1
|
Gingrich AA, Razmara AM, Gingrich PW, Rebhun RB, Murphy WJ, Kent MS, Brown CT, Siegel JB, Canter RJ. Missing a "Missing Self" Mechanism: Modeling and Detection of Ly49 Expression in Canine NK Cells. Immunohorizons 2023; 7:760-770. [PMID: 37971282 PMCID: PMC10696421 DOI: 10.4049/immunohorizons.2300092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/19/2023] Open
Abstract
NK cells are a key focus in immuno-oncology, based on their ability to eliminate malignant cells without prior sensitization. Dogs are valuable models for translational immunotherapy studies, especially for NK cells, where critical species differences exist between mice and humans. Given that the mechanism for recognition of "self" by canine NK cells is currently unknown, we sought to evaluate expression of Ly49 in canine NK cells using in silico and high-throughput techniques. We interrogated the identified polymorphism/mutation in canine Ly49 and assessed the potential impact on structure using computational modeling of three-dimensional protein structure and protein-protein docking of canine Ly49 with MHC class I (MHC-I). Bulk and single-cell RNA-sequencing analysis was performed to detect gene expression of Ly49/KLRA1 in resting and activated NK cells. Tertiary protein structure demonstrated significant structural similarity to the known murine system. Molecular docking of canine Ly49 with MHC-I was favorable, converging at a single low-energy conformation. RNA sequencing revealed expression of Ly49/KLRA1 in both resting and activated NK cells and demonstrated almost exclusive expression of the gene in the NK cluster at the single-cell level. Despite prior reports of a mutated, nonfunctional canine Ly49, our data support that the protein product is predicted to bind to MHC-I in a comparable conformation to the murine system and is expressed in canine NK cells with upregulation following activation. Taken together, these data suggest that Ly49 is capable of recognizing MHC-I and therefore regulating NK cell function in dogs.
Collapse
Affiliation(s)
- Alicia A. Gingrich
- Department of Surgery, University of California, Davis School of Medicine, Sacramento, CA
| | - Aryana M. Razmara
- Department of Surgery, University of California, Davis School of Medicine, Sacramento, CA
| | - Phillip W. Gingrich
- Department of Biochemistry and Molecular Medicine, University of California, Davis School of Medicine, Sacramento, CA
| | - Robert B. Rebhun
- Department of Surgical and Radiological Sciences, University of California, Davis School of Veterinary Medicine, Davis, CA
| | - William J. Murphy
- Department of Dermatology, University of California, Davis School of Medicine, Sacramento, CA
| | - Michael S. Kent
- Department of Surgical and Radiological Sciences, University of California, Davis School of Veterinary Medicine, Davis, CA
| | - C. Titus Brown
- Department of Population Health and Reproduction, University of California, Davis School of Veterinary Medicine, Davis, CA
| | - Justin B. Siegel
- Department of Biochemistry and Molecular Medicine, University of California, Davis School of Medicine, Sacramento, CA
| | - Robert J. Canter
- Department of Surgery, University of California, Davis School of Medicine, Sacramento, CA
| |
Collapse
|
2
|
Proksch SF, Matthysen CP, Jardine JE, Wyatt KM, Finlay JR, Nelson DJ. Developing a translational murine-to-canine pathway for an IL-2/agonist anti-CD40 antibody cancer immunotherapy. Vet Comp Oncol 2022; 20:602-612. [PMID: 35315197 PMCID: PMC9540797 DOI: 10.1111/vco.12813] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 03/14/2022] [Accepted: 03/14/2022] [Indexed: 11/29/2022]
Abstract
Human and canine sarcomas are difficult to treat soft tissue malignancies with an urgent need for new improved therapeutic options. Local recurrence rates for humans are between 10%-30%, and 30%-40% develop metastases. Outcomes for dogs with sarcoma vary with grade but can be similar. Pet dogs share the human environment and represent human cancer with genetic variation in hosts and tumours. We asked if our murine studies using genetically identical mice and cloned tumour cells were translatable to larger, genetically diverse domestic dogs with naturally occurring tumours, to (i) develop a canine cancer therapeutic, and (ii) to use as a translational pathway to humans. Our murine studies showed that intra-tumoral delivery of interleukin-2 (IL-2) plus an agonist anti-CD40 antibody (Ab) induces long-term curative responses ranging from 30% to 100%, depending on tumour type. We developed an agonist anti-canine-CD40 Ab and conducted a phase I dose finding/toxicology 3 + 3 clinical trial in dogs (n = 27) with soft tissue sarcomas on account of suitability for intratumoral injection and straightforward monitoring. Dogs were treated with IL-2 plus anti-CD40 antibody for 2 weeks. Three dose levels induced tumour regression with minimal side effects, measured by monitoring, haematological and biochemical assays. Importantly, our mouse and canine studies provide encouraging fundamental proof-of-concept data upon which we can develop veterinary and human immunotherapeutic strategies.
Collapse
Affiliation(s)
- Stephen Francis Proksch
- Curtin Medical SchoolCurtin UniversityBentleyWestern AustraliaAustralia
- CHIRI BiosciencesCurtin UniversityBentleyWestern AustraliaAustralia
- Selvax Pty LtdWest PerthWestern AustraliaAustralia
| | - Clinton Petrus Matthysen
- Curtin Medical SchoolCurtin UniversityBentleyWestern AustraliaAustralia
- CHIRI BiosciencesCurtin UniversityBentleyWestern AustraliaAustralia
| | | | - Ken Mark Wyatt
- Perth Veterinary Specialists (PVS)Osborne ParkWestern AustraliaAustralia
| | | | - Delia Jane Nelson
- Curtin Medical SchoolCurtin UniversityBentleyWestern AustraliaAustralia
- CHIRI BiosciencesCurtin UniversityBentleyWestern AustraliaAustralia
| |
Collapse
|
3
|
Peart CR, Williams C, Pophaly SD, Neely BA, Gulland FMD, Adams DJ, Ng BL, Cheng W, Goebel ME, Fedrigo O, Haase B, Mountcastle J, Fungtammasan A, Formenti G, Collins J, Wood J, Sims Y, Torrance J, Tracey A, Howe K, Rhie A, Hoffman JI, Johnson J, Jarvis ED, Breen M, Wolf JBW. Hi-C scaffolded short- and long-read genome assemblies of the California sea lion are broadly consistent for syntenic inference across 45 million years of evolution. Mol Ecol Resour 2021; 21:2455-2470. [PMID: 34097816 PMCID: PMC9732816 DOI: 10.1111/1755-0998.13443] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/06/2021] [Accepted: 05/26/2021] [Indexed: 12/13/2022]
Abstract
With the advent of chromatin-interaction maps, chromosome-level genome assemblies have become a reality for a wide range of organisms. Scaffolding quality is, however, difficult to judge. To explore this gap, we generated multiple chromosome-scale genome assemblies of an emerging wild animal model for carcinogenesis, the California sea lion (Zalophus californianus). Short-read assemblies were scaffolded with two independent chromatin interaction mapping data sets (Hi-C and Chicago), and long-read assemblies with three data types (Hi-C, optical maps and 10X linked reads) following the "Vertebrate Genomes Project (VGP)" pipeline. In both approaches, 18 major scaffolds recovered the karyotype (2n = 36), with scaffold N50s of 138 and 147 Mb, respectively. Synteny relationships at the chromosome level with other pinniped genomes (2n = 32-36), ferret (2n = 34), red panda (2n = 36) and domestic dog (2n = 78) were consistent across approaches and recovered known fissions and fusions. Comparative chromosome painting and multicolour chromosome tiling with a panel of 264 genome-integrated single-locus canine bacterial artificial chromosome probes provided independent evaluation of genome organization. Broad-scale discrepancies between the approaches were observed within chromosomes, most commonly in translocations centred around centromeres and telomeres, which were better resolved in the VGP assembly. Genomic and cytological approaches agreed on near-perfect synteny of the X chromosome, and in combination allowed detailed investigation of autosomal rearrangements between dog and sea lion. This study presents high-quality genomes of an emerging cancer model and highlights that even highly fragmented short-read assemblies scaffolded with Hi-C can yield reliable chromosome-level scaffolds suitable for comparative genomic analyses.
Collapse
Affiliation(s)
- Claire R. Peart
- Division of Evolutionary Biology, Faculty of Biology, LMU Munich, Munchen, Germany
| | - Christina Williams
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Saurabh D. Pophaly
- Division of Evolutionary Biology, Faculty of Biology, LMU Munich, Munchen, Germany
- Max Planck institute for Plant Breeding Research, Cologne, Germany
| | - Benjamin A. Neely
- National Institute of Standards and Technology, NIST Charleston, Charleston, South Carolina, USA
| | - Frances M. D. Gulland
- Karen Dryer Wildlife Health Center, University of California Davis, Davis, California, USA
| | - David J. Adams
- Cytometry Core Facility, Wellcome Sanger Institute, Cambridge, UK
| | - Bee Ling Ng
- Cytometry Core Facility, Wellcome Sanger Institute, Cambridge, UK
| | - William Cheng
- Cytometry Core Facility, Wellcome Sanger Institute, Cambridge, UK
| | - Michael E. Goebel
- Institute of Marine Science, University of California Santa Cruz, Santa Cruz, California, USA
| | - Olivier Fedrigo
- Vertebrate Genome Lab, The Rockefeller University, New York City, New York, USA
| | - Bettina Haase
- Vertebrate Genome Lab, The Rockefeller University, New York City, New York, USA
| | | | | | - Giulio Formenti
- Vertebrate Genome Lab, The Rockefeller University, New York City, New York, USA
- Laboratory of Neurogenetics of Language, The Rockefeller University, New York City, New York, USA
| | - Joanna Collins
- Tree of Life Programme, Wellcome Sanger Institute, Cambridge, UK
| | - Jonathan Wood
- Tree of Life Programme, Wellcome Sanger Institute, Cambridge, UK
| | - Ying Sims
- Tree of Life Programme, Wellcome Sanger Institute, Cambridge, UK
| | - James Torrance
- Tree of Life Programme, Wellcome Sanger Institute, Cambridge, UK
| | - Alan Tracey
- Tree of Life Programme, Wellcome Sanger Institute, Cambridge, UK
| | - Kerstin Howe
- Tree of Life Programme, Wellcome Sanger Institute, Cambridge, UK
| | - Arang Rhie
- Genome Informatics Section, Computational and Statistical Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, USA
| | - Joseph I. Hoffman
- Department of Animal Behaviour, Bielefeld University, Bielefeld, Germany
- British Antarctic Survey, Cambridge, UK
| | - Jeremy Johnson
- Broad Institute of Harvard and Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA
| | - Erich D. Jarvis
- Vertebrate Genome Lab, The Rockefeller University, New York City, New York, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Matthew Breen
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Jochen B. W. Wolf
- Division of Evolutionary Biology, Faculty of Biology, LMU Munich, Munchen, Germany
| |
Collapse
|
4
|
Krane GA, O'Dea CA, Malarkey DE, Miller AD, Miller CR, Tokarz DA, Jensen HL, Janardhan KS, Shockley KR, Flagler N, Rainess BA, Mariani CL. Immunohistochemical evaluation of immune cell infiltration in canine gliomas. Vet Pathol 2021; 58:952-963. [PMID: 34196247 PMCID: PMC11404454 DOI: 10.1177/03009858211023946] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Evasion of the immune response is an integral part of the pathogenesis of glioma. In humans, important mechanisms of immune evasion include recruitment of regulatory T cells (Tregs) and polarization of macrophages toward an M2 phenotype. Canine glioma has a robust immune cell infiltrate that has not been extensively characterized. The purpose of this study was to determine the distribution of immune cells infiltrating spontaneous intracranial canine gliomas. Seventy-three formalin-fixed, paraffin-embedded tumor samples were evaluated using immunohistochemistry for CD3, forkhead box 3 (FOXP3), CD20, Iba1, calprotectin (Mac387), CD163, and indoleamine 2,3-dioxygenase (IDO). Immune cell infiltration was present in all tumors. Low-grade and high-grade gliomas significantly differed in the numbers of FoxP3+ cells, Mac387+ cells, and CD163+ cells (P = .006, .01, and .01, respectively). Considering all tumors, there was a significant increase in tumor area fraction of CD163 compared to Mac387 (P < .0001), and this ratio was greater in high-grade tumors than in low-grade tumors (P = .005). These data warrant further exploration into the roles of macrophage repolarization or Treg interference therapy in canine glioma.
Collapse
Affiliation(s)
- Gregory A Krane
- 6857National Institute of Environmental Health Sciences, National Toxicology Program, Cellular and Molecular Pathology Branch, Research Triangle Park, NC, USA
- Department of Clinical Sciences, North Carolina State University, Raleigh, NC, USA
- Comparative Neuroimmunology and Neuro-Oncology Laboratory, North Carolina State University, Raleigh, NC, USA
| | | | - David E Malarkey
- 6857National Institute of Environmental Health Sciences, National Toxicology Program, Cellular and Molecular Pathology Branch, Research Triangle Park, NC, USA
| | | | | | - Debra A Tokarz
- Experimental Pathology Laboratories Inc, Research Triangle Park, NC, USA
| | - Heather L Jensen
- 6857National Institute of Environmental Health Sciences, National Toxicology Program, Cellular and Molecular Pathology Branch, Research Triangle Park, NC, USA
| | | | - Keith R Shockley
- Division of Intramural Research, Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Norris Flagler
- 6857National Institute of Environmental Health Sciences, National Toxicology Program, Cellular and Molecular Pathology Branch, Research Triangle Park, NC, USA
| | - Brittani A Rainess
- Comparative Neuroimmunology and Neuro-Oncology Laboratory, North Carolina State University, Raleigh, NC, USA
| | - Christopher L Mariani
- Comparative Neuroimmunology and Neuro-Oncology Laboratory, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
5
|
Tawa GJ, Braisted J, Gerhold D, Grewal G, Mazcko C, Breen M, Sittampalam G, LeBlanc AK. Transcriptomic profiling in canines and humans reveals cancer specific gene modules and biological mechanisms common to both species. PLoS Comput Biol 2021; 17:e1009450. [PMID: 34570764 PMCID: PMC8523068 DOI: 10.1371/journal.pcbi.1009450] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 10/18/2021] [Accepted: 09/14/2021] [Indexed: 12/25/2022] Open
Abstract
Understanding relationships between spontaneous cancer in companion (pet) canines and humans can facilitate biomarker and drug development in both species. Towards this end we developed an experimental-bioinformatic protocol that analyzes canine transcriptomics data in the context of existing human data to evaluate comparative relevance of canine to human cancer. We used this protocol to characterize five canine cancers: melanoma, osteosarcoma, pulmonary carcinoma, B- and T-cell lymphoma, in 60 dogs. We applied an unsupervised, iterative clustering method that yielded five co-expression modules and found that each cancer exhibited a unique module expression profile. We constructed cancer models based on the co-expression modules and used the models to successfully classify the canine data. These canine-derived models also successfully classified human tumors representing the same cancers, indicating shared cancer biology between canines and humans. Annotation of the module genes identified cancer specific pathways relevant to cells-of-origin and tumor biology. For example, annotations associated with melanin production (PMEL, GPNMB, and BACE2), synthesis of bone material (COL5A2, COL6A3, and COL12A1), synthesis of pulmonary surfactant (CTSH, LPCAT1, and NAPSA), ribosomal proteins (RPL8, RPS7, and RPLP0), and epigenetic regulation (EDEM1, PTK2B, and JAK1) were unique to melanoma, osteosarcoma, pulmonary carcinoma, B- and T-cell lymphoma, respectively. In total, 152 biomarker candidates were selected from highly expressing modules for each cancer type. Many of these biomarker candidates are under-explored as drug discovery targets and warrant further study. The demonstrated transferability of classification models from canines to humans enforces the idea that tumor biology, biomarker targets, and associated therapeutics, discovered in canines, may translate to human medicine.
Collapse
Affiliation(s)
- Gregory J. Tawa
- National Institutes of Health, National Center for Advancing Translational Sciences, Division of Preclinical Innovation, Therapeutic Development Branch, Rockville, Maryland, United States of America
| | - John Braisted
- National Institutes of Health, National Center for Advancing Translational Sciences, Division of Preclinical Innovation, Therapeutic Development Branch, Rockville, Maryland, United States of America
| | - David Gerhold
- National Institutes of Health, National Center for Advancing Translational Sciences, Division of Preclinical Innovation, Therapeutic Development Branch, Rockville, Maryland, United States of America
| | - Gurmit Grewal
- National Institutes of Health, National Center for Advancing Translational Sciences, Division of Preclinical Innovation, Therapeutic Development Branch, Rockville, Maryland, United States of America
| | - Christina Mazcko
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Comparative Oncology Program, Bethesda, Maryland, United States of America
| | - Matthew Breen
- Department of Molecular Biomedical Sciences, North Carolina State University, College of Veterinary Medicine, Raleigh, North Carolina, United States of America
| | - Gurusingham Sittampalam
- National Institutes of Health, National Center for Advancing Translational Sciences, Division of Preclinical Innovation, Therapeutic Development Branch, Rockville, Maryland, United States of America
| | - Amy K. LeBlanc
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Comparative Oncology Program, Bethesda, Maryland, United States of America
| |
Collapse
|
6
|
Gross C, Ramirez DA, McGrath S, Gustafson DL. Cannabidiol Induces Apoptosis and Perturbs Mitochondrial Function in Human and Canine Glioma Cells. Front Pharmacol 2021; 12:725136. [PMID: 34456736 PMCID: PMC8385407 DOI: 10.3389/fphar.2021.725136] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 07/31/2021] [Indexed: 11/13/2022] Open
Abstract
Cannabidiol (CBD), the major non-psychoactive compound found in cannabis, is frequently used both as a nutraceutical and therapeutic. Despite anecdotal evidence as an anticancer agent, little is known about the effect CBD has on cancer cells. Given the intractability and poor prognoses of brain cancers in human and veterinary medicine, we sought to characterize the in vitro cytotoxicity of CBD on human and canine gliomas. Glioma cells treated with CBD showed a range of cytotoxicity from 4.9 to 8.2 μg/ml; canine cells appeared to be more sensitive than human. Treatment with >5 μg/ml CBD invariably produced large cytosolic vesicles. The mode of cell death was then interrogated using pharmacologic inhibitors. Inhibition of apoptosis was sufficient to rescue CBD-mediated cytotoxicity. Inhibition of RIPK3, a classical necroptosis kinase, also rescued cells from death and prevented the formation of the large cytosolic vesicles. Next, cellular mitochondrial activity in the presence of CBD was assessed and within 2 hours of treatment CBD reduced oxygen consumption in a dose dependent manner with almost complete ablation of activity at 10 μg/ml CBD. Fluorescent imaging with a mitochondrial-specific dye revealed that the large cytosolic vesicles were, in fact, swollen mitochondria. Lastly, calcium channels were pharmacologically inhibited and the effect on cell death was determined. Inhibition of mitochondrial channel VDAC1, but not the TRPV1 channel, rescued cells from CBD-mediated cytotoxicity. These results demonstrate the cytotoxic nature of CBD in human and canine glioma cells and suggest a mechanism of action involving dysregulation of calcium homeostasis and mitochondrial activity.
Collapse
Affiliation(s)
- Chase Gross
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Dominique A Ramirez
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Stephanie McGrath
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Daniel L Gustafson
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, United States.,University of Colorado Cancer Center, Aurora, CO, United States
| |
Collapse
|
7
|
José‐López R, Gutierrez‐Quintana R, de la Fuente C, Manzanilla EG, Suñol A, Pi Castro D, Añor S, Sánchez‐Masian D, Fernández‐Flores F, Ricci E, Marioni‐Henry K, Mascort J, Matiasek LA, Matiasek K, Brennan PM, Pumarola M. Clinical features, diagnosis, and survival analysis of dogs with glioma. J Vet Intern Med 2021; 35:1902-1917. [PMID: 34117807 PMCID: PMC8295679 DOI: 10.1111/jvim.16199] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Gliomas in dogs remain poorly understood. OBJECTIVES To characterize the clinicopathologic findings, diagnostic imaging features and survival of a large sample of dogs with glioma using the Comparative Brain Tumor Consortium diagnostic classification. ANIMALS Ninety-one dogs with histopathological diagnosis of glioma. METHODS Multicentric retrospective case series. Signalment, clinicopathologic findings, diagnostic imaging characteristics, treatment, and outcome were used. Tumors were reclassified according to the new canine glioma diagnostic scheme. RESULTS No associations were found between clinicopathologic findings or survival and tumor type or grade. However, definitive treatments provided significantly (P = .03) improved median survival time (84 days; 95% confidence interval [CI], 45-190) compared to palliative treatment (26 days; 95% CI, 11-54). On magnetic resonance imaging (MRI), oligodendrogliomas were associated with smooth margins and T1-weighted hypointensity compared to astrocytomas (odds ratio [OR], 42.5; 95% CI, 2.42-744.97; P = .04; OR, 45.5; 95% CI, 5.78-333.33; P < .001, respectively) and undefined gliomas (OR, 84; 95% CI, 3.43-999.99; P = .02; OR, 32.3; 95% CI, 2.51-500.00; P = .008, respectively) and were more commonly in contact with the ventricles than astrocytomas (OR, 7.47; 95% CI, 1.03-53.95; P = .049). Tumor spread to neighboring brain structures was associated with high-grade glioma (OR, 6.02; 95% CI, 1.06-34.48; P = .04). CONCLUSIONS AND CLINICAL IMPORTANCE Dogs with gliomas have poor outcomes, but risk factors identified in survival analysis inform prognosis and the newly identified MRI characteristics could refine diagnosis of tumor type and grade.
Collapse
Affiliation(s)
- Roberto José‐López
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgowUK
- Department of Animal Medicine and Surgery, Veterinary FacultyUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Rodrigo Gutierrez‐Quintana
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgowUK
| | - Cristian de la Fuente
- Department of Animal Medicine and Surgery, Veterinary FacultyUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Edgar G. Manzanilla
- School of Veterinary Medicine, University College DublinDublinIreland
- TEAGASC, The Irish Food and Agriculture AuthorityCorkIreland
| | - Anna Suñol
- ARS VeterinariaBarcelonaSpain
- Present address:
Royal (Dick) School of Veterinary Studies, University of EdinburghEdinburghUK
| | - Dolors Pi Castro
- Department of Animal Medicine and Surgery, Veterinary FacultyUniversitat Autònoma de BarcelonaBarcelonaSpain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER‐BBN), Universitat Autònoma de BarcelonaBarcelonaSpain
- Present address:
Anicura Arvivet Hospital VeterinariBarcelonaSpain
| | - Sonia Añor
- Department of Animal Medicine and Surgery, Veterinary FacultyUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Daniel Sánchez‐Masian
- Institute of Veterinary Science, University of LiverpoolNestonUK
- Present address:
Anderson Moores Veterinary SpecialistsWinchesterUK
| | | | - Emanuele Ricci
- Institute of Veterinary Science, University of LiverpoolNestonUK
| | - Katia Marioni‐Henry
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of EdinburghEdinburghUK
| | | | - Lara A. Matiasek
- Tierklinik HaarHaarGermany
- Present address:
Anicura Small Animal ClinicBabenhausenGermany
| | - Kaspar Matiasek
- Centre for Clinical Veterinary Medicine, Ludwig‐Maximilians‐UniversitaetMunichGermany
| | - Paul M. Brennan
- Translational Neurosurgery, Centre for Clinical Brain Sciences, University of EdinburghEdinburghUK
| | - Martí Pumarola
- Department of Animal Medicine and Surgery, Veterinary FacultyUniversitat Autònoma de BarcelonaBarcelonaSpain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER‐BBN), Universitat Autònoma de BarcelonaBarcelonaSpain
| |
Collapse
|
8
|
Kent M, Song RB, Glass EN, Warren JD, de Lahunta A, Miller A. Pathology in Practice. J Am Vet Med Assoc 2021; 257:1241-1244. [PMID: 33269964 DOI: 10.2460/javma.257.12.1241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
9
|
Abstract
Comparative oncology clinical trials play an important and growing role in cancer research and drug development efforts. These trials, typically conducted in companion (pet) dogs, allow assessment of novel anticancer agents and combination therapies in a veterinary clinical setting that supports serial biologic sample collections and exploration of dose, schedule and corresponding pharmacokinetic/pharmacodynamic relationships. Further, an intact immune system and natural co-evolution of tumour and microenvironment support exploration of novel immunotherapeutic strategies. Substantial improvements in our collective understanding of the molecular landscape of canine cancers have occurred in the past 10 years, facilitating translational research and supporting the inclusion of comparative studies in drug development. The value of the approach is demonstrated in various clinical trial settings, including single-agent or combination response rates, inhibition of metastatic progression and randomized comparison of multiple agents in a head-to-head fashion. Such comparative oncology studies have been purposefully included in the developmental plan for several US FDA-approved and up-and-coming anticancer drugs. Challenges for this field include keeping pace with technology and data dissemination/harmonization, improving annotation of the canine genome and immune system, and generation of canine-specific validated reagents to support integration of correlative biology within clinical trial efforts.
Collapse
Affiliation(s)
- Amy K LeBlanc
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Christina N Mazcko
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
10
|
Overgaard NH, Fan TM, Schachtschneider KM, Principe DR, Schook LB, Jungersen G. Of Mice, Dogs, Pigs, and Men: Choosing the Appropriate Model for Immuno-Oncology Research. ILAR J 2019; 59:247-262. [PMID: 30476148 DOI: 10.1093/ilar/ily014] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 07/30/2018] [Indexed: 02/06/2023] Open
Abstract
The immune system plays dual roles in response to cancer. The host immune system protects against tumor formation via immunosurveillance; however, recognition of the tumor by immune cells also induces sculpting mechanisms leading to a Darwinian selection of tumor cell variants with reduced immunogenicity. Cancer immunoediting is the concept used to describe the complex interplay between tumor cells and the immune system. This concept, commonly referred to as the three E's, is encompassed by 3 distinct phases of elimination, equilibrium, and escape. Despite impressive results in the clinic, cancer immunotherapy still has room for improvement as many patients remain unresponsive to therapy. Moreover, many of the preclinical results obtained in the widely used mouse models of cancer are lost in translation to human patients. To improve the success rate of immuno-oncology research and preclinical testing of immune-based anticancer therapies, using alternative animal models more closely related to humans is a promising approach. Here, we describe 2 of the major alternative model systems: canine (spontaneous) and porcine (experimental) cancer models. Although dogs display a high rate of spontaneous tumor formation, an increased number of genetically modified porcine models exist. We suggest that the optimal immuno-oncology model may depend on the stage of cancer immunoediting in question. In particular, the spontaneous canine tumor models provide a unique platform for evaluating therapies aimed at the escape phase of cancer, while genetically engineered swine allow for elucidation of tumor-immune cell interactions especially during the phases of elimination and equilibrium.
Collapse
Affiliation(s)
- Nana H Overgaard
- Department of Micro- and Nanotechnology, Technical University of Denmark, Kgs Lyngby, Denmark
| | - Timothy M Fan
- Department of Veterinary Clinical Medicine, University of Illinois, Urbana-Champaign, Illinois
| | | | - Daniel R Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, Illinois
| | - Lawrence B Schook
- Department of Radiology, University of Illinois, Chicago, Illinois.,Department of Animal Sciences, University of Illinois, Urbana-Champaign, Illinois
| | - Gregers Jungersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
11
|
Koehler JW, Miller AD, Miller CR, Porter B, Aldape K, Beck J, Brat D, Cornax I, Corps K, Frank C, Giannini C, Horbinski C, Huse JT, O'Sullivan MG, Rissi DR, Mark Simpson R, Woolard K, Shih JH, Mazcko C, Gilbert MR, LeBlanc AK. A Revised Diagnostic Classification of Canine Glioma: Towards Validation of the Canine Glioma Patient as a Naturally Occurring Preclinical Model for Human Glioma. J Neuropathol Exp Neurol 2019; 77:1039-1054. [PMID: 30239918 DOI: 10.1093/jnen/nly085] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The National Cancer Institute-led multidisciplinary Comparative Brain Tumor Consortium (CBTC) convened a glioma pathology board, comprising both veterinarian and physician neuropathologists, and conducted a comprehensive review of 193 cases of canine glioma. The immediate goal was to improve existing glioma classification methods through creation of a histologic atlas of features, thus yielding greater harmonization of phenotypic characterization. The long-term goal was to support future incorporation of clinical outcomes and genomic data into proposed simplified diagnostic schema, so as to further bridge the worlds of veterinary and physician neuropathology and strengthen validity of the dog as a naturally occurring, translationally relevant animal model of human glioma. All cases were morphologically reclassified according to a new schema devised by the entire board, yielding a majority opinion diagnosis of astrocytoma (43, 22.3%), 19 of which were low-grade and 24 high-grade, and oligodendroglioma (134, 69.4%), 35 of which were low-grade and 99 were high-grade. Sixteen cases (8.3%) could not be classified as oligodendroglioma or astrocytoma based on morphology alone and were designated as undefined gliomas. The simplified classification scheme proposed herein provides a tractable means for future addition of molecular data, and also serves to highlight histologic similarities and differences between human and canine glioma.
Collapse
Affiliation(s)
- Jennifer W Koehler
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Andrew D Miller
- Department of Biomedical Sciences, Section of Anatomic Pathology, College of Veterinary Medicine, Cornell University, Ithaca, New York
| | - C Ryan Miller
- Department of Pathology and Laboratory Medicine.,Department of Neurology.,Department of Pharmacology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Brian Porter
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jessica Beck
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Daniel Brat
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Ingrid Cornax
- Department of Pediatrics, University of California-San Diego, San Diego California
| | - Kara Corps
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Viral Immunology and Intravital Imaging Section, Bethesda, Maryland
| | - Chad Frank
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, Colorado
| | - Caterina Giannini
- Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Craig Horbinski
- Department of Pathology.,Department of Neurosurgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jason T Huse
- Departments of Pathology and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - M Gerard O'Sullivan
- Masonic Cancer Center Comparative Pathology Shared Resource and Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
| | - Daniel R Rissi
- Department of Pathology and Athens Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Georgia, Athens, Georgia
| | - R Mark Simpson
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Molecular Pathology Unit, Laboratory of Cancer Biology and Genetics, Bethesda, Maryland
| | - Kevin Woolard
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California-Davis, Davis, California
| | - Joanna H Shih
- Biometrics Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Christina Mazcko
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mark R Gilbert
- National Institute of Neurological Disorders and Stroke and the Center for Cancer Research, National Cancer Institute, National Institutes of Health, NeuroOncology Branch, Bethesda, Maryland
| | - Amy K LeBlanc
- National Cancer Institute, National Institutes of Health, Comparative Oncology Program, Center for Cancer Research, Bethesda, Maryland
| |
Collapse
|
12
|
Mitchell D, Chintala S, Fetcko K, Henriquez M, Tewari BN, Ahmed A, Bentley RT, Dey M. Common Molecular Alterations in Canine Oligodendroglioma and Human Malignant Gliomas and Potential Novel Therapeutic Targets. Front Oncol 2019; 9:780. [PMID: 31475119 PMCID: PMC6702544 DOI: 10.3389/fonc.2019.00780] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/31/2019] [Indexed: 01/05/2023] Open
Abstract
Spontaneous canine (Canis lupus) oligodendroglioma (ODG) holds tremendous potential as an immunocompetent large animal model of human malignant gliomas (MG). However, the feasibility of utilizing this model in pre-clinical studies depends on a thorough understanding of the similarities and differences of the molecular pathways associated with gliomas between the two species. We have previously shown that canine ODG has an immune landscape and expression pattern of commonly described oncogenes similar to that of human MG. In the current study, we performed a comprehensive analysis of canine ODG RNAseq data from 4 dogs with ODG and 2 normal controls to identify highly dysregulated genes in canine tumors. We then evaluated the expression of these genes in human MG using Xena Browser, a publicly available database. STRING-database inquiry was used in order to determine the suggested protein associations of these differentially expressed genes as well as the dysregulated pathways commonly enriched by the protein products of these genes in both canine ODG and human MG. Our results revealed that 3,712 (23%) of the 15,895 differentially expressed genes demonstrated significant up- or downregulation (log2-fold change > 2.0). Of the 3,712 altered genes, ~50% were upregulated (n = 1858) and ~50% were downregulated (n = 1854). Most of these genes were also found to have altered expression in human MG. Protein association and pathway analysis revealed common pathways enriched by members of the up- and downregulated gene categories in both species. In summary, we demonstrate that a similar pattern of gene dysregulation characterizes both human MG and canine ODG and provide additional support for the use of the canine model in order to therapeutically target these common genes. The results of such therapeutic targeting in the canine model can serve to more accurately predict the efficacy of anti-glioma therapies in human patients.
Collapse
Affiliation(s)
- Dana Mitchell
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sreenivasulu Chintala
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Kaleigh Fetcko
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Mario Henriquez
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Brij N Tewari
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Atique Ahmed
- Department of Neurological Surgery, Northwestern University, Chicago, IL, United States
| | - R Timothy Bentley
- Department of Veterinary Clinical Sciences, Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Mahua Dey
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
13
|
Fan TM, Selting KA. Exploring the Potential Utility of Pet Dogs With Cancer for Studying Radiation-Induced Immunogenic Cell Death Strategies. Front Oncol 2019; 8:680. [PMID: 30697532 PMCID: PMC6340932 DOI: 10.3389/fonc.2018.00680] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/27/2018] [Indexed: 01/21/2023] Open
Abstract
Radiotherapy serves as a foundational pillar for the therapeutic management of diverse solid tumors through the generation of lethal DNA damage and induction of cell death. While the direct cytotoxic effects of radiation therapy remain a cornerstone for cancer management, in the era of immunooncology there is renewed and focused interest in exploiting the indirect bystander activities of radiation, termed abscopal effects. In radioimmunobiologic terms, abscopal effects describe the radiotherapy-induced regression of cancerous lesions distant from the primary site of radiation delivery and rely upon the induction of immunogenic cell death and consequent systemic anticancer immune activation. Despite the promise of radiation therapy for awaking potent anticancer immune responses, the purposeful harnessing of abscopal effects with radiotherapy remain clinically elusive. In part, failure to fully leverage and clinically implement the promise of radiation-induced abscopal effects stems from limitations associated with existing conventional tumor models which inadequately recapitulate the complexity of malignant transformation and the dynamic nature of tumor immune surveillance. To supplement this existing gap in modeling systems, pet dogs diagnosed with solid tumors including melanoma and osteosarcoma, which are both metastatic and immunogenic in nature, could potentially serve as unique resources for exploring the fundamental underpinnings required for maximizing radiation-induced abscopal effects. Given the spontaneous course of cancer development in the context of operative immune mechanisms, pet dogs treated with radiotherapy for metastatic solid tumors might be leveraged as valuable model systems for realizing the science and best clinical practices necessary to generate potent abscopal effects with anti-metastatic immune activities.
Collapse
Affiliation(s)
- Timothy M Fan
- Comparative Oncology Research Laboratory, Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois at Urbana-Champaign Urbana, IL, United States
| | - Kimberly A Selting
- Comparative Oncology Research Laboratory, Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois at Urbana-Champaign Urbana, IL, United States
| |
Collapse
|
14
|
Gadau SD. Tubulin post-translational modifications in developing dog primary neurons obtained with methods according to the 3Rs principles. Res Vet Sci 2018; 122:56-63. [PMID: 30458355 DOI: 10.1016/j.rvsc.2018.11.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 10/19/2018] [Accepted: 11/12/2018] [Indexed: 11/30/2022]
Abstract
Microtubules play a crucial role during neuronal morphogenesis regulating many functions. In the study of these phenomena in vitro cellular models have been employed, mainly resorting to housed experimental animals. Among alternative models in neurobiological study, recently dog caught particular attention. In fact, the complexity of the canine brain, the life long span and the neurodegenerative pathologies render the dog a species more close to humans than rodents. Lately, growing interest in the limitation of the use of experimental animals, has stimulated the search for alternative experimental protocols. Starting from fetal dog brain, obtained by alternative way of sampling, we set neuronal primary cultures. Through immunofluorescence, we examined the presence and the cellular distribution of tubulin post-translational modifications as tyrosinated and acetylated α-tubulin, as markers of dynamic and stable microtubule respectively. In addition, we evaluated the pattern of two associated proteins which may slide on these two tubulin modifications, i.e. CLIP-170 and Kinesin-1. A clear positivity for tyrosinated and acetylated α-tubulin, was found. As far as the motor proteins are concerned, we detected a prevalence of CLIP-170 compared to kinesin-1 with a better overlapping between tyrosinated α-tubulin and CLIP-170. Our findings highlighted some original data about the role of the microtubular network during early phases of canine neuronal morphogenesis. In addition, the experimental protocol underlined the utility of this alternative model that allows to bypass both the scarcity of commercial canine neuronal cell lines and the need to resort to experimental dogs, respecting the 3Rs principles (reduction, refinement, and replacement).
Collapse
Affiliation(s)
- Sergio Domenico Gadau
- Department of Veterinary Medicine, University of Sassari, Via Vienna 2, 07100, Italy.
| |
Collapse
|
15
|
Hubbard ME, Arnold S, Bin Zahid A, McPheeters M, Gerard O’Sullivan M, Tabaran AF, Hunt MA, Pluhar GE. Naturally Occurring Canine Glioma as a Model for Novel Therapeutics. Cancer Invest 2018; 36:415-423. [DOI: 10.1080/07357907.2018.1514622] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Molly E. Hubbard
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
| | - Susan Arnold
- College of Veterinary Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Abdullah Bin Zahid
- Division of Neurosurgery, Hennepin County Medical Center, Minneapolis, MN, USA
| | | | - M. Gerard O’Sullivan
- College of Veterinary Medicine, University of Minnesota, Minneapolis, MN, USA
- Comparitive Pathology Shared Resource at Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Alexandru-Flaviu Tabaran
- College of Veterinary Medicine, University of Minnesota, Minneapolis, MN, USA
- Comparitive Pathology Shared Resource at Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Matthew A. Hunt
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
| | - G. Elizabeth Pluhar
- College of Veterinary Medicine, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
16
|
In vitro effect of chlorambucil on human glioma cell lines (SF767 and U87-MG), and human microvascular endothelial cell (HMVEC) and endothelial progenitor cells (ECFCs), in the context of plasma chlorambucil concentrations in tumor-bearing dogs. PLoS One 2018; 13:e0203517. [PMID: 30192852 PMCID: PMC6128565 DOI: 10.1371/journal.pone.0203517] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 08/22/2018] [Indexed: 12/12/2022] Open
Abstract
The objective of this study was to investigate a possible mechanism of action of metronomic chlorambucil on glioma by studying the in vitro cytotoxicity and anti-angiogenic effects on glioma and endothelial cells, respectively. The in vitro LD50 and IC50 of chlorambucil were determined using human SF767 and U87-MG glioma cell lines, human microvascular endothelial cells (HMVECs) and human endothelial colony forming cells (ECFCs). Results were analyzed in the context of chlorambucil concentrations measured in the plasma of tumor-bearing dogs receiving 4 mg m-2 metronomic chlorambucil. The LD50 and IC50 of chlorambucil were 270 μM and 114 μM for SF767, and 390 μM and 96 μM for U87-MG, respectively. The IC50 of chlorambucil was 0.53 μM and 145 μM for the HMVECs and ECFCs, respectively. In pharmacokinetic studies, the mean plasma Cmax of chlorambucil was 0.06 μM. Results suggest that metronomic chlorambucil in dogs does not achieve plasma concentrations high enough to cause direct cytotoxic or growth inhibitory effects on either glioma or endothelial cells.
Collapse
|
17
|
Nakamoto Y, Fukunaga D, Uchida K, Mori T, Kishimoto T, Ozawa T. Anaplastic oligodendroglioma with leptomeningeal dissemination in a french bulldog. J Vet Med Sci 2018; 80:1590-1595. [PMID: 30185721 PMCID: PMC6207505 DOI: 10.1292/jvms.17-0652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A 2.5-year-old male French Bulldog was evaluated for seizures. Cranial magnetic resonance imaging (MRI) suggested a glioma in the left piriform area. Radiation therapy (RT) and continuous chemotherapy were administered. Although the lesion had regressed significantly 2 months after RT, a follow-up MRI revealed meningeal enhancement in the left piriform area, which expanded further, with hydrocephalus, by day 310 (8 months after RT). Because of the poor prognosis, the dog was euthanized on day 356 and necropsy was performed. Histopathological examination confirmed anaplastic oligodendroglioma with leptomeningeal dissemination. This case suggests that the possibility of leptomeningeal dissemination and hydrocephalus should be considered even after RT and chemotherapy for anaplastic oligodendroglioma.
Collapse
Affiliation(s)
- Yuya Nakamoto
- Kyoto Animal Referral Medical Center, 208-4, Shinarami, Tai, Kumiyama-cho, Kuze-gun, Kyoto 613-0036, Japan.,Department of Bioartificial Organs, Institute for Frontier Life and Medical Sciences, Kyoto University, 53, Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Daisuke Fukunaga
- Crea Animal Hospital, 5-13-21, Aoyama, Otsu, Shiga 520-2101, Japan
| | - Kazuyuki Uchida
- Department of Veterinary Pathology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Takashi Mori
- Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, 1-1, Yanagido, Gifu 501-1193, Japan
| | - Takuya Kishimoto
- Department of Veterinary Pathology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Tsuyoshi Ozawa
- Kyoto Animal Referral Medical Center, 208-4, Shinarami, Tai, Kumiyama-cho, Kuze-gun, Kyoto 613-0036, Japan
| |
Collapse
|
18
|
Young JS, Bernal G, Polster SP, Nunez L, Larsen GF, Mansour N, Podell M, Yamini B. Convection-Enhanced Delivery of Polymeric Nanoparticles Encapsulating Chemotherapy in Canines with Spontaneous Supratentorial Tumors. World Neurosurg 2018; 117:e698-e704. [PMID: 29960096 DOI: 10.1016/j.wneu.2018.06.114] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Despite aggressive multimodal treatment, survival for patients with glioblastoma remains dismal. One obstacle to improving patient outcomes is the difficulty in delivering adequate therapeutic to the central nervous system due to the presence of the blood-brain barrier. Although direct drug infusion by convection-enhanced delivery (CED) can bypass the blood-brain barrier and facilitate delivery to intracranial tumors, determining the distribution of delivered therapeutic remains problematic. Image guidance is a strategy that can optimize the accuracy of therapeutic delivery. METHODS Here we performed an open-label clinical trial in 10 pet dogs with spontaneous intracranial tumors to examine the target coverage accuracy of delivering polymeric magnetite nanoparticles (PMNPs) encapsulating temozolomide (TMZ). A modified small animal frame was applied to the head of each subject, and PMNPs were delivered stereotactically to the center of the tumor. Magnetic resonance imaging (MRI) was performed immediately postoperatively to examine PMNP distribution, and the animals were followed until death. RESULTS Nine of the 10 dogs underwent PMNP infusion without complications. No infusate backflow was observed during any procedure. In 70% of the cases, the infusion accurately targeted the tumor mass, as determined by the presence of PMNP signal in the tumor on immediate postoperative MRI. CONCLUSIONS These data suggest that CED of PMNPs carrying TMZ is safe in dogs with intracranial tumors and can lead to nanoparticle distribution in the region of the target. Image guidance is an important adjunct to CED, because distribution is unpredictable, with the potential for missed target delivery.
Collapse
Affiliation(s)
- Jacob S Young
- Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Giovanna Bernal
- Section of Neurosurgery, Department of Surgery, The University of Chicago, Chicago, Illinois, USA
| | - Sean P Polster
- Section of Neurosurgery, Department of Surgery, The University of Chicago, Chicago, Illinois, USA
| | - Luis Nunez
- LNK Chemsolutions LLC, Lincoln, Nebraska, USA
| | | | - Nassir Mansour
- Section of Neurosurgery, Department of Surgery, The University of Chicago, Chicago, Illinois, USA
| | - Michael Podell
- Section of Neurosurgery, Department of Surgery, The University of Chicago, Chicago, Illinois, USA; Medvet Chicago, Chicago, Illinois, USA
| | - Bakhtiar Yamini
- Section of Neurosurgery, Department of Surgery, The University of Chicago, Chicago, Illinois, USA.
| |
Collapse
|
19
|
Immunologic and gene expression profiles of spontaneous canine oligodendrogliomas. J Neurooncol 2018; 137:469-479. [PMID: 29330750 DOI: 10.1007/s11060-018-2753-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 01/07/2018] [Indexed: 10/18/2022]
Abstract
Malignant glioma (MG), the most common primary brain tumor in adults, is extremely aggressive and uniformly fatal. Several treatment strategies have shown significant preclinical promise in murine models of glioma; however, none have produced meaningful clinical responses in human patients. We hypothesize that introduction of an additional preclinical animal model better approximating the complexity of human MG, particularly in interactions with host immune responses, will bridge the existing gap between these two stages of testing. Here, we characterize the immunologic landscape and gene expression profiles of spontaneous canine glioma and evaluate its potential for serving as such a translational model. RNA in situ hybridization, flowcytometry, and RNA sequencing were used to evaluate immune cell presence and gene expression in healthy and glioma-bearing canines. Similar to human MGs, canine gliomas demonstrated increased intratumoral immune cell infiltration (CD4+, CD8+ and CD4+Foxp3+ T cells). The peripheral blood of glioma-bearing dogs also contained a relatively greater proportion of CD4+Foxp3+ regulatory T cells and plasmacytoid dendritic cells. Tumors were strongly positive for PD-L1 expression and glioma-bearing animals also possessed a greater proportion of immune cells expressing the immune checkpoint receptors CTLA-4 and PD-1. Analysis of differentially expressed genes in our canine populations revealed several genetic changes paralleling those known to occur in human disease. Naturally occurring canine glioma has many characteristics closely resembling human disease, particularly with respect to genetic dysregulation and host immune responses to tumors, supporting its use as a translational model in the preclinical testing of prospective anti-glioma therapies proven successful in murine studies.
Collapse
|
20
|
Abstract
Case series summary This study aimed to evaluate the pathologic and diagnostic features of 13 cases of feline glioma diagnosed at two veterinary diagnostic institutions over 16 years. A retrospective search was conducted using the laboratory information system. Selected cases were reviewed, tumors were classified according to the 2007 World Health Organization Classification of Tumours of the Central Nervous System, and tissue sections were submitted to an immunohistochemistry panel for further characterization. Affected cats were adults (average age 8 years), and male neutered (9/13) domestic shorthair cats (12/13) were over-represented. Clinical signs had acute onset and were progressive, lasting from 1-60 days (average course 20.7 days). Euthanasia was elected in all but one case, owing to a poor prognosis or because of a suspicion of rabies. Tumors occurred in the telencephalon (8/13), spinal cord (3/13) and brainstem (2/13). Gross changes in seven cases consisted of well- or poorly demarcated, gray-to-brown, soft, gelatinous masses that often caused secondary changes to the brain. The final diagnosis and classification were made based on histopathology and immunohistochemistry. Diagnoses consisted of oligodendroglioma (six cases), anaplastic astrocytoma (two cases), oligoastrocytoma, anaplastic ependymoma, gliomatosis cerebri, glioblastoma and anaplastic oligodendroglioma (one case each). Relevance and novel information This article confirms the need for a full neurologic and neuropathologic examination in cats with clinical neurologic disease. In addition, it highlights and proposes a complete routine immunohistochemical panel for diagnostic confirmation and differentiation between glioma and other morphologically similar central nervous system neoplasms in cats.
Collapse
Affiliation(s)
- Daniel R Rissi
- Department of Pathology and Athens Veterinary Diagnostic Laboratory, College of Veterinary Medicine, The University of Georgia, Athens, GA, USA
| | - Andrew D Miller
- Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, NY, USA
| |
Collapse
|
21
|
Grenier JK, Foureman PA, Sloma EA, Miller AD. RNA-seq transcriptome analysis of formalin fixed, paraffin-embedded canine meningioma. PLoS One 2017; 12:e0187150. [PMID: 29073243 PMCID: PMC5658167 DOI: 10.1371/journal.pone.0187150] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 10/14/2017] [Indexed: 12/21/2022] Open
Abstract
Meningiomas are the most commonly reported primary intracranial tumor in dogs and humans and between the two species there are similarities in histology and biologic behavior. Due to these similarities, dogs have been proposed as models for meningioma pathobiology. However, little is known about specific pathways and individual genes that are involved in the development and progression of canine meningioma. In addition, studies are lacking that utilize RNAseq to characterize gene expression in clinical cases of canine meningioma. The primary objective of this study was to develop a technique for which high quality RNA can be extracted from formalin-fixed, paraffin embedded tissue and then used for transcriptome analysis to determine patterns of gene expression. RNA was extracted from thirteen canine meningiomas-eleven from formalin fixed and two flash-frozen. These represented six grade I and seven grade II meningiomas based on the World Health Organization classification system for human meningioma. RNA was also extracted from fresh frozen leptomeninges from three control dogs for comparison. RNAseq libraries made from formalin fixed tissue were of sufficient quality to successfully identify 125 significantly differentially expressed genes, the majority of which were related to oncogenic processes. Twelve genes (AQP1, BMPER, FBLN2, FRZB, MEDAG, MYC, PAMR1, PDGFRL, PDPN, PECAM1, PERP, ZC2HC1C) were validated using qPCR. Among the differentially expressed genes were oncogenes, tumor suppressors, transcription factors, VEGF-related genes, and members of the WNT pathway. Our work demonstrates that RNA of sufficient quality can be extracted from FFPE canine meningioma samples to provide biologically relevant transcriptome analyses using a next-generation sequencing technique, such as RNA-seq.
Collapse
Affiliation(s)
- Jennifer K. Grenier
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, New York, United States of America
| | - Polly A. Foureman
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, New York, United States of America
- Division of Biological Sciences, Chandler-Gilbert Community College, Chandler, Arizona, United States of America
| | - Erica A. Sloma
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, New York, United States of America
| | - Andrew D. Miller
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, New York, United States of America
| |
Collapse
|
22
|
Mazzatenta A, Carluccio A, Robbe D, Giulio CD, Cellerino A. The companion dog as a unique translational model for aging. Semin Cell Dev Biol 2017; 70:141-153. [DOI: 10.1016/j.semcdb.2017.08.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/06/2017] [Accepted: 08/07/2017] [Indexed: 10/19/2022]
|
23
|
Bentley RT, Ahmed AU, Yanke AB, Cohen-Gadol AA, Dey M. Dogs are man's best friend: in sickness and in health. Neuro Oncol 2017; 19:312-322. [PMID: 27298310 DOI: 10.1093/neuonc/now109] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/08/2016] [Indexed: 01/17/2023] Open
Abstract
With the median survival of 14.6 months following best available standard of care, malignant gliomas (MGs) remain one of the biggest therapeutic challenges of the modern time. Although the last several decades have witnessed tremendous advancement in our understanding of MG and evolution of many successful preclinical therapeutic strategies, even the most successful preclinical therapeutic strategies often fail to cross the phase I/II clinical trial threshold. One of the significant, but less commonly discussed, barriers in developing effective glioma therapy is the lack of a robust preclinical model. For the last 30 years, rodent orthotopic xenograft models have been extensively used in the preclinical setting. Although they provide a good basic model for understanding tumor biology, their value in successfully translating preclinical therapeutic triumph into clinical success is extremely poor. Companion dogs, which share the same environmental stress as their human counterparts, also spontaneously develop MGs. Dog gliomas that develop spontaneously in an immunocompetent host are very similar to human gliomas and potentially provide a stronger platform for validating the efficacy of therapeutic strategies proven successful in preclinical mouse models. Integrating this model can accelerate development of effective therapeutic options that will benefit both human subjects and pet dogs.
Collapse
Affiliation(s)
- R Timothy Bentley
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, Indiana, USA
| | | | - Amy B Yanke
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, Indiana, USA
| | | | - Mahua Dey
- Department of Neurosurgery, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|
24
|
Joshi AD, Botham RC, Schlein LJ, Roth HS, Mangraviti A, Borodovsky A, Tyler B, Joslyn S, Looper JS, Podell M, Fan TM, Hergenrother PJ, Riggins GJ. Synergistic and targeted therapy with a procaspase-3 activator and temozolomide extends survival in glioma rodent models and is feasible for the treatment of canine malignant glioma patients. Oncotarget 2017; 8:80124-80138. [PMID: 29113289 PMCID: PMC5655184 DOI: 10.18632/oncotarget.19085] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/09/2017] [Indexed: 12/17/2022] Open
Abstract
Purpose Glioblastoma is a deadly brain cancer with a median survival time of ∼15 months. Ionizing radiation plus the DNA alkylator temozolomide (TMZ) is the current standard therapy. PAC-1, a procaspase-3 activating small molecule, is blood-brain barrier penetrant and has previously demonstrated ability to synergize with diverse pro-apoptotic chemotherapeutics. We studied if PAC-1 could enhance the activity of TMZ, and whether addition of PAC-1 to standard treatment would be feasible in spontaneous canine malignant gliomas. Experimental Design Using cell lines and online gene expression data, we identified procaspase-3 as a potential molecular target for most glioblastomas. We investigated PAC-1 as a single agent and in combination with TMZ against glioma cells in culture and in orthotopic rodent models of glioma. Three dogs with spontaneous gliomas were treated with an analogous human glioblastoma treatment protocol, with concurrent PAC-1. Results Procaspase-3 is expressed in gliomas, with higher gene expression correlating with increased tumor grade and decreased prognosis. PAC-1 is cytotoxic to glioma cells in culture and active in orthotopic rodent glioma models. PAC-1 added to TMZ treatments in cell culture increases apoptotic death, and the combination significantly increases survival in orthotopic glioma models. Addition of PAC-1 to TMZ and radiation was well-tolerated in 3 out of 3 pet dogs with spontaneous glioma, and partial to complete tumor reductions were observed. Conclusions Procaspase-3 is a clinically relevant target for treatment of glioblastoma. Synergistic activity of PAC-1/TMZ in rodent models and the demonstration of feasibility of the combined regime in canine patients suggest potential for PAC-1 in the treatment of glioblastoma.
Collapse
Affiliation(s)
- Avadhut D Joshi
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Rachel C Botham
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Lisa J Schlein
- Department of Pathobiology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Howard S Roth
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Antonella Mangraviti
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Alexandra Borodovsky
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Betty Tyler
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | | - Jayme S Looper
- Department of Veterinary Clinical Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - Michael Podell
- Department of Neurology, MedVet Chicago, Chicago, IL, USA
| | - Timothy M Fan
- Department of Veterinary Clinical Medicine, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Paul J Hergenrother
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Gregory J Riggins
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
25
|
Genomic profiling of canine mast cell tumors identifies DNA copy number aberrations associated with KIT mutations and high histological grade. Chromosome Res 2017; 25:129-143. [DOI: 10.1007/s10577-016-9543-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 12/13/2016] [Accepted: 12/15/2016] [Indexed: 10/20/2022]
|
26
|
Rozov O, Piñeyro PE, Zimmerman KL, Herring IP, Matusow R, Rossmeisl JH, Jortner BS, Dreyfus J. Optic nerve astrocytoma in a dog. Clin Case Rep 2016; 4:855-60. [PMID: 27648262 PMCID: PMC5018588 DOI: 10.1002/ccr3.612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 04/14/2016] [Accepted: 04/30/2016] [Indexed: 12/03/2022] Open
Abstract
Intracranial astrocytomas are relatively uncommon in dogs and optic nerve astrocytomas even more so. This neoplasm should be considered as differential in canine patients with vision loss, retinal detachment, ocular mass, and histopathologic findings of infiltrative fusiform to polygonal glial cells possibly associated with glomeruloid vascular proliferation.
Collapse
Affiliation(s)
- Orr Rozov
- Department of Biomedical Sciences and Pathobiology Virginia-Maryland College of Veterinary Medicine Virginia Tech Blacksburg Virginia 24061-0442 USA
| | - Pablo E Piñeyro
- Department of Veterinary Diagnostic and Production Animal Medicine College of Veterinary Medicine Iowa State University Ames Iowa 50011-1250 USA
| | - Kurt L Zimmerman
- Department of Biomedical Sciences and Pathobiology Virginia-Maryland College of Veterinary Medicine Virginia Tech Blacksburg Virginia 24061-0442 USA
| | - Ian P Herring
- Department of Small Animal Clinical Sciences Virginia-Maryland College of Veterinary Medicine Virginia Tech Blacksburg Virginia 24061-0442 USA
| | - Rachel Matusow
- Department of Small Animal Clinical Sciences Virginia-Maryland College of Veterinary Medicine Virginia Tech Blacksburg Virginia 24061-0442 USA
| | - John H Rossmeisl
- Department of Small Animal Clinical Sciences Virginia-Maryland College of Veterinary Medicine Virginia Tech Blacksburg Virginia 24061-0442 USA
| | - Bernard S Jortner
- Department of Biomedical Sciences and Pathobiology Virginia-Maryland College of Veterinary Medicine Virginia Tech Blacksburg Virginia 24061-0442 USA
| | - Jennifer Dreyfus
- Department of Biomedical Sciences and Pathobiology Virginia-Maryland College of Veterinary Medicine Virginia Tech Blacksburg Virginia 24061-0442 USA; Department of Pathobiological Sciences School of Veterinary Medicine University of Wisconsin - Madison Madison Wisconsin 53706 USA
| |
Collapse
|
27
|
Embracing Integrative Multiomics Approaches. Int J Genomics 2016; 2016:1715985. [PMID: 27689071 PMCID: PMC5027047 DOI: 10.1155/2016/1715985] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 06/22/2016] [Indexed: 11/26/2022] Open
Abstract
As “-omics” data technology advances and becomes more readily accessible to address complex biological questions, increasing amount of cross “-omics” dataset is inspiring the use and development of integrative bioinformatics analysis. In the current review, we discuss multiple options for integrating data across “-omes” for a range of study designs. We discuss established methods for such analysis and point the reader to in-depth discussions for the various topics. Additionally, we discuss challenges and new directions in the area.
Collapse
|
28
|
LeBlanc AK, Mazcko C, Brown DE, Koehler JW, Miller AD, Miller CR, Bentley RT, Packer RA, Breen M, Boudreau CE, Levine JM, Simpson RM, Halsey C, Kisseberth W, Rossmeisl JH, Dickinson PJ, Fan TM, Corps K, Aldape K, Puduvalli V, Pluhar GE, Gilbert MR. Creation of an NCI comparative brain tumor consortium: informing the translation of new knowledge from canine to human brain tumor patients. Neuro Oncol 2016; 18:1209-18. [PMID: 27179361 PMCID: PMC4999002 DOI: 10.1093/neuonc/now051] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/27/2016] [Indexed: 12/14/2022] Open
Abstract
On September 14-15, 2015, a meeting of clinicians and investigators in the fields of veterinary and human neuro-oncology, clinical trials, neuropathology, and drug development was convened at the National Institutes of Health campus in Bethesda, Maryland. This meeting served as the inaugural event launching a new consortium focused on improving the knowledge, development of, and access to naturally occurring canine brain cancer, specifically glioma, as a model for human disease. Within the meeting, a SWOT (strengths, weaknesses, opportunities, and threats) assessment was undertaken to critically evaluate the role that naturally occurring canine brain tumors could have in advancing this aspect of comparative oncology aimed at improving outcomes for dogs and human beings. A summary of this meeting and subsequent discussion are provided to inform the scientific and clinical community of the potential for this initiative. Canine and human comparisons represent an unprecedented opportunity to complement conventional brain tumor research paradigms, addressing a devastating disease for which innovative diagnostic and treatment strategies are clearly needed.
Collapse
Affiliation(s)
- Amy K LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Christina Mazcko
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Diane E Brown
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Jennifer W Koehler
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Andrew D Miller
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - C Ryan Miller
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - R Timothy Bentley
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Rebecca A Packer
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Matthew Breen
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - C Elizabeth Boudreau
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Jonathan M Levine
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - R Mark Simpson
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Charles Halsey
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - William Kisseberth
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - John H Rossmeisl
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Peter J Dickinson
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Timothy M Fan
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Kara Corps
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Kenneth Aldape
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Vinay Puduvalli
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - G Elizabeth Pluhar
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Mark R Gilbert
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| |
Collapse
|
29
|
Dickinson PJ, York D, Higgins RJ, LeCouteur RA, Joshi N, Bannasch D. Chromosomal Aberrations in Canine Gliomas Define Candidate Genes and Common Pathways in Dogs and Humans. J Neuropathol Exp Neurol 2016; 75:700-10. [PMID: 27251041 DOI: 10.1093/jnen/nlw042] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Indexed: 12/16/2022] Open
Abstract
Spontaneous gliomas in dogs occur at a frequency similar to that in humans and may provide a translational model for therapeutic development and comparative biological investigations. Copy number alterations in 38 canine gliomas, including diffuse astrocytomas, glioblastomas, oligodendrogliomas, and mixed oligoastrocytomas, were defined using an Illumina 170K single nucleotide polymorphism array. Highly recurrent alterations were seen in up to 85% of some tumor types, most notably involving chromosomes 13, 22, and 38, and gliomas clustered into 2 major groups consisting of high-grade IV astrocytomas, or oligodendrogliomas and other tumors. Tumor types were characterized by specific broad and focal chromosomal events including focal loss of the INK4A/B locus in glioblastoma and loss of the RB1 gene and amplification of the PDGFRA gene in oligodendrogliomas. Genes associated with the 3 critical pathways in human high-grade gliomas (TP53, RB1, and RTK/RAS/PI3K) were frequently associated with canine aberrations. Analysis of oligodendrogliomas revealed regions of chromosomal losses syntenic to human 1p involving tumor suppressor genes, such as CDKN2C, as well as genes associated with apoptosis, autophagy, and response to chemotherapy and radiation. Analysis of high frequency chromosomal aberrations with respect to human orthologues may provide insight into both novel and common pathways in gliomagenesis and response to therapy.
Collapse
Affiliation(s)
- Peter J Dickinson
- From the Departments of Surgical and Radiological Sciences (PJD, DY, RAL), Pathology, Microbiology and Immunology (RJH), and Population Health & Reproduction (DB), School of Veterinary Medicine, University of California, Davis, and Bioinformatics Core, UC Davis Genome Center (NJ) University of California, Davis, California.
| | - Dan York
- From the Departments of Surgical and Radiological Sciences (PJD, DY, RAL), Pathology, Microbiology and Immunology (RJH), and Population Health & Reproduction (DB), School of Veterinary Medicine, University of California, Davis, and Bioinformatics Core, UC Davis Genome Center (NJ) University of California, Davis, California
| | - Robert J Higgins
- From the Departments of Surgical and Radiological Sciences (PJD, DY, RAL), Pathology, Microbiology and Immunology (RJH), and Population Health & Reproduction (DB), School of Veterinary Medicine, University of California, Davis, and Bioinformatics Core, UC Davis Genome Center (NJ) University of California, Davis, California
| | - Richard A LeCouteur
- From the Departments of Surgical and Radiological Sciences (PJD, DY, RAL), Pathology, Microbiology and Immunology (RJH), and Population Health & Reproduction (DB), School of Veterinary Medicine, University of California, Davis, and Bioinformatics Core, UC Davis Genome Center (NJ) University of California, Davis, California
| | - Nikhil Joshi
- From the Departments of Surgical and Radiological Sciences (PJD, DY, RAL), Pathology, Microbiology and Immunology (RJH), and Population Health & Reproduction (DB), School of Veterinary Medicine, University of California, Davis, and Bioinformatics Core, UC Davis Genome Center (NJ) University of California, Davis, California
| | - Danika Bannasch
- From the Departments of Surgical and Radiological Sciences (PJD, DY, RAL), Pathology, Microbiology and Immunology (RJH), and Population Health & Reproduction (DB), School of Veterinary Medicine, University of California, Davis, and Bioinformatics Core, UC Davis Genome Center (NJ) University of California, Davis, California
| |
Collapse
|
30
|
Rogers N. Canine clues: Dog genomes explored in effort to bring human cancer to heel. Nat Med 2016; 21:1374-5. [PMID: 26646483 DOI: 10.1038/nm1215-1374] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
31
|
Schiffman JD, Breen M. Comparative oncology: what dogs and other species can teach us about humans with cancer. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0231. [PMID: 26056372 DOI: 10.1098/rstb.2014.0231] [Citation(s) in RCA: 251] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Over 1.66 million humans (approx. 500/100,000 population rate) and over 4.2 million dogs (approx. 5300/100,000 population rate) are diagnosed with cancer annually in the USA. The interdisciplinary field of comparative oncology offers a unique and strong opportunity to learn more about universal cancer risk and development through epidemiology, genetic and genomic investigations. Working across species, researchers from human and veterinary medicine can combine scientific findings to understand more quickly the origins of cancer and translate these findings to novel therapies to benefit both human and animals. This review begins with the genetic origins of canines and their advantage in cancer research. We next focus on recent findings in comparative oncology related to inherited, or genetic, risk for tumour development. We then detail the somatic, or genomic, changes within tumours and the similarities between species. The shared cancers between humans and dogs that we discuss include sarcoma (osteosarcoma, soft tissue sarcoma, histiocytic sarcoma, hemangiosarcoma), haematological malignancies (lymphoma, leukaemia), bladder cancer, intracranial neoplasms (meningioma, glioma) and melanoma. Tumour risk in other animal species is also briefly discussed. As the field of genomics advances, we predict that comparative oncology will continue to benefit both humans and the animals that live among us.
Collapse
Affiliation(s)
- Joshua D Schiffman
- Department of Pediatrics and Oncological Sciences, Primary Children's Hospital, Intermountain Healthcare, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Matthew Breen
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, Center for Comparative Medicine and Translational Research, Center for Human Health and the Environment, Cancer Genetics, UNC Lineberger Comprehensive Cancer Center, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
32
|
Rossmeisl JH, Garcia PA, Pancotto TE, Robertson JL, Henao-Guerrero N, Neal RE, Ellis TL, Davalos RV. Safety and feasibility of the NanoKnife system for irreversible electroporation ablative treatment of canine spontaneous intracranial gliomas. J Neurosurg 2015; 123:1008-25. [DOI: 10.3171/2014.12.jns141768] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECT
Irreversible electroporation (IRE) is a novel nonthermal ablation technique that has been used for the treatment of solid cancers. However, it has not been evaluated for use in brain tumors. Here, the authors report on the safety and feasibility of using the NanoKnife IRE system for the treatment of spontaneous intracranial gliomas in dogs.
METHODS
Client-owned dogs with a telencephalic glioma shown on MRI were eligible. Dog-specific treatment plans were generated by using MRI-based tissue segmentation, volumetric meshing, and finite element modeling. After biopsy confirmation of glioma, IRE treatment was delivered stereotactically with the NanoKnife system using pulse parameters and electrode configurations derived from therapeutic plans. The primary end point was an evaluation of safety over the 14 days immediately after treatment. Follow-up was continued for 12 months or until death with serial physical, neurological, laboratory, and MRI examinations.
RESULTS
Seven dogs with glioma were treated. The mean age of the dogs was 9.3 ± 1.6 years, and the mean pretreatment tumor volume was 1.9 ± 1.4 cm3. The median preoperative Karnofsky Performance Scale score was 70 (range 30–75). Severe posttreatment toxicity was observed in 2 of the 7 dogs; one developed fatal (Grade 5) aspiration pneumonia, and the other developed treatment-associated cerebral edema, which resulted in transient neurological deterioration. Results of posttreatment diagnostic imaging, tumor biopsies, and neurological examinations indicated that tumor ablation was achieved without significant direct neurotoxicity in 6 of the 7 dogs. The median 14-day post-IRE Karnofsky Performance Scale score of the 6 dogs that survived to discharge was 80 (range 60–90), and this score was improved over the pretreatment value in every case. Objective tumor responses were seen in 4 (80%) of 5 dogs with quantifiable target lesions. The median survival was 119 days (range 1 to > 940 days).
CONCLUSION
With the incorporation of additional therapeutic planning procedures, the NanoKnife system is a novel technology capable of controlled IRE ablation of telencephalic gliomas.
Collapse
Affiliation(s)
- John H. Rossmeisl
- 1Department of Small Animal Clinical Sciences and
- 2Veterinary and Comparative Neurooncology Laboratory, Virginia-Maryland Regional College of Veterinary Medicine, and
- 3Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, Virginia; and
| | - Paulo A. Garcia
- 2Veterinary and Comparative Neurooncology Laboratory, Virginia-Maryland Regional College of Veterinary Medicine, and
- 3Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, Virginia; and
| | | | - John L. Robertson
- 1Department of Small Animal Clinical Sciences and
- 2Veterinary and Comparative Neurooncology Laboratory, Virginia-Maryland Regional College of Veterinary Medicine, and
- 3Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, Virginia; and
| | | | - Robert E. Neal
- 3Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, Virginia; and
| | - Thomas L. Ellis
- 4Department of Neurosurgery and Deep Brain Stimulation Program, School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Rafael V. Davalos
- 3Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, Virginia; and
| |
Collapse
|
33
|
Comparative Aspects of BRAF Mutations in Canine Cancers. Vet Sci 2015; 2:231-245. [PMID: 29061943 PMCID: PMC5644641 DOI: 10.3390/vetsci2030231] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 08/14/2015] [Accepted: 08/19/2015] [Indexed: 01/07/2023] Open
Abstract
Activating mutations of the BRAF gene lead to constitutive activation of the MAPK pathway. The characterization and discovery of BRAF mutations in a variety of human cancers has led to the development of specific inhibitors targeting the BRAF/MAPK pathway and dramatically changed clinical outcomes in BRAF-mutant melanoma patients. Recent discovery of BRAF mutation in canine cancers underscores the importance of MAPK pathway activation as an oncogenic molecular alteration evolutionarily conserved between species. A comparative approach using the domestic dog as a spontaneous cancer model will provide new insights into the dysregulation of BRAF/MAPK pathway in carcinogenesis and facilitate in vivo studies to evaluate therapeutic strategies targeting this pathway's molecules for cancer therapy. The BRAF mutation in canine cancers may also represent a molecular marker and therapeutic target in veterinary oncology. This review article summarizes the current knowledge on BRAF mutations in human and canine cancers and discusses the potential applications of this abnormality in veterinary oncology.
Collapse
|
34
|
Dieterich S, Zwingenberger A, Hansen K, Pfeiffer I, Théon A, Kent MS. INTER- AND INTRAFRACTION MOTION FOR STEREOTACTIC RADIOSURGERY IN DOGS AND CATS USING A MODIFIED BRAINLAB FRAMELESS STEREOTACTIC MASK SYSTEM. Vet Radiol Ultrasound 2015; 56:563-9. [DOI: 10.1111/vru.12271] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 04/05/2015] [Indexed: 12/27/2022] Open
Affiliation(s)
- Sonja Dieterich
- Department of Radiation Oncology; UC Davis School of Medicine; Sacramento CA
| | - Allison Zwingenberger
- Department of Surgical and Radiological Sciences; UC Davis School of Veterinary Medicine 1 Shields Ave; Davis CA
| | - Katherine Hansen
- Department of Surgical and Radiological Sciences; UC Davis School of Veterinary Medicine 1 Shields Ave; Davis CA
| | - Isabella Pfeiffer
- The William R. Prichard Veterinary Medical Teaching Hospital; UC Davis School of Veterinary Medicine 1 Shields Ave; Davis CA
| | - Alain Théon
- Department of Surgical and Radiological Sciences; UC Davis School of Veterinary Medicine 1 Shields Ave; Davis CA
| | - Michael S. Kent
- Department of Surgical and Radiological Sciences; UC Davis School of Veterinary Medicine 1 Shields Ave; Davis CA
| |
Collapse
|
35
|
Roode SC, Rotroff D, Avery AC, Suter SE, Bienzle D, Schiffman JD, Motsinger-Reif A, Breen M. Genome-wide assessment of recurrent genomic imbalances in canine leukemia identifies evolutionarily conserved regions for subtype differentiation. Chromosome Res 2015; 23:681-708. [PMID: 26037708 DOI: 10.1007/s10577-015-9475-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/02/2015] [Accepted: 05/05/2015] [Indexed: 11/30/2022]
Abstract
Leukemia in dogs is a heterogeneous disease with survival ranging from days to years, depending on the subtype. Strides have been made in both human and canine leukemia to improve classification and understanding of pathogenesis through immunophenotyping, yet classification and choosing appropriate therapy remains challenging. In this study, we assessed 123 cases of canine leukemia (28 ALLs, 24 AMLs, 25 B-CLLs, and 46 T-CLLs) using high-resolution oligonucleotide array comparative genomic hybridization (oaCGH) to detect DNA copy number alterations (CNAs). For the first time, such data were used to identify recurrent CNAs and inclusive genes that may be potential drivers of subtype-specific pathogenesis. We performed predictive modeling to identify CNAs that could reliably differentiate acute subtypes (ALL vs. AML) and chronic subtypes (B-CLL vs. T-CLL) and used this model to differentiate cases with up to 83.3 and 95.8 % precision, respectively, based on CNAs at only one to three genomic regions. In addition, CGH datasets for canine and human leukemia were compared to reveal evolutionarily conserved copy number changes between species, including the shared gain of HSA 21q in ALL and ∼25 Mb of shared gain of HSA 12 and loss of HSA 13q14 in CLL. These findings support the use of canine leukemia as a relevant in vivo model for human leukemia and justify the need to further explore the conserved genomic regions of interest for their clinical impact.
Collapse
Affiliation(s)
- Sarah C Roode
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, Raleigh, NC, 27607, USA
| | - Daniel Rotroff
- Bioinformatics Research Center, Department of Statistics, North Carolina State University, Raleigh, NC, USA
| | - Anne C Avery
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Science, Colorado State University, Fort Collins, CO, USA
| | - Steven E Suter
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.,Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, NC, USA.,Cancer Genetics Program, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Dorothee Bienzle
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - Joshua D Schiffman
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA.,Department of Oncological Sciences, Center for Children's Cancer Research, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Alison Motsinger-Reif
- Bioinformatics Research Center, Department of Statistics, North Carolina State University, Raleigh, NC, USA.,Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, NC, USA
| | - Matthew Breen
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, Raleigh, NC, 27607, USA. .,Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, NC, USA. .,Cancer Genetics Program, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
36
|
Hicks J, Platt S, Kent M, Haley A. Canine brain tumours: a model for the human disease? Vet Comp Oncol 2015; 15:252-272. [PMID: 25988678 DOI: 10.1111/vco.12152] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 04/04/2015] [Accepted: 04/06/2015] [Indexed: 01/10/2023]
Abstract
Canine brain tumours are becoming established as naturally occurring models of disease to advance diagnostic and therapeutic understanding successfully. The size and structure of the dog's brain, histopathology and molecular characteristics of canine brain tumours, as well as the presence of an intact immune system, all support the potential success of this model. The limited success of current therapeutic regimens such as surgery and radiation for dogs with intracranial tumours means that there can be tremendous mutual benefit from collaboration with our human counterparts resulting in the development of new treatments. The similarities and differences between the canine and human diseases are described in this article, emphasizing both the importance and limitations of canines in brain tumour research. Recent clinical veterinary therapeutic trials are also described to demonstrate the areas of research in which canines have already been utilized and to highlight the important potential benefits of translational research to companion dogs.
Collapse
Affiliation(s)
- J Hicks
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - S Platt
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - M Kent
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - A Haley
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| |
Collapse
|
37
|
Omeir R, Thomas R, Teferedegne B, Williams C, Foseh G, Macauley J, Brinster L, Beren J, Peden K, Breen M, Lewis AM. A novel canine kidney cell line model for the evaluation of neoplastic development: karyotype evolution associated with spontaneous immortalization and tumorigenicity. Chromosome Res 2015; 23:663-80. [PMID: 25957863 PMCID: PMC4666904 DOI: 10.1007/s10577-015-9474-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 04/12/2015] [Accepted: 04/14/2015] [Indexed: 01/01/2023]
Abstract
The molecular mechanisms underlying spontaneous neoplastic transformation in cultured mammalian cells remain poorly understood, confounding recognition of parallels with the biology of naturally occurring cancer. The broad use of tumorigenic canine cell lines as research tools, coupled with the accumulation of cytogenomic data from naturally occurring canine cancers, makes the domestic dog an ideal system in which to investigate these relationships. We developed a canine kidney cell line, CKB1-3T7, which allows prospective examination of the onset of spontaneous immortalization and tumorigenicity. We documented the accumulation of cytogenomic aberrations in CKB1-3T7 over 24 months in continuous culture. The majority of aberrations emerged in parallel with key phenotypic changes in cell morphology, growth kinetics, and tumor incidence and latency. Focal deletion of CDKN2A/B emerged first, preceding the onset and progression of tumorigenic potential, and progressed to a homozygous deletion across the cell population during extended culture. Interestingly, CKB1-3T7 demonstrated a tumorigenic phenotype in vivo prior to exhibiting loss of contact inhibition in vitro. We also performed the first genome-wide characterization of the canine tumorigenic cell line MDCK, which also exhibited CDKN2A/B deletion. MDCK and CKB1-3T7 cells shared several additional aberrations that we have reported previously as being highly recurrent in spontaneous canine cancers, many of which, as with CDKN2A/B deletion, are evolutionarily conserved in their human counterparts. The conservation of these molecular events across multiple species, in vitro and in vivo, despite their contrasting karyotypic architecture, is a powerful indicator of a common mechanism underlying emerging neoplastic activity. Through integrated cytogenomic and phenotypic characterization of serial passages of CKB1-3T7 from initiation to development of a tumorigenic phenotype, we present a robust and readily accessible model (to be made available through the American Type Culture Collection) of spontaneous neoplastic transformation that overcomes many of the limitations of earlier studies.
Collapse
Affiliation(s)
- R Omeir
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - R Thomas
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, Raleigh, NC, 27607, USA.,Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, NC, 27607, USA
| | - B Teferedegne
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - C Williams
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, Raleigh, NC, 27607, USA
| | - G Foseh
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - J Macauley
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - L Brinster
- Division of Veterinary Resources, National Institutes of Health, Bethesda, MD, 20892, USA
| | - J Beren
- Office of Counter-Terrorism and Emergency Coordination, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - K Peden
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - M Breen
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, Raleigh, NC, 27607, USA. .,Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, NC, 27607, USA. .,Cancer Genetics Program, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, 27599, USA. .,Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, 27607, USA.
| | - A M Lewis
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA.
| |
Collapse
|
38
|
Borge KS, Nord S, Van Loo P, Lingjærde OC, Gunnes G, Alnæs GIG, Solvang HK, Lüders T, Kristensen VN, Børresen-Dale AL, Lingaas F. Canine Mammary Tumours Are Affected by Frequent Copy Number Aberrations, including Amplification of MYC and Loss of PTEN. PLoS One 2015; 10:e0126371. [PMID: 25955013 PMCID: PMC4425491 DOI: 10.1371/journal.pone.0126371] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 04/01/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Copy number aberrations frequently occur during the development of many cancers. Such events affect dosage of involved genes and may cause further genomic instability and progression of cancer. In this survey, canine SNP microarrays were used to study 117 canine mammary tumours from 69 dogs. RESULTS We found a high occurrence of copy number aberrations in canine mammary tumours, losses being more frequent than gains. Increased frequency of aberrations and loss of heterozygosity were positively correlated with increased malignancy in terms of histopathological diagnosis. One of the most highly recurrently amplified regions harbored the MYC gene. PTEN was located to a frequently lost region and also homozygously deleted in five tumours. Thus, deregulation of these genes due to copy number aberrations appears to be an important event in canine mammary tumour development. Other potential contributors to canine mammary tumour pathogenesis are COL9A3, INPP5A, CYP2E1 and RB1. The present study also shows that a more detailed analysis of chromosomal aberrations associated with histopathological parameters may aid in identifying specific genes associated with canine mammary tumour progression. CONCLUSIONS The high frequency of copy number aberrations is a prominent feature of canine mammary tumours as seen in other canine and human cancers. Our findings share several features with corresponding studies in human breast tumours and strengthen the dog as a suitable model organism for this disease.
Collapse
Affiliation(s)
- Kaja S. Borge
- Section of Genetics, Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences (NMBU),Oslo, Norway
| | - Silje Nord
- Department of Genetics, Institute for Cancer Research, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Peter Van Loo
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
- Human Genome Laboratory, Department of Human Genetics, VIB and University of Leuven, Leuven, Belgium
| | - Ole C. Lingjærde
- Department of Genetics, Institute for Cancer Research, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital Radiumhospitalet, Oslo, Norway
- Biomedical Informatics, Department of Informatics, University of Oslo, Oslo, Norway
- Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway
| | - Gjermund Gunnes
- Section of Anatomy and Pathology, Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences (NMBU), Oslo, Norway
| | - Grethe I. G. Alnæs
- Department of Genetics, Institute for Cancer Research, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Hiroko K. Solvang
- Marine Mammals Research Group, Institute of Marine Research, Bergen, Norway
| | - Torben Lüders
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Molecular Biology and Laboratory Sciences (EpiGen), Akershus University Hospital, Lørenskog, Norway
| | - Vessela N. Kristensen
- Department of Genetics, Institute for Cancer Research, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital Radiumhospitalet, Oslo, Norway
- The K. G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Molecular Biology, Division of Medicine, Akershus University Hospital, Ahus, Norway
| | - Anne-Lise Børresen-Dale
- Department of Genetics, Institute for Cancer Research, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital Radiumhospitalet, Oslo, Norway
- The K. G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Frode Lingaas
- Section of Genetics, Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences (NMBU),Oslo, Norway
| |
Collapse
|
39
|
Rossmeisl JH. New treatment modalities for brain tumors in dogs and cats. Vet Clin North Am Small Anim Pract 2014; 44:1013-38. [PMID: 25441624 DOI: 10.1016/j.cvsm.2014.07.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Despite advancements in standard therapies, intracranial tumors remain a significant source of morbidity and mortality in veterinary and human medicine. Several newer approaches are gaining more widespread acceptance or are currently being prepared for translation from experimental to routine therapeutic use. Clinical trials in dogs with spontaneous brain tumors have contributed to the development and human translation of several novel therapeutic brain tumor approaches.
Collapse
Affiliation(s)
- John H Rossmeisl
- Neurology and Neurosurgery, Department of Small Animal Clinical Sciences, VA-MD Regional College of Veterinary Medicine, Virginia Tech, 215 Duckpond Drive, Mail Code 0442, Blacksburg, VA 24061, USA.
| |
Collapse
|
40
|
Rotroff DM, Thomas R, Breen M, Motsinger-Reif AA. Naturally occuring canine cancers: powerful models for stimulating pharmacogenomic advancement in human medicine. Pharmacogenomics 2014; 14:1929-31. [PMID: 24279843 DOI: 10.2217/pgs.13.178] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Daniel M Rotroff
- Bioinformatics Research Center, Department of Statistics, Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695, USA
| | | | | | | |
Collapse
|
41
|
Dickinson P. Advances in diagnostic and treatment modalities for intracranial tumors. J Vet Intern Med 2014; 28:1165-85. [PMID: 24814688 PMCID: PMC4857954 DOI: 10.1111/jvim.12370] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 02/24/2014] [Accepted: 03/25/2014] [Indexed: 12/23/2022] Open
Abstract
Intracranial neoplasia is a common clinical condition in domestic companion animals, particularly in dogs. Application of advances in standard diagnostic and therapeutic modalities together with a broad interest in the development of novel translational therapeutic strategies in dogs has resulted in clinically relevant improvements in outcome for many canine patients. This review highlights the status of current diagnostic and therapeutic approaches to intracranial neoplasia and areas of novel treatment currently in development.
Collapse
Affiliation(s)
- P.J. Dickinson
- Department of Surgical and Radiological SciencesSchool of Veterinary MedicineUniversity of California DavisDavisCA
| |
Collapse
|
42
|
Genomic profiling reveals extensive heterogeneity in somatic DNA copy number aberrations of canine hemangiosarcoma. Chromosome Res 2014; 22:305-19. [PMID: 24599718 DOI: 10.1007/s10577-014-9406-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 01/22/2014] [Accepted: 01/23/2014] [Indexed: 01/08/2023]
Abstract
Canine hemangiosarcoma is a highly aggressive vascular neoplasm associated with extensive clinical and anatomical heterogeneity and a grave prognosis. Comprehensive molecular characterization of hemangiosarcoma may identify novel therapeutic targets and advanced clinical management strategies, but there are no published reports of tumor-associated genome instability and disrupted gene dosage in this cancer. We performed genome-wide microarray-based somatic DNA copy number profiling of 75 primary intra-abdominal hemangiosarcomas from five popular dog breeds that are highly predisposed to this disease. The cohort exhibited limited global genomic instability, compared to other canine sarcomas studied to date, and DNA copy number aberrations (CNAs) were predominantly of low amplitude. Recurrent imbalances of several key cancer-associated genes were evident; however, the global penetrance of any single CNA was low and no distinct hallmark aberrations were evident. Copy number gains of dog chromosomes 13, 24, and 31, and loss of chromosome 16, were the most recurrent CNAs involving large chromosome regions, but their relative distribution within and between cases suggests they most likely represent passenger aberrations. CNAs involving CDKN2A, VEGFA, and the SKI oncogene were identified as potential driver aberrations of hemangiosarcoma development, highlighting potential targets for therapeutic modulation. CNA profiles were broadly conserved between the five breeds, although subregional variation was evident, including a near twofold lower incidence of VEGFA gain in Golden Retrievers versus other breeds (22 versus 40 %). These observations support prior transcriptional studies suggesting that the clinical heterogeneity of this cancer may reflect the existence of multiple, molecularly distinct subtypes of canine hemangiosarcoma.
Collapse
|
43
|
Olin MR, Pluhar GE, Andersen BM, Shaver R, Waldron NN, Moertel CL. Victory and defeat in the induction of a therapeutic response through vaccine therapy for human and canine brain tumors: a review of the state of the art. Crit Rev Immunol 2014; 34:399-432. [PMID: 25404047 PMCID: PMC4485925 DOI: 10.1615/critrevimmunol.2014011577] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Anti-tumor immunotherapy using tumor lysate-based vaccines has made great advances over recent decades. Cancer vaccines aim to elicit adaptive immune responses through various pathways by providing tumor and tumor-associated antigens with an immune stimulant or adjuvant. These anti-tumor vaccines are therefore developed as personalized treatments. Utilizing tumors as a source of vaccine antigens in immunotherapy has demonstrated promising results with minimal toxicity. However, to date, researchers have failed to overcome the overpowering immune suppressive effects within the tumor microenvironment. Immune suppression occurs naturally via multiple mechanisms. These mechanisms serve an important homeostatic role restoring a normal tissue microenvironment following an inflammatory response. Due to these suppressive mechanisms and the inherent heterogeneity of tumors, it is imperative to then elicit and maintain a specific tumoricidal response if vaccine therapy or some other combination of reagents is chosen. In this review, we focus on the historical use of tumors as a source of antigens to elicit a tumoricidal response and the limitations encountered that prevent greater success in immunotherapy. We describe the advantages and disadvantages of various vaccines and their ineffectiveness due to tumor-induced immune suppression.
Collapse
Affiliation(s)
- Michael R. Olin
- Department of Pediatrics. University of Minnesota, Minneapolis, MN 55445
| | - G. Elizabeth Pluhar
- Department of Veterinary Medicine, College of Veterinary Medicine. University of Minnesota, St. Paul, MN 55108
| | - Brian M. Andersen
- Department of Pediatrics. University of Minnesota, Minneapolis, MN 55445
| | - Rob Shaver
- Department of Pediatrics. University of Minnesota, Minneapolis, MN 55445
| | - Nate N. Waldron
- Department of Pediatrics. University of Minnesota, Minneapolis, MN 55445
| | | |
Collapse
|
44
|
Johnson GC, Coates JR, Wininger F. Diagnostic immunohistochemistry of canine and feline intracalvarial tumors in the age of brain biopsies. Vet Pathol 2013; 51:146-60. [PMID: 24280940 DOI: 10.1177/0300985813509387] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The focus of immunohistochemistry as applied to nervous system tumors is in identifying the neoplasm present and evaluating margins between normal and neoplastic tissue. Although not always utilized by specialists in neuropathology, immunohistochemistry remains useful to resolve concerns about the differentiation and rate of tumor growth. The aims of this review are to discuss the utility of immunohistochemical reagents currently used in diagnosis of canine and feline intracalvarial tumors, to indicate the applicability of some tests currently used in human nervous system tumors for domestic species, and to evaluate a few less commonly used reagents. A panel of biomarkers is usually needed to confirm a diagnosis, with groups of reagents for leptomeningeal, intraparenchymal, and ventricular neoplasms. In the future, signature genetic alterations found among feline and canine brain tumors--as correlated prospectively with diagnosis, rate of enlargement, or response to treatment--may result in new immunohistochemical reagents to simplify the task of diagnosis. Prospective studies determining the type and proportion of stem cell marker expression on patient longevity are likely to be fruitful and suggest new therapies. Due to increased frequency of biopsy or partial resection of tumors from the living patient, biomarkers are needed to serve as accurate prognostic indicators and assist in determining the efficacy of developing therapeutic options in nervous system tumors of dogs and cats.
Collapse
Affiliation(s)
- G C Johnson
- Department of Veterinary Pathobiology, Veterinary Medical Diagnostic Laboratory, University of Missouri, 1600 East Rollins Street, Columbia MO 65211, USA.
| | | | | |
Collapse
|
45
|
Rossmeisl JH, Garcia PA, Daniel GB, Bourland JD, Debinski W, Dervisis N, Klahn S. Invited review--neuroimaging response assessment criteria for brain tumors in veterinary patients. Vet Radiol Ultrasound 2013; 55:115-32. [PMID: 24219161 DOI: 10.1111/vru.12118] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 09/07/2013] [Indexed: 12/28/2022] Open
Abstract
The evaluation of therapeutic response using cross-sectional imaging techniques, particularly gadolinium-enhanced MRI, is an integral part of the clinical management of brain tumors in veterinary patients. Spontaneous canine brain tumors are increasingly recognized and utilized as a translational model for the study of human brain tumors. However, no standardized neuroimaging response assessment criteria have been formulated for use in veterinary clinical trials. Previous studies have found that the pathophysiologic features inherent to brain tumors and the surrounding brain complicate the use of the response evaluation criteria in solid tumors (RECIST) assessment system. Objectives of this review are to describe strengths and limitations of published imaging-based brain tumor response criteria and propose a system for use in veterinary patients. The widely used human Macdonald and response assessment in neuro-oncology (RANO) criteria are reviewed and described as to how they can be applied to veterinary brain tumors. Discussion points will include current challenges associated with the interpretation of brain tumor therapeutic responses such as imaging pseudophenomena and treatment-induced necrosis, and how advancements in perfusion imaging, positron emission tomography, and magnetic resonance spectroscopy have shown promise in differentiating tumor progression from therapy-induced changes. Finally, although objective endpoints such as MR imaging and survival estimates will likely continue to comprise the foundations for outcome measures in veterinary brain tumor clinical trials, we propose that in order to provide a more relevant therapeutic response metric for veterinary patients, composite response systems should be formulated and validated that combine imaging and clinical assessment criteria.
Collapse
Affiliation(s)
- John H Rossmeisl
- Department of Small Animal Clinical Sciences, Virginia-Maryland Regional College of Veterinary Medicine, VA, 24061; Biomechanical Systems and Veterinary and Comparative Neuro-oncology Laboratories, Department of Biomedical Engineering, Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, VA, 24061
| | | | | | | | | | | | | |
Collapse
|
46
|
Gillard M, Cadieu E, De Brito C, Abadie J, Vergier B, Devauchelle P, Degorce F, Dréano S, Primot A, Dorso L, Lagadic M, Galibert F, Hédan B, Galibert MD, André C. Naturally occurring melanomas in dogs as models for non-UV pathways of human melanomas. Pigment Cell Melanoma Res 2013; 27:90-102. [PMID: 24112648 DOI: 10.1111/pcmr.12170] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 09/18/2013] [Indexed: 01/10/2023]
Abstract
Spontaneously occurring melanomas are frequent in dogs. They appear at the same localizations as in humans, i.e. skin, mucosal sites, nail matrix and eyes. They display variable behaviors: tumors at oral localizations are more frequent and aggressive than at other anatomical sites. Interestingly, dog melanomas are associated with strong breed predispositions and overrepresentation of black-coated dogs. Epidemiological analysis of 2350 affected dogs showed that poodles are at high risk of developing oral melanoma, while schnauzers or Beauce shepherds mostly developped cutaneous melanoma. Clinical and histopathological analyses were performed on a cohort of 153 cases with a 4-yr follow-up. Histopathological characterization showed that most canine tumors are intradermal and homologous to human rare morphological melanomas types - 'nevocytoid type' and 'animal type'-. Tumor cDNA sequencing data, obtained from 95 dogs for six genes, relevant to human melanoma classification, detected somatic mutations in oral melanoma, in NRAS and PTEN genes, at human hotspot sites, but not in BRAF. Altogether, these findings support the relevance of the dog model for comparative oncology of melanomas, especially for the elucidation of non-UV induced pathways.
Collapse
Affiliation(s)
- Marc Gillard
- CNRS, UMR 6290, Institut Génétique et Développement de Rennes, Rennes, France; Faculté de Médecine, SFR Biosit, Université Rennes 1, Rennes, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lim JH, Koh S, Olby NJ, Piedrahita J, Mariani CL. Isolation and characterization of neural progenitor cells from adult canine brains. Am J Vet Res 2012; 73:1963-8. [DOI: 10.2460/ajvr.73.12.1963] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
48
|
Koh S, Thomas R, Tsai S, Bischoff S, Lim JH, Breen M, Olby NJ, Piedrahita JA. Growth requirements and chromosomal instability of induced pluripotent stem cells generated from adult canine fibroblasts. Stem Cells Dev 2012; 22:951-63. [PMID: 23016947 DOI: 10.1089/scd.2012.0393] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In mice and humans, it has been shown that embryonic and adult fibroblasts can be reprogrammed into pluripotency by introducing 4 transcription factors, Oct3/4, Klf4, Sox2, and c-Myc (OKSM). Here, we report the derivation of induced pluripotent stem cells (iPSCs) from adult canine fibroblasts by retroviral OKSM transduction. The isolated canine iPSCs (ciPSCs) were expanded in 3 different culture media [fibroblast growth factor 2 (FGF2), leukemia inhibitory factor (LIF), or FGF2 plus LIF]. Cells cultured in both FGF2 and LIF expressed pluripotency markers [POU5F1 (OCT4), SOX2, NANOG, and LIN28] and embryonic stem cell (ESC)-specific genes (PODXL, DPPA5, FGF5, REX1, and LAMP1) and showed strong levels of alkaline phosphatase expression. In vitro differentiation by formation of embryoid bodies and by directed differentiation generated cell derivatives of all 3 germ layers as confirmed by mRNA and protein expression. In vivo, the ciPSCs created solid tumors, which failed to reach epithelial structure formation, but expressed markers for all 3 germ layers. Array comparative genomic hybridization and chromosomal fluorescence in situ hybridization analyses revealed that while retroviral transduction per se did not result in significant DNA copy number imbalance, there was evidence for the emergence of low-level aneuploidy during prolonged culture or tumor formation. In summary, we were able to derive ciPSCs from adult fibroblasts by using 4 transcription factors. The isolated iPSCs have similar characteristics to ESCs from other species, but the exact cellular mechanisms behind their unique co-dependency on both FGF2 and LIF are still unknown.
Collapse
Affiliation(s)
- Sehwon Koh
- Genomics Program, North Carolina State University, Raleigh, NC 27607, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Motta L, Mandara MT, Skerritt GC. Canine and feline intracranial meningiomas: An updated review. Vet J 2012; 192:153-65. [DOI: 10.1016/j.tvjl.2011.10.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 10/10/2011] [Accepted: 10/11/2011] [Indexed: 12/24/2022]
|
50
|
Seiser EL, Thomas R, Richards KL, Kelley MK, Moore P, Suter SE, Breen M. Reading between the lines: molecular characterization of five widely used canine lymphoid tumour cell lines. Vet Comp Oncol 2011; 11:30-50. [PMID: 22236332 DOI: 10.1111/j.1476-5829.2011.00299.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Molecular characterization of tumour cell lines is increasingly regarded as a prerequisite for defining their validity as models of in vivo neoplasia. We present the first comprehensive catalogue of genomic and transcriptional characteristics of five widely used canine lymphoid tumour cell lines. High-resolution microarray-based comparative genomic hybridization defined their unique profiles of genomic DNA copy number imbalance. Multicolour fluorescence in situ hybridization identified aberrant gains of MYC, KIT and FLT3 and deletions of PTEN and CDKN2 in individual cell lines, and also revealed examples of extensive structural chromosome reorganization. Gene expression profiling and RT-PCR analyses defined the relationship between genomic imbalance and transcriptional dysregulation in each cell line, clarifying their relevance as models of discrete functional pathways with biological and therapeutic significance. In combination, these data provide an extensive resource of molecular data for directing the appropriate use of these cell lines as tools for studying canine lymphoid neoplasia.
Collapse
Affiliation(s)
- E L Seiser
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27606, USA
| | | | | | | | | | | | | |
Collapse
|