, double-blind, randomised phase 3 trial. Lancet. 2014 Aug 23. 384(9944):665-73. [View Abstract]
  123. Fidias PM, Dakhil SR, Lyss AP, et al. Phase III study of immediate compared with delayed docetaxel after front-line therapy with gemcitabine plus carboplatin in advanced non-small-cell lung cancer. J Clin Oncol. 2009 Feb 1. 27(4):591-8. [View Abstract]
  124. Hanna N, Shepherd FA, Fossella FV, et al. Randomized phase III trial of pemetrexed versus docetaxel in patients with non-small-cell lung cancer previously treated with chemotherapy. J Clin Oncol. 2004 May 1. 22(9):1589-97. [View Abstract]
  125. Ramlau R, Gorbunova V, Ciuleanu TE, et al. Aflibercept and Docetaxel versus Docetaxel alone after platinum failure in patients with advanced or metastatic non-small-cell lung cancer: a randomized, controlled phase III trial. J Clin Oncol. 2012 Oct 10. 30(29):3640-7. [View Abstract]
  126. Dillman RO, Seagren SL, Propert KJ, et al. A randomized trial of induction chemotherapy plus high-dose radiation versus radiation alone in stage III non-small-cell lung cancer. N Engl J Med. 1990 Oct 4. 323(14):940-5. [View Abstract]
  127. Le Chevalier T, Arriagada R, Quoix E, et al. Radiotherapy alone versus combined chemotherapy and radiotherapy in nonresectable non-small-cell lung cancer: first analysis of a randomized trial in 353 patients. J Natl Cancer Inst. 1991 Mar 20. 83(6):417-23. [View Abstract]
  128. Albain KS, Rusch VW, Crowley JJ, et al. Concurrent cisplatin/etoposide plus chest radiotherapy followed by surgery for stages IIIA (N2) and IIIB non-small-cell lung cancer: mature results of Southwest Oncology Group phase II study 8805. J Clin Oncol. 1995 Aug. 13(8):1880-92. [View Abstract]
  129. Schaake-Koning C, van den Bogaert W, Dalesio O, et al. Effects of concomitant cisplatin and radiotherapy on inoperable non- small-cell lung cancer. N Engl J Med. 1992 Feb 20. 326(8):524-30. [View Abstract]
  130. Albain KS, Swann RS, Rusch VR, et al. Phase III study of concurrent chemotherapy and radiotherapy (CT/RT) vs CT/RT followed by surgical resection for stage IIIA(pN2) non-small cell lung cancer (NSCLC): Outcomes update of North American Intergroup 0139 (RTOG 9309). J Clin Oncol. 2005. 23:624s.
  131. Curran WJ, Scott C, Langer C, et al. Long-term benefit is observed in a phase III comparison of sequential vs concurrent chemo-radiation for patients with unresected stage III non small cell lung cancer: RTOG 9410 (abstract). Proc Am Soc Clin Oncol. 2003. 22:621a.
  132. Curran WJ, Scott C, Langer C, et al. Phase III Comparison of Sequential vs Concurrent Chemoradiation for Patients (Pts) with Unresected Stage III Non-Small Cell Lung Cancer (NSCLC): Initial Report of Radiation Therapy Oncology Group (RTOG) 9410. Proc Am Soc Clin Oncol. 2000. 19:484a.
  133. Belani CP, Choy H, Bonomi P, et al. Combined chemoradiotherapy regimens of paclitaxel and carboplatin for locally advanced non-small-cell lung cancer: a randomized phase II locally advanced multi-modality protocol. J Clin Oncol. 2005 Sep 1. 23(25):5883-91. [View Abstract]
  134. Johnson DH, Paul DM, Hande KR, et al. Paclitaxel plus carboplatin in advanced non-small-cell lung cancer: a phase II trial. J Clin Oncol. 1996 Jul. 14(7):2054-60. [View Abstract]
  135. Langer CJ, Leighton JC, Comis RL, et al. Paclitaxel and carboplatin in combination in the treatment of advanced non-small-cell lung cancer: a phase II toxicity, response, and survival analysis. J Clin Oncol. 1995 Aug. 13(8):1860-70. [View Abstract]
  136. Hanna N, Neubauer M, Yiannoutsos C, et al. Phase III study of cisplatin, etoposide, and concurrent chest radiation with or without consolidation docetaxel in patients with inoperable stage III non-small-cell lung cancer: the Hoosier Oncology Group and U.S. Oncology. J Clin Oncol. 2008 Dec 10. 26(35):5755-60. [View Abstract]
  137. Curran WJ Jr, Paulus R, Langer CJ, et al. Sequential vs. concurrent chemoradiation for stage III non-small cell lung cancer: randomized phase III trial RTOG 9410. J Natl Cancer Inst. 2011 Oct 5. 103(19):1452-60. [View Abstract]
  138. Antonia SJ, et al; PACIFIC Investigators. Durvalumab after Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. N Engl J Med. 2017 Nov 16. 377 (20):1919-1929. [View Abstract]
  139. Schumacher A, Riesenbeck D, Braunheim M, et al. Combined modality treatment for locally advanced non-small cell lung cancer: preoperative chemoradiation does not result in a poorer quality of life. Lung Cancer. 2004 Apr. 44(1):89-97. [View Abstract]
  140. van Meerbeeck JP, Kramer GW, Van Schil PE, Legrand C, Smit EF, Schramel F, et al. Randomized controlled trial of resection versus radiotherapy after induction chemotherapy in stage IIIA-N2 non-small-cell lung cancer. J Natl Cancer Inst. 2007 Mar 21. 99 (6):442-50. [View Abstract]
  141. Daly BD, Ebright MI, Walkey AJ, et al. Impact of neoadjuvant chemoradiotherapy followed by surgical resection on node-negative T3 and T4 non-small cell lung cancer. J Thorac Cardiovasc Surg. 2011 Jun. 141(6):1392-7. [View Abstract]
  142. Ahn MJ, BCC, Siena S, Drilon A, et al. Entrectinib in patients with locally advanced or metastatic ROS1 fusion-positive non-small cell lung cancer (NSCLC) Abstract OA14.06. Presented at the IASLC 18th World Conference on Lung Cancer; Yokohama, Japan. 2017. J Thoracic Onc. 2017 Nov;12(11; suppl 2):S1783.
  143. Drilon A, Barlesi F, DeBraud F, Cho BC, Ahn MJ, Seina S, et al. Entrectinib in locally advanced or metastatic ROS1 fusion-positive non-small cell lung cancer (NSCLC): Integrated analysis of ALKA-372-001, STARTRK-1 and STARTRK-2 (abstract CT192). Presented at the American Association for Cancer Research (AACR) Annual Meeting 2019 March 29-April 3. Atlanta, GA.
  144. Rosell R, Moran T, Queralt C, et al. Screening for epidermal growth factor receptor mutations in lung cancer. N Engl J Med. 2009 Sep 3. 361(10):958-67. [View Abstract]
  145. Sequist LV, Yang JC, Yamamoto N, et al. Phase III Study of Afatinib or Cisplatin Plus Pemetrexed in Patients With Metastatic Lung Adenocarcinoma With EGFR Mutations. J Clin Oncol. 2013 Jul 1. [View Abstract]
  146. Gilotrif (afatinib [package insert]. Ridgefield, CT: Boehringer Ingelheim Pharmaceuticals, Inc. January, 2018. Available at
  147. Soria JC, Felip E, Cobo M, Lu S, Syrigos K, Lee KH, et al. Afatinib versus erlotinib as second-line treatment of patients with advanced squamous cell carcinoma of the lung (LUX-Lung 8): an open-label randomised controlled phase 3 trial. Lancet Oncol. 2015 Aug. 16 (8):897-907. [View Abstract]
  148. Lynch TJ, Bell DW, Sordella R. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004 May 20. 350(21):2129-39. [View Abstract]
  149. Thatcher N, Chang A, Parikh P, et al. Gefitinib plus best supportive care in previously treated patients with refractory advanced non-small-cell lung cancer: results from a randomised, placebo-controlled, multicentre study (Iressa Survival Evaluation in Lung Cancer). Lancet. 2005 Oct 29-Nov 4. 366(9496):1527-37. [View Abstract]
  150. Douillard JY, Ostoros G, Cobo M, Ciuleanu T, McCormack R, Webster A, et al. First-line gefitinib in Caucasian EGFR mutation-positive NSCLC patients: a phase-IV, open-label, single-arm study. Br J Cancer. 2014 Jan 7. 110 (1):55-62. [View Abstract]
  151. Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, Isobe H, et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med. 2010 Jun 24. 362 (25):2380-8. [View Abstract]
  152. Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009 Sep 3. 361(10):947-57. [View Abstract]
  153. Fukuoka M, Wu YL, Thongprasert S, et al. Biomarker Analyses and Final Overall Survival Results From a Phase III, Randomized, Open-Label, First-Line Study of Gefitinib Versus Carboplatin/Paclitaxel in Clinically Selected Patients With Advanced Non-Small-Cell Lung Cancer in Asia (IPASS). J Clin Oncol. 2011 Jul 20. 29(21):2866-74. [View Abstract]
  154. Shepherd FA, Pereira J, Ciuleanu TE, et al. A randomized placebo-controlled trial of erlotinib in patients with advanced non-small cell lung cancer (NSCLC) following failure of 1st line or 2nd line chemotherapy. A National Cancer Institute of Canada Clinical Trials Group (NCIC CTG) trial. J Clin Oncol. 2004. Vol 22, No 14S (July 15 Supplement):Abstract 7022.
  155. Shepherd FA, Rodrigues Pereira J, Ciuleanu T, et al. Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med. 2005 Jul 14. 353(2):123-32. [View Abstract]
  156. Zhou C, Wu YL, Chen G, et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol. 2011 Aug. 12(8):735-42. [View Abstract]
  157. Gridelli C, Ciardiello F, Gallo C, et al. First-Line Erlotinib Followed by Second-Line Cisplatin-Gemcitabine Chemotherapy in Advanced Non-Small-Cell Lung Cancer: The TORCH Randomized Trial. J Clin Oncol. 2012 Aug 20. 30(24):3002-11. [View Abstract]
  158. Herbst RS, Ansari R, Bustin F, et al. Efficacy of bevacizumab plus erlotinib versus erlotinib alone in advanced non-small-cell lung cancer after failure of standard first-line chemotherapy (BeTa): a double-blind, placebo-controlled, phase 3 trial. Lancet. 2011 May 28. 377(9780):1846-54. [View Abstract]
  159. Hirsch FR, Kabbinavar F, Eisen T, et al. A randomized, phase II, biomarker-selected study comparing erlotinib to erlotinib intercalated with chemotherapy in first-line therapy for advanced non-small-cell lung cancer. J Clin Oncol. 2011 Sep 10. 29(26):3567-73. [View Abstract]
  160. Tagrisso (osimertinib) [package insert]. Wilmington, DE 19850: AstraZeneca Pharmaceuticals LP. April 2018. Available at
  161. Soria JC, Ohe Y, Vansteenkiste J, Reungwetwattana T, Chewaskulyong B, et al. Osimertinib in Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. N Engl J Med. 2018 Jan 11. 378 (2):113-125. [View Abstract]
  162. Soria JC, et al. Osimertinib in Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. N Engl J Med. 2018 Jan 11. 378 (2):113-125. [View Abstract]
  163. Wu YL, Cheng Y, Zhou X, Lee KH, Nakagawa K, Niho S, et al. Dacomitinib versus gefitinib as first-line treatment for patients with EGFR-mutation-positive non-small-cell lung cancer (ARCHER 1050): a randomised, open-label, phase 3 trial. Lancet Oncol. 2017 Nov. 18 (11):1454-1466. [View Abstract]
  164. Mok TS, Cheng Y, Zhou X, Lee KH, Nakagawa K, Niho S, et al. Improvement in Overall Survival in a Randomized Study That Compared Dacomitinib With Gefitinib in Patients With Advanced Non-Small-Cell Lung Cancer and EGFR-Activating Mutations. J Clin Oncol. 2018 Aug 1. 36 (22):2244-2250. [View Abstract]
  165. Pirker R, Szczesna A, von Pawel J, et al. FLEX: A randomized, multicenter, phase III study of cetuximab in combination with cisplatin/vinorelbine (CV) versus CV alone in the first-line treatment of patients with advanced non-small cell lung cancer (NSCLC). J Clin Oncol. 2008. 26:1006s (Abstract).
  166. Pirker R, Pereira JR, von Pawel J, et al. EGFR expression as a predictor of survival for first-line chemotherapy plus cetuximab in patients with advanced non-small-cell lung cancer: analysis of data from the phase 3 FLEX study. Lancet Oncol. 2012 Jan. 13(1):33-42. [View Abstract]
  167. FDA Approves First Targeted Therapy to Treat Aggressive Form of Lung Cancer. U.S. Food & Drug Administration. Available at https://www.fda.gov/news-events/press-announcements/fda-approves-first-targeted-therapy-treat-aggressive-form-lung-cancer. May 6, 2020; Accessed: May 12, 2020.
  168. Thatcher N, Hirsch FR, Luft AV, Szczesna A, Ciuleanu TE, Dediu M, et al. Necitumumab plus gemcitabine and cisplatin versus gemcitabine and cisplatin alone as first-line therapy in patients with stage IV squamous non-small-cell lung cancer (SQUIRE): an open-label, randomised, controlled phase 3 trial. Lancet Oncol. 2015 Jul. 16 (7):763-74. [View Abstract]
  169. Paz-Ares L, Luft A, Vicente D, Tafreshi A, Gümüş M, Mazières J, et al. Pembrolizumab plus Chemotherapy for Squamous Non-Small-Cell Lung Cancer. N Engl J Med. 2018 Nov 22. 379 (21):2040-2051. [View Abstract]
  170. de Boer RH, Arrieta O, Yang CH, et al. Vandetanib Plus Pemetrexed for the Second-Line Treatment of Advanced Non-Small-Cell Lung Cancer: A Randomized, Double-Blind Phase III Trial. J Clin Oncol. 2011 Mar 10. 29(8):1067-74. [View Abstract]
  171. Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati A. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med. 2006 Dec 14. 355(24):2542-50. [View Abstract]
  172. Reck M, von Pawel J, Zatloukal P, et al. Phase III trial of cisplatin plus gemcitabine with either placebo or bevacizumab as first-line therapy for nonsquamous non-small-cell lung cancer: AVAil. J Clin Oncol. 2009 Mar 10. 27(8):1227-34. [View Abstract]
  173. Soria JC, Mauguen A, Reck M, et al. Systematic review and meta-analysis of randomised, phase II/III trials adding bevacizumab to platinum-based chemotherapy as first-line treatment in patients with advanced non-small-cell lung cancer. Ann Oncol. 2012 Nov 23. [View Abstract]
  174. Stark, Angela. FDA approves first biosimilar for the treatment of cancer. FDA News Release. 09/14/2017. Available at https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm576112.htm
  175. Garon EB, Ciuleanu TE, Arrieta O, Prabhash K, Syrigos KN, et al. Ramucirumab plus docetaxel versus placebo plus docetaxel for second-line treatment of stage IV non-small-cell lung cancer after disease progression on platinum-based therapy (REVEL): a multicentre, double-blind, randomised phase 3 trial. Lancet. 2014 Aug 23. 384 (9944):665-73. [View Abstract]
  176. Kwak EL, Bang YJ, Camidge DR, et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med. 2010 Oct 28. 363(18):1693-703. [View Abstract]
  177. Crizotinib now standard of care for ALK+ NSCLC. Medscape Medical News. Available at http://www.medscape.com/viewarticle/771853. Accessed: Oct 16 2012.
  178. Shaw AT, Kim DW, Nakagawa K, et al. Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med. 2013 Jun 20. 368(25):2385-94. [View Abstract]
  179. Shaw AT, Ou SH, Bang YJ, Camidge DR, Solomon BJ, Salgia R, et al. Crizotinib in ROS1-rearranged non-small-cell lung cancer. N Engl J Med. 2014 Nov 20. 371 (21):1963-71. [View Abstract]
  180. Kim DW, Tiseo M, Ahn MJ, Reckamp KL, Hansen KH, Kim SW, et al. Brigatinib in Patients With Crizotinib-Refractory Anaplastic Lymphoma Kinase-Positive Non-Small-Cell Lung Cancer: A Randomized, Multicenter Phase II Trial. J Clin Oncol. 2017 May 5. JCO2016715904. [View Abstract]
  181. Shaw AT, Kim DW, Mehra R, et al. Ceritinib in ALK-rearranged non-small-cell lung cancer. N Engl J Med. 2014 Mar 27. 370(13):1189-97. [View Abstract]
  182. Soria JC, Tan DS, Chiari R, Wu YL, Paz-Ares L, Wolf J, et al. First-line ceritinib versus platinum-based chemotherapy in advanced ALK-rearranged non-small-cell lung cancer (ASCEND-4): a randomised, open-label, phase 3 study. Lancet. 2017 Mar 4. 389 (10072):917-929. [View Abstract]
  183. Cho BC, et al. ASCEND-8: A Randomized Phase 1 Study of Ceritinib, 450 mg or 600 mg, Taken with a Low-Fat Meal versus 750 mg in Fasted State in Patients with Anaplastic Lymphoma Kinase (ALK)-Rearranged Metastatic Non-Small Cell Lung Cancer (NSCLC). J Thorac Oncol. 2017 Sep. 12 (9):1357-1367. [View Abstract]
  184. Peters S, Camidge DR, Shaw AT, Gadgeel S, Ahn JS, Kim DW, et al. Alectinib versus Crizotinib in Untreated ALK-Positive Non-Small-Cell Lung Cancer. N Engl J Med. 2017 Aug 31. 377 (9):829-838. [View Abstract]
  185. Hida T, Nokihara H, Kondo M, Kim YH, Azuma K, Seto T, et al. Alectinib versus crizotinib in patients with ALK-positive non-small-cell lung cancer (J-ALEX): an open-label, randomised phase 3 trial. Lancet. 2017 Jul 1. 390 (10089):29-39. [View Abstract]
  186. Solomon BJ, Besse B, Bauer TM, Felip E, Soo RA, Camidge DR, et al. Lorlatinib in patients with ALK-positive non-small-cell lung cancer: results from a global phase 2 study. Lancet Oncol. 2018 Nov 6. [View Abstract]
  187. Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. N Engl J Med. 2015 Jul 9. 373 (2):123-35. [View Abstract]
  188. Reck M, Schenker M, Lee KH, Provencio M, Nishio M, Lesniewski-Kmak K, et al. Nivolumab plus ipilimumab versus chemotherapy as first-line treatment in advanced non-small-cell lung cancer with high tumour mutational burden: patient-reported outcomes results from the randomised, open-label, phase III CheckMate 227 trial. Eur J Cancer. 2019 Jul. 116:137-147. [View Abstract]
  189. Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015 May 21. 372 (21):2018-28. [View Abstract]
  190. Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med. 2016 Oct 8. [View Abstract]
  191. Mok TSK, Wu YL, Kudaba I, Kowalski DM, Cho BC, Turna HZ, et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial. Lancet. 2019 Apr 4. [View Abstract]
  192. Langer CJ, Gadgeel SM, Borghaei H, Papadimitrakopoulou VA, Patnaik A, Powell SF, et al. Carboplatin and pemetrexed with or without pembrolizumab for advanced, non-squamous non-small-cell lung cancer: a randomised, phase 2 cohort of the open-label KEYNOTE-021 study. Lancet Oncol. 2016 Nov. 17 (11):1497-1508. [View Abstract]
  193. Herbst RS, Baas P, Kim DW, Felip E, Pérez-Gracia JL, Han JY, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016 Apr 9. 387 (10027):1540-50. [View Abstract]
  194. Gandhi L, et al; KEYNOTE-189 Investigators. Pembrolizumab plus Chemotherapy in Metastatic Non-Small-Cell Lung Cancer. N Engl J Med. 2018 May 31. 378(22):2078-2092. [View Abstract]
  195. Fehrenbacher L, Spira A, Ballinger M, Kowanetz M, Vansteenkiste J, Mazieres J, et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet. 2016 Apr 30. 387 (10030):1837-46. [View Abstract]
  196. Barlesi F, Park K, Ciardiello F, von Pawel J, Gadgeel S, Hida T, et al. A randomized phase 3 study of atezolizumab (an engineered anti-PDL1 antibody) compared to docetaxel in patients with locally advanced or metastatic non-small cell lung cancer who have failed platinum therapy - "OAK". ClinicalTrials.gov. Available at https://clinicaltrials.gov/ct2/show/NCT02008227. January 9, 2019; Accessed: June 15, 2019.
  197. Tecentriq (atezolizumab) [package insert]. South San Francisco, CA: Genentech, Inc. May 2020. Available at
  198. Karp DD, Paz-Ares LG, Novello S, et al. Phase II study of the anti-insulin-like growth factor type 1 receptor antibody CP-751,871 in combination with paclitaxel and carboplatin in previously untreated, locally advanced, or metastatic non-small-cell lung cancer. J Clin Oncol. 2009 May 20. 27(15):2516-22. [View Abstract]
  199. FDA grants regular approval to dabrafenib and trametinib combination for metastatic NSCLC with BRAF V600E mutation. FDA.gov. Available at https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm564331.htm. June 22, 2017; Accessed: June 15, 2019.
  200. Retevmo (selpercatinib) [package insert]. Indianapolis, IN: Eli Lilly. 2020 May. Available at
  201. Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med. 2016 Nov 10. 375 (19):1823-1833. [View Abstract]
  202. Berner F, Bomze D, Diem S, et al. Association of Checkpoint Inhibitor-Induced Toxic Effects With Shared Cancer and Tissue Antigens in Non-Small Cell Lung Cancer. JAMA Oncol. 2019 Apr 25. [View Abstract]
  203. [Guideline] National Comprehensive Cancer Network. Management of Immunotherapy-Related Toxicities. NCCN. Available at https://www.nccn.org/professionals/physician_gls/pdf/immunotherapy.pdf. Version 2.2019 — April 8, 2019; Accessed: August 1, 2019.
  204. Pignon JP, Tribodet H, Scagliotti GV, et al. Lung adjuvant cisplatin evaluation: a pooled analysis by the LACE Collaborative Group. J Clin Oncol. 2008 Jul 20. 26(21):3552-9. [View Abstract]
  205. Strauss GM, Herndon JE 2nd, Maddaus MA, et al. Adjuvant paclitaxel plus carboplatin compared with observation in stage IB non-small-cell lung cancer: CALGB 9633 with the Cancer and Leukemia Group B, Radiation Therapy Oncology Group, and North Central Cancer Treatment Group Study Groups. J Clin Oncol. 2008 Nov 1. 26(31):5043-51. [View Abstract]
  206. Gore EM, Bae K, Wong SJ, et al. Phase III comparison of prophylactic cranial irradiation versus observation in patients with locally advanced non-small-cell lung cancer: primary analysis of radiation therapy oncology group study RTOG 0214. J Clin Oncol. 2011 Jan 20. 29(3):272-8. [View Abstract]
  207. Sun A, Bae K, Gore EM, et al. Phase III trial of prophylactic cranial irradiation compared with observation in patients with locally advanced non-small-cell lung cancer: neurocognitive and quality-of-life analysis. J Clin Oncol. 2011 Jan 20. 29(3):279-86. [View Abstract]
  208. Lynch TJ, Patel T, Dreisbach L, et al. Cetuximab and first-line taxane/carboplatin chemotherapy in advanced non-small-cell lung cancer: results of the randomized multicenter phase III trial BMS099. J Clin Oncol. 2010 Feb 20. 28(6):911-7. [View Abstract]
  209. Biesma B, Wymenga AN, Vincent A, et al. Quality of life, geriatric assessment and survival in elderly patients with non-small-cell lung cancer treated with carboplatin-gemcitabine or carboplatin-paclitaxel: NVALT-3 a phase III study. Ann Oncol. 2011 Jul. 22(7):1520-7. [View Abstract]
  210. Raz DJ, Lanuti M, Gaissert HC, et al. Outcomes of patients with isolated adrenal metastasis from non-small cell lung carcinoma. Ann Thorac Surg. 2011 Nov. 92(5):1788-92; discussion 1793. [View Abstract]
  211. Ciuleanu T, Brodowicz T, Zielinski C, et al. Maintenance pemetrexed plus best supportive care versus placebo plus best supportive care for non-small-cell lung cancer: a randomised, double-blind, phase 3 study. Lancet. 2009 Oct 24. 374(9699):1432-40. [View Abstract]
  212. Belani CP, Brodowicz T, Ciuleanu TE, et al. Quality of life in patients with advanced non-small-cell lung cancer given maintenance treatment with pemetrexed versus placebo (H3E-MC-JMEN): results from a randomised, double-blind, phase 3 study. Lancet Oncol. 2012 Mar. 13(3):292-9. [View Abstract]
  213. Paz-Ares L, de Marinis F, Dediu M, et al. Maintenance therapy with pemetrexed plus best supportive care versus placebo plus best supportive care after induction therapy with pemetrexed plus cisplatin for advanced non-squamous non-small-cell lung cancer (PARAMOUNT): a double-blind, phase 3, randomised controlled trial. Lancet Oncol. 2012 Mar. 13(3):247-55. [View Abstract]
  214. [Guideline] Masters GA, Temin S, Azzoli CG, Giaccone G, Baker S Jr, Brahmer JR, et al. Systemic Therapy for Stage IV Non-Small-Cell Lung Cancer: American Society of Clinical Oncology Clinical Practice Guideline Update. J Clin Oncol. 2015 Oct 20. 33 (30):3488-515. [View Abstract]
  215. Jorenby DE, Leischow SJ, Nides MA, et al. A controlled trial of sustained-release bupropion, a nicotine patch, or both for smoking cessation. N Engl J Med. 1999 Mar 4. 340(9):685-91. [View Abstract]
  216. Mallin R. Smoking cessation: integration of behavioral and drug therapies. Am Fam Physician. 2002 Mar 15. 65(6):1107-14. [View Abstract]
  217. Moysich KB, Menezes RJ, Ronsani A, et al. Regular aspirin use and lung cancer risk. BMC Cancer. 2002 Nov 26. 2:31. [View Abstract]
  218. Jaklitsch MT, Jacobson FL, Austin JH, Field JK, Jett JR, Keshavjee S, et al. The American Association for Thoracic Surgery guidelines for lung cancer screening using low-dose computed tomography scans for lung cancer survivors and other high-risk groups. J Thorac Cardiovasc Surg. 2012 Jul. 144 (1):33-8. [View Abstract]
  219. [Guideline] Kalemkerian GP, Narula N, Kennedy EB, Biermann WA, Donington J, Leighl NB, et al. Molecular Testing Guideline for the Selection of Patients With Lung Cancer for Treatment With Targeted Tyrosine Kinase Inhibitors: American Society of Clinical Oncology Endorsement of the College of American Pathologists/International Association for the Study of Lung Cancer/Association for Molecular Pathology Clinical Practice Guideline Update. J Clin Oncol. 2018 Mar 20. 36 (9):911-919. [View Abstract]
  220. [Guideline] Planchard D, Popat S, Kerr K, Novello S, Smit EF, Faivre-Finn C, et al. Metastatic non-small cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. ESMO. Available at https://www.esmo.org/guidelines/lung-and-chest-tumours/metastatic-non-small-cell-lung-cancer. September 18, 2019; Accessed: May 19, 2020.
  221. Hanna N, Johnson D, Temin S, Baker S Jr, Brahmer J, Ellis PM, et al. Systemic Therapy for Stage IV Non-Small-Cell Lung Cancer: American Society of Clinical Oncology Clinical Practice Guideline Update. J Clin Oncol. 2017 Aug 14. JCO2017746065. [View Abstract]
  222. [Guideline] Rodrigues G, Choy H, Bradley J, Rosenzweig KE, Bogart J, Curran WJ Jr, et al. Adjuvant radiation therapy in locally advanced non-small cell lung cancer: Executive summary of an American Society for Radiation Oncology (ASTRO) evidence-based clinical practice guideline. Pract Radiat Oncol. 2015 May-Jun. 5 (3):149-55. [View Abstract]
  223. [Guideline] Rodrigues G, Choy H, Bradley J, Rosenzweig KE, Bogart J, Curran WJ Jr, et al. Definitive radiation therapy in locally advanced non-small cell lung cancer: Executive summary of an American Society for Radiation Oncology (ASTRO) evidence-based clinical practice guideline. Pract Radiat Oncol. 2015 May-Jun. 5 (3):141-8. [View Abstract]
  224. [Guideline] Bezjak A, Temin S, Franklin G, Giaccone G, Govindan R, Johnson ML, et al. Definitive and Adjuvant Radiotherapy in Locally Advanced Non-Small-Cell Lung Cancer: American Society of Clinical Oncology Clinical Practice Guideline Endorsement of the American Society for Radiation Oncology Evidence-Based Clinical Practice Guideline. J Clin Oncol. 2015 Jun 20. 33 (18):2100-5. [View Abstract]
  225. Planchard D, Kim TM, Mazieres J, Quoix E, Riely G, Barlesi F, et al. Dabrafenib in patients with BRAF(V600E)-positive advanced non-small-cell lung cancer: a single-arm, multicentre, open-label, phase 2 trial. Lancet Oncol. 2016 May. 17 (5):642-50. [View Abstract]
  226. Steuer CE, Khuri FR, Ramalingam SS. The next generation of epidermal growth factor receptor tyrosine kinase inhibitors in the treatment of lung cancer. Cancer. 2014 Dec 17. [View Abstract]
  227. Alecensa (alectinib) [package insert]. South San Francisco, CA: Genentech USA, Inc. December, 2015. Available at
  228. Ou SI, Ahn JS, De Petris L, Govindan R, Yang JC, Hughes B, et al. Alectinib in Crizotinib-Refractory ALK-Rearranged Non-Small-Cell Lung Cancer: A Phase II Global Study. J Clin Oncol. 2015 Nov 23. [View Abstract]
  229. Gohagan J, Marcus P, Fagerstrom R, Pinsky P, Kramer B, Prorok P. Baseline findings of a randomized feasibility trial of lung cancer screening with spiral CT scan vs chest radiograph: the Lung Screening Study of the National Cancer Institute. Chest. 2004 Jul. 126(1):114-21. [View Abstract]
  230. Mekinist (trametinib) [package insert]. East Hanover, New Jersey 07936: Novartis Pharmaceuticals Corporation. 06/2017. Available at
  231. Tafinlar (dabrafenib) [package insert]. East Hanover, New Jersey 07936: Novartis Pharmaceuticals Corporation. June 2017. Available at
  232. [Guideline] COVID-19 Guidelines for Triage of Thoracic Patients. American College of Surgeons. Available at https://www.facs.org/covid-19/clinical-guidance/elective-case/thoracic-cancer. March 24, 2020; Accessed: April 10, 2020.

Non–small cell lung cancer. A cavitating right lower lobe squamous cell carcinoma.

Non–small cell lung cancer. A cavitating right lower lobe squamous cell carcinoma.

Non–small cell lung cancer. Prognostic factors for lung cancer.

Non–small cell lung cancer. Symptoms and signs of lung cancer.

Non–small cell lung cancer. Symptoms and signs of lung cancer.

Non–small cell lung cancer. Diagnostic approach for possible lung cancer.

Staging workup for non–small cell lung cancer.

Non–small cell lung cancer. Symptoms and signs of lung cancer.

Non–small cell lung cancer. Bronchoscopy. A large central lesion was diagnosed as non–small cell carcinoma.

Non–small cell lung cancer. Left pleural effusion and volume loss secondary to non–small cell carcinoma of the left lower lobe. The pleural effusion was sampled and found to be malignant; therefore, the lesion is inoperable.

Lung cancer, small cell. Contrast-enhanced CT scan of the chest shows a large left lung and a hilar mass, with invasion of the left pulmonary artery.

Lung cancer, small cell. Whole-body nuclear medicine bone scanning with anterior and posterior images reveal multiple abnormal areas of increased radiotracer activity in the pelvis, spine, ribs, and left scapula. These findings are consistent with bony metastatic disease. The bones are commonly affected in patients with small-cell lung cancer.

Lung cancer, small cell. Coronal positron emission tomogram shows abnormal areas of increased metabolic activity in the left hilar and left adrenal regions consistent with a hilar tumor with left adrenal metastasis.

Non–small cell lung cancer. Performance status scales for patients with cancer.

Non–small cell lung cancer. Symptoms and signs of lung cancer.

Non–small cell lung cancer. Diagnostic approach for possible lung cancer.

Staging workup for non–small cell lung cancer.

Treatment recommendations and future research directions in the management of non–small cell lung cancer.

Non–small cell lung cancer. Performance status scales for patients with cancer.

Non–small cell lung cancer. Prognostic factors for lung cancer.

Non–small cell lung cancer. Bronchoscopy. A large central lesion was diagnosed as non–small cell carcinoma.

Non–small cell lung cancer. Left pleural effusion and volume loss secondary to non–small cell carcinoma of the left lower lobe. The pleural effusion was sampled and found to be malignant; therefore, the lesion is inoperable.

Non–small cell lung cancer. Left upper collapse is almost always secondary to endobronchial bronchogenic carcinoma.

Non–small cell lung cancer. Complete left lung collapse secondary to bronchogenic carcinoma of left mainstem bronchus.

Non–small cell lung cancer. A cavitating right lower lobe squamous cell carcinoma.

Non–small cell lung cancer. CT scan shows cavitation and air-fluid level.

Non–small cell lung cancer. Patient has right lower lobe opacity. This is not well circumscribed and was found to be a squamous cell carcinoma.

Lung cancer, small cell. Contrast-enhanced CT scan of the chest shows a large left lung and a hilar mass, with invasion of the left pulmonary artery.

Lung cancer, small cell. Coronal positron emission tomogram shows abnormal areas of increased metabolic activity in the left hilar and left adrenal regions consistent with a hilar tumor with left adrenal metastasis.

Lung cancer, small cell. Whole-body nuclear medicine bone scanning with anterior and posterior images reveal multiple abnormal areas of increased radiotracer activity in the pelvis, spine, ribs, and left scapula. These findings are consistent with bony metastatic disease. The bones are commonly affected in patients with small-cell lung cancer.

Lung squamous carcinoma 4x: low power magnification of moderately differentiated squamous cell carcinoma showing irregular nests of tumor cells with focal areas of keratinization (pink-orange areas).

Lung squamous carcinoma 20x: higher power magnification of moderately differentiated squamous cell carcinoma showing focal areas of keratinization (pink-orange areas) just to the right of center.

Lung adenocarcinoma 4x: low power magnification of moderately differentiated adenocarcinoma showing rounded nests of pale staining tumor cells with gland lumina within some of the clusters.