901
|
Venkatesan S, Swanton C. Tumor Evolutionary Principles: How Intratumor Heterogeneity Influences Cancer Treatment and Outcome. Am Soc Clin Oncol Educ Book 2016; 35:e141-9. [PMID: 27249716 DOI: 10.1200/edbk_158930] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Recent studies have shown that intratumor heterogeneity contributes to drug resistance in advanced disease. Intratumor heterogeneity may foster the selection of a resistant subclone, sometimes detectable prior to treatment. Next-generation sequencing is enabling the phylogenetic reconstruction of a cancer's life history and has revealed different modes of cancer evolution. These studies have shown that cancer evolution is not always stochastic and has certain constraints. Consideration of cancer evolution may enable the better design of clinical trials and cancer therapeutics. In this review, we summarize the different modes of cancer evolution and how this might impact clinical outcomes. Furthermore, we will discuss several therapeutic strategies for managing emergent intratumor heterogeneity.
Collapse
Affiliation(s)
- Subramanian Venkatesan
- From the UCL Cancer Institute, CRUK Lung Cancer Centre of Excellence, London, United Kingdom; The Francis Crick Institute, London, United Kingdom
| | - Charles Swanton
- From the UCL Cancer Institute, CRUK Lung Cancer Centre of Excellence, London, United Kingdom; The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
902
|
Bai H, Harmancı AS, Erson-Omay EZ, Li J, Coşkun S, Simon M, Krischek B, Özduman K, Omay SB, Sorensen EA, Turcan Ş, Bakırcığlu M, Carrión-Grant G, Murray PB, Clark VE, Ercan-Sencicek AG, Knight J, Sencar L, Altınok S, Kaulen LD, Gülez B, Timmer M, Schramm J, Mishra-Gorur K, Henegariu O, Moliterno J, Louvi A, Chan TA, Tannheimer SL, Pamir MN, Vortmeyer AO, Bilguvar K, Yasuno K, Günel M. Integrated genomic characterization of IDH1-mutant glioma malignant progression. Nat Genet 2016; 48:59-66. [PMID: 26618343 PMCID: PMC4829945 DOI: 10.1038/ng.3457] [Citation(s) in RCA: 235] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 11/06/2015] [Indexed: 12/13/2022]
Abstract
Gliomas represent approximately 30% of all central nervous system tumors and 80% of malignant brain tumors. To understand the molecular mechanisms underlying the malignant progression of low-grade gliomas with mutations in IDH1 (encoding isocitrate dehydrogenase 1), we studied paired tumor samples from 41 patients, comparing higher-grade, progressed samples to their lower-grade counterparts. Integrated genomic analyses, including whole-exome sequencing and copy number, gene expression and DNA methylation profiling, demonstrated nonlinear clonal expansion of the original tumors and identified oncogenic pathways driving progression. These include activation of the MYC and RTK-RAS-PI3K pathways and upregulation of the FOXM1- and E2F2-mediated cell cycle transitions, as well as epigenetic silencing of developmental transcription factor genes bound by Polycomb repressive complex 2 in human embryonic stem cells. Our results not only provide mechanistic insight into the genetic and epigenetic mechanisms driving glioma progression but also identify inhibition of the bromodomain and extraterminal (BET) family as a potential therapeutic approach.
Collapse
Affiliation(s)
- Hanwen Bai
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
- Program in Brain Tumor Research, Yale School of Medicine, New Haven, Connecticut, USA
| | - Akdes Serin Harmancı
- Program in Brain Tumor Research, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - E Zeynep Erson-Omay
- Program in Brain Tumor Research, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jie Li
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Süleyman Coşkun
- Program in Brain Tumor Research, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Matthias Simon
- Department of Neurosurgery, University of Bonn Medical School, Bonn, Germany
| | - Boris Krischek
- Department of General Neurosurgery, University Hospital of Cologne, Cologne, Germany
| | - Koray Özduman
- Department of Neurosurgery, Acıbadem University School of Medicine, Istanbul, Turkey
| | - S Bülent Omay
- Program in Brain Tumor Research, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Eric A Sorensen
- Translational Medicine, Biomarkers, Gilead Sciences, Inc., Foster City, California, USA
| | - Şevin Turcan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mehmet Bakırcığlu
- Department of Neurosurgery, Acıbadem University School of Medicine, Istanbul, Turkey
| | - Geneive Carrión-Grant
- Program in Brain Tumor Research, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Phillip B Murray
- Department of Pharmacology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Victoria E Clark
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
- Program in Brain Tumor Research, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - A Gulhan Ercan-Sencicek
- Program in Brain Tumor Research, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - James Knight
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
- Yale Center for Genome Analysis, Yale School of Medicine, Orange, Connecticut, USA
| | - Leman Sencar
- Program in Brain Tumor Research, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Selin Altınok
- Program in Brain Tumor Research, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Leon D Kaulen
- Program in Brain Tumor Research, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Burcu Gülez
- Program in Brain Tumor Research, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Marco Timmer
- Department of General Neurosurgery, University Hospital of Cologne, Cologne, Germany
| | - Johannes Schramm
- Department of Neurosurgery, University of Bonn Medical School, Bonn, Germany
| | - Ketu Mishra-Gorur
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
- Program in Brain Tumor Research, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurobiology, Yale School of Medicine, New Haven, Connecticut, USA
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Octavian Henegariu
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
- Program in Brain Tumor Research, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurobiology, Yale School of Medicine, New Haven, Connecticut, USA
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jennifer Moliterno
- Program in Brain Tumor Research, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Angeliki Louvi
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurobiology, Yale School of Medicine, New Haven, Connecticut, USA
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Timothy A Chan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Stacey L Tannheimer
- Translational Medicine, Biomarkers, Gilead Sciences, Inc., Foster City, California, USA
| | - M Necmettin Pamir
- Department of Neurosurgery, Acıbadem University School of Medicine, Istanbul, Turkey
| | | | - Kaya Bilguvar
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
- Program in Brain Tumor Research, Yale School of Medicine, New Haven, Connecticut, USA
- Yale Center for Genome Analysis, Yale School of Medicine, Orange, Connecticut, USA
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Katsuhito Yasuno
- Program in Brain Tumor Research, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Murat Günel
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
- Program in Brain Tumor Research, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurobiology, Yale School of Medicine, New Haven, Connecticut, USA
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, Connecticut, USA
- Yale Comprehensive Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
903
|
Panichnantakul P, Bourgey M, Montpetit A, Bourque G, Riazalhosseini Y. RNA-Seq as a Tool to Study the Tumor Microenvironment. Methods Mol Biol 2016; 1458:311-37. [PMID: 27581031 DOI: 10.1007/978-1-4939-3801-8_22] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The transcriptome is composed of different types of RNA molecules including mRNAs, tRNAs, rRNAs, and other noncoding RNAs that are found inside a cell at a given time. Analyzing transcriptome patterns can shed light on the functional state of the cell as well as on the dynamics of cellular behavior associated with genomic and environmental changes. Likewise, transcriptome analysis has been a major help in solving biological issues and understanding the molecular basis of many diseases including human cancers. Specifically, since targeted and whole genome sequencing studies are becoming more common in identifying the driving factors of cancer, a comprehensive and high-resolution analysis of the transcriptome, as provided by RNA-Sequencing (RNA-Seq), plays a key role in investigating the functional relevance of the identified genomic aberrations. Here, we describe experimental procedures of RNA-Seq and downstream data processing and analysis, with a focus on the identification of abnormally expressed transcripts and genes.
Collapse
Affiliation(s)
- Pudchalaluck Panichnantakul
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada
| | - Mathieu Bourgey
- McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada
| | | | - Guillaume Bourque
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada
| | - Yasser Riazalhosseini
- Department of Human Genetics, McGill University, Montreal, QC, Canada.
- McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada.
| |
Collapse
|
904
|
de la Fuente MI, Young RJ, Rubel J, Rosenblum M, Tisnado J, Briggs S, Arevalo-Perez J, Cross JR, Campos C, Straley K, Zhu D, Dong C, Thomas A, Omuro AA, Nolan CP, Pentsova E, Kaley TJ, Oh JH, Noeske R, Maher E, Choi C, Gutin PH, Holodny AI, Yen K, DeAngelis LM, Mellinghoff IK, Thakur SB. Integration of 2-hydroxyglutarate-proton magnetic resonance spectroscopy into clinical practice for disease monitoring in isocitrate dehydrogenase-mutant glioma. Neuro Oncol 2015; 18:283-90. [PMID: 26691210 DOI: 10.1093/neuonc/nov307] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 11/14/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The majority of WHO grades II and III gliomas harbor a missense mutation in the metabolic gene isocitrate dehydrogenase (IDH) and accumulate the metabolite R-2-hydroxyglutarate (R-2HG). Prior studies showed that this metabolite can be detected in vivo using proton magnetic-resonance spectroscopy (MRS), but the sensitivity of this methodology and its clinical implications are unknown. METHODS We developed an MR imaging protocol to integrate 2HG-MRS into routine clinical glioma imaging and examined its performance in 89 consecutive glioma patients. RESULTS Detection of 2-hydroxyglutarate (2HG) in IDH-mutant gliomas was closely linked to tumor volume, with sensitivity ranging from 8% for small tumors (<3.4 mL) to 91% for larger tumors (>8 mL). In patients undergoing 2HG-MRS prior to surgery, tumor levels of 2HG corresponded with tumor cellularity but not with tumor grade or mitotic index. Cytoreductive therapy resulted in a gradual decrease in 2HG levels with kinetics that closely mirrored changes in tumor volume. CONCLUSIONS Our study demonstrates that 2HG-MRS can be linked with routine MR imaging to provide quantitative measurements of 2HG in glioma and may be useful as an imaging biomarker to monitor the abundance of IDH-mutant tumor cells noninvasively during glioma therapy and disease monitoring.
Collapse
Affiliation(s)
- Macarena I de la Fuente
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Robert J Young
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Jennifer Rubel
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Marc Rosenblum
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Jamie Tisnado
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Samuel Briggs
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Julio Arevalo-Perez
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Justin R Cross
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Carl Campos
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Kimberly Straley
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Dongwei Zhu
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Chuanhui Dong
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Alissa Thomas
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Antonio A Omuro
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Craig P Nolan
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Elena Pentsova
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Thomas J Kaley
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Jung H Oh
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Ralph Noeske
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Elizabeth Maher
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Changho Choi
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Philip H Gutin
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Andrei I Holodny
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Katharine Yen
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Lisa M DeAngelis
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Ingo K Mellinghoff
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| | - Sunitha B Thakur
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (M.I.d., S.B., A.T., A.A.O., C.P.N., E.P., T.J.K., L.M.D., I.K.M.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York (R.J.Y., J.R., J.T., J.A.-P., A.I.H., S.B.T.); Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (M.R.); Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, New York (J.R.C.); Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York (C.C., I.K.M.); Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York (P.H.G.); Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York (J.H.O., S.B.T.); Agios Pharmaceuticals, Cambridge, Massachusetts (K.S., D.Z., K.Y.); Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami, Miami, Florida (C.D.); Advanced Imaging Research Center, University Of Texas Southwestern Medical Center, Dallas, Texas (E.M., C.C.); GE Healthcare, Berlin, Germany (R.N.); Department of Pharmacology, Weill-Cornell Graduate School of Biomedical Sciences, New York, New York (I.K.M.)
| |
Collapse
|
905
|
Comparative transcriptomics reveals similarities and differences between astrocytoma grades. BMC Cancer 2015; 15:952. [PMID: 26673168 PMCID: PMC4682229 DOI: 10.1186/s12885-015-1939-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 11/01/2015] [Indexed: 11/23/2022] Open
Abstract
Background Astrocytomas are the most common primary brain tumors distinguished into four histological grades. Molecular analyses of individual astrocytoma grades have revealed detailed insights into genetic, transcriptomic and epigenetic alterations. This provides an excellent basis to identify similarities and differences between astrocytoma grades. Methods We utilized public omics data of all four astrocytoma grades focusing on pilocytic astrocytomas (PA I), diffuse astrocytomas (AS II), anaplastic astrocytomas (AS III) and glioblastomas (GBM IV) to identify similarities and differences using well-established bioinformatics and systems biology approaches. We further validated the expression and localization of Ang2 involved in angiogenesis using immunohistochemistry. Results Our analyses show similarities and differences between astrocytoma grades at the level of individual genes, signaling pathways and regulatory networks. We identified many differentially expressed genes that were either exclusively observed in a specific astrocytoma grade or commonly affected in specific subsets of astrocytoma grades in comparison to normal brain. Further, the number of differentially expressed genes generally increased with the astrocytoma grade with one major exception. The cytokine receptor pathway showed nearly the same number of differentially expressed genes in PA I and GBM IV and was further characterized by a significant overlap of commonly altered genes and an exclusive enrichment of overexpressed cancer genes in GBM IV. Additional analyses revealed a strong exclusive overexpression of CX3CL1 (fractalkine) and its receptor CX3CR1 in PA I possibly contributing to the absence of invasive growth. We further found that PA I was significantly associated with the mesenchymal subtype typically observed for very aggressive GBM IV. Expression of endothelial and mesenchymal markers (ANGPT2, CHI3L1) indicated a stronger contribution of the micro-environment to the manifestation of the mesenchymal subtype than the tumor biology itself. We further inferred a transcriptional regulatory network associated with specific expression differences distinguishing PA I from AS II, AS III and GBM IV. Major central transcriptional regulators were involved in brain development, cell cycle control, proliferation, apoptosis, chromatin remodeling or DNA methylation. Many of these regulators showed directly underlying DNA methylation changes in PA I or gene copy number mutations in AS II, AS III and GBM IV. Conclusions This computational study characterizes similarities and differences between all four astrocytoma grades confirming known and revealing novel insights into astrocytoma biology. Our findings represent a valuable resource for future computational and experimental studies. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1939-9) contains supplementary material, which is available to authorized users.
Collapse
|
906
|
Gatson NTN, Weathers SPS, de Groot JF. ReACT Phase II trial: a critical evaluation of the use of rindopepimut plus bevacizumab to treat EGFRvIII-positive recurrent glioblastoma. CNS Oncol 2015; 5:11-26. [PMID: 26670466 DOI: 10.2217/cns.15.38] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma is the most deadly primary brain tumor in adults and has long represented a therapeutic challenge. Disease recurrence is inevitable, and the management of recurrent disease is complicated by spontaneous or induced tumor heterogeneity which confers resistance to therapy and increased oncogenicity. EGFR and the tumor-specific mutation EGFRvIII is commonly altered in glioblastoma making it an appealing therapeutic target. Immunotherapy is an emerging and promising therapeutic approach to glioma and the EGFRvIII vaccine, rindopepimut, is the first immunotherapeutic drug to enter Phase III clinical trials for glioblastoma. Rindopepimut activates a specific immune response against tumor cells harboring the EGFRvIII protein. This review evaluates the recently completed ReACT Phase II trial using rindopepimut plus bevacizumab in the setting of EGFRvIII-positive recurrent glioblastoma (Clinical Trials identifier: NCT01498328).
Collapse
Affiliation(s)
- Na Tosha N Gatson
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 0431, Houston, TX 77054, USA
| | - Shiao-Pei S Weathers
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 0431, Houston, TX 77054, USA
| | - John F de Groot
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 0431, Houston, TX 77054, USA
| |
Collapse
|
907
|
Tateishi K, Wakimoto H, Iafrate AJ, Tanaka S, Loebel F, Lelic N, Wiederschain D, Bedel O, Deng G, Zhang B, He T, Shi X, Gerszten RE, Zhang Y, Yeh JRJ, Curry WT, Zhao D, Sundaram S, Nigim F, Koerner MVA, Ho Q, Fisher DE, Roider EM, Kemeny LV, Samuels Y, Flaherty KT, Batchelor TT, Chi AS, Cahill DP. Extreme Vulnerability of IDH1 Mutant Cancers to NAD+ Depletion. Cancer Cell 2015; 28:773-784. [PMID: 26678339 PMCID: PMC4684594 DOI: 10.1016/j.ccell.2015.11.006] [Citation(s) in RCA: 329] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 07/09/2015] [Accepted: 11/14/2015] [Indexed: 01/08/2023]
Abstract
Heterozygous mutation of IDH1 in cancers modifies IDH1 enzymatic activity, reprogramming metabolite flux and markedly elevating 2-hydroxyglutarate (2-HG). Here, we found that 2-HG depletion did not inhibit growth of several IDH1 mutant solid cancer types. To identify other metabolic therapeutic targets, we systematically profiled metabolites in endogenous IDH1 mutant cancer cells after mutant IDH1 inhibition and discovered a profound vulnerability to depletion of the coenzyme NAD+. Mutant IDH1 lowered NAD+ levels by downregulating the NAD+ salvage pathway enzyme nicotinate phosphoribosyltransferase (Naprt1), sensitizing to NAD+ depletion via concomitant nicotinamide phosphoribosyltransferase (NAMPT) inhibition. NAD+ depletion activated the intracellular energy sensor AMPK, triggered autophagy, and resulted in cytotoxicity. Thus, we identify NAD+ depletion as a metabolic susceptibility of IDH1 mutant cancers.
Collapse
Affiliation(s)
- Kensuke Tateishi
- Department of Neurosurgery, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Hiroaki Wakimoto
- Department of Neurosurgery, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - A John Iafrate
- Department of Pathology, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Shota Tanaka
- Divisions of Neuro-Oncology and Hematology/Oncology, Department of Neurology, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Franziska Loebel
- Department of Neurosurgery, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Nina Lelic
- Department of Neurosurgery, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | - Timothy He
- Sanofi Oncology, Cambridge, MA 02139, USA
| | - Xu Shi
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Robert E Gerszten
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Yiyun Zhang
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jing-Ruey J Yeh
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - William T Curry
- Department of Neurosurgery, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Dan Zhao
- Divisions of Neuro-Oncology and Hematology/Oncology, Department of Neurology, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Sudhandra Sundaram
- Divisions of Neuro-Oncology and Hematology/Oncology, Department of Neurology, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Fares Nigim
- Department of Neurosurgery, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Mara V A Koerner
- Department of Neurosurgery, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Quan Ho
- Department of Pathology, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - David E Fisher
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Elisabeth M Roider
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lajos V Kemeny
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | - Keith T Flaherty
- Division of Hematology/Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Tracy T Batchelor
- Divisions of Neuro-Oncology and Hematology/Oncology, Department of Neurology, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Andrew S Chi
- Divisions of Neuro-Oncology and Hematology/Oncology, Department of Neurology, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Daniel P Cahill
- Department of Neurosurgery, Translational Neuro-Oncology Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
908
|
Gallo M, Coutinho FJ, Vanner RJ, Gayden T, Mack SC, Murison A, Remke M, Li R, Takayama N, Desai K, Lee L, Lan X, Park NI, Barsyte-Lovejoy D, Smil D, Sturm D, Kushida MM, Head R, Cusimano MD, Bernstein M, Clarke ID, Dick JE, Pfister SM, Rich JN, Arrowsmith CH, Taylor MD, Jabado N, Bazett-Jones DP, Lupien M, Dirks PB. MLL5 Orchestrates a Cancer Self-Renewal State by Repressing the Histone Variant H3.3 and Globally Reorganizing Chromatin. Cancer Cell 2015; 28:715-729. [PMID: 26626085 DOI: 10.1016/j.ccell.2015.10.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/13/2015] [Accepted: 10/12/2015] [Indexed: 02/04/2023]
Abstract
Mutations in the histone 3 variant H3.3 have been identified in one-third of pediatric glioblastomas (GBMs), but not in adult tumors. Here we show that H3.3 is a dynamic determinant of functional properties in adult GBM. H3.3 is repressed by mixed lineage leukemia 5 (MLL5) in self-renewing GBM cells. MLL5 is a global epigenetic repressor that orchestrates reorganization of chromatin structure by punctuating chromosomes with foci of compacted chromatin, favoring tumorigenic and self-renewing properties. Conversely, H3.3 antagonizes self-renewal and promotes differentiation. We exploited these epigenetic states to rationally identify two small molecules that effectively curb cancer stem cell properties in a preclinical model. Our work uncovers a role for MLL5 and H3.3 in maintaining self-renewal hierarchies in adult GBM.
Collapse
Affiliation(s)
- Marco Gallo
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Fiona J Coutinho
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Robert J Vanner
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Tenzin Gayden
- Departments of Pediatrics and Human Genetics, McGill University and McGill University Health Centre Research Institute, Montreal, QC H3H 1P4, Canada
| | - Stephen C Mack
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland, OH 44195, USA; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Alex Murison
- Ontario Institute for Cancer Research and Princess Margaret Cancer Centre-University Health Network, Toronto, ON M5G 1L7, Canada
| | - Marc Remke
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Ren Li
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Naoya Takayama
- Ontario Institute for Cancer Research and Princess Margaret Cancer Centre-University Health Network, Toronto, ON M5G 1L7, Canada
| | - Kinjal Desai
- Department of Genetics, Norris Cotton Cancer Center, Dartmouth Medical School, Lebanon, NH 03755, USA
| | - Lilian Lee
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Xiaoyang Lan
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Nicole I Park
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Dalia Barsyte-Lovejoy
- Ontario Institute for Cancer Research and Princess Margaret Cancer Centre-University Health Network, Toronto, ON M5G 1L7, Canada; Structural Genomics Consortium, Toronto, ON M5G 1L7, Canada
| | - David Smil
- Ontario Institute for Cancer Research and Princess Margaret Cancer Centre-University Health Network, Toronto, ON M5G 1L7, Canada; Structural Genomics Consortium, Toronto, ON M5G 1L7, Canada
| | - Dominik Sturm
- Division of Pediatric Neurooncology, German Cancer Research Centre (DKFZ), Heidelberg 69120, Germany
| | - Michelle M Kushida
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Renee Head
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Michael D Cusimano
- Division of Neurosurgery, University of Toronto, Toronto, ON M5S 1A8, Canada; St. Michael's Hospital, Toronto, ON M5B 1W8, Canada
| | - Mark Bernstein
- Division of Neurosurgery, University of Toronto, Toronto, ON M5S 1A8, Canada; Toronto Western Hospital, Toronto, ON M5T 2S8, Canada
| | - Ian D Clarke
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - John E Dick
- Ontario Institute for Cancer Research and Princess Margaret Cancer Centre-University Health Network, Toronto, ON M5G 1L7, Canada
| | - Stefan M Pfister
- Division of Pediatric Neurooncology, German Cancer Research Centre (DKFZ), Heidelberg 69120, Germany
| | - Jeremy N Rich
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Cheryl H Arrowsmith
- Ontario Institute for Cancer Research and Princess Margaret Cancer Centre-University Health Network, Toronto, ON M5G 1L7, Canada; Structural Genomics Consortium, Toronto, ON M5G 1L7, Canada
| | - Michael D Taylor
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Division of Neurosurgery, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Nada Jabado
- Departments of Pediatrics and Human Genetics, McGill University and McGill University Health Centre Research Institute, Montreal, QC H3H 1P4, Canada
| | - David P Bazett-Jones
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Mathieu Lupien
- Ontario Institute for Cancer Research and Princess Margaret Cancer Centre-University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
| | - Peter B Dirks
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Division of Neurosurgery, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
909
|
Understanding the Genetic Mechanisms of Cancer Drug Resistance Using Genomic Approaches. Trends Genet 2015; 32:127-137. [PMID: 26689126 DOI: 10.1016/j.tig.2015.11.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 11/03/2015] [Accepted: 11/16/2015] [Indexed: 12/14/2022]
Abstract
A major obstacle in precision cancer medicine is the inevitable resistance to targeted therapies. Tremendous effort and progress has been made over the past few years to understand the biochemical and genetic mechanisms underlying drug resistance, with the goal to eventually overcome such daunting challenges. Diverse mechanisms, such as secondary mutations, oncogene bypass, and epigenetic alterations, can all lead to drug resistance, and the number of known involved genes is growing rapidly, thus providing many possibilities to overcome resistance. The finding of these mechanisms and genes invariably requires the application of genomic and functional genomic approaches to tumors or cancer models. In this review, we briefly highlight the major drug-resistance mechanisms known today, and then focus primarily on the technological approaches leading to the advancement of this field.
Collapse
|
910
|
Targeting the Hippo pathway: Clinical implications and therapeutics. Pharmacol Res 2015; 103:270-8. [PMID: 26678601 DOI: 10.1016/j.phrs.2015.11.025] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 11/30/2015] [Accepted: 11/30/2015] [Indexed: 12/12/2022]
Abstract
The Hippo pathway plays a critical role in tissue and organ size regulation by restraining cell proliferation and apoptosis under homeostatic conditions. Deregulation of this pathway can promote tumorigenesis in multiple malignant human tumor types, including sarcoma, breast, lung and liver cancers. In this review, we summarize the current understanding of Hippo pathway function, it's role in human cancer, and address the potential of Hippo pathway member proteins as therapeutic targets for a variety of tumors.
Collapse
|
911
|
Abstract
Glioblastoma is a refractory malignancy with limited treatment options at tumor recurrence. Only a small proportion of patients survive 2 years or longer with the current standard of care. Gene expression profiling can segregate newly diagnosed patients into groups with different prognoses, and these biomarkers are being incorporated into a new generation of personalized clinical trials. Using the experience from recently completed large scale, multi-faceted, randomized glioblastoma clinical trials, a new clinical trial paradigm is being established to move promising therapies forward into the newly diagnosed treatment setting. Upcoming trials using the immune check-point inhibitors are an example of this changing paradigm and these and other immunotherapies have potential as promising new treatment modalities for newly diagnosed GB patients.
Collapse
Affiliation(s)
- Brett J Theeler
- Department of Neurology and John P. Murtha Cancer Center, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Building 19, Bethesda, MD, 20889, USA.
| | - Mark R Gilbert
- National Institutes of Health, 9030 Old Georgetown Road, Bethesda, MD, 20892, USA.
| |
Collapse
|
912
|
Zhang C, Burger MC, Jennewein L, Genßler S, Schönfeld K, Zeiner P, Hattingen E, Harter PN, Mittelbronn M, Tonn T, Steinbach JP, Wels WS. ErbB2/HER2-Specific NK Cells for Targeted Therapy of Glioblastoma. J Natl Cancer Inst 2015; 108:djv375. [PMID: 26640245 DOI: 10.1093/jnci/djv375] [Citation(s) in RCA: 281] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 11/02/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common and malignant intracranial tumor in adults and currently incurable. To specifically target natural killer (NK) cell activity to GBM, we employed NK-92/5.28.z cells that are continuously expanding human NK cells expressing an ErbB2-specific chimeric antigen receptor (CAR). METHODS ErbB2 expression in 56 primary tumors, four primary cell cultures, and seven established cell lines was assessed by immunohistochemistry and flow cytometry. Cell killing activity of NK-92/5.28.z cells was analyzed in in vitro cytotoxicity assays. In vivo antitumor activity was evaluated in NOD-SCID IL2Rγ(null) (NSG) mice carrying orthotopic human GBM xenografts (6 to 11 mice per group) and C57BL/6 mice carrying subcutaneous and orthotopic ErbB2-expressing murine GBM tumors (5 to 8 mice per group). Statistical tests were two-sided. RESULTS We found elevated ErbB2 protein expression in 41% of primary GBM samples and in the majority of GBM cell lines investigated. In in vitro assays, NK-92/5.28.z in contrast to untargeted NK-92 cells lysed all ErbB2-positive established and primary GBM cells analyzed. Potent in vivo antitumor activity of NK-92/5.28.z was observed in orthotopic GBM xenograft models in NSG mice, leading to a marked extension of symptom-free survival upon repeated stereotactic injection of CAR NK cells into the tumor area (median survival of 200.5 days upon treatment with NK-92/5.28.z vs 73 days upon treatment with parental NK-92 cells, P < .001). In immunocompetent mice, local therapy with NK-92/5.28.z cells resulted in cures of transplanted syngeneic GBM in four of five mice carrying subcutaneous tumors and five of eight mice carrying intracranial tumors, induction of endogenous antitumor immunity, and long-term protection against tumor rechallenge at distant sites. CONCLUSIONS Our data demonstrate the potential of ErbB2-specific NK-92/5.28.z cells for adoptive immunotherapy of glioblastoma, justifying evaluation of this approach for the treatment of ErbB2-positive GBM in clinical studies.
Collapse
Affiliation(s)
- Congcong Zhang
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Michael C Burger
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Lukas Jennewein
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Sabrina Genßler
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Kurt Schönfeld
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Pia Zeiner
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Elke Hattingen
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Patrick N Harter
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Michel Mittelbronn
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Torsten Tonn
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Joachim P Steinbach
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| | - Winfried S Wels
- Affiliations of authors:Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main , Germany (CZ, SG, KS, WSW); German Cancer Consortium (DKTK), partner site Frankfurt/Mainz , Germany (CZ, MCB, JPS, WSW); German Cancer Research Center (DKFZ), Heidelberg , Germany (CZ, MCB); Institute for Neurooncology (MCB, JPS), Edinger Institute (LJ, PZ, PNH, MM), Department of Neurology (PZ), and Institute of Neuroradiology (EH), Goethe University, Frankfurt am Main , Germany ; Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East and Medical Faculty Carl Gustav Carus, TU Dresden , Dresden , Germany (TT)
| |
Collapse
|
913
|
Mazzocco P, Barthélémy C, Kaloshi G, Lavielle M, Ricard D, Idbaih A, Psimaras D, Renard M, Alentorn A, Honnorat J, Delattre J, Ducray F, Ribba B. Prediction of Response to Temozolomide in Low-Grade Glioma Patients Based on Tumor Size Dynamics and Genetic Characteristics. CPT Pharmacometrics Syst Pharmacol 2015; 4:728-37. [PMID: 26904387 PMCID: PMC4759703 DOI: 10.1002/psp4.54] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 04/23/2015] [Accepted: 05/04/2015] [Indexed: 01/27/2023] Open
Abstract
Both molecular profiling of tumors and longitudinal tumor size data modeling are relevant strategies to predict cancer patients' response to treatment. Herein we propose a model of tumor growth inhibition integrating a tumor's genetic characteristics (p53 mutation and 1p/19q codeletion) that successfully describes the time course of tumor size in patients with low-grade gliomas treated with first-line temozolomide chemotherapy. The model captures potential tumor progression under chemotherapy by accounting for the emergence of tissue resistance to treatment following prolonged exposure to temozolomide. Using information on individual tumors' genetic characteristics, in addition to early tumor size measurements, the model was able to predict the duration and magnitude of response, especially in those patients in whom repeated assessment of tumor response was obtained during the first 3 months of treatment. Combining longitudinal tumor size quantitative modeling with a tumor''s genetic characterization appears as a promising strategy to personalize treatments in patients with low-grade gliomas.
Collapse
Affiliation(s)
- P Mazzocco
- Inria, project‐team Numed, Ecole Normale Supérieure de LyonLyonFrance
| | - C Barthélémy
- Inria, project‐team Popix, Université Paris‐SudOrsayFrance
| | - G Kaloshi
- AP‐HP, Groupe Hospitalier Pitié‐Salpêtrière, Service de Neurologie Mazarin; INSERM, U975, Centre de Recherche de l'Institut du Cerveau et de la Moelle, Université Pierre & Marie Curie Paris VI, Faculté de Médecine Pitié‐Salpêtrière, CNRS UMR 7225 and UMR‐S975ParisFrance
| | - M Lavielle
- Inria, project‐team Popix, Université Paris‐SudOrsayFrance
| | - D Ricard
- Hôpital d'instruction des Armées du Val‐de‐GrâceParisFrance
| | - A Idbaih
- AP‐HP, Groupe Hospitalier Pitié‐Salpêtrière, Service de Neurologie Mazarin; INSERM, U975, Centre de Recherche de l'Institut du Cerveau et de la Moelle, Université Pierre & Marie Curie Paris VI, Faculté de Médecine Pitié‐Salpêtrière, CNRS UMR 7225 and UMR‐S975ParisFrance
| | - D Psimaras
- AP‐HP, Groupe Hospitalier Pitié‐Salpêtrière, Service de Neurologie Mazarin; INSERM, U975, Centre de Recherche de l'Institut du Cerveau et de la Moelle, Université Pierre & Marie Curie Paris VI, Faculté de Médecine Pitié‐Salpêtrière, CNRS UMR 7225 and UMR‐S975ParisFrance
| | - M‐A Renard
- AP‐HP, Groupe Hospitalier Pitié‐Salpêtrière, Service de Neurologie Mazarin; INSERM, U975, Centre de Recherche de l'Institut du Cerveau et de la Moelle, Université Pierre & Marie Curie Paris VI, Faculté de Médecine Pitié‐Salpêtrière, CNRS UMR 7225 and UMR‐S975ParisFrance
| | - A Alentorn
- AP‐HP, Groupe Hospitalier Pitié‐Salpêtrière, Service de Neurologie Mazarin; INSERM, U975, Centre de Recherche de l'Institut du Cerveau et de la Moelle, Université Pierre & Marie Curie Paris VI, Faculté de Médecine Pitié‐Salpêtrière, CNRS UMR 7225 and UMR‐S975ParisFrance
| | - J Honnorat
- Hospices Civils de Lyon, Hôpital Neurologique, Neuro‐oncologie; Université de Lyon, Claude Bernard Lyon 1, Lyon Neuroscience Research Center INSERM U1028/CNRS UMRLyonFrance
| | - J‐Y Delattre
- AP‐HP, Groupe Hospitalier Pitié‐Salpêtrière, Service de Neurologie Mazarin; INSERM, U975, Centre de Recherche de l'Institut du Cerveau et de la Moelle, Université Pierre & Marie Curie Paris VI, Faculté de Médecine Pitié‐Salpêtrière, CNRS UMR 7225 and UMR‐S975ParisFrance
| | - F Ducray
- Hospices Civils de Lyon, Hôpital Neurologique, Neuro‐oncologie; Université de Lyon, Claude Bernard Lyon 1, Lyon Neuroscience Research Center INSERM U1028/CNRS UMRLyonFrance
| | - B Ribba
- Inria, project‐team Numed, Ecole Normale Supérieure de LyonLyonFrance
| |
Collapse
|
914
|
Abstract
Glioblastoma, the most aggressive of the gliomas, has a high recurrence and mortality rate. The nature of this poor prognosis resides in the molecular heterogeneity and phenotypic features of this tumor. Despite research advances in understanding the molecular biology, it has been difficult to translate this knowledge into effective treatment. Nearly all will have tumor recurrence, yet to date very few therapies have established efficacy as salvage regimens. This challenge is further complicated by imaging confounders and to an even greater degree by the ever increasing molecular heterogeneity that is thought to be both sporadic and treatment-induced. The development of novel clinical trial designs to support the development and testing of novel treatment regimens and drug delivery strategies underscore the need for more precise techniques in imaging and better surrogate markers to help determine treatment response. This review summarizes recent approaches to treat patients with recurrent glioblastoma and considers future perspectives.
Collapse
Affiliation(s)
- Carlos Kamiya-Matsuoka
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | |
Collapse
|
915
|
Hedberg ML, Goh G, Chiosea SI, Bauman JE, Freilino ML, Zeng Y, Wang L, Diergaarde BB, Gooding WE, Lui VWY, Herbst RS, Lifton RP, Grandis JR. Genetic landscape of metastatic and recurrent head and neck squamous cell carcinoma. J Clin Invest 2015; 126:169-80. [PMID: 26619122 DOI: 10.1172/jci82066] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 10/22/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Recurrence and/or metastasis occurs in more than half of patients with head and neck squamous cell carcinoma (HNSCC), and these events pose the greatest threats to long-term survival. We set out to identify genetic alterations that underlie recurrent/metastatic HNSCC. METHODS Whole-exome sequencing (WES) was performed on genomic DNA extracted from fresh-frozen whole blood and patient-matched tumor pairs from 13 HNSCC patients with synchronous lymph node metastases and 10 patients with metachronous recurrent tumors. Mutational concordance within and between tumor pairs was used to analyze the spatiotemporal evolution of HNSCC in individual patients and to identify potential therapeutic targets for functional evaluation. RESULTS Approximately 86% and 60% of single somatic nucleotide variants (SSNVs) identified in synchronous nodal metastases and metachronous recurrent tumors, respectively, were transmitted from the primary index tumor. Genes that were mutated in more than one metastatic or recurrent tumor, but not in the respective primary tumors, include C17orf104, inositol 1,4,5-trisphosphate receptor, type 3 (ITPR3), and discoidin domain receptor tyrosine kinase 2 (DDR2). Select DDR2 mutations have been shown to confer enhanced sensitivity to SRC-family kinase (SFK) inhibitors in other malignancies. Similarly, HNSCC cell lines harboring endogenous and engineered DDR2 mutations were more sensitive to the SFK inhibitor dasatinib than those with WT DDR2. CONCLUSION In this WES study of patient-matched tumor pairs in HNSCC, we found synchronous lymph node metastases to be genetically more similar to their paired index primary tumors than metachronous recurrent tumors. This study outlines a compendium of somatic mutations in primary, metastatic, and/or recurrent HNSCC cancers, with potential implications for precision medicine approaches. FUNDING National Cancer Institute, American Cancer Society, Agency for Science, Technology and Research of Singapore, and Gilead Sciences Inc.
Collapse
|
916
|
Jacoby MA, Duncavage EJ, Walter MJ. Implications of Tumor Clonal Heterogeneity in the Era of Next-Generation Sequencing. Trends Cancer 2015; 1:231-241. [PMID: 28741514 DOI: 10.1016/j.trecan.2015.10.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 10/24/2015] [Accepted: 10/26/2015] [Indexed: 02/05/2023]
Abstract
Recent whole-genome sequencing (WGS) studies have demonstrated that tumors typically comprise a founding clone and multiple subclones (i.e., clonal heterogeneity is common). The possible combination of mutations in each tumor clone is enormous, making each tumor genetically unique. Clonal heterogeneity likely has a role in cancer progression, relapse, metastasis, and chemoresistance due to functional differences in genetically unique subclones. In current clinical practice, gene mutations are only classified as being present or absent, ignoring the clonal complexity of cancers. In this review, we address how tumor clonality is measured using next-generation sequencing (NGS) data, highlight that clonal heterogeneity is common across multiple tumor types, and discuss the potential clinical implications of tumor clonal heterogeneity.
Collapse
Affiliation(s)
- Meagan A Jacoby
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - Eric J Duncavage
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA; Alvin J. Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA
| | - Matthew J Walter
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO, USA; Alvin J. Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
917
|
Karunasena E, McIver LJ, Rood BR, Wu X, Zhu H, Bavarva JH, Garner HR. Somatic intronic microsatellite loci differentiate glioblastoma from lower-grade gliomas. Oncotarget 2015; 5:6003-14. [PMID: 25153720 PMCID: PMC4171608 DOI: 10.18632/oncotarget.2076] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Genomic studies of glioma sub-types have amassed new disease specific mutations, yet these only partially explain how mutations are linked to predisposition or progression. We hypothesized that microsatellite variation could expand the understanding of glioma etiology. Furthermore, germline markers for gliomas are typically undetectable; therefore we also hypothesize that the predictability of cancer-associated microsatellite loci in germline DNA may support the current hypothesis of a glioma cell of origin. In this study, “normal” germline exome sequenced DNA from the 1000 Genomes Project (n=390) were compared with exome sequences from germlines of subjects with WHO grade II and III lower-grade glioma (LGG, n=136) and WHO grade IV glioblastoma (GBM, n=252) from The Cancer Genome Atlas to identify microsatellite loci non-randomly associated with glioma. From germline data, we identified 48 GBM-specific loci, 42 Lower-grade glioma specific loci and 29 loci that distinguish GBM from LGG (p≤ 0.01). We then attempted to distinguish WHO grade II glioma (n=67) from GBM resulting in 8 informative loci. Significantly, in all glioma grades, comparisons between tumor and matched germline sequences demonstrated no significant differences in these variants (p≥ 0.01). Therefore, these microsatellite loci are considered to be components of grade-specific signatures for glioma which distinguish germline sequences of individuals with cancer from those of individuals that are “normal”. In order to better understand the significance of these loci, we identified biological processes enriched in genes with these variants. Most strikingly, six helicase genes were enriched in the GBM cohort (p≤ 1.0 ×10−3). The preservation of these glioma-specific loci could therefore serve as valuable diagnostic and therapeutic markers; especially since the heterogeneity of tumor cell populations can obscure the identification of mutations preceding a metastatic phenotype.
Collapse
Affiliation(s)
- Enusha Karunasena
- Virginia Bioinformatics Institute, Medical Informatics and Systems Division; Blacksburg, VA; These authors contributed equally to this work
| | - Lauren J McIver
- Virginia Bioinformatics Institute, Medical Informatics and Systems Division; Blacksburg, VA; These authors contributed equally to this work
| | - Brian R Rood
- Center for Cancer and Blood Disorders at Children's National Medical Center; Washington, D.C
| | - Xiaowei Wu
- Department of Statistics at Virginia Tech; Blacksburg, VA
| | - Hongxiao Zhu
- Department of Statistics at Virginia Tech; Blacksburg, VA
| | - Jasmin H Bavarva
- Virginia Bioinformatics Institute, Medical Informatics and Systems Division; Blacksburg, VA
| | - Harold R Garner
- Virginia Bioinformatics Institute, Medical Informatics and Systems Division; Blacksburg, VA
| |
Collapse
|
918
|
Roelcke U, Wyss MT, Nowosielski M, Rudà R, Roth P, Hofer S, Galldiks N, Crippa F, Weller M, Soffietti R. Amino acid positron emission tomography to monitor chemotherapy response and predict seizure control and progression-free survival in WHO grade II gliomas. Neuro Oncol 2015; 18:744-51. [PMID: 26578622 DOI: 10.1093/neuonc/nov282] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 10/12/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Patients with WHO grade II glioma may respond to chemotherapy that is currently not standardized regarding timing and treatment duration. Metabolic changes during chemotherapy may precede structural tumor volume reductions. We therefore compared time courses of amino acid PET and MRI responses to temozolomide (TMZ) and assessed whether responses correlated with seizure control and progression-free survival (PFS). METHODS PET and MRI were performed before and during TMZ chemotherapy. Tumor volumes were calculated using regions-of-interest analysis. Amino acid uptake was also quantified as metabolically active tumor volume and tumor-to-cerebellum uptake ratio. RESULTS One hundred twenty-five PET and 125 MRI scans from 33 patients were analyzed. Twenty-five patients showed metabolic responses that exhibited an exponential time course with a 25% reduction of the active volume on average after 2.3 months. MRI responses followed a linear course with a 25% reduction after 16.8 months. Reduction of metabolically active tumor volumes, but not reduction of PET uptake ratios or MRI tumor volumes, correlated with improved seizure control following chemotherapy (P = .012). Receiver-operating-characteristic curve analysis showed that a decrease of the active tumor volume of ≥80.5% predicts a PFS of ≥60 months (P = .018) and a decrease of ≥64.5% a PFS of ≥48 months (P = .037). CONCLUSIONS Amino acid PET is superior to MRI for evaluating TMZ responses in WHO grade II glioma patients. The response delay between both imaging modalities favors amino acid PET for individually tailoring the duration of chemotherapy. Additional studies should investigate whether this personalized approach is appropriate with regard to outcome.
Collapse
Affiliation(s)
- Ulrich Roelcke
- Department of Neurology and Brain Tumor Center, Cantonal Hospital, Aarau, Switzerland (U.R.); Institute for Pharmacology and Toxicology, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Neuroscience Center, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Department of Neurology, Medical University, Innsbruck, Austria (M.N.); Department of Neuro-Oncology, University Hospital, Torino, Italy (R.R., R.S.); Department of Neurology, University Hospital, Switzerland (P.R., M.W.); Department of Oncology, University Hospital, Zürich, Switzerland (S.H.); Department of Neurology, University Hospital, Cologne, Germany, and Research Center, Jülich, Germany (N.G.); Medicina Nucleare, Istituto Nazionale dei Tumori, Milano, Italy (F.C.)
| | - Matthias T Wyss
- Department of Neurology and Brain Tumor Center, Cantonal Hospital, Aarau, Switzerland (U.R.); Institute for Pharmacology and Toxicology, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Neuroscience Center, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Department of Neurology, Medical University, Innsbruck, Austria (M.N.); Department of Neuro-Oncology, University Hospital, Torino, Italy (R.R., R.S.); Department of Neurology, University Hospital, Switzerland (P.R., M.W.); Department of Oncology, University Hospital, Zürich, Switzerland (S.H.); Department of Neurology, University Hospital, Cologne, Germany, and Research Center, Jülich, Germany (N.G.); Medicina Nucleare, Istituto Nazionale dei Tumori, Milano, Italy (F.C.)
| | - Martha Nowosielski
- Department of Neurology and Brain Tumor Center, Cantonal Hospital, Aarau, Switzerland (U.R.); Institute for Pharmacology and Toxicology, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Neuroscience Center, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Department of Neurology, Medical University, Innsbruck, Austria (M.N.); Department of Neuro-Oncology, University Hospital, Torino, Italy (R.R., R.S.); Department of Neurology, University Hospital, Switzerland (P.R., M.W.); Department of Oncology, University Hospital, Zürich, Switzerland (S.H.); Department of Neurology, University Hospital, Cologne, Germany, and Research Center, Jülich, Germany (N.G.); Medicina Nucleare, Istituto Nazionale dei Tumori, Milano, Italy (F.C.)
| | - Roberta Rudà
- Department of Neurology and Brain Tumor Center, Cantonal Hospital, Aarau, Switzerland (U.R.); Institute for Pharmacology and Toxicology, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Neuroscience Center, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Department of Neurology, Medical University, Innsbruck, Austria (M.N.); Department of Neuro-Oncology, University Hospital, Torino, Italy (R.R., R.S.); Department of Neurology, University Hospital, Switzerland (P.R., M.W.); Department of Oncology, University Hospital, Zürich, Switzerland (S.H.); Department of Neurology, University Hospital, Cologne, Germany, and Research Center, Jülich, Germany (N.G.); Medicina Nucleare, Istituto Nazionale dei Tumori, Milano, Italy (F.C.)
| | - Patrick Roth
- Department of Neurology and Brain Tumor Center, Cantonal Hospital, Aarau, Switzerland (U.R.); Institute for Pharmacology and Toxicology, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Neuroscience Center, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Department of Neurology, Medical University, Innsbruck, Austria (M.N.); Department of Neuro-Oncology, University Hospital, Torino, Italy (R.R., R.S.); Department of Neurology, University Hospital, Switzerland (P.R., M.W.); Department of Oncology, University Hospital, Zürich, Switzerland (S.H.); Department of Neurology, University Hospital, Cologne, Germany, and Research Center, Jülich, Germany (N.G.); Medicina Nucleare, Istituto Nazionale dei Tumori, Milano, Italy (F.C.)
| | - Silvia Hofer
- Department of Neurology and Brain Tumor Center, Cantonal Hospital, Aarau, Switzerland (U.R.); Institute for Pharmacology and Toxicology, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Neuroscience Center, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Department of Neurology, Medical University, Innsbruck, Austria (M.N.); Department of Neuro-Oncology, University Hospital, Torino, Italy (R.R., R.S.); Department of Neurology, University Hospital, Switzerland (P.R., M.W.); Department of Oncology, University Hospital, Zürich, Switzerland (S.H.); Department of Neurology, University Hospital, Cologne, Germany, and Research Center, Jülich, Germany (N.G.); Medicina Nucleare, Istituto Nazionale dei Tumori, Milano, Italy (F.C.)
| | - Norbert Galldiks
- Department of Neurology and Brain Tumor Center, Cantonal Hospital, Aarau, Switzerland (U.R.); Institute for Pharmacology and Toxicology, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Neuroscience Center, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Department of Neurology, Medical University, Innsbruck, Austria (M.N.); Department of Neuro-Oncology, University Hospital, Torino, Italy (R.R., R.S.); Department of Neurology, University Hospital, Switzerland (P.R., M.W.); Department of Oncology, University Hospital, Zürich, Switzerland (S.H.); Department of Neurology, University Hospital, Cologne, Germany, and Research Center, Jülich, Germany (N.G.); Medicina Nucleare, Istituto Nazionale dei Tumori, Milano, Italy (F.C.)
| | - Flavio Crippa
- Department of Neurology and Brain Tumor Center, Cantonal Hospital, Aarau, Switzerland (U.R.); Institute for Pharmacology and Toxicology, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Neuroscience Center, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Department of Neurology, Medical University, Innsbruck, Austria (M.N.); Department of Neuro-Oncology, University Hospital, Torino, Italy (R.R., R.S.); Department of Neurology, University Hospital, Switzerland (P.R., M.W.); Department of Oncology, University Hospital, Zürich, Switzerland (S.H.); Department of Neurology, University Hospital, Cologne, Germany, and Research Center, Jülich, Germany (N.G.); Medicina Nucleare, Istituto Nazionale dei Tumori, Milano, Italy (F.C.)
| | - Michael Weller
- Department of Neurology and Brain Tumor Center, Cantonal Hospital, Aarau, Switzerland (U.R.); Institute for Pharmacology and Toxicology, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Neuroscience Center, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Department of Neurology, Medical University, Innsbruck, Austria (M.N.); Department of Neuro-Oncology, University Hospital, Torino, Italy (R.R., R.S.); Department of Neurology, University Hospital, Switzerland (P.R., M.W.); Department of Oncology, University Hospital, Zürich, Switzerland (S.H.); Department of Neurology, University Hospital, Cologne, Germany, and Research Center, Jülich, Germany (N.G.); Medicina Nucleare, Istituto Nazionale dei Tumori, Milano, Italy (F.C.)
| | - Riccardo Soffietti
- Department of Neurology and Brain Tumor Center, Cantonal Hospital, Aarau, Switzerland (U.R.); Institute for Pharmacology and Toxicology, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Neuroscience Center, ETH and University of Zürich, Zürich, Switzerland (M.T.W.); Department of Neurology, Medical University, Innsbruck, Austria (M.N.); Department of Neuro-Oncology, University Hospital, Torino, Italy (R.R., R.S.); Department of Neurology, University Hospital, Switzerland (P.R., M.W.); Department of Oncology, University Hospital, Zürich, Switzerland (S.H.); Department of Neurology, University Hospital, Cologne, Germany, and Research Center, Jülich, Germany (N.G.); Medicina Nucleare, Istituto Nazionale dei Tumori, Milano, Italy (F.C.)
| |
Collapse
|
919
|
Ellis HP, Greenslade M, Powell B, Spiteri I, Sottoriva A, Kurian KM. Current Challenges in Glioblastoma: Intratumour Heterogeneity, Residual Disease, and Models to Predict Disease Recurrence. Front Oncol 2015; 5:251. [PMID: 26636033 PMCID: PMC4644939 DOI: 10.3389/fonc.2015.00251] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 10/29/2015] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma (GB) is the most common primary malignant brain tumor, and despite the availability of chemotherapy and radiotherapy to combat the disease, overall survival remains low with a high incidence of tumor recurrence. Technological advances are continually improving our understanding of the disease, and in particular, our knowledge of clonal evolution, intratumor heterogeneity, and possible reservoirs of residual disease. These may inform how we approach clinical treatment and recurrence in GB. Mathematical modeling (including neural networks) and strategies such as multiple sampling during tumor resection and genetic analysis of circulating cancer cells, may be of great future benefit to help predict the nature of residual disease and resistance to standard and molecular therapies in GB.
Collapse
Affiliation(s)
- Hayley P Ellis
- Brain Tumour Research Group, Institute of Clinical Neurosciences, University of Bristol , Bristol , UK
| | - Mark Greenslade
- Bristol Genetics Laboratory, North Bristol NHS Trust , Bristol , UK
| | - Ben Powell
- School of Mathematics, University of Bristol , Bristol , UK
| | - Inmaculada Spiteri
- Centre for Evolution and Cancer, The Institute of Cancer Research , London , UK
| | - Andrea Sottoriva
- Centre for Evolution and Cancer, The Institute of Cancer Research , London , UK
| | - Kathreena M Kurian
- Brain Tumour Research Group, Institute of Clinical Neurosciences, University of Bristol , Bristol , UK
| |
Collapse
|
920
|
De Mattos-Arruda L, Mayor R, Ng CKY, Weigelt B, Martínez-Ricarte F, Torrejon D, Oliveira M, Arias A, Raventos C, Tang J, Guerini-Rocco E, Martínez-Sáez E, Lois S, Marín O, de la Cruz X, Piscuoglio S, Towers R, Vivancos A, Peg V, Ramon y Cajal S, Carles J, Rodon J, González-Cao M, Tabernero J, Felip E, Sahuquillo J, Berger MF, Cortes J, Reis-Filho JS, Seoane J. Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma. Nat Commun 2015; 6:8839. [PMID: 26554728 PMCID: PMC5426516 DOI: 10.1038/ncomms9839] [Citation(s) in RCA: 602] [Impact Index Per Article: 60.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 10/08/2015] [Indexed: 12/16/2022] Open
Abstract
Cell-free circulating tumour DNA (ctDNA) in plasma has been shown to be informative of the genomic alterations present in tumours and has been used to monitor tumour progression and response to treatments. However, patients with brain tumours do not present with or present with low amounts of ctDNA in plasma precluding the genomic characterization of brain cancer through plasma ctDNA. Here we show that ctDNA derived from central nervous system tumours is more abundantly present in the cerebrospinal fluid (CSF) than in plasma. Massively parallel sequencing of CSF ctDNA more comprehensively characterizes the genomic alterations of brain tumours than plasma, allowing the identification of actionable brain tumour somatic mutations. We show that CSF ctDNA levels longitudinally fluctuate in time and follow the changes in brain tumour burden providing biomarkers to monitor brain malignancies. Moreover, CSF ctDNA is shown to facilitate and complement the diagnosis of leptomeningeal carcinomatosis. DNA circulating in the plasma of cancer patients carries features of the primary tumour, however such DNA is found in low levels in brain cancer patients. Here, the authors show that circulating tumour DNA can be detected in the cerebral spinal fluid of cancer patients and that this better recapitulates the primary tumour compared to DNA from the plasma.
Collapse
Affiliation(s)
- Leticia De Mattos-Arruda
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, P. Vall d'Hebron 119-129, 08035 Barcelona, Spain.,Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA.,Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Regina Mayor
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, P. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Charlotte K Y Ng
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| | - Francisco Martínez-Ricarte
- Universitat Autònoma de Barcelona, 08193 Barcelona, Spain.,Vall d'Hebron Institute of Research, Vall d'Hebron University Hospital, Ps Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Davis Torrejon
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, P. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Mafalda Oliveira
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, P. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Alexandra Arias
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, P. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Carolina Raventos
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, P. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Jiabin Tang
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Elena Guerini-Rocco
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| | - Elena Martínez-Sáez
- Vall d'Hebron Institute of Research, Vall d'Hebron University Hospital, Ps Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Sergio Lois
- Vall d'Hebron Institute of Research, Vall d'Hebron University Hospital, Ps Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Oscar Marín
- Vall d'Hebron Institute of Research, Vall d'Hebron University Hospital, Ps Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Xavier de la Cruz
- Vall d'Hebron Institute of Research, Vall d'Hebron University Hospital, Ps Vall d'Hebron 119-129, 08035 Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Salvatore Piscuoglio
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| | - Russel Towers
- Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Ana Vivancos
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, P. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Vicente Peg
- Vall d'Hebron Institute of Research, Vall d'Hebron University Hospital, Ps Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Santiago Ramon y Cajal
- Universitat Autònoma de Barcelona, 08193 Barcelona, Spain.,Vall d'Hebron Institute of Research, Vall d'Hebron University Hospital, Ps Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Joan Carles
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, P. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Jordi Rodon
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, P. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | | | - Josep Tabernero
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, P. Vall d'Hebron 119-129, 08035 Barcelona, Spain.,Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Enriqueta Felip
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, P. Vall d'Hebron 119-129, 08035 Barcelona, Spain.,Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Joan Sahuquillo
- Universitat Autònoma de Barcelona, 08193 Barcelona, Spain.,Vall d'Hebron Institute of Research, Vall d'Hebron University Hospital, Ps Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Michael F Berger
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Javier Cortes
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, P. Vall d'Hebron 119-129, 08035 Barcelona, Spain.,Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| | - Joan Seoane
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, P. Vall d'Hebron 119-129, 08035 Barcelona, Spain.,Universitat Autònoma de Barcelona, 08193 Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
921
|
Cooley M, Fang P, Fang F, Nephew KP, P Nephew K, Chien J. Molecular determinants of chemotherapy resistance in ovarian cancer. Pharmacogenomics 2015; 16:1763-7. [PMID: 26554863 DOI: 10.2217/pgs.15.130] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Megan Cooley
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Pingping Fang
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Fang Fang
- Department of Cellular & Integrative Physiology, Medical Sciences Program, Indiana University, Bloomington, IN 47405, USA
| | | | - Kenneth P Nephew
- Department of Cellular & Integrative Physiology, Medical Sciences Program, Indiana University, Bloomington, IN 47405, USA
| | - Jeremy Chien
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
922
|
Gatto F, Nielsen J. In search for symmetries in the metabolism of cancer. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2015; 8:23-35. [PMID: 26538017 DOI: 10.1002/wsbm.1321] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 09/18/2015] [Accepted: 09/23/2015] [Indexed: 12/12/2022]
Abstract
Even though aerobic glycolysis, or the Warburg effect, is arguably the most common trait of metabolic reprogramming in cancer, it is unobserved in certain tumor types. Systems biology advocates a global view on metabolism to dissect which traits are consistently reprogrammed in cancer, and hence likely to constitute an obligate step for the evolution of cancer cells. We refer to such traits as symmetric. Here, we review early systems biology studies that attempted to reveal symmetric traits in the metabolic reprogramming of cancer, discuss the symmetry of reprogramming of nucleotide metabolism, and outline the current limitations that, if unlocked, could elucidate whether symmetries in cancer metabolism may be claimed.
Collapse
Affiliation(s)
- Francesco Gatto
- Department of Biology and Biological Engineering, Chalmers University of Technology, Göteborg, Sweden
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Göteborg, Sweden
| |
Collapse
|
923
|
Parmiani G, Russo V, Maccalli C, Parolini D, Rizzo N, Maio M. Peptide-based vaccines for cancer therapy. Hum Vaccin Immunother 2015; 10:3175-8. [PMID: 25483658 DOI: 10.4161/hv.29418] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Interest for cancer vaccination started more than 30 years ago after the demonstration that both in animal models and later on in patients it is possible to generate anti-tumor immune responses. The clinical application of this knowledge, however, was disappointing. In this review we summarize results on peptides epitopes recognized by T cells that have been studied thanks to their easy synthesis and the lack of significant side effects when administered in-vivo. To improve the clinical efficacy, peptides were modified in their aminoacid sequence to augment their immunogenicity. Peptides vaccines were recently shown to induce a high frequency of immune response in patients that were accompanied by clinical efficacy. These data are discussed at the light of recent progression of immunotherapy caused by the addition of check-point antibodies thus providing a general picture of the potential therapeutic efficacy of the peptide-based vaccines and their combination with other biological agents.
Collapse
|
924
|
Schroeder B, Shah N, Rostad S, McCullough B, Aguedan B, Foltz G, Cobbs C. Genetic investigation of multicentric glioblastoma multiforme: case report. J Neurosurg 2015; 124:1353-8. [PMID: 26473785 DOI: 10.3171/2015.4.jns142231] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The authors report a case of multicentric glioblastoma multiforme (GBM) in which all 4 tumor foci were resected and evaluated using both comparative genomic hybridization array and RNA sequencing. Genetic analysis showed that the tumors shared a common origin, although each had its own unique set of genetic aberrations. The authors note that the genetic heterogeneity of multicentric GBM likely contributes to the failures of current treatments. The case underscores the necessity of increased genetic investigation.
Collapse
Affiliation(s)
- Brett Schroeder
- Michigan State University College of Medicine, Grand Rapids, Michigan;,Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute
| | - Nameeta Shah
- Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute
| | - Steve Rostad
- CellNetix Pathology & Laboratories, Seattle; and
| | | | - Brian Aguedan
- Department of Radiology, Radia Inc., Everett, Washington
| | - Greg Foltz
- Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute
| | - Charles Cobbs
- Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute
| |
Collapse
|
925
|
Recurrent Glioblastomas Reveal Molecular Subtypes Associated with Mechanistic Implications of Drug-Resistance. PLoS One 2015; 10:e0140528. [PMID: 26466313 PMCID: PMC4605710 DOI: 10.1371/journal.pone.0140528] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/28/2015] [Indexed: 01/04/2023] Open
Abstract
Previously, transcriptomic profiling studies have shown distinct molecular subtypes of glioblastomas. It has also been suggested that the recurrence of glioblastomas could be achieved by transcriptomic reprograming of tumors, however, their characteristics are not yet fully understood. Here, to gain the mechanistic insights on the molecular phenotypes of recurrent glioblastomas, gene expression profiling was performed on the 43 cases of glioblastomas including 15 paired primary and recurrent cases. Unsupervised clustering analyses revealed two subtypes of G1 and G2, which were characterized by proliferation and neuron-like gene expression traits, respectively. While the primary tumors were classified as G1 subtype, the recurrent glioblastomas showed two distinct expression types. Compared to paired primary tumors, the recurrent tumors in G1 subtype did not show expression alteration. By contrast, the recurrent tumors in G2 subtype showed expression changes from proliferation type to neuron-like one. We also observed the expression of stemness-related genes in G1 recurrent tumors and the altered expression of DNA-repair genes (i.e., AURK, HOX, MGMT, and MSH6) in the G2 recurrent tumors, which might be responsible for the acquisition of drug resistance mechanism during tumor recurrence in a subtype-specific manner. We suggest that recurrent glioblastomas may choose two different strategies for transcriptomic reprograming to escape the chemotherapeutic treatment during tumor recurrence. Our results might be helpful to determine personalized therapeutic strategy against heterogeneous glioma recurrence.
Collapse
|
926
|
Lee Y, Kim KH, Kim DG, Cho HJ, Kim Y, Rheey J, Shin K, Seo YJ, Choi YS, Lee JI, Lee J, Joo KM, Nam DH. FoxM1 Promotes Stemness and Radio-Resistance of Glioblastoma by Regulating the Master Stem Cell Regulator Sox2. PLoS One 2015; 10:e0137703. [PMID: 26444992 PMCID: PMC4596841 DOI: 10.1371/journal.pone.0137703] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 08/19/2015] [Indexed: 01/19/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive and most lethal brain tumor. As current standard therapy consisting of surgery and chemo-irradiation provides limited benefit for GBM patients, novel therapeutic options are urgently required. Forkhead box M1 (FoxM1) transcription factor is an oncogenic regulator that promotes the proliferation, survival, and treatment resistance of various human cancers. The roles of FoxM1 in GBM remain incompletely understood, due in part to pleotropic nature of the FoxM1 pathway. Here, we show the roles of FoxM1 in GBM stem cell maintenance and radioresistance. ShRNA-mediated FoxM1 inhibition significantly impeded clonogenic growth and survival of patient-derived primary GBM cells with marked downregulation of Sox2, a master regulator of stem cell phenotype. Ectopic expression of Sox2 partially rescued FoxM1 inhibition-mediated effects. Conversely, FoxM1 overexpression upregulated Sox2 expression and promoted clonogenic growth of GBM cells. These data, with a direct binding of FoxM1 in the Sox2 promoter region in GBM cells, suggest that FoxM1 regulates stemness of primary GBM cells via Sox2. We also found significant increases in FoxM1 and Sox2 expression in GBM cells after irradiation both in vitro and in vivo orthotopic tumor models. Notably, genetic or a small-molecule FoxM1 inhibitor-mediated FoxM1 targeting significantly sensitized GBM cells to irradiation, accompanying with Sox2 downregulation. Finally, FoxM1 inhibition combined with irradiation in a patient GBM-derived orthotopic model significantly impeded tumor growth and prolonged the survival of tumor bearing mice. Taken together, these results indicate that the FoxM1-Sox2 signaling axis promotes clonogenic growth and radiation resistance of GBM, and suggest that FoxM1 targeting combined with irradiation is a potentially effective therapeutic approach for GBM.
Collapse
Affiliation(s)
- Yeri Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Kang Ho Kim
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, South Korea
| | - Dong Geon Kim
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Hee Jin Cho
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Yeonghwan Kim
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States of America
| | - Jinguen Rheey
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, South Korea
| | - Kayoung Shin
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Yun Jee Seo
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yeon-Sook Choi
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jung-Il Lee
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jeongwu Lee
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States of America
| | - Kyeung Min Joo
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, South Korea
- Department of Anatomy and cell biology, Sungkyunkwan University School of Medicine, Suwon, South Korea
- * E-mail: (DHN); (KMJ)
| | - Do-Hyun Nam
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- * E-mail: (DHN); (KMJ)
| |
Collapse
|
927
|
SubClonal Hierarchy Inference from Somatic Mutations: Automatic Reconstruction of Cancer Evolutionary Trees from Multi-region Next Generation Sequencing. PLoS Comput Biol 2015; 11:e1004416. [PMID: 26436540 PMCID: PMC4593588 DOI: 10.1371/journal.pcbi.1004416] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 06/27/2015] [Indexed: 01/10/2023] Open
Abstract
Recent improvements in next-generation sequencing of tumor samples and the ability to identify somatic mutations at low allelic fractions have opened the way for new approaches to model the evolution of individual cancers. The power and utility of these models is increased when tumor samples from multiple sites are sequenced. Temporal ordering of the samples may provide insight into the etiology of both primary and metastatic lesions and rationalizations for tumor recurrence and therapeutic failures. Additional insights may be provided by temporal ordering of evolving subclones—cellular subpopulations with unique mutational profiles. Current methods for subclone hierarchy inference tightly couple the problem of temporal ordering with that of estimating the fraction of cancer cells harboring each mutation. We present a new framework that includes a rigorous statistical hypothesis test and a collection of tools that make it possible to decouple these problems, which we believe will enable substantial progress in the field of subclone hierarchy inference. The methods presented here can be flexibly combined with methods developed by others addressing either of these problems. We provide tools to interpret hypothesis test results, which inform phylogenetic tree construction, and we introduce the first genetic algorithm designed for this purpose. The utility of our framework is systematically demonstrated in simulations. For most tested combinations of tumor purity, sequencing coverage, and tree complexity, good power (≥ 0.8) can be achieved and Type 1 error is well controlled when at least three tumor samples are available from a patient. Using data from three published multi-region tumor sequencing studies of (murine) small cell lung cancer, acute myeloid leukemia, and chronic lymphocytic leukemia, in which the authors reconstructed subclonal phylogenetic trees by manual expert curation, we show how different configurations of our tools can identify either a single tree in agreement with the authors, or a small set of trees, which include the authors’ preferred tree. Our results have implications for improved modeling of tumor evolution and the importance of multi-region tumor sequencing. Cancer is a genetic disease, driven by DNA mutations. Each tumor is composed of millions of cells with differing genetic profiles that compete with each other for resources in a process similar to Darwinian evolution. We describe a computational framework to model tumor evolution on the cellular level, using next-generation sequencing. The framework is the first to apply a rigorous statistical hypothesis test designed to inform a new search algorithm. Both the test and the algorithm are based on evolutionary principles. The utility of the framework is shown in computer simulations and by automated reconstruction of the cellular evolution underlying murine small cell lung cancers, acute myeloid leukemias and chronic lymophocytic leukemias, from three recent published studies.
Collapse
|
928
|
Apostoli AJ, Ailles L. Clonal evolution and tumor-initiating cells: New dimensions in cancer patient treatment. Crit Rev Clin Lab Sci 2015; 53:40-51. [PMID: 26397062 DOI: 10.3109/10408363.2015.1083944] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Human cancer is not a uniform disease but a plethora of disparate tumor types and subtypes. The differences that exist between individual tumors (intertumoral heterogeneity) present a significant roadblock to the eradication of cancer. It has also become increasingly clear that variations across individual tumors (intratumoral heterogeneity) have important implications to cancer progression and treatment efficacy. Therefore, in order to improve patient care and develop novel chemotherapeutics, the evolving tumor landscape needs to be further explored. Next-generation sequencing (NGS) technologies are revolutionizing the cancer research arena by providing state-of-the-art, high-speed methods of genome sequencing at single-nucleotide resolution, thus enabling an unprecedented detection of tumor-specific genetic abnormalities. These anomalies can be quantified to reveal specific frequencies of DNA alterations that correspond to distinct clonal populations within a given tumor. As such, NGS approaches have also been utilized to explore the heterogeneous landscape of patient tumors as well as to match metastatic and/or recurrent growths and patient-derived engrafts. By sequencing in this manner--through time so to speak--cancer researchers can track shifting clonal populations, make important inferences about tumor evolution and potentially identify tumor subclones that could be viably targeted. This exciting new territory has important implications for the competing clonal evolution and cancer stem cell models of tumor heterogeneity, and also offers a new dimension for cancer treatment and profound hope for patients in the coming years.
Collapse
Affiliation(s)
- Anthony J Apostoli
- a Princess Margaret Cancer Centre, University Health Network , Toronto , Ontario , Canada and
| | - Laurie Ailles
- a Princess Margaret Cancer Centre, University Health Network , Toronto , Ontario , Canada and.,b Department of Medical Biophysics , University of Toronto , Toronto , Ontario , Canada
| |
Collapse
|
929
|
Kim J, Lee IH, Cho HJ, Park CK, Jung YS, Kim Y, Nam SH, Kim BS, Johnson MD, Kong DS, Seol HJ, Lee JI, Joo KM, Yoon Y, Park WY, Lee J, Park PJ, Nam DH. Spatiotemporal Evolution of the Primary Glioblastoma Genome. Cancer Cell 2015; 28:318-28. [PMID: 26373279 DOI: 10.1016/j.ccell.2015.07.013] [Citation(s) in RCA: 236] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 06/10/2015] [Accepted: 07/28/2015] [Indexed: 12/11/2022]
Abstract
Tumor recurrence following treatment is the major cause of mortality for glioblastoma multiforme (GBM) patients. Thus, insights on the evolutionary process at recurrence are critical for improved patient care. Here, we describe our genomic analyses of the initial and recurrent tumor specimens from each of 38 GBM patients. A substantial divergence in the landscape of driver alterations was associated with distant appearance of a recurrent tumor from the initial tumor, suggesting that the genomic profile of the initial tumor can mislead targeted therapies for the distally recurred tumor. In addition, in contrast to IDH1-mutated gliomas, IDH1-wild-type primary GBMs rarely developed hypermutation following temozolomide (TMZ) treatment, indicating low risk for TMZ-induced hypermutation for these tumors under the standard regimen.
Collapse
Affiliation(s)
- Jinkuk Kim
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul 06351, Korea; Samsung Advanced Institute of Technology, Samsung Electronics Co., Ltd., Seoul 06351, Korea
| | - In-Hee Lee
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul 06351, Korea; Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Korea
| | - Hee Jin Cho
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul 06351, Korea; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Science and Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Korea
| | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 06351, Korea
| | - Yang-Soon Jung
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Yanghee Kim
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul 06351, Korea
| | - So Hee Nam
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul 06351, Korea
| | - Byung Sup Kim
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Mark D Johnson
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Doo-Sik Kong
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Ho Jun Seol
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Jung-Il Lee
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Kyeung Min Joo
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul 06351, Korea; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Science and Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Korea; Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Yeup Yoon
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul 06351, Korea; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Science and Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Korea
| | - Woong-Yang Park
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul 06351, Korea; Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Korea; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Science and Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Korea
| | - Jeongwu Lee
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Peter J Park
- Division of Genetics, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA.
| | - Do-Hyun Nam
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul 06351, Korea; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Science and Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Korea; Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.
| |
Collapse
|
930
|
Mazor T, Pankov A, Johnson BE, Hong C, Hamilton EG, Bell RJ, Smirnov IV, Reis GF, Phillips JJ, Barnes MJ, Idbaih A, Alentorn A, Kloezeman JJ, Lamfers MLM, Bollen AW, Taylor BS, Molinaro AM, Olshen AB, Chang SM, Song JS, Costello JF. DNA Methylation and Somatic Mutations Converge on the Cell Cycle and Define Similar Evolutionary Histories in Brain Tumors. Cancer Cell 2015; 28:307-317. [PMID: 26373278 PMCID: PMC4573399 DOI: 10.1016/j.ccell.2015.07.012] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 05/21/2015] [Accepted: 07/28/2015] [Indexed: 12/20/2022]
Abstract
The evolutionary history of tumor cell populations can be reconstructed from patterns of genetic alterations. In contrast to stable genetic events, epigenetic states are reversible and sensitive to the microenvironment, prompting the question whether epigenetic information can similarly be used to discover tumor phylogeny. We examined the spatial and temporal dynamics of DNA methylation in a cohort of low-grade gliomas and their patient-matched recurrences. Genes transcriptionally upregulated through promoter hypomethylation during malignant progression to high-grade glioblastoma were enriched in cell cycle function, evolving in parallel with genetic alterations that deregulate the G1/S cell cycle checkpoint. Moreover, phyloepigenetic relationships robustly recapitulated phylogenetic patterns inferred from somatic mutations. These findings highlight widespread co-dependency of genetic and epigenetic events throughout brain tumor evolution.
Collapse
Affiliation(s)
- Tali Mazor
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94158, USA
| | - Aleksandr Pankov
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94158, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Brett E. Johnson
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94158, USA
| | - Chibo Hong
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94158, USA
| | - Emily G. Hamilton
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94158, USA
| | - Robert J.A. Bell
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94158, USA
| | - Ivan V. Smirnov
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94158, USA
| | - Gerald F. Reis
- Department of Pathology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Joanna J. Phillips
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94158, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Michael J. Barnes
- Department of Pathology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Ahmed Idbaih
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Service de Neurologie 2-Mazarin, F-75013, Paris, France
| | - Agusti Alentorn
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Service de Neurologie 2-Mazarin, F-75013, Paris, France
| | - Jenneke J. Kloezeman
- Department of Neurosurgery, Brain Tumor Center, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Martine L. M. Lamfers
- Department of Neurosurgery, Brain Tumor Center, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Andrew W. Bollen
- Department of Pathology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Barry S. Taylor
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Annette M. Molinaro
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94158, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Adam B. Olshen
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158, USA
| | - Susan M. Chang
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94158, USA
| | - Jun S. Song
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA 94158, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Champaign, IL 61801, USA
- Department of Physics, University of Illinois at Urbana-Champaign, Champaign, IL 61801, USA
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, IL 61801, USA
| | - Joseph F. Costello
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94158, USA
- Correspondence: (J.F.C)
| |
Collapse
|
931
|
Abstract
In this issue of Cancer Cell, studies from Mazor and colleagues and Kim and colleagues use a combination of epigenetic and genetic approaches to reveal a complex evolutionary process underlying two of the biggest challenges facing neuro-oncology, specifically glioblastoma malignant progression and treatment resistance.
Collapse
Affiliation(s)
- Vijay Ramaswamy
- Division of Haematology/Oncology, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Michael D Taylor
- Division of Neurosurgery, Program in Developmental and Stem Cell Biology, Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada.
| |
Collapse
|
932
|
Shiroishi MS, Boxerman JL, Pope WB. Physiologic MRI for assessment of response to therapy and prognosis in glioblastoma. Neuro Oncol 2015; 18:467-78. [PMID: 26364321 DOI: 10.1093/neuonc/nov179] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 08/01/2015] [Indexed: 02/06/2023] Open
Abstract
Aside from bidimensional measurements from conventional contrast-enhanced MRI, there are no validated or FDA-qualified imaging biomarkers for high-grade gliomas. However, advanced functional MRI techniques, including perfusion- and diffusion-weighted MRI, have demonstrated much potential for determining prognosis, predicting therapeutic response, and assessing early treatment response. They may also prove useful for differentiating pseudoprogression from true progression after temozolomide chemoradiation and pseudoresponse from true response after anti-angiogenic therapy. This review will highlight recent developments using these techniques and emphasize the need for technical standardization and validation in prospective studies in order for these methods to become incorporated into standard-of-care imaging for brain tumor patients.
Collapse
Affiliation(s)
- Mark S Shiroishi
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California (M.S.S.); Department of Diagnostic Imaging, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.); Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, California (W.B.P.)
| | - Jerrold L Boxerman
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California (M.S.S.); Department of Diagnostic Imaging, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.); Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, California (W.B.P.)
| | - Whitney B Pope
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California (M.S.S.); Department of Diagnostic Imaging, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.); Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, California (W.B.P.)
| |
Collapse
|
933
|
Sherborne AL, Davidson PR, Yu K, Nakamura AO, Rashid M, Nakamura JL. Mutational Analysis of Ionizing Radiation Induced Neoplasms. Cell Rep 2015; 12:1915-26. [PMID: 26344771 DOI: 10.1016/j.celrep.2015.08.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 07/08/2015] [Accepted: 08/05/2015] [Indexed: 10/25/2022] Open
Abstract
Ionizing radiation (IR) is a mutagen that promotes tumorigenesis in multiple exposure contexts. One severe consequence of IR is the development of second malignant neoplasms (SMNs), a radiotherapy-associated complication in survivors of cancers, particularly pediatric cancers. SMN genomes are poorly characterized, and the influence of genetic background on genotoxin-induced mutations has not been examined. Using our mouse models of SMNs, we performed whole exome sequencing of neoplasms induced by fractionated IR in wild-type and Nf1 mutant mice. Using non-negative matrix factorization, we identified mutational signatures that did not segregate by genetic background or histology. Copy-number analysis revealed recurrent chromosomal alterations and differences in copy number that were background dependent. Pathway analysis identified enrichment of non-synonymous variants in genes responsible for cell assembly and organization, cell morphology, and cell function and maintenance. In this model system, ionizing radiation and Nf1 heterozygosity each exerted distinct influences on the mutational landscape.
Collapse
Affiliation(s)
- Amy L Sherborne
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Philip R Davidson
- Department of Finance and Statistical Analysis, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Katharine Yu
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Alice O Nakamura
- Department of Finance and Statistical Analysis, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Mamunur Rashid
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Jean L Nakamura
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
934
|
Zhang RR, Pointer KB, Kuo JS, Dempsey RJ. Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma. Neurosurgery 2015; 75:N9-10. [PMID: 25406627 DOI: 10.1227/neu.0000000000000580] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
935
|
Devarakonda S, Govindan R. Clonal Evolution: Multiregion Sequencing of Esophageal Adenocarcinoma Before and After Chemotherapy. Cancer Discov 2015; 5:796-8. [DOI: 10.1158/2159-8290.cd-15-0739] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
936
|
Kang H, Salomon MP, Sottoriva A, Zhao J, Toy M, Press MF, Curtis C, Marjoram P, Siegmund K, Shibata D. Many private mutations originate from the first few divisions of a human colorectal adenoma. J Pathol 2015; 237:355-62. [PMID: 26119426 DOI: 10.1002/path.4581] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/23/2015] [Accepted: 06/24/2015] [Indexed: 01/08/2023]
Abstract
Intratumoural mutational heterogeneity (ITH) or the presence of different private mutations in different parts of the same tumour is commonly observed in human tumours. The mechanisms generating such ITH are uncertain. Here we find that ITH can be remarkably well structured by measuring point mutations, chromosome copy numbers, and DNA passenger methylation from opposite sides and individual glands of a 6 cm human colorectal adenoma. ITH was present between tumour sides and individual glands, but the private mutations were side-specific and subdivided the adenoma into two major subclones. Furthermore, ITH disappeared within individual glands because the glands were clonal populations composed of cells with identical mutant genotypes. Despite mutation clonality, the glands were relatively old, diverse populations when their individual cells were compared for passenger methylation and by FISH. These observations can be organized into an expanding star-like ancestral tree with co-clonal expansion, where many private mutations and multiple related clones arise during the first few divisions. As a consequence, most detectable mutational ITH in the final tumour originates from the first few divisions. Much of the early history of a tumour, especially the first few divisions, may be embedded within the detectable ITH of tumour genomes.
Collapse
Affiliation(s)
- Haeyoun Kang
- Department of Pathology, CHA University, Seongnam-si, South Korea
| | - Matthew P Salomon
- Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Andrea Sottoriva
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Junsong Zhao
- Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Morgan Toy
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Michael F Press
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Christina Curtis
- Department of Medicine, Stanford University, Stanford, California, USA
| | - Paul Marjoram
- Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Kimberly Siegmund
- Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Darryl Shibata
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| |
Collapse
|
937
|
Molecular subtypes, stem cells and heterogeneity: Implications for personalised therapy in glioma. J Clin Neurosci 2015; 22:1219-26. [DOI: 10.1016/j.jocn.2015.02.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 02/14/2015] [Indexed: 01/08/2023]
|
938
|
Piccirillo SGM, Spiteri I. Intratumor heterogeneity and transcriptional profiling in glioblastoma: translational opportunities. FUTURE NEUROLOGY 2015. [DOI: 10.2217/fnl.15.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The study of phenotypic and genetic intratumor heterogeneity in glioblastoma is attracting a lot of attention. Recent studies have demonstrated that transcriptional profiling analysis can help interpret the complexity of this disease. Previously proposed molecular classifiers have been recently challenged due to the unexpected degree of intratumor heterogeneity that has been described spatially and at single-cell level. Different computational methods have been employed to analyze this huge amount of data, but new experimental designs including multisampling from individual patients and single-cell experiments require new specific approaches. In light of these results, there is hope that integration of genetic, phenotypic and transcriptional data coupled with functional experiments might help define new therapeutic strategies and classify patients according to key pathways and molecular targets that can be further investigated to develop personalized and combinatorial treatment strategies.
Collapse
Affiliation(s)
- Sara GM Piccirillo
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Inmaculada Spiteri
- The Institute of Cancer Research, Centre for Evolution and Cancer, 15 Cotswold Road, Sutton SM2 5NG, UK
| |
Collapse
|
939
|
Gleize V, Alentorn A, Connen de Kérillis L, Labussière M, Nadaradjane AA, Mundwiller E, Ottolenghi C, Mangesius S, Rahimian A, Ducray F, Mokhtari K, Villa C, Sanson M. CIC inactivating mutations identify aggressive subset of 1p19q codeleted gliomas. Ann Neurol 2015; 78:355-74. [PMID: 26017892 DOI: 10.1002/ana.24443] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 05/25/2015] [Accepted: 05/26/2015] [Indexed: 11/10/2022]
Abstract
OBJECTIVE CIC gene is frequently mutated in oligodendroglial tumors with 1p19q codeletion. However, clinical and biological impact remain poorly understood. METHODS We sequenced the CIC gene on 127 oligodendroglial tumors (109 with the 1p19q codeletion) and analyzed patients' outcome. We compared magnetic resonance imaging, transcriptomic profile, and CIC protein expression of CIC wild-type (WT) and mutant gliomas. We compared the level of expression of CIC target genes on Hs683-IDH1(R132H) cells transfected with lentivirus encoding mutant and WT CIC. RESULTS We found 63 mutations affecting 60 of 127 patients, virtually all 1p19q codeleted and IDH mutated (59 of 60). In the 1p19q codeleted gliomas, CIC mutations were associated with a poorer outcome by uni- (p = 0.001) and multivariate analysis (p < 0.016). CIC mutation prognostic impact was validated on the TCGA cohort. CIC mutant grade II codeleted gliomas spontaneously grew faster than WTs. Transcriptomic analysis revealed an enrichment of proliferative pathways and oligodendrocyte precursor cell gene expression profile in CIC mutant gliomas, with upregulation of normally CIC repressed genes ETV1, ETV4, ETV5, and CCND1. Various missense mutations resulted in CIC protein expression loss. Moreover, a truncating CIC mutation resulted in a defect of nuclear targeting of CIC protein to the nucleus in a human glioma cell line expressing IDH1(R132H) and overexpression of CCND1 and other new target genes of CIC, such as DUSP4 and SPRED1. INTERPRETATION CIC mutations result in protein inactivation with upregulation of CIC target genes, activation of proliferative pathways, inhibition of differentiation, and poorer outcome in patients with a 1p19q codeleted glioma.
Collapse
Affiliation(s)
- Vincent Gleize
- Sorbonne Université, UPMC Univ Paris 06, Inserm, CNRS, UM 75, U 1127, UMR 7225, ICM, Paris, France
| | - Agusti Alentorn
- Sorbonne Université, UPMC Univ Paris 06, Inserm, CNRS, UM 75, U 1127, UMR 7225, ICM, Paris, France.,AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service de Neurologie 2, Paris, France
| | - Léa Connen de Kérillis
- Sorbonne Université, UPMC Univ Paris 06, Inserm, CNRS, UM 75, U 1127, UMR 7225, ICM, Paris, France
| | - Marianne Labussière
- Sorbonne Université, UPMC Univ Paris 06, Inserm, CNRS, UM 75, U 1127, UMR 7225, ICM, Paris, France
| | - Aravidan A Nadaradjane
- Sorbonne Université, UPMC Univ Paris 06, Inserm, CNRS, UM 75, U 1127, UMR 7225, ICM, Paris, France
| | - Emeline Mundwiller
- Institut du Cerveau et de la Moelle Epinière, Plateforme de Génotypage et Séquençage, Paris, France
| | - Chris Ottolenghi
- Biochimie Métabolique, Université Paris Descartes et Inserm U1124, Paris, France
| | - Stephanie Mangesius
- Sorbonne Université, UPMC Univ Paris 06, Inserm, CNRS, UM 75, U 1127, UMR 7225, ICM, Paris, France
| | | | | | | | - Karima Mokhtari
- Sorbonne Université, UPMC Univ Paris 06, Inserm, CNRS, UM 75, U 1127, UMR 7225, ICM, Paris, France.,AP-HP, Onconeurothèque, Paris, France.,AP-HP, Groupe Hospitalier Pitié Salpêtrière, Laboratoire de Neuropathologie R Escourolle, Paris, France
| | - Chiara Villa
- AP-HP, Groupe Hospitalier Pitié Salpêtrière, Laboratoire de Neuropathologie R Escourolle, Paris, France
| | - Marc Sanson
- Sorbonne Université, UPMC Univ Paris 06, Inserm, CNRS, UM 75, U 1127, UMR 7225, ICM, Paris, France.,AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service de Neurologie 2, Paris, France.,AP-HP, Onconeurothèque, Paris, France
| |
Collapse
|
940
|
Young RM, Jamshidi A, Davis G, Sherman JH. Current trends in the surgical management and treatment of adult glioblastoma. ANNALS OF TRANSLATIONAL MEDICINE 2015. [PMID: 26207249 DOI: 10.3978/j.issn.2305-5839.2015.05.10] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
This manuscript discusses the current surgical management of glioblastoma. This paper highlights the common pathophysiology attributes of glioblastoma, surgical options for diagnosis/treatment, current thoughts of extent of resection (EOR) of tumor, and post-operative (neo)adjuvant treatment. Glioblastoma is not a disease that can be cured with surgery alone, however safely performed maximal surgical resection is shown to significantly increase progression free and overall survival while maximizing quality of life. Upon invariable tumor recurrence, re-resection also is shown to impact survival in a select group of patients. As adjuvant therapy continues to improve survival, the role of surgical resection in the treatment of glioblastoma looks to be further defined.
Collapse
Affiliation(s)
- Richard M Young
- Department of Neurological Surgery, George Washington University Medical Center, Washington, DC 20037, USA
| | - Aria Jamshidi
- Department of Neurological Surgery, George Washington University Medical Center, Washington, DC 20037, USA
| | - Gregory Davis
- Department of Neurological Surgery, George Washington University Medical Center, Washington, DC 20037, USA
| | - Jonathan H Sherman
- Department of Neurological Surgery, George Washington University Medical Center, Washington, DC 20037, USA
| |
Collapse
|
941
|
Ramakrishna R, Pisapia D. Recent Molecular Advances in Our Understanding of Glioma. Cureus 2015; 7:e287. [PMID: 26244119 PMCID: PMC4523144 DOI: 10.7759/cureus.287] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 07/23/2015] [Indexed: 12/18/2022] Open
Abstract
Our molecular understanding of glioma has undergone a sea change over the last decade. In this review, we discuss two recent articles that employed whole genome sequencing to subclassify gliomas vis-à-vis known molecular alterations. We further discuss the relevance of these findings vis-à-vis current treatment paradigms.
Collapse
Affiliation(s)
- Rohan Ramakrishna
- Neurological Surgery, Weill Cornell Medical College ; Neurological Surgery, NewYork-Presbyterian/Weill Cornell Medical Center
| | - David Pisapia
- Pathology, Weill Cornell Medical College ; Pathology, New York Presbyterian Hospital
| |
Collapse
|
942
|
Tafe LJ, Gorlov IP, de Abreu FB, Lefferts JA, Liu X, Pettus JR, Marotti JD, Bloch KJ, Memoli VA, Suriawinata AA, Dragnev KH, Fadul CE, Schwartz GN, Morgan CR, Holderness BM, Peterson JD, Tsongalis GJ, Miller TW, Chamberlin MD. Implementation of a Molecular Tumor Board: The Impact on Treatment Decisions for 35 Patients Evaluated at Dartmouth-Hitchcock Medical Center. Oncologist 2015. [PMID: 26205736 DOI: 10.1634/theoncologist.2015-0097] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Although genetic profiling of tumors is a potentially powerful tool to predict drug sensitivity and resistance, its routine use has been limited because clinicians are often unfamiliar with interpretation and incorporation of the information into practice. We established a Molecular Tumor Board (MTB) to interpret individual patients' tumor genetic profiles and provide treatment recommendations. PATIENTS AND METHODS DNA from tumor specimens was sequenced in a Clinical Laboratory Improvement Amendments-certified laboratory to identify coding mutations in a 50-gene panel (n = 34) or a 255-gene panel (n = 1). Cases were evaluated by a multidisciplinary MTB that included pathologists, oncologists, hematologists, basic scientists, and genetic counselors. RESULTS During the first year, 35 cases were evaluated by the MTB, with 32 presented for recommendations on targeted therapies, and 3 referred for potential germline mutations. In 56.3% of cases, MTB recommended treatment with a targeted agent based on evaluation of tumor genetic profile and treatment history. Four patients (12.5%) were subsequently treated with a MTB-recommended targeted therapy; 3 of the 4 patients remain on therapy, 2 of whom experienced clinical benefit lasting >10 months. CONCLUSION For the majority of cases evaluated, the MTB was able to provide treatment recommendations based on targetable genetic alterations. The most common reasons that MTB-recommended therapy was not administered stemmed from patient preferences and genetic profiling at either very early or very late stages of disease; lack of drug access was rarely encountered. Increasing awareness of molecular profiling and targeted therapies by both clinicians and patients will improve acceptance and adherence to treatments that could significantly improve outcomes. IMPLICATIONS FOR PRACTICE Case evaluation by a multidisciplinary Molecular Tumor Board (MTB) is critical to benefit from individualized genetic data and maximize clinical impact. MTB recommendations shaped treatment options for the majority of cases evaluated. In the few patients treated with MTB-recommended therapy, disease outcomes were positive and support genetically informed treatment.
Collapse
Affiliation(s)
- Laura J Tafe
- Departments of Pathology, Community and Family Medicine, Medicine, and Pharmacology & Toxicology, Comprehensive Breast Program, and Familial Cancer Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Ivan P Gorlov
- Departments of Pathology, Community and Family Medicine, Medicine, and Pharmacology & Toxicology, Comprehensive Breast Program, and Familial Cancer Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Francine B de Abreu
- Departments of Pathology, Community and Family Medicine, Medicine, and Pharmacology & Toxicology, Comprehensive Breast Program, and Familial Cancer Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Joel A Lefferts
- Departments of Pathology, Community and Family Medicine, Medicine, and Pharmacology & Toxicology, Comprehensive Breast Program, and Familial Cancer Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Xiaoying Liu
- Departments of Pathology, Community and Family Medicine, Medicine, and Pharmacology & Toxicology, Comprehensive Breast Program, and Familial Cancer Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Jason R Pettus
- Departments of Pathology, Community and Family Medicine, Medicine, and Pharmacology & Toxicology, Comprehensive Breast Program, and Familial Cancer Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Jonathan D Marotti
- Departments of Pathology, Community and Family Medicine, Medicine, and Pharmacology & Toxicology, Comprehensive Breast Program, and Familial Cancer Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Kasia J Bloch
- Departments of Pathology, Community and Family Medicine, Medicine, and Pharmacology & Toxicology, Comprehensive Breast Program, and Familial Cancer Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Vincent A Memoli
- Departments of Pathology, Community and Family Medicine, Medicine, and Pharmacology & Toxicology, Comprehensive Breast Program, and Familial Cancer Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Arief A Suriawinata
- Departments of Pathology, Community and Family Medicine, Medicine, and Pharmacology & Toxicology, Comprehensive Breast Program, and Familial Cancer Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Konstantin H Dragnev
- Departments of Pathology, Community and Family Medicine, Medicine, and Pharmacology & Toxicology, Comprehensive Breast Program, and Familial Cancer Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Camilo E Fadul
- Departments of Pathology, Community and Family Medicine, Medicine, and Pharmacology & Toxicology, Comprehensive Breast Program, and Familial Cancer Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Gary N Schwartz
- Departments of Pathology, Community and Family Medicine, Medicine, and Pharmacology & Toxicology, Comprehensive Breast Program, and Familial Cancer Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Clinton R Morgan
- Departments of Pathology, Community and Family Medicine, Medicine, and Pharmacology & Toxicology, Comprehensive Breast Program, and Familial Cancer Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Britt M Holderness
- Departments of Pathology, Community and Family Medicine, Medicine, and Pharmacology & Toxicology, Comprehensive Breast Program, and Familial Cancer Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Jason D Peterson
- Departments of Pathology, Community and Family Medicine, Medicine, and Pharmacology & Toxicology, Comprehensive Breast Program, and Familial Cancer Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Gregory J Tsongalis
- Departments of Pathology, Community and Family Medicine, Medicine, and Pharmacology & Toxicology, Comprehensive Breast Program, and Familial Cancer Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Todd W Miller
- Departments of Pathology, Community and Family Medicine, Medicine, and Pharmacology & Toxicology, Comprehensive Breast Program, and Familial Cancer Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Mary D Chamberlin
- Departments of Pathology, Community and Family Medicine, Medicine, and Pharmacology & Toxicology, Comprehensive Breast Program, and Familial Cancer Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| |
Collapse
|
943
|
Agnihotri S, Aldape KD, Zadeh G. Isocitrate dehydrogenase status and molecular subclasses of glioma and glioblastoma. Neurosurg Focus 2015; 37:E13. [PMID: 25434382 DOI: 10.3171/2014.9.focus14505] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Diffuse gliomas and secondary glioblastomas (GBMs) that develop from low-grade gliomas are a common and incurable class of brain tumor. Mutations in the metabolic enzyme glioblastomas (IDH1) represent a distinguishing feature of low-grade gliomas and secondary GBMs. IDH1 mutations are one of the most common and earliest detectable genetic alterations in low-grade diffuse gliomas, and evidence supports this mutation as a driver of gliomagenesis. Here, the authors highlight the biological consequences of IDH1 mutations in gliomas, the clinical and therapeutic/diagnostic implications, and the molecular subtypes of these tumors. They also explore, in brief, the non-IDH1-mutated gliomas, including primary GBMs, and the molecular subtypes and drivers of these tumors. A fundamental understanding of the diversity of GBMs and lower-grade gliomas will ultimately allow for more effective treatments and predictors of survival.
Collapse
Affiliation(s)
- Sameer Agnihotri
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children
| | | | | |
Collapse
|
944
|
Abstract
PURPOSE OF REVIEW The primary treatment of low-grade gliomas is still claimed to lack robust supporting evidence. Yet, several investigations were performed in the last 2 decades. To critically review these studies could help in further clarifying the role of surgery aimed at maximal resection. RECENT FINDINGS Despite the lack of randomized clinical trials hampering the performance of appropriate meta-analyses, the increasing amount of evidence pointed toward an aggressive surgical strategy to low-grade glioma. Low-grade glioma surgery has to be performed with the appropriate armamentarium, which is the availability of intraoperative stimulation mapping, especially for those lesions occurring in cortical and subcortical eloquent sites. SUMMARY According to the recently published guidelines, surgical treatment has been increasingly recognized as the initial therapeutic act of choice for patients diagnosed with a presumed low-grade glioma, given that total resection can improve seizure control, progression-free survival and overall survival, while reducing the risk of malignant transformation and preserving patients' functional status.
Collapse
|
945
|
Aum DJ, Kim DH, Beaumont TL, Leuthardt EC, Dunn GP, Kim AH. Molecular and cellular heterogeneity: the hallmark of glioblastoma. Neurosurg Focus 2015; 37:E11. [PMID: 25434380 DOI: 10.3171/2014.9.focus14521] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
There has been increasing awareness that glioblastoma, which may seem histopathologically similar across many tumors, actually represents a group of molecularly distinct tumors. Emerging evidence suggests that cells even within the same tumor exhibit wide-ranging molecular diversity. Parallel to the discoveries of molecular heterogeneity among tumors and their individual cells, intense investigation of the cellular biology of glioblastoma has revealed that not all cancer cells within a given tumor behave the same. The identification of a subpopulation of brain tumor cells termed "glioblastoma cancer stem cells" or "tumor-initiating cells" has implications for the management of glioblastoma. This focused review will therefore summarize emerging concepts on the molecular and cellular heterogeneity of glioblastoma and emphasize that we should begin to consider each individual glioblastoma to be an ensemble of molecularly distinct subclones that reflect a spectrum of dynamic cell states.
Collapse
|
946
|
Agnihotri S, Zadeh G. Metabolic reprogramming in glioblastoma: the influence of cancer metabolism on epigenetics and unanswered questions. Neuro Oncol 2015; 18:160-72. [PMID: 26180081 DOI: 10.1093/neuonc/nov125] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 06/15/2015] [Indexed: 12/21/2022] Open
Abstract
A defining hallmark of glioblastoma is altered tumor metabolism. The metabolic shift towards aerobic glycolysis with reprogramming of mitochondrial oxidative phosphorylation, regardless of oxygen availability, is a phenomenon known as the Warburg effect. In addition to the Warburg effect, glioblastoma tumor cells also utilize the tricarboxylic acid cycle/oxidative phosphorylation in a different capacity than normal tissue. Altered metabolic enzymes and their metabolites are oncogenic and not simply a product of tumor proliferation. Here we highlight the advantages of why tumor cells, including glioblastoma cells, require metabolic reprogramming and how tumor metabolism can converge on tumor epigenetics and unanswered questions in the field.
Collapse
Affiliation(s)
- Sameer Agnihotri
- MacFeeters-Hamilton Brain Tumor Centre, Toronto, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada (S.A., G.Z.); Department of Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, Canada (G.Z)
| | - Gelareh Zadeh
- MacFeeters-Hamilton Brain Tumor Centre, Toronto, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada (S.A., G.Z.); Department of Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, Canada (G.Z)
| |
Collapse
|
947
|
van den Bent MJ, Gao Y, Kerkhof M, Kros JM, Gorlia T, van Zwieten K, Prince J, van Duinen S, Sillevis Smitt PA, Taphoorn M, French PJ. Changes in the EGFR amplification and EGFRvIII expression between paired primary and recurrent glioblastomas. Neuro Oncol 2015; 17:935-41. [PMID: 25691693 PMCID: PMC5762005 DOI: 10.1093/neuonc/nov013] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 01/13/2015] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The efficacy of novel targeted therapies is often tested at the time of tumor recurrence. However, for glioblastoma (GBM) patients, surgical resections at recurrence are performed only in a minority of patients; therefore, molecular data are predominantly derived from the initial tumor. Molecular data of the initial tumor for patient selection into personalized medicine trials can therefore be used only when the specific genetic change is retained in the recurrent tumor. METHODS In this study we determined whether EGFR amplification and expression of the most common mutation in GBMs (EGFRvIII) is retained at tumor recurrence. Because retention of genetic changes may be dependent on the initial treatment, we only used a cohort of GBM samples that were uniformly treated according to the current standard of care (ie, chemo-irradiation with temozolomide). RESULTS Our data show that, in spite of some quantitative differences, the EGFR amplification status remains stable in the majority (84%) of tumors evaluated. EGFRvIII expression remained similar in 79% of GBMs. However, within the tumors expressing EGFRvIII at initial diagnosis, approximately one-half lose their EGFRvIII expression at tumor recurrence. CONCLUSIONS The relative stability of EGFR amplification indicates that molecular data obtained in the primary tumor can be used to predict the EGFR status of the recurrent tumor, but care should be taken in extrapolating EGFRvIII expression from the primary tumor, particularly when expressed at first diagnosis.
Collapse
Affiliation(s)
- Martin J. van den Bent
- Department of Neurology, Erasmus MC, Rotterdam, Netherlands (M.J.v.d.B., Y.G., K.v.Z., J.P., P.A.S.S., P.J.F.); Department of Pathology, Erasmus MC, Rotterdam, Netherlands (J.M.K.); Department of Neurology, Haaglanden MC, The Hague, Netherlands (M.K., M.T.); EORTC Headquarters, Brussels, Belgium (T.G.); Pathology Department, Leiden University Medical Center, Leiden, Netherlands (S.v.D.)
| | - Ya Gao
- Department of Neurology, Erasmus MC, Rotterdam, Netherlands (M.J.v.d.B., Y.G., K.v.Z., J.P., P.A.S.S., P.J.F.); Department of Pathology, Erasmus MC, Rotterdam, Netherlands (J.M.K.); Department of Neurology, Haaglanden MC, The Hague, Netherlands (M.K., M.T.); EORTC Headquarters, Brussels, Belgium (T.G.); Pathology Department, Leiden University Medical Center, Leiden, Netherlands (S.v.D.)
| | - Melissa Kerkhof
- Department of Neurology, Erasmus MC, Rotterdam, Netherlands (M.J.v.d.B., Y.G., K.v.Z., J.P., P.A.S.S., P.J.F.); Department of Pathology, Erasmus MC, Rotterdam, Netherlands (J.M.K.); Department of Neurology, Haaglanden MC, The Hague, Netherlands (M.K., M.T.); EORTC Headquarters, Brussels, Belgium (T.G.); Pathology Department, Leiden University Medical Center, Leiden, Netherlands (S.v.D.)
| | - Johan M. Kros
- Department of Neurology, Erasmus MC, Rotterdam, Netherlands (M.J.v.d.B., Y.G., K.v.Z., J.P., P.A.S.S., P.J.F.); Department of Pathology, Erasmus MC, Rotterdam, Netherlands (J.M.K.); Department of Neurology, Haaglanden MC, The Hague, Netherlands (M.K., M.T.); EORTC Headquarters, Brussels, Belgium (T.G.); Pathology Department, Leiden University Medical Center, Leiden, Netherlands (S.v.D.)
| | - Thierry Gorlia
- Department of Neurology, Erasmus MC, Rotterdam, Netherlands (M.J.v.d.B., Y.G., K.v.Z., J.P., P.A.S.S., P.J.F.); Department of Pathology, Erasmus MC, Rotterdam, Netherlands (J.M.K.); Department of Neurology, Haaglanden MC, The Hague, Netherlands (M.K., M.T.); EORTC Headquarters, Brussels, Belgium (T.G.); Pathology Department, Leiden University Medical Center, Leiden, Netherlands (S.v.D.)
| | - Kitty van Zwieten
- Department of Neurology, Erasmus MC, Rotterdam, Netherlands (M.J.v.d.B., Y.G., K.v.Z., J.P., P.A.S.S., P.J.F.); Department of Pathology, Erasmus MC, Rotterdam, Netherlands (J.M.K.); Department of Neurology, Haaglanden MC, The Hague, Netherlands (M.K., M.T.); EORTC Headquarters, Brussels, Belgium (T.G.); Pathology Department, Leiden University Medical Center, Leiden, Netherlands (S.v.D.)
| | - Jory Prince
- Department of Neurology, Erasmus MC, Rotterdam, Netherlands (M.J.v.d.B., Y.G., K.v.Z., J.P., P.A.S.S., P.J.F.); Department of Pathology, Erasmus MC, Rotterdam, Netherlands (J.M.K.); Department of Neurology, Haaglanden MC, The Hague, Netherlands (M.K., M.T.); EORTC Headquarters, Brussels, Belgium (T.G.); Pathology Department, Leiden University Medical Center, Leiden, Netherlands (S.v.D.)
| | - Sjoerd van Duinen
- Department of Neurology, Erasmus MC, Rotterdam, Netherlands (M.J.v.d.B., Y.G., K.v.Z., J.P., P.A.S.S., P.J.F.); Department of Pathology, Erasmus MC, Rotterdam, Netherlands (J.M.K.); Department of Neurology, Haaglanden MC, The Hague, Netherlands (M.K., M.T.); EORTC Headquarters, Brussels, Belgium (T.G.); Pathology Department, Leiden University Medical Center, Leiden, Netherlands (S.v.D.)
| | - Peter A. Sillevis Smitt
- Department of Neurology, Erasmus MC, Rotterdam, Netherlands (M.J.v.d.B., Y.G., K.v.Z., J.P., P.A.S.S., P.J.F.); Department of Pathology, Erasmus MC, Rotterdam, Netherlands (J.M.K.); Department of Neurology, Haaglanden MC, The Hague, Netherlands (M.K., M.T.); EORTC Headquarters, Brussels, Belgium (T.G.); Pathology Department, Leiden University Medical Center, Leiden, Netherlands (S.v.D.)
| | - Martin Taphoorn
- Department of Neurology, Erasmus MC, Rotterdam, Netherlands (M.J.v.d.B., Y.G., K.v.Z., J.P., P.A.S.S., P.J.F.); Department of Pathology, Erasmus MC, Rotterdam, Netherlands (J.M.K.); Department of Neurology, Haaglanden MC, The Hague, Netherlands (M.K., M.T.); EORTC Headquarters, Brussels, Belgium (T.G.); Pathology Department, Leiden University Medical Center, Leiden, Netherlands (S.v.D.)
| | - Pim J. French
- Department of Neurology, Erasmus MC, Rotterdam, Netherlands (M.J.v.d.B., Y.G., K.v.Z., J.P., P.A.S.S., P.J.F.); Department of Pathology, Erasmus MC, Rotterdam, Netherlands (J.M.K.); Department of Neurology, Haaglanden MC, The Hague, Netherlands (M.K., M.T.); EORTC Headquarters, Brussels, Belgium (T.G.); Pathology Department, Leiden University Medical Center, Leiden, Netherlands (S.v.D.)
| |
Collapse
|
948
|
Swanton C, McGranahan N, Starrett GJ, Harris RS. APOBEC Enzymes: Mutagenic Fuel for Cancer Evolution and Heterogeneity. Cancer Discov 2015; 5:704-12. [PMID: 26091828 PMCID: PMC4497973 DOI: 10.1158/2159-8290.cd-15-0344] [Citation(s) in RCA: 373] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 05/14/2015] [Indexed: 12/16/2022]
Abstract
UNLABELLED Deep sequencing technologies are revealing the complexities of cancer evolution, casting light on mutational processes fueling tumor adaptation, immune escape, and treatment resistance. Understanding mechanisms driving cancer diversity is a critical step toward developing strategies to attenuate tumor evolution and adaptation. One emerging mechanism fueling tumor diversity and subclonal evolution is genomic DNA cytosine deamination catalyzed by APOBEC3B and at least one other APOBEC family member. Deregulation of APOBEC3 enzymes causes a general mutator phenotype that manifests as diverse and heterogeneous tumor subclones. Here, we summarize knowledge of the APOBEC DNA deaminase family in cancer, and their role as driving forces for intratumor heterogeneity and a therapeutic target to limit tumor adaptation. SIGNIFICANCE APOBEC mutational signatures may be enriched in tumor subclones, suggesting APOBEC cytosine deaminases fuel subclonal expansions and intratumor heterogeneity. APOBEC family members might represent a new class of drug target aimed at limiting tumor evolution, adaptation, and drug resistance.
Collapse
Affiliation(s)
- Charles Swanton
- The Francis Crick Institute, London, United Kingdom. UCL Cancer Institute, CRUK Lung Cancer Centre of Excellence, London, United Kingdom.
| | - Nicholas McGranahan
- The Francis Crick Institute, London, United Kingdom. Centre for Mathematics & Physics in the Life Sciences & Experimental Biology (CoMPLEX), University College London, London, United Kingdom
| | - Gabriel J Starrett
- Department of Biochemistry, Molecular Biology and Biophysics, Institute for Molecular Virology, and Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Reuben S Harris
- Department of Biochemistry, Molecular Biology and Biophysics, Institute for Molecular Virology, and Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
949
|
Kaloshi G, Roci E, Rroji A, Ducray F, Petrela M. Kinetic evaluation of low-grade gliomas in adults before and after treatment with CCNU alone. J Neurosurg 2015; 123:1244-6. [PMID: 26115464 DOI: 10.3171/2014.12.jns141068] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT The aim of this study was to evaluate the impact of CCNU chemotherapy alone on low-grade glioma (LGG) growth dynamics. METHODS The authors measured the evolution of the mean tumor diameter (MTD) in adult patients with LGG before (n=28 patients) and after (n=38 patients) CCNU administration. RESULTS Natural (spontaneous) growth of LGG in the present study was 4.3 mm/year (range 2.1-6.6 mm/year). The median MTD decrease after CCNU was 5.1 mm/year (range 1-8.9 mm/year). MTD decrease was noted in 30 patients (late decrease in 4 patients, and ongoing decrease in 24 patients with oligodendroglial tumors and 2 with astrocytic tumors). The median duration it took for the MTD to decrease after initiation of CCNU treatment was 619 days (1038 days for oligodendroglial tumors vs 377 days for astrocytic tumors; p=0.003). CONCLUSIONS These results show that CCNU as a single agent has a significant impact on LGG tumor growth. The impact of CCNU seems to be comparable to the previously reported impact of temozolomide therapy and of combined procarbazine, CCNU, and vincristine chemotherapy.
Collapse
Affiliation(s)
- Gentian Kaloshi
- Department of Neurosurgery, University Hospital Center "Mother Theresa," Tirana, Albania; and
| | - Ermir Roci
- Department of Neurosurgery, University Hospital Center "Mother Theresa," Tirana, Albania; and
| | - Arben Rroji
- Department of Neurosurgery, University Hospital Center "Mother Theresa," Tirana, Albania; and
| | - Francois Ducray
- Department of Neuro-oncology, Hospices Civils de Lyon, Hôpital Neurologique, Lyon, France
| | - Mentor Petrela
- Department of Neurosurgery, University Hospital Center "Mother Theresa," Tirana, Albania; and
| |
Collapse
|
950
|
Brat DJ, Verhaak RGW, Aldape KD, Yung WKA, Salama SR, Cooper LAD, Rheinbay E, Miller CR, Vitucci M, Morozova O, Robertson AG, Noushmehr H, Laird PW, Cherniack AD, Akbani R, Huse JT, Ciriello G, Poisson LM, Barnholtz-Sloan JS, Berger MS, Brennan C, Colen RR, Colman H, Flanders AE, Giannini C, Grifford M, Iavarone A, Jain R, Joseph I, Kim J, Kasaian K, Mikkelsen T, Murray BA, O'Neill BP, Pachter L, Parsons DW, Sougnez C, Sulman EP, Vandenberg SR, Van Meir EG, von Deimling A, Zhang H, Crain D, Lau K, Mallery D, Morris S, Paulauskis J, Penny R, Shelton T, Sherman M, Yena P, Black A, Bowen J, Dicostanzo K, Gastier-Foster J, Leraas KM, Lichtenberg TM, Pierson CR, Ramirez NC, Taylor C, Weaver S, Wise L, Zmuda E, Davidsen T, Demchok JA, Eley G, Ferguson ML, Hutter CM, Mills Shaw KR, Ozenberger BA, Sheth M, Sofia HJ, Tarnuzzer R, Wang Z, Yang L, Zenklusen JC, Ayala B, Baboud J, Chudamani S, Jensen MA, Liu J, Pihl T, Raman R, Wan Y, Wu Y, Ally A, Auman JT, Balasundaram M, Balu S, Baylin SB, Beroukhim R, Bootwalla MS, Bowlby R, Bristow CA, Brooks D, Butterfield Y, Carlsen R, Carter S, Chin L, Chu A, et alBrat DJ, Verhaak RGW, Aldape KD, Yung WKA, Salama SR, Cooper LAD, Rheinbay E, Miller CR, Vitucci M, Morozova O, Robertson AG, Noushmehr H, Laird PW, Cherniack AD, Akbani R, Huse JT, Ciriello G, Poisson LM, Barnholtz-Sloan JS, Berger MS, Brennan C, Colen RR, Colman H, Flanders AE, Giannini C, Grifford M, Iavarone A, Jain R, Joseph I, Kim J, Kasaian K, Mikkelsen T, Murray BA, O'Neill BP, Pachter L, Parsons DW, Sougnez C, Sulman EP, Vandenberg SR, Van Meir EG, von Deimling A, Zhang H, Crain D, Lau K, Mallery D, Morris S, Paulauskis J, Penny R, Shelton T, Sherman M, Yena P, Black A, Bowen J, Dicostanzo K, Gastier-Foster J, Leraas KM, Lichtenberg TM, Pierson CR, Ramirez NC, Taylor C, Weaver S, Wise L, Zmuda E, Davidsen T, Demchok JA, Eley G, Ferguson ML, Hutter CM, Mills Shaw KR, Ozenberger BA, Sheth M, Sofia HJ, Tarnuzzer R, Wang Z, Yang L, Zenklusen JC, Ayala B, Baboud J, Chudamani S, Jensen MA, Liu J, Pihl T, Raman R, Wan Y, Wu Y, Ally A, Auman JT, Balasundaram M, Balu S, Baylin SB, Beroukhim R, Bootwalla MS, Bowlby R, Bristow CA, Brooks D, Butterfield Y, Carlsen R, Carter S, Chin L, Chu A, Chuah E, Cibulskis K, Clarke A, Coetzee SG, Dhalla N, Fennell T, Fisher S, Gabriel S, Getz G, Gibbs R, Guin R, Hadjipanayis A, Hayes DN, Hinoue T, Hoadley K, Holt RA, Hoyle AP, Jefferys SR, Jones S, Jones CD, Kucherlapati R, Lai PH, Lander E, Lee S, Lichtenstein L, Ma Y, Maglinte DT, Mahadeshwar HS, Marra MA, Mayo M, Meng S, Meyerson ML, Mieczkowski PA, Moore RA, Mose LE, Mungall AJ, Pantazi A, Parfenov M, Park PJ, Parker JS, Perou CM, Protopopov A, Ren X, Roach J, Sabedot TS, Schein J, Schumacher SE, Seidman JG, Seth S, Shen H, Simons JV, Sipahimalani P, Soloway MG, Song X, Sun H, Tabak B, Tam A, Tan D, Tang J, Thiessen N, Triche T, Van Den Berg DJ, Veluvolu U, Waring S, Weisenberger DJ, Wilkerson MD, Wong T, Wu J, Xi L, Xu AW, Yang L, Zack TI, Zhang J, Aksoy BA, Arachchi H, Benz C, Bernard B, Carlin D, Cho J, DiCara D, Frazer S, Fuller GN, Gao J, Gehlenborg N, Haussler D, Heiman DI, Iype L, Jacobsen A, Ju Z, Katzman S, Kim H, Knijnenburg T, Kreisberg RB, Lawrence MS, Lee W, Leinonen K, Lin P, Ling S, Liu W, Liu Y, Liu Y, Lu Y, Mills G, Ng S, Noble MS, Paull E, Rao A, Reynolds S, Saksena G, Sanborn Z, Sander C, Schultz N, Senbabaoglu Y, Shen R, Shmulevich I, Sinha R, Stuart J, Sumer SO, Sun Y, Tasman N, Taylor BS, Voet D, Weinhold N, Weinstein JN, Yang D, Yoshihara K, Zheng S, Zhang W, Zou L, Abel T, Sadeghi S, Cohen ML, Eschbacher J, Hattab EM, Raghunathan A, Schniederjan MJ, Aziz D, Barnett G, Barrett W, Bigner DD, Boice L, Brewer C, Calatozzolo C, Campos B, Carlotti CG, Chan TA, Cuppini L, Curley E, Cuzzubbo S, Devine K, DiMeco F, Duell R, Elder JB, Fehrenbach A, Finocchiaro G, Friedman W, Fulop J, Gardner J, Hermes B, Herold-Mende C, Jungk C, Kendler A, Lehman NL, Lipp E, Liu O, Mandt R, McGraw M, Mclendon R, McPherson C, Neder L, Nguyen P, Noss A, Nunziata R, Ostrom QT, Palmer C, Perin A, Pollo B, Potapov A, Potapova O, Rathmell WK, Rotin D, Scarpace L, Schilero C, Senecal K, Shimmel K, Shurkhay V, Sifri S, Singh R, Sloan AE, Smolenski K, Staugaitis SM, Steele R, Thorne L, Tirapelli DPC, Unterberg A, Vallurupalli M, Wang Y, Warnick R, Williams F, Wolinsky Y, Bell S, Rosenberg M, Stewart C, Huang F, Grimsby JL, Radenbaugh AJ, Zhang J. Comprehensive, Integrative Genomic Analysis of Diffuse Lower-Grade Gliomas. N Engl J Med 2015; 372:2481-98. [PMID: 26061751 PMCID: PMC4530011 DOI: 10.1056/nejmoa1402121] [Show More Authors] [Citation(s) in RCA: 2319] [Impact Index Per Article: 231.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Diffuse low-grade and intermediate-grade gliomas (which together make up the lower-grade gliomas, World Health Organization grades II and III) have highly variable clinical behavior that is not adequately predicted on the basis of histologic class. Some are indolent; others quickly progress to glioblastoma. The uncertainty is compounded by interobserver variability in histologic diagnosis. Mutations in IDH, TP53, and ATRX and codeletion of chromosome arms 1p and 19q (1p/19q codeletion) have been implicated as clinically relevant markers of lower-grade gliomas. METHODS We performed genomewide analyses of 293 lower-grade gliomas from adults, incorporating exome sequence, DNA copy number, DNA methylation, messenger RNA expression, microRNA expression, and targeted protein expression. These data were integrated and tested for correlation with clinical outcomes. RESULTS Unsupervised clustering of mutations and data from RNA, DNA-copy-number, and DNA-methylation platforms uncovered concordant classification of three robust, nonoverlapping, prognostically significant subtypes of lower-grade glioma that were captured more accurately by IDH, 1p/19q, and TP53 status than by histologic class. Patients who had lower-grade gliomas with an IDH mutation and 1p/19q codeletion had the most favorable clinical outcomes. Their gliomas harbored mutations in CIC, FUBP1, NOTCH1, and the TERT promoter. Nearly all lower-grade gliomas with IDH mutations and no 1p/19q codeletion had mutations in TP53 (94%) and ATRX inactivation (86%). The large majority of lower-grade gliomas without an IDH mutation had genomic aberrations and clinical behavior strikingly similar to those found in primary glioblastoma. CONCLUSIONS The integration of genomewide data from multiple platforms delineated three molecular classes of lower-grade gliomas that were more concordant with IDH, 1p/19q, and TP53 status than with histologic class. Lower-grade gliomas with an IDH mutation either had 1p/19q codeletion or carried a TP53 mutation. Most lower-grade gliomas without an IDH mutation were molecularly and clinically similar to glioblastoma. (Funded by the National Institutes of Health.).
Collapse
|