Accepted Manuscript Title: Parallel screening for ALK, MET and ROS1 alterations in non-small cell lung cancer with implications for daily routine testing* Author: Philipp Jurmeister Dido Lenze Erika Berg Stefanie Mende Frank Sch¨aper Udo Kellner Hermann Herbst Christine Sers Jan Budczies Manfred Dietel Michael Hummel Maximilian von Laffert PII: DOI: Reference:

S0169-5002(14)00491-7 http://dx.doi.org/doi:10.1016/j.lungcan.2014.11.018 LUNG 4739

To appear in:

Lung Cancer

Received date: Revised date: Accepted date:

17-9-2014 8-11-2014 30-11-2014

Please cite this article as: Jurmeister P, Lenze D, Berg E, Mende S, Sch¨aper F, Kellner U, Herbst H, Sers C, Budczies J, Dietel M, Hummel M, von Laffert M, Parallel screening for ALK, MET and ROS1 alterations in non-small cell lung cancer with implications for daily routine testing*, Lung Cancer (2014), http://dx.doi.org/10.1016/j.lungcan.2014.11.018 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

1

Parallel screening for ALK, MET and ROS1 alterations in non-small cell lung

2

cancer with implications for daily routine testing*

ip t

3

Philipp Jurmeistera, Dido Lenzea, Erika Berga, Stefanie Mendea, † Frank Schäper*b,

5

Udo Kellnerc, Hermann Herbstd, Christine Sersa, Jan Budcziesa, Manfred Dietela,

6

Michael Hummela and Maximilian von Lafferta

us

cr

4

7

* Dedicated to Frank Schäper (‫٭‬05.04.1963 - †17.01.2014)

an

8

M

9 10

a

11

Charitéplatz 1, 10117 Berlin, Germany

12

b

13

Lindenberger Weg 27, 13125 Berlin, Germany

14

c

15

32429 Minden, Germany

16

d

17

Berlin, Germany

te

d

Institute of Pathology, Campus Charité Mitte, Charité Universitätsmedizin Berlin,

Ac ce p

Pathology-Berlin, Bioptisches Institut Gemeinschaftspraxis für Pathologie,

Institute of Pathology, Johannes Wesling Klinikum Minden, Hans-Nolte-Straße 1,

Institute of Pathology, Vivantes Klinikum Berlin, Oranienburger Straße 285, 13437

18 19

Address of Correspondence

20

Maximilian von Laffert, MD

21

Institute of Pathology

22

Humboldt University Berlin, Campus Charité Mitte 1

Page 1 of 33

Charité Universitätsmedizin Berlin

24

Charitéplatz 1, 10117 Berlin, Germany

25

Tel.: +49 30 450 536116

26

Fax: +49 30 450 536943

27

E-mail: [email protected]

ip t

23

cr

28

30

us

29

Highlights

d

M

 This is the first report on parallel FISH and IHC screening for ALK, MET and ROS1  Concordant results for ALK IHC and ALK FISH were seen in all cases  MET FISH and IHC showed discordant results in 10.0% of cases  MET overexpression can occur in ALK rearranged tumors  Besides rearrangements, ROS1 amplifications can also occur in NSCLCs

te

32 33 34 35 36 37 38

an

31

Ac ce p

39

40

Abstract

41

Objectives: ALK, MET and ROS1 are prognostic and predictive markers in NSCLC,

42

which need to be implemented in daily routine. To evaluate different detection

43

approaches and scoring systems for optimal stratification of patients eligible for

44

mutation testing in the future, we screened a large and unselected cohort of NSCLCs

45

for all three alterations.

46

Material and Methods: Using tissue microarrays, 473 surgically resected NSCLCs

47

were tested for ALK and MET expression by IHC and genomic alterations in the ALK,

48

MET and ROS1 gene by FISH. For MET IHC, two different criteria (MetMab and H-

2

Page 2 of 33

score), for MET FISH, three different scoring systems (UCCC, Cappuzzo,

50

PathVysion) were investigated.

51

Results: ALK and ROS1 positivity was seen in 2.6% and 1.3% of all ADCs,

52

respectively, but not in pure SCCs. One ROS1 translocated tumor showed additional

53

ROS1 amplification. MET IHC+/FISH+ cases were found in both histological

54

subtypes (8.6% in all NSCLCs; 10.6% in ADCs; 5.0% in SCCs) and were associated

55

with pleural invasion, lymphatic vessel invasion and lymph node metastasis. MET

56

altered ADCs more frequently showed a papillary growth pattern. Whereas ALK

57

testing revealed homogenous results in IHC and FISH, we saw discordant results for

58

MET in about 10% of cases. Both MET-IHC scoring systems revealed almost

59

identical results. We did not encounter any combined FISH positivity for ALK, MET or

60

ROS1. However, three ALK positive cases harbored MET overexpression.

61

Conclusion: In daily routine, IHC could support FISH in the identification of ALK

62

altered NSCLCs. Further research is needed to assess the role of discordant MET

63

results by means of IHC and FISH as well as the relevance of tumors with an

64

increased ROS1 gene copy number.

cr

us

an

M

d

te

Ac ce p

65

ip t

49

3

Page 3 of 33

Introduction

66

In the last decade, the treatment modalities for non-small cell lung cancer (NSCLC)

67

have changed tremendously. In addition to surgery and standard chemotherapy,

68

personalized medicine such as tyrosine kinase inhibitors (TKIs) against the epidermal

69

growth factor receptor (EGFR) pose new therapeutic options with significantly

70

improved progression-free survival rates in a subset of patients [1]. Over the years,

71

additional targetable drivers have been identified in NSCLC such as the anaplastic

72

lymphoma kinase (ALK). It took only a few years from its discovery as a driver

73

mutation in NSCLC [2] to the approval of an ALK-TKI (crizotinib) in 2011. However,

74

crizotinib does not only target ALK but also MET and ROS1. This is supported by

75

recent reports that showed promising results in patients with MET amplification or

76

ROS1 translocation treated with crizotinib [3-6]. Additionally, MET was also shown to

77

be targetable by other inhibitors [7-9]. However, a clinical phase III trial was stopped

78

recently and the role of MET will need further evaluation [10]. Regardless, MET plays

79

an important role in the development of therapy resistance, especially in the context

80

of anti-EGFR-therapy [11]. Therefore, the identification of patients harboring

81

alterations in the ALK, ROS1 and still the MET gene seems of high interest.

82

In this study, we analyzed the frequency of ALK and ROS1 translocations as well as

83

MET amplifications in an unselected cohort of 473 surgically resected NSCLC

84

samples. We compared immunohistochemistry (IHC) and fluorescence in-situ

85

hybridization (FISH) as potential diagnostic assays. For MET FISH, we investigated

86

all three scoring systems so far described in literature [7,9,12-17]. Those were

87

compared among each other and correlated with two MET IHC scoring systems

88

(MetMAb and H-score). In the future, this could help to implement reliable and

89

efficient

Ac ce p

te

d

M

an

us

cr

ip t

65

screening

algorithms for

these

4

alterations

in routine

diagnostics.

Page 4 of 33

Materials and Methods

91

Patients and Samples

92

Tumor tissue of 473 NSCLC patients (289 men (61.1%) and 184 women (38.9%);

93

Table 1) who underwent pulmonary resection between 2000 and 2013 were obtained

94

from the archives of several Institutes of Pathology in Berlin (Charité University

95

Medicine Berlin, Pathology-Berlin Bioptisches Institut Gemeinschaftspraxis für

96

Pathologie, Vivantes Klinikum Berlin) and Minden (Johannes Wesling Klinikum

97

Minden). Detailed clinicopathological data was available for all patients, except for

98

two cases in which the staging was not documented. The median age was 67 years

99

(range: 29-85 years). Stage I was diagnosed in 261 (55.4%), stage II in 102 (21.7%),

100

stage III in 84 (17.8%) and stage IV in 24 (5.1%) cases. 335 (70.8%) tumors were

101

classified as adenocarcinomas (ADC). The predominant growth pattern was acinar in

102

144 (43.0%), solid in 114 (34.0%), papillary in 37 (11.0%), lepidic in 32 (9.6%) and

103

micropapillary in eight (2.4%) cases. 108 (22.8%) tumors were classified as

104

squamous cell carcinomas (SCC). The predominant growth pattern was non-

105

keratinizing in 61 (56.5%), keratinizing in 38 (35.2%) and basaloid in nine (8.3%)

106

cases. 13 (2.8%) samples were categorized as NSCLC not otherwise specified

107

(NOS), ten (2.1%) as large-cell carcinomas (LCC) and seven (1.5%) as

108

adenosquamous carcinomas (ADSC). Representative tumor areas were identified

109

and a total of ten tissue microarrays (TMA) were constructed using two cores for

110

each case (one mm diameter per core).

Ac ce p

te

d

M

an

us

cr

ip t

90

111 112

Immunohistochemistry (IHC)

113

IHC was performed as described previously on the VENTANA BenchMark XT 5

Page 5 of 33

automated slide stainer, using the anti-ALK-D5F3 Optiview kit [18] and the anti-total

115

c-MET-SP44 antibody (both Ventana Medical Systems, USA) [7]. ALK expression

116

was scored as negative (ALK IHC-) or positive (ALK IHC+); the latter was defined as

117

strong granular cytoplasmatic staining pattern [18]. MET expression was scored

118

according to the MetMAb trial criteria [7,19]: samples with no (IHC score 0) or weak

119

(IHC score 1+) membranous and/or cytoplasmatic staining in ≥50% of tumor cells

120

were considered MET IHC negative (MET IHC-). Tumors with moderate (IHC score

121

2+) or strong (IHC score 3+) membranous and/or cytoplasmatic staining in ≥50% of

122

tumor cells were considered MET IHC positive (MET IHC+). Furthermore, for MET

123

IHC we calculated a continuous H-score by multiplying the staining intensity (0: no

124

staining; 1: weak staining; 2: moderate staining; 3: strong staining) with the

125

respective percentage of tumor cells to reach a value between 0 and 300.

M

an

us

cr

ip t

114

d

126

Fluorescence in-situ hybridization (FISH)

128

FISH analysis was performed according to manufacturer’s instructions. Briefly, 4 µm

129

sections of TMA blocks were deparaffinized, dehydrated and then incubated in pre-

130

treatment solution (Dako, Denmark) for 10 minutes at 95-99°C. Samples were

131

immersed in pepsin solution for 3-6 minutes at 37°C. Following washing and

132

dehydration, slides were air dried and the respective DNA probe (ALK: Vysis LSI ALK

133

Dual Color, Abbott Molecular, USA; MET: MET/CEP7 dual-color probe, Abbott

134

Molecular, USA; ROS1: RF POSEIDON ROS1 Break probe, Kreatech, The

135

Netherlands; CEP6: CEP6 SpectrumAqua Probe, Abbott Molecular, USA) was

136

applied. The sections were sealed, denaturalized in humidified atmosphere at 82°C

137

for 5 minutes and then incubated overnight at 45°C to achieve hybridization. After

138

post-hybridization washing, slides were counterstained with 4’,6-diamidino-2-

Ac ce p

te

127

6

Page 6 of 33

139

phenylindole (DAPI).

140

Fluorescence in-situ hybridization (FISH) scoring

142

For each case, signals were enumerated in at least 50 non-overlapping tumor cells

143

using a fluorescence microscope (Axio Imager Z1, Zeiss, Germany). Computer-

144

based documentation and image analysis was performed with the ISIS imaging

145

system (MetaSystems, Germany).

146

ALK FISH positivity (ALK FISH+) was defined as the presence of split signals (SS)

147

and/or single red signals (SRS) in ≥15% of tumor cells [20-22].

148

For MET we used the three different scoring systems so far described in literature:

149

(1) UCCC (University of Colorado Cancer Center): cases with (A) MET/CEP7-ratio ≥2

150

or (B) small gene clusters (4-10 copies) or innumerable tight gene clusters in >10%

151

of tumor cells or (C) larger and brighter MET signals than CEP7 signals in >10% of

152

tumor cells or (D) >15 MET signals in >10% of tumor cells were considered as gene

153

amplification (GA). Specimens with ≥4 MET signals in ≥40% of tumor cells were

154

defined as high polysomy (HP). GA and HP were both considered to be MET FISH

155

positive (MET FISH+) [23,24]. However, as HP might be discussed as not being a

156

“true” amplification, we further discriminated between GA and HP.

157

(2) Cappuzzo: specimens with a mean gene copy number (GCN) of ≥5 MET signals

158

per tumor cell were classified as MET FISH+ [25].

159

(3) PathVysion: cases with a MET/CEP7-ratio ≥2 were defined as MET FISH+

160

[9,13,26].

161

For ROS1, tumors harboring SS and/or single green signals (SGS) in ≥15% of tumor

Ac ce p

te

d

M

an

us

cr

ip t

141

7

Page 7 of 33

162

cells were considered as ROS1 FISH positive (ROS1 FISH+) [27-30].

163

Statistical Analysis

165

Statistical analysis was performed using SPSS 21 software (IBM Corporation, USA).

166

Significance for cross tables larger than 2x2 was calculated using the statistical

167

language R. Comparison of clinicopathological data with IHC and FISH results was

168

performed using Fisher’s exact test (categorical data) and Mann-Whitney U test (age

169

and

size).

Ac ce p

te

d

M

an

tumor

us

cr

ip t

164

8

Page 8 of 33

170

Results

172

ALK IHC and ALK FISH

173

444 samples (93.9%) were evaluable by means of IHC and FISH. Concordant results

174

were seen in all 444 cases. A total of nine (2.0%) samples (2.6% of ADC) displayed

175

positivity for IHC and FISH (Figure 1). ALK rearrangements more frequently occurred

176

in younger patients with a median age of 54 years (range: 29-75 years; p=0.009). Six

177

(66.7%) of the ALK altered tumors showed lymphatic vessel invasion (p=0.023) and

178

lymph node metastasis (p=0.004). There was no significant trend (p=0.199) towards

179

advanced tumor stage (stage I: one case; stage II: three cases; stage III: two cases;

180

stage IV: one case). ALK IHC+/FISH+ tumors were mainly classified as ADC with

181

predominant acinar (five cases), solid (two cases) or papillary (one case) growth

182

pattern. One sample was classified as ADSC without keratinization in the SCC

183

component and with a solid growth pattern in the ADC component.

cr

us

an

M

d

te

Ac ce p

184

ip t

171

185

MET

186

MET IHC MetMAb criteria and MET FISH

187

440 samples (93.0%) were evaluable by means of IHC and FISH. Using the MetMAb

188

criteria for IHC and the UCCC scoring system including HP and GA for FISH,

189

homogenous results were seen in 396 cases (90.0%), with IHC-/FISH- in 358

190

(81.4%) and IHC+/FISH+ in 38 (8.6%) samples. 37 (8.4%) IHC+/FISH- and seven

191

(1.6%) IHC-/FISH+ cases were observed (Figure 2). Using the UCCC GA, Cappuzzo

9

Page 9 of 33

192

and PathVysion scoring system, the correlation was weaker with homogenous results

193

in in 383 (87,0%), 378 (85.9%) and 371 cases (84.3%), respectively (Table 3).

194

MET IHC H-score and MET FISH

196

The additionally applied MET IHC H-score showed a range between 0 and 280. A

197

good comparability of both tested IHC criteria was seen at an H-score threshold of

198

≥105, with discordant results in ten cases (2.2%; Supplemental Figure 1). Five of

199

these cases were classified as MetMAb positive and H-score negative, five as

200

MetMAb negative and H-score positive.

201

An H-score threshold of ≥180 encompassed 18 cases, all of them were considered

202

as MET FISH+ (Figure 4). Nine specimens were classified as HP and nine as GA.

203

These samples were also identified using the MetMAb criteria (IHC score 2+: 6

204

cases; IHC score 3+: 12 cases). However, this cut-off would not cover 29 (35.5%)

205

MET FISH+ samples.

cr

us

an

M

d

te

Ac ce p

206

ip t

195

207

Correlation of MET+ tumors (MetMab and UCCC) with clinicopathological data

208

MET IHC+/FISH+ samples were not associated with sex (25 men (65.8%), 13

209

women (34.2%); p=0.603) or age (median age: 67 years; range: 48-84 years;

210

p=0.694). Pleural invasion was present in in 47.2% of all IHC+/FISH+ cases

211

compared to 23.6% in tumors without MET IHC+/FISH+ results (p=0.006). MET

212

IHC+/FISH+ samples more frequently harbored lymphatic vessel invasion (60.5% vs.

213

41.0%; p=0.026) and lymph node metastasis (47.4% vs. 30.7%; p=0.048). There was

214

a non-significant trend towards advanced tumor stages (p=0.111) as alterations also

10

Page 10 of 33

occurred in early stages (stage I: 16 cases (6.8%); stage II: nine cases (9.9%); stage

216

III: eight cases (9.9%); stage IV: five cases (23.8%)). IHC+/FISH+ cases included 33

217

ADCs and five SCCs. MET alterations were more common in ADC (10.6%) than in

218

SCC (5.0%) and other NSCLC subtypes, although this difference was not significant

219

(p=0.051). We observed papillary (eleven cases), acinar (ten cases), solid (nine

220

cases) and lepidic (three cases) predominant growth patterns among the MET

221

altered ADCs with a significant trend towards papillary growth pattern (p=0.006). The

222

IHC+/FISH+ SCCs were classified as both keratinizing (one case) and non-

223

keratinizing (four cases; p=0.743).

224

Furthermore, we performed a separate analysis of the particular FISH scoring

225

systems with clinicopathological data (Supplementary Table 1). Pleural invasion and

226

advanced stage were significantly more common in the UCCC HP, UCCC GA and

227

Cappuzzo group. Blood vessel invasion was more frequent in the UCCC GA

228

(p=0.037) and Cappuzzo (p=0.005) group, while cases from the UCCC HP group

229

were significantly more likely to harbor lymphatic vessel invasion (p=0.007) and

230

lymph node metastasis (p=0.007). However, there were no significant correlations

231

with tumors that were considered positive according to the PathVysion criteria.

cr

us

an

M

d

te

Ac ce p

232

ip t

215

233

ROS1 FISH

234

ROS1 FISH was evaluable in 450 cases (94.5%). We discovered ROS1

235

rearrangements in four ADCs (0.9%), accounting for 1.3% of all ADCs. One ROS1

236

translocated tumor also showed an increased ROS1 GCN (5.4 copies per cell).

237

Another sample displayed 7.8 copies per cell but no ROS1 rearrangement (Figure 3).

11

Page 11 of 33

ROS1 translocations occurred in both men and women with a median age of 70

239

years (range 60-77 years). Three tumors showed lymphatic vessel invasion, lymph

240

node metastasis were present in two of them. Two cases were classified as stage I

241

and the other two samples as stage III. ROS1 rearranged tumors were all classified

242

as ADCs with predominantly solid (three cases) or papillary (one case) growth

243

pattern.

244

We reevaluated the two cases with an increased ROS1 GCN using an additional

245

CEP6 FISH probe. The ROS1 rearranged tumor showed amplification of the ROS1

246

gene locus with a ROS1/CEP6 ratio of 3.6, while the copy number gain in the second

247

sample was due to polysomy of chromosome 6 (Figure 3).

an

us

cr

ip t

238

M

248

Co-occurrence of ALK, MET and ROS1 alterations

250

Results for ALK, MET and ROS1 were available in 405 samples (85.6%) There was

251

no concomitant FISH positivity for any of the evaluated alterations. However, one

252

MET IHC+/FISH+ sample with gene amplification (MET/CEP7 ratio: 2.1; MetMAb

253

IHC score 2+; H-score 170) showed an increased ROS1 GCN due to polysomy of

254

chromosome 6 (Figure

255

overexpression (Figure 1). MET overexpression was more common in ALK altered

256

tumors (33% vs. 17.2%), although this was not significant (p=0.197).

257

One ALK IHC+ and MET IHC- case did not yield any evaluable FISH signals for ALK,

258

MET and ROS1, even though FISH was repeated several times, including

259

reevaluation of whole-slide tissue sections and different tumor blocks.

Ac ce p

te

d

249

3). Three ALK

IHC+/FISH+

cases showed

MET

260

12

Page 12 of 33

Discussion

261

Approximately 90% of all NSCLC diagnoses are performed on biopsy specimens

262

[31]. As the amount of tumor tissue available for molecular analysis is often limited

263

and the number of predictive targets increases steadily, the establishment of reliable

264

tests and adequate algorithms is of utmost importance. In this study we performed a

265

parallel screening for ALK, MET and ROS1 alterations and discuss the

266

consequences of our results for daily routine practice. ALK and ROS1

267

rearrangements were found in 2.6% and 1.3% of ADCs, respectively, but not in pure

268

SCCs. However, one ALK positive case showed adenosquamous histology. MET

269

IHC+/FISH+ results, on the other hand, were found in 8.6% of all NSCLCs and were

270

associated with pleural invasion. MET altered ADCs more frequently showed a

271

papillary growth pattern. Whereas ALK testing revealed reliable and concordant

272

results in IHC and FISH, the role of these two methods to identify MET amplifications

273

requires further investigation, as our data revealed discordant results in about 10.0%.

cr

us

an

M

d

te

Ac ce p

274

ip t

260

275

ALK alterations were described to be more frequent in younger patients [32-34]

276

which is consistent with the relatively low median age of 54 years in our cohort.

277

However, we encountered three ALK positive patients older than 60 years. Thus,

278

elderly patients should not generally be excluded from ALK testing [35]. For ALK, we

279

saw a clear correlation between IHC and FISH. All nine FISH+ cases were identified

280

by means of IHC and no false positive IHC results were encountered. Moreover, ALK

281

protein expression was helpful in two so-called “ALK FISH borderline” samples with

282

split signals in only 15-20% of tumor cells. These cases are particularly challenging in

283

routine diagnostics as they might be diagnosed as false negative in FISH [32,36,37].

284

Of note, we also encountered one ALK IHC+ case that was not evaluable by FISH. 13

Page 13 of 33

This case illustrates the urgent need for alternative methods to identify ALK

286

rearrangements besides FISH, as a therapy decision must be drawn under these

287

circumstances as well. In summary, our data support results of previous studies

288

[18,34,38] underlining the potential role of IHC as an alternative assay or at least as a

289

pre-screening tool to identify ALK rearrangements.

ip t

285

cr

290

Regarding MET, IHC+/FISH+ results were seen in 38 tumors (8.6%) when using the

292

MetMAb criteria for IHC and the UCCC scoring system for FISH. At a cut-off value of

293

≥105, the results from the IHC MetMAb criteria and the H-score were very similar

294

with homogenous results in 97.8% of cases. This is not surprising, as the definition of

295

a case with MetMAb IHC score 2+ (moderate staining in ≥50% of tumor cells)

296

corresponds to an H-score of at least 100. Furthermore, all tumors with an H-score

297

≥180 (n= 18) also harbored alterations of the MET gene in FISH. Although

298

statements on the predictive value of this finding is beyond the focus of this paper, an

299

IHC assay using an H-score with this threshold value could be examined as a

300

possible predictive test in future clinical trials.

an

M

d

te

Ac ce p

301

us

291

302

In accordance with literature, MET IHC+/FISH+ predominantly occurred in ADCs but

303

were also found in SCCs [17,39]. As already described by Nakamura et al. [40] we

304

observed that MET alterations were significantly more common in ADCs with

305

papillary growth pattern. Other authors described a significant correlation between

306

MET amplification and non-lepidic growth patterns [14,41]. Interestingly, activating

307

MET mutations also frequently occur in renal and thyroid carcinomas with papillary

308

growth [40]. Therefore, further research is needed to assess if MET might play an

14

Page 14 of 33

important role in the formation of papillary structures as proposed by Nakamura et al.

310

[40]. In accordance with previous studies we saw that MET alterations occurred more

311

frequently in tumors with pleural invasion, lymphatic vessel invasion and lymph node

312

metastasis [14,17]. Even though MET IHC+/FISH+ seem to be more common in

313

advanced tumors, they have also been described to occur in early stages [41].

314

Consistent with these reports, we observed MET alterations in 6.8% of all stage I

315

patients. Thus, from our data, it appears that MET testing should neither be limited to

316

a certain clinical subgroup nor to a certain histology.

317

Besides possible links with clinicopathological characteristics, the identification of the

318

most suitable method to screen for patients harboring MET amplifications is crucial

319

for routine diagnostics. The best correlation between IHC and FISH was achieved by

320

using the MetMAb criteria for IHC and the UCCC scoring system including HP for

321

FISH with concordant results in 90.0% compared to 85.9% and 84.3% when using

322

the Cappuzzo or PathVysion criteria, respectively (Table 3). The UCCC scoring

323

system was able to identify all tumors that were classified as positive according to

324

one of the other criteria. Therefore, the UCCC scoring system seems to be superior

325

to the Cappuzzo and PathVysion criteria in identifying IHC+ cases. Still, 44 samples

326

revealed contradictory results with 37 IHC+/FISH- and seven IHC-/FISH+ tumors.

327

These heterogeneous results might explain the huge differences between the

328

frequencies of MET overexpression (25-54%) and MET amplification (11.1-16.7%

329

when using the UCCC criteria) in earlier studies [7,8,12,24,39]. IHC-/FISH+ cases

330

have been reported before although they have never been described in detail [8,12].

331

It is unknown whether these tumors with a significantly increased MET GCN actually

332

do not overexpress MET or if the weak immunoreactivity in IHC was due to a specific

333

lack of antibody affinity or other mechanisms that might reduce staining intensity.

334

However, it should be noted that all MET IHC-/FISH+ specimens had been evaluable

Ac ce p

te

d

M

an

us

cr

ip t

309

15

Page 15 of 33

for other IHC markers such as TTF-1 or p63, stained in the process of the routine

336

diagnostics (data not shown). While the clinical outcome of patients with MET IHC-

337

/FISH+ tumors has not yet been investigated in detail, two recent studies showed

338

that MET IHC+/FISH- patients could also benefit from therapy with onartuzumab or

339

tivantinib [7,8]. Thus, there is a need to investigate (A) the underlying mechanisms

340

that lead to MET overexpression in FISH negative tumors; (B) potential reasons why

341

some FISH positive tumors show no or only weak protein expression in IHC; (C) the

342

clinical relevance of cases with conflicting results in IHC and FISH, especially of IHC-

343

/FISH+ patients.

an

us

cr

ip t

335

344

Recently, ROS1 has been identified as a treatable target in lung cancer [29,42,43]. In

346

our investigation, we saw an overall frequency of 0.9% (1.3% in ADCs) which is in

347

line with previous reports [29,42,43]. Recent trials and case reports discovered that

348

patients with ROS1 translocation in FISH could profit from anti-ROS1 therapy [4,5].

349

Regarding histology, we confirmed that ROS1 translocations mainly occur in ADCs

350

[42]. However, ROS1 might also be affected in SCCs and LCCs as previously

351

described [27,42,43]. Due to the low frequency of ROS1 alterations in our study, any

352

links with clinicopathological characteristics should be interpreted with care.

353

Furthermore, we encountered one case with polysomy of chromosome 6 and one

354

tumor with ROS1 amplification, the latter in addition to ROS1 translocation (Figure 3).

355

To our knowledge, “true” amplifications of the ROS1 gene locus have not been

356

described yet in NSCLC. Interestingly, tumors with an increased ROS1 GCN were

357

reported to show strong ROS1 expression in IHC [44]. These patterns need further

358

investigation as they might also benefit from targeted therapy.

Ac ce p

te

d

M

345

16

Page 16 of 33

359

Regarding concomitant alterations, we did not encounter any combined FISH+

361

results for ALK, MET or ROS1. For IHC, we observed three ALK rearranged tumors

362

showing MET overexpression. However, in contrast to recently published data

363

[17,45], MET IHC positivity was not significantly more common in ALK rearranged

364

tumors. This might be due to the lower number of ALK positive samples in our cohort.

365

Regardless, the clinical role of such cases is unknown and requires further

366

investigation (therapy resistance?).

367

While other publications showed that ROS1 alterations may overlap with KRAS,

368

EGFR and BRAF mutations [42,46], we observed no co-occurrence of ROS1

369

rearrangements with ALK or MET alterations although these results might not be

370

representative due to the low number of ROS1 translocated tumors in our study. One

371

case with an increased ROS1 GCN due to polysomy of chromosome 6 showed a

372

concomitant MET amplification (Figure 3). The clinical and biological role is unclear

373

and needs further evaluation.

cr

us

an

M

d

te

Ac ce p

374

ip t

360

375

In summary, our study is the first to report on parallel testing for ALK, MET and

376

ROS1 alterations in a large and non-preselected NSCLC-cohort. We may conclude

377

from our data that ALK and ROS1 screening should first of all be performed in ADCs

378

of the lung, whereas MET screening might be established as a further predictive

379

diagnostic option not only in ADCs but also in SCCs. In daily routine, IHC could

380

complement FISH in the diagnosis of ALK altered NSCLCs, especially with regards

381

to borderline cases or tumors without evaluable FISH signals. For MET, future

382

research is needed to evaluate the predictive value of IHC and FISH as well as the

17

Page 17 of 33

383

clinical role of patients with discordant results. Additionally, the clinical relevance of

384

tumors with increased ROS1 copy number due to polysomy of chromosome 6 or

385

amplification

the

ROS1

gene

locus

requires

further

evaluation.

Ac ce p

te

d

M

an

us

cr

ip t

of

18

Page 18 of 33

386

Acknowledgement

388

Part of this work was supported by PFIZER within the framework of the FALKE-

389

project (detection of ALK-rearrangements by FISH in NSCLC).

392

None

us

Conflict of interest

Ac ce p

te

d

M

an

391

cr

390

ip t

387

19

Page 19 of 33

393 394

References

395

[1]

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.

397

N Engl J Med 2010;362:2380–8. [2]

Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, et al.

cr

398

ip t

396

Identification of the transforming EML4–ALK fusion gene in non-small-cell

400

lung cancer. Nature 2007;448:561–6.

401

[3]

us

399

Schwab R, Petak I, Kollar M, Pinter F, Varkondi E, Kohanka A, et al. Major partial response to crizotinib, a dual MET/ALK inhibitor, in a squamous cell

403

lung (SCC) carcinoma patient with de novo c-MET amplification in the

404

absence of ALK rearrangement. Lung Cancer 2013;83:109–11. [4]

Ou S-HI, Camidge R, Riely GJ, Salgia R, Shapiro G, Clark JW, et al. Efficacy

M

405

an

402

406

and safety of crizotinib in patients with advanced ROS1-rearranged non-small

407

cell lung cancer (NSCLC). Ann Oncol 2013;24:ix43–3. [5]

Bos M, Gardizi M, Schildhaus HU, Heukamp LC, Geist T, Kaminsky B, et al.

d

408

Complete metabolic response in a patient with repeatedly relapsed non-small

410

cell lung cancer harboring ROS1 gene rearrangement after treatment with

411

crizotinib. Lung Cancer 2013;81:142–3. [6]

and anaplastic lymphoma kinase (ALK) inhibitor, in a non-small cell lung

414

cancer patient with de novo MET amplification. J Thorac Oncol 2011;6:942–6.

415

[7]

Spigel DR, Ervin TJ, Ramlau RA, Daniel DB, Goldschmidt JH, Blumenschein GR, et al. Randomized Phase II Trial of Onartuzumab in Combination With

417

Erlotinib in Patients With Advanced Non-Small-Cell Lung Cancer. J Clin

418

Oncol 2013;31:4105–14.

419 420

Ou S-HI, Kwak EL, Siwak-Tapp C, Dy J, Bergethon K, Clark JW, et al. Activity of crizotinib (PF02341066), a dual mesenchymal-epithelial transition (MET)

413

416

Ac ce p

412

te

409

[8]

Rodig SJ, Sequist LV, Schiller JH, Chen Y, Halim A, Waghorne C, et al.

421

Abstract 1729: An exploratory biomarker analysis evaluating the effect of the

422

c-MET inhibitor tivantinib (ARQ 197) and erlotinib in NSCLC patients in a

423

randomized, double-blinded phase 2 study. Cancer Res 2012;72:1729.

424

[9]

Sequist LV, Sequist LV, Pawel von J, Pawel von J, Garmey EG, Garmey EG, 20

Page 20 of 33

425

et al. Randomized Phase II Study of Erlotinib Plus Tivantinib Versus Erlotinib

426

Plus Placebo in Previously Treated Non-Small-Cell Lung Cancer. J Clin

427

Oncol 2011;29:3307–15. [10]

come and gone? Clin Cancer Res 2014;20:4422–4.

429 430

Hirsch FR, Bunn PA, Herbst RS. “Companion diagnostics”: has their time

[11]

Bean J, Brennan C, Shih J-Y, Riely G, Viale A, Wang L, et al. MET

ip t

428

amplification occurs with or without T790M mutations in EGFR mutant lung

432

tumors with acquired resistance to gefitinib or erlotinib. Proc Natl Acad Sci U

433

S A 2007;104:20932–7. [12]

Dziadziuszko R, Wynes MW, Singh S, Asuncion BR, Ranger-Moore J,

us

434

cr

431

Konopa K, et al. Correlation between MET gene copy number by silver in situ

436

hybridization and protein expression by immunohistochemistry in non-small

437

cell lung cancer. J Thorac Oncol 2012;7:340–7.

438

[13]

an

435

Yu HA, Arcila ME, Rekhtman N, Sima CS, Zakowski MF, Pao W, et al. Analysis of tumor specimens at the time of acquired resistance to EGFR-TKI

440

therapy in 155 patients with EGFR-mutant lung cancers. Clin Cancer Res

441

2013;19:2240–7. [14]

Tachibana K, Minami Y, Shiba-Ishii A, Kano J, Nakazato Y, Sato Y, et al.

d

442

M

439

Abnormality of the hepatocyte growth factor/MET pathway in pulmonary

444

adenocarcinogenesis. Lung Cancer 2012;75:181–8.

446 447 448 449 450 451 452

[15]

MET Increased Gene Copy Number and Primary Resistance to Gefitinib Therapy in Non-Small Cell Lung Cancer Patients. Ann Oncol 2008;20:298– 304.

[16]

Minuti G, D'Incecco A, Cappuzzo F. Targeted therapy for NSCLC with driver mutations. Expert Opin Biol Ther 2013;13:1401–12.

[17]

Tsuta K, Kozu Y, Mimae T, Yoshida A, Kohno T, Sekine I, et al. cMET/phospho-MET protein expression and MET gene copy number in nonsmall cell lung carcinomas. J Thorac Oncol 2012;7:331–9.

453 454

Cappuzzo F, Janne PA, Skokan M, Finocchiaro G, Rossi E, Ligorio C, et al.

Ac ce p

445

te

443

[18]

Nitta H, Tsuta K, Yoshida A, Ho SN, Kelly BD, Murata LB, et al. New methods

455

for ALK status diagnosis in non-small-cell lung cancer: an improved ALK

456

immunohistochemical assay and a new, Brightfield, dual ALK IHC-in situ

457

hybridization assay. J Thorac Oncol 2013;8:1019–31.

458

[19]

Spigel DR, Ervin TJ, Ramlau R, Daniel DB. Final efficacy results from 21

Page 21 of 33

459

OAM4558g, a randomized phase II study evaluating MetMAb or placebo in

460

combination with erlotinib in advanced NSCLC. J Clin Oncol 2011.

461

[20]

McLeer-Florin AA, Moro-Sibilot DD, Melis AA, Salameire DD, Lefebvre CC, Ceccaldi FF, et al. Dual IHC and FISH testing for ALK gene rearrangement in

463

lung adenocarcinomas in a routine practice: a French study. J Thorac Oncol

464

2012;7:348–54.

465

[21]

ip t

462

Thunnissen E, Bubendorf L, Dietel M, Elmberger G, Kerr K, López-Ríos F, et al. EML4-ALK testing in non-small cell carcinomas of the lung: a review with

467

recommendations. Virchows Arch 2012;461:245–57. [22]

Yi ES, Boland JM, Maleszewski JJ, Roden AC, Oliveira AM, Aubry M-C, et al.

us

468

cr

466

Correlation of IHC and FISH for ALK Gene Rearrangement in Non-small Cell

470

Lung Carcinoma IHC Score Algorithm for FISH. J Thorac Oncol 2011;6:459–

471

65.

472

[23]

an

469

Varella-Garcia M. Stratification of non-small cell lung cancer patients for therapy with epidermal growth factor receptor inhibitors: the EGFR

474

fluorescence in situ hybridization assay. Diagn Pathol 2006;1:19.

475

[24]

M

473

Go H, Jeon YK, Park HJ, Sung S-W, Seo J-W, Chung DH. High MET gene copy number leads to shorter survival in patients with non-small cell lung

477

cancer. J Thorac Oncol 2010;5:305–13.

480 481 482 483 484 485 486

te

479

[25]

Cappuzzo F, Marchetti A, Skokan M, Rossi E, Gajapathy S, Felicioni L, et al. Increased MET Gene Copy Number Negatively Affects Survival of Surgically

Ac ce p

478

d

476

Resected Non-Small Cell Lung Cancer Patients. J Clin Oncol 2009;27:1667– 74.

[26]

Tanaka A, Tanaka A, Sueoka-Aragane N, Sueoka-Aragane N, Nakamura T, Nakamura T, et al. Co-existence of positive MET FISH status with EGFR mutations signifies poor prognosis in lung adenocarcinoma patients. Lung Cancer 2012;75:89–94.

[27]

Davies KD, Davies KD, Le AT, Le AT, Theodoro MF, Theodoro MF, et al.

487

Identifying and targeting ROS1 gene fusions in non-small cell lung cancer.

488

Clin Cancer Res 2012;18:4570–9.

489

[28]

Kim MH, Shim HS, Kang DR, Jung JY, Lee CY, Kim DJ, et al. Clinical and

490

prognostic implications of ALK and ROS1 rearrangements in never-smokers

491

with surgically resected lung adenocarcinoma. Lung Cancer 2014;83:1–7.

492

[29]

Bergethon K, Bergethon K, Shaw AT, Shaw AT, Ignatius Ou SH, Ignatius Ou 22

Page 22 of 33

493

SH, et al. ROS1 Rearrangements Define a Unique Molecular Class of Lung

494

Cancers. J Clin Oncol 2012;30:863–70.

495

[30]

Yoshida A, Tsuta K, Wakai S, Arai Y, Asamura H, Shibata T, et al.

496

Immunohistochemical detection of ROS1 is useful for identifying ROS1

497

rearrangements inlung cancers. Mod Pathol 2013:1–10. Histopathology 2009;54:12–27.

499 500

Kerr KM. Pulmonary adenocarcinomas: classification and reporting.

ip t

[31] [32]

Camidge DR, Theodoro M, Maxson DA, Skokan M, O'Brien T, Lu X, et al.

cr

498

Correlations between the percentage of tumor cells showing an ALK gene

502

rearrangement, ALK signal copy number, and response to crizotinib therapy

503

in ALK FISH positive non-small cell lung cancer. Cancer Res 2012;118:4486–

504

94. [33]

an

505

us

501

Camidge DR, Hirsch FRF, Varella-Garcia MM, Franklin WAW. Finding ALKpositive lung cancer: what are we really looking for? J Thorac Oncol

507

2011;6:411–3.

508

[34]

M

506

Sholl LM, Weremowicz S, Gray SW, Wong K-K, Chirieac LR, Lindeman NI, et al. Combined use of ALK immunohistochemistry and FISH for optimal

510

detection of ALK-rearranged lung adenocarcinomas. J Thorac Oncol

511

2013;8:322–8.

514 515 516 517 518 519 520 521

te

513

[35]

Lindeman NI, Cagle PT, Beasley MB, Chitale DA, Dacic S, Giaccone G, et al. Molecular testing guideline for selection of lung cancer patients for EGFR and

Ac ce p

512

d

509

ALK tyrosine kinase inhibitors: guideline from the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. J Thorac Oncol 2013;8:823–59.

[36]

V Laffert M, Warth A, Penzel R, Schirmacher P, Jonigk D, Kreipe H, et al. Anaplastic lymphoma kinase (ALK) gene rearrangement in non-small cell lung cancer (NSCLC): results of a multi-centre ALK-testing. Lung Cancer 2013;81:200–6.

[37]

V Laffert M, Warth A, Penzel R, Schirmacher P, Kerr K, Elmberger G, et al.

522

Anaplastic lymphoma kinase (ALK)-detection in non-small cell lung cancer:

523

results of the first European IHC-based (D5F3-Optiview) panel test within 16

524

institutes. J Thorac Oncol 2013;8:S484.

525 526

[38]

Savic S, Bode B, Diebold J, Tosoni I, Barascud A, Baschiera B, et al. Detection of ALK-Positive Non-Small-Cell Lung Cancers on Cytological 23

Page 23 of 33

527

Specimens: High Accuracy of Immunocytochemistry with the 5A4 Clone. J

528

Thorac Oncol 2013;8:1004–11.

529

[39]

Park S, Choi Y-L, Sung CO, An J, Seo J, Ahn M-J, et al. High MET copy

530

number and MET overexpression: poor outcome in non-small cell lung cancer

531

patients. Histol Histopathol 2012;27:197–207. [40]

Nakamura Y, Niki T, Goto A, Morikawa T, Miyazawa K, Nakajima J, et al. c-

ip t

532

Met activation in lung adenocarcinoma tissues: an immunohistochemical

534

analysis. Cancer Sci 2007;98:1006–13.

535

[41]

cr

533

Jin Y, Sun P-L, Kim H, Seo AN, Jheon S, Lee C-T, et al. MET Gene Copy Number Gain is an Independent Poor Prognostic Marker in Korean Stage I

537

Lung Adenocarcinomas. Ann Surg Oncol 2013. [42]

Warth A, Muley T, Dienemann H, Goeppert B, Stenzinger A, Schnabel PA, et

an

538

us

536

539

al. ROS1 Expression and Translocations in Non-Small Cell Lung Cancer:

540

Clinicopathological Analysis of 1478 Cases. Histopathology 2014. [43]

Rimkunas VM, Crosby KE, Li D, Hu Y, Kelly ME, Gu TL, et al. Analysis of

M

541

Receptor Tyrosine Kinase ROS1-Positive Tumors in Non-Small Cell Lung

543

Cancer: Identification of a FIG-ROS1 Fusion. Clin Cancer Res 2012;18:4449–

544

57. [44]

Lee HJ, Seol HS, Kim JY, Chun S-M, Suh Y-A, Park Y-S, et al. ROS1

te

545

d

542

Receptor Tyrosine Kinase, a Druggable Target, is Frequently Overexpressed

547

in Non-Small Cell Lung Carcinomas Via Genetic and Epigenetic Mechanisms.

548 549 550 551 552 553 554

Ac ce p

546

Ann Surg Oncol 2012;20:200–8.

[45]

Feng Y, Minca EC, Lanigan C, Liu A, Zhang W, Yin L, et al. High MET receptor expression but not gene amplification in ALK 2p23 rearrangement positive non-small-cell lung cancer. J Thorac Oncol 2014;9:646–53.

[46]

Stumpfova M, Janne PA. Zeroing in on ROS1 Rearrangements in Non-Small Cell Lung Cancer. Clin Cancer Res 2012;18:4222–4.

555

24

Page 24 of 33

Titles and legends to figures

562

Figure 1 Example of an adenocarcinoma (a; hematoxylin and eosin stain) with

563

positive results in ALK IHC (b) and ALK FISH (c; red: 3’ ALK probe, green: 5’ ALK

564

probe). Additionally, this tumor showed MET overexpression (d; MetMAb IHC score

565

2+; H-score 150) but no amplification in MET FISH (e; red: MET gene locus, cyan:

566

centromere 7).

cr

ip t

561

us

567

Figure 2 Examples of tumors with equivocal and discordant results in MET IHC and

569

FISH: cases with homogenous results with IHC-/FISH- in case 1 (a-c) and

570

IHC+/FISH+ in case 2 (d-f). Cases 3 (g-I) showed MET overexpression but revealed

571

no amplification in FISH while case 4 (j-l) showed a significant MET copy number

572

gain but lacked any MET expression. Pictures a, d and h show hematoxylin and

573

eosin stains of representative tumor areas for each case.

M

d

te Ac ce p

574

an

568

575

Figure 3 Case 1 (a-c; green: 3’ ROS1 probe, red: 5’ ROS1 probe) showed ROS1

576

translocation. Picture c shows magnification of representative tumor cells from the

577

same case. Case 2 (d-g; cyan: centromere 6) revealed amplification and

578

translocation of the ROS1 gene locus with a gene-to-chromosome ratio of 3.6 and at

579

least one split signal or single green signal (e and f, white arrows indicate rearranged

580

signals) in 30% of tumor cells. Case 3 (h-k) showed ROS1 copy number gain due to

581

polysomy of chromosome 6 and had an additional MET amplification (j; red: MET

582

gene locus, cyan: centromere 7; MET/CEP7 ratio: 2.1) with MET overexpression in

583

IHC (data not shown). Pictures a, d and h show hematoxylin and eosin stains of

584

representative tumor areas for each case. 29

Page 25 of 33

585

Figure 4 Plot of MET IHC H-score and MET FISH results. The black line indicates a

587

threshold value of ≥180. All cases with an H-score ≥180 also revealed MET alteration

588

in FISH. For reasons of clarity, cases with an H-score of 0 are only shown if they

589

were classified as FISH+. The total number of cases with an H-score ≥180 was 18

590

and they were all classified as 2+ (6 cases) or 3+ (12 cases) according to the

591

MetMAb criteria.

Ac ce p

te

d

M

an

us

cr

ip t

586

30

Page 26 of 33

Ac

ce

pt

ed

M

an

us

cr

i

Figure 1

Page 27 of 33

Ac

ce

pt

ed

M

an

us

cr

i

Figure 2

Page 28 of 33

Ac

ce

pt

ed

M

an

us

cr

i

Figure 3

Page 29 of 33

300 UCCC high polysomy UCCC gene amplification UCCC negative

250

≥180

us

cr

ip

t

200

100

ce pt

ed

M

an

150

Ac

H-Score

Figure 4

50 0

Page 30 of 33

Table 1

Ac ce pt e

d

M

an

us

cr

ip t

Table 1 Correlation between MET amplification, ALK and ROS1 translocation and clinicopathological characteristics. a MET FISH+/IHC+ ALK FISH+/IHC+ ROS1 FISH+ All cases Positive p-value Positive p-value Positive p-value Total 473 38 (8.6%) 9 (1.9%) 4 (0.9%) Sex Male 289 25 5 2 0.603 0.739 0.643 Female 184 13 4 2 Age (years) Median (standard deviation) 67 ± 10 66 ± 8.7 54 ± 14.2 70 ± 7.6 0.694 0.009 0.386 Range 29 – 85 48 – 84 29 – 75 60 – 77 Stage I-II 363 25 4 2 0.111 0.199 0.220 III-IV 108 13 3 2 Lymph-node metastasis Absent 324 20 1 2 0.048 0.004 0.588 Present 147 18 6 2 Lymphatic vessel invasion Absent 276 15 1 1 0.026 0.023 0.308 Present 195 23 6 3 Blood vessel invasion Absent 369 26 4 2 0.158 0.195 0.202 Present 102 12 3 2 Pleural invasion Absent 352 19 5 3 0.006 1.000 1.000 Present 119 17 2 1 Tumor size (centimeter) Median (standard deviation) 3.0 ± 2.6 3.5 ± 2.0 0.960 2.3 ± 3.4 0.505 2,4 ± 0.8 0.321 Range 0.8 – 17.0 1.0 – 9.0 1.2 – 9.0 1.9 – 3.7 Histology ADC 335 33 8 4 SCC 108 5 0.051 0 0.182 0 0.673 b Others 30 0 1 0 ADC predominant growth pattern ADC solid 114 9 2 3 ADC acinar 144 10 5 0 ADC papillary 37 11 0.006 1 0.816 1 0.238 ADC lepidic 32 3 0 0 ADC micropapillary 8 0 0 0 SCC growth pattern Keratinizing 38 1 0 0 Non-Keratinizing 61 4 0.743 0 0 Basaloid 9 0 0 0 ALK, anaplastic lymphoma kinase; MET, mesenchymal-epithelial transition factor; ROS1, repressor of silencing 1; IHC, immunohistochemistry; FISH, fluorescence in-situ hybridization; ADC, adenocarcinoma; SCC, squamous cell carcinoma a Using the University of Colorado Cancer center (UCCC) scoring system for FISH and the MetMAb trial criteria for IHC b 13 non-small cell lung cancers not otherwise specified, ten large cell carcinomas, seven adenosquamous carcinomas

Page 31 of 33

Table 2

Ac ce pt e

d

M

an

us

cr

ip t

Table 2 Molecular pathological characteristics of patients harboring ALK or ROS1 translocation. Case # Sex Age Histology, FISH pattern ALK IHC MET MET IHC predominant (% of altered FISH MetMAb cells) criteria ALK 1 M 62 ADC, solid SS, SRS (69%) Positive Negative Positive (2+) ALK 2 M 69 ADC, acinar SRS (70%) Positive Negative Negative (0) ALK 3 F 39 ADSC, solid SS, SRS (18%) Positive Negative Negative (0) ALK 4 F 50 ADC, solid SS (20%) Positive Negative Negative (0) ALK 5 M 75 ADC, acinar SS (40%) Positive Negative Negative (1+) ALK 6 M 54 ADC, acinar SS (40%) Positive Negative Negative (0) ALK 7 M 41 ADC, papillary SS, SRS (62%) Positive Negative Positive (2+) ALK 8 F 58 ADC, acinar SS (70%) Positive Negative Negative (0) ALK 9 F 29 ADC, acinar SS, SRS (70%) Positive Negative Positive (2+) ROS1 M 60 ADC, acinar SS (71%) Negative Negative Negative (1+) 1 ROS1 F 77 ADC, solid SS (40%) Negative Negative Negative (1+) 2 ROS1 M 67 ADC, solid SS, SGS Negative Negative Negative (0) 3 (30%), A ROS1 F 74 ADC, solid SS, SGS (90%) Negative Negative Negative (1+) 4 ALK, anaplastic lymphoma kinase, ROS1, repressor of silencing 1; MET, mesenchymal-epithelial transition factor; FISH, fluorescence in-situ hybridization; IHC, immunohistochemistry; M, male; F, female; ADC, adenocarcinoma; ADSC, adenosquamous carcinoma; SS, split signals; SRS, single red signals; SGS, single green signals; A, amplification

Page 32 of 33

Table 3

Ac ce pt e

d

M

an

us

cr

ip t

Table 3 Correlation of different MET FISH amplification scoring systems and MET IHC scores. MET FISH MET IHC negative MET IHC positive All MET IHC 0 MET IHC 1+ MET IHC 2+ MET IHC 3+ a UCCC negative 395 250 108 37 0 a UCCC positive 45 5 2 26 12 UCCC HP negative 417 251 109 50 7 UCCC HP positive 23 4 1 13 5 UCCC GA negative 418 254 109 50 5 UCCC GA positive 22 1 13 7 Cappuzzo negative 423 254 109 54 6 Cappuzzo positive 17 1 1 9 6 PathVysion negative 436 255 110 59 10 PathVysion positive 6 0 0 4 2 MET, mesenchymal-epithelial transition; FISH, Fluorescence in-situ hybridization; IHC, immunohistochemistry; UCCC, University of Colorado Cancer Center; HP, high polysomy; GA, gene amplification a : Including high polysomy and gene amplification

Page 33 of 33

Parallel screening for ALK, MET and ROS1 alterations in non-small cell lung cancer with implications for daily routine testing.

ALK, MET and ROS1 are prognostic and predictive markers in NSCLC, which need to be implemented in daily routine. To evaluate different detection appro...
1MB Sizes 0 Downloads 7 Views