Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Apr 4.
Published in final edited form as: J Clin Oncol. 2011 Apr 11;29(15):1943–1945. doi: 10.1200/JCO.2010.34.1974

Genetic Testing for Lung Cancer: Reflex vs. Clinical Selection

Paul A Bunn Jr 1, Robert C Doebele 1
PMCID: PMC4819165  NIHMSID: NIHMS372980  PMID: 21483017

Interest in activating “driver” mutations in tyrosine kinase genes as therapeutic targets in non-small cell lung cancer (NSCLC) has peaked recently due to the discovery that EGFR tyrosine kinase inhibitors (TKIs) produce higher response rates, longer progression free survival (PFS) times, less toxicity, improved symptom control, improved quality of life, and greater convenience compared to cytotoxic chemotherapy in the first line treatment of advanced NSCLC patients harboring EGFR activating mutations (14). A more recent study showed that advanced NSCLC patients whose tumors harbor EML4-ALK fusion genes, who are treated with an ALK TKI termed crizotinib, have high response rates and long progression free survival times that are similar to those of NSCLC patients harboring EGFR mutations treated with erlotinib (5,6).

Recent genomic studies in adenocarcinoma of the lung identified other mutations in tyrosine kinase genes including KRAS, NRAS, BRAF, PI3KCA and others in frequencies exceeding 1% (715). These findings raised the possibility that other mutation specific TKIs could produce similar benefits in NSCLC patients whose tumors harbor these specific “driver” mutations. BRAF is one of the genes identified in these early reports with studies indicating the presence of BRAF mutations in 0% to 17% of tumors and cell lines. In these reports, 20 of 805 NSCLC tumors harbored BRAF mutations (2.5%) and many of these mutations were not the typical V600E mutations observed in malignant melanoma (815). Lung cancer cell lines had even higher rates of BRAF mutations with rates of 2% to 17% (14, 15). Preclinical model studies demonstrated that activating BRAF mutations were sufficient for the development of lung adenocarcinomas in mice (16, 17).

V600E BRAF mutations in malignant melanoma were shown to be present in an even higher percentage of cases (50–70%) (13, 18). Several studies reported that melanomas harboring BRAF mutations are dependent on MEK/ERK signaling (17, 19, 20). PLX4032, a potent inhibitor of oncogenic BRAF, was developed through structure guided discovery and selectively blocked RAF/MEK/ERK signaling in BRAF mutant cells and caused regression of BRAF mutant tumor xenografts (21). In a phase 1 study, PLX4032 inhibited ERK phosphorylation and produced an 81% response rate in melanoma patients with BRAF mutant tumors (22). These findings led to an extension phase of the PLX4032 study in 32 melanoma patients whose tumors had V600E BRAF mutations. There were 24 objective partial responses and 2 complete responses and the median PFS exceeded 7 months (23). Subsequent studies showed that acquired resistance to PLX 4032 developed by mutually exclusive PDGFR beta up-regulation and NRAS mutation and not through secondary mutations in BRAF (24).

In this issue of the JCO, Paik et al. report finding BRAF mutations in 18 of 673 (3%) adenocarcinomas of the lung (25). Although the frequency of BRAF mutations in NSCLC is lower than in melanoma, it is high enough to warrant evaluation BRAF specific inhibitors in this population. The 3% figure is in keeping with the 2.5% observed in other studies and the finding that half of the mutations were non-V600E is also consistent with previous reports. As the authors point out, a 3% rate could mean as many as 5600 new cases annually in the US and 35,000 annually worldwide. However, it may be imperative to distinguish between the V600E and non-V600E mutations. If there are differences in therapeutic response to current BRAF inhibitors between V600E and non-V600E, only about 1.5% of all NSCLCs may have each of these classes of BRAF mutation. Cell lines such as NCIH1755 that has a non-V600E, G469A BRAF mutation has been reported to be resistant to PLX4032 (21). Cells with non-V600E BRAF mutations may also be resistant to other V600E BRAF inhibitors such as GSK2118436. However, tumor cells with other BRAF mutations are sensitive to downstream pathways inhibitors such as MEK inhibitors (20). Thus, it is likely that it will be critical to determine the exact type of BRAF mutation.

Paik et al. also report association between clinical characteristics, BRAF mutation, and survival outcome. Unlike other oncogenic mutations reported to date, they found BRAF mutations occurred most often in former or current smokers compared to never smokers. It should be noted that previous studies have identified BRAF mutations in non-smokers (26, 27). Although EGFR activating mutations were initially identified in a subset of patients that shared clinical and histologic characteristics, a recent comprehensive study has shown that approximately 39% of all EGFR mutations occur in current and former smokers (28); thus basing clinical testing on unreliable clinical characteristics can deny patients the benefit of targeted therapy. The authors did not find a sex-specific association with BRAF mutations. One major limitation of this study is that only adenocarcinomas were evaluated for BRAF mutations. Previous studies have shown the presence of both exon 11 and exon 15 BRAF mutations in squamous cell carcinoma of the lung (26). There is poor concordance on histologic diagnosis, even among expert pathologists (29). Therefore, testing based on histology cannot be recommended. The survival outcome seems to be slightly superior compared to patients without such driver mutations and similar to the survival of patients with EGFR mutations but the number of patients was quite small and there was no uniformity of treatment.

One of the major implications of the association with clinical features is to determine a priority for molecular testing. Many argue that it may be most cost effective to test only the patients most likely to have a specific mutation. However, if one conducts serial analysis of each mutation individually, it may take a long time to rule out the presence of any of the mutations. Enrichment strategies may make sense when trying to identify patients for clinical trials with limited resources to screen large numbers of patients (30); however, newer technologies such as SNapshot and Sequenom allow testing of all of these mutations at the same time and with little additional expense compared to testing single mutations. Furthermore, there are reports of NSCLC patients with more than one oncogenic driver mutations, (e.g., EML4-ALK and EGFR or KRAS mutations) (31, 32). Patients with BRAF (or KRAS) mutations can also have inactivating mutations in the tumor suppressor gene, LKB1, and this may influence clinical benefit from BRAF TKIs (33). As more driver mutations are identified, we can expect the complexity of overlapping mutations to increase. This multiplexed approach is clearly practicable as the NCI-funded Lung Cancer mutation consortium is currently conducting simultaneous genetic testing for 10 oncogenic changes for which there are specific therapies. For example, there is a trial of GSK 2118436 for patients with BRAF V600E mutations and a trial of the MEK inhibitor GSK1120212 in patients with non-V600E BRAF mutations.

This study of mutation testing in NSCLC patients and others measuring other genetic changes, signal a new era of personalized medicine for patients with advanced lung cancer where is will be imperative to match the specific mutations of a patients tumor with a specific therapy. One challenge will be obtaining adequate tumor material at the time of diagnostic biopsies to ensure successful testing. Another challenge will be developing testing platforms that can analyze for the presence of somatic mutations, gene fusions, or other genetic changes simultaneously. It is likely that routine simultaneous (reflex) testing of multiple markers will be conducted on all patients prior to initiation of therapy, irrespective of their clinical features.

Acknowledgments

Supported in part by NCI SPORE CA 5 P50 CA 058187-15 And NCI GO CA RC2 CA 148394-02

References

  • 1.Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or caboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009;361:947–957. doi: 10.1056/NEJMoa0810699. [DOI] [PubMed] [Google Scholar]
  • 2.Maemondo M, Inoue A, Kobayashi K, et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med. 2010;362:2380–2388. doi: 10.1056/NEJMoa0909530. [DOI] [PubMed] [Google Scholar]
  • 3.Lee JS, Park K, Kim S, et al. A randomized phase III study of gefitinib (IRESSATM) versus standard chemotherapy (gemcitabine plus cisplatin) as a first-line treatment for never-smokers with advanced or metastatic adenocarcinoma of the lung. J Thorac Oncol. 2009;4(9):S283–S284. [Google Scholar]
  • 4.Zhou C, et al. Program and abstracts of the 35th European Society of Medical Oncology Congress. Milan, Italy: 2010. Oct, Efficacy results from the randomized phase 3 OPTIMAL (CTONG 0802) study comparing first-line erlotinib versus carboplatin (CBDCA) plus gemcitabine (GEM) in Chinese advanced non-small-cell lung cancer (NSCLC) patients (pts) with EGFR activating mutations. Abstr. LBA13. [Google Scholar]
  • 5.Kwak EL, Bang YJ, Camidge DR, et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med. 2010;363:1693–1703. doi: 10.1056/NEJMoa1006448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ou SH, Bazhenova L, Camidge DR, et al. Rapid and dramatic radiographic and clinical response to an ALK inhibitor (crizotinib, PF02341066) in an ALK translocation-positive patient with non-small cell lung cancer. J Thorac Oncol. 2010;5:2044–2046. doi: 10.1097/JTO.0b013e318200f9ff. [DOI] [PubMed] [Google Scholar]
  • 7.Ding L, Getz G, Wheeler DA, et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature. 2008;455:1069–1075. doi: 10.1038/nature07423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Sun Y, Ren Y, Fang Z, et al. Lung adenocarcinoma from East Asian never smokers is a disease largely defined by targetable oncogenic mutant kinases. J Clin Oncol. 2010;28:4616–4620. doi: 10.1200/JCO.2010.29.6038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Schmid K, Oehl N, Wiba F, et al. EGFR/KRAS/BRAF mutations in primary lung adenocarcinomas and corresponding locoregional lymph node metastases. Clin Cancer Res. 2009;5:4554–4560. doi: 10.1158/1078-0432.CCR-09-0089. [DOI] [PubMed] [Google Scholar]
  • 10.Yousem SA, Nikiforova M, Nikiforov Y. The histopathology of BRAFV600E mutated lung adenocarcinoma. Am J Surg Pathol. 2008;32:1317–1321. doi: 10.1097/PAS.0b013e31816597ca. [DOI] [PubMed] [Google Scholar]
  • 11.Sasaki H, Kawano O, Endo K, et al. Uncommon V599E mutations in Japanese patients with lung cancer. J Surg Res. 2006;133:203–206. doi: 10.1016/j.jss.2005.10.022. [DOI] [PubMed] [Google Scholar]
  • 12.Naoki K, Chen TH, Richards WG, et al. Missense mutations in the BRAF gene in human lung adenocarcinoma. Cancer Res. 2002;62:7001–7003. [PubMed] [Google Scholar]
  • 13.Brose MS, Volpe P, Feldman M, et al. BRAF and KRAS mutations in lung cancer and melanoma. Cancer Res. 2002;62:6997–7000. [PubMed] [Google Scholar]
  • 14.Soh J, Okumura N, Lockwood WW, et al. Oncogenic mutations, copy number gains and mutant allele specific imbalance (MASI) frequently occur together in tumor cells. PLoS One. 2009;4:e7464. doi: 10.1371/journal.pone.0007464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Blanco R, Iwakawa R, Tang M, et al. A gene alteration profile of human lung cancer cell lines. Hum Mutat. 2009;30:1199–1206. doi: 10.1002/humu.21028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Ji H, Wang Z, Perera SA, et al. Mutations in BRAF and KRAS converge on activation of the mitogen activated protein kinase pathway in lung cancer mouse models. Cancer Res. 2007;67:4993–4999. doi: 10.1158/0008-5472.CAN-06-4592. [DOI] [PubMed] [Google Scholar]
  • 17.Dankfort D, Filenova E, Collado M, et al. A new mouse model to explore the initiation, progression and therapy of BRAFV600E-induced lung tumors. Genes Dev. 2007;23:379–384. doi: 10.1101/gad.1516407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Wellbrock C, Hursstone A. BRAF as therapeutic target in melanoma. Biochem Pharmacology. 2010;80:561–567. doi: 10.1016/j.bcp.2010.03.019. [DOI] [PubMed] [Google Scholar]
  • 19.Solit DB, Garraway LA, Pratilas CA, et al. BRAF mutations predict sensitivity to MEK inhibition. Nature. 2006;49:358–362. doi: 10.1038/nature04304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Hatzivassilliou G, song K, Yen I, et al. RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth. Nature. 2010;464:431–435. doi: 10.1038/nature08833. [DOI] [PubMed] [Google Scholar]
  • 21.Yang H, Higquis B, Kolisky K, et al. RG 7204 (PL4032), a selective BRAF V600E inhibitor, dsiplays potent antitumor activity in preclinical melanoma models. Cancer Res. 2010;70:5518–5527. doi: 10.1158/0008-5472.CAN-10-0646. [DOI] [PubMed] [Google Scholar]
  • 22.Bollag G, Hirth P, Tsai J, et al. Clinical efficacy of a BRAF inhibitor needs broad target blockade in BRAF mutant melanoma. Nature. 2010;467:596–599. doi: 10.1038/nature09454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Flaherty KT, Puzanov I, Kim KB, et al. Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med. 2010;363:809–819. doi: 10.1056/NEJMoa1002011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Nazarian R, Shi H, Wang Q, et al. Melanomas acquire resistance to BRAF(V600E) inhibition by RTK or N-RAS upregulation. Nature. 2010;468:973–977. doi: 10.1038/nature09626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Pail PK, et al. Clinical characteristics of patients with lung adenocarcinomas harboring BRAF mutations. J Clin Oncol. 2010 doi: 10.1200/JCO.2010.33.1280. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Pratilas CA, Hanrahan AJ, Halillovic E, et al. Genetic predictors of MEK dependence in non-small cell lung cancer. Cancer Res. 2008;68:9375–9383. doi: 10.1158/0008-5472.CAN-08-2223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Yamamoto H, Shigematsu H, Nomura M, et al. PIK3CA mutations and copy number gains in human lung cancers. Cancer Res. 2008;68:6913–6921. doi: 10.1158/0008-5472.CAN-07-5084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Pietanza M, D’Angelo SP, Johnson ML, et al. EGFR mutations in men and cigarette smokers with lung adenocarcinoma. J Clin Oncol. 2010;28 doi: 10.1200/JCO.2010.32.6181. abstr. 10538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Field RW, Smith BJ, Platz CE, et al. Lung cancer histologic type in the surveillance, epidemiology and end results registry versus independent review. J Natl Cancer Inst. 2004;96:1105–1007. doi: 10.1093/jnci/djh189. [DOI] [PubMed] [Google Scholar]
  • 30.Camidge DR, Kone SA, Flacco A, et al. Optimizing the detection of lung cancer patients harboring anaplastic lymphoma kinase (ALK) gene rearrangements potentially suitable for ALK inhibitor treatment. Clin Cancer Res. 2010;16:5581–5590. doi: 10.1158/1078-0432.CCR-10-0851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Koivunen JP, Mermel C, Zejnullahu K, et al. EML4-ALK fusion gene and efficacy of an ALK kinase inhibitor in lung cancer. Clin Cancer Res. 2008;14:4275–4283. doi: 10.1158/1078-0432.CCR-08-0168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Martelli MP, Sozzi G, Hernandez L, et al. EML4-ALK rearrangement in non-small cell lung cancer and non-tumor lung tissues. Am J Pathol. 2009;174:661–670. doi: 10.2353/ajpath.2009.080755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Koivunen JP, Kim J, Lee J, et al. Mutations in the LKB1 tumour suppressor are frequently detected in tumours from Caucasian but not Asian lung cancer patients. Br J Cancer. 2008;99:245–252. doi: 10.1038/sj.bjc.6604469. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES