You are on page 1of 14

A Genomics-Based Classification of Human Lung Tumors

The Clinical Lung Cancer Genome Project (CLCGP) and Network Genomic
Medicine (NGM),
Sci Transl Med 5, 209ra153 (2013);
DOI: 10.1126/scitranslmed.3006802

Editor's Summary

Lung Tumors Great and Small

The treatment of lung cancer and many other malignancies has traditionally been determined by the histology of
a tumor. Small cell, large cell, squamous cell, or epithelial cell −like (for adenocarcinoma) microscopic appearance
was known to correlate with patient outcome and determine the course of therapy. In recent years, scientists have
made increasing progress toward understanding specific mutations that lead to cancer and targeting them with
appropriate drugs. However, many questions still remain, and cancer diagnosis is still primarily based on histology.

Now, Seidel and coauthors present data from two cohorts totaling more than 6000 lung cancer patients,
demonstrating the correlation between lung tumor subtype and its predominant mutations, and the benefit of genetic

Downloaded from stm.sciencemag.org on October 30, 2013


testing and targeted therapy in these patients. With the addition of genetic classification, some tumors were found to
belong to different biological groups than initially suspected. In particular, the group of large cell lung cancers almost
disappeared and was reclassified into different subtypes, some even changing over to the small cell category. It
remains to be seen how this reclassification of large cell cancers and others will ultimately affect patient management
and prognostic accuracy in wider clinical use, but so far, the study showed a clear survival benefit in patients who
received therapy informed by genetic testing of their tumors.

A complete electronic version of this article and other services, including high-resolution figures,
can be found at:
http://stm.sciencemag.org/content/5/209/209ra153.full.html
Supplementary Material can be found in the online version of this article at:
http://stm.sciencemag.org/content/suppl/2013/10/28/5.209.209ra153.DC1.html
Related Resources for this article can be found online at:
http://stm.sciencemag.org/content/scitransmed/5/186/186ra66.full.html
http://stm.sciencemag.org/content/scitransmed/4/136/136ra70.full.html
http://stm.sciencemag.org/content/scitransmed/4/120/120ra17.full.html
http://stm.sciencemag.org/content/scitransmed/3/111/111ra121.full.html
Information about obtaining reprints of this article or about obtaining permission to reproduce this
article in whole or in part can be found at:
http://www.sciencemag.org/about/permissions.dtl

Science Translational Medicine (print ISSN 1946-6234; online ISSN 1946-6242) is published weekly, except the
last week in December, by the American Association for the Advancement of Science, 1200 New York Avenue
NW, Washington, DC 20005. Copyright 2013 by the American Association for the Advancement of Science; all
rights reserved. The title Science Translational Medicine is a registered trademark of AAAS.
RESEARCH ARTICLE

CANCER

A Genomics-Based Classification of
Human Lung Tumors
The Clinical Lung Cancer Genome Project (CLCGP) and Network Genomic Medicine (NGM)*†

We characterized genome alterations in 1255 clinically annotated lung tumors of all histological subgroups
to identify genetically defined and clinically relevant subtypes. More than 55% of all cases had at least one
oncogenic genome alteration potentially amenable to specific therapeutic intervention, including several
personalized treatment approaches that are already in clinical evaluation. Marked differences in the pattern
of genomic alterations existed between and within histological subtypes, thus challenging the original his-
tomorphological diagnosis. Immunohistochemical studies confirmed many of these reassigned subtypes.
The reassignment eliminated almost all cases of large cell carcinomas, some of which had therapeutically
relevant alterations. Prospective testing of our genomics-based diagnostic algorithm in 5145 lung cancer
patients enabled a genome-based diagnosis in 3863 (75%) patients, confirmed the feasibility of rational
reassignments of large cell lung cancer, and led to improvement in overall survival in patients with EGFR-
mutant or ALK-rearranged cancers. Thus, our findings provide support for broad implementation of genome-
based diagnosis of lung cancer.

INTRODUCTION RESULTS
Lung cancer is traditionally classified into non–small cell lung cancer
(NSCLC), small cell lung cancer (SCLC), and carcinoid (CA). NSCLC Cancer genome alterations in human lung tumors
is further divided into adenocarcinoma (AD), squamous cell carcinoma Recent studies have provided analyses of genome alterations in lung
(SQ), and large cell carcinoma (LC), which also includes tumors with cancer (20, 21, 24, 25). To establish a systematic relationship of such
neuroendocrine differentiation [large cell neuroendocrine carcinoma alterations across the different cancer subtypes, we collected a total of
(LCNEC)] (1). These categories have been enriched with detailed his- 1882 surgically resected, fresh-frozen human lung tumor specimens
tomorphological and immunohistochemical characteristics leading to with clinical annotation, yielding 1255 specimens suitable for genetic
the 2004 World Health Organization (WHO) classification. The most analysis (Table 1, fig. S1A, and table S1). Visual inspection of somatic
detailed subcategories exist for AD, mostly defined by growth patterns copy number alterations (SCNAs) in those tumors for which both
(2). This rather descriptive taxonomy has been complicated in the past single-nucleotide polymorphism (SNP) array and histology data were
decade because of the recognition of somatic genetic alterations oc- available (922 of 1032) revealed distinctive patterns in cases sorted
curring in some of these subtypes: EGFR mutations, KRAS mutations, according to their initial histological subtype (Fig. 1A): some SCNAs
and EML4-ALK fusions occur mainly in lung AD (3–7), whereas muta- were present in all subtypes (for example, gains affecting 5p), whereas
tions in DDR2, FGFR2, and NFE2L2 or amplifications of FGFR1 and other SCNAs were subtype-specific (for example, amplifications of 3q
SOX2 mainly affect SQ (8–11). The fact that some of these are asso- containing SOX2 in SQs) (8). In addition, the overall pattern of SCNAs
ciated with clinical response to molecularly targeted therapeutics differed across histological entities; for example, SCLCs and some LCs
(3–5, 12, 13) emphasizes the importance of adding genetic annota- exhibited a predominance of chromosome arm–level events, in contrast
tion to the current taxonomy. Systematic efforts to comprehensively to focal events in AD and SQ. Stromal admixture could mask the de-
characterize the cancer genome (14) constantly add genome altera- tection and amplitude of SCNAs (26), but only partly accounted for the
tions to the compendium of such potentially actionable alterations diversity of SCNA patterns between subtypes (Fig. 1A). LC, in contrast
(9, 15–21). Similarly, immunohistochemical analyses have chal- to other subtypes, did not exhibit a specific SCNA pattern. To identify
lenged some of the original histomorphological diagnoses, in partic- significant copy number alterations across lung tumor subtypes, we ap-
ular in the case of LC (22, 23). We therefore sought to assess cancer plied a rank sum–based method that is insensitive to tumor purity and
genome alterations linked to histomorphological and immuno- identified 8 regions of amplification and 12 regions of deletion (fig. S2
histochemical features of the disease as well as to patient outcome to and tables S2 and S3) (11). In cases with only one deletion, CDKN2A
identify genetically defined subtypes of lung tumors and optimize (located at 9p21) was affected in 38% (fig. S3) (11, 27).
them for biologically informed patient stratification for personalized Similarly, most mutations showed histological subtype specificity
therapeutic approaches. We then tested the clinical relevance of these (Fig. 1B). The most frequently mutated genes were TP53 (53.6%), KRAS
molecularly defined patient subgroups prospectively in a diagnostic (16.1%), STK11 (9.8%), EGFR (7.2%), KEAP1 (6.6%), and NFE2L2
outreach setting. (4.5%) (20, 21, 28). Seventeen genes were altered in at least two samples
(figs. S4 and S5 and tables S1, S4, and S5). The NFE2L2/KEAP1 axis was
one of the most frequently mutated oncogenic pathways in lung cancer.
*Lists of participants and their affiliations appear at the end of the paper.
†Corresponding authors. E-mail: roman.thomas@uni-koeln.de (R.T.); reinhard.buett- Furthermore, gene set enrichment analysis revealed significant changes
ner@uk-koeln.de (R.B.); juergen.wolf@uk-koeln.de (J.W.) in the expression of NFE2L2 and KEAP1 target genes in cases harboring

www.ScienceTranslationalMedicine.org 30 October 2013 Vol 5 Issue 209 209ra153 1


RESEARCH ARTICLE

Table 1. Clinical characteristics of lung cancer patients in the retrospective [Clinical Lung Cancer Genome Project (CLCGP)] and prospective (NGM)
data sets. UICC, Union Internationale Contre le Cancer.

Retrospective CLCGP Prospective NGM

Total n 1255 3863


Age at diagnosis n 1225 3019
Median (range) 65 (18–94) 67 (28–96)
Sex, n (%) Female 419 (33.5) 1397 (37.2)
Male 831 (66.5) 2354 (62.8)
Unknown 5 112
Histology (WHO 2004), n (%) Adenocarcinoma 537 (44.5) 2250 (62.7)
Carcinoid 71 (6) 3 (0.08)
Large cell carcinoma* 129 (10.7) 54 (1.5)
Small cell carcinoma 65 (5.4) 265 (7.4)
Squamous cell carcinoma 403 (33.4) 1018 (28.3)
Other/unknown 50 273
Stage (UICC), n (%) IA 305 (25.4) 105 (10.4)
IB 316 (26.3) 72 (7.1)
IIA 33 (2.8) 56 (5.5)
IIB 190 (15.8) 49 (4.8)
IIIA 195 (16.2) 131 (13.0)
IIIB 89 (7.4) 115 (11.4)
IV 74 (6.1) 479 (47.5)
Unknown 53 2856
Survival, median no. of months I 152 Not reached
II 66 49†
III 34 28
IV 18 9.5
Unknown 58 11.6
Smoking history, n (%) Current/former 896 (86) 102 (89)
Never 147 (14) 13 (11)
Unknown 212 3748

*Including LCNEC. †Median follow-up time for stage II patients was 16.3 months. Because of short median follow-up time for stage II patients, accuracy of median overall survival in this
stage was low (fig. S19).

such mutations (false discovery rate, q = 0.0008) (29). Either NFE2L2 or which was partly driven by the higher stage of these patients. Because
KEAP1 was mutated in 10.4% of AD (21) and 16.9% of SQ (20) in mu- most of these patients were diagnosed before EGFR inhibitors were
tually exclusive fashion. NFE2L2 mutations were mainly found in SQ broadly available, this observation is likely to reflect a general aggressive
(30–32) (Fig. 1B and fig. S4). In addition to known frequent driver mu- behavior of EGFR/TP53 double-mutant tumors (36). Similarly, patients
tations, we also found rare mutations that are possibly relevant for therapy. with concurrent mutations of TP53 and RB1 loss had a particularly poor
For example, we found an R248C mutation in the fibroblast growth factor prognosis (Fig. 1C), independent of stage and histology (Cox regression
(FGF)–binding domain of FGFR3 (33, 34) in 3 of 365 SQ (0.8%), all of analysis, P = 0.023).
which were negative for FGFR2 mutations (35). FGFR3-R248C was onco-
genic in vitro and associated with sensitivity to FGFR inhibition (fig. S6). Subtype-specific genome alterations
Overall, more than 55% of all malignant lung tumors harbored at least As a next step, we sought to determine which of the genomic alterations
one genetic alteration with features of a possibly tractable target (fig. S7). were significant in each subtype. Across all lung tumor cases, we identified
We observed clinically relevant associations related to histology amplified and deleted regions (fig. S2) that included known proto-
and stage (1). The most frequent genome alterations had no significant oncogenes or tumor suppressor genes, which could be assigned to specific
impact on survival in AD, LC, and SQ (Fig. 1C). TP53 mutations were histological subtypes. Significantly amplified chromosomal regions in AD
associated with inferior survival in EGFR-mutant patients (P = 0.028), were 5p, 7p (EGFR), 8q (MYC), 11q (CCND1), 12q (MDM2), 14q (NKX2-1),

www.ScienceTranslationalMedicine.org 30 October 2013 Vol 5 Issue 209 209ra153 2


RESEARCH ARTICLE

A B

NFE2L2

PIK3CA
ERBB2

FGFR3

FGFR2
KEAP1
STK11
EGFR

HRAS
NRAS
KRAS
DDR2
BRAF

CDK4

TP53
ALK*
2 4 6 8 10 12 14 16 18 20 22 Purity %
1 3 5 7 9 11 13 15 17 19 21
0 50 100
AD
CA
SCLC LC

3.4
2
Copy number 1.2
SQ

Median
1
frequency %
Gains
Frequency %

20 * *
Alteration

* * * *
10
0 10

5 100
1.

4.

4.
1.

1.

16
Losses
7.

9. .1
6.

53
9

9
8

1
2

8
6

.6
C Histology Genetics - all cases EGFR-mutant TP53-mutant
Probability of survival

1.0 1.0 1.0 1.0


TP53wt
0.8 0.8 0.8 0.8 RB1no loss
0.6 0.6 0.6 0.6
BRAF Stage
TP53mut
0.4 AD 0.4 EGFR 0.4 0.4
CA FGFR1 amp
KRAS
I/II III/IV
RB1loss
0.2 LC 0.2 PIK3CA
0.2 3 7 1 6 TP53 wt 0.2
SCLC Wild type 12 1 5 TP53 mut
0.0 SQ 0.0 NA 0.0 P = 0.028 P = 0.01 0.0 P = 0.003
0 10 20 30 40 50 60 0 10 20 30 40 50 6 0 0 10 20 30 40 50 60 0 10 20 30 40 50 60

Month
Fig. 1. A global view of the lung cancer genome. (A) Copy number chosen consistent with lung cancer subtypes. Total mutation frequencies
profiles of lung cancer specimens of the major histological subtypes per gene expressed as a percentage of all cases are shown as a bar graph
(n = 992) (red, increases; blue, decreases) are plotted along the genome at the bottom. Frequencies below 1% are marked with an asterisk. (C)
(horizontal axis: chromosomes as indicated, centromeres in red). Verti- Kaplan-Meier curves for overall survival are shown for the overall pop-
cal colored bars on the left indicate lung cancer subtypes. Bottom: The ulation per histological subtype (LC includes LCNEC), per genotype, for
frequencies ( y axis) of copy number gains (red; cutoff, 2.7) and losses EGFR-mutant cases according to their TP53 mutation status, and for
(blue; cutoff, 1.3) across all samples, calculated for adjoining 1-Mb frag- TP53-mutant cases according to their RB1 alteration status (from left
ments using segmented copy number data, are represented along the to right) (P values for survival were calculated using the log-rank test).
genome. Purity of tumor samples determined through SNP array–derived Numbers of cases with wild-type (wt) and mutant (mut) TP53 in early
copy number data (26) is shown on the right, with the median purity stages (I and II) and late stages (III and IV) are given for EGFR-mutant
calculated for each histological subgroup indicated in red. (B) Mutations cases (inset; P value was calculated using the Pearson c2 test). Color
and ALK rearrangements (ALK*) are depicted per sample per gene as code for histology: orange, AD; black, CA; green, LC (including LCNEC);
colored ellipses. Sample order was conserved from (A), and colors were red, SCLC; blue, SQ.

www.ScienceTranslationalMedicine.org 30 October 2013 Vol 5 Issue 209 209ra153 3


RESEARCH ARTICLE

A AD CA LC SCLC SQ
q-value amplifications
1 2e-07 1e-58 1 0.002 5e-26 1 2e-06 2e-46 1 0.007 2e-17 1 1e-09 1e-99

MYCL1 1
MYCN
2

FHIT 3
SOX2
4
5
6
EGFR 7
PTPRD FGFR1 8
CDKN2A MYC 9
10
CCND1 11
KRAS
RB1 MDM2
12
13
NKX2-1 14
15
16
ERBB2 17
18
CCNE1 19
20
21 22

1e-17 4e-21 0.02 5e-26 0.002 1 9e-12 2e-170.02 2e-17 0.007 1 1e-99 1e-09 1
q-value deletions

CDKN2A°
B C
FGFR1°
NFE2L2

PIK3CA

KE
NKX2-1

ST
STK11°

CCND1

CCNE1
MYCL1
ERBB2
ERBB2

KEAP1
EGFR°

KRAS°

SOX2°
MDM2

MYCN

EGFR

TP53*
K1

KRAS
BRAF

AP
FHIT*

RB1*

MYC
1
TP5
AD SOX2

CA
-1
NKX2
LC
S
7
EG
SCLC A FR
KR

EG AF
ND

BR
CDKN2A
FGFR1

FR
CC
SQ
q value 0.05 <1–10

Fig. 2. Genomic alterations in histological subgroups of lung cancer. and SOX2 were adapted by dividing values by two (circles)]. Significant mu-
(A) Significantly amplified (red) and deleted (blue) regions calculated using tations were determined using a binomial test with a background mutation
a rank sum–based algorithm (24) are plotted along the genome (y axis) for rate of 0.5%. P values were adjusted for multiple hypothesis testing using
the five major lung cancer subtypes [AD (n = 421), CA (n = 69), LC (n = 101), the Benjamini and Hochberg method across each histological subtype. q
SCLC (n = 63), and SQ (n = 338)]. Statistical significance, expressed by q values of significant results (q < 0.05) are indicated by the color code of the
values (x axes: amplification, upper scale; deletion, lower scale), was com- symbols (color key provided below the chart). (C) Associations of copy
puted for each genomic location. Known or potential oncogenes (red) or number alterations and mutations calculated using Fisher’s exact test
tumor suppressor genes (blue) are given at respective locations. Vertical followed by Benjamini and Hochberg adjustment are represented with a
lines indicate level of significance of q = 0.01. (B) Frequencies of significant Circos plot. Involved genes are named at corresponding genomic locations
genomic alterations are given per gene per histological subtype. Colors of (copy number gains in red, copy number deletions in blue, and mutations
gene names are encoded as follows: red, amplified; blue, deleted; and in black) outside the ring representing the genome. Internal lines show
black, mutated. Frequencies of alterations correspond to circle size [fre- significant co-occurring (red) and mutually exclusive (blue) events (q < 0.05)
quencies of deletions of FHIT and RB1 and mutations in TP53 were adapted between two copy number alterations or two frequently mutated genes
by dividing values by three (asterisks); frequencies of mutations in EGFR, (solid lines) or between a copy number alteration and a mutation (dashed
KRAS, and STK11, of deletions in CDKN2A, and of amplifications in FGFR1 lines) found in lung cancer.

and 17q (ERBB2) (21, 37); in SCLC were 1p (MYCL1), 2p (MYCN), 5p, 8p on genomics alone. We integrated data on mutations and SCNAs that
(FGFR1), and 19q (CCNE1) (24); and in SQ were 1p (MYCL1), 3q (SOX2), were significant above the assumed background mutation rate of 0.005
7p (EGFR), 8p (FGFR1), and 11q (CCND1) (8, 11, 20). CA harbored (binomial test) to identify significant signature alterations for each his-
no significant SCNA. As before, in LC, we observed amplifications tological subtype (Fig. 2B). Some of these alterations mainly occurred
typical of other histologies (for example, amplification of NKX2-1, in a certain subtype, such as alterations in ALK (3.4%), BRAF (2.7%),
which is specific for AD, or of SOX2, which is typical of SQ). Dele- EGFR (15.3%), ERBB2 (1.7%), KRAS (32.6%), and STK11 (17.4%) in
tions in 9p (CDKN2A) were present in all subtypes except SCLC, AD; MYCN amplifications (6.5%) in SCLC; and mutations in DDR2
which had deletions of 3p (FHIT) and 13q (RB1) as its hallmarks (1.1%), FGFR3 (0.8%), and NFE2L2 (10.6%) in SQ (Figs. 1B and 2B and
(Fig. 2A). fig. S4). Others were not only enriched in a given subtype (for example,
The specific co-occurrence of several of the alterations in individual NKX2-1 amplification in AD, MYCL1 amplification and RB1 deletion in
lung cancer subtypes suggested that it might be possible to determine SCLC, or SOX2 and FGFR1 amplification in SQ) but also present in other
patterns of alterations that could be used to identify subtypes based histologies (Fig. 2B). By contrast, LC harbored alterations typical of all

www.ScienceTranslationalMedicine.org 30 October 2013 Vol 5 Issue 209 209ra153 4


RESEARCH ARTICLE

other subtypes (amplification of ERBB2 and NKX2-1 and mutations in Finally, LCNEC shared the significantly mutated genes TP53, RB1,
KRAS and STK11 as in AD; amplification of MYCL1 and RB1 as in SCLC; and EP300 with SCLC, as determined by whole-exome sequencing of
amplification of CCND1, FGFR1, and SOX2 as in SQ) (Fig. 2B) but had 15 and transcriptome sequencing of 10 pathologically reviewed LCNECs
no significant signature alterations. (Fig. 3C and tables S8 and S9). We also found additional mutated genes
The availability of large genomic data sets enabled us to conduct a in LCNEC that typically occurred in AD or SQ but did not reach sig-
systematic analysis of co-occurrence and exclusivity of genome altera- nificance in LCNEC (tables S8 and S9). Thus, LCNEC is most similar to
tions (Fig. 2C and fig. S8). In 5 of 21 lung tumors with BRAF mutations SCLC, with individual cases bearing mutations of other subtypes. The
affecting residues other than V600E, either NRAS or KRAS was mutated genetic similarity between LCNEC and SCLC is also reflected by a simi-
(38); such cases might be particularly sensitive to mitogen-activated pro- lar overall survival (Fig. 3D). In summary, LC exhibits a general diag-
tein kinase kinase (MEK) inhibition (39). Furthermore, ERBB2 muta- nostic plasticity when considering data on chromosomal copy number,
tions never co-occurred with mutations in BRAF, HRAS, KRAS, NRAS, or gene mutations, gene expression, and immunohistochemistry. Com-
STK11 (table S1). FGFR2 mutations did not co-occur with FGFR1 ampli- bined immunohistochemical and genomic analysis is therefore ideal
fications, in support of both alterations being oncogenic drivers (fig. S6). In to classify this heterogeneous group as AD, SQ, or NEC.
our data set, EGFR amplifications correlated with EGFR mutations in
AD (P = 0.0009), but not in SQ (fig. S9). EGFR amplification predicted Automated genomics-based lung tumor classification
treatment response and outcome of patients receiving EGFR inhibitors Given the strong correlation of specific genome alterations with certain
in some studies (40, 41), although this could not be confirmed in a study histological subtypes, we devised a statistical model to test if subtypes
focusing exclusively on ADs (42). Thus, whether patients with EGFR- could be predicted robustly based on such alterations alone. The di-
amplified SQ may benefit from EGFR inhibition is currently unclear. agnosis predicted by our model and the original diagnosis (Fig. 4A, left)
Similarly, loss of PTEN may influence the dependency of tumors on or the diagnosis obtained from pathology review were highly similar
mutant receptor tyrosine kinases (43, 44). PTEN was homozygously or for AD and SQ (Fig. 4A, right, fig. S15, and table S10). Only a few of the
hemizygously deleted in 5.5 and 11.1% of EGFR-mutant ADs, respec- AD or SQ cases were reclassified according to the predominant genome
tively (fig. S10). In summary, genome alterations that define specific lung alterations in these cases (fig. S15). Feature selection and automated
tumor subgroups were determined in the major histological subgroups, reclassification using a similar model was highly stable when applied
except for LC, which has genomic features of all other subtypes. to validation data sets of 382 AD (21), SQ (20), and SCLC (25) cases
(Fig. 4B, fig. S16, and table S11). Review of individual discrepant cases
The heterogeneity of large cell lung cancer revealed subtype-specific alterations, such as in one case that was pre-
Immunohistochemistry has become an indispensable method for dicted to be AD but classified by central pathology review as SQ, which
lung cancer diagnosis. We therefore performed an independent harbored the oncogenic S310F ERBB2 mutation (48); ERBB2 mutations
immunohistochemistry-based pathology review of 583 cases confirm- were typical of AD. Furthermore, one of three cases predicted to be SQ
ing all subclasses of lung cancer except LC (fig. S11). In 42% of all LC but classified as AD by pathology review exhibited amplification of
cases, pathology review led to reclassification to the other subtypes FGFR1 and CCND1, both predictive of SQ in our data set (fig. S16A).
sharing similar immunohistochemical and genetic features. Nevertheless, most initial diagnoses of AD, SCLC, and SQ were con-
To gain further insight into the heterogeneity of LC, we applied gene firmed by both our model and pathology review (Fig. 4, A and B, and
expression–based unsupervised hierarchical clustering to 261 lung fig. S11), but most of the LCs with at least one genomic alteration were
tumors, including 31 initially diagnosed LCs. Whereas 87% of ADs, reassigned to either AD, SQ, or aggressive neuroendocrine lung cancer/
92% of CA, 84% of SCLCs, and 76% of SQs formed distinct clusters, SCLC in accordance with the pathological review (Fig. 4C, fig. S15, and
LCs were dispersed across all other clusters (Fig. 3A and tables S1 and table S10). Even in those cases where immunohistochemistry did not
S6). Except for one case, all LCs clustered with those tumors of the other yield an unequivocal diagnosis, most remaining LC cases could be re-
subtypes that shared the same histology-defining signature alterations assigned genetically to the other lung cancer subtypes (Fig. 4C).
(Fig. 3, A and B, and table S7). Similar results were obtained when
applying consensus clustering, where almost 98% of AD cases formed Clinical evaluation of genomics-based
a distinct cluster, as well as 84% of SCLCs and 77% of SQs. Sample lung cancer diagnoses
overlap between clusters obtained by the two methods was 95%, 93%, To evaluate our combined genomic and immunohistochemical diagnostic
and 75% for the clusters, which mainly included tumors with neuro- approach, we enrolled 5145 lung cancer patients in a molecular screening
endocrine differentiation (NEC), AD, or SQ cases, respectively (table outreach program run by Network Genomic Medicine (NGM) in the re-
S1). Applying recently described transcriptional classifiers of sub- gion of our cancer center between January 2010 and April 2013 (Table 1,
types of AD (45) and SQ (46) revealed comparable results (figs. fig. S1B, and table S12). The addition of immunohistochemistry to the di-
S12 and S13 and table S1). An integrative analysis of 209 cases in- agnostic workup of tumors with LC features reduced the prevalence of this
cluding copy number and expression data using iCluster defined two subgroup from 5.9% in the retrospective analysis (table S1) to 1.3% in the
subgroups that were mainly driven by amplifications on chromosome 3 diagnostic NGM data set (Fig. 5A). The expression of TTF-1 and CK7 as
(3q13.31-3q29) and chromosome 12 (12p13.33-12q15) and thus did immunohistochemical markers of AD, as well as p63 and CK5 as markers
not reveal distinct clinically relevant subtypes (47). of SQ, allowed such cases to be assigned to either AD or SQ. Expression of
LCNEC exhibited substantial transcriptional similarity to SCLC neuroendocrine markers CD56, chromogranin A, and synaptophysin
when analyzed with unsupervised hierarchical clustering and classifica- was tested to identify tumors with neuroendocrine differentiation.
tion (Fig. 3A, gray triangles, and figs. S12 and S13). Furthermore, similar We performed central genotyping for key alterations identified
amplified and deleted regions were observed in LCNEC and SCLC within our retrospective genomic study (ALK, BRAF, DDR2, EGFR,
when compared with other histological subtypes (fig. S14). ERBB2, FGFR1, KRAS, and PIK3CA) (fig. S17). Genomic testing was fea-

www.ScienceTranslationalMedicine.org 30 October 2013 Vol 5 Issue 209 209ra153 5


RESEARCH ARTICLE

sible in 3863 (75%) paraffin-embedded tumor


A
samples obtained by routine diagnostic proce-
Cluster I II III IV dures, yielding 1481 genomic alterations (table
S12). Treatment recommendations were
LC provided to the network partners to enable ge-
RB1 netically tailored cancer therapy either with
NKX2-1 approved drugs or within clinical trials. Six-
BRAF/ERBB/KRAS ty-four of 84 advanced-stage (IIIB or IV) pa-
SOX2 tients with an EGFR mutation (76%) received
DDR2/NFE2L2
erlotinib or gefitinib, and 15 of 30 advanced-
B stage patients with ALK translocation (50%) re-
H&E TTF-1 p63 CD56 Alteration
50 µm
ceived crizotinib. Furthermore, 34 patients with
KRAS G12C alterations in BRAF (n = 4), KRAS (n = 10), or
STK11 P321fs
AD-like

FGFR1 (n = 20) were enrolled in clinical trials.


TP53 L265_N268del
The frequencies of genomic alterations in the
S01654 diagnostic NGM data set were similar to those
of the retrospective discovery data set (Fig. 5, A
50 µm
PIK3CA E542K and B). BRAF mutations were predominantly
TP53 R273L activating (49, 50) (60%, possibly associated
SQ-like

CCND1 amp with sensitivity to BRAF or MEK inhibition).


FGFR1 amp In 24% of the cases, BRAF mutations were pre-
S00110
dicted to be inactivating (50, 51), which might
50 µm
TP53 splice 6i-1 predict sensitivity to dasatinib (52) (fig. S18).
RB1 loss In 34% of the remaining LC cases, which were
NEC

not otherwise classifiable, we found signature


S00530 alterations of AD or SQ (Fig. 5, A and B) that
provided both a genetic diagnosis and a ratio-
nale for genetically tailored therapy. Thus,
50 µm
EGFR K714N
KRAS G12C
combined immunohistochemical and genet-
NOS

TP53 R213Q ic diagnosis reduced the subgroup of LC to


CDKN2A del 1.1%.
S00195 We next compared the survival of patients in
SOX2 amp
our historical and prospective data sets. Survival
C D of stage IIIB/IV patients in the retrospective
S00567
S00587
S00640
S00710
S00725
S00732
S01318
S01407
S01499
S01505
S01508
S01517

S01581
S01586
S01580

1.0 data set was better than that in the prospective


Probability of survival

0.8
data set (P < 0.02, fig. S19). The most likely rea-
son for this difference is a higher proportion of
TP53
Loss Mutation

0.6 patients undergoing surgery with curative in-


RB1
EP300 0.4 AD tention in the retrospective data set (which
CA consisted of surgically resected cases only),
LC
TP53 0.2 LCNEC whereas very few patients underwent surgery
SCLC
RB1 0.0 SQ in the prospective data set. In contrast, the im-
Missense Nonsense Indel Loss 0 10 20 30 40 50 60 proved survival of patients with earlier-stage dis-
Month ease in the prospective data set compared to the
Fig. 3. Genetic features typical of other lung cancer subtypes in LC. (A) Unsupervised hierarchical retrospective data set most likely reflects treat-
clustering using 294 highly variable (SD/mean >2.1) expressed genes identified four gene expression ment improvements. Histology had no major
subgroups containing mainly CA (I), SCLC (II), AD (III), and SQ (IV). LC samples are indicated as triangles impact on survival of advanced-stage patients
at corresponding positions below the cluster dendrogram. They are colored orange if they have AD- in both data sets (Fig. 5C).
specific alterations, blue if they have SQ-specific alterations, gray if the case was initially diagnosed as We next performed analyses within the
an LCNEC, and green if they have no known alteration. Genetic alterations (label: red, amplified; blue, prospective NGM cohort alone. When com-
deleted; black, mutated; ERBB includes mutation in EGFR or ERBB2) are given for selected genes per paring the survival of patients in our prospec-
sample as vertical lines (LC cases in green; others in black). (B) Typical immunohistochemistry is shown tive data set, whose tumors had been genotyped
for LC specimens with immunohistochemical and genetic characteristics of AD (AD-like), SQ (SQ-like),
(n = 975) to that of patients in the same cohort,
and NEC, as well as LC lacking features of other lung cancer subtypes (NOS, not otherwise specified). The
corresponding genetic alterations are indicated on the right. H&E, hematoxylin and eosin. (C) Distribu- in whom genetic diagnosis was not feasible
tion of mutations (in red, symbols according to type of mutation: diamond for missense, square for (for example, because of insufficient tissue;
nonsense, and circle for indel) and copy number loss (in blue) of TP53, RB1, and EP300 across all whole n = 277), genotyping alone had stage- and
exome–sequenced LCNECs. (D) Overall survival corresponding to each histological lung cancer subtype, histology-independent impact on overall sur-
with LC separated into LCs with neuroendocrine (gray) and without neuroendocrine (green) features. vival in multivariate analyses (P = 0.002) (Fig.

www.ScienceTranslationalMedicine.org 30 October 2013 Vol 5 Issue 209 209ra153 6


RESEARCH ARTICLE

Fig. 4. Genomics-based classification of A


lung cancer. (A) Semisupervised reclassification
Histology Prediction Agreement of prediction
of lung tumor samples. The relative proportion
of cases per histological subtype (left; the LC
original with CPR
group includes LCNEC cases) that were reclassi- AD LC SCLC SQ
incl LCNEC
fied on the basis of 18 genetic alterations (table 0.8
AD AD 0.6
S11) to a certain subgroup (labels in the middle) 91.4% 0.4
is illustrated as lines. The weight of the lines is n = 393 0.2
0
proportional to the fraction of cases classified 0.8
0.6
48% LC

Frequency
to the respective subgroup. All cases that were LC 0.4
0.2
predicted to be LC were histological LCNEC. Bars incl LCNEC LCNEC 0 * *
in the right graph give the concordance of each n = 80 0.8
predicted class with the central pathological re- 0.6
0.4
view (CPR). Subtypes for which no CPR was avail- SCLC 81% SCLC 0.2
0
able are denoted with asterisks. (B) Supervised n = 48
in silico classification of lung cancer specimens 0.8
0.6
based on genetic features for 637 tumor samples 0.4
SQ 79% SQ 0.2
with at least one genetic alteration and vali- n = 245 0 *
dation of the classifier in independent data * CPR not available
sets of all three subgroups (20, 21, 25). Original
histological subtypes defined groups for su- B

91.7
pervised learning. Bars indicate classification

84.2
77.1

79.9
100
71.3
100

71.5
prediction in %

frequencies relative to the original histology.


Model-based

Classification results for the CLCGP data set 75 75


are shown on the left, and results of the three
50 50

20.8
17.8

validation data sets are on the right. (C) Semi-


10.8

11.9

10.5
10.1
10.1
8.3
supervised genetics-based reclassification of

11

7.7
25 25

5.3
0

LC specimens without neuroendocrine features. 0 0


For each sample (rows), prediction to a certain Histology:
subtype (color per row in accordance to the AD SCLC SQ AD SCLC SQ
color code used for histological subtypes, see Training data set Validation data set
below) is given (lower graph). Degree of su-
pervision (x axis, upper graph) decreases con-
tinuously from left to right, the farthest right C Degree of supervision
representing a genetics-based prediction. High (histology) Low (genetics)

Chromogranin A
prediction with CPR

1.0

Synaptophysin
Agreement of the prediction with the CPR is
Agreement of

0.8
plotted for each stage of supervision (upper 0.6

CDKN2A
NFE2L2
part). Detailed information is given in Supple-
NKX2-1

CCND1
MYCL1
ERBB2

FGFR1
0.4
STK11

MDM2

MYCN
EGFR

EGFR

CK5/6
KRAS

KRAS

TTF-1
BRAF

SOX2

CD56
MYC
mentary Materials and Methods. Genome
CPR

RB1

CK7
0.2

p63
alterations (black lines) and immunohistochem- 0
istry results (black, positive; brown, negative;
LC without LCNEC

Negative
thin gray line, not available) are indicated for
each sample (middle and right panels). Genes
are sorted according to their predictive value
for histological subtypes. For the cases in the

Positive
lower part of the figure, immunohistochemistry
was not performed. Color code for predicted
classes and CPR: orange, AD; black, CA; green,
LC; gray, LCNEC; red, SCLC; blue, SQ; combina- Mutated NA
tion of colors, mixed subtype; white, no CPR.

5D, upper left). Although this observation most likely results from the hort. Patients with EGFR-mutant lung cancer treated with EGFR inhib-
favorable outcome in patients treated with kinase inhibitors, it dem- itors survived longer than those not receiving EGFR inhibitors (median
onstrates that genotyping is mandatory for patients to benefit from tar- overall survival, 31.5 versus 9.6 months; P < 0.001) (Fig. 5D, lower left).
geted therapeutic intervention. Accordingly, among the different genotypes Similarly, the overall survival of patients with ALK-rearranged lung
determined within the prospective NGM cohort, EGFR mutations were cancer treated with crizotinib was significantly better compared to
associated with improved survival (Fig. 5D, upper right; hazard ratio, ALK-positive patients not receiving crizotinib (median overall survival,
0.617; 95% confidence interval, 0.442 to 0.859; P = 0.004). We note, 23 versus 11 months; P = 0.024) (Fig. 5D, lower right). No difference in
however, that by the time ALK fusion testing was introduced, ALK in- the number of therapeutic regimens and the number of platinum-based
hibitors were not yet broadly available, which may explain why ALK treatment regimens existed between patients with EGFR-mutant lung
fusions were not generally associated with improved survival in our co- cancer treated with EGFR inhibitors and patients not treated with EGFR

www.ScienceTranslationalMedicine.org 30 October 2013 Vol 5 Issue 209 209ra153 7


RESEARCH ARTICLE

Fig. 5. Clinically relevant genome A


alterations in lung cancer subtypes. Retrospective Prospective
(A) Genetic alterations per histological
subtype in retrospective and prospective

AD

AD
sample sets. Each chart represents the
overall population with proportions of
histological subtypes color-coded in the LC LC ALK fusion
outer ring. Frequencies of alterations (wild BRAF
DDR2
type: no alteration in ALK, EGFR, FGFR1, EGFR
FGFR1 amp
KRAS, or PIK3CA) are given per gene rela- HER2 amp

LC
KRAS

LC
tive to all cases within each histological

SC
PIK3CA

SC
subtype. Distribution of alterations for Various

SQ
SQ Wild type
the LC population is shown separately.
(B) Genotyping results of 3590 patients
enrolled in the prospective screening ef- B C Retrospective stages IIIB/IV Prospective stages IIIB/IV
fort are sorted according to the histolog-

amp
mut
1.0 AD 1.0 AD
mut
amp
Histology

mut
mut
mut
fusion
ical subtype [AD (2250), CA (3), SCLC LC LC

PIK3CA
FGFR1

Probability of survival
SCLC SCLC
EGFR
KRAS

DDR2
HER2

BRAF
(265), SQ (1018), LC (47), and LCNEC (7)]. 0.8 SQ 0.8 SQ
ALK

Colored lines indicate alterations, and


0.6 0.6
gray lines indicate wild type. Frequencies
Lung tumor cases

of alterations for AD (orange) and SQ 0.4 0.4


(blue) are plotted below the respective
genes, comparing the mutation frequen- 0.2 0.2
cy in the prospective data set (dark colors) 0.0 0.0
to the retrospective data set (light colors). 0 10 20 30 40 50 60 0 10 20 30 40 50 60
No significant difference between the
data sets was seen (q values are given D All cases Stages IIIB/IV
on the graph; P values were adjusted 1.0 HR 0.719 (CI 95 0.588-0.879) 1.0 ALK fusion
mOS
(14)
for multiple hypothesis testing using mOS 31.6 vs 15.1, P = 0.001 EGFR (30.1)
FGFR1 amp (9.4)
Frequency % LCNEC LC SQ SCLC CA AD

the Benjamini and Hochberg method). 0.8 0.8


KRAS (8.3)
Color code: orange, AD; black, CA; green, PIK3CA (7.7)
0.6 0.6 Wild type (11.1)
LC; gray, LCNEC; red, SCLC; blue, SQ. (C)
Kaplan-Meier curves for overall survival 0.4 0.4
are shown for stage IIIB/IV patients per
0.2 0.2
Probability of survival

histological subtype for the retrospec- Genotyped


tive (left) and prospective (right) sample 0.0 Not genotyped 0.0
sets. No significant difference was seen 0 10 20 30 40 50 60 0 10 20 30 40 50 60
between subtypes within each data
set. (D) Prospective sample set: Kaplan- EGFR mutation positive ALK fusion positive
Meier curves for overall survival are shown 1.0 HR 0.169 (CI 95 0.07-0.41) 1.0 HR 0.235 (CI95 0.074-0.742)
mOS 31.5 vs 9.6, P < 0.001 mOS 23 vs 11, P = 0.024
for all patients who were genetically tested 0.8 0.8
versus those without available genetic
0.6 0.6
0

information (top left). Overall survival is


shown for stage IIIB/IV patients with altera-
*0

0.4 0.4
.6
*0

*0
*0

*0

tions in given genes versus patients with


10

7
.4

.4
.3

.3
8

8
7

wild type in the given genes (top right). 0.2 EGFRi 0.2 Crizotinib
*0

*0
.3

Standard Standard
.3
8
*0
100

Overall survival was statistically significantly


7

chemotherapy 0.0 chemotherapy


.4

0.0
8

longer in EGFR-mutant cases compared to Prospective *q value


AD

SQ

Retrospective 0 10 20 30 40 50 60 0 10 20 30 40 50 60
all other (log-rank test, P < 0.05) except
Month
ALK-rearranged cases (P = 0.065). Overall
survival is shown for patients with EGFR mutation treated with an EGFR inhibitor survival in the patient group treated with kinase inhibitors versus standard
or standard chemotherapy (bottom left) and patients with ALK translocations chemotherapy is given by the median overall survival (mOS). P values were
treated with crizotinib or standard chemotherapy (bottom right). Gain of overall corrected using the Bonferroni adjustment. HR, hazard ratio.

inhibitors (two-sided t test, P = 0.43). Patients with lung cancer bles, only treatment with kinase inhibitors had a significant impact on
harboring ALK rearrangements treated with crizotinib did not differ survival (P = 0.043).
in the number of platinum-based chemotherapy regimens, but differed In summary, we validated the frequencies of signature alterations
in the total number of treatment regimens received: patients treated with across lung cancer subtypes, demonstrated the feasibility of thorough
crizotinib received an average of 2.17 regimens, compared to 1.3 regi- and broad genome diagnostics in an academic-to-nonacademic out-
mens in patients who did not receive crizotinib (two-sided t test, P = reach setting, confirmed the almost universal reassignment of LC to
0.034) (table S12). However, in a Cox regression analysis including treat- the other biologically relevant diagnoses, and showed that the introduc-
ment with kinase inhibitors and number of treatment regimens as varia- tion of a molecular diagnosis coupled to specific therapeutic interven-

www.ScienceTranslationalMedicine.org 30 October 2013 Vol 5 Issue 209 209ra153 8


RESEARCH ARTICLE

tion improves the overall survival of patients with alterations in EGFR In summary, we provide a blueprint for genomic diagnosis of lung
and ALK compared to standard chemotherapy (53, 54). tumors. Determination of immunohistochemical, genomic, and clinical
features may thus be combined to yield classes of tumors that are bio-
logically relevant, afford genomically tailored stratification of patients
DISCUSSION into clinical trials, and improve overall survival of patients with lung
cancer.
Here, we defined a minimal set of genome alterations for genomic lung
cancer diagnosis using comprehensive copy number analyses in com-
bination with focused sequencing and RNA expression analysis. This MATERIALS AND METHODS
approach yielded robust frequencies of genome alterations and afforded
reassignment of LC to therapeutically relevant groups such as AD or Study design
SQ. Finally, introduction of genome diagnostics in an outreach setting Detailed information on materials and methods, including study design
not only confirmed these observations but also resulted in substantial and statistics, is given in the Supplementary Materials. In brief, we col-
improvement in overall survival of patients receiving genetically in- lected frozen tissue or genomic DNA from 1882 resected primary lung
formed therapeutic intervention compared to standard chemothera- tumors (table S1) after obtaining informed consent. Mutations in 28 genes
peutic treatment. (ABL1, AKT2, ALK, BRAF, CDK4, DDR2, EGFR, EPHA3, EPHA5, ERBB2,
A major advantage of this combined histopathological and genetic FGFR1, FGFR2, FGFR3, FLT3, HRAS, JAK2, KEAP1, KIT, KRAS, NFE2L2,
analysis is the classification of the group of LCs, a subtype that is poorly NRAS, NTRK1, NTRK3, PDGFRA, PIK3CA, STK11, TP53, and RET)
defined, mainly because of a lack of specific morphologic features of were analyzed in 1127 tumor specimens using multiple technologies
AD, SCLC, or SQ. Applying immunohistochemistry (22, 23) (for exam- (table S13, A and B) (9, 28, 38). Rearrangements of ALK (n = 602), RET
ple, TTF-1 for AD and p63 for SQ) helped assigning some of the LC (n = 362), and ROS1 (n = 211) were detected by fluorescence in situ
cases to other categories. However, the addition of genome annotation hybridization. Gene copy number of 1032 and whole-exome sequenc-
not only confirmed several of the immunohistochemical assignments ing data of 15 LCNEC tumors were analyzed as described previously
but also added information on possibly therapeutically relevant altera- (11, 24). Gene expression analyses were performed in 261 samples. Five
tions and afforded classification in cases where definite pathological hundred eighty-three cases were independently reviewed by lung
diagnosis was not possible. We have also observed a marked similarity pathologists (E.B. and W.D.T.) (1, 2) (fig. S1). For the prospective diag-
between several LCNEC tumors and SCLC that shared the same pattern nostic cohort, 5145 lung cancer patients were included for central geno-
of SCNAs, the particularly poor survival, and the most significant gene typing for alterations in ALK, BRAF, DDR2, EGFR, ERBB2, FGFR1,
mutations in this data set of limited size. KRAS, or PIK3CA, and written reports were provided to the treating
We also found (exceptionally rare) SQ tumors bearing EGFR muta- oncologists, containing the detected mutation and a treatment recom-
tions or ALK rearrangements, which might be treatable with targeted mendation (fig. S17 and table S12).
therapies as well. Thus, genomic diagnosis should include all subtypes
and all genome alterations to provide a therapeutic rationale for all pos-
SUPPLEMENTARY MATERIALS
sible patients. We propose to capture both genomic and immunohis-
tochemical data to link these taxa to treatment benefit in trials (fig. S20). www.sciencetranslationalmedicine.org/cgi/content/full/5/209/209ra153/DC1
In our outreach study, genotyping alone—as a prerequisite for per- Materials and Methods
sonalized treatment—was associated with improved patient survival. Fig. S1. Overview of sample processing.
Fig. S2. Significantly amplified and deleted regions in lung cancer.
These epidemiological results emphasize the need for broad availability Fig. S3. 9p21 is the most frequently deleted region in lung cancer.
of systematic and comprehensive genomic lung cancer diagnoses. Fig. S4. Mutation frequencies by histological subtype.
We note that the prospective part of our study was not a randomized Fig. S5. Distribution of mutations within genes.
clinical trial but an observational diagnostic intervention study. Un- Fig. S6. FGFR alterations in lung tumors.
Fig. S7. Frequencies of alterations in genes with known or potential clinical relevance in lung cancer.
fortunately, obtaining overall survival data in a randomized fashion
Fig. S8. Genetic associations in lung cancer.
requires prohibiting patients from crossing to the other treatment Fig. S9. EGFR mutations and amplifications frequently co-occur in lung AD but not in SQ.
arm—an irresponsible measure in this setting. Thus, registry data Fig. S10. Homozygous and hemizygous deletions of PTEN in EGFR-mutant ADs.
from observational studies like ours may be an approach to demonstrate Fig. S11. Central pathological review.
differences in survival between two different therapeutic strategies. Fig. S12. Gene expression subtypes of AD according to Wilkerson.
Fig. S13. Gene expression subtypes of SQ according to Wilkerson.
Furthermore, the two cohorts analyzed in this study differ in impor- Fig. S14. Genome-wide comparison of LCNEC and SCLC.
tant aspects: Whereas patient registration and genotyping occurred in a Fig. S15. Semisupervised reclassification of lung cancer specimens.
central study office in the prospective cohort, tumors of the retrospective Fig. S16. Automated reclassification of AD, SCLC, and SQ.
cohort were obtained from multiple centers before targeted therapies Fig. S17. Prospective testing of genomics-based diagnosis of lung cancer.
Fig. S18. BRAF mutations in lung cancer.
became broadly available, thus giving us limited control over the quality
Fig. S19. Overall survival by stage.
of the clinical data. Additional differences existed in the time of treat- Fig. S20. Diagnostic algorithm for lung tumors.
ment and distribution of stages and rate of surgical treatment. Finally, Table S1. Clinical and genetic features of all CLCGP cases.
clinical annotation was not complete, and data gaps exist for perform- Table S2. Significant copy number alterations in lung cancer for each histological subtype.
ance status, smoking status, and treatment. We note, however, that all Table S3. Median copy number per patient for chromosome regions with significantly altered
copy number.
major class-related genotypic and immunohistochemical findings were Table S4. Annotation of genetic alterations in lung cancer patients.
consistent across both cohorts, thus underscoring the validity of our Table S5. Statistical results comparing alteration frequencies between histological subtypes.
approach. Table S6. Gene expression data used for hierarchical clustering.

www.ScienceTranslationalMedicine.org 30 October 2013 Vol 5 Issue 209 209ra153 9


RESEARCH ARTICLE

Table S7. Reclassification of LCs. 13. E. L. Kwak, Y. J. Bang, D. R. Camidge, A. T. Shaw, B. Solomon, R. G. Maki, S. H. Ou, B. J. Dezube,
Table S8. Mutations detected in 15 whole exome–sequenced LCNECs of the lung. P. A. Jänne, D. B. Costa, M. Varella-Garcia, W. H. Kim, T. J. Lynch, P. Fidias, H. Stubbs, J. A. Engelman,
Table S9. RNAseq-derived gene expression data for 10 LCNECs of the lung. L. V. Sequist, W. Tan, L. Gandhi, M. Mino-Kenudson, G. C. Wei, S. M. Shreeve, M. J. Ratain,
Table S10. Results of the unsupervised genetics-based classification of lung tumors. J. Settleman, J. G. Christensen, D. A. Haber, K. Wilner, R. Salgia, G. I. Shapiro, J. W. Clark,
Table S11. Automated supervised prediction of lung cancer subtypes based on genetic alterations. A. J. Iafrate, Anaplastic lymphoma kinase inhibition in non–small-cell lung cancer. N. Engl. J. Med.
Table S12. Clinical and genetic features of all NGM cases. 363, 1693–1703 (2010).
Table S13. Primer sequences. 14. M. Meyerson, S. Gabriel, G. Getz, Advances in understanding cancer genomes through
References (55–62) second-generation sequencing. Nat. Rev. Genet. 11, 685–696 (2010).
15. K. Bergethon, A. T. Shaw, S. H. Ou, R. Katayama, C. M. Lovly, N. T. McDonald, P. P. Massion,
C. Siwak-Tapp, A. Gonzalez, R. Fang, E. J. Mark, J. M. Batten, H. Chen, K. D. Wilner, E. L. Kwak,
J. W. Clark, D. P. Carbone, H. Ji, J. A. Engelman, M. Mino-Kenudson, W. Pao, A. J. Iafrate,
REFERENCES AND NOTES ROS1 rearrangements define a unique molecular class of lung cancers. J. Clin. Oncol. 30,
863–870 (2012).
1. W. Travis, E. Brambilla, H. Mueller-Hermelink, C. Harris, World Health Organization Classification 16. Y. S. Ju, W. C. Lee, J. Y. Shin, S. Lee, T. Bleazard, J. K. Won, Y. T. Kim, J. I. Kim, J. H. Kang, J. S. Seo,
of Tumours. Pathology and Genetics: Tumours of the Lung, Pleura, Thymus and Heart (World A transforming KIF5B and RET gene fusion in lung adenocarcinoma revealed from whole-
Health Organization, Geneva, 2004). genome and transcriptome sequencing. Genome Res. 22, 436–445 (2012).
2. W. D. Travis, E. Brambilla, M. Noguchi, A. G. Nicholson, K. Geisinger, Y. Yatabe, C. A. Powell, 17. T. Kohno, H. Ichikawa, Y. Totoki, K. Yasuda, M. Hiramoto, T. Nammo, H. Sakamoto, K. Tsuta,
D. Beer, G. Riely, K. Garg, J. H. Austin, V. W. Rusch, F. R. Hirsch, J. Jett, P. C. Yang, M. Gould, K. Furuta, Y. Shimada, R. Iwakawa, H. Ogiwara, T. Oike, M. Enari, A. J. Schetter, H. Okayama,
American Thoracic Society, International Association for the Study of Lung Cancer/ A. Haugen, V. Skaug, S. Chiku, I. Yamanaka, Y. Arai, S. Watanabe, I. Sekine, S. Ogawa, C. C. Harris,
American Thoracic Society/European Respiratory Society: International multidisciplinary clas- H. Tsuda, T. Yoshida, J. Yokota, T. Shibata, KIF5B-RET fusions in lung adenocarcinoma. Nat. Med.
sification of lung adenocarcinoma: Executive summary. Proc. Am. Thorac. Soc. 8, 381–385 18, 375–377 (2012).
(2011). 18. D. Lipson, M. Capelletti, R. Yelensky, G. Otto, A. Parker, M. Jarosz, J. A. Curran, S. Balasubramanian,
3. T. J. Lynch, D. W. Bell, R. Sordella, S. Gurubhagavatula, R. A. Okimoto, B. W. Brannigan, P. L. Harris, T. Bloom, K. W. Brennan, A. Donahue, S. R. Downing, G. M. Frampton, L. Garcia, F. Juhn,
S. M. Haserlat, J. G. Supko, F. G. Haluska, D. N. Louis, D. C. Christiani, J. Settleman, D. A. Haber, K. C. Mitchell, E. White, J. White, Z. Zwirko, T. Peretz, H. Nechushtan, L. Soussan-Gutman,
Activating mutations in the epidermal growth factor receptor underlying responsiveness of J. Kim, H. Sasaki, H. R. Kim, S. I. Park, D. Ercan, C. E. Sheehan, J. S. Ross, M. T. Cronin, P. A. Jänne,
non–small-cell lung cancer to gefitinib. N. Engl. J. Med. 350, 2129–2139 (2004). P. J. Stephens, Identification of new ALK and RET gene fusions from colorectal and lung
4. J. G. Paez, P. A. Jänne, J. C. Lee, S. Tracy, H. Greulich, S. Gabriel, P. Herman, F. J. Kaye, N. Lindeman, cancer biopsies. Nat. Med. 18, 382–384 (2012).
T. J. Boggon, K. Naoki, H. Sasaki, Y. Fujii, M. J. Eck, W. R. Sellers, B. E. Johnson, M. Meyerson, 19. K. Takeuchi, M. Soda, Y. Togashi, R. Suzuki, S. Sakata, S. Hatano, R. Asaka, W. Hamanaka,
EGFR mutations in lung cancer: Correlation with clinical response to gefitinib therapy. Science H. Ninomiya, H. Uehara, Y. Lim Choi, Y. Satoh, S. Okumura, K. Nakagawa, H. Mano, Y. Ishikawa,
304, 1497–1500 (2004). RET, ROS1 and ALK fusions in lung cancer. Nat. Med. 18, 378–381 (2012).
5. W. Pao, N. Girard, New driver mutations in non-small-cell lung cancer. Lancet Oncol. 12, 20. Cancer Genome Atlas Research Network, Comprehensive genomic characterization of
175–180 (2011). squamous cell lung cancers. Nature 489, 519–525 (2012).
6. S. Rodenhuis, R. J. Slebos, A. J. Boot, S. G. Evers, W. J. Mooi, S. S. Wagenaar, P. C. van Bodegom, 21. M. Imielinski, A. H. Berger, P. S. Hammerman, B. Hernandez, T. J. Pugh, E. Hodis, J. Cho, J. Suh,
J. L. Bos, Incidence and possible clinical significance of K-ras oncogene activation in adeno- M. Capelletti, A. Sivachenko, C. Sougnez, D. Auclair, M. S. Lawrence, P. Stojanov, K. Cibulskis,
carcinoma of the human lung. Cancer Res. 48, 5738–5741 (1988). K. Choi, L. de Waal, T. Sharifnia, A. Brooks, H. Greulich, S. Banerji, T. Zander, D. Seidel, F. Leenders,
7. M. Soda, Y. L. Choi, M. Enomoto, S. Takada, Y. Yamashita, S. Ishikawa, S. Fujiwara, H. Watanabe, S. Ansén, C. Ludwig, W. Engel-Riedel, E. Stoelben, J. Wolf, C. Goparju, K. Thompson, W. Winckler,
K. Kurashina, H. Hatanaka, M. Bando, S. Ohno, Y. Ishikawa, H. Aburatani, T. Niki, Y. Sohara, D. Kwiatkowski, B. E. Johnson, P. A. Jänne, V. A. Miller, W. Pao, W. D. Travis, H. I. Pass, S. B. Gabriel,
Y. Sugiyama, H. Mano, Identification of the transforming EML4-ALK fusion gene in non- E. S. Lander, R. K. Thomas, L. A. Garraway, G. Getz, M. Meyerson, Mapping the hallmarks of
small-cell lung cancer. Nature 448, 561–566 (2007). lung adenocarcinoma with massively parallel sequencing. Cell 150, 1107–1120 (2012).
8. A. J. Bass, H. Watanabe, C. H. Mermel, S. Yu, S. Perner, R. G. Verhaak, S. Y. Kim, L. Wardwell, 22. J. Pardo, A. M. Martinez-Peñuela, J. J. Sola, A. Panizo, A. Gúrpide, J. M. Martinez-Peñuela,
P. Tamayo, I. Gat-Viks, A. H. Ramos, M. S. Woo, B. A. Weir, G. Getz, R. Beroukhim, M. O’Kelly, M. D. Lozano, Large cell carcinoma of the lung: An endangered species? Appl. Immu-
A. Dutt, O. Rozenblatt-Rosen, P. Dziunycz, J. Komisarof, L. R. Chirieac, C. J. Lafargue, V. Scheble, nohistochem. Mol. Morphol. 17, 383–392 (2009).
T. Wilbertz, C. Ma, S. Rao, H. Nakagawa, D. B. Stairs, L. Lin, T. J. Giordano, P. Wagner, J. D. Minna, 23. N. Rekhtman, D. C. Ang, C. S. Sima, W. D. Travis, A. L. Moreira, Immunohistochemical
A. F. Gazdar, C. Q. Zhu, M. S. Brose, I. Cecconello, U. Ribeiro Jr., S. K. Marie, O. Dahl, R. A. algorithm for differentiation of lung adenocarcinoma and squamous cell carcinoma based
Shivdasani, M. S. Tsao, M. A. Rubin, K. K. Wong, A. Regev, W. C. Hahn, D. G. Beer, A. K. Rustgi, on large series of whole-tissue sections with validation in small specimens. Mod. Pathol.
M. Meyerson, SOX2 is an amplified lineage-survival oncogene in lung and esophageal squa- 24, 1348–1359 (2011).
mous cell carcinomas. Nat. Genet. 41, 1238–1242 (2009). 24. M. Peifer, L. Fernández-Cuesta, M. L. Sos, J. George, D. Seidel, L. H. Kasper, D. Plenker,
9. P. S. Hammerman, M. L. Sos, A. H. Ramos, C. Xu, A. Dutt, W. Zhou, L. E. Brace, B. A. Woods, F. Leenders, R. Sun, T. Zander, R. Menon, M. Koker, I. Dahmen, C. Müller, V. Di Cerbo,
W. Lin, J. Zhang, X. Deng, S. M. Lim, S. Heynck, M. Peifer, J. R. Simard, M. S. Lawrence, H. U. Schildhaus, J. Altmüller, I. Baessmann, C. Becker, B. de Wilde, J. Vandesompele,
R. C. Onofrio, H. B. Salvesen, D. Seidel, T. Zander, J. M. Heuckmann, A. Soltermann, H. Moch, D. Böhm, S. Ansén, F. Gabler, I. Wilkening, S. Heynck, J. M. Heuckmann, X. Lu, S. L. Carter,
M. Koker, F. Leenders, F. Gabler, S. Querings, S. Ansén, E. Brambilla, C. Brambilla, P. Lorimier, K. Cibulskis, S. Banerji, G. Getz, K. S. Park, D. Rauh, C. Grütter, M. Fischer, L. Pasqualucci,
O. T. Brustugun, A. Helland, I. Petersen, J. H. Clement, H. Groen, W. Timens, H. Sietsma, G. Wright, Z. Wainer, P. Russell, I. Petersen, Y. Chen, E. Stoelben, C. Ludwig, P. Schnabel,
E. Stoelben, J. Wolf, D. G. Beer, M. S. Tsao, M. Hanna, C. Hatton, M. J. Eck, P. A. Janne, H. Hoffmann, T. Muley, M. Brockmann, W. Engel-Riedel, L. A. Muscarella, V. M. Fazio, H. Groen,
B. E. Johnson, W. Winckler, H. Greulich, A. J. Bass, J. Cho, D. Rauh, N. S. Gray, K. K. Wong, W. Timens, H. Sietsma, E. Thunnissen, E. Smit, D. A. Heideman, P. J. Snijders, F. Cappuzzo,
E. B. Haura, R. K. Thomas, M. Meyerson, Mutations in the DDR2 kinase gene identify a C. Ligorio, S. Damiani, J. Field, S. Solberg, O. T. Brustugun, M. Lund-Iversen, J. Sänger,
novel therapeutic target in squamous cell lung cancer. Cancer Discov. 1, 78–89 (2011). J. H. Clement, A. Soltermann, H. Moch, W. Weder, B. Solomon, J. C. Soria, P. Validire, B. Besse,
10. A. Dutt, A. H. Ramos, P. S. Hammerman, C. Mermel, J. Cho, T. Sharifnia, A. Chande, K. E. Tanaka, E. Brambilla, C. Brambilla, S. Lantuejoul, P. Lorimier, P. M. Schneider, M. Hallek, W. Pao,
N. Stransky, H. Greulich, N. S. Gray, M. Meyerson, Inhibitor-sensitive FGFR1 amplification in M. Meyerson, J. Sage, J. Shendure, R. Schneider, R. Büttner, J. Wolf, P. Nürnberg, S. Perner,
human non-small cell lung cancer. PLoS One 6, e20351 (2011). L. C. Heukamp, P. K. Brindle, S. Haas, R. K. Thomas, Integrative genome analyses identify
11. J. Weiss, M. L. Sos, D. Seidel, M. Peifer, T. Zander, J. M. Heuckmann, R. T. Ullrich, R. Menon, key somatic driver mutations of small-cell lung cancer. Nat. Genet. 44, 1104–1110 (2012).
S. Maier, A. Soltermann, H. Moch, P. Wagener, F. Fischer, S. Heynck, M. Koker, J. Schöttle, 25. C. M. Rudin, S. Durinck, E. W. Stawiski, J. T. Poirier, Z. Modrusan, D. S. Shames, E. A. Bergbower,
F. Leenders, F. Gabler, I. Dabow, S. Querings, L. C. Heukamp, H. Balke-Want, S. Ansén, D. Rauh, Y. Guan, J. Shin, J. Guillory, C. S. Rivers, C. K. Foo, D. Bhatt, J. Stinson, F. Gnad, P. M. Haverty,
I. Baessmann, J. Altmüller, Z. Wainer, M. Conron, G. Wright, P. Russell, B. Solomon, E. Brambilla, R. Gentleman, S. Chaudhuri, V. Janakiraman, B. S. Jaiswal, C. Parikh, W. Yuan, Z. Zhang,
C. Brambilla, P. Lorimier, S. Sollberg, O. T. Brustugun, W. Engel-Riedel, C. Ludwig, I. Petersen, H. Koeppen, T. D. Wu, H. M. Stern, R. L. Yauch, K. E. Huffman, D. D. Paskulin, P. B. Illei, M. Varella-Garcia,
J. Sänger, J. Clement, H. Groen, W. Timens, H. Sietsma, E. Thunnissen, E. Smit, D. Heideman, A. F. Gazdar, F. J. de Sauvage, R. Bourgon, J. D. Minna, M. V. Brock, S. Seshagiri, Comprehen-
F. Cappuzzo, C. Ligorio, S. Damiani, M. Hallek, R. Beroukhim, W. Pao, B. Klebl, M. Baumann, sive genomic analysis identifies SOX2 as a frequently amplified gene in small-cell lung
R. Buettner, K. Ernestus, E. Stoelben, J. Wolf, P. Nürnberg, S. Perner, R. K. Thomas, Frequent cancer. Nat. Genet. 44, 1111–1116 (2012).
and focal FGFR1 amplification associates with therapeutically tractable FGFR1 dependency 26. S. L. Carter, K. Cibulskis, E. Helman, A. McKenna, H. Shen, T. Zack, P. W. Laird, R. C. Onofrio,
in squamous cell lung cancer. Sci. Transl. Med. 2, 62ra93 (2010). W. Winckler, B. A. Weir, R. Beroukhim, D. Pellman, D. A. Levine, E. S. Lander, M. Meyerson,
12. W. Pao, J. Chmielecki, Rational, biologically based treatment of EGFR-mutant non-small-cell G. Getz, Absolute quantification of somatic DNA alterations in human cancer. Nat. Biotechnol.
lung cancer. Nat. Rev. Cancer 10, 760–774 (2010). 30, 413–421 (2012).

www.ScienceTranslationalMedicine.org 30 October 2013 Vol 5 Issue 209 209ra153 10


RESEARCH ARTICLE

27. R. Iwakawa, T. Kohno, Y. Anami, M. Noguchi, K. Suzuki, Y. Matsuno, K. Mishima, R. Nishikawa, R. J. Lee, A. K. Joe, J. L. Troy, R. Gonzalez, T. E. Hutson, I. Puzanov, B. Chmielowski, C. J. Springer,
F. Tashiro, J. Yokota, Association of p16 homozygous deletions with clinicopathologic char- G. A. McArthur, J. A. Sosman, R. S. Lo, A. Ribas, R. Marais, RAS mutations in cutaneous
acteristics and EGFR/KRAS/p53 mutations in lung adenocarcinoma. Clin. Cancer Res. 14, squamous-cell carcinomas in patients treated with BRAF inhibitors. N. Engl. J. Med. 366,
3746–3753 (2008). 207–215 (2012).
28. L. Ding, G. Getz, D. A. Wheeler, E. R. Mardis, M. D. McLellan, K. Cibulskis, C. Sougnez, H. Greulich, 40. F. Cappuzzo, F. R. Hirsch, E. Rossi, S. Bartolini, G. L. Ceresoli, L. Bemis, J. Haney, S. Witta,
D. M. Muzny, M. B. Morgan, L. Fulton, R. S. Fulton, Q. Zhang, M. C. Wendl, M. S. Lawrence, K. Danenberg, I. Domenichini, V. Ludovini, E. Magrini, V. Gregorc, C. Doglioni, A. Sidoni,
D. E. Larson, K. Chen, D. J. Dooling, A. Sabo, A. C. Hawes, H. Shen, S. N. Jhangiani, L. R. Lewis, M. Tonato, W. A. Franklin, L. Crino, P. A. Bunn Jr., M. Varella-Garcia, Epidermal growth factor
O. Hall, Y. Zhu, T. Mathew, Y. Ren, J. Yao, S. E. Scherer, K. Clerc, G. A. Metcalf, B. Ng, receptor gene and protein and gefitinib sensitivity in non-small-cell lung cancer. J. Natl.
A. Milosavljevic, M. L. Gonzalez-Garay, J. R. Osborne, R. Meyer, X. Shi, Y. Tang, D. C. Koboldt, Cancer Inst. 97, 643–655 (2005).
L. Lin, R. Abbott, T. L. Miner, C. Pohl, G. Fewell, C. Haipek, H. Schmidt, B. H. Dunford-Shore, 41. M. S. Tsao, A. Sakurada, J. C. Cutz, C. Q. Zhu, S. Kamel-Reid, J. Squire, I. Lorimer, T. Zhang, N. Liu,
A. Kraja, S. D. Crosby, C. S. Sawyer, T. Vickery, S. Sander, J. Robinson, W. Winckler, J. Baldwin, M. Daneshmand, P. Marrano, G. da Cunha Santos, A. Lagarde, F. Richardson, L. Seymour,
L. R. Chirieac, A. Dutt, T. Fennell, M. Hanna, B. E. Johnson, R. C. Onofrio, R. K. Thomas, G. Tonon, M. Whitehead, K. Ding, J. Pater, F. A. Shepherd, Erlotinib in lung cancer—Molecular and
B. A. Weir, X. Zhao, L. Ziaugra, M. C. Zody, T. Giordano, M. B. Orringer, J. A. Roth, M. R. Spitz, clinical predictors of outcome. N. Engl. J. Med. 353, 133–144 (2005).
I. I. Wistuba, B. Ozenberger, P. J. Good, A. C. Chang, D. G. Beer, M. A. Watson, M. Ladanyi, 42. M. Fukuoka, Y. L. Wu, S. Thongprasert, P. Sunpaweravong, S. S. Leong, V. Sriuranpong,
S. Broderick, A. Yoshizawa, W. D. Travis, W. Pao, M. A. Province, G. M. Weinstock, H. E. Varmus, T. Y. Chao, K. Nakagawa, D. T. Chu, N. Saijo, E. L. Duffield, Y. Rukazenkov, G. Speake, H. Jiang,
S. B. Gabriel, E. S. Lander, R. A. Gibbs, M. Meyerson, R. K. Wilson, Somatic mutations affect A. A. Armour, K. F. To, J. C. Yang, T. S. Mok, Biomarker analyses and final overall survival
key pathways in lung adenocarcinoma. Nature 455, 1069–1075 (2008). results from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/
29. D. Malhotra, E. Portales-Casamar, A. Singh, S. Srivastava, D. Arenillas, C. Happel, C. Shyr, paclitaxel in clinically selected patients with advanced non–small-cell lung cancer in Asia (IPASS).
N. Wakabayashi, T. W. Kensler, W. W. Wasserman, S. Biswal, Global mapping of binding J. Clin. Oncol. 29, 2866–2874 (2011).
sites for Nrf2 identifies novel targets in cell survival response through ChIP-Seq profiling 43. I. K. Mellinghoff, M. Y. Wang, I. Vivanco, D. A. Haas-Kogan, S. Zhu, E. Q. Dia, K. V. Lu, K. Yoshimoto,
and network analysis. Nucleic Acids Res. 38, 5718–5734 (2010). J. H. Huang, D. J. Chute, B. L. Riggs, S. Horvath, L. M. Liau, W. K. Cavenee, P. N. Rao, R. Beroukhim,
30. Y. R. Kim, J. E. Oh, M. S. Kim, M. R. Kang, S. W. Park, J. Y. Han, H. S. Eom, N. J. Yoo, S. H. Lee, T. C. Peck, J. C. Lee, W. R. Sellers, D. Stokoe, M. Prados, T. F. Cloughesy, C. L. Sawyers, P. S. Mischel,
Oncogenic NRF2 mutations in squamous cell carcinomas of oesophagus and skin. J. Pathol. Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N. Engl.
220, 446–451 (2010). J. Med. 353, 2012–2024 (2005).
31. T. Shibata, T. Ohta, K. I. Tong, A. Kokubu, R. Odogawa, K. Tsuta, H. Asamura, M. Yamamoto, 44. M. L. Sos, M. Koker, B. A. Weir, S. Heynck, R. Rabinovsky, T. Zander, J. M. Seeger, J. Weiss,
S. Hirohashi, Cancer related mutations in NRF2 impair its recognition by Keap1-Cul3 E3 F. Fischer, P. Frommolt, K. Michel, M. Peifer, C. Mermel, L. Girard, M. Peyton, A. F. Gazdar,
ligase and promote malignancy. Proc. Natl. Acad. Sci. U.S.A. 105, 13568–13573 (2008). J. D. Minna, L. A. Garraway, H. Kashkar, W. Pao, M. Meyerson, R. K. Thomas, PTEN loss
32. T. Ohta, K. Iijima, M. Miyamoto, I. Nakahara, H. Tanaka, M. Ohtsuji, T. Suzuki, A. Kobayashi, contributes to erlotinib resistance in EGFR-mutant lung cancer by activation of Akt and EGFR.
J. Yokota, T. Sakiyama, T. Shibata, M. Yamamoto, S. Hirohashi, Loss of Keap1 function activates Nrf2 Cancer Res. 69, 3256–3261 (2009).
and provides advantages for lung cancer cell growth. Cancer Res. 68, 1303–1309 (2008). 45. M. D. Wilkerson, X. Yin, V. Walter, N. Zhao, C. R. Cabanski, M. C. Hayward, C. R. Miller, M. A. Socinski,
33. D. Cappellen, C. De Oliveira, D. Ricol, S. de Medina, J. Bourdin, X. Sastre-Garau, D. Chopin, A. M. Parsons, L. B. Thorne, B. E. Haithcock, N. K. Veeramachaneni, W. K. Funkhouser, S. H. Randell,
J. P. Thiery, F. Radvanyi, Frequent activating mutations of FGFR3 in human bladder and P. S. Bernard, C. M. Perou, D. N. Hayes, Differential pathogenesis of lung adenocarcinoma
cervix carcinomas. Nat. Genet. 23, 18–20 (1999). subtypes involving sequence mutations, copy number, chromosomal instability, and meth-
34. M. Karoui, H. Hofmann-Radvanyi, U. Zimmermann, A. Couvelard, C. Degott, L. Faridoni-Laurens, ylation. PLoS One 7, e36530 (2012).
J. C. Ahomadegbe, S. Gazzeri, E. Brambilla, T. Clerici, P. Charbonnier, C. Tresallet, E. Mitry, 46. M. D. Wilkerson, X. Yin, K. A. Hoadley, Y. Liu, M. C. Hayward, C. R. Cabanski, K. Muldrew, C. R. Miller,
C. Penna, P. Rougier, C. Boileau, J. P. Thiery, B. Nordlinger, B. Franc, F. Radvanyi, No evidence S. H. Randell, M. A. Socinski, A. M. Parsons, W. K. Funkhouser, C. B. Lee, P. J. Roberts, L. Thorne,
of somatic FGFR3 mutation in various types of carcinoma. Oncogene 20, 5059–5061 (2001). P. S. Bernard, C. M. Perou, D. N. Hayes, Lung squamous cell carcinoma mRNA expression
35. A. Dutt, H. B. Salvesen, T. H. Chen, A. H. Ramos, R. C. Onofrio, C. Hatton, R. Nicoletti, W. Winckler, subtypes are reproducible, clinically important, and correspond to normal cell types. Clin.
R. Grewal, M. Hanna, N. Wyhs, L. Ziaugra, D. J. Richter, J. Trovik, I. B. Engelsen, I. M. Stefansson, Cancer Res. 16, 4864–4875 (2010).
T. Fennell, K. Cibulskis, M. C. Zody, L. A. Akslen, S. Gabriel, K. K. Wong, W. R. Sellers, M. Meyerson, 47. R. Shen, A. B. Olshen, M. Ladanyi, Integrative clustering of multiple genomic data types
H. Greulich, Drug-sensitive FGFR2 mutations in endometrial carcinoma. Proc. Natl. Acad. Sci. using a joint latent variable model with application to breast and lung cancer subtype
U.S.A. 105, 8713–8717 (2008). analysis. Bioinformatics 25, 2906–2912 (2009).
36. J. S. Seo, Y. S. Ju, W. C. Lee, J. Y. Shin, J. K. Lee, T. Bleazard, J. Lee, Y. J. Jung, J. O. Kim, J. Y. Shin, 48. H. Greulich, B. Kaplan, P. Mertins, T. H. Chen, K. E. Tanaka, C. H. Yun, X. Zhang, S. H. Lee, J. Cho,
S. B. Yu, J. Kim, E. R. Lee, C. H. Kang, I. K. Park, H. Rhee, S. H. Lee, J. I. Kim, J. H. Kang, Y. T. Kim, L. Ambrogio, R. Liao, M. Imielinski, S. Banerji, A. H. Berger, M. S. Lawrence, J. Zhang, N. H. Pho,
The transcriptional landscape and mutational profile of lung adenocarcinoma. Genome Res. S. R. Walker, W. Winckler, G. Getz, D. Frank, W. C. Hahn, M. J. Eck, D. R. Mani, J. D. Jaffe, S. A. Carr,
22, 2109–2119 (2012). K. K. Wong, M. Meyerson, Functional analysis of receptor tyrosine kinase mutations in lung
37. B. A. Weir, M. S. Woo, G. Getz, S. Perner, L. Ding, R. Beroukhim, W. M. Lin, M. A. Province, cancer identifies oncogenic extracellular domain mutations of ERBB2. Proc. Natl. Acad. Sci.
A. Kraja, L. A. Johnson, K. Shah, M. Sato, R. K. Thomas, J. A. Barletta, I. B. Borecki, S. Broderick, U.S.A. 109, 14476–14481 (2012).
A. C. Chang, D. Y. Chiang, L. R. Chirieac, J. Cho, Y. Fujii, A. F. Gazdar, T. Giordano, H. Greulich, 49. K. B. Dahlman, J. Xia, K. Hutchinson, C. Ng, D. Hucks, P. Jia, M. Atefi, Z. Su, S. Branch, P. L. Lyle,
M. Hanna, B. E. Johnson, M. G. Kris, A. Lash, L. Lin, N. Lindeman, E. R. Mardis, J. D. McPherson, D. J. Hicks, V. Bozon, J. A. Glaspy, N. Rosen, D. B. Solit, J. L. Netterville, C. L. Vnencak-Jones,
J. D. Minna, M. B. Morgan, M. Nadel, M. B. Orringer, J. R. Osborne, B. Ozenberger, A. H. Ramos, J. A. Sosman, A. Ribas, Z. Zhao, W. Pao, BRAFL597 mutations in melanoma are associated
J. Robinson, J. A. Roth, V. Rusch, H. Sasaki, F. Shepherd, C. Sougnez, M. R. Spitz, M. S. Tsao, with sensitivity to MEK inhibitors. Cancer Discov. 2, 791–797 (2012).
D. Twomey, R. G. Verhaak, G. M. Weinstock, D. A. Wheeler, W. Winckler, A. Yoshizawa, S. Yu, 50. P. T. Wan, M. J. Garnett, S. M. Roe, S. Lee, D. Niculescu-Duvaz, V. M. Good, C. M. Jones,
M. F. Zakowski, Q. Zhang, D. G. Beer, I. I. Wistuba, M. A. Watson, L. A. Garraway, M. Ladanyi, C. J. Marshall, C. J. Springer, D. Barford, R. Marais; Cancer Genome Project, Mechanism
W. D. Travis, W. Pao, M. A. Rubin, S. B. Gabriel, R. A. Gibbs, H. E. Varmus, R. K. Wilson, E. S. Lander, of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell
M. Meyerson, Characterizing the cancer genome in lung adenocarcinoma. Nature 450, 116, 855–867 (2004).
893–898 (2007). 51. S. J. Heidorn, C. Milagre, S. Whittaker, A. Nourry, I. Niculescu-Duvas, N. Dhomen,
38. R. K. Thomas, A. C. Baker, R. M. Debiasi, W. Winckler, T. Laframboise, W. M. Lin, M. Wang, J. Hussain, J. S. Reis-Filho, C. J. Springer, C. Pritchard, R. Marais, Kinase-dead BRAF and onco-
W. Feng, T. Zander, L. MacConaill, J. C. Lee, R. Nicoletti, C. Hatton, M. Goyette, L. Girard, genic RAS cooperate to drive tumor progression through CRAF. Cell 140, 209–221 (2010).
K. Majmudar, L. Ziaugra, K. K. Wong, S. Gabriel, R. Beroukhim, M. Peyton, J. Barretina, 52. B. Sen, S. Peng, X. Tang, H. S. Erickson, H. Galindo, T. Mazumdar, D. J. Stewart, I. Wistuba,
A. Dutt, C. Emery, H. Greulich, K. Shah, H. Sasaki, A. Gazdar, J. Minna, S. A. Armstrong, F. M. Johnson, Kinase-impaired BRAF mutations in lung cancer confer sensitivity to dasatinib.
I. K. Mellinghoff, F. S. Hodi, G. Dranoff, P. S. Mischel, T. F. Cloughesy, S. F. Nelson, L. M. Liau, Sci. Transl. Med. 4, 136ra70 (2012).
K. Mertz, M. A. Rubin, H. Moch, M. Loda, W. Catalona, J. Fletcher, S. Signoretti, F. Kaye, 53. R. Rosell, E. Carcereny, R. Gervais, A. Vergnenegre, B. Massuti, E. Felip, R. Palmero, R. Garcia-Gomez,
K. C. Anderson, G. D. Demetri, R. Dummer, S. Wagner, M. Herlyn, W. R. Sellers, M. Meyerson, C. Pallares, J. M. Sanchez, R. Porta, M. Cobo, P. Garrido, F. Longo, T. Moran, A. Insa, F. De Marinis,
L. A. Garraway, High-throughput oncogene mutation profiling in human cancer. Nat. Genet. R. Corre, I. Bover, A. Illiano, E. Dansin, J. de Castro, M. Milella, N. Reguart, G. Altavilla, U. Jimenez,
39, 347–351 (2007). M. Provencio, M. A. Moreno, J. Terrasa, J. Muñoz-Langa, J. Valdivia, D. Isla, M. Domine, O. Molinier,
39. F. Su, A. Viros, C. Milagre, K. Trunzer, G. Bollag, O. Spleiss, J. S. Reis-Filho, X. Kong, R. C. Koya, J. Mazieres, N. Baize, R. Garcia-Campelo, G. Robinet, D. Rodriguez-Abreu, G. Lopez-Vivanco,
K. T. Flaherty, P. B. Chapman, M. J. Kim, R. Hayward, M. Martin, H. Yang, Q. Wang, H. Hilton, V. Gebbia, L. Ferrera-Delgado, P. Bombaron, R. Bernabe, A. Bearz, A. Artal, E. Cortesi, C. Rolfo,
J. S. Hang, J. Noe, M. Lambros, F. Geyer, N. Dhomen, I. Niculescu-Duvaz, A. Zambon, M. Sanchez-Ronco, A. Drozdowskyj, C. Queralt, I. de Aguirre, J. L. Ramirez, J. J. Sanchez,
D. Niculescu-Duvaz, N. Preece, L. Robert, N. J. Otte, S. Mok, D. Kee, Y. Ma, C. Zhang, M. A. Molina, M. Taron, L. Paz-Ares, Spanish Lung Cancer Group in collaboration with Groupe
G. Habets, E. A. Burton, B. Wong, H. Nguyen, M. Kockx, L. Andries, B. Lestini, K. B. Nolop, Français de Pneumo-Cancérologie and Associazione Italiana Oncologia Toracica, Erlotinib

www.ScienceTranslationalMedicine.org 30 October 2013 Vol 5 Issue 209 209ra153 11


RESEARCH ARTICLE

versus standard chemotherapy as first-line treatment for European patients with advanced Roche, Bayer, Novartis, and Boehringer Ingelheim. M.L.S. is a fellow of the International Associ-
EGFR mutation-positive non-small-cell lung cancer (EURTAC): A multicentre, open-label, ran- ation for the Study of Lung Cancer. M.S. received funds from Lilly, Roche, Boehringer Ingelheim,
domised phase 3 trial. Lancet Oncol. 13, 239–246 (2012). AstraZeneca, and Pfizer. P.N. is a founder, CEO, and shareholder of ATLAS Biolabs GmbH. W.P.
54. A. T. Shaw, D. W. Kim, K. Nakagawa, T. Seto, L. Crinó, M. J. Ahn, T. De Pas, B. Besse, B. J. Solomon, received research funding from Enzon Xcovery, AstraZeneca, Symphogen, Clovis Oncology, and
F. Blackhall, Y. L. Wu, M. Thomas, K. J. O’Byrne, D. Moro-Sibilot, D. R. Camidge, T. Mok, V. Hirsh, Bristol-Myers Squibb and had paid consulting relationships with MolecularMD, AstraZeneca,
G. J. Riely, S. Iyer, V. Tassell, A. Polli, K. D. Wilner, P. A. Jänne, Crizotinib versus chemotherapy Bristol-Myers Squibb, Symphony Evolution, Clovis Oncology, Exelixis, and Clarient. Rights to EGFR
in advanced ALK-positive lung cancer. N. Engl. J. Med. 368, 2385–2394 (2013). T790M testing were licensed on behalf of W.P. and others by Memorial Sloan-Kettering Cancer
55. C. Greenman, P. Stephens, R. Smith, G. L. Dalgliesh, C. Hunter, G. Bignell, H. Davies, J. Teague, Center to MolecularMD. M.P. is a founder and shareholder of Blackfield AG, J.H.C. is a member of
A. Butler, C. Stevens, S. Edkins, S. O’Meara, I. Vastrik, E. E. Schmidt, T. Avis, S. Barthorpe, the Advisory Board of Boehringer Ingelheim Pharma GmbH & Co KG. H.G. is a member of the
G. Bhamra, G. Buck, B. Choudhury, J. Clements, J. Cole, E. Dicks, S. Forbes, K. Gray, K. Halliday, Advisory Boards of Roche, Eli Lilly, and Pfizer. H.-U.S. has an advisory relationship with Roche,
R. Harrison, K. Hills, J. Hinton, A. Jenkinson, D. Jones, A. Menzies, T. Mironenko, J. Perry, K. Raine, Abbott Molecular, and Pfizer and has received honoraria from Novartis, Roche, Abbott Molec-
D. Richardson, R. Shepherd, A. Small, C. Tofts, J. Varian, T. Webb, S. West, S. Widaa, A. Yates, ular, and Pfizer. B.S. is a member of the Advisory Boards for Pfizer, Novartis, Roche, Boehringer
D. P. Cahill, D. N. Louis, P. Goldstraw, A. G. Nicholson, F. Brasseur, L. Looijenga, B. L. Weber, Ingelheim, and AstraZeneca. J.W. is a member of the Advisory Boards for Novartis, Roche,
Y. E. Chiew, A. DeFazio, M. F. Greaves, A. R. Green, P. Campbell, E. Birney, D. F. Easton, AstraZeneca, Boehringer Ingelheim, Lilly, Bristol-Myers Squibb, and Bayer and has received
G. Chenevix-Trench, M. H. Tan, S. K. Khoo, B. T. Teh, S. T. Yuen, S. Y. Leung, R. Wooster, speaking fees from Novartis, Roche, AstraZeneca, Boehringer Ingelheim, and Lilly and research
P. A. Futreal, M. R. Stratton, Patterns of somatic mutation in human cancer genomes. Nature support from Roche, Boehringer Ingelheim, Novartis, and Bayer. T.Z. has performed advisory work
446, 153–158 (2007). for Merck, Lilly, Amgen, Roche, and Boehringer Ingelheim. D.B. is a member of regional advisory
56. J. T. Ney, S. Froehner, A. Roesler, R. Buettner, S. Merkelbach-Bruse, High-resolution melting boards for Roche, Lilly, and Boehringer Ingelheim. F.L. received consulting fees from Blackfield
analysis as a sensitive prescreening diagnostic tool to detect KRAS, BRAF, PIK3CA, and AG. S.M.-B. received honoraria and grants from Roche and Novartis. L.N. received speaking fees
AKT1 mutations in formalin-fixed, paraffin-embedded tissues. Arch. Pathol. Lab. Med. from Roche and Novartis and honoraria from Pfizer. M.D.W. had paid consulting relationships with
136, 983–992 (2012). GeneCentric Cancer Therapeutics Innovation Group. Data and materials availability: Segmented
57. S. Querings, J. Altmüller, S. Ansén, T. Zander, D. Seidel, F. Gabler, M. Peifer, E. Markert, copy number data, gene expression data from expression arrays and RNAseq, and binary
K. Stemshorn, B. Timmermann, B. Saal, S. Klose, K. Ernestus, M. Scheffler, W. Engel-Riedel, sequence alignment data of 300–base pair regions around all somatic mutations that were identified
E. Stoelben, E. Brambilla, J. Wolf, P. Nürnberg, R. K. Thomas, Benchmarking of mutation di- from whole-exome sequencing data are available at http://www.uni-koeln.de/med-fak/clcgp/. The
agnostics in clinical lung cancer specimens. PLoS One 6, e19601 (2011). data are also available through ArrayExpress at accession number E-MTAB-1999. Author contribu-
58. F. Göke, S. Perner, Translational research and diagnostics in lung cancer. Pathologe 33, tions: The CLCGP and NGM Initiatives contributed collectively to this study. The Biospecimens and
(Suppl. 2), 269–272 (2012). Data Source Sites provided biospecimens or data for further analysis. Data were generated and
59. H. U. Schildhaus, L. C. Heukamp, S. Merkelbach-Bruse, K. Riesner, K. Schmitz, E. Binot, E. Paggen, analyzed by the biospecimen core resources and data analysis team led by D.S., T.Z., L.C.H., M.P.,
K. Albus, W. Schulte, Y. D. Ko, A. Schlesinger, S. Ansén, W. Engel-Riedel, M. Brockmann, M. Serke, M.B., R.B., J.W., and R.K.T.
U. Gerigk, S. Huss, F. Göke, S. Perner, K. Hekmat, K. F. Frank, M. Reiser, R. Schnell, M. Bos,
C. Mattonet, M. Sos, E. Stoelben, J. Wolf, T. Zander, R. Buettner, Definition of a fluorescence in-situ
hybridization score identifies high- and low-level FGFR1 amplification types in squamous cell Data analysis team: Danila Seidel,1,2* Thomas Zander,1,3,52* Lukas C. Heukamp,8,52*
lung cancer. Mod. Pathol. 25, 1473–1480 (2012). Martin Peifer,1* Marc Bos,3,52* Lynnette Fernández-Cuesta,1 Frauke Leenders,1,2 Xin Lu,1
60. S. Perner, P. L. Wagner, F. Demichelis, R. Mehra, C. J. Lafargue, B. J. Moss, S. Arbogast, Sascha Ansén,3 Masyar Gardizi,3,52 Chau Nguyen,5,11 Johannes Berg,5 Prudence Russell,46
A. Soltermann, W. Weder, T. J. Giordano, D. G. Beer, D. S. Rickman, A. M. Chinnaiyan, H. Moch, Zoe Wainer,45 Hans-Ulrich Schildhaus,8,52,76 Toni-Maree Rogers,13 Benjamin Solomon,44
M. A. Rubin, EML4-ALK fusion lung cancer: A rare acquired event. Neoplasia 10, 298–302 William Pao,38 Scott L. Carter,42 Gad Getz,42 D. Neil Hayes,51 Matthew D. Wilkerson,51,77
(2008). Erik Thunnissen,24 William D. Travis,26 Sven Perner,14 Gavin Wright,45,78 Elisabeth Brambilla,36,37
61. S. Perner, F. Demichelis, R. Beroukhim, F. H. Schmidt, J. M. Mosquera, S. Setlur, J. Tchinda, Reinhard Büttner,2,8,52 Jürgen Wolf,2,3,52 and Roman K. Thomas1,2,8,52
S. A. Tomlins, M. D. Hofer, K. G. Pienta, R. Kuefer, R. Vessella, X. W. Sun, M. Meyerson, C. Lee, Biospecimens core resources: Franziska Gabler,2,4 Ines Wilkening,4 Christian Müller,1
W. R. Sellers, A. M. Chinnaiyan, M. A. Rubin, TMPRSS2:ERG fusion-associated deletions provide Ilona Dahmen,1 Roopika Menon,14 Katharina König,8,52 Kerstin Albus,8,52 Sabine Merkelbach-Bruse,8,52
insight into the heterogeneity of prostate cancer. Cancer Res. 66, 8337–8341 (2006). Jana Fassunke,8,52 Katja Schmitz,8,52 Helen Kuenstlinger,8,52 Michaela A. Kleine,8,52 Elke Binot,8,52
62. A. Subramanian, P. Tamayo, V. K. Mootha, S. Mukherjee, B. L. Ebert, M. A. Gillette, A. Paulovich, Silvia Querings,2,4 Janine Altmüller,6,10 Ingelore Bäßmann,6 Peter Nürnberg,6,9,10 Peter M. Schneider,12
S. L. Pomeroy, T. R. Golub, E. S. Lander, J. P. Mesirov, Gene set enrichment analysis: A knowledge- and Magdalena Bogus12
based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. U.S.A.
Pathology committee: Reinhard Büttner,2,8 Sven Perner, 14 Prudence Russell, 46
Erik Thunnissen,24 William D. Travis,26 and Elisabeth Brambilla36,37
102, 15545–15550 (2005).
Biospecimens and data source sites: Alex Soltermann,33 Holger Moch,33 Odd Terje
Brustugun,31,32 Steinar Solberg,30 Marius Lund-Iversen,73 Åslaug Helland,31,32 Thomas Muley,18,48
Acknowledgments: We thank W. Vogel, C. Beschorner, C. Becker, B. Pinther, and P. Löhrer for
Hans Hoffmann,18 Philipp A. Schnabel,17,48 Yuan Chen,15 Harry Groen,22 Wim Timens,23
their technical assistance and M. Knowles for providing the FGFR3b pBabe plasmids. We also Hannie Sietsma,23 Joachim H. Clement,71 Walter Weder,33 Jörg Sänger,19 Erich Stoelben,7
thank the regional computing center of the University of Cologne (Regional Computing Cen- Corinna Ludwig,7 Walburga Engel-Riedel,7 Egbert Smit,24 Daniëlle A. M. Heideman,24
tre) for providing the CPU time on the German Research Foundation (DFG)–funded super- Peter J. F. Snijders,24 Lucia Nogova,3,52 Martin L. Sos,3,52,53 Christian Mattonet,3,52 Karin Töpelt,3,52
computer “CHEOPS” as well as for support. The Victorian Cancer Biobank stored and Matthias Scheffler,3,52 Eray Goekkurt,52,55 Rainer Kappes,52,56,62 Stefan Krüger,52,56 Kato Kambartel,52,57
processed the Australian biospecimens used in this study. Funding: This work was Dirk Behringer,52,58 Wolfgang Schulte,52,59 Wolfgang Galetke,52,60 Winfried Randerath,52,61
supported by the EU-Framework Programme CURELUNG (HEALTH-F2-2010-258677 to R.K.T., Matthias Heldwein,52,63 Andreas Schlesinger,52,64 Monika Serke,52,65 Khosro Hekmat,52,63
R.B., and J.W.), by the Deutsche Forschungsgemeinschaft through TH1386/3-1 (to R.K.T. and M.L.S.) Konrad F. Frank,52,66 Roland Schnell,52,67 Marcel Reiser,52,68 Ali-Nuri Hünerlitürkoglu,52,69
and through SFB832 (TP6 to R.K.T. and J.W.; TP5 to L.C.H.) and SB680 (to J.B.), by the German Stephan Schmitz,52,70 Lisa Meffert,52,40 Yon-Dschun Ko,52,40 Markus Litt-Lampe,52,72 Ulrich Gerigk,52,41
Ministry of Science and Education as part of the NGFNplus program (grant 01GS08100 to R.K.T. Rainer Fricke,54 Benjamin Besse,36 Christian Brambilla,37 Sylvie Lantuejoul,36,75 Philippe Lorimier,36
and J.W.; grant 01GS08101 to P.N.), by the Deutsche Krebshilfe as part of the Oncology Centers Denis Moro-Sibilot,37 Federico Cappuzzo,27 Claudia Ligorio,28 Stefania Damiani,28 John K. Field,29
of Excellence funding program (to R.K.T., R.B., and J.W.) and SyBaCol (to J.B.), by the Max Planck Russell Hyde,29 Pierre Validire,35 Philippe Girard,35 Lucia A. Muscarella,47 Vito M. Fazio,47,74
Society, by the Behrens-Weise Foundation (M.I.F.A.NEUR8061 to R.K.T.), by Stand Up To Michael Hallek,2,3 Jean-Charles Soria,34 Scott L. Carter,42 Gad Getz,42 D. Neil Hayes,51
Cancer–American Association for Cancer Research Innovative Research Grant (SU2C-AACR- Matthew D. Wilkerson,51,77 Viktor Achter,50 and Ulrich Lang49,50
IR60109 to W.P. and R.K.T.), by the Roy Castle Lung Cancer Foundation UK (to J.K.F.), and by
Writing committee: Danila Seidel,1,2* Thomas Zander,1,3,52* Lukas C. Heukamp,8,52*
Martin Peifer,1* Marc Bos,3,52* William Pao,38 William D. Travis,26 Elisabeth Brambilla,36,37
an anonymous foundation (to R.K.T.). T.M. was supported by 82DZL00402 (DZL biobank plat-
Reinhard Büttner,2,8,52 Jürgen Wolf,2,3,52 and Roman K Thomas1,2,8,52
form) from the German Centre for Lung Research. Funding of supercomputer CHEOPS was
*These authors contributed equally to this work.
provided by the DFG and the Ministry of Research of the State of North Rhine–Westphalia.
Study leaders: Reinhard Büttner,2,8,52 Jürgen Wolf,2,3,52 and Roman K. Thomas1,2,8,52
Competing interests: R.K.T. is a founder and shareholder of Blackfield AG. R.K.T. received 1
Department of Translational Genomics, Center of Integrated Oncology Köln-Bonn,
consulting and lecture fees (Sanofi-Aventis, Merck, Roche, Lilly, Boehringer Ingelheim, AstraZeneca, University of Cologne, 50931 Cologne, Germany. 2Laboratory of Translational Cancer
Atlas-Biolabs, Daiichi-Sankyo, and Blackfield AG) as well as research support (Merck, EOS, and Genomics, Center of Integrated Oncology Köln-Bonn, University of Cologne, 50937
AstraZeneca). R.B. is a cofounder and owner of Targos Molecular Diagnostics and received Cologne, Germany. 3Department I of Internal Medicine, Center of Integrated Oncology
honoraria for consulting and lecturing from AstraZeneca, Boehringer Ingelheim, Merck, Roche, Köln-Bonn, University of Cologne, 50937 Cologne, Germany. 4Max Planck Institute for
Novartis, Lilly, Qiagen, and Pfizer. J.W. received consulting and lecture fees from Roche, Novartis, Neurological Research with Klaus-Joachim-Zülch Laboratories of the Max Planck Society
Boehringer Ingelheim, AstraZeneca, Bayer, Lilly, Merck, and Amgen and research support from and the Medical Faculty of the University of Cologne, 50931 Cologne, Germany. 5Insti-

www.ScienceTranslationalMedicine.org 30 October 2013 Vol 5 Issue 209 209ra153 12


RESEARCH ARTICLE

tute for Theoretical Physics, University of Cologne, 50937 Cologne, Germany. 6Cologne 3065 Victoria, Australia. 46Department of Pathology, St. Vincent’s Hospital, Melbourne,
Center for Genomics, University of Cologne, 50931 Cologne, Germany. 7Thoracic Surgery, 3065 Victoria, Australia. 47Laboratory of Oncology, Istituto di Ricovero e Cura a Carattere
Lungenklinik Merheim, Kliniken der Stadt Köln gGmbH, 51109 Cologne, Germany. 8Institute Scientifico Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy. 48Translational
of Pathology, Center of Integrated Oncology Köln-Bonn, University of Cologne, 50937 Lung Research Center Heidelberg, Member of the German Center for Lung Research,
Cologne, Germany. 9Center for Molecular Medicine Cologne, University of Cologne, Heidelberg, Germany. 49Department of Informatics, University of Cologne, 50931
Cologne, Germany. 10Cologne Excellence Cluster on Cellular Stress Responses in Aging- Cologne, Germany. 50Computing Center, University of Cologne, 50931 Cologne, Germany.
Associated Diseases, University of Cologne, Cologne, Germany. 11Bonn-Cologne Graduate 51
Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill,
School of Physics and Astronomy, 53115 Bonn, Germany. 12Institute of Legal Medicine, Chapel Hill, NC, USA. 52Network Genomic Medicine, University Hospital Cologne, Center
University of Cologne, 50823 Cologne, Germany. 13Department of Pathology, Peter of Integrated Oncology Köln Bonn, 50937 Cologne, Germany. 53Howard Hughes Medical
MacCallum Cancer Centre, Melbourne, 3002 Victoria, Australia. 14Department of Prostate Institute, Department of Cellular and Molecular Pharmacology, University of California, San
Cancer Research at the Institute of Pathology, University Hospital of Bonn, 53127 Bonn, Francisco, San Francisco, CA 94158, USA. 54Epidemiologisches Krebsregister NRW gGmbH,
Germany. 15Institute of Pathology, Jena University Hospital, Friedrich-Schiller-University, 48149 Münster, Germany. 55Hämatologisch-Onkologische Praxis Eppendorf, 20249
07743 Jena, Germany. 16Technical University Dortmund, Department of Chemical Biol- Hamburg, Germany. 56Klinik für Pneumologie, Florence Nightingale Krankenhaus, 40489
ogy, 44227 Dortmund, Germany. 17Institute of Pathology, University of Heidelberg, 69120 Düsseldorf, Germany. 57Lungenklinik, Krankenhaus Bethanien Moers, 47441 Moers, Germany.
Heidelberg, Germany. 18Department of Thoracic Surgery, Thoraxklinik am Universitätsklinikum 58
Klinik für Hämatologie, Onkologie und Palliativmedizin, Augusta Kliniken Bochum, 44791
Heidelberg, 69126 Heidelberg, Germany. 19Institute for Pathology Bad Berka, 99438 Bochum, Lungenkrebszentrum Herne-Bochum, Germany. 59Abteilung für Pneumologie,
Bad Berka, Germany. 20Department for Internal Medicine II, Jena University Hospital, Malteser Krankenhaus, 53123 Bonn, Germany. 60Klinik für Innere Medizin, Krankenhaus
Friedrich-Schiller University, 07740 Jena, Germany. 21Center for Medical Genetics, Ghent der Augustinerinnen, 50678 Köln, Germany. 61Klinik für Pneumologie, Krankenhaus
University, 9000 Ghent, Belgium. 22Department of Pulmonary Diseases, University of Bethanien Solingen, 42669 Solingen, Germany. 62Schwerpunktpraxis für Lungen- und
Groningen, University Medical Centre Groningen, 9713 GZ Groningen, the Netherlands. Bronchialheilkunde, Allergologie, Friedrichstr. 33-35, 40217 Düsseldorf, Germany. 63Depart-
23
Department Pathology and Medical Biology, University of Groningen, University Med- ment of Cardiothoracic Surgery, University Hospital Cologne, Center of Integrated Oncol-
ical Center Groningen, 9713 GZ Groningen, the Netherlands. 24Department of Pathology, ogy Köln Bonn, 50924 Cologne, Germany. 64Abteilung für Pneumologie, Evangelisches
VU University Medical Center Amsterdam, 1007 MB Amsterdam, the Netherlands. 25De- Krankenhaus Kalk, 51103 Köln, Germany. 65Abteilung Pneumologie III/Thorakale Onkologie,
partment of Pulmonary Diseases, VU University Medical Center Amsterdam, 1007 MB Lungenklinik Hemer, 58675 Hemer, Germany. 66Department III of Internal Medicine, Uni-
Amsterdam, the Netherlands. 26Department of Pathology, Memorial Sloan-Kettering versity Hospital Cologne, Center of Integrated Oncology Köln Bonn, 50924 Cologne,
Cancer Center, New York, NY 10065, USA. 27Department of Medical Oncology, Ospedale Germany. 67PIOH Frechen, Kölner Straße 9, 50226 Frechen, Germany. 68PIOH Köln, Richard
Civile, 57100 Livorno, Italy. 28Department of Experimental, Diagnostic and Specialty Med- Wagner Straße 13-17, 50674 Köln, Germany. 69Lukaskrankenhaus Neuss, Innere Medizin II,
icine, University of Bologna, 40138 Bologna, Italy. 29Roy Castle Lung Cancer Research 41464 Neuss, Germany. 70Gemeinschaftspraxis für Hämatologie und Onkologie, Sachsenring
Programme, The University of Liverpool Cancer Research Centre, University of Liverpool 69, 50677 Köln, Germany. 71Department Hematology and Oncology, Jena University Hospital,
Cancer Research Centre, Department of Molecular and Clinical Cancer Medicine, Institute Friedrich-Schiller University, 07747 Jena, Germany. 72Innere Medizin II Gastroenterologie/
of Translational Medicine, The University of Liverpool, Liverpool L3 9TA, UK. 30Department Pneumologie/Onkologie, Marienhospital Brühl, 50321 Brühl, Germany. 73Department
of Thoracic Surgery, Rikshospitalet, Oslo University Hospital, N-0027 Oslo, Norway. 31Insti- of Pathology, Norwegian Radium Hospital, Oslo University Hospital, N-0310 Oslo, Norway.
74
tute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0424 Oslo, Norway. University Campus Bio-Medico of Roma, Center for Integrated Research, Laboratory for
32
Department of Oncology, Norwegian Radium Hospital, Oslo University Hospital, N-0310 Molecular Medicine and Biotechnology, 00128 Rome, Italy. 75Université Joseph Fourier-
Oslo, Norway. 33Institute for Surgical Pathology, University Hospital Zurich, 8091 Zurich, INSERM U 823 Institut Albert Bonniot, 38700 La Tronche, France. 76Institute of Pathology,
Switzerland. 34Phase I Unit–Department of Medicine, Institute Gustave Roussy, 94800 Villejuif, University Hospital Goettingen, Goettingen, Germany. 77Department of Genetics, Univer-
France. 35France Service d’Anatomie-Pathologie, Institut Mutualiste Montsouris, 75014 Paris, sity of North Carolina at Chapel Hill, Chapel Hill, NC, USA. 78Division of Cancer Surgery,
France. 36Department of Cancer Medicine, Institute Gustave Roussy, 94800 Villejuif, France. Peter MacCallum Cancer Centre, East Melbourne, 3002 Victoria, Australia.
37
Pole de Cancérologie et Médecine Aigue Communautaire, CHU Grenoble, CS 10217,
38043 Grenoble, France and INSERM U823, Grenoble, France. 38Vanderbilt-Ingram Cancer Submitted 14 June 2013
Center, Nashville, TN, USA. 39Department of Genome Sciences, University of Washington, Accepted 17 October 2013
Seattle, WA 98195–5065, USA. 40Klinik für Internistische Onkologie Evangelische Kliniken Published 30 October 2013
Bonn, 53113 Bonn, Germany. 41Klinik für Thoraxchirurgie, Malteser Krankenhaus, 53123 Bonn, 10.1126/scitranslmed.3006802
Germany. 42The Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA. 43Depart-
ment of Genetics, Stanford University, Stanford, CA, USA. 44Department of Haematology and Citation: The Clinical Lung Cancer Genome Project (CLCGP) and Network Genomic Medicine
Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, 3002 Victoria, Australia. (NGM), A genomics-based classification of human lung tumors. Sci Transl. Med 5, 209ra153
45
The University of Melbourne Department of Surgery, St. Vincent’s Hospital, Melbourne, (2013).

Submitted 14 June 2013 10.1126/scitranslmed.3006802


Accepted 17 October 2013 Citation: The Clinical Lung Cancer Genome Project (CLCGP) and Network Genomic Medicine
Published 30 October 2013 (NGM), A genomics-based classification of human lung tumors. Sci. Transl. Med.Sci Transl. Med
10.1126/scitranslmed.3006802 5, 209ra153 (2013).

www.ScienceTranslationalMedicine.org 30 October 2013 Vol 5 Issue 209 209ra153 13

You might also like