1
|
Farrell KB, Das S, Nordeen SK, Lambert JR, Thamm DH. VDX-111 targets proliferative pathways in canine cancer cell lines. PLoS One 2024; 19:e0303470. [PMID: 38771847 PMCID: PMC11108205 DOI: 10.1371/journal.pone.0303470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/15/2024] [Indexed: 05/23/2024] Open
Abstract
VDX-111 (also identified as AMPI-109) is a vitamin D derivative which has shown anticancer activity. To further assess the function of this compound against multiple cancer types, we examined the efficacy of VDX-111 against a panel of 30 well characterized canine cancer cell lines. Across a variety of cancer types, VDX-111 induced widely variable growth inhibition, cell death, and migration inhibition, at concentrations ranging from 10 nM to 1 μM. Growth inhibition sensitivity did not correlate strongly with tumor cell histotype; however, it was significantly correlated with the expression of genes in multiple cell signaling pathways, including the MAPK and PI3K-AKT pathways. We confirmed inhibition of these signaling pathways as likely participants in the effects of VDX-111. These results suggest that a subset of canine tumors may be sensitive to treatment with VDX-111, and suggests possible predictive markers of drug sensitivity and pharmacodynamic biomarkers of drug exposure that could be employed in future clinical trials.
Collapse
Affiliation(s)
- Kristen B. Farrell
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, United States of America
| | - Sunetra Das
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, United States of America
| | - Steven K. Nordeen
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - James R. Lambert
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Douglas H. Thamm
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, United States of America
| |
Collapse
|
2
|
Schlein LJ, Brill SA, Brady RV, Farrell KB, Rose BJ, Meuten TK, Jordan CT, Thamm DH. Parthenolide As a Therapeutic for Disseminated Canine Neoplasms. J Pharmacol Exp Ther 2024; 388:774-787. [PMID: 38135509 PMCID: PMC10877710 DOI: 10.1124/jpet.123.001851] [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: 08/08/2023] [Revised: 11/09/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
This study provides a unique translational research opportunity to help both humans and dogs diagnosed with diseases that carry dismal prognoses in both species: histiocytic sarcoma (HS), hemangiosarcoma (HSA), and disseminated mastocytosis/mast cell tumor (MCT). Although exceedingly rare in humans, these so called "orphan diseases" are relatively more common in dogs. For these and other more commonplace cancers like lymphoma (Lym), dogs are an excellent translational model for human disease due to remarkably similar disease biology. In this study, assays were performed to assess the therapeutic potential of parthenolide (PTL), a known canonical nuclear factor kappa B (NF-κB) signaling inhibitor with additional mechanisms of antineoplastic activity, including alteration of cellular reduction-oxidation balance. Canine cell lines and primary cells are sensitive to PTL and undergo dose-dependent apoptosis after exposure to drug. PTL exposure also leads to glutathione depletion, reactive oxygen species generation, and NF-κB inhibition in canine cells. Standard-of-care therapeutics broadly synergize with PTL. In two canine HS cell lines, expression of NF-κB pathway signaling partners is downregulated with PTL therapy. Preliminary data suggest that PTL inhibits NF-κB activity of cells and extends survival time in a mouse model of disseminated canine HS. These data support further investigation of compounds that can antagonize canonical NF-κB pathway signaling in these cancers and pave the way for clinical trials of PTL in affected dogs. As dogs are an excellent natural disease model for these cancers, these data will ultimately improve our understanding of their human disease counterparts and hopefully improve care for both species. SIGNIFICANCE STATEMENT: Disseminated neoplasms in human and canine cancers are challenging to treat, and novel therapeutic approaches are needed to improve outcomes. Parthenolide is a promising treatment for histiocytic sarcoma, hemangiosarcoma, and mast cell neoplasia.
Collapse
Affiliation(s)
- Lisa J Schlein
- Flint Animal Cancer Center (L.J.S., S.A.B., R.V.B., K.B.F., B.J.R., T.K.M., D.H.T.) and Cell and Molecular Biology Graduate Program (L.J.S., S.A.B., R.V.B.), Colorado State University, Fort Collins, Colorado; and Anschutz Medical Campus, University of Colorado, Aurora, Colorado (C.T.J.)
| | - Samuel A Brill
- Flint Animal Cancer Center (L.J.S., S.A.B., R.V.B., K.B.F., B.J.R., T.K.M., D.H.T.) and Cell and Molecular Biology Graduate Program (L.J.S., S.A.B., R.V.B.), Colorado State University, Fort Collins, Colorado; and Anschutz Medical Campus, University of Colorado, Aurora, Colorado (C.T.J.)
| | - Rachel V Brady
- Flint Animal Cancer Center (L.J.S., S.A.B., R.V.B., K.B.F., B.J.R., T.K.M., D.H.T.) and Cell and Molecular Biology Graduate Program (L.J.S., S.A.B., R.V.B.), Colorado State University, Fort Collins, Colorado; and Anschutz Medical Campus, University of Colorado, Aurora, Colorado (C.T.J.)
| | - Kristen B Farrell
- Flint Animal Cancer Center (L.J.S., S.A.B., R.V.B., K.B.F., B.J.R., T.K.M., D.H.T.) and Cell and Molecular Biology Graduate Program (L.J.S., S.A.B., R.V.B.), Colorado State University, Fort Collins, Colorado; and Anschutz Medical Campus, University of Colorado, Aurora, Colorado (C.T.J.)
| | - Barbara J Rose
- Flint Animal Cancer Center (L.J.S., S.A.B., R.V.B., K.B.F., B.J.R., T.K.M., D.H.T.) and Cell and Molecular Biology Graduate Program (L.J.S., S.A.B., R.V.B.), Colorado State University, Fort Collins, Colorado; and Anschutz Medical Campus, University of Colorado, Aurora, Colorado (C.T.J.)
| | - Travis K Meuten
- Flint Animal Cancer Center (L.J.S., S.A.B., R.V.B., K.B.F., B.J.R., T.K.M., D.H.T.) and Cell and Molecular Biology Graduate Program (L.J.S., S.A.B., R.V.B.), Colorado State University, Fort Collins, Colorado; and Anschutz Medical Campus, University of Colorado, Aurora, Colorado (C.T.J.)
| | - Craig T Jordan
- Flint Animal Cancer Center (L.J.S., S.A.B., R.V.B., K.B.F., B.J.R., T.K.M., D.H.T.) and Cell and Molecular Biology Graduate Program (L.J.S., S.A.B., R.V.B.), Colorado State University, Fort Collins, Colorado; and Anschutz Medical Campus, University of Colorado, Aurora, Colorado (C.T.J.)
| | - Douglas H Thamm
- Flint Animal Cancer Center (L.J.S., S.A.B., R.V.B., K.B.F., B.J.R., T.K.M., D.H.T.) and Cell and Molecular Biology Graduate Program (L.J.S., S.A.B., R.V.B.), Colorado State University, Fort Collins, Colorado; and Anschutz Medical Campus, University of Colorado, Aurora, Colorado (C.T.J.)
| |
Collapse
|
3
|
Kelly B, Thamm D, Rosengren RJ. Utility of the second-generation curcumin analogue RL71 in canine histiocytic sarcoma. Vet Res Commun 2024; 48:563-568. [PMID: 37597087 PMCID: PMC10811103 DOI: 10.1007/s11259-023-10201-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 08/15/2023] [Indexed: 08/21/2023]
Abstract
Canine histiocytic sarcoma is an aggressive cancer, with a high rate of metastasis. Thus, novel therapeutic approaches are needed. Synthetic analogues of curcumin have elicited potent anti-cancer activity in multiple in vitro and in vivo models of human cancer. Furthermore, the compound 3,5-bis(3,4,5-trimethoxybenzylidene)-1-methylpiperidine-4-one (RL71) has recently exhibited potent cell cycle arrest and apoptotic induction in a canine osteosarcoma cell line. To determine its potency in canine histiocytic sarcoma cells, cell viability of DH82 and Nike cells was measured using the sulforhodamine B assay. Flow cytometry was then used to analyse both cell cycle distribution and apoptosis. Of the five curcumin analogues examined, RL71, had the highest potency with EC50 values of 0.66 ± 0.057 µM and 0.79 ± 0.13 µM in the DH82 and Nike cell lines, respectively. Furthermore, RL71 at the 1x EC50 concentration increased G2/M cell cycle arrest 2-fold, and at the 2x EC50 concentration increased the number of apoptotic cells 4-fold. These findings are consistent with previous work using RL71 in both canine and human cancer cell lines. Future research should be directed on time-dependent changes, and mechanistic investigation in greater detail to elucidate RL71 mechanisms of action.
Collapse
Affiliation(s)
- Barnaby Kelly
- Department of Pharmacology and Toxicology, Unversity of Otago, 18 Frederick Street, Dunedin, 9085, New Zealand
| | - Douglas Thamm
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, USA
| | - Rhonda J Rosengren
- Department of Pharmacology and Toxicology, Unversity of Otago, 18 Frederick Street, Dunedin, 9085, New Zealand.
| |
Collapse
|
4
|
Schlein LJ, Thamm DH. Immunohistochemical evidence of NF-kB activation in canine lymphomas, histiocytic sarcomas, hemangiosarcomas, and mast cell tumors. Vet Pathol 2024; 61:20-31. [PMID: 37357953 DOI: 10.1177/03009858231180484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
Increased or constitutive activation of nuclear factor kappa B (NF-kB) is a feature of many chronic disease processes, including cancer. While NF-kB overactivation has been documented extensively in human oncology, there is a relative paucity of data documenting the same phenomenon in veterinary medicine. To assess NF-kB activity, antibodies to p65 and p100/p52, which are components of NF-kB heterodimers, were first validated for specificity and canine cross-reactivity via Western blot and labeling of immortalized cell pellets. Then, nuclear labeling for these antibodies was assessed via QuPath software in over 200 tumor tissue samples (10 hemangiosarcomas, 94 histiocytic sarcomas, 71 lymphomas, and 28 mast cell tumors) and compared to immunolabeling in appropriate normal tissue counterparts. Greater than 70% of spontaneous canine tumors evaluated in this study had more nuclear p65 and p100/p52 immunoreactivity than was observed in comparable normal cell populations. Specifically, 144/204 (70.58%) of tumors evaluated had positive p65 nuclear labeling and 179/195 (91.79%) had positive p100/p52 nuclear labeling. Surprisingly, greater nuclear p100/p52 reactivity was associated with a longer progression-free survival (PFS) and overall survival (OS) in canine lymphomas. These results provide support and preliminary data to investigate the role of NF-kB signaling in different types of canine cancer.
Collapse
|
5
|
Kelly B, Thamm D, Rosengren RJ. The second-generation curcumin analogue RL71 elicits G2/M cell cycle arrest and apoptosis in canine osteosarcoma cells. Vet Comp Oncol 2023; 21:595-604. [PMID: 37435770 DOI: 10.1111/vco.12922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 07/13/2023]
Abstract
Canine osteosarcoma is an aggressive cancer, comprising 85% of canine bone neoplasms. Current treatment practices of surgery and chemotherapy increase 1-year survival by only 45%. The curcumin analogue RL71, has demonstrated potent in vitro and in vivo efficacy in several models of human breast cancer through increased apoptosis and cell cycle arrest. Thus, the present study aimed to investigate efficacy of curcumin analogues in two canine osteosarcoma cell lines. Osteosarcoma cell viability was assessed using the sulforhodamine B assay and mechanisms of action were determined by analysing the levels of cell cycle and apoptotic regulatory proteins via Western blotting. Further evidence was obtained using flow cytometry to detect cell cycle distribution and the number of apoptotic cells. RL71 was the most potent curcumin analogue with EC50 values of 0.64 ± 0.04 and 0.38 ± 0.009 μM (n = 3) in D-17 (commercial) and Gracie canine osteosarcoma cells, respectively. RL71 significantly increased the ratio of cleaved-caspase 3 to pro-caspase 3 and the level of apoptotic cells at the 2× and 5× EC50 concentration (p < 0.001, n = 3). Furthermore, at the same concentration, RL71 significantly increased the number of cells in the G2/M phase. In conclusion, RL71 has potent cytotoxic activity in canine osteosarcoma cells triggering G2/M arrest and apoptosis at concentrations achievable in vivo. Future research should further investigate molecular mechanisms for these changes in other canine osteosarcoma cell lines prior to in vivo investigation.
Collapse
Affiliation(s)
- Barnaby Kelly
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Douglas Thamm
- Flint Animal Cancer Center, Colorado State University, Ft. Collins, Colorado, USA
| | - Rhonda J Rosengren
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
6
|
Gustafson DL, Viola LO, Towers CG, Das S, Duval DL, Van Eaton KM. Sensitivity of osteosarcoma cell lines to autophagy inhibition as determined by pharmacologic and genetic manipulation. Vet Comp Oncol 2023; 21:726-738. [PMID: 37724007 PMCID: PMC11470750 DOI: 10.1111/vco.12937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/05/2023] [Accepted: 09/05/2023] [Indexed: 09/20/2023]
Abstract
Pharmacologic inhibition of autophagy can be achieved using lysosomotropic agents such as hydroxychloroquine (HCQ) that interfere with fusion of the autophagosome to the lysosome thus preventing completion of the recycling process. The goal of the present study is to determine the sensitivity of eight canine (cOSA) and four human (hOSA) osteosarcoma tumour cell lines to antiproliferative and cytotoxic effects of lysosomal autophagy inhibitors, and to compare these results to the autophagy-dependence measured using a CRISPR/Cas9 live-cell imaging assay in OSA and other tumour cell lines. Antiproliferative and cytotoxic response to HCQ and Lys05 was determined using live cell imaging and YOYO-1 staining. CRISPR/Cas9 live cell imaging screen was done using species specific guide RNA's and transfection of reagents into cells. Response to autophagy core genes was compared to response to an essential (PCNA) and non-essential (FOXO3A) gene. cOSA and hOSA cell lines showed similar antiproliferative and cytotoxic responses to HCQ and Lys05 with median lethal dose (Dm ) values ranging from 4.6-15.8 μM and 2.1-5.1 μM for measures of anti-proliferative response, respectively. A relationship was observed between antiproliferative responses to HCQ and Lys05 and VPS34 CRISPR score with Dm values correlating with VPS34 response (r = 0.968 and 0.887) in a species independent manner. The results show that a subset of cOSA and hOSA cell lines are autophagy-dependent and sensitive to HCQ at pharmacologically-relevant exposures.
Collapse
Affiliation(s)
- Daniel L. Gustafson
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, USA
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
- Developmental Therapeutics Program, University of Colorado Cancer Center, Aurora, Colorado, USA
| | - Lindsey O. Viola
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
| | - Christina G. Towers
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Sciences, La Jolla, California, USA
| | - Sunetra Das
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
| | - Dawn L. Duval
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
- Developmental Therapeutics Program, University of Colorado Cancer Center, Aurora, Colorado, USA
| | - Kristen M. Van Eaton
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
7
|
Das S, Idate R, Lana SE, Regan DP, Duval DL. Integrated analysis of canine soft tissue sarcomas identifies recurrent mutations in TP53, KMT genes and PDGFB fusions. Sci Rep 2023; 13:10422. [PMID: 37369741 PMCID: PMC10300023 DOI: 10.1038/s41598-023-37266-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/19/2023] [Indexed: 06/29/2023] Open
Abstract
Soft tissue sarcomas (STS) are a heterogenous group of mesenchymal tumors representing over 50 distinct types with overlapping histological features and non-specific anatomical locations. Currently, localized sarcomas are treated with surgery + / - radiation in both humans and dogs with few molecularly targeted therapeutic options. However, to improve precision-based cancer therapy through trials in pet dogs with naturally occurring STS tumors, knowledge of genomic profiling and molecular drivers in both species is essential. To this purpose, we sought to characterize the transcriptomic and genomic mutation profiles of canine STS subtypes (fibrosarcoma, undifferentiated pleomorphic sarcoma, and peripheral nerve sheath tumors), by leveraging RNAseq, whole exome sequencing, immunohistochemistry, and drug assays. The most common driver mutations were in cell cycle/DNA repair (31%, TP53-21%) and chromatin organization/binding (41%, KMT2D-21%) genes. Similar to a subset of human sarcomas, we identified fusion transcripts of platelet derived growth factor B and collagen genes that predict sensitivity to PDGFR inhibitors. Transcriptomic profiling grouped these canine STS tumors into 4 clusters, one PNST group (H1), and 3 FSA groups selectively enriched for extracellular matrix interactions and PDFGB fusions (H2), homeobox transcription factors (H3), and elevated T-cell infiltration (H4). This multi-omics approach provides insights into canine STS sub-types at a molecular level for comparison to their human counterparts, to improve diagnosis, and may provide additional targets for chemo- and immuno-therapy.
Collapse
Affiliation(s)
- Sunetra Das
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, 80523, USA
| | - Rupa Idate
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, 80523, USA
| | - Susan E Lana
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, 80523, USA
| | - Daniel P Regan
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, 80523, USA
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, 80523, USA
| | - Dawn L Duval
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA.
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, 80523, USA.
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
8
|
SONI PANKAJ, PRADHAN PRAVATAK, SOOD NEERAJ. Development, characterization and in vitro applications of a thymus cell line from Pangasianodon hypophthalmus (Sauvage 1878). THE INDIAN JOURNAL OF ANIMAL SCIENCES 2023. [DOI: 10.56093/ijans.v93i2.128796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Pangasianodon hypophthalmus is an economically important catfish species cultured in India. In the present study, a continuous cell line from the thymus of P. hypophthalmus (PHT) was established using the explant method and subcultured 52 times since development. PHT cells showed optimal growth in L-15 medium containing 20% fetal bovine serum at 28°C. The species of the cell line from striped catfish was confirmed through PCR amplification and sequencing of 16S, and COI genes. The cell line was found to be Mycoplasma free. The modal chromosome number of PHT cells was 60 (2n). Immunophenotyping using different antibodies showed the epithelial nature of the cells. Cytotoxicity of arsenic and mercury was assessed using Neutral red and MTT assay, which revealed reduced cell survival with an increase in toxicant concentration. Cells transfected successfully with the GFP reporter gene using lipofectamine reagent indicated the suitability of the cell line for expression studies. The cell line has been submitted to NRFC, Lucknow with accession no. NRFC-078 at ICAR-NBFGR, Lucknow. The developed cell line will have applications in suspected viral disease investigation, transgenic, and immunological studies.
Collapse
|
9
|
Gardner HL, Fenger JM, Roberts RD, London CA. Characterizing the metabolic role of STAT3 in canine osteosarcoma. Vet Comp Oncol 2022; 20:817-824. [PMID: 35608271 PMCID: PMC9669091 DOI: 10.1111/vco.12841] [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: 02/06/2022] [Revised: 05/05/2022] [Accepted: 05/19/2022] [Indexed: 12/30/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) dysregulation has been characterized in canine OS, with previous data suggesting that constitutive STAT3 activation contributes to survival and proliferation in OS cell lines in vitro. Recently, the contribution of STAT3 to tumour metabolism has been described across several tumour histologies, and understanding the metabolic implications of STAT3 loss may elucidate novel therapeutic approaches with synergistic activity. The objective of this work was to characterize metabolic benchmarks associated with STAT3 loss in canine OS. STAT3 expression and activation was evaluated using western blotting in canine OS cell lines OSCA8 and Abrams. STAT3 was deleted from these OS cell lines using CRISPR-Cas9, and the effects on proliferation, invasion and metabolism (respirometry, intracellular lactate) were determined. Loss of STAT3 was associated with decreased basal and compensatory glycolysis in canine OS cell lines, without modulation of cellular proliferation. Loss of STAT3 also resulted in diminished invasive capacity in vitro. Interestingly, the absence of STAT3 did not impact sensitivity to doxorubicin in vitro. Our data demonstrate that loss of STAT3 modulates features of aerobic glycolysis in canine OS impacting capacities for cellular invasions, suggesting a role for this transcription factor in metastasis.
Collapse
Affiliation(s)
- Heather L. Gardner
- Cummings School of Veterinary MedicineTufts UniversityNorth GraftonMassachusettsUSA
| | - Joelle M. Fenger
- College of Veterinary MedicineThe Ohio State UniversityColumbusOhioUSA,Present address:
Ethos Veterinary Health and Ethos Discovery (501c3)WoburnMassachusettsUSA
| | - Ryan D. Roberts
- Research Institute at Nationwide Children's HospitalColumbusOhioUSA
| | - Cheryl A. London
- Cummings School of Veterinary MedicineTufts UniversityNorth GraftonMassachusettsUSA
| |
Collapse
|
10
|
Megquier K, Turner-Maier J, Morrill K, Li X, Johnson J, Karlsson EK, London CA, Gardner HL. The genomic landscape of canine osteosarcoma cell lines reveals conserved structural complexity and pathway alterations. PLoS One 2022; 17:e0274383. [PMID: 36099278 PMCID: PMC9469990 DOI: 10.1371/journal.pone.0274383] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/25/2022] [Indexed: 01/09/2023] Open
Abstract
The characterization of immortalized canine osteosarcoma (OS) cell lines used for research has historically been based on phenotypic features such as cellular morphology and expression of bone specific markers. With the increasing use of these cell lines to investigate novel therapeutic approaches prior to in vivo translation, a much more detailed understanding regarding the genomic landscape of these lines is required to ensure accurate interpretation of findings. Here we report the first whole genome characterization of eight canine OS cell lines, including single nucleotide variants, copy number variants and other structural variants. Many alterations previously characterized in primary canine OS tissue were observed in these cell lines, including TP53 mutations, MYC copy number gains, loss of CDKN2A, PTEN, DLG2, MAGI2, and RB1 and structural variants involving SETD2, DLG2 and DMD. These data provide a new framework for understanding how best to incorporate in vitro findings generated using these cell lines into the design of future clinical studies involving dogs with spontaneous OS.
Collapse
Affiliation(s)
- Kate Megquier
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Jason Turner-Maier
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Kathleen Morrill
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Xue Li
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Jeremy Johnson
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Elinor K. Karlsson
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Cheryl A. London
- Cummings School of Veterinary Medicine at Tufts University, North Grafton, Massachusetts, United States of America
| | - Heather L. Gardner
- Cummings School of Veterinary Medicine at Tufts University, North Grafton, Massachusetts, United States of America
| |
Collapse
|
11
|
Curley TL, Thamm DH, Johnson SW, Boscan P. Effects of morphine on histamine release from two cell lines of canine mast cell tumor and on plasma histamine concentrations in dogs with cutaneous mast cell tumor. Am J Vet Res 2021; 82:1013-1018. [PMID: 34714766 DOI: 10.2460/ajvr.20.08.0137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To determine the effects of morphine on histamine release from 2 canine mast cell tumor (MCT) cell lines and on plasma histamine concentrations in dogs with cutaneous MCTs. ANIMALS 10 dogs with cutaneous MCT and 10 dogs with soft tissue sarcoma (STS). PROCEDURES The study consisted of 2 phases. First, 2 canine MCT cell lines were exposed to 3 pharmacologically relevant morphine concentrations, and histamine concentrations were determined by an ELISA. Second, dogs with MCT or STS received 0.5 mg of morphine/kg, IM, before surgery for tumor excision. Clinical signs, respiratory rate, heart rate, arterial blood pressure, rectal temperature, and plasma histamine concentrations were recorded before and 5, 15, 30, and 60 minutes after morphine administration but prior to surgery. Data were compared by use of a 2-way ANOVA with the Sidak multiple comparisons test. RESULTS In the first phase, canine MCT cell lines did not release histamine when exposed to pharmacologically relevant morphine concentrations. In the second phase, no differences were noted for heart rate, arterial blood pressure, and rectal temperature between MCT and STS groups. Plasma histamine concentrations did not significantly differ over time within groups and between groups. CONCLUSIONS AND CLINICAL RELEVANCE No significant changes in histamine concentrations were noted for both in vitro and in vivo study phases, and no hemodynamic changes were noted for the in vivo study phase. These preliminary results suggested that morphine may be used safely in some dogs with MCT.
Collapse
Affiliation(s)
- Taylor L Curley
- From the Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80521
| | - Douglas H Thamm
- From the Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80521
| | - Sam W Johnson
- From the Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80521
| | - Pedro Boscan
- From the Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80521
| |
Collapse
|
12
|
Das S, Idate R, Regan DP, Fowles JS, Lana SE, Thamm DH, Gustafson DL, Duval DL. Immune pathways and TP53 missense mutations are associated with longer survival in canine osteosarcoma. Commun Biol 2021; 4:1178. [PMID: 34635775 PMCID: PMC8505454 DOI: 10.1038/s42003-021-02683-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 09/15/2021] [Indexed: 12/20/2022] Open
Abstract
Osteosarcoma affects about 2.8% of dogs with cancer, with a one-year survival rate of approximately 45%. The purpose of this study was to characterize mutation and expression profiles of osteosarcoma and its association with outcome in dogs. The number of somatic variants identified across 26 samples ranged from 145 to 2,697 with top recurrent mutations observed in TP53 and SETD2. Additionally, 47 cancer genes were identified with copy number variations. Missense TP53 mutation status and low pre-treatment blood monocyte counts were associated with a longer disease-free interval (DFI). Patients with longer DFI also showed increased transcript levels of anti-tumor immune response genes. Although, T-cell and myeloid cell quantifications were not significantly associated with outcome; immune related genes, PDL-1 and CD160, were correlated with T-cell abundance. Overall, the association of gene expression and mutation profiles to outcome provides insights into pathogenesis and therapeutic interventions in osteosarcoma patients.
Collapse
Affiliation(s)
- Sunetra Das
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA.
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, 80523, USA.
| | - Rupa Idate
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, 80523, USA
| | - Daniel P Regan
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, 80523, USA
- Department of Microbiology, Immunology, & Pathology, Colorado State University, Fort Collins, CO, 80523, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Jared S Fowles
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, 80523, USA
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, 80523, USA
| | - Susan E Lana
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, 80523, USA
| | - Douglas H Thamm
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, 80523, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, 80045, USA
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, 80523, USA
| | - Daniel L Gustafson
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, 80523, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, 80045, USA
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, 80523, USA
| | - Dawn L Duval
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA.
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, 80523, USA.
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, 80523, USA.
| |
Collapse
|
13
|
Park H, Gladstone M, Shanley C, Goodrich R, Guth A. A novel cancer immunotherapy utilizing autologous tumour tissue. Vox Sang 2020; 115:525-535. [PMID: 32378223 PMCID: PMC8344074 DOI: 10.1111/vox.12935] [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: 12/02/2019] [Revised: 02/18/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND With the recent interest in personalized medicine for cancer patients and immune therapy, the field of cancer vaccines has been resurrected. Previous autologous, whole cell tumour vaccine trials have not produced convincing results due, in part to poor patient selection and inactivation methos that are harsh on the cells. These methods can alter protein structure and antigenic profiles making vaccine candidates ineffective in stimulating immune response to autochthonous tumour cells. MATERIALS AND METHODS We investigated a novel method for inactivating tumour cells that uses UVA/UVB light and riboflavin (vitamin B2) (RF + UV). RF + UV inactivates the tumour cells' ability to replicate, yet preserves tumour cell integrity and antigenicity. RESULTS Our results demonstrate that proteins are preserved on the surface of RF + UV-inactivated tumour cells and that they are immunogenic via induction of dendritic cell maturation, increase in IFNγ production and generation of tumour cell-specific IgG. Moreover, when formulated with an adjuvant ('Innocell vaccine') and tested in different murine tumour primary and metastatic disease models, decreased tumour growth, decreased metastatic disease and prolonged survival were observed. In addition, immune cells obtained from tumour tissue following vaccination had decreased exhausted and regulatory T cells, suggesting that activation of intra-tumoural T cells may be playing a role leading to reduced tumour growth. CONCLUSIONS These data suggest that the RF + UV inactivation of tumour cells may provide an efficacious method for generating autologous whole tumour cell vaccines for use in cancer patients.
Collapse
Affiliation(s)
- Haemin Park
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, USA
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | | | - Crystal Shanley
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Raymond Goodrich
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Amanda Guth
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
14
|
Das S, Idate R, Cronise KE, Gustafson DL, Duval DL. Identifying Candidate Druggable Targets in Canine Cancer Cell Lines Using Whole-Exome Sequencing. Mol Cancer Ther 2019; 18:1460-1471. [PMID: 31175136 DOI: 10.1158/1535-7163.mct-18-1346] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/15/2019] [Accepted: 05/24/2019] [Indexed: 12/15/2022]
Abstract
Cancer cell culture has been a backbone in cancer research, in which analysis of human cell line mutational profiles often correlates with oncogene addiction and drug sensitivity. We have conducted whole-exome sequence analyses on 33 canine cancer cell lines from 10 cancer types to identify somatic variants that contribute to pathogenesis and therapeutic sensitivity. A total of 66,344 somatic variants were identified. Mutational load ranged from 15.79 to 129.37 per Mb, and 13.2% of variants were located in protein-coding regions (PCR) of 5,085 genes. PCR somatic variants were identified in 232 genes listed in the Cancer Gene Census (COSMIC). Cross-referencing variants with human driving mutations on cBioPortal identified 61 variants as candidate cancer drivers in 30 cell lines. The most frequently mutated cancer driver was TP53 (15 mutations in 12 cell lines). No drivers were identified in three cell lines. We identified 501 non-COSMIC genes with PCR variants that functionally annotate with COSMIC genes. These genes frequently mapped to the KEGG MAPK and PI3K-AKT pathways. We evaluated the cell lines for ERK1/2 and AKT(S473) phosphorylation and sensitivity to the MEK1/2 inhibitor, trametinib. Twelve of the 33 cell lines were trametinib-sensitive (IC50 < 32 nmol/L), all 12 exhibited constitutive or serum-activated ERK1/2 phosphorylation, and 8 carried MAPK pathway cancer driver variants: NF1(2), BRAF(3), N/KRAS(3). This functionally annotated database of canine cell line variants will inform hypothesis-driven preclinical research to support the use of companion animals in clinical trials to test novel combination therapies.
Collapse
Affiliation(s)
- Sunetra Das
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado. .,Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado
| | - Rupa Idate
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado.,Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado
| | - Kathryn E Cronise
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado.,Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado.,Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, Colorado
| | - Daniel L Gustafson
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado.,Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado.,University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado
| | - Dawn L Duval
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado.,Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado.,University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
15
|
Cronise KE, Hernandez BG, Gustafson DL, Duval DL. Identifying the ErbB/MAPK Signaling Cascade as a Therapeutic Target in Canine Bladder Cancer. Mol Pharmacol 2019; 96:36-46. [PMID: 31048548 DOI: 10.1124/mol.119.115808] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/27/2019] [Indexed: 12/19/2022] Open
Abstract
Transitional cell carcinoma (TCC) of the bladder comprises 2% of diagnosed canine cancers. TCC tumors are generally inoperable and unresponsive to traditional chemotherapy, indicating a need for more effective therapies. BRAF, a kinase in the mitogen-activated protein kinase (MAPK) pathway, is mutated in 70% of canine TCCs. In this study, we use BRAF mutant and wild-type TCC cell lines to characterize the role of BRAF mutations in TCC pathogenesis and assess the efficacy of inhibition of the MAPK pathway alone and in combination with other gene targets as a treatment for canine TCC. Analysis of MAPK target gene expression and assessment of extracellular signal-regulated kinase (ERK) 1/2 phosphorylation following serum starvation indicated constitutive MAPK activity in all TCC cell lines. BRAF mutant TCC cell lines were insensitive to the BRAF inhibitor vemurafenib, with IC50 values greater than 5 μM, but exhibited greater sensitivity to a paradox-breaking BRAF inhibitor (IC50: 0.2-1 μM). All TCC cell lines had IC50 values less than 7 nM to the mitogen-activated protein kinase kinase (MEK) 1/2 inhibitor trametinib independent of their BRAF mutation status. ERK1/2 phosphorylation decreased after 6-hour treatments with MAPK inhibitors, but rebounded by 24 hours, suggesting the presence of resistance mechanisms. Microarray analysis identified elevated expression of the ErbB family of receptors and ligands in TCC cell lines. The pan-ErbB inhibitor sapitinib synergized with BRAF inhibition in BRAF mutant Bliley TCC cells and synergized with MEK1/2 inhibition in Bliley and BRAF wild-type Kinsey cells. These findings suggest the potential for combined MAPK and ErbB receptor inhibition as a therapy for canine TCC. SIGNIFICANCE STATEMENT: The results of this study (1) identify a novel combination strategy for canine bladder cancer treatment: targeting the ErbB/MAPK signaling cascade and (2) establish the utility of canine bladder cancer as a naturally-occurring model for human MAPK-driven cancers.
Collapse
Affiliation(s)
- Kathryn E Cronise
- Flint Animal Cancer Center, Department of Clinical Sciences (K.E.C., B.G.H., D.L.G., D.L.D.), and Cell and Molecular Biology Graduate Program (K.E.C., D.L.G., D.L.D.), Colorado State University, Fort Collins, Colorado; and University of Colorado Cancer Center, Aurora, Colorado (D.L.G., D.L.D.)
| | - Belen G Hernandez
- Flint Animal Cancer Center, Department of Clinical Sciences (K.E.C., B.G.H., D.L.G., D.L.D.), and Cell and Molecular Biology Graduate Program (K.E.C., D.L.G., D.L.D.), Colorado State University, Fort Collins, Colorado; and University of Colorado Cancer Center, Aurora, Colorado (D.L.G., D.L.D.)
| | - Daniel L Gustafson
- Flint Animal Cancer Center, Department of Clinical Sciences (K.E.C., B.G.H., D.L.G., D.L.D.), and Cell and Molecular Biology Graduate Program (K.E.C., D.L.G., D.L.D.), Colorado State University, Fort Collins, Colorado; and University of Colorado Cancer Center, Aurora, Colorado (D.L.G., D.L.D.)
| | - Dawn L Duval
- Flint Animal Cancer Center, Department of Clinical Sciences (K.E.C., B.G.H., D.L.G., D.L.D.), and Cell and Molecular Biology Graduate Program (K.E.C., D.L.G., D.L.D.), Colorado State University, Fort Collins, Colorado; and University of Colorado Cancer Center, Aurora, Colorado (D.L.G., D.L.D.)
| |
Collapse
|
16
|
Borlle L, Dergham A, Wund Z, Zumbo B, Southard T, Hume KR. Salinomycin decreases feline sarcoma and carcinoma cell viability when combined with doxorubicin. BMC Vet Res 2019; 15:36. [PMID: 30678671 PMCID: PMC6346515 DOI: 10.1186/s12917-019-1780-5] [Citation(s) in RCA: 5] [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: 02/06/2018] [Accepted: 01/14/2019] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Cancer is a significant health threat in cats. Chemoresistance is prevalent in solid tumors. The ionophore salinomycin has anti-cancer properties and may work synergistically with chemotherapeutics. The purpose of our study was to determine if salinomycin could decrease cancer cell viability when combined with doxorubicin in feline sarcoma and carcinoma cells. RESULTS We established two new feline injection-site sarcoma cell lines, B4 and C10, and confirmed their tumorigenic potential in athymic nude mice. B4 was more resistant to doxorubicin than C10. Dose-dependent effects were not observed until 92 μM in B4 cells (p = 0.0006) vs. 9.2 μM (p = 0.0004) in C10 cells. Dose-dependent effects of salinomycin were observed at 15 μM in B4 cells (p = 0.025) and at 10 μM in C10 cells (p = 0.020). Doxorubicin plus 5 μM salinomycin decreased viability of B4 cells compared to either agent alone, but only at supra-pharmacological doxorubicin concentrations. However, doxorubicin plus 5 μM salinomycin decreased viability of C10 cells compared to either agent alone at doxorubicin concentrations that can be achieved in vivo (1.84 and 4.6 μM, p < 0.004). In SCCF1 cells, dose-dependent effects of doxorubicin and salinomycin were observed at 9.2 (p = 0.036) and 2.5 (p = 0.0049) μM, respectively. When doxorubicin was combined with either 1, 2.5, or 5 μM of salinomycin in SCCF1 cells, dose-dependent effects of doxorubicin were observed at 9.2 (p = 0.0021), 4.6 (p = 0.0042), and 1.84 (p = 0.0021) μM, respectively. Combination index calculations for doxorubicin plus 2.5 and 5 μM salinomycin in SCCF1 cells were 0.4 and 0.6, respectively. CONCLUSIONS We have developed two new feline sarcoma cell lines that can be used to study chemoresistance. We observed that salinomycin may potentiate (C10 cells) or work synergistically (SCCF1 cells) with doxorubicin in certain feline cancer cells. Further research is indicated to understand the mechanism of action of salinomycin in feline cancer cells as well as potential tolerability and toxicity in normal feline tissues.
Collapse
Affiliation(s)
- Lucia Borlle
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853 USA
- Department of Animal Sciences, Cornell University College of Agricultural and Life Sciences, Ithaca, NY 14853 USA
| | - Abdo Dergham
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853 USA
| | - Zacharie Wund
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853 USA
| | - Brittany Zumbo
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853 USA
| | - Teresa Southard
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853 USA
| | - Kelly R. Hume
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853 USA
| |
Collapse
|
17
|
Hartley G, Elmslie R, Murphy B, Hopkins L, Guth A, Dow S. Cancer stem cell populations in lymphoma in dogs and impact of cytotoxic chemotherapy. Vet Comp Oncol 2018; 17:69-79. [PMID: 30238600 DOI: 10.1111/vco.12447] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 12/12/2022]
Abstract
Cancer relapse following chemotherapy has been attributed in part to the presence of cancer stem cells (CSC), which drive tumour growth and metastasis and are highly resistant to the effects of cytotoxic chemotherapy. As a result, treatment with cytotoxic chemotherapy selects for drug-resistant CSC populations that eventually drive tumour recurrence. Little is known currently regarding the role of CSC in dogs with lymphoma, nor the impact of chemotherapy on CSC populations. Therefore, we prospectively quantitated CSC populations in dogs with B-cell (BCL) and T-cell lymphoma (TCL), using tumour aspirates and flow cytometric analysis with a panel of CSC markers. In addition, in vitro studies were carried out to determine the impact of chemotherapy resistance on the stem cell phenotype and stem cell properties of lymphoma cells. We found that the percentages of tumour cells expressing CSC markers were significantly increased in dogs with BCL, compared with B cells from normal lymph nodes. Similar findings were observed in dogs with TCL. In vitro studies revealed that lymphoma cells selected for resistance to CHOP chemotherapy had significantly upregulated expression of CSC markers, formed spheroids in culture more readily, and expressed significantly greater aldehyde dehydrogenase activity compared with chemotherapy-sensitive tumour cells. Similar results were observed in tumour samples dogs with relapsed BCL. These findings suggest that cytotoxic chemotherapy can lead to a relative enrichment of tumour cells with CSC properties, which may be associated with lymphoma recurrence.
Collapse
Affiliation(s)
- Genevieve Hartley
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado
| | - Robyn Elmslie
- Veterinary Specialty and Emergency Hospital, Englewood, Colorado
| | - Brent Murphy
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado
| | - Leone Hopkins
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado
| | - Amanda Guth
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado
| | - Steven Dow
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
18
|
Pyuen AA, Meuten T, Rose BJ, Thamm DH. In vitro effects of PI3K/mTOR inhibition in canine hemangiosarcoma. PLoS One 2018; 13:e0200634. [PMID: 30011343 PMCID: PMC6047806 DOI: 10.1371/journal.pone.0200634] [Citation(s) in RCA: 6] [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: 11/08/2017] [Accepted: 06/29/2018] [Indexed: 02/07/2023] Open
Abstract
While extremely rare in humans, hemangiosarcoma (HSA) accounts for nearly 2% of canine neoplasia, and is characterized by both aggressive local growth/invasion and a high rate of metastasis. Both canine and human HSA exhibit sustained aberrant PI3K/Akt/mTOR pathway signaling. The purpose of this study was to examine the in vitro effects of a novel dual PI3K/mTOR inhibitor, VDC-597, in three canine HSA cell lines (DEN-, CIN-, and SB-HSA). VDC-597 suppressed activation of both Akt and 4eBP1 in canine HSA cells in a dose-dependent fashion, with an IC50 of approximately 0.3 uM, a concentration predicted to be clinically achievable based on preliminary early-phase canine and human studies. VDC-597 dose-dependently reduced proliferation, migration, and vascular endothelial growth factor production in HSA cells, while promoting tumor cell apoptosis. VDC-597 demonstrated additive antiproliferative effects when combined with doxorubicin. These results suggest that inhibitors of the PI3K/mTOR pathway may act against multiple components of the neoplastic process, including proliferation/apoptosis, chemosensitivity, migration, and angiogenesis, and justify the evaluation of PI3K/mTOR inhibitors in canine, and potentially human, HSA.
Collapse
Affiliation(s)
- Alex A. Pyuen
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, United States of America
| | - Travis Meuten
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
| | - Barbara J. Rose
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
| | - Douglas H. Thamm
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, United States of America
- Comprehensive Cancer Center, University of Colorado, Aurora, CO, United States of America
- * E-mail:
| |
Collapse
|
19
|
Abstract
The Cellosaurus is a knowledge resource on cell lines. It aims to describe all cell lines used in biomedical research. Its scope encompasses both vertebrates and invertebrates. Currently, information for >100,000 cell lines is provided. For each cell line, it provides a wealth of information, cross-references, and literature citations. The Cellosaurus is available on the ExPASy server (https://web.expasy.org/cellosaurus/) and can be downloaded in a variety of formats. Among its many uses, the Cellosaurus is a key resource to help researchers identify potentially contaminated/misidentified cell lines, thus contributing to improving the quality of research in the life sciences.
Collapse
Affiliation(s)
- Amos Bairoch
- Computer and Laboratory Investigation of Proteins of Human Origin Group, Faculty of Medicine, Swiss Institute of Bioinformatics, University of Geneva, Geneva 4, Switzerland
| |
Collapse
|
20
|
Zhang H, Rose BJ, Pyuen AA, Thamm DH. In vitro antineoplastic effects of auranofin in canine lymphoma cells. BMC Cancer 2018; 18:522. [PMID: 29724201 PMCID: PMC5934856 DOI: 10.1186/s12885-018-4450-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 04/27/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The orally available gold complex auranofin (AF) has been used in humans, primarily as an antirheumatic/immunomodulatory agent. It has been safely administered to healthy dogs to establish pharmacokinetic parameters for oral administration, and has also been used as a treatment in some dogs with immune-mediated conditions. Multiple in vitro studies have recently suggested that AF may possess antineoplastic properties. Spontaneous canine lymphoma may be a very useful translational model for the study of human lymphoma, prompting the evaluation of AF in canine lymphoma cells. METHODS We investigated the antineoplastic activity of AF in 4 canine lymphoid tumor derived cell lines through measurements of proliferation, apoptosis, thioredoxin reductase (TrxR) activity and generation of reactive oxygen species (ROS), and detected the effects of AF when combined with conventional cytotoxic drugs using the Chou and Talalay method. We also evaluated the antiproliferative effects of AF in primary canine lymphoma cells using a bioreductive fluorometric assay. RESULTS At concentrations that appear clinically achievable in humans, AF demonstrated potent antiproliferative and proapoptotic effects in canine lymphoid tumor cell lines. TrxR inhibition and increased ROS production was observed following AF treatment. Moreover, a synergistic antiproliferative effect was observed when AF was combined with lomustine or doxorubicin. CONCLUSIONS Auranofin appears to inhibit the growth and initiate apoptosis in canine lymphoma cells in vitro at clinically achievable concentrations. Therefore, this agent has the potential to have near-term benefit for the treatment of canine lymphoma, as well as a translational model for human lymphoma. Decreased TrxR activity and increasing ROS production may be useful biomarkers of drug exposure.
Collapse
Affiliation(s)
- Hong Zhang
- Department of Veterinary Clinical Science, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Barbara J Rose
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, 300 West Drake Road, Fort Collins, CO, 80523-1620, USA
| | - Alex A Pyuen
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, 300 West Drake Road, Fort Collins, CO, 80523-1620, USA.,Present Address: Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, The University of Georgia, 2200 College Station Rd, Athens, GA, 30602, USA
| | - Douglas H Thamm
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, 300 West Drake Road, Fort Collins, CO, 80523-1620, USA. .,Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, USA. .,Comprehensive Cancer Center, University of Colorado, Aurora, CO, USA.
| |
Collapse
|
21
|
Parrales A, McDonald P, Ottomeyer M, Roy A, Shoenen FJ, Broward M, Bruns T, Thamm DH, Weir SJ, Neville KA, Iwakuma T, Fulbright JM. Comparative oncology approach to drug repurposing in osteosarcoma. PLoS One 2018; 13:e0194224. [PMID: 29579058 PMCID: PMC5868798 DOI: 10.1371/journal.pone.0194224] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 02/27/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Osteosarcoma is an orphan disease for which little improvement in survival has been made since the late 1980s. New drug discovery for orphan diseases is limited by the cost and time it takes to develop new drugs. Repurposing already approved FDA-drugs can help overcome this limitation. Another limitation of cancer drug discovery is the lack of preclinical models that accurately recapitulate what occurs in humans. For OS using dogs as a model can minimize this limitation as OS in canines develops spontaneously, is locally invasive and metastasizes to the lungs as it does in humans. METHODS In our present work we used high-throughput screens to identify drugs from a library of 2,286 FDA-approved drugs that demonstrated selective growth inhibition against both human and canine OS cell lines. The identified lead compound was then tested for synergy with 7 other drugs that have demonstrated activity against OS. These results were confirmed with in vitro assays and an in vivo murine model of OS. RESULTS We identified 13 drugs that demonstrated selective growth inhibition against both human and canine OS cell lines. Auranofin was selected for further in vitro combination drug screens. Auranofin showed synergistic effects with vorinostat and rapamycin on OS viability and apoptosis induction. Auranofin demonstrated single-agent growth inhibition in both human and canine OS xenografts, and cooperative growth inhibition was observed in combination with rapamycin or vorinostat. There was a significant decrease in Ki67-positive cells and an increase in cleaved caspase-3 levels in tumor tissues treated with a combination of auranofin and vorinostat or rapamycin. CONCLUSIONS Auranofin, alone or in combination with rapamycin or vorinostat, may be useful new treatment strategies for OS. Future studies may evaluate the efficacy of auranofin in dogs with OS as a prelude to human clinical evaluation.
Collapse
Affiliation(s)
- Alejandro Parrales
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Peter McDonald
- High Throughput Screening Laboratory, University of Kansas Cancer Center, University of Kansas, Lawrence, Kansas, United States of America
| | - Megan Ottomeyer
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Anuradha Roy
- High Throughput Screening Laboratory, University of Kansas Cancer Center, University of Kansas, Lawrence, Kansas, United States of America
- University of Kansas Cancer Center, Kansas City, Kansas, United States of America
| | - Frank J. Shoenen
- University of Kansas Cancer Center, Kansas City, Kansas, United States of America
- Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas, United States of America
| | - Melinda Broward
- University of Kansas Cancer Center, Kansas City, Kansas, United States of America
- Institute for Advancing Medical Innovation, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Tyce Bruns
- Institute for Advancing Medical Innovation, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Douglas H. Thamm
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, United States of America
- University of Colorado Comprehensive Cancer Center, Aurora, Colorado, United States of America
| | - Scott J. Weir
- University of Kansas Cancer Center, Kansas City, Kansas, United States of America
- Institute for Advancing Medical Innovation, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- Department of Pediatrics, University of Missouri Kansas City, Kansas City, Missouri, United States of America
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas, Kansas City, Kansas, United States of America
| | - Kathleen A. Neville
- Arkansas Children’s Hospital, Little Rock, Arkansas, United States of America
| | - Tomoo Iwakuma
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- University of Kansas Cancer Center, Kansas City, Kansas, United States of America
- Division of Hematology and Oncology, Children’s Mercy Hospital and Clinics, Kansas City, Missouri, United States of America
| | - Joy M. Fulbright
- University of Kansas Cancer Center, Kansas City, Kansas, United States of America
- Department of Pediatrics, University of Missouri Kansas City, Kansas City, Missouri, United States of America
- Division of Hematology and Oncology, Children’s Mercy Hospital and Clinics, Kansas City, Missouri, United States of America
| |
Collapse
|
22
|
Regulation of PD-L1 expression on murine tumor-associated monocytes and macrophages by locally produced TNF-α. Cancer Immunol Immunother 2017; 66:523-535. [PMID: 28184968 DOI: 10.1007/s00262-017-1955-5] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/15/2017] [Indexed: 12/22/2022]
Abstract
PD-L1 is an immune checkpoint protein that has emerged as a major signaling molecule involved with tumor escape from T cell immune responses. Studies have shown that intra-tumoral expression of PD-L1 can inhibit antitumor immune responses. However, it has recently been shown that expression of PD-L1 on myeloid cells from the tumor is a stronger indicator of prognosis than tumor cell PD-L1 expression. Therefore, it is important to understand the factors that govern the regulation of PD-L1 expression on tumor-infiltrating myeloid cells. We found that immature bone marrow monocytes in tumor-bearing mice had low levels of PD-L1 expression, while higher levels of expression were observed on monocytes in circulation. In contrast, macrophages found in tumor tissues expressed much higher levels of PD-L1 than circulating monocytes, implying upregulation by the tumor microenvironment. We demonstrated that tumor-conditioned media strongly induced increased PD-L1 expression by bone marrow-derived monocytes and TNF-α to be a cytokine that causes an upregulation of PD-L1 expression by the monocytes. Furthermore, we found production of TNF-α by the monocytes themselves to be a TLR2-dependent response to versican secreted by tumor cells. Thus, PD-L1 expression by tumor macrophages appears to be regulated in a different manner than by tumor cells themselves.
Collapse
|
23
|
Thamm DH, Hayes DF, Meuten T, Laver T, Thomas DG. Epithelial Cell Adhesion Molecule Expression in Canine Tumours. J Comp Pathol 2016; 155:299-304. [PMID: 27567927 DOI: 10.1016/j.jcpa.2016.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/09/2016] [Accepted: 07/21/2016] [Indexed: 01/12/2023]
Abstract
Epithelial cell adhesion molecule (EpCAM) is expressed in most human normal and neoplastic tissues of epithelial derivation and may have an association with tumour cell aggressiveness, a stem cell-like phenotype and clinical outcome. Antibody-based strategies for the targeting and capture of EpCAM-expressing tumour cells are showing promise, both as diagnostic tools and potential therapies. The aim of this study was to assess EpCAM expression in canine tumours. EpCAM expression was assessed in tumour cell lines via gene expression profiling and in formalin-fixed and paraffin wax-embedded tissues from canine carcinomas representing various anatomical sites by immunohistochemistry. EpCAM mRNA expression was higher in cell lines from carcinomas than those derived from sarcomas or haemopoietic tumours. EpCAM was expressed by >2/3 of tumour cells in 71% of canine carcinomas evaluated, irrespective of histotype, with the exception of carcinomas of the adrenal gland. Canine sarcomas and haemopoietic tumours were uniformly negative. Most canine carcinomas express EpCAM and so could be suitable for the study of EpCAM-directed diagnostics and therapeutics.
Collapse
Affiliation(s)
- D H Thamm
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; Cell and Molecular Biology Graduate Program, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; University of Colorado Comprehensive Cancer Center, 1665 N. Ursula St., Aurora, CO, USA.
| | - D F Hayes
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive SPC 5942, Ann Arbor, MI, USA
| | - T Meuten
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - T Laver
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - D G Thomas
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive SPC 5942, Ann Arbor, MI, USA
| |
Collapse
|
24
|
Combination Therapy with Zoledronic Acid and Parathyroid Hormone Improves Bone Architecture and Strength following a Clinically-Relevant Dose of Stereotactic Radiation Therapy for the Local Treatment of Canine Osteosarcoma in Athymic Rats. PLoS One 2016; 11:e0158005. [PMID: 27332712 PMCID: PMC4917251 DOI: 10.1371/journal.pone.0158005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/08/2016] [Indexed: 01/24/2023] Open
Abstract
Clinical studies using definitive-intent stereotactic radiation therapy (SRT) for the local treatment of canine osteosarcoma (OSA) have shown canine patients achieving similar median survival times as the current standard of care (amputation and adjuvant chemotherapy). Despite this, there remains an unacceptable high risk of pathologic fracture following radiation treatment. Zoledronic acid (ZA) and parathyroid hormone (PTH) are therapeutic candidates for decreasing this fracture risk post-irradiation. Due to differing mechanisms, we hypothesized that the combined treatment with ZA and PTH would significantly improve bone healing more than ZA or PTH treatment alone. Using an orthotopic model of canine osteosarcoma in athymic rats, we evaluated bone healing following clinically-relevant doses of radiation therapy (12 Gy x 3 fractions, 36 Gy total). Groups included 36 Gy SRT only, 36 Gy SRT plus ZA, 36 Gy SRT plus ZA and PTH, 36 Gy SRT plus PTH, and 36 Gy SRT plus localized PTH treatment. Our study showed significant increases in bone volume and increased polar moments of inertia (in the distal femoral metaphysis) 8 weeks after radiation in the combined (ZA/PTH) treatment group as compared to radiation treatment alone. Histomorphometric analysis revealed evidence of active mineralization at the study endpoint as well as successful tumor-cell kill across all treatment groups. This work provides further evidence for the expanding potential indications for ZA and PTH therapy, including post-irradiated bone disease due to osteosarcoma.
Collapse
|
25
|
Fowles JS, Dailey DD, Gustafson DL, Thamm DH, Duval DL. The Flint Animal Cancer Center (FACC) Canine Tumour Cell Line Panel: a resource for veterinary drug discovery, comparative oncology and translational medicine. Vet Comp Oncol 2016; 15:481-492. [PMID: 27197945 DOI: 10.1111/vco.12192] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 10/09/2015] [Accepted: 10/12/2015] [Indexed: 01/22/2023]
Abstract
Mammalian cell tissue culture has been a critical tool leading to our current understanding of cancer including many aspects of cellular transformation, growth and response to therapies. The current use of large panels of cell lines with associated phenotypic and genotypic information now allows for informatics approaches and in silico screens to rapidly test hypotheses based on simple as well as complex relationships. Current cell line panels with large amounts of associated drug sensitivity and genomics data are comprised of human cancer cell lines (i.e. NCI60 and GDSC). There is increased recognition of the contribution of canine cancer to comparative cancer research as a spontaneous large animal model with application in basic and translational studies. We have assembled a panel of canine cancer cell lines to facilitate studies in canine cancer and report here phenotypic and genotypic data associated with these cells.
Collapse
Affiliation(s)
- J S Fowles
- Cell and Molecular Biology Program, Department of Clinical Sciences, Colorado State University, Fort Collins, CO, USA.,Flint Animal Cancer Center, Veterinary Medical Center, Colorado State University, Fort Collins, CO, USA
| | - D D Dailey
- Cell and Molecular Biology Program, Department of Clinical Sciences, Colorado State University, Fort Collins, CO, USA.,Flint Animal Cancer Center, Veterinary Medical Center, Colorado State University, Fort Collins, CO, USA
| | - D L Gustafson
- Cell and Molecular Biology Program, Department of Clinical Sciences, Colorado State University, Fort Collins, CO, USA.,Flint Animal Cancer Center, Veterinary Medical Center, Colorado State University, Fort Collins, CO, USA.,University of Colorado Comprehensive Cancer Center, University of Colorado, Denver, CO, USA
| | - D H Thamm
- Cell and Molecular Biology Program, Department of Clinical Sciences, Colorado State University, Fort Collins, CO, USA.,Flint Animal Cancer Center, Veterinary Medical Center, Colorado State University, Fort Collins, CO, USA.,University of Colorado Comprehensive Cancer Center, University of Colorado, Denver, CO, USA
| | - D L Duval
- Cell and Molecular Biology Program, Department of Clinical Sciences, Colorado State University, Fort Collins, CO, USA.,Flint Animal Cancer Center, Veterinary Medical Center, Colorado State University, Fort Collins, CO, USA.,University of Colorado Comprehensive Cancer Center, University of Colorado, Denver, CO, USA
| |
Collapse
|
26
|
Fowles JS, Brown KC, Hess AM, Duval DL, Gustafson DL. Intra- and interspecies gene expression models for predicting drug response in canine osteosarcoma. BMC Bioinformatics 2016; 17:93. [PMID: 26892349 PMCID: PMC4759767 DOI: 10.1186/s12859-016-0942-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 02/10/2016] [Indexed: 01/05/2023] Open
Abstract
Background Genomics-based predictors of drug response have the potential to improve outcomes associated with cancer therapy. Osteosarcoma (OS), the most common primary bone cancer in dogs, is commonly treated with adjuvant doxorubicin or carboplatin following amputation of the affected limb. We evaluated the use of gene-expression based models built in an intra- or interspecies manner to predict chemosensitivity and treatment outcome in canine OS. Models were built and evaluated using microarray gene expression and drug sensitivity data from human and canine cancer cell lines, and canine OS tumor datasets. The “COXEN” method was utilized to filter gene signatures between human and dog datasets based on strong co-expression patterns. Models were built using linear discriminant analysis via the misclassification penalized posterior algorithm. Results The best doxorubicin model involved genes identified in human lines that were co-expressed and trained on canine OS tumor data, which accurately predicted clinical outcome in 73 % of dogs (p = 0.0262, binomial). The best carboplatin model utilized canine lines for gene identification and model training, with canine OS tumor data for co-expression. Dogs whose treatment matched our predictions had significantly better clinical outcomes than those that didn’t (p = 0.0006, Log Rank), and this predictor significantly associated with longer disease free intervals in a Cox multivariate analysis (hazard ratio = 0.3102, p = 0.0124). Conclusions Our data show that intra- and interspecies gene expression models can successfully predict response in canine OS, which may improve outcome in dogs and serve as pre-clinical validation for similar methods in human cancer research. Electronic supplementary material The online version of this article (doi:10.1186/s12859-016-0942-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jared S Fowles
- Cell and Molecular Biology Program, Department of Clinical Sciences, Colorado State University, Fort Collins, CO, USA.,Flint Animal Cancer Center, Veterinary Medical Center, Colorado State University, Fort Collins, CO, USA
| | - Kristen C Brown
- Cell and Molecular Biology Program, Department of Biology, Colorado State University, Fort Collins, CO, USA
| | - Ann M Hess
- Department of Statistics, Colorado State University, Fort Collins, CO, USA
| | - Dawn L Duval
- Cell and Molecular Biology Program, Department of Clinical Sciences, Colorado State University, Fort Collins, CO, USA.,Flint Animal Cancer Center, Veterinary Medical Center, Colorado State University, Fort Collins, CO, USA
| | - Daniel L Gustafson
- Cell and Molecular Biology Program, Department of Clinical Sciences, Colorado State University, Fort Collins, CO, USA. .,Flint Animal Cancer Center, Veterinary Medical Center, Colorado State University, Fort Collins, CO, USA. .,Shipley University Chair in Comparative Oncology, Flint Animal Cancer Center, Room 246, Colorado State University VMC, 300 West Drake Road, Fort Collins, CO, 80523-1620, USA.
| |
Collapse
|
27
|
Hartley G, Faulhaber E, Caldwell A, Coy J, Kurihara J, Guth A, Regan D, Dow S. Immune regulation of canine tumour and macrophage PD-L1 expression. Vet Comp Oncol 2016; 15:534-549. [PMID: 26842912 DOI: 10.1111/vco.12197] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 10/08/2015] [Accepted: 10/25/2015] [Indexed: 01/08/2023]
Abstract
Expression of programmed cell death receptor ligand 1 (PD-L1) on tumor cells has been associated with immune escape in human and murine cancers, but little is known regarding the immune regulation of PD-L1 expression by tumor cells and tumor-infiltrating macrophages in dogs. Therefore, 14 canine tumor cell lines, as well as primary cultures of canine monocytes and macrophages, were evaluated for constitutive PD-L1 expression and for responsiveness to immune stimuli. We found that PD-L1 was expressed constitutively on all canine tumor cell lines evaluated, although the levels of basal expression were very variable. Significant upregulation of PD-L1 expression by all tumor cell lines was observed following IFN-γ exposure and by exposure to a TLR3 ligand. Canine monocytes and monocyte-derived macrophages did not express PD-L1 constitutively, but did significantly upregulate expression following treatment with IFN-γ. These findings suggest that most canine tumors express PD-L1 constitutively and that both innate and adaptive immune stimuli can further upregulate PD-L1 expression. Therefore the upregulation of PD-L1 expression by tumor cells and by tumor-infiltrating macrophages in response to cytokines such as IFN-γ may represent an important mechanism of tumor-mediated T-cell suppression in dogs as well as in humans.
Collapse
Affiliation(s)
- G Hartley
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Ft. Collins, CO, USA
| | - E Faulhaber
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Ft. Collins, CO, USA
| | - A Caldwell
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Ft. Collins, CO, USA
| | - J Coy
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Ft. Collins, CO, USA
| | - J Kurihara
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Ft. Collins, CO, USA
| | - A Guth
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Ft. Collins, CO, USA
| | - D Regan
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Ft. Collins, CO, USA
| | - S Dow
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Ft. Collins, CO, USA
| |
Collapse
|
28
|
Sood N, Chaudhary DK, Pradhan PK, Verma DK, Raja Swaminathan T, Kushwaha B, Punia P, Jena JK. Establishment and characterization of a continuous cell line from thymus of striped snakehead, Channa striatus (Bloch 1793). In Vitro Cell Dev Biol Anim 2015; 51:787-96. [DOI: 10.1007/s11626-015-9891-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Accepted: 03/09/2015] [Indexed: 12/31/2022]
|
29
|
Gustafson TL, Kitchell BE, Biller B. Hedgehog signaling is activated in canine transitional cell carcinoma and contributes to cell proliferation and survival. Vet Comp Oncol 2015; 15:174-183. [PMID: 25864514 DOI: 10.1111/vco.12149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 02/11/2015] [Accepted: 03/04/2015] [Indexed: 01/11/2023]
Abstract
Transitional cell carcinoma (TCC) is the most commonly diagnosed tumor of the canine urinary system. Hedgehog (HH) signaling represents one possible novel therapeutic target, based on its recently identified central role in human urothelial carcinoma. The purpose of this study was to determine if HH mediators are expressed in canine TCC and the effect of inhibition of this pathway on cell growth and survival. HH pathway mediators were found to be expressed in five canine TCC cell lines. Indian HH was expressed in tumor cells in five canine bladder tumor tissues, but not in normal canine bladder tissue. Inhibition of HH signaling with cyclopamine and GANT61 led to significantly decreased cell proliferation but had a smaller effect on apoptosis. These results support future investigation of inhibitors of HH signaling in the treatment of canine TCC.
Collapse
Affiliation(s)
- T L Gustafson
- Colorado State University, Animal Cancer Center, Fort Collins, CO, USA
| | - B E Kitchell
- VCA Veterinary Care Animal Hospital and Referral Center, Oncology, Albuquerque, NM, USA
| | - B Biller
- Colorado State University, CVMBS-VTH, Animal Cancer Center, Fort Collins, CO, USA
| |
Collapse
|
30
|
The effect of Zhangfei/CREBZF on cell growth, differentiation, apoptosis, migration, and the unfolded protein response in several canine osteosarcoma cell lines. BMC Vet Res 2015; 11:22. [PMID: 25890299 PMCID: PMC4326286 DOI: 10.1186/s12917-015-0331-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 01/20/2015] [Indexed: 01/15/2023] Open
Abstract
Background We had previously shown that the bLZip domain-containing transcription factor, Zhangfei/CREBZF inhibits the growth and the unfolded protein response (UPR) in cells of the D–17 canine osteosarcoma (OS) line and that the effects of Zhangfei are mediated by it stabilizing the tumour suppressor protein p53. To determine if our observations with D-17 cells applied more universally to canine OS, we examined three other independently isolated canine OS cell lines—Abrams, McKinley and Gracie. Results Like D–17, the three cell lines expressed p53 proteins that were capable of activating promoters with p53 response elements on their own, and synergistically with Zhangfei. Furthermore, as with D–17 cells, Zhangfei suppressed the growth and UPR-related transcripts in the OS cell lines. Zhangfei also induced the activation of osteocalcin expression, a marker of osteoblast differentiation and triggered programmed cell death. Conclusions Osteosarcomas are common malignancies in large breeds of dogs. Although there has been dramatic progress in their treatment, these therapies often fail, leading to recurrence of the tumour and metastatic spread. Our results indicate that induction of the expression of Zhangfei in OS, where p53 is functional, may be an effective modality for the treatment of OS.
Collapse
|
31
|
Geraghty RJ, Capes-Davis A, Davis JM, Downward J, Freshney RI, Knezevic I, Lovell-Badge R, Masters JRW, Meredith J, Stacey GN, Thraves P, Vias M. Guidelines for the use of cell lines in biomedical research. Br J Cancer 2014; 111:1021-46. [PMID: 25117809 PMCID: PMC4453835 DOI: 10.1038/bjc.2014.166] [Citation(s) in RCA: 258] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 03/05/2014] [Indexed: 01/13/2023] Open
Abstract
Cell-line misidentification and contamination with microorganisms, such as mycoplasma, together with instability, both genetic and phenotypic, are among the problems that continue to affect cell culture. Many of these problems are avoidable with the necessary foresight, and these Guidelines have been prepared to provide those new to the field and others engaged in teaching and instruction with the information necessary to increase their awareness of the problems and to enable them to deal with them effectively. The Guidelines cover areas such as development, acquisition, authentication, cryopreservation, transfer of cell lines between laboratories, microbial contamination, characterisation, instability and misidentification. Advice is also given on complying with current legal and ethical requirements when deriving cell lines from human and animal tissues, the selection and maintenance of equipment and how to deal with problems that may arise.
Collapse
Affiliation(s)
- R J Geraghty
- Cancer Research UK Cambridge
Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way,
Cambridge
CB2 0RE, UK
| | - A Capes-Davis
- CellBank Australia, Children's
Medical Research Institute, Locked Bag 23,
Wentworthville, New South Wales
2145, Australia
| | - J M Davis
- School of Life and Medical Sciences,
University of Hertfordshire, College Lane, Hatfield,
Hertfordshire
AL10 9AB, UK
| | - J Downward
- Cancer Research UK, London Research
Institute, 44 Lincoln's Inn Fields, London
WC2A 3LY, UK
| | - R I Freshney
- Institute for Cancer Sciences,
University of Glasgow, 24 Greenwood Drive, Bearsden,
Glasgow
G61 2HA, UK
| | - I Knezevic
- Department of Essential Medicines and
Health Products, Quality, Safety and Standards Team, World Health
Organization, 20 Avenue Appia, 1211
Geneva 27, Switzerland
| | - R Lovell-Badge
- MRC National Institute for Medical
Research, The Ridgeway, Mill Hill, London
NW7 1AA, UK
| | - J R W Masters
- University College London, 67 Riding
House Street, London
W1W 7EJ, UK
| | - J Meredith
- Cancer Research UK, Angel Building,
407 St John Street, London
EC1V 4AD, UK
| | - G N Stacey
- National Institute for Biological
Standards and Control, A Centre of the Medicines and Healthcare Products
Regulatory Agency, Blanche Lane, South Mimms, Herts
EN6 3QG, UK
| | - P Thraves
- Culture Collections, Public Health
England, Porton Down, Salisbury
SP4 0JG, UK
| | - M Vias
- Cancer Research UK Cambridge
Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way,
Cambridge
CB2 0RE, UK
| |
Collapse
|
32
|
Abstract
Cell-line misidentification and contamination with microorganisms, such as mycoplasma, together with instability, both genetic and phenotypic, are among the problems that continue to affect cell culture. Many of these problems are avoidable with the necessary foresight, and these Guidelines have been prepared to provide those new to the field and others engaged in teaching and instruction with the information necessary to increase their awareness of the problems and to enable them to deal with them effectively. The Guidelines cover areas such as development, acquisition, authentication, cryopreservation, transfer of cell lines between laboratories, microbial contamination, characterisation, instability and misidentification. Advice is also given on complying with current legal and ethical requirements when deriving cell lines from human and animal tissues, the selection and maintenance of equipment and how to deal with problems that may arise.
Collapse
|
33
|
Wirth KA, Kow K, Salute ME, Bacon NJ, Milner RJ. In vitroeffects ofYunnan Baiyaoon canine hemangiosarcoma cell lines. Vet Comp Oncol 2014; 14:281-94. [DOI: 10.1111/vco.12100] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 05/01/2014] [Accepted: 05/05/2014] [Indexed: 02/02/2023]
Affiliation(s)
- K. A. Wirth
- Department of Clinical Sciences; University of Florida; Gainesville FL USA
| | - K. Kow
- Department of Clinical Sciences; University of Florida; Gainesville FL USA
| | - M. E. Salute
- Department of Clinical Sciences; University of Florida; Gainesville FL USA
| | - N. J. Bacon
- Department of Clinical Sciences; University of Florida; Gainesville FL USA
| | - R. J. Milner
- Department of Clinical Sciences; University of Florida; Gainesville FL USA
| |
Collapse
|
34
|
Shoeneman JK, Ehrhart EJ, Charles JB, Thamm DH. Survivin inhibition via EZN-3042 in canine lymphoma and osteosarcoma. Vet Comp Oncol 2014; 14:e45-57. [PMID: 24923332 DOI: 10.1111/vco.12104] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/08/2014] [Accepted: 05/15/2014] [Indexed: 12/21/2022]
Abstract
Canine lymphoma (LSA) and osteosarcoma (OS) have high mortality rates and remain in need of more effective therapeutic approaches. Survivin, an inhibitor of apoptosis (IAP) family member protein that inhibits apoptosis and drives cell proliferation, is commonly elevated in human and canine cancer. Survivin expression is a negative prognostic factor in dogs with LSA and OS, and canine LSA and OS cell lines express high levels of survivin. In this study, we demonstrate that survivin downregulation in canine LSA and OS cells using a clinically applicable locked nucleic acid antisense oligonucleotide (EZN-3042, Enzon Pharmaceuticals, Piscataway Township, NJ, USA) inhibits growth, induces apoptosis and enhances chemosensitivity in vitro, and inhibits survivin transcription and protein production in orthotopic canine OS xenografts. Our findings strongly suggest that survivin-directed therapies might be effective in treatment of canine LSA and OS and support evaluation of EZN-3042 in dogs with cancer.
Collapse
Affiliation(s)
- J K Shoeneman
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.,Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, USA
| | - E J Ehrhart
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.,Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, USA.,Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - J B Charles
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.,Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - D H Thamm
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.,Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
35
|
Schwartz AL, Custis JT, Harmon JF, Powers BE, Chubb LS, LaRue SM, Ehrhart NP, Ryan SD. Orthotopic model of canine osteosarcoma in athymic rats for evaluation of stereotactic radiotherapy. Am J Vet Res 2013; 74:452-8. [PMID: 23438123 DOI: 10.2460/ajvr.74.3.452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To develop an orthotopic model of canine osteosarcoma in athymic rats as a model for evaluating the effects of stereotactic radiotherapy (SRT) on osteosarcoma cells. ANIMALS 26 athymic nude rats. PROCEDURES 3 experiments were performed. In the first 2 experiments, rats were injected with 1 × 10(6) Abrams canine osteosarcoma cells into the proximal aspect of the tibia (n = 12) or distal aspect of the femur (6). Tumor engraftment and progression were monitored weekly via radiography, luciferase imaging, and measurement of urine pyridinoline concentration for 5 weeks and histologic evaluation after euthanasia. In the third experiment, 8 rats underwent canine osteosarcoma cell injection into the distal aspect of the femur and SRT was administered to the affected area in three 12-Gy fractions delivered on consecutive days (total radiation dose, 36 Gy). Percentage tumor necrosis and urinary pyridinoline concentrations were used to assess local tumor control. The short-term effect of SRT on skin was also evaluated. RESULTS Tumors developed in 10 of 12 tibial sites and all 14 femoral sites. Administration of SRT to rats with femoral osteosarcoma was feasible and successful. Mean tumor necrosis of 95% was achieved histologically, and minimal adverse skin effects were observed. CONCLUSIONS AND CLINICAL RELEVANCE The orthotopic model of canine osteosarcoma in rats developed in this study was suitable for evaluating the effects of local tumor control and can be used in future studies to evaluate optimization of SRT duration, dose, and fractionation schemes. The model could also allow evaluation of other treatments in combination with SRT, such as chemotherapy or bisphosphonate, radioprotectant, or parathyroid hormone treatment.
Collapse
Affiliation(s)
- Anthony L Schwartz
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Dailey DD, Anfinsen KP, Pfaff LE, Ehrhart EJ, Charles JB, Bønsdorff TB, Thamm DH, Powers BE, Jonasdottir TJ, Duval DL. HES1, a target of Notch signaling, is elevated in canine osteosarcoma, but reduced in the most aggressive tumors. BMC Vet Res 2013; 9:130. [PMID: 23816051 PMCID: PMC3701487 DOI: 10.1186/1746-6148-9-130] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 06/24/2013] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Hairy and enhancer of split 1 (HES1), a basic helix-loop-helix transcriptional repressor, is a downstream target of Notch signaling. Notch signaling and HES1 expression have been linked to growth and survival in a variety of human cancer types and have been associated with increased metastasis and invasiveness in human osteosarcoma cell lines. Osteosarcoma (OSA) is an aggressive cancer demonstrating both high metastatic rate and chemotherapeutic resistance. The current study examined expression of Notch signaling mediators in primary canine OSA tumors and canine and human osteosarcoma cell lines to assess their role in OSA development and progression. RESULTS Reverse transcriptase - quantitative PCR (RT-qPCR) was utilized to quantify HES1, HEY1, NOTCH1 and NOTCH2 gene expression in matched tumor and normal metaphyseal bone samples taken from dogs treated for appendicular OSA at the Colorado State University Veterinary Teaching Hospital. Gene expression was also assessed in tumors from dogs with a disease free interval (DFI) of <100 days compared to those with a DFI > 300 days following treatment with surgical amputation followed by standard chemotherapy. Immunohistochemistry was performed to confirm expression of HES1. Data from RT-qPCR and immunohistochemical (IHC) experiments were analyzed using REST2009 software and survival analysis based on IHC expression employed the Kaplan-Meier method and log rank analysis. Unbiased clustered images were generated from gene array analysis data for Notch/HES1 associated genes. Gene array analysis of Notch/HES1 associated genes suggested alterations in the Notch signaling pathway may contribute to the development of canine OSA. HES1 mRNA expression was elevated in tumor samples relative to normal bone, but decreased in tumor samples from dogs with a DFI < 100 days relative to those with a DFI > 300 days. NOTCH2 and HEY1 mRNA expression was also elevated in tumors relative to normal bone, but was not differentially expressed between the DFI tumor groups. Survival analysis confirmed an association between decreased HES1 immunosignal and shorter DFI. CONCLUSIONS Our findings suggest that activation of Notch signaling occurs and may contribute to the development of canine OSA. However, association of low HES1 expression and shorter DFI suggests that mechanisms that do not alter HES1 expression may drive the most aggressive tumors.
Collapse
|
37
|
Capes-Davis A, Reid YA, Kline MC, Storts DR, Strauss E, Dirks WG, Drexler HG, MacLeod RA, Sykes G, Kohara A, Nakamura Y, Elmore E, Nims RW, Alston-Roberts C, Barallon R, Los GV, Nardone RM, Price PJ, Steuer A, Thomson J, Masters JR, Kerrigan L. Match criteria for human cell line authentication: Where do we draw the line? Int J Cancer 2012; 132:2510-9. [DOI: 10.1002/ijc.27931] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Accepted: 09/26/2012] [Indexed: 12/18/2022]
|
38
|
Lyles SE, Milner RJ, Kow K, Salute ME. In vitroeffects of the tyrosine kinase inhibitor, masitinib mesylate, on canine hemangiosarcoma cell lines. Vet Comp Oncol 2012; 10:223-35. [DOI: 10.1111/j.1476-5829.2012.00335.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 04/18/2012] [Accepted: 04/25/2012] [Indexed: 12/17/2022]
Affiliation(s)
- S. E. Lyles
- Department of Small Animal Clinical Sciences; University of Florida; Gainesville FL USA
| | - R. J. Milner
- Department of Small Animal Clinical Sciences; University of Florida; Gainesville FL USA
| | - K. Kow
- Department of Small Animal Clinical Sciences; University of Florida; Gainesville FL USA
| | - M. E. Salute
- Department of Small Animal Clinical Sciences; University of Florida; Gainesville FL USA
| |
Collapse
|
39
|
Gentschev I, Adelfinger M, Josupeit R, Rudolph S, Ehrig K, Donat U, Weibel S, Chen NG, Yu YA, Zhang Q, Heisig M, Thamm D, Stritzker J, MacNeill A, Szalay AA. Preclinical evaluation of oncolytic vaccinia virus for therapy of canine soft tissue sarcoma. PLoS One 2012; 7:e37239. [PMID: 22615950 PMCID: PMC3352892 DOI: 10.1371/journal.pone.0037239] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 04/18/2012] [Indexed: 12/27/2022] Open
Abstract
Virotherapy using oncolytic vaccinia virus (VACV) strains is one promising new strategy for canine cancer therapy. In this study we describe the establishment of an in vivo model of canine soft tissue sarcoma (CSTS) using the new isolated cell line STSA-1 and the analysis of the virus-mediated oncolytic and immunological effects of two different Lister VACV LIVP1.1.1 and GLV-1h68 strains against CSTS. Cell culture data demonstrated that both tested VACV strains efficiently infected and destroyed cells of the canine soft tissue sarcoma line STSA-1. In addition, in our new canine sarcoma tumor xenograft mouse model, systemic administration of LIVP1.1.1 or GLV-1h68 viruses led to significant inhibition of tumor growth compared to control mice. Furthermore, LIVP1.1.1 mediated therapy resulted in almost complete tumor regression and resulted in long-term survival of sarcoma-bearing mice. The replication of the tested VACV strains in tumor tissues led to strong oncolytic effects accompanied by an intense intratumoral infiltration of host immune cells, mainly neutrophils. These findings suggest that the direct viral oncolysis of tumor cells and the virus-dependent activation of tumor-associated host immune cells could be crucial parts of anti-tumor mechanism in STSA-1 xenografts. In summary, the data showed that both tested vaccinia virus strains and especially LIVP1.1.1 have great potential for effective treatment of CSTS.
Collapse
Affiliation(s)
- Ivaylo Gentschev
- Genelux Corporation, San Diego Science Center, San Diego, California, United States of America
- Department of Biochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - Marion Adelfinger
- Department of Biochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - Rafael Josupeit
- Department of Biochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - Stephan Rudolph
- Department of Biochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - Klaas Ehrig
- Genelux Corporation, San Diego Science Center, San Diego, California, United States of America
- Department of Biochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - Ulrike Donat
- Department of Biochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - Stephanie Weibel
- Department of Biochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - Nanhai G. Chen
- Genelux Corporation, San Diego Science Center, San Diego, California, United States of America
- Department of Radiation Oncology, Moores Cancer Center, University of California San Diego, La Jolla, California, United States of America
| | - Yong A. Yu
- Genelux Corporation, San Diego Science Center, San Diego, California, United States of America
- Department of Radiation Oncology, Moores Cancer Center, University of California San Diego, La Jolla, California, United States of America
| | - Qian Zhang
- Genelux Corporation, San Diego Science Center, San Diego, California, United States of America
- Department of Radiation Oncology, Moores Cancer Center, University of California San Diego, La Jolla, California, United States of America
| | - Martin Heisig
- Institut für Medizinische Strahlenkunde und Zellforschung (MSZ), University of Wuerzburg, Wuerzburg, Germany
| | - Douglas Thamm
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Jochen Stritzker
- Genelux Corporation, San Diego Science Center, San Diego, California, United States of America
- Department of Biochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - Amy MacNeill
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois, Urbana, Illinois, United States of America
| | - Aladar A. Szalay
- Genelux Corporation, San Diego Science Center, San Diego, California, United States of America
- Department of Biochemistry, University of Wuerzburg, Wuerzburg, Germany
- Rudolf Virchow Center for Experimental Biomedicine, University of Wuerzburg, Wuerzburg, Germany
- Institute for Molecular Infection Biology, University of Wuerzburg, Wuerzburg, Germany
- Department of Radiation Oncology, Moores Cancer Center, University of California San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
40
|
Shoeneman JK, Ehrhart EJ, Eickhoff JC, Charles JB, Powers BE, Thamm DH. Expression and function of survivin in canine osteosarcoma. Cancer Res 2011; 72:249-59. [PMID: 22068035 DOI: 10.1158/0008-5472.can-11-2315] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Osteosarcoma has a high mortality rate and remains in need of more effective therapeutic approaches. Survivin is an inhibitor of apoptosis family member protein that blocks apoptosis and drives proliferation in human cancer cells where it is commonly elevated. In this study, we illustrate the superiority of a canine osteosarcoma model as a translational tool for evaluating survivin-directed therapies, owing to the striking similarities in gross and microscopic appearance, biologic behavior, gene expression, and signaling pathway alterations. Elevated survivin expression in primary canine osteosarcoma tissue correlated with increased histologic grade and mitotic index and a decreased disease-free interval (DFI). Survivin attenuation in canine osteosarcoma cells inhibited cell-cycle progression, increased apoptosis, mitotic arrest, and chemosensitivity, and cooperated with chemotherapy to significantly improve in vivo tumor control. Our findings illustrate the utility of a canine system to more accurately model human osteosarcoma and strongly suggest that survivin-directed therapies might be highly effective in its treatment.
Collapse
Affiliation(s)
- Jenette K Shoeneman
- The Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, USA
| | | | | | | | | | | |
Collapse
|