throbber
ARTICLE
`
`OPEN
`
`doi:10.1038/nature12634
`
`Mutational landscape and significance
`across 12 major cancer types
`
`Cyriac Kandoth1*, Michael D. McLellan1*, Fabio Vandin2, Kai Ye1,3, Beifang Niu1, Charles Lu1, Mingchao Xie1, Qunyuan Zhang1,3,
`Joshua F. McMichael1, Matthew A. Wyczalkowski1, Mark D. M. Leiserson2, Christopher A. Miller1, John S. Welch4,5,
`Matthew J. Walter4,5, Michael C. Wendl1,3,6, Timothy J. Ley1,3,4,5, Richard K. Wilson1,3,5, Benjamin J. Raphael2 & Li Ding1,3,4,5
`
`The Cancer Genome Atlas (TCGA) has used the latest sequencing and analysis methods to identify somatic variants across
`thousands of tumours. Here we present data and analytical results for point mutations and small insertions/deletions
`from 3,281 tumours across 12 tumour types as part of the TCGA Pan-Cancer effort. We illustrate the distributions of
`mutation frequencies, types and contexts across tumour types, and establish their links to tissues of origin, environmental/
`carcinogen influences, and DNA repair defects. Using the integrated data sets, we identified 127 significantly mutated genes
`from well-known (for example, mitogen-activated protein kinase, phosphatidylinositol-3-OH kinase, Wnt/b-catenin and
`receptor tyrosine kinase signalling pathways, and cell cycle control) and emerging (for example, histone, histone
`modification, splicing, metabolism and proteolysis) cellular processes in cancer. The average number of mutations in
`these significantly mutated genes varies across tumour types; most tumours have two to six, indicating that the number of
`driver mutations required during oncogenesis is relatively small. Mutations in transcriptional factors/regulators show
`tissue specificity, whereas histone modifiers are often mutated across several cancer types. Clinical association analysis
`identifies genes having a significant effect on survival, and investigations of mutations with respect to clonal/subclonal
`architecture delineate their temporal orders during tumorigenesis. Taken together, these results lay the groundwork for
`developing new diagnostics and individualizing cancer treatment.
`
`The advancement of DNA sequencing technologies now enables the
`processing of thousands of tumours of many types for systematic
`mutation discovery. This expansion of scope, coupled with appreciable
`progress in algorithms1–5, has led directly to characterization of signifi-
`cant functional mutations, genes and pathways6–18. Cancer encompasses
`more than 100 related diseases19, making it crucial to understand the
`commonalities and differences among various types and subtypes.
`TCGA was founded to address these needs, and its large data sets
`are providing unprecedented opportunities for systematic, integrated
`analysis.
`We performed a systematic analysis of 3,281 tumours from 12 cancer
`types to investigate underlying mechanisms of cancer initiation and
`progression. We describe variable mutation frequencies and contexts
`and their associations with environmental factors and defects in DNA
`repair. We identify 127 significantly mutated genes (SMGs) from diverse
`signalling and enzymatic processes. The finding of a TP53-driven
`breast, head and neck, and ovarian cancer cluster with a dearth of other
`mutations in SMGs suggests common therapeutic strategies might be
`applied for these tumours. We determined interactions among muta-
`tions and correlated mutations in BAP1, FBXW7 and TP53 with det-
`rimental phenotypes across several cancer types. The subclonal structure
`and transcription status of underlying somatic mutations reveal the
`trajectory of tumour progression in patients with cancer.
`
`Standardization of mutation data
`Stringent filters (Methods) were applied to ensure high quality muta-
`tion calls for 12 cancer types: breast adenocarcinoma (BRCA), lung
`adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC),
`uterine corpus endometrial carcinoma (UCEC), glioblastoma multiforme
`
`(GBM), head and neck squamous cell carcinoma (HNSC), colon and
`rectal carcinoma (COAD, READ), bladder urothelial carcinoma (BLCA),
`kidney renal clear cell carcinoma (KIRC), ovarian serous carcinoma
`(OV) and acute myeloid leukaemia (LAML; conventionally called
`AML) (Supplementary Table 1). A total of 617,354 somatic mutations,
`consisting of 398,750 missense, 145,488 silent, 36,443 nonsense, 9,778
`splice site, 7,693 non-coding RNA, 523 non-stop/readthrough, 15,141
`frameshift insertions/deletions (indels) and 3,538 inframe indels, were
`included for downstream analyses (Supplementary Table 2).
`
`Distinct mutation frequencies and sequence context
`Figure 1a shows that AML has the lowest median mutation frequency
`and LUSC the highest (0.28 and 8.15 mutations per megabase (Mb),
`respectively). Besides AML, all types average over 1 mutation per Mb,
`substantially higher than in paediatric tumours20. Clustering21 illus-
`trates that mutation frequencies for KIRC, BRCA, OV and AML are
`normally distributed within a single cluster, whereas other types have
`several clusters (for example, 5 and 6 clusters in UCEC and COAD/
`READ, respectively) (Fig. 1a and Supplementary Table 3a, b). In UCEC,
`the largest patient cluster has a frequency of approximately 1.5 muta-
`tions per Mb, and the cluster with the highest frequency is more than
`150 times greater. Multiple clusters suggest that factors other than age
`contribute to development in these tumours14,16. Indeed, there is a
`significant correlation between high mutation frequency and DNA
`repair pathway genes (for example, PRKDC, TP53 and MSH6) (Sup-
`plementary Table 3c). Notably, PRKDC mutations are associated with
`high frequency in BLCA, COAD/READ, LUAD and UCEC, whereas
`TP53 mutations are related with higher frequencies in AML, BLCA,
`BRCA, HNSC, LUAD, LUSC and UCEC (all P , 0.05). Mutations in
`
`1The Genome Institute, Washington University in St Louis, Missouri 63108, USA. 2Department of Computer Science, Brown University, Providence, Rhode Island 02912, USA. 3Department of Genetics,
`Washington University in St Louis, Missouri 63108, USA. 4Department of Medicine, Washington University in St Louis, Missouri 63108, USA. 5Siteman Cancer Center, Washington University in St Louis,
`Missouri 63108, USA. 6Department of Mathematics, Washington University in St Louis, Missouri 63108, USA.
`*These authors contributed equally to this work.
`
`1 7 O C T O B E R 2 0 1 3 | V O L 5 0 2 | N A T U R E | 3 3 3
`Macmillan Publishers Limited. All rights reserved
`
`©2013
`
`Genome Ex. 1021
`Page 1 of 20
`
`

`

`of C.T transitions are between 59% and 67%, substantially higher
`than the approximately 40% in other cancer types. Higher frequencies
`of transition mutations at CpG in gastrointestinal tumours, including
`colorectal, were previously reported22. We found three additional
`cancer types (GBM, AML and UCEC) clustered in the C.T mutation
`at CpG, consistent with previous findings of aberrant DNA methyla-
`tion in endometrial cancer23 and glioblastoma24. BLCA has a unique
`signature for C.T transitions compared to the other types (enriched
`for TC) (Extended Data Fig. 1).
`
`Significantly mutated genes
`Genes under positive selection, either in individual or multiple tumour
`types, tend to display higher mutation frequencies above background.
`Our statistical analysis3, guided by expression data and curation (Methods),
`identified 127 such genes (SMGs; Supplementary Table 4). These SMGs
`are involved in a wide range of cellular processes, broadly classified
`into 20 categories (Fig. 2), including transcription factors/regulators,
`histone modifiers, genome integrity, receptor tyrosine kinase signal-
`ling, cell cycle, mitogen-activated protein kinases (MAPK) signalling,
`phosphatidylinositol-3-OH kinase (PI(3)K) signalling, Wnt/b-catenin
`signalling, histones, ubiquitin-mediated proteolysis, and splicing (Fig. 2).
`The identification of MAPK, PI(3)K and Wnt/b-catenin signalling path-
`ways is consistent with classical cancer studies. Notably, newer categories
`(for example, splicing, transcription regulators, metabolism, proteolysis
`and histones) emerge as exciting guides for the development of new
`therapeutic targets. Genes categorized as histone modifiers (Z 5 0.57),
`PI(3)K signalling (Z 5 1.03), and genome integrity (Z 5 0.66) all relate
`to more than one cancer type, whereas transcription factor/regulator
`(Z 5 0.40), TGF-b signalling (Z 5 0.66), and Wnt/b-catenin signalling
`(Z 5 0.55) genes tend to associate with single types (Methods).
`Notably, 3,053 out of 3,281 total samples (93%) across the Pan-
`Cancer collection had at least one non-synonymous mutation in at
`least one SMG. The average number of point mutations and small
`indels in these genes varies across tumour types, with the highest (,6
`mutations per tumour) in UCEC, LUAD and LUSC, and the lowest
`(,2 mutations per tumour) in AML, BRCA, KIRC and OV. This
`suggests that the numbers of both cancer-related genes (only 127
`identified in this study) and cooperating driver mutations required
`during oncogenesis are small (most cases only had 2–6) (Fig. 3),
`although large-scale structural rearrangements were not included in
`this analysis.
`
`Common mutations
`The most frequently mutated gene in the Pan-Cancer cohort is TP53
`(42% of samples). Its mutations predominate in serous ovarian (95%)
`and serous endometrial carcinomas (89%) (Fig. 2). TP53 mutations
`are also associated with basal subtype breast tumours. PIK3CA is the
`second most commonly mutated gene, occurring frequently (.10%)
`in most cancer types except OV, KIRC, LUAD and AML. PIK3CA
`mutations frequented UCEC (52%) and BRCA (33.6%), being speci-
`fically enriched in luminal subtype tumours. Tumours lacking PIK3CA
`mutations often had mutations in PIK3R1, with the highest occur-
`rences in UCEC (31%) and GBM (11%) (Fig. 2).
`Many cancer types carried mutations in chromatin re-modelling
`genes. In particular, histone-lysine N-methyltransferase genes (MLL2
`(also known as KMT2D), MLL3 (KMT2C) and MLL4 (KMT2B)) clus-
`ter in bladder, lung and endometrial cancers, whereas the lysine (K)-
`specific demethylase KDM5C is prevalently mutated in KIRC (7%).
`Mutations in ARID1A are frequent in BLCA, UCEC, LUAD and
`LUSC, whereas mutations in ARID5B predominate in UCEC (10%)
`(Fig. 2).
`KRAS and NRAS mutations are typically mutually exclusive, with
`recurrent activating mutations (KRAS (Gly 12), KRAS (Gly 13) and
`NRAS (Gln 61)) common in COAD/READ (30%, 5% and 5%, respect-
`ively), UCEC (15%, 4% and 2%) and LUAD (24%, 1% and 2%). EGFR
`mutations are frequent in GBM (27%) and LUAD (11%). Recurrent,
`
`RESEARCH ARTICLE
`
`AML BRCA OV
`
`KIRC UCEC GBM COAD/
`READ
`
`HNSC BLCA LUAD LUSC
`
`C>T
`
`C>G
`
`C>A
`
`A>T
`
`A>G
`
`A>C
`
`A>G
`
`A>T
`
`100
`
`1
`
`a
`
`Number of mutations per Mb
`
`0.01
`
`100
`75
`50
`25
`0
`
`b
`
`Ti/Tv frequency (%)
`
`c
`
`A>C
`
`C>A
`
`C>G
`
`C>T
`
`Correlation coefficient
`
`0.75 0.85 0.95
`
`Figure 1 | Mutation frequencies, spectra and contexts across 12 cancer
`types. a, Distribution of mutation frequencies across 12 cancer types. Dashed
`grey and solid white lines denote average across cancer types and median for
`each type, respectively. b, Mutation spectrum of six transition (Ti) and
`transversion (Tv) categories for each cancer type. c, Hierarchically clustered
`mutation context (defined by the proportion of A, T, C and G nucleotides
`within 62 bp of variant site) for six mutation categories. Cancer types
`correspond to colours in a. Colour denotes degree of correlation: yellow
`(r 5 0.75) and red (r 5 1).
`
`POLQ and POLE associate with high frequencies in multiple cancer types;
`POLE association in UCEC is consistent with previous observations14.
`Comparison of spectra across the 12 types (Fig. 1b and Supplemen-
`tary Table 3d) reveals that LUSC and LUAD contain increased C.A
`transversions, a signature of cigarette smoke exposure10. Sequence
`context analysis across 12 types revealed the largest difference being
`in C.T transitions and C.G transversions (Fig. 1c). The frequency
`of thymine 1-bp (base pair) upstream of C.G transversions is mark-
`edly higher in BLCA, BRCA and HNSC than in other cancer types
`(Extended Data Fig. 1). GBM, AML, COAD/READ and UCEC have
`similar contexts in that the proportions of guanine 1 base downstream
`
`3 3 4 | N A T U R E | V O L 5 0 2 | 1 7 O C T O B E R 2 0 1 3
`©2013
`Macmillan Publishers Limited. All rights reserved
`
`Genome Ex. 1021
`Page 2 of 20
`
`

`

`ARTICLE RESEARCH
`
`KRAS
`NF1
`MAP3K1
`BRAF
`NRAS
`MAP2K4
`MAPK8IP1
`PIK3CA
`PTEN
`PIK3R1
`TLR4
`PIK3CG
`AKT1
`SMAD4
`TGFBR2
`ACVR1B
`SMAD2
`ACVR2A
`APC
`CTNNB1
`AXIN2
`TBL1XR1
`SOX17
`HIST1H1C
`H3F3C
`HIST1H2BD
`FBXW7
`KEAP1
`SPOP
`SF3B1
`U2AF1
`PCBP1
`CDH1
`AJUBA
`DNMT3A
`TET2
`IDH1
`IDH2
`NFE2L2
`NFE2L3
`PPP2R1A
`PTPN11
`RPL22
`RPL5
`MTOR
`STK11
`NAV3
`NOTCH1
`LRRK2
`MALAT1
`ARHGAP35
`POLQ
`NCOR1
`USP9X
`NPM1
`HGF
`EPPK1
`AR
`LIFR
`PRX
`CRIPAK
`EGR3
`B4GALT3
`MIR142
`
`Pan−Cancer
`
`UCEC
`
`OV
`
`LUSC
`
`LUAD
`
`AML
`
`KIRC
`
`HNSC
`
`GBM
`
`COAD/READ
`
`BRCA
`
`BLCA
`
`0.0 0.8 45.1 0.7 0.3 0.2 4.0 26.3 1.2 0.6 20.0 6.7
`7.1 2.5 1.0 11.0 2.7 1.7 1.0 11.8 10.3 3.8 3.5 4.4
`3.1 7.2 0.0 2.1 1.0 1.2 0.0 1.8 1.7 0.3 3.5 2.7
`2.0 0.4 3.6 2.1 1.0 0.2 0.0 6.6 4.6 0.6 0.9 1.5
`2.0 0.1 8.8 0.3 0.0 0.0 7.5 1.8 0.6 0.6 2.6 1.5
`0.0 4.1 2.6 0.0 0.3 0.0 0.0 1.3 0.6 0.3 1.3 1.4
`2.0 0.3 2.1 0.7 0.7 0.5 0.0 1.8 1.2 0.3 0.4 0.7
`17.4 33.6 17.6 11.0 20.6 2.9 0.0 4.4 14.9 0.6 52.2 17.8
`3.1 3.8 1.0 30.7 1.3 4.3 0.0 2.2 8.1 0.6 63.5 9.7
`1.0 2.5 2.1 11.4 1.7 0.5 0.0 1.3 0.6 0.3 30.9 4.4
`2.0 1.2 0.0 0.3 2.0 0.5 0.5 11.4 5.8 1.0 0.4 1.9
`2.0 0.4 0.5 2.4 2.7 0.7 0.0 5.3 7.5 1.0 1.3 1.7
`0.0 2.5 0.0 0.3 0.7 0.5 0.0 0.0 0.6 0.0 1.3 0.9
`2.0 0.4 9.8 0.3 2.0 0.5 0.0 3.1 2.9 0.0 0.0 1.4
`3.1 0.4 2.6 0.7 3.0 0.2 0.0 0.9 1.7 1.0 1.3 1.1
`0.0 0.7 3.6 0.0 1.3 1.0 0.0 2.2 1.2 0.3 1.7 1.0
`1.0 0.5 5.7 0.0 1.0 0.5 0.0 0.9 1.2 0.0 1.3 0.9
`1.0 0.5 2.6 0.0 0.7 0.2 0.0 0.9 1.2 0.0 0.4 0.6
`4.1 0.5 81.9 0.3 4.0 1.4 0.0 9.2 4.0 2.2 5.7 7.3
`2.0 0.1 4.7 0.3 0.7 0.2 0.0 3.5 1.7 0.6 28.3 2.9
`3.1 0.1 3.6 0.3 1.7 0.2 0.0 0.9 0.6 0.3 2.6 0.9
`2.0 1.1 0.0 0.0 1.0 0.7 0.0 2.2 1.2 0.3 1.3 0.8
`0.0 0.0 0.5 0.3 0.3 0.0 0.0 0.4 0.0 0.0 3.0 0.3
`1.0 0.4 1.0 0.7 1.3 0.2 0.0 0.4 0.6 1.3 0.0 0.6
`0.0 0.0 0.0 0.7 0.7 0.0 0.0 1.8 1.2 0.0 0.9 0.4
`1.0 0.0 0.0 0.0 1.3 0.0 0.0 0.0 1.2 0.3 2.6 0.4
`9.2 0.8 11.4 0.3 5.0 0.2 0.0 1.3 5.2 1.0 11.7 3.0
`3.1 0.1 0.0 0.0 4.0 0.5 0.0 17.1 12.1 0.3 1.3 2.6
`1.0 0.1 0.0 0.0 1.0 0.0 0.0 0.4 0.6 0.3 6.5 0.7
`4.1 1.8 1.0 0.7 0.7 1.0 0.5 2.2 2.3 0.0 2.2 1.3
`1.0 0.3 0.5 0.0 1.3 0.0 4.0 2.6 0.0 0.0 0.9 0.8
`1.0 0.0 2.6 0.0 0.0 0.2 0.0 0.4 0.0 0.0 0.9 0.3
`5.1 7.0 0.5 0.3 1.3 0.5 0.0 1.3 1.7 0.3 3.0 2.5
`2.0 0.1 0.0 0.3 6.0 0.5 0.0 0.9 0.0 0.0 0.0 0.8
`0.0 0.5 1.0 0.0 1.7 1.2 25.5 4.0 4.0 1.0 1.3 2.8
`3.1 0.4 0.0 0.7 0.3 1.9 8.5 3.1 2.3 0.0 2.2 1.6
`3.1 0.3 0.0 5.2 0.3 0.5 9.5 0.9 1.2 0.0 0.9 1.5
`0.0 0.0 1.6 0.0 0.0 0.0 10.0 0.4 0.0 0.0 0.4 0.8
`9.2 0.1 0.0 0.0 5.3 1.2 0.0 2.2 14.9 0.0 5.2 2.3
`3.1 0.8 0.0 0.3 1.3 0.2 0.0 0.0 2.3 0.3 1.7 0.8
`1.0 0.1 1.6 0.0 1.3 1.2 0.0 1.3 4.6 1.3 8.7 1.5
`0.0 0.1 1.0 1.7 0.3 0.2 4.5 2.6 1.7 0.3 0.9 1.0
`0.0 0.0 0.0 0.3 0.7 0.5 0.0 0.4 0.0 0.0 10.9 1.0
`0.0 0.4 0.0 2.8 0.0 1.4 0.0 0.4 1.2 0.0 0.9 0.7
`2.0 1.4 3.6 1.4 1.3 6.0 0.0 7.5 4.6 1.9 5.2 3.0
`0.0 0.3 0.0 0.0 0.3 0.2 0.0 8.8 1.7 0.0 0.4 0.9
`5.1 1.4 2.1 1.0 7.3 1.4 0.0 21.5 19.0 1.3 5.2 4.6
`5.1 0.4 0.0 0.0 19.3 1.0 0.5 3.1 8.1 0.6 1.7 3.1
`5.1 0.7 2.6 1.0 5.0 1.4 0.0 6.6 11.5 2.9 3.5 2.8
`15.3 1.1 0.0 0.0 6.3 1.9 0.0 9.7 5.8 1.0 0.0 2.7
`5.1 0.9 0.5 0.7 3.7 1.2 0.5 4.0 5.8 1.6 10.0 2.5
`7.1 0.8 0.5 1.0 4.3 1.2 0.0 5.7 9.2 1.0 3.9 2.4
`8.2 3.9 0.5 0.7 3.3 0.7 0.0 2.6 3.5 0.3 1.3 2.2
`3.1 1.2 0.0 0.7 4.3 1.0 0.5 5.3 4.6 0.3 6.5 2.1
`0.0 0.0 0.0 0.3 0.3 0.0 27.0 0.9 0.0 0.0 0.4 1.8
`1.0 0.5 0.0 0.3 2.7 0.2 0.0 10.5 5.8 0.6 1.3 1.7
`2.0 0.3 0.0 2.8 2.7 0.7 0.0 3.1 4.0 0.3 3.0 1.4
`1.0 0.7 2.1 0.0 2.3 0.5 0.0 1.8 3.5 0.3 3.5 1.2
`1.0 0.8 5.2 0.0 2.7 0.5 0.0 0.9 1.7 0.6 1.7 1.2
`5.1 0.5 0.5 0.7 1.7 1.2 0.5 0.4 1.2 0.3 1.3 0.9
`2.0 0.3 0.0 0.3 1.0 0.5 0.0 5.3 0.0 0.0 0.4 0.7
`1.0 0.3 0.0 0.3 0.0 0.2 0.0 0.4 1.2 0.0 0.4 0.3
`0.0 0.1 0.5 0.0 0.0 0.2 0.0 0.0 1.2 0.0 0.9 0.2
`0.0 0.1 0.0 0.0 0.0 0.0 2.0 0.0 0.0 0.0 0.0 0.2
`
`MAPK signalling
`
`PI(3)K signalling
`
`TGF-β signalling
`
`Wnt/β-catenin
`signalling
`
`Histone
`
`Proteolysis
`
`Splicing
`
`HIPPO signalling
`
`DNA methylation
`
`Metabolism
`
`NFE2L
`
`Protein phosphatase
`
`Ribosome
`
`TOR signalling
`Other
`
`VHL
`GATA3
`TSHZ3
`EP300
`CTCF
`TAF1
`TSHZ2
`RUNX1
`MECOM
`TBX3
`SIN3A
`WT1
`EIF4A2
`FOXA1
`PHF6
`CBFB
`SOX9
`ELF3
`VEZF1
`CEBPA
`FOXA2
`MLL3
`MLL2
`ARID1A
`PBRM1
`SETD2
`NSD1
`SETBP1
`KDM5C
`KDM6A
`MLL4
`ARID5B
`ASXL1
`EZH2
`TP53
`ATM
`ATRX
`BRCA2
`ATR
`STAG2
`BAP1
`BRCA1
`SMC1A
`SMC3
`CHEK2
`RAD21
`ERCC2
`EGFR
`FLT3
`EPHA3
`ERBB4
`PDGFRA
`EPHB6
`FGFR2
`KIT
`FGFR3
`CDKN2A
`RB1
`CDK12
`CDKN1B
`CCND1
`CDKN1A
`CDKN2C
`
`Pan−Cancer
`
`UCEC
`
`OV
`
`LUSC
`
`LUAD
`
`AML
`
`KIRC
`
`HNSC
`
`GBM
`
`COAD/READ
`
`BRCA
`
`BLCA
`
`0.0 0.0 0.0 0.0 0.0 52.3 0.0 0.0 0.6 0.0 0.9 6.9
`1.0 10.6 1.0 0.0 2.0 0.0 0.0 2.6 2.9 0.3 0.4 3.2
`2.0 0.7 3.1 0.7 1.3 1.2 0.5 14.9 6.3 1.0 3.9 2.6
`17.4 0.8 2.1 0.3 8.0 1.4 0.0 0.9 4.6 0.3 5.2 2.5
`2.0 2.4 1.6 0.0 3.3 0.5 0.5 1.3 0.0 0.3 16.5 2.4
`2.0 1.1 1.6 1.4 2.3 1.2 0.0 4.0 6.9 1.6 8.7 2.3
`4.1 0.9 3.1 2.4 1.3 0.7 0.0 6.6 3.5 1.0 1.7 1.8
`1.0 3.3 1.0 0.0 0.7 0.0 9.0 0.4 0.0 0.0 1.3 1.6
`5.1 0.5 1.0 1.4 1.7 1.0 0.0 3.5 4.6 0.6 3.0 1.5
`3.1 2.4 1.0 0.0 0.7 0.0 0.0 4.4 2.9 1.0 1.3 1.4
`1.0 0.5 0.5 0.7 0.7 0.5 0.0 1.8 2.9 0.6 5.2 1.1
`0.0 0.1 1.0 0.7 0.0 0.7 6.0 3.5 2.3 0.0 0.4 1.0
`2.0 0.5 2.6 0.0 0.0 0.7 0.0 1.8 1.2 0.6 1.3 0.8
`4.1 1.7 0.0 1.0 0.7 0.0 0.0 0.4 0.6 0.0 0.0 0.8
`3.1 0.4 0.0 0.3 0.3 0.5 3.0 0.9 1.2 0.3 1.3 0.8
`1.0 2.1 0.0 0.0 0.0 0.2 1.0 0.4 0.6 0.0 0.4 0.7
`0.0 0.1 4.2 1.0 0.7 0.7 0.0 1.3 0.6 0.0 0.4 0.7
`8.2 0.1 3.6 0.0 0.3 0.0 0.0 0.4 0.0 0.3 0.4 0.6
`2.0 0.9 0.0 0.7 0.7 0.0 0.0 0.9 1.7 0.0 0.0 0.6
`0.0 0.0 0.0 0.0 0.0 0.2 6.5 0.0 0.6 0.0 0.0 0.5
`1.0 0.0 0.5 0.0 0.0 0.0 0.0 0.0 0.0 0.6 4.8 0.5
`24.5 6.4 2.6 3.1 7.3 3.6 0.5 18.4 15.5 1.9 5.2 6.6
`25.5 1.6 1.6 1.7 17.9 3.1 0.5 8.8 20.1 0.6 8.3 5.9
`27.6 2.0 5.7 0.7 3.0 2.9 0.5 6.1 6.3 1.0 30.0 5.4
`6.1 0.4 0.0 0.7 2.3 32.9 0.0 1.8 3.5 0.3 2.6 5.4
`6.1 1.2 2.6 1.7 2.3 11.5 0.5 7.9 2.9 1.9 2.6 3.6
`6.1 0.3 0.5 0.3 10.6 1.0 0.0 3.1 5.2 0.6 5.7 2.4
`2.0 0.4 1.6 1.4 3.0 1.4 1.0 12.7 5.2 0.0 2.2 2.2
`1.0 0.5 0.5 0.7 1.0 6.5 0.0 4.8 2.9 1.9 2.2 2.0
`26.5 1.1 0.0 1.0 2.7 1.0 1.5 0.9 4.0 0.0 0.9 2.0
`7.1 0.7 2.1 2.1 2.7 1.0 0.0 1.8 4.0 0.3 8.3 2.0
`3.1 0.4 0.0 0.3 3.3 0.7 0.0 2.2 1.7 0.6 9.6 1.6
`3.1 0.4 1.6 0.0 3.0 1.0 2.5 1.3 5.2 0.0 0.9 1.3
`1.0 0.1 0.0 1.0 0.3 0.7 1.5 2.2 2.3 0.0 1.7 0.8
`50.0 32.9 58.6 28.3 69.8 2.2 7.5 51.8 79.3 94.6 27.8 42.0
`11.2 2.1 5.7 1.4 2.7 2.9 0.0 7.9 4.0 1.3 6.5 3.3
`8.2 1.2 1.0 5.5 4.3 1.9 0.0 6.1 5.8 0.6 3.0 2.8
`6.1 1.7 1.6 1.4 3.7 1.9 0.0 5.7 5.8 3.2 4.4 2.7
`4.1 0.8 2.1 1.4 5.3 1.2 0.0 5.7 4.0 0.6 7.0 2.4
`10.2 0.9 1.0 4.1 0.7 1.7 3.0 2.6 3.5 1.0 3.9 2.2
`4.1 0.3 0.0 0.7 1.0 10.1 0.0 1.3 0.6 0.6 2.2 2.0
`4.1 1.6 0.0 1.0 2.7 1.0 0.0 3.5 5.2 3.5 0.9 1.9
`3.1 0.8 1.6 1.7 1.0 0.5 3.5 1.3 0.6 1.3 4.4 1.5
`1.0 0.4 0.0 1.4 1.7 1.2 3.5 2.6 2.3 0.3 0.4 1.2
`2.0 0.4 0.0 1.7 2.3 0.7 0.0 0.9 1.2 0.3 1.3 0.9
`2.0 0.5 1.0 0.3 1.0 0.0 2.5 2.6 1.2 0.3 0.9 0.9
`12.2 0.1 0.5 0.0 0.3 0.2 0.0 1.3 0.0 0.3 0.4 0.7
`1.0 0.7 1.6 26.6 4.7 1.7 1.0 11.4 2.9 1.9 1.3 4.6
`2.0 0.4 0.0 1.7 0.7 0.5 26.5 4.0 4.0 1.0 0.9 2.7
`1.0 0.5 3.1 1.0 3.7 0.5 0.5 8.8 6.3 1.0 2.2 2.1
`2.0 0.8 3.6 0.3 4.3 1.4 0.0 7.5 5.2 0.0 2.6 2.1
`6.1 0.4 1.0 3.8 1.0 1.4 0.5 6.6 4.0 1.0 1.3 1.9
`3.1 0.4 0.0 1.4 1.3 1.2 0.0 9.7 3.5 0.3 1.7 1.6
`2.0 0.9 0.0 0.3 0.7 0.2 0.0 3.1 2.3 0.0 10.4 1.5
`1.0 0.5 1.0 1.0 1.0 0.7 4.0 1.8 3.5 1.9 2.2 1.4
`8.2 0.1 0.5 1.4 1.7 1.4 0.0 0.4 2.3 0.3 0.4 1.0
`4.1 0.0 0.5 0.7 21.3 1.0 0.0 6.6 14.9 0.0 0.4 3.6
`14.3 1.8 0.5 8.3 3.0 0.2 0.0 5.3 6.9 1.9 3.9 3.2
`4.1 0.9 1.6 0.3 1.7 1.4 0.0 3.1 0.6 2.9 2.2 1.5
`2.0 0.9 1.0 0.3 0.7 0.0 0.0 1.8 0.0 0.3 0.9 0.7
`2.0 0.1 0.0 0.0 0.3 0.0 0.0 0.9 0.6 0.0 5.2 0.6
`12.2 0.0 0.0 0.3 0.0 0.2 0.0 0.4 1.2 0.3 0.0 0.6
`0.0 0.0 0.0 1.0 0.0 0.2 0.0 0.0 0.6 0.6 0.0 0.2
`
`Transcription
`factor/regulator
`
`Histone modifier
`
`Genome integrity
`
`RTK signalling
`
`Cell cycle
`
`Figure 2 | The 127 SMGs from 20 cellular processes in cancer identified in
`12 cancer types. Percentages of samples mutated in individual tumour types
`
`and Pan-Cancer are shown, with the highest percentage in each gene among 12
`cancer types in bold.
`
`gain-of-function mutations in IDH1 (Arg 132) and/or IDH2 (Arg 172)
`typify GBM and AML (Supplementary Table 2 and Fig. 2). Although
`KRAS residues Gly 12 and Gly 13 are commonly mutated in LUAD,
`COAD/READ and UCEC, the proportion of Gly12Cys changes is
`significantly higher in lung cancer (P , 3.2 3 10210), resulting from
`the high C.A transversion rate (Extended Data Fig. 2).
`
`Tumour-type-specific mutations
`Signature mutations exclusive to KIRC include those affecting VHL
`(52%) and PBRM1 (33%) (Fig. 2). Mutations to BAP1 (10%) and
`SETD2 (12%) are also most common in KIRC. Transcription factor
`CTCF, ribosome component RPL22, and histone modifiers ARID1A
`and ARID5B have the highest frequencies in UCEC. Predominant
`COAD/READ-specific mutations are those affecting APC (82%) and
`
`Wnt/b-catenin signalling (93% of samples). Several mutations occur
`exclusively in AML, including recurrent mutations in NPM1 (27%)
`and FLT3 (27%), and rare mutations affecting MIR142 (Fig. 2).
`Mutations of methylation and chromatin modifiers are also typical
`in AML, mostly affecting DNMT3A and TET2. BRCA-specific muta-
`tions include GATA3 and MAP3K1, whereas KEAP1 mutations pre-
`dominate in lung cancer (LUAD 17%, LUSC 12%). EPHA3 (9%),
`SETBP1 (13%) and STK11 (9%) are characteristic in LUAD.
`
`Shared and cancer type-specific mutation signatures
`Cluster analysis on mutations in SMGs (Fig. 4 and Extended Data Fig. 3)
`showed 72% (1,881 of 2,611) of tumours are adjacent to those from the
`same tissue type. Notably, clustering identified several dominant groups
`in UCEC, COAD, GBM, AML, KIRC, OV and BRCA. Two major
`
`1 7 O C T O B E R 2 0 1 3 | V O L 5 0 2 | N A T U R E | 3 3 5
`Macmillan Publishers Limited. All rights reserved
`
`©2013
`
`Genome Ex. 1021
`Page 3 of 20
`
`

`

`Mutual exclusivity and co-occurrence among SMGs
`Pairwise exclusivity and co-occurrence analysis for the 127 SMGs
`found 14 mutually exclusive (false discovery rate (FDR) , 0.05) and
`148 co-occurring (FDR , 0.05) pairs (Supplementary Table 6). TP53
`and CDH1 are exclusive in BRCA, with mutations enriched in different
`subtypes13, as are TP53 and CTNNB1 in UCEC. Cohort analysis iden-
`tified pairs where at least one gene has mutations strongly associated
`(corrected P , 0.05) to one cancer type, and also identifies TP53 and
`PIK3CA with significant exclusivity (Extended Data Fig. 4). Pairs with
`significant co-occurrence include IDH1 and ATRX in GBM29, TP53
`and CDKN2A in HNSC, and TBX3 and MLL4 in LUAD.
`Dendrix30 identified a set of five genes (TP53, PTEN, VHL, NPM1
`and GATA3) having strong mutual exclusivity (P , 0.01) (Extended
`Data Fig. 5a and Supplementary Table 7). Not surprisingly, many are
`associated (P , 0.05) with one cancer type (for example, VHL muta-
`tions in KIRC), demonstrating a strong relationship between exclus-
`ivity and tissue of origin. When 600 non-cancer-type-specific genes
`were added to the analysis (Methods), we identified another set con-
`sisting of TP53, PIK3CA, PIK3R1, SETD2 and WT1 (P , 0.01;
`Extended Data Fig. 5b and Supplementary Table 7). Dendrix also
`finds genes with strong mutual exclusivity from each cancer type
`separately (Extended Data Fig. 5c), allowing calculation of ‘cancer
`exclusivity’. KIRC has the strongest exclusivity from the other 11
`cancer types, followed by AML with clear exclusivity from BRCA
`and UCEC. Conversely, COAD/READ displayed the greatest co-
`occurrence with other cancer types (Extended Data Fig. 5d).
`
`Clinical correlation across tumour types
`We examined how clinical features (Supplementary Table 8) correlate
`with somatic events in 127 SMGs within tumour types. Some findings
`are unsurprising, such as the correlation of TP53 mutations with gene-
`rally unfavourable indicators, for example in tumour stage (P 5 0.01,
`Fisher’s exact test) and elapsed time to death (P 5 0.006, Wilcoxon) in
`HNSC, age (P 5 0.002, Wilcoxon rank test) and time to death (P 5
`0.09, Wilcoxon) in AML, and vital status in OV (P 5 0.04, Fisher). In
`UCEC, mutations in several genes are correlated with the endometrioid
`rather than serous subtype: PTEN, CTNNB1, PIK3R1, KRAS, ARID1A,
`CTCF, RPL22 and ARID5B (all P , 0.03) (Supplementary Table 9).
`
`RESEARCH ARTICLE
`
`12
`
`10
`
`02468
`
`Number of non-synonymous mutations in SMGs
`
`OV
`
`BRCA
`
`GBM
`
`KIRC
`
`AML
`
`(n = 316)
`
`(n = 763)
`
`(n = 290)
`
`(n = 471)
`
`(n = 200)
`
`COAD/
`READ
`(n = 193)
`
`HNSC
`
`LUAD
`
`LUSC
`
`BLCA
`
`UCEC
`
`(n = 301)
`
`(n = 228)
`
`(n = 174)
`
`(n = 98)
`
`(n = 230)
`
`Figure 3 | Distribution of mutations in 127 SMGs across Pan-Cancer
`cohort. Box plot displays median numbers of non-synonymous mutations,
`with outliers shown as dots. In total, 3,210 tumours were used for this analysis
`(hypermutators excluded).
`
`endometrial endometroid clusters were found, one having mutations
`in PIK3CA, PTEN and ARID1A, and the other containing mutations in
`two additional genes (PIK3R1 and CTNNB1). Five major breast cancer
`clusters were observed, with mutations in CDH1, GATA3, MAP3K1,
`PIK3CA and TP53 as drivers for respective clusters. The TP53-driven
`cluster is adjacent to an HNSC cluster and an ovarian cancer cluster, all
`having a dearth of other SMG mutations (Fig. 4). The glioblastoma
`cluster is characterized by mutations in EGFR. Two kidney clear cell
`cancer clusters were detected; both have VHL as the common driver
`and one has additional mutations in PBRM1 and/or BAP1 (refs 25–27).
`PBRM1 and BAP1 mutations are mutually exclusive in KIRC (P 5
`0.006), consistent with previous reports26,28. AML has three major clus-
`ters represented by various combinations of DNMT3A, NPM1 and
`FLT3 mutations, and one cluster dominated by RUNX1 mutations.
`One cluster having APC and KRAS mutations was almost exclusively
`detected in COAD/READ. Tumours from BLCA, HNSC, LUAD and
`LUSC are largely scattered over the Pan-Cancer cohort, indicating
`extensive heterogeneity in these diseases.
`
`BLCA
`
`BRCA
`
`COAD/READ
`
`GBM
`
`HNSC
`
`KIRC
`
`AML
`
`LUAD
`
`LUSC
`
`OV
`
`UCEC
`
`TP53
`PIK3CA
`PTEN
`PBRM1
`VHL
`KRAS
`APC
`MLL3
`EGFR
`ARID1A
`CTNNB1
`PIK3R1
`MLL2
`NF1
`GATA3
`SETD2
`MAP3K1
`NOTCH1
`CDKN2A
`ATM
`RB1
`CDH1
`NAV3
`FBXW7
`DNMT3A
`NPM1
`FLT3
`MTOR
`BAP1
`IDH1
`ATRX
`BRCA2
`CTCF
`NCOR1
`STAG2
`TAF1
`RUNX1
`
`Figure 4 | Unsupervised clustering based on mutation status of SMGs.
`Tumours having no mutation or more than 500 mutations were excluded. A
`mutation status matrix was constructed for 2,611 tumours. Major clusters of
`
`mutations detected in UCEC, COAD, GBM, AML, KIRC, OV and BRCA were
`highlighted. Complete gene list shown in Extended Data Fig. 3.
`
`3 3 6 | N A T U R E | V O L 5 0 2 | 1 7 O C T O B E R 2 0 1 3
`©2013
`Macmillan Publishers Limited. All rights reserved
`
`Genome Ex. 1021
`Page 4 of 20
`
`

`

`ARTICLE RESEARCH
`
`We examined which genes correlate with survival using the Cox
`proportional hazards model, first analysing individual cancer types
`using age and gender as covariates; an average of 2 genes (range: 0–4)
`with mutation frequency $2% were significant (P # 0.05) in each
`type (Supplementary Table 10a and Extended Data Fig. 6). KDM6A
`and ARID1A mutations correlate with better survival
`in BLCA
`(P 5 0.03, hazard ratio (HR) 5 0.36, 95% confidence interval (CI):
`0.14–0.92) and UCEC (P 5 0.03, HR 5 0.11, 95% CI: 0.01–0.84),
`respectively, but mutations in SETBP1, recently identified with worse
`prognosis in atypical chronic myeloid leukaemia (aCML)31, have a
`significant detrimental effect in HNSC (P 5 0.006, HR 5 3.21, 95% CI:
`1.39–7.44). BAP1 strongly correlates with poor survival (P 5 0.00079,
`HR 5 2.17, 95% CI: 1.38–3.41) in KIRC. Conversely, BRCA2 muta-
`tions (P 5 0.02, HR 5 0.31, 95% CI: 0.12–0.85) associate with better
`survival in ovarian cancer, consistent with previous reports32,33; BRCA1
`mutations showed positive correlation with better survival, but did not
`reach significance here.
`We extended our survival analysis across cancer types, restricting
`our attention to the subset of 97 SMGs whose mutations appeared
`in $2% of patients having survival data in $2 tumour types. Taking
`type, age and gender as covariates, we found 7 significant genes: BAP1,
`DNMT3A, HGF, KDM5C, FBXW7, BRCA2 and TP53 (Extended Data
`Table 1). In particular, BAP1 was highly significant (P 5 0.00013,
`
`HR 5 2.20, 95% CI: 1.47–3.29, more than 53 mutated tumours out
`of 888 total), with mutations associating with detrimental outcome in
`four tumour types and notable associations in KIRC (P 5 0.00079),
`consistent with a recent report28, and in UCEC (P 5 0.066). Mutations in
`several other genes are detrimental, including DNMT3A (HR 5 1.59),
`previously identified with poor prognosis in AML34, and KDM5C
`(HR 5 1.63), FBXW7 (HR 5 1.57) and TP53 (HR 5 1.19). TP53 has
`significant associations with poor outcome in KIRC (P 5 0.012), AML
`(P 5 0.0007) and HNSC (P 5 0.00007). Conversely, BRCA2 (P 5 0.05,
`HR 5 0.62, 95% CI: 0.38 to 0.99) correlates with survival benefit in six
`types, including OV and UCEC (Supplementary Table 10a, b). IDH1
`mutations are associated with improved prognosis across the Pan-
`Cancer set (HR 5 0.67, P 5 0.16) and also in GBM (HR 5 0.42,
`P 5 0.09) (Supplementary Table 10a, b), consistent with previous work35.
`
`Driver mutations and tumour clonal architecture
`To understand the temporal order of somatic events, we analysed the
`variant allele fraction (VAF) distribution of mutations in SMGs across
`AML, BRCA and UCEC (Fig. 5a and Supplementary Table 11a) and
`other tumour types (Extended Data Fig. 7). To minimize the effect of
`copy number alterations, we focused on mutations in copy neutral
`segments. Mutations in TP53 have higher VAFs on average in all three
`cancer types, suggesting early appearance during tumorigenesis, although
`
`UCEC
`
`100
`
`75
`
`50
`
`VAF
`
`25
`
`0
`
`BRCA
`
`ChrX
`
`100
`
`75
`
`50
`
`VAF
`
`25
`
`0
`
`a
`
`100
`
`AML
`
`75
`
`50
`
`VAF
`
`25
`
`0
`
`TP53
`SOX17
`PPP2R1A
`ARID1A
`PTEN
`CCND1
`FBXW7
`FGFR2
`FOXA2
`PIK3CA
`RPL22
`PIK3R1
`SPOP
`CTNNB1
`MECOM
`CTCF
`ARHGAP35
`TAF1
`KRAS
`HIST1H2BD
`ARID5B
`MLL4
`AR
`NRAS
`ATR
`SIN3A
`NFE2L2
`USP9X
`
`AKT1
`TP53
`MAP2K4
`NCOR1
`TBX3
`ARID1A
`FOXA1
`CDH1
`RB1
`PIK3CA
`PTEN
`MAP3K1
`CTCF
`SF3B1
`GATA3
`RUNX1
`NF1
`KRAS
`MLL3
`PIK3R1
`TBL1XR1
`
`PHF6
`SMC1A
`SMC3
`TP53
`DNMT3A
`U2AF1
`IDH2
`STAG2
`RUNX1
`IDH1
`WT1
`RAD21
`CEBPA
`FLT3
`KRAS
`TET2
`PTPN11
`KIT
`NRAS
`
`AML TCGA-AB-2968
`
`VAF
`
`b
`
`BRCA TCGA-BH-A18P
`
`VAF
`
`UCEC TCGA-B5-A0JV
`
`VAF
`
`80
`
`100
`
`0
`
`20
`
`40
`
`60
`
`80
`
`100
`
`0
`
`20
`
`40
`
`60
`
`80
`
`100
`
`2 copies
`Model fit
`Component fits
`
`Density (a.u.)
`
`2 copies
`Model fit
`Component fits
`
`Density (a.u.)
`
`2 copies
`Model fit
`Component fits
`
`0
`
`20
`
`40
`
`60
`
`Density (a.u.)
`
`5
`5
`
`2
`2
`
`6
`6
`
`40
`5 CTCF (R566H)
`6 PIK3CA (E542K)
`7 PMS2 (N432S)
`
`60
`
`80
`
`100
`
`Cluster 1
`Cluster 2
`
`ChrX variants
`Autosome
`variants
`
`17
`17
`
`3
`3
`
`4
`4
`
`2,000
`1,000
`500
`200
`100
`50
`20
`10
`5
`20
`0
`1 NRAS (Q61K)
`2 ARID1A (R693*)
`3 PTEN (P96T)
`4 KRAS (G12D)
`
`Tumour coverage
`
`20
`10
`5
`20
`0
`1 FOXA1 (I176M)
`2 PIK3R1 (E635K)
`3 MLL3 (E1992Q)
`
`40
`Cluster 1
`Cluster 2
`
`80
`
`60
`ChrX variants
`Autosome variants
`
`100
`
`Tumour coverage
`
`
`
`33
`
`
`
`22
`
`40
`Cluster 1
`Cluster 2
`Cluster 3
`Cluster 4
`
`80
`60
`ChrX variants
`Autosome variants
`
`100
`
`
`
`11
`
`2,000
`1,000
`500
`200
`100
`50
`20
`10
`5
`20
`0
`1 NRAS (Q61H)
`2 DNMT3A (E477*)
`3 DNMT3A (A741B)
`
`Tumour coverage
`
`2,000
`1,000
`500
`
`200
`100
`50
`
`2
`2
`
`3
`3
`
`1
`1
`
`Figure 5 | Driver initiation and progression mutations and tumour clonal
`architecture. a, Variant allele fraction (VAF) distribution of mutations in
`SMGs across tumours from AML, BRCA and UCEC for mutations ($203
`coverage) in copy neutral segments. SMGs having $5 mutation data points
`were included. ChrX, chromosome X. b, In AML sample TCGA-AB-2968
`(WGS), two DNMT3A mutations are in the founding clone, and one NRAS
`
`mutation is in the subclone. In BRCA tumour TCGA-BH-A18P (exome), one
`FOXA1 mutation is in the founding clone, and PIK3R1 and MLL3 mutations
`are in the subclone. In UCEC tumour TCGA-B5-A0JV (exome), PIK3CA,
`ARID1A and CTCF mutations are in the founding clone, and NRAS, PTEN and
`KRAS mutations are in the secondary clone. Asterisk denotes stop codon.
`
`1 7 O C T O B E R 2 0 1 3 | V O L 5 0 2 | N A T U R E | 3 3 7
`Macmillan Publishers Limited. All rights reserved
`
`©2013
`
`Genome Ex. 1021
`Page 5 of 20
`
`

`

`RESEARCH ARTICLE
`
`it is possible that a later mutation contributing to tumour cell expan-
`sion might have a high VAF. It is worth noting that copy neutral loss of
`heterozygosity is commonly found in classical tumour suppressors
`such as TP53, BRCA1, BRCA2 and PTEN, leading to increased VAFs
`in these genes. In AML, DNMT3A (permutation test P 5 0), RUNX1
`(P 5 0.0003) and SMC3 (P 5 0.05) have significantly higher VAFs
`than average among SMGs (Fig. 5a and Supplementary Table 11b).
`In breast cancer, AKT1, CBFB, MAP2K4, ARID1A, FOXA1 and PIK3CA
`have relatively high average VAFs. For endometrial cancer, multiple
`SMGs (for example, PIK3CA, PIK3R1, PTEN, FOXA2 and ARID1A)
`have similar median VAFs. Conversely, KRAS and/or NRAS mutations
`tend to have lower VAFs in all three tumour types (Fig. 5a), suggesting
`NRAS (for example, P 5 0 in AML) and KRAS (for example, P 5 0.02
`in BRCA) have a

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket