5501
|
Malapelle U, Carlomagno C, de Luca C, Bellevicine C, Troncone G. KRAS testing in metastatic colorectal carcinoma: challenges, controversies, breakthroughs and beyond. J Clin Pathol 2013; 67:1-9. [DOI: 10.1136/jclinpath-2013-201835] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
5502
|
Coelho TR, Gaspar R, Figueiredo P, Mendonça C, Lazo PA, Almeida L. Human JC polyomavirus in normal colorectal mucosa, hyperplastic polyps, sporadic adenomas, and adenocarcinomas in Portugal. J Med Virol 2013; 85:2119-27. [DOI: 10.1002/jmv.23705] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2013] [Indexed: 11/09/2022]
Affiliation(s)
- Tatiana Rasteiro Coelho
- Faculty of Medicine, Pharmacology and Experimental Therapeutics Service (Unit of Therapeutics); University of Coimbra; Coimbra Portugal
| | - Rita Gaspar
- Faculty of Medicine, Pharmacology and Experimental Therapeutics Service (Unit of Therapeutics); University of Coimbra; Coimbra Portugal
| | - Pedro Figueiredo
- Faculty of Medicine; University of Coimbra; Coimbra Portugal
- Gastroenterology Department; Hospital of the University of Coimbra; Coimbra Portugal
| | - Cristina Mendonça
- Faculty of Medicine; University of Coimbra; Coimbra Portugal
- Center Department; National Institute of Forensic Medicine and Forensic Sciences; Coimbra Portugal
| | - Pedro A. Lazo
- Instituto de Biología Molecular y Celular del Cáncer; CSIC-Universidad de Salamanca; Salamanca Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL); Hospital Universitario de Salamanca; Salamanca Spain
| | - Luís Almeida
- Faculty of Medicine, Pharmacology and Experimental Therapeutics Service (Unit of Therapeutics); University of Coimbra; Coimbra Portugal
- Faculty of Medicine; University of Coimbra; Coimbra Portugal
| |
Collapse
|
5503
|
Hosgood HD, Pao W, Rothman N, Hu W, Pan YH, Kuchinsky K, Jones KD, Xu J, Vermeulen R, Simko J, Lan Q. Driver mutations among never smoking female lung cancer tissues in China identify unique EGFR and KRAS mutation pattern associated with household coal burning. Respir Med 2013; 107:1755-62. [PMID: 24055406 DOI: 10.1016/j.rmed.2013.08.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 07/31/2013] [Accepted: 08/22/2013] [Indexed: 01/30/2023]
Abstract
Lung cancer in never smokers, which has been partially attributed to household solid fuel use (i.e., coal), is etiologically and clinically different from lung cancer attributed to tobacco smoking. To explore the spectrum of driver mutations among lung cancer tissues from never smokers, specifically in a population where high lung cancer rates have been attributed to indoor air pollution from domestic coal use, multiplexed assays were used to detect >40 point mutations, insertions, and deletions (EGFR, KRAS, BRAF, HER2, NRAS, PIK3CA, MEK1, AKT1, and PTEN) among the lung tumors of confirmed never smoking females from Xuanwei, China [32 adenocarcinomas (ADCs), 7 squamous cell carcinomas (SCCs), 1 adenosquamous carcinoma (ADSC)]. EGFR mutations were detected in 35% of tumors. 46% of these involved EGFR exon 18 G719X, while 14% were exon 21 L858R mutations. KRAS mutations, all of which were G12C_34G>T, were observed in 15% of tumors. EGFR and KRAS mutations were mutually exclusive, and no mutations were observed in the other tested genes. Most point mutations were transversions and were also found in tumors from patients who used coal in their homes. Our high mutation frequencies in EGFR exon 18 and KRAS and low mutation frequency in EGFR exon 21 are strikingly divergent from those in other smoking and never smoking populations from Asia. Given that our subjects live in a region where coal is typically burned indoors, our findings provide new insights into the pathogenesis of lung cancer among never smoking females exposed to indoor air pollution from coal.
Collapse
Affiliation(s)
- H Dean Hosgood
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, NCI, NIH, DHHS, Bethesda, MD, USA; Division of Epidemiology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5504
|
Lewis NE, Abdel-Haleem AM. The evolution of genome-scale models of cancer metabolism. Front Physiol 2013; 4:237. [PMID: 24027532 PMCID: PMC3759783 DOI: 10.3389/fphys.2013.00237] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 08/13/2013] [Indexed: 01/30/2023] Open
Abstract
The importance of metabolism in cancer is becoming increasingly apparent with the identification of metabolic enzyme mutations and the growing awareness of the influence of metabolism on signaling, epigenetic markers, and transcription. However, the complexity of these processes has challenged our ability to make sense of the metabolic changes in cancer. Fortunately, constraint-based modeling, a systems biology approach, now enables one to study the entirety of cancer metabolism and simulate basic phenotypes. With the newness of this field, there has been a rapid evolution of both the scope of these models and their applications. Here we review the various constraint-based models built for cancer metabolism and how their predictions are shedding new light on basic cancer phenotypes, elucidating pathway differences between tumors, and dicovering putative anti-cancer targets. As the field continues to evolve, the scope of these genome-scale cancer models must expand beyond central metabolism to address questions related to the diverse processes contributing to tumor development and metastasis.
Collapse
Affiliation(s)
- Nathan E Lewis
- Department of Biology, Brigham Young University Provo, UT, USA
| | | |
Collapse
|
5505
|
Abstract
The zebrafish is a recent addition to animal models of human cancer, and studies using this model are rapidly contributing major insights. Zebrafish develop cancer spontaneously, after mutagen exposure and through transgenesis. The tumours resemble human cancers at the histological, gene expression and genomic levels. The ability to carry out in vivo imaging, chemical and genetic screens, and high-throughput transgenesis offers a unique opportunity to functionally characterize the cancer genome. Moreover, increasingly sophisticated modelling of combinations of genetic and epigenetic alterations will allow the zebrafish to complement what can be achieved in other models, such as mouse and human cell culture systems.
Collapse
Affiliation(s)
- Richard White
- Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA.
| | | | | |
Collapse
|
5506
|
Doble B, Harris A, Thomas DM, Fox S, Lorgelly P. Multiomics medicine in oncology: assessing effectiveness, cost–effectiveness and future research priorities for the molecularly unique individual. Pharmacogenomics 2013; 14:1405-17. [DOI: 10.2217/pgs.13.142] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The development of genomic technologies has ushered in the era of pharmacogenomics. However, discoveries and clinical use of targeted therapies are still in their infancy. A focus on monogenic pharmacogenetic traits may contribute to this lack of progress. Variation in drug response is likely a complex paradigm involving not only genomic factors but proteomic, metabolomic and epigenomic influences. The incorporation of these omics elements into pharmaceutical development and clinical decision-making will ultimately require the use of methods to determine clinical and economic value. Current methodologies and guidelines for determining clinical effectiveness and cost–effectiveness may have limited applicability to the increasingly personalized nature of omics treatment strategies. Using examples from oncology, this article argues for the adaptation and tailoring of three existing methods for ensuring development and clinical use of multiomics-guided therapies that are effective, safe and offer value for money.
Collapse
Affiliation(s)
- Brett Doble
- Centre for Health Economics, Faculty of Business & Economics, Room 278, Level 2, Building 75, Monash University, Clayton, Victoria 3800, Australia
| | - Anthony Harris
- Centre for Health Economics, Faculty of Business & Economics, Room 278, Level 2, Building 75, Monash University, Clayton, Victoria 3800, Australia
| | - David M Thomas
- Division of Cancer Medicine, Sir Peter MacCallum Department of Oncology, University of Melbourne, East Melbourne, Victoria, Australia
| | - Stephen Fox
- Molecular Pathology Research & Development Laboratory, Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Paula Lorgelly
- Centre for Health Economics, Faculty of Business & Economics, Room 278, Level 2, Building 75, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
5507
|
Abstract
Two review articles published in 2000 and 2011 by Hanahan and Weinberg have dominated the discourse about carcinogenesis among researchers in the recent past. The basic tenets of their arguments favour considering cancer as a cell-based, genetic disease whereby DNA mutations cause uncontrolled cell proliferation. Their explanation of cancer phenotypes is based on the premises adopted by the somatic mutation theory (SMT) and its cell-centered variants. From their perspective, eight broad features have been identified as so-called 'Hallmarks of Cancer'. Here, we criticize the value of these features based on the numerous intrinsic inconsistencies in the data and in the rationale behind SMT. An alternative interpretation of the same data plus data mostly ignored by Hanahan and Weinberg is proposed, based instead on evolutionarily relevant premises. From such a perspective, cancer is viewed as a tissue-based disease. This alternative, called the tissue organization field theory, incorporates the premise that proliferation and motility are the default state of all cells, and that carcinogenesis is due to alterations on the reciprocal interactions among cells and between cells and their extracellular matrix. In this view, cancer is development gone awry.
Collapse
Affiliation(s)
- Carlos Sonnenschein
- Tufts University School of Medicine, Program on Cellular, Developmental and Molecular Biology, Boston, MA 02111, USA
| | - Ana M. Soto
- Tufts University School of Medicine, Program on Cellular, Developmental and Molecular Biology, Boston, MA 02111, USA
| |
Collapse
|
5508
|
Zhang G, Hoersch S, Amsterdam A, Whittaker CA, Beert E, Catchen JM, Farrington S, Postlethwait JH, Legius E, Hopkins N, Lees JA. Comparative oncogenomic analysis of copy number alterations in human and zebrafish tumors enables cancer driver discovery. PLoS Genet 2013; 9:e1003734. [PMID: 24009526 PMCID: PMC3757083 DOI: 10.1371/journal.pgen.1003734] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 07/05/2013] [Indexed: 01/11/2023] Open
Abstract
The identification of cancer drivers is a major goal of current cancer research. Finding driver genes within large chromosomal events is especially challenging because such alterations encompass many genes. Previously, we demonstrated that zebrafish malignant peripheral nerve sheath tumors (MPNSTs) are highly aneuploid, much like human tumors. In this study, we examined 147 zebrafish MPNSTs by massively parallel sequencing and identified both large and focal copy number alterations (CNAs). Given the low degree of conserved synteny between fish and mammals, we reasoned that comparative analyses of CNAs from fish versus human MPNSTs would enable elimination of a large proportion of passenger mutations, especially on large CNAs. We established a list of orthologous genes between human and zebrafish, which includes approximately two-thirds of human protein-coding genes. For the subset of these genes found in human MPNST CNAs, only one quarter of their orthologues were co-gained or co-lost in zebrafish, dramatically narrowing the list of candidate cancer drivers for both focal and large CNAs. We conclude that zebrafish-human comparative analysis represents a powerful, and broadly applicable, tool to enrich for evolutionarily conserved cancer drivers.
Collapse
Affiliation(s)
- GuangJun Zhang
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts, United States of America
| | - Sebastian Hoersch
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts, United States of America
- Bioinformatics Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Adam Amsterdam
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts, United States of America
| | - Charles A. Whittaker
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts, United States of America
| | - Eline Beert
- Department of Human Genetics, Catholic University Leuven, Leuven, Belgium
| | - Julian M. Catchen
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
| | - Sarah Farrington
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts, United States of America
| | - John H. Postlethwait
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
| | - Eric Legius
- Department of Human Genetics, Catholic University Leuven, Leuven, Belgium
| | - Nancy Hopkins
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts, United States of America
| | - Jacqueline A. Lees
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
5509
|
Lichtman MA. Does a diagnosis of myelogenous leukemia require 20% marrow myeloblasts, and does <5% marrow myeloblasts represent a remission? The history and ambiguity of arbitrary diagnostic boundaries in the understanding of myelodysplasia. Oncologist 2013; 18:973-80. [PMID: 23982763 DOI: 10.1634/theoncologist.2013-0099] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
5510
|
Abstract
Non-small-cell lung cancer is often diagnosed at the metastatic stage, with median survival of just 1 year. The identification of driver mutations in the epidermal growth factor receptor (EGFR) as the primary oncogenic event in a subset of lung adenocarcinomas led to a model of targeted treatment and genetic profiling of the disease. EGFR tyrosine kinase inhibitors confer remission in 60% of patients, but responses are short-lived. The pre-existing EGFR Thr790Met mutation could be a subclonal driver responsible for these transient responses. Overexpression of AXL and reduced MED12 function are hallmarks of resistance to tyrosine kinase inhibitors in EGFR-mutant non-small-cell lung cancer. Crosstalk between signalling pathways is another mechanism of resistance; therefore, identification of the molecular components involved could lead to the development of combination therapies cotargeting these molecules instead of EGFR tyrosine kinase inhibitor monotherapy. Additionally, novel biomarkers could be identified through deep sequencing analysis of serial rebiopsies before and during treatment.
Collapse
|
5511
|
Heitzer E, Auer M, Ulz P, Geigl JB, Speicher MR. Circulating tumor cells and DNA as liquid biopsies. Genome Med 2013; 5:73. [PMID: 23998943 PMCID: PMC3979149 DOI: 10.1186/gm477] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
For cancer patients, the current approach to prognosis relies on clinicopathological staging, but usually this provides little information about the individual response to treatment. Therefore, there is a tremendous need for protein and genetic biomarkers with predictive and prognostic information. As biomarkers are identified, the serial monitoring of tumor genotypes, which are instable and prone to changes under selection pressure, is becoming increasingly possible. To this end, circulating tumor cells (CTCs) or circulating tumor DNA (ctDNA) shed from primary and metastatic cancers may allow the non-invasive analysis of the evolution of tumor genomes during treatment and disease progression through 'liquid biopsies'. Here we review recent progress in the identification of CTCs among thousands of other cells in the blood and new high-resolution approaches, including recent microfluidic platforms, for dissecting the genomes of CTCs and obtaining functional data. We also discuss new ctDNA-based approaches, which may become a powerful alternative to CTC analysis. Together, these approaches provide novel biological insights into the process of metastasis and may elucidate signaling pathways involved in cell invasiveness and metastatic competence. In medicine these liquid biopsies may emerge to be powerful predictive and prognostic biomarkers and could therefore be instrumental for areas such as precision or personalized medicine.
Collapse
Affiliation(s)
- Ellen Heitzer
- Institute of Human Genetics, Medical University of Graz, Harrachgasse 21/8, A-8010 Graz, Austria
| | - Martina Auer
- Institute of Human Genetics, Medical University of Graz, Harrachgasse 21/8, A-8010 Graz, Austria
| | - Peter Ulz
- Institute of Human Genetics, Medical University of Graz, Harrachgasse 21/8, A-8010 Graz, Austria
| | - Jochen B Geigl
- Institute of Human Genetics, Medical University of Graz, Harrachgasse 21/8, A-8010 Graz, Austria
| | - Michael R Speicher
- Institute of Human Genetics, Medical University of Graz, Harrachgasse 21/8, A-8010 Graz, Austria
| |
Collapse
|
5512
|
Ajani JA, Xiao L, Roth JA, Hofstetter WL, Walsh G, Komaki R, Liao Z, Rice DC, Vaporciyan AA, Maru DM, Lee JH, Bhutani MS, Eid A, Yao JC, Phan AP, Halpin A, Suzuki A, Taketa T, Thall PF, Swisher SG. A phase II randomized trial of induction chemotherapy versus no induction chemotherapy followed by preoperative chemoradiation in patients with esophageal cancer. Ann Oncol 2013; 24:2844-9. [PMID: 23975663 DOI: 10.1093/annonc/mdt339] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The contribution of induction chemotherapy (IC) before preoperative chemoradiation for esophageal cancer (EC) is not known. We hypothesized that IC would increase the rate of pathologic complete response (pathCR). METHODS Trimodality-eligibile patients were randomized to receive no IC (Arm A) or IC (oxaliplatin/FU; Arm B) before oxaliplatin/FU/radiation. Surgery was attempted ∼5-6 weeks after chemoradiation. The pathCR rate, post-surgery 30-day mortality, overall survival (OS), and toxic effects were assessed. Bayesian methods and Fisher's exact test were used. RESULTS One hundred twenty-six patients were randomized dynamically to balance the two arms for histology, baseline stage, gender, race, and age. Fifty-five patients in Arm A and 54 in Arm B underwent surgery. The median actuarial OS for all patients (54 deaths) was 45.62 months [95% confidence interval (CI), 27.63-NA], with median OS 45.62 months (95% CI 25.56-NA) in Arm A and 43.68 months (95% CI 27.63-NA) in Arm B (P = 0.69). The pathCR rate in Arm A was 13% (7 of 55) and 26% (14 of 54) in Arm B (two-sided Fisher's exact test, P = 0.094). Safety was similar in both arms. CONCLUSIONS These data suggest that IC produces non-significant increase in the pathCR rate and does not prolong OS. Further development of IC before chemoradiation may not be beneficial. Clinical trial no.: NCT 00525915 (www.clinicaltrials.gov).
Collapse
Affiliation(s)
- J A Ajani
- Departments of Gastrointestinal Medical Oncology
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5513
|
Ow TJ, Sandulache VC, Skinner HD, Myers JN. Integration of cancer genomics with treatment selection: from the genome to predictive biomarkers. Cancer 2013; 119:3914-28. [PMID: 24037788 DOI: 10.1002/cncr.28304] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 07/02/2013] [Accepted: 07/02/2013] [Indexed: 12/11/2022]
Abstract
The field of cancer genomics is rapidly advancing as new technology provides detailed genetic and epigenetic profiling of human cancers. The amount of new data available describing the genetic make-up of tumors is paralleled by rapid advances in drug discovery and molecular therapy currently under investigation to treat these diseases. This review summarizes the challenges and approaches associated with the integration of genomic data into the development of new biomarkers in the management of cancer.
Collapse
Affiliation(s)
- Thomas J Ow
- Department of Otorhinolaryngology-Head and Neck Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York; Department of Pathology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York
| | | | | | | |
Collapse
|
5514
|
A nanoparticle formulation that selectively transfects metastatic tumors in mice. Proc Natl Acad Sci U S A 2013; 110:14717-22. [PMID: 23959886 DOI: 10.1073/pnas.1313330110] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Nanoparticle gene therapy holds great promise for the treatment of malignant disease in light of the large number of potent, tumor-specific therapeutic payloads potentially available for delivery. To be effective, gene therapy vehicles must be able to deliver their therapeutic payloads to metastatic lesions after systemic administration. Here we describe nanoparticles comprised of a core of high molecular weight linear polyethylenimine (LPEI) complexed with DNA and surrounded by a shell of polyethyleneglycol-modified (PEGylated) low molecular weight LPEI. Compared with a state-of-the-art commercially available in vivo gene delivery formulation, i.v. delivery of the core/PEGylated shell (CPS) nanoparticles provided more than a 16,000-fold increase in the ratio of tumor to nontumor transfection. The vast majority of examined liver and lung metastases derived from a colorectal cancer cell line showed transgene expression after i.v. CPS injection in an animal model of metastasis. Histological examination of tissues from transfected mice revealed that the CPS nanoparticles selectively transfected neoplastic cells rather than stromal cells within primary and metastatic tumors. However, only a small fraction of neoplastic cells (<1%) expressed the transgene, and the extent of delivery varied with the tumor cell line, tumor site, and host mouse strain used. Our results demonstrate that these CPS nanoparticles offer substantial advantages over previously described formulations for in vivo nanoparticle gene therapeutics. At the same time, they illustrate that major increases in the effectiveness of such approaches are needed for utility in patients with metastatic cancer.
Collapse
|
5515
|
Shugay M, Ortiz de Mendíbil I, Vizmanos JL, Novo FJ. Oncofuse: a computational framework for the prediction of the oncogenic potential of gene fusions. ACTA ACUST UNITED AC 2013; 29:2539-46. [PMID: 23956304 DOI: 10.1093/bioinformatics/btt445] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
MOTIVATION Gene fusions resulting from chromosomal aberrations are an important cause of cancer. The complexity of genomic changes in certain cancer types has hampered the identification of gene fusions by molecular cytogenetic methods, especially in carcinomas. This is changing with the advent of next-generation sequencing, which is detecting a substantial number of new fusion transcripts in individual cancer genomes. However, this poses the challenge of identifying those fusions with greater oncogenic potential amid a background of 'passenger' fusion sequences. RESULTS In the present work, we have used some recently identified genomic hallmarks of oncogenic fusion genes to develop a pipeline for the classification of fusion sequences, namely, Oncofuse. The pipeline predicts the oncogenic potential of novel fusion genes, calculating the probability that a fusion sequence behaves as 'driver' of the oncogenic process based on features present in known oncogenic fusions. Cross-validation and extensive validation tests on independent datasets suggest a robust behavior with good precision and recall rates. We believe that Oncofuse could become a useful tool to guide experimental validation studies of novel fusion sequences found during next-generation sequencing analysis of cancer transcriptomes. AVAILABILITY AND IMPLEMENTATION Oncofuse is a naive Bayes Network Classifier trained and tested using Weka machine learning package. The pipeline is executed by running a Java/Groovy script, available for download at www.unav.es/genetica/oncofuse.html.
Collapse
Affiliation(s)
- Mikhail Shugay
- Department of Genetics, University of Navarra. 31008 Pamplona, Spain
| | | | | | | |
Collapse
|
5516
|
Huang X, Withers BR, Dickson RC. Sphingolipids and lifespan regulation. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:657-64. [PMID: 23954556 DOI: 10.1016/j.bbalip.2013.08.006] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 08/01/2013] [Accepted: 08/06/2013] [Indexed: 02/08/2023]
Abstract
Diseases including cancer, type 2 diabetes, cardiovascular and immune dysfunction and neurodegeneration become more prevalent as we age, and combined with the increase in average human lifespan, place an ever increasing burden on the health care system. In this chapter we focus on finding ways of modulating sphingolipids to prevent the development of age-associated diseases or delay their onset, both of which could improve health in elderly, fragile people. Reducing the incidence of or delaying the onset of diseases of aging has blossomed in the past decade because of advances in understanding signal transduction pathways and cellular processes, especially in model organisms, that are largely conserved in most eukaryotes and that can be modulated to reduce signs of aging and increase health span. In model organisms such interventions must also increase lifespan to be considered significant, but this is not a requirement for use in humans. The most encouraging interventions in model organisms involve lowering the concentration of one or more sphingolipids so as to reduce the activity of key signaling pathways, one of the most promising being the Target of Rapamycin Complex 1 (TORC1) protein kinase pathway. Other potential ways in which modulating sphingolipids may contribute to improving the health profile of the elderly is by reducing oxidative stresses, inflammatory responses and growth factor signaling. Lastly, perhaps the most interesting way to modulate sphingolipids and promote longevity is by lowering the activity of serine palmitoyltransferase, the first enzyme in the de novo sphingolipid biosynthesis pathway. Available data in yeasts and rodents are encouraging and as we gain insights into molecular mechanisms the strategies for improving human health by modulating sphingolipids will become more apparent. This article is part of a Special Issue entitled New Frontiers in Sphingolipid Biology.
Collapse
Affiliation(s)
- Xinhe Huang
- Department of Molecular and Cellular Biochemistry and the Lucille Markey Cancer Center, University of Kentucky College of Medicine, 741 S. Limestone, Lexington, KY 40536, USA
| | - Bradley R Withers
- Department of Molecular and Cellular Biochemistry and the Lucille Markey Cancer Center, University of Kentucky College of Medicine, 741 S. Limestone, Lexington, KY 40536, USA
| | - Robert C Dickson
- Department of Molecular and Cellular Biochemistry and the Lucille Markey Cancer Center, University of Kentucky College of Medicine, 741 S. Limestone, Lexington, KY 40536, USA.
| |
Collapse
|
5517
|
Poon SL, Pang ST, McPherson JR, Yu W, Huang KK, Guan P, Weng WH, Siew EY, Liu Y, Heng HL, Chong SC, Gan A, Tay ST, Lim WK, Cutcutache I, Huang D, Ler LD, Nairismagi ML, Lee MH, Chang YH, Yu KJ, Chan-on W, Li BK, Yuan YF, Qian CN, Ng KF, Wu CF, Hsu CL, Bunte RM, Stratton MR, Futreal PA, Sung WK, Chuang CK, Ong CK, Rozen SG, Tan P, Teh BT. Genome-Wide Mutational Signatures of Aristolochic Acid and Its Application as a Screening Tool. Sci Transl Med 2013; 5:197ra101. [DOI: 10.1126/scitranslmed.3006086] [Citation(s) in RCA: 202] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
5518
|
Hoang ML, Chen CH, Sidorenko VS, He J, Dickman KG, Yun BH, Moriya M, Niknafs N, Douville C, Karchin R, Turesky RJ, Pu YS, Vogelstein B, Papadopoulos N, Grollman AP, Kinzler KW, Rosenquist TA. Mutational signature of aristolochic acid exposure as revealed by whole-exome sequencing. Sci Transl Med 2013; 5:197ra102. [PMID: 23926200 PMCID: PMC3973132 DOI: 10.1126/scitranslmed.3006200] [Citation(s) in RCA: 202] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In humans, exposure to aristolochic acid (AA) is associated with urothelial carcinoma of the upper urinary tract (UTUC). Exome sequencing of UTUCs from 19 individuals with documented exposure to AA revealed a remarkably large number of somatic mutations and an unusual mutational signature attributable to AA. Most of the mutations (72%) in these tumors were A:T-to-T:A transversions, located predominantly on the nontranscribed strand, with a strong preference for deoxyadenosine in a consensus sequence (T/CAG). This trinucleotide motif overlaps the canonical splice acceptor site, possibly accounting for the excess of splice site mutations observed in these tumors. The AA mutational fingerprint was found frequently in oncogenes and tumor suppressor genes in AA-associated UTUC. The AA mutational signature was observed in one patient's tumor from a UTUC cohort without previous indication of AA exposure. Together, these results directly link an established environmental mutagen to cancer through genome-wide sequencing and highlight its power to reveal individual exposure to carcinogens.
Collapse
Affiliation(s)
- Margaret L. Hoang
- Ludwig Center for Cancer Genetics and Therapeutics and the Howard Hughes Medical Institute at Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21231, USA
| | - Chung-Hsin Chen
- Department of Urology, National Taiwan University Hospital and College of Medicine, Taipei 10002, Taiwan
| | - Viktoriya S. Sidorenko
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jian He
- Ludwig Center for Cancer Genetics and Therapeutics and the Howard Hughes Medical Institute at Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21231, USA
| | - Kathleen G. Dickman
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
- Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Byeong Hwa Yun
- Division of Environmental Health Sciences, Wadsworth Center, New York State Department of Health, Empire State Plaza, Albany, NY 12201, USA
| | - Masaaki Moriya
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| | - Noushin Niknafs
- Department of Biomedical Engineering, Institute for Computational Medicine, the Johns Hopkins University, Baltimore, MD 21218, USA
| | - Christopher Douville
- Department of Biomedical Engineering, Institute for Computational Medicine, the Johns Hopkins University, Baltimore, MD 21218, USA
| | - Rachel Karchin
- Department of Biomedical Engineering, Institute for Computational Medicine, the Johns Hopkins University, Baltimore, MD 21218, USA
| | - Robert J. Turesky
- Division of Environmental Health Sciences, Wadsworth Center, New York State Department of Health, Empire State Plaza, Albany, NY 12201, USA
| | - Yeong-Shiau Pu
- Department of Urology, National Taiwan University Hospital and College of Medicine, Taipei 10002, Taiwan
| | - Bert Vogelstein
- Ludwig Center for Cancer Genetics and Therapeutics and the Howard Hughes Medical Institute at Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21231, USA
| | - Nickolas Papadopoulos
- Ludwig Center for Cancer Genetics and Therapeutics and the Howard Hughes Medical Institute at Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21231, USA
| | - Arthur P. Grollman
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
- Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Kenneth W. Kinzler
- Ludwig Center for Cancer Genetics and Therapeutics and the Howard Hughes Medical Institute at Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21231, USA
| | - Thomas A. Rosenquist
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
5519
|
Gospodinov A, Herceg Z. Chromatin structure in double strand break repair. DNA Repair (Amst) 2013; 12:800-10. [PMID: 23919923 DOI: 10.1016/j.dnarep.2013.07.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 07/11/2013] [Indexed: 12/16/2022]
Abstract
Cells are under constant assault by endogenous and environmental DNA damaging agents. DNA double strand breaks (DSBs) sever entire chromosomes and pose a major threat to genome integrity as a result of chromosomal fragment loss or chromosomal rearrangements. Exogenous factors such as ionizing radiation, crosslinking agents, and topoisomerase poisons, contribute to break formation. DSBs are associated with oxidative metabolism, form during the normal S phase, when replication forks collapse and are generated during physiological processes such as V(D)J recombination, yeast mating type switching and meiosis. It is estimated that in mammalian cells ∼10 DSBs per cell are formed daily. If left unrepaired DSBs can lead to cell death or deregulated growth, and cancer development. Cellular response to DSB damage includes mechanisms to halt the progression of the cell cycle and to restore the structure of the broken chromosome. Changes in chromatin adjacent to DNA break sites are instrumental to the DNA damage response (DDR) with two apparent ends: to control compaction and to bind repair and signaling molecules to the lesion. Here, we review the key findings related to each of these functions and examine their cross-talk.
Collapse
Affiliation(s)
- Anastas Gospodinov
- Institute of Molecular Biology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str. 21, 1113 Sofia, Bulgaria.
| | | |
Collapse
|
5520
|
Yan B, Broek RV, Saleh AD, Mehta A, Van Waes C, Chen Z. Signaling Networks of Activated Oncogenic and Altered Tumor Suppressor Genes in Head and Neck Cancer. JOURNAL OF CARCINOGENESIS & MUTAGENESIS 2013; Suppl 7:4. [PMID: 25587491 PMCID: PMC4289631 DOI: 10.4172/2157-2518.s7-004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) arises from the upper aerodigestive tract and is the six most common cancers worldwide. HNSCC is associated with high morbidity and mortality, as standard surgery, radiation, and chemotherapy can cause significant disfigurement and only provide 5-year survival rates of ~50-60%. The heterogeneity of HNSCC subsets with different potentials for recurrence and metastasis challenges the traditional pathological classification system, thereby increasing demand for the development of new diagnostic, prognostic, and therapeutic tools based on global molecular signatures of HNSCC. Historically, using classical biological techniques, it has been extremely difficult and time-consuming to survey hundreds or thousands of genes in a given disease. However, the development of high throughput technologies and high-powered computation throughout the last two decades has enabled us to investigate hundreds or thousands of genes simultaneously. Using high throughput technologies, our laboratory has identified the gene signatures and protein networks, which significantly affect HNSCC malignant phenotypes, including TP53/p63/p73 family members, IL-1/TNF-β/NF-κB, PI3K/AKT/mTOR, IL-6/IL-6R/JAK/STAT3, EGFR/MAPK/AP1, HGF/cMET/EGR1, and TGFβ/TGFβR/TAK1/SMAD pathways. This review summarizes the results from high-throughput technological assays conducted on HNSCC samples, including microarray, DNA methylation, miRNA profiling, and protein array, using primarily experimental data and conclusions generated in our own laboratory. The use of bioinformatics and integrated analyses of data sets from different platforms, as well as meta-analysis of large datasets pulled from multiple publicly available studies, provided significantly higher statistical power to extract biologically relevant information. The data suggested that the heterogeneity of HNSCC genotype and phenotype are much more complex than we previously thought. Understanding of global molecular signatures and disease classification for specific subsets of HNSCC will be essential to provide accurate diagnoses for targeted therapy and personalized treatment, which is an important effort toward improving patient outcomes.
Collapse
Affiliation(s)
- Bin Yan
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong, China
| | - Robert Vander Broek
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD USA
- NIH Medical Research Scholars Program, Bethesda, MD USA
| | - Anthony D Saleh
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD USA
| | - Arpita Mehta
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD USA
| | - Carter Van Waes
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD USA
| | - Zhong Chen
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD USA
| |
Collapse
|
5521
|
Claus J, Cameron AJM, Parker PJ. Pseudokinase drug intervention: a potentially poisoned chalice. Biochem Soc Trans 2013; 41:1083-8. [PMID: 23863183 DOI: 10.1042/bst20130078] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Pseudokinases, the catalytically impaired component of the kinome, have recently been found to share more properties with active kinases than previously thought. In many pseudokinases, ATP binding and even some activity is preserved, highlighting these proteins as potential drug targets. In both active kinases and pseudokinases, binding of ATP or drugs in the nucleotide-binding pocket can stabilize specific conformations required for activity and protein-protein interactions. We discuss the implications of locking particular conformations in a selection of (pseudo)kinases and the dual potential impact on the druggability of these proteins.
Collapse
Affiliation(s)
- Jeroen Claus
- Cancer Research UK, London Research Institute, Lincoln's Inn Fields, London WC2A 3LY, UK.
| | | | | |
Collapse
|
5522
|
Marx SJ. Multiplicity of hormone-secreting tumors: common themes about cause, expression, and management. J Clin Endocrinol Metab 2013; 98:3139-48. [PMID: 23771922 PMCID: PMC3733851 DOI: 10.1210/jc.2013-1511] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CONTEXT Multiplicity of hormone-secreting tumors occurs in a substantial portion of hormone-excess states. Multiplicity increases the difficulty of management and drives the selection of special strategies. EVIDENCE ACQUISITION This is a synthesis from publications about tumor development and expression, and also about types of clinical strategy for hormone-secreting tumors. EVIDENCE SYNTHESIS Comparisons were made between patient groups with solitary tumors vs those with multiple tumors. Major themes with clinical relevance emerged. Usually, tumor multiplicity develops from a genetic susceptibility in all cells of a tissue. This applies to hormone-secreting tumors that begin as either polyclonal (such as in the parathyroids of familial hypocalciuric hypercalcemia) or monoclonal tumors (such as in the parathyroids of multiple endocrine neoplasia type 1 [MEN1]). High penetrance of a hereditary tumor frequently results in bilaterality and in several other types of multiplicity. Managements are better for the hormone excess than for the associated cancers. Management strategies can be categorized broadly as ablation that is total, subtotal, or zero. Examples are discussed for each category, and 1 example of each category is named here: 1) total ablation of the entire tissue with effort to replace ablated functions (for example, in C-cell neoplasia of multiple endocrine neoplasia type 2); 2) subtotal ablation with increased likelihood of persistent disease or recurrent disease (for example, in the parathyroid tumors of MEN1); or 3) no ablation of tissue with or without the use of pharmacotherapy (for example, with blockers for secretion of stomach acid in gastrinomas of MEN1). CONCLUSIONS Tumor multiplicity usually arises from defects in all cells of the precursor tissue. Even the optimized managements involve compromises. Still, an understanding of pathophysiology and of therapeutic options should guide optimized management.
Collapse
Affiliation(s)
- Stephen J Marx
- Genetics and Endocrinology Section, Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA.
| |
Collapse
|
5523
|
Melanoma genotypes and phenotypes get personal. J Transl Med 2013; 93:858-67. [PMID: 23817084 DOI: 10.1038/labinvest.2013.84] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 05/28/2013] [Accepted: 06/01/2013] [Indexed: 01/23/2023] Open
Abstract
Traditionally, the diagnosis of metastatic melanoma was terminal to most patients. However, the advancements towards understanding the fundamental etiology, pathophysiology, and treatment have raised melanoma to the forefront of contemporary medicine. Indeed, the evidence of durable remissions are being heard ever more frequently in clinics around the globe. Despite having more gene mutations per cell than any other type of cancer, investigators are overcoming complex genomic landscapes, signaling pathways, and immune checkpoints by generating novel technological methods and clinical protocols with breath-taking speed. Significant progress in deciphering molecular genetics, epigenetics, kinase-driven networks, metabolomics, and immune-enhancing pathways to achieve personalized and positive outcomes has truly provided new hope for melanoma patients. However, obstacles requiring breakthroughs include understanding the influence of sunlight exposure on melanoma etiology, and overcoming all too frequently acquired drug resistance, complicating targeted therapy. Pathologists continue to have critically important roles in advancing the field, particularly in the area of transitioning from microscope-based diagnostic reports to pharmacogenomics through molecularly informed tumor boards. Although melanoma is no longer considered just 'one disease', pathologists will continue this rapidly progressing and exciting journey to identify tumor subtypes, to utilize tumorgraft or so-called patient-derived xenograft (PDX) models, and to develop companion diagnostics to keep pace with the bewildering breakthroughs occurring on a regular basis. Exactly which combination of drugs will ultimately be required to eradicate melanoma cells remains to be determined. However, it is clear that pathologists who are as dedicated to melanoma as the pioneering pathologist Dr Sidney Farber was committed to childhood cancers, will be required as the battle against melanoma continues. In this review, we describe what sets melanoma apart from other tumors, and demonstrate how lessons learned in the melanoma clinic are being transferred to many other types of aggressive neoplasms.
Collapse
|
5524
|
Workman P, Al-Lazikani B, Clarke PA. Genome-based cancer therapeutics: targets, kinase drug resistance and future strategies for precision oncology. Curr Opin Pharmacol 2013; 13:486-96. [DOI: 10.1016/j.coph.2013.06.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 06/09/2013] [Indexed: 01/09/2023]
|
5525
|
Kopelovich L, Shea-Herbert B. Heritable one-hit events defining cancer prevention? Cell Cycle 2013; 12:2553-7. [PMID: 23907126 DOI: 10.4161/cc.25690] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Over 100 years ago (1902-1914) Theodor Boveri suggested a role for mutations in cancer. Boveri's ideas were derived from the then "just-emerging" chromosome theory of inheritance. While demonstrating chromosomal aberrations as a cause of genetic imbalance, Boveri suggested that possible causes of malignancy may include events such as aneuploidy that are now defined as gene mutations, asserting all the while that malignancy occurs at the cellular level. Indeed, studies to date essentially uniformly show that cancer is a genetic disease.
Collapse
Affiliation(s)
- Levy Kopelovich
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | | |
Collapse
|
5526
|
Abstract
Generating an anti-tumor immune response is a multi-step process that is executed by effector T cells that can recognize and kill tumor targets. However, tumors employ multiple strategies to attenuate the effectiveness of T-cell-mediated attack. They achieve this by interfering with nearly every step required for effective immunity, from deregulation of antigen-presenting cells to establishment of a physical barrier at the vasculature that prevents homing of effector tumor-rejecting cells and the suppression of effector lymphocytes through the recruitment and activation of immunosuppressive cells such as myeloid-derived suppressor cells, tolerogenic monocytes, and T regulatory cells. Here, we review the ways in which tumors exert immune suppression and highlight the new therapies that seek to reverse this phenomenon and promote anti-tumor immunity. Understanding anti-tumor immunity, and how it becomes disabled by tumors, will ultimately lead to improved immune therapies and prolonged survival of patients.
Collapse
Affiliation(s)
- Greg T Motz
- Ovarian Cancer Research Center, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | |
Collapse
|
5527
|
Farrell J, Petrovics G, McLeod DG, Srivastava S. Genetic and molecular differences in prostate carcinogenesis between African American and Caucasian American men. Int J Mol Sci 2013; 14:15510-31. [PMID: 23892597 PMCID: PMC3759870 DOI: 10.3390/ijms140815510] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 07/08/2013] [Accepted: 07/10/2013] [Indexed: 12/18/2022] Open
Abstract
Prostate cancer is the most common non-skin cancer and the second leading cause of cancer-related death for men in the United States. Prostate cancer incidence and associated mortality are highest in African American men in comparison to other races. The observed differences in incidence and disease aggressiveness at presentation support a potential role for different pathways of prostate carcinogenesis between African American and Caucasian men. This review focuses on some of the recent molecular biology discoveries, which have been investigated in prostate carcinogenesis and their likely contribution to the known discrepancies across race and ethnicity. Key discussion points include the androgen receptor gene structure and function, genome-wide association studies and epigenetics. The new observations of the ethnic differences of the ERG oncogene, the most common prostate cancer gene, are providing new insights into ERG based stratification of prostate cancers in the context of ethnically diverse patient populations. This rapidly advancing knowledge has the likely potential to benefit clinical practice. Current and future work will improve the ability to sub-type prostate cancers by molecular alterations and lead to targeted therapy against this common malignancy.
Collapse
Affiliation(s)
- James Farrell
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland, 1530 E. Jefferson St., Rockville, MD 20852, USA; E-Mails: (J.F.); (G.P.); (D.G.M.)
- Urology Service, Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
| | - Gyorgy Petrovics
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland, 1530 E. Jefferson St., Rockville, MD 20852, USA; E-Mails: (J.F.); (G.P.); (D.G.M.)
| | - David G. McLeod
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland, 1530 E. Jefferson St., Rockville, MD 20852, USA; E-Mails: (J.F.); (G.P.); (D.G.M.)
- Urology Service, Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
| | - Shiv Srivastava
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland, 1530 E. Jefferson St., Rockville, MD 20852, USA; E-Mails: (J.F.); (G.P.); (D.G.M.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-240-543-8952; Fax: +1-240-453-8912
| |
Collapse
|
5528
|
|
5529
|
Evidence for APOBEC3B mutagenesis in multiple human cancers. Nat Genet 2013; 45:977-83. [PMID: 23852168 DOI: 10.1038/ng.2701] [Citation(s) in RCA: 607] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Accepted: 06/20/2013] [Indexed: 12/13/2022]
Abstract
Thousands of somatic mutations accrue in most human cancers, and their causes are largely unknown. We recently showed that the DNA cytidine deaminase APOBEC3B accounts for up to half of the mutational load in breast carcinomas expressing this enzyme. Here we address whether APOBEC3B is broadly responsible for mutagenesis in multiple tumor types. We analyzed gene expression data and mutation patterns, distributions and loads for 19 different cancer types, with over 4,800 exomes and 1,000,000 somatic mutations. Notably, APOBEC3B is upregulated, and its preferred target sequence is frequently mutated and clustered in at least six distinct cancers: bladder, cervix, lung (adenocarcinoma and squamous cell carcinoma), head and neck, and breast. Interpreting these findings in the light of previous genetic, cellular and biochemical studies, the most parsimonious conclusion from these global analyses is that APOBEC3B-catalyzed genomic uracil lesions are responsible for a large proportion of both dispersed and clustered mutations in multiple distinct cancers.
Collapse
|
5530
|
Melisi D, Piro G, Tamburrino A, Carbone C, Tortora G. Rationale and clinical use of multitargeting anticancer agents. Curr Opin Pharmacol 2013; 13:536-42. [PMID: 23856428 DOI: 10.1016/j.coph.2013.06.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 06/26/2013] [Accepted: 06/28/2013] [Indexed: 02/05/2023]
Abstract
Human solid tumors contain genetically distinct subpopulations of tumor cells that can be enriched under selective pressure of specific treatments. This heterogeneous nature reflects the dynamism of drug response and it represents a fundamental driver of resistance. Moreover, the complexity of cancer disease is increased by the activity of cross-talking, redundant signaling pathways, escape pathways and compensatory events, which triggers activation of secondary growth and survival. Broad multi-targeted approaches are requested to overcome a complex, heterogeneous, and dynamic disease such as cancer.
Collapse
Affiliation(s)
- Davide Melisi
- Digestive Molecular Clinical Oncology Research Unit, Medical Oncology, Azienda Ospedaliera Universitaria Integrata and University of Verona, Verona, Italy
| | | | | | | | | |
Collapse
|
5531
|
Challenges imposed by the complexity of cancer genome. Lancet Oncol 2013; 14:e291-2. [PMID: 23816294 DOI: 10.1016/s1470-2045(13)70224-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
5532
|
Francke U. How will genomic information become integrated into the health care system? Mol Genet Genomic Med 2013; 1:67-70. [PMID: 24498603 PMCID: PMC3865570 DOI: 10.1002/mgg3.26] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 06/11/2013] [Indexed: 11/17/2022] Open
Affiliation(s)
- Uta Francke
- Departments of Genetics and Pediatrics, Stanford University School of Medicine Stanford, California
| |
Collapse
|
5533
|
Abstract
Integration of clinical evaluations and whole-genome sequence data from eight individuals in a recent study demonstrates that genetic and clinical information can be combined and applied to preventive medicine. Statistical and graphical tools were developed to assess and visualize the genetic risk of common chronic conditions and to show the changes in disease risk that result from monitoring clinical symptoms over time. This approach provides a direction to consider in the adoption of genetic information in health care, but, like all provocative scientific articles, it raises as many questions as it answers. Please see related Research: http://genomemedicine.com/content/5/6/58
Collapse
Affiliation(s)
- Nicholas J Schork
- Department of Molecular and Experimental Medicine and the Scripps Translational Science Institute, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
5534
|
Wilson JL, Hemann MT, Fraenkel E, Lauffenburger DA. Integrated network analyses for functional genomic studies in cancer. Semin Cancer Biol 2013; 23:213-8. [PMID: 23811269 DOI: 10.1016/j.semcancer.2013.06.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Revised: 06/11/2013] [Accepted: 06/13/2013] [Indexed: 11/24/2022]
Abstract
RNA-interference (RNAi) studies hold great promise for functional investigation of the significance of genetic variations and mutations, as well as potential synthetic lethalities, for understanding and treatment of cancer, yet technical and conceptual issues currently diminish the potential power of this approach. While numerous research groups are usefully employing this kind of functional genomic methodology to identify molecular mediators of disease severity, response, and resistance to treatment, findings are generally confounded by "off-target" effects. These effects arise from a variety of issues beyond non-specific reagent behavior, such as biological cross-talk and feedback processes so thus can occur even with specific perturbation. Interpreting RNAi results in a network framework instead of merely as individual "hits" or "targets" leverages contributions from all hit/target contributions to pathways via their relationships with other network nodes. This interpretation can ameliorate dependence upon individual reagent performance and increase confidence in biological validation. Here we provide background on RNAi studies in cancer applications, review key challenges with functional genomics, and motivate the use of network models grounded in pathway analyses.
Collapse
Affiliation(s)
- Jennifer L Wilson
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, USA.
| | | | | | | |
Collapse
|
5535
|
Marquardt JU, Thorgeirsson SS. Linking MLL and the HGF-MET signaling pathway in liver cancer. J Clin Invest 2013; 123:2780-3. [PMID: 23934122 DOI: 10.1172/jci70235] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mixed-lineage leukemia (MLL; also known as myeloid/lymphoid), the human homolog of trithorax in Drosophila, is a transcriptional coactivator that plays an essential role during early development and hematopoiesis. Furthermore, MLL is critically involved in the epigenetic regulation of cell cycle, senescence, DNA damage, and stem cell self-renewal. Chromosomal aberrations of MLL in acute leukemias are well documented, but the role of this gene in solid malignancies remains unclear. In this issue of the JCI, Takeda et al. describe a novel epigenetic link between MLL and the HGF-MET signaling pathway conferring invasive and metastatic properties to hepatocellular carcinoma cells.
Collapse
Affiliation(s)
- Jens U Marquardt
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
5536
|
Affiliation(s)
- Janet D Rowley
- Department of Medicine, University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637, USA.
| |
Collapse
|
5537
|
A capture-sequencing strategy identifies IRF8, EBF1, and APRIL as novel IGH fusion partners in B-cell lymphoma. Blood 2013; 122:726-33. [PMID: 23775715 DOI: 10.1182/blood-2013-04-495804] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The characterization of immunoglobulin heavy chain (IGH) translocations provides information on the diagnosis and guides therapeutic decisions in mature B-cell malignancies while enhancing our understanding of normal and malignant B-cell biology. However, existing methodologies for the detection of IGH translocations are labor intensive, often require viable cells, and are biased toward known IGH fusions. To overcome these limitations, we developed a capture sequencing strategy for the identification of IGH rearrangements at nucleotide level resolution and tested its capabilities as a diagnostic and discovery tool in 78 primary diffuse large B-cell lymphomas (DLBCLs). We readily identified IGH-BCL2, IGH-BCL6, IGH-MYC, and IGH-CCND1 fusions and discovered IRF8, EBF1, and TNFSF13 (APRIL) as novel IGH partners in these tumors. IRF8 and TNFSF13 expression was significantly higher in lymphomas with IGH rearrangements targeting these loci. Modeling the deregulation of IRF8 and EBF1 in vitro defined a lymphomagenic profile characterized by up-regulation of AID and/or BCL6, down-regulation of PRMD1, and resistance to apoptosis. Using a capture sequencing strategy, we discovered the B-cell relevant genes IRF8, EBF1, and TNFSF13 as novel targets for IGH deregulation. This methodology is poised to change how IGH translocations are identified in clinical settings while remaining a powerful tool to uncover the pathogenesis of B-cell malignancies.
Collapse
|
5538
|
Dudek AM, Grotenhuis AJ, Vermeulen SH, Kiemeney LALM, Verhaegh GW. Urinary bladder cancer susceptibility markers. What do we know about functional mechanisms? Int J Mol Sci 2013; 14:12346-66. [PMID: 23752272 PMCID: PMC3709789 DOI: 10.3390/ijms140612346] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 05/23/2013] [Accepted: 05/30/2013] [Indexed: 12/24/2022] Open
Abstract
Genome-wide association studies (GWAS) have been successful in the identification of the several urinary bladder cancer (UBC) susceptibility loci, pointing towards novel genes involved in tumor development. Despite that, functional characterization of the identified variants remains challenging, as they mostly map to poorly understood, non-coding regions. Recently, two of the UBC risk variants (PSCA and UGT1A) were confirmed to have functional consequences. They were shown to modify bladder cancer risk by influencing gene expression in an allele-specific manner. Although the role of the other UBC risk variants is unknown, it can be hypothesized-based on studies from different cancer types-that they influence cancer susceptibility by alterations in regulatory networks. The insight into UBC heritability gained through GWAS and further functional studies can impact on cancer prevention and screening, as well as on the development of new biomarkers and future personalized therapies.
Collapse
Affiliation(s)
- Aleksandra M. Dudek
- Department of Urology, Radboud University Medical Centre, Geert Grooteplein 16, Nijmegen 6525 GA, The Netherlands; E-Mails: (L.A.L.M.K.); (G.W.V.)
- Department for Health Evidence, Radboud University Medical Centre, Geert Grooteplein 21, Nijmegen 6525 EZ, The Netherlands; E-Mails: (A.J.G.); (S.H.V.)
- Nijmegen Centre for Molecular Life Sciences, Geert Grooteplein 28, Nijmegen 6525 GA, The Netherlands
| | - Anne J. Grotenhuis
- Department for Health Evidence, Radboud University Medical Centre, Geert Grooteplein 21, Nijmegen 6525 EZ, The Netherlands; E-Mails: (A.J.G.); (S.H.V.)
- Nijmegen Centre for Evidence Based Practice, Geert Grooteplein 21, Nijmegen 6525 GA, The Netherlands
| | - Sita H. Vermeulen
- Department for Health Evidence, Radboud University Medical Centre, Geert Grooteplein 21, Nijmegen 6525 EZ, The Netherlands; E-Mails: (A.J.G.); (S.H.V.)
- Nijmegen Centre for Evidence Based Practice, Geert Grooteplein 21, Nijmegen 6525 GA, The Netherlands
| | - Lambertus A. L. M. Kiemeney
- Department of Urology, Radboud University Medical Centre, Geert Grooteplein 16, Nijmegen 6525 GA, The Netherlands; E-Mails: (L.A.L.M.K.); (G.W.V.)
- Department for Health Evidence, Radboud University Medical Centre, Geert Grooteplein 21, Nijmegen 6525 EZ, The Netherlands; E-Mails: (A.J.G.); (S.H.V.)
- Nijmegen Centre for Evidence Based Practice, Geert Grooteplein 21, Nijmegen 6525 GA, The Netherlands
| | - Gerald W. Verhaegh
- Department of Urology, Radboud University Medical Centre, Geert Grooteplein 16, Nijmegen 6525 GA, The Netherlands; E-Mails: (L.A.L.M.K.); (G.W.V.)
- Nijmegen Centre for Molecular Life Sciences, Geert Grooteplein 28, Nijmegen 6525 GA, The Netherlands
| |
Collapse
|
5539
|
Targeting protein-protein interactions as an anticancer strategy. Trends Pharmacol Sci 2013; 34:393-400. [PMID: 23725674 DOI: 10.1016/j.tips.2013.04.007] [Citation(s) in RCA: 273] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 04/23/2013] [Accepted: 04/29/2013] [Indexed: 02/07/2023]
Abstract
The emergence and convergence of cancer genomics, targeted therapies, and network oncology have significantly expanded the landscape of protein-protein interaction (PPI) networks in cancer for therapeutic discovery. Extensive biological and clinical investigations have led to the identification of protein interaction hubs and nodes that are critical for the acquisition and maintenance of characteristics of cancer essential for cell transformation. Such cancer-enabling PPIs have become promising therapeutic targets. With technological advances in PPI modulator discovery and validation of PPI-targeting agents in clinical settings, targeting of PPI interfaces as an anticancer strategy has become a reality. Future research directed at genomics-based PPI target discovery, PPI interface characterization, PPI-focused chemical library design, and patient-genomic subpopulation-driven clinical studies is expected to accelerate the development of the next generation of PPI-based anticancer agents for personalized precision medicine. Here we briefly review prominent PPIs that mediate cancer-acquired properties, highlight recognized challenges and promising clinical results in targeting PPIs, and outline emerging opportunities.
Collapse
|
5540
|
Sabaawy HE. Genetic Heterogeneity and Clonal Evolution of Tumor Cells and their Impact on Precision Cancer Medicine. ACTA ACUST UNITED AC 2013; 1:1000124. [PMID: 24558642 PMCID: PMC3927925 DOI: 10.4172/2329-6917.1000124] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The efficacy of targeted therapies in leukemias and solid tumors depends upon the accurate detection and sustained targeting of initial and evolving driver mutations and/or aberrations in cancer cells. Tumor clonal evolution of the diverse populations of cancer cells during cancer progression contributes to the longitudinal variations of clonal, morphological, anatomical, and molecular heterogeneity of tumors. Moreover, drug-resistant subclones present at initiation of therapy or emerging as a result of targeted therapies represent major challenges for achieving success of personalized therapies in providing meaningful improvement in cancer survival rates. Here, I briefly portray tumor cell clonal evolution at the cellular and molecular levels, and present the multiple types of genetic heterogeneity in tumors, with a focus on their impact on the implementation of personalized or precision cancer medicine.
Collapse
Affiliation(s)
- Hatem E Sabaawy
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, USA ; Department of Cellular and Molecular Pharmacology, Rutgers-Robert Wood Johnson Medical School, USA ; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903-2681, USA
| |
Collapse
|
5541
|
Frye SV. Drug discovery in academic institutions. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2013; 2013:300-305. [PMID: 24319195 DOI: 10.1182/asheducation-2013.1.300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Although academic science has always provided a fundamental understanding of the biological and clinical basis of disease, the opportunity and imperative for academics to contribute more directly to the discovery of new medicines continues to grow. Embedding medicinal chemists with cancer biologists creates collaborative opportunities for drug discovery and the design and synthesis of chemical biology tool compounds (chemical probes) to better elucidate the role of specific proteins and pathways in biology and disease. Two case studies are presented here: (1) the discovery of inhibitors of mer kinase to treat acute lymphoblastic leukemia in children and (2) the discovery of chemical probes targeting epigenetic regulators. These case studies provide lessons in target selection strategies, the requirement for iterative optimization of lead compounds (useful drugs/probes rarely come directly from a screen), and the value of mutually dependent collaborations between medicinal chemists and cancer biologists.
Collapse
Affiliation(s)
- Stephen V Frye
- 1Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|