Abstract
Background
Globally and in the United States, lung cancer has been the most common cancer for the past several decades. In addition to the well-established geographical- and sex-specific differences in lung cancer incidence, mortality and survival, there is also growing evidence for racial and ethnic differences.
Methods
Based on available published data, we present a summary of the current knowledge and substantive findings related to racial and ethnic differences in lung cancer.
Results
Although this report is not a systematic review, we summarized the current knowledge and substantive findings related to racial and ethnic differences in lung cancer with a particular focus on lung cancer statistics(incidence, mortality, and survival), cigarette smoking, prevention and early detection, and the lung cancer genome. Finally, we summarize some the systems-level and provider-related issues that likely contribute to racial and ethnic-specific health disparities and provide some suggestions for future strategies that may reduce the disproportionate burden of lung cancer.
Conclusions
Although lung carcinogenesis is a multifactorial process driven by exogenous exposures (e.g., cigarette smoking), inherited genetic variations, and an accumulation of somatic genetic events, this multifactorial process appears to have racial and ethnic differences which in turn impacts the observed epidemiologic differences in incidence, mortality, and survival.
Keywords: Lung cancer, race, ethnicity, cancer epidemiology, precision medicine, health disparities, risk factors, screening and prevention
Burden of Lung Cancer Globally and in the United States
Incidence and Mortality
Globally, lung cancer has been the most common cancer for the past several decades1,2. Based on data from the GLOBOCAN project from the International Agency for Research on Cancer (IARC)2, in 2012 there were approximately 1.8 million new lung cancer diagnoses worldwide accounting for 12.9% of the global cancer burden. Among men, lung cancer remains the most common cancer diagnosis with approximately 1.2 million cases in 2012. The highest incidence rates occur in Central and Eastern Europe (53.5 per 100,000) and Eastern Asia (50.4 per 100,000), and the lowest rates in Central Africa (2.0 per 100,000) and Western Africa(1.7 per 100,000 respectively). Incidence rates are generally lower among women with approximately 583,000 new lung cancer diagnoses in 2012. The geographical variations for incidence and mortality differ between men and women, which is largely attributed to geographical differences in cigarette smoking between the two sexes2,3. The highest incidence rates in women are observed in North America (33.8 per 100,000) and Northern Europe (23.7per 100,000), while the lowest rates are found in Western Africa (1.1 per 100,000) and Central Africa (0.8 per 100,000)2.
In the United States, lung cancer is the second most common cancer in men after prostate cancer and the second most common cancer in women after breast cancer4. In 2016 an estimated 224,390 new cases of lung cancer are expected. The incidence rate has been declining in men over the past two decades, but has recently started to decrease in the mid-2000s among women. The incidence rates from 2008 to 2012 have decreased by 3.0% per year among men and by 1.9% per year among women. During this time period, the incidence rate among men was76.7 per 100,000 and 54.1 per 100,000for women. While lung cancer is the second most common cancer among both sexes, it is the leading cause of cancer-related death and accounts for more deaths than prostate, breast, colon, and pancreatic cancer combined. In 2016 an estimated 158,080 lung cancer-related deaths are expected to occur. From 2008 to 2012 the mortality rate among men was59.8 per 100,000 and 37.8 per 100,000 for women4.
Survival
Despite considerable improvements in patient survival over the last several decades for other cancer types including breast and prostate cancer, there have been little improvements in lung cancer survival. The lack of improvement in lung cancer survival is largely attributed to by the time a diagnosis is made, the cancer is often advanced stage and treatment options are limited. Because of the high fatality rate of this disease, there is relative lack of variability in survival in different world regions.2 In the United States, the five-year relative survival rate for all lung cancers is 17% (i.e., non-small cell lung cancer [NSCLC] and small cell lung cancer combined [SCLC])4.Among NSCLCs, the five-year relative survival rate is 21% for all stages combined4 and SCLCs have an overall 5-year survival rate of approximately 6%5. The overall prognosis for NSCLC remains poor and prognostic factors associated with poor survival include late stage diagnosis, current smoking status, advanced age, male sex, poor pulmonary function, presence of cardiovascular disease, non-squamous cell histology, and pneumonectomy6–10.Among SCLC patients, poor prognosis is associated with age greater than 70 year, male sex, relapsed disease, extensive-stage disease, weight loss greater than 10% of body weight at diagnosis, and poor performance status11–15.Despite the poor patient outcomes associated with a lung cancer diagnosis, the emergence of immunotherapy/immune checkpoint inhibitors have demonstrated durable long-term survival in some patients16,17. As such, these therapies may hold the key in improving lung cancer survival and possibly making lung cancer curable in the early-stage setting and/or a chronic disease for patients with metastatic disease18.
In addition to the well-established geographical- and sex-specific differences in lung cancer incidence, mortality, and survival, there are also racial and ethnic differences for which this review will present and discuss. Although this report is not a systematic review, we present a summary of the current knowledge and substantive findings related to racial and ethnic differences in lung cancer.
Racial and Ethnic Differences in Lung Cancer
Definition of Race and Ethnicity
Although the definitions of race and ethnic background are often applied inconsistently19, acommonly used definition describes race as a social construct that incorporates beliefs about language, history, and culture20 and forms the basis on which social identity, traditions, and politics are built19. In the United States, the race classification scheme used in the2000 U.S. Census, which is often used in biomedical research, includes five major groups: White, Black or African American, Asian, Native Hawaiian or other Pacific Islander, and American Indian or Alaska native21. Broadly, this classification scheme emphasizes the geographic region of origin of a person’s ancestry. Ethnicity background is a broader construct that takes into consideration cultural tradition, common history, religion, and often a shared genetic heritage20. In all cited studies, we use race and ethnicity as reported in the original study to describe the population.
Incidence and Mortality
Although race and ethnicity are complex social and cultural constructs, they are also often associated with socioeconomic status. Racial and ethnic differences in disease burden can reveal specific issues of a particular population or subpopulations. Racial and ethnic differences in lung cancer incidence, mortality, and survival are well-documented. The most comprehensive report22 in the United States are based on data from Surveillance, Epidemiology, and End Results Program (SEER) revealed that Blacks have a higher incidence and mortality rates than any other racial or ethnic group. A report from the Disease Control and Prevention (CDC)23 assessed potential racial and ethnic disparities and geographic differences in lung cancer incidence from 1998 to 2006 using data from SEER and CDC’s National Program of Cancer Registries (NPCR). In this report, the annual incidence of lung cancer was highest among Blacks (76.1 per 100,000), followed by Whites (69.7 per 100,000), American Indians/Alaska Natives (48.4 per 100,000), and Asian/Pacific Islanders (38.4 per 100,000). Hispanics had lower lung cancer incidence (37.3 per 100,000) than non-Hispanics (71.9 per 100,000). Regionally in the United States, the highest incidence was found in the South (76.0 per 100,000) and the lowest incidence was in the West (58.8 per 100,000). Among Whites, the highest lung cancer incidence was in the South (76.3 per 100,000). The highest incidence among Blacks (88.9 per 100,000), American Indians/Alaska Natives (64.2 per 100,000), and Hispanics (40.6 per 100,000) were in the Midwest, and the highest incidence among Asian/Pacific Islanders was in the West (42.5 per 100,000). The identification of geographical differences in incidence among racial/ethnic populations presents novel opportunities for targeted efforts in primary prevention and early detection23.
Survival
Published data has also demonstrated that the survival of lung cancer patients also differs significantly based on race and ethnicity. For examples, studies have reported Black patients are less likely to receive surgical resection than Whites patients24–26 which likely contributes to data that shows Black patients lower survival for early-stage NSCLC27. Using SEER data linked to Medicare claims data during the time period of 1995 to 1999, Shugarman et al26 reported that Black patients were 66% less likely to receive timely and appropriate treatment than Whites patients, and Black men were least likely to receive resection (22% for Black men versus 43.7% for White men). The authors also reported that Black patients were 34% less likely to receive timely surgery, chemotherapy, or radiation for stage III disease and were 51% less likely to receive chemotherapy in a timely fashion for stage IV disease relative to White patients. Additionally, Howington et al reported that Native American Indians and Native Alaskans have worse survival than non-Hispanic Whites28. With regard to potential ethnic difference, a study by Lin et al29 compared rates of stage-appropriate treatment among Blacks, Hispanics, and non minority patients and did not observe treatment disparities among Hispanic patients. However, the authors did find that Blacks were less likely than non-minorities to receive stage-appropriate treatment as other studies reported30–32.
Cigarette Smoking
Unequivocally, tobacco smoking is the most important and prevalent lung cancer risk factor33–35. Lung cancer is one of the first chronic diseases to be causally linked to tobacco smoking. Approximately 90% of lung cancer diagnoses in the United States are attributed to tobacco smoking8,9. Cigarette smoke contains more than 7,000 chemicals including over 60 established carcinogens and other toxicants associated with chronic diseases36. Although only around one in nine smokers develops lung cancer, the relative risk of lung cancer in long-term smokers is estimated to be between 10- and 30-fold higher than that of a lifetime never smoker8.
The percentage of cigarette smoking among adults in the United States has been a steady decline over the last several decades from 20.9% in 2005 to 16.8% in 201437. In 2014, prevalence of smoking was higher among males (18.8%) than females (14.8%). By racial and ethnic groups, smoking prevalence was highest among American Indian/Alaska Natives (29.2%) and multiracial adults (27.9%), and lowest among Asians (9.5%). For other racial and ethnic groups, smoking prevalence was 18.2% for Whites, 17.5% for Blacks, and 11.2% for Hispanics. As described above, the annual incidence is highest among Blacks compared to any other race, yet the prevalence of smoking among Blacks is lower than American Indian/Alaska Natives, multiracial adults, and Whites. Moreover, data from the Multiethnic Cohort Study38 reported significant differences in the association between cigarette smoking and the risk of lung cancer among five ethnic and racial groups. Interestingly, among those who smoked no more than 30 cigarettes per day, African Americans and Native Hawaiians had significantly greater risks of lung cancer than did the other groups. As such, these data provide further support for ethnic and racial differences, which could be attributed to innate genetic difference, in the smoking-associated risk of lung cancer.
Other established and putative risk factors lung cancer risk factors, that will not be reviewed here, include exposure to secondhand smoke (i.e., passive smoke), history of chronic obstructive pulmonary disease (COPD), family history of lung cancer, radon exposure, and occupational exposures such as asbestos, arsenic, diesel exhaust, and chromium8,39,40. There is conflicting evidence regarding the impact of exogenous hormones among women, diet, and BMI, on lung cancer risk40–47. Moreover, there are limited studies that have assessed potential racial and ethnic differences for these established and putative risk factors lung cancer risk factors.
Prevention and Early Detection
Until recently, no screening method has been shown to decrease mortality rates for NSCLC. The National Lung Screening Trial (NLST) randomized 53,452 former and current cigarette smokers who were 55 to 74 years old with a 30 or more pack-year smoking history into two arms, low-dose helical computed tomography (LDCT) and standard chest radiography, for three annual screens (a baseline screen and two follow-up screens approximately 12 months apart)48. After a median follow-up of 6.4 years, a 20% relative reduction in lung cancer mortality was observed for LDCT compared to standard chest radiography. Screen-detected incidence lung cancers (i.e., diagnosed on the follow-up screens)accounted for 58% of all LDCT-detected lung cancers, were 2.7-fold higher in the CT arm versus the chest radiography arm, were associated with a stage shift from late stage to more early stage lung cancers, and demonstrated improved 5-year survival compared to cancers diagnosed at the baseline prevalence screen48. An important benefit of early detection is the ability to detect more early stage lung cancers. A meta-analysis49 revealed that the rate of detection of stage I lung cancers was 70% with LDCT screening compared to 16% detected during routine care. Thus, screen-detected incidence lung cancers diagnosed as a result of LDCT screening have a better possibility of surgical cure and improved 5-year survival compared to prevalence- and routine care detected lung cancers that are diagnosed when patients develop symptoms from lung cancers that are associated with later stages8. Based on the findings from the NLST, the United States Preventive Services Task Force (USPSTF) issued a recommendation in December 2013 for annual screening for lung cancer with LDCT in adults aged 55 to 80 years who have a 30 pack-year smoking history and currently smoke or have quit within the past 15 years50.
Of the 53,452 participants in the NLST, 90.8% were White (N = 48,549), 4.4% were Black (N = 2,376), 1.7% were Asian (N = 895),2.0% were of other racial groups or reported more than one race or ethnic group (N = 1062), and 1.7% were Hispanic or Latino (N = 935). Although the original report48 did not provide ethnic or racial-specific results, a recent post hoc analysis by Tanner et al51 assessed racial differences in outcomes between Blacks and White individuals who participated in the NLST. Although demographics associated with improved lung cancer survival (i.e., higher education, former smoking status, and fewer comorbidities) were less commonly found among Blacks in the NLST, their analysis revealed that screening with LDCT reduced lung cancer mortality in all racial groups but more so in Black individuals (hazard ratio [HR] = 0.61 for Blacks vs. HR = 0.86 for Whites). The authors also noted that among all racial groups, current smokers had worse lung cancer-specific mortality; however, the risk was two-fold higher in Black current smokers compared to White current smokers. Additionally, all-cause mortality was 1.35-times higher in Blacks versus White, but black individuals screened with LDCT had a statistically significant reduction in all-cause mortality when compared with White individuals. As such, the authors note that screening for lung cancer is beneficial in Black individuals but that different strategies will be needed to achieve a significant reduction in lung cancer mortality in this population51. On-going disparities in access to screening and resolution of abnormal results underscore the importance of delivering lung cancer screening within the context of health care systems that offer screening and resolve abnormal findings quickly.
In a separate post hoc analysis of the NLST, Kumar et al52 examined racial differences in smoking behaviors among White and Black participants in the NLST who were current smokers at screening. They analyzed data from a follow-up survey on 24-hour and 7-day quit attempts,6-month continuous abstinence, and the use of smoking cessation programs and aids at 12 months after screening. The authors reported that Blacks were more likely than Whites to have 24-hour and 7-day quit attempts; however, these attempts did not translate to increased rates of 6-month continuous abstinence among Black smokers. Specifically, at 12 months after screening, Blacks were more likely to report a 24-hour (52.7% for Blacks vs. 41.2% for Whites, p < 0.01) or 7-day (33.6% for Blacks vs. 27.2% for Whites, p < 0.01) quit attempt. However, there were no statistically significant racial differences were found in 6-month continuous abstinence (5.6% for Blacks vs. 7.2% for Whites). In multivariable analyses, Black race was statistically significant predictive of a higher likelihood of a 24- and 7-day quit attempt; however, race was not associated with 6-month continuous abstinence. Only a positive screening result for lung cancer was significantly predictive of successful 6-month continuous abstinence. Presently, there is a gap in knowledge about the efficacy of LDCT lung cancer screening in other racial and ethnic groups.
Certainly the NLST demonstrated that screening with LDCT can mitigate the risk of lung cancer via early detection. Indeed, screening with LDCT appears to be second only to primary prevention (i.e., smoking prevention/cessation) for mitigating lung-cancer mortality—and the only remaining option for those who have already quit smoking. Lung cancer risk caused by smoking is reduced following smoking cessation, but it always remains elevated compared to never-smokers.
Lung Cancer Genomics
Fundamentally cancer is a genomic disorder and racial and ethnic differences in cancer incidence, mortality, and patient outcomes can be attributed, in part, to diversity in inherited genetic variations(germline) and an accumulation of somatic genetic events including mutations, rearrangements, or amplification that lead to uncontrolled cell proliferation, evasion of apoptosis, and eventually angiogenesis. Based on data53 from the Clinical Lung Cancer Genome Project (CLCGP) and Network Genomic Medicine (NGM), the most frequently mutated genes in lung cancer are TP53 (53.6%), KRAS (16.1%), STK11 (9.8%), EGFR (7.2%), KEAP1 (6.6%), and NFE2L2 (4.5%). Advances in tumor genomic profiling has resulted in a paradigm shift whereby lung cancers are characterized and classified by genetic alternations in oncogenes and tumor suppressor genes that are critical to tumor growth and survival and can be exploited with specific targeted agents54. Because most lung cancer harbor somatic mutations and/or alterations, targeted therapy, or precision medicine55, is important to improve outcomes of this disease. Unfortunately, to date there has been limited research on the impact of race and ethnicity in targeted therapies. Additionally, the impact and frequency of tumor mutations are not as well characterized in Black and of Hispanic/Latino patients to the same extent as they are in Asian and White populations. Nonetheless, in the following section we will highlight examples of how the frequency of mutations in oncogenes and tumor suppressor differ across racial and ethnic populations (Table 1). Though there is evidence that suggests racial and ethnicity differences in markers of susceptibility56,57, this review focused on somatic mutations because of their clinical implications and observed racial and ethnic differences, and because germline variations in lung cancer have yet to be deemed clinically actionable.
Table 1.
Gene | Frequency (%) |
---|---|
EGFR | |
By region | |
Western populations | ~10% |
Asian populations | ~35% |
By Country | |
Argentina | 19.3% |
Colombia | 24.8% |
Mexico | 31.2% |
Peru | 67.0% |
Costa Rica | 26.0 to 35.3% |
Panama | 24.8% |
KRAS | |
By region | |
Western populations | 18.0 to 26.0% |
Asian populations | 3.8 to 8.0% |
STK11/LKB1 | |
Western populations | 9.0 to 17.0% |
Asian populations | 3.0 to 7.0% |
BRAFa | |
Overall | 1.0 to 3.5% |
PIK3CAa | |
Overall | 1.0 to 3.0% |
ALK fusion | |
Overall | 3.0 to 7.0% |
By region | |
Western populations | 1.0 to 3.0% |
Asian populations | 2.3 to 6.7% |
No published reports assessing racial and ethnic differences
EGFR
Epidermal growth factor receptor (EGFR) is a transmembrane protein with cytoplasmic kinase activity that facilitates critical growth factor signaling from the extracellular milieu to the cell58. EGFR is expressed on the cell surface and EGFR tyrosine kinase inhibitors (TKIs), inhibitors that target the kinase domain of EGFR have been developed and are clinically active. TKIs are especially effective in patients whose tumors harbor activating mutations in the tyrosine kinase domain of the EGFR gene. Therefore, mutation testing is routinely performed to identify patients harboring targetable EGFR mutations, given that selection based only on clinical and pathologic characteristics is inadequate. Overall, approximately 10% of NSCLC patients in Western Countries and 35% in East Asia have a tumor that exhibit EGFR mutations59,60. The most frequent EGFR mutations occur in exons 18–21, which encodes a portion of the EGFR kinase domain; and, EGFR mutations are often heterozygous with the mutant allele also showing gene amplification61. Approximately 90% of these mutations are exon 19 deletions or exon 21 L858R point mutations62. Irrespective of ethnicity, EGFR mutations are more often found in females who are never smokers with adenocarcinoma histology59,60. However, EGFR mutations can also be found in other subsets of NSCLC, including in former and current smokers as well as in other lung cancer histological subtypes.
In frame deletions and insertions in exon 19 and point mutation L858R in exon 21 are the most common activating EGFR mutations, and their prevalence is significantly higher in Japanese, Korean, and Chinese patients than White patients from the United States or Europe63. Based on data from the Iressa Pan-Asia Study (IPASS) study, the frequency of activating EGFR mutations in Asian never smokers or light smokers with advanced-stage adenocarcinoma were 59.5%. In contrast, the frequency was only 15% in non-Asian patients based on data from an early phase III trials that performed retrospective EGFR mutation testing on archival samples64,65. The IGNITE study compared EGFR mutations in Asian and Russian patients with advanced NSCLC and reported that the frequency of EGFR mutations in adenocarcinoma to be 49% East Asian patients and 18% in Russian patients. The frequency of EGFR mutation non-adenocarcinoma lung cancers were 14% and 4%, respectively.66 There has also been published studies exploring whether the frequency of EGFR mutation differ by race and ethnicity. There have been several studies that compared the frequency of EGFR mutation between Black and White patients with NSCLC, but the results are conflicting. Specifically, Yang et al67 and Leidner et al68 found a lower frequency of EGFR mutation among Black patients, while Riely et al69, Cote et al70 and Reinersman et al71 did not find a statistically significant difference between the two groups.
Until recently, few studies have addressed the frequency of lung cancer mutations among Hispanics/Latinos. Arrieta et al72 analyzed 1,150 biopsies of NSCLC patients from Latin America (Argentina, Colombia, Peru, and Mexico) and found that frequency of EGFR mutations among all four countries was 32.5%. The frequency of mutations by country was 19.3% in Argentina, 24.8% in Colombia, 31.2% in Mexico, and 67% in Peru. Our group has previously speculated56 that the particularly high frequency of EGFR mutations in Peru is attributed to Asian migration. A study presented at the Latin American Lung Conference (LALCA) found the frequency of EGFR mutation ranging from 26% to 35.3% in Costa Rican patients73,74 and 24.8% in Panamanian patients75. However, in contrast, several studies have reported a lower frequency of EGFR mutations among Hispanics/Latinos while other studies found no statistically significant difference between Hispanics/Latinos and Whites (reviewed in56).
KRAS
The RAS gene family, which includes HRAS, NRAS, and KRAS, and encodes for membrane-bound guanosine-triphosphate-(GTP)-binding proteins that regulate cell growth, differentiation and apoptosis by interacting with mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase (PI3K) and signal transducer and activator of transcription (STAT) cascades76,77. The three RAS genes encode have 85% sequence identity proteins and the KRAS protein is a potent tumor initiator when aberrantly activated when a point mutation in the gene replaces an amino acid at codons 12 or 13 in Exon 2 or codon 61 in Exon 378. These mutations results in impaired GTPase activity and a constitutive activation of RAS signaling79. Mutations in KRAS gene occur frequently in NSCLC especially in adenocarcinoma (20% to 30%) and they are less common in squamous cell carcinoma (about 7%)80,81. Though mutationally activated KRAS tumors were originally identified in 198278, to date there are no successful treatment strategies that target these tumors81 and their impact on lung cancer survival and prognosis is unclear and remains controversial82,83. To date there have been over 50 published studies evaluating KRAS mutations on clinical outcome in lung cancer82,83 and the results from individual studies vary greatly and are inconsistent. A meta-analysis of 41 studies83 concluded that KRAS mutations are associated with a poor prognosis in patients with NSCLS, especially in patients with adenocarcinoma and early stage NSCLC. Published studies have suggested that the frequency of KRAS mutations differ by race and ethnicity. Specifically, KRAS mutations are less common in Asians compared White lung cancer patients84,85. Frequency of KRAS mutations ranged between 3.8% and 8% in studies comprised of Chinese NSCLC patients which is substantially lower compared to the White patients which range from 18% to 26%57,68,86. There are also very few studies that have assess KRAS mutational status among Black patients, and the published data are inconsistent. Three studies have reported no statistically significant difference in frequency of KRAS mutations between Whites and Blacks68,71,87; however, the frequency in Hispanics may be lower72,88.
STK11
Theserine/threonine kinase 11 (STK11) gene encodes a tumor suppressor located on chromosome 19p13.3 that encodes the serine/threonine protein kinase also known as liver kinase β1 (LKB1). The gene spans 23 kb and is made up of nine coding exons (exons 1 to 9) and a final non-coding exon (exon 10). The STK11 gene product regulates cellular energy metabolism and cell polarity by activating AMP-activated protein kinase (AMPK) and other members of the AMPK family89,90. As a multi-functional kinase, STK11 is involved in a broad spectrum of cellular activity including metabolism, polarity and epithelial-mesenchymal transition, cell cycle regulation, apoptosis, and autophagy.91 Germline mutations in STK11 were first identified in patients with Peutz-Jeghers syndrome92, a rare autosomal dominant disorder which is associated with an increased risk of gastrointestinal and other malignancies93. Studies have also reported that STK11 somatic mutations are quite common in NSCLCs with prevalence of inactivating mutations ranging up to 50% revealing an important role of STK11 in lung tumorigenesis91,94–100. The frequency of STK11 mutations in these studies range from 0.6% to 44.4%. Previously published data have reported that STK11 inactivating mutations occur in other lung cancer histology subtypes including 19% of squamous cell carcinomas, 14% of large cell carcinomas, and 25% of adenosquamous carcinomas91,97,98,101–103. Like KRAS and EGFR, there appear to be ethnic and racial differences in STK11 mutations. Studies in Asian populations including Japanese, Korean, and Chinese have reported much lower STK11 mutation rates compared to Whites, ranging from 3% to 7%103–106. This observation is similar to KRAS mutations in lung cancer, which frequently co-occur with STK11, where it has been noted that lung tumors in Western populations harbor a higher frequency of KRAS mutation compared to Asian populations. Asian populations have also been found to express an STK11 germline F354L polymorphism at approximately 10% frequency105. This allele has been called a nonfunctional polymorphism in lung cancer, but has also previously been reported to affect cell polarity maintenance in an AMPK-dependent manner.107 At present there are no published data on the frequency of STK11 mutations among Blacks and Hispanics/Latinos.
BRAF
BRAF kinase is a proto-oncogene that belongs to a family of serine-threonine protein kinases that includes ARAF, BRAF, and CRAF (RAF1). Mutant BRAF has been implicated in the pathogenesis of several cancers and the most commonly identified BRAF mutation is V600E accounting for 90% of BRAF mutations in melanoma. By contrast, in NSCLC the frequency of BRAF gene mutations ranges from 1% to 3.5%108–111. There have been no published reports assessing racial and ethnic differences of BRAF mutations in lung cancer patients, likely attributed to its rarity in lung cancer.
PIK3CA
The PIK3CA gene belongs to a family of lipid kinases involved in many cellular processes, including cell growth, proliferation, differentiation, motility, and survival. PIK3CA was found to be mutated in over 30% of colorectal cancers112. By contrast, mutations in PIK3CA have been found only in 1% to 3% of all NSCLCs112,113. As noted with BRAF, there have been no published reports assessing racial and ethnic of PIK3CA mutations in lung cancer which again can attributed its rarity in this disease.
ALK
Anaplastic lymphoma kinase (ALK)is a tyrosine kinase receptor that is can be abnormally expressed by forming a fusion gene with one of several other genes, by gaining additional gene copies, or by somatic mutations. The EML4-ALK fusion was first documented in 2007 in NSCLC114 as a novel potential oncogenic driver mutant kinase. Studies have reported that approximately 3% to 7% of all lung tumors harbor ALK fusions114,115 and EML4-ALK fusions are usually found in light or never smokers of patients diagnosed at a younger age116–118. EML4-ALK rearrangements have also reported to differ across different racial groups. Studies have reported that the frequency of translocation to between 2.3% to 6.7% among Asians 119–121. In contrast, EML4-ALK rearrangement was found to be much lower in Whites ranging 1% to 3%114,117,122. One study analyzed a cohort of NSCLC patient samples collected from Italy and Spain and found that 7.5% of the patients expressed EML4-ALK transcripts which is more similar to Asians that Whites123.
Summary of Lung Cancer Genomics
In the previous section we focused on specific oncogenes and tumor suppressor genes where published studies have assessed potential racial and ethnic differences. Although other genes have also been studied, including MET, PI3KCA, PTEN, andROS1, at present the published limited data suggests there are no differences56
The genomic diversity of oncogenes and tumor suppressor genes across racial and ethnic groups poses unique but important challenges for therapeutic opportunities in providing personalized medicine. Molecular genomic profiling for specific alterations is necessary to identify which patients will benefit from targeted therapeutic intervention. As such, identifying novel, but actionable target able mutations that may be exclusive to specific racial and ethnic groups is critical to ensure these populations benefit from such therapies.
Conclusion
Although lung carcinogenesis is a multifactorial process driven by exogenous exposures (e.g., cigarette smoking), inherited genetic variations, and an accumulation of somatic genetic events, this multifactorial process also appears to have racial and ethnic differences. In this review, we highlighted examples of racial and ethnic differences in smoking and lung cancer genome which in turn may play a role in the observed epidemiologic differences in incidence, mortality, and survival. However, many of the observed racial and ethnic differences are attributed to underlying racial and ethnic disparities in lung cancer are not fully understood. Broadly speaking, health disparities, can also occur at the systems-level issues, such as access to care, insurance, and hospital-level factors24,32,124. Other potential sources of racial and ethnic disparities are derived from provider-related factors such as limited cultural sensitivity, stereotyping, and poor patient–physician communication may also be involved125. Studies have shown that even after controlling for these factors126,127, treatment disparities persist, suggesting that patient-specific factors may also contribute to lung cancer differences.
The lung cancer epidemic peaked in the 20th century in the United States and most Western Nations2, but began to decline by the century’s end and continues to decline today due to successful tobacco-control efforts. In the United States smoking rates have steadily declined since the 1960s128; however, today nearly 17% of adults still continue to smoke cigarettes129. Even after smoking cessation is successfully accomplished, former smokers remain at significant risk of developing lung cancer. Certainly the NLST demonstrated that screening with LDCT can mitigate the risk of lung cancer via early detection. As such, screening with LDCT appears to be second only to primary prevention (i.e., smoking prevention/cessation) for mitigating lung-cancer mortality—and the only remaining option for those who have already quit smoking. Lung cancer risk caused by smoking is reduced following smoking cessation, but it always remains elevated compared to never-smokers. As such, lung cancer will remain a major global public health burden for the 21st century.
Although effective smoking cessation programs and early detection will reduce the overall lung cancer burden, improvements to the access of these modalities and affordability of health care is particularly important to address lung cancer-related racial and ethnic disparities. As such, identifying and addressing cultural factors may also help improve implementing racially/ethnicity-sensitive population-based interventions, such as surveillance and screening programs and smoking cessation interventions, and lung cancer treatment rates among minorities. Finally, identifying and eliminating regional and racial/ethnic differences in lung cancer may contribute to more effective preventive and treatment strategies that will ultimately reduce the disproportionate burden of lung cancer in the United States126.
Acknowledgments
None
Funding: This work has been supported in part by the James & Esther King Biomedical Research Program (5JK06) and by a Cancer Center Support Grant (CCSG) at the H. Lee Moffitt Cancer Center and Research Institute; an NCI designated Comprehensive Cancer Center (P30-CA76292).
Footnotes
Conflict of interest: None
References
- 1.American Cancer Society. Global Cancer Facts & Figures. 2nd. Atlanta: American Cancer Society; 2011. [Google Scholar]
- 2.International Agency for Research on Cancer. GLOBOCAN 2012: Estimated Cancer Incidence, Morality, and Prevalence Worldwide in 2012. http://globocan.iarc.fr/Pages/fact_sheets_cancer.aspx.
- 3.Islami F, Torre LA, Jemal A. Global trends of lung cancer mortality and smoking prevalence. Transl Lung Cancer Res. 2015;4(4):327–338. doi: 10.3978/j.issn.2218-6751.2015.08.04. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.American Cancer Society. Cancer Facts & Figures 2016. Atlanta: American Cancer Society; 2016. [Google Scholar]
- 5.Harris K, Khachaturova I, Azab B, Maniatis T, Murukutla S, Chalhoub M, Hatoum H, Kilkenny T, Elsayegh D, Maroun R, Alkaied H. Small cell lung cancer doubling time and its effect on clinical presentation: a concise review. Clin Med Insights Oncol. 2012;6:199–203. doi: 10.4137/CMO.S9633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Schabath MB, Thompson ZJ, Gray JE. Temporal trends in demographics and overall survival of non-small-cell lung cancer patients at Moffitt Cancer Center from 1986 to 2008. Cancer Control. 2014;21(1):51–56. doi: 10.1177/107327481402100107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Kachroo S, Tong L, Spitz MR, Xing Y, Merriman K, Zhu DK, Fueger J, Amos CI, Etzel CJ. Trends in prevalence of prognostic factors and survival in lung cancer patients from 1985 to 2004 at a tertiary care center. Cancer Detect Prev. 2008;32(2):101–108. doi: 10.1016/j.cdp.2008.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Dela Cruz CS, Tanoue LT, Matthay RA. Lung cancer: epidemiology, etiology, and prevention. Clin Chest Med. 2011;32(4):605–644. doi: 10.1016/j.ccm.2011.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Birim O, Kappetein AP, van Klaveren RJ, Bogers AJ. Prognostic factors in non-small cell lung cancer surgery. Eur J Surg Oncol. 2006;32(1):12–23. doi: 10.1016/j.ejso.2005.10.001. [DOI] [PubMed] [Google Scholar]
- 10.Ries LAG. Cancer survival among adults : U.S. SEER program, 1988–2001, patient and tumor characteristics. Bethesda, MD: U.S. Department of Health and Human Services, National Institutes of Health, National Cancer Institute; 2007. [Google Scholar]
- 11.Paesmans M, Sculier JP, Lecomte J, Thiriaux J, Libert P, Sergysels R, Bureau G, Dabouis G, Van Cutsem O, Mommen P, Ninane V, Klastersky J. Prognostic factors for patients with small cell lung carcinoma: analysis of a series of 763 patients included in 4 consecutive prospective trials with a minimum follow-up of 5 years. Cancer. 2000;89(3):523–533. doi: 10.1002/1097-0142(20000801)89:3<523::aid-cncr7>3.0.co;2-6. [DOI] [PubMed] [Google Scholar]
- 12.Spiegelman D, Maurer LH, Ware JH, Perry MC, Chahinian AP, Comis R, Eaton W, Zimmer B, Green M. Prognostic factors in small-cell carcinoma of the lung: an analysis of 1,521 patients. J Clin Oncol. 1989;7(3):344–354. doi: 10.1200/JCO.1989.7.3.344. [DOI] [PubMed] [Google Scholar]
- 13.Albain KS, Crowley JJ, LeBlanc M, Livingston RB. Determinants of improved outcome in small-cell lung cancer: an analysis of the 2,580-patient Southwest Oncology Group data base. J Clin Oncol. 1990;8(9):1563–1574. doi: 10.1200/JCO.1990.8.9.1563. [DOI] [PubMed] [Google Scholar]
- 14.Osterlind K, Andersen PK. Prognostic factors in small cell lung cancer: multivariate model based on 778 patients treated with chemotherapy with or without irradiation. Cancer Res. 1986;46(8):4189–4194. [PubMed] [Google Scholar]
- 15.Schabath MB, Nguyen A, Wilson P, Sommerer KR, Thompson ZJ, Chiappori AA. Temporal trends from 1986 to 2008 in overall survival of small cell lung cancer patients. Lung Cancer. 2014;86(1):14–21. doi: 10.1016/j.lungcan.2014.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Santarpia M, Giovannetti E, Rolfo C, Karachaliou N, Gonzalez-Cao M, Altavilla G, Rosell R. Recent developments in the use of immunotherapy in non-small cell lung cancer. Expert Rev Respir Med. 2016 doi: 10.1080/17476348.2016.1182866. [DOI] [PubMed] [Google Scholar]
- 17.Tchekmedyian N, Gray JE, Creelan BC, Chiappori AA, Beg AA, Soliman H, Perez BA, Antonia SJ. Propelling Immunotherapy Combinations Into the Clinic. Oncology (Williston Park) 2015;29(12) [PubMed] [Google Scholar]
- 18.Brahmer JR. Harnessing the immune system for the treatment of non-small-cell lung cancer. J Clin Oncol. 2013;31(8):1021–1028. doi: 10.1200/JCO.2012.45.8703. [DOI] [PubMed] [Google Scholar]
- 19.Sankar P, Cho MK. Genetics. Toward a new vocabulary of human genetic variation. Science. 2002;298(5597):1337–1338. doi: 10.1126/science.1074447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Witzig R. The medicalization of race: scientific legitimization of a flawed social construct. Ann Intern Med. 1996;125(8):675–679. doi: 10.7326/0003-4819-125-8-199610150-00008. [DOI] [PubMed] [Google Scholar]
- 21.Grieco EM, Cassidy RC. Overview of race and Hispanic origin : 2000. Washington, D.C.: U.S. Dept. of Commerce, Economics and Statistics Administration, U.S. Census Bureau; 2001. United States. Bureau of the Census. [Google Scholar]
- 22.U.S. National Institutes of Health. National Cancer Institute. SEER Cancer Statistics Review. 1975–2012 [Google Scholar]
- 23.Centers for Disease Control and Prevention. Racial/Ethnic Disparities and Geographic Differences in Lung Cancer Incidence --- 38 States and the District of Columbia, 1998--2006. Morbidity and Mortality Weekly Report. 2010;59(44):1434–1438. [PubMed] [Google Scholar]
- 24.Farjah F, Wood DE, Yanez ND, 3rd, Vaughan TL, Symons RG, Krishnadasan B, Flum DR. Racial disparities among patients with lung cancer who were recommended operative therapy. Arch Surg. 2009;144(1):14–18. doi: 10.1001/archsurg.2008.519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Margolis ML, Christie JD, Silvestri GA, Kaiser L, Santiago S, Hansen-Flaschen J. Racial differences pertaining to a belief about lung cancer surgery: results of a multicenter survey. Ann Intern Med. 2003;139(7):558–563. doi: 10.7326/0003-4819-139-7-200310070-00007. [DOI] [PubMed] [Google Scholar]
- 26.Shugarman LR, Mack K, Sorbero ME, Tian H, Jain AK, Ashwood JS, Asch SM. Race and sex differences in the receipt of timely and appropriate lung cancer treatment. Med Care. 2009;47(7):774–781. doi: 10.1097/MLR.0b013e3181a393fe. [DOI] [PubMed] [Google Scholar]
- 27.Zullig LL, Carpenter WR, Provenzale DT, Weinberger M, Reeve BB, Williams CD, Jackson GL. The association of race with timeliness of care and survival among Veterans Affairs health care system patients with late-stage non-small cell lung cancer. Cancer Manag Res. 2013;5:157–163. doi: 10.2147/CMAR.S46688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Howington JA, Blum MG, Chang AC, Balekian AA, Murthy SC. Treatment of stage I and II non-small cell lung cancer: Diagnosis and management of lung cancer, 3rd ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2013;143(5 Suppl):e278S–e313S. doi: 10.1378/chest.12-2359. [DOI] [PubMed] [Google Scholar]
- 29.Lin JJ, Mhango G, Wall MM, Lurslurchachai L, Bond KT, Nelson JE, Berman AR, Salazar-Schicchi J, Powell C, Keller SM, Halm EA, Leventhal H, Wisnivesky JP. Cultural factors associated with racial disparities in lung cancer care. Ann Am Thorac Soc. 2014;11(4):489–495. doi: 10.1513/AnnalsATS.201402-055OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Hardy D, Liu CC, Xia R, Cormier JN, Chan W, White A, Burau K, Du XL. Racial disparities and treatment trends in a large cohort of elderly black and white patients with nonsmall cell lung cancer. Cancer. 2009;115(10):2199–2211. doi: 10.1002/cncr.24248. [DOI] [PubMed] [Google Scholar]
- 31.Hardy D, Xia R, Liu CC, Cormier JN, Nurgalieva Z, Du XL. Racial disparities and survival for nonsmall-cell lung cancer in a large cohort of black and white elderly patients. Cancer. 2009;115(20):4807–4818. doi: 10.1002/cncr.24521. [DOI] [PubMed] [Google Scholar]
- 32.Steele CB, Pisu M, Richardson LC. Urban/rural patterns in receipt of treatment for non-small cell lung cancer among black and white Medicare beneficiaries, 2000–2003. J Natl Med Assoc. 2011;103(8):711–718. doi: 10.1016/s0027-9684(15)30410-7. [DOI] [PubMed] [Google Scholar]
- 33.U.S. Department of Health and Human Services. The Health Consequences of Smoking: 50 Years of Progress. A Report of the Surgeon General. Atlanta, GA: U.S. Department of Health and Human Services, Centers for Disease Control and Prevention, National Center for Chronic Disease Prevention and Health Promotion, Office on Smoking and Health; 2014. [Google Scholar]
- 34.Carter BD, Freedman ND, Jacobs EJ. Smoking and mortality--beyond established causes. N Engl J Med. 2015;372(22):2170. doi: 10.1056/NEJMc1503675. [DOI] [PubMed] [Google Scholar]
- 35.Pesch B, Kendzia B, Gustavsson P, Jockel KH, Johnen G, Pohlabeln H, Olsson A, Ahrens W, Gross IM, Bruske I, Wichmann HE, Merletti F, Richiardi L, Simonato L, Fortes C, Siemiatycki J, Parent ME, Consonni D, Landi MT, Caporaso N, Zaridze D, Cassidy A, Szeszenia-Dabrowska N, Rudnai P, Lissowska J, Stucker I, Fabianova E, Dumitru RS, Bencko V, Foretova L, Janout V, Rudin CM, Brennan P, Boffetta P, Straif K, Bruning T. Cigarette smoking and lung cancer--relative risk estimates for the major histological types from a pooled analysis of case-control studies. Int J Cancer. 2012;131(5):1210–1219. doi: 10.1002/ijc.27339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.U.S. Department of Health and Human Services. How Tobacco Smoke Causes Disease: The Biology and Behavioral Basis for Smoking-Attributable Disease: A Report of the Surgeon General. Atlanta, GA: U.S. Department of Health and Human Services, Centers for Disease Control and Prevention, National Center for Chronic Disease Prevention and Health Promotion, Office on Smoking and Health; 2010. [Google Scholar]
- 37.Centers for Disease Control and Prevention. Current Cigarette Smoking Among Adults—United States, 2005–2014. Morbidity and Mortality Weekly Report. 2015;64(44):1233–1240. doi: 10.15585/mmwr.mm6444a2. [DOI] [PubMed] [Google Scholar]
- 38.Haiman CA, Stram DO, Wilkens LR, Pike MC, Kolonel LN, Henderson BE, Le Marchand L. Ethnic and racial differences in the smoking-related risk of lung cancer. N Engl J Med. 2006;354(4):333–342. doi: 10.1056/NEJMoa033250. [DOI] [PubMed] [Google Scholar]
- 39.Driscoll T, Nelson DI, Steenland K, Leigh J, Concha-Barrientos M, Fingerhut M, Pruss-Ustun A. The global burden of disease due to occupational carcinogens. Am J Ind Med. 2005;48(6):419–431. doi: 10.1002/ajim.20209. [DOI] [PubMed] [Google Scholar]
- 40.Alberg AJ, Brock MV, Ford JG, Samet JM, Spivack SD. Epidemiology of lung cancer: Diagnosis and management of lung cancer, 3rd ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2013;143(5 Suppl):e1S–e29S. doi: 10.1378/chest.12-2345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Brinton LA, Gierach GL, Andaya A, Park Y, Schatzkin A, Hollenbeck AR, Spitz MR. Reproductive and hormonal factors and lung cancer risk in the NIH-AARP Diet and Health Study cohort. Cancer Epidemiol Biomarkers Prev. 2011;20(5):900–911. doi: 10.1158/1055-9965.EPI-10-1325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Yao Y, Gu X, Zhu J, Yuan D, Song Y. Hormone replacement therapy in females can decrease the risk of lung cancer: a meta-analysis. PLoS One. 2013;8(8):e71236. doi: 10.1371/journal.pone.0071236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Yang Y, Dong J, Sun K, Zhao L, Zhao F, Wang L, Jiao Y. Obesity and incidence of lung cancer: a meta-analysis. Int J Cancer. 2013;132(5):1162–1169. doi: 10.1002/ijc.27719. [DOI] [PubMed] [Google Scholar]
- 44.Smith L, Brinton LA, Spitz MR, Lam TK, Park Y, Hollenbeck AR, Freedman ND, Gierach GL. Body mass index and risk of lung cancer among never, former, and current smokers. J Natl Cancer Inst. 2012;104(10):778–789. doi: 10.1093/jnci/djs179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Wu SH, Liu Z. Soy food consumption and lung cancer risk: a meta-analysis using a common measure across studies. Nutr Cancer. 2013;65(5):625–632. doi: 10.1080/01635581.2013.795983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Wu QJ, Xie L, Zheng W, Vogtmann E, Li HL, Yang G, Ji BT, Gao YT, Shu XO, Xiang YB. Cruciferous vegetables consumption and the risk of female lung cancer: a prospective study and a meta-analysis. Ann Oncol. 2013;24(7):1918–1924. doi: 10.1093/annonc/mdt119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Song J, Su H, Wang BL, Zhou YY, Guo LL. Fish consumption and lung cancer risk: systematic review and meta-analysis. Nutr Cancer. 2014;66(4):539–549. doi: 10.1080/01635581.2014.894102. [DOI] [PubMed] [Google Scholar]
- 48.Aberle DR, Adams AM, Berg CD, Black WC, Clapp JD, Fagerstrom RM, Gareen IF, Gatsonis C, Marcus PM, Sicks JD. 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]
- 49.Slatore CG, Baumann C, Pappas M, Humphrey LL. Smoking behaviors among patients receiving computed tomography for lung cancer screening. Systematic review in support of the U.S. Preventive Services Task Force. Annals of the American Thoracic Society. 2014;11(4):619–627. doi: 10.1513/AnnalsATS.201312-460OC. [DOI] [PubMed] [Google Scholar]
- 50.Final Recommendation Statement: Lung Cancer: Screening. U.S. Preventive Services Task Force. 2014 Oct; http://www.uspreventiveservicestaskforce.org/Page/Document/RecommendationStatementFinal/lung-cancer-screening.
- 51.Tanner NT, Gebregziabher M, Hughes Halbert C, Payne E, Egede LE, Silvestri GA. Racial Differences in Outcomes within the National Lung Screening Trial. Implications for Widespread Implementation. Am J Respir Crit Care Med. 2015;192(2):200–208. doi: 10.1164/rccm.201502-0259OC. [DOI] [PubMed] [Google Scholar]
- 52.Kumar P, Gareen IF, Lathan C, Sicks JD, Perez GK, Hyland KA, Park ER. Racial Differences in Tobacco Cessation and Treatment Usage After Lung Screening: An Examination of the National Lung Screening Trial. Oncologist. 2016;21(1):40–49. doi: 10.1634/theoncologist.2015-0325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Clinical Lung Cancer Genome P, Network Genomic M. A genomics-based classification of human lung tumors. Sci Transl Med. 2013;5(209):209ra153. doi: 10.1126/scitranslmed.3006802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Pao W, Girard N. New driver mutations in non-small-cell lung cancer. Lancet Oncol. 2011;12(2):175–180. doi: 10.1016/S1470-2045(10)70087-5. [DOI] [PubMed] [Google Scholar]
- 55.Politi K, Herbst RS. Lung cancer in the era of precision medicine. Clin Cancer Res. 2015;21(10):2213–2220. doi: 10.1158/1078-0432.CCR-14-2748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Cress WD, Chiappori A, Santiago P, Munoz-Antonia T. Lung cancer mutations and use of targeted agents in Hispanics. Rev Recent Clin Trials. 2014;9(4):225–232. doi: 10.2174/1574887110666150127103555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Zhou W, Christiani DC. East meets West: ethnic differences in epidemiology and clinical behaviors of lung cancer between East Asians and Caucasians. Chin J Cancer. 2011;30(5):287–292. doi: 10.5732/cjc.011.10106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.da Cunha Santos G, Shepherd FA, Tsao MS. EGFR mutations and lung cancer. Annu Rev Pathol. 2011;6:49–69. doi: 10.1146/annurev-pathol-011110-130206. [DOI] [PubMed] [Google Scholar]
- 59.Pao W, Miller V, Zakowski M, Doherty J, Politi K, Sarkaria I, Singh B, Heelan R, Rusch V, Fulton L, Mardis E, Kupfer D, Wilson R, Kris M, Varmus H. EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc Natl Acad Sci U S A. 2004;101(36):13306–13311. doi: 10.1073/pnas.0405220101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Paez JG, Janne PA, Lee JC, Tracy S, Greulich H, Gabriel S, Herman P, Kaye FJ, Lindeman N, Boggon TJ, Naoki K, Sasaki H, Fujii Y, Eck MJ, Sellers WR, Johnson BE, Meyerson M. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 2004;304(5676):1497–1500. doi: 10.1126/science.1099314. [DOI] [PubMed] [Google Scholar]
- 61.Soh J, Okumura N, Lockwood WW, Yamamoto H, Shigematsu H, Zhang W, Chari R, Shames DS, Tang X, MacAulay C, Varella-Garcia M, Vooder T, Wistuba II, Lam S, Brekken R, Toyooka S, Minna JD, Lam WL, Gazdar AF. Oncogene mutations, copy number gains and mutant allele specific imbalance (MASI) frequently occur together in tumor cells. PLoS One. 2009;4(10):e7464. doi: 10.1371/journal.pone.0007464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Ladanyi M, Pao W. Lung adenocarcinoma: guiding EGFR-targeted therapy and beyond. Mod Pathol. 2008;21(Suppl 2):S16–S22. doi: 10.1038/modpathol.3801018. [DOI] [PubMed] [Google Scholar]
- 63.Shigematsu H, Lin L, Takahashi T, Nomura M, Suzuki M, Wistuba II, Fong KM, Lee H, Toyooka S, Shimizu N, Fujisawa T, Feng Z, Roth JA, Herz J, Minna JD, Gazdar AF. Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers. J Natl Cancer Inst. 2005;97(5):339–346. doi: 10.1093/jnci/dji055. [DOI] [PubMed] [Google Scholar]
- 64.Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, Sunpaweravong P, Han B, Margono B, Ichinose Y, Nishiwaki Y, Ohe Y, Yang JJ, Chewaskulyong B, Jiang H, Duffield EL, Watkins CL, Armour AA, Fukuoka M. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009;361(10):947–957. doi: 10.1056/NEJMoa0810699. [DOI] [PubMed] [Google Scholar]
- 65.Zhu CQ, da Cunha Santos G, Ding K, Sakurada A, Cutz JC, Liu N, Zhang T, Marrano P, Whitehead M, Squire JA, Kamel-Reid S, Seymour L, Shepherd FA, Tsao MS. National Cancer Institute of Canada Clinical Trials Group Study BR. Role of KRAS and EGFR as biomarkers of response to erlotinib in National Cancer Institute of Canada Clinical Trials Group Study BR.21. J Clin Oncol. 2008;26(26):4268–4275. doi: 10.1200/JCO.2007.14.8924. [DOI] [PubMed] [Google Scholar]
- 66.Han B, Tjulandin S, Hagiwara K, Normanno N, Wulandari L, Konstantinovich LK, Hudoyo A, Ratcliffe M, McCormack R, Reck M. Determining the Prevalence of Egfr Mutations in Asian and Russian Patients (Pts) with Advanced Non-Small-Cell Lung Cancer (Ansclc) of Adenocarcinoma (Adc) and Non-Adc Histology: Ignite Study. Annals of Oncology. 2015;26 [Google Scholar]
- 67.Yang SH, Mechanic LE, Yang P, Landi MT, Bowman ED, Wampfler J, Meerzaman D, Hong KM, Mann F, Dracheva T, Fukuoka J, Travis W, Caporaso NE, Harris CC, Jen J. Mutations in the tyrosine kinase domain of the epidermal growth factor receptor in non-small cell lung cancer. Clin Cancer Res. 2005;11(6):2106–2110. doi: 10.1158/1078-0432.CCR-04-1853. [DOI] [PubMed] [Google Scholar]
- 68.Leidner RS, Fu P, Clifford B, Hamdan A, Jin C, Eisenberg R, Boggon TJ, Skokan M, Franklin WA, Cappuzzo F, Hirsch FR, Varella-Garcia M, Halmos B. Genetic abnormalities of the EGFR pathway in African American Patients with non-small-cell lung cancer. J Clin Oncol. 2009;27(33):5620–5626. doi: 10.1200/JCO.2009.23.1431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Riely GJ, Pao W, Pham D, Li AR, Rizvi N, Venkatraman ES, Zakowski MF, Kris MG, Ladanyi M, Miller VA. Clinical course of patients with non-small cell lung cancer and epidermal growth factor receptor exon 19 and exon 21 mutations treated with gefitinib or erlotinib. Clin Cancer Res. 2006;12(3 Pt 1):839–844. doi: 10.1158/1078-0432.CCR-05-1846. [DOI] [PubMed] [Google Scholar]
- 70.Cote ML, Haddad R, Edwards DJ, Atikukke G, Gadgeel S, Soubani AO, Lonardo F, Bepler G, Schwartz AG, Ethier SP. Frequency and type of epidermal growth factor receptor mutations in African Americans with non-small cell lung cancer. J Thorac Oncol. 2011;6(3):627–630. doi: 10.1097/JTO.0b013e31820a0ec0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Reinersman JM, Johnson ML, Riely GJ, Chitale DA, Nicastri AD, Soff GA, Schwartz AG, Sima CS, Ayalew G, Lau C, Zakowski MF, Rusch VW, Ladanyi M, Kris MG. Frequency of EGFR and KRAS mutations in lung adenocarcinomas in African Americans. J Thorac Oncol. 2011;6(1):28–31. doi: 10.1097/JTO.0b013e3181fb4fe2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Arrieta O, Cardona AF, Federico Bramuglia G, Gallo A, Campos-Parra AD, Serrano S, Castro M, Aviles A, Amorin E, Kirchuk R, Cuello M, Borbolla J, Riemersma O, Becerra H, Rosell R, Clica P. Genotyping non-small cell lung cancer (NSCLC) in Latin America. J Thorac Oncol. 2011;6(11):1955–1959. doi: 10.1097/JTO.0b013e31822f655f. [DOI] [PubMed] [Google Scholar]
- 73.Corrales-Rodriguez L, Abbas J, Ramos-Esquivel A, Araya MAMC. Immunohistochemistry as a predictor of EGFR mutations: a retrospective case-control study. J Thorac Oncol. 2014;9(9):S176. [Google Scholar]
- 74.Rojas-Vigott R, Ugalde-Gamboa LJRS. Thoracic Oncology Clinic: Our First 200 cases. San Jose, Costa Rica. J Thorac Oncol. 2014;9(9):S182. [Google Scholar]
- 75.Castillo-Fernandez O, Arauz E, Cabreja A. al. e. EGFR Mutation and outcomes in aptients with metastatic NSCLC treated with erlotinib at Instituto Oncologico Nacional. Panama. J Thorac Oncol. 2014;9(9):S189. [Google Scholar]
- 76.Riely GJ, Marks J, Pao W. KRAS mutations in non-small cell lung cancer. Proc Am Thorac Soc. 2009;6(2):201–205. doi: 10.1513/pats.200809-107LC. [DOI] [PubMed] [Google Scholar]
- 77.Adjei AA. Blocking oncogenic Ras signaling for cancer therapy. Journal of the National Cancer Institute. 2001;93(14):1062–1074. doi: 10.1093/jnci/93.14.1062. [DOI] [PubMed] [Google Scholar]
- 78.Cox AD, Der CJ. Ras history: The saga continues. Small GTPases. 2010;1(1):2–27. doi: 10.4161/sgtp.1.1.12178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Castellano E, Santos E. Functional specificity of ras isoforms: so similar but so different. Genes Cancer. 2011;2(3):216–231. doi: 10.1177/1947601911408081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Pao W, Girard N. New driver mutations in non-small-cell lung cancer. Lancet Oncol. 2011;12(2):175–180. doi: 10.1016/S1470-2045(10)70087-5. [DOI] [PubMed] [Google Scholar]
- 81.Ding L, Getz G, Wheeler DA, Mardis ER, McLellan MD, Cibulskis K, Sougnez C, Greulich H, Muzny DM, Morgan MB, Fulton L, Fulton RS, Zhang Q, Wendl MC, Lawrence MS, Larson DE, Chen K, Dooling DJ, Sabo A, Hawes AC, Shen H, Jhangiani SN, Lewis LR, Hall O, Zhu Y, Mathew T, Ren Y, Yao J, Scherer SE, Clerc K, Metcalf GA, Ng B, Milosavljevic A, Gonzalez-Garay ML, Osborne JR, Meyer R, Shi X, Tang Y, Koboldt DC, Lin L, Abbott R, Miner TL, Pohl C, Fewell G, Haipek C, Schmidt H, Dunford-Shore BH, Kraja A, Crosby SD, Sawyer CS, Vickery T, Sander S, Robinson J, Winckler W, Baldwin J, Chirieac LR, Dutt A, Fennell T, Hanna M, Johnson BE, Onofrio RC, Thomas RK, Tonon G, Weir BA, Zhao X, Ziaugra L, Zody MC, Giordano T, Orringer MB, Roth JA, Spitz MR, Wistuba II, Ozenberger B, Good PJ, Chang AC, Beer DG, Watson MA, Ladanyi M, Broderick S, Yoshizawa A, Travis WD, Pao W, Province MA, Weinstock GM, Varmus HE, Gabriel SB, Lander ES, Gibbs RA, Meyerson M, Wilson RK. 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]
- 82.Califano R, Landi L, Cappuzzo F. Prognostic and predictive value of K-RAS mutations in non-small cell lung cancer. Drugs. 2012;72(Suppl 1):28–36. doi: 10.2165/1163012-S0-000000000-00000. [DOI] [PubMed] [Google Scholar]
- 83.Meng D, Yuan M, Li X, Chen L, Yang J, Zhao X, Ma W, Xin J. Prognostic value of K-RAS mutations in patients with non-small cell lung cancer: a systematic review with meta-analysis. Lung Cancer. 2013;81(1):1–10. doi: 10.1016/j.lungcan.2013.03.019. [DOI] [PubMed] [Google Scholar]
- 84.Riely GJ, Kris MG, Rosenbaum D, Marks J, Li A, Chitale DA, Nafa K, Riedel ER, Hsu M, Pao W, Miller VA, Ladanyi M. Frequency and distinctive spectrum of KRAS mutations in never smokers with lung adenocarcinoma. Clin Cancer Res. 2008;14(18):5731–5734. doi: 10.1158/1078-0432.CCR-08-0646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Sun Y, Ren Y, Fang Z, Li C, Fang R, Gao B, Han X, Tian W, Pao W, Chen H, Ji H. Lung adenocarcinoma from East Asian never-smokers is a disease largely defined by targetable oncogenic mutant kinases. J Clin Oncol. 2010;28(30):4616–4620. doi: 10.1200/JCO.2010.29.6038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.El-Telbany A, Ma PC. Cancer genes in lung cancer: racial disparities: are there any? Genes Cancer. 2012;3(7–8):467–480. doi: 10.1177/1947601912465177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Bauml J, Mick R, Zhang Y, Watt CD, Vachani A, Aggarwal C, Evans T, Langer C. Frequency of EGFR and KRAS mutations in patients with non small cell lung cancer by racial background: do disparities exist? Lung Cancer. 2013;81(3):347–353. doi: 10.1016/j.lungcan.2013.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.McQuitty E, Zhang W, Hendrickson H, Tio FO, Jagirdar J, Olsen R, Cagle PT. Lung adenocarcinoma biomarker incidence in Hispanic versus non-Hispanic white patients. Arch Pathol Lab Med. 2014;138(3):390–394. doi: 10.5858/arpa.2013-0225-OA. [DOI] [PubMed] [Google Scholar]
- 89.Shaw RJ, Kosmatka M, Bardeesy N, Hurley RL, Witters LA, DePinho RA, Cantley LC. The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(10):3329–3335. doi: 10.1073/pnas.0308061100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Cao Y, Li H, Liu H, Zheng C, Ji H, Liu X. The serine/threonine kinase LKB1 controls thymocyte survival through regulation of AMPK activation and Bcl-XL expression. Cell Res. 2010;20(1):99–108. doi: 10.1038/cr.2009.141. [DOI] [PubMed] [Google Scholar]
- 91.Gao Y, Ge G, Ji H. LKB1 in lung cancerigenesis: a serine/threonine kinase as tumor suppressor. Protein Cell. 2011;2(2):99–107. doi: 10.1007/s13238-011-1021-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Hemminki A, Markie D, Tomlinson I, Avizienyte E, Roth S, Loukola A, Bignell G, Warren W, Aminoff M, Hoglund P, Jarvinen H, Kristo P, Pelin K, Ridanpaa M, Salovaara R, Toro T, Bodmer W, Olschwang S, Olsen AS, Stratton MR, de la Chapelle A, Aaltonen LA. A serine/threonine kinase gene defective in Peutz-Jeghers syndrome. Nature. 1998;391(6663):184–187. doi: 10.1038/34432. [DOI] [PubMed] [Google Scholar]
- 93.Lim W, Hearle N, Shah B, Murday V, Hodgson SV, Lucassen A, Eccles D, Talbot I, Neale K, Lim AG, O’Donohue J, Donaldson A, Macdonald RC, Young ID, Robinson MH, Lee PW, Stoodley BJ, Tomlinson I, Alderson D, Holbrook AG, Vyas S, Swarbrick ET, Lewis AA, Phillips RK, Houlston RS. Further observations on LKB1/STK11 status and cancer risk in Peutz-Jeghers syndrome. Br J Cancer. 2003;89(2):308–313. doi: 10.1038/sj.bjc.6601030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Sanchez-Cespedes M, Parrella P, Esteller M, Nomoto S, Trink B, Engles JM, Westra WH, Herman JG, Sidransky D. Inactivation of LKB1/STK11 is a common event in adenocarcinomas of the lung. Cancer Res. 2002;62(13):3659–3662. [PubMed] [Google Scholar]
- 95.Shah U, Sharpless NE, Hayes DN. LKB1 and lung cancer: more than the usual suspects. Cancer Res. 2008;68(10):3562–3565. doi: 10.1158/0008-5472.CAN-07-6620. [DOI] [PubMed] [Google Scholar]
- 96.Sanchez-Cespedes M. The role of LKB1 in lung cancer. Fam Cancer. 2011;10(3):447–453. doi: 10.1007/s10689-011-9443-0. [DOI] [PubMed] [Google Scholar]
- 97.Matsumoto S, Iwakawa R, Takahashi K, Kohno T, Nakanishi Y, Matsuno Y, Suzuki K, Nakamoto M, Shimizu E, Minna JD, Yokota J. Prevalence and specificity of LKB1 genetic alterations in lung cancers. Oncogene. 2007;26(40):5911–5918. doi: 10.1038/sj.onc.1210418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Ji H, Ramsey MR, Hayes DN, Fan C, McNamara K, Kozlowski P, Torrice C, Wu MC, Shimamura T, Perera SA, Liang MC, Cai D, Naumov GN, Bao L, Contreras CM, Li D, Chen L, Krishnamurthy J, Koivunen J, Chirieac LR, Padera RF, Bronson RT, Lindeman NI, Christiani DC, Lin X, Shapiro GI, Janne PA, Johnson BE, Meyerson M, Kwiatkowski DJ, Castrillon DH, Bardeesy N, Sharpless NE, Wong KK. LKB1 modulates lung cancer differentiation and metastasis. Nature. 2007;448(7155):807–810. doi: 10.1038/nature06030. [DOI] [PubMed] [Google Scholar]
- 99.O’Neill GM, Seo S, Serebriiskii IG, Lessin SR, Golemis EA. A new central scaffold for metastasis: parsing HEF1/Cas-L/NEDD9. Cancer Res. 2007;67(19):8975–8979. doi: 10.1158/0008-5472.CAN-07-1328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Gao Y, Xiao Q, Ma H, Li L, Liu J, Feng Y, Fang Z, Wu J, Han X, Zhang J, Sun Y, Wu G, Padera R, Chen H, Wong KK, Ge G, Ji H. LKB1 inhibits lung cancer progression through lysyl oxidase and extracellular matrix remodeling. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(44):18892–18897. doi: 10.1073/pnas.1004952107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Wingo SN, Gallardo TD, Akbay EA, Liang MC, Contreras CM, Boren T, Shimamura T, Miller DS, Sharpless NE, Bardeesy N, Kwiatkowski DJ, Schorge JO, Wong KK, Castrillon DH. Somatic LKB1 mutations promote cervical cancer progression. PloS one. 2009;4(4):e5137. doi: 10.1371/journal.pone.0005137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Sanchez-Cespedes M. A role for LKB1 gene in human cancer beyond the Peutz-Jeghers syndrome. Oncogene. 2007;26(57):7825–7832. doi: 10.1038/sj.onc.1210594. [DOI] [PubMed] [Google Scholar]
- 103.Koivunen JP, Kim J, Lee J, Rogers AM, Park JO, Zhao X, Naoki K, Okamoto I, Nakagawa K, Yeap BY, Meyerson M, Wong KK, Richards WG, Sugarbaker DJ, Johnson BE, Janne PA. Mutations in the LKB1 tumour suppressor are frequently detected in tumours from Caucasian but not Asian lung cancer patients. Br J Cancer. 2008;99(2):245–252. doi: 10.1038/sj.bjc.6604469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Onozato R, Kosaka T, Achiwa H, Kuwano H, Takahashi T, Yatabe Y, Mitsudomi T. LKB1 gene mutations in Japanese lung cancer patients. Cancer Sci. 2007;98(11):1747–1751. doi: 10.1111/j.1349-7006.2007.00585.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Gao B, Sun Y, Zhang J, Ren Y, Fang R, Han X, Shen L, Liu XY, Pao W, Chen H, Ji H. Spectrum of LKB1, EGFR, and KRAS mutations in chinese lung adenocarcinomas. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2010;5(8):1130–1135. doi: 10.1097/JTO.0b013e3181e05016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Sun YH, Ren Y, Fang ZY, Li CG, Fang R, Gao B, Han XK, Tian WD, Pao W, Chen HQ, Ji HB. Lung Adenocarcinoma From East Asian Never-Smokers Is a Disease Largely Defined by Targetable Oncogenic Mutant Kinases. Journal of Clinical Oncology. 2010;28(30):4616–4620. doi: 10.1200/JCO.2010.29.6038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Forcet C, Etienne-Manneville S, Gaude H, Fournier L, Debilly S, Salmi M, Baas A, Olschwang S, Clevers H, Billaud M. Functional analysis of Peutz-Jeghers mutations reveals that the LKB1 C-terminal region exerts a crucial role in regulating both the AMPK pathway and the cell polarity. Human molecular genetics. 2005;14(10):1283–1292. doi: 10.1093/hmg/ddi139. [DOI] [PubMed] [Google Scholar]
- 108.Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ, Bottomley W, Davis N, Dicks E, Ewing R, Floyd Y, Gray K, Hall S, Hawes R, Hughes J, Kosmidou V, Menzies A, Mould C, Parker A, Stevens C, Watt S, Hooper S, Wilson R, Jayatilake H, Gusterson BA, Cooper C, Shipley J, Hargrave D, Pritchard-Jones K, Maitland N, Chenevix-Trench G, Riggins GJ, Bigner DD, Palmieri G, Cossu A, Flanagan A, Nicholson A, Ho JW, Leung SY, Yuen ST, Weber BL, Seigler HF, Darrow TL, Paterson H, Marais R, Marshall CJ, Wooster R, Stratton MR, Futreal PA. Mutations of the BRAF gene in human cancer. Nature. 2002;417(6892):949–954. doi: 10.1038/nature00766. [DOI] [PubMed] [Google Scholar]
- 109.Naoki K, Chen TH, Richards WG, Sugarbaker DJ, Meyerson M. Missense mutations of the BRAF gene in human lung adenocarcinoma. Cancer Res. 2002;62(23):7001–7003. [PubMed] [Google Scholar]
- 110.Brose MS, Volpe P, Feldman M, Kumar M, Rishi I, Gerrero R, Einhorn E, Herlyn M, Minna J, Nicholson A, Roth JA, Albelda SM, Davies H, Cox C, Brignell G, Stephens P, Futreal PA, Wooster R, Stratton MR, Weber BL. BRAF and RAS mutations in human lung cancer and melanoma. Cancer Res. 2002;62(23):6997–7000. [PubMed] [Google Scholar]
- 111.Marchetti A, Felicioni L, Malatesta S, Grazia Sciarrotta M, Guetti L, Chella A, Viola P, Pullara C, Mucilli F, Buttitta F. Clinical features and outcome of patients with non-small-cell lung cancer harboring BRAF mutations. J Clin Oncol. 2011;29(26):3574–3579. doi: 10.1200/JCO.2011.35.9638. [DOI] [PubMed] [Google Scholar]
- 112.Samuels Y, Wang Z, Bardelli A, Silliman N, Ptak J, Szabo S, Yan H, Gazdar A, Powell SM, Riggins GJ, Willson JK, Markowitz S, Kinzler KW, Vogelstein B, Velculescu VE. High frequency of mutations of the PIK3CA gene in human cancers. Science. 2004;304(5670):554. doi: 10.1126/science.1096502. [DOI] [PubMed] [Google Scholar]
- 113.Kawano O, Sasaki H, Endo K, Suzuki E, Haneda H, Yukiue H, Kobayashi Y, Yano M, Fujii Y. PIK3CA mutation status in Japanese lung cancer patients. Lung Cancer. 2006;54(2):209–215. doi: 10.1016/j.lungcan.2006.07.006. [DOI] [PubMed] [Google Scholar]
- 114.Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, Fujiwara S, Watanabe H, Kurashina K, Hatanaka H, Bando M, Ohno S, Ishikawa Y, Aburatani H, Niki T, Sohara Y, Sugiyama Y, Mano H. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature. 2007;448(7153):561–566. doi: 10.1038/nature05945. [DOI] [PubMed] [Google Scholar]
- 115.Koivunen JP, Mermel C, Zejnullahu K, Murphy C, Lifshits E, Holmes AJ, Choi HG, Kim J, Chiang D, Thomas R, Lee J, Richards WG, Sugarbaker DJ, Ducko C, Lindeman N, Marcoux JP, Engelman JA, Gray NS, Lee C, Meyerson M, Janne PA. EML4-ALK fusion gene and efficacy of an ALK kinase inhibitor in lung cancer. Clin Cancer Res. 2008;14(13):4275–4283. doi: 10.1158/1078-0432.CCR-08-0168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Wong DW, Leung EL, So KK, Tam IY, Sihoe AD, Cheng LC, Ho KK, Au JS, Chung LP, Pik Wong M University of Hong Kong Lung Cancer Study G. 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]
- 117.Inamura K, Takeuchi K, Togashi Y, Nomura K, Ninomiya H, Okui M, Satoh Y, Okumura S, Nakagawa K, Soda M, Choi YL, Niki T, Mano H, Ishikawa Y. EML4-ALK fusion is linked to histological characteristics in a subset of lung cancers. J Thorac Oncol. 2008;3(1):13–17. doi: 10.1097/JTO.0b013e31815e8b60. [DOI] [PubMed] [Google Scholar]
- 118.Inamura K, Takeuchi K, Togashi Y, Hatano S, Ninomiya H, Motoi N, Mun MY, Sakao Y, Okumura S, Nakagawa K, Soda M, Choi YL, Mano H, Ishikawa Y. EML4-ALK lung cancers are characterized by rare other mutations, a TTF-1 cell lineage, an acinar histology, and young onset. Mod Pathol. 2009;22(4):508–515. doi: 10.1038/modpathol.2009.2. [DOI] [PubMed] [Google Scholar]
- 119.Ren S, Kuang P, Zheng L, Su C, Li J, Li B, Chen X, Wang Y, KimCurran V, Liu L, Hu Q, Zhang J, Tang L, Zhou C. Analysis of driver mutations in female non-smoker Asian patients with pulmonary adenocarcinoma. Cell Biochem Biophys. 2012;64(2):155–160. doi: 10.1007/s12013-012-9384-8. [DOI] [PubMed] [Google Scholar]
- 120.Li C, Fang R, Sun Y, Han X, Li F, Gao B, Iafrate AJ, Liu XY, Pao W, Chen H, Ji H. Spectrum of oncogenic driver mutations in lung adenocarcinomas from East Asian never smokers. PLoS One. 2011;6(11):e28204. doi: 10.1371/journal.pone.0028204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Kim HR, Shim HS, Chung JH, Lee YJ, Hong YK, Rha SY, Kim SH, Ha SJ, Kim SK, Chung KY, Soo R, Kim JH, Cho BC. Distinct clinical features and outcomes in never-smokers with nonsmall cell lung cancer who harbor EGFR or KRAS mutations or ALK rearrangement. Cancer. 2012;118(3):729–739. doi: 10.1002/cncr.26311. [DOI] [PubMed] [Google Scholar]
- 122.Shinmura K, Kageyama S, Tao H, Bunai T, Suzuki M, Kamo T, Takamochi K, Suzuki K, Tanahashi M, Niwa H, Ogawa H, Sugimura H. EML4-ALK fusion transcripts, but no NPM-, TPM3-, CLTC-, ATIC-, or TFG-ALK fusion transcripts, in non-small cell lung carcinomas. Lung Cancer. 2008;61(2):163–169. doi: 10.1016/j.lungcan.2007.12.013. [DOI] [PubMed] [Google Scholar]
- 123.Martelli MP, Sozzi G, Hernandez L, Pettirossi V, Navarro A, Conte D, Gasparini P, Perrone F, Modena P, Pastorino U, Carbone A, Fabbri A, Sidoni A, Nakamura S, Gambacorta M, Fernandez PL, Ramirez J, Chan JK, Grigioni WF, Campo E, Pileri SA, Falini B. EML4-ALK rearrangement in non-small cell lung cancer and non-tumor lung tissues. Am J Pathol. 2009;174(2):661–670. doi: 10.2353/ajpath.2009.080755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Esnaola NF, Gebregziabher M, Knott K, Finney C, Silvestri GA, Reed CE, Ford ME. Underuse of surgical resection for localized, non-small cell lung cancer among whites and African Americans in South Carolina. Ann Thorac Surg. 2008;86(1):220–226. doi: 10.1016/j.athoracsur.2008.02.072. discussion 227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.van Ryn M. Research on the provider contribution to race/ethnicity disparities in medical care. Med Care. 2002;40(1 Suppl):I140–I151. doi: 10.1097/00005650-200201001-00015. [DOI] [PubMed] [Google Scholar]
- 126.Underwood JM, Townsend JS, Tai E, Davis SP, Stewart SL, White A, Momin B, Fairley TL. Racial and regional disparities in lung cancer incidence. Cancer. 2012;118(7):1910–1918. doi: 10.1002/cncr.26479. [DOI] [PubMed] [Google Scholar]
- 127.Yang R, Cheung MC, Byrne MM, Huang Y, Nguyen D, Lally BE, Koniaris LG. Do racial or socioeconomic disparities exist in lung cancer treatment? Cancer. 2010;116(10):2437–2447. doi: 10.1002/cncr.24986. [DOI] [PubMed] [Google Scholar]
- 128.Centers for Disease C, Prevention. Cigarette smoking among adults--United States, 2006. MMWR Morb Mortal Wkly Rep. 2007;56(44):1157–1161. [PubMed] [Google Scholar]
- 129.Centers for Disease Control and Prevention. Current Cigarette Smoking Among Adults—United States, 2005–2013. Morbidity and Mortality Weekly Report. 2014;63(47):1108–1112. [PMC free article] [PubMed] [Google Scholar]