throbber
RAPID COMMUNICATION
`
`Human Mutation
`
`OFFICIAL JOURNAL
`
`IDH1 Mutations at Residue p.R132 (IDH1R132) Occur
`Frequently in High-Grade Gliomas But Not in Other Solid
`Tumors
`
`www.hgvs.org
`
`Fonnet E. Bleeker,1,2 Simona Lamba,2 Sieger Leenstra,3,4 Dirk Troost,5 Theo Hulsebos,6 W. Peter Vandertop,1,7
`Milo Frattini,8,9 Francesca Molinari,9 Margaret Knowles,10 Aniello Cerrato,11 Monica Rodolfo,8 Aldo Scarpa,12
`Lara Felicioni,13 Fiamma Buttitta,13 Sara Malatesta,13 Antonio Marchetti,13 and Alberto Bardelli2,14
`1Neurosurgical Center Amsterdam, Location Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
`2Laboratory of Molecular Genetics, The OncoGenomics Center, Institute for Cancer Research and Treatment, University of Torino, Medical
`School, Candiolo, Italy
`3Department of Neurosurgery, St. Elisabeth Ziekenhuis, Tilburg, The Netherlands
`4Department of Neurosurgery, Erasmus Medical Center, Rotterdam, The Netherlands
`5Departments of Neuropathology, The Netherlands
`6Department of Neurogenetics, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
`7Neurosurgical Center Amsterdam, Location VU University Medical Center, Amsterdam, The Netherlands
`8Department of Experimental Oncology, Istituto Nazionale Tumori; Milan, Italy
`9Laboratory of Molecular Diagnostic Institute of Pathology, Locarno, Switzerland
`10Section of Experimental Oncology, Leeds Institute for Molecular Medicine, Leeds, United Kingdom
`11Institute of Endocrinology and Experimental Oncology, National Council of Research, Naples, Italy
`12Department of Pathology, Section of Anatomic Pathology, University of Verona, Verona, Italy
`13Clinical Research Center, Center of Excellence on Aging, University-Foundation, Chieti, Italy
`14FIRC Institute of Molecular Oncology, Milan, Italy
`
`Communicated by Richard Wooster
`Received 1 October 2008; accepted revised manuscript 10 October 2008.
`Published online 31 December 2008 in Wiley InterScience (www.interscience.wiley.com). DOI 10.1002/humu.20937
`
`ABSTRACT: Systematic sequence profiling of the Glio-
`blastoma Multiforme (GBM) genome has recently led to
`the identification of somatic mutations in the isocitrate
`dehydrogenase 1 (IDH1) gene. Interestingly, only the
`evolutionarily conserved residue R132 located in the
`substrate binding site of IDH1 was found mutated in
`GBM. At present, the occurrence and the relevance of
`p.R132 (IDH1R132) variants in tumors other than GBMs
`is largely unknown. We searched for mutations at
`position R132 of the IDH1 gene in a panel of 672 tumor
`samples. These included high-grade glioma, gastrointest-
`inal stromal tumors (GIST), melanoma, bladder, breast,
`colorectal, lung, ovarian, pancreas, prostate, and thyroid
`carcinoma specimens. In addition, we assessed a panel of
`84 cell
`lines from different tumor lineages. Somatic
`mutations affecting the IDH1R132 residue were detected
`
`Additional Supporting Information may be found in the online version of this
`
`article.
`
`
`Correspondence to: Alberto Bardelli, Laboratory of Molecular Genetics, Institute
`
`for Cancer Research and Treatment, University of Torino; Medical School, Str prov
`
`142Km 3.95; Candiolo (TO), 10060, Italy. E-mail: a.bardelli@unito.it.
`
`Contract grant sponsor: The Italian Association for Cancer Research (AIRC; A.B.);
`
`Italian Ministry of Health, Regione Piemonte (A.B.); Italian Ministry of University and
`
`Research, CRT Progetto Alfieri (A.B.); Fondazione Monte dei Paschi di Siena, Siena,
`
`Italy (A.S.); Association for International Cancer Research (AICR-UK; A.B.) and EU
`
`FP6; Grant number: 037297 (A.B.).
`
`in 20% (23 of 113) high-grade glioma samples. In
`addition to the previously reported p.R132H and
`p.R132S alleles, we identified three novel
`somatic
`mutations (p.R132C, p.R132G, and p.R132L) affecting
`residue IDH1R132 in GBM. Strikingly, no IDH1 muta-
`tions were detected in the other tumor types. These data
`indicate that cancer mutations affecting IDH1R132 are
`tissue-specific, and suggest that it plays a unique role in
`the development of high-grade gliomas.
`Hum Mutat 30, 7–11, 2009.
`& 2008 Wiley-Liss, Inc.
`
`KEY WORDS: cancer; somatic mutation; IDH1; GBM;
`HGG
`
`Introduction
`
`tumor genomes is taking an
`The molecular profiling of
`enormous spurt these days. Genome-wide sequencing analyses
`have been performed in colorectal and breast cancer (Sjo¨blom
`et al., 2006; Wood et al., 2007), and most recently the same
`approach has been performed in pancreatic ductal adenocarcino-
`ma (PDAC) (Jones et al., 2008) and glioblastoma multiforme
`(GBM) (Parsons et al., 2008). These mutational efforts have led to
`the identification of novel somatic mutations in genes that had
`not been previously linked to tumorigenesis. Of particular interest
`
`& 2008 WILEY-LISS, INC.
`
`Rigel Exhibit 1017
`Page 1 of 5
`
`

`

`screen revealed 12% somatic
`the GBM mutational
`that
`is
`mutations in the IDH1 gene (MIM# 147700) (Parsons et al.,
`2008). The IDH1 mutations were found predominantly in the
`group of secondary GBMs and younger patients. Furthermore, the
`patients with mutated IDH1 had a significantly longer survival
`(Parsons et al., 2008). Other tumor type datasets analyzed for this
`gene have been relatively small thus far, and the reported mutation
`frequencies are generally low (0 of 11 breast, 1 of 11 colon) (Wood
`et al., 2007) (http://www.sanger.ac.uk/cosmic).
`IDH1 encodes isocitrate dehydrogenase 1 (Geisbrecht and
`Gould, 1999), an enzyme that catalyzes the oxidative decarboxy-
`lation of isocitrate to a-ketoglutarate (Koshland et al., 1985). This
`reaction leads to NADPH production, and is thought to play a
`role in the cellular control of oxidative damage (Lee et al., 2002).
`IDH1 is
`localized within the cytoplasm and peroxisomes
`(Geisbrecht and Gould, 1999). Two different mutations in IDH1
`have been described in GBM, both affecting the amino acid
`arginine at position 132 and leading to amino acid residue
`substitutions (p.R132H and p.R132S). R132 is evolutionarily
`highly conserved, and is localized in the substrate binding site of
`IDH1, where hydrophilic interactions between R132 and both the
`a- and b-carboxylate of isocitrate are formed (Xu et al., 2004). The
`IDH1p:R132H and p:R132S changes might affect these interactions and
`its enzymatic activity (Parsons et al., 2008). In this study we have
`investigated the mutational status of IDH1 in 672 tumor samples
`and 84 cancer cell lines.
`
`Materials and Methods
`
`High-grade glioma (HGG; WHO grade III and grade IV) tumor
`samples (GBM, anaplastic astrocytoma, and anaplastic oligoden-
`droglioma) and the matched normal DNA samples were obtained
`from the tumor bank maintained by the Departments of
`
`the Academic Medical
`Neurosurgery and Neuropathology at
`Center (Amsterdam, The Netherlands). DNA of melanoma,
`colorectal cancer, and gastrointestinal stromal tumors (GIST)
`samples was obtained from the Department of Experimental
`Oncology at the Istituto Nazionale Tumouri (Milan, Italy). DNA
`of PDAC xenografts was obtained from the Department of
`Pathology, Section of Anatomic Pathology at the University of
`Verona (Verona, Italy). DNA of breast, lung, ovarian, and thyroid
`(papillary carcinoma) cancer samples was obtained from the
`Clinical Research Center, Center of Excellence on Aging at the
`University-Foundation (Chieti, Italy). Additional DNA samples of
`thyroid carcinomas (medullary histotype), extracted from frozen
`tissues, were obtained from the Department of Cellular Biology
`and Molecular Pathology at the University of Naples (Naples,
`Italy). DNA of bladder cancer samples was obtained from the
`Section of Experimental Oncology at the Leeds Institute for
`Molecular Medicine (Leeds, UK). Tumor databases are listed in
`Table 1, and have been previously validated by showing that
`somatic mutations in common cancer genes could be detected at
`the expected frequencies.
`In addition, a panel consisting of 84 cell lines from multiple
`tumor lineages was screened for IDH1R132
`mutations. Cell line
`details are shown in Supp. Table S1. The NCI-60 panel (the 60
`human cancer cell lines of the National Cancer Institute) was
`obtained from ATCC (Middlesex, UK). In addition, 16 astro-
`cytoma cell lines were included: the cell lines CCF-STTG1, Hs683,
`U87MG, U118MG, U251MG, U373MG, T98G (ATCC, Middlesex,
`UK), GAMG (Deutsche Sammlung von Mikroorganismen und
`Zellkulturen, Braunschweig, Germany), SKMG-3 (a gift of Dr. C.Y.
`Thomas, University of Virginia Division of Hematology/Oncol-
`ogy, Charlottesville, VA), D384MG, SF763 (gifts of Dr. M.L.
`Lamfers, Department of Neurosurgery, Free University, Amster-
`dam, The Netherlands), SF126 (a gift of Dr. C. Van Bree,
`
`Table 1.
`
`IDH1R132 mutations are specific for high grade gliomas
`
`Tumor type
`
`Histotype
`
`Number of samples analysed
`
`Number of mutated samples
`
`P-value (Fisher’s exact test)
`
`High grade glioma
`
`Bladder
`Breast
`
`Colorectal
`GIST
`Lung
`
`Melanoma
`Thyroid
`
`Ovary
`Pancreas
`Prostate
`
`Total
`GBM primary
`GBM secondary
`AA
`AO
`Transitional Cell
`Total
`Ductal
`Lobular
`Medullary
`Mucinous
`Adenocarcinoma
`
`Total
`Adenocarcinoma
`Carcinoid
`Small Cell
`
`Total
`Medullary
`Papillary
`Adenocarcinoma
`Ductal Adenocarcinoma
`
`113
`94
`15
`2
`2
`34
`127
`48
`45
`17
`17
`128
`25
`107
`84
`7
`16
`23
`42
`21
`21
`46
`23
`4
`
`23
`11
`11
`0
`1
`0
`0
`
`0
`0
`0
`
`0
`0
`
`0
`0
`
`0.002179
`8,03E-09
`0.000292
`0.000295
`0.041578
`0.041578
`7,26E-09
`0.014124
`8,20E-09
`0.000001
`0.343849
`0.073930
`0.013501
`0.000576
`0.023904
`0.023904
`0.000284
`0.013501
`0.584147
`
`Tumor samples, tumor type, histotype, the number of samples analysed and the number of mutated samples are indicated. In addition, P-values of the Fisher’s exact test, used
`to determine the tissue specificity for IDH1R132
`mutations in high grade gliomas, are listed. Abbreviations: AA; Anaplastic Astrocytoma, AO; Anaplastic Oligodendroglioma,
`GBM; Glioblastoma Multiforme, GIST; Gastrointestinal Stromal Tumors.
`
`8
`
`HUMAN MUTATION, Vol. 30, No. 1, 7–11, 2009
`
`Rigel Exhibit 1017
`Page 2 of 5
`
`

`

`Table 2. Mutations affecting IDH1R132 identified in high grade
`gliomas
`
`IDH1 mutation
`
`Nucleotide
`
`Amino
`Acid
`
`Number of
`mutated samples
`
`Histology of
`mutated samples
`
`Previously
`described
`
`c.394C4T p.R132C
`c.394C4G p.R132G
`c.394C4A p.R132S
`c.395G4T p.R132L
`c.395G4A p.R132H
`
`3
`1
`1
`1
`17
`
`GBM
`GBM
`AO
`GBM
`GBM
`
`yes
`no
`yes
`no
`yes
`
`The nucleotide and amino acid changes are listed alongside the number and
`histology of the mutated samples. In addition, we indicate whether the mutation has
`been described before in high grade gliomas. The nucleotide numbering uses the A of
`the ATG translation initiation start site as nucleotide 11, based on reference sequence
`NM__005896.2. All mutations are heterozygous. Abbreviations: AO; Anaplastic
`Oligodendroglioma, GBM; Glioblastoma Multiforme.
`
`Cancer cell lines in which the IDH1R132 mutations
`Table 3.
`were analyzed
`
`Tumor type
`
`Astrocytoma
`Bladder
`Breast
`Cervix
`Colon
`HNSCC (tongue)
`Kidney
`Leukemia
`Lung
`Melanoma
`Mesothelioma
`Oesophageus
`Ovary
`Prostate
`Thyroid
`
`Number of cell lines analysed
`
`20
`4
`7
`1
`9
`1
`6
`5
`9
`8
`1
`2
`8
`2
`1
`
`Cell lines are listed according to the tumor lineage from which they were originated.
`Abbreviation: HNSCC; Head and neck squamous cell carcinoma.
`
`University of Amsterdam, Laboratory for Experimental Oncology
`and Radiation Biology, Amsterdam, The Netherlands), A58 and
`A60 (gifts of Dr. A. van Tilborg and Dr. P. De Witt Hamer,
`Department of Neurosurgery, Academic Medical Center, Am-
`sterdam, The Netherlands), the xenograft cell line IGRG121 (a gift
`of Dr. B. Geoerger, Institut Gustave Roussy, Villejuif, France).
`Genomic DNA of the cell lines A1847, DU145, JAMA2, MCF7,
`ME180, MSTO-211H, NCI-H1299, NCI-H69, OE19, OE33,
`OVCA433, SCC9, SKCO1, and ZR-75-1 was provided by Dr. F.
`Di Nicolantonio (OncoGenomics Center, Institute for Cancer
`Research and Treatment, Italy). DNA from other cell lines was
`derived from our own laboratories.
`Genomic DNA was isolated as previously described (Balakrish-
`nan et al., 2007). PCR primers
`for
`the genomic region
`corresponding to IDH1 (NM_005896.2) exon 4, which encodes
`codon R132, and the flanking intronic sequences,
`including
`splicing donor and acceptor regions were designed using Primer 3
`(http://frodo.wi.mit.edu/cgi-bin/primer3/primer3_www.cgi). The
`0
`0
`primers (forward 5
`-AATGAGCTCTATATGCCATCACTG-3
`, re-
`0
`0
`0
`verse 5
`-TTCATACCTTGCTTAATGGGTGT-3
`and sequence 5
`-
`0
`GCCATCACTGCAGTTGTAGGTTA-3
`) were synthesized by In-
`vitrogen/Life Technologies, Inc. (Paisley, England). PCRs were
`
`performed in 96-well formats in 10 ml reaction volumes, contain-
`ing 0.25 mmol/l deoxynucleotide triphosphates, 1 mmol/l each of
`the forward and reverse primers, 6% DMSO, 1  PCR buffer,
`1 ng/ml DNA, and 0.05 unit/ml Platinum Taq (Invitrogen/Life
`Technologies). A touchdown PCR program was used for PCR
`amplification (Peltier Thermocycler, PTC-200, MJ Research, Bio-
`Rad Laboratories, Inc., Italy).
`PCR conditions were as follows: 941C for 2 min; three cycles of
`941C for 15 sec, 641C for 30 sec, 701C for 30 sec; three cycles
`of 941C for 15 sec, 611C for 30 sec, 701C for 30 sec; three cycles of
`941C for 15 sec, 581C for 30 sec, 701C for 30 sec; and 35 cycles of
`941C for 15 sec, 571C for 30 sec, and 701C for 30 sec, followed by
`701C for 5 min, and 121C thereafter. PCR products were purified
`using AMPure (Agencourt Bioscience Corp., Beckman Coulter
`S.p.A, Milan, Italy). Cycle sequencing was carried out using BigDye
`Terminator v3.1 Cycle Sequencing kit (Applied Biosystems, Foster
`City, CA) with an initial denaturation at 971C for 3 min, followed
`by 28 cycles of 971C for 10 sec, 501C for 20 sec, and 601C for 2 min.
`Sequencing products were purified using CleanSeq (Agencourt
`Bioscience, Beckman Coulter) and analyzed on a 3730 DNA
`Analyzer, ABI capillary electrophoresis system (Applied Biosys-
`tems). Sequence traces were analyzed using the Mutation Surveyor
`software package (SoftGenetics, State College, PA).
`tumor
`A total of 756 PCR products, spanning 367 kb of
`genomic DNA, were generated and subjected to direct sequencing.
`Changes previously described as SNPs were excluded from further
`analyses (http://www.ensembl.org/index.html). To ensure that the
`observed mutations were not PCR or
`sequencing artifacts,
`amplicons were independently reamplified and resequenced in
`the corresponding tumors. All verified tumor changes were
`resequenced in parallel with the matched normal DNA to
`distinguish between somatic mutations and SNPs not previously
`described. For samples in which mutations were found, matching
`between germ-line and tumor DNA was verified by direct
`sequencing of 26 single nucleotide polymorphism (SNP) at 24
`loci (data not shown), to ensure that the observed changes are
`somatic mutations. Nucleotide and amino acid numbering uses
`the A of the ATG translation initiation start site (codon 1) as
`nucleotide 11, based on reference sequence NM_005896.2.
`The Fisher’s exact test (http://www.langsrud.com/fisher.htm)
`was used to determine the tissue specificity of IDH1R132
`mutations
`in HGG. In this test, the absence of mutations of different tumor
`types was compared with the number of mutations found in HGG
`samples.
`
`Results and Discussion
`We determined the occurrence of IDH1R132
`sequence variants in
`a panel of 672 tumor samples. These included 113 HGG samples
`(109 GBM, 2 anaplastic astrocytoma, and 2 anaplastic oligoden-
`droglioma), 25 GIST, 23 melanoma, 34 bladder cancer, 127 breast
`cancer, 128 colorectal cancer, 107 lung cancer, 46 ovarian cancer, 4
`prostate cancer, 42 thyroid cancer, and 23 PDAC specimens
`(Table 1). In addition, a panel consisting of 84 cell lines from
`multiple tumor lineages was screened for IDH1R132
`mutations.
`Out of the 756 samples analyzed, 23 displayed heterozygous
`the IDH1 gene. Strikingly,
`mutations at position R132 of
`mutations were only found in HGG (23 out of 113 samples
`corresponding to 20%; see Table 1). In agreement with previous
`results (Parsons et al., 2008), the most common change detected
`in our GBM tumor database is the IDH1p:R132H mutation. In
`addition to the reported p.R132 H and p.R132S variants, we
`detected three novel heterozygous somatic mutations affecting
`
`HUMAN MUTATION, Vol. 30, No. 1, 7–11, 2009
`
`9
`
`Rigel Exhibit 1017
`Page 3 of 5
`
`

`

`Examples of novel somatic IDH1R132 mutations identified in GBM. Top, chromatogram of the sequence of a tumor sample; bottom,
`Figure 1.
`chromatogram of the matched normal. Arrows, location of missense somatic mutations. Nucleotide and amino acid alterations are below the
`traces. The nucleotide numbering uses the A of the ATG translation initiation start site as nucleotide 11, based on reference sequence
`NM_005896.2. Numbers above the sequences are part of the software output. A: p.R132G mutation; B: p.R132L mutation.
`
`residue R132 (p.R132C, p.R132G, and p.R132L) in GBM (Fig. 1,
`Table 2). As previously reported, most of the mutations were
`detected in glioblastomas. However, we also found a single
`mutated anaplastic oligodendroglioma. None of the other human
`solid tumor types displayed IDH1R132
`variants, and this was in
`most cases statistically significant (see Table 1). Although one
`colorectal cancer sample has been previously described to have an
`IDH1p:R132C allele (Sjo¨blom et al., 2006),
`in our set of 128
`colorectal cancer samples R132 variants were never detected.
`Cancer cell lines represent unique tools for multiple aspects of
`biomedical research including the evaluation of the functional
`relevance of cancer alleles. We therefore searched for mutations at
`position R132 of IDH1 in a panel of cancer cell lines, including 20
`astrocytoma (Table 3 and Supp. Table S1). None of the cell lines
`displayed IDH1R132
`variants. However, we found two previously
`unreported IDH1 alleles in three cell lines: p.V71I was detected in
`the plasma cell myeloma line RPMI-8226, while p.G97D was
`found in the colorectal cancer cell lines DLD-1 and HCT-15. These
`two colorectal cancer cell lines are suggested to be genetically
`identical, and therefore may be derived from the same patient
`(Chen et al., 1995). As matched normal samples are not available
`for these tumor cell lines, we cannot assess whether the nature of
`these mutations is somatic. Neither the p.V71I nor the p.G97D
`variants have been reported previously as SNPs (http://www.
`ensembl.org/index.html). Considering that we did not find these
`alleles in any of the 672 tumor samples and 84 cell lines that we
`sequenced, we suspect that they are either very rare SNPs or novel
`IDH1 somatic changes. Compared to the frequency (20%) that we
`found in HGG samples, the lack of IDH1 mutations in our panel
`of 20 high-grade astrocytoma cell
`lines appears statistically
`significant (p-value 5 0.024, Fisher’s exact test). It is possible,
`however, that GBM cell lines are predominantly derived from
`primary GBM tumors, thus explaining our results.
`GBM, WHO grade IV with predominant astrocytic differentia-
`tion, is the most common and most aggressive primary brain
`tumor (Louis et al., 2007). Most glioblastoma manifest rapidly de
`novo, without recognizable precursor lesions. These so-called
`primary GBMs typically present in middle age to elderly patients
`
`10
`
`HUMAN MUTATION, Vol. 30, No. 1, 7–11, 2009
`
`with a brief clinical history and show rapid progression and short
`survival time (Ohgaki and Kleihues, 2007; Scherer, 1940). In
`contrast, secondary GBMs are typically seen in younger patients,
`with a history of epilepsy caused by low-grade gliomas, which in
`years progress to GBM (Ohgaki and Kleihues, 2007; Scherer,
`1940). Secondary GBMs are rare (5%) in comparison to primary
`GBM (Ohgaki et al., 2004), and can only be diagnosed with
`clinical (neuroimaging) or histological evidence of evolution from
`a less malignant astrocytoma (Ohgaki and Kleihues, 2007). Both
`subtypes are considered histopathologically indistinguishable.
`However, the classification in primary and secondary is nicely
`reflected by molecular mechanisms. Primary GBMs have a high
`rate of EGFR alterations, MDM2 duplications, PTEN mutations,
`and homozygous P16INK4A
`deletions, whereas TP53 mutations are
`most prevalent in secondary GBMs (Ohgaki and Kleihues, 2007;
`IDH1 mutations
`Ohgaki
`et al., 2004). We observed the
`predominantly in secondary GBM (11 of 94 vs. 11 of 15, p-
`value 5 0.0000016, Fisher’s exact test; see Table 1), in accordance
`with the results of Parsons and colleagues (2008). In addition to
`the IDH1p:R132C=G=H=L mutations in GBM, we identified an
`IDH1p:R132S mutation in an anaplastic oligodendroglioma sample.
`Interestingly, GBM patients with an IDH1R132
`mutation have been
`reported to have a better survival (Parsons et al., 2008). No
`information on IDH1 mutations in low-grade gliomas is available
`thus far; therefore, assessment of whether lower grade gliomas
`display IDH1R132
`mutations and if they have a survival advantage
`are critical questions that should be addressed.
`In conclusion, our data support the evidence that IDH1 is a
`pivotal GBM cancer gene mutated predominantly in secondary
`glioblastomas. The identification of three novel mutations in
`IDH1 affecting amino acid R132 may allow further structural and
`functional analysis of the function of this residue on the catalytic
`activity of isocitrate dehydrogenase 1. Our most relevant finding
`entails the unique and striking tissue-specific pattern of the
`IDH1R132 mutations in human solid cancer. The tissue specificity
`of cancer mutations has been observed in multiple cancer genes
`(e.g., APC, AKT1) (Bleeker et al., 2008a; Bleeker et al., 2008b).
`Why some genes are mutated in specific tumor types remains an
`
`Rigel Exhibit 1017
`Page 4 of 5
`
`

`

`unsettled issue whose solution will be relevant for basic and
`clinical cancer research. As IDH1 is involved in a specific
`metabolic pathway, its mutations may potentially be exploited
`for therapeutic purposes. However, to therapeutically challenge
`the IDH1 cancer variants it must first be assessed whether they
`functionally operate as oncogenes or tumor suppressor genes. The
`fact that we and others only found heterozygous mutations at one
`specific IDH1 residue involved in its catalytic activity, strongly
`suggests that these mutations could activate IDH1 in a pro-
`oncogenic (dominant) fashion in cancer cells. Studies revealing
`the functional role of the IDH1R132
`mutations are vital to confirm
`this hypothesis and to provide insights in the potential of mutated
`IDH1 as therapeutic target.
`
`Acknowledgments
`
`The authors thank Dr. C. Zanon for help with sequencing, and Dr. S.
`Thorlacius and Dr. F. Di Nicolantonio for critical reading of the manuscript.
`F.B. is supported by a Netherlands Genomic Initiative Fellowship.
`
`References
`
`Balakrishnan A, Bleeker FE, Lamba S, Rodolfo M, Daniotti M, Scarpa A, van Tilborg
`AA, Leenstra S, Zanon C, Bardelli A. 2007. Novel somatic and germline
`mutations in cancer candidate genes in glioblastoma, melanoma, and pancreatic
`carcinoma. Cancer Res 67:3545–3550.
`Bleeker FE, Felicioni L, Buttitta F, Lamba S, Cardone L, Rodolfo M, Scarpa A,
`Leenstra S, Frattini M, Barbareschi M, Grammastro MD, Sciarrotta MG, Zanon
`C, Marchetti A, Bardelli A. 2008a. AKT1(E17K) in human solid tumours.
`Oncogene May 26 [Epub ahead of print].
`Bleeker FE, Lamba S, Rodolfo M, Scarpa A, Leenstra S, Vandertop WP, Bardelli A.
`2008b. Mutational profiling of cancer candidate genes
`in glioblastoma,
`melanoma and pancreatic carcinoma reveals a snapshot of their genomic
`landscapes. Hum Mutat, in press.
`Chen TR, Dorotinsky CS, McGuire LJ, Macy ML, Hay RJ. 1995. DLD-1 and HCT-15
`cell lines derived separately from colorectal carcinomas have totally different
`chromosome changes but the same genetic origin. Cancer Genet Cytogenet
`81:103–108.
`Geisbrecht BV, Gould SJ. 1999. The human PICD gene encodes a cytoplasmic and
`peroxisomal NADP(1)-dependent
`isocitrate dehydrogenase.
`J Biol Chem
`274:30527–30533.
`
`Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H,
`Kamiyama H, Jimeno A, Hong SM, Fu B, Lin MT, Calhoun ES, Kamiyama M,
`Walter K, Nikolskaya T, Nikolsky Y, Hartigan J, Smith DR, Hidalgo M, Leach
`SD, Klein AP, Jaffee EM, Goggins M, Maitra A, Iacobuzio-Donahue C, Eshleman
`JR, Kern SE, Hruban RH, Karchin R, Papadopoulos N, Parmigiani G, Vogelstein
`B, Velculescu VE, Kinzler KW. 2008. Core signaling pathways in human
`pancreatic cancers revealed by global genomic analyses. Science 321:1801–1806.
`Koshland Jr DE, Walsh K, LaPorte DC. 1985. Sensitivity of metabolic fluxes to
`covalent control. Curr Top Cell Regul 27:13–22.
`Lee SM, Koh HJ, Park DC, Song BJ, Huh TL, Park JW. 2002. Cytosolic NADP(1)-
`dependent isocitrate dehydrogenase status modulates oxidative damage to cells.
`Free Radic Biol Med 32:1185–1196.
`Louis DN, Ohgaki H, Wiestler OD, Cavenee WK. 2007. WHO classification of tumours
`of the central nervous system. Lyon: IARC Press.
`Ohgaki H, Dessen P, Jourde B, Horstmann S, Nishikawa T, Di Patre PL, Burkhard C,
`Schuler D, Probst-Hensch NM, Maiorka PC et al. 2004. Genetic pathways to
`glioblastoma: a population-based study. Cancer Res 64:6892–6899.
`Ohgaki H, Kleihues P. 2007. Genetic pathways
`to primary and secondary
`glioblastoma. Am J Pathol 170:1445–1453.
`Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H,
`Siu IM, Gallia GL, Olivi A, McLendon R, Rasheed BA, Keir S, Nikolskaya T,
`Nikolsky Y, Busam DA, Tekleab H, Diaz Jr LA, Hartigan J, Smith DR, Strausberg
`RL, Marie SK, Shinjo SM, Yan H, Riggins GJ, Bigner DD, Karchin R,
`Papadopoulos N, Parmigiani G, Vogelstein B, Velculescu VE, Kinzler KW. 2008.
`An integrated genomic analysis of human glioblastoma multiforme. Science
`321:1807–1812.
`Scherer H. 1940. Cerebral astrocytomas and their derivatives. Am J Cancer
`40:159–198.
`Sjo¨blom T, Jones S, Wood LD, Parsons DW, Lin J, Barber TD, Mandelker D, Leary RJ,
`Ptak J, Silliman N, Szabo S, Buckhaults P, Farrell C, Meeh P, Markowitz SD,
`Willis J, Dawson D, Willson JK, Gazdar AF, Hartigan J, Wu L, Liu C, Parmigiani
`G, Park BH, Bachman KE, Papadopoulos N, Vogelstein B, Kinzler KW,
`Velculescu VE. 2006. The consensus coding sequences of human breast and
`colorectal cancers. Science 314:268–274.
`Wood LD, Parsons DW, Jones S, Lin J, Sjo¨blom T, Leary RJ, Shen D, Boca SM, Barber
`T, Ptak J, Silliman N, Szabo S, Dezso Z, Ustyanksky V, Nikolskaya T, Nikolsky Y,
`Karchin R, Wilson PA, Kaminker JS, Zhang Z, Croshaw R, Willis J, Dawson D,
`Shipitsin M, Willson JK, Sukumar S, Polyak K, Park BH, Pethiyagoda CL, Pant
`PV, Ballinger DG, Sparks AB, Hartigan J, Smith DR, Suh E, Papadopoulos N,
`Buckhaults P, Markowitz SD, Parmigiani G, Kinzler KW, Velculescu VE,
`Vogelstein B. 2007. The genomic landscapes of human breast and colorectal
`cancers. Science 318:1108–1113.
`Xu X, Zhao J, Xu Z, Peng B, Huang Q, Arnold E, Ding J. 2004. Structures of human
`cytosolic NADP-dependent
`isocitrate dehydrogenase reveal a novel
`self-
`regulatory mechanism of activity. J Biol Chem 279:33946–33957.
`
`HUMAN MUTATION, Vol. 30, No. 1, 7–11, 2009
`
`11
`
`Rigel Exhibit 1017
`Page 5 of 5
`
`

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