SlideShare ist ein Scribd-Unternehmen logo
1 von 11
Downloaden Sie, um offline zu lesen
The new engl and jour nal of medicine
n engl j med nejm.org 1
The authors’ full names and academic
degrees are listed in the Appendix. Ad-
dress reprint requests to Dr. Linehan at
the Urologic Oncology Branch, National
Cancer Institute, Bldg. 10 CRC Rm.
1-5940, Bethesda, MD 20892-1107, or at
­wml@​­nih​.­gov.
*The authors are members of the Cancer
Genome Atlas Research Network, and
their names, affiliations, and roles are
listed in Supplementary Appendix 1,
available at NEJM.org.
This article was published on November 4,
2015, at NEJM.org.
DOI: 10.1056/NEJMoa1505917
Copyright © 2015 Massachusetts Medical Society.
BACKGROUND
Papillary renal-cell carcinoma, which accounts for 15 to 20% of renal-cell carcino-
mas, is a heterogeneous disease that consists of various types of renal cancer,
including tumors with indolent, multifocal presentation and solitary tumors with
an aggressive, highly lethal phenotype. Little is known about the genetic basis of
sporadic papillary renal-cell carcinoma, and no effective forms of therapy for ad-
vanced disease exist.
METHODS
We performed comprehensive molecular characterization of 161 primary papillary
renal-cell carcinomas, using whole-exome sequencing, copy-number analysis, mes-
senger RNA and microRNA sequencing, DNA-methylation analysis, and proteomic
analysis.
RESULTS
Type 1 and type 2 papillary renal-cell carcinomas were shown to be different types
of renal cancer characterized by specific genetic alterations, with type 2 further
classified into three individual subgroups on the basis of molecular differences
associated with patient survival. Type 1 tumors were associated with MET altera-
tions, whereas type 2 tumors were characterized by CDKN2A silencing, SETD2
mutations, TFE3 fusions, and increased expression of the NRF2–antioxidant re-
sponse element (ARE) pathway. A CpG island methylator phenotype (CIMP) was
observed in a distinct subgroup of type 2 papillary renal-cell carcinomas that was
characterized by poor survival and mutation of the gene encoding fumarate hydra-
tase (FH).
CONCLUSIONS
Type 1 and type 2 papillary renal-cell carcinomas were shown to be clinically and
biologically distinct. Alterations in the MET pathway were associated with type 1,
and activation of the NRF2-ARE pathway was associated with type 2; CDKN2A loss
and CIMP in type 2 conveyed a poor prognosis. Furthermore, type 2 papillary re-
nal-cell carcinoma consisted of at least three subtypes based on molecular and
phenotypic features. (Funded by the National Institutes of Health.)
ABSTR ACT
Comprehensive Molecular Characterization
of Papillary Renal-Cell Carcinoma
The Cancer Genome Atlas Research Network*
Original Article
The New England Journal of Medicine
Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission.
Copyright © 2015 Massachusetts Medical Society. All rights reserved.
n engl j med nejm.org2
The new engl and jour nal of medicine
K
idney cancer, or renal-cell carci-
noma, is not a single disease but is made
up of various types of cancer that are
characterized by different genetic drivers; each
type has distinct histologic features and a dis-
tinct clinical course and response to therapy.1,2
Papillary renal-cell carcinoma, which accounts
for 15 to 20% of kidney cancers, is a heteroge-
neous disease with histologic subtypes and varia-
tions in both disease progression and patient
outcomes. Papillary renal-cell carcinoma has two
main subtypes: type 1, which is often multifocal,
is characterized by papillae and tubular struc-
tures covered with small cells containing baso-
philic cytoplasm and small, uniform, oval nuclei,3
whereas type 2, which is more heterogeneous, is
characterized by papillae covered with large cells
containing eosinophilic cytoplasm and large,
spherical nuclei with prominent nucleoli.3,4
Al-
though in some patients papillary renal-cell
carcinoma is indolent, bilateral, and multifocal,
other patients present with solitary lesions that
have an aggressive clinical course. Little is
known about the genetic basis of the sporadic
forms of papillary renal-cell carcinoma, and there
are currently no effective forms of therapy for
patients with advanced disease.
Much of our knowledge of the genetic basis
of papillary renal-cell carcinoma has been based
on the study of the inherited form of the disease.
Hereditary papillary renal-cell carcinoma, a rare
disorder that is associated with an increased risk
of type 1 disease,4
is characterized by activating
germline mutations of MET.5
Somatic MET muta-
tions occur in 13 to 15% of nonhereditary papil-
lary renal-cell carcinomas.6,7
The hereditary leio-
myomatosis and renal-cell cancer syndrome,
which confers a predisposition to an aggressive
form of type 2 papillary renal-cell carcinoma,8,9
is caused by germline mutation of the gene en-
coding fumarate hydratase (FH), an enzyme of
the tricarboxylic acid cycle.10
These aggressive
tumors are characterized by increased oxidative
stress11
and activation of the NRF2–antioxidant
response element (ARE) pathway.12
Mutations in
the genes that regulate the NRF2-ARE pathway,
such as CUL3 and NFE2L2 (which encodes NRF2),
have also been observed in sporadic papillary
renal-cell carcinoma.13
We performed an integrative genomic analy-
sis of 161 papillary renal-cell carcinoma tumors
to provide molecular insights into tumor classi-
fication, inform clinical recommendations, and
suggest paths to the development of mechanisti-
cally based therapies.
Methods
Patients
Tumors were selected from 161 patients. Patho-
logical review was performed to classify the tu-
mors as type 1, type 2, or uncharacterized papil-
lary renal-cell carcinoma (see the Experimental
Procedures section in Supplementary Appendix 1,
available with the full text of this article at
NEJM.org). The clinical and genetic characteris-
tics of these patients are described in Supple-
mentary Appendix 2.
Analytic Platforms
We performed whole-exome sequencing and
analyses to determine copy number, microRNA
and messenger RNA (mRNA) expression, protein
expression, and DNA methylation at CpG sites
(Supplementary Appendix 3). Details of all the
analyses are available in the Experimental Proce-
dures section in Supplementary Appendix 1. All
data sets are available at the Cancer Genome At-
las data portal (https:/​­/​­tcga-data​.­nci​.­nih​.­gov/​­tcga).
Results
Histologic Subtyping
Pathological review of the 161 tumors identified
75 type 1 tumors, 60 type 2 tumors, and 26 tu-
mors that could not be classified as type 1 or
type 2. The type 1 tumors were predominantly
stage I, whereas the type 2 tumors were fre-
quently stage III or IV (Fig. S1 in Supplementary
Appendix 1); these findings were consistent with
those of previous studies.3,14
Role of Somatic Alterations in Molecular
Differences between Type 1 and Type 2 Tumors
Copy-Number Alterations
Single-nucleotide-polymorphism array–based
profiling of somatic copy-number alterations
revealed distinctive patterns across three main
tumor subgroups. One subgroup, composed
predominantly of type 1 and lower-grade tumors,
was defined by multiple chromosomal gains (of
at least one complete copy of the chromosome),
The New England Journal of Medicine
Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission.
Copyright © 2015 Massachusetts Medical Society. All rights reserved.
n engl j med nejm.org 3
Papillary Renal-Cell Carcinoma
including nearly universal gain of chromosomes
7 and 17 and less frequent gain of chromosomes
2, 3, 12, 16, and 20 (Fig. 1A, and Fig. S2 in
Supplementary Appendix 1). The other two sub-
groups were predominantly type 2 tumors; al-
though one of these subgroups had few copy-
number alterations, the other was characterized
by a high degree of aneuploidy with multiple
chromosomal losses, including frequent loss of
chromosome 9p, and was associated with poorer
survival (P<0.001) (Fig. 1A, and Fig. S2 in Sup-
plementary Appendix 1).
Whole-Exome Sequencing
Whole-exome sequencing identified 10,380 puta-
tive somatic mutations in 157 tumors with an
average of 1.45 nonsilent mutations per mega-
base (see the Experimental Procedures section in
Supplementary Appendix 1). An initial screen for
significantly mutated genes with q values of less
than 0.1 (q values range from 0.0 to 1.0), with
the use of MutSigCV, version 2.0, identified five
such genes (MET, SETD2, NF2, KDM6A, and
SMARCB1) that were recurrently mutated in pap-
illary renal-cell carcinoma, representing 24% of
cases (Fig. 1B). Further analysis, performed with
restriction of multiple hypothesis testing to
genes previously associated with cancer in the
PanCan21 data set,15
identified six additional
significantly mutated genes (FAT1, BAP1, PBRM1,
STAG2, NFE2L2, and TP53), with 36% of cases
showing mutation of at least one of these genes
(Fig. 1B). Mutation of these significantly mutat-
ed genes showed no evidence of subclonality
(Supplementary Appendix 4).
Hippo and Chromatin Modifier Pathways
Several significantly mutated genes in papillary
renal-cell carcinoma are components of well-
known cancer-associated pathways or complexes,
including NF2 in the Hippo signaling pathway,
SMARCB1 and PBRM1 in the SWI/SNF complex,
and SETD2, KDM6A, and BAP1 in several chroma-
tin modifier pathways. Assessment of genes in
these pathways (Supplementary Appendix 5)
showed a high number of mutations in both
type 1 and type 2 tumors involving the SWI/SNF
complex (20% and 27%, respectively), chromatin
modifier pathways (35% and 38%, respectively),
and the Hippo signaling pathway (3% and 10%,
respectively) (Fig. 1C).
TFE3 and TFEB Gene Fusions
Gene fusions involving TFE3 or TFEB have previ-
ously been associated with papillary renal-cell
carcinoma (reviewed in Kauffman et al.16
). We
identified gene fusions in 17 tumors (10.6%),
including 8 involving TFE3 or TFEB (Supplemen-
tary Appendix 6). Four of the TFE3 fusions in-
volved known fusion partners, PRCC and SFPQ,
and 2 involved novel fusion partners, RBM10 and
DVL2 (Fig. 1D). The tumors with TFE3 fusions
showed varying degrees of increased mRNA ex-
pression for known TFE3 transcriptional targets,
including CTSK, BIRC7, DIAPH1, and HIF1A (Fig.
S3 in Supplementary Appendix 1). The two TFEB
fusions involved novel fusion partners, COL21A1
and CADM2, with the COL21A1–TFEB fusion re-
sulting in a construct similar to the known
MALAT1–TFEB fusions16
and the TFEB–CADM2 fu-
sion resulting in a novel truncated version of
TFEB that had lost several microRNA binding
sites (Fig. 1D). The tumors with TFEB fusions
showed high mRNA expression of the TFEB tran-
scription factor and a known target gene, CTSK
(Fig. S4 in Supplementary Appendix 1). Seven of
the fusions involving TFE3 or TFEB were identi-
fied in the type 2 tumors (7 of 60 [12%]).
Alterations Specific to Types of Papillary
Renal-Cell Carcinoma
MET Mutation in Type 1 Tumors
We found mutation of MET in 17 tumors, includ-
ing germline mutation in 3 tumors. A total of 14
of the 17 MET mutations were in the tyrosine
kinase domain, and 13 of these mutations were
observed in type 1 tumors (17% of the 75 type 1
tumors) (Fig. 2A and 2B). In addition, an alter-
nate MET RNA transcript that replaces canonical
exons 1 and 2 with a novel exon 1 spliced to
canonical exon 3 (Fig. 2A) was identified in
8 tumors (4 type 1 tumors, 3 type 2 tumors, and
1 unclassified tumor). This isoform represented
the majority of transcripts in 2 tumors and a
fraction in the remaining 6 tumors and was re-
cently observed to produce a stable, shortened
protein in gastric-cancer cell lines (Fig. S5A in
Supplementary Appendix 1).19
Exons 1 and 2 of
MET encode the ligand-binding domain of he-
patocyte growth factor receptor; this isoform,
analogous to the epidermal growth factor recep-
tor variant III isoform,20
may result in ligand-
independent MET activation. In addition, gene
The New England Journal of Medicine
Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission.
Copyright © 2015 Massachusetts Medical Society. All rights reserved.
n engl j med nejm.org4
The new engl and jour nal of medicine
B
A
No.ofMutations
16
12
14
10
8
4
2
6
0
C
D
Missense
Splice site
Nonsense
Frame shift
In-frame indel
MET gene
Hippo pathway
NRF2 pathway
Chromatin modifiers
SWI/SNF complex
mTOR pathway
p53-related genes
Chr 7 gain
Type 1 PRCC
(N=75)
Type 2 PRCC
(N=60)
Unclassified
PRCC
(N=26)
Somatic
Mutation SMG (current study) No dataOther related geneOther pan-cancer SMG
1 2 3 4 5 6 7 8 9 10 11 12 13 15 17 19 22
Chromosome
Copy gain
Type 1 PRCC
Type 2 PRCC
Copy loss
M
ETFAT1SETD
2
N
F2KD
M
6ABAP1PBRM
1
SM
ARCB1N
FE2L2STAG
2TP53
All PRCC (N=157)
Type 1 PRCC (N=71)
Type 2 PRCC (N=60)
−log10 P value
0 2 4 6 8
q<0.10 for pan-cancer
SMGs
*
*
* *
Genome unstable,
Chr 9p deleted
Arm-level
alterations,
Chr 7 gain
Genome stable,
sporadic SCNAs,
few Chr 9p deletions
* * *
**
* *
†
†
†
†
†
q<0.05 for all genes†
MAD2L2
DVL2
DIX
RBM10
SFPQ
TFEB
microRNA binding sites
COL21A1
TFEB
3x PRCC
TFE3
TFE3
TFE3
TFE3
AD bHLH-LZ
AD bHLH-LZ
AD bHLH-LZ
AD bHLH-LZ
AD bHLH-LZ
AD bHLH-LZ
RRM1RRM2Q/E/P-Rich
1 4 5 6 78 9 10
2 3 4 5 6 78 9 102 3 4 5 61
1 2 3 4 5 3 4 5 6 78 9 10
1 2 3 4 56 78 9 10 5 6 78 9 101112 13 14151617
3' UTR
2
3 4 5 6 78 9 10
1
2
3 4 5 6 7 8 9
PRCC-associated
TFE3 fusions (4)
Additional
PRCC-associated
TFE3 fusions (2)
Additional
PRCC-associated
TFEB fusions (2)
CADM2
Mutation Type
The New England Journal of Medicine
Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission.
Copyright © 2015 Massachusetts Medical Society. All rights reserved.
n engl j med nejm.org 5
Papillary Renal-Cell Carcinoma
fusions involving MET were observed in 3 tumors
(Supplementary Appendix 6). Levels of MET
mRNA expression and of protein phosphoryla-
tion (pY1235) were significantly higher in type 1
tumors than in type 2 tumors (P<1×10−9
and
P=0.007, respectively, by t-test) (Fig. S5B in
Supplementary Appendix 1) — a finding poten-
tially driven in part by trisomy of chromosome 7
in type 1 tumors. Altered MET status (defined as
mutation, splice variant, or gene fusion) or in-
creased chromosome 7 copy number (which en-
codes MET but may also involve other genes) was
identified in 81% of type 1 papillary renal-cell
carcinomas. Analysis by means of Genomic
Identification of Significant Targets in Cancer
(GISTIC), version 2.0, determined that the loss of
1p36 observed in 18 papillary renal-cell carcino-
mas (11.2%) included the candidate tumor sup-
pressor ERRFI1, a negative regulator of EGFR
Figure 1 (facing page). Somatic Alterations in Papillary
Renal-Cell Carcinoma and Molecular Differences between
Type 1 and Type 2 Cancers.
Unsupervised clustering of DNA copy profiles of 161
papillary renal-cell carcinomas (PRCCs) (Panel A) re-
vealed three molecular subtypes, one of which was
highly enriched for type 1 tumors and the other two
for type 2 tumors. SCNA denotes somatic copy-number
alterations. Significantly mutated genes (SMGs) in
PRCC (Panel B) were determined by considering all
genes (q<0.1 [range, 0.0 to 1.0]) or focusing on the set
of 260 genes previously implicated in cancer by large-
scale, pan-cancer exome analyses15
(q<0.1). P values
were calculated with the MutSigCV algorithm, version
2.0. A pathway-centric view of gene mutations in PRCC
(Panel C) shows key pathways and genes implicated in
cancer, either in the current study or elsewhere.15
The
tumors were classified according to histologic type
(from left to right) and according to gene or pathway
altered (from top to bottom). Pathways and genes rep-
resented include MET, the Hippo pathway (NF2, SAV1,
and WWC1), the NRF2 pathway (NFE2L2, KEAP1, CUL3,
SIRT1, and FH), chromatin modification (CREBBP,
DOTL1, EHMT1/2, EP300, EZH1/2, KAT2A/B, KDM1A/B,
KDM4A/B, KDM5A/B/C, KDM6A/B, MLL1/2/3/4/5,
NSD1, SETD2, SMYD4, and SRCAP), the SWI/SNF
complex (ACTB, ACTL6A/B, ARID1A/B, ARID2, BCL6A/
B/C, BCL11A/B, BRD7/9, DPF1/2/3, PHF10, PBRM1,
SMARCA2/4, SMARCB1, SMARCC1/2, SMARCD1/2/3,
and SMARCE1), the mammalian target of rapamycin
(mTOR) pathway (MTOR, PIK3CA, PTEN, STK11, TSC1,
and TSC2), and the p53 pathway (ATM, CDKN1A,
CDKN2A, FBXW7, RB1, and TP53). Fusion gene analy-
sis (Panel D) identified TFE3 or TFEB fusions in eight
PRCC tumors, including two novel gene-fusion partners
for TFE3 (DVL2 and RBM10) and two novel gene-fusion
partners for TFEB (COL21A1 and CADM2). Schematic
versions of these fusions show the exons and functional
domains that are present in the different gene fusions
and the position of potential microRNA binding sites in
TFEB. The retained exons of TFE3 or TFEB are colored in
shades of blue. Thin regions represent noncoding se-
quence, thick regions represent the translated reading
frame, and white strips indicate that the region is no
longer to scale. AD denotes strong transcription acti-
vation domain, bHLH basic helix-loop-helix domain,
DIX dishevelled and axin domain, LZ leucine zipper
domain, MAD2L2 mitotic arrest deficient–like 2 inter-
action domain, and RRM RNA-recognition motif.
Figure 2. Alterations in Papillary Renal-Cell Carcinoma Involving the MET
Oncogene.
Panel A is a schematic representation of somatic mutations in MET, along
with germline variant H1112R, which was previously implicated in hereditary
papillary renal-cell carcinoma,17
and the novel RNA transcript variant of MET
lacking the canonical exons 1 and 2 but containing a novel exon 1 that splices
to the canonical exon 3. IPT denotes immunoglobulin-like, plexins, and
transcription factors, and PSI plexins, semaphorins, and integrins. Panel B
shows the crystal structure for the MET tyrosine kinase catalytic domain
(RCSB-PDB 3I5 N18
), on which are mapped the residues that are altered in
papillary renal-cell carcinoma. All numbering of amino acids is based on the
MET protein sequences.
Activation loop
Somatic mutation
observed in
unclassified PRCC
Somatic mutation
observed in type 1 PRCC
L296P
F1218
Catalytic loop
Y1248
ATP-
binding
regionV1110
H1112
V1088
D1246
S1254
M1268
p+1 Loop
I639L F970V
F1218IH1112Y/R
V1110I
V1088E/R
D1246H
Y1248H
S1254R
M1268T
Tyrosine kinase
catalytic domain
IPT domainSema domain PSI
domain
Germline mutation
observed in type 1 PRCC Somatic mutation
observed in
type 2 PRCC
Encoded by
alternate exon
(~65 aa)
A
B
The New England Journal of Medicine
Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission.
Copyright © 2015 Massachusetts Medical Society. All rights reserved.
n engl j med nejm.org6
The new engl and jour nal of medicine
(Fig. S6 in Supplementary Appendix 1). Dele-
tions of 1p36 co-occurred significantly with
gain of chromosome 7 and EGFR amplification
(P = 0.02 by Fisher’s exact test).
CDKN2A Mutation in Type 2 Tumors
Analysis by GISTIC, version 2.0, identified focal
loss of 9p21 in 13 papillary renal-cell carcino-
mas (8.1%), resulting in loss of CDKN2A (Fig. S7A
in Supplementary Appendix 1). We found muta-
tion or promoter hypermethylation of CDKN2A in
11 tumors (Fig. S7B in Supplementary Appendix
1), including 3 of the tumors with focal loss of
9p21, resulting in 21 tumors (13.0%) defined as
having CDKN2A alteration (Fig. S7C in Supple-
mentary Appendix 1). CDKN2A alteration was
strongly associated with type 2 histologic fea-
tures, with 25% of type 2 tumors (15 of 60)
showing alterations. CDKN2A-altered tumors
showed both increased levels of phosphorylated
retinoblastoma protein (Rb) and increased ex-
pression of cell-cycle–related genes, findings
consistent with the predicted consequences of
CDKN2A loss (Fig. S7D in Supplementary Appen-
dix 1). In a univariate analysis, patients with
CDKN2A-altered tumors had a significantly lower
rate of overall survival than those without CDKN2A-
altered tumors (P<1×10−10
) (Fig. S7E in Supple-
mentary Appendix 1). The findings were similar
when the analysis was limited to patients with
type 2 tumors (P<0.001) (Fig. S7F in Supplemen-
tary Appendix 1). In addition, increased expres-
sion of microRNA miR-10b-5p correlated with de-
creased expression of its target, CDKN2A (Fig. S8
in Supplementary Appendix 1).
SETD2, BAP1, and PBRM1 Mutation in Type 2
Tumors
Type 2 tumors were associated with mutations
in the chromatin-modifying genes SETD2, BAP1,
and PBRM1, which are frequently mutated in
clear-cell kidney tumors in combination with
loss of chromosome 3p.21
Mutations of BAP1 and
PBRM1 were mutually exclusive, but PBRM1 muta-
tions were frequently concurrent with SETD2
mutations (Fig. S9 in Supplementary Appendix
1). Although loss of chromosome 3p was also
associated with type 2 papillary renal-cell carci-
noma, only 3 of 13 type 2 tumors with SETD2,
BAP1, or PBRM1 mutation showed such loss, and
no promoter hypermethylation was observed
(Fig. S9 in Supplementary Appendix 1).
CpG Island Methylator Phenotype (CIMP) in Type 2
Tumors
Nine tumors (5.6%) had increased DNA methyla-
tion at loci that were unmethylated in matched
normal tissue. This represents a novel kidney-
associated CIMP22
that included universal hyper-
methylation of the CDKN2A promoter (Fig. 3A).
Eight of the nine tumors were type 2 papillary
renal-cell carcinomas. In five tumors, we found
germline or somatic mutation of FH (56%). We
found decreased expression of FH mRNA and
increased expression of genes associated with
cell-cycle progression and response to hypoxia
in all nine tumors (Fig. 3A). Patients with CIMP-
associated tumors were younger at the time of
presentation and had a lower probability of over-
all survival than other patients with papillary
renal-cell carcinoma (Fig. 3B). Fumarate hydra-
tase–deficient type 2 tumors in patients with the
hereditary leiomyomatosis and renal-cell cancer
syndrome are characterized by a Warburg-like
metabolic shift to glycolysis-dependent metabo-
lism and an increased expression of hypoxia-
related genes.25,26
Similarly, CIMP-associated
tumors showed increased expression of key genes
involved in glycolysis (HK1, LDHA, and PDK1),
the pentose phosphate pathway (G6PD), and fat-
ty-acid synthesis (FASN) (Fig. 3C, and Fig. S10 in
Supplementary Appendix 1). In addition, there
was decreased expression of the majority of
genes involved in the Krebs cycle and the ade-
nosine monophosphate–activated protein kinase
(AMPK) complex, a suppressor of fatty-acid syn-
thesis (Fig. 3D). Data on the expression of pro-
teins G6PD, FASN, and AMPK correlated with
the data on mRNA expression (Fig. 3D).
Identification of Papillary Renal-Cell
Carcinoma Subgroups by Multiplatform
Analysis
Cluster-of-Clusters Analysis
As was the case with the copy-number analysis
and DNA-methylation analysis, the profiles of
mRNA expression and microRNA expression
and the data on protein expression clustered the
cases of papillary renal-cell carcinoma into sep-
arate groups with distinct overall outcomes (Fig.
S11, S12, and S13 in Supplementary Appendix
1). The five data types were combined to per-
form a cluster-of-clusters analysis27,28
that identi-
fied four tumor clusters: C1 (enriched for type 1
tumors), C2a and C2b (both enriched for type 2
The New England Journal of Medicine
Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission.
Copyright © 2015 Massachusetts Medical Society. All rights reserved.
n engl j med nejm.org 7
Papillary Renal-Cell Carcinoma
Figure 3. A Subgroup of Papillary Renal-Cell Carcinoma That Manifests a CpG Island Methylator Phenotype (CIMP).
As depicted in Panel A, molecular subtyping by means of a DNA methylation platform revealed three subtypes of papillary renal-cell car-
cinoma (PRCC), one of which showed widespread DNA hypermethylation patterns characteristic of CIMP-associated tumors (the other
subtypes are identified as cluster 1 and cluster 2). Corresponding data tracks highlight molecular features associated with CIMP tumors
(nine cases), including CDKN2A silencing, germline or somatic mutations of FH, type 2 histologic status, and expression of both cell-
cycle–related genes23
and hypoxia-related genes.24
Panel B shows differences in patient age and overall survival among the three sub-
types. Data on survival were not available for two patients in the cluster 2 group. Panel C shows differential messenger RNA (mRNA)
expression patterns for key genes involved in metabolism among CIMP-associated PRCC, type 1 PRCC, non–CIMP-associated type 2
PRCC, and normal kidney. Panel D shows differential expression patterns of CIMP-associated tumors versus type 1 tumors in metabo-
lism-related pathways, with a focus on gene-expression and protein-expression patterns previously associated with Warburg-like effects
in kidney cancer.21
P values were calculated with the use of a t-test.
C D
A B
ProbabilityofOverall
Survival
1.0
0.8
0.6
0.4
0.2
0.0
0.9
0.7
0.5
0.3
0.1
0 50 100 150 200 250
Months
AgeofPatient(yr)
80
60
40
0
70
50
30
Subtype Based on DNA Methylation
CIMP Cluster 1 Cluster 2
PRCC (N=161 cases)
Tumor type
Normal kidney
(N=50)
CIM
P
Cluster 1 Cluster 2
Subtype Based on
DNA Methylation
343GenomicLoci
CDKN2A silencing
CDKN2A loss
CDKN2A mutation
FH mutation
DNAMethylation(βvalue)
High
Low
0
0.5
1
CIMP (N=9)
Cluster 1 (N=30)
Cluster 2 (N=120)
Censored
P=1×10−16 for overall
comparison by
log-rank test
FH expr.
Cell cycle expr.
Hypoxia expr.
HighLow
CIMP-
Associated
PRCC
(N=9)
Other Type 2
PRCC
(N=52)
Type 1 PRCC
(N=75)
Normal
Kidney
(N=30)
HK1
G6PD
LDHA
PDK1
FASN
PRKAB1
ACO2
OGDH
SUCLA2
SDHA
FH
MDH2
mRNA Expression
Somatic
mutation
Germline
mutation
Higher
expression
Lower
expression
Type 1 Type 2
Glycolysis
Pentose
phosphate
pathway
Fatty acids and lipids
FASNPDK1
G6PD
HK1
Glucose-
6-phosphate
PRKAB1
PRKAA2
PyruvateLactate
LDHA
OGDH
IDH2
ACO2
Glutamate
Acetyl CoA
Citrate
Isocitrate
α-ketoglutarate
Succinyl CoA
Krebs cycle
Succinate
Malate
Fumarate
FH
MDH2
Oxaloacetate
SDHA SUCLA2
G6PD
Glucose
FASN
ProteinRNA
>1.5×
>2×
>1.2×
Higher in CIMP (P<0.01)
Lower in CIMP (P<0.01)
CIMP-Associated PRCC
vs Type 1 PRCC
AMPK
The New England Journal of Medicine
Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission.
Copyright © 2015 Massachusetts Medical Society. All rights reserved.
n engl j med nejm.org8
The new engl and jour nal of medicine
tumors), and C2c (consisting solely of CIMP-
associated papillary renal-cell carcinoma) (Fig. 4A).
Cluster C1 was predominantly type 1 papil-
lary renal-cell carcinoma and was strongly as-
sociated with gain of chromosome 7, MET muta-
tion, mRNA cluster 1, and an early stage of
tumor development (stage I or II) (Fig. 4A, and
Fig. S14 in Supplementary Appendix 1). Cluster
C2a was predominantly type 2 papillary renal-
cell carcinoma and was associated with an early
stage of tumor development and DNA methyla-
tion cluster 2. Cluster C2b consisted exclusively
of type 2 and unclassified papillary renal-cell
carcinomas and was associated with DNA meth-
ylation cluster 1, a later stage of tumor develop-
ment (stage III or IV), and mutation of SETD2.
The CIMP-associated tumor subtype that was
observed previously in DNA-methylation analysis
was preserved as cluster C2c. Patients with clus-
ter C1 or cluster C2a tumors had the highest
probability of overall survival, patients with
cluster C2b tumors had a lower probability, and
patients with cluster C2c tumors had the lowest
probability (Fig. 4B).
NRF2-ARE Pathway in Type 2 Tumors
Pathway analysis was performed to compare the
microRNA and mRNA signatures of type 1 tu-
mors with those of type 2 tumors (Fig. S15, S16,
and S17 in Supplementary Appendix 1, and Sup-
plementary Appendixes 7, 8, and 9), and data on
mRNA expression highlighted the NRF2-ARE
pathway as a distinguishing feature of type 2
tumors (Fig. S17A in Supplementary Appendix 1).
Expression of NQO1, a gene activated by the
NRF2-ARE pathway,29
was lowest in cluster C1,
intermediate in clusters C2a and C2b, and high-
est in the CIMP cluster C2c (P = 1×10−18
by analy-
sis of variance) (Fig. S18A in Supplementary
Appendix 1), and increased NQO1 expression was
associated with decreased survival (P = 0.001)
(Fig. S18C in Supplementary Appendix 1). These
findings are consistent with those of studies
showing increased activation of the NRF2-ARE
pathway in type 2 tumors and mutations in
Figure 4. Multiplatform-Based Subtype Discovery
in Papillary Renal-Cell Carcinoma.
As shown in Panel A, integration of subtype classifica-
tions from five genomic data platforms with the use of
a cluster-of-clusters analysis identified four major groups
of papillary renal-cell carcinoma: C1 (enriched for type 1),
C2a and C2b (enriched for type 2), and C2c (represent-
ing the CIMP-associated papillary renal-cell carcinomas).
The heat map (center of panel) displays the subtypes
defined independently by DNA methylation (pink),
chromosomal copy number (black), microRNA expres-
sion (blue), mRNA expression (red), and protein (RPPA)
expression (green); samples with missing data for pro-
tein expression are shown in gray. Clinical features as-
sociated with the multiplatform-based subtypes are
also shown. Panel B shows differences in overall sur-
vival according to subtype. Data on survival were not
available for two patients in the C1 group.
Months
B
A
Histologic Type
Stage of Tumor
No. of Cases 9 22 95 35
RPPA 2
microRNA 1
Copy number 2
mRNA 1
DNA methylation 2
mRNA 2
DNA methylation 1
Copy number 1
CIMP
RPPA 3
microRNA 2
microRNA 3
Copy number 3
RPPA 1
microRNA 4
mRNA 3
C2c
(CIMP)
C2b C1 C2a
ProbabilityofOverallSurvival
1.0
0.8
0.9
0.7
0.6
0.4
0.3
0.1
0.5
0.2
0.0
0 50 100 150 200 250
C2b (N=22)
Censored
C1 (N=93)
C2a (N=35)
C2c (N=9)
Histologic Type
Type 1 PRCC Type 2 PRCC
Unclassified PRCC
Stage of Tumor
II
III
I
IV
P=1×10−16 for overall comparison
by log-rank test
C1 vs. C2a, P=0.26
C1 vs. C2b, P<0.001
C1 vs. C2c, P=1×10−22
The New England Journal of Medicine
Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission.
Copyright © 2015 Massachusetts Medical Society. All rights reserved.
n engl j med nejm.org 9
Papillary Renal-Cell Carcinoma
NRF2-ARE pathway genes (NFE2L2, CUL3, KEAP1,
and SIRT1).12,13
Four NFE2L2 (NRF2) mutations in
known activating hotspots were identified, as
well as mutations in both CUL3 (five mutations)
and KEAP1 (one). These mutations in NFE2L2,
CUL3, and KEAP1 correlated with high levels of
NQO1 expression (P<1×10−6
by t-test) but did not
solely account for the observed differences in
NQO1 expression among subtypes (Fig. S18A in
Supplementary Appendix 1).
Integrated Analysis of Low-Frequency
Candidate Driver Mutations
Some tumors (most relatively small) lacked high-
confidence candidate cancer-driving events. Man-
ual pathway analysis identified candidate driver
mutations in known cancer-associated genes,
such as PTEN, NRAS, KRAS, TP53, TSC2, and
those in the MLL and ARID families, in an addi-
tional 27% of the cases (Fig. S19A in Supplemen-
tary Appendix 1, and Supplementary Appendix
10). For the remaining 37 tumors (23%), low-
frequency somatic events in genes identified by
HotNet2 analysis (Fig. S19 in Supplementary
Appendix 1) or associated with cancer in either
the PanCan21 data set15
or the Catalogue of So-
matic Mutations in Cancer database were pro-
posed as potential drivers and are listed in Sup-
plementary Appendix 10. In comparison with
the tumors with candidate cancer-driving events,
the remaining 37 papillary renal-cell carcinomas
showed a higher percentage of type 1 tumors (26
of 37 [70%]) (P = 0.001 by Fisher’s exact test), and
most (21 of 26 [81%]) showed a gain of chromo-
some 7, which includes MET. This gain of chro-
mosome 7, which is seen in a number of tumors
(e.g., Wilms’ tumor and papillary thyroid can-
cer), could be considered a driver event, but it
does not identify a specific driver. Although gain
of chromosome 7 was associated with increased
MET expression in papillary renal-cell carcinoma
(P<0.001 by two-factor analysis of variance) (Fig.
S20 in Supplementary Appendix 1), other poten-
tial driver genes on chromosome 7, such as EGFR,
could influence tumorigenesis.
Discussion
We used a comprehensive genomics approach to
characterize the biologic foundation of papillary
renal-cell carcinoma and found that type 1 and
type 2 papillary renal-cell carcinoma are dis-
tinctly different diseases and that type 2 papil-
lary renal-cell carcinoma is a heterogeneous dis-
ease with multiple distinct subgroups. Common
driver mutations among the different subtypes
were relatively rare, as had been observed in two
recent studies.7,30
Molecular and phenotypic dif-
ferences between type 1 and type 2 papillary
renal-cell carcinoma were reflected in individual
and combined analyses of various data plat-
forms. The usefulness of CDKN2A alterations as
an independent prognostic marker associated
with type 2 tumors requires validation. This study
suggests that gene fusions involving TFE3 or
TFEB are underappreciated in type 2 tumors in
adults and should be considered in any patient
with type 2 disease. Although papillary renal-
cell carcinomas with fusions involving TFE3 or
TFEB are generally considered to be diseases of
children and young adults,16
the mean age in our
study was 52 years, and we found tumors with
TFEB fusions in patients 64 and 71 years of age.
The most distinct of the three type 2 sub-
groups was the subgroup defined by the CIMP,
which was associated with the worst overall
survival. CIMP hypermethylation patterns have
been observed in a number of other cancer sub-
types, including glioblastoma,31
lung adenocar-
cinoma,32
and gastric adenocarcinoma.33
The
CIMP-associated tumors showed low levels of FH
mRNA expression, and five had germline or
somatic mutation of FH. Germline mutation of
FH has been observed in the aggressive type 2
tumor associated with the hereditary leiomyoma-
tosis and renal-cell cancer syndrome.9,34
In this
syndrome, the high levels of fumarate accumu-
lating from loss of fumarate hydratase enzyme
activity result in impaired function of enzymes
such as the TET family of enzymes, which play
a role in maintaining appropriate DNA methyla-
tion within the genome.35
The subgrouping of
type 2 tumors according to molecular features
and the presence of specific subsets of type 2
tumors, such as those with TFE3 fusions or
CIMP, suggest that substratification of type 2
papillary renal-cell carcinoma according to spe-
cific molecular markers may allow more accurate
diagnosis that could lead to the development of
mechanistic, disease-specific targeted therapies.
This classification of papillary renal-cell car-
cinoma could potentially have a substantial ef-
fect on clinical and therapeutic management and
on the design of clinical trials. Alteration of MET
or gain of chromosome 7 was observed in a
large percentage (81%) of type 1 tumors. Anti-
The New England Journal of Medicine
Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission.
Copyright © 2015 Massachusetts Medical Society. All rights reserved.
n engl j med nejm.org10
The new engl and jour nal of medicine
tumor activity of an agent targeting the MET
and VEGFR2 pathways has been shown in a
phase 2 trial involving patients with papillary
renal-cell carcinoma, with a particularly high
response rate among patients who had tumors
with MET mutations.36
Mutation of the Hippo
pathway tumor suppressor, NF2, was observed in
a number of papillary renal-cell carcinomas.
This pathway has been targeted in other cancers
with agents such as dasatinib, an inhibitor of
the YES1 kinase that interacts with the YAP tran-
scription factor that is up-regulated with Hippo
pathway dysregulation.37
The CIMP-associated
tumors showed a Warburg-like metabolic shift,
similar to that observed in fumarate hydratase–
deficient tumors in patients with the hereditary
leiomyomatosis and renal-cell cancer syn-
drome.11,25,26
A clinical trial targeting this meta-
bolic shift in papillary renal-cell carcinoma is
currently under way (ClinicalTrials.gov number,
NCT01130519). Increased expression of the
NRF2-ARE pathway has been observed in both
hereditary and sporadic type 2 papillary renal-
cell carcinomas.12
Immunohistochemical analy-
sis for NQO1 could provide a valuable marker of
activation of the NRF2-ARE pathway. Currently,
there is intense interest in the NRF2-ARE path-
way in cancer,38
and novel strategies have re-
cently been developed to target this pathway.39
The identification of altered genes and path-
ways provides a comprehensive foundation for
an understanding of the molecular basis of pap-
illary renal-cell carcinoma. This refined classifi-
cation more accurately reflects the genotypic
and phenotypic differences among the various
types of these tumors and may lead to more ap-
propriate clinical management and development
of more effective forms of therapy.
Supported by grants (U54 HG003273, U54 HG003067, U54
HG003079, U24 CA143799, U24 CA143835, U24 CA143840, U24
CA143843, U24 CA143845, U24 CA143848, U24 CA143858, U24
CA143866, U24 CA143867, U24 CA143882, U24 CA143883, U24
CA144025, and P30 CA016672) from the National Institutes of
Health.
Disclosure forms provided by the authors are available with
the full text of this article at NEJM.org.
Appendix
The authors’ full names and academic degrees are as follows: W. Marston Linehan, M.D., Paul T. Spellman, Ph.D., Christopher J.
Ricketts, Ph.D., Chad J. Creighton, Ph.D., Suzanne S. Fei, Ph.D., Caleb Davis, Ph.D., David A. Wheeler, Ph.D., Bradley A. Murray, Ph.D.,
Laura Schmidt, Ph.D., Cathy D. Vocke, Ph.D., Myron Peto, Ph.D., Abu Amar M. Al Mamun, Ph.D., Eve Shinbrot, Ph.D., Anurag Sethi,
Ph.D., Samira Brooks, B.S., W. Kimryn Rathmell, M.D., Ph.D., Angela N. Brooks, Ph.D., Katherine A. Hoadley, Ph.D., A. Gordon
Robertson, Ph.D., Denise Brooks, Ph.D., Reanne Bowlby, B.S., Sara Sadeghi, Ph.D., Hui Shen, Ph.D., Daniel J. Weisenberger, Ph.D.,
Moiz Bootwalla, M.S., Stephen B. Baylin, M.D., Peter W. Laird, Ph.D., Andrew D. Cherniack, Ph.D., Gordon Saksena, M.Eng., Scott
Haake, M.D., Jun Li, Ph.D., Han Liang, Ph.D., Yiling Lu, M.D., Gordon B. Mills, M.D., Ph.D., Rehan Akbani, Ph.D., Mark D.M. Leiser-
son, Ph.D., Benjamin J. Raphael, Ph.D., Pavana Anur, M.S., Donald Bottaro, Ph.D., Laurence Albiges, M.D., Ph.D., Nandita Barnabas,
Ph.D., Toni K. Choueiri, M.D., Bogdan Czerniak, M.D., Andrew K. Godwin, Ph.D., A. Ari Hakimi, M.D., Thai H. Ho, M.D., Ph.D.,
James Hsieh, M.D., Michael Ittmann, M.D., William Y. Kim, M.D., Bhavani Krishnan, Ph.D., Maria J. Merino, M.S., Kenna R. Mills
Shaw, Ph.D., Victor E. Reuter, M.D., Ed Reznik, Ph.D., Carl S. Shelley, D.Phil., Brian Shuch, M.D., Sabina Signoretti, M.D., Ramaprasad
Srinivasan, M.D., Ph.D., Pheroze Tamboli, M.D., George Thomas, M.D., Satish Tickoo, M.D., Kenneth Burnett, M.S., Daniel Crain,
M.B.A., Johanna Gardner, C.T.R., Kevin Lau, B.S., David Mallery, M.B.A., J.D., Scott Morris, P.S.M., Ph.D., Joseph D. Paulauskis, Ph.D.,
Robert J. Penny, M.D., Ph.D., Candace Shelton, C.C.R.A., C.C.D.M., W. Troy Shelton, M.S., P.M.P., Mark Sherman, Ph.D., Eric Thomp-
son, Ph.D., Peggy Yena, B.S., Melissa T. Avedon, B.S., Jay Bowen, M.S., Julie M. Gastier‑Foster, Ph.D., Mark Gerken, B.S., Kristen M.
Leraas, M.S., Tara M. Lichtenberg, B.A., Nilsa C. Ramirez, M.D., Tracie Santos, M.S., Lisa Wise, C.T.R., Erik Zmuda, Ph.D., John A.
Demchok, M.S., Ina Felau, B.S., Carolyn M. Hutter, Ph.D., Margi Sheth, B.S., Heidi J. Sofia, Ph.D., M.P.H., Roy Tarnuzzer, Ph.D., Zhin-
ing Wang, Ph.D., Liming Yang, Ph.D., Jean C. Zenklusen, Ph.D., Jiashan Zhang, B.A., Brenda Ayala, B.S., Julien Baboud, B.S., Sudha
Chudamani, M.Phil., Jia Liu, Ph.D., Laxmi Lolla, Rashi Naresh, M.S., Todd Pihl, Ph.D., Qiang Sun, M.S., Yunhu Wan, Ph.D., Ye Wu,
Ph.D., Adrian Ally, B.S., Miruna Balasundaram, B.S., Saianand Balu, M.S., Rameen Beroukhim, M.D., Ph.D., Tom Bodenheimer, M.S.,
Christian Buhay, B.S., Yaron S.N. Butterfield, B.S., Rebecca Carlsen, M.S., Scott L. Carter, Ph.D., Hsu Chao, D.V.M., Eric Chuah, B.S.,
Amanda Clarke, T.D., Kyle R. Covington, Ph.D., Mahmoud Dahdouli, B.Sc., Ninad Dewal, Ph.D., Noreen Dhalla, B.S., Harsha V. Dod-
dapaneni, Ph.D., Jennifer A. Drummond, B.A., Stacey B. Gabriel, Ph.D., Richard A. Gibbs, Ph.D., Ranabir Guin, B.S., Walker Hale, B.A.,
Alicia Hawes, B.S., D. Neil Hayes, M.D., M.P.H., Robert A. Holt, Ph.D., Alan P. Hoyle, B.S., Stuart R. Jefferys, Ph.D., Steven J.M. Jones,
Ph.D., Corbin D. Jones, Ph.D., Divya Kalra, M.S., Christie Kovar, B.S., Lora Lewis, B.S., Jie Li, M.D., Ph.D., Yussanne Ma, Ph.D., Mar-
co A. Marra, Ph.D., Michael Mayo, B.S., Shaowu Meng, Ph.D., Matthew Meyerson, M.D., Ph.D., Piotr A. Mieczkowski, Ph.D., Richard A.
Moore, Ph.D., Donna Morton, M.B.A., Lisle E. Mose, B.S., Andrew J. Mungall, Ph.D., Donna Muzny, M.S., Joel S. Parker, Ph.D.,
Charles M. Perou, Ph.D., Jeffrey Roach, Ph.D., Jacqueline E. Schein, M.S., Steven E. Schumacher, M.S., Yan Shi, Ph.D., Janae V. Simons,
B.S., Payal Sipahimalani, M.S., Tara Skelly, B.S., Matthew G. Soloway, B.S., Carrie Sougnez, B.S., Angela Tam, B.S., Donghui Tan, M.S.,
Nina Thiessen, M.S., Umadevi Veluvolu, M.Phil., Min Wang, Ph.D., Matthew D. Wilkerson, Ph.D., Tina Wong, B.S., Junyuan Wu, M.S.,
Liu Xi, M.S., Jane Zhou, Jason Bedford, B.S., Fengju Chen, Ph.D., Yao Fu, Ph.D., Mark Gerstein, Ph.D., David Haussler, Ph.D., Katayoon
Kasaian, B.S., Phillip Lai, B.S., Shiyun Ling, Ph.D., Amie Radenbaugh, B.S., David Van Den Berg, Ph.D., John N. Weinstein, M.D.,
Ph.D., Jingchun Zhu, Ph.D., Monique Albert, M.Sc., Iakovina Alexopoulou, M.D., Jeremiah J. Andersen, M.D., J. Todd Auman, Ph.D.,
John Bartlett, Ph.D., Sheldon Bastacky, M.D., Julie Bergsten, R.N., Michael L. Blute, M.D., Lori Boice, N.S., Roni J. Bollag, M.D., Ph.D.,
Jeff Boyd, Ph.D., Erik Castle, M.D., Ying‑Bei Chen, M.D., John C. Cheville, M.D., Erin Curley, M.B.A., Benjamin Davies, M.D., April
DeVolk, B.A., Rajiv Dhir, M.B., B.S., Laura Dike, Ph.D., John Eckman, P.A. (A.S.C.P.), Jay Engel, M.D., Jodi Harr, B.S., Ronald Hre-
binko, M.D., Mei Huang, Ph.D., Lori Huelsenbeck‑Dill, Mary Iacocca, M.D., Bruce Jacobs, M.D, M.P.H., Michael Lobis, M.D., Jodi K.
The New England Journal of Medicine
Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission.
Copyright © 2015 Massachusetts Medical Society. All rights reserved.
n engl j med nejm.org 11
Papillary Renal-Cell Carcinoma
Maranchie, M.D., Scott McMeekin, M.D., Jerome Myers, M.D., Joel Nelson, M.D., Jeremy Parfitt, M.D., Anil Parwani, M.D., Ph.D.,
Nicholas Petrelli, M.D., Brenda Rabeno, M.B.A., Somak Roy, M.D., Andrew L. Salner, M.D., Joel Slaton, M.D., Melissa Stanton, M.D.,
R. Houston Thompson, M.D., Leigh Thorne, M.D., Kelinda Tucker, B.A., B.S.N., Paul M. Weinberger, M.D., Cynthia Winemiller,
Leigh Anne Zach, and Rosemary Zuna, M.D.
References
1.	 Linehan WM, Srinivasan R, Schmidt
LS. The genetic basis of kidney cancer: a
metabolic disease. Nat Rev Urol 2010;​7:​
277-85.
2.	 Linehan WM. Genetic basis of kidney
cancer: role of genomics for the develop-
ment of disease-based therapeutics. Ge-
nome Res 2012;​22:​2089-100.
3.	 Delahunt B, Eble JN. Papillary renal
cell carcinoma: a clinicopathologic and
immunohistochemical study of 105 tu-
mors. Mod Pathol 1997;​10:​537-44.
4.	 Zbar B, Tory K, Merino MJ, et al. He-
reditary papillary renal cell carcinoma. J
Urol 1994;​151:​561-6.
5.	 Schmidt LS, Duh FM, Chen F, et al.
Germline and somatic mutations in the
tyrosine kinase domain of the MET proto-
oncogene in papillary renal carcinomas.
Nat Genet 1997;​16:​68-73.
6.	 Schmidt LS, Junker K, Nakaigawa N,
et al. Novel mutations of the MET proto-
oncogene in papillary renal carcinomas.
Oncogene 1999;​18:​2343-50.
7.	 Durinck S, Stawiski EW, Pavía-Jimé-
nez A, et al. Spectrum of diverse genomic
alterations define non-clear cell renal car-
cinoma subtypes. Nat Genet 2015;​47:​13-
21.
8.	 Launonen V, Vierimaa O, Kiuru M, et
al. Inherited susceptibility to uterine leio-
myomas and renal cell cancer. Proc Natl
Acad Sci U S A 2001;​98:​3387-92.
9.	 Grubb RL III, Franks ME, Toro J, et al.
Hereditary leiomyomatosis and renal cell
cancer: a syndrome associated with an ag-
gressive form of inherited renal cancer. J
Urol 2007;​177:​2074-9.
10.	 Tomlinson IP, Alam NA, Rowan AJ, et
al. Germline mutations in FH predispose
to dominantly inherited uterine fibroids,
skin leiomyomata and papillary renal cell
cancer. Nat Genet 2002;​30:​406-10.
11.	 Sudarshan S, Sourbier C, Kong HS, et
al. Fumarate hydratase deficiency in renal
cancer induces glycolytic addiction and
HIF-1 alpha stabilization by glucose-de-
pendent generation of reactive oxygen
species. Mol Cell Biol 2009;​15:​4080-90.
12.	Ooi A, Wong JC, Petillo D, et al. An
antioxidant response phenotype shared
between hereditary and sporadic type 2
papillary renal cell carcinoma. Cancer
Cell 2011;​20:​511-23.
13.	Ooi A, Dykema K, Ansari A, et al.
CUL3 and NRF2 mutations confer an
NRF2 activation phenotype in a sporadic
form of papillary renal cell carcinoma.
Cancer Res 2013;​73:​2044-51.
14.	Jiang F, Richter J, Schraml P, et al.
Chromosomal imbalances in papillary re-
nal cell carcinoma: genetic differences be-
tween histological subtypes. Am J Pathol
1998;​153:​1467-73.
15.	 Lawrence MS, Stojanov P, Mermel CH,
et al. Discovery and saturation analysis of
cancer genes across 21 tumour types. Na-
ture 2014;​505:​495-501.
16.	Kauffman EC, Ricketts CJ, Rais-Bah-
rami S, et al. Molecular genetics and cel-
lular features of TFE3 and TFEB fusion
kidney cancers. Nat Rev Urol 2014;​11:​465-
75.
17.	Schmidt LS, Junker K, Weirich G, et
al. Two North American families with he-
reditary papillary renal carcinoma and
identical novel mutations in the MET
proto-oncogene. Cancer Res 1998;​58:​
1719-22.
18.	 Boezio AA, Berry L, Albrecht BK, et al.
Discovery and optimization of potent and
selective triazolopyridazine series of c-
Met inhibitors. Bioorg Med Chem Lett
2009;​19:​6307-12.
19.	Muratani M, Deng N, Ooi WF, et al.
Nanoscale chromatin profiling of gastric
adenocarcinoma reveals cancer-associat-
ed cryptic promoters and somatically ac-
quired regulatory elements. Nat Commun
2014;​5:​4361.
20.	Gan HK, Cvrljevic AN, Johns TG. The
epidermal growth factor receptor variant
III (EGFRvIII): where wild things are al-
tered. FEBS J 2013;​280:​5350-70.
21.	Cancer Genome Atlas Research Net-
work. Comprehensive molecular charac-
terization of clear cell renal cell carcino-
ma. Nature 2013;​499:​43-9.
22.	Shen H, Laird PW. Interplay between
the cancer genome and epigenome. Cell
2013;​153:​38-55.
23.	Whitfield ML, Sherlock G, Saldanha
AJ, et al. Identification of genes periodi-
cally expressed in the human cell cycle
and their expression in tumors. Mol Biol
Cell 2002;​13:​1977-2000.
24.	Harris AL. Hypoxia — a key regula-
tory factor in tumour growth. Nat Rev
Cancer 2002;​2:​38-47.
25.	Tong WH, Sourbier C, Kovtunovych
G, et al. The glycolytic shift in fumarate-
hydratase-deficient kidney cancer lowers
AMPK levels, increases anabolic propen-
sities and lowers cellular iron levels. Can-
cer Cell 2011;​20:​315-27.
26.	Yang Y, Lane AN, Ricketts CJ, et al.
Metabolic reprogramming for producing
energy and reducing power in fumarate
hydratase null cells from hereditary leio-
myomatosis renal cell carcinoma. PLoS
One 2013;​8(8):​e72179.
27.	 Cancer Genome Atlas Network. Com-
prehensive molecular portraits of human
breast tumours. Nature 2012;​490:​61-70.
28.	Hoadley KA, Yau C, Wolf DM, et al.
Multiplatform analysis of 12 cancer types
reveals molecular classification within
and across tissues of origin. Cell 2014;​
158:​929-44.
29.	Venugopal R, Jaiswal AK. Nrf1 and
Nrf2 positively and c-Fos and Fra1 nega-
tively regulate the human antioxidant re-
sponse element-mediated expression of
NAD(P)H:quinone oxidoreductase1 gene.
Proc Natl Acad Sci U S A 1996;​93:​14960-5.
30.	Kovac M, Navas C, Horswell S, et al.
Recurrent chromosomal gains and het-
erogeneous driver mutations characterise
papillary renal cancer evolution. Nat
Commun 2015;​6:​6336.
31.	Noushmehr H, Weisenberger DJ,
Diefes K, et al. Identification of a CpG is-
land methylator phenotype that defines a
distinct subgroup of glioma. Cancer Cell
2010;​17:​510-22.
32.	Cancer Genome Atlas Research Net-
work. Comprehensive molecular profiling
of lung adenocarcinoma. Nature 2014;​
511:​543-50.
33.	Cancer Genome Atlas Research Net-
work. Comprehensive molecular charac-
terization of gastric adenocarcinoma.
Nature 2014;​513:​202-9.
34.	Linehan WM, Rouault TA. Molecular
pathways: fumarate hydratase-deficient
kidney cancer — targeting the Warburg
effect in cancer. Clin Cancer Res 2013;​19:​
3345-52.
35.	Xiao M, Yang H, Xu W, et al. Inhibi-
tion of α-KG-dependent histone and DNA
demethylases by fumarate and succinate
that are accumulated in mutations of FH
and SDH tumor suppressors. Genes Dev
2012;​26:​1326-38.
36.	 Choueiri TK, Vaishampayan U, Rosen-
berg JE, et al. Phase II and biomarker
study of the dual MET/VEGFR2 inhibitor
foretinib in patients with papillary renal
cell carcinoma. J Clin Oncol 2013;​31:​
181-6.
37.	 Johnson R, Halder G. The two faces of
Hippo: targeting the Hippo pathway for
regenerative medicine and cancer treat-
ment. Nat Rev Drug Discov 2014;​13:​63-79.
38.	 Sporn MB, Liby KT. NRF2 and cancer:
the good, the bad and the importance of
context. Nat Rev Cancer 2012;​12:​564-71.
39.	 Sourbier C, Ricketts CJ, Matsumoto S,
et al. Targeting ABL1-mediated oxidative
stress adaptation in fumarate hydratase-
deficient cancer. Cancer Cell 2014;​26:​840-
50.
Copyright © 2015 Massachusetts Medical Society.
The New England Journal of Medicine
Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission.
Copyright © 2015 Massachusetts Medical Society. All rights reserved.

Weitere ähnliche Inhalte

Was ist angesagt?

Comparison of risk factors and molecular analysis of right-sided colon and le...
Comparison of risk factors and molecular analysis of right-sided colon and le...Comparison of risk factors and molecular analysis of right-sided colon and le...
Comparison of risk factors and molecular analysis of right-sided colon and le...M. Luisetto Pharm.D.Spec. Pharmacology
 
Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...
Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...
Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...Sheldon Stein
 
Advances in lung cancer research
Advances in lung cancer research Advances in lung cancer research
Advances in lung cancer research Rohan Jagdale
 
Molecular Genetics of Cancer
Molecular Genetics of CancerMolecular Genetics of Cancer
Molecular Genetics of CancerNeha Vats
 
Clin Cancer Res-2012-Lechner-4549-59
Clin Cancer Res-2012-Lechner-4549-59Clin Cancer Res-2012-Lechner-4549-59
Clin Cancer Res-2012-Lechner-4549-59Karolina Megiel
 
Invasion and metastasis.kiran
Invasion and metastasis.kiranInvasion and metastasis.kiran
Invasion and metastasis.kiranKiran Ramakrishna
 
cancer stomach
cancer stomachcancer stomach
cancer stomachErenyPoles
 
Introduction to cancer biology nerdy scientists
Introduction to cancer biology nerdy scientistsIntroduction to cancer biology nerdy scientists
Introduction to cancer biology nerdy scientiststazib rahaman
 
Inhibition of KPNA4 Attenuates Prostate Cancer Metastasis
Inhibition of KPNA4 Attenuates Prostate Cancer MetastasisInhibition of KPNA4 Attenuates Prostate Cancer Metastasis
Inhibition of KPNA4 Attenuates Prostate Cancer MetastasisXin Li
 
Poster_Template_Horizontal_Red
Poster_Template_Horizontal_RedPoster_Template_Horizontal_Red
Poster_Template_Horizontal_RedAlexander Seutin
 
Overexpression of YAP 1 contributes to progressive features and poor prognosi...
Overexpression of YAP 1 contributes to progressive features and poor prognosi...Overexpression of YAP 1 contributes to progressive features and poor prognosi...
Overexpression of YAP 1 contributes to progressive features and poor prognosi...Enrique Moreno Gonzalez
 
Proteomics Exploration of Chronic Lymphocytic Leukemia_Crimson Publishers
Proteomics Exploration of Chronic Lymphocytic Leukemia_Crimson PublishersProteomics Exploration of Chronic Lymphocytic Leukemia_Crimson Publishers
Proteomics Exploration of Chronic Lymphocytic Leukemia_Crimson PublishersCrimsonpublishersCancer
 
Molecular biology of oral cancer, ppt
Molecular biology of oral cancer, pptMolecular biology of oral cancer, ppt
Molecular biology of oral cancer, pptRaghda Ibrahim
 

Was ist angesagt? (20)

Comparison of risk factors and molecular analysis of right-sided colon and le...
Comparison of risk factors and molecular analysis of right-sided colon and le...Comparison of risk factors and molecular analysis of right-sided colon and le...
Comparison of risk factors and molecular analysis of right-sided colon and le...
 
Austin Biochemistry
Austin BiochemistryAustin Biochemistry
Austin Biochemistry
 
Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...
Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...
Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...
 
presentation
presentationpresentation
presentation
 
Advances in lung cancer research
Advances in lung cancer research Advances in lung cancer research
Advances in lung cancer research
 
Molecular Genetics of Cancer
Molecular Genetics of CancerMolecular Genetics of Cancer
Molecular Genetics of Cancer
 
Clin Cancer Res-2012-Lechner-4549-59
Clin Cancer Res-2012-Lechner-4549-59Clin Cancer Res-2012-Lechner-4549-59
Clin Cancer Res-2012-Lechner-4549-59
 
Neoplasia 4
Neoplasia 4Neoplasia 4
Neoplasia 4
 
Dr g vassiliou
Dr g vassiliouDr g vassiliou
Dr g vassiliou
 
ACC Cancer Cell May 2016
ACC Cancer Cell May 2016ACC Cancer Cell May 2016
ACC Cancer Cell May 2016
 
Cancer biochemistry
Cancer biochemistryCancer biochemistry
Cancer biochemistry
 
Invasion and metastasis.kiran
Invasion and metastasis.kiranInvasion and metastasis.kiran
Invasion and metastasis.kiran
 
cancer stomach
cancer stomachcancer stomach
cancer stomach
 
Medi 99-e18811 (1)
Medi 99-e18811 (1)Medi 99-e18811 (1)
Medi 99-e18811 (1)
 
Introduction to cancer biology nerdy scientists
Introduction to cancer biology nerdy scientistsIntroduction to cancer biology nerdy scientists
Introduction to cancer biology nerdy scientists
 
Inhibition of KPNA4 Attenuates Prostate Cancer Metastasis
Inhibition of KPNA4 Attenuates Prostate Cancer MetastasisInhibition of KPNA4 Attenuates Prostate Cancer Metastasis
Inhibition of KPNA4 Attenuates Prostate Cancer Metastasis
 
Poster_Template_Horizontal_Red
Poster_Template_Horizontal_RedPoster_Template_Horizontal_Red
Poster_Template_Horizontal_Red
 
Overexpression of YAP 1 contributes to progressive features and poor prognosi...
Overexpression of YAP 1 contributes to progressive features and poor prognosi...Overexpression of YAP 1 contributes to progressive features and poor prognosi...
Overexpression of YAP 1 contributes to progressive features and poor prognosi...
 
Proteomics Exploration of Chronic Lymphocytic Leukemia_Crimson Publishers
Proteomics Exploration of Chronic Lymphocytic Leukemia_Crimson PublishersProteomics Exploration of Chronic Lymphocytic Leukemia_Crimson Publishers
Proteomics Exploration of Chronic Lymphocytic Leukemia_Crimson Publishers
 
Molecular biology of oral cancer, ppt
Molecular biology of oral cancer, pptMolecular biology of oral cancer, ppt
Molecular biology of oral cancer, ppt
 

Andere mochten auch

Keep a Public Diary of Your Achievements
Keep a Public Diary of Your AchievementsKeep a Public Diary of Your Achievements
Keep a Public Diary of Your AchievementsBrandon Schaefer
 
Algonquin Social Media for Recreation and Leisure Presentation
Algonquin Social Media for Recreation and Leisure PresentationAlgonquin Social Media for Recreation and Leisure Presentation
Algonquin Social Media for Recreation and Leisure PresentationPreston Martelly
 
HISTORY OF INTEREST RATES
HISTORY OF INTEREST RATESHISTORY OF INTEREST RATES
HISTORY OF INTEREST RATESDieter Drews
 
SolPresent Solution Overview - Smarter Presentation Management and Use
SolPresent Solution Overview - Smarter Presentation Management and UseSolPresent Solution Overview - Smarter Presentation Management and Use
SolPresent Solution Overview - Smarter Presentation Management and UseFrank Liggett
 
Fed Policy, Inflation, and Interest Rate Risk
Fed Policy, Inflation, and Interest Rate RiskFed Policy, Inflation, and Interest Rate Risk
Fed Policy, Inflation, and Interest Rate RiskDieter Drews
 
Algonquin Social Media Presentation- Slides
Algonquin Social Media Presentation- SlidesAlgonquin Social Media Presentation- Slides
Algonquin Social Media Presentation- SlidesPreston Martelly
 
Kitchen Cabinet company order processing procedures
Kitchen Cabinet company order processing proceduresKitchen Cabinet company order processing procedures
Kitchen Cabinet company order processing proceduresDieter Drews
 
No. 2 how to build real wealth with property - an easy guide for beginners ...
No. 2   how to build real wealth with property - an easy guide for beginners ...No. 2   how to build real wealth with property - an easy guide for beginners ...
No. 2 how to build real wealth with property - an easy guide for beginners ...Don Hannay
 
Always Put a Why Behind Your Ask
Always Put a Why Behind Your AskAlways Put a Why Behind Your Ask
Always Put a Why Behind Your AskBrandon Schaefer
 
Sample Powerpoint presentation for sales associates
Sample Powerpoint presentation for sales associatesSample Powerpoint presentation for sales associates
Sample Powerpoint presentation for sales associatesDieter Drews
 
Knowledge Retention: A Successful Bottom-up Approach
Knowledge Retention: A Successful Bottom-up ApproachKnowledge Retention: A Successful Bottom-up Approach
Knowledge Retention: A Successful Bottom-up ApproachRob Mallens (-500)
 
TECTONIC TURTLE OUTREACH PLAN
TECTONIC TURTLE OUTREACH PLANTECTONIC TURTLE OUTREACH PLAN
TECTONIC TURTLE OUTREACH PLANMonica Kuhon
 

Andere mochten auch (14)

Keep a Public Diary of Your Achievements
Keep a Public Diary of Your AchievementsKeep a Public Diary of Your Achievements
Keep a Public Diary of Your Achievements
 
Algonquin Social Media for Recreation and Leisure Presentation
Algonquin Social Media for Recreation and Leisure PresentationAlgonquin Social Media for Recreation and Leisure Presentation
Algonquin Social Media for Recreation and Leisure Presentation
 
HISTORY OF INTEREST RATES
HISTORY OF INTEREST RATESHISTORY OF INTEREST RATES
HISTORY OF INTEREST RATES
 
SolPresent Solution Overview - Smarter Presentation Management and Use
SolPresent Solution Overview - Smarter Presentation Management and UseSolPresent Solution Overview - Smarter Presentation Management and Use
SolPresent Solution Overview - Smarter Presentation Management and Use
 
Fed Policy, Inflation, and Interest Rate Risk
Fed Policy, Inflation, and Interest Rate RiskFed Policy, Inflation, and Interest Rate Risk
Fed Policy, Inflation, and Interest Rate Risk
 
Algonquin Social Media Presentation- Slides
Algonquin Social Media Presentation- SlidesAlgonquin Social Media Presentation- Slides
Algonquin Social Media Presentation- Slides
 
Journal of Surgical Oncology - Barnabas
Journal of Surgical Oncology - BarnabasJournal of Surgical Oncology - Barnabas
Journal of Surgical Oncology - Barnabas
 
Kitchen Cabinet company order processing procedures
Kitchen Cabinet company order processing proceduresKitchen Cabinet company order processing procedures
Kitchen Cabinet company order processing procedures
 
No. 2 how to build real wealth with property - an easy guide for beginners ...
No. 2   how to build real wealth with property - an easy guide for beginners ...No. 2   how to build real wealth with property - an easy guide for beginners ...
No. 2 how to build real wealth with property - an easy guide for beginners ...
 
Enumerators
EnumeratorsEnumerators
Enumerators
 
Always Put a Why Behind Your Ask
Always Put a Why Behind Your AskAlways Put a Why Behind Your Ask
Always Put a Why Behind Your Ask
 
Sample Powerpoint presentation for sales associates
Sample Powerpoint presentation for sales associatesSample Powerpoint presentation for sales associates
Sample Powerpoint presentation for sales associates
 
Knowledge Retention: A Successful Bottom-up Approach
Knowledge Retention: A Successful Bottom-up ApproachKnowledge Retention: A Successful Bottom-up Approach
Knowledge Retention: A Successful Bottom-up Approach
 
TECTONIC TURTLE OUTREACH PLAN
TECTONIC TURTLE OUTREACH PLANTECTONIC TURTLE OUTREACH PLAN
TECTONIC TURTLE OUTREACH PLAN
 

Ähnlich wie Publication NJEM (2015)

Overview of New Targets For Anti-tumor Drugs.pdf
Overview of New Targets For Anti-tumor Drugs.pdfOverview of New Targets For Anti-tumor Drugs.pdf
Overview of New Targets For Anti-tumor Drugs.pdfDoriaFang
 
Breast N C C Nguidlinesms1
Breast N C C Nguidlinesms1Breast N C C Nguidlinesms1
Breast N C C Nguidlinesms1guest108e832
 
High Sensitivity Detection of Tumor Gene Mutations-v3
High Sensitivity Detection of Tumor Gene Mutations-v3High Sensitivity Detection of Tumor Gene Mutations-v3
High Sensitivity Detection of Tumor Gene Mutations-v3Michael Powell
 
hMSH2 Gly322Asp (rs4987188) Single nucleotide polymorphism and the risk of br...
hMSH2 Gly322Asp (rs4987188) Single nucleotide polymorphism and the risk of br...hMSH2 Gly322Asp (rs4987188) Single nucleotide polymorphism and the risk of br...
hMSH2 Gly322Asp (rs4987188) Single nucleotide polymorphism and the risk of br...Agriculture Journal IJOEAR
 
Molecular classification of endometrial cancer
Molecular classification of endometrial cancerMolecular classification of endometrial cancer
Molecular classification of endometrial cancerMohammed Nassar
 
Highlights from asco gu 2017
Highlights from asco gu 2017   Highlights from asco gu 2017
Highlights from asco gu 2017 Mohamed Abdulla
 
Molecular Biology of Esophageal and Gastric Cancer
Molecular Biology of Esophageal and Gastric CancerMolecular Biology of Esophageal and Gastric Cancer
Molecular Biology of Esophageal and Gastric CancerSiddharth Sreemahadevan
 
Using biomarkers to monitor the dynamics of tumor
Using biomarkers to monitor the dynamics of tumorUsing biomarkers to monitor the dynamics of tumor
Using biomarkers to monitor the dynamics of tumorsummer elmorshidy
 
Immunotherapy in endometrial
Immunotherapy in endometrialImmunotherapy in endometrial
Immunotherapy in endometrialOrmias Pratama
 
Cancer Genetics - Denise Sheer
Cancer Genetics - Denise SheerCancer Genetics - Denise Sheer
Cancer Genetics - Denise SheerDenise Sheer
 
Sex-Based Difference in Gene Alterations and Biomarkers in Anal Squamous Cell...
Sex-Based Difference in Gene Alterations and Biomarkers in Anal Squamous Cell...Sex-Based Difference in Gene Alterations and Biomarkers in Anal Squamous Cell...
Sex-Based Difference in Gene Alterations and Biomarkers in Anal Squamous Cell...semualkaira
 
Next Generation Sequencing in Diagnosis of Reed’s Syndrome, or Hereditary Lei...
Next Generation Sequencing in Diagnosis of Reed’s Syndrome, or Hereditary Lei...Next Generation Sequencing in Diagnosis of Reed’s Syndrome, or Hereditary Lei...
Next Generation Sequencing in Diagnosis of Reed’s Syndrome, or Hereditary Lei...CrimsonpublishersCancer
 
Michael's IUCRL Poster 2014 Close to Final with CDW edits
Michael's IUCRL Poster 2014 Close to Final with CDW editsMichael's IUCRL Poster 2014 Close to Final with CDW edits
Michael's IUCRL Poster 2014 Close to Final with CDW editsMichael Araya
 
SCT60103 Group 2 Presentation
SCT60103 Group 2 PresentationSCT60103 Group 2 Presentation
SCT60103 Group 2 PresentationJayden On
 
Sitagliptina e proteção em ca diferenciado da tireóide.x
Sitagliptina e proteção em ca diferenciado da tireóide.xSitagliptina e proteção em ca diferenciado da tireóide.x
Sitagliptina e proteção em ca diferenciado da tireóide.xRuy Pantoja
 
The KRAS-Variant and miRNA Expression in RTOG Endometrial Cancer Clinical Tri...
The KRAS-Variant and miRNA Expression in RTOG Endometrial Cancer Clinical Tri...The KRAS-Variant and miRNA Expression in RTOG Endometrial Cancer Clinical Tri...
The KRAS-Variant and miRNA Expression in RTOG Endometrial Cancer Clinical Tri...UCLA
 

Ähnlich wie Publication NJEM (2015) (20)

Overview of New Targets For Anti-tumor Drugs.pdf
Overview of New Targets For Anti-tumor Drugs.pdfOverview of New Targets For Anti-tumor Drugs.pdf
Overview of New Targets For Anti-tumor Drugs.pdf
 
Breast N C C Nguidlinesms1
Breast N C C Nguidlinesms1Breast N C C Nguidlinesms1
Breast N C C Nguidlinesms1
 
High Sensitivity Detection of Tumor Gene Mutations-v3
High Sensitivity Detection of Tumor Gene Mutations-v3High Sensitivity Detection of Tumor Gene Mutations-v3
High Sensitivity Detection of Tumor Gene Mutations-v3
 
hMSH2 Gly322Asp (rs4987188) Single nucleotide polymorphism and the risk of br...
hMSH2 Gly322Asp (rs4987188) Single nucleotide polymorphism and the risk of br...hMSH2 Gly322Asp (rs4987188) Single nucleotide polymorphism and the risk of br...
hMSH2 Gly322Asp (rs4987188) Single nucleotide polymorphism and the risk of br...
 
Molecular classification of endometrial cancer
Molecular classification of endometrial cancerMolecular classification of endometrial cancer
Molecular classification of endometrial cancer
 
Highlights from asco gu 2017
Highlights from asco gu 2017   Highlights from asco gu 2017
Highlights from asco gu 2017
 
Molecular Biology of Esophageal and Gastric Cancer
Molecular Biology of Esophageal and Gastric CancerMolecular Biology of Esophageal and Gastric Cancer
Molecular Biology of Esophageal and Gastric Cancer
 
Using biomarkers to monitor the dynamics of tumor
Using biomarkers to monitor the dynamics of tumorUsing biomarkers to monitor the dynamics of tumor
Using biomarkers to monitor the dynamics of tumor
 
Immunotherapy in endometrial
Immunotherapy in endometrialImmunotherapy in endometrial
Immunotherapy in endometrial
 
Cancer Genetics - Denise Sheer
Cancer Genetics - Denise SheerCancer Genetics - Denise Sheer
Cancer Genetics - Denise Sheer
 
Session 2.1 Kinkhabwala
Session 2.1 KinkhabwalaSession 2.1 Kinkhabwala
Session 2.1 Kinkhabwala
 
Session 2.1: Kinkhabwala
Session 2.1: KinkhabwalaSession 2.1: Kinkhabwala
Session 2.1: Kinkhabwala
 
Sex-Based Difference in Gene Alterations and Biomarkers in Anal Squamous Cell...
Sex-Based Difference in Gene Alterations and Biomarkers in Anal Squamous Cell...Sex-Based Difference in Gene Alterations and Biomarkers in Anal Squamous Cell...
Sex-Based Difference in Gene Alterations and Biomarkers in Anal Squamous Cell...
 
Next Generation Sequencing in Diagnosis of Reed’s Syndrome, or Hereditary Lei...
Next Generation Sequencing in Diagnosis of Reed’s Syndrome, or Hereditary Lei...Next Generation Sequencing in Diagnosis of Reed’s Syndrome, or Hereditary Lei...
Next Generation Sequencing in Diagnosis of Reed’s Syndrome, or Hereditary Lei...
 
Michael's IUCRL Poster 2014 Close to Final with CDW edits
Michael's IUCRL Poster 2014 Close to Final with CDW editsMichael's IUCRL Poster 2014 Close to Final with CDW edits
Michael's IUCRL Poster 2014 Close to Final with CDW edits
 
SCT60103 Group 2 Presentation
SCT60103 Group 2 PresentationSCT60103 Group 2 Presentation
SCT60103 Group 2 Presentation
 
PIIS0016508514604509
PIIS0016508514604509PIIS0016508514604509
PIIS0016508514604509
 
408131
408131408131
408131
 
Sitagliptina e proteção em ca diferenciado da tireóide.x
Sitagliptina e proteção em ca diferenciado da tireóide.xSitagliptina e proteção em ca diferenciado da tireóide.x
Sitagliptina e proteção em ca diferenciado da tireóide.x
 
The KRAS-Variant and miRNA Expression in RTOG Endometrial Cancer Clinical Tri...
The KRAS-Variant and miRNA Expression in RTOG Endometrial Cancer Clinical Tri...The KRAS-Variant and miRNA Expression in RTOG Endometrial Cancer Clinical Tri...
The KRAS-Variant and miRNA Expression in RTOG Endometrial Cancer Clinical Tri...
 

Publication NJEM (2015)

  • 1. The new engl and jour nal of medicine n engl j med nejm.org 1 The authors’ full names and academic degrees are listed in the Appendix. Ad- dress reprint requests to Dr. Linehan at the Urologic Oncology Branch, National Cancer Institute, Bldg. 10 CRC Rm. 1-5940, Bethesda, MD 20892-1107, or at ­wml@​­nih​.­gov. *The authors are members of the Cancer Genome Atlas Research Network, and their names, affiliations, and roles are listed in Supplementary Appendix 1, available at NEJM.org. This article was published on November 4, 2015, at NEJM.org. DOI: 10.1056/NEJMoa1505917 Copyright © 2015 Massachusetts Medical Society. BACKGROUND Papillary renal-cell carcinoma, which accounts for 15 to 20% of renal-cell carcino- mas, is a heterogeneous disease that consists of various types of renal cancer, including tumors with indolent, multifocal presentation and solitary tumors with an aggressive, highly lethal phenotype. Little is known about the genetic basis of sporadic papillary renal-cell carcinoma, and no effective forms of therapy for ad- vanced disease exist. METHODS We performed comprehensive molecular characterization of 161 primary papillary renal-cell carcinomas, using whole-exome sequencing, copy-number analysis, mes- senger RNA and microRNA sequencing, DNA-methylation analysis, and proteomic analysis. RESULTS Type 1 and type 2 papillary renal-cell carcinomas were shown to be different types of renal cancer characterized by specific genetic alterations, with type 2 further classified into three individual subgroups on the basis of molecular differences associated with patient survival. Type 1 tumors were associated with MET altera- tions, whereas type 2 tumors were characterized by CDKN2A silencing, SETD2 mutations, TFE3 fusions, and increased expression of the NRF2–antioxidant re- sponse element (ARE) pathway. A CpG island methylator phenotype (CIMP) was observed in a distinct subgroup of type 2 papillary renal-cell carcinomas that was characterized by poor survival and mutation of the gene encoding fumarate hydra- tase (FH). CONCLUSIONS Type 1 and type 2 papillary renal-cell carcinomas were shown to be clinically and biologically distinct. Alterations in the MET pathway were associated with type 1, and activation of the NRF2-ARE pathway was associated with type 2; CDKN2A loss and CIMP in type 2 conveyed a poor prognosis. Furthermore, type 2 papillary re- nal-cell carcinoma consisted of at least three subtypes based on molecular and phenotypic features. (Funded by the National Institutes of Health.) ABSTR ACT Comprehensive Molecular Characterization of Papillary Renal-Cell Carcinoma The Cancer Genome Atlas Research Network* Original Article The New England Journal of Medicine Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission. Copyright © 2015 Massachusetts Medical Society. All rights reserved.
  • 2. n engl j med nejm.org2 The new engl and jour nal of medicine K idney cancer, or renal-cell carci- noma, is not a single disease but is made up of various types of cancer that are characterized by different genetic drivers; each type has distinct histologic features and a dis- tinct clinical course and response to therapy.1,2 Papillary renal-cell carcinoma, which accounts for 15 to 20% of kidney cancers, is a heteroge- neous disease with histologic subtypes and varia- tions in both disease progression and patient outcomes. Papillary renal-cell carcinoma has two main subtypes: type 1, which is often multifocal, is characterized by papillae and tubular struc- tures covered with small cells containing baso- philic cytoplasm and small, uniform, oval nuclei,3 whereas type 2, which is more heterogeneous, is characterized by papillae covered with large cells containing eosinophilic cytoplasm and large, spherical nuclei with prominent nucleoli.3,4 Al- though in some patients papillary renal-cell carcinoma is indolent, bilateral, and multifocal, other patients present with solitary lesions that have an aggressive clinical course. Little is known about the genetic basis of the sporadic forms of papillary renal-cell carcinoma, and there are currently no effective forms of therapy for patients with advanced disease. Much of our knowledge of the genetic basis of papillary renal-cell carcinoma has been based on the study of the inherited form of the disease. Hereditary papillary renal-cell carcinoma, a rare disorder that is associated with an increased risk of type 1 disease,4 is characterized by activating germline mutations of MET.5 Somatic MET muta- tions occur in 13 to 15% of nonhereditary papil- lary renal-cell carcinomas.6,7 The hereditary leio- myomatosis and renal-cell cancer syndrome, which confers a predisposition to an aggressive form of type 2 papillary renal-cell carcinoma,8,9 is caused by germline mutation of the gene en- coding fumarate hydratase (FH), an enzyme of the tricarboxylic acid cycle.10 These aggressive tumors are characterized by increased oxidative stress11 and activation of the NRF2–antioxidant response element (ARE) pathway.12 Mutations in the genes that regulate the NRF2-ARE pathway, such as CUL3 and NFE2L2 (which encodes NRF2), have also been observed in sporadic papillary renal-cell carcinoma.13 We performed an integrative genomic analy- sis of 161 papillary renal-cell carcinoma tumors to provide molecular insights into tumor classi- fication, inform clinical recommendations, and suggest paths to the development of mechanisti- cally based therapies. Methods Patients Tumors were selected from 161 patients. Patho- logical review was performed to classify the tu- mors as type 1, type 2, or uncharacterized papil- lary renal-cell carcinoma (see the Experimental Procedures section in Supplementary Appendix 1, available with the full text of this article at NEJM.org). The clinical and genetic characteris- tics of these patients are described in Supple- mentary Appendix 2. Analytic Platforms We performed whole-exome sequencing and analyses to determine copy number, microRNA and messenger RNA (mRNA) expression, protein expression, and DNA methylation at CpG sites (Supplementary Appendix 3). Details of all the analyses are available in the Experimental Proce- dures section in Supplementary Appendix 1. All data sets are available at the Cancer Genome At- las data portal (https:/​­/​­tcga-data​.­nci​.­nih​.­gov/​­tcga). Results Histologic Subtyping Pathological review of the 161 tumors identified 75 type 1 tumors, 60 type 2 tumors, and 26 tu- mors that could not be classified as type 1 or type 2. The type 1 tumors were predominantly stage I, whereas the type 2 tumors were fre- quently stage III or IV (Fig. S1 in Supplementary Appendix 1); these findings were consistent with those of previous studies.3,14 Role of Somatic Alterations in Molecular Differences between Type 1 and Type 2 Tumors Copy-Number Alterations Single-nucleotide-polymorphism array–based profiling of somatic copy-number alterations revealed distinctive patterns across three main tumor subgroups. One subgroup, composed predominantly of type 1 and lower-grade tumors, was defined by multiple chromosomal gains (of at least one complete copy of the chromosome), The New England Journal of Medicine Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission. Copyright © 2015 Massachusetts Medical Society. All rights reserved.
  • 3. n engl j med nejm.org 3 Papillary Renal-Cell Carcinoma including nearly universal gain of chromosomes 7 and 17 and less frequent gain of chromosomes 2, 3, 12, 16, and 20 (Fig. 1A, and Fig. S2 in Supplementary Appendix 1). The other two sub- groups were predominantly type 2 tumors; al- though one of these subgroups had few copy- number alterations, the other was characterized by a high degree of aneuploidy with multiple chromosomal losses, including frequent loss of chromosome 9p, and was associated with poorer survival (P<0.001) (Fig. 1A, and Fig. S2 in Sup- plementary Appendix 1). Whole-Exome Sequencing Whole-exome sequencing identified 10,380 puta- tive somatic mutations in 157 tumors with an average of 1.45 nonsilent mutations per mega- base (see the Experimental Procedures section in Supplementary Appendix 1). An initial screen for significantly mutated genes with q values of less than 0.1 (q values range from 0.0 to 1.0), with the use of MutSigCV, version 2.0, identified five such genes (MET, SETD2, NF2, KDM6A, and SMARCB1) that were recurrently mutated in pap- illary renal-cell carcinoma, representing 24% of cases (Fig. 1B). Further analysis, performed with restriction of multiple hypothesis testing to genes previously associated with cancer in the PanCan21 data set,15 identified six additional significantly mutated genes (FAT1, BAP1, PBRM1, STAG2, NFE2L2, and TP53), with 36% of cases showing mutation of at least one of these genes (Fig. 1B). Mutation of these significantly mutat- ed genes showed no evidence of subclonality (Supplementary Appendix 4). Hippo and Chromatin Modifier Pathways Several significantly mutated genes in papillary renal-cell carcinoma are components of well- known cancer-associated pathways or complexes, including NF2 in the Hippo signaling pathway, SMARCB1 and PBRM1 in the SWI/SNF complex, and SETD2, KDM6A, and BAP1 in several chroma- tin modifier pathways. Assessment of genes in these pathways (Supplementary Appendix 5) showed a high number of mutations in both type 1 and type 2 tumors involving the SWI/SNF complex (20% and 27%, respectively), chromatin modifier pathways (35% and 38%, respectively), and the Hippo signaling pathway (3% and 10%, respectively) (Fig. 1C). TFE3 and TFEB Gene Fusions Gene fusions involving TFE3 or TFEB have previ- ously been associated with papillary renal-cell carcinoma (reviewed in Kauffman et al.16 ). We identified gene fusions in 17 tumors (10.6%), including 8 involving TFE3 or TFEB (Supplemen- tary Appendix 6). Four of the TFE3 fusions in- volved known fusion partners, PRCC and SFPQ, and 2 involved novel fusion partners, RBM10 and DVL2 (Fig. 1D). The tumors with TFE3 fusions showed varying degrees of increased mRNA ex- pression for known TFE3 transcriptional targets, including CTSK, BIRC7, DIAPH1, and HIF1A (Fig. S3 in Supplementary Appendix 1). The two TFEB fusions involved novel fusion partners, COL21A1 and CADM2, with the COL21A1–TFEB fusion re- sulting in a construct similar to the known MALAT1–TFEB fusions16 and the TFEB–CADM2 fu- sion resulting in a novel truncated version of TFEB that had lost several microRNA binding sites (Fig. 1D). The tumors with TFEB fusions showed high mRNA expression of the TFEB tran- scription factor and a known target gene, CTSK (Fig. S4 in Supplementary Appendix 1). Seven of the fusions involving TFE3 or TFEB were identi- fied in the type 2 tumors (7 of 60 [12%]). Alterations Specific to Types of Papillary Renal-Cell Carcinoma MET Mutation in Type 1 Tumors We found mutation of MET in 17 tumors, includ- ing germline mutation in 3 tumors. A total of 14 of the 17 MET mutations were in the tyrosine kinase domain, and 13 of these mutations were observed in type 1 tumors (17% of the 75 type 1 tumors) (Fig. 2A and 2B). In addition, an alter- nate MET RNA transcript that replaces canonical exons 1 and 2 with a novel exon 1 spliced to canonical exon 3 (Fig. 2A) was identified in 8 tumors (4 type 1 tumors, 3 type 2 tumors, and 1 unclassified tumor). This isoform represented the majority of transcripts in 2 tumors and a fraction in the remaining 6 tumors and was re- cently observed to produce a stable, shortened protein in gastric-cancer cell lines (Fig. S5A in Supplementary Appendix 1).19 Exons 1 and 2 of MET encode the ligand-binding domain of he- patocyte growth factor receptor; this isoform, analogous to the epidermal growth factor recep- tor variant III isoform,20 may result in ligand- independent MET activation. In addition, gene The New England Journal of Medicine Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission. Copyright © 2015 Massachusetts Medical Society. All rights reserved.
  • 4. n engl j med nejm.org4 The new engl and jour nal of medicine B A No.ofMutations 16 12 14 10 8 4 2 6 0 C D Missense Splice site Nonsense Frame shift In-frame indel MET gene Hippo pathway NRF2 pathway Chromatin modifiers SWI/SNF complex mTOR pathway p53-related genes Chr 7 gain Type 1 PRCC (N=75) Type 2 PRCC (N=60) Unclassified PRCC (N=26) Somatic Mutation SMG (current study) No dataOther related geneOther pan-cancer SMG 1 2 3 4 5 6 7 8 9 10 11 12 13 15 17 19 22 Chromosome Copy gain Type 1 PRCC Type 2 PRCC Copy loss M ETFAT1SETD 2 N F2KD M 6ABAP1PBRM 1 SM ARCB1N FE2L2STAG 2TP53 All PRCC (N=157) Type 1 PRCC (N=71) Type 2 PRCC (N=60) −log10 P value 0 2 4 6 8 q<0.10 for pan-cancer SMGs * * * * Genome unstable, Chr 9p deleted Arm-level alterations, Chr 7 gain Genome stable, sporadic SCNAs, few Chr 9p deletions * * * ** * * † † † † † q<0.05 for all genes† MAD2L2 DVL2 DIX RBM10 SFPQ TFEB microRNA binding sites COL21A1 TFEB 3x PRCC TFE3 TFE3 TFE3 TFE3 AD bHLH-LZ AD bHLH-LZ AD bHLH-LZ AD bHLH-LZ AD bHLH-LZ AD bHLH-LZ RRM1RRM2Q/E/P-Rich 1 4 5 6 78 9 10 2 3 4 5 6 78 9 102 3 4 5 61 1 2 3 4 5 3 4 5 6 78 9 10 1 2 3 4 56 78 9 10 5 6 78 9 101112 13 14151617 3' UTR 2 3 4 5 6 78 9 10 1 2 3 4 5 6 7 8 9 PRCC-associated TFE3 fusions (4) Additional PRCC-associated TFE3 fusions (2) Additional PRCC-associated TFEB fusions (2) CADM2 Mutation Type The New England Journal of Medicine Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission. Copyright © 2015 Massachusetts Medical Society. All rights reserved.
  • 5. n engl j med nejm.org 5 Papillary Renal-Cell Carcinoma fusions involving MET were observed in 3 tumors (Supplementary Appendix 6). Levels of MET mRNA expression and of protein phosphoryla- tion (pY1235) were significantly higher in type 1 tumors than in type 2 tumors (P<1×10−9 and P=0.007, respectively, by t-test) (Fig. S5B in Supplementary Appendix 1) — a finding poten- tially driven in part by trisomy of chromosome 7 in type 1 tumors. Altered MET status (defined as mutation, splice variant, or gene fusion) or in- creased chromosome 7 copy number (which en- codes MET but may also involve other genes) was identified in 81% of type 1 papillary renal-cell carcinomas. Analysis by means of Genomic Identification of Significant Targets in Cancer (GISTIC), version 2.0, determined that the loss of 1p36 observed in 18 papillary renal-cell carcino- mas (11.2%) included the candidate tumor sup- pressor ERRFI1, a negative regulator of EGFR Figure 1 (facing page). Somatic Alterations in Papillary Renal-Cell Carcinoma and Molecular Differences between Type 1 and Type 2 Cancers. Unsupervised clustering of DNA copy profiles of 161 papillary renal-cell carcinomas (PRCCs) (Panel A) re- vealed three molecular subtypes, one of which was highly enriched for type 1 tumors and the other two for type 2 tumors. SCNA denotes somatic copy-number alterations. Significantly mutated genes (SMGs) in PRCC (Panel B) were determined by considering all genes (q<0.1 [range, 0.0 to 1.0]) or focusing on the set of 260 genes previously implicated in cancer by large- scale, pan-cancer exome analyses15 (q<0.1). P values were calculated with the MutSigCV algorithm, version 2.0. A pathway-centric view of gene mutations in PRCC (Panel C) shows key pathways and genes implicated in cancer, either in the current study or elsewhere.15 The tumors were classified according to histologic type (from left to right) and according to gene or pathway altered (from top to bottom). Pathways and genes rep- resented include MET, the Hippo pathway (NF2, SAV1, and WWC1), the NRF2 pathway (NFE2L2, KEAP1, CUL3, SIRT1, and FH), chromatin modification (CREBBP, DOTL1, EHMT1/2, EP300, EZH1/2, KAT2A/B, KDM1A/B, KDM4A/B, KDM5A/B/C, KDM6A/B, MLL1/2/3/4/5, NSD1, SETD2, SMYD4, and SRCAP), the SWI/SNF complex (ACTB, ACTL6A/B, ARID1A/B, ARID2, BCL6A/ B/C, BCL11A/B, BRD7/9, DPF1/2/3, PHF10, PBRM1, SMARCA2/4, SMARCB1, SMARCC1/2, SMARCD1/2/3, and SMARCE1), the mammalian target of rapamycin (mTOR) pathway (MTOR, PIK3CA, PTEN, STK11, TSC1, and TSC2), and the p53 pathway (ATM, CDKN1A, CDKN2A, FBXW7, RB1, and TP53). Fusion gene analy- sis (Panel D) identified TFE3 or TFEB fusions in eight PRCC tumors, including two novel gene-fusion partners for TFE3 (DVL2 and RBM10) and two novel gene-fusion partners for TFEB (COL21A1 and CADM2). Schematic versions of these fusions show the exons and functional domains that are present in the different gene fusions and the position of potential microRNA binding sites in TFEB. The retained exons of TFE3 or TFEB are colored in shades of blue. Thin regions represent noncoding se- quence, thick regions represent the translated reading frame, and white strips indicate that the region is no longer to scale. AD denotes strong transcription acti- vation domain, bHLH basic helix-loop-helix domain, DIX dishevelled and axin domain, LZ leucine zipper domain, MAD2L2 mitotic arrest deficient–like 2 inter- action domain, and RRM RNA-recognition motif. Figure 2. Alterations in Papillary Renal-Cell Carcinoma Involving the MET Oncogene. Panel A is a schematic representation of somatic mutations in MET, along with germline variant H1112R, which was previously implicated in hereditary papillary renal-cell carcinoma,17 and the novel RNA transcript variant of MET lacking the canonical exons 1 and 2 but containing a novel exon 1 that splices to the canonical exon 3. IPT denotes immunoglobulin-like, plexins, and transcription factors, and PSI plexins, semaphorins, and integrins. Panel B shows the crystal structure for the MET tyrosine kinase catalytic domain (RCSB-PDB 3I5 N18 ), on which are mapped the residues that are altered in papillary renal-cell carcinoma. All numbering of amino acids is based on the MET protein sequences. Activation loop Somatic mutation observed in unclassified PRCC Somatic mutation observed in type 1 PRCC L296P F1218 Catalytic loop Y1248 ATP- binding regionV1110 H1112 V1088 D1246 S1254 M1268 p+1 Loop I639L F970V F1218IH1112Y/R V1110I V1088E/R D1246H Y1248H S1254R M1268T Tyrosine kinase catalytic domain IPT domainSema domain PSI domain Germline mutation observed in type 1 PRCC Somatic mutation observed in type 2 PRCC Encoded by alternate exon (~65 aa) A B The New England Journal of Medicine Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission. Copyright © 2015 Massachusetts Medical Society. All rights reserved.
  • 6. n engl j med nejm.org6 The new engl and jour nal of medicine (Fig. S6 in Supplementary Appendix 1). Dele- tions of 1p36 co-occurred significantly with gain of chromosome 7 and EGFR amplification (P = 0.02 by Fisher’s exact test). CDKN2A Mutation in Type 2 Tumors Analysis by GISTIC, version 2.0, identified focal loss of 9p21 in 13 papillary renal-cell carcino- mas (8.1%), resulting in loss of CDKN2A (Fig. S7A in Supplementary Appendix 1). We found muta- tion or promoter hypermethylation of CDKN2A in 11 tumors (Fig. S7B in Supplementary Appendix 1), including 3 of the tumors with focal loss of 9p21, resulting in 21 tumors (13.0%) defined as having CDKN2A alteration (Fig. S7C in Supple- mentary Appendix 1). CDKN2A alteration was strongly associated with type 2 histologic fea- tures, with 25% of type 2 tumors (15 of 60) showing alterations. CDKN2A-altered tumors showed both increased levels of phosphorylated retinoblastoma protein (Rb) and increased ex- pression of cell-cycle–related genes, findings consistent with the predicted consequences of CDKN2A loss (Fig. S7D in Supplementary Appen- dix 1). In a univariate analysis, patients with CDKN2A-altered tumors had a significantly lower rate of overall survival than those without CDKN2A- altered tumors (P<1×10−10 ) (Fig. S7E in Supple- mentary Appendix 1). The findings were similar when the analysis was limited to patients with type 2 tumors (P<0.001) (Fig. S7F in Supplemen- tary Appendix 1). In addition, increased expres- sion of microRNA miR-10b-5p correlated with de- creased expression of its target, CDKN2A (Fig. S8 in Supplementary Appendix 1). SETD2, BAP1, and PBRM1 Mutation in Type 2 Tumors Type 2 tumors were associated with mutations in the chromatin-modifying genes SETD2, BAP1, and PBRM1, which are frequently mutated in clear-cell kidney tumors in combination with loss of chromosome 3p.21 Mutations of BAP1 and PBRM1 were mutually exclusive, but PBRM1 muta- tions were frequently concurrent with SETD2 mutations (Fig. S9 in Supplementary Appendix 1). Although loss of chromosome 3p was also associated with type 2 papillary renal-cell carci- noma, only 3 of 13 type 2 tumors with SETD2, BAP1, or PBRM1 mutation showed such loss, and no promoter hypermethylation was observed (Fig. S9 in Supplementary Appendix 1). CpG Island Methylator Phenotype (CIMP) in Type 2 Tumors Nine tumors (5.6%) had increased DNA methyla- tion at loci that were unmethylated in matched normal tissue. This represents a novel kidney- associated CIMP22 that included universal hyper- methylation of the CDKN2A promoter (Fig. 3A). Eight of the nine tumors were type 2 papillary renal-cell carcinomas. In five tumors, we found germline or somatic mutation of FH (56%). We found decreased expression of FH mRNA and increased expression of genes associated with cell-cycle progression and response to hypoxia in all nine tumors (Fig. 3A). Patients with CIMP- associated tumors were younger at the time of presentation and had a lower probability of over- all survival than other patients with papillary renal-cell carcinoma (Fig. 3B). Fumarate hydra- tase–deficient type 2 tumors in patients with the hereditary leiomyomatosis and renal-cell cancer syndrome are characterized by a Warburg-like metabolic shift to glycolysis-dependent metabo- lism and an increased expression of hypoxia- related genes.25,26 Similarly, CIMP-associated tumors showed increased expression of key genes involved in glycolysis (HK1, LDHA, and PDK1), the pentose phosphate pathway (G6PD), and fat- ty-acid synthesis (FASN) (Fig. 3C, and Fig. S10 in Supplementary Appendix 1). In addition, there was decreased expression of the majority of genes involved in the Krebs cycle and the ade- nosine monophosphate–activated protein kinase (AMPK) complex, a suppressor of fatty-acid syn- thesis (Fig. 3D). Data on the expression of pro- teins G6PD, FASN, and AMPK correlated with the data on mRNA expression (Fig. 3D). Identification of Papillary Renal-Cell Carcinoma Subgroups by Multiplatform Analysis Cluster-of-Clusters Analysis As was the case with the copy-number analysis and DNA-methylation analysis, the profiles of mRNA expression and microRNA expression and the data on protein expression clustered the cases of papillary renal-cell carcinoma into sep- arate groups with distinct overall outcomes (Fig. S11, S12, and S13 in Supplementary Appendix 1). The five data types were combined to per- form a cluster-of-clusters analysis27,28 that identi- fied four tumor clusters: C1 (enriched for type 1 tumors), C2a and C2b (both enriched for type 2 The New England Journal of Medicine Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission. Copyright © 2015 Massachusetts Medical Society. All rights reserved.
  • 7. n engl j med nejm.org 7 Papillary Renal-Cell Carcinoma Figure 3. A Subgroup of Papillary Renal-Cell Carcinoma That Manifests a CpG Island Methylator Phenotype (CIMP). As depicted in Panel A, molecular subtyping by means of a DNA methylation platform revealed three subtypes of papillary renal-cell car- cinoma (PRCC), one of which showed widespread DNA hypermethylation patterns characteristic of CIMP-associated tumors (the other subtypes are identified as cluster 1 and cluster 2). Corresponding data tracks highlight molecular features associated with CIMP tumors (nine cases), including CDKN2A silencing, germline or somatic mutations of FH, type 2 histologic status, and expression of both cell- cycle–related genes23 and hypoxia-related genes.24 Panel B shows differences in patient age and overall survival among the three sub- types. Data on survival were not available for two patients in the cluster 2 group. Panel C shows differential messenger RNA (mRNA) expression patterns for key genes involved in metabolism among CIMP-associated PRCC, type 1 PRCC, non–CIMP-associated type 2 PRCC, and normal kidney. Panel D shows differential expression patterns of CIMP-associated tumors versus type 1 tumors in metabo- lism-related pathways, with a focus on gene-expression and protein-expression patterns previously associated with Warburg-like effects in kidney cancer.21 P values were calculated with the use of a t-test. C D A B ProbabilityofOverall Survival 1.0 0.8 0.6 0.4 0.2 0.0 0.9 0.7 0.5 0.3 0.1 0 50 100 150 200 250 Months AgeofPatient(yr) 80 60 40 0 70 50 30 Subtype Based on DNA Methylation CIMP Cluster 1 Cluster 2 PRCC (N=161 cases) Tumor type Normal kidney (N=50) CIM P Cluster 1 Cluster 2 Subtype Based on DNA Methylation 343GenomicLoci CDKN2A silencing CDKN2A loss CDKN2A mutation FH mutation DNAMethylation(βvalue) High Low 0 0.5 1 CIMP (N=9) Cluster 1 (N=30) Cluster 2 (N=120) Censored P=1×10−16 for overall comparison by log-rank test FH expr. Cell cycle expr. Hypoxia expr. HighLow CIMP- Associated PRCC (N=9) Other Type 2 PRCC (N=52) Type 1 PRCC (N=75) Normal Kidney (N=30) HK1 G6PD LDHA PDK1 FASN PRKAB1 ACO2 OGDH SUCLA2 SDHA FH MDH2 mRNA Expression Somatic mutation Germline mutation Higher expression Lower expression Type 1 Type 2 Glycolysis Pentose phosphate pathway Fatty acids and lipids FASNPDK1 G6PD HK1 Glucose- 6-phosphate PRKAB1 PRKAA2 PyruvateLactate LDHA OGDH IDH2 ACO2 Glutamate Acetyl CoA Citrate Isocitrate α-ketoglutarate Succinyl CoA Krebs cycle Succinate Malate Fumarate FH MDH2 Oxaloacetate SDHA SUCLA2 G6PD Glucose FASN ProteinRNA >1.5× >2× >1.2× Higher in CIMP (P<0.01) Lower in CIMP (P<0.01) CIMP-Associated PRCC vs Type 1 PRCC AMPK The New England Journal of Medicine Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission. Copyright © 2015 Massachusetts Medical Society. All rights reserved.
  • 8. n engl j med nejm.org8 The new engl and jour nal of medicine tumors), and C2c (consisting solely of CIMP- associated papillary renal-cell carcinoma) (Fig. 4A). Cluster C1 was predominantly type 1 papil- lary renal-cell carcinoma and was strongly as- sociated with gain of chromosome 7, MET muta- tion, mRNA cluster 1, and an early stage of tumor development (stage I or II) (Fig. 4A, and Fig. S14 in Supplementary Appendix 1). Cluster C2a was predominantly type 2 papillary renal- cell carcinoma and was associated with an early stage of tumor development and DNA methyla- tion cluster 2. Cluster C2b consisted exclusively of type 2 and unclassified papillary renal-cell carcinomas and was associated with DNA meth- ylation cluster 1, a later stage of tumor develop- ment (stage III or IV), and mutation of SETD2. The CIMP-associated tumor subtype that was observed previously in DNA-methylation analysis was preserved as cluster C2c. Patients with clus- ter C1 or cluster C2a tumors had the highest probability of overall survival, patients with cluster C2b tumors had a lower probability, and patients with cluster C2c tumors had the lowest probability (Fig. 4B). NRF2-ARE Pathway in Type 2 Tumors Pathway analysis was performed to compare the microRNA and mRNA signatures of type 1 tu- mors with those of type 2 tumors (Fig. S15, S16, and S17 in Supplementary Appendix 1, and Sup- plementary Appendixes 7, 8, and 9), and data on mRNA expression highlighted the NRF2-ARE pathway as a distinguishing feature of type 2 tumors (Fig. S17A in Supplementary Appendix 1). Expression of NQO1, a gene activated by the NRF2-ARE pathway,29 was lowest in cluster C1, intermediate in clusters C2a and C2b, and high- est in the CIMP cluster C2c (P = 1×10−18 by analy- sis of variance) (Fig. S18A in Supplementary Appendix 1), and increased NQO1 expression was associated with decreased survival (P = 0.001) (Fig. S18C in Supplementary Appendix 1). These findings are consistent with those of studies showing increased activation of the NRF2-ARE pathway in type 2 tumors and mutations in Figure 4. Multiplatform-Based Subtype Discovery in Papillary Renal-Cell Carcinoma. As shown in Panel A, integration of subtype classifica- tions from five genomic data platforms with the use of a cluster-of-clusters analysis identified four major groups of papillary renal-cell carcinoma: C1 (enriched for type 1), C2a and C2b (enriched for type 2), and C2c (represent- ing the CIMP-associated papillary renal-cell carcinomas). The heat map (center of panel) displays the subtypes defined independently by DNA methylation (pink), chromosomal copy number (black), microRNA expres- sion (blue), mRNA expression (red), and protein (RPPA) expression (green); samples with missing data for pro- tein expression are shown in gray. Clinical features as- sociated with the multiplatform-based subtypes are also shown. Panel B shows differences in overall sur- vival according to subtype. Data on survival were not available for two patients in the C1 group. Months B A Histologic Type Stage of Tumor No. of Cases 9 22 95 35 RPPA 2 microRNA 1 Copy number 2 mRNA 1 DNA methylation 2 mRNA 2 DNA methylation 1 Copy number 1 CIMP RPPA 3 microRNA 2 microRNA 3 Copy number 3 RPPA 1 microRNA 4 mRNA 3 C2c (CIMP) C2b C1 C2a ProbabilityofOverallSurvival 1.0 0.8 0.9 0.7 0.6 0.4 0.3 0.1 0.5 0.2 0.0 0 50 100 150 200 250 C2b (N=22) Censored C1 (N=93) C2a (N=35) C2c (N=9) Histologic Type Type 1 PRCC Type 2 PRCC Unclassified PRCC Stage of Tumor II III I IV P=1×10−16 for overall comparison by log-rank test C1 vs. C2a, P=0.26 C1 vs. C2b, P<0.001 C1 vs. C2c, P=1×10−22 The New England Journal of Medicine Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission. Copyright © 2015 Massachusetts Medical Society. All rights reserved.
  • 9. n engl j med nejm.org 9 Papillary Renal-Cell Carcinoma NRF2-ARE pathway genes (NFE2L2, CUL3, KEAP1, and SIRT1).12,13 Four NFE2L2 (NRF2) mutations in known activating hotspots were identified, as well as mutations in both CUL3 (five mutations) and KEAP1 (one). These mutations in NFE2L2, CUL3, and KEAP1 correlated with high levels of NQO1 expression (P<1×10−6 by t-test) but did not solely account for the observed differences in NQO1 expression among subtypes (Fig. S18A in Supplementary Appendix 1). Integrated Analysis of Low-Frequency Candidate Driver Mutations Some tumors (most relatively small) lacked high- confidence candidate cancer-driving events. Man- ual pathway analysis identified candidate driver mutations in known cancer-associated genes, such as PTEN, NRAS, KRAS, TP53, TSC2, and those in the MLL and ARID families, in an addi- tional 27% of the cases (Fig. S19A in Supplemen- tary Appendix 1, and Supplementary Appendix 10). For the remaining 37 tumors (23%), low- frequency somatic events in genes identified by HotNet2 analysis (Fig. S19 in Supplementary Appendix 1) or associated with cancer in either the PanCan21 data set15 or the Catalogue of So- matic Mutations in Cancer database were pro- posed as potential drivers and are listed in Sup- plementary Appendix 10. In comparison with the tumors with candidate cancer-driving events, the remaining 37 papillary renal-cell carcinomas showed a higher percentage of type 1 tumors (26 of 37 [70%]) (P = 0.001 by Fisher’s exact test), and most (21 of 26 [81%]) showed a gain of chromo- some 7, which includes MET. This gain of chro- mosome 7, which is seen in a number of tumors (e.g., Wilms’ tumor and papillary thyroid can- cer), could be considered a driver event, but it does not identify a specific driver. Although gain of chromosome 7 was associated with increased MET expression in papillary renal-cell carcinoma (P<0.001 by two-factor analysis of variance) (Fig. S20 in Supplementary Appendix 1), other poten- tial driver genes on chromosome 7, such as EGFR, could influence tumorigenesis. Discussion We used a comprehensive genomics approach to characterize the biologic foundation of papillary renal-cell carcinoma and found that type 1 and type 2 papillary renal-cell carcinoma are dis- tinctly different diseases and that type 2 papil- lary renal-cell carcinoma is a heterogeneous dis- ease with multiple distinct subgroups. Common driver mutations among the different subtypes were relatively rare, as had been observed in two recent studies.7,30 Molecular and phenotypic dif- ferences between type 1 and type 2 papillary renal-cell carcinoma were reflected in individual and combined analyses of various data plat- forms. The usefulness of CDKN2A alterations as an independent prognostic marker associated with type 2 tumors requires validation. This study suggests that gene fusions involving TFE3 or TFEB are underappreciated in type 2 tumors in adults and should be considered in any patient with type 2 disease. Although papillary renal- cell carcinomas with fusions involving TFE3 or TFEB are generally considered to be diseases of children and young adults,16 the mean age in our study was 52 years, and we found tumors with TFEB fusions in patients 64 and 71 years of age. The most distinct of the three type 2 sub- groups was the subgroup defined by the CIMP, which was associated with the worst overall survival. CIMP hypermethylation patterns have been observed in a number of other cancer sub- types, including glioblastoma,31 lung adenocar- cinoma,32 and gastric adenocarcinoma.33 The CIMP-associated tumors showed low levels of FH mRNA expression, and five had germline or somatic mutation of FH. Germline mutation of FH has been observed in the aggressive type 2 tumor associated with the hereditary leiomyoma- tosis and renal-cell cancer syndrome.9,34 In this syndrome, the high levels of fumarate accumu- lating from loss of fumarate hydratase enzyme activity result in impaired function of enzymes such as the TET family of enzymes, which play a role in maintaining appropriate DNA methyla- tion within the genome.35 The subgrouping of type 2 tumors according to molecular features and the presence of specific subsets of type 2 tumors, such as those with TFE3 fusions or CIMP, suggest that substratification of type 2 papillary renal-cell carcinoma according to spe- cific molecular markers may allow more accurate diagnosis that could lead to the development of mechanistic, disease-specific targeted therapies. This classification of papillary renal-cell car- cinoma could potentially have a substantial ef- fect on clinical and therapeutic management and on the design of clinical trials. Alteration of MET or gain of chromosome 7 was observed in a large percentage (81%) of type 1 tumors. Anti- The New England Journal of Medicine Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission. Copyright © 2015 Massachusetts Medical Society. All rights reserved.
  • 10. n engl j med nejm.org10 The new engl and jour nal of medicine tumor activity of an agent targeting the MET and VEGFR2 pathways has been shown in a phase 2 trial involving patients with papillary renal-cell carcinoma, with a particularly high response rate among patients who had tumors with MET mutations.36 Mutation of the Hippo pathway tumor suppressor, NF2, was observed in a number of papillary renal-cell carcinomas. This pathway has been targeted in other cancers with agents such as dasatinib, an inhibitor of the YES1 kinase that interacts with the YAP tran- scription factor that is up-regulated with Hippo pathway dysregulation.37 The CIMP-associated tumors showed a Warburg-like metabolic shift, similar to that observed in fumarate hydratase– deficient tumors in patients with the hereditary leiomyomatosis and renal-cell cancer syn- drome.11,25,26 A clinical trial targeting this meta- bolic shift in papillary renal-cell carcinoma is currently under way (ClinicalTrials.gov number, NCT01130519). Increased expression of the NRF2-ARE pathway has been observed in both hereditary and sporadic type 2 papillary renal- cell carcinomas.12 Immunohistochemical analy- sis for NQO1 could provide a valuable marker of activation of the NRF2-ARE pathway. Currently, there is intense interest in the NRF2-ARE path- way in cancer,38 and novel strategies have re- cently been developed to target this pathway.39 The identification of altered genes and path- ways provides a comprehensive foundation for an understanding of the molecular basis of pap- illary renal-cell carcinoma. This refined classifi- cation more accurately reflects the genotypic and phenotypic differences among the various types of these tumors and may lead to more ap- propriate clinical management and development of more effective forms of therapy. Supported by grants (U54 HG003273, U54 HG003067, U54 HG003079, U24 CA143799, U24 CA143835, U24 CA143840, U24 CA143843, U24 CA143845, U24 CA143848, U24 CA143858, U24 CA143866, U24 CA143867, U24 CA143882, U24 CA143883, U24 CA144025, and P30 CA016672) from the National Institutes of Health. Disclosure forms provided by the authors are available with the full text of this article at NEJM.org. Appendix The authors’ full names and academic degrees are as follows: W. Marston Linehan, M.D., Paul T. Spellman, Ph.D., Christopher J. Ricketts, Ph.D., Chad J. Creighton, Ph.D., Suzanne S. Fei, Ph.D., Caleb Davis, Ph.D., David A. Wheeler, Ph.D., Bradley A. Murray, Ph.D., Laura Schmidt, Ph.D., Cathy D. Vocke, Ph.D., Myron Peto, Ph.D., Abu Amar M. Al Mamun, Ph.D., Eve Shinbrot, Ph.D., Anurag Sethi, Ph.D., Samira Brooks, B.S., W. Kimryn Rathmell, M.D., Ph.D., Angela N. Brooks, Ph.D., Katherine A. Hoadley, Ph.D., A. Gordon Robertson, Ph.D., Denise Brooks, Ph.D., Reanne Bowlby, B.S., Sara Sadeghi, Ph.D., Hui Shen, Ph.D., Daniel J. Weisenberger, Ph.D., Moiz Bootwalla, M.S., Stephen B. Baylin, M.D., Peter W. Laird, Ph.D., Andrew D. Cherniack, Ph.D., Gordon Saksena, M.Eng., Scott Haake, M.D., Jun Li, Ph.D., Han Liang, Ph.D., Yiling Lu, M.D., Gordon B. Mills, M.D., Ph.D., Rehan Akbani, Ph.D., Mark D.M. Leiser- son, Ph.D., Benjamin J. Raphael, Ph.D., Pavana Anur, M.S., Donald Bottaro, Ph.D., Laurence Albiges, M.D., Ph.D., Nandita Barnabas, Ph.D., Toni K. Choueiri, M.D., Bogdan Czerniak, M.D., Andrew K. Godwin, Ph.D., A. Ari Hakimi, M.D., Thai H. Ho, M.D., Ph.D., James Hsieh, M.D., Michael Ittmann, M.D., William Y. Kim, M.D., Bhavani Krishnan, Ph.D., Maria J. Merino, M.S., Kenna R. Mills Shaw, Ph.D., Victor E. Reuter, M.D., Ed Reznik, Ph.D., Carl S. Shelley, D.Phil., Brian Shuch, M.D., Sabina Signoretti, M.D., Ramaprasad Srinivasan, M.D., Ph.D., Pheroze Tamboli, M.D., George Thomas, M.D., Satish Tickoo, M.D., Kenneth Burnett, M.S., Daniel Crain, M.B.A., Johanna Gardner, C.T.R., Kevin Lau, B.S., David Mallery, M.B.A., J.D., Scott Morris, P.S.M., Ph.D., Joseph D. Paulauskis, Ph.D., Robert J. Penny, M.D., Ph.D., Candace Shelton, C.C.R.A., C.C.D.M., W. Troy Shelton, M.S., P.M.P., Mark Sherman, Ph.D., Eric Thomp- son, Ph.D., Peggy Yena, B.S., Melissa T. Avedon, B.S., Jay Bowen, M.S., Julie M. Gastier‑Foster, Ph.D., Mark Gerken, B.S., Kristen M. Leraas, M.S., Tara M. Lichtenberg, B.A., Nilsa C. Ramirez, M.D., Tracie Santos, M.S., Lisa Wise, C.T.R., Erik Zmuda, Ph.D., John A. Demchok, M.S., Ina Felau, B.S., Carolyn M. Hutter, Ph.D., Margi Sheth, B.S., Heidi J. Sofia, Ph.D., M.P.H., Roy Tarnuzzer, Ph.D., Zhin- ing Wang, Ph.D., Liming Yang, Ph.D., Jean C. Zenklusen, Ph.D., Jiashan Zhang, B.A., Brenda Ayala, B.S., Julien Baboud, B.S., Sudha Chudamani, M.Phil., Jia Liu, Ph.D., Laxmi Lolla, Rashi Naresh, M.S., Todd Pihl, Ph.D., Qiang Sun, M.S., Yunhu Wan, Ph.D., Ye Wu, Ph.D., Adrian Ally, B.S., Miruna Balasundaram, B.S., Saianand Balu, M.S., Rameen Beroukhim, M.D., Ph.D., Tom Bodenheimer, M.S., Christian Buhay, B.S., Yaron S.N. Butterfield, B.S., Rebecca Carlsen, M.S., Scott L. Carter, Ph.D., Hsu Chao, D.V.M., Eric Chuah, B.S., Amanda Clarke, T.D., Kyle R. Covington, Ph.D., Mahmoud Dahdouli, B.Sc., Ninad Dewal, Ph.D., Noreen Dhalla, B.S., Harsha V. Dod- dapaneni, Ph.D., Jennifer A. Drummond, B.A., Stacey B. Gabriel, Ph.D., Richard A. Gibbs, Ph.D., Ranabir Guin, B.S., Walker Hale, B.A., Alicia Hawes, B.S., D. Neil Hayes, M.D., M.P.H., Robert A. Holt, Ph.D., Alan P. Hoyle, B.S., Stuart R. Jefferys, Ph.D., Steven J.M. Jones, Ph.D., Corbin D. Jones, Ph.D., Divya Kalra, M.S., Christie Kovar, B.S., Lora Lewis, B.S., Jie Li, M.D., Ph.D., Yussanne Ma, Ph.D., Mar- co A. Marra, Ph.D., Michael Mayo, B.S., Shaowu Meng, Ph.D., Matthew Meyerson, M.D., Ph.D., Piotr A. Mieczkowski, Ph.D., Richard A. Moore, Ph.D., Donna Morton, M.B.A., Lisle E. Mose, B.S., Andrew J. Mungall, Ph.D., Donna Muzny, M.S., Joel S. Parker, Ph.D., Charles M. Perou, Ph.D., Jeffrey Roach, Ph.D., Jacqueline E. Schein, M.S., Steven E. Schumacher, M.S., Yan Shi, Ph.D., Janae V. Simons, B.S., Payal Sipahimalani, M.S., Tara Skelly, B.S., Matthew G. Soloway, B.S., Carrie Sougnez, B.S., Angela Tam, B.S., Donghui Tan, M.S., Nina Thiessen, M.S., Umadevi Veluvolu, M.Phil., Min Wang, Ph.D., Matthew D. Wilkerson, Ph.D., Tina Wong, B.S., Junyuan Wu, M.S., Liu Xi, M.S., Jane Zhou, Jason Bedford, B.S., Fengju Chen, Ph.D., Yao Fu, Ph.D., Mark Gerstein, Ph.D., David Haussler, Ph.D., Katayoon Kasaian, B.S., Phillip Lai, B.S., Shiyun Ling, Ph.D., Amie Radenbaugh, B.S., David Van Den Berg, Ph.D., John N. Weinstein, M.D., Ph.D., Jingchun Zhu, Ph.D., Monique Albert, M.Sc., Iakovina Alexopoulou, M.D., Jeremiah J. Andersen, M.D., J. Todd Auman, Ph.D., John Bartlett, Ph.D., Sheldon Bastacky, M.D., Julie Bergsten, R.N., Michael L. Blute, M.D., Lori Boice, N.S., Roni J. Bollag, M.D., Ph.D., Jeff Boyd, Ph.D., Erik Castle, M.D., Ying‑Bei Chen, M.D., John C. Cheville, M.D., Erin Curley, M.B.A., Benjamin Davies, M.D., April DeVolk, B.A., Rajiv Dhir, M.B., B.S., Laura Dike, Ph.D., John Eckman, P.A. (A.S.C.P.), Jay Engel, M.D., Jodi Harr, B.S., Ronald Hre- binko, M.D., Mei Huang, Ph.D., Lori Huelsenbeck‑Dill, Mary Iacocca, M.D., Bruce Jacobs, M.D, M.P.H., Michael Lobis, M.D., Jodi K. The New England Journal of Medicine Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission. Copyright © 2015 Massachusetts Medical Society. All rights reserved.
  • 11. n engl j med nejm.org 11 Papillary Renal-Cell Carcinoma Maranchie, M.D., Scott McMeekin, M.D., Jerome Myers, M.D., Joel Nelson, M.D., Jeremy Parfitt, M.D., Anil Parwani, M.D., Ph.D., Nicholas Petrelli, M.D., Brenda Rabeno, M.B.A., Somak Roy, M.D., Andrew L. Salner, M.D., Joel Slaton, M.D., Melissa Stanton, M.D., R. Houston Thompson, M.D., Leigh Thorne, M.D., Kelinda Tucker, B.A., B.S.N., Paul M. Weinberger, M.D., Cynthia Winemiller, Leigh Anne Zach, and Rosemary Zuna, M.D. References 1. Linehan WM, Srinivasan R, Schmidt LS. The genetic basis of kidney cancer: a metabolic disease. Nat Rev Urol 2010;​7:​ 277-85. 2. Linehan WM. Genetic basis of kidney cancer: role of genomics for the develop- ment of disease-based therapeutics. Ge- nome Res 2012;​22:​2089-100. 3. Delahunt B, Eble JN. Papillary renal cell carcinoma: a clinicopathologic and immunohistochemical study of 105 tu- mors. Mod Pathol 1997;​10:​537-44. 4. Zbar B, Tory K, Merino MJ, et al. He- reditary papillary renal cell carcinoma. J Urol 1994;​151:​561-6. 5. Schmidt LS, Duh FM, Chen F, et al. Germline and somatic mutations in the tyrosine kinase domain of the MET proto- oncogene in papillary renal carcinomas. Nat Genet 1997;​16:​68-73. 6. Schmidt LS, Junker K, Nakaigawa N, et al. Novel mutations of the MET proto- oncogene in papillary renal carcinomas. Oncogene 1999;​18:​2343-50. 7. Durinck S, Stawiski EW, Pavía-Jimé- nez A, et al. Spectrum of diverse genomic alterations define non-clear cell renal car- cinoma subtypes. Nat Genet 2015;​47:​13- 21. 8. Launonen V, Vierimaa O, Kiuru M, et al. Inherited susceptibility to uterine leio- myomas and renal cell cancer. Proc Natl Acad Sci U S A 2001;​98:​3387-92. 9. Grubb RL III, Franks ME, Toro J, et al. Hereditary leiomyomatosis and renal cell cancer: a syndrome associated with an ag- gressive form of inherited renal cancer. J Urol 2007;​177:​2074-9. 10. Tomlinson IP, Alam NA, Rowan AJ, et al. Germline mutations in FH predispose to dominantly inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer. Nat Genet 2002;​30:​406-10. 11. Sudarshan S, Sourbier C, Kong HS, et al. Fumarate hydratase deficiency in renal cancer induces glycolytic addiction and HIF-1 alpha stabilization by glucose-de- pendent generation of reactive oxygen species. Mol Cell Biol 2009;​15:​4080-90. 12. Ooi A, Wong JC, Petillo D, et al. An antioxidant response phenotype shared between hereditary and sporadic type 2 papillary renal cell carcinoma. Cancer Cell 2011;​20:​511-23. 13. Ooi A, Dykema K, Ansari A, et al. CUL3 and NRF2 mutations confer an NRF2 activation phenotype in a sporadic form of papillary renal cell carcinoma. Cancer Res 2013;​73:​2044-51. 14. Jiang F, Richter J, Schraml P, et al. Chromosomal imbalances in papillary re- nal cell carcinoma: genetic differences be- tween histological subtypes. Am J Pathol 1998;​153:​1467-73. 15. Lawrence MS, Stojanov P, Mermel CH, et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Na- ture 2014;​505:​495-501. 16. Kauffman EC, Ricketts CJ, Rais-Bah- rami S, et al. Molecular genetics and cel- lular features of TFE3 and TFEB fusion kidney cancers. Nat Rev Urol 2014;​11:​465- 75. 17. Schmidt LS, Junker K, Weirich G, et al. Two North American families with he- reditary papillary renal carcinoma and identical novel mutations in the MET proto-oncogene. Cancer Res 1998;​58:​ 1719-22. 18. Boezio AA, Berry L, Albrecht BK, et al. Discovery and optimization of potent and selective triazolopyridazine series of c- Met inhibitors. Bioorg Med Chem Lett 2009;​19:​6307-12. 19. Muratani M, Deng N, Ooi WF, et al. Nanoscale chromatin profiling of gastric adenocarcinoma reveals cancer-associat- ed cryptic promoters and somatically ac- quired regulatory elements. Nat Commun 2014;​5:​4361. 20. Gan HK, Cvrljevic AN, Johns TG. The epidermal growth factor receptor variant III (EGFRvIII): where wild things are al- tered. FEBS J 2013;​280:​5350-70. 21. Cancer Genome Atlas Research Net- work. Comprehensive molecular charac- terization of clear cell renal cell carcino- ma. Nature 2013;​499:​43-9. 22. Shen H, Laird PW. Interplay between the cancer genome and epigenome. Cell 2013;​153:​38-55. 23. Whitfield ML, Sherlock G, Saldanha AJ, et al. Identification of genes periodi- cally expressed in the human cell cycle and their expression in tumors. Mol Biol Cell 2002;​13:​1977-2000. 24. Harris AL. Hypoxia — a key regula- tory factor in tumour growth. Nat Rev Cancer 2002;​2:​38-47. 25. Tong WH, Sourbier C, Kovtunovych G, et al. The glycolytic shift in fumarate- hydratase-deficient kidney cancer lowers AMPK levels, increases anabolic propen- sities and lowers cellular iron levels. Can- cer Cell 2011;​20:​315-27. 26. Yang Y, Lane AN, Ricketts CJ, et al. Metabolic reprogramming for producing energy and reducing power in fumarate hydratase null cells from hereditary leio- myomatosis renal cell carcinoma. PLoS One 2013;​8(8):​e72179. 27. Cancer Genome Atlas Network. Com- prehensive molecular portraits of human breast tumours. Nature 2012;​490:​61-70. 28. Hoadley KA, Yau C, Wolf DM, et al. Multiplatform analysis of 12 cancer types reveals molecular classification within and across tissues of origin. Cell 2014;​ 158:​929-44. 29. Venugopal R, Jaiswal AK. Nrf1 and Nrf2 positively and c-Fos and Fra1 nega- tively regulate the human antioxidant re- sponse element-mediated expression of NAD(P)H:quinone oxidoreductase1 gene. Proc Natl Acad Sci U S A 1996;​93:​14960-5. 30. Kovac M, Navas C, Horswell S, et al. Recurrent chromosomal gains and het- erogeneous driver mutations characterise papillary renal cancer evolution. Nat Commun 2015;​6:​6336. 31. Noushmehr H, Weisenberger DJ, Diefes K, et al. Identification of a CpG is- land methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell 2010;​17:​510-22. 32. Cancer Genome Atlas Research Net- work. Comprehensive molecular profiling of lung adenocarcinoma. Nature 2014;​ 511:​543-50. 33. Cancer Genome Atlas Research Net- work. Comprehensive molecular charac- terization of gastric adenocarcinoma. Nature 2014;​513:​202-9. 34. Linehan WM, Rouault TA. Molecular pathways: fumarate hydratase-deficient kidney cancer — targeting the Warburg effect in cancer. Clin Cancer Res 2013;​19:​ 3345-52. 35. Xiao M, Yang H, Xu W, et al. Inhibi- tion of α-KG-dependent histone and DNA demethylases by fumarate and succinate that are accumulated in mutations of FH and SDH tumor suppressors. Genes Dev 2012;​26:​1326-38. 36. Choueiri TK, Vaishampayan U, Rosen- berg JE, et al. Phase II and biomarker study of the dual MET/VEGFR2 inhibitor foretinib in patients with papillary renal cell carcinoma. J Clin Oncol 2013;​31:​ 181-6. 37. Johnson R, Halder G. The two faces of Hippo: targeting the Hippo pathway for regenerative medicine and cancer treat- ment. Nat Rev Drug Discov 2014;​13:​63-79. 38. Sporn MB, Liby KT. NRF2 and cancer: the good, the bad and the importance of context. Nat Rev Cancer 2012;​12:​564-71. 39. Sourbier C, Ricketts CJ, Matsumoto S, et al. Targeting ABL1-mediated oxidative stress adaptation in fumarate hydratase- deficient cancer. Cancer Cell 2014;​26:​840- 50. Copyright © 2015 Massachusetts Medical Society. The New England Journal of Medicine Downloaded from nejm.org on November 23, 2015. For personal use only. No other uses without permission. Copyright © 2015 Massachusetts Medical Society. All rights reserved.