Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Jul 1.
Published in final edited form as: Curr Opin Pulm Med. 2013 Jul;19(4):331–339. doi: 10.1097/MCP.0b013e328362075c

How and when to use genetic markers for non-small cell lung cancer

Donald R Lazarus 1, David E Ost 1
PMCID: PMC3926417  NIHMSID: NIHMS529066  PMID: 23715289

Abstract

Purpose of review

Many driver mutations that determine the malignant behavior of lung cancer have been identified in recent years. The promise of therapies targeted to the specific molecular pathways altered by such mutations has made genetic testing in non-small cell lung cancer (NSCLC) attractive to clinicians. We review recent research on clinically relevant genetic and molecular tests for patients with NSCLC, with an emphasis on the tests linked to actionable mutations that influence therapy and improve outcomes.

Recent findings

Mutations in the epidermal growth factor receptor (EGFR) and translocations involving the anaplastic lymphoma kinase (ALK) gene have been shown to be common driver mutations in lung adenocarcinoma. The presence or absence of these mutations has been demonstrated to predict response to targeted therapy in many recent studies. Targeted therapies for patients with mutations in the EGFR domain or the EML4-ALK translocation have been shown to be effective and are approved for use. Ongoing studies continue to define the extent of their utility and may continue to expand their indications. Sufficient tissue for genetic analysis can be obtained from cytologic samples, including those obtained from endobronchial ultrasound-guided transbronchial needle aspiration.

Summary

Genetic testing for driver mutations is useful in identifying patients with NSCLC who are likely to respond to targeted therapy. These tests are best used in patients with adenocarcinoma who have advanced-stage cancer.

Keywords: Lung cancer, genetic test, targeted therapy, epidermal growth factor receptor, anaplastic lymphoma kinase

Introduction

Lung cancer is responsible for more deaths than any other form of cancer in the United States; more than 150,000 people died from lung cancer in 2008 alone. Non-small cell lung cancer (NSCLC) is much more common than small cell lung cancer (1). Historically, all types of NSCLC were treated in a similar manner, determined primarily by clinicopathologic stage, and patients with advanced NSCLC received cytotoxic chemotherapy as the mainstay of treatment (2). As recent research has improved knowledge of the molecular pathways that determine the behavior of cancer cells, it has become clear that NSCLC is a biologically heterogeneous group of cancers driven by differing molecular pathways. As the mutations causing the oncogenic alterations to these pathways have been discovered and genetic tests for them have become available, researchers have been developing treatments that target these aberrant pathways in the hope that such targeted treatments will provide better outcomes with fewer adverse effects than traditional cytotoxic chemotherapy (3)*.

In this review, we will first discuss the characteristics of a useful genetic marker in patients with NSCLC. We will then review the genetic tests that have been linked to driver mutations in NSCLC for which targeted treatment is available. Finally, we will briefly discuss the approved targeted treatments for NSCLC that harbors these mutations. A full discussion of the many other mutations that have been found to play a role in malignant behavior in NSCLC for which effective targeted treatments have not yet been discovered or approved will not be provided in this review.

Characteristics of a Useful Genetic Marker

Not all mutations harbored in cancer cells make useful genetic markers. Useful genetic markers are those that indicate driver mutations, can be identified accurately using genetic testing, are common in the population of interest, and point to an oncogenic pathway for which effective targeted therapy exists

A genetic marker is useful only if it is associated with an oncogenic pathway whose disruption would result in inhibited cellular growth and survival. Most of the genetic mutations found in cancer cells do not contribute to the development of cancer. These “passenger mutations” occur incidentally, are not of themselves responsible for malignant transformation of the cell, and are not important for maintaining the malignant phenotype (4). Although passenger mutations may often be related to pathways important to the growth and survival of the cell, they are typically found downstream from other genes that stimulate multiple pathways promoting growth and survival. Mutations in these genes, or “driver mutations,” initiate the transformation of a benign cell to a malignant cell. Driver mutations are not found in the germline genome, and the transformed cell comes to rely on the driver mutation's signaling for survival, a process known as oncogene addiction (4-6). For this reason, driver mutations are often good candidates for targeted therapy. Disrupting the signaling of driver mutations can have far-reaching effects on multiple metabolic pathways within the cancer cell. Disrupting the pathway regulated by a single passenger mutation is generally less useful because driver mutations continue to stimulate multiple additional pathways promoting growth and survival (4, 6).

In addition to identifying a driver mutation, a useful genetic marker should have an accurate test available that identifies the presence of the mutation of interest. Various methods are used to test samples of cancers for mutations, including polymerase chain reaction, immunohistochemistry (IHC), and fluorescence in situ hybridization (FISH). However, not every type of genetic test predicts tumor response to targeted therapy for each specific mutation, and the correct test must be chosen to properly identify the sought-after mutation. For example, the 2 most commonly tested markers in NSCLC are those for mutations in the epidermal growth factor receptor (EGFR) and for the echinoderm microtubule-associated protein-like 4 anaplastic lymphoma kinase (EML4-ALK) translocation. Mutational analysis is currently the preferred test for EGFR mutations, whereas the EML4-ALK translocation is best identified via FISH (7, 8)*.

Many types of histologic and cytologic samples have been used to test for EGFR mutations and the EML4-ALK translocation. Histologic specimens obtained via surgery or core needle biopsy have been historically preferred because of the large amount of tissue provided, but biopsies of this size are difficult to obtain in some cases (9)*. Nakajima et al first reported the use of cytologic specimens obtained during endobronchial ultrasound (EBUS) for EGFR mutation testing, and several other groups have confirmed the utility of cytologic specimens for this purpose (10-12). Billah et al reported only a 4% rate of insufficiency for EGFR mutation testing of tissue obtained via EBUS (9)*. A more recent large multicenter study of 774 cytologic specimens obtained via EBUS reported that EGFR mutation analysis was possible in 90% of specimens for which it was requested (13)*. ALK fusion genes can also be identified on cytologic specimens obtained via EBUS (14). Even multiple gene mutation analyses are feasible using cytologic specimens (15, 16). The literature supports the use of cytologic specimens of good quality, including those taken during bronchoscopy, for molecular testing in NSCLC.

A useful genetic marker must also be found commonly enough for its use to be practical. Genetic testing is costly, and testing large numbers of patients when the prevalence of the mutation of interest is vanishingly low would produce relatively little benefit for the cost involved. One way to enrich the population chosen to undergo genetic testing in lung cancer is to narrow the group of patients to be tested to a more manageable size with a higher prevalence of the mutation of interest. For example, the genetic alteration in EGFR that is susceptible to targeted therapy is found almost exclusively in non-squamous NSCLC. The prevalence of the EGFR mutation is less than 5% in patients with squamous cell cancer, and is estimated to be about 15-20% for patients with adenocarcinoma (17-19)*. The group of patients referred for EGFR mutation analysis should also include patients with histologically adenosquamous tumors, because small studies have demonstrated a significant response of adenosquamous tumors with the EGFR mutation to agents targeting EGFR (20)*. Using tumor histologic findings to determine when genetic testing should be performed can allow the clinician to use genetic testing in a population in which the prevalence of a genetic mutation justifies the expense of the test.

Finally, a useful genetic marker must indicate an oncogenic pathway for which an effective targeted therapy exists. It is reasonable to test for a broad array of mutations that may not be associated with a targeted therapy as part of epidemiologic studies of lung cancer or to assess patients for entry into a clinical trial. However, it is much less useful to do so in general clinical practice because most mutations have no effective targeted therapy, making the information that would be obtained not actionable. It is more appropriate and less costly to perform genetic testing only for mutations that mark oncogenic pathways that are susceptible to available and effective targeted treatments. The genetic markers in NSCLC that predict susceptibility to currently available targeted therapy are few and are found almost exclusively in adenocarcinoma of the lung.

The following sections review the genetic markers that are considered useful markers in NSCLC, with a brief discussion of the currently available targeted therapies for patients with these mutations.

EGFR Mutations

Mutations in the EGFR family of receptor tyrosine kinases are found in 10-23% of lung adenocarcinomas and rarely if ever in squamous cancers (18, 19, 21, 22)*. EGFR tyrosine kinases are involved in regulating multiple cellular activities, including growth, migration, and survival (21). The 2 most common EGFR mutations are small deletions in exon 19 and the L858R missense mutation in exon 21. Together these mutations account for more than 90% of EGFR mutations found in lung adenocarcinoma, and both mutations activate cellular survival pathways that inhibit apoptosis and have been shown to be transforming mutations in cultured cells (19, 23-26)*. Because cells with transforming EGFR mutations have also been shown to be sensitive to the effects of targeted EGFR tyrosine kinase inhibitors (TKIs), tumors with EGFR mutations are good subjects for targeted therapy (26-30).

Although EGFR mutations are reported to be more common in never-smokers, women, and Asians, EGFR mutations are not found exclusively in these groups (18, 31, 32). Two large studies recently assessed the genetic and clinical characteristics of patients with lung adenocarcinoma. D'Angelo et al examined 2142 lung adenocarcinoma specimens for EGFR mutations and found that although EGFR mutations were more common in specimens from never-smokers and women, significant numbers of specimens from smokers and men also harbored the mutations (22)*. Dogan et al examined 3026 lung adenocarcinoma specimens and found that it was impossible to clinically identify a subgroup of patients with adenocarcinoma of the lung that had a low enough (less than 1%) prevalence of EGFR mutations to obviate the need for genetic testing (19)*.

No strong consensus has been reached on the best way to test for EGFR mutations. FISH and IHC can be used to measure the EGFR gene copy number, but neither method has yet been consistently shown to produce results that correlate with sensitivity to therapy targeted to EGFR (7)*. One recent study did compare FISH, IHC, and mutation analysis by direct sequencing in specimens from 40 patients with NSCLC treated with EGFR TKI. Only results from EGFR mutation analysis by direct sequencing were found to correlate with response to therapy in this study, so the mutation analysis method is currently recommended for testing for EGFR mutations (31).

Mutations in EGFR also predict responsiveness to treatment with EGFR TKIs (30, 33-38)*, and 2 medications in this class, gefitinib and erlotinib, are currently available. Both are orally administered EGFR TKIs that are approved for the treatment of advanced NSCLC for the indications shown in Table 1 (39-41). Several recent studies have continued to refine criteria used to select patients for testing for EGFR mutations as well as potentially expand the indications for using EGFR TKIs to treat NSCLC. The most important of these for gefitinib was the Iressa Pan-Asia Study, which compared gefitinib with carboplatin plus paclitaxel in 1217 previously untreated patients with advanced NSCLC in Asia. Patients were eligible to participate in the study if they had clinical features associated with response to EGFR TKIs in prior studies: histologically indicated adenocarcinoma and nonsmoker or former light smoker. Although no difference in mean progression-free survival duration was noted between the study groups, differences in outcome were observed when the patients were stratified by EGFR mutation status. Among patients whose NSCLC harbored the EGFR mutation, the mean progression-free survival duration was longer among those treated with gefitinib than among those treated with carboplatin-paclitaxel. Conversely, among patients whose NSCLC did not harbor the EGFR mutation, the mean progression-free survival duration was shorter among those treated with gefitinib than among those treated with carboplatin-paclitaxel (see Figure 1). These findings demonstrate the primacy of the EGFR mutation status in determining which patients responded to treatment with gefitinib. All of the patients in this study had clinical characteristics that were thought to predict a good response to treatment with gefitinib, but only EGFR mutation status accurately predicted response to targeted therapy (42). A subsequent, similar study in Korea confirmed these findings in a smaller group of patients (43)*.

Table 1. Indications for approved targeted therapies in patients with non-small cell lung cancer (NSCLC) [33-35].

Targeted therapy Indications
Gefitinib Formerly for advanced NSCLC that has not responded to either platinum-based or docetaxel chemotherapy; in the United States now restricted to clinical trials or for patients who have benefitted from previous treatment with gefitinib
Erlotinib For advanced or metastatic NSCLC that has not responded to at least 1 other chemotherapy regimen or maintenance therapy for stage IIIB/IV NSCLC that has not progressed after 4 cycles of standard chemotherapy
Crizotinib For advanced NSCLC harboring an anaplastic lymphoma kinase translocation

Figure 1.

Figure 1

Results from the Iressa Pan-Asia Study [42] showing that in patients with non-small cell lung cancer harboring in the epidermal growth factor receptor (EGFR) mutation, the mean progression-free survival duration was longer among those treated with gefitinib than among those treated with carboplatin plus paclitaxel (B). However, the opposite was true among patients whose cancer did not harbor the EGFR mutation (C). (Figure reprinted with permission.)

Other recent studies assessing gefitinib in patients selected by mutation status rather than by clinical characteristics have shown promising results. The Western Japan Oncology Group compared gefitinib with cisplatin plus docetaxel in 172 patients with advanced NSCLC with the EGFR mutation and found that patients in the gefitinib group had longer progression-free survival durations than those in the cisplatin-docetaxel group (44). The North-East Japan Study Group also compared gefitinib with carboplatin-paclitaxel in patients with advanced NSCLC with the EGFR mutation and found that those in the gefitinib group had longer progression-free survival durations and better quality of life than those in the carboplatin-paclitaxel group (45). Although neither study was able to demonstrate a significant improvement in overall survival durations in those treated with gefitinib, this may have been due to crossover of patients in the control group to the gefitinib group after they experienced disease progression (44, 45).

Other recent trials have assessed extended indications for gefitinib. One retrospective case-control study suggested a survival benefit for patients with lung adenocarcinoma who have experienced recurrence after complete resection (46). Another recent trial demonstrated improved progression-free survival durations in patients with stage III or IV adenocarcinoma who received gefitinib as maintenance therapy after an initial response to standard chemotherapy and who had clinical characteristics predicting the presence of an EGFR mutation (47)*. Additional trials will be needed to confirm the utility of both of these potential indications.

Erlotinib was initially shown to improve overall survival rates in unselected patients with advanced NSCLC whose disease progressed despite prior first- or second-line chemotherapy. These patients all had stage III or IV NSCLC with residual disease after having received 1 or 2 standard chemotherapeutic regimens and were not eligible for further chemotherapy. Overall survival durations, progression-free survival durations, and response rates were all significantly greater in the group receiving erlotinib compared with the group receiving a placebo. Subgroup analysis suggested similar clinical predictors of response to erlotinib to the clinical predictors of response to gefitinib (48).

Although trials have not shown erlotinib to be as effective of a first-line treatment as standard chemotherapy for unselected patients with NSCLC (49)*, other recent trials in China and Europe have shown that first-line treatment with erlotinib resulted in better progression-free survival than first-line treatment with standard chemotherapy in patients with advanced NSCLC that harbored the EGFR mutation (50, 51)*. In the OPTIMAL study, Zhou et al randomized 165 patients in China with stage IIIB or IV NSCLC who were known to have EGFR mutations to receive initial treatment with either erlotinib or gemcitabine plus carboplatin. Patients in the erlotinib group had longer progression-free survival durations and fewer adverse effects than patients in the chemotherapy group (50)*. Rosell et al assessed erlotinib as a first-line treatment for advanced NSCLC in European patients in the EURTAC trial: 173 patients with advanced NSCLC with an EGFR mutation were randomized to receive initial treatment with either erlotinib or cisplatin plus docetaxel. The median progression-free survival duration was significantly longer in the erlotinib group, and the erlotinib group also had a lower rate of severe treatment-related adverse events (51)*. These trials suggest a role for erlotinib as a first-line treatment for selected patients with NSCLC, although formal approval for this indication in the United States is still pending.

Other recent studies have explored additional indications for erlotinib. One group retrospectively analyzed a cohort of patients with stage I or II lung adenocarcinoma with an EGFR mutation who underwent complete resection as primary therapy. Patients who received erlotinib as adjuvant therapy had slightly improved disease-free survival durations, but randomized trials will be needed to confirm this finding (52)*. Erlotinib has also been reported to be effective in patients with adenosquamous carcinoma with an EGFR mutation (20)*, and a phase II trial has suggested that erlotinib may also sensitize tumors to radiotherapy (53). Erlotinib has not been shown to be effective in patients with an EGFR mutation whose disease has already demonstrated resistance to gefitinib (54), and a recent phase II trial did not demonstrate additional benefit from administering chemotherapy to selected patients who had a high probability of harboring an EGFR mutation and were already receiving erlotinib as a first-line treatment for lung adenocarcinoma (55)*. The common thread in all of these studies is the importance of EGFR mutation analysis in predicting the response to targeted therapy. More research in this field should continue to clarify and possibly expand the indications for targeted EGFR TKIs.

EML4-ALK Translocation

A more recently characterized mutation is an inversion of the short arm of chromosome 2 that can lead to the EML4-ALK translocation (56). This translocation causes constitutive activation of the ALK receptor tyrosine kinase and has been shown to be transforming in cells and in vivo (56, 57)*. The EML4-ALK translocation is found in 2-7% of lung adenocarcinomas (57-60). It is more common in patients younger than 55 years and nonsmokers, and it is nearly always found in non-squamous NSCLC (56, 61, 62). The current standard for genetic testing for the EML4-ALK translocation is the break-apart FISH assay. This test is favored currently because it was the exclusive test used in the initial trials of crizotinib and its results correlate to response to targeted therapy. The FISH assay can identify any rearrangement involving ALK, allowing it to identify rare and previously unseen ALK rearrangements. This means that it has a lower false negative rate than the currently available reverse transcriptase polymerase chain reaction test for ALK, which cannot identify previously unknown ALK rearrangements (8).

The presence of the EML4-ALK translocation predicts sensitivity of NSCLC to crizotinib, an orally administered inhibitor of the ALK kinase as well as several other kinases within the cell (3, 62). Crizotinib received accelerated US Food and Drug Administration approval in 2011 for use in patients with advanced NSCLC who were confirmed to have the EML4-ALK translocation according to FISH assay results (8). Kwak et al evaluated 82 patients with ALK translocations in the first published trial of crizotinib for the treatment of NSCLC. An overall response rate of 57% was demonstrated in this single-arm, open-label study (62). Another phase I trial reported in abstract form in 2011 reported a 61% overall response rate (63). A retrospective analysis demonstrated improved overall survival durations after treatment with crizotinib among patients with advanced NSCLC with the ALK translocation compared with ALK-positive controls, but prospective studies are lacking (64)*. Other ongoing prospective trials are assessing survival benefits of treatment with crizotinib. Although less information is available regarding crizotinib than for EGFR TKIs, the activity of crizotinib against ALK and several other kinases that may act as driver mutations holds great promise.

Practical Clinical Application of Genetic Testing in NSCLC

As outlined above, the ideal useful genetic marker in NSCLC indicates a driver mutation that responds to targeted therapy and is present in a significant proportion of the population to be tested, and for which an accurate test is available. The practicing clinician has little influence on which genetic tests or targeted therapies are available; however, the clinician can decide which patients with NSCLC to test for genetic markers. A simple, clinically based algorithm as outlined in Figure 2 can help physicians to identify patients for whom the above detailed genetic tests should be ordered.

Figure 2.

Figure 2

Suggested algorithm for genetic testing in patients with non-small cell lung cancer.

First, the clinical stage of the tumor should be considered. None of the available targeted therapies are currently indicated for limited-stage lung cancer; surgery remains the mainstay of treatment for limited-stage NSCLC (65). When surgery or surgery followed by adjuvant chemotherapy is the initial plan for definitive treatment, testing for genetic markers predicting response to targeted therapy is not clearly indicated. However, between 9% and 40% of patients who appear to have limited-stage disease at the time of diagnosis experience cancer recurrence after complete resection (66-68). Targeted therapy may be useful for certain patients who experience such recurrences, but clinical factors alone cannot predict which patients are likely to respond to targeted treatment (8, 19). For this reason, the practice of tissue banking, if available, is a good way to allow for genetic testing at a later date without the need for a repeat biopsy for patients who experience recurrence after resection.

For patients who have advanced or metastatic NSCLC at the time of diagnosis, the histologic type of the tumor should also be used as an early screening step to determine which patients should receive genetic testing. Because the currently available genetic tests that predict response to available targeted therapies are few and largely restricted to adenocarcinoma of the lung, patients with adenocarcinoma (or non-squamous NSCLC) should be considered for additional genetic testing. Patients with squamous cell lung cancer have not yet been conclusively shown to derive benefit from targeted therapy, so genetic testing should be deferred for these patients. However, patients with squamous cell lung cancer may also benefit from tissue banking; if ongoing research leads to new targeted therapies for squamous cell lung cancer, then genetic testing could be performed at a later date if needed without a repeat biopsy.

Patients who have locally advanced or metastatic adenocarcinoma of the lung should undergo genetic testing. EGFR mutations are much more common than the EML4-ALK translocation in lung adenocarcinoma, and the 2 mutations are very unlikely to be found in the same tumor (18, 19, 21, 22, 60). Therefore, a good argument can be made for performing EGFR testing first because it is more likely to be positive, which would avoid the cost of performing the EML4-ALK test. If EGFR mutation analysis is positive for a mutation predicting susceptibility to targeted therapy, then no additional testing is needed and the patient can receive treatment with erlotinib or gefitinib. If EGFR mutation testing is negative, then testing the tumor for the EML4-ALK translocation using the break-apart FISH assay is the next appropriate step. If FISH assay results for the EML4-ALK translocation are positive, then the patient can then receive treatment with crizotinib. If the test results are negative, then standard cytotoxic chemotherapy is appropriate.

It should also be noted that genetic testing in patients with NSCLC that does not fit the parameters of this recommendation (e.g., limited stage or squamous tumors) may be appropriate in the context of clinical trials.

Conclusion

NSCLC is a heterogeneous disease with varying biological behavior driven by differing genetic alterations that determine malignant behavior. Although many of the mutations involved in oncogenic pathways have been found, few of these mutations are truly useful genetic markers for clinical practice, and currently the useful mutations are found primarily in lung adenocarcinoma. Outside the setting of clinical trials, clinicians should order only genetic tests that yield information that can lead to a change in treatment and thereby potentially improve outcomes. We recommend genetic testing for locally advanced and metastatic adenocarcinomas of the lung as outlined above in Figure 2. In appropriately selected patients, we recommend testing for EGFR mutations and then for the EML4-ALK translocation if EGFR testing is negative to determine whether targeted therapy with EGFR TKIs or crizotinib is indicated. We also recommend tissue banking, if it is available, for patients with early-stage lung cancer or squamous cell lung cancer so that tissue is available for future testing in case of recurrence or new advances in targeted therapy. As clinical trials of more therapies targeted toward driver mutations in NSCLC are conducted, more such genetic tests will likely be incorporated into routine clinical practice.

Key points.

  • Useful genetic tests identify driver mutations which are common in the population of interest and indicate a pathway for which effective targeted therapy exists

  • The identified useful genetic tests in non-small cell lung cancer are found predominantly in adenocarcinoma of the lung

  • Clinical characteristics in the absence of mutation testing are not sufficient to predict response to targeted therapy, but clinical characteristics can be used to select patients who are appropriate for genetic testing

  • Erlotinib and gefitinib target cancers harboring the EGFR mutation and crizotinib targets those harboring the EML4-ALK translocation

Acknowledgments

The University of Texas MD Anderson Cancer Center is supported in part by a Cancer Center Support Grant (CA016672) from the National Institutes of Health.

Footnotes

The authors report no relevant conflicts of interest related to the content of this article.

References

  • 1.Group USCSW. United States Cancer Statistics:1999-2008 Incidence and Mortality Web-based Report. Atlanta, GA: U. S. Department of Health and Human Services, Centers for Disease Control and Prevention, and National Cancer Institute; Jan 12, 2012. Available from: www.cdc.gov/uscs. [Google Scholar]
  • 2.Alberts WM. Diagnosis and management of lung cancer executive summary: ACCP evidence-based clinical practice guidelines. Chest. (2nd) 2007 Sep;132(3 Suppl):1S–19S. doi: 10.1378/chest.07-1860. [DOI] [PubMed] [Google Scholar]
  • *3.Riess JW, Wakelee HA. Metastatic non-small cell lung cancer management: novel targets and recent clinical advances. Clin Adv Hematol Oncol. 2012 Apr;10(4):226–34. A brief review focusing on current targeted therapy, promising mutations being studied as potential targets for future personalized therapy, and maintenance therapy. [PubMed] [Google Scholar]
  • 4.Torkamani A, Schork NJ. Prediction of cancer driver mutations in protein kinases. Cancer Res. 2008 Mar 15;68(6):1675–82. doi: 10.1158/0008-5472.CAN-07-5283. [DOI] [PubMed] [Google Scholar]
  • 5.Ma PC. Personalized targeted therapy in advanced non-small cell lung cancer. Cleve Clin J Med. 2012 May;79(Electronic Suppl 1):eS56–60. doi: 10.3949/ccjm.79.s2.12. [DOI] [PubMed] [Google Scholar]
  • 6.Pao W. New approaches to targeted therapy in lung cancer. Proc Am Thorac Soc. 2012 May;9(2):72–3. doi: 10.1513/pats.201112-054MS. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *7.Beasley MB, Milton DT. ASCO Provisional Clinical Opinion: Epidermal Growth Factor Receptor Mutation Testing in Practice. J Oncol Pract. 2011 May;7(3):202–4. doi: 10.1200/JOP.2010.000166. Short review covering various methods of EGFR mutation testing from a pathologist's perspective. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *8.Shaw AT, Solomon B, Kenudson MM. Crizotinib and testing for ALK. J Natl Compr Canc Netw. 2011 Dec;9(12):1335–41. doi: 10.6004/jnccn.2011.0115. Overview of the available methods of performing ALK translocation testing. [DOI] [PubMed] [Google Scholar]
  • *9.Billah S, Stewart J, Staerkel G, Chen S, Gong Y, Guo M. EGFR and KRAS mutations in lung carcinoma: molecular testing by using cytology specimens. Cancer Cytopathol. 2011 Apr 25;119(2):111–7. doi: 10.1002/cncy.20151. Retrospective study demonstrating the suitability of cytologic specimens for EGFR and KRAS mutation testing in lung cancer. [DOI] [PubMed] [Google Scholar]
  • 10.Nakajima T, Yasufuku K, Suzuki M, Hiroshima K, Kubo R, Mohammed S, et al. Assessment of epidermal growth factor receptor mutation by endobronchial ultrasound-guided transbronchial needle aspiration. Chest. 2007 Aug;132(2):597–602. doi: 10.1378/chest.07-0095. [DOI] [PubMed] [Google Scholar]
  • 11.Allegrini S, Antona J, Mezzapelle R, Miglio U, Paganotti A, Veggiani C, et al. Epidermal growth factor receptor gene analysis with a highly sensitive molecular assay in routine cytologic specimens of lung adenocarcinoma. Am J Clin Pathol. 2012 Sep;138(3):377–81. doi: 10.1309/AJCPVAGIUC1AHC3Y. [DOI] [PubMed] [Google Scholar]
  • 12.Garcia-Olive I, Monso E, Andreo F, Sanz-Santos J, Taron M, Molina-Vila MA, et al. Endobronchial ultrasound-guided transbronchial needle aspiration for identifying EGFR mutations. Eur Respir J. 2010 Feb;35(2):391–5. doi: 10.1183/09031936.00028109. [DOI] [PubMed] [Google Scholar]
  • *13.Navani N, Brown JM, Nankivell M, Woolhouse I, Harrison RN, Jeebun V, et al. Suitability of endobronchial ultrasound-guided transbronchial needle aspiration specimens for subtyping and genotyping of non-small cell lung cancer: a multicenter study of 774 patients. Am J Respir Crit Care Med. 2012 Jun 15;185(12):1316–22. doi: 10.1164/rccm.201202-0294OC. Very large multicenter study confirming the suitability of cytologic specimens obtained via endobronchial ultrasound with needle aspiration for EGFR mutation testing and histologic subtyping in patients with non-small cell lung cancer. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Sakairi Y, Nakajima T, Yasufuku K, Ikebe D, Kageyama H, Soda M, et al. EML4-ALK fusion gene assessment using metastatic lymph node samples obtained by endobronchial ultrasound-guided transbronchial needle aspiration. Clin Cancer Res. 2010 Oct 15;16(20):4938–45. doi: 10.1158/1078-0432.CCR-10-0099. [DOI] [PubMed] [Google Scholar]
  • 15.Nakajima T, Yasufuku K, Nakagawara A, Kimura H, Yoshino I. Multigene mutation analysis of metastatic lymph nodes in non-small cell lung cancer diagnosed by endobronchial ultrasound-guided transbronchial needle aspiration. Chest. 2011 Nov;140(5):1319–24. doi: 10.1378/chest.10-3186. [DOI] [PubMed] [Google Scholar]
  • 16.Santis G, Angell R, Nickless G, Quinn A, Herbert A, Cane P, et al. Screening for EGFR and KRAS mutations in endobronchial ultrasound derived transbronchial needle aspirates in non-small cell lung cancer using COLD-PCR. PLoS One. 2011;6(9):e25191. doi: 10.1371/journal.pone.0025191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *17.Perez-Moreno P, Brambilla E, Thomas R, Soria JC. Squamous cell carcinoma of the lung: molecular subtypes and therapeutic opportunities. Clin Cancer Res. 2012 May 1;18(9):2443–51. doi: 10.1158/1078-0432.CCR-11-2370. Review reporting molecular profiles of squamous cell cancers and potential future targets for personalized therapy in squamous lung cancers. [DOI] [PubMed] [Google Scholar]
  • 18.Rosell R, Moran T, Queralt C, Porta R, Cardenal F, Camps C, et al. Screening for epidermal growth factor receptor mutations in lung cancer. N Engl J Med. 2009 Sep 3;361(10):958–67. doi: 10.1056/NEJMoa0904554. [DOI] [PubMed] [Google Scholar]
  • *19.Dogan S, Shen R, Ang DC, Johnson ML, D'Angelo SP, Paik PK, et al. Molecular Epidemiology of EGFR and KRAS Mutations in 3,026 Lung Adenocarcinomas: Higher Susceptibility of Women to Smoking-Related KRAS-Mutant Cancers. Clin Cancer Res. 2012 Nov 15;18(22):6169–77. doi: 10.1158/1078-0432.CCR-11-3265. Study of 3026 patients with lung adenocarcinoma reporting the epidemiology of EGFR and KRAS mutations in this cohort. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *20.Paik PK, Varghese AM, Sima CS, Moreira AL, Ladanyi M, Kris MG, et al. Response to Erlotinib in Patients with EGFR Mutant Advanced Non-Small Cell Lung Cancers with a Squamous or Squamous-like Component. Mol Cancer Ther. 2012 Nov;11(11):2535–40. doi: 10.1158/1535-7163.MCT-12-0163. Brief paper reporting the promising response of adenosquamous lung cancer to erlotinib in a small group of patients. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Marchetti A, Martella C, Felicioni L, Barassi F, Salvatore S, Chella A, et al. EGFR mutations in non-small-cell lung cancer: analysis of a large series of cases and development of a rapid and sensitive method for diagnostic screening with potential implications on pharmacologic treatment. J Clin Oncol. 2005 Feb 1;23(4):857–65. doi: 10.1200/JCO.2005.08.043. [DOI] [PubMed] [Google Scholar]
  • *22.D'Angelo SP, Pietanza MC, Johnson ML, Riely GJ, Miller VA, Sima CS, et al. Incidence of EGFR exon 19 deletions and L858R in tumor specimens from men and cigarette smokers with lung adenocarcinomas. J Clin Oncol. 2011 May 20;29(15):2066–70. doi: 10.1200/JCO.2010.32.6181. Paper in which 2142 patients with lung adenocarcinoma were evaluated for EGFR mutation and the correlation between EGFR mutation status and clinical characteristics was calculated. Although EGFR mutations were more common in smokers and women, a significant proportion were still found in men and smokers. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Sordella R, Bell DW, Haber DA, Settleman J. Gefitinib-sensitizing EGFR mutations in lung cancer activate anti-apoptotic pathways. Science. 2004 Aug 20;305(5687):1163–7. doi: 10.1126/science.1101637. [DOI] [PubMed] [Google Scholar]
  • 24.Riely GJ, Politi KA, Miller VA, Pao W. Update on epidermal growth factor receptor mutations in non-small cell lung cancer. Clin Cancer Res. 2006 Dec 15;12(24):7232–41. doi: 10.1158/1078-0432.CCR-06-0658. [DOI] [PubMed] [Google Scholar]
  • 25.Haura EB, Zheng Z, Song L, Cantor A, Bepler G. Activated epidermal growth factor receptor-Stat-3 signaling promotes tumor survival in vivo in non-small cell lung cancer. Clin Cancer Res. 2005 Dec 1;11(23):8288–94. doi: 10.1158/1078-0432.CCR-05-0827. [DOI] [PubMed] [Google Scholar]
  • 26.Greulich H, Chen TH, Feng W, Janne PA, Alvarez JV, Zappaterra M, et al. Oncogenic transformation by inhibitor-sensitive and -resistant EGFR mutants. PLoS Med. 2005 Nov;2(11):e313. doi: 10.1371/journal.pmed.0020313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Eberhard DA, Johnson BE, Amler LC, Goddard AD, Heldens SL, Herbst RS, et al. Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. J Clin Oncol. 2005 Sep 1;23(25):5900–9. doi: 10.1200/JCO.2005.02.857. [DOI] [PubMed] [Google Scholar]
  • 28.Jackman DM, Miller VA, Cioffredi LA, Yeap BY, Janne PA, Riely GJ, et al. Impact of epidermal growth factor receptor and KRAS mutations on clinical outcomes in previously untreated non-small cell lung cancer patients: results of an online tumor registry of clinical trials. Clin Cancer Res. 2009 Aug 15;15(16):5267–73. doi: 10.1158/1078-0432.CCR-09-0888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Paz-Ares L, Soulieres D, Melezinek I, Moecks J, Keil L, Mok T, et al. Clinical outcomes in non-small-cell lung cancer patients with EGFR mutations: pooled analysis. J Cell Mol Med. 2010 Jan;14(1-2):51–69. doi: 10.1111/j.1582-4934.2009.00991.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Tsao MS, Sakurada A, Cutz JC, Zhu CQ, Kamel-Reid S, Squire J, et al. Erlotinib in lung cancer - molecular and clinical predictors of outcome. N Engl J Med. 2005 Jul 14;353(2):133–44. doi: 10.1056/NEJMoa050736. [DOI] [PubMed] [Google Scholar]
  • 31.Sholl LM, Xiao Y, Joshi V, Yeap BY, Cioffredi LA, Jackman DM, et al. EGFR mutation is a better predictor of response to tyrosine kinase inhibitors in non-small cell lung carcinoma than FISH, CISH, and immunohistochemistry. Am J Clin Pathol. 2010 Jun;133(6):922–34. doi: 10.1309/AJCPST1CTHZS3PSZ. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Zheng Z, Bepler G, Cantor A, Haura EB. Small tumor size and limited smoking history predicts activated epidermal growth factor receptor in early-stage non-small cell lung cancer. Chest. 2005 Jul;128(1):308–16. doi: 10.1378/chest.128.1.308. [DOI] [PubMed] [Google Scholar]
  • 33.Mitsudomi T, Kosaka T, Endoh H, Horio Y, Hida T, Mori S, et al. Mutations of the epidermal growth factor receptor gene predict prolonged survival after gefitinib treatment in patients with non-small-cell lung cancer with postoperative recurrence. J Clin Oncol. 2005 Apr 10;23(11):2513–20. doi: 10.1200/JCO.2005.00.992. [DOI] [PubMed] [Google Scholar]
  • 34.Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004 May 20;350(21):2129–39. doi: 10.1056/NEJMoa040938. [DOI] [PubMed] [Google Scholar]
  • 35.Paez JG, Janne PA, Lee JC, Tracy S, Greulich H, Gabriel S, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 2004 Jun 4;304(5676):1497–500. doi: 10.1126/science.1099314. [DOI] [PubMed] [Google Scholar]
  • 36.Pao W, Miller V, Zakowski M, Doherty J, Politi K, Sarkaria I, et al. 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 Sep 7;101(36):13306–11. doi: 10.1073/pnas.0405220101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Marks JL, Broderick S, Zhou Q, Chitale D, Li AR, Zakowski MF, et al. Prognostic and therapeutic implications of EGFR and KRAS mutations in resected lung adenocarcinoma. J Thorac Oncol. 2008 Feb;3(2):111–6. doi: 10.1097/JTO.0b013e318160c607. [DOI] [PubMed] [Google Scholar]
  • *38.D'Angelo SP, Janjigian YY, Ahye N, Riely GJ, Chaft JE, Sima CS, et al. Distinct Clinical Course of EGFR-Mutant Resected Lung Cancers: Results of Testing of 1118 Surgical Specimens and Effects of Adjuvant Gefitinib and Erlotinib. J Thorac Oncol. 2012 Dec;7(12):1815–22. doi: 10.1097/JTO.0b013e31826bb7b2. Retrospective analysis of patients with resected lung adenocarcinoma suggests improved survival in the presence of EGFR mutations, especially if erlotinib or gefitinib are used as adjuvant therapy. KRAS mutations did not predict outcomes in this cohort. [DOI] [PubMed] [Google Scholar]
  • 39.Cohen MH, Williams GA, Sridhara R, Chen G, Pazdur R. FDA drug approval summary: gefitinib (ZD1839) (Iressa) tablets. Oncologist. 2003;8(4):303–6. doi: 10.1634/theoncologist.8-4-303. [DOI] [PubMed] [Google Scholar]
  • 40.Cohen MH, Johnson JR, Chen YF, Sridhara R, Pazdur R. FDA drug approval summary: erlotinib (Tarceva) tablets. Oncologist. 2005 Aug;10(7):461–6. doi: 10.1634/theoncologist.10-7-461. [DOI] [PubMed] [Google Scholar]
  • 41.Cohen MH, Johnson JR, Chattopadhyay S, Tang S, Justice R, Sridhara R, et al. Approval summary: erlotinib maintenance therapy of advanced/metastatic non-small cell lung cancer (NSCLC) Oncologist. 2010;15(12):1344–51. doi: 10.1634/theoncologist.2010-0257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009 Sep 3;361(10):947–57. doi: 10.1056/NEJMoa0810699. [DOI] [PubMed] [Google Scholar]
  • *43.Han JY, Park K, Kim SW, Lee DH, Kim HY, Kim HT, et al. First-SIGNAL: first-line single-agent iressa versus gemcitabine and cisplatin trial in never-smokers with adenocarcinoma of the lung. J Clin Oncol. 2012 Apr 1;30(10):1122–8. doi: 10.1200/JCO.2011.36.8456. This study showed that first-line treatment with gefitinib for advanced lung carcinoma was not significantly more effective than standard chemotherapy in clinically selected patients, but gefitinib was more effective in patients whose tumors harbored the EGFR mutation. This outcome echoes the results of the landmark IPASS trial. [DOI] [PubMed] [Google Scholar]
  • 44.Mitsudomi T, Morita S, Yatabe Y, Negoro S, Okamoto I, Tsurutani J, et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol. 2010 Feb;11(2):121–8. doi: 10.1016/S1470-2045(09)70364-X. [DOI] [PubMed] [Google Scholar]
  • 45.Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, Isobe H, et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med. 2010 Jun 24;362(25):2380–8. doi: 10.1056/NEJMoa0909530. [DOI] [PubMed] [Google Scholar]
  • 46.Katayama T, Matsuo K, Kosaka T, Sueda T, Yatabe Y, Mitsudomi T. Effect of gefitinib on the survival of patients with recurrence of lung adenocarcinoma after surgery: a retrospective case-matching cohort study. Surg Oncol. 2010 Dec;19(4):e144–9. doi: 10.1016/j.suronc.2010.07.002. [DOI] [PubMed] [Google Scholar]
  • *47.Zhang L, Ma S, Song X, Han B, Cheng Y, Huang C, et al. Gefitinib versus placebo as maintenance therapy in patients with locally advanced or metastatic non-small-cell lung cancer (INFORM; C-TONG 0804): a multicentre, double-blind randomised phase 3 trial. Lancet Oncol. 2012 May;13(5):466–75. doi: 10.1016/S1470-2045(12)70117-1. This trial demonstrated improved progression-free survival in clinically selected patients with advanced lung adenocarcinoma whose tumors responded to initial standard chemotheray and who received gefinitib as maintenance therapy. [DOI] [PubMed] [Google Scholar]
  • 48.Shepherd FA, Rodrigues Pereira J, Ciuleanu T, Tan EH, Hirsh V, Thongprasert S, et al. Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med. 2005 Jul 14;353(2):123–32. doi: 10.1056/NEJMoa050753. [DOI] [PubMed] [Google Scholar]
  • *49.Gridelli C, Ciardiello F, Gallo C, Feld R, Butts C, Gebbia V, et al. First-line erlotinib followed by second-line cisplatin-gemcitabine chemotherapy in advanced non-small-cell lung cancer: the TORCH randomized trial. J Clin Oncol. 2012 Aug 20;30(24):3002–11. doi: 10.1200/JCO.2011.41.2056. This trial assessed erlotinib as a first-line treatment for advanced non-small cell lung cancer in unselected patients and demonstrated that it was inferior to standard chemotherapy in that population. [DOI] [PubMed] [Google Scholar]
  • *50.Zhou C, Wu YL, Chen G, Feng J, Liu XQ, Wang C, et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol. 2011 Aug;12(8):735–42. doi: 10.1016/S1470-2045(11)70184-X. This randomized phase III trial demonstrated the superiority of erlotinib over standard chemotherapy as a first-line treatment for Asian patients with advanced non-small cell lung cancer in the presence of a sensitizing EGFR mutation. [DOI] [PubMed] [Google Scholar]
  • *51.Rosell R, Carcereny E, Gervais R, Vergnenegre A, Massuti B, Felip E, et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2012 Mar;13(3):239–46. doi: 10.1016/S1470-2045(11)70393-X. This randomized trial confirmed the superiority of erlotinib over standard chemotherapy as a first-line treatment for European patients with advanced non-small cell lung cancer and EGFR mutations. [DOI] [PubMed] [Google Scholar]
  • *52.Janjigian YY, Park BJ, Zakowski MF, Ladanyi M, Pao W, D'Angelo SP, et al. Impact on disease-free survival of adjuvant erlotinib or gefitinib in patients with resected lung adenocarcinomas that harbor EGFR mutations. J Thorac Oncol. 2011 Mar;6(3):569–75. doi: 10.1097/JTO.0b013e318202bffe. Retrospective study of a cohort of patients with lung adenocarcinomas that were fully resected. Patients in this cohort whose tumors harbored EGFR mutations who received targeted therapy had slightly increased disease-free survival durations compared with the other groups. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Komaki R, Blumenschein GR, Wistuba II, Lee JJ, Allen P, Wei X, et al. Phase II trial of erlotinib and radiotherapy following chemotherapy for patients with stage III non-small cell lung cancer. J Clin Oncol. 2011;29(suppl) abstr 7020. [Google Scholar]
  • 54.Costa DB, Nguyen KS, Cho BC, Sequist LV, Jackman DM, Riely GJ, et al. Effects of erlotinib in EGFR mutated non-small cell lung cancers with resistance to gefitinib. Clin Cancer Res. 2008 Nov 1;14(21):7060–7. doi: 10.1158/1078-0432.CCR-08-1455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *55.Janne PA, Wang X, Socinski MA, Crawford J, Stinchcombe TE, Gu L, et al. Randomized phase II trial of erlotinib alone or with carboplatin and paclitaxel in patients who were never or light former smokers with advanced lung adenocarcinoma: CALGB 30406 trial. J Clin Oncol. 2012 Jun 10;30(17):2063–9. doi: 10.1200/JCO.2011.40.1315. This trial comparing erlotinib alone versus erlotinib in combination with chemotherapy in clinically selected patients found similar progression-free survival durations in both arms, reinforcing the idea that using clinical predictors without mutation analysis is insufficient to predict which patients will respond to targeted therapy with EGFR tyrosine kinase inhibitors. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *56.Pillai RN, Ramalingam SS. The biology and clinical features of non-small cell lung cancers with EML4-ALK translocation. Curr Oncol Rep. 2012 Apr;14(2):105–10. doi: 10.1007/s11912-012-0213-4. Exclellent review covering the clinical features and biological characteristics of non-small cell lung cancer with the EML4-ALK translocation. [DOI] [PubMed] [Google Scholar]
  • 57.Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature. 2007 Aug 2;448(7153):561–6. doi: 10.1038/nature05945. [DOI] [PubMed] [Google Scholar]
  • 58.Inamura K, Takeuchi K, Togashi Y, Nomura K, Ninomiya H, Okui M, et al. EML4-ALK fusion is linked to histological characteristics in a subset of lung cancers. J Thorac Oncol. 2008 Jan;3(1):13–7. doi: 10.1097/JTO.0b013e31815e8b60. [DOI] [PubMed] [Google Scholar]
  • 59.Martelli MP, Sozzi G, Hernandez L, Pettirossi V, Navarro A, Conte D, et al. EML4-ALK rearrangement in non-small cell lung cancer and non-tumor lung tissues. Am J Pathol. 2009 Feb;174(2):661–70. doi: 10.2353/ajpath.2009.080755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Takahashi T, Sonobe M, Kobayashi M, Yoshizawa A, Menju T, Nakayama E, et al. Clinicopathologic features of non-small-cell lung cancer with EML4-ALK fusion gene. Ann Surg Oncol. 2010 Mar;17(3):889–97. doi: 10.1245/s10434-009-0808-7. [DOI] [PubMed] [Google Scholar]
  • 61.Shaw AT, Yeap BY, Mino-Kenudson M, Digumarthy SR, Costa DB, Heist RS, et al. Clinical features and outcome of patients with non-small-cell lung cancer who harbor EML4-ALK. J Clin Oncol. 2009 Sep 10;27(26):4247–53. doi: 10.1200/JCO.2009.22.6993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kwak EL, Bang YJ, Camidge DR, Shaw AT, Solomon B, Maki RG, et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med. 2010 Oct 28;363(18):1693–703. doi: 10.1056/NEJMoa1006448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Camidge DR, Bang YJ, Kwak EL, Shaw AT, Iafrate AJ, Maki RG, et al. Progression-free survival (PFS) from a phase I study of crizotinib (PF-02341066) in patients with ALK-positive non-small cell lung cancer (NSCLC) J Clin Oncol. 2011;29(suppl) Meeting abstract. abstr 2501. [Google Scholar]
  • *64.Shaw AT, Yeap BY, Solomon BJ, Riely GJ, Gainor J, Engelman JA, et al. Effect of crizotinib on overall survival in patients with advanced non-small-cell lung cancer harbouring ALK gene rearrangement: a retrospective analysis. Lancet Oncol. 2011 Oct;12(11):1004–12. doi: 10.1016/S1470-2045(11)70232-7. This retrospective analysis is the first study to show improved survivial with treatment with crizotinib in patients with non-small cell lung cancer and the ALK translocation. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Ettinger DS, Akerly W, Borghaei H, Chang AC, Cheney RT, Chirieac LR, et al. National Comprehensive Cancer Network Guidelines: Non-Small Cell Lung Cancer v.1.2013. Clinical Practice Guidelines. 2012 [Google Scholar]
  • 66.Martini N, Bains MS, Burt ME, Zakowski MF, McCormack P, Rusch VW, et al. Incidence of local recurrence and second primary tumors in resected stage I lung cancer. J Thorac Cardiovasc Surg. 1995 Jan;109(1):120–9. doi: 10.1016/S0022-5223(95)70427-2. [DOI] [PubMed] [Google Scholar]
  • 67.Maeda R, Yoshida J, Hishida T, Aokage K, Nishimura M, Nishiwaki Y, et al. Late recurrence of non-small cell lung cancer more than 5 years after complete resection: incidence and clinical implications in patient follow-up. Chest. 2010 Jul;138(1):145–50. doi: 10.1378/chest.09-2361. [DOI] [PubMed] [Google Scholar]
  • 68.Lopez Guerra JL, Gomez DR, Lin SH, Levy LB, Zhuang Y, Komaki R, et al. Risk factors for local and regional recurrence in patients with resected N0-N1 non-small-cell lung cancer, with implications for patient selection for adjuvant radiation therapy. Ann Oncol. 2012 doi: 10.1093/annonc/mds274. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES