Skip to main content
Translational Lung Cancer Research logoLink to Translational Lung Cancer Research
. 2018 Jun;7(3):416–427. doi: 10.21037/tlcr.2018.05.01

Genome-wide copy number analyses of samples from LACE-Bio project identify novel prognostic and predictive markers in early stage non-small cell lung cancer

Federico Rotolo 1,2,3, Chang-Qi Zhu 4, Elisabeth Brambilla 5, Stephen L Graziano 6, Ken Olaussen 7, Thierry Le-Chevalier 8, Jean-Pierre Pignon 1,2,3, Robert Kratzke 9, Jean-Charles Soria 7,8, Frances A Shepherd 4,10, Lesley Seymour 11, Stefan Michiels 1,2,3, Ming-Sound Tsao 4,12,, on behalf of the LACE-Bio Consortium
PMCID: PMC6037976  PMID: 30050779

Abstract

Background

Adjuvant chemotherapy (ACT) provides modest benefit in resected non-small cell lung cancer (NSCLC) patients. Genome-wide studies have identified gene copy number aberrations (CNA), but their prognostic implication is unknown.

Methods

DNA from 1,013 FFPE tumor samples from three pivotal multicenter randomized trials (ACT vs. control) in the LACE-Bio consortium (median follow-up: 5.2 years) was successfully extracted, profiled using a molecular inversion probe SNP assay, normalized relative to a pool of normal tissues and segmented. Minimally recurrent regions were identified. P values were adjusted to control the false discovery rate (Q values).

Results

A total of 976 samples successfully profiled, 414 (42%) adenocarcinoma (ADC), 430 (44%) squamous cell carcinoma (SCC) and 132 (14%) other NSCLC; 710 (73%) males. We identified 431 recurrent regions, with on average 51 gains and 43 losses; 253 regions (59%) were ≤3 Mb. Most frequent gains (up to 48%) were on chr1, 3q, 5p, 6p, 8q, 22q; most frequent losses (up to 40%) on chr3p, 8p, 9p. CNA frequency of 195 regions was significantly different (Q≤0.05) between ADC and SCC. Fourteen regions (7p11–12, 9p21, 18q12, and 19p11–13) were associated with disease-free survival (DFS) (univariate P≤0.005, Q<0.142), with poorer DFS for losses of regions including CDKN2A/B [hazard ratio (HR) for 2-fold lower CN: 1.5 (95% CI: 1.2–1.9), P<0.001, Q=0.020] and STK11 [HR =2.4 (1.3–4.3), P=0.005, Q=0.15]. Chromosomal instability was associated with poorer DFS (HR =1.5, P=0.015), OS (HR =1.2, P=0.189) and lung-cancer specific survival (HR =1.7, P=0.003).

Conclusions

These large-scale genome-wide analyses of gene CNA provide new candidate prognostic markers for stage I–III NSCLC.

Keywords: Copy number aberrations (CNA), non-small cell lung cancer (NSCLC), platinum-based chemotherapy, biomarkers, phase III

Introduction

Lung cancer is the leading cause of cancer death worldwide. Non-small cell lung cancer (NSCLC), accounting for 85% of all lung cancers, has a 5-year survival of 59% for early resectable disease, but only 15% for cancers in advances stages (1). However, great differences within individual stages suggest the existence of unknown tumor factors. In the era of personalized medicine, the assessment of prognostic factors is crucial for individual treatment decision making. The activation of oncogenes (i.e., EGFR and KRAS) and the inhibition of tumor-suppressors (TP53) drive tumor progression. While targeting some of these genes is a promising therapeutic strategy in adenocarcinoma (ADC), most lung cancers lack proven (targetable) driver genes and identification of additional ones is critical. Recent developments of genome-wide profiling have identified new genes, but the studies reported to date are underpowered or lack a control arm. Bass et al. (2) profiled 40 esophageal squamous cell carcinomas (SCC) (29 primary and 11 cell lines) and 47 primary lung SCC for DNA copy number (CN) change. They reported that SOX2 (chr.3q26.33) was significantly amplified and that it was a lineage-survival oncogene by knockdown experiments in cell lines. However, the small sample size hindered assessment of the prognostic value of CN aberrations (CNA). The Cancer Genome Atlas (TCGA) recruited 10,000 samples from 33 cancer types and profiled alterations from genomic DNA, RNA, and protein. However, due to the inclusion criteria (≥70% tumor cellularity), advanced stages were underrepresented. Furthermore, the samples used in these studies were snap-frozen tissues whereas most of the samples in clinical settings are formalin-fixed and paraffin-embedded (FFPE). Thus, identifying prognostic markers from FFPE samples may be clinically relevant.

The Lung Adjuvant Cisplatin Evaluation (LACE-Bio) project comprises FFPE samples from four LACE adjuvant chemotherapy (ACT) trials and evaluated the prognostic and predictive role of biomarkers including ERCC1 (3), tumor infiltrating lymphocytes (TILs) (4), mucin (5), beta-tubulin (6), KRAS (7), EGFR (8) and TP53 (9). Importantly, 1,013 samples from three trials were profiled for their DNA CNAs. Since the trials were randomized and controlled, the data were fit for evaluating markers associated with the magnitude of ACT benefit.

Methods

Patients and samples

The LACE-Bio2 consortium includes patients from four pivotal trials comparing platinum-based ACT to observation after complete resection of stage I–III NSCLC (10-15). Of these, 1,013 patients from three trials had FFPE samples available, whereas samples in one trial (15) were exhausted. All individual trials including tissue collection for future research were approved by institutional review boards at each participating site.

DNA isolation and profiling

DNA was successfully extracted from 976 FFPE samples using the AllPrep DNA/RNA FFPE Kit (Quagen, Germantown, MD, USA), and profiled using the OncoScan CNV Plus Assay (ThermoFisher, Carlsbad, California, USA), a molecular inversion probe SNP assay (16). The platform algorithm delivered the median of the absolute values of all pairwise differences (MAPD) (17,18) as quality metrics; 777 samples with MAPD ≤0.3 were classified as optimal quality.

Statistical analyses

The data were normalized relative to a pool of reference normal samples and segmented using circular binary segmentation (19,20). Minimal recurrent regions were identified via the CGHregions algorithm (21). Tumor clonal composition number was estimated by using the OncoClone composition program (22). The primary endpoint was disease-free survival (DFS). Secondary endpoints were overall survival (OS) and lung-cancer specific survival (LCSS). CNAs were correlated to endpoints using Cox models stratified by trial and adjusted for treatment and clinicopathological factors. The regression models estimated the hazard ratio HRgain for a 2-fold higher CN, with HRloss = 1/HRgain the relative hazard for a 2-fold lower CN. The predictive role of CNAs was estimated by further adding a treatment-by-CN interaction to the models. We performed univariate (by region) and two multivariate analyses (stepwise selection and penalized regression) (23,24). Q values were used to correct P values for multiple comparisons (25).

Preplanned sensitivity analyses included: histologic subgroups (ADC vs. SCC), optimal quality subgroup. CN differences between histologies were assessed by t-tests, with P values corrected via step-down multiple testing procedures (26,27). We compared results to those from our reanalysis of the TCGA (28,29) using exactly the same method. Known tumor suppressors and oncogenes were obtained from literature (30).

The association of the number of breakpoints (BPs), quantifying chromosomal instability, with clinicopathological factors was tested in univariate analyses, then in multivariate log-linear models. The association of chromosomal instability and of clonality with outcomes and treatment effect was studied in Cox models.

Full details of statistical methods are provided in the supplementary material.

Results

Three samples (Figure S1) were partially processed; 1 failed linkage to the clinical database; the inferred gender of 32 patients was incorrect; 1 sample was duplicate. In total, 976 samples were analyzed: 414 (42%) ADC, 430 (44%) SCC, 132 (14%) other NSCLC; 485 were in the control and 491 in the ACT groups (Table 1).

Figure S1.

Figure S1

Flowchart. FFPE, formalin fixation and paraffin embedding; CALBG, Cancer and Leukemia Group B trial 9633 (8); IALT, International Adjuvant Lung Trial (4,5); JBR.10, National Cancer Institute of Canada intergroup (6,7); CAN, copy number aberration.

Table 1. Demographic characteristics of patients with OncoScan analysis results.

Characteristics Control group (N=485) (No., %) Chemotherapy group (N=491) (No., %) Total (N=976) (No., %)
Trial
   CALGB 66 [14] 58 [12] 124 [13]
   IALT 258 [53] 266 [54] 524 [54]
   JBR 10 161 [33] 167 [34] 328 [34]
Age
   ≤55 137 [28] 137 [28] 274 [28]
   55–64 202 [42] 205 [42] 407 [42]
   ≥65 146 [30] 149 [30] 295 [30]
Sex
   Female 139 [29] 127 [26] 266 [27]
   Male 346 [71] 364 [74] 710 [73]
PS
   0 248 [51] 252 [51] 500 [51]
   1–2 235 [49] 238 [49] 473 [49]
Histology
   Adenocarcinoma 207 [43] 207 [42] 414 [42]
   Squamous cell carcinoma 218 [45] 212 [43] 430 [44]
   Other 60 [12] 72 [15] 132 [14]
T
   T1 57 [12] 64 [13] 121 [12]
   T2 372 [77] 365 [75] 737 [76]
   T3/T4 54 [11] 60 [12] 114 [12]
N
   N0 250 [52] 251 [51] 501 [52]
   N1 167 [35] 175 [36] 342 [35]
   N2 66 [14] 63 [13] 129 [13]
Surgery
   Lobectomy/other 344 [71] 333 [68] 677 [69]
   Pneumonectomy 141 [29] 157 [32] 298 [31]

The 217,611 array probes were grouped into 431 common-CN regions; 253 regions (59%) were ≤3 Mb, 340 (79%) were ≤10 Mb (Figure 1); 166 regions had a loss (177 a gain) in ≥10% patients. On average, patients had 94 CNAs (standard deviation 69), 51 gains and 43 losses.

Figure 1.

Figure 1

The landscape of copy number aberrations in all 976 LACE-Bio patients available for OncoScan assay analysis.

The most frequent CN gains (Table S1) were in 1q21–23, 3q22–26, 5p13–15, 6p24, 8q21–24, 22q11, containing genes TERT, PIK3CA, MECOM, CCNL1 among others. The most frequent CN losses were in chromosomes 3p21.31, 8p23, and 9p21.3, containing CDKN2A/B. These results remained consistent in the optimal quality samples subset (N=777; Figure S2, Table S2).

Table S1. Most frequent copy number aberrations in all the samples (N=976).

Chr Region ID Loss Freq Gain Freq Start End Mb Genes cytoBands
1 6 15.8% 44.7% 110 231 909 110 240 929 9.0E−3 p13.3
13 0.5% 31.8% 145 394 955 148 544 968 3.0E+0 BCL9, TXNIP q21.1–2
15 0.5% 32.8% 149 742 045 161 515 326 1.0E+1 MLLT11, NTRK1, PRCC, TPM3, PYHIN1, EFNA1, MUC1, PLEKHO1, AIM2 q21.2–q23.3
16 3.8% 30.6% 161 591 477 161 607 441 2.0E−2 q23.3
17 1.7% 31.7% 161 609 660 161 622 701 1.0E−2 q23.3
3 48 30.0% 0.2% 46 804 388 46 831 840 3.0E−2 p21.31
50 31.0% 1.6% 75 444 906 75 554 646 1.0E−1 p12.3
64 3.5% 31.2% 134 402 484 143 774 592 9.0E+0 XRN1 q22.2–q24
65 3.0% 36.9% 143 794 370 151 504 070 8.0E+0 WWTR1 q24, q25.1
66 3.6% 38.2% 151 520 944 151 546 041 3.0E−2 q25.1
67 2.8% 39.8% 151 563 527 162 500 864 1.0E+1 CCNL1, GMPS q25.1–q26.1
68 12.7% 40.0% 162 540 700 162 602 984 6.0E−2 q26.1
69 2.6% 41.5% 162 640 497 162 702 814 6.0E−2 q26.1
70 2.2% 41.8% 162 719 684 165 245 914 3.0E+0 q26.1
71 2.3% 42.6% 165 270 444 165 296 562 3.0E−2 q26.1
72 1.8% 44.8% 165 314 375 169 905 944 5.0E+0 MECOM q26.1–2
73 2.2% 45.4% 169 918 311 175 861 931 6.0E+0 PRKCI, PRKCI q26.2–32
74 2.6% 45.1% 175 889 230 175 905 626 2.0E−2 q26.32
75 2.4% 45.5% 175 920 884 187 866 388 1.0E+1 PIK3CA, DCUN1D1, BCL6 q26.32–q27.3
76 3.2% 43.1% 187 870 778 189 361 993 1.0E+0 q27.3–q28
77 3.5% 43.0% 189 365 570 189 367 551 2.0E−3 q28
78 3.1% 42.7% 189 370 963 195 341 037 6.0E+0 q28–q29
79 3.3% 42.0% 195 419 229 197 852 564 2.0E+0 q29
5 101 0.3% 47.6% 38 139 685 504 6.0E−1 SDHA p15.33
102 2.4% 47.1% 718 972 766 213 5.0E−2 p15.33
103 0.2% 45.7% 776 473 8 685 711 8.0E+0 TERT p15.33–31
104 1.5% 45.6% 8 704 021 8 737 812 3.0E−2 p15.31
105 0.3% 45.9% 8 753 733 17 516 734 9.0E+0 p15.31–p15.1
106 1.5% 45.9% 17 602 685 17 634 942 3.0E−2 p15.1
107 0.3% 44.0% 17 648 614 32 164 826 1.0E+1 p15.1–p14.3
108 0.2% 43.4% 32 168 437 45 893 362 1.0E+1 DAB2 p13.3–p12
109 1.2% 40.3% 45 895 885 45 915 513 2.0E−2 p12
110 1.5% 39.0% 45 939 674 46 381 782 4.0E−1 p12–p11
6 122 1.5% 40.3% 11 488 926 11 492 749 4.0E−3 p24.2
8 159 30.4% 2.6% 172 417 2 232 383 2.0E+0 p23.3–2
160 31.8% 2.5% 2 254 703 2 260 986 6.0E−3 p23.2
177 33.2% 14.0% 39 274 995 39 383 000 1.0E−1 p11.22
199 0.7% 31.7% 91 055 345 114 039 680 2.0E+1 RUNX1T1, TP53INP1 q21.3–q23.3
200 2.6% 31.1% 114 041 368 114 044 217 3.0E−3 q23.3
201 0.9% 33.2% 114 052 153 123 551 840 9.0E+0 EXT1 q23.3–q24.13
202 0.7% 38.2% 123 567 563 130 055 981 6.0E+0 MYC, MTSS1 q24.13–21
203 1.0% 35.6% 130 070 130 137 656 246 8.0E+0 WISP1 q24.21–23
204 4.3% 32.6% 137 693 433 137 855 026 2.0E−1 q24.23
205 1.0% 33.7% 137 862 600 146 114 526 8.0E+0 MAFA, MAFA q24.3–23
9 207 31.9% 4.0% 204 738 12 433 357 1.0E+1 JAK2, KANK1, PTPRD p24.3–p23
208 32.3% 4.0% 12 445 364 13 036 438 6.0E−1 p23
209 32.8% 3.9% 13 059 473 16 048 844 3.0E+0 p23–p22.3
210 31.4% 3.9% 16 060 347 20 876 513 5.0E+0 MLLT3 p22.3–p21.3
211 34.8% 3.4% 20 890 669 21 179 174 3.0E−1 p21.3
212 35.9% 3.3% 21 194 379 21 778 976 6.0E−1 p21.3
213 38.7% 3.2% 21 785 018 21 845 577 6.0E−2 p21.3
214 40.2% 3.0% 21 853 221 22 176 560 3.0E−1 CDKN2A, CDKN2B p21.3
215 36.3% 3.3% 22 202 151 23 953 634 2.0E+0 p21.3
216 34.7% 4.1% 23 971 815 24 725 697 8.0E−1 p21.3
217 35.0% 3.9% 24 741 204 24 750 179 9.0E−3 p21.3
218 32.8% 4.5% 24 769 948 25 268 867 5.0E−1 p21.3
219 30.5% 4.6% 25 294 701 27 670 083 2.0E+0 p21.3–2
220 30.1% 4.8% 27 678 194 27 700 539 2.0E−2 p21.2
22 425 21.3% 45.5% 24 346 428 24 390 318 4.0E−2 q11.23

Figure S2.

Figure S2

Copy number aberrations in optimal quality (MAPD ≤0.3) samples only (N=777).

Table S2. Most frequent copy number aberrations in the optimal quality samples only (N=777).

Chr Region ID Loss Freq Gain Freq Start End Mb Genes cytoBands
1 13 1% 34% 145 394 955 148 544 968 3.0E+0 BCL9, TXNIP q21.1–2
1 15 0% 35% 149 742 045 161 515 326 1.0E+1 MLLT11, NTRK1, PRCC, TPM3, PYHIN1, EFNA1, MUC1, PLEKHO1, AIM2 q21.2–q23.3
1 16 1% 32% 161 591 477 161 607 441 2.0E−2 q23.3
1 17 1% 34% 161 609 660 161 622 701 1.0E−2 q23.3
1 18 0% 32% 161 641 596 196 703 707 4.0E+1 FCGR2B, PBX1, TPR, LHX4, CDC73 q23.3–q31.3
3 65 3% 32% 143 794 370 151 504 070 8.0E+0 WWTR1 q24–q25.1
3 66 3% 33% 151 520 944 151 546 041 3.0E−2 q25.1
3 67 3% 34% 151 563 527 162 500 864 1.0E+1 CCNL1, GMPS q25.1–q26.1
3 69 2% 34% 162 640 497 162 702 814 6.0E−2 q26.1
3 70 2% 34% 162 719 684 165 245 914 3.0E+0 q26.1
3 71 2% 34% 165 270 444 165 296 562 3.0E−2 q26.1
3 72 2% 35% 165 314 375 169 905 944 5.0E+0 MECOM q26.1–2
3 73 2% 34% 169 918 311 175 861 931 6.0E+0 PRKCI, PRKCI q26.2–32
3 74 2% 33% 175 889 230 175 905 626 2.0E−2 q26.32
3 75 2% 33% 175 920 884 187 866 388 1.0E+1 PIK3CA, DCUN1D1, BCL6 q26.32–q27.3
3 76 3% 33% 187 870 778 189 361 993 1.0E+0 q27.3–q28
3 77 3% 32% 189 365 570 189 367 551 2.0E−3 q28
3 78 3% 34% 189 370 963 195 341 037 6.0E+0 q28–q29
3 79 3% 34% 195 419 229 197 852 564 2.0E+0 q29
5 101 0% 47% 38 139 685 504 6.0E−1 SDHA p15.33
5 102 1% 45% 718 972 766 213 5.0E−2 p15.33
5 103 0% 46% 776 473 8 685 711 8.0E+0 TERT p15.33–31
5 104 0% 45% 8 704 021 8 737 812 3.0E−2 p15.31
5 105 0% 45% 8 753 733 17 516 734 9.0E+0 p15.31–p15.1
5 106 0% 45% 17 602 685 17 634 942 3.0E−2 p15.1
5 107 0% 44% 17 648 614 32 164 826 1.0E+1 p15.1–p13.3
5 108 0% 42% 32 168 437 45 893 362 1.0E+1 DAB2 p13.3–p12
5 109 1% 41% 45 895 885 45 915 513 2.0E−2 p12
5 110 1% 39% 45 939 674 46 381 782 4.0E−1 p12–p11
8 199 1% 32% 91 055 345 114 039 680 2.0E+1 RUNX1T1, TP53INP1 q21.3–q23.3
8 200 1% 32% 114 041 368 114 044 217 3.0E−3 q23.3
8 201 1% 34% 114 052 153 123 551 840 9.0E+0 EXT1 q23.3–q24.13
8 202 1% 37% 123 567 563 130 055 981 6.0E+0 MYC, MTSS1 q24.13–21
8 203 1% 36% 130 070 130 137 656 246 8.0E+0 WISP1 q24.21–23
8 204 2% 33% 137 693 433 137 855 026 2.0E−1 q24.23
8 205 1% 34% 137 862 600 146 114 526 8.0E+0 MAFA q24.3–23
9 209 30% 4% 13 059 473 16 048 844 3.0E+0 p23–p22.3
9 213 30% 3% 21 785 018 21 845 577 6.0E−2 p21.3
9 214 31% 3% 21 853 221 22 176 560 3.0E−1 CDKN2A, CDKN2B p21.3
9 215 30% 3% 22 202 151 23 953 634 2.0E+0 p21.3

The CN profile was heterogeneous across histology and results were confirmed in our reanalysis of the TCGA data (Figure S3). The frequency of 195 regions (49% were ≤3 Mb and 71% ≤10 Mb; Table S3) was significantly different between ADC and SCC (Q≤0.05). The most significant differences were: more gains in 3q (including genes PIK3CA, MECOM, CCNL1), 22q (NF2, PDGFB) and 12p (KRAS) in SCC; more losses in 3p (RASSF1), 4 (PTTG2, NKX2-1), and 5q in SCC.

Figure S3.

Figure S3

Copy number aberrations in adenocarcinomas (A and D) and squamous cell carcinomas (B and E) in the LACE-Bio (A,B,C) and the Cancer Genome Atlas (TCGA) data (D,E,F).

Table S3. Regions [195] with significantly (Q ≤0.05) different copy number aberration frequency between adenocarcinomas and squamous cell carcinomas in all the samples (N=976).

Chr Region ID Loss Freq Gain Freq Q Start End Mb Genes cytoBands
ADC SCC ADC SCC
1 1 7% 18% 1% 1% 0.001 754 192 12 833 428 1.0E+0 SKI, PARK7, CHD5, ERRFI1, TP73 p36.21–33
3 5% 15% 2% 1% 0.001 13 181 849 72 758 707 6.0E+0 EPS15, FGR, JUN, LCK, PAX7, STIL, TAL1, NBL1, EPHB2, MUTYH, NBL1, ARNT p36.21–p31.1
5 8% 17% 2% 1% 0.001 72 814 783 110 222 219 4.0E+0 BCL10 p31.1–p13.3
7 10% 21% 1% 0% 0.001 110 246 359 110 761 020 5.0E−2 p13.3
8 11% 18% 2% 1% 0.001 110 777 105 120 508 803 1.0E+0 NRAS, RBM15, RAP1A p13.3–p12
11 1% 6% 26% 13% 0.001 144 852 910 145 095 477 2.0E−2 q21.1
12 1% 3% 31% 16% 0.001 145 115 883 145 382 341 3.0E−2 q21.1
13 0% 1% 43% 22% 0.001 145 394 955 148 544 968 3.0E−1 BCL9, TXNIP q21.1–2
15 0% 1% 45% 22% 0.001 149 742 045 161 515 326 1.0E+0 MLLT11, NTRK1, PRCC, TPM3, PYHIN1, EFNA1, MUC1, PLEKHO1, AIM2 q21.2–q23.3
16 3% 5% 41% 21% 0.027 161 591 477 161 607 441 2.0E−3 q23.3
17 1% 2% 42% 22% 0.001 161 609 660 161 622 701 1.0E−3 q23.3
18 0% 0% 37% 22% 0.001 161 641 596 196 703 707 4.0E+0 FCGR2B, PBX1, TPR, LHX4, CDC73 q23.3–q31.3
20 1% 1% 37% 20% 0.001 196 823 613 196 882 344 6.0E−3 q31.3
21 1% 1% 37% 19% 0.001 196 922 021 248 687 952 5.0E+0 FH, PHLDA3, LIN9, LGR6, RASSF5 q31.3–q44
22 2% 3% 37% 20% 0.001 248 773 062 249 212 878 4.0E−2 q44
2 23 1% 1% 2% 7% 0.001 21 494 34 689 435 3.0E+0 ALK, MYCN, NCOA1, RHOB p25.3–p22.3
25 1% 0% 2% 10% 0.001 34 741 001 89 572 881 5.0E+0 REL, MSH2 p22.3–p11.2
32 4% 14% 3% 5% 0.006 141 786 613 141 882 709 1.0E−2 q22.1
33 4% 17% 4% 5% 0.001 141 893 894 142 075 788 2.0E−2 q22.1
42 1% 11% 3% 2% 0.001 206 472 683 220 020 335 1.0E+0 IDH1 q33.3–q35
43 3% 15% 3% 1% 0.001 220 035 105 220 042 675 8.0E−4 q35
44 3% 13% 3% 1% 0.001 220 056 954 242 834 648 2.0E+0 PAX3, DIS3L2 q35–q37.3
3 46 15% 36% 1% 1% 0.001 63 411 30 625 123 3.0E+0 RAF1, RARB, VHL p26.3–p24.1
47 16% 38% 1% 0% 0.001 30 638 028 46 778 842 2.0E+0 MLH1, LIMD1, DLEC1 p24.1–p21.31
48 18% 41% 1% 0% 0.004 46 804 388 46 831 840 3.0E−3 p21.31
49 17% 41% 1% 1% 0.001 46 852 679 75 394 787 3.0E+0 RHOA, NCKIPSD, TCTA, USP4, NPRL2, RASSF1, TUSC2, FHIT, NAT6, PBRM1, PRKCD p21.31–p12.3
50 18% 43% 2% 1% 0.001 75 444 906 75 554 646 1.0E−2 p12.3
51 16% 40% 1% 1% 0.001 75 815 879 78 927 132 3.0E−1 p12.3
52 16% 40% 1% 2% 0.001 78 930 451 78 937 737 7.0E−4 p12.3
53 17% 39% 2% 2% 0.001 78 939 727 84 046 628 5.0E−1 p12.3–1
54 15% 35% 3% 6% 0.001 84 068 424 89 392 778 5.0E−1 p12.1–p11.1
56 13% 33% 5% 7% 0.001 89 423 343 90 418 473 1.0E−1 p11.1
58 9% 5% 9% 34% 0.001 93 530 364 100 327 532 7.0E−1 q11.1–q12.2
59 8% 4% 10% 36% 0.001 100 351 896 111 493 739 1.0E+0 TFG q12.2–q13.13
60 7% 4% 11% 39% 0.001 111 521 268 111 555 200 3.0E−3 q13.2
61 7% 3% 10% 40% 0.001 111 567 238 129 762 859 2.0E+0 q13.2–q22.1
62 6% 2% 12% 43% 0.001 129 784 388 129 810 022 3.0E−3 q22.1
63 6% 2% 12% 46% 0.001 129 823 705 134 398 090 5.0E−1 q22.1–2
64 5% 1% 14% 50% 0.001 134 402 484 143 774 592 9.0E−1 XRN1 q22.2–q24
65 5% 0% 14% 61% 0.001 143 794 370 151 504 070 8.0E−1 WWTR1 q24–q25.1
66 5% 1% 15% 64% 0.001 151 520 944 151 546 041 3.0E−3 q25.1
67 5% 0% 15% 67% 0.001 151 563 527 162 500 864 1.0E+0 CCNL1, GMPS q25.1–q26.1
68 15% 11% 21% 61% 0.001 162 540 700 162 602 984 6.0E−3 q26.1
69 4% 1% 18% 68% 0.001 162 640 497 162 702 814 6.0E−3 q26.1
70 4% 0% 17% 69% 0.001 162 719 684 165 245 914 3.0E−1 q26.1
71 4% 0% 18% 70% 0.001 165 270 444 165 296 562 3.0E−3 q26.1
72 3% 0% 18% 74% 0.001 165 314 375 169 905 944 5.0E−1 MECOM q26.1–2
73 4% 0% 17% 77% 0.001 169 918 311 175 861 931 6.0E−1 PRKCI, PRKCI q26.2–32
74 4% 1% 16% 77% 0.001 175 889 230 175 905 626 2.0E−3 q26.32
75 4% 0% 16% 78% 0.001 175 920 884 187 866 388 1.0E+0 PIK3CA, DCUN1D1, BCL6 q26.32–q27.3
76 5% 1% 15% 74% 0.001 187 870 778 189 361 993 1.0E−1 q27.3–q28
77 5% 1% 15% 74% 0.001 189 365 570 189 367 551 2.0E−4 q28
78 5% 0% 15% 73% 0.001 189 370 963 195 341 037 6.0E−1 q28–q29
79 6% 0% 15% 72% 0.001 195 419 229 197 852 564 2.0E−1 q29
4 80 5% 21% 3% 1% 0.001 69 404 9 153 037 9.0E−1 WHSC1 p16.3–1
82 5% 22% 3% 0% 0.001 9 586 764 34 772 494 3.0E+0 p16.1–p15.1
84 3% 20% 3% 1% 0.001 34 847 676 48 061 771 1.0E+0 PTTG2 p15.1–p12
85 3% 16% 5% 4% 0.001 48 083 885 49 085 053 1.0E−1 p12–p11
86 3% 16% 4% 3% 0.001 49 085 414 49 092 454 7.0E−4 p11
92 9% 23% 2% 3% 0.001 87 465 741 88 195 494 7.0E−2 AFF1 q21.3–q22.1
93 5% 19% 2% 4% 0.001 88 208 266 90 739 539 3.0E−1 q22.1
95 6% 21% 1% 2% 0.001 90 784 528 122 271 282 3.0E+0 TET2 q22.1–q27
96 7% 25% 1% 2% 0.001 122 282 972 122 288 144 5.0E−4 q27
97 6% 25% 2% 2% 0.001 122 299 078 134 264 058 1.0E+0 IL2 q27–q28.3
98 6% 26% 2% 2% 0.001 134 271 747 134 295 157 2.0E−3 q28.3
99 6% 27% 3% 2% 0.001 134 304 705 166 810 338 3.0E+0 q28.3–q32.3
100 12% 30% 1% 1% 0.001 166 830 580 190 915 650 2.0E+0 HPGD q32.3–q35.2
5 111 5% 17% 27% 22% 0.005 49 441 966 49 562 291 1.0E−2 q11.1
112 10% 26% 16% 9% 0.001 49 597 497 49 608 094 1.0E−3 q11.1
113 10% 29% 13% 7% 0.001 49 640 141 51 484 497 2.0E−1 q11.1–2
114 13% 35% 8% 2% 0.001 51 505 665 60 219 800 9.0E−1 PLK2, PDE4D q11.2–q12.1
115 15% 36% 6% 1% 0.001 60 241 946 68 828 372 9.0E−1 q12.1–q13.2
116 17% 36% 5% 1% 0.001 70 306 678 112 950 805 4.0E+0 FER, RASA1, APC, MCC q13.2–q22.2
118 18% 36% 5% 1% 0.001 112 997 656 139 372 404 3.0E+0 AFF4, IRF1 q22.2–q31.2
119 15% 31% 11% 6% 0.010 139 379 707 139 400 093 2.0E−3 q31.2
120 16% 35% 5% 1% 0.001 139 411 703 180 698 312 4.0E+0 CSF1R, ARHGAP26, NPM1, PDGFRB, NSD1, PTTG1 q31.2–q35.3
6 121 1% 7% 9% 4% 0.001 204 909 11 474 632 1.0E+0 p25.3–p24.2
123 0% 6% 10% 5% 0.001 11 521 599 31 276 175 2.0E+0 DEK p24.2–p21.33
125 1% 6% 10% 2% 0.001 31 297 365 32 528 026 1.0E−1 p21.33–32
126 1% 7% 11% 2% 0.001 32 561 716 32 577 756 2.0E−3 p21.32
127 1% 5% 9% 2% 0.001 32 581 816 42 552 548 1.0E+0 PIM1 p21. 32–p21.1
128 0% 3% 10% 7% 0.026 42 572 859 51 038 424 8.0E−1 VEGFA p21.1–p12.3
133 6% 2% 4% 5% 0.017 64 281 705 65 202 867 9.0E−2 q12
134 11% 3% 2% 3% 0.001 65 286 066 78 962 125 1.0E+0 q12–q14.1
136 16% 6% 1% 1% 0.001 79 042 157 103 728 158 2.0E+0 UFL1 q14.1–q16.3
138 14% 7% 1% 2% 0.001 103 766 477 170 913 051 7.0E+0 FOXO3, FYN, MAS1, MLLT4, MYB, ROS1, SASH1, FRK, RPS6KA2, LATS1 q16.3–q27
7 152 2% 1% 14% 20% 0.001 88 259 445 100 958 270 1.0E+0 q21.13–q22.1
8 163 25% 36% 4% 2% 0.023 16 025 118 25 073 138 9.0E−1 PCM1, LZTS1, DMTN, NKX3-1, MTUS1 p22–p21.2
169 19% 20% 8% 20% 0.047 36 025 847 36 621 588 6.0E−2 p12–p11.23
170 18% 15% 10% 25% 0.001 36 633 318 37 651 477 1.0E−1 p11.23
171 18% 13% 11% 28% 0.001 37 667 018 37 767 527 1.0E−2 p11.23
172 17% 11% 11% 30% 0.001 37 786 457 38 127 768 3.0E−2 PPAPDC1B p11.23
173 17% 10% 11% 31% 0.001 38 137 530 38 139 729 2.0E−4 WHSC1L1 p11.23
174 17% 9% 10% 32% 0.001 38 143 357 38 528 508 4.0E−2 WHSC1L1 p11.23–22
175 17% 10% 10% 30% 0.001 38 552 757 39 217 074 7.0E−2 p11.22
178 15% 11% 11% 27% 0.001 39 412 457 39 969 006 6.0E−2 p11.22–21
179 15% 12% 11% 23% 0.001 39 976 970 41 058 677 1.0E−1 p11.21
180 14% 11% 12% 22% 0.001 41 077 098 41 261 544 2.0E−2 p11.21
181 13% 10% 13% 22% 0.003 41 280 152 42 559 586 1.0E−1 KAT6A p11.21
182 12% 10% 13% 20% 0.012 42 574 931 43 157 099 6.0E−2 p11.21–1
9 211 28% 42% 3% 3% 0.003 20 890 669 21 179 174 3.0E−2 p21.3
212 29% 43% 3% 3% 0.001 21 194 379 21 778 976 6.0E−2 p21.3
213 32% 46% 3% 3% 0.001 21 785 018 21 845 577 6.0E−3 p21.3
214 33% 48% 3% 2% 0.001 21 853 221 22 176 560 3.0E−2 CDKN2A, CDKN2B p21.3
215 30% 44% 3% 3% 0.012 22 202 151 23 953 634 2.0E−1 p21.3
216 28% 41% 4% 4% 0.017 23 971 815 24 725 697 8.0E−2 p21.3
232 21% 16% 1% 4% 0.001 71 035 938 91 434 530 2.0E+0 q21.11–q22.1
233 19% 14% 2% 5% 0.003 91 440 590 96 341 196 5.0E−1 FAM120A q22.1–31
234 18% 14% 1% 4% 0.005 96 382 906 141 054 761 4.0E+0 ABL1, SET, TAL2, NR4A3, NUP214, GFI1B, DAB2IP, DEC1, PTCH1, TSC1 q22.31–q34.3
10 235 3% 8% 5% 3% 0.001 126 070 35 317 317 4.0E+0 BMI1, NET1, MAP3K8, BMI1, MLLT10, ZMYND11 p15.3–p11.21
237 3% 6% 6% 3% 0.001 35 351 249 42 608 180 7.0E−1 p11.21–q11.21
238 4% 9% 4% 5% 0.005 42 614 561 46 177 093 4.0E−1 RET, RASSF4 q11.21–22
239 7% 14% 8% 9% 0.024 46 965 151 47 127 279 2.0E−2 q11.22
241 6% 12% 4% 3% 0.001 48 302 618 51 594 462 3.0E−1 NCOA4 q11.22–23
242 7% 15% 2% 2% 0.001 51 785 728 63 841 130 1.0E+0 CCDC6 q11.23–q21.2
243 7% 16% 3% 2% 0.001 63 861 600 68 064 594 4.0E−1 q21.2–3
244 8% 17% 2% 2% 0.001 68 077 764 68 114 481 4.0E−3 q21.3
245 8% 17% 3% 2% 0.001 68 128 614 83 887 644 2.0E+0 q21.3–q23.1
246 8% 20% 3% 1% 0.001 83 894 966 91 400 384 8.0E−1 PTEN q23.1–q23.31
247 9% 18% 3% 1% 0.001 91 422 054 131 496 457 4.0E+0 FRAT1, DMBT1, FGFR2, TLX1, MXI1, NFKB2, WDR11, C10orf90, DMBT1, PDCD4, SUFU, NEURL1 q23.31–q26.3
249 10% 19% 3% 2% 0.001 131 540 611 135 434 303 4.0E−1 q26.3
11 250 3% 12% 0% 0% 0.001 192 764 27 362 359 3.0E+0 RRAS2, CSNK2A3, HRAS, LMO1, AKIP1, CARS, CDKN1C, HTATIP2, PRKCDBP p15.5–p14.1
258 4% 9% 4% 3% 0.015 65 186 348 65 271 832 9.0E−3 q13.1
259 1% 5% 7% 5% 0.004 65 283 143 67 656 861 2.0E−1 BRMS1 q13.1–2
264 3% 8% 7% 5% 0.001 71 620 699 77 051 820 5.0E−1 q13.4–5
265 4% 10% 8% 3% 0.001 77 055 862 89 473 402 1.0E+0 PICALM q13.5–q14.3
266 8% 12% 5% 2% 0.007 89 654 897 118 621 410 3.0E+0 YAP1, DDX6, KMT2A, POU2AF1, ZBTB16, PDGFD, CADM1, ATM, ARHGAP20 q14.3–q23.3
12 268 6% 1% 11% 22% 0.001 189 400 866 208 7.0E−2 p13.33
270 7% 1% 9% 24% 0.001 876 288 9 623 841 9.0E−1 FGF6, ING4 p13.33–31
273 8% 1% 9% 24% 0.001 9 739 885 11 501 856 2.0E−1 STYK1 p13.31–2
274 14% 7% 8% 22% 0.001 11 515 281 11 543 338 3.0E−3 p13.2
275 8% 1% 7% 23% 0.001 11 555 467 17 613 438 6.0E−1 CDKN1B, ETV6, CREBL2 p12.3–p13.2
276 5% 1% 12% 23% 0.001 17 635 521 31 241 345 1.0E+0 KRAS p12.3–p11.21
277 4% 1% 10% 20% 0.001 31 296 219 33 982 162 3.0E−1 p11.21–1
278 4% 2% 9% 18% 0.001 33 994 599 34 749 065 8.0E−2 p11.1
279 3% 2% 9% 15% 0.026 34 755 947 34 828 211 7.0E−3 p11.1
285 3% 1% 5% 6% 0.004 72 362 265 93 623 290 2.0E+0 BTG1, ZDHHC17 q21.1–q22
13 294 13% 23% 4% 3% 0.018 81 036 784 103 258 600 2.0E+0 q31.1–q33.1
295 13% 24% 4% 2% 0.001 103 273 465 103 286 814 1.0E−3 q33.1
14 303 3% 11% 17% 8% 0.001 28 292 800 34 485 112 6.0E−1 q12–q13.1
304 2% 10% 22% 9% 0.001 34 490 900 35 596 092 1.0E−1 q13.1–2
305 2% 10% 25% 11% 0.001 35 605 465 38 959 413 3.0E−1 NKX2-1, MBIP q13.2–q21.1
306 2% 10% 21% 10% 0.001 38 976 380 39 121 670 1.0E−2 q21.1
307 2% 10% 20% 9% 0.001 39 186 659 39 246 264 6.0E−3 q21.1
308 3% 10% 18% 8% 0.001 39 311 307 41 601 018 2.0E−1 PNN q21.1
310 3% 10% 16% 8% 0.001 41 673 714 43 083 557 1.0E−1 q21.1
311 4% 11% 12% 7% 0.022 43 093 389 61 786 976 2.0E+0 q21.1–q23.1
312 4% 12% 10% 7% 0.014 61 799 920 61 917 178 1.0E−2 q23.1
15 327 13% 8% 1% 2% 0.001 20 161 372 34 671 061 1.0E+0 q11.1–q14
329 14% 7% 1% 2% 0.001 34 870 223 38 622 707 4.0E−1 q14
330 14% 7% 1% 1% 0.001 38 623 948 43 995 380 5.0E−1 ZFYVE19, BUB1B q14–q15.3
331 10% 4% 2% 3% 0.001 44 016 417 76 738 959 3.0E+0 PML, ARID3B, PML q15.3–q24.3
332 8% 3% 2% 5% 0.001 76 752 698 93 407 788 2.0E+0 AKAP13, FES, FES, ST20, IDH2 q24.3–q26.1
333 7% 2% 3% 7% 0.001 93 429 646 102 397 317 9.0E−1 q26.1–3
16 334 4% 10% 4% 0% 0.001 83 887 6 988 411 7.0E−1 TSC2, AXIN1 p13.3
335 4% 9% 13% 4% 0.008 6 999 231 7 013 483 1.0E−3 p13.3
336 4% 7% 7% 1% 0.001 7 023 927 28 609 205 2.0E+0 MYH11, TNFRSF17, LITAF, PALB2 p13.3–p11.2
338 2% 5% 6% 2% 0.002 28 628 225 32 137 965 4.0E−1 FUS, PYCARD p11.2
17 346 16% 24% 1% 1% 0.004 400 959 18 928 388 2.0E+0 CRK, ELAC2, GAS7, USP6, TP53, KCTD11, DPH1, FLCN, HIC1, XAF1 p13.3–p11.2
348 8% 17% 3% 3% 0.001 21 690 667 22 217 883 5.0E−2 p11.2–1
352 2% 4% 12% 7% 0.001 34 815 264 36 854 507 2.0E−1 q12
353 1% 6% 10% 8% 0.001 36 861 302 45 005 703 8.0E−1 ERBB2, MLLT6, RARA, WNT3, BRCA1 q12–q21.32
360 1% 7% 15% 11% 0.001 80 185 188 80 263 427 8.0E−3 q25.3
18 361 7% 5% 4% 12% 0.001 12 842 14 240 269 1.0E+0 YES1, EPB41L3 p11.32–21
362 7% 5% 4% 10% 0.001 14 270 974 15 377 471 1.0E−1 p11.21
363 8% 5% 5% 9% 0.026 18 554 999 21 648 788 3.0E−1 q11.1–2
364 11% 6% 4% 10% 0.001 21 659 508 24 123 575 2.0E−1 ZNF521, SS18 q11.2
365 14% 7% 3% 10% 0.001 24 143 454 27 670 629 4.0E−1 q11.2–q12.1
366 15% 7% 4% 11% 0.001 27 678 287 29 104 698 1.0E−1 q12.1
367 15% 8% 3% 9% 0.001 29 119 357 29 715 321 6.0E−2 q12.1
368 16% 9% 3% 9% 0.001 29 736 017 29 737 077 1.0E−4 q12.1
369 14% 8% 3% 9% 0.001 29 754 749 29 779 205 2.0E−3 q12.1
370 14% 8% 3% 9% 0.001 29 790 889 30 339 291 5.0E−2 q12.1
371 15% 9% 2% 7% 0.001 30 358 394 33 590 529 3.0E−1 q12.1–2
19 378 11% 6% 0% 0% 0.001 1 335 531 9 051 725 8.0E−1 MLLT1, SH3GL1, TCF3, VAV1 p13.3–2
380 11% 4% 1% 3% 0.001 9 059 232 20 499 493 1.0E+0 LYL1, RAB8A, ELL, CDKN2D p13.2–p12
382 11% 4% 2% 5% 0.001 20 723 899 20 758 368 3.0E−3 p12
383 10% 5% 2% 3% 0.001 20 769 956 24 505 466 4.0E−1 p12–p11
20 399 6% 3% 10% 21% 0.001 69 094 13 595 807 1.0E+0 RASSF2 p13–p12.1
400 5% 3% 11% 20% 0.004 13 618 382 14 780 319 1.0E−1 p12.1
402 5% 4% 12% 20% 0.008 14 827 680 15 557 228 7.0E−2 p12.1
403 5% 2% 12% 22% 0.001 15 560 791 23 693 161 8.0E−1 p12.1–p11.21
404 3% 3% 13% 22% 0.024 23 700 872 25 672 987 2.0E−1 p11.21–p11.1
21 419 10% 18% 1% 1% 0.014 9 648 315 10 964 139 1.0E−1 p11.2–1
420 8% 16% 1% 1% 0.002 14 344 537 34 787 312 2.0E+0 OLIG2, TCP10L q11.2–q22.11
421 8% 19% 1% 1% 0.001 34 796 886 48 097 610 1.0E+0 ERG, ETS2, RUNX1, SIK1 q22.11–q22.3
22 422 6% 5% 2% 9% 0.001 16 054 713 19 009 167 3.0E−1 q11.1–21
423 5% 2% 3% 14% 0.001 19 026 877 21 462 601 2.0E−1 q11.21
424 5% 3% 3% 11% 0.001 21 804 610 24 338 651 3.0E−1 BCR, SMARCB1 q11.21–23
426 6% 6% 12% 18% 0.026 24 394 088 24 396 598 3.0E−4 q11.23
427 7% 3% 3% 11% 0.001 24 398 768 25 917 803 2.0E−1 q11.23–q12.1
428 9% 3% 1% 11% 0.001 25 942 595 36 907 098 1.0E+0 EWSR1, PATZ1, RASL10A, CHEK2, MN1, NF2 q12.1–3
429 8% 3% 2% 10% 0.001 36 919 447 39 343 292 2.0E−1 q12.3–q13.1
430 9% 4% 1% 11% 0.001 39 363 830 42 517 758 3.0E−1 PDGFB, MKL1 q13.1–2
431 9% 5% 1% 9% 0.001 42 518 382 51 213 826 9.0E−1 PIM3, PRR5 q13.2–33

Copy-number aberrations associated with prognosis

The median follow-up for DFS (510 events) was 5.3 years. In univariate analyses (Table 2), 14 focal regions (11 ≤3 Mb, 14 ≤10 Mb) in loci 7p11–12, 9p21, 18q12, 19p11–13 were prognostic (P≤0.005) with Q≤0.142. Losses associated with shorter DFS were in: 8 regions in 9p21.3 (loss frequency: 31–40%, including CDKN2A/B), with HRloss =1.5 (95% CI: 1.2–1.9) (P<0.001, Q=0.02); one region in 19p13 [STK11, 11%, HRloss =2.4 (1.3–4.3), P=0.005, Q=0.15]; one in 18q12.1 [12%, HRloss =1.6 (1.2–2.3), P=0.004, Q=0.12]. Other seemingly deleterious losses were found in 19p11–13 (MLLT1, SH3GL1, TCF3, VAV1). Gains in 7p11–12 (frequency: 17%) were associated with shorter DFS [HRgain =2.0 (1.2–3.2), P=0.005, Q=0.14]. Two of these regions (7p12.3 and 9p21.3) remained significant in multivariate analyses (Table S4), which also suggested a benefit [HRloss =0.32 (0.16–0.61), P<0.001] for losses in a region in 1p31–36 (9.8%), including EPS15, FGR, JUN, LCK, PAX7, STIL, TAL1, NBL1, EPHB2, MUTYH, ARNT. Penalized regression confirmed the prognostic role of the region in 9p21.3, plus another one containing CDKN2A/B (Table S5).

Table 2. Genomic regions with prognostic effect of copy number aberrations (CNA).

Region ID Chr cytoBands Mb CNA frequency Disease-free survival Overall survival Lung-cancer specific survival Genes
Loss Gain HR for loss* (95% CI) HR for gain** (95% CI) P Q HR for loss* (95% CI) HR for gain** (95% CI) P Q HR for loss* (95% CI) HR for gain** (95% CI) P Q
142 7 p12.3–p11.2 8.0E+0 0.7% 17% 0.51 (0.31–0.82) 2.0 (1.2–3.2) 0.005 0.142
185 8 p11.1–q11.1 4.0E+0 7.3% 17.4% 2.0 (1.2–3.1) 0.51 (0.32–0.82) 0.005 0.130
211 9 p21.3 3.0E−1 34.8% 3.4% 1.7 (1.3–2.3) 0.57 (0.44–0.76) <0.001 0.020 1.8 (1.4–2.5) 0.55 (0.41–0.74) <0.001 0.029 1.8 (1.4–2.4) 0.55 (0.41–0.74) <0.001 0.019
212 9 p21.3 6.0E−1 35.9% 3.3% 1.6 (1.2–2.0) 0.64 (0.49–0.84) 0.001 0.076 1.7 (1.3–2.2) 0.59 (0.44–0.79) <0.001 0.044 1.6 (1.2–2.1) 0.62 (0.47–0.83) 0.001 0.062
213 9 p21.3 6.0E−2 38.7% 3.2% 1.5 (1.2–1.9) 0.67 (0.53–0.86) 0.001 0.076 1.5 (1.2–2.0) 0.66 (0.51–0.86) 0.002 0.079 1.5 (1.2–2.0) 0.65 (0.50–0.85) 0.001 0.062
214 9 p21.3 3.0E−1 40.2% 3.0% 1.5 (1.2–1.9) 0.66 (0.53–0.81) <0.001 0.020 1.5 (1.2–1.9) 0.67 (0.54–0.84) <0.001 0.049 1.6 (1.2–2.0) 0.64 (0.51–0.80) <0.001 0.021 CDKN2A, CDKN2B
215 9 p21.3 2.0E+0 36.3% 3.3% 1.6 (1.2–2.1) 0.62 (0.48–0.81) <0.001 0.034 1.7 (1.3–2.2) 0.59 (0.45–0.79) <0.001 0.044 1.7 (1.3–2.2) 0.61 (0.46–0.80) <0.001 0.029
217 9 p21.3 9.0E−3 35.0% 3.9% 1.7 (1.3–2.2) 0.60 (0.46–0.79) <0.001 0.026 1.5 (1.2–2.1) 0.65 (0.48–0.87) 0.004 0.084 1.7 (1.3–2.3) 0.58 (0.44–0.78) <0.001 0.026
218 9 p21.3 5.0E−1 32.8% 4.5% 1.6 (1.2–2.2) 0.61 (0.45–0.82) 0.001 0.076 1.7 (1.2–2.3) 0.60 (0.43–0.83) 0.002 0.079 1.7 (1.2–2.4) 0.59 (0.42–0.82) 0.002 0.064
219 9 p21.3–2 2.0E+0 30.5% 4.6% 1.8 (1.2–2.6) 0.57 (0.39–0.82) 0.003 0.106 1.8 (1.2–2.7) 0.56 (0.37–0.83) 0.004 0.084 1.9 (1.2–2.8) 0.54 (0.36–0.80) 0.002 0.085
220 9 p21.2 2.0E−2 30.1% 4.8% 1.6 (1.2–2.1) 0.64 (0.47–0.86) 0.004 0.11
222 9 p21.1 2.0E+0 27.8% 5.2% 1.9 (1.2–2.8) 0.54 (0.36–0.81) 0.003 0.084
223 9 p21.1 7.0E−1 26.0% 6.4% 1.8 (1.2–2.8) 0.54 (0.36–0.82) 0.003 0.084
312 14 q23.1 1.0E−1 8.5% 8.9% 2.2 (1.3–3.6) 0.46 (0.28–0.76) 0.002 0.079
366 18 q12.1 1.0E+0 10.8% 8.6% 1.9 (1.2–3.1) 0.52 (0.32–0.82) 0.005 0.105
367 18 q12.1 6.0E−1 10.9% 7.1% 2.0 (1.2–3.3) 0.49 (0.30–0.80) 0.005 0.092
368 18 q12.1 1.0E−3 12.1% 6.6% 1.6 (1.2–2.3) 0.61 (0.44–0.85) 0.004 0.119 1.7 (1.2–2.5) 0.58 (0.41–0.82) 0.002 0.079 1.7 (1.2–2.4) 0.59 (0.42–0.85) 0.004 0.11
376 19 p13.3 1.0E+0 10.7% 0.4% 2.4 (1.3–4.3) 0.42 (0.23–0.77) 0.005 0.142 FSTL3, STK11
378 19 p13.3–2 8.0E+0 9.2% 0.3% 2.6 (1.4–4.8) 0.38 (0.21–0.72) 0.003 0.106 2.9 (1.5–5.6) 0.34 (0.18–0.66) 0.001 0.078 3.4 (1.7–6.6) 0.29 (0.15–0.58) <0.001 0.029 MLLT1, SH3GL1, TCF3, VAV1
379 19 p13.2 3.0E−3 10.3% 0.9% 2.2 (1.3–3.8) 0.45 (0.26–0.76) 0.003 0.106 2.3 (1.3–4.2) 0.43 (0.24–0.77) 0.004 0.092 2.6 (1.4–4.6) 0.39 (0.22–0.69) 0.001 0.062
380 19 p13.2–p12 1.0E+1 8.0% 2.2% 2.7 (1.4–5.1) 0.38 (0.20–0.73) 0.004 0.084 LYL1, RAB8A, ELL, CDKN2D
383 19 p12–p11 4.0E+0 8.0% 2.5% 2.4 (1.4–4.2) 0.42 (0.24–0.74) 0.003 0.106 2.7 (1.5–5.0) 0.36 (0.2–0.66) <0.001 0.066 2.5 (1.3–4.5) 0.41 (0.22–0.75) 0.004 0.11

The univariate hazard ratio (HR) for loss shows the relative risk of a patient with a 2-fold lower CN, for example one copy as compared to two copies. The HR for gain shows the relative risk of a patient with a 2-fold higher CN, for example four copies as compared to two copies. Of note, HR for gain is 1/HR for loss. CI, confidence interval; Chr, chromosome. *, hazard ratio for a 2-fold lower copy number; **, hazard ratio for a 2-fold higher copy number.

Table S4. Prognostic effect of the copy number of genomic regions. Multivariate results.

Chr Region ID CNA frequency Disease-free survival Overall survival Lung-cancer specific survival Mb Genes cytoBands
Losses Gains HR for loss* (95% CI) HR for gain** (95% CI) P HR for loss* (95% CI) HR for gain** (95% CI) P HR for loss* (95% CI) HR for gain** (95% CI) P
1 3 9.8% 1.6% 0.32 (0.16–0.61) 3.2 (1.6–6.1) <0.001 0.21 (0.10–0.43) 4.8 (2.3–10) <0.001 6.0E+1 EPS15, FGR, JUN, LCK, PAX7, STIL, TAL1, NBL1, EPHB2, MUTYH, ARNT p36.21–p31.1
3 72 1.8% 44.8% 1.8 (1.3–2.6) 0.55 (0.38–0.79) 0.001 5.0E+0 MECOM q26.1–2
6 122 1.5% 40.3% 0.78 (0.66–0.91) 1.3 (1.1–1.5) 0.002 4.0E−3 p24.2
7 142 0.7% 17.3% 0.46 (0.29–0.75) 2.1 (1.3–3.5) 0.002 8.0E+0 p12.3–p11.2
8 159 30.4% 2.6% 0.53 (0.34–0.83) 1.9 (1.2–3.0) 0.005 2.0E+0 p23.3–2
9 211 34.8% 3.4% 1.9 (1.4–2.5) 0.54 (0.41–0.72) <0.001 2.1 (1.5–2.8) 0.49 (0.36–0.66) <0.001 2.1 (1.5–3.0) 0.47 (0.34–0.65) <0.001 3.0E−1 p21.3
19 378 9.2% 0.3% 3.7 (1.7–7.7) 0.27 (0.13–0.58) <0.001 8.0E+0 MLLT1, SH3GL1, TCF3, VAV1 p13.3–2
20 409 0.8% 20.2% 2.3 (1.2–4.2) 0.44 (0.24–0.81) 0.009 1.0E−1 q11.21

*, hazard ratio for a 2-fold lower copy number; **, hazard ratio for a 2-fold higher copy number.

Table S5. Prognostic effect of the copy number of genomic regions. Multivariate results obtained via penalized regression.

Chr Region ID CNA frequency Disease-free survival Overall survival Lung-cancer specific survival Mb Genes cytoBands
Losses Gains HR for loss* (95% CI) HR for gain** (95% CI) P HR for loss* (95% CI) HR for gain** (95% CI) P HR for loss* (95% CI) HR for gain** (95% CI) P
1 3 9.8% 1.6% 0.92 (0.83–1.0) 1.1 (0.98–1.2) 0.129 6.0E+1 EPS15, FGR, JUN, LCK, PAX7, STIL, TAL1, NBL1, EPHB2, MUTYH, NBL1, ARNT p36.21–p31.1
1 14 15.6% 25.9% 1.0 (0.92–1.1) 0.99 (0.90–1.1) 0.837 2.0E−1 q21.2
3 71 2.3% 42.6% 1.0 (0.90–1.1) 0.99 (0.88–1.1) 0.919 3.0E−2 q26.1
6 122 1.5% 40.3% 0.97 (0.87–1.1) 1.0 (0.93–1.1) 0.554 4.0E−3 p24.2
7 142 0.7% 17.3% 0.97 (0.87–1.1) 1.0 (0.93–1.1) 0.531 8.0E+0 p12.3–p11.2
8 166 23.5% 7.4% 0.98 (0.89–1.1) 1.0 (0.91–1.1) 0.771 1.0E+0 p12
8 185 7.3% 17.4% 1.0 (0.94–1.1) 0.96 (0.87–1.1) 0.460 4.0E+0 p11.1–q11.1
9 211 34.8% 3.4% 1.0 (0.91–1.2) 0.98 (0.86–1.1) 0.686 1.1 (0.92–1.2) 0.94 (0.81–1.1) 0.427 3.0E−1 p21.3
9 214 40.2% 3.0% 1.0 (0.88–1.1) 1.0 (0.88–1.1) 0.984 1.0 (0.89–1.2) 0.98 (0.85–1.1) 0.724 3.0E−1 CDKN2A, CDKN2B p21.3
9 217 35.0% 3.9% 1.0 (0.90–1.2) 0.98 (0.85–1.1) 0.717 9.0E−3 p21.3
12 270 3.9% 16.4% 0.98 (0.88–1.1) 1.0 (0.92–1.1) 0.705 9.0E+0 FGF6, ING4 p13.33–31
14 312 8.5% 8.9% 1.0 (0.92–1.1) 0.99 (0.90–1.1) 0.844 1.0E−1 q23.1
18 368 12.1% 6.6% 1.1 (0.95–1.2) 0.95 (0.86–1.1) 0.321 1.0E−3 q12.1
19 378 9.2% 0.3% 1.1 (0.92–1.2) 0.95 (0.84–1.1) 0.443 8.0E+0 MLLT1, SH3GL1, TCF3, VAV1 p13.3–2
19 379 10.3% 0.9% 1.0 (0.90–1.1) 0.99 (0.87–1.1) 0.821 3.0E−3 p13.2
19 383 8.0% 2.5% 1.0 (0.91–1.1) 0.99 (0.88–1.1) 0.802 4.0E+0 p12–p11
20 410 1.1% 20.8% 1.0 (0.9–1.1) 0.99 (0.89–1.1) 0.895 2.0E+0 HCK q11.21–22

Results from a model adjuster by treatment arm, patient age, sex, performance status (PS), histology, T, and N stage. *, hazard ratio for a 2-fold lower copy number; **, hazard ratio for a 2-fold higher copy number.

The median follow-up for OS (451 events) was 5.3 years. The above-mentioned CN losses in 9p21, 18q12, 19p13 were also prognostic of shorter OS (P≤0.005, Q≤0.092; Table 2), together with 5 additional regions in 9p21.1, 18q12.1, and 19p12–13 (ELL). One further focal region on 14q23.1 (8.5% of losses, 89% of gains) was prognostic for OS (P=0.002, Q=0.079), with HRloss =2.2 (1.3–3.6), corresponding to HRgain =0.46 (0.28–0.76). The prognostic role of a region in 9p12.3 was confirmed in multivariate analyses (Table S4), together with the possible benefit for gains in 3q26 [MECOM, 45%, HRgain =0.55 (0.38–0.79), P=0.001]. Penalized regression (Table S5) did not select any region for OS.

The median follow-up for LCSS (427 events) was 5.0 years. Results were similar to DFS, with the addition of one region in chr8, for which gains (17%) were associated with longer LCSS [HRgain =0.51 (0.32–0.82), P=0.005, Q=0.13]. In multivariate analyses (Table S4), two of the three regions associated with DFS (chr3 and 9) were also associated with LCSS, in addition to regions in 6p24.2 [HRgain =1.3 (1.1–1.5), P=0.002], 8p23 [HRloss =0.53 (0.34–0.83), P=0.005], 19p13 [MLLT1, SH3GL1, TCF3, VAV1; HRloss =3.7 (1.7–7.7), P<0.001], and 20q11.21 [HRgain =0.44 (0.24–0.81), P=0.009]. Penalized regression (Table S5) selected 17 prognostic regions for LCSS on chr1 (EPS15, FGR, JUN, LCK, PAX7, STIL, TAL1, NBL1, EPHB2, MUTYH, NBL1, ARNT), chr9 (CDKN2A/B), chr12 (FGF6, ING4), chr19 (MLLT1, SH3GL1, TCF3, VAV1), and chr20 (HCK).

Copy-number aberrations associated with the effect of ACT

The average ACT effect on DFS estimated within the 976 patients with CN data was HRACT =0.85 (0.71–1.0) (P=0.06). Univariate analyses (Table 3) identified five regions in 14q32.33 as potentially predictive of better response to ACT (P<0.05), but with very high Q values. The effect of CNAs in these regions was similar. CN loss in one region in 14q32.33 had HRloss for interaction of 0.42 (0.22–0.83) (P=0.012, Q=0.010), corresponding to HRgain for interaction of 2.4 (1.2–4.6). This means that, given a treatment effect (ACT vs. control) of HR[ACT|CN=2] =0.85 for a patient with CN=2, such an effect is stronger for a patient with CN=1 (HR[ACT|CN=1] =0.42×0.85=0.36) and reversed with CN=4 (HR[ACT|CN=4] =2.4×0.85=2.0). The predictive role of this region was the only confirmed in multivariate analyses (Table S6), with HRloss for interaction of 0.39 (0.20–0.79) (P=0.009).

Table 3. Predictive effect of the copy number aberration (CNA) at various genomic regions for the magnitude of the effect of adjuvant chemotherapy. Univariate results.

Region ID Chr cytoBands Mb CNA Frequency Disease-free survival Overall survival Lung-cancer specific survival Genes
Losses Gains HR for loss* (95% CI) HR for gain** (95% CI) P Q HR for loss* (95% CI) HR for gain** (95% CI) P Q HR for loss* (95% CI) HR for gain** (95% CI) P Q
160 8 p23.2 6.0E−3 31.8% 2.5% 0.73 (0.57–0.95) 1.4 (1.1–1.8) 0.019 0.76
235 10 p15.3–p11.21 4.0E+1 5.9% 4.0% 0.27 (0.09–0.82) 3.6 (1.2–11) 0.021 0.76 BMI1, NET1, MAP3K8, BMI1, MLLT10, ZMYND11
237 10 p11.21–q11.21 7.0E+0 4.6% 4.5% 0.28 (0.09–0.83) 3.6 (1.2–11) 0.022 0.76
238 10 q11.21–22 4.0E+0 7.2% 4.6% 0.26 (0.08–0.8) 3.9 (1.3–12) 0.019 0.76 RET, RASSF4
318 14 q32.33 2.0E−2 8.9% 10.0% 0.40 (0.19–0.88) 2.5 (1.1–5.4) 0.022 >0.99 0.33 (0.14–0.78) 3.0 (1.3–7.1) 0.011 0.90
319 14 q32.33 1.0E−1 10.8% 11.4% 0.42 (0.22–0.83) 2.4 (1.2–4.6) 0.012 0.99 0.38 (0.18–0.79) 2.6 (1.3–5.4) 0.009 0.90
324 14 q32.33 5.0E−2 9.1% 9.0% 0.37 (0.15–0.87) 2.7 (1.2–6.5) 0.022 0.99
325 14 q32.33 3.0E−2 16.3% 8.3% 0.68 (0.51–0.92) 1.5 (1.1–2.0) 0.012 0.99 0.66 (0.48–0.91) 1.5 (1.1–2.1) 0.012 0.90
326 14 q32.33 4.0E−1 8.7% 9.2% 0.33 (0.14–0.77) 3.1 (1.3–7.2) 0.011 0.99
333 15 q26.1–3 9.0E+0 4.7% 4.5% 0.21 (0.06–0.71) 4.8 (1.4–16) 0.012 0.76
408 20 q11.21 5.0E−1 0.8% 20.8% 0.18 (0.06–0.52) 5.6 (1.9–16) 0.002 0.57
409 20 q11.21 1.0E−1 0.8% 20.2% 0.17 (0.05–0.54) 5.9 (1.9–19) 0.003 0.57

*, hazard ratio for a 2-fold lower copy number; **, hazard ratio for a 2-fold higher copy number.

Table S6. Predictive effect of the copy number of genomic regions. Multivariate results.

Chr Region ID CNA frequency Disease-free survival Overall survival Lung-cancer specific survival Mb cytoBands
Losses Gains HR for loss* (95% CI) HR for gain** (95% CI) P HR for loss* (95% CI) HR for gain** (95% CI) P HR for loss* (95% CI) HR for gain** (95% CI) P
8 159 30.4% 2.6% 0.42 (0.19–0.93) 2.4 (1.1–5.2) 0.032 2.0E+0 p23.3–2
14 319 10.8% 11.4% 0.39 (0.20–0.79) 2.5 (1.3–5.1) 0.009 0.35 (0.17–0.74) 2.8 (1.4–6.0) 0.006 0.37 (0.17–0.82) 2.7 (1.2–5.9) 0.015 1.0E−1 q32.33
20 409 0.8% 20.2% 0.11 (0.03–0.39) 8.8 (2.6–30) <0.001 1.0E−1 q11.21

Results from a model adjuster by treatment arm, patient age, sex, performance status (PS), histology, T, and N stage. The multivariate model has been obtained via stepwise selection (αin =0.10 and αout =0.01). Only regions with P<0.005 are shown. *, hazard ratio for a 2-fold lower copy number; **, hazard ratio for a 2-fold higher copy number.

The average effect of ACT on OS was HRACT=0.95 (0.79–1.1) (P=0.58). At a raw P<0.05, 5 regions were possibly associated to the ACT effect for OS, but with very high Q values (Table 3). One region in 8p23.2 showed a treatment effect enhanced for the 31.8% of patients with a CN loss [HRloss for interaction 0.73 (0.57–0.95), P=0.019, Q=0.76), meaning that the HR for a patient with CN=1 was HR[ACT|CN=1] =0.73×0.95=0.69. An adjacent region in 8p23 was selected in multivariate analyses (Table S6), with a similar effect [HRloss for interaction 0.42 (0.19–0.93), P=0.032). In univariate analyses, 3 regions in chr10 (BMI1, NET1, MAP3K8, BMI1, MLLT10, ZMYND11, RET, RASSF4) with 5–7% losses and 4–5% gains showed predictive effects with HRloss for interaction 0.26–0.28 and CNgain for interaction 3.6–3.9. One region in 15q26 (losses: 4.7%, gains: 4.5%) had HRloss for interaction 0.21 (0.06–0.71) (P=0.012, Q=0.76) corresponding to HRgain for interaction 4.8 (1.4–16). In multivariate analyses (Table S6) one region in 14q32.33 was predictive (P=0.006), with HRloss for interaction 0.35 (0.17–0.74) and HRgain for interaction 2.8 (1.4–6.0).

The average effect of ACT on LCSS was HRACT =0.83 (0.68–1.0) (P=0.05). Three of the above-mentioned regions in 14q32 predictive of ACT effect for DFS were also predictive for LCSS (Table 3). Two additional regions in 20q11.21 (gain frequency: 20%) had possibly significant interaction with ACT, with HRgain for interaction 5.6 (1.9–16) (P=0.002, Q=0.57) and 5.9 (1.9–19) (P=0.003, Q=0.57), respectively. Two of them (14q32 and 20q11) were confirmed in multivariate analyses (Table S6).

Penalized regression did not select any predictive region for either endpoint.

Sensitivity analyses

The results within the optimal quality sample subgroup (Tables S7-S10) were consistent with those of the whole population. Table S11 shows the genomic regions for which the prognostic effect was significantly different between ADC and SCC (interaction P<0.005). CN gains in two regions in 1q23–31 (FCGR2B, PBX1, TPR, LHX4, CDC73) were associated to shorter DFS in ADC [HR =2.8 (1.3–5.8) and 2.3 (1.1–4.7)] and longer DFS in SCC [HR =0.44 (0.18–1.1) and 0.53 (0.27–1.0)]. One of these regions showed similar results for LCSS. Similar results were observed for 3 regions in 7p11 (also for LCSS and including EGFR), one in 7q11, one in 11p14 (also for OS), and one in 20q11, with increased risk in ADC and reduced risk in SCC for CN gains. Of note, only one region (chr11p14) had quite low interaction Q-value and only for OS (Q=0.056). Conversely, CN gains in 3 further regions [1p13, 4p12–15 (PTTG2), 4q27] were associated to longer OS in ADC [HR =0.50 (0.19–1.3), 0.20 (0.07–0.57), and 0.51 (0.28–0.92), respectively] than in SCC [HR =2.4 (1.0–5.6), 2.1 (0.9–4.8), and 1.6 (0.97–2.6), respectively].

Table S7. Prognostic effect of the copy number of genomic regions. Univariate results in optimal quality samples only.

Chr Region ID CNA frequency Disease-free survival Overall survival Lung-cancer specific survival Mb Genes cytoBands
Losses Gains HR for loss* (95% CI) HR for gain** (95% CI) P Q HR for loss* (95% CI) HR for gain** (95% CI) P Q HR for loss* (95% CI) HR for gain** (95% CI) P Q
3 71 2.1% 34.0% 1.8 (1.2–2.6) 0.56 (0.39–0.81) 0.002 0.096 3.0E−2 q26.1
72 2.1% 34.7% 1.8 (1.2–2.6) 0.57 (0.39–0.83) 0.004 0.164 2.1 (1.4–3.1) 0.48 (0.32–0.73) <0.001 0.096 1.9 (1.2–2.8) 0.54 (0.35–0.81) 0.003 0.123 5.0E+0 MECOM q26.1–2
9 210 29.0% 3.6% 2.3 (1.4–3.7) 0.44 (0.27–0.72) 0.001 0.077 2.2 (1.3–3.8) 0.45 (0.27–0.76) 0.003 0.107 2.6 (1.5–4.3) 0.39 (0.23–0.66) <0.001 0.051 5.0E+0 MLLT3 p22.3–p21.3
211 34.8% 3.4% 1.9 (1.4–2.7) 0.52 (0.37–0.72) <0.001 0.053 2.0 (1.4–2.9) 0.50 (0.34–0.72) <0.001 0.075 2.1 (1.5–3) 0.48 (0.33–0.68) <0.001 0.016 3.0E−1 p21.3
212 35.9% 3.3% 1.6 (1.2–2.2) 0.62 (0.45–0.85) 0.003 0.147 1.7 (1.2–2.4) 0.57 (0.41–0.81) 0.001 0.096 1.7 (1.2–2.4) 0.59 (0.42–0.83) 0.002 0.100 6.0E−1 p21.3
213 38.7% 3.2% 1.5 (1.1–2) 0.67 (0.5–0.88) 0.004 0.164 1.6 (1.2–2.1) 0.64 (0.47–0.86) 0.003 0.123 6.0E−2 p21.3
214 40.2% 3.0% 1.5 (1.2–1.9) 0.66 (0.52–0.84) <0.001 0.077 1.5 (1.2–2.0) 0.65 (0.50–0.84) 0.001 0.066 3.0E−1 CDKN2A, CDKN2B p21.3
215 36.3% 3.3% 1.7 (1.2–2.2) 0.61 (0.45–0.82) 0.001 0.077 1.7 (1.2–2.4) 0.59 (0.42–0.81) 0.001 0.096 1.7 (1.3–2.4) 0.58 (0.42–0.80) <0.001 0.058 2.0E+0 p21.3
217 35.0% 3.9% 1.7 (1.3–2.4) 0.58 (0.42–0.79) <0.001 0.077 1.9 (1.4–2.7) 0.52 (0.38–0.73) <0.001 0.027 9.0E−3 p21.3
218 32.8% 4.5% 1.7 (1.2–2.3) 0.60 (0.43–0.83) 0.002 0.136 1.7 (1.2–2.4) 0.59 (0.41–0.85) 0.004 0.133 1.8 (1.3–2.6) 0.54 (0.38–0.77) <0.001 0.051 5.0E−1 p21.3
219 30.5% 4.6% 2.0 (1.3–3.0) 0.51 (0.34–0.76) 0.001 0.077 2.0 (1.3–3.1) 0.49 (0.32–0.77) 0.002 0.096 2.1 (1.4–3.3) 0.47 (0.3–0.73) <0.001 0.051 2.0E+0 p21.3–2
220 30.1% 4.8% 1.6 (1.2–2.2) 0.63 (0.46–0.86) 0.004 0.133 2.0E−2 p21.2
222 27.8% 5.2% 2.1 (1.3–3.3) 0.48 (0.30–0.77) 0.002 0.096 2.0E+0 p21.1
223 26.0% 6.4% 1.9 (1.2–2.9) 0.53 (0.35–0.82) 0.004 0.164 2.1 (1.3–3.3) 0.48 (0.3–0.77) 0.002 0.096 7.0E−1 p21.1
19 383 8.0% 2.5% 2.5 (1.3–4.6) 0.41 (0.22–0.76) 0.005 0.164 2.8 (1.4–5.3) 0.36 (0.19–0.7) 0.003 0.103 4.0E+0 p12–p11
386 4.6% 7.3% 2.2 (1.3–3.9) 0.45 (0.26–0.79) 0.005 0.161 3.0E−1 q11

Results from a model adjuster by treatment arm, patient age, sex, performance status (PS), histology, T, and N stage. *, hazard ratio for a 2-fold lower copy number; **, hazard ratio for a 2-fold higher copy number.

Table S8. Prognostic effect of the copy number of genomic regions. Multivariate results in optimal quality samples only.

Chr Region ID CNA frequency Disease-free survival Overall survival Lung-cancer specific survival Mb Genes cytoBands
Losses Gains HR for loss* (95% CI) HR for gain** (95% CI) P HR for loss* (95% CI) HR for gain** (95% CI) P Q HR for loss* (95% CI) HR for gain** (95% CI) P
3 72 1.8% 44.8% 2.4 (1.6–3.5) 0.42 (0.28–0.63) <0.001 2.8 (1.8–4.4) 0.35 (0.23–0.55) <0.001 2.5 (1.6–3.8) 0.40 (0.26–0.62) <0.001 5.0E+0 MECOM q26.1–2
9 211 34.8% 3.4% 2.4 (1.7–3.3) 0.43 (0.30–0.60) <0.001 2.8 (1.9–4.1) 0.36 (0.24–0.53) <0.001 3.0E−1 p21.3
12 277 2.4% 14.9% 0.47 (0.27–0.81) 2.1 (1.2–3.7) 0.007 3.0E+0 p11.21–1
17 354 2.7% 10.3% 0.45 (0.23–0.86) 2.2 (1.2–4.3) 0.016 0.41 (0.21–0.82) 2.4 (1.2–4.9) 0.012 2.0E−2 q21.32
19 396 4.6% 10.5% 0.64 (0.46–0.88) 1.6 (1.1–2.2) 0.006 2.0E−2 q13.32

Results from a model adjuster by treatment arm, patient age, sex, performance status (PS), histology, T, and N stage. *, hazard ratio for a 2-fold lower copy number; **, hazard ratio for a 2-fold higher copy number.

Table S9. Predictive effect of the copy number of genomic regions. Univariate results in optimal quality samples only.

Chr Region ID CNA frequency Disease-free survival Overall survival Lung-cancer specific survival Mb Genes cytoBands
Losses Gains HR for loss* (95% CI) HR for gain** (95% CI) P Q HR for loss* (95% CI) HR for gain** (95% CI) P Q HR for loss* (95% CI) HR for gain** (95% CI) P Q
5 102 0.6% 45.3% 0.45 (0.22–0.88) 2.2 (1.1–4.4) 0.021 0.705 5.0E−2 p15.33
9 232 17.2% 2.4% 4.0 (1.2–14) 0.25 (0.07–0.85) 0.026 0.979 2.0E+1 q21.11–q22.1
233 15.6% 3.2% 4.1 (1.2–14) 0.25 (0.07–0.84) 0.025 0.979 5.0E+0 FAM120A q22.1–31
10 238 7.2% 4.6% 0.22 (0.06–0.82) 4.6 (1.2–17) 0.024 0.705 4.0E+0 RET, RASSF4 q11.21–22
14 318 8.9% 10.0% 0.29 (0.11–0.75) 3.4 (1.3–8.8) 0.010 0.979 0.22 (0.08–0.62) 4.5 (1.6–13) 0.004 0.614 2.0E−2 q32.33
319 10.8% 11.4% 0.37 (0.17–0.81) 2.7 (1.2–6.0) 0.013 0.979 1.0E−1 q32.33
325 16.3% 8.3% 0.66 (0.45–0.95) 1.5 (1.0–2.2) 0.028 0.979 3.0E−2 q32.33
299 13.5% 9.4% 0.33 (0.13–0.83) 3.0 (1.2–7.6) 0.019 0.705 5.0E−1 q11.2
302 6.4% 9.5% 0.29 (0.10–0.84) 3.4 (1.2–9.8) 0.022 0.705 4.0E+0 q11.2–q12
17 360 2.1% 14.2% 0.23 (0.07–0.82) 4.3 (1.2–15) 0.023 0.705 8.0E−2 q25.3
20 408 0.8% 20.8% 0.19 (0.06–0.63) 5.3 (1.6–18) 0.007 0.614 5.0E−1 q11.21
409 0.8% 20.2% 0.19 (0.06–0.65) 5.3 (1.5–18) 0.008 0.614 1.0E−1 q11.21
411 3.0% 17.4% 0.18 (0.05–0.60) 5.7 (1.7–19) 0.006 0.614 7.0E+0 SRC, MAFB, RBL1, MAFB q11.22–q12
412 3.1% 17.5% 0.21 (0.07–0.68) 4.7 (1.5–15) 0.009 0.614 3.0E−2 q12

Results from a model adjuster by treatment arm, patient age, sex, performance status (PS), histology, T, and N stage. *, hazard ratio for a 2-fold lower copy number; **, hazard ratio for a 2-fold higher copy number.

Table S10. Predictive effect of the copy number of genomic regions. Multivariate results in optimal quality samples only.

Chr Region ID CNA frequency Disease-free survival Overall survival Lung-cancer specific survival Mb cytoBands
Losses Gains HR for loss* (95% CI) HR for gain** (95% CI) P HR for loss* (95% CI) HR for gain** (95% CI) P HR for loss* (95% CI) HR for gain** (95% CI) P
14 319 10.8% 11.4% 0.25 (0.10–0.62) 4.0 (1.6–10) 0.003 1.0E−1 q32.33
18 368 12.1% 6.6% 2.6 (1.1–6.1) 0.39 (0.16–0.91) 0.029 1.0E−3 q12.1
20 407 1.3% 18.4% 0.19 (0.05– 0.77) 5.2 (1.3–21) 0.020 5.0E−2 q11.21

Results from a model adjuster by treatment arm, patient age, sex, performance status (PS), histology, T, and N stage. *, hazard ratio for a 2-fold lower copy number; **, hazard ratio for a 2-fold higher copy number.

Table S11. Genomic regions with differential prognostic effect according to the histologic subtype.

Chr Region ID Disease free survival Overall survival Lung cancer specific survival Mb Genes cytoBands
HR for ADC (95% CI) HR for SCC (95% CI) P inter Q inter HR for ADC (95% CI) HR for SCC (95% CI) P inter Q inter HR for ADC (95% CI) HR for SCC (95% CI) P inter Q inter
1 7 0.50 (0.19–1.3) 2.4 (1.0–5.6) 0.003 0.252 5.0E−1 p13.3
16 1.6 (0.93–2.9) 0.68 (0.47–0.99) 0.004 0.213 2.0E−2 q23.3
18 2.8 (1.3–5.8) 0.44 (0.18–1.1) 0.003 0.169 3.7 (1.7–8.1) 0.55 (0.20–1.5) 0.005 0.213 4.0E+1 FCGR2B, PBX1, TPR, LHX4, CDC73 q23.3–q31.3
20 2.3 (1.1–4.7) 0.53 (0.27–1.0) 0.004 0.169 6.0E−2 q31.3
4 84 0.20 (0.07–0.57) 2.1 (0.9–4.8) 0.001 0.184 1.0E+1 PTTG2 p15.1–p12
96 0.51 (0.28–0.92) 1.6 (0.97–2.6) 0.005 0.252 5.0E−3 q27
7 144 1.6 (1.0–2.4) 0.63 (0.40–0.97) 0.002 0.169 1.6 (1.0–2.5) 0.55 (0.33–0.94) 0.002 0.213 5.0E−1 EGFR p11.2
145 2.3 (1.3–4.0) 0.63 (0.33–1.2) 0.001 0.169 2.5 (1.4–4.6) 0.55 (0.26–1.1) 0.001 0.212 2.0E+0 p11.2
146 2.6 (1.4–4.9) 0.59 (0.28–1.2) <0.001 0.169 3.1 (1.6–5.9) 0.56 (0.24–1.3) 0.001 0.212 1.0E−2 p11.1
147 2.5 (1.3–4.9) 0.62 (0.29–1.4) 0.004 0.169 8.0E−1 q11.1–21
11 251 1.3 (1.0–1.6) 0.78 (0.62–0.98) 0.002 0.169 1.4 (1.1–1.7) 0.72 (0.56–0.93) <0.001 0.056 1.0E−2 p14.1
20 407 1.8 (0.79–4.3) 0.33 (0.15–0.74) 0.005 0.169 5.0E−2 q11.21

Results from a model adjuster by treatment arm, patient age, sex, performance status (PS), histology, T, and N stage. All the hazard ratios (HR) are for a 2-fold higher copy number.

Chromosomal instability

The number of BPs was heterogeneous across trials, higher for men and possibly for high performance status (Table S12). Patients with a very high number of BPs (≥314) had shorter DFS than patients with very few (≤109) [HR =1.5 (1.1–2.0), P=0.015). This result was weaker for OS [HR =1.2 (0.90–1.7), P=0.19), but stronger for LCSS [HR =1.7 (1.2–2.3), P=0.003). Flexible models (Figure S4) in all patients showed that the BP effect can be considered log-linear. Such an effect was HR =1.1 (0.99–1.2, P=0.084, Table 4) on DFS for a patient as compared to another having a two times fewer BPs; this log-linear effect was similar LCSS [HR =1.1 (1.0–1.3), P=0.036) and statistically not significant on OS [HR =1.0 (0.93–1.2), P=0.51). The treatment effect was independent of the number of BPs both when comparing extreme groups (HR range: 0.96 to 1.1, P range: 0.78 to 0.93) and in terms of log-linear effects (HR: 0.93 to 0.99, P: 0.53 to 0.93).

Table S12. Chromosomal instability.

Factor nBP ratio LCI UCI uP value mP value
Trial
   CALGB 1.0 0.001 <0.001
   IALT 1.0 0.89 1.2
   JBR 10 1.3 1.1 1.5
Age
   ≤55 0.96 0.86 1.1 0.631 0.327
   55–64 1.0
   ≥65 1.0 0.93 1.2
Arm
   Control 1.00 0.512 0.999
   Chemotherapy 1.00 0.92 1.1
Sex
   Woman 1.0 0.013 0.009
   Men 1.1 0.99 1.2
PS
   0 1.0 0.002 0.047
   1–2 1.1 0.99 1.2
Surgery
   Lobectomy/other 1.0 0.016 0.163
   Pneumonectomy 1.0 0.94 1.2
Histology
   Adenocarcinoma 1.0 <0.001 0.194
   Squamous cell carcinoma 1.1 0.99 1.2
   Other 1.1 0.93 1.2
T stage
   T1 1.1 0.93 1.2 0.117 0.470
   T2 1.0
   T3/T4 1.1 0.92 1.2
N stage
   N0 0.99 0.89 1.1 0.050 0.775
   N1 1.0
   N2 1.0 0.90 1.2

nBP ratio, the ratio between the expected number of breakpoints (BPs) as compared to the reference class; LCI and UCI, lower and upper bounds of the 95% confidence interval; uP value, P value in the univariate analyses (Kruskal-Wallis test); mP value, P value in the multivariate analysis (likelihood ratio test).

Figure S4.

Figure S4

Flexible model (splines) to account for the possibly non-linear effect of the number of breakpoints (BPs) on the patient outcomes in all patients (prognostic effects, left) and within each arm (predictive effect, right). The two vertical lines are the tertiles of the number of BPs.

Table 4. Association between chromosomal instability and patient outcomes.

Variables HR (95% CI) P value
Prognostic effect of the number of BPs
   DFS
      Comparison between extreme classes* 1.5 (1.1–2.0) 0.015
      Comparison on all the sample range** 1.1 (0.99–1.2) 0.084
   OS
        Comparison between extreme classes* 1.2 (0.90–1.7) 0.19
        Comparison on all the sample range** 1.0 (0.93–1.2) 0.51
   LCSS
      Comparison between extreme classes* 1.7 (1.2–2.3) 0.003
      Comparison on all the sample range** 1.1 (1.0–1.3) 0.036
Predictive effect of the number of BPs
   DFS
      Comparison between extreme classes* 0.96 (0.54–1.7) 0.91
      Comparison on all the sample range** 0.95 (0.54–1.7) 0.62
   OS
      Comparison between extreme classes* 0.97 (0.52–1.8) 0.93
      Comparison on all the sample range** 0.93 (0.76–1.2) 0.53
   LCSS
      Comparison between extreme classes* 1.1 (0.57–2.1) 0.77
      Comparison on all the sample range** 0.99 (0.80–1.2) 0.93

*, HR between the high-number-of-breakpoints group (≥314, N=200) and the low-number-of-breakpoints group (≤109, N=197); **, Log2-linear effect, i.e., the HR is the ratio of the risk of a patient with a given number of breakpoints (BPs) as compared to a patient with a 2-fold lower number of BPs. DFS, disease-free survival; OS, overall survival; LCSS, lung-cancer specific survival; HR, hazard ratio.

Clonality

Patients with 2+ clones (N=518) had shorter DFS and LCSS [HR =1.2 (1.0–1.4 and 1.0–1.3), Table S13] than patients with 0–1 clones (N=456). This result was statistically non-significant (P=0.054 and 0.051, respectively) notably for OS [HR =1.1 (0.88–1.3), P=0.48]. The treatment effect was not associated to clonality [P=0.63 (DFS), 0.47 (OS), 0.52 (LCSS)].

Table S13. Association between clonality and patient outcomes.

Variables HR LCI UCI P value
Prognostic effect of the number of BPs
   Disease-free survival (DFS)
      Stratified 1.2 0.99 1.4 0.063
      Stratified + adjusted 1.2 1.0 1.4 0.054
   Overall survival (OS)
      Stratified 1.1 0.90 1.3 0.38
      Stratified + adjusted 1.1 0.88 1.3 0.48
   Lung-cancer specific survival (LCSS)
      Stratified 1.2 0.99 1.45 0.068
      Stratified + adjusted 1.2 1.0 1.5 0.051
Predictive effect of the number of BPs
   Disease-free survival (DFS)
      Stratified 1.2 0.81 1.6 0.42
      Stratified + adjusted 1.1 0.76 1.6 0.63
   Overall survival (OS)
      Stratified 1.2 0.82 1.7 0.35
      Stratified + adjusted 1.2 0.79 1.7 0.47
   Lung-cancer specific survival (LCSS)
      Stratified 1.3 0.86 1.9 0.25
      Stratified + adjusted 1.1 0.77 1.7 0.52

HR, hazard ratio between the patients with 2 or more clones (N=518) and patients with 0 or 1 clones (N=456); LCI and UCI: lower and upper bounds of the 95% confidence interval; stratified, model stratified on the trial; adjusted, model adjusted on clinicopathological factors.

Discussion

Increased understanding of the genomic changes of NSCLC facilitates the identification of prognostic and predictive biomarkers and provides vital information for personalized therapy, potentially allowing tailored treatments for individual patients. We utilized NSCLC FFPE samples from the LACE-Bio project to profile DNA CNAs. The most frequent CN gains were found on 1p13, 1q21, 3q22–26, 5p13–15, 6p24, and 22q11, the most frequent losses on 3p21.31, 8p23, and 9p21.3. The more focal and less frequent losses might be due to harder identification of losses in tumors with stromal cell contamination. Telomerase reverse transcriptase (TERT), among the most frequently amplified genes, is the catalytic subunit of the enzyme telomerase; its overexpression has been associated with poor prognosis (31). Among the loss genes, cyclin-dependent kinase Inhibitor 2A (CDKN2A), reported to be deleted in many tumors including lung cancer (32), codes for two proteins, p16 (or p16INK4a) and p14arf, which act as tumor suppressors by regulating the cell cycle.

The different spectrum of CNAs between ADC and SCC has been reported previously (33,34). Genes such as PIK3CA (33) and PDGFB (35) were amplified in lung SCC. Cyclin L (CCNL1) has been identified as oncogene in head and neck cancer (36). Mutations in CHEK2 (37) and NF2 (38) have been reported to be associated with SCC. CN loss and promoter hypermethylation of RASSF1 was reported in SCCHN (39) and in early stage NSCLC (40). NKX2-1 amplification was significantly less frequent than in ADC (33).

Our analyses confirmed some of the prognostic genes reported in the literature, such as shorter survival with CN loss of CDKN2A/B (32). In the present study, CDKN2A/B CN loss occurred in 40% of the cases and was significantly associated with shorter DFS. CDKN2A/B CN loss was also prognostic in ADC. Copy number loss of the tumor suppressor STK11 (or LKB1) has been associated with increased risk of brain metastasis (41). We were not able to confirm this due to incomplete reporting of metastatic sites. NSCLC patients with STK11 exon 1 or 2 mutations have shorter survival (42). A recent meta-analysis (14 studies, 1915 patients with solid tumors) revealed that decreased expression of STK11 was a prognostic factor [HR =2.2 (1.5–3.2), P<0.001] (43). In the present study, STK11 CN loss was found in 11% of samples and was significantly associated with shorter DFS [HR =2.4 (1.3–4.3), P=0.005]. We also identified novel prognostic genes, such as FSTL3, which encodes a secreted glycoprotein, and transcriptional factors MLLT1, SH3GL1, and TCF3, and the guanine nucleotide exchange factor (GEF) gene VAV1. Its overexpression significantly increased the risk of death [HR =1.81 (1.39–2.36), P<0.001) (44). However, in the present study, the CN loss frequency of the region containing these genes was 9%. Additional studies on their prognostic value are warranted.

The LACE-bio study has the unique possibility to identify biomarkers that predict efficacy of ACT in NSCLC by comparison to observation arms. Three regions had significant differences in multivariate analyses between the two study arms, but they came with high false discovery rate (Table S6). Particularly, 8p23.3–2 losses were significantly associated with increased ACT efficacy for OS. The frequent gains of 20q11.21 strongly were associated with no benefit from ACT for LCSS. This deleterious effect from ACT was in strong contrast with the small group (0.8%) of patients with 20q11.21 loss where ACT lead to a notably high survival benefit. The 20q11.21 region is rich in genes that might have a potential role in cancer such as HCK (tyrosine kinase), BCL2L1 (apoptotic regulator), MAPRE1 and TPX2 (microtubule associated factors), DNMT3B (epigenetic modifier) and transcriptional regulators. It is even more striking to find the p53 and DNA damage-regulated gene named PDRG1 in 20q11.21. PDRG1 is an oncogene in lung cancer cell lines, is selectively regulated by DNA damaging agents such as UV, and promotes radioresistance (45,46). Whatsoever, its exact role in mediating resistance to ACT in NSCLC remains to be confirmed. Finally, the 14q32.33 region also had differential HR (loss predictive of ACT efficacy, gain predictive of inefficacy), but the proportion of patients with losses and gains were equally high (10.8% and 11.4% respectively), making interpretation more difficult in the context of prediction of ACT efficacy.

In exploratory analyses in the LACE-Bio2 samples, the prognosis of patients with very high chromosomal instability was significantly worse than for patients with very low, independently of the clinical factors. Chromosomal instability could likely be associated with the risk of relapses rather than to death. We found no association with the magnitude of the ACT effect.

The LACE-Bio data and tissue bank provided a valuable source for studying the prognostic and predictive role of the CN of genomic regions in stage I–III NSCLC. These large-scale genome-wide analyses were consistent with previous results and provide new candidate prognostic markers. Furthermore, as the data come from randomized controlled trials, we propose new markers which could predict the effect of ACT.

Detailed bioinformatics and statistical methods

Bioinformatics pre-processing

The CGH data were normalized relative to an internal pool of 390 reference normal tissues, segmented using circular binary segmentation (CBS) (17,18), and minimal recurrent regions were identified via the CGHregions algorithm (19). We planned to discard regions with <20 CNAs. The proportion of probes on the X-chromosome with called allelic imbalance allowed inferring patient gender. The inferred gender was compared to the actual gender and inconsistent samples were discarded.

Endpoints

The primary endpoint was:

  • ❖ Disease-free survival (DFS), defined as the time from randomization to first recurrence (loco-regional or distant) or death from any cause.

Secondary endpoints were:

  • ❖ Overall survival (OS), defined as the time from randomization to death from any cause, and;

  • ❖ Lung-cancer specific survival (LCSS), defined as the time from randomization to death from lung cancer. Death without evidence of cancer relapse was treated as censoring for LCSS.

Statistical analyses

The called copy number (CN) of each region was correlated to survival endpoints via Cox models stratified by trial and adjusted for treatment arm, patient age, sex, performance status, histology, type of surgery, T, and N stage. The CN entered in the regression models as log2(CN). Thus, the estimated hazard ratio (HRgain) expresses the relative hazard for a 2-fold higher CN of a given region. Its reciprocal (HRloss = 1/HRgain) is the relative hazard for a 2-fold lower CN. To evaluate the predictive role of CNAs, a treatment-by-log2(CN) interaction was further added. In both the prognostic and the predictive models, the Cox model was stratified by trial and adjusted for treatment arm and clinical variables.

We performed both univariate (each region separately) and multivariate analyses (several regions jointly). The P values were corrected to control the false discovery rate [Q values (20)]. Multivariate models were built by stepwise selection (αin =0.10 and αout =0.01) and using a penalized regression approach (21,22) with lasso penalty for prognostic analyses and adaptive lasso for predictive analyses.

Preplanned sensitivity analyses were

The analyses were repeated, in addition to the entire study population, within the following subgroups:

  • ❖ Histological subtypes (ADC vs. SCC);

  • ❖ Optimal quality subgroup (MAPD ≤0.3).

The significance of the CN differences between histologic subtypes was assessed by t-tests; the P values were corrected via step- down multiple testing procedures (23,24). We compared the obtained results to those from TCGA (25,26). Known tumor suppressor genes and oncogenes were obtained from previously published results (27).

Chromosomal instability

The number of breakpoints (BPs) in the CN was used as measure of chromosomal instability. Its association with clinicopathological factors was first tested in univariate analyses (Kruskal-Wallis tests), then in a multivariate analysis using a log-linear quasi-Poisson model. Its association with outcomes and treatment effect was studied in Cox models comparing the 20% of patients with the highest number of BPs (≥314) to the 20% of patients with the lowest (≤109).

Software

The bioinformatics pre-processing and the statistical analyses were performed using R software v3.3, with the following packages: biospear, CGHbase, CGHcall, CGHregions, DNAcopy, glmnet, gplots, parallel, qvalue, scales, survival, TxDb.Hsapiens.UCSC.hg19.knownGene, XLConnect.

Acknowledgements

The authors would like to acknowledge Ni Liu (Princess Margaret Cancer Centre), Nicolas Lemaitre (Institut Albert Bonniot) and Shakeel Virk (Canadian Cancer Trials Group) for technical assistance. Grants from the US NCI R01 grant, Ligue Nationale Contre le Cancer (France), le Programme National d’Excellence Spécialisé cancer du poumon de l’Institut National du Cancer (INCa) (France), Canadian Cancer Society, the Gustave Roussy Foundation, the Princess Margaret Cancer Foundation and the European contract EU-FP7 Curelung.

Ethical Statement: The study was approved by institutional review boards (No. UHN 04-0333-T).

Footnotes

Conflicts of Interest: The authors have no conflicts of interest to declare.

References

  • 1.Howlader N, Noone A, Krapcho M, et al. SEER Cancer Statistics Review, 1975-2014. Bethesda, MD; 2017. Available online: https://seer.cancer.gov/csr/1975_2014/. Based on November 2016 SEER data submission, posted to the SEER web site, April 2017.
  • 2.Bass AJ, Watanabe H, Mermel CH, et al. SOX2 is an amplified lineage-survival oncogene in lung and esophageal squamous cell carcinomas. Nat Genet 2009;41:1238-42. 10.1038/ng.465 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Friboulet L, Olaussen KA, Pignon JP, et al. ERCC1 Isoform Expression and DNA Repair in Non–Small-Cell Lung Cancer. N Engl J Med 2013;368:1101-10. 10.1056/NEJMoa1214271 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Brambilla E, Le Teuff G, Marguet S, et al. Prognostic Effect of Tumor Lymphocytic Infiltration in Resectable Non-Small-Cell Lung Cancer. J Clin Oncol 2016;34:1223-30. 10.1200/JCO.2015.63.0970 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Graziano SL, Lacas B, Vollmer R, et al. Cross-validation analysis of the prognostic significance of mucin expression in patients with resected non-small cell lung cancer treated with adjuvant chemotherapy: Results from IALT, JBR.10 and ANITA. Lung Cancer 2013;82:149-55. 10.1016/j.lungcan.2013.06.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Reiman T, Lai R, Veillard AS, et al. Cross-validation study of class III beta-tubulin as a predictive marker for benefit from adjuvant chemotherapy in resected non-small-cell lung cancer: analysis of four randomized trials. Ann Oncol 2012;23:86-93. 10.1093/annonc/mdr033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Shepherd FA, Domerg C, Hainaut P, et al. Pooled Analysis of the Prognostic and Predictive Effects of KRAS Mutation Status and KRAS Mutation Subtype in Early-Stage Resected Non-Small-Cell Lung Cancer in Four Trials of Adjuvant Chemotherapy. J Clin Oncol 2013;31:2173-81. 10.1200/JCO.2012.48.1390 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Shepherd FA, Lacas B, Le Teuff G, et al. Pooled Analysis of the Prognostic and Predictive Effects of TP53 Comutation Status Combined With KRAS or EGFR Mutation in Early-Stage Resected Non–Small-Cell Lung Cancer in Four Trials of Adjuvant Chemotherapy. J Clin Oncol 2017;35:2018-27. 10.1200/JCO.2016.71.2893 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Tsao M-S, Marguet S, Le Teuff G, et al. Subtype Classification of Lung Adenocarcinoma Predicts Benefit From Adjuvant Chemotherapy in Patients Undergoing Complete Resection. J Clin Oncol 2015;33:3439-46. 10.1200/JCO.2014.58.8335 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Arriagada R, Bergman B, Dunant A, et al. Cisplatin-Based Adjuvant Chemotherapy in Patients with Completely Resected Non–Small-Cell Lung Cancer. N Engl J Med 2004;350:351-60. 10.1056/NEJMoa031644 [DOI] [PubMed] [Google Scholar]
  • 11.Arriagada R, Dunant A, Pignon JP, et al. Long-Term Results of the International Adjuvant Lung Cancer Trial Evaluating Adjuvant Cisplatin-Based Chemotherapy in Resected Lung Cancer. J Clin Oncol 2010;28:35-42. 10.1200/JCO.2009.23.2272 [DOI] [PubMed] [Google Scholar]
  • 12.Winton T, Livingston R, Johnson D, et al. Vinorelbine plus cisplatin vs. observation in resected non-small-cell lung cancer. N Engl J Med 2005;352:2589-97. 10.1056/NEJMoa043623 [DOI] [PubMed] [Google Scholar]
  • 13.Butts CA, Ding K, Seymour L, et al. Randomized phase III trial of vinorelbine plus cisplatin compared with observation in completely resected stage IB and II non-small-cell lung cancer: updated survival analysis of JBR-10. J Clin Oncol 2010;28:29-34. 10.1200/JCO.2009.24.0333 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Strauss GM, Herndon JE, Maddaus MA, et al. Adjuvant Paclitaxel Plus Carboplatin Compared With Observation in Stage IB Non-Small-Cell Lung Cancer: CALGB 9633 With the Cancer and Leukemia Group B, Radiation Therapy Oncology Group, and North Central Cancer Treatment Group Study Groups. J Clin Oncol 2008;26:5043-51. 10.1200/JCO.2008.16.4855 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Douillard JY, Rosell R, De Lena M, et al. Adjuvant vinorelbine plus cisplatin versus observation in patients with completely resected stage IB–IIIA non-small-cell lung cancer (Adjuvant Navelbine International Trialist Association [ANITA]): a randomised controlled trial. Lancet Oncol 2006;7:719-27. 10.1016/S1470-2045(06)70804-X [DOI] [PubMed] [Google Scholar]
  • 16.Foster JM, Oumie A, Togneri FS, et al. Cross-laboratory validation of the OncoScan® FFPE Assay, a multiplex tool for whole genome tumour profiling. BMC Med Genomics 2015;8:5. 10.1186/s12920-015-0079-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wang Y, Carlton VE, Karlin-Neumann G, et al. High quality copy number and genotype data from FFPE samples using Molecular Inversion Probe (MIP) microarrays. BMC Med Genomics 2009;2:8. 10.1186/1755-8794-2-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Xie T, d’Ario G, Lamb JR, et al. A Comprehensive Characterization of Genome-Wide Copy Number Aberrations in Colorectal Cancer Reveals Novel Oncogenes and Patterns of Alterations. Corvalan AH, ed. PLoS One 2012;7:e42001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Olshen AB, Venkatraman ES, Lucito R, et al. Circular binary segmentation for the analysis of array-based DNA copy number data. Biostatistics 2004;5:557-72. 10.1093/biostatistics/kxh008 [DOI] [PubMed] [Google Scholar]
  • 20.Venkatraman ES, Olshen AB. A faster circular binary segmentation algorithm for the analysis of array CGH data. Bioinformatics 2007;23:657-63. 10.1093/bioinformatics/btl646 [DOI] [PubMed] [Google Scholar]
  • 21.van de Wiel MA, Van Wieringen WN. CGHregions: Dimension reduction for array CGH data with minimal information loss. Cancer Inform 2007;3:55-63. 10.1177/117693510700300031 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sakai K, Ukita M, Schmidt J, et al. Clonal composition of human ovarian cancer based on copy number analysis reveals a reciprocal relation with oncogenic mutation status. Cancer Lett 2017;405:22-8. 10.1016/j.canlet.2017.07.013 [DOI] [PubMed] [Google Scholar]
  • 23.Ternès N, Rotolo F, Michiels S. Empirical extensions of the lasso penalty to reduce the false discovery rate in high-dimensional Cox regression models. Stat Med 2016;35:2561-73. 10.1002/sim.6927 [DOI] [PubMed] [Google Scholar]
  • 24.Ternès N, Rotolo F, Heinze G, et al. Identification of biomarker-by-treatment interactions in randomized clinical trials with survival outcomes and high-dimensional spaces. Biom J 2017;59:685-701. 10.1002/bimj.201500234 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Storey JD. A direct approach to false discovery rates. J R Stat Soc Ser B (Statistical Methodol) 2002;64:479-98. 10.1111/1467-9868.00346 [DOI] [Google Scholar]
  • 26.Westfall PH, Young SS. Resampling-Based Multiple Testing: Examples and Methods for P-Value Adjustment. New York, NY: John Wiley & Sons, 1993. [Google Scholar]
  • 27.Dudoit S, Shaffer JP, Boldrick JC. Multiple Hypothesis Testing in Microarray Experiments. Stat Sci 2003;18:71-103. 10.1214/ss/1056397487 [DOI] [Google Scholar]
  • 28.Cancer Genome Atlas Research Network Comprehensive genomic characterization of squamous cell lung cancers. Nature 2012;489:519-25. 10.1038/nature11404 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Cancer Genome Atlas Research Network Comprehensive molecular profiling of lung adenocarcinoma. Nature 2014;511:543-50. 10.1038/nature13385 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Weir BA, Woo MS, Getz G, et al. Characterizing the cancer genome in lung adenocarcinoma. Nature 2007;450:893-98. 10.1038/nature06358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Aviel-Ronen S, Coe BP, Lau SK, et al. Genomic markers for malignant progression in pulmonary adenocarcinoma with bronchioloalveolar features. Proc Natl Acad Sci 2008;105:10155-60. 10.1073/pnas.0709618105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Nobori T, Miura K, Wu DJ, et al. Deletions of the cyclin-dependent kinase-4 inhibitor gene in multiple human cancers. Nature 1994;368:753-6. 10.1038/368753a0 [DOI] [PubMed] [Google Scholar]
  • 33.Inoue Y, Matsuura S, Kurabe N, et al. Clinicopathological and Survival Analysis of Japanese Patients with Resected Non-Small-Cell Lung Cancer Harboring NKX2-1, SETDB1, MET, HER2, SOX2, FGFR1, or PIK3CA Gene Amplification. J Thorac Oncol 2015;10:1590-600. 10.1097/JTO.0000000000000685 [DOI] [PubMed] [Google Scholar]
  • 34.Lee E, Moon JW, Wang X, et al. Genomic Copy Number Signatures Uncovered a Genetically Distinct Group from Adenocarcinoma and Squamous Cell Carcinoma in Non-Small Cell Lung Cancer. Hum Pathol 2015;46:1111-20. 10.1016/j.humpath.2015.04.009 [DOI] [PubMed] [Google Scholar]
  • 35.Donnem T, Al-Shibli K, Al-Saad S, et al. Prognostic Impact of Fibroblast Growth Factor 2 in Non-small Cell Lung Cancer: Coexpression with VEGFR-3 and PDGF-B Predicts Poor Survival. J Thorac Oncol 2009;4:578-85. 10.1097/JTO.0b013e31819f2e38 [DOI] [PubMed] [Google Scholar]
  • 36.Redon R, Hussenet T, Bour G, et al. Amplicon mapping and transcriptional analysis pinpoint cyclin L as a candidate oncogene in head and neck cancer. Cancer Res 2002;62:6211-7. [PubMed] [Google Scholar]
  • 37.Wang Y, McKay JD, Rafnar T, et al. Rare variants of large effect in BRCA2 and CHEK2 affect risk of lung cancer. Nat Genet 2014;46:736-41. 10.1038/ng.3002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Yoo NJ, Park SW, Lee SH. Mutational analysis of tumour suppressor gene NF2 in common solid cancers and acute leukaemias. Pathology 2012;44:29-32. 10.1097/PAT.0b013e32834c3599 [DOI] [PubMed] [Google Scholar]
  • 39.Hogg RP, Honorio S, Martinez A, et al. Frequent 3p allele loss and epigenetic inactivation of the RASSF1A tumour suppressor gene from region 3p21.3 in head and neck squamous cell carcinoma. Eur J Cancer 2002;38:1585-92. 10.1016/S0959-8049(01)00422-1 [DOI] [PubMed] [Google Scholar]
  • 40.Buckingham L, Penfield Faber L, Kim A, et al. PTEN, RASSF1 and DAPK site-specific hypermethylation and outcome in surgically treated stage I and II nonsmall cell lung cancer patients. Int J Cancer 2010;126:1630-9. [DOI] [PubMed] [Google Scholar]
  • 41.Zhao N, Wilkerson MD, Shah U, et al. Alterations of LKB1 and KRAS and risk of brain metastasis: Comprehensive characterization by mutation analysis, copy number, and gene expression in non-small-cell lung carcinoma. Lung Cancer 2014;86:255-61. 10.1016/j.lungcan.2014.08.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Pécuchet N, Laurent-Puig P, Mansuet-Lupo A, et al. Different prognostic impact of STK11 mutations in non-squamous non-small-cell lung cancer. Oncotarget 2017;8:23831-40. 10.18632/oncotarget.6379 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Xiao J, Zou Y, Chen X, et al. The Prognostic Value of Decreased LKB1 in Solid Tumors: A Meta-Analysis. PLoS One 2016;11:e0152674. 10.1371/journal.pone.0152674 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Qi Y, Kong FM, Deng Q, et al. Clinical significance and prognostic value of Vav1 expression in Non-small cell lung cancer. Am J Cancer Res 2015;5:2491-7. [PMC free article] [PubMed] [Google Scholar]
  • 45.Jiang L, Luo X, Shi J, et al. PDRG1, a novel tumor marker for multiple malignancies that is selectively regulated by genotoxic stress. Cancer Biol Ther 2011;11:567-73. 10.4161/cbt.11.6.14412 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Tao Z, Chen S, Mao G, et al. The PDRG1 is an oncogene in lung cancer cells, promoting radioresistance via the ATM-P53 signaling pathway. Biomed Pharmacother 2016;83:1471-77. 10.1016/j.biopha.2016.08.034 [DOI] [PubMed] [Google Scholar]

Articles from Translational Lung Cancer Research are provided here courtesy of AME Publications

RESOURCES