ALK ambiguous-positive non-small cell lung cancers are tumors challenged by diagnostic and therapeutic issues

  • Authors:
    • Arnaud Uguen
    • Sophie Andrieu‑Key
    • Florence Vergne
    • Renaud Descourt
    • Gilles Quéré
    • Isabelle Quintin‑Roué
    • Stéphane Key
    • Paul Guéguen
    • Matthieu Talagas
    • Marc De Braekeleer
    • Pascale Marcorelles
  • View Affiliations

  • Published online on: July 21, 2016     https://doi.org/10.3892/or.2016.4962
  • Pages: 1427-1434
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Searching for ALK rearrangements using the approved fluorescent in situ hybridization (FISH) test and complementary immunohistochemistry (IHC) has become the rule to treat patients with advanced non‑small cell lung cancer (NSCLC) with anti‑ALK targeted therapy. The concordance between the two techniques is reported to be strong but imperfect. We report our experience with cases of ALK‑rearranged lung adenocarcinomas pointing out particularly ambiguous cases. FISH and IHC data on ALK but also c‑MET IHC as well as EGFR and KRAS mutation screening are considered, together with response to crizotinib treatment. We classified the 55 FISH ALK‑rearranged tumors into two groups according to the FISH and IHC results: a concordant FISH+IHC+ group (31 tumors) and an ambiguous group (24 tumors). These tumors were considered as ‘ambiguous’ ALK‑positive due to negative (21 tumors) or non‑contributive (3 tumors) IHC. In addition, the percentage of FISH-positive nuclei was between 15 and 20% in 17 tumors belonging to one or the other group (now called borderline tumors). We discuss the accuracy of the different tests with intent to determine whether ambiguous and borderline tumors are real positive ALK‑rearranged tumors. To conclude, ambiguous ALK‑positive lung cancers are challenging tumors with diagnosis and therapeutic issues that can justify parallel FISH, IHC and molecular screening strategy.

Introduction

Lung cancer remains a major cause of human mortality. Treatment of non-small cell lung cancer (NSCLC) is being improved by a better understanding of the molecular mechanisms involved in tumor initiation and progression, mainly in adenocarcinoma. The discovery of EGFR activating mutations and ALK rearrangements in a subset of NSCLC has led to major changes in the therapeutic strategy. Anti-EGFR and anti-ALK therapies achieve disease regression and improvement in survival in some patients (1,2). As a consequence, detection of ALK rearrangements, present in ~3–5% of NSCLC, has become mandatory to screen for patients who may benefit anti-ALK targeted therapy. Searching for an ALK-rearrangement using the Vysis LSI ALK Dual Color Break Apart fluorescence in situ hybridization (FISH) probe (Abbott Molecular, Rungis, France) is the Food and Drug Administration (FDA)-approved molecular test and is considered as the 'gold-standard'. ALK-rearranged NSCLC are defined as tumors with 15% or more nuclei with rearranged signals (first count in 50 nuclei and if considered as equivocal, i.e., 5–25 positive cells among these 50 first nuclei, the count must include 50 additional tumor nuclei) (3,4). In addition to FISH testing, many studies have suggested the use of ALK immunohistochemistry (IHC) and RT-PCR to detect ALK-rearranged cancers especially under the European guidelines. Although most of the studies have reported a close correlation between FISH and IHC, many of them, including the largest ones, have reported some discordance between both techniques (521). These discordances are all relevant as ALK FISH+IHC and ALK FISHIHC+ patients may respond to anti-ALK therapy (6,10,15). Additional methods such as next generation sequencing and real-time polymerase chain reaction have been proposed as complementary or even replacement techniques for ALK screening. IHC with different antibodies or FISH with other probes or brightfield combined IHC-in situ hybridization were also proposed to improve the diagnostic accuracy (8,15,19,2226). Nevertheless, discrepant cases are still described. Recently, some authors introduced the concept of 'borderline' ALK-positive because of ALK FISH percentages of rearranged nuclei close to the threshold of 15% and of 'ALK-equivocal' tumors to describe tumors with challenging ALK FISH and/or ALK IHC analysis results, and/or discrepancies between FISH and IHC (10,15). Some, but not all, of these 'ambiguous' ALK tumors respond to crizotinib treatment, all the more if they also strongly express c-MET, another potential target of crizotinib (6,10,15). ALK screening strategy is still debated to maximize ALK-rearranged NSCLC detection and to minimize ALK false positivity. Ambiguous ALK-rearranged tumors represent a major diagnostic and therapeutic challenge.

In this study, we report our experience in ALK rearrangement screening in lung adenocarcinoma using the FDA-approved FISH probe and IHC. Clinical outcomes of the crizotinib-treated patients were also reported. This study identifies and describes the issues concerning ambiguous ALK-rearranged tumors.

Materials and methods

Cases studied

We included all the ALK-rearranged adenocarcinoma cases identified by FISH and diagnosed at the University Hospital Morvan cancer molecular genetics platform from January 2010 to December 2014 for which sufficient tumor material was available to perform IHC analyses. These specimens (primary tumors and metastases) were formalin-fixed and paraffin embedded (FFPE). ALK analyses were conducted as part of the diagnostic work-up for the therapeutic management of patients with advanced stages of NSCLC according the French National Cancer Institute guidelines, together with EGFR and KRAS mutation screening. c-MET and complementary anti-ALK IHC analyses with different antibodies were also performed on samples with sufficient amount of tumor cells. The present study was conducted following our national and institutional guidelines. All samples were included in a registered tumour tissue collection and the present study was conducted in compliance with the Helsinki Declaration and after approval by our Institutional Review Board (CHRU Brest, CPP n° DC-2008-214). Response to crizotinib treatment was provided by the oncologists in charge of the therapeutic management of the ALK-positive patients. Therapy response was quoted by the oncologists assuming the clinical follow-up of the patients using clinical and radiological criteria, as used in other ALK-NSCLC dedicated studies.

ALK fluorescent in situ hybridization (FISH)

Tissue sections 3-µm thick were laid on SuperFrost® Plus slides. After deparaffinization, the slides were pre-treated with Dako Histology FISH Accessory kit (Dako, Glostrup, Denmark) following the manufacturer's instructions. Slides were washed in distilled water and dehydrated in increasing concentrations of alcohol (70, 90 and 100%) and air-dried at room temperature. Ten microliters of the Vysis LSI ALK Dual Color Break Apart Rearrangement Probe was placed onto the tissue sections. Slides were denaturated at 73°C for 5 min and then hybridized at 37°C for 16 h on a Dako Hybridizer. Following hybridization, the slides were washed with buffer, counter-colored with 4′,6-diamidino-2-phenylindole (DAPI) solution and coverslipped. They were then read using an epifluorescence microscope (Zeiss, Le Pecq, France) connected to a CCD camera and software for analyzing fluorescent signals (ISIS software; MetaSystems, Altlussheim, Germany).

At least 50 tumor nuclei (and, if required, 100 tumor nuclei following the FISH test guidelines) were assessed for each case considering the following criteria: ALK FISH was considered positive (i.e., ALK-rearranged) if there was a split between the orange (3′-end) and the green (5′-end) signals (i.e., orange and green signals being two or more signals apart) or an isolated single orange signal in ≥15% of tumor nuclei. We also noted the mean ALK copy number in tumor nuclei (counting both fused ALK and single 3′ALK signals).

ALK and c-MET immunohistochemistry

First line IHC was performed using the monoclonal antibody anti-ALK p80 (clone 5A4; CliniSciences, Nanterre, France) at a dilution of 1:25. Immunohistochemistry was performed on Ventana Benchmark XT® automated slide preparation system using OptiView DAB IHC Detection kit (both from Roche Diagnostics, Meylan, France). This IHC has successfully obtained European and French external quality controls. Briefly, IHC was performed on 3-µm thick tissue sections. OptiView® DAB IHC Detection kit was used according to Ventana staining procedure including pre-treatment with cell conditioner 1 for 92 min, followed by incubation with diluted antibody at 37°C for 1 h. Antibody incubation and signal amplification steps were followed by counterstaining with one drop of hematoxylin for 20 min and one drop of bluing reagent for 4 min. Subsequently, the slides were removed from the immunostainer, washed in water with dishwashing detergent, and mounted. Immunostaining was scored as negative (score 0), or as positive with faint staining (score 1+), moderate (score 2+) or intense (score 3+) staining of the tumor cells.

Samples with a sufficient amount of tumors cells were analyzed with additional IHC using the same IHC protocol with two other anti-ALK antibodies (clone D5F3, prediluted, Ventana, Roche Diagnostics; clone 1A4, 1:100, Origene, Rockville, MD, USA) and with anti-c-MET antibody (clone SP44, prediluted, Ventana, Roche Diagnostics) following the manufacturer's instructions.

Results

Cases included

Fifty-five ALK FISH-positive tumors from 55 patients, including 24 treated with crizotinib, were included in our study. Data concerning these 55 tumors and patients, including their response to crizotinib, are summarized in Table I. The 55 patients consisted of 30 men and 25 women with a mean age of 61 years (range, 28 to 88 years). Thirty-seven patients had a history of present or past smoking and 14 were never-smokers (no data for 4 patients). In addition to ALK FISH positivity, a KRAS mutation was identified in 7 tumors and a EGFRL858R in another. Complete response to crizotinib was observed only in one patient (case 10). A partial response (i.e., tumor regression or stable disease) was noted in 16 patients. The disease continued to progress despite crizotinib treatment in the other 7 patients.

Table I

Summary of the 55 ALK-positive cases in fluorescent in situ hybridization.

Table I

Summary of the 55 ALK-positive cases in fluorescent in situ hybridization.

CaseGenderAgeSmokerKRASEGFRc-MET IHCALK FISH nuclei positive (%)Main ALK FISH positive patternALK IHC (5A4 clone)ALK IHC (D5F3 clone)ALK IHC (1A4 clone)Response to crizotinib
1M74Yes080SRS3+3+3+Progression
2M54NoNC75SRS3+NCNCPartial response
3F62No2+70SS3+3+3+Partial response
4M40No2+65SRS3+3+NCPartial response
5M51No3+31SS3+3+3+Partial response
6F65Yes3+95SRS3+3+NCNo crizotinib
7M64YesNC40SRS2+3+3+No crizotinib
8M74Yes2+72SS2+NCNCStable disease
9F32YesNC30SRS3+3+3+Progression
10F61Yes3+50SS2+3+3+Complete response
11F74No3+80SS2+2+3+Partial response
12F35No2+80SS2+3+3+Partial response
13F57Yes2+70SS2+NCNCPartial response
14F48NoNC45SRS2+NCNCPartial response
15F67No2+16 (B)SS2+3+3+Partial response
16F65Yes1+80SRS2+NCNCNo crizotinib
17F60ND2+70SS2+3+2+No crizotinib
18F70YesNC70SS2+3+2+No crizotinib
19M36YesNC64SRS2+3+2+No crizotinib
20M77NoNC63SS2+3+NCNo crizotinib
21F61YesNC40SS2+3+NCNo crizotinib
22M75NoNC35SS2+2+NCNo crizotinib
23F28Yes1+30SS2+3+3+No crizotinib
24M61NoNC15 (B)SS2+3+NCNo crizotinib
25M70YesNCNC3+15 (B)SS2+NCNCNo crizotinib
26M60YesNCNCNC30SS1+NCNCPartial response
27F80NoNC30SS1+NCNCPartial response
28M55YesNC99SRS1+2+2+No crizotinib
29M65YesNC25SRS1+NCNCNo crizotinib
30F78YesNC20 (B)SS1+1+NCNo crizotinib
31M60YesNCNC2+20 (B)SS1+NCNCNo crizotinib
32 (A)F62YesG12SNC20 (B)SS0NCNCStable disease
33 (A)M58YesG13C1+30SS000Progression
34 (A)M49YesNCNCNC25SS0NCNCProgression
35 (A)M74Yes3+17 (B)SRS001+Progression
36 (A)F51YesG12V2+16 (B)SRS0NC1+Progression
37 (A)M73Yes2+25SS000Partial response
38 (A)F64NoNCNCNC20(B)SS0NCNCPartial response
39 (A)M75NDNC80SS001+No crizotinib
40 (A)M63YesNC60SS001+No crizotinib
41 (A)M54YesNC47SS0NCNCNo crizotinib
42 (A)M58YesNC40SRS0NCNCNo crizotinib
43 (A)M53YesNC30SS000No crizotinib
44 (A)F80YesG12C3+30SS00NCNo crizotinib
45 (A)M58YesNC25SS00NCNo crizotinib
46 (A)F43YesNC20 (B)SS001+No crizotinib
47 (A)F78NoL858R3+20 (B)SRS0NCNCNo crizotinib
48 (A)M65NDNC18 (B)SS0NCNCNo crizotinib
49 (A)M78NDNC18 (B)SS00NCNo crizotinib
50 (A)M88YesG12DNC17 (B)SRS0NCNCNo crizotinib
51 (A)M61YesNC17 (B)SS001+No crizotinib
52 (A)F58YesNC16 (B)SS001+No crizotinib
53 (A)F57YesNC40SSNCNCNCPartial response
54 (A)F50YesG12V017 (B)SSNCNCNCProgression
55 (A)M45YesG13DNC25SSNCNCNCNo crizotinib

[i] F, female; M, male; ND, no data; NC, non-contributive analysis; -, no mutation; (A), so-called 'ambiguous case'; (B), so-called 'borderline' percentage; SS, split signals; SRS, single red signals.

ALK fluorescent in situ hydridization

Table II summarizes the main FISH results. The mean percentage of positive nuclei per tumor was 41.4% (from 15 to 99%). Split and isolated 3′ signals co-existed in most of the tumors (mostly split signal in 39 tumors and isolated 3′ signal in 16 samples). The percentage of positive nuclei were between 15 and 20% in 17 tumors. Only one (1/55-1.8%) tumor presented a high ALK copy number (i.e., >6 ALK copy numbers per nucleus). Fig. 1 presents examples of ALK FISH positive patterns.

Table II

Summary of the ALK FISH patterns and correlation with immunohistochemistry (5A4 clone) and response to anti-ALK targeted therapy.

Table II

Summary of the ALK FISH patterns and correlation with immunohistochemistry (5A4 clone) and response to anti-ALK targeted therapy.

Number of casesMain ALK rearrangement signals
Mean ALK copy no. per nucleus
SplitSingle 3′<6>6
ALK FISH+IHC+ Response+13103130
ALK FISH+IHC+ Response20220
ALK FISH+IHC+ Not treated16106160
ALK FISH+IHC Response+33030
ALK FISH+IHC Response42240
ALK FISH+IHC Not treated14113131a
ALK FISH+IHC NC Response+11010
ALK FISH+IHC NC Response11010
ALK FISH+IHC NC Not treated11010
Total563917551

a Case 47. Response includes stable disease.

ALK immunohistochemistry

ALK IHC using clone 5A4 was non-contributive in three cases (Table I). Twenty-one tumors (38.2%) were immunonegative. Thirty-one tumors were considered ALK positive (56.3%) with a 3+ staining in 7/31 cases, a 2+ staining in 18/31 cases and a 1+ staining in 6/31 cases. Additional IHC using clones D5F3 and 1A4 was contributive for only 33 and 24 cases, respectively, because of progressive cell depletion in small biopsies. IHC with clone D5F3 was positive in 21/33 (63.6%) tumors with the higher rate of strong 3+ staining intensity in 17/33 (51.5%) tumors. IHC with clone 1A4 was positive in 21/24 (87.5%) tumors. Twelve and three tumors remained immunonegative with clones D5F3 and 1A4, respectively. Table III summarizes the results of ALK IHC with different antibodies and examples of staining are shown in Fig. 2.

Table III

Summary of the results of ALK immunohistochemistry analyses.

Table III

Summary of the results of ALK immunohistochemistry analyses.

ALK antibodyInsufficient materialNegative (−)Score 1+Score 2+Score 3+No of positive cases/total (%)
Clone 5A4321618731/52 (59.6)
Clone D5F32212131721/33 (63.6)
Clone 1A4313741021/24 (87.5)
c-MET expression

c-MET IHC was performed in only 23/55 tumors because of cell depletion. Among these 23 samples, 18 samples were considered positive (i.e., 2+ or 3+ staining intensity) and 5 samples were considered negative (i.e., 1+ or 0 staining). Indeed, in our daily practice, we also screen every NSCLC patient for c-MET expression but not for c-MET amplification which is only performed when a clinician requires this information for inclusion in a c-MET-related specific treatment trial. Therefore, c-MET FISH was not performed in these ALK-positive NSCLC cases.

Correlation between response to crizotinib, FISH and IHC results

Table II summarizes the distribution of patients according to their response to crizotinib and ALK FISH and IHC results. Thirty-one patients had a concordant FISH+IHC+ status. Of note, two of the 15 treated ALK FISH+IHC+ patients did not respond to crizotinib (cases 1 and 9). Three of the 21 ALK FISH+IHC patients had a response to anti-ALK therapy. A partial response was observed in case 37 with 25% of positive tumor nuclei also expressing a 2+ staining with c-MET IHC. A stable disease was observed in case 32 having a 20% ALK FISH rearranged status and a KRAS G12S mutation. A partial response was observed in case 38 with 20% ALK FISH rearranged nuclei without contributive c-MET IHC and EGFR and KRAS mutational analyses.

Discussion

ALK-rearranged NSCLC are classically reported to be adenocarcinomas involving young and never-smoker patients, characterized by mucinous and cribriform histopathological features and the absence of association to EGFR or KRAS mutations (2731). Most of these ALK-rearranged NSCLC respond to crizotinib (32). A strong correlation between the FISH-rearranged status of the tumor and the expression of the ALK protein detected by IHC was reported in many studies (512,1421,30). In addition, the mean copy number of the ALK gene in ALK-rearranged tumor is admitted to be usually low, with <6 ALK copies per nucleus, in contrast to tumors lacking ALK rearrangement in which high ALK copy gain is frequent (33).

We classified the ALK-rearranged tumors in our study into two groups. The first group included ALK-rearranged tumors by both FISH and IHC positivity (FISH+IHC+), without high ALK copy gain and no EGFR and KRAS mutation. The second group, designated as ambiguous ALK-positive, contained those ALK positive tumors that did not correspond to these criteria (in fact mainly FISH+IHCcases). Seventeen tumors, so-called borderline tumors, had a percentage of rearranged nuclei ≤20% and were included in the first or second group. Most of these 'borderline' tumors were FISH+IHC but some were FISH+IHC+ (Table I) (10,15).

Ambiguous ALK phenotype is presented by tumors being positive for only FISH or IHC. Some large studies have pointed out a significant rate of discrepancies between FISH and IHC (6,11,12). In our study, 24 tumors could be considered as ALK 'ambiguous'-positive tumors because they were IHC negative or non-contributive. Four of the 9 patients among these 24 cases treated with crizotinib showed a response. In a large French study, only 53.3% (80/150) of ALK-positive tumors were FISH+IHC+ and 24% (36/150) were FISH+IHC; 19 tumors were FISHIHC+ and 15 FISH non-contributive IHC (6). Crizotinib-responders are reported among FISH+IHC and FISHIHC+ cases, pointing out that combining FISH and IHC is important to minimize the risk of ALK-testing false-negativity. Indeed, examples of crizotinib-responders are reported even in patients with rearrangement rates as high as 60% by FISH although they are IHC (6).

Confrontation of ALK status with EGFR and KRAS mutational status speaks in favor of an accurate screening strategy. In the study by Cabillic et al on 3,244 NSCLC, 8 (5.3%) and 14 (9.3%) of the 150 ALK-positive tumors were also mutated for EGFR and KRAS, respectively (6). Another French study reported a 7% rate (11/150) of ALK FISH+IHC tumors mutated for EGFR and KRAS genes (11). In our opinion, even if the concept of mutually exclusive mutations/rearrangements concerning ALK, EGFR and KRAS is widely accepted, the challenging cases of double mutants justify parallel analyses of these three genes instead of a multistep algorithm that would lead to analyze ALK only in EGFR and KRAS wild-type tumors. Moreover, ALK inhibitors are reported to be effective in patients with co-alterations in ALK and EGFR (34).

Tumors having a percentage of ALK-rearranged nuclei between 15 and 20%, in the so-called 'borderline' or 'equivocal' grey-zone, face a particular analytic issue. In our study, 17 tumors could be considered as borderline tumors. Three of the 6 patients treated with crizotinib showed a response. A study by Camidge et al on 13 ALK-positive patients among 73 patients was concordant with the threshold of 15% FISH-rearranged nuclei to consider a tumor as ALK-rearranged or not (35). In this study, the lowest percentage of rearranged nuclei in the so-called ALK-positive tumors was ~22% and the highest percentage in the ALK-negative tumors was ~10%. No tumor had a percentage of rearranged nuclei between 10 and 20%. Of note, up to 11% of rearranged nuclei were encountered within non-tumor areas (35). More recently, many studies reported tumors within this 'grey-zone' from 10 to 20% of rearranged tumor nuclei, with various combinations of discordance between FISH and IHC results. These studies also discussed the interest of using different FISH probes and anti-ALK antibodies (6,10,11,15). Detection of potential ALK-rearranged tumors that could benefit anti-ALK therapy beyond the threshold of 15% remains a challenging issue that justifies a systematic use of anti-ALK IHC complementary to ALK FISH to detect ALK FISHIHC+ cases (17,18). Indeed, a few cases with a rate as low as 5% of ALK-positive nuclei associated with IHC positivity are reported to respond to crizotinib therapy (11,15). As this grey-zone is really close to the percentage of ALK-rearranged nuclei observed in non-tumor tissue, one can hypothesize that some of these tumors with rearranged nuclei from 15 to 20% could be technical false-positive results. Nevertheless, crizotinib-responders were reported in these grey-zone borderline tumors supporting the biological significance of the FISH positivity (6,10,15). Some authors hypothesized that a high ALK copy number, and/or a c-MET expression in these ALK 'borderline' tumors could explain the response or absence of response of the patients to anti-ALK therapy. However, the biological relevance of these two additional molecular defects is still not clearly demonstrated (10). Intra-tumor heterogeneity was proposed to have implications in the detection of ALK-rearrangements (7,36). A combination of multiple FISH analyses with different probes was also proposed to allow enhancement of the detection of ALK rearrangements in borderline and ambiguous tumors (15). In our study, most of the ALK borderline tumors within this grey-zone were also ambiguous FISH+IHC tumors. Even if the IHC negative feature could be corrected using different antibodies in some samples, cell depletion can prevent efficient comparison of antibodies, as in our study. We tested the three supplementary antibodies in only half of the cases. Nevertheless, 7 samples initially considered FISH+IHC were weakly positive (1+) for at least one additional antibody.

Furthermore, cell depletion in small biopsies can hamper the carrying out of EGFR and KRAS molecular analyses, and in the near future from analyzing other oncogenes such as ROS1. The diagnostic strategy must take into account the problem of tiny biopsies, in concomitant molecular and IHC analyses. Tissue handling, processing and sectioning must be optimal to minimize tumor wastage (4).

To conclude, it is crucial to be aware of the therapeutic implications despite discordances between FISH and IHC in ALK ambiguous and borderline positive tumors. These lesions - with diagnostic and therapeutic issues because of potential response to anti-ALK targeted therapies - must be studied further to facilitate the diagnosis of ALK-rearranged tumors in an intent-to-treat strategy. Additional FISH analyses with bacterial artificial chromosome clones or reverse transcriptase-polymerase chain reaction targeting already known ALK fusion partners could be helpful to solve the issue of borderline and/or ambiguous ALK-positive tumors.

In the meantime, the issue remains partially unsolved. Nevertheless, our data clearly emphasize that, besides using different FISH probes to solve certain ambiguous cases, using different IHC could also help to elucidate some of the first-appearing discrepant data. Still, some discrepant cases remain unsolved and the prediction of a response or progression following crizotinib treatment in these challenging cases remains difficult. Clinicians and pathologists must be aware of these potential issues to reach a personalized diagnostic strategy in the era of personalized medicine. New sampling and additional FISH and IHC analyses are parts of this personalized diagnostic strategy.

Acknowledgments

This study was supported by the 'Omnium group'. The authors wish to thank Mrs. Stéphanie Bouvier, Ms. Sandrine Duigou and the Brest Biobank for their technical assistance in this study.

References

1 

Kwak EL, Bang YJ, Camidge DR, Shaw AT, Solomon B, Maki RG, Ou SH, Dezube BJ, Jänne PA, Costa DB, et al: Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med. 363:1693–1703. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, Fujiwara S, Watanabe H, Kurashina K, Hatanaka H, et al: Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature. 448:561–566. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Lindeman NI, Cagle PT, Beasley MB, Chitale DA, Dacic S, Giaccone G, Jenkins RB, Kwiatkowski DJ, Saldivar JS, Squire J, et al: Molecular testing guideline for selection of lung cancer patients for EGFR and ALK tyrosine kinase inhibitors: Guideline from the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. Arch Pathol Lab Med. 137:828–860. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Thunnissen E, Bubendorf L, Dietel M, Elmberger G, Kerr K, Lopez-Rios F, Moch H, Olszewski W, Pauwels P, Penault-Llorca F, et al: EML4-ALK testing in non-small cell carcinomas of the lung: A review with recommendations. Virchows Arch. 461:245–257. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Alì G, Proietti A, Pelliccioni S, Niccoli C, Lupi C, Sensi E, Giannini R, Borrelli N, Menghi M, Chella A, et al: ALK rearrangement in a large series of consecutive non-small cell lung cancers: Comparison between a new immunohistochemical approach and fluorescence in situ hybridization for the screening of patients eligible for crizotinib treatment. Arch Pathol Lab Med. 138:1449–1458. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Cabillic F, Gros A, Dugay F, Begueret H, Mesturoux L, Chiforeanu DC, Dufrenot L, Jauffret V, Dachary D, Corre R, et al: Parallel FISH and immunohistochemical studies of ALK status in 3244 non-small-cell lung cancers reveal major discordances. J Thorac Oncol. 9:295–306. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Conklin CM, Craddock KJ, Have C, Laskin J, Couture C and Ionescu DN: Immunohistochemistry is a reliable screening tool for identification of ALK rearrangement in non-small-cell lung carcinoma and is antibody dependent. J Thorac Oncol. 8:45–51. 2013. View Article : Google Scholar

8 

Hofman P, Ilie M, Hofman V, Roux S, Valent A, Bernheim A, Alifano M, Leroy-Ladurie F, Vaylet F, Rouquette I, et al: Immunohistochemistry to identify EGFR mutations or ALK rearrangements in patients with lung adenocarcinoma. Ann Oncol. 23:1738–1743. 2012. View Article : Google Scholar

9 

Hutarew G, Hauser-Kronberger C, Strasser F, Llenos IC and Dietze O: Immunohistochemistry as a screening tool for ALK rearrangement in NSCLC: Evaluation of five different ALK antibody clones and ALK FISH. Histopathology. 65:398–407. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Ilie MI, Bence C, Hofman V, Long-Mira E, Butori C, Bouhlel L, Lalvée S, Mouroux J, Poudenx M, Otto J, et al: Discrepancies between FISH and immunohistochemistry for assessment of the ALK status are associated with ALK 'borderline'-positive rearrangements or a high copy number: A potential major issue for anti-ALK therapeutic strategies. Ann Oncol. 26:238–244. 2015. View Article : Google Scholar

11 

Lantuejoul S, Rouquette I, Blons H, Le Stang N, Ilie M, Begueret H, Grégoire V, Hofman P, Gros A, Garcia S, et al: French multicentric validation of ALK rearrangement diagnostic in 547 lung adenocarcinomas. Eur Respir J. 46:207–218. 2015. View Article : Google Scholar : PubMed/NCBI

12 

McLeer-Florin A, Moro-Sibilot D, Melis A, Salameire D, Lefebvre C, Ceccaldi F, de Fraipont F, Brambilla E and Lantuejoul S: Dual IHC and FISH testing for ALK gene rearrangement in lung adenocarcinomas in a routine practice: A French study. J Thorac Oncol. 7:348–354. 2012. View Article : Google Scholar

13 

Paik JH, Choe G, Kim H, Choe JY, Lee HJ, Lee CT, Lee JS, Jheon S and Chung JH: Screening of anaplastic lymphoma kinase rearrangement by immunohistochemistry in non-small cell lung cancer: Correlation with fluorescence in situ hybridization. J Thorac Oncol. 6:466–472. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Savic S, Diebold J, Zimmermann AK, Jochum W, Baschiera B, Grieshaber S, Tornillo L, Bisig B, Kerr K and Bubendorf L: Screening for ALK in non-small cell lung carcinomas: 5A4 and D5F3 antibodies perform equally well, but combined use with FISH is recommended. Lung Cancer. 89:104–109. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Selinger C, Cooper W, Lum T, McNeil C, Morey A, Waring P, Amanuel B, Millward M, Peverall J, Van Vliet C, et al: Equivocal ALK fluorescence in-situ hybridization (FISH) cases may benefit from ancillary ALK FISH probe testing. Histopathology. 67:654–663. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Selinger CI, Rogers TM, Russell PA, O'Toole S, Yip P, Wright GM, Wainer Z, Horvath LG, Boyer M, McCaughan B, et al: Testing for ALK rearrangement in lung adenocarcinoma: A multicenter comparison of immunohistochemistry and fluorescent in situ hybridization. Mod Pathol. 26:1545–1553. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Sholl LM, Aisner DL, Varella-Garcia M, Berry LD, Dias-Santagata D, Wistuba II, Chen H, Fujimoto J, Kugler K, Franklin WA, et al LCMC Investigators: Multi-institutional oncogenic driver mutation analysis in lung adenocarcinoma: the lung cancer mutation consortium experience. J Thorac Oncol. 10:768–777. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Sullivan HC, Fisher KE, Hoffa AL, Wang J, Saxe D, Siddiqui MT and Cohen C: The role of immunohistochemical analysis in the evaluation of EML4-ALK gene rearrangement in lung cancer. Appl Immunohistochem Mol Morphol. 23:239–244. 2015. View Article : Google Scholar

19 

Teixidó C, Karachaliou N, Peg V, Gimenez-Capitan A and Rosell R: Concordance of IHC, FISH and RT-PCR for EML4-ALK rearrangements. Transl Lung Cancer Res. 3:70–74. 2014.

20 

Wynes MW, Sholl LM, Dietel M, Schuuring E, Tsao MS, Yatabe Y, Tubbs RR and Hirsch FR: An international interpretation study using the ALK IHC antibody D5F3 and a sensitive detection kit demonstrates high concordance between ALK IHC and ALK FISH and between evaluators. J Thorac Oncol. 9:631–638. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Zwaenepoel K, Van Dongen A, Lambin S, Weyn C and Pauwels P: Detection of ALK expression in non-small-cell lung cancer with ALK gene rearrangements - comparison of multiple immunohistochemical methods. Histopathology. 65:539–548. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Gruber K, Horn H, Kalla J, Fritz P, Rosenwald A, Kohlhäufl M, Friedel G, Schwab M, Ott G and Kalla C: Detection of rearrangements and transcriptional up-regulation of ALK in FFPE lung cancer specimens using a novel, sensitive, quantitative reverse transcription polymerase chain reaction assay. J Thorac Oncol. 9:307–315. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Gruber K, Kohlhäufl M, Friedel G, Ott G and Kalla C: A novel, highly sensitive ALK antibody 1A4 facilitates effective screening for ALK rearrangements in lung adenocarcinomas by standard immunohistochemistry. J Thorac Oncol. 10:713–716. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Kim H, Yoo SB, Choe JY, Paik JH, Xu X, Nitta H, Zhang W, Grogan TM, Lee CT, Jheon S, et al: Detection of ALK gene rearrangement in non-small cell lung cancer: A comparison of fluorescence in situ hybridization and chromogenic in situ hybridization with correlation of ALK protein expression. J Thorac Oncol. 6:1359–1366. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Nitta H, Tsuta K, Yoshida A, Ho SN, Kelly BD, Murata LB, Kosmeder J, White K, Ehser S, Towne P, et al: New methods for ALK status diagnosis in non-small-cell lung cancer: An improved ALK immunohistochemical assay and a new, Brightfield, dual ALK IHC-in situ hybridization assay. J Thorac Oncol. 8:1019–1031. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Pekar-Zlotin M, Hirsch FR, Soussan-Gutman L, Ilouze M, Dvir A, Boyle T, Wynes M, Miller VA, Lipson D, Palmer GA, et al: Fluorescence in situ hybridization, immunohistochemistry, and next-generation sequencing for detection of EML4-ALK rearrangement in lung cancer. Oncologist. 20:316–322. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Gainor JF, Varghese AM, Ou SH, Kabraji S, Awad MM, Katayama R, Pawlak A, Mino-Kenudson M, Yeap BY, Riely GJ, et al: ALK rearrangements are mutually exclusive with mutations in EGFR or KRAS: An analysis of 1,683 patients with non-small cell lung cancer. Clin Cancer Res. 19:4273–4281. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Jokoji R, Yamasaki T, Minami S, Komuta K, Sakamaki Y, Takeuchi K and Tsujimoto M: Combination of morphological feature analysis and immunohistochemistry is useful for screening of EML4-ALK-positive lung adenocarcinoma. J Clin Pathol. 63:1066–1070. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Just PA, Cazes A, Audebourg A, Cessot A, Pallier K, Danel C, Vacher-Lavenu MC, Laurent-Puig P, Terris B and Blons H: Histologic subtypes, immunohistochemistry, FISH or molecular screening for the accurate diagnosis of ALK-rearrangement in lung cancer: A comprehensive study of Caucasian non-smokers. Lung Cancer. 76:309–315. 2012. View Article : Google Scholar

30 

Paik JH, Choi CM, Kim H, Jang SJ, Choe G, Kim DK, Kim HJ, Yoon H, Lee CT, Jheon S, et al: Clinicopathologic implication of ALK rearrangement in surgically resected lung cancer: A proposal of diagnostic algorithm for ALK-rearranged adenocarcinoma. Lung Cancer. 76:403–409. 2012. View Article : Google Scholar

31 

Shaw AT, Yeap BY, Mino-Kenudson M, Digumarthy SR, Costa DB, Heist RS, Solomon B, Stubbs H, Admane S, McDermott U, et al: Clinical features and outcome of patients with non-small-cell lung cancer who harbor EML4-ALK. J Clin Oncol. 27:4247–4253. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Solomon BJ, Mok T, Kim DW, Wu YL, Nakagawa K, Mekhail T, Felip E, Cappuzzo F, Paolini J, Usari T, et al PROFILE 1014 Investigators: First-line crizotinib versus chemotherapy in ALK-positive lung cancer. N Engl J Med. 371:2167–2177. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Salido M, Pijuan L, Martínez-Avilés L, Galván AB, Cañadas I, Rovira A, Zanui M, Martínez A, Longarón R, Sole F, et al: Increased ALK gene copy number and amplification are frequent in non-small cell lung cancer. J Thorac Oncol. 6:21–27. 2011. View Article : Google Scholar

34 

Won JK, Keam B, Koh J, Cho HJ, Jeon YK, Kim TM, Lee SH, Lee DS, Kim DW and Chung DH: Concomitant ALK translocation and EGFR mutation in lung cancer: A comparison of direct sequencing and sensitive assays and the impact on responsiveness to tyrosine kinase inhibitor. Ann Oncol. 26:348–354. 2015. View Article : Google Scholar

35 

Camidge DR, Kono SA, Flacco A, Tan AC, Doebele RC, Zhou Q, Crino L, Franklin WA and Varella-Garcia M: Optimizing the detection of lung cancer patients harboring anaplastic lymphoma kinase (ALK) gene rearrangements potentially suitable for ALK inhibitor treatment. Clin Cancer Res. 16:5581–5590. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Abe H, Kawahara A, Azuma K, Taira T, Takase Y, Fukumitsu C, Murata K, Yamaguchi T, Akiba J, Ishii H, et al: Heterogeneity of anaplastic lymphoma kinase gene rearrangement in non-small-cell lung carcinomas: A comparative study between small biopsy and excision samples. J Thorac Oncol. 10:800–805. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2016
Volume 36 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Uguen A, Andrieu‑Key S, Vergne F, Descourt R, Quéré G, Quintin‑Roué I, Key S, Guéguen P, Talagas M, De Braekeleer M, De Braekeleer M, et al: ALK ambiguous-positive non-small cell lung cancers are tumors challenged by diagnostic and therapeutic issues. Oncol Rep 36: 1427-1434, 2016
APA
Uguen, A., Andrieu‑Key, S., Vergne, F., Descourt, R., Quéré, G., Quintin‑Roué, I. ... Marcorelles, P. (2016). ALK ambiguous-positive non-small cell lung cancers are tumors challenged by diagnostic and therapeutic issues. Oncology Reports, 36, 1427-1434. https://doi.org/10.3892/or.2016.4962
MLA
Uguen, A., Andrieu‑Key, S., Vergne, F., Descourt, R., Quéré, G., Quintin‑Roué, I., Key, S., Guéguen, P., Talagas, M., De Braekeleer, M., Marcorelles, P."ALK ambiguous-positive non-small cell lung cancers are tumors challenged by diagnostic and therapeutic issues". Oncology Reports 36.3 (2016): 1427-1434.
Chicago
Uguen, A., Andrieu‑Key, S., Vergne, F., Descourt, R., Quéré, G., Quintin‑Roué, I., Key, S., Guéguen, P., Talagas, M., De Braekeleer, M., Marcorelles, P."ALK ambiguous-positive non-small cell lung cancers are tumors challenged by diagnostic and therapeutic issues". Oncology Reports 36, no. 3 (2016): 1427-1434. https://doi.org/10.3892/or.2016.4962