Skip to main content
UKPMC Funders Author Manuscripts logoLink to UKPMC Funders Author Manuscripts
. Author manuscript; available in PMC: 2025 Nov 18.
Published in final edited form as: JAMA. 2025 Nov 25;334(20):1836–1845. doi: 10.1001/jama.2025.17695

Lung cancer In non-smokers

Cian Murphy 1,2,#, Tej Pandya 1,3,#, Charles Swanton 1,4,5, Benjamin J Solomon 6,7,
PMCID: PMC7618360  EMSID: EMS210578  PMID: 41114991

Abstract

Importance

Lung cancer in non-smokers (defined as people who have smoked fewer than 100 cigarettes in their lifetime) accounts for 15% to 25% of all lung cancer cases worldwide. In the US, the annual incidence of lung cancer in non-smokers is 14-20 per 100,000 person-years in women and 4-13 per 100,000 person-years in men.

Observations

Most lung cancers in non-smokers (60-80%) are adenocarcinomas, compared with 40% among current or former smokers with lung cancer. The remainder are adenosquamous (10-20%), squamous (10-15%) and small cell lung cancer (0-5%). Risk factors include exposure to passive smoking (percentage attributable fraction: 15%), radon exposure (9%), air pollution (8%), asbestos (8%), and history of lung cancer in a first-degree family member (26%). Therapeutically targetable genomic variants such as EGFR variants or ALK gene rearrangements are more common in tumors from non smokers compared to those with a smoking history (43% vs 11% for EGFR and 12% vs 2% for ALK). In contrast, tumor mutation burden, the number of somatic mutations in a tumor cell, is lower in lung cancer among non-smokers (0-3 mutations per Megabase (Mb) vs 0-30 mutations/Mb). Similar to individuals with a history of smoking, non-smokers with lung cancer may present with wheeze, chest pain, dyspnea, hemoptysis or symptoms attributable to metastatic disease (eg, bone pain and headache) or be diagnosed with incidentally detected disease.The US Preventive Services Task Force (USPSTF) does not currently recommend lung cancer screening with low-dose CT scans for non-smokers, although screening guidelines vary globally.

Treatment typically involves a combination of surgery, radiotherapy and systemic therapies depending on stage, performance status and molecular features of the tumor. Comprehensive next-generation sequencing should be performed on Stage Ib to IIIa lung cancer tumor tissue from non-smokers because actionable genomic alterations such as EGFR variants or ALK gene rearrangements are treated with targeted therapy such as the tyrosine kinase inhibitors osimertinib or lorlatinib, respectively. Median survival among non-smokers with advanced NSCLC (Stage IIIb or higher) and actionable genomic alterations can exceed 3-5 years, while survival without these genomic alterations is similiar to lung cancer in smokers (1-2 years).

Conclusions

Lung cancer in non-smokers accounts for 15-20% of lung cancer cases worldwide. Among patients with lung cancer, non-smokers are more likely to have genomic alterations such as EGFR variants or ALK gene rearrangements, and these patients have improved survival when treated with tyrosine kinase inhibitors, compared with chemotherapy.

Introduction

Lung cancer is the leading cause of cancer-related mortality worldwide, causing approximately 1.8 million deaths in 20221. Although tobacco smoking is the predominant risk factor for lung cancer, present in 80–85% of cases, smoking rates are declining in the US and in many other parts of the world. In parallel with a decrease in adult US smoking rates (23.3% in 2000 vs 11.5% in 20212), lung cancer incidence in the US declined from 68/100,000 in 2000 to 47/100,000 in 20213.

Lung cancer among non-smokers (individuals who have smoked fewer than 100 cigarettes in their lifetime) accounts for 15-25% of all lung cancer4,5, and may be related to factors including age, air pollution, passive smoking, radon exposure, asbestos exposure and germline genetic risk68. The misperception that lung cancer is almost invariably caused by smoking may delay assessment and diagnosis9,10. This review will discuss the epidemiology, risk factors, screening, clinical presentation, assessment, diagnosis, treatment and prognosis of lung cancer in non-smokers.

Methods

A PubMed search for English-language articles describing studies on lung cancer in non-smokers was conducted from January 1, 2005 to August 1st, 2025. In total, 902 studies were retrieved from this search and 92 were included in this review, consisting of 6 meta-analyses or systematic reviews, 16 randomised clinical trials, 8 prospective cohort studies, 7 retrospective cohort studies, 3 cross-sectional studies, 4 observational or case-control studies, 13 genomic or molecular studies, 11 narrative reviews, 10 statistical or surveillance reports, 11 guidelines or recommendation statements, 2 registry or database studies, and 1 preclinical or experimental study.

Epidemiology and Screening

Most lung cancers in non-smokers are adenocarcinomas (60-80%), and are diagnosed at a median age of 67 years, compared with age 70 years in former or current smokers (Table 1)11. The absolute incidence of lung cancer in non-smokers in the US and worldwide is increasing. A retrospective study of 10,000 cases from 3 US hospital networks reported that the proportion of lung cancer among non-smokers increased from 8% to 14.9% from 1990-20134. A pooled analysis of 7 Finnish cohorts reported an absolute increase in lung cancer among non-smokers from 6.9 per 100,000 person-years in 1972 to 12.9 per 100,000 person-years in 201512. Lung cancer is the third most diagnosed cancer worldwide, and if lung cancer in non-smokers were classified as a distinct entity, it would be the 7th most common cancer13.

Table 1.

The principal differences between lung cancer in people who have never smoked (LCINS) and lung cancer in people who have smoked (LCIPS). Mb; Megabase. SBS4: The Catalogue of Somatic Mutations in Cancer (COSMIC) mutational profile associated with smoking, characterised by a transcriptional bias for C > A mutations.

Characteristic Lung cancer in people who have never smoked Lung cancer in people who have smoked
Median age at diagnosis 67 years21 70 years21
Proportion of new lung cancer
diagnoses worldwide
15%4,6 85%4,6
Absolute Incidence 0.16 per 1000 person years85 2.99 per 1000 person years
Lung Adenocarcinoma Risk
factors (% of total cases the risk
factor is thought to contribute to)
Targetable Genetic Alterations (70-80%)5,11,86
Second hand smoke (15%)
Occupational exposures (13%)
Radon (9%)
Air pollution (8%)
Household cooking and heating (4%)
Prior Radiotherapy (0.3% of those exposed)
Female sex (Up to 80% in some cohorts)
Smoking (100%)
Targetable Genetic Alterations (49.5%)11
Second hand smoke (15%)*
Occupational exposures (10%)*
Radon (9%)*
Air pollution (8%)*
Prior Radiotherapy (4% of those exposed)
Female sex (40%)
Screening recommendations No national screening programs other than Taiwan87 US: Annual low dose CT screen for those aged 50-80 with ≥20 pack year history and smoked within last 15 years44
Sex Difference 19% of females with lung cancer
9% of males with lung cancer21
81% of females with lung cancer
91% of males with lung cancer
Survival Benefit with
Immunotherapy
Hazard Ratio (HR)= 0.87, Confidence Interval (CI)
0.74-1.7474
HR 0.74, CI 0.69-0.81
Lung Adenocarcinoma Frequency
of oncogenic drivers (see Table 2)
78-92%11 49.5%88
Tumor Mutation Burden 0-3 mutations / Mb89 0-30 mutations / Mb89
Genomic signature None Smoking signature (SBS4)90
Histology Lung Adenocarcinoma (60-80%), Small Cell (8%), Squamous Cell (17%)91 Lung Adenocarcinoma (24%), Small Cell (17%), Squamous Cell (41%)91
*

Effects are additive in those who have smoked.

There are substantial ethnic, racial and geographic differences in the incidence of lung cancer in non-smokers. The age-adjusted incidence rate of lung cancer in non-smokers among female Asians in the US between 2000 and 2013 was 17.5 per 100,000 individuals (95% CI, 15.0-20.2) compared with 10.1 per 100,000 (95% CI 9.0-11.3) in non-Hispanic White females14.

The incidence of lung cancer in non-smokers worldwide is higher among women than men, however this may be confounded by the difference in the population at risk15,16. In Taiwan, up to 83% of non-smokers with lung cancer cases occur in women5,17,18. Smoking has historically has been higher among males than females and there are concerns about the accuracy of data on smoking status18. For example, a study of electronic healthcare records of 16,874 individuals in the US reported 80% had inaccuracies in their smoking history19. In addition, the US Surveillance, Epidemiology, and End Results (SEER) program, which aggregates survival data from lung cancers registered in the US, does not record smoking status20.

A pooled analysis of 7 Finnish cohorts demonstrated an age-standardized increase in lung cancer among female non-smokers (0.4 per 100,000 person-years in 1972 vs 6.2 per 100,000 person-years in 2015) compared with a stable rate of lung cancer among male non-smokers (6.5 per 100,000 person-years in 1972 vs 6.7 per 100,000 person years in 201512. A study that included 6 population-based cohorts (n=1,364,658 individuals, n=5379 incident lung cancer case) reported that the age-adjusted incident rate ranged was 14.4-20.8 per 100,000 person-years in non-smoking females and 4.8 -12.7 per 100,000 person-years in non-smoking males21.

Environmental Risk Factors

Several environmental exposures are associated with an increased risk of lung cancer in non-smokers (Table 1). Radon, a colorless gas that arises from the radioactive decay of uranium found in rocks and soil, is classified as a class I carcinogen by the International Agency for Research on Cancer (IARC)22. Up to 21,000 cases of lung cancer-related deaths in the US annually are due to radon, leading the US Centers for Disease Control and Prevention (CDC) to recommend that all homes be tested for radon, but radon testing is not federally mandated as a requirement for a home sale23. A meta-analysis containing estimates from 4 pooled studies (incorporating data from 24 individual case-control studies), 1 case-control study and 1 cohort study reported an adjusted excess relative risk of lung cancer of 0.15 (95% CI 0.06–0.25) per 100 becquerels per cubic meter (Bq/m3) increase in radon24. For homes that have radon levels at or above 4 picocuries per liter (pCi/L), or 150 Bq/m3, the Environmental Protection Agency (EPA) recommends reducing radon levels using measures such as active soil depressurization and improved ventilation. Because no level of radon exposure is considered safe, the EPA also advises homeowners to consider mitigation for levels between 2 and 4 pCi/L (75–150 Bq/m3)25.

In 2022, worldwide, approximately 200,000 cases of lung adenocarcinoma, the predominant histologic subtype of lung cancer, were attributed to ambient air pollution, estimated using a population-attributable fraction (PAF) model8. Specifically, particulate matter with a diameter of less than 2.5 micrometres (PM2.5), found in diesel exhaust or smoke from indoor cooking, can penetrate the alveoli and enter the bloodstream after inhalation. PM2.5, a known cause of lung cancer, promotes tumorigenesis via an influx of macrophages and release of interleukin-1β26. Approximately 99% of people worldwide live in areas that exceed World Health Organization (WHO) guidelines on PM2.5 (< 5μg m–3 annually). A recent multi-country study (6799 lung cancers, including 3615 lung cancer in non-smokers and 26807 controls without lung cancer) reported an association between PM2.5 levels and EGFR-driven lung cancer incidence, with the relative incidence rates increasing by 0.63-1.82 (per 100,000 population), per 1μg m–3 increase of PM2.527.

Other environmental particulates associated with lung cancer in non-smokers include asbestos and silica, which are considered Group 1 carcinogens by the IARC28. Second hand smoke exposure is also associated with an increased risk of lung cancer among non-smokers. A systematic review of a retrospective observational cohort and 5 case-control studies that included 622469 individuals reported a HR of 1.28 (95% CI: 1.10–1.48) for lung cancer in non-smokers29. A secondary analysis of the Global Burden of Disease Study data reported that second hand smoke was responsible for nearly 100,000 deaths worldwide in 202130. Prior radiotherapy to the chest is another risk factor for lung cancer; a retrospective observational study that found 1.74% of 613,746 patients (smoking status unavailable) who received radiotherapy during breast cancer treatment subsequently developed lung cancer31.

Familial and Genetic Risk

Data from a case-control study of 24,380 individuals with lung cancer and 23,399 controls indicate that individuals with a first-degree relative with lung cancer had an odds ratio of 1.51 (95% CI 1.39-1.63) for developing lung cancer compared to those without this family history32.

Large scale genome-wide association studies (GWAS) have identifed a pattern of low penetrance germline variants in regions such as 5p15.33 and 3q28 associated with increased risk of lung cancer in non-smokers. These variants involve multiple genes associated with cell cycle regulation, DNA damage, immune response and genomic stability33. Singular, highly penetrant germline variants in genes such as EGFR and YAP1 are rare, but have been reported in familial lung cancers from Western and Asian populations, respectively34,35. Clonal hematopoiesis (CH), the age-related expansion of a subpopulation of hematopoietic cells with acquired somatic mutations, has been associated with the incidence of many diseases, including lung cancer. In a CH case-control study of 104 cases of incident lung cancers and 343 age-, smoking status- and sex-matched controls, the odds ratio for risk of incident lung cancer was 1.43 (95% CI 1.06-1.94). Although limited by the relatively small number of non-smokers (11 cases and 35 controls) included in the analysis this effect may be independent of smoking history36.

Compared with current or former smokers with lung cancer, non-smokers with lung cancer have higher rates of somatic variants and gene rearrangements that activate genes that directly contribute to carcinogenesis and represent potential molecular targets. For example, in a study of 17,712 patients with lung cancer, somatic variants in the EGFR gene were identified in 40-60% of lung cancer tumor tissue in non-smokers compared with approximately 10% of smokers with lung cancer37. A study of 121 patients with lung cancer younger than age 40 years (73% non-smokers) indicated that 84% carried an actionable oncogenic variant38 (Table 2). Rearrangements in the ALK and ROS1 genes are found in 5-14% and 1-2% in tumors of patients with lung cancer, but with higher rates in non-smokers (OR = 3.57 95%CI=2.04-6.25 for RET fusions in non-smokers compared to smokers)39,40. Other actionable genomic alterations found more frequently in lung cancer biopsy specimens from non-smokers compared with current or former smokers include somatic mutations in HER2 (ERBB2), and gene fusions or rearrangements in RET, NTRK1/2/3 and NRG140,41. A study of non-smoking patients with lung adenocarcinomas (n=160) reported 78%-92% had clinically actionable driver mutations compared with 49.5% in ever smokers (n=299)11. A study of 188 lung adenocarcinomas that included 20 non-smokers found that non-smokers had a 10-fold lower tumor mutational burden, defined as the total number of DNA mutations in cancer cells, than tumors from smokers42.

Table 2. Driver mutation frequency and current availability of targeted FDA approved drugs.

Frequency data from whole exome sequencing profiling of 160 people with lung cancer. Smoking values are weighted averages among the current and former smoker categories11. NR = Not Reached meaning data too immature to calculate data from.

Oncogene Mutation
Frequency
in LCINS
Mutation
Frequency
in LCIPS
Targetable
alteration
FDA Approved
Drugs (Target
alteration)
Year of
First
approval
Median Overall
Survival (months)
Median
Progression Free
Survival (months)
Delivery route
EGFR 43% 11% 1) Exon 19 Deletion or Exon 21 L858R Afatanib (1,2) 2018 27.9 11 Oral
Amivantamab (3) 2024 NR 11.4 Intravenous
Dacomitinib (1,2) 2018 34.1 14.7 Oral
Erlotinib (1,2) 2016 84.2 10 Oral
Gefitinib (1,2) 2003 27 9.2 Oral
2) S768I, L861Q, and/or G719X Osimertinib (1,2) 2015 38.6 18.9 Oral
3) Exon 20
Insertion
Mutation
Amivantamab + chemotherapy 2024 38.9 20.6 Oral
ALK 12% 2% Rearrangement Alectinib 2015 NR 34.8 Oral
Brigatinib 2017 NR 16.7 Oral
Ceritinib 2014 51.3 16.6 Oral
Crizotinib 2015 NR 10.9 Oral
Ensartinib 2024 NR 25.8 Oral
Lorlatinib 2018 NR NR Oral
KRAS 9.10% 29% G12C Adagrasib 2022 NR 7.4 Oral
Sotorasib 2021 12.5 6.3 Oral
ROS1 3.22% 1.11% Rearrangement Entrectinib 2019 47.8 15.7 Oral
Crizotinib 2011 NR 10.9 Oral
Repotrectinib 2023 NR 35.7 Oral
Taletrectinib 2025 NR 45.6 Oral
ERBB2
(HER2)
2.22% 1.34% Mutation Fam-Trastuzumab
Deruxtecan
2022 17.8 8.2 Intravenous
RET 2% 0.5% Rearrangement Pralsetinib 2022 21.2 10.7 Oral
Selpercatinib 2022 NR 24.8 Oral
BRAF 1.83% 4.12% V600E Encorafenib/
Binimetinib
2018 33.6 14.9 Oral
Dabrafenib/ Trametinib 2013 25.9 11.1 Oral
MET 1.5% 2.1% Exon 14 Skipping Capmatinib 2020 20.8 10.8 Oral
Tepotinib 2021 29.7 15.9 Oral
NRG1 0.18% 0.08% Gene Fusion Zenocutuzumab 2024 Not reported 6.8 Intravenous
NTRK ~0.17%* Gene Fusion Entrectinib 2019 41.5 28 Oral
Repotrectinib 2024 NR NR Oral
*

= reliable data by smoking status not available.

Screening

To reduce lung cancer mortality, US Preventive Services Task Force (USPSTF) guidelines recommend low-dose CT scanning for individuals aged 50-80 years with a ≥ 20 pack-year history of smoking who currently smoke or quit within the past 15 years4346. However, the USPSTF does not currently recommend screening for lung cancer in non-smokers47. In contrast, Taiwan initiated a national lung cancer early detection program in 2022, offering biennial low-dose CT scans to non-smokers (aged 45-74 years for females and 50-74 years for males) with a family history of lung cancer48. This screening program was based on results from a study that performed CT screening for 12,011 asymptomatic Taiwanese non-smokers aged 55-75 years with risk factors, such as having a first degree relative with lung cancer, passive smoking exposure, or cooking without ventilation. This study reported that 2.7% with a family history of lung cancer were diagnosed with incident lung cancer, in contrast to 1.6% without a family history (p <0.0001), and 77.4% of lung cancers were diagnosed at Stage I48.

Clinical Presentation

Among individuals diagnosed with lung cancer, non-smokers have a similar clinical presentation as individuals with a history of tobacco consumption11. In a registry of 9876 patients diagnosed with lung cancer in Spain ,1177 (11.9%) were non smokers. Among non smokers, the most common symptoms at diagnosis were cough (34%), dyspnea (29%), pain (28%) and weight loss (19%). The percentage who were asymptomatic (31.5% of all patients) did not differ when stratified by smoking status49. In a retrospective series of 539 patients who underwent surgical resection for primary lung cancer at the University of California, Los Angeles, 345 (64%) were asymptomatic and had their tumors discovered incidentally on CT imaging and 143 (41%) were never-smokers50.

Assessment and Diagnosis

Imaging

The recommended imaging study for patients with chest symptoms concerning for lung cancer or with an abnormality detected on chest radiograph, is a contrast-enhanced chest CT to evaluate the primary lesion and assess for nodal involvement and metastases.

Brain imaging, preferably MRI, should be performed in all patients diagnosed with lung cancer, as 10-25% have brain metastases at diagnosis51. This rate may be higher in patients with genomic alterations such as EGFR variants and ALK or RET fusions with 52% of 50 patients presenting with brain metastases having an EGFR mutation in 1 study51. Fluorodeoxyglucose (FDG) Positron emission tomography (PET)-CT should be performed for patients being considered for potentially curative local therapy (such as radiotherapy or surgery) to determine extent of nodal involvement and to exclude metastatic disease52.

Patients with indeterminate pulmonary nodules (a focal distinct radiographic density surrounded by lung tissue) should undergo follow-up imaging based on the Fleischner Society guidelines (Supplementary Table 1). More than 95% of indeterminate pulmonary nodules are benign and the probability of malignancy is less than 1% for all nodules smaller than 6 mm (Supplementary Table 1)53.

Tissue Diagnosis and Molecular Testing

Biopsy of tumor tissue in the lung and/or lymph nodes performed with percutaneous biopsy or bronchoscopic biopsy, often guided by endobronchial ultrasound, confirms the diagnosis and may be useful for staging. Molecular testing should be performed for all non-smokers with lung cancer to aid decision making about use of targeted therapy and immunotherapy54. High throughput next generation sequencing (NGS) with DNA and RNA panels is currently recommended to identify genetic variants or rearrangements in genes including EGFR, ALK, ROS1 and RET for consideration of targeted therapies54. Characterization of circulating tumor DNA (ctDNA) or “liquid biopsy” is emerging as a test for detecting actionable genomic variants in plasma and may be particularly useful if limited tissue is available for molecular analyses. A study of 171 individuals with lung cancer reported that compared with patients with ctDNA-high status (n = 38), those with a pre-operative ctDNA-negative status (n = 18) had improved 5-year overall survival: 100%; 95% CI, 100%–100% vs 48.8%; 95% CI, 34.7%–68.7% (p=0.0024)55. Among patients who develop resistance to treatment, re-biopsy of lung cancer or liquid biopsy can reveal mechanisms of resistance such as alterations in genes encoding components of the PI3K-AKT MTOR pathway and alterations in RAS, which can guide subsequent treatment strategies56.

Staging

Most non-smokers with lung cancer are diagnosed at an advanced stage, typically with unresectable locally advanced disease (stage III) or distant metastases (stage IV). In a retrospective cohort study of 254 non-smoking patients with lung cancer, 62.9% were diagnosed with stage III or IV disease57. In another retrospective cohort study of 795 non-smoking patients, 43.4% were diagnosed with Stage IIIB-IV at diagnosis57. Early dectection of lung cancer in non-smokers may be difficult due to absence of routine screening in this population and the often non-specific symptoms such as cough and fatigue. However, staging at diagnosis can vary significantly by geography and by use of CT screening. For example, in the targeted low-dose CT screening trial for high-risk never-smokers in Taiwan, 246/257 (95.7%) were diagnosed with stage IA or IB lung cancer, demonstrating the potential effect of screening in shifting diagnosis toward earlier stages48.

Treatment

Treatment for lung cancer is guided by the stage, histology and molecular status of the tumor, patient performance status, comorbidities, and preferences, and does not differ by smoking status. Treatment options primarily include surgery, radiation therapy (RT), and systemic therapy often given in combination depending on disease stage and often involving multidisciplinary assessment and treatment by surgeons, medical and radiation oncologists, radiologists, pathologists, and nurses to deliver personalized treatment recommendations65,66.

Early-stage and locally advanced lung cancer

Surgical resection is preferred treatment for patients with anatomically resectable lung cancer (Stage I-III) who are medically eligible for surgery, with follow-up CT screening recommended every 6 months for 2-3 years, and then annually. For patients with stage IB-IIIA and resected EGFR variant positive or ALK rearranged NSCLC, targeted therapies are effective in both non-smokers and smokers. A phase 3 trial of 682 patients with stage IB-IIIA completely resected EGFR-variant NSCLC reported that among patients with stage II-IIIA disease randomized to 3 years of treatment with the EGFR Tyrosine Kinase Inhibitor (TKI) osimertinib 80 mg once-daily or placebo, 4-year disease-free survival (DFS) was 70% in the osimertinib group vs 29% in the placebo group, with DFS HR of 0.23 (95% CI, 0.18 to 0.30) at a median follow-up of 44.2 months (osimertinib) and 19.6 months (placebo)58.The Hazard Ratio (HR) for death or recurrence at 2 years was 0.23 [95% CI, 0.15-0.34] in non-smokers (n=488), and 0.10 [95% CI, 0.04-0.22] in those who had smoked (n=194)59. A follow-up study of this cohort of patients with stage IB to IIIA disease reported a 5-year overall survival (OS) of 85% in the osimertinib group vs 73% in the placebo group (overall HR for death, 0.49; 95% CI, 0.33 to 0.73; P<0.001)60. In a trial of 257 patients with stage IB, II or III resected ALK rearrangement positive NSCLC, after 2 years of adjuvant TKI (alectinib) or platinum-based chemotherapy, among patients with stage II or IIIA lung cancer, DFS at 2 years was 93.8% in the alectinib group and 63.0% in the chemotherapy group (hazard ratio for disease recurrence or death, 0.24; 95% CI, 0.13 to 0.45; P<0.001)61.

A trial that included 216 patients unresectable EGFR-mutated stage III NSCLC without progression during or after chemoradiotherapy reported improved median PFS with adjuvant osimertinib vs placebo (39.1 months vs 5.6 months), with a HR for disease progression or death of 0.22 (95% CI 0.14-0.34) in non-smokers (n=102), and 0.26 (95% CI 0.14-0.48) in smokers (n=41)62.

Immunotherapy with PD-1 or PD-L1 inhibitors are commonly prescribed neoadjuvant or adjuvant treatments for patients with early-stage lung cancer. However, their role for treating non-smokers with lung cancer is less clear due to a lower likelihood of response to immunotherapy, particularly in the context of EGFR gene variants or ALK gene rearrangements. A pooled analysis of 3 randomized trials that included treating individuals with nivolumab (n=292), pembrolizumab (n=691), or atezolimuab (n=144),vs docetaxel (n=776) reported that single-agent immunotherapy did not improve overall survival in EGFR-variant NSCLC, regardless of smoking status (HR=1.05, 95% CI: 0.70-1.55)63. Additionally, patients with EGFR gene variants and ALK gene rearrangements have often been excluded from peri-operative and adjuvant studies of immunotherapy for patients with lung cancer; few non-smokers have therefore been included in these studies64,65. In one trial, 797 patients with stage II, IIIA, or IIIB NSCLC were randomized to the PD-1 inhibitor pembrolizumab 200mg (n=397), or placebo (n=400) given with chemotherapy prior to surgery and for 12 months after surgery vs pre-operative chemotherapy alone.66 OS in all patients at 36-months was 71% (95% CI 66–76) in the pembrolizumab group and 64% (95% CI 58–69) in the placebo group (hazard ratio 0.72 [95% CI 0.56–0.93])66. The greatest benefit for perioperative pembrolizumab were seen in current smokers (HR for OS 0.59 (95% CI 0.38-0.93), with less benefit seen in non-smokers (HR 1.00 (95% CI 0.41-2.46)67.

Unresectable locally advanced or Metastatic Lung Cancer

For patients with advanced stage lung cancer, treatment typically consists of a combination of chemotherapy, immunotherapy or targeted therapies.Treatment with targeted agents have shown benefits over chemotherapy for indviduals who have EGFR variants, ALK rearrangements and RET rearrangements. A trial that randomized 556 patients with advanced stage EGFR-variant lung cancer to osimertinib or a first generation oral EGFR-tyrosine kinase inhibitor reported the hazard ratio for disease progression or death was significant in both those who smoked (n=199), HR 0.48 for osimertinib vs EGFR-tyrosine kinase inhibitor, (95% CI 0.34-0.68) and non-smokers (n=357) HR 0.45, (95% CI 0.34-0.59)67. Overall survival was also improved with firstline osimertinib (38.6 months vs 31.8 months, HR 0.80)68. A post hoc analysis of 5-year outcomes of the CROWN study, in which 296 patients with advanced ALK-positive non-small cell lung cancer were randomized to lorlatanib (an ALK inhibitor) or crizotinib (a TKI), reported improved outcomes with lorlatinib.69 In the lorlatinib group, median PFS was not reached (NR [95% CI, 64.3 to NR]) vs 9.1 months (95% CI, 7.4 to 10.9) in the crizotinib group ([HR], 0.19 [95% CI, 0.13 to 0.27]); 5-year PFS was 60% (95% CI, 51 to 68) in the lorlatinib group vs 8% (95% CI, 3 to 14) in the crizotinib group69. Other FDA-approved oncogene driven therapies such as repotrectinib or taletrectinib for ROS-1 positive tumors have demonstrated higher response rates and durations of response than seen historically with chemotherapy in both smokers and non-smokers with objective responses rates, defined as the percentage of patients with a partial or complete response to therapy, seen in 89% of individuals (n=152) for taletrectinib and 79% of individuals with repotrectinib (n=71)70,71 (Table 2).

For lung cancers without actionable genomic alterations that can be treated with targeted therapy, chemotherapy combined with immunotherapy can be considered72,73. However, single-agent immunotherapy with inhibitors of PD-1 or PD-L1 such as pembrolizumab have limited efficacy in non-smokers, particularly with EGFR or ALK alterations, and should not be used for non-smoking patients with these variants and unresectable locally advanced or metastatic lung cancer74.

Local therapy, such as radiotherapy or surgery may be used to treat sites of metastatic disease. For brain metastases, stereotactic radiosurgery can be used in preference to whole brain radiotherapy to minimize CNS toxicity including cognitive effects75. TKIs with excellent CNS penetration such as the EGFR inhibitor, osimertinib, or the ALK inhibitor, lorlatinib, may be used instead of radiotherapy for brain metastasis in many patients62. Stereotactic ablative radiotherapy (SABR) is recommended for oligometastatic disease in conjunction with systemic treatment due to its ability to deliver more targeted radiotherapy to a region76. In an open-label phase 2 trial of 99 patients with a controlled primary tumor and 1 to 5 metastases, median OS was 41 months (95% CI 26-not reached) in patients randomized to SABR vs 28 months (95% CI 19-33) with standard care77. However, adverse events (grade 2 or higher) occurred in 19/99 (29%) of patients in the SABR group vs 3(9%) of 33 in the standard care group (p=0·026), and treatment-related deaths occurred in 3/66 (4·5%) patients after SABR, compared with 0 in the standard care group.77

Prognosis

Lung cancer survival primarily depends on stage at diagnosis (5-year survival rate of 65% for Stage I vs < 10% for stage IV), performance status and presence of actionable genetic alterations for treatment78. A prospective cohort study of 5594 patients with NSCLC (61.8% lung adenocarcinoma) reported a median OS of 58.9 months (95% CI 51.9-67.4) for 795 non-smokers (55.8% Stage 1A -IIIA, 43.4% Stage IIIB-IV), a median OS of 51.2 months (95% CI 47.7-54.6) for 3308 former smokers (68.9% Stage 1A-IIIA, 30.1% Stage IIIB -IV) and a median OS of 34 months (95% CI 29.1-42.3) for 1491current smokers (63.6% Stage 1A-IIIA and 35.9% Stage IIIB-IV)57.

Significant improvements in PFS and OS among patients with lung cancer have occurred with use of targeted therapies directed at oncogenic drivers52,79,80. A population-based study using the Surveillance, Epidemiology, and End Results (SEER) data reported that 2-year lung cancer-specific survival improved from 26% for patients diagnosed in 2001 to 35% in 2014, a change largely attributable to the advent of targeted therapies. A French study reported that median OS among patients diagnosed with lung adenocarcinoma increased from 8.5 months in 2000 (n=1684) to 20.7 months in 2020 (n=5015), although no information on smoking status was provided81. The median OS for advanced EGFR variant positive lung cancer treated with EGFR inhibitors is currently 38.6 months and the median OS for patients with advanced ALK positive NSCLC treated with ALK inhibitors exceeds 5 years among smokers and non-smokers (Table 2)68,82.

Practical Considerations

Individuals with lung cancer may face stigma due to the disease's association with smoking, which can lead to feelings of isolation74. Patient education and psychosocial support for non-smokers with lung cancer enhances well-being and may improve outcomes through greater adherence to treatment as found in a systematic review of 18 cohorts73. A trial of 151 patients with metastatic lung cancer randomized to early palliative care (n=77 with 18 non-smokers) vs standard of care (n=74 with 16 non-smokers) found that early palliative care led to significant improvement in quality of life, reduced depressive symptoms and improved median survival (11.6 months vs 8. 9 months, P=0.02)83. Many support and advocacy groups exist, including those focusing on specific molecular subtypes of lung cancer (Table 3). Proactive follow-up of respiratory symptoms with CT imaging, regardless of a patient’s smoking history, could lead to earlier-stage lung-cancer diagnosis, a message echoed by awareness campaigns such as “See Through the Symptoms” and “All You Need Is Lungs9.

Table 3. List of lung cancer support groups available to patients, clinicians and scientists for advocacy, support and funding.

Support Group Role/Focus
ALK-Positive Support and advocacy for patients with ALK-positive lung cancer, focusing on research and education.
BRAF-Bombers Dedicated to supporting and educating patients with BRAF-mutated lung cancer.
EGFR-Resisters Support and advocacy group for patients with EGFR-mutated lung cancer, focusing on treatment resistance.
Exon20 Group Specialises in supporting patients with EGFR Exon 20 insertion mutations, advocating for research and treatments.
KRAS Kickers Supports patients with KRAS-mutated lung cancer, promoting research and sharing treatment information.
LUNGevity
Foundation
Offers support, education, and funding for lung cancer research, focusing on early detection and treatment.
MET crusaders Advocacy for those affected by lung cancer with a MET mutation
RET positive Supports patient with forms of RET positive cancers
ROS1ders Seeks to improve outcomes for those with ROS1 positive cancers

Future Directions

Ongoing trials, such as the Female Asian Nonsmoker Screening Study (FANSS) study in the US are evaluating potential benefits of screening for lung cancer among high-risk groups of non-smokers84. In addition, studies of biomarker-based early detection approaches in blood (such as multi-cancer early detection tests and proteomic approaches) are in progress to determine if lung cancers can be detected at an early and more curable stage.

Limitations

This review has several limitations. First, smoking history is often not included in many databases, cancer registries and clinical trials, making it difficult to accurately determine the incidence and prevalence of lung cancer in non-smokers and to evaluate treatment outcomes based on smoking status. Second, accurate quantification of environmental exposures, such as air pollution, is challenging. Third, the quality of the evidence was not formally evaluated. Fourth, some articles may have been missed.

Conclusions

Lung Cancer in non-smokers accounts for 15-20% of the lung cancer cases worldwide. Among patients with lung cancer, non-smokers are more likely to have genomic alterations such as EGFR variants or ALK gene rearrangements, and these patients have improved survival when treated with tyrosine kinase inhibitors, compared with chemotherapy.

Supplementary Material

Supplementary table

Box.

QUESTION: “How common is lung cancer in non-smokers and what are known risk factors?

ANSWER: “Worldwide, lung cancer in non-smokers accounts for 15-20% of all lung cancers. Exposure to passive smoking, radon, air pollution and asbestos, and having a first-degree relative with lung cancer are common risk factors for lung cancer in non-smokers.”

QUESTION: Among patients with lung cancer, which genetic variants are more common among non-smokers compared with former or current smokers?

ANSWER: Non-smokers with lung cancer have higher rates of genomic alterations such as EGFR variants or ALK gene rearrangements that directly contribute to carcinogenesis and represent potential molecular targets. Among patients with these genetic alterations, use of tyrosine kinase inhibitors (osimertinib and lorlatinib) is associated with improved survival.

Question: What are the recommendations about screening for lung cancer in non-smokers?

ANSWER: The US Preventive Services Task Force (USPSTF) does not recommend screening non-smokers for lung cancer with low-dose CT scans. In contrast, Taiwan initiated a national lung cancer early detection program in 2022 that offers biennial low-dose CT scans to non-smokers with a family history of lung cancer.

Acknowledgements

C.M. is supported by the Rosetrees Trust. T.P is supported by a UCL UKRI Centre for Doctoral Training in AI-enabled Healthcare studentship (EP/S021612/1), a Francis Crick Institute Idea to Innovation (i2i) grant and by the Ruth Strauss Foundation. C.S. is a Royal Society Napier Research Professor (RSRP\R\210001). His work is supported by the Francis Crick Institute, which receives its core funding from Cancer Research UK (CC2041), the UK Medical Research Council (CC2041) and the Wellcome Trust (CC2041). C.S. is funded by Cancer Research UK (TRACERx (C11496/A17786), PEACE (C416/A21999) and CRUK Cancer Immunotherapy Catalyst Network); Cancer Research UK Lung Cancer Centre of Excellence (C11496/A30025); the Rosetrees Trust, the Ruth Strauss Foundation, Future Dreams, Gustave Roussy Foundation, Butterfield and Stoneygate Trusts; NovoNordisk Foundation (ID16584); Royal Society Professorship Enhancement Award (RP/EA/180007 and RF\ERE\231118)); National Institute for Health Research (NIHR) University College London Hospitals Biomedical Research Centre; the Cancer Research UK-University College London Centre; Experimental Cancer Medicine Centre; the Breast Cancer Research Foundation (US) (BCRF-22-157); Cancer Research UK Early Detection an Diagnosis Primer Award (Grant EDDPMA-Nov21/100034); and The Mark Foundation for Cancer Research Aspire Award (Grant 21-029-ASP) and ASPIRE Phase II award (Grant 23-034-ASP). C.S. received an ERC Advanced Grant (PROTEUS) from the European Research Council under the European Union’s Horizon 2020 research and innovation program (grant agreement no. 835297). B.J.S is supported by an National Health and Medical Research Council Investigator Grant

References

  • 1.Lung Cancer Awareness Month 2024. [Accessed January 17, 2025]. https://www.iarc.who.int/featured-news/lung-cancer-awareness-month-2024 .
  • 2.American Lung Association. Tobacco trends brief: Overall tobacco trends brief. Published online 2024 https://www.lung.org/research/trends-in-lung-disease/tobacco-trends-brief/overall-smoking-trends.
  • 3.Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA: A Cancer Journal for Clinicians. 2024;74(1):12–49. doi: 10.3322/caac.21820. [DOI] [PubMed] [Google Scholar]
  • 4.Pelosof L, Ahn C, Gao A, et al. Proportion of Never-Smoker Non–Small Cell Lung Cancer Patients at Three Diverse Institutions. J Natl Cancer Inst. 2017;109(7):djw295. doi: 10.1093/jnci/djw295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.LoPiccolo J, Gusev A, Christiani DC, Jänne PA. Lung cancer in patients who have never smoked — an emerging disease. Nature Reviews Clinical Oncology. doi: 10.1038/s41571-023-00844-0. Published online 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Lung cancer. [Accessed January 16, 2025]. https://www.who.int/news-room/fact-sheets/detail/lung-cancer .
  • 7.Bray F, Laversanne M, Sung H, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74(3):229–263. doi: 10.3322/caac.21834. [DOI] [PubMed] [Google Scholar]
  • 8.Luo G, Zhang Y, Rumgay H, et al. Estimated worldwide variation and trends in incidence of lung cancer by histological subtype in 2022 and over time: a population-based study. The Lancet Respiratory Medicine. 2025 doi: 10.1016/S2213-2600(24)00428-4. 0(0) [DOI] [PubMed] [Google Scholar]
  • 9.All you need is lungs. ALL YOU NEED IS LUNGS. [Accessed January 17, 2025]. https://www.allyouneedislungs.org.uk .
  • 10.See through the symptoms. [Accessed January 17, 2025]. See through the symptoms https://www.seethroughthesymptoms.uk.
  • 11.Devarakonda S, Li Y, Martins Rodrigues F, et al. Genomic Profiling of Lung Adenocarcinoma in Never-Smokers. J Clin Oncol. 2021;39(33):3747–3758. doi: 10.1200/JCO.21.01691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Rissanen E, Heikkinen S, Seppä K, et al. Incidence trends and risk factors of lung cancer in never smokers: Pooled analyses of seven cohorts. Int J Cancer. 2021;149(12):2010–2019. doi: 10.1002/ijc.33765. [DOI] [PubMed] [Google Scholar]
  • 13.Kerpel-Fronius A, Tammemägi M, Cavic M, et al. Screening for Lung Cancer in Individuals Who Never Smoked: An International Association for the Study of Lung Cancer Early Detection and Screening Committee Report. J Thorac Oncol. 2022;17(1):56–66. doi: 10.1016/j.jtho.2021.07.031. [DOI] [PubMed] [Google Scholar]
  • 14.DeRouen MC, Canchola AJ, Thompson CA, et al. Incidence of Lung Cancer Among Never-Smoking Asian American, Native Hawaiian, and Pacific Islander Females. J Natl Cancer Inst. 2022;114(1):78–86. doi: 10.1093/jnci/djab143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Jemal A, Schafer EJ, Star J, et al. Lung cancer incidence rates in young women and men by state in the United States. Intl Journal of Cancer. 2025;156(3):499–504. doi: 10.1002/ijc.35188. [DOI] [PubMed] [Google Scholar]
  • 16.Wu Z, Tan F, Yang Z, et al. Sex disparity of lung cancer risk in non-smokers: a multicenter population-based prospective study based on China National Lung Cancer Screening Program. Chin Med J (Engl) 2022;135(11):1331–1339. doi: 10.1097/CM9.0000000000002161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Tseng CH, Tsuang BJ, Chiang CJ, et al. The Relationship Between Air Pollution and Lung Cancer in Nonsmokers in Taiwan. J Thorac Oncol. 2019;14(5):784–792. doi: 10.1016/j.jtho.2018.12.033. [DOI] [PubMed] [Google Scholar]
  • 18.Noronha V, Budukh A, Chaturvedi P, et al. Uniqueness of lung cancer in Southeast Asia. The Lancet Regional Health - Southeast Asia. 2024;27 doi: 10.1016/j.lansea.2024.100430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Kukhareva PV, Caverly TJ, Li H, et al. Inaccuracies in electronic health records smoking data and a potential approach to address resulting underestimation in determining lung cancer screening eligibility. J Am Med Inform Assoc. 2022;29(5):779–788. doi: 10.1093/jamia/ocac020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.SEER*Explorer: An interactive website for SEER cancer statistics. Surveillance Research Program, National Cancer Institute; [Accessed 10/11/2024]. [Internet] https://seer.cancer.gov/statistics-network/explorer/ [Google Scholar]
  • 21.Wakelee HA, Chang ET, Gomez SL, et al. Lung cancer incidence in never smokers. J Clin Oncol. 2007;25(5):472–478. doi: 10.1200/JCO.2006.07.2983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.US EPA O. What is EPA’s Action Level for Radon and What Does it Mean? 2019. Feb 19, [Accessed May 8, 2025]. https://www.epa.gov/radon/what-epas-action-level-radon-and-what-does-it-mean .
  • 23.US EPA O. Health Risk of Radon. 2024. Feb 27, [Accessed October 29, 2024]. https://www.epa.gov/radon/health-risk-radon .
  • 24.Cheng ES, Egger S, Hughes S, et al. Systematic review and meta-analysis of residential radon and lung cancer in never-smokers. Eur Respir Rev. 2021;30(159):200230. doi: 10.1183/16000617.0230-2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Bagnoli P. Consumer’s Guide to Radon Reduction - How to Fix Your Home.
  • 26.Huang F, Pan B, Wu J, Chen E, Chen L. Relationship between exposure to PM2.5 and lung cancer incidence and mortality: A meta-analysis. Oncotarget. 2017;8(26):43322–43331. doi: 10.18632/oncotarget.17313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hill W, Lim EL, Weeden CE, et al. Lung adenocarcinoma promotion by air pollutants. Nature. 2023;616(7955):159–167. doi: 10.1038/s41586-023-05874-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. [Accessed March 27, 2025];Agents Classified by the <em>IARC Monographs</em>. 1–137 https://monographs.iarc.who.int/agents-classified-by-the-iarc . [Google Scholar]
  • 29.Hori M, Tanaka H, Wakai K, Sasazuki S, Katanoda K. Secondhand smoke exposure and risk of lung cancer in Japan: a systematic review and meta-analysis of epidemiologic studies. Japanese Journal of Clinical Oncology. 2016;46(10):942–951. doi: 10.1093/jjco/hyw091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Yu Y, Aili A, Wu B, Zhao W, Yang M, Yuan X. Trends, differences, and future projections of lung cancer attributable to secondhand smoke across 204 countries and territories from 1990 to 2036. Tob Induc Dis. 2025 March;23:1–13. doi: 10.18332/tid/202228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wong LY, Kapula N, He H, et al. Risk of developing subsequent primary lung cancer after receiving radiation for breast cancer. JTCVS Open. 2023;16:919–928. doi: 10.1016/j.xjon.2023.10.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Coté ML, Liu M, Bonassi S, et al. Increased risk of lung cancer in individuals with a family history of the disease: A pooled analysis from the International Lung Cancer Consortium. Eur J Cancer. 2012;48(13):1957–1968. doi: 10.1016/j.ejca.2012.01.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.J B, Y H, Y L, et al. Cross-ancestry genome-wide meta-analysis of 61,047 cases and 947,237 controls identifies new susceptibility loci contributing to lung cancer. Nature genetics. 2022;54(8) doi: 10.1038/s41588-022-01115-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bell DW, Gore I, Okimoto RA, et al. Inherited susceptibility to lung cancer may be associated with the T790M drug resistance mutation in EGFR. Nat Genet. 2005;37(12):1315–1316. doi: 10.1038/ng1671. [DOI] [PubMed] [Google Scholar]
  • 35.Chen HY, Yu SL, Ho BC, et al. R331W Missense Mutation of Oncogene YAP1 Is a Germline Risk Allele for Lung Adenocarcinoma With Medical Actionability. JCO. 2015;33(20):2303–2310. doi: 10.1200/JCO.2014.59.3590. [DOI] [PubMed] [Google Scholar]
  • 36.Tian R, Wiley B, Liu J, et al. Clonal Hematopoiesis and Risk of Incident Lung Cancer. J Clin Oncol. 2023;41(7):1423–1433. doi: 10.1200/JCO.22.00857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Prabhakar CN. Epidermal growth factor receptor in non-small cell lung cancer. Transl Lung Cancer Res. 2015;4(2):110–118. doi: 10.3978/j.issn.2218-6751.2015.01.01. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Gitlitz BJ, Novello S, Vavalà T, et al. The Genomics of Young Lung Cancer: Comprehensive Tissue Genomic Analysis in Patients Under 40 With Lung Cancer. JTO Clin Res Rep. 2021;2(7):100194. doi: 10.1016/j.jtocrr.2021.100194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Wong DWS, Leung ELH, So KKT, et al. The EML4-ALK fusion gene is involved in various histologic types of lung cancers from nonsmokers with wild-type EGFR and KRAS. Cancer. 2009;115(8):1723–1733. doi: 10.1002/cncr.24181. [DOI] [PubMed] [Google Scholar]
  • 40.Lin C, Wang S, Xie W, Chang J, Gan Y. The RET fusion gene and its correlation with demographic and clinicopathological features of non-small cell lung cancer: a meta-analysis. Cancer Biol Ther. 2015;16(7):1019–1028. doi: 10.1080/15384047.2015.1046649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Park S, Ahn BC, Lim SW, et al. Characteristics and Outcome of ROS1-Positive Non-Small Cell Lung Cancer Patients in Routine Clinical Practice. J Thorac Oncol. 2018;13(9):1373–1382. doi: 10.1016/j.jtho.2018.05.026. [DOI] [PubMed] [Google Scholar]
  • 42.Ding L, Getz G, Wheeler DA, et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature. 2008;455(7216):1069–1075. doi: 10.1038/nature07423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Bhamani A, Creamer A, Verghese P, et al. Low-dose CT for lung cancer screening in a high-risk population (SUMMIT): a prospective, longitudinal cohort study. The Lancet Oncology. 2025 doi: 10.1016/S1470-2045(25)00082-8. 0(0) [DOI] [PubMed] [Google Scholar]
  • 44.Krist AH, Davidson KW, US Preventive Services Task Force et al. Screening for Lung Cancer: US Preventive Services Task Force Recommendation Statement. JAMA. 2021;325(10):962. doi: 10.1001/jama.2021.1117. [DOI] [PubMed] [Google Scholar]
  • 45.Aberle DR, Adams AM, National Lung Screening Trial Research Team et al. Reduced lung-cancer mortality with low-dose computed tomographic screening. N Engl J Med. 2011;365(5):395–409. doi: 10.1056/NEJMoa1102873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.de Koning HJ, van der Aalst CM, de Jong PA, et al. Reduced Lung-Cancer Mortality with Volume CT Screening in a Randomized Trial. New England Journal of Medicine. 2020;382(6):503–513. doi: 10.1056/NEJMoa1911793. [DOI] [PubMed] [Google Scholar]
  • 47.Khan S, Hatton N, Tough D, et al. Lung cancer in never smokers (LCINS): development of a UK national research strategy. BJC Rep. 2023;1:21. doi: 10.1038/s44276-023-00006-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Chang GC, Chiu CH, Yu CJ, et al. Low-dose CT screening among never-smokers with or without a family history of lung cancer in Taiwan: a prospective cohort study. The Lancet Respiratory Medicine. 2024;12(2):141–152. doi: 10.1016/S2213-2600(23)00338-7. [DOI] [PubMed] [Google Scholar]
  • 49.Ruano-Raviña A, Provencio M, Calvo De Juan V, et al. Lung cancer symptoms at diagnosis: results of a nationwide registry study. ESMO Open. 2020;5(6):e001021. doi: 10.1136/esmoopen-2020-001021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Zhou N, Deng J, Faltermeier C, et al. The majority of patients with resectable incidental lung cancers are ineligible for lung cancer screening. JTCVS Open. 2023;13:379–388. doi: 10.1016/j.xjon.2022.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Rangachari D, Yamaguchi N, VanderLaan PA, et al. Brain metastases in patients with EGFR-mutated or ALK-rearranged non-small-cell lung cancers. Lung Cancer. 2015;88(1):108–111. doi: 10.1016/j.lungcan.2015.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Riba, et al. NCCN Guidelines® Insights: Distress Management, Version 2.2023. [Accessed July 23, 2024];Journal of the National Comprehensive Cancer Network. 2023 21(5):2023. doi: 10.6004/jnccn.2023.0026. https://jnccn.org/view/journals/jnccn/21/5/article-p450.xml . [DOI] [PubMed] [Google Scholar]
  • 53.Mazzone PJ, Lam L. Evaluating the Patient With a Pulmonary Nodule: A Review. JAMA. 2022;327(3):264–273. doi: 10.1001/jama.2021.24287. [DOI] [PubMed] [Google Scholar]
  • 54.Riely GJ, Wood DE, Ettinger DS, et al. Non–Small Cell Lung Cancer, Version 4.2024, NCCN Clinical Practice Guidelines in Oncology. Journal of the National Comprehensive Cancer Network. 2024;22(4):249–274. doi: 10.6004/jnccn.2204.0023. [DOI] [PubMed] [Google Scholar]
  • 55.Black JRM, Bartha G, Abbott CW, et al. Ultrasensitive ctDNA detection for preoperative disease stratification in early-stage lung adenocarcinoma. Nat Med. 2025;31(1):70–76. doi: 10.1038/s41591-024-03216-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Jee J, Lebow ES, Yeh R, et al. Overall survival with circulating tumor DNA-guided therapy in advanced non-small-cell lung cancer. Nat Med. 2022;28(11):2353–2363. doi: 10.1038/s41591-022-02047-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Wang X, Romero-Gutierrez CW, Kothari J, Shafer A, Li Y, Christiani DC. Prediagnosis Smoking Cessation and Overall Survival Among Patients With Non-Small Cell Lung Cancer. JAMA Netw Open. 2023;6(5):e2311966. doi: 10.1001/jamanetworkopen.2023.11966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Herbst RS, Wu YL, John T, et al. Adjuvant Osimertinib for Resected EGFR-Mutated Stage IB-IIIA Non–Small-Cell Lung Cancer: Updated Results From the Phase III Randomized ADAURA Trial. J Clin Oncol. 2023;41(10):1830–1840. doi: 10.1200/JCO.22.02186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wu YL, Tsuboi M, He J, et al. Osimertinib in Resected EGFR-Mutated Non–Small-Cell Lung Cancer. New England Journal of Medicine. 2020;383(18):1711–1723. doi: 10.1056/NEJMoa2027071. [DOI] [PubMed] [Google Scholar]
  • 60.Tsuboi M, Herbst RS, John T, et al. Overall Survival with Osimertinib in Resected EGFR-Mutated NSCLC. New England Journal of Medicine. 2023;389(2):137–147. doi: 10.1056/NEJMoa2304594. [DOI] [PubMed] [Google Scholar]
  • 61.Wu YL, Dziadziuszko R, Ahn JS, et al. Alectinib in Resected ALK -Positive Non–Small-Cell Lung Cancer. N Engl J Med. 2024;390(14):1265–1276. doi: 10.1056/NEJMoa2310532. [DOI] [PubMed] [Google Scholar]
  • 62.Lu S, Kato T, Dong X, et al. Osimertinib after Chemoradiotherapy in Stage III EGFR-Mutated NSCLC. New England Journal of Medicine. 2024;391(7):585–597. doi: 10.1056/NEJMoa2402614. [DOI] [PubMed] [Google Scholar]
  • 63.Lee CK, Man J, Lord S, et al. Checkpoint Inhibitors in Metastatic EGFR-Mutated Non-Small Cell Lung Cancer-A Meta-Analysis. J Thorac Oncol. 2017;12(2):403–407. doi: 10.1016/j.jtho.2016.10.007. [DOI] [PubMed] [Google Scholar]
  • 64.Cascone T, Awad MM, Spicer JD, et al. Perioperative Nivolumab in Resectable Lung Cancer. N Engl J Med. 2024;390(19):1756–1769. doi: 10.1056/NEJMoa2311926. [DOI] [PubMed] [Google Scholar]
  • 65.Heymach JV, Harpole D, Mitsudomi T, et al. Perioperative Durvalumab for Resectable Non-Small-Cell Lung Cancer. N Engl J Med. 2023;389(18):1672–1684. doi: 10.1056/NEJMoa2304875. [DOI] [PubMed] [Google Scholar]
  • 66.Spicer JD, Garassino MC, Wakelee H, et al. Neoadjuvant pembrolizumab plus chemotherapy followed by adjuvant pembrolizumab compared with neoadjuvant chemotherapy alone in patients with early-stage non-small-cell lung cancer (KEYNOTE-671): a randomised, double-blind, placebo-controlled, phase 3 trial. The Lancet. 2024;404(10459):1240–1252. doi: 10.1016/S0140-6736(24)01756-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Soria JC, Ohe Y, Vansteenkiste J, et al. Osimertinib in Untreated EGFR-Mutated Advanced Non–Small-Cell Lung Cancer. New England Journal of Medicine. 2018;378(2):113–125. doi: 10.1056/NEJMoa1713137. [DOI] [PubMed] [Google Scholar]
  • 68.Ramalingam SS, Vansteenkiste J, Planchard D, et al. Overall Survival with Osimertinib in Untreated, EGFR-Mutated Advanced NSCLC. New England Journal of Medicine. 2020;382(1):41–50. doi: 10.1056/NEJMoa1913662. [DOI] [PubMed] [Google Scholar]
  • 69.Solomon BJ, Liu G, Felip E, et al. Lorlatinib Versus Crizotinib in Patients With Advanced ALK -Positive Non–Small Cell Lung Cancer: 5-Year Outcomes From the Phase III CROWN Study. JCO. :JCO.24.00581. doi: 10.1200/JCO.24.00581. Published online May 31, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Pérol M, Li W, Pennell NA, et al. Taletrectinib in ROS1+ Non-Small Cell Lung Cancer: TRUST. J Clin Oncol. 2025;43(16):1920–1929. doi: 10.1200/JCO-25-00275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Drilon A, Camidge DR, Lin JJ, et al. Repotrectinib in ROS1 Fusion–Positive Non–Small-Cell Lung Cancer. New England Journal of Medicine. 2024;390(2):118–131. doi: 10.1056/NEJMoa2302299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Mazieres J, Drilon A, Lusque A, et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Annals of Oncology. 2019;30(8):1321–1328. doi: 10.1093/annonc/mdz167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Vokes NI, Pan K, Le X. Efficacy of immunotherapy in oncogene-driven non-small-cell lung cancer. Therapeutic Advances in Medical Oncology. 2023;15:17588359231161409. doi: 10.1177/17588359231161409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Dai L, Jin B, Liu T, Chen J, Li G, Dang J. The effect of smoking status on efficacy of immune checkpoint inhibitors in metastatic non-small cell lung cancer: A systematic review and meta-analysis. eClinicalMedicine. 2021;38 doi: 10.1016/j.eclinm.2021.100990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Vogelbaum MA, Brown PD, Messersmith H, et al. Treatment for Brain Metastases: ASCO-SNO-ASTRO Guideline. JCO. 2022;40(5):492–516. doi: 10.1200/JCO.21.02314. [DOI] [PubMed] [Google Scholar]
  • 76.Iyengar P, All S, Berry MF, et al. Treatment of Oligometastatic Non-Small Cell Lung Cancer: An ASTRO/ESTRO Clinical Practice Guideline. Pract Radiat Oncol. 2023;13(5):393–412. doi: 10.1016/j.prro.2023.04.004. [DOI] [PubMed] [Google Scholar]
  • 77.Palma DA, Olson R, Harrow S, et al. Stereotactic ablative radiotherapy versus standard of care palliative treatment in patients with oligometastatic cancers (SABR-COMET): a randomised, phase 2, open-label trial. Lancet. 2019;393(10185):2051–2058. doi: 10.1016/S0140-6736(18)32487-5. [DOI] [PubMed] [Google Scholar]
  • 78.Cancer Survival in England, cancers diagnosed 2016 to 2020, followed up to 2021. NHS England Digital. [Accessed June 8, 2025]. https://digital.nhs.uk/data-and-information/publications/statistical/cancer-survival-in-england/cancers-diagnosed-2016-to-2020-followed-up-to-2021 .
  • 79.ASCO Guidelines: Thoracic Cancer | ASCO Publications. American Society of Clinical Oncology. [Accessed July 2, 2025]. https://ascopubs.org/topics/asco-guidelines/thoracic-cancer .
  • 80.ESMO Clinical Practice Guidelines: Lung and Chest Tumours | ESMO. [Accessed July 2, 2025]. https://www.esmo.org/guidelines/esmo-clinical-practice-guidelines-lung-and-chest-tumours .
  • 81.Debieuvre D, Falchero L, Molinier O, et al. Survival of Patients with Lung Adenocarcinoma Diagnosed in 2000, 2010, and 2020. NEJM Evid. 2025;4(7):EVIDoa2400443. doi: 10.1056/EVIDoa2400443. [DOI] [PubMed] [Google Scholar]
  • 82.Mok T, Camidge DR, Gadgeel SM, et al. Updated overall survival and final progression-free survival data for patients with treatment-naive advanced ALK-positive non-small-cell lung cancer in the ALEX study. Ann Oncol. 2020;31(8):1056–1064. doi: 10.1016/j.annonc.2020.04.478. [DOI] [PubMed] [Google Scholar]
  • 83.Temel JS, Greer JA, Muzikansky A, et al. Early Palliative Care for Patients with Metastatic Non–Small-Cell Lung Cancer. New England Journal of Medicine. 2010;363(8):733–742. doi: 10.1056/NEJMoa1000678. [DOI] [PubMed] [Google Scholar]
  • 84.Shum E, Li W, Ou SHI, Goldberg JD, Chachoua A, Wong KK. EP01.06-003 Female Asian Nonsmoker Screening Study (FANSS): Exploring Lung Cancer Screening in a High Risk Population. Journal of Thoracic Oncology. 2022;17(9, Supplement):S189. doi: 10.1016/j.jtho.2022.07.315. [DOI] [Google Scholar]
  • 85.Zhang P, Chen PL, Li ZH, et al. Association of smoking and polygenic risk with the incidence of lung cancer: a prospective cohort study. Br J Cancer. 2022;126(11):1637–1646. doi: 10.1038/s41416-022-01736-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Mack PC, Klein MI, Ayers KL, et al. Targeted Next-Generation Sequencing Reveals Exceptionally High Rates of Molecular Driver Mutations in Never-Smokers With Lung Adenocarcinoma. Oncologist. 2022;27(6):476–486. doi: 10.1093/oncolo/oyac035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Hardavella G, Frille A, Sreter KB, et al. Lung cancer screening: where do we stand? Breathe. 2024;20(2) doi: 10.1183/20734735.0190-2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Alexandrov LB, Ju YS, Haase K, et al. Mutational signatures associated with tobacco smoking in human cancer. Science. 2016;354(6312):618–622. doi: 10.1126/science.aag0299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Govindan R, Ding L, Griffith M, et al. Genomic landscape of non-small cell lung cancer in smokers and never-smokers. Cell. 2012;150(6):1121–1134. doi: 10.1016/j.cell.2012.08.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Australian Pancreatic Cancer Genome Initiative, ICGC Breast Cancer Consortium, ICGC MMML-Seq Consortium et al. Signatures of mutational processes in human cancer. Nature. 2013;500(7463):415–421. doi: 10.1038/nature12477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Pesch B, Kendzia B, Gustavsson P, et al. Cigarette smoking and lung cancer—relative risk estimates for the major histological types from a pooled analysis of case–control studies. Intl Journal of Cancer. 2012;131(5):1210–1219. doi: 10.1002/ijc.27339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.MacMahon H, Naidich DP, Goo JM, et al. Guidelines for Management of Incidental Pulmonary Nodules Detected on CT Images: From the Fleischner Society 2017. Radiology. 2017;284(1):228–243. doi: 10.1148/radiol.2017161659. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary table

RESOURCES