National Academies Press: OpenBook
« Previous: 5 Breast Cancer
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

6

Lung Cancer

Lung cancer is the most common cause of cancer mortality in the United States, despite reductions in smoking over the past 50 years that have led to a decrease in smoking-related lung cancer incidence and mortality. Lung cancers are broadly classified as non-small-cell lung cancer (NSCLC) or small-cell lung cancer (SCLC). NSCLC accounts for the majority (80–85%) of all lung cancer cases in the United States, while SCLC, which has a much poorer prognosis, represents approximately 10–15% of lung cancers (ACS, 2019). For more about the epidemiology of lung cancer, see Chapter 3.

Recent advances in lung cancer care (i.e., prevention, screening, diagnosis, and treatment) have contributed to improved outcomes for patients, including longer survival and less mortality for patients with NSCLC. Nonetheless, the morbidity and mortality associated with lung cancer remain high. The impairments and subsequent functional limitations experienced by lung cancer survivors are a consequence of the adverse effects of the cancer itself; of the adverse effects of its treatment; and of the high risk of recurrence, metastases, or development of new primary cancers, as well as the presence of major comorbid conditions often associated with smoking and aging. To help manage these impairments and functional limitations as well as the adverse effects from both the cancer and its treatment, palliative and supportive care may be recommended for patients at the time of their diagnosis or during the course of treatment. Two of the most common comorbidities in lung cancer survivors are chronic obstructive pulmonary disease (COPD) and cardiovascular disease. Metastases of the primary lung cancer and new primary cancers are also highly likely to occur in this population. Adults with either NSCLC or SCLC, who are often current or

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

former smokers, have a particularly high risk of developing other tobacco-associated malignancies, including another lung cancer (NSCLC, SCLC) or other cancers of the head and neck, bladder, esophagus, or pancreas. Ongoing monitoring for new primary cancers, disease progression, and cancer recurrence requires frequent physical examination and imaging and is the standard of care for lung cancer (see Figure 4-1).

This chapter describes the screening, diagnosis, treatment, and survivorship of lung cancer in adults beginning with risk factors and survival rates. This is followed by information about the diagnosis and staging of lung cancer, and the treatment of NSCLC and SCLC. The final section describes the range of lung cancer-related impairments that survivors may experience and identifies the potential adverse effects on survivor functioning.

RISK FACTORS

Cigarette smoking, along with cigar and pipe smoking, is by far the leading risk factor for lung cancer. In smokers, the risk of developing lung cancer increases with age and with both the quantity and the duration of smoking. The second-leading cause of lung cancer in the United States is exposure to radon gas, which is released from soil and can accumulate in indoor air (NCI, 2011). Other risk factors include exposure to second-hand smoke, asbestos, certain metals (e.g., chromium, cadmium, arsenic), some organic chemicals, radiation, air pollution, and diesel exhaust. Increased risk is also associated with specific occupational exposures (i.e., rubber manufacturing, paving, roofing, painting, and chimney sweeping) (Field and Withers, 2012). A personal or family history of lung cancer is another risk factor for lung cancer (Cannon-Albright et al., 2019). There are certain genes and chromosomes (e.g., TP53 germline sequence variations and a marker on chromosome 15) that are linked to an increased risk of lung cancer (Zappa and Mousa, 2016).

LUNG CANCER SCREENING

Lung cancer typically presents in an advanced stage (stages are defined below), so the development of a screening test for high-risk individuals is an important advancement in lung cancer care (de Koning et al., 2020; National Lung Screening Trial Research Team et al., 2011). In 2013 the U.S. Preventive Services Task Force (USPSTF) recommended annual screening for lung cancer with low-dose computed tomography (LDCT) in adults ages 55 through 80 years who have at least a 30-pack-a-year smoking history and currently smoke or have quit within the past 15 years (USPSTF, 2013). At the time of this report’s publication, USPSTF was in the process

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

of updating its 2013 lung cancer screening recommendations (USPSTF, 2020). On July 7, 2020, USPSTF released a draft decision analysis about lung cancer screening for public comment. The proposed screening guideline broadens the target population by expanding the age range to include ages 50 to 80 years and by reducing the number of pack-years of smoking exposure to 20 years (see Table 4-1). USPSTF states that such changes would potentially increase the eligible population beyond that defined by the current criteria and may reduce disparities in eligibility by sex and race/ethnicity (CISNET, 2020).

Despite guideline recommendations and insurance coverage for those at high risk of lung cancer (under the Patient Protection and Affordable Care Act, Medicare Part B, and some state Medicaid programs), lung cancer screening is not widely performed. Overall, only 5.7% of high-risk individuals were screened in 2019 (ALA, 2020). Moreover, in 2020 only 38 states covered screening of high-risk Medicaid enrollees (ALA, 2020). Barriers to the implementation of lung cancer screening are the subject of research investigations; for example, some studies are examining clinician-related factors affecting screening practices (Ersek et al., 2016; Lewis et al., 2015).

Lung cancer may be suspected on the basis of screening, incidentally detected lung nodules when imaging is performed for other purposes, or patient-reported symptoms. When it is suspected, protocols for diagnosis and cancer staging, which often occur simultaneously, are the foundations for clinical decisions about appropriate lung cancer treatment.

DIAGNOSIS AND STAGING OF LUNG CANCER

Diagnosing lung cancer, classifying the type, and determining the extent of the disease (cancer stage) are critically important to treatment management. Current approaches to the diagnosis of lung cancer include not only conventional histology (the microscopic examination of body tissues), but also immunohistochemistry1 and molecular and immune panels.

The most common presenting symptoms of lung cancer include cough, hemoptysis, dyspnea (shortness of breath), anorexia, fatigue, weight loss, and neurologic symptoms caused by paraneoplastic symptoms. In most cases the choice of diagnostic test is driven by the patients’ presenting symptoms, which may suggest an advanced stage (bone pain, headache, or weight loss) or point to a more local stage (cough, pneumonia). Tests and procedures determine the presence of lung cancer and identify the lung cancer stage.

Generally, lung cancer patients receive a computed tomography (CT) scan and a positron emission tomography (PET) scan; a brain magnetic

___________________

1 Immunohistochemistry is a laboratory method that uses antibodies to check for certain antigens (biomarkers) in a sample of tissue.

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

resonance imaging (MRI) scan is recommended in stage II and higher, is optional for patients with stage IB disease, and is not recommended for patients with stage IA disease (NCCN, 2020b). These procedures are used to detect the possible presence of sub-clinical or asymptomatic distant metastases. Patients with lung cancer stages II–III may undergo bronchoscopy, endobronchial ultrasound, or percutaneous lung needle biopsy to radiologically or pathologically confirm indeterminate findings of PET or CT scans. Other diagnostic tools may include image-guided transthoracic needle core biopsy, thoracentesis, and mediastinoscopy (NCCN, 2020b). Because both NSCLC and SCLC frequently spread to the liver, adrenal glands, bone, and brain, staging has traditionally been with a CT scan with intravenous contrast of the chest and abdomen along with brain imaging using an MRI scan. When an MRI scan is contraindicated, a contrast enhanced CT scan of the brain is acceptable. If cancer appears localized with a CT scan, a fluorodeoxyglucose positron emission tomography (FDG-PET) scan is performed because it images the whole body including bones and is the most sensitive test for cancer outside of the brain.

A diagnosis of lung cancer is confirmed by a pathologist primarily based on light microscopy of tumor tissue, obtained by a needle biopsy or surgical resection. This type of histologic work-up is usually adequate to distinguish SCLC from NSCLC and to differentiate among the subtypes of NSCLC. Specifically, NSCLC is further classified as either squamous (25%) or non-squamous lung cancer; the majority of non-squamous lung cancer is adenocarcinoma (40% of lung cancers), and there are other types (large cell, poorly differentiated NSCLC-not otherwise specified, other rare) that account for small percentages of all lung cancers (PDQ® Adult Treatment Editorial Board, 2020). Further testing with immunohistochemistry (CK7, CD20, TTF-1, P63, and/or napsin) may be needed to confirm lung cancer subtypes or to distinguish metastatic cancers that may mimic primary lung cancer. In addition, there are molecular and immunologic techniques for more complete pathological profiling of NSCLC that not only aid lung cancer diagnosis but that are essential for treatment decisions and prognosis.

In the current molecular era, more complete pathological profiling for NSCLC requires a large tissue sample, which can be challenging to obtain in advanced disease. When the cancer is localized (early stage), the tumor is resected, which usually produces enough tissue for pathologic diagnosis. In the case of a late-stage cancer that cannot be resected due to excessive risk or metastases, a needle biopsy is performed instead, which yields a small volume of tissue that may not support the diagnostic assessment necessary for selecting appropriate treatment in advanced disease. In such instances the emerging practice is to obtain multiple tissue cores at the time of core needle biopsy, or to perform the analysis on tumor genetic material in the blood, which is referred to as a “liquid biopsy.”

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

Lung Cancer Stages

As discussed in Chapter 4, the American Joint Committee on Cancer’s (AJCC’s) AJCC Cancer Staging Manual, Eighth Edition (AJCC, 2017; Goldstraw et al., 2016; Kay et al., 2017) is the staging model used for most cancers, including lung cancer, to characterize the malignancy. In the AJCC staging system, TNM stands for tumor (T), node (N), and distant metastases (M) (see Table 6-1). In lung cancer staging, the T stage characterizes the size of the tumor and the degree of local extension to other structures within the lung. The N stage characterizes the location of lymph node involvement within the chest as follows: N0 indicates no nodal spread; N1 indicates cancer involved nodes limited within the lung proper that could

TABLE 6-1 Anatomic Staging of Lung Cancer

Stage Detailed Stage TMN Stage
0 Stage 0
  • Tis, N0, M0
I Stage IA
  • T1mi, N0, M0
  • T1, N0, M0
Stage IB
  • T2, N0, M0
II Stage IIA
  • T2, N0, M0
Stage IIB
  • T1, N1, M0
  • T2, N1, M0
  • T3, N0, M0
III Stage IIIA
  • T1, N2, M0
  • T2, N2, M0
  • T3, N1, M0
  • T4, N0, M0
  • T4, N1, M0
Stage IIIB
  • T1, N3, M0
  • T2, N3, M0
  • T3, N2, M0
  • T4, N2, M0
Stage IIIC
  • T3, N3, M0
  • T4, N3, M0
IV Stage IVA
  • Any T, any N, M1
Stage IVB
  • Any T, any N, M1

NOTE: M = presence or absence of distant metastasis; N = extent of regional lymph node spread; T = size or extent of the primary tumor; Tis = carcinoma in situ; T1mi = minimally invasive adenocarcinoma.

SOURCES: Used with the permission of the American College of Surgeons, Chicago, Illinois. The original source for this information is the AJCC Cancer Staging Manual, Eighth Edition (2017) published by Springer International Publishing: the American College of Surgeons. Amin, M.B., Edge, S.B., Greene, F.L., et al. (Eds.). AJCC Cancer Staging Manual, 8th Ed. Springer New York, 2017, with permission.

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

be removed with the primary tumor; and N2 and N3 indicate regionally involved nodes limited to the mediastinum (area between the lungs in the middle of the chest) that could be potentially resected separately from the tumor, or distant nodal spread that is metastatic (outside of the thorax). The M stage distinguishes intrathoracic metastases (M1a) from extrathoracic ones, with the latter split into single-site metastasis (M1b) or multiple-site metastases (M1c) (Kay et al., 2017).

TNM classifications for lung cancer are translated to stages I, II, III and IV. Stage I and stage II are limited to the lung itself with no lymph node involvement or else involvement of only the hilar lymph nodes (N1 region). Stage III is defined by larger cancers and those that have spread to the mediastinum or low neck lymph node regions outside of the lung (N2 or N3 regions). A stage IV cancer is widespread with systemic metastatic disease. A significant proportion of NSCLC cases (40%) are stage IV at the time of diagnosis (PDQ® Adult Treatment Editorial Board, 2020).

As with other solid tumors, the TNM staging can be used to diagnosis SCLC. However, the predominant basis for SCLC treatment decisions is the Veterans Administration Lung Group 2-stage system, which defines limited-stage (LS) and extensive-stage (ES) SCLC (Kalemkerian, 2012). LS-SCLC, which occurs in approximately one-third of SCLC patients, is defined as disease that is confined to the hemithorax (one side of the chest) of origin, the mediastinum, or the supraclavicular nodes (nodes located above the collarbone) and that can be encompassed within a tolerable radiation field. ES-SCLC, which occurs in approximately two-thirds of SCLC patients, is defined as disease that has spread beyond the supraclavicular areas and lung and is too widespread to be included within a tolerable radiation field. Patients with distant metastases by definition have extensive-stage disease (Wang et al., 2019).

Survival Rates

Lung cancer is one of the most lethal cancers: more than half of people with lung cancer die within 1 year of being diagnosed (SEER, 2020, n.d.c). The all-stage 5-year survival rate for NSCLC is 24.9% and only 6.5% for SCLC (SEER, n.d.-a,-b). Table 6-2 shows the 5-year survival rates for NSCLC and SCLC by stage at the time of diagnosis.

Although lung cancer stages I–III are potentially curable, the reason for this high mortality rate is two-fold: First, most patients are diagnosed in advanced stages of the disease, when the cancer has metastasized to distant organs, and is incurable (stage IV). Second, the recurrence rate for those diagnosed with earlier stages (stages I–III) approaches 50% for NSCLC and 80% for SCLC. If lung cancer is diagnosed at an early (localized) stage, the overall 5-year survival improves dramatically to 59%; however, only 17% of lung cancers are diagnosed at this stage (SEER, n.d.-d).

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

TABLE 6-2 5-Year Survival Rates Since Diagnosis (diagnosed between 2010 and 2016)

Stage Percent Surviving NSCLC Percent Surviving SCLC
Stage I or II (localized) 63.1 27.2
Stage III (regional spread) 35.4 16.4
Stage IV (metastatic) 6.9 2.9
All stages combined 24.9 6.5

SOURCE: Howlader et al., 2020.

SCLC typically grows quickly, is highly metastatic, and is usually rapidly fatal. In 2012 the U.S. Congress classified SCLC as a recalcitrant cancer—that is, a cancer with a 5-year relative survival rate of less than 20% and estimated to cause the death of at least 30,000 individuals per year in the United States—and it authorized the National Cancer Institute to specifically dedicate resources to combat this disease.2

LUNG CANCER TREATMENT

In general, treatment options for lung cancer are determined by the histology, stage, and molecular characteristics of the tumor and by the patient’s general health and comorbidities. This section presents an overview of the current treatments for NSCLC and for SCLC. It reflects the National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology®, which are the leading evidence-based, consensus driven recommendations for the management of cancer in the United States (NCCN, 2020a,b). The NCCN guidelines state that clinical trials provide the best management option for patients, but the committee recognizes that such trials are not always available or accessible for an individual patient diagnosed with lung cancer.

Lung cancer treatments are generally categorized as local/regional therapies (surgery, radiation therapy, ablation) or systemic therapies (chemotherapy, immunotherapy, targeted therapy), and the various types are often used in combination. Improved survival of patients with lung cancer have followed from recent advances in genetics and biomarker testing, particularly for NSCLC, which have led to personalized medicine approaches with targeted treatments (Riely et al., 2009). As described in Chapter 4, targeted therapies are based on the identification of specific features of a patient’s tumor (e.g., an abnormal ALK gene) that may respond to the therapy. Chemotherapy, the foundation of medical oncology, is no longer the most

___________________

2 Recalcitrant Cancer Act of 2012, HR 733, 112th Congress.

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

effective systemic treatment for either NSCLC or SCLC (Mok et al., 2016; Reck et al., 2016; Solomon et al., 2014; Zhou et al., 2011).

Treatment of Non-Small-Cell Lung Cancer

This section describes the standard therapies that are used to treat NSCLC and is organized into treatments for cancer stages I–II, stage III, and stage IV.

Stages I–II NSCLC

Surgery is the standard-of-care approach for patients with stage I or stage II NSCLC who are eligible for surgery (NCCN, 2020b). Patients need to undergo an extensive preoperative evaluation of heart and lung function to determine if they are medically fit for surgery, which is typically done by a thoracic surgeon. While surgical resection is the treatment of choice for stages I–II NSCLC, many patients are not candidates for surgery either because of medical comorbidities, or deficits in functional status or in pulmonary function, or because of refusal to undergo surgery. For these patients, radiation therapy, ablation, and chemotherapy are alternatives to surgery.

Surgery

The surgical resection of a NSCLC stages I–II tumor involves the removal of the primary cancer within the lung, usually done by removing a single lobe of the lung (lobectomy), plus the removal of several lymph nodes within the mediastinum. Traditionally, the removal of a lobe of a lung required a thoracotomy, which involves a large incision and resulting scar extending from the patient’s back to the side. More recently, less invasive techniques have emerged and a growing number of patients may undergo video-assisted thorascopic surgery (VATS), which requires only small incisions to insert a camera and surgical instruments to remove the specimen. A recent analysis of the Society of Thoracic Surgeons General Thoracic Surgery Database of patients aged 65 or older who had undergone lobectomy for stage I lung cancer showed that 42% underwent a thoracotomy and 58% received a VATS procedure (Boffa et al., 2018). VATS procedures are also being done with robotic assistance. VATS lobectomy has equivalent or slightly improved safety and efficacy, as well as better patient outcomes, compared with traditional open lobectomy (Aoki et al., 2007; Yan et al., 2009). The removal of the entire lung (pneumonectomy) may be required in patients with tumors that are located near critical structures in the center of the chest such as the major airways, or that cross a lung fissure to involve more than one lobe. (James and Faber, 1999). Pneumonectomy is associated with a number of potential complications that involve not only

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

the respiratory system, but also the cardiovascular system and the pleural space, and that may result in a significant risk of perioperative mortality and long-term morbidity (Kopec et al., 1998). For these reasons the rate of pneumonectomy is declining. Sublobular resection, either segmentectomy or wedge resection, may be an option for some patients, particularly those with poor pulmonary reserve and with small peripheral nodules (<2 cm) (NCCN, 2020b).

Mediastinal staging, removal, and evaluation of lymph nodes between the lungs is a key element of lung cancer surgery mainly for prognosis and treatment planning. The optimal extent of lymph node surgery remains uncertain. A meta-analysis demonstrated that the dissection of the most at-risk lymph nodes results in a small improvement in survival compared with no lymph node sampling (Manser et al., 2005). A greater number of lymph nodes removed at the time of surgery improves the accuracy of staging (Gajra et al., 2003). Those found to have incidental tumor involvement of mediastinal lymph nodes (N2 or N3) are, by definition, upstaged to stage III.

Radiation therapy

When surgical resection of a NSCLC stage I or node-negative stage II tumor is not feasible, radiation as a single treatment modality is often used. Over the past 20 years, radiation therapy has improved considerably, and the current state-of-the-art technique is called stereotactic body radiation therapy (SBRT) (also referred to as stereotactic ablative radiotherapy; see Chapter 4). For patients with stages I–II NSCLC who are not surgical candidates, SBRT is the treatment of choice (Schneider et al., 2018). SBRT delivers a dose of 10–34 Gy as one fraction for small, peripheral tumors, and up to 60–70 Gy total dose in 8–10 fractions for central tumors (NCCN, 2020b). SBRT is suitable for localized tumors because features of the technology (such as the ability to adjust patient position) ensure that high doses of radiation are delivered to a precise location. SBRT has resulted in excellent local tumor control for stages I–II NSCLC in prospective studies with long-term primary tumor control rates of greater than 90% (Hobbs et al., 2018; Senthi et al., 2012). Nevertheless, patients still need close monitoring for recurrences elsewhere in the same lobe or lung, in the lymph node regions, and especially in distant organs (e.g., liver, bones, adrenal glands, brain, opposite lung).

Clinical trials are under way comparing surgical resection to SBRT. Should SBRT demonstrate better outcomes than surgery, it is possible that the standard of care for patients with stages I–II NSCLC would change (Chang et al., 2015; University of Texas Southwestern Medical Center, 2015; VA ORD, 2016).

The acute effects of SBRT are minimal and generally do not peak until 1 week or longer after treatment. Fatigue is the most commonly reported

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

acute effect and skin irritation (radiation dermatitis) is typically not seen or is quite mild. Some patients may experience cough from transient inflammation in the lung or airways. Additional sequelae of SBRT, such as scarring of the lung (lung fibrosis), are discussed in the section on Life After Lung Cancer Diagnosis.

Percutaneous image-guided ablation approaches

Although radiation therapy is the preferred treatment approach for patients with stages I–II NSCLC when surgery is not an option, in some cases (e.g., small tumors in stage I NSCLC), interventional radiologists can remove the tumors, with CT guidance, using techniques such as radiofrequency ablation, microwave ablation, or cryoablation (see Chapter 4 for more detail on ablation techniques). To induce tumor necrosis, these local ablative techniques rely on thermal energy: radiofrequency ablation (Ambrogi et al., 2011) and microwave ablation to apply heat to the tumor, whereas cryoablation applies intense cold (Gage and Baust, 1998).

Patients who require treatment of multiple synchronous or metachronous primary tumors are often good candidates for percutaneous ablation techniques. The best outcomes with percutaneous ablation have been reported for tumors less than 3 cm. With larger tumors, there are added complexities to the procedure, thus increasing the risk of complications.

Pneumothorax, pleural effusion, and hemothorax are the most common complications of ablation (Kashima et al., 2011). In different studies the rate of pneumothorax ranges from 1.3% to 60%, and requires thoracostomy tube placement in roughly 15% of patients (Kashima et al., 2011; Zheng et al., 2014). Other complications are less common, but include aseptic pleuritis (2.3%), pneumonia (<2.0%), and bleeding requiring blood transfusion (1.6%). In one study, deaths following ablation were 0.4% (Dupuy et al., 2006). Complication rates are influenced by longer ablation times, the use of multiple probes, the size and location of the lesion, and underlying lung disease.

Chemotherapy for high-probability of recurrence

Some patients with stage I–II NSCLC have a substantial probability of recurrence even after surgical resection, SBRT, or local ablation techniques. The probability that subclinical cancer has been left behind at the time of surgery is proportional to the stage, tumor size, nodal spread, and degree of invasion. For example, Brandt et al. (2018) found a distance recurrence rate of 20–50% with increasing T tumor stage in node-negative disease even after complete resection. A systematic review by the Cochrane Collaborative supports the use of adjuvant chemotherapy with a platinum-based agent following lung resection in early-stage lung cancer (Burdett et al., 2015). Adjuvant chemotherapy following surgery was found to result in an absolute increase

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

in survival of 4% (from 60% to 64%) at 5 years; a similar improvement was seen when adjuvant chemotherapy was given after surgery and postoperative radiation (from 29% to 33%) (Burdett et al., 2015). The NCCN guidelines recommend adjuvant chemotherapy with cisplatin plus a second chemotherapy agent (e.g., pemetrexed, gemcitabine, or docetaxel) given every 3 weeks for four cycles (NCCN, 2020b). Recurrence-free survival improved by 15%, time to locoregional recurrence improved by 21%, and time to distant recurrence improved by 25% as a result of adjuvant chemotherapy (Burdett et al., 2015).

In some cases, the administration of neoadjuvant therapy can also result in a reduction of tumor mass and thereby make some surgeries more likely to achieve a complete resection of the tumor. Neoadjuvant chemotherapy may also remove micro-metastases and improve patient treatment completion (Subramanian and Puri, 2019). One study found that neoadjuvant chemotherapy increased 5-year recurrence-free survival by 6% (from 30% to 36%) and yielded a 10% absolute benefit for distant recurrence (NSCLC Meta-analysis Collaborative Group, 2014). Although there are fewer studies on the use of neoadjuvant chemotherapy for early-stage NSCLC compared with adjuvant chemotherapy, Brandt et al. (2019) found that among patients with cT2-4N0-1M0 NSCLC, there is no significant difference in 5-year disease-free survival or overall survival between patients receiving adjuvant or neoadjuvant platinum-based chemotherapy, however, those receiving neoadjuvant chemotherapy were more likely to complete their therapy and had fewer severe toxicities from it.

Several studies are evaluating immunotherapy alone or in combination with chemotherapy as neoadjuvant or adjuvant therapy for early-stage NSCLC. It is likely that these agents may be introduced into the management of resectable NSCLC in the near future (Cascone et al., 2019; Forde et al., 2018; Kwiatkowski et al., 2019).

Stage III NSCLC

Stage III NSCLC represents a heterogeneous group of diseases divided into three major categories (see Table 6-1). Stage IIIA NSCLC can range from very large tumors with or without small nodes in the ipsilateral hilum (T3 N1 or T4 ± N1) to very small tumors with ipsilateral mediastinal nodes (T1–T2, N2). Stage IIIB NSCLC ranges from large tumors with ipsilateral mediastinal nodes (T3–T4, N2) to small tumors with contralateral or supraclavicular nodes (T1–T2, N3). Stage IIIC NSCLC consists of large tumors with contralateral or supraclavicular nodes (T3–T4, N3) (AJCC, 2017).

Due to the high probability of relapse, stage III NSCLC is treated with multiple treatment modalities in order to address the various malignant

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

behaviors of the cancer. Presently this includes combinations of local/regional treatments such as surgery and radiation with systemic treatments such as chemotherapy and/or immunotherapy. In short, stage III NSCLC lung cancer is quite diverse and so is its treatment.

Surgery

For stage III NCSLC, surgery is sometimes used in conjunction with chemotherapy and radiation therapy (trimodal therapy). Patients with larger or more involved nodes may not be candidates for surgery. Patients with one or more small (<3 cm in size) involved lymph nodes may be candidates for surgical resection (NCCN, 2020b). Patients with a greater number of metastatic nodes are not candidates for surgery. A randomized clinical trial demonstrated that patients in stage IIIA (pN2) disease, who received neoadjuvant chemotherapy (cisplatin plus etoposide) with radiation therapy followed by resection did not have improved survival, but those who were treated with a lobectomy rather than a pneumonectomy had longer median and 5-year progression-free survival (12.8 months and 22.4%, respectively) compared with patients who received chemotherapy and radiation therapy but did not undergo surgery (10.5 months and 11.1%, respectively); 5-year overall survival was also significantly improved in those receiving lobectomy after chemotherapy and radiation compared with those who did not (27.2% versus 20.3%) (Albain et al., 2009). N0 status at thoracotomy was predictive of overall survival. A majority of patients have extensive lymph node involvement of the mediastinum or do not have the physiologic reserve to tolerate such surgery. The use of surgery remains controversial in stage III NSCLC. Ultimately, a multidisciplinary approach is recommended for patients with stage III lung cancer that is resectable.

Concurrent chemotherapy and radiation therapy

Chemotherapy given at the same time as radiation therapy (concurrent chemoradiation) is the standard-of-care treatment for unresectable stage III NSCLC and may also be administered prior to, or following, surgical resection in patients for whom surgery may be curative.

More than 40 years ago patients with unresectable stage III NSCLC were treated with radiation therapy alone (60 Gy in 30 treatments) (Calikusu and Altinok, 2018) and had a poor chance of survival. Subsequent research provided evidence supporting the use of both radiation therapy and chemotherapy (Kubota et al., 1994). More recently, several randomized trials have demonstrated that concurrent chemoradiation resulted in superior survival rates compared with chemotherapy followed by radiation therapy (sequential chemoradiation) (Auperin et al., 2010; O’Rourke et al., 2010).

The radiation dose used with concurrent chemotherapy remains 60 Gy in 30 fractions. This dose of radiation results in better survival rates

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

than does a higher dose of radiation (74 Gy in 37 fractions) (Bradley et al., 2015, 2020).

Several chemotherapy regimens may be used for concurrent chemoradiation. A common strategy uses the histology of the lung cancer to choose the most appropriate regimen. For patients with non-squamous cell histologies, the preferred regimen is carboplatin or cisplatin plus pemetrexed. For patients with squamous cell histology, the preferred regimens are carboplatin plus paclitaxel or cisplatin plus etoposide (NCCN, 2020b). Each regimen has a standardized administration protocol, with a specified number of days and cycles.

Consolidation therapy

Despite the gradual improvements in survival rates that have been seen in patients with unresectable stage III NSCLC over the past few decades, it is still the case that more than 50% of these patients will ultimately develop distant metastases (Bradley et al., 2015). Consolidation therapy involves giving additional therapies upon completion of concurrent chemotherapy and radiation therapy to try to reduce the risk of distant metastases, and ultimately improve survival rates. Numerous studies have investigated consolidation chemotherapy (Bradley et al., 2015, 2020; Kelly et al., 2008; Tsujino et al., 2013). A landmark randomized study in 2017 demonstrated that stage III NSCLC patients treated with concurrent chemotherapy and radiation followed by 1 year of immunotherapy with durvalumab achieved a significant reduction in the risk of disease progression or death compared with those who did not receive durvalumab (Antonia et al., 2017). In addition, the overall survival improved with consolidation durvalumab, with two-thirds of the patients alive at 2 years and a median overall survival time that has not yet been reached (Gray et al., 2020).

Acute toxicities of chemoradiation

For most patients with stage III NSCLC, a common side effect of concurrent chemoradiation, in addition to fatigue and skin irritation, is radiation esophagitis resulting from radiation targeting lymph nodes located in close proximity to the esophagus. This irritation of the esophagus can manifest itself as heartburn, difficulty swallowing, or pain with swallowing. Radiation esophagitis is managed with oral medications and rinses, but if it is severe enough it can lead to severe dehydration and weight loss that may necessitate hospital admission, possibly with the placement of a feeding tube. The risk of developing radiation esophagitis and its severity are directly related to radiation dose-volume parameters including the mean dose to the esophagus, and factors such as the volume of the esophagus that receives 60 Gy or higher (Verma et al., 2017).

In addition to esophagitis, patients who receive chemoradiation are also at risk of decreased blood counts (white blood cells, red blood cells, and

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

platelets). Depending on the severity of the decrease in these blood counts, the chemotherapy may need to be dose-reduced or withheld at some point during treatment. Radiation therapy generally continues even in the setting of decreased blood counts unless the patient is hospitalized with fevers and low blood counts or the platelets or red blood cells get to dangerously low levels that require transfusions. Radiation dose-volume parameters to the vertebral bodies (spine bones) may be associated with the risk of developing decreased blood counts, and this is an area of active investigation with the goal of reducing the risk of patients developing hematologic toxicities (Verma et al., 2017).

Cough and shortness of breath are additional possible acute effects of chemoradiation. If these symptoms develop during treatment, they are usually indicative of an acute inflammatory reaction in the lung or airways and are managed conservatively with cough medications, mucolytics, and sometimes short-acting inhalers. Radiation pneumonitis refers to a severe inflammatory reaction in the lungs that can develop anywhere from 1 to 6 months after radiation. Radiation pneumonitis is considered a subacute reaction because it generally does not develop in the acute setting (during chemoradiation therapy) and it does not develop in the late setting (>6 months after the end of radiation). The incidence of radiation pneumonitis is strongly dependent on radiation dose-volume parameters. Symptomatic radiation pneumonitis is managed with high doses of corticosteroids with a prolonged taper. Some patients may need to be hospitalized for supplemental oxygen. Radiation pneumonitis is usually not fatal if it is detected early and treated appropriately (Verma et al., 2017).

Stage IV NSCLC

Until recently, all patients with metastatic stage IV NSCLC were treated with similar courses of systemic chemotherapy, but treatment has evolved to a more personalized or precision approach (see Chapter 4). In these approaches, tumor features that drive the malignant behavior of the cancer are targeted, when possible, so that each cancer is treated with the most effective drug for that tumor. The identification of the molecular and immunologic features of the cancer has become the most important element for treatment of late-stage NSCLC.

Targeted therapy

The molecular assay for NSCLC typically consists of a broad next-generation sequencing gene panel that evaluates genetic aberrations (e.g., DNA mutations and translocations) known to be drivers of lung cancer. Drugs that inhibit the abnormal protein products of these driver genes are used to treat the specific cancer with the driver gene.

FDA has approved a number of therapies that target molecular abnormalities in NSCLC. Most of these agents are oral, and most of the mutations where effective agents exist occur in patients with no smoking

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

history. These therapies have very different side effects than conventional chemotherapy. Currently, FDA-approved inhibitors for EGFR, ALK, ROS, BRAF, MET and RET are included in national guidelines and are standard of care for NSCLC (ACS, 2020; Bironzo and Di Maio, 2018; PDQ® Adult Treatment Editorial Board, 2020). EFGR, the most commonly identified actionable mutation in patients with metastatic NSCLC, occurs in approximately 13% of such patients (Eberhard et al., 2005). EGFR is more common in lung cancers in nonsmokers than in smokers (51% versus 10%), East Asians, women, and NSCLC adenocarcinomas (Shigematsu et al., 2005). ALK is the second most common actionable mutation occurring at a frequency of 5% with ROS, BRAF, MET and RET occurring at frequencies of less than 5% (Griffin and Ramirez, 2017). Many additional inhibitors of genetic drivers of cancer are under development.

Osimertinib is an EGFR inhibitor approved for EGFR-driven lung cancer. In a randomized clinical trial of EGFR-mutated NSCLC, comparing osimertinib with erlotinib (a EFGR-tyrosine kinase inhibitor [TKI]), the median progression-free survival was significantly longer with osimertinib than with standard EGFR-TKIs (18.9 months versus 10.2 months) (Soria et al., 2017). In addition, adverse events were less frequent with osimertinib than with standard EGFR-TKIs (34% versus 45%) and there were fewer neurologic events for osimertinib. Osimertinib has also been tested against conventional chemotherapy and has been shown to be superior (Mok et al., 2016; Soria et al., 2017). Molecular studies of resistance to EGFR inhibitors continue to be evaluated.

Immunotherapies

The concept of an immune panel is evolving. Presently, there are only two immune biomarkers approved for the treatment of NSCLC: programmed death (PD) ligand-1 (PD-L1) (PDQ® Adult Treatment Editorial Board, 2020) and the tumor mutational burden (FDA, 2020). The programed death pathway is a protective pathway of normal tissue for immune regulation that is commonly hijacked by tumor tissue. NSCLC and other tumors over-expressing PD-L1 are insensitive to T-cell killing, but that sensitivity can be restored by anti-PD therapy (see Chapter 8 for more information on immunotherapy). The tumor mutational burden measures the number of mutations per megabyte of DNA and is a measure of how susceptible a tumor is to recognition by the immune system.

When the protein PD-L1 is present on greater than 50% of lung cancer cells, immunotherapy with the programmed cell death-1 (PD-1) inhibitor pembrolizumab has been shown to be superior to combination chemotherapy with a cisplatin-based regimen (Reck et al., 2016). Follow-up data at 3 years post-treatment showed the overall survival rate was 43.7% among patients in the pembrolizumab arm versus 24.9% in the chemotherapy arm (ASCO Post Staff, 2019). It is estimated that 25% of patients who were treated with pembrolizumab may be alive at 5 years (Garon et al., 2019).

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

In addition, in patients with NSCLC whose cancer had progressed after chemotherapy, the PD-1 inhibitors, nivolumab and pembrolizumab, as well as the PD-L1 inhibitor, atezolizumab, all demonstrated improved survival compared with conventional chemotherapy. Approximately 15% were alive at 5 years (Garon et al., 2019; Gettinger et al., 2018).

In the absence of a targetable DNA biomarker or elevated PD-L1, randomized clinical studies have shown positive results from concurrent chemoimmunotherapy versus sequential chemotherapy followed by standard-of-care immunotherapy (Gandhi et al., 2018). Survival data showed an improvement in overall survival across all PD-L1 categories that were evaluated. Median progression-free survival was 9 months in the pembrolizumab-combination group and 4.9 months in the placebo-combination group (Gadgeel et al., 2020).

Acute toxicities related to PD-1- and PD-L1-based immunotherapies for lung cancer are less frequent than and different from those associated with conventional chemotherapy (e.g., nausea, hair loss, infections, neuropathy). PD-1 inhibitors inhibit an inhibitor of the immune system and, as such, provide a general, non-specific activation of the immune system. The goal is to stimulate immune activation against the cancer only, but this activation can also stimulate autoimmunity against normal organs. The organs most commonly affected by excessive immune stimulation are the skin (dermatitis), bowel (colitis), endocrine organs (hypophysitis, thyroiditis, diabetes), and lung (pneumonitis), among others. Such nonspecific immune stimulation is treated with immune suppression, most commonly with steroids. While the side effects may be life-threatening, they are generally reversible and easily managed in most cases. Endocrine organ dysfunction may be remedied by replacing the associated hormone deficiency (NCCN, 2020a).

Oligometastatic

NSCLC Oligometastatic lung cancer is a unique subtype of NSCLC in which the metastases are limited in number and organ sites. While there are no universally accepted-criteria for defining the oligometastatic state, a European Consensus definition indicates that it includes up to five metastases from three organ sites (Dingemans et al., 2019). Oligometastatic disease is an important category of metastatic lung cancer as it represents a clinical scenario in which metastasis-directed local therapies (radiation or surgery), in addition to systemic therapy, may play a role in potentially curing patients with otherwise incurable stage IV disease.

The question of whether oligometastatic disease is potentially curable is particularly important in NSCLC because the incidence of oligometastases may range from 26% to 55% (Parikh et al., 2014; Torok et al., 2017; Yano et al., 2013). Data from prospective randomized clinical trials of oligometastatic NSCLC support aggressive local regional therapy and ablation of metastases (Gomez et al., 2016, 2019; Iyengar et al., 2018; Palma et al., 2019).

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

Based on these studies, integrating local therapy with systemic therapies should be strongly considered for patients with oligometastatic NSCLC, with a multidisciplinary discussion to help guide the appropriate timing of local therapy (before or after first-line systemic therapy) and choice of local therapy (radiation, thermal ablation, or surgery) for each case.

Treatment of Small-Cell Lung Cancer

Unlike other common solid tumors, SCLC is very sensitive to chemotherapy and radiation therapy when it is initially diagnosed. However, resistance to the chemotherapy develops rapidly in more than 90% of patients, leading to disease recurrence. Second-line chemotherapies are relatively ineffective, so the mortality rate for this disease is high (Farago and Keane, 2018). The prognosis of SCLC is poor, with survival measured in months (Poirier et al., 2020); the median overall survival of patients with LS-SCLC is 15–20 months and 8–13 months for those diagnosed with ES-SCLC (Zhong et al., 2020).

Because SCLC is considered a systemic disease, chemotherapy is a cornerstone of its treatment even in cases where the disease is limited to a single lesion in the lungs. In addition, surgery is typically not used in SCLC. While surgical resection followed by adjuvant chemotherapy is a reasonable option for early-stage disease (Zhong et al., 2020), most surgical cases of SCLC occur because of an initial misdiagnosis as NSCLC.

Treatment of LS-SCLC, which has not changed over the past four decades, consists of a standard first-line chemotherapy regimen with concurrent radiation. The chemotherapy typically consists of a platinum-based agent and etoposide (NCCN, 2020b). The treatment of ES-SCLC uses the same systemic chemotherapy without concurrent radiation; again, the chemotherapy would include a platinum agent and etoposide or irinotecan (NCCN, 2020a). For the first time in over a decade, the survival of patients with ES-SCLC recently showed improvement, with survival increasing by a median of 2 months when standard chemotherapy was combined with the PD-L1 inhibitors atezolizumab (Horn et al., 2018) or durvalumab (Paz-Ares et al., 2019).

Approximately 20% of SCLC patients are diagnosed with brain metastases at the initial diagnosis (Goncalves et al., 2016). Importantly, many of these patients have no neurologic symptoms indicative of brain metastases (Hochstenbag et al., 2000). The rate of brain metastases increases significantly among patients who survive for more than 1 year after treatment (Schouten et al., 2002). To counteract this predilection of SCLC to metastasize to the brain, prophylactic cranial irradiation in the absence of detectable brain metastases, is a component of therapy for LS-SCLC patients who achieve a complete or near-complete response to systemic

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

chemotherapy and thoracic radiation. This treatment reduces the risk of brain metastases and improves survival (Rudin et al., 2015). However, this approach is currently being questioned because of late cognitive effects of prophylactic cranial irradiation and because of the option of effective MRI surveillance and stereotactic radiation therapy, which can effectively manage brain metastases after they are diagnosed (Lukas et al., 2017).

When SCLC recurs, treatment is relatively ineffective. A recurrence of ES-SCLC after first-line chemotherapy is categorized as refractory (i.e., occurs within 60–90 days after first-line chemotherapy ends) or sensitive (i.e., occurs at least 60–90 days after end of first-line chemotherapy). Topotecan, a topoisomerase 1 inhibitor and the only FDA-approved agent for recurrent SCLC, is associated with a 1-year overall survival of 9% for refractory recurrent SCLC and 27% for sensitive recurrent SCLC (Horita et al., 2015). Topotecan resulted in a mean survival rate of 33–35 weeks and a 1-year survival rate of 29–33% in patients with either ES- or LS-SCLC, although the type of recurrence was not specified (Eckardt et al., 2007). Tumor shrinkage following third-line chemotherapy in metastatic SCLC is uncommon; however, two PD-1 inhibitors, nivolumab and pembrolizumab, have been approved for this indication (Chung et al., 2020; Ready et al., 2020).

Acute treatment-related impairments are those associated with chemotherapy and radiation (see Acute Toxicities of Chemoradiation above) because most patients with LS-SCLC receive concurrent chemoradiation and some patients with ES-SCLC receive consolidative chest radiation after the completion of chemotherapy (Slotman et al., 2015). Similarly, late-onset and long-term impairments are associated with chemotherapy (peripheral and sensory neuropathy) and radiation therapy (pulmonary and cardiac complications) as well as with cognitive impairment stemming from prophylactic cranial radiation. These impairments are discussed in the section on Life After Lung Cancer Diagnosis.

SUMMARY OF LUNG CANCER DIAGNOSTIC EVALUATION AND TREATMENT

Table 6-3 summarizes the information presented in the preceding sections on the diagnosis and treatment of stage I–II, stage III, and stage IV NSCLC and LS- and ES-SCLC.

LIFE AFTER LUNG CANCER DIAGNOSIS

A lung cancer survivor’s health status and quality of life (QOL) are often degraded not only by the cancer and its treatment, but also by various comorbid medical conditions, including COPD and emphysema as well

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

TABLE 6-3 Summary of Lung Cancer Diagnosis and Treatment

Stage Diagnostic Evaluation Treatment
Non-small-cell lung cancer
Stage I or II (localized) CT scan; PET scan; MRI scan; endobronchial ultrasound; mediastinoscopy; percutaneous lung biopsy

Resectable tumors:

  • Surgery: open, video- or robot-assisted lobectomy; or pneumonectomy
  • Adjuvant chemotherapy if risk of relapse is >25% Unresectable tumors due to limited organ function:
  • Stereotactic radiotherapy
  • Radiofrequency ablation
Stage III (regional spread) CT scan; PET scan; MRI scan; endobronchial ultrasound; mediastinoscopy; percutaneous lung biopsy

Resectable tumors:

  • Surgery ± radiation ± neoadjuvant chemotherapy Unresectable tumors:
  • Chemotherapy + radiation + immunotherapy
Stage IV (metastatic) CT scan; PET scan; MRI scan; needle biopsy; molecular and DNA biomarkers
  • Chemotherapy ± immunotherapy ± targeted therapy
  • Radiation for palliation
Small-cell lung cancer
Limited stage CT scan; MRI scan; bone scintigraphy; FDG-PET scan
  • Chemotherapy with radiation ± prophylactic cranial irradiation
Extensive stage CT scan; MRI scan; bone scintigraphy; FDG-PET scan
  • Chemotherapy ± immunotherapy ± radiation

NOTES: The symbol ± in the treatment column means “with or without.” The exact treatment regimen is determined on a case-by-case basis. CT = computed tomography; DNA = deoxyribonucleic acid; FDG-PET = fluorodeoxyglucose positron emission tomography; MRI = magnetic resonance imaging; PET = positron emission tomography.

as recurrent or new cancers. Therefore, in addition to an accurate diagnosis and appropriate treatment regimen, supporting the optimal health and functioning of lung cancer survivors involves care for the adverse sequelae of the primary lung cancer and its treatment along with health promotion and disease-prevention activities, such as smoking cessation, and monitoring for evidence of recurrence and new primary cancers (Vijayvergia et al., 2015). For example, the American Society of Clinical Oncology currently recommends that patients with stage I–III NSCLC and SCLC who have been curatively treated and who have no symptoms of recurrent disease should undergo imaging for recurrence every 6 months for 2 years and then once per year in order to detect any new primary lung cancers (Schneider et al., 2020). See Chapter 10 for more information about cancer survivorship care.

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

Often persistent and overlapping, cancer-related sequelae impair a lung cancer survivor’s ability to maintain good health and function. Some of these sequelae may resolve within 6 months (acute effects), others may become chronic (long-term effects), and still others may develop in the months to years after treatment ends (late-onset effects) (Vijayvergia et al., 2015). The sections below described the range of late-onset and long-term physical, psychological, and psychosocial impairments that may adversely affect a lung cancer survivor’s overall QOL and increase the risk for functional limitations. See Chapter 9 for more on functional limitations.

Physical Effects

In general, the physical effects caused by lung cancer and its treatments include respiratory problems, fatigue, pain, reduced appetite, neuropathy, and ototoxicity (e.g., inner ear problems from drug toxicity resulting in hearing loss or tinnitus) (Ha et al., 2020; Hung et al., 2011; Yang et al., 2012). These physical impairments tend to become chronic. For example, a 7-year longitudinal study of 477 lung cancer survivors (mostly of NSCLC) showed persistent physical symptoms that did not lessen over time and that diminished the survivors’ QOL (Yang et al., 2012). Among survivors reporting declines in QOL (n = 155), significant symptoms were fatigue (69%), pain (59%), dyspnea (58%), depressed appetite (49%), and coughing (42%).

Fatigue is the most commonly reported symptom among survivors of lung cancer, affecting up to 90% of them and it may persist for months to years after treatment (Hung et al., 2011). There is an association between moderate to severe fatigue and impairment in daily functioning. Among NSCLC survivors with moderate to severe fatigue, one in four (23.7%) was functionally impaired, that is, their ability to work or to independently care for their personal needs outside the home was limited (Hung et al., 2011). Contributing factors to fatigue include comorbid health conditions, pain, depression, anxiety, sleep disturbances, and changes in breathing capacity (Hung et al., 2011; Sugimura and Yang, 2006). Studies suggest that fatigue can resolve in 3 to 6 months in some cases, such as after lung cancer surgery (Brunelli et al., 2007; Win et al., 2005), or it can persist for months to years after treatment completion (Kenny et al., 2008; Sarna et al., 2008).

Between 80% and 100% of all cancer survivors experience pain (Rausch et al., 2012), and pain significantly diminishes their QOL (Sugimura and Yang, 2006). Chronic pain was reported by 25% of long-term NSCLC survivors followed at a survivorship clinic (Huang et al., 2014). Many NSCLC survivors who undergo surgery may experience post-thoracotomy pain—a pain syndrome involving burning, dysesthesia, and aching along the

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

thoracotomy incision—that can persist for a prolonged period (>2 months) after surgery (Rogers and Duffy, 2000). Thirty percent of patients report continued pain up to 5 years after surgery (Dajczman et al., 1991). Pain may also be due to chemotherapy-induced peripheral neuropathy, as described below (and in Chapter 9).

In one study, nearly two-thirds of lung cancer patients experienced dyspnea, cough, wheezing, phlegm, pain, and decreased functionality (Sarna et al., 2004). Physical, cognitive, and emotional functions may be adversely affected by dyspnea (Sugimura and Yang, 2006). Persistent cough can interfere with speech, eating, and sleeping and thus significantly affect psychological, social, and physical QOL (Harle et al., 2012).

Psychological and Psychosocial Effects

Lung cancer and its treatment put survivors at risk for various effects that affect their psychosocial well-being, including psychological health (e.g., anxiety, depression, fear of recurrence), interpersonal connections (e.g., relationships, role in family), financial security (e.g., loss of employment or insurance, cost of cancer care), and behavioral health (e.g., capacity for a healthy lifestyle).

At least 40% of NSCLC survivors report psychological distress, which is significantly higher than the rates seen in survivors of other cancers (Chapple et al., 2004; Zabora et al., 2001). Even mild symptoms of emotional and psychological distress can affect daily functioning and QOL and are associated with a decline in both cognitive and social functioning (Fox and Lyon, 2006; Shi et al., 2011).

Psychological distress in long-term cancer survivors has been associated with non-adherence to cancer surveillance screening recommendations (Katz et al., 2009) and a lack of engagement in healthful behaviors, such as exercise (Courneya et al., 2008) and smoking cessation (Schnoll et al., 2010). Patients with lung cancer who continue to smoke in the perioperative period have higher postoperative pulmonary morbidity and mortality (Vaporciyan et al., 2002). In long-term NSCLC survivors, greater motivational readiness for physical activity was associated with improved QOL (Clark et al., 2008), although nearly 66% of survivors did not meet national physical activity guidelines during the post-treatment period (Carmack et al., 2011; Coups et al., 2009).

Lung cancer significantly increases the risk of departure from the workforce compared with other cancers such as colon cancer (odds ratio 2.8 for stage II disease and 6.1 for stage III disease) (Earle et al., 2010). One study of 5-year cancer survivors showed that lung cancer survivors had the greatest decline in employment rates of all cancer survivors (Torp et al., 2013). Loss of employment after a lung cancer diagnosis can eliminate

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

health insurance benefits, leading to the survivor missing follow-up care and recommended treatments. One study found that among patients with lung cancer, 40% had limited financial reserves (<2 months) and that there was a strong association between greater financial strain and poor physical and mental well-being (Lathan et al., 2016).

Effects by Treatment Modality

This section summarizes specific long-term and late-onset effects related to lung cancer and its treatment with surgery, chemotherapy, or radiation therapy or a combination of them. The long-term effects of immunotherapy are not known despite the fact that it has become a mainstay of lung cancer treatment (Vijayvergia et al., 2015).

Surgery

Lung cancer patients undergoing lobectomy show improved QOL scores (except for pain) within 3 months postoperatively, indicating good recovery. After pneumonectomy, physical functioning, pain, shoulder function, and dyspnea had not returned to baseline even 12 months after surgery (Balduyck et al., 2007). After surgical resection of NSCLC, QOL indicators for physical functioning, pain, and dyspnea are significantly impaired even at 24 months. QOL—as determined by physical function, social function, role function, global health, and pain—was better after lobectomy than after pneumonectomy (Bezjak et al., 2008; Schulte et al., 2009). QOL may improve sooner in lung cancer patients who undergo VATS procedures than in those who undergo open thoracotomy (Aoki et al., 2007). Evaluation of pain and musculoskeletal function after surgery, with referral to and treatment from appropriate rehabilitation specialists, can improve function.

Chemotherapy

The long-term toxicities of adjuvant cisplatin-based chemotherapy include sensory neuropathy and hearing loss, which have been estimated to occur in approximately 48% and 21% of patients, respectively (Winton et al., 2005). These toxicities may persist for up to 30 months post-treatment (Bezjak et al., 2008) and can affect overall QOL during this time (Aziz, 2002). Adjuvant therapy increases the chance of survival but leads to a decline in overall QOL, although QOL tends to improve once the treatment is completed. For example, a retrospective study of NSCLC patients who underwent surgery found that adjuvant therapy improved long-term QOL at a median follow-up of 4.8 years (Ilonen et al., 2013). Chapter 9

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

provides additional information about chemotherapy-related toxicities and management.

Radiation Therapy

Scarring in the lung (lung fibrosis) can develop in an area that received radiation years after the treatment is complete. The treatment for radiation-induced lung fibrosis is often supportive, with supplemental oxygen and pulmonary medications. In lung cancer survivors who receive adjuvant radiation, advanced age and higher radiotherapy doses predict an increased risk of death from intercurrent diseases (i.e., a disease occurring at the same time as another unrelated disease) (Machtay et al., 2001). Additional long-term or late-onset toxicities of chemoradiation therapy include esophageal strictures, pulmonary toxicity, and cardiac toxicity. Recent studies on cardiotoxicity from chemoradiation for patients with stage III NSCLC suggest that major cardiac events are occurring within 1–2 years of treatment and that increased radiation doses to the heart are associated with a significantly increased risk of mortality (Atkins et al., 2019; Ming et al., 2016). More research on the cardiotoxicity of chemoradiation for stage III NSCLC is needed. Other acute effects of chemoradiation such as radiation pneumonitis were discussed earlier in the chapter. These effects are further described in Chapter 9.

SBRT can also damage normal body tissues, and the risk of damage depends on whether the tumor is located in the periphery of the lung (near the ribs or chest wall) or in the central portion of the lung (near the major airways, heart, or esophagus). SBRT to peripheral tumors can cause chest wall pain resulting from rib fractures or from inflammation of the chest wall muscles; the incidence of pain is about 8% (Coroller et al., 2014). This pain is managed conservatively with oral medications and generally resolves over time. SBRT for the treatment of central tumors is potentially life-threatening and has the risk of long-term complications that can significantly impair physical functioning, including lung collapse and resulting pneumonias; damage to blood vessels; tracheo-esophageal fistulas; fatal radiation pneumonitis; and narrowing or perforation of the esophagus. Overall, the incidence of fatal adverse effects attributable to SBRT for central tumors has been reported to be 3.7% (Haseltine et al., 2016).

In the case of SCLC, treatment may include radiation to the patient’s whole brain to prevent metastases from lung cancer. This prophylactic cranial irradiation is associated with cognitive impairment; an oral medication, memantine, has been shown to modestly improve neurocognitive function in patients receiving whole brain radiation therapy (Brown et al., 2013). Early studies show that avoiding the hippocampus when using whole brain radiation therapy plus memantine results in modest improvements

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

in neurocognitive function compared to standard whole brain radiation therapy plus memantine (Brown et al., 2020). Currently, there is an ongoing randomized clinical trial of hippocampal avoidance prophylactic cranial irradiation versus standard prophylactic cranial irradiation for patients with SCLC (The Netherlands Cancer Institute and Dutch Cancer Society, 2013). Cranial radiation may also affect the neuro-endocrine system in the brain, and if long-term survival is achieved, there is a risk of visual, hearing, or dental complications and new benign or malignant tumors (Chowdhary et al., 2012; Rahman et al., 2020).

FINDINGS AND CONCLUSIONS

Findings

  1. Lung cancer is broadly classified into NSCLC and SCLC. NSCLC is more prevalent, but SCLC has a poorer prognosis.
  2. Smoking and age are major risk factors for lung cancer. Other risk factors include exposure to radon gas, second-hand exposure to smoking, asbestos, radiation, air pollution, and some metals.
  3. Lung cancer death rates have declined due to reductions in smoking and, particularly for NSCLC, from recent advances in genetics and biomarkers testing.
  4. Standard diagnostic procedures for lung cancer include CT scans, MRIs, PET scans, and endobronchial ultrasound. In certain cases, additional diagnostic profiling may be necessary using a FDG-PET scan, mediastinoscopy, percutaneous lung biopsy, immunohistochemistry, or gene profiling.
  5. Most lung cancers are diagnosed at an advanced stage (stage IV NSCLC or extensive-stage SCLC), with metastatic spread to other organ systems in the body.
  6. Treatment strategies are determined on the basis of tumor histology, tumor location/size, stage, molecular characteristics, as well as the patient’s general health and comorbidities.
  7. Surgical resection (lobectomy) is the standard of care for patients diagnosed with NSCLC stage I and II who are medically fit for surgery. In certain cases of stage I or stage II NSCLC, the addition of neoadjuvant chemotherapy has survival benefits. The potential for long-term benefits with the addition of neoadjuvant immunotherapy is still being investigated for this group of patients.
  8. Stage III NSCLC is treated with multiple modalities depending on tumor characteristics (e.g., the possibility of surgical resection) and patient factors (e.g., other medical conditions). Possible treatments
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
  1. include chemotherapy, radiation, immunotherapy, and targeted therapy.
  2. Stage IV NSCLC requires systemic therapies, with local therapies (radiation, surgery) generally used to improve symptoms; however, when metastases are limited in number and organ (oligometastatic), local therapies may play a role in improving outcomes.
  3. Treatment of SCLC includes the use of chemotherapy, radiation, and prophylactic cranial radiation. SCLC develops resistance to chemotherapy in the vast majority of patients, leading to recurrence, which is usually fatal.
  4. The diagnosis and treatment of lung cancer results in numerous acute as well as late-onset and long-term effects. The pulmonary and cardiovascular systems are most often affected.
  5. Adverse effect profiles coincide with treatment regimens and their known adverse effects (e.g., cisplatin—neuropathy, ototoxicity). These adverse effects may have long-lasting impacts on the survivors’ physical, psychological, psychosocial, and behavioral well-being; reduce their quality of life; and affect their ability to work.
  6. Common impairments reported by lung cancer survivors that can interfere with their quality of life include respiratory problems, fatigue, pain, and neuropathy. Pain is the most common cause of disability and often is associated with depression, anxiety, and sleep disturbances.
  7. Psychosocial effects of lung cancer can result in decrements to psychological health (anxiety, depression, fear of recurrence), interpersonal relationships (connectedness), financial security (job insecurity, disability), and behavioral health (capacity for a healthy life style). These effects may have a significant impact on work ability, return to work, and productivity.

Conclusions

  1. Reductions in smoking and improvements in screening, diagnosis, and treatment options have contributed to lower lung cancer death rates; however, survivors experience numerous adverse effects and impairments that can result in functional limitations.
  2. As a result of recent advances in immunotherapy and targeted therapy, chemotherapy is no longer the most effective systemic treatment for many cases of lung cancer.
  3. Changes in the treatment of NSCLC in recent years have produced improvements in survival. Some patients with metastatic NSCLC are now achieving 5-year survival; however, this longevity is still extremely rare for SCLC.
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
  1. Survivors of lung cancer may have cancer-related impairments as a consequence of their cancer and its treatment. The high risk of lung cancer recurrence, metastases, or the development of new primary cancers, and the major comorbid conditions often associated with smoking and aging, such as chronic obstructive pulmonary disease, can increase the number and severity of any impairments.

REFERENCES

ACS (American Cancer Society). 2019. What is lung cancer? https://www.cancer.org/cancer/lung-cancer/about/what-is.html (accessed November 13, 2020).

ACS. 2020. Targeted drug therapy for non-small cell lung cancer. https://www.cancer.org/cancer/lung-cancer/treating-non-small-cell/targeted-therapies.html (accessed November 12, 2020).

AJCC (American Joint Committee on Cancer). 2017. AJCC cancer staging manual, eighth edition. New York: Springer International Publishing.

ALA (American Lung Association). 2020. State of lung cancer 2020: Lung cancer key findings. https://www.lung.org/research/state-of-lung-cancer/key-findings (accessed October 12, 2020).

Albain, K.S., R.S. Swann, V.W. Rusch, A.T. Turrisi, 3rd, F.A. Shepherd, C. Smith, Y. Chen, R.B. Livingston, R.H. Feins, D.R. Gandara, W.A. Fry, G. Darling, D.H. Johnson, M.R. Green, R.C. Miller, J. Ley, W.T. Sause, and J.D. Cox. 2009. Radiotherapy plus chemotherapy with or without surgical resection for stage III non-small-cell lung cancer: A phase III randomised controlled trial. The Lancet 374(9687):379–386.

Ambrogi, M.C., O. Fanucchi, R. Cioni, P. Dini, A. De Liperi, C. Cappelli, F. Davini, C. Bartolozzi, and A. Mussi. 2011. Long-term results of radiofrequency ablation treatment of stage I non-small cell lung cancer: A prospective intention-to-treat study. Journal of Thoracic Oncology 6(12):2044–2051.

Amin, M.B., F.L. Greene, S.B. Edge, C.C. Compton, J.E. Gershenwald, R.K. Brookland, L. Meyer, D.M. Gress, D.R. Byrd, and D.P. Winchester. 2017. The Eighth Edition AJCC Cancer Staging Manual: Continuing to build a bridge from a population-based to a more “personalized” approach to cancer staging. CA: A Cancer Journal for Clinicians 67(2):93–99.

Antonia, S.J., A. Villegas, D. Daniel, D. Vicente, S. Murakami, R. Hui, T. Yokoi, A. Chiappori, K.H. Lee, M. de Wit, B.C. Cho, M. Bourhaba, X. Quantin, T. Tokito, T. Mekhail, D. Planchard, Y.C. Kim, C.S. Karapetis, S. Hiret, G. Ostoros, K. Kubota, J.E. Gray, L. Paz-Ares, J. de Castro Carpeno, C. Wadsworth, G. Melillo, H. Jiang, Y. Huang, P.A. Dennis, M. Ozguroglu, for the PACIFIC Investigators. 2017. Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. New England Journal of Medicine 377(20):1919–1929.

Aoki, T., M. Tsuchida, T. Hashimoto, M. Saito, T. Koike, and J.-I. Hayashi. 2007. Quality of life after lung cancer surgery: Video-assisted thoracic surgery versus thoracotomy. Heart, Lung and Circulation 16(4):285–289.

The ASCO Post staff. 2019. WCLC 2019: Keynote-024 survival update shows benefit with pembrolizumab vs chemotherapy in advanced NSCLS. https://ascopost.com/news/september-2019/keynote-024-survival-update (accessed November 11, 2020).

Atkins, K.M., B. Rawal, T.L. Chaunzwa, N. Lamba, D.S. Bitterman, C.L. Williams, D.E. Kozono, E.H. Baldini, A.B. Chen, P.L. Nguyen, A.V. D’Amico, A. Nohria, U. Hoffmann, H.J.W.L. Aerts, and R.H. Mak. 2019. Cardiac radiation dose, cardiac disease, and mortality in patients with lung cancer. Journal of the American College of Cardiology 73(23):2976–2987.

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

Auperin, A., C. Le Pechoux, E. Rolland, W.J. Curran, K. Furuse, P. Fournel, J. Belderbos, G. Clamon, H.C. Ulutin, R. Paulus, T. Yamanaka, M.C. Bozonnat, A. Uitterhoeve, X. Wang, L. Stewart, R. Arriagada, S. Burdett, and J.P. Pignon. 2010. Meta-analysis of concomitant versus sequential radiochemotherapy in locally advanced non-small-cell lung cancer. Journal of Clinical Oncology 28(13):2181–2190.

Aziz, N.M. 2002. Cancer survivorship research: Challenge and opportunity. Journal of Nutrition 132(11 Suppl):3494s–3503s.

Balduyck, B., J. Hendriks, P. Lauwers, and P. Van Schil. 2007. Quality of life evolution after lung cancer surgery: A prospective study in 100 patients. Lung Cancer 56(3):423–431.

Bezjak, A., C.W. Lee, K. Ding, M. Brundage, T. Winton, B. Graham, M. Whitehead, D.H. Johnson, R.B. Livingston, and L. Seymour. 2008. Quality-of-life outcomes for adjuvant chemotherapy in early-stage non-small-cell lung cancer: Results from a randomized trial, JBR.10. Journal of Clinical Oncology 26(31):5052–5059.

Bironzo, P., and M. Di Maio. 2018. A review of guidelines for lung cancer. Journal of Thoracic Diseases 10(Suppl 13):S1556–S1563.

Boffa, D.J., A.S. Kosinski, A.P. Furnary, S. Kim, M.W. Onaitis, B.C. Tong, P.A. Cowper, J.R. Hoag, J.P. Jacobs, C.D. Wright, J.B. Putnam, Jr., and F.G. Fernandez. 2018. Minimally invasive lung cancer surgery performed by thoracic surgeons as effective as thoracotomy. Journal of Clinical Oncology 36(23):2378–2385.

Bradley, J.D., R. Paulus, R. Komaki, G. Masters, G. Blumenschein, S. Schild, J. Bogart, C. Hu, K. Forster, A. Magliocco, V. Kavadi, Y.I. Garces, S. Narayan, P. Iyengar, C. Robinson, R.B. Wynn, C. Koprowski, J. Meng, J. Beitler, R. Gaur, W. Curran, Jr., and H. Choy. 2015. Standard-dose versus high-dose conformal radiotherapy with concurrent and consolidation carboplatin plus paclitaxel with or without cetuximab for patients with stage IIIa or IIIb non-small-cell lung cancer (RTOG 0617): A randomised, two-by-two factorial phase 3 study. The Lancet Oncology 16(2):187–199.

Bradley, J.D., C. Hu, R.R. Komaki, G.A. Masters, G.R. Blumenschein, S.E. Schild, J.A. Bogart, K.M. Forster, A.M. Magliocco, V.S. Kavadi, S. Narayan, P. Iyengar, C.G. Robinson, R.B. Wynn, C.D. Koprowski, M.R. Olson, J. Meng, R. Paulus, W.J. Curran, Jr., and H. Choy. 2020. Long-term results of NRG oncology RTOG 0617: Standard- versus high-dose chemoradiotherapy with or without cetuximab for unresectable stage III non-small-cell lung cancer. Journal of Clinical Oncology 38(7):706–714.

Brandt, W.S., I. Bouabdallah, K.S. Tan, B.J. Park, P.S. Adusumilli, D. Molena, M.S. Bains, J. Huang, J.M. Isbell, M.J. Bott, and D.R. Jones. 2018. Factors associated with distant recurrence following R0 lobectomy for pN0 lung adenocarcinoma. Journal of Thoracic and Cardiovascular Surgery 155(3):1212–1224.

Brandt, W.S., W. Yan, J. Zhou, K.S. Tan, J. Montecalvo, B.J. Park, P.S. Adusumilli, J. Huang, M.J. Bott, V.W. Rusch, D. Molena, W.D. Travis, M.G. Kris, J.E. Chaft, and D.R. Jones. 2019. Outcomes after neoadjuvant or adjuvant chemotherapy for cT2-4N0-1 non-small cell lung cancer: A propensity-matched analysis. Journal of Thoracic and Cardiovascular Surgery 157(2):743–753.

Brown, P.D., S. Pugh, N.N. Laack, J.S. Wefel, D. Khuntia, C. Meyers, A. Choucair, S. Fox, J.H. Suh, D. Roberge, V. Kavadi, S.M. Bentzen, M.P. Mehta, and D. Watkins-Bruner. 2013. Memantine for the prevention of cognitive dysfunction in patients receiving whole-brain radiotherapy: A randomized, double-blind, placebo-controlled trial. Neuro-Oncology 15(10):1429–1437.

Brown, P.D., V. Gondi, S. Pugh, W.A. Tome, J.S. Wefel, T.S. Armstrong, J.A. Bovi, C. Robinson, A. Konski, D. Khuntia, D. Grosshans, T.L.S. Benzinger, D. Bruner, M.R. Gilbert, D. Roberge, V. Kundapur, K. Devisetty, S. Shah, K. Usuki, B.M. Anderson, B. Stea, H. Yoon, J. Li, N.N. Laack, T.J. Kruser, S.J. Chmura, W. Shi, S. Deshmukh, M.P. Mehta, and L.A. Kachnic. 2020. Hippocampal avoidance during whole-brain radiotherapy plus memantine for patients with brain metastases: Phase III trial NRG oncology CC001. Journal of Clinical Oncology 38(10):1019–1029.

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

Brunelli, A., L. Socci, M. Refai, M. Salati, F. Xiumé, and A. Sabbatini. 2007. Quality of life before and after major lung resection for lung cancer: A prospective follow-up analysis. Annals of Thoracic Surgery 84(2):410–416.

Burdett, S., J.P. Pignon, J. Tierney, H. Tribodet, L. Stewart, C. Le Pechoux, A. Aupérin, T. Le Chevalier, R.J. Stephens, R. Arriagada, J.P.T. Higgins, D.H. Johnson, J. Van Meerbeeck, M.K.B. Parmar, R.L. Souhami, B. Bergman, J.Y. Douillard, A. Dunant, C. Endo, D. Girling, H. Kato, S.M. Keller, H. Kimura, A. Knuuttila, K. Kodama, R. Komaki, M.G. Kris, T. Lad, T. Mineo, S. Piantadosi, R. Rosell, G. Scagliotti, L.K. Seymour, F.A. Shepherd, R. Sylvester, H. Tada, F. Tanaka, V. Torri, D. Waller, Y. Liang, for the Non-Small Cell Lung Cancer Collaborative Group. 2015. Adjuvant chemotherapy for resected early-stage non-small cell lung cancer. Cochrane Database of Systematic Reviews 2015(3):CD011430.

Calikusu, Z., and P. Altinok. 2018. Treatment of locally advanced, unresectable or medically inoperable stage III non-small-cell lung cancer; the past, present and future of chemoradiotherapy. Journal of Oncological Sciences 4(1):49–52.

Cannon-Albright, L.A., S.R. Carr, and W. Akerley. 2019. Population-based relative risks for lung cancer based on complete family history of lung cancer. Journal of Thoracic Oncology 14(7):1184–1191.

Carmack, C.L., K. Basen-Engquist, and E.R. Gritz. 2011. Survivors at higher risk for adverse late outcomes due to psychosocial and behavioral risk factors. Cancer Epidemiology, Biomarkers & Prevention 20(10):2068–2077.

Cascone, T., W.N. William, A. Weissferdt, H.Y. Lin, C.H. Leung, B.W. Carter, F.V. Fossella, F. Mott, V. Papadimitrakopoulou, G.R. Blumenschein, X.N. Le, L. Federico, E.R.P. Cuentas, C. Bernatchez, I.I. Wistuba, A.A. Vaporciyan, D.L. Gibbons, S. Swisher, J. Heymach, and B. Sepesi. 2019. Neoadjuvant nivolumab (N) or nivolumab plus ipilimumab (NI) for resectable non-small cell lung cancer (NSCLC): Clinical and correlative results from the NEOSTAR study. Journal of Clinical Oncology 37(15):8504.

Chang, J.Y., S. Senan, M.A. Paul, R.J. Mehran, A.V. Louie, P. Balter, H.J. Groen, S.E. McRae, J. Widder, L. Feng, B.E. van den Borne, M.F. Munsell, C. Hurkmans, D.A. Berry, E. van Werkhoven, J.J. Kresl, A.M. Dingemans, O. Dawood, C.J. Haasbeek, L.S. Carpenter, K. De Jaeger, R. Komaki, B.J. Slotman, E.F. Smit, and J.A. Roth. 2015. Stereotactic ablative radiotherapy versus lobectomy for operable stage I non-small-cell lung cancer: A pooled analysis of two randomised trials. The Lancet Oncology 16(6):630–637.

Chapple, A., S. Ziebland, and A. McPherson. 2004. Stigma, shame, and blame experienced by patients with lung cancer: Qualitative study. BMJ 328(7454):1470.

Chowdhary, A., A.M. Spence, L. Sales, R.C. Rostomily, J.K. Rockhill, and D.L. Silbergeld. 2012. Radiation associated tumors following therapeutic cranial radiation. Surgical Neurology International 3:48.

Chung, H.C., S.A. Piha-Paul, J. Lopez-Martin, J.H.M. Schellens, S. Kao, W.H. Miller, Jr., J.P. Delord, B. Gao, D. Planchard, M. Gottfried, A. Zer, S.I. Jalal, N. Penel, J.M. Mehnert, I. Matos, J. Bennouna, D.W. Kim, L. Xu, S. Krishnan, K. Norwood, and P.A. Ott. 2020. Pembrolizumab after two or more lines of previous therapy in patients with recurrent or metastatic SCLC: Results from the Keynote-028 and Keynote-158 studies. Journal of Thoracic Oncology 15(4):618–627.

CISNET (Cancer Intervention and Surveillance Modeling Network). 2020. Evaluation of the benefits and harms of lung cancer screening with low-dose computed tomography: A collaborative modeling study for the U.S. Preventive Services Task Force. AHRQ Publication No. 20-05266-EF-2. Rockville, MD: Agency for Healthcare Research and Quality.

Clark, M.M., P.J. Novotny, C.A. Patten, S.M. Rausch, Y.I. Garces, A. Jatoi, J.A. Sloan, and P. Yang. 2008. Motivational readiness for physical activity and quality of life in long-term lung cancer survivors. Lung Cancer 61(1):117–122.

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

Coroller, T.P., R.H. Mak, J.H. Lewis, E.H. Baldini, A.B. Chen, and Y.L. Colson. 2014. Low incidence of chest wall pain with a risk-adapted lung stereotactic body radiation therapy approach using three or five fractions based on chest wall dosimetry. PLOS ONE 9(4):e94859.

Coups, E.J., B.J. Park, M.B. Feinstein, R.M. Steingart, B.L. Egleston, D.J. Wilson, and J.S. Ostroff. 2009. Physical activity among lung cancer survivors: Changes across the cancer trajectory and associations with quality of life. Cancer Epidemiology, Biomarkers & Prevention 18(2):664–672.

Courneya, K.S., R.J. Segal, K. Gelmon, R.D. Reid, J.R. Mackey, C.M. Friedenreich, C. Proulx, K. Lane, A.B. Ladha, and J.K. Vallance. 2008. Predictors of supervised exercise adherence during breast cancer chemotherapy. Medicine & Science in Sports & Exercise 40(6):1180–1187.

Dajczman, E., A. Gordon, H. Kreisman, and N. Wolkove. 1991. Long-term postthoracotomy pain. Chest 99(2):270–274.

de Koning, H.J., C.M. van der Aalst, P.A. de Jong, E.T. Scholten, K. Nackaerts, M.A. Heuvelmans, J.-W.J. Lammers, C. Weenink, U. Yousaf-Khan, N. Horeweg, S. van ’t Westeinde, M. Prokop, W.P. Mali, F.A.A. Mohamed Hoesein, P.M.A. van Ooijen, J.G.J.V. Aerts, M.A. den Bakker, E. Thunnissen, J. Verschakelen, R. Vliegenthart, J.E. Walter, K. ten Haaf, H.J.M. Groen, and M. Oudkerk. 2020. Reduced lung-cancer mortality with volume CT screening in a randomized trial. New England Journal of Medicine 382(6):503–513.

Dingemans, A.C., L.E.L. Hendriks, T. Berghmans, A. Levy, B. Hasan, C. Faivre-Finn, M. Giaj-Levra, N. Giaj-Levra, N. Girard, L. Greillier, S. Lantuejoul, J. Edwards, M. O’Brien, M. Reck, E.F. Smit, P. Van Schil, P.E. Postmus, S. Ramella, Y. Lievens, M. Gaga, N. Peled, G.V. Scagliotti, S. Senan, L. Paz-Ares, M. Guckenberger, F. McDonald, S. Ekman, T. Cufer, H. Gietema, M. Infante, R. Dziadziuszko, S. Peters, R.R. Porta, J. Vansteenkiste, C. Dooms, D. de Ruysscher, B. Besse, and S. Novello. 2019. Definition of synchronous oligometastatic non-small cell lung cancer—A consensus report. Journal of Thoracic Oncology 14(12):2109–2119.

Dupuy, D.E., T. DiPetrillo, S. Gandhi, N. Ready, T. Ng, W. Donat, and W.W. Mayo-Smith. 2006. Radiofrequency ablation followed by conventional radiotherapy for medically inoperable stage I non-small cell lung cancer. Chest 129(3):738–745.

Earle, C.C., Y. Chretien, C. Morris, J.Z. Ayanian, N.L. Keating, L.A. Polgreen, R. Wallace, P.A. Ganz, and J.C. Weeks. 2010. Employment among survivors of lung cancer and colorectal cancer. Journal of Clinical Oncology 28(10):1700.

Eberhard, D.A., B.E. Johnson, L.C. Amler, A.D. Goddard, S.L. Heldens, R.S. Herbst, W.L. Ince, P.A. Jänne, T. Januario, D.H. Johnson, P. Klein, V.A. Miller, M.A. Ostland, D.A. Ramies, D. Sebisanovic, J.A. Stinson, Y.R. Zhang, S. Seshagiri, and K.J. Hillan. 2005. Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. Journal of Clinical Oncology 23(25):5900–5909.

Eckardt, J.R., J. von Pawel, J.-L. Pujol, Z. Papai, E. Quoix, A. Ardizzoni, R. Poulin, A.J. Preston, G. Dane, and G. Ross. 2007. Phase III study of oral compared with intravenous topotecan as second-line therapy in small-cell lung cancer. Journal of Clinical Oncology 25(15):2086–2092.

Ersek, J.L., J.M. Eberth, K.K. McDonnell, S.M. Strayer, E. Sercy, K.B. Cartmell, and D.B. Friedman. 2016. Knowledge of, attitudes toward, and use of low-dose computed tomography for lung cancer screening among family physicians. Cancer 122(15):2324–2331.

Farago, A.F., and F.K. Keane. 2018. Current standards for clinical management of small cell lung cancer. Translational Lung Cancer Research 7(1):69–79.

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

FDA (U.S. Food and Drug Administration). 2020. FDA approves pembrolizumab for adults and children with TMB-H solid tumors. https://www.fda.gov/drugs/drug-approvals-anddatabases/fda-approves-pembrolizumab-adults-and-children-tmb-h-solid-tumors (accessed August 14, 2020).

Field, R.W., and B.L. Withers. 2012. Occupational and environmental causes of lung cancer. Clinics in Chest Medicine 33(4):681–703.

Forde, P.M., J.E. Chaft, K.N. Smith, V. Anagnostou, T.R. Cottrell, M.D. Hellmann, M. Zahurak, S.C. Yang, D.R. Jones, S. Broderick, R.J. Battafarano, M.J. Velez, N. Rekhtman, Z. Olah, J. Naidoo, K.A. Marrone, F. Verde, H. Guo, J. Zhang, J.X. Caushi, H.Y. Chan, J.W. Sidhom, R.B. Scharpf, J. White, E. Gabrielson, H. Wang, G.L. Rosner, V. Rusch, J.D. Wolchok, T. Merghoub, J.M. Taube, V.E. Velculescu, S.L. Topalian, J.R. Brahmer, and D.M. Pardoll. 2018. Neoadjuvant PD-1 blockade in resectable lung cancer. New England Journal of Medicine 378(21):1976–1986.

Fox, S.W., and D.E. Lyon. 2006. Symptom clusters and quality of life in survivors of lung cancer. Oncology Nursing Forum 33(5):931–936.

Gadgeel, S., D. Rodríguez-Abreu, G. Speranza, E. Esteban, E. Felip, M. Dómine, R. Hui, M.J. Hochmair, P. Clingan, S.F. Powell, S.Y.-S. Cheng, H.G. Bischoff, N. Peled, F. Grossi, R.R. Jennens, M. Reck, E.B. Garon, S. Novello, B. Rubio-Viqueira, M. Boyer, T. Kurata, J.E. Gray, J. Yang, T. Bas, M.C. Pietanza, and M.C. Garassino. 2020. Updated analysis from Keynote-189: Pembrolizumab or placebo plus pemetrexed and platinum for previously untreated metastatic nonsquamous non-small-cell lung cancer. Journal of Clinical Oncology 38(14):1505–1517.

Gage, A.A., and J. Baust. 1998. Mechanisms of tissue injury in cryosurgery. Cryobiology 37(3):171–186.

Gajra, A., N. Newman, G. P. Gamble, L.J. Kohman, and S.L. Graziano. 2003. Effect of number of lymph nodes sampled on outcome in patients with stage I non-small-cell lung cancer. Journal of Clinical Oncology 21(6):1029–1034.

Gandhi, L., D. Rodríguez-Abreu, S. Gadgeel, E. Esteban, E. Felip, F. De Angelis, M. Domine, P. Clingan, M.J. Hochmair, S.F. Powell, S.Y.-S. Cheng, H.G. Bischoff, N. Peled, F. Grossi, R.R. Jennens, M. Reck, R. Hui, E.B. Garon, M. Boyer, B. Rubio-Viqueira, S. Novello, T. Kurata, J.E. Gray, J. Vida, Z. Wei, J. Yang, H. Raftopoulos, M.C. Pietanza, and M.C. Garassino. 2018. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. New England Journal of Medicine 378(22):2078–2092.

Garon, E.B., M.D. Hellmann, N.A. Rizvi, E. Carcereny, N.B. Leighl, M.J. Ahn, J.P. Eder, A.S. Balmanoukian, C. Aggarwal, L. Horn, A. Patnaik, M. Gubens, S.S. Ramalingam, E. Felip, J.W. Goldman, C. Scalzo, E. Jensen, D.A. Kush, and R. Hui. 2019. Five-year overall survival for patients with advanced non small-cell lung cancer treated with pembrolizumab: Results from the phase I KEYNOTE-001 Study. Journal of Clinical Oncology 37(28):2518–2527.

Gettinger, S., L. Horn, D. Jackman, D. Spigel, S. Antonia, M. Hellmann, J. Powderly, R. Heist, L.V. Sequist, D.C. Smith, P. Leming, W.J. Geese, D. Yoon, A. Li, and J. Brahmer. 2018. Five-year follow-up of nivolumab in previously treated advanced non-small-cell lung cancer: Results from the CA209-003 study. Journal of Clinical Oncology 36(17):1675–1684.

Goldstraw, P., K. Chansky, J. Crowley, R. Rami-Porta, H. Asamura, W.E. Eberhardt, A.G. Nicholson, P. Groome, A. Mitchell, V. Bolejack, International Association for the Study of Lung Cancer Staging and Prognostic Factors Committee, Advisory Boards, and Participating Institutions, International Association for the Study of Lung Cancer Staging and Prognostic Factors Committee Advisory Boards and Participating Institutions. 2016. The IASLC Lung Cancer Staging Project: Proposals for revision of the TNM stage groupings in the forthcoming (eighth) edition of the TNM Classification for Lung Cancer. Journal of Thoracic Oncology 11(1):39–51.

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

Gomez, D.R., G.R. Blumenschein, Jr., J.J. Lee, M. Hernandez, R. Ye, D.R. Camidge, R.C. Doebele, F. Skoulidis, L.E. Gaspar, D.L. Gibbons, J.A. Karam, B.D. Kavanagh, C. Tang, R. Komaki, A.V. Louie, D.A. Palma, A.S. Tsao, B. Sepesi, W.N. William, J. Zhang, Q. Shi, X.S. Wang, S.G. Swisher, and J.V. Heymach. 2016. Local consolidative therapy versus maintenance therapy or observation for patients with oligometastatic non-small-cell lung cancer without progression after first-line systemic therapy: A multicentre, randomised, controlled, phase 2 study. The Lancet Oncology 17(12):1672–1682.

Gomez, D.R., C. Tang, J. Zhang, G.R. Blumenschein, Jr., M. Hernandez, J.J. Lee, R. Ye, D.A. Palma, A.V. Louie, D.R. Camidge, R.C. Doebele, F. Skoulidis, L.E. Gaspar, J.W. Welsh, D.L. Gibbons, J.A. Karam, B.D. Kavanagh, A.S. Tsao, B. Sepesi, S.G. Swisher, and J.V. Heymach. 2019. Local consolidative therapy vs. maintenance therapy or observation for patients with oligometastatic non-small-cell lung cancer: Long-term results of a multi-institutional, phase II, randomized study. Journal of Clinical Oncology 37(18):1558–1565.

Goncalves, P.H., S.L. Peterson, F.D. Vigneau, R.D. Shore, W.O. Quarshie, K. Islam, A.G. Schwartz, A.J. Wozniak, and S.M. Gadgeel. 2016. Risk of brain metastases in patients with nonmetastatic lung cancer: Analysis of the Metropolitan Detroit Surveillance, Epidemiology, and End Results (SEER) data. Cancer 122(12):1921–1927.

Gray, J.E., A. Villegas, D. Daniel, D. Vicente, S. Murakami, R. Hui, T. Kurata, A. Chiappori, K.H. Lee, B.C. Cho, D. Planchard, L. Paz-Ares, C. Faivre-Finn, J.F. Vansteenkiste, D.R. Spigel, C. Wadsworth, M. Taboada, P.A. Dennis, M. Ozguroglu, and S.J. Antonia. 2020. Three-year overall survival with durvalumab after chemoradiotherapy in stage III NSCLC-update from PACIFIC. Journal of Thoracic Oncology 15(2):288–293.

Griffin, R., and R.A. Ramirez. 2017. Molecular targets in non-small cell lung cancer. The Ochsner Journal 17(4):388–392.

Ha, D., A.L. Ries, S.M. Lippman, and M.M. Fuster. 2020. Effects of curative-intent lung cancer therapy on functional exercise capacity and patient-reported outcomes. Supportive Care in Cancer 28(10):4707–4720.

Harle, A.S., F.H. Blackhall, J.A. Smith, and A. Molassiotis. 2012. Understanding cough and its management in lung cancer. Current Opinions in Supportive and Palliative Care 6(2):153–162.

Haseltine, J.M., A. Rimner, D.Y. Gelblum, A. Modh, K.E. Rosenzweig, A. Jackson, E.D. Yorke, and A.J. Wu. 2016. Fatal complications after stereotactic body radiation therapy for central lung tumors abutting the proximal bronchial tree. Practical Radiation Oncology 6(2):e27–e33.

Hobbs, C.J., S.J. Ko, N.N. Paryani, J.M. Accurso, K.R. Olivier, Y.I. Garces, S.S. Park, C.L. Hallemeier, S.E. Schild, S.A. Vora, J.B. Ashman, W.G. Rule, J.R. Bowers, M.G. Heckman, N.N. Diehl, and R.C. Miller. 2018. Stereotactic body radiotherapy for medically inoperable stage I–II non-small cell lung cancer: The Mayo Clinic experience. Mayo Clinic Proceedings in Innovation, Quality & Outcomes 2(1):40–48.

Hochstenbag, M.M., A. Twijnstra, J.T. Wilmink, E.F. Wouters, and G.P. ten Velde. 2000. Asymptomatic brain metastases (BM) in small cell lung cancer (SCLC): MR-imaging is useful at initial diagnosis. Journal of Neuro-Oncology 48(3):243–248.

Horita, N., M. Yamamoto, T. Sato, T. Tsukahara, H. Nagakura, K. Tashiro, Y. Shibata, H. Watanabe, K. Nagai, M. Inoue, K. Nakashima, R. Ushio, M. Shinkai, M. Kudo, and T. Kaneko. 2015. Topotecan for relapsed small-cell lung cancer: Systematic review and meta-analysis of 1347 patients. Science Reports 5:15437.

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

Horn, L., A.S. Mansfield, A. Szczesna, L. Havel, M. Krzakowski, M.J. Hochmair, F. Huemer, G. Losonczy, M.L. Johnson, M. Nishio, M. Reck, T. Mok, S. Lam, D.S. Shames, J. Liu, B. Ding, A. Lopez-Chavez, F. Kabbinavar, W. Lin, A. Sandler, and S.V. Liu. 2018. First-line atezolizumab plus chemotherapy in extensive-stage small-cell lung cancer. New England Journal of Medicine 379(23):2220–2229.

Howlader, N., A.M. Noone, M. Krapcho, D. Miller, A. Brest, M. Yu, J. Ruhl, Z. Tatalovich, A. Mariotto, D.R. Lewis, H.S. Chen, E.J. Feuer, and K.A. Cronin. 2020. SEER cancer statistics review,1975–2017. https://seer.cancer.gov/csr/1975_2017 (accessed September 1, 2020).

Huang, J., A.E. Logue, J.S. Ostroff, B.J. Park, M. McCabe, D.R. Jones, M.S. Bains, N.P. Rizk, M.G. Kris, and V.W. Rusch. 2014. Comprehensive long-term care of patients with lung cancer: Development of a novel thoracic survivorship program. Annals of Thoracic Surgery 98(3):955–961.

Hung, R., P. Krebs, E.J. Coups, M.B. Feinstein, B.J. Park, J. Burkhalter, and J.S. Ostroff. 2011. Fatigue and functional impairment in early-stage non-small cell lung cancer survivors. Journal of Pain and Symptom Management 41(2):426–435.

Ilonen, I., V. Rauma, J. Räsänen, and J. Salo. 2013. Long-term lung cancer survivors have permanently decreased quality of life following surgery. Interactive CardioVascular and Thoracic Surgery 17(Suppl 2):S132.

Iyengar, P., Z. Wardak, D.E. Gerber, V. Tumati, C. Ahn, R.S. Hughes, J.E. Dowell, N. Cheedella, L. Nedzi, K.D. Westover, S. Pulipparacharuvil, H. Choy, and R.D. Timmerman. 2018. Consolidative radiotherapy for limited metastatic non-small-cell lung cancer: A phase 2 randomized clinical trial. JAMA Oncology 4(1):e173501.

James, T.W., and L.P. Faber. 1999. Indications for pneumonectomy. Pneumonectomy for malignant disease. Chest Surgery Clinics of North America 9(2):291–309.

Kalemkerian, G.P. 2012. Staging and imaging of small cell lung cancer. Cancer Imaging 11(1):253–258.

Kashima, M., K. Yamakado, H. Takaki, H. Kodama, T. Yamada, J. Uraki, and A. Nakatsuka. 2011. Complications after 1000 lung radiofrequency ablation sessions in 420 patients: A single center’s experiences. American Journal Roentgenology 197(4):W576–W580.

Katz, M.L., K.A. Donohue, C.M. Alfano, J.M. Day, J.E. Herndon, and E.D. Paskett. 2009. Cancer surveillance behaviors and psychosocial factors among long-term survivors of breast cancer: Cancer and leukemia group B 79804. Cancer 115(3):480–488.

Kay, F.U., A. Kandathil, K. Batra, S.S. Saboo, S. Abbara, and P. Rajiah. 2017. Revisions to the tumor, node, metastasis staging of lung cancer (8th edition): Rationale, radiologic findings and clinical implications. World Journal of Radiology 9(6):269–279.

Kelly, K., K. Chansky, L.E. Gaspar, K.S. Albain, J. Jett, Y.C. Ung, D.H. Lau, J.J. Crowley, and D.R. Gandara. 2008. Phase III trial of maintenance gefitinib or placebo after concurrent chemoradiotherapy and docetaxel consolidation in inoperable stage III non-small-cell lung cancer: SWOG S0023. Journal of Clinical Oncology 26(15):2450–2456.

Kenny, P.M., M.T. King, R.C. Viney, M.J. Boyer, C.A. Pollicino, J.M. McLean, M.J. Fulham, and B.C. McCaughan. 2008. Quality of life and survival in the 2 years after surgery for non-small-cell lung cancer. Journal of Clinical Oncology 26(2):233–241.

Kopec, S.E., R.S. Irwin, C.B. Umali-Torres, J.P. Balikian, and A.A. Conlan. 1998. The post-pneumonectomy state. Chest 114(4):1158–1184.

Kubota, K., K. Furuse, M. Kawahara, N. Kodama, M. Yamamoto, M. Ogawara, S. Negoro, N. Masuda, M. Takada, and K. Matsui. 1994. Role of radiotherapy in combined modality treatment of locally advanced non-small-cell lung cancer. Journal of Clinical Oncology 12(8):1547–1552.

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

Kwiatkowski, D.J., V.W. Rusch, J.E. Chaft, B.E. Johnson, A. Nicholas, I.I. Wistuba, R. Merritt, J.M. Lee, P.A. Bunn, Y. Tang, S.C. Phan, S.N. Waqar, A. Patterson, E.B. Haura, E.M. Toloza, K.L. Reckamp, D. Raz, K. Schulze, A. Johnson, and D.P. Carbone. 2019. Neoadjuvant atezolizumab in resectable non-small cell lung cancer (NSCLC): Interim analysis and biomarker data from a multicenter study (LCMC3). Journal of Clinical Oncology 37(15):8503.

Lathan, C.S., A. Cronin, R. Tucker-Seeley, S.Y. Zafar, J.Z. Ayanian, and D. Schrag. 2016. Association of financial strain with symptom burden and quality of life for patients with lung or colorectal cancer. Journal of Clinical Oncology 34(15):1732–1740.

Lewis, J.A., W.J. Petty, J.A. Tooze, D.P. Miller, C. Chiles, A.A. Miller, C. Bellinger, and K.E. Weaver. 2015. Low-dose CT lung cancer screening practices and attitudes among primary care providers at an academic medical center. Cancer Epidemiology, Biomarkers & Prevention 24(4):664–670.

Lukas, R.V., V. Gondi, D.O. Kamson, P. Kumthekar, and R. Salgia. 2017. State-of-the-art considerations in small cell lung cancer brain metastases. Oncotarget 8(41):71223–71233.

Machtay, M., J.H. Lee, J.B. Shrager, L.R. Kaiser, and E. Glatstein. 2001. Risk of death from intercurrent disease is not excessively increased by modern postoperative radiotherapy for high-risk resected non-small-cell lung carcinoma. Journal of Clinical Oncology 19(19):3912–3917.

Manser, R., G. Wright, D. Hart, G. Byrnes, and D.A. Campbell. 2005. Surgery for early stage non-small cell lung cancer. Cochrane Database of Systemic Reviews 2005(1):CD004699.

Ming, X., Y. Feng, C. Yang, W. Wang, P. Wang, and J. Deng. 2016. Radiation-induced heart disease in lung cancer radiotherapy: A dosimetric update. Medicine 95(41):e5051.

Mok, T.S., Y.-L. Wu, M.-J. Ahn, M.C. Garassino, H.R. Kim, S.S. Ramalingam, F.A. Shepherd, Y. He, H. Akamatsu, W.S.M.E. Theelen, C.K. Lee, M. Sebastian, A. Templeton, H. Mann, M. Marotti, S. Ghiorghiu, and V.A. Papadimitrakopoulou. 2016. Osimertinib or platinum–pemetrexed in EGFR T790m–positive lung cancer. New England Journal of Medicine 376(7):629–640.

National Lung Screening Trial Research Team, D.R. Aberle, A.M. Adams, C.D. Berg, W.C. Black, J.D. Clapp, R.M. Fagerstrom, I.F. Gareen, C. Gatsonis, P.M. Marcus, and J.D. Sicks. 2011. Reduced lung-cancer mortality with low-dose computed tomographic screening. New England Journal of Medicine 365(5):395–409.

NCCN (National Comprehensive Cancer Network). 2020a. NCCN clinical practice guidelines in oncology: Small cell lung cancer. https://www.nccn.org/professionals/physician_gls/pdf/sclc.pdf (accessed April 26, 2020).

NCCN. 2020b. NCCN clinical practice guidelines in oncology. Non-small cell lung cancer. https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf (accessed April 26, 2020).

NCI (National Cancer Institute). 2011. Radon and cancer. https://www.cancer.gov/about-cancer/causes-prevention/risk/substances/radon/radon-fact-sheet#how-does-radon-cause-cancer (accessed December 30, 2020).

NSCLC Meta-analysis Collaborative Group. 2014. Preoperative chemotherapy for non-small-cell lung cancer: A systematic review and meta-analysis of individual participant data. The Lancet 383(9928):1561–1571.

O’Rourke, N., I.F.M. Roque, N. Farre Bernado, and F. Macbeth. 2010. Concurrent chemoradiotherapy in non-small cell lung cancer. Cochrane Database of Systematic Reviews 2010(6):CD002140.

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

Palma, D.A., R. Olson, S. Harrow, S. Gaede, A.V. Louie, C. Haasbeek, L. Mulroy, M. Lock, G.B. Rodrigues, B.P. Yaremko, D. Schellenberg, B. Ahmad, G. Griffioen, S. Senthi, A. Swaminath, N. Kopek, M. Liu, K. Moore, S. Currie, G.S. Bauman, A. Warner, and S. Senan. 2019. Stereotactic ablative radiotherapy versus standard of care palliative treatment in patients with oligometastatic cancers (SABR-COMET): A randomised, phase 2, open-label trial. The Lancet 393(10185):2051–2058.

Parikh, R.B., A.M. Cronin, D.E. Kozono, G.R. Oxnard, R.H. Mak, D.M. Jackman, P.C. Lo, E.H. Baldini, B.E. Johnson, and A.B. Chen. 2014. Definitive primary therapy in patients presenting with oligometastatic non-small cell lung cancer. International Journal of Radiation Oncology, Biology, Physics 89(4):880–887.

Paz-Ares, L., M. Dvorkin, Y. Chen, N. Reinmuth, K. Hotta, D. Trukhin, G. Statsenko, M.J. Hochmair, M. Ozguroglu, J.H. Ji, O. Voitko, A. Poltoratskiy, S. Ponce, F. Verderame, L. Havel, I. Bondarenko, A. Kazarnowicz, G. Losonczy, N.V. Conev, J. Armstrong, N. Byrne, N. Shire, H. Jiang, and J.W. Goldman. 2019. Durvalumab plus platinum-etoposide versus platinum-etoposide in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): A randomised, controlled, open-label, phase 3 trial. The Lancet 394(10212):1929–1939.

PDQ® Adult Treatment Editorial Board. 2020. PDQ non-small cell lung cancer treatment. https://www.cancer.gov/types/lung/hp/non-small-cell-lung-treatment-pdq (accessed April 22, 2020).

Poirier, J.T., J. George, T.K. Owonikoko, A. Berns, E. Brambilla, L.A. Byers, D. Carbone, H.J. Chen, C.L. Christensen, C. Dive, A.F. Farago, R. Govindan, C. Hann, M.D. Hellmann, L. Horn, J.E. Johnson, Y.S. Ju, S. Kang, M. Krasnow, J. Lee, S.H. Lee, J. Lehman, B. Lok, C. Lovly, D. MacPherson, D. McFadden, J. Minna, M. Oser, K. Park, K.S. Park, Y. Pommier, V. Quaranta, N. Ready, J. Sage, G. Scagliotti, M.L. Sos, K.D. Sutherland, W.D. Travis, C.R. Vakoc, S.J. Wait, I. Wistuba, K.K. Wong, H. Zhang, J. Daigneault, J. Wiens, C.M. Rudin, and T.G. Oliver. 2020. New approaches to SCLC therapy: From the laboratory to the clinic. Journal of Thoracic Oncology 15(4):520–540.

Rahman, R., B.M. Alexander, and P.Y. Wen. 2020. Neurologic complications of cranial radiation therapy and strategies to prevent or reduce radiation toxicity. Current Neurology and Neuroscience Reports 20(8):34.

Rausch, S.M., B.D. Gonzalez, M.M. Clark, C. Patten, S. Felten, H. Liu, Y. Li, J. Sloan, and P. Yang. 2012. SNPs in PTGS2 and LTA predict pain and quality of life in long term lung cancer survivors. Lung Cancer 77(1):217–223.

Ready, N.E., P.A. Ott, M.D. Hellmann, J. Zugazagoitia, C.L. Hann, F. de Braud, S.J. Antonia, P.A. Ascierto, V. Moreno, A. Atmaca, S. Salvagni, M. Taylor, A. Amin, D.R. Camidge, L. Horn, E. Calvo, A. Li, W.H. Lin, M.K. Callahan, and D.R. Spigel. 2020. Nivolumab monotherapy and nivolumab plus ipilimumab in recurrent small cell lung cancer: Results from the CheckMate 032 randomized cohort. Journal of Thoracic Oncology 15(3):426–435.

Reck, M., D. Rodríguez-Abreu, A.G. Robinson, R. Hui, T. Cs szi, A. Fülöp, M. Gottfried, N. Peled, A. Tafreshi, S. Cuffe, M. O’Brien, S. Rao, K. Hotta, M.A. Leiby, G.M. Lubiniecki, Y. Shentu, R. Rangwala, and J.R. Brahmer. 2016. Pembrolizumab versus chemotherapy for PD-L1–positive non-small-cell lung cancer. New England Journal of Medicine 375(19):1823–1833.

Riely, G.J., J. Marks, and W. Pao. 2009. KRAS mutations in non-small cell lung cancer. Proceedings of the American Thoracic Society 6(2):201–205.

Rogers, M.L., and J.P. Duffy. 2000. Surgical aspects of chronic post-thoracotomy pain. European Journal of Cardiothoracic Surgery 18(6):711–716.

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

Rudin, C.M., N. Ismaila, C.L. Hann, N. Malhotra, B. Movsas, K. Norris, M.C. Pietanza, S.S. Ramalingam, A.T. Turrisi, 3rd, and G. Giaccone. 2015. Treatment of small-cell lung cancer: American Society of Clinical Oncology endorsement of the American College of Chest Physicians guideline. Journal of Clinical Oncology 33(34):4106–4111.

Sarna, L., L. Evangelista, D. Tashkin, G. Padilla, C. Holmes, M.L. Brecht, and F. Grannis. 2004. Impact of respiratory symptoms and pulmonary function on quality of life of long-term survivors of non-small cell lung cancer. Chest 125(2):439–445.

Sarna, L., M.E. Cooley, J.K. Brown, C. Chernecky, D. Elashoff, and J. Kotlerman. 2008. Symptom severity 1 to 4 months after thoracotomy for lung cancer. American Journal of Critical Care 17(5):455–467.

Schneider, B.J., M.E. Daly, E.B. Kennedy, M.B. Antonoff, S. Broderick, J. Feldman, S. Jolly, B. Meyers, G. Rocco, C. Rusthoven, B.J. Slotman, D.H. Sterman, and B.M. Stiles. 2018. Stereotactic body radiotherapy for early-stage non-small-cell lung cancer: American Society of Clinical Oncology endorsement of the American Society for Radiation Oncology evidence-based guideline. Journal of Clinical Oncology 36(7):710–719.

Schneider, B.J., N. Ismaila, J. Aerts, C. Chiles, M.E. Daly, F.C. Detterbeck, J.W.D. Hearn, S.I. Katz, N.B. Leighl, B. Levy, B. Meyers, S. Murgu, L. Nekhlyudov, E.S. Santos, N. Singh, J. Tashbar, D. Yankelevitz, and N. Altorki. 2020. Lung cancer surveillance after definitive curative-intent therapy: ASCO guideline. Journal of Clinical Oncology 38(7):753–766.

Schnoll, R.A., E. Martinez, K.L. Tatum, D.M. Weber, N. Kuzla, M. Glass, J.A. Ridge, C. Langer, C. Miyamoto, and E.P. Wileyto. 2010. A bupropion smoking cessation clinical trial for cancer patients. Cancer Causes & Control 21(6):811–820.

Schouten, L.J., J. Rutten, H.A. Huveneers, and A. Twijnstra. 2002. Incidence of brain metastases in a cohort of patients with carcinoma of the breast, colon, kidney, and lung and melanoma. Cancer 94(10):2698–2705.

Schulte, T., B. Schniewind, P. Dohrmann, T. Küchler, and R. Kurdow. 2009. The extent of lung parenchyma resection significantly impacts long-term quality of life in patients with non-small cell lung cancer. Chest 135(2):322–329.

SEER (Surveillance, Epidemiology, and End Results). n.d.-a. Cancer statistics review 1975–2017: Table 15.14 Non-small cell cancer of the lung and bronchus (invasive). https://seer.cancer.gov/csr/1975_2017/browse_csr.php?sectionSEL=15&pageSEL=sect_15_table.14#table5 (accessed November 12, 2020).

SEER. n.d.-b. Cancer statistics review 1975–2017: Table 15.13 Small cell cancer of the lung and bronchus (invasive). https://seer.cancer.gov/csr/1975_2017/browse_csr.php?sectionSEL=15&pageSEL=sect_15_table.13 (accessed November 12, 2020).

SEER. n.d.-c. Cancer statistics review 1975–2017: Table 15.15 Cancer of the lung and bronchus (invasive). https://seer.cancer.gov/csr/1975_2017/browse_csr.php?sectionSEL=15&pageSEL=sect_15_table.15 (accessed November 17, 2020).

SEER. n.d.-d. Cancer stat facts: Lung and bronchus cancer. https://seer.cancer.gov/statfacts/html/lungb.html (accessed November 12, 2020).

Senthi, S., F.J. Lagerwaard, C.J. Haasbeek, B.J. Slotman, and S. Senan. 2012. Patterns of disease recurrence after stereotactic ablative radiotherapy for early stage non-small-cell lung cancer: A retrospective analysis. The Lancet Oncology 13(8):802–809.

Shi, Q., T.G. Smith, J.D. Michonski, K.D. Stein, C. Kaw, and C.S. Cleeland. 2011. Symptom burden in cancer survivors 1 year after diagnosis: A report from the American Cancer Society’s studies of cancer survivors. Cancer 117(12):2779–2790.

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

Shigematsu, H., L. Lin, T. Takahashi, M. Nomura, M. Suzuki, I.I. Wistuba, K.M. Fong, H. Lee, S. Toyooka, N. Shimizu, T. Fujisawa, Z. Feng, J.A. Roth, J. Herz, J.D. Minna, and A.F. Gazdar. 2005. Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers. Journal of the National Cancer Institute 97(5):339–346.

Slotman, B.J., H. van Tinteren, J.O. Praag, J.L. Knegjens, S.Y. El Sharouni, M. Hatton, A. Keijser, C. Faivre-Finn, and S. Senan. 2015. Use of thoracic radiotherapy for extensive stage small-cell lung cancer: A phase 3 randomised controlled trial. The Lancet 385(9962):36–42.

Solomon, B.J., T. Mok, D.-W. Kim, Y.-L. Wu, K. Nakagawa, T. Mekhail, E. Felip, F. Cappuzzo, J. Paolini, T. Usari, S. Iyer, A. Reisman, K.D. Wilner, J. Tursi, and F. Blackhall. 2014. First-line crizotinib versus chemotherapy in ALK-positive lung cancer. New England Journal of Medicine 371(23):2167–2177.

Soria, J.-C., Y. Ohe, J. Vansteenkiste, T. Reungwetwattana, B. Chewaskulyong, K.H. Lee, A. Dechaphunkul, F. Imamura, N. Nogami, T. Kurata, I. Okamoto, C. Zhou, B.C. Cho, Y. Cheng, E.K. Cho, P.J. Voon, D. Planchard, W.-C. Su, J.E. Gray, S.-M. Lee, R. Hodge, M. Marotti, Y. Rukazenkov, and S.S. Ramalingam. 2017. Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer. New England Journal of Medicine 378(2):113–125.

Subramanian, M.P., and V. Puri. 2019. Neoadjuvant vs. adjuvant chemotherapy in locally advanced non-small cell lung cancer—is timing everything? Journal of Thoracic Disease 11(12):5674–5676.

Sugimura, H., and P. Yang. 2006. Long-term survivorship in lung cancer: A review. Chest 129(4):1088–1097.

The Netherlands Cancer Institute and Dutch Cancer Society. 2013. Prophylactic cranial irradiation with or without hippocampal avoidance in SCLC: A randomized phase III trial. https://clinicaltrials.gov/ct2/show/NCT01780675 (accessed October 27, 2020).

Torok, J.A., L. Gu, D.J. Tandberg, X. Wang, D.H. Harpole, Jr., C.R. Kelsey, and J.K. Salama. 2017. Patterns of distant metastases after surgical management of non-small-cell lung cancer. Clinics in Lung Cancer 18(1):e57–e70.

Torp, S., R.A. Nielsen, S.D. Fosså, S.B. Gudbergsson, and A.A. Dahl. 2013. Change in employment status of 5-year cancer survivors. European Journal of Public Health 23(1):116–122.

Tsujino, K., T. Kurata, S. Yamamoto, T. Kawaguchi, A. Kubo, S. Isa, Y. Hasegawa, S.H. Ou, M. Takada, and M. Ando. 2013. Is consolidation chemotherapy after concurrent chemoradiotherapy beneficial for patients with locally advanced non-small-cell lung cancer? A pooled analysis of the literature. Journal of Thoracic Oncology 8(9):1181–1189.

University of Texas Southwestern Medical Center. 2015. JoLT-Ca sublobar resection (SR) versus Stereotactic Ablative Radiotherapy (SAbR) for lung cancer (STABLE-MATES). https://www.clinicaltrials.gov/ct2/show/NCT02468024?term=STABLE-MATES&rank=1 (accessed April 26, 2020).

USPSTF (U.S. Preventive Services Task Force). 2013. U.S. Preventive Services Task Force recommendation statement: Screening for lung cancer. https://www.uspreventiveservicestaskforce.org/uspstf/recommendation/lung-cancer-screening (accessed April 26, 2020).

USPSTF. 2020. Draft recommendation statement: Lung cancer: Screening. https://www.uspreventiveservicestaskforce.org/uspstf/draft-recommendation/lung-cancer-screening-2020 (accessed November 10, 2020).

VA ORD (U.S. Department of Veterans Affairs Office of Research and Development). 2016. Veterans Affairs Lung Cancer Surgery or Stereotactic Radiotherapy (VALOR). https://www.clinicaltrials.gov/ct2/show/NCT02984761?term=NCT02984761&draw=2&rank=1 (accessed April 26, 2020).

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

Vaporciyan, A.A., K.W. Merriman, F. Ece, J.A. Roth, W.R. Smythe, S.G. Swisher, G.L. Walsh, J.C. Nesbitt, and J.B. Putnam Jr. 2002. Incidence of major pulmonary morbidity after pneumonectomy: Association with timing of smoking cessation. Annals of Thoracic Surgery 73(2):420–426.

Verma, V., C.B. Simone, 2nd, and M. Werner-Wasik. 2017. Acute and late toxicities of concurrent chemoradiotherapy for locally-advanced non-small cell lung cancer. Cancers 9(9):120.

Vijayvergia, N., P.C. Shah, and C.S. Denlinger. 2015. Survivorship in non-small cell lung cancer: Challenges faced and steps forward. Journal of the National Comprehensive Cancer Network 13(9):1151–1161.

Wang, S., S. Zimmermann, K. Parikh, A.S. Mansfield, and A.A. Adjei. 2019. Current diagnosis and management of small-cell lung cancer. Mayo Clinic Proceedings 94(8):1599–1622.

Win, T., L. Sharples, F. Wells, A. Ritchie, H. Munday, and C. Laroche. 2005. Effect of lung cancer surgery on quality of life. Thorax 60(3):234–238.

Winton, T., R. Livingston, D. Johnson, J. Rigas, M. Johnston, C. Butts, Y. Cormier, G. Goss, R. Inculet, and E. Vallieres. 2005. Vinorelbine plus cisplatin vs. observation in resected non-small-cell lung cancer. New England Journal of Medicine 352(25):2589–2597.

Yan, T.D., D. Black, P.G. Bannon, and B.C. McCaughan. 2009. Systematic review and meta-analysis of randomized and nonrandomized trials on safety and efficacy of video-assisted thoracic surgery lobectomy for early-stage non-small-cell lung cancer. Journal of Clinical Oncology 27(15):2553–2562.

Yang, P., A.L. Cheville, J.A. Wampfler, Y.I. Garces, A. Jatoi, M.M. Clark, S.D. Cassivi, D.E. Midthun, R.S. Marks, M.-C. Aubry, S.H. Okuno, B.A. Williams, F.C. Nichols, V.F. Trastek, H. Sugimura, L. Sarna, M.S. Allen, C. Deschamps, and J.A. Sloan. 2012. Quality of life and symptom burden among long-term lung cancer survivors. Journal of Thoracic Oncology 7(1):64–70.

Yano, T., T. Okamoto, A. Haro, S. Fukuyama, T. Yoshida, M. Kohno, and Y. Maehara. 2013. Local treatment of oligometastatic recurrence in patients with resected non-small cell lung cancer. Lung Cancer 82(3):431–435.

Zabora, J., K. BrintzenhofeSzoc, B. Curbow, C. Hooker, and S. Piantadosi. 2001. The prevalence of psychological distress by cancer site. Psycho Oncology 10(1):19–28.

Zappa, C., and S.A. Mousa. 2016. Non-small cell lung cancer: Current treatment and future advances. Translational Lung Cancer Research 5(3):288–300.

Zheng, A., X. Wang, X. Yang, W. Wang, G. Huang, Y. Gai, and X. Ye. 2014. Major complications after lung microwave ablation: A single-center experience on 204 sessions. Annals of Thoracic Surgery 98(1):243–248.

Zhong, L., J. Suo, Y. Wang, J. Han, H. Zhou, H. Wei, and J. Zhu. 2020. Prognosis of limited-stage small cell lung cancer with comprehensive treatment including radical resection. World Journal of Surgical Oncology 18:27.

Zhou, C., Y.L. Wu, G. Chen, J. Feng, X.Q. Liu, C. Wang, S. Zhang, J. Wang, S. Zhou, S. Ren, S. Lu, L. Zhang, C. Hu, C. Hu, Y. Luo, L. Chen, M. Ye, J. Huang, X. Zhi, Y. Zhang, Q. Xiu, J. Ma, L. Zhang, and C. You. 2011. 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. The Lancet Oncology 12(8):735–742.

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×

This page intentionally left blank.

Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 155
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 156
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 157
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 158
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 159
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 160
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 161
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 162
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 163
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 164
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 165
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 166
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 167
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 168
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 169
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 170
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 171
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 172
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 173
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 174
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 175
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 176
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 177
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 178
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 179
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 180
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 181
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 182
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 183
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 184
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 185
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 186
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 187
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 188
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 189
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 190
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 191
Suggested Citation:"6 Lung Cancer." National Academies of Sciences, Engineering, and Medicine. 2021. Diagnosing and Treating Adult Cancers and Associated Impairments. Washington, DC: The National Academies Press. doi: 10.17226/25956.
×
Page 192
Next: 7 Selected Topics in Other Cancers »
Diagnosing and Treating Adult Cancers and Associated Impairments Get This Book
×
 Diagnosing and Treating Adult Cancers and Associated Impairments
Buy Paperback | $80.00 Buy Ebook | $64.99
MyNAP members save 10% online.
Login or Register to save!
Download Free PDF

Cancer is the second leading cause of death among adults in the United States after heart disease. However, improvements in cancer treatment and earlier detection are leading to growing numbers of cancer survivors. As the number of cancer survivors grows, there is increased interest in how cancer and its treatments may affect a person's ability to work, whether the person has maintained employment throughout the treatment or is returning to work at a previous, current, or new place of employment. Cancer-related impairments and resulting functional limitations may or may not lead to disability as defined by the U.S. Social Security Administration (SSA), however, adults surviving cancer who are unable to work because of cancer-related impairments and functional limitations may apply for disability benefits from SSA.

At the request of SSA, Diagnosing and Treating Adult Cancers and Associated Impairments provides background information on breast cancer, lung cancer, and selected other cancers to assist SSA in its review of the listing of impairments for disability assessments. This report addresses several specific topics, including determining the latest standards of care as well as new technologies for understanding disease processes, treatment modalities, and the effect of cancer on a person's health and functioning, in order to inform SSA's evaluation of disability claims for adults with cancer.

READ FREE ONLINE

  1. ×

    Welcome to OpenBook!

    You're looking at OpenBook, NAP.edu's online reading room since 1999. Based on feedback from you, our users, we've made some improvements that make it easier than ever to read thousands of publications on our website.

    Do you want to take a quick tour of the OpenBook's features?

    No Thanks Take a Tour »
  2. ×

    Show this book's table of contents, where you can jump to any chapter by name.

    « Back Next »
  3. ×

    ...or use these buttons to go back to the previous chapter or skip to the next one.

    « Back Next »
  4. ×

    Jump up to the previous page or down to the next one. Also, you can type in a page number and press Enter to go directly to that page in the book.

    « Back Next »
  5. ×

    Switch between the Original Pages, where you can read the report as it appeared in print, and Text Pages for the web version, where you can highlight and search the text.

    « Back Next »
  6. ×

    To search the entire text of this book, type in your search term here and press Enter.

    « Back Next »
  7. ×

    Share a link to this book page on your preferred social network or via email.

    « Back Next »
  8. ×

    View our suggested citation for this chapter.

    « Back Next »
  9. ×

    Ready to take your reading offline? Click here to buy this book in print or download it as a free PDF, if available.

    « Back Next »
Stay Connected!