101
|
Beadling C, Wald AI, Warrick A, Neff TL, Zhong S, Nikiforov YE, Corless CL, Nikiforova MN. A Multiplexed Amplicon Approach for Detecting Gene Fusions by Next-Generation Sequencing. J Mol Diagn 2016; 18:165-75. [DOI: 10.1016/j.jmoldx.2015.10.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 10/25/2015] [Accepted: 10/30/2015] [Indexed: 01/05/2023] Open
|
102
|
Sancho-Martinez I, Nivet E, Xia Y, Hishida T, Aguirre A, Ocampo A, Ma L, Morey R, Krause MN, Zembrzycki A, Ansorge O, Vazquez-Ferrer E, Dubova I, Reddy P, Lam D, Hishida Y, Wu MZ, Esteban CR, O'Leary D, Wahl GM, Verma IM, Laurent LC, Izpisua Belmonte JC. Establishment of human iPSC-based models for the study and targeting of glioma initiating cells. Nat Commun 2016; 7:10743. [PMID: 26899176 PMCID: PMC4764898 DOI: 10.1038/ncomms10743] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/13/2016] [Indexed: 01/06/2023] Open
Abstract
Glioma tumour-initiating cells (GTICs) can originate upon the transformation of neural progenitor cells (NPCs). Studies on GTICs have focused on primary tumours from which GTICs could be isolated and the use of human embryonic material. Recently, the somatic genomic landscape of human gliomas has been reported. RTK (receptor tyrosine kinase) and p53 signalling were found dysregulated in ∼90% and 86% of all primary tumours analysed, respectively. Here we report on the use of human-induced pluripotent stem cells (hiPSCs) for modelling gliomagenesis. Dysregulation of RTK and p53 signalling in hiPSC-derived NPCs (iNPCs) recapitulates GTIC properties in vitro. In vivo transplantation of transformed iNPCs leads to highly aggressive tumours containing undifferentiated stem cells and their differentiated derivatives. Metabolic modulation compromises GTIC viability. Last, screening of 101 anti-cancer compounds identifies three molecules specifically targeting transformed iNPCs and primary GTICs. Together, our results highlight the potential of hiPSCs for studying human tumourigenesis. Glioma can originate from the transformation of neural progenitor cells into glioma initiating cells. Here, the authors demonstrate the use of induced pluripotent stem cells as a suitable model for generating neural progenitor cells, which can be subsequently transformed to glioma initiating cells that are able to the generate human glioma-like tumours in mice.
Collapse
Affiliation(s)
- Ignacio Sancho-Martinez
- Gene Expression Laboratory Belmonte, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Emmanuel Nivet
- Gene Expression Laboratory Belmonte, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Yun Xia
- Gene Expression Laboratory Belmonte, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Tomoaki Hishida
- Gene Expression Laboratory Belmonte, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Aitor Aguirre
- Gene Expression Laboratory Belmonte, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Alejandro Ocampo
- Gene Expression Laboratory Belmonte, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Li Ma
- Gene Expression Laboratory Belmonte, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA.,Universidad Católica San Antonio de Murcia (UCAM) Campus de los Jerónimos, N° 135 Guadalupe, Murcia 30107, Spain
| | - Robert Morey
- Department of Reproductive Medicine, University of California, San Diego, Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, California 92037, USA
| | - Marie N Krause
- Gene Expression Laboratory Belmonte, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Andreas Zembrzycki
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Olaf Ansorge
- Department of Neuropathology, West Wing, Level 1, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Eric Vazquez-Ferrer
- Gene Expression Laboratory Belmonte, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Ilir Dubova
- Gene Expression Laboratory Belmonte, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA.,Universidad Católica San Antonio de Murcia (UCAM) Campus de los Jerónimos, N° 135 Guadalupe, Murcia 30107, Spain
| | - Pradeep Reddy
- Gene Expression Laboratory Belmonte, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| | - David Lam
- Gene Expression Laboratory Belmonte, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Yuriko Hishida
- Gene Expression Laboratory Belmonte, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Min-Zu Wu
- Gene Expression Laboratory Belmonte, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Concepcion Rodriguez Esteban
- Gene Expression Laboratory Belmonte, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Dennis O'Leary
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Geoffrey M Wahl
- Gene Expression Laboratory Wahl, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Inder M Verma
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Louise C Laurent
- Department of Reproductive Medicine, University of California, San Diego, Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, California 92037, USA
| | - Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory Belmonte, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| |
Collapse
|
103
|
Kanagal-Shamanna R, Singh RR, Routbort MJ, Patel KP, Medeiros LJ, Luthra R. Principles of analytical validation of next-generation sequencing based mutational analysis for hematologic neoplasms in a CLIA-certified laboratory. Expert Rev Mol Diagn 2016; 16:461-72. [PMID: 26765348 DOI: 10.1586/14737159.2016.1142374] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Targeted therapy based on mutational profiles is the current standard of practice for the management of patients with hematologic malignancies. Next-generation sequencing (NGS)- based analysis has been adopted by clinical laboratories for high-throughput mutational profiling of myeloid and lymphoid neoplasms. The technology is fairly novel and complex, hence both validation and test implementation in a CLIA-certified laboratory differ substantially from traditional sequencing platforms. Recently, organizations such as the American College of Medical Genetics, Centers for Disease Control and Prevention and College of American Pathologists have published principles and guidelines for NGS test development to ensure standardization of testing across institutions. Summarized here are the recommendations from these organizations as they pertain to targeted NGS-based testing of hematologic malignancies ('liquid tumors'), with particular emphasis on myeloid neoplasms.
Collapse
Affiliation(s)
- Rashmi Kanagal-Shamanna
- a Department of Hematopathology , The University of Texas at M.D. Anderson Cancer Center , Houston , TX , USA
| | - Rajesh R Singh
- a Department of Hematopathology , The University of Texas at M.D. Anderson Cancer Center , Houston , TX , USA
| | - Mark J Routbort
- a Department of Hematopathology , The University of Texas at M.D. Anderson Cancer Center , Houston , TX , USA
| | - Keyur P Patel
- a Department of Hematopathology , The University of Texas at M.D. Anderson Cancer Center , Houston , TX , USA
| | - L Jeffrey Medeiros
- a Department of Hematopathology , The University of Texas at M.D. Anderson Cancer Center , Houston , TX , USA
| | - Rajyalakshmi Luthra
- a Department of Hematopathology , The University of Texas at M.D. Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
104
|
Yamagata K, Yamanishi A, Kokubu C, Takeda J, Sese J. COSMOS: accurate detection of somatic structural variations through asymmetric comparison between tumor and normal samples. Nucleic Acids Res 2016; 44:e78. [PMID: 26833260 PMCID: PMC4856976 DOI: 10.1093/nar/gkw026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 01/11/2016] [Indexed: 11/13/2022] Open
Abstract
An important challenge in cancer genomics is precise detection of structural variations (SVs) by high-throughput short-read sequencing, which is hampered by the high false discovery rates of existing analysis tools. Here, we propose an accurate SV detection method named COSMOS, which compares the statistics of the mapped read pairs in tumor samples with isogenic normal control samples in a distinct asymmetric manner. COSMOS also prioritizes the candidate SVs using strand-specific read-depth information. Performance tests on modeled tumor genomes revealed that COSMOS outperformed existing methods in terms of F-measure. We also applied COSMOS to an experimental mouse cell-based model, in which SVs were induced by genome engineering and gamma-ray irradiation, followed by polymerase chain reaction-based confirmation. The precision of COSMOS was 84.5%, while the next best existing method was 70.4%. Moreover, the sensitivity of COSMOS was the highest, indicating that COSMOS has great potential for cancer genome analysis.
Collapse
Affiliation(s)
- Koichi Yamagata
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, 135-0064, Japan
| | - Ayako Yamanishi
- Department of Genome Biology, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Chikara Kokubu
- Department of Genome Biology, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Junji Takeda
- Department of Genome Biology, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Jun Sese
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, 135-0064, Japan Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, 135-0064, Japan
| |
Collapse
|
105
|
Ren G, Krawetz R. Applying computation biology and "big data" to develop multiplex diagnostics for complex chronic diseases such as osteoarthritis. Biomarkers 2016; 20:533-9. [PMID: 26809774 PMCID: PMC4819822 DOI: 10.3109/1354750x.2015.1105499] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The data explosion in the last decade is revolutionizing diagnostics research and the healthcare industry, offering both opportunities and challenges. These high-throughput “omics” techniques have generated more scientific data in the last few years than in the entire history of mankind. Here we present a brief summary of how “big data” have influenced early diagnosis of complex diseases. We will also review some of the most commonly used “omics” techniques and their applications in diagnostics. Finally, we will discuss the issues brought by these new techniques when translating laboratory discoveries to clinical practice.
Collapse
Affiliation(s)
- Guomin Ren
- a McCaig Institute for Bone & Joint Health, University of Calgary , Calgary , AB , Canada
| | - Roman Krawetz
- a McCaig Institute for Bone & Joint Health, University of Calgary , Calgary , AB , Canada .,b Department of Surgery , University of Calgary , Calgary , AB , Canada , and.,c Department of Anatomy and Cell Biology , University of Calgary , Calgary , AB , Canada
| |
Collapse
|
106
|
De Maeyer D, Weytjens B, De Raedt L, Marchal K. Network-Based Analysis of eQTL Data to Prioritize Driver Mutations. Genome Biol Evol 2016; 8:481-94. [PMID: 26802430 PMCID: PMC4825419 DOI: 10.1093/gbe/evw010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
In clonal systems, interpreting driver genes in terms of molecular networks helps understanding how these drivers elicit an adaptive phenotype. Obtaining such a network-based understanding depends on the correct identification of driver genes. In clonal systems, independent evolved lines can acquire a similar adaptive phenotype by affecting the same molecular pathways, a phenomenon referred to as parallelism at the molecular pathway level. This implies that successful driver identification depends on interpreting mutated genes in terms of molecular networks. Driver identification and obtaining a network-based understanding of the adaptive phenotype are thus confounded problems that ideally should be solved simultaneously. In this study, a network-based eQTL method is presented that solves both the driver identification and the network-based interpretation problem. As input the method uses coupled genotype-expression phenotype data (eQTL data) of independently evolved lines with similar adaptive phenotypes and an organism-specific genome-wide interaction network. The search for mutational consistency at pathway level is defined as a subnetwork inference problem, which consists of inferring a subnetwork from the genome-wide interaction network that best connects the genes containing mutations to differentially expressed genes. Based on their connectivity with the differentially expressed genes, mutated genes are prioritized as driver genes. Based on semisynthetic data and two publicly available data sets, we illustrate the potential of the network-based eQTL method to prioritize driver genes and to gain insights in the molecular mechanisms underlying an adaptive phenotype. The method is available at http://bioinformatics.intec.ugent.be/phenetic_eqtl/index.html
Collapse
Affiliation(s)
- Dries De Maeyer
- Deptartment of Information Technology (INTEC, iMINDS), UGent, 9052 Ghent, Belgium Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark 927, 9052 Gent, Belgium Bioinformatics Institute Ghent, Technologiepark 927, 9052 Ghent, Belgium Department of Microbial and Molecular Systems, KU Leuven, Kasteelpark Arenberg 20, B-3001 Leuven, Belgium
| | - Bram Weytjens
- Deptartment of Information Technology (INTEC, iMINDS), UGent, 9052 Ghent, Belgium Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark 927, 9052 Gent, Belgium Bioinformatics Institute Ghent, Technologiepark 927, 9052 Ghent, Belgium Department of Microbial and Molecular Systems, KU Leuven, Kasteelpark Arenberg 20, B-3001 Leuven, Belgium
| | - Luc De Raedt
- Department of Computer Science, KU Leuven, Celestijnenlaan 200A, B-3001 Leuven, Belgium
| | - Kathleen Marchal
- Deptartment of Information Technology (INTEC, iMINDS), UGent, 9052 Ghent, Belgium Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark 927, 9052 Gent, Belgium Bioinformatics Institute Ghent, Technologiepark 927, 9052 Ghent, Belgium Department of Genetics, University of Pretoria, Hatfield Campus, Pretoria 0028, South Africa Department of Microbial and Molecular Systems, KU Leuven, Kasteelpark Arenberg 20, B-3001 Leuven, Belgium
| |
Collapse
|
107
|
Abstract
Despite the enormous medical impact of cancers and intensive study of their biology, detailed characterization of tumor growth and development remains elusive. This difficulty occurs in large part because of enormous heterogeneity in the molecular mechanisms of cancer progression, both tumor-to-tumor and cell-to-cell in single tumors. Advances in genomic technologies, especially at the single-cell level, are improving the situation, but these approaches are held back by limitations of the biotechnologies for gathering genomic data from heterogeneous cell populations and the computational methods for making sense of those data. One popular way to gain the advantages of whole-genome methods without the cost of single-cell genomics has been the use of computational deconvolution (unmixing) methods to reconstruct clonal heterogeneity from bulk genomic data. These methods, too, are limited by the difficulty of inferring genomic profiles of rare or subtly varying clonal subpopulations from bulk data, a problem that can be computationally reduced to that of reconstructing the geometry of point clouds of tumor samples in a genome space. Here, we present a new method to improve that reconstruction by better identifying subspaces corresponding to tumors produced from mixtures of distinct combinations of clonal subpopulations. We develop a nonparametric clustering method based on medoidshift clustering for identifying subgroups of tumors expected to correspond to distinct trajectories of evolutionary progression. We show on synthetic and real tumor copy-number data that this new method substantially improves our ability to resolve discrete tumor subgroups, a key step in the process of accurately deconvolving tumor genomic data and inferring clonal heterogeneity from bulk data.
Collapse
Affiliation(s)
- Theodore Roman
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, 5000 Forbes Ave, Pittsburgh, 15213, PA, USA. .,Joint Carnegie Mellon/University of Pittsburgh Ph.D. Program in Computational Biology, 5000 Forbes Ave, Pittsburgh, 15213, PA, USA.
| | - Lu Xie
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, 5000 Forbes Ave, Pittsburgh, 15213, PA, USA. .,Joint Carnegie Mellon/University of Pittsburgh Ph.D. Program in Computational Biology, 5000 Forbes Ave, Pittsburgh, 15213, PA, USA.
| | - Russell Schwartz
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, 5000 Forbes Ave, Pittsburgh, 15213, PA, USA. .,Department of Biological Sciences, Mellon College of Science, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, 15213, PA, USA.
| |
Collapse
|
108
|
Abstract
Understanding the molecular landscape of cancer has facilitated the development of diagnostic, prognostic, and predictive biomarkers for clinical oncology. Developments in next-generation DNA sequencing technologies have increased the speed and reduced the cost of sequencing the nucleic acids of cancer cells. This has unlocked opportunities to characterize the genomic and transcriptomic landscapes of cancer for basic science research through projects like The Cancer Genome Atlas. The cancer genome includes DNA-based alterations, such as point mutations or gene duplications. The cancer transcriptome involves RNA-based alterations, including changes in messenger RNAs. Together, the genome and transcriptome can provide a comprehensive view of an individual patient's cancer that is beginning to impact real-time clinical decision-making. The authors discuss several opportunities for translating this basic science knowledge into clinical practice, including a molecular classification of cancer, heritable risk of cancer, eligibility for targeted therapies, and the development of innovative, genomic-based clinical trials. In this review, key applications and new directions are outlined for translating the cancer genome and transcriptome into patient care in the clinic.
Collapse
Affiliation(s)
- Sameek Roychowdhury
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210 USA
- Department of Pharmacology, The Ohio State University, Columbus, Ohio 43210 USA
| | - Arul M. Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Howard Hughes Medical Institute, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI 48109 USA
- Center for Computational Medicine and Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
109
|
Araya CL, Cenik C, Reuter JA, Kiss G, Pande VS, Snyder MP, Greenleaf WJ. Identification of significantly mutated regions across cancer types highlights a rich landscape of functional molecular alterations. Nat Genet 2015; 48:117-25. [PMID: 26691984 PMCID: PMC4731297 DOI: 10.1038/ng.3471] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 11/20/2015] [Indexed: 02/07/2023]
Abstract
Cancer sequencing studies have primarily identified cancer-driver genes by the accumulation of protein-altering mutations. An improved method would be annotation-independent, sensitive to unknown distributions of functions within proteins, and inclusive of non-coding drivers. We employed density-based clustering methods in 21 tumor types to detect variably-sized significantly mutated regions (SMRs). SMRs reveal recurrent alterations across a spectrum of coding and non-coding elements, including transcription factor binding sites and untranslated regions mutated in up to ∼15% of specific tumor types. SMRs reveal spatial clustering of mutations at molecular domains and interfaces, often with associated changes in signaling. Mutation frequencies in SMRs demonstrate that distinct protein regions are differentially mutated among tumor types, as exemplified by a linker region of PIK3CA in which biophysical simulations suggest mutations affect regulatory interactions. The functional diversity of SMRs underscores both the varied mechanisms of oncogenic misregulation and the advantage of functionally-agnostic driver identification.
Collapse
Affiliation(s)
- Carlos L Araya
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Can Cenik
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Jason A Reuter
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Gert Kiss
- Department of Chemistry, Stanford University, Stanford, California, USA
| | - Vijay S Pande
- Department of Chemistry, Stanford University, Stanford, California, USA
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - William J Greenleaf
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA.,Department of Applied Physics, Stanford University, Stanford, California, USA
| |
Collapse
|
110
|
Vicini P, Fields O, Lai E, Litwack ED, Martin AM, Morgan TM, Pacanowski MA, Papaluca M, Perez OD, Ringel MS, Robson M, Sakul H, Vockley J, Zaks T, Dolsten M, Søgaard M. Precision medicine in the age of big data: The present and future role of large-scale unbiased sequencing in drug discovery and development. Clin Pharmacol Ther 2015; 99:198-207. [PMID: 26536838 DOI: 10.1002/cpt.293] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 10/30/2015] [Indexed: 12/15/2022]
Abstract
High throughput molecular and functional profiling of patients is a key driver of precision medicine. DNA and RNA characterization has been enabled at unprecedented cost and scale through rapid, disruptive progress in sequencing technology, but challenges persist in data management and interpretation. We analyze the state-of-the-art of large-scale unbiased sequencing in drug discovery and development, including technology, application, ethical, regulatory, policy and commercial considerations, and discuss issues of LUS implementation in clinical and regulatory practice.
Collapse
Affiliation(s)
- P Vicini
- Pfizer Worldwide Research & Development, La Jolla, California, Collegeville, Pennsylvania, and New York, New York, USA
| | - O Fields
- Pfizer Worldwide Research & Development, La Jolla, California, Collegeville, Pennsylvania, and New York, New York, USA
| | - E Lai
- Takeda Pharmaceuticals International, Deerfield, Illinois, USA
| | - E D Litwack
- Food and Drug Administration, Silver Spring, Maryland, USA
| | - A-M Martin
- GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - T M Morgan
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, and East Hanover, New Jersey, USA
| | - M A Pacanowski
- Food and Drug Administration, Silver Spring, Maryland, USA
| | | | - O D Perez
- Pfizer Worldwide Research & Development, La Jolla, California, Collegeville, Pennsylvania, and New York, New York, USA
| | - M S Ringel
- Boston Consulting Group, Boston, Massachusetts, USA
| | - M Robson
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, and East Hanover, New Jersey, USA
| | - H Sakul
- Pfizer Worldwide Research & Development, La Jolla, California, Collegeville, Pennsylvania, and New York, New York, USA
| | - J Vockley
- Inova Translational Medicine Institute, Falls Church, Virginia, USA
| | - T Zaks
- Sanofi, Cambridge, Massachusetts, USA
| | - M Dolsten
- Pfizer Worldwide Research & Development, La Jolla, California, Collegeville, Pennsylvania, and New York, New York, USA
| | - M Søgaard
- Pfizer Worldwide Research & Development, La Jolla, California, Collegeville, Pennsylvania, and New York, New York, USA
| |
Collapse
|
111
|
Yang R, Nelson AC, Henzler C, Thyagarajan B, Silverstein KAT. ScanIndel: a hybrid framework for indel detection via gapped alignment, split reads and de novo assembly. Genome Med 2015; 7:127. [PMID: 26643039 PMCID: PMC4671222 DOI: 10.1186/s13073-015-0251-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/18/2015] [Indexed: 12/30/2022] Open
Abstract
Comprehensive identification of insertions/deletions (indels) across the full size spectrum from second generation sequencing is challenging due to the relatively short read length inherent in the technology. Different indel calling methods exist but are limited in detection to specific sizes with varying accuracy and resolution. We present ScanIndel, an integrated framework for detecting indels with multiple heuristics including gapped alignment, split reads and de novo assembly. Using simulation data, we demonstrate ScanIndel’s superior sensitivity and specificity relative to several state-of-the-art indel callers across various coverage levels and indel sizes. ScanIndel yields higher predictive accuracy with lower computational cost compared with existing tools for both targeted resequencing data from tumor specimens and high coverage whole-genome sequencing data from the human NIST standard NA12878. Thus, we anticipate ScanIndel will improve indel analysis in both clinical and research settings. ScanIndel is implemented in Python, and is freely available for academic use at https://github.com/cauyrd/ScanIndel.
Collapse
Affiliation(s)
- Rendong Yang
- Supercomputing Institute for Advanced Computational Research, University of Minnesota, 117 Pleasant St. SE, RM 541, Minneapolis, MN, 55455, USA.
| | - Andrew C Nelson
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Christine Henzler
- Supercomputing Institute for Advanced Computational Research, University of Minnesota, 117 Pleasant St. SE, RM 541, Minneapolis, MN, 55455, USA.
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Kevin A T Silverstein
- Supercomputing Institute for Advanced Computational Research, University of Minnesota, 117 Pleasant St. SE, RM 541, Minneapolis, MN, 55455, USA.
| |
Collapse
|
112
|
Abstract
Cancer drugs are broadly classified into two categories: cytotoxic chemotherapies and targeted therapies that specifically modulate the activity of one or more proteins involved in cancer. Major advances have been achieved in targeted cancer therapies in the past few decades, which is ascribed to the increasing understanding of molecular mechanisms for cancer initiation and progression. Consequently, monoclonal antibodies and small molecules have been developed to interfere with a specific molecular oncogenic target. Targeting gain-of-function mutations, in general, has been productive. However, it has been a major challenge to use standard pharmacologic approaches to target loss-of-function mutations of tumor suppressor genes. Novel approaches, including synthetic lethality and collateral vulnerability screens, are now being developed to target gene defects in p53, PTEN, and BRCA1/2. Here, we review and summarize the recent findings in cancer genomics, drug development, and molecular cancer biology, which show promise in targeting tumor suppressors in cancer therapeutics.
Collapse
Affiliation(s)
- Yunhua Liu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaoxiao Hu
- State Key Laboratory for Chemo/Bio Sensing and Chemometrics, College of Biology, Hunan University, Changsha, China
| | - Cecil Han
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Liana Wang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xinna Zhang
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaoming He
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | - Xiongbin Lu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
113
|
Verigos J, Magklara A. Revealing the Complexity of Breast Cancer by Next Generation Sequencing. Cancers (Basel) 2015; 7:2183-200. [PMID: 26561834 PMCID: PMC4695885 DOI: 10.3390/cancers7040885] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 10/18/2015] [Accepted: 10/26/2015] [Indexed: 02/06/2023] Open
Abstract
Over the last few years the increasing usage of "-omic" platforms, supported by next-generation sequencing, in the analysis of breast cancer samples has tremendously advanced our understanding of the disease. New driver and passenger mutations, rare chromosomal rearrangements and other genomic aberrations identified by whole genome and exome sequencing are providing missing pieces of the genomic architecture of breast cancer. High resolution maps of breast cancer methylomes and sequencing of the miRNA microworld are beginning to paint the epigenomic landscape of the disease. Transcriptomic profiling is giving us a glimpse into the gene regulatory networks that govern the fate of the breast cancer cell. At the same time, integrative analysis of sequencing data confirms an extensive intertumor and intratumor heterogeneity and plasticity in breast cancer arguing for a new approach to the problem. In this review, we report on the latest findings on the molecular characterization of breast cancer using NGS technologies, and we discuss their potential implications for the improvement of existing therapies.
Collapse
Affiliation(s)
- John Verigos
- Laboratory of Clinical Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina 45110, Greece.
- Department of Biomedical Research, Institute of Molecular Biology & Biotechnology,Foundation for Research & Technology-Hellas, Ioannina 45110, Greece.
| | - Angeliki Magklara
- Laboratory of Clinical Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina 45110, Greece.
- Department of Biomedical Research, Institute of Molecular Biology & Biotechnology,Foundation for Research & Technology-Hellas, Ioannina 45110, Greece.
| |
Collapse
|
114
|
Next-Generation Sequencing Approaches in Cancer: Where Have They Brought Us and Where Will They Take Us? Cancers (Basel) 2015; 7:1925-58. [PMID: 26404381 PMCID: PMC4586802 DOI: 10.3390/cancers7030869] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/15/2015] [Indexed: 12/20/2022] Open
Abstract
Next-generation sequencing (NGS) technologies and data have revolutionized cancer research and are increasingly being deployed to guide clinicians in treatment decision-making. NGS technologies have allowed us to take an “omics” approach to cancer in order to reveal genomic, transcriptomic, and epigenomic landscapes of individual malignancies. Integrative multi-platform analyses are increasingly used in large-scale projects that aim to fully characterize individual tumours as well as general cancer types and subtypes. In this review, we examine how NGS technologies in particular have contributed to “omics” approaches in cancer research, allowing for large-scale integrative analyses that consider hundreds of tumour samples. These types of studies have provided us with an unprecedented wealth of information, providing the background knowledge needed to make small-scale (including “N of 1”) studies informative and relevant. We also take a look at emerging opportunities provided by NGS and state-of-the-art third-generation sequencing technologies, particularly in the context of translational research. Cancer research and care are currently poised to experience significant progress catalyzed by accessible sequencing technologies that will benefit both clinical- and research-based efforts.
Collapse
|
115
|
Cheng F, Zhao J, Zhao Z. Advances in computational approaches for prioritizing driver mutations and significantly mutated genes in cancer genomes. Brief Bioinform 2015; 17:642-56. [PMID: 26307061 DOI: 10.1093/bib/bbv068] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Indexed: 12/27/2022] Open
Abstract
Cancer is often driven by the accumulation of genetic alterations, including single nucleotide variants, small insertions or deletions, gene fusions, copy-number variations, and large chromosomal rearrangements. Recent advances in next-generation sequencing technologies have helped investigators generate massive amounts of cancer genomic data and catalog somatic mutations in both common and rare cancer types. So far, the somatic mutation landscapes and signatures of >10 major cancer types have been reported; however, pinpointing driver mutations and cancer genes from millions of available cancer somatic mutations remains a monumental challenge. To tackle this important task, many methods and computational tools have been developed during the past several years and, thus, a review of its advances is urgently needed. Here, we first summarize the main features of these methods and tools for whole-exome, whole-genome and whole-transcriptome sequencing data. Then, we discuss major challenges like tumor intra-heterogeneity, tumor sample saturation and functionality of synonymous mutations in cancer, all of which may result in false-positive discoveries. Finally, we highlight new directions in studying regulatory roles of noncoding somatic mutations and quantitatively measuring circulating tumor DNA in cancer. This review may help investigators find an appropriate tool for detecting potential driver or actionable mutations in rapidly emerging precision cancer medicine.
Collapse
|
116
|
Niroula A, Vihinen M. Harmful somatic amino acid substitutions affect key pathways in cancers. BMC Med Genomics 2015; 8:53. [PMID: 26282678 PMCID: PMC4539680 DOI: 10.1186/s12920-015-0125-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 07/30/2015] [Indexed: 12/14/2022] Open
Abstract
Background Cancer is characterized by the accumulation of large numbers of genetic variations and alterations of multiple biological phenomena. Cancer genomics has largely focused on the identification of such genetic alterations and the genes containing them, known as ‘cancer genes’. However, the non-functional somatic variations out-number functional variations and remain as a major challenge. Recurrent somatic variations are thought to be cancer drivers but they are present in only a small fraction of patients. Methods We performed an extensive analysis of amino acid substitutions (AASs) from 6,861 cancer samples (whole genome or exome sequences) classified into 30 cancer types and performed pathway enrichment analysis. We also studied the overlap between the cancers based on proteins containing harmful AASs and pathways affected by them. Results We found that only a fraction of AASs (39.88 %) are harmful even in known cancer genes. In addition, we found that proteins containing harmful AASs in cancers are often centrally located in protein interaction networks. Based on the proteins containing harmful AASs, we identified significantly affected pathways in 28 cancer types and indicate that proteins containing harmful AASs can affect pathways despite the frequency of AASs in them. Our cross-cancer overlap analysis showed that it would be more beneficial to identify affected pathways in cancers rather than individual genes and variations. Conclusion Pathways affected by harmful AASs reveal key processes involved in cancer development. Our approach filters out the putative benign AASs thus reducing the list of cancer variations allowing reliable identification of affected pathways. The pathways identified in individual cancer and overlap between cancer types open avenues for further experimental research and for developing targeted therapies and interventions. Electronic supplementary material The online version of this article (doi:10.1186/s12920-015-0125-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Abhishek Niroula
- Department of Experimental Medical Science, Lund University, BMC B13, SE-22184, Lund, Sweden.
| | - Mauno Vihinen
- Department of Experimental Medical Science, Lund University, BMC B13, SE-22184, Lund, Sweden.
| |
Collapse
|
117
|
Arsuaga J, Borrman T, Cavalcante R, Gonzalez G, Park C. Identification of Copy Number Aberrations in Breast Cancer Subtypes Using Persistence Topology. MICROARRAYS (BASEL, SWITZERLAND) 2015; 4:339-69. [PMID: 27600228 PMCID: PMC4996377 DOI: 10.3390/microarrays4030339] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 08/03/2015] [Indexed: 01/01/2023]
Abstract
DNA copy number aberrations (CNAs) are of biological and medical interest because they help identify regulatory mechanisms underlying tumor initiation and evolution. Identification of tumor-driving CNAs (driver CNAs) however remains a challenging task, because they are frequently hidden by CNAs that are the product of random events that take place during tumor evolution. Experimental detection of CNAs is commonly accomplished through array comparative genomic hybridization (aCGH) assays followed by supervised and/or unsupervised statistical methods that combine the segmented profiles of all patients to identify driver CNAs. Here, we extend a previously-presented supervised algorithm for the identification of CNAs that is based on a topological representation of the data. Our method associates a two-dimensional (2D) point cloud with each aCGH profile and generates a sequence of simplicial complexes, mathematical objects that generalize the concept of a graph. This representation of the data permits segmenting the data at different resolutions and identifying CNAs by interrogating the topological properties of these simplicial complexes. We tested our approach on a published dataset with the goal of identifying specific breast cancer CNAs associated with specific molecular subtypes. Identification of CNAs associated with each subtype was performed by analyzing each subtype separately from the others and by taking the rest of the subtypes as the control. Our results found a new amplification in 11q at the location of the progesterone receptor in the Luminal A subtype. Aberrations in the Luminal B subtype were found only upon removal of the basal-like subtype from the control set. Under those conditions, all regions found in the original publication, except for 17q, were confirmed; all aberrations, except those in chromosome arms 8q and 12q were confirmed in the basal-like subtype. These two chromosome arms, however, were detected only upon removal of three patients with exceedingly large copy number values. More importantly, we detected 10 and 21 additional regions in the Luminal B and basal-like subtypes, respectively. Most of the additional regions were either validated on an independent dataset and/or using GISTIC. Furthermore, we found three new CNAs in the basal-like subtype: a combination of gains and losses in 1p, a gain in 2p and a loss in 14q. Based on these results, we suggest that topological approaches that incorporate multiresolution analyses and that interrogate topological properties of the data can help in the identification of copy number changes in cancer.
Collapse
Affiliation(s)
- Javier Arsuaga
- Department of Mathematics, University of California Davis, 1 Shields Avenue, Davis, CA 95616, USA.
- Department of Molecular and Cellular Biology, University of California Davis, 1 Shields Avenue, Davis, CA 95616, USA.
| | - Tyler Borrman
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Raymond Cavalcante
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Georgina Gonzalez
- Department of Mathematics, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 96132, USA.
| | - Catherine Park
- Helen Diller Comprehensive Cancer Center,University of California San Francisco, 1600 Divisadero Street, San Francisco, CA 94143, USA.
| |
Collapse
|
118
|
Leiserson MDM, Gramazio CC, Hu J, Wu HT, Laidlaw DH, Raphael BJ. MAGI: visualization and collaborative annotation of genomic aberrations. Nat Methods 2015; 12:483-4. [PMID: 26020500 DOI: 10.1038/nmeth.3412] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Mark D M Leiserson
- 1] Department of Computer Science, Brown University, Providence, Rhode Island, USA. [2] Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, USA
| | - Connor C Gramazio
- Department of Computer Science, Brown University, Providence, Rhode Island, USA
| | - Jason Hu
- Department of Computer Science, Brown University, Providence, Rhode Island, USA
| | - Hsin-Ta Wu
- 1] Department of Computer Science, Brown University, Providence, Rhode Island, USA. [2] Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, USA
| | - David H Laidlaw
- 1] Department of Computer Science, Brown University, Providence, Rhode Island, USA. [2] Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, USA
| | - Benjamin J Raphael
- 1] Department of Computer Science, Brown University, Providence, Rhode Island, USA. [2] Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
119
|
Wei L, Liu LT, Conroy JR, Hu Q, Conroy JM, Morrison CD, Johnson CS, Wang J, Liu S. MAC: identifying and correcting annotation for multi-nucleotide variations. BMC Genomics 2015; 16:569. [PMID: 26231518 PMCID: PMC4521406 DOI: 10.1186/s12864-015-1779-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 07/14/2015] [Indexed: 12/17/2022] Open
Abstract
Background Next-Generation Sequencing (NGS) technologies have rapidly advanced our understanding of human variation in cancer. To accurately translate the raw sequencing data into practical knowledge, annotation tools, algorithms and pipelines must be developed that keep pace with the rapidly evolving technology. Currently, a challenge exists in accurately annotating multi-nucleotide variants (MNVs). These tandem substitutions, when affecting multiple nucleotides within a single protein codon of a gene, result in a translated amino acid involving all nucleotides in that codon. Most existing variant callers report a MNV as individual single-nucleotide variants (SNVs), often resulting in multiple triplet codon sequences and incorrect amino acid predictions. To correct potentially misannotated MNVs among reported SNVs, a primary challenge resides in haplotype phasing which is to determine whether the neighboring SNVs are co-located on the same chromosome. Results Here we describe MAC (Multi-Nucleotide Variant Annotation Corrector), an integrative pipeline developed to correct potentially mis-annotated MNVs. MAC was designed as an application that only requires a SNV file and the matching BAM file as data inputs. Using an example data set containing 3024 SNVs and the corresponding whole-genome sequencing BAM files, we show that MAC identified eight potentially mis-annotated SNVs, and accurately updated the amino acid predictions for seven of the variant calls. Conclusions MAC can identify and correct amino acid predictions that result from MNVs affecting multiple nucleotides within a single protein codon, which cannot be handled by most existing SNV-based variant pipelines. The MAC software is freely available and represents a useful tool for the accurate translation of genomic sequence to protein function.
Collapse
Affiliation(s)
- Lei Wei
- Department of Biostatistics & Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA.
| | - Lu T Liu
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA.
| | - Jacob R Conroy
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA.
| | - Qiang Hu
- Department of Biostatistics & Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA.
| | - Jeffrey M Conroy
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA.
| | - Carl D Morrison
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA.
| | - Candace S Johnson
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA.
| | - Jianmin Wang
- Department of Biostatistics & Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA.
| | - Song Liu
- Department of Biostatistics & Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA.
| |
Collapse
|
120
|
Gross A, Schoendube J, Zimmermann S, Steeb M, Zengerle R, Koltay P. Technologies for Single-Cell Isolation. Int J Mol Sci 2015; 16:16897-919. [PMID: 26213926 PMCID: PMC4581176 DOI: 10.3390/ijms160816897] [Citation(s) in RCA: 269] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 07/06/2015] [Accepted: 07/14/2015] [Indexed: 11/16/2022] Open
Abstract
The handling of single cells is of great importance in applications such as cell line development or single-cell analysis, e.g., for cancer research or for emerging diagnostic methods. This review provides an overview of technologies that are currently used or in development to isolate single cells for subsequent single-cell analysis. Data from a dedicated online market survey conducted to identify the most relevant technologies, presented here for the first time, shows that FACS (fluorescence activated cell sorting) respectively Flow cytometry (33% usage), laser microdissection (17%), manual cell picking (17%), random seeding/dilution (15%), and microfluidics/lab-on-a-chip devices (12%) are currently the most frequently used technologies. These most prominent technologies are described in detail and key performance factors are discussed. The survey data indicates a further increasing interest in single-cell isolation tools for the coming years. Additionally, a worldwide patent search was performed to screen for emerging technologies that might become relevant in the future. In total 179 patents were found, out of which 25 were evaluated by screening the title and abstract to be relevant to the field.
Collapse
Affiliation(s)
- Andre Gross
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
- Cytena GmbH, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
| | - Jonas Schoendube
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
- Cytena GmbH, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
| | - Stefan Zimmermann
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
| | - Maximilian Steeb
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
| | - Roland Zengerle
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
- Hahn-Schickard, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
- BIOSS-Centre for Biological Signalling Studies, University of Freiburg, Freiburg 79110, Germany.
| | - Peter Koltay
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
- Cytena GmbH, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
| |
Collapse
|
121
|
Kang X, Chen K, Li Y, Li J, D'Amico TA, Chen X. Personalized targeted therapy for esophageal squamous cell carcinoma. World J Gastroenterol 2015; 21:7648-58. [PMID: 26167067 PMCID: PMC4491954 DOI: 10.3748/wjg.v21.i25.7648] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 03/19/2015] [Accepted: 04/28/2015] [Indexed: 02/06/2023] Open
Abstract
Esophageal squamous cell carcinoma continues to heavily burden clinicians worldwide. Researchers have discovered the genomic landscape of esophageal squamous cell carcinoma, which holds promise for an era of personalized oncology care. One of the most pressing problems facing this issue is to improve the understanding of the newly available genomic data, and identify the driver-gene mutations, pathways, and networks. The emergence of a legion of novel targeted agents has generated much hope and hype regarding more potent treatment regimens, but the accuracy of drug selection is still arguable. Other problems, such as cancer heterogeneity, drug resistance, exceptional responders, and side effects, have to be surmounted. Evolving topics in personalized oncology, such as interpretation of genomics data, issues in targeted therapy, research approaches for targeted therapy, and future perspectives, will be discussed in this editorial.
Collapse
|
122
|
Lim JS, Lee JH. Molecular genetic decoding of malformations of cortical development. ACTA ACUST UNITED AC 2015. [DOI: 10.5734/jgm.2015.12.1.12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Jae Seok Lim
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Korea
| | - Jeong Ho Lee
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Korea
| |
Collapse
|
123
|
Wills QF, Mead AJ. Application of single-cell genomics in cancer: promise and challenges. Hum Mol Genet 2015; 24:R74-84. [PMID: 26113645 PMCID: PMC4571998 DOI: 10.1093/hmg/ddv235] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 06/18/2015] [Indexed: 12/13/2022] Open
Abstract
Recent advances in single-cell genomics are opening up unprecedented opportunities to transform cancer genomics. While bulk tissue genomic analysis across large populations of tumour cells has provided key insights into cancer biology, this approach does not provide the resolution that is critical for understanding the interaction between different genetic events within the cellular hierarchy of the tumour during disease initiation, evolution, relapse and metastasis. Single-cell genomic approaches are uniquely placed to definitively unravel complex clonal structures and tissue hierarchies, account for spatiotemporal cell interactions and discover rare cells that drive metastatic disease, drug resistance and disease progression. Here we present five challenges that need to be met for single-cell genomics to fulfil its potential as a routine tool alongside bulk sequencing. These might be thought of as being challenges related to samples (processing and scale for analysis), sensitivity and specificity of mutation detection, sources of heterogeneity (biological and technical), synergies (from data integration) and systems modelling. We discuss these in the context of recent advances in technologies and data modelling, concluding with implications for moving cancer research into the clinic.
Collapse
Affiliation(s)
- Quin F Wills
- Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK and
| | - Adam J Mead
- Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK, NIHR Biomedical Research Centre, Churchill Hospital, Oxford OX3 7LE, UK
| |
Collapse
|
124
|
Tian R, Basu MK, Capriotti E. Computational methods and resources for the interpretation of genomic variants in cancer. BMC Genomics 2015; 16 Suppl 8:S7. [PMID: 26111056 PMCID: PMC4480958 DOI: 10.1186/1471-2164-16-s8-s7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The recent improvement of the high-throughput sequencing technologies is having a strong impact on the detection of genetic variations associated with cancer. Several institutions worldwide have been sequencing the whole exomes and or genomes of cancer patients in the thousands, thereby providing an invaluable collection of new somatic mutations in different cancer types. These initiatives promoted the development of methods and tools for the analysis of cancer genomes that are aimed at studying the relationship between genotype and phenotype in cancer. In this article we review the online resources and computational tools for the analysis of cancer genome. First, we describe the available repositories of cancer genome data. Next, we provide an overview of the methods for the detection of genetic variation and computational tools for the prioritization of cancer related genes and causative somatic variations. Finally, we discuss the future perspectives in cancer genomics focusing on the impact of computational methods and quantitative approaches for defining personalized strategies to improve the diagnosis and treatment of cancer.
Collapse
|
125
|
Proteomics in cancer biomarkers discovery: challenges and applications. DISEASE MARKERS 2015; 2015:321370. [PMID: 25999657 PMCID: PMC4427011 DOI: 10.1155/2015/321370] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 01/15/2015] [Accepted: 02/18/2015] [Indexed: 01/28/2023]
Abstract
With the introduction of recent high-throughput technologies to various fields of science and medicine, it is becoming clear that obtaining large amounts of data is no longer a problem in modern research laboratories. However, coherent study designs, optimal conditions for obtaining high-quality data, and compelling interpretation, in accordance with the evidence-based systems biology, are critical factors in ensuring the emergence of good science out of these recent technologies. This review focuses on the proteomics field and its new perspectives on cancer research. Cornerstone publications that have tremendously helped scientists and clinicians to better understand cancer pathogenesis; to discover novel diagnostic and/or prognostic biomarkers; and to suggest novel therapeutic targets will be presented. The author of this review aims at presenting some of the relevant literature data that helped as a step forward in bridging the gap between bench work results and bedside potentials. Undeniably, this review cannot include all the work that is being produced by expert research groups all over the world.
Collapse
|
126
|
Hastings RJ, Bown N, Tibiletti MG, Debiec-Rychter M, Vanni R, Espinet B, van Roy N, Roberts P, van den Berg-de-Ruiter E, Bernheim A, Schoumans J, Chatters S, Zemanova Z, Stevens-Kroef M, Simons A, Heim S, Salido M, Ylstra B, Betts DR. Guidelines for cytogenetic investigations in tumours. Eur J Hum Genet 2015; 24:6-13. [PMID: 25804401 DOI: 10.1038/ejhg.2015.35] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 01/19/2015] [Accepted: 02/03/2015] [Indexed: 12/31/2022] Open
Affiliation(s)
- Rosalind J Hastings
- Women's Centre, John Radcliffe Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Nick Bown
- Northern Genetics Service, Institute of Genomic Medicine, Central Parkway, Newcastle, UK
| | - Maria G Tibiletti
- UO Anatomia Patologica, Ospedale di Circolo-Polo Universitario, Varese, Italy
| | - Maria Debiec-Rychter
- Laboratory for Genetics of Malignant Disorders, Department of Human Genetics, University Hospital Gasthuisberg, UZ Leuven, Leuven, Belgium
| | - Roberta Vanni
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, Italy
| | - Blanca Espinet
- Laboratori de Citogenètica Molecular, Servei de Patologia, Hospital del Mar, Barcelona, Spain
| | - Nadine van Roy
- Centre for Medical Genetics, University Hospital Ghent, Ghent, Belgium
| | - Paul Roberts
- Cytogenetics Department, St James's Hospital, Leeds, UK
| | - Eva van den Berg-de-Ruiter
- Department of Genetics, University of Groningen, University Medical Centre Groningen, RB Groningen, The Netherlands
| | - Alain Bernheim
- Génétique des tumeurs (INSERM U985), Laboratoire de Cytogénétique, Pathologie Moléculaire Gustave Roussy, Paris-Villejuif, France
| | - Jacqueline Schoumans
- Cancer Cytogenetic Unit, Lausanne University Hospital, CHUV, Lausanne, Switzerland
| | - Steve Chatters
- Haematology, Cellular and Molecular Diagnostic Service, Great Ormond St Hospital, London, UK
| | - Zuzana Zemanova
- Center of Oncocytogenetics, Institute of Clinical Biochemistry and Laboratory Diagnostics, General University Hospital and First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Marian Stevens-Kroef
- Radboud University Nijmegen Medical Centre Department of Human Genetics, HB Nijmegen, The Netherlands
| | - Annet Simons
- Radboud University Nijmegen Medical Centre Department of Human Genetics, HB Nijmegen, The Netherlands
| | - Sverre Heim
- Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Marta Salido
- Laboratori de Citogenètica Molecular, Servei de Patologia, Hospital del Mar, Barcelona, Spain
| | - Bauke Ylstra
- Department of Pathology, Cancer Center Amsterdam, VU University Medical Center, MB Amsterdam, The Netherlands
| | - David R Betts
- Department of Clinical Genetics, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| | | | | |
Collapse
|
127
|
Beck AH. Open access to large scale datasets is needed to translate knowledge of cancer heterogeneity into better patient outcomes. PLoS Med 2015; 12:e1001794. [PMID: 25710538 PMCID: PMC4339838 DOI: 10.1371/journal.pmed.1001794] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In this guest editorial, Andrew Beck discusses the importance of open access to big data for translating knowledge of cancer heterogeneity into better outcomes for cancer patients.
Collapse
Affiliation(s)
- Andrew H. Beck
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
128
|
McGranahan N, Swanton C. Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer Cell 2015; 27:15-26. [PMID: 25584892 DOI: 10.1016/j.ccell.2014.12.001] [Citation(s) in RCA: 781] [Impact Index Per Article: 86.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Indexed: 10/24/2022]
Abstract
Precision medicine requires an understanding of cancer genes and mutational processes, as well as an appreciation of the extent to which these are found heterogeneously in cancer cells during tumor evolution. Here, we explore the processes shaping the cancer genome, placing these within the context of tumor evolution and their impact on intratumor heterogeneity and drug development. We review evidence for constraints and contingencies to tumor evolution and highlight the clinical implications of diversity within tumors. We outline the limitations of genome-driven targeted therapies and explore future strategies, including immune and adaptive approaches, to address this therapeutic challenge.
Collapse
Affiliation(s)
- Nicholas McGranahan
- Cancer Research UK London Research Institute, London WC2A 3LY, UK; Centre for Mathematics & Physics in the Life Science & Experimental Biology (CoMPLEX), University College London, London WC1E 6BT, UK
| | - Charles Swanton
- Cancer Research UK London Research Institute, London WC2A 3LY, UK; UCL Cancer Institute, Paul O'Gorman Building, Huntley Street, London WC1E 6DD, UK.
| |
Collapse
|
129
|
Abstract
A system characterized by redundancy has various elements that are able to act in the same biologic or dynamic manner, where the inhibition of one of those elements has no significant effect on the global biologic outcome or on the system's dynamic behavior. Methods that aim to predict the effectiveness of cancer therapies must include evolutionary and dynamic features that would change the static view that is widely accepted. Here, we explore several important issues about mechanisms of redundancy, heterogeneity, biologic importance, and drug resistance and describe methodologic challenges that, if overcome, would significantly contribute to cancer research.
Collapse
Affiliation(s)
- Orit Lavi
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
130
|
Somatic mosaicism in the human genome. Genes (Basel) 2014; 5:1064-94. [PMID: 25513881 PMCID: PMC4276927 DOI: 10.3390/genes5041064] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 11/26/2014] [Accepted: 11/28/2014] [Indexed: 12/17/2022] Open
Abstract
Somatic mosaicism refers to the occurrence of two genetically distinct populations of cells within an individual, derived from a postzygotic mutation. In contrast to inherited mutations, somatic mosaic mutations may affect only a portion of the body and are not transmitted to progeny. These mutations affect varying genomic sizes ranging from single nucleotides to entire chromosomes and have been implicated in disease, most prominently cancer. The phenotypic consequences of somatic mosaicism are dependent upon many factors including the developmental time at which the mutation occurs, the areas of the body that are affected, and the pathophysiological effect(s) of the mutation. The advent of second-generation sequencing technologies has augmented existing array-based and cytogenetic approaches for the identification of somatic mutations. We outline the strengths and weaknesses of these techniques and highlight recent insights into the role of somatic mosaicism in causing cancer, neurodegenerative, monogenic, and complex disease.
Collapse
|
131
|
Bennett NC, Farah CS. Next-generation sequencing in clinical oncology: next steps towards clinical validation. Cancers (Basel) 2014; 6:2296-312. [PMID: 25412366 PMCID: PMC4276967 DOI: 10.3390/cancers6042296] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 11/07/2014] [Accepted: 11/10/2014] [Indexed: 02/05/2023] Open
Abstract
Compelling evidence supports the transition of next generation sequencing (NGS) technology from a research environment into clinical practice. Before NGS technologies are fully adopted in the clinic, they should be thoroughly scrutinised for their potential as powerful diagnostic and prognostic tools. The importance placed on generating accurate NGS data, and consequently appropriate clinical interpretation, has stimulated much international discussion regarding the creation and implementation of strict guidelines and regulations for NGS clinical use. In the context of clinical oncology, NGS technologies are currently transitioning from a clinical research background into a setting where they will contribute significantly to individual patient cancer management. This paper explores the steps that have been taken, and those still required, for the transition of NGS into the clinical area, with particular emphasis placed on validation in the setting of clinical oncology.
Collapse
Affiliation(s)
- Nigel C Bennett
- The University of Queensland, UQ Centre for Clinical Research, Herston Qld 4029, Australia.
| | - Camile S Farah
- The University of Queensland, UQ Centre for Clinical Research, Herston Qld 4029, Australia.
| |
Collapse
|