1
|
Elliott J. Early-Delayed Radiotoxicity Associated With Stereotactic Radiation Therapy Following a Daily 6 Gy × 5 (30 Gy) Protocol for Presumed Canine Intracranial Meningiomas. Vet Comp Oncol 2025; 23:152-160. [PMID: 39828659 DOI: 10.1111/vco.13040] [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] [Received: 08/29/2024] [Revised: 11/22/2024] [Accepted: 01/04/2025] [Indexed: 01/22/2025]
Abstract
Early-delayed side effects (EDSEs) following treatment of canine intracranial meningiomas with 1-3-fraction stereotactic radiation therapy (SRT) can cause worsening neurologic signs, and one potential method of mitigating this toxicity is reducing the dose per fraction. Twenty dogs with imaging-diagnosed intracranial meningiomas and telephone follow-up of at least 6 months received a protocol of 6 Gy × 5, daily (30 Gy). A 'possible EDSE' was defined as mental dullness, neurologic exacerbation of existing neurologic signs or new neurologic signs occurring within 1-4 months of completing SRT, regardless of the response to steroids and even if an MRI was not performed. A 'probable EDSE' was defined as mental dullness, neurologic exacerbation of existing neurologic signs or new neurologic signs occurring within 1-4 months of completing SRT. These signs were either reversible with the initiation or increased doses of prednisolone, or follow-up MRI revealed no evidence of an alternate explanation. No dogs experienced acute radiotoxicity or clinical signs compatible with EDSEs. The protocol appears to result in limited acute radiotoxicity, and further evaluation of the frequency of long-term toxicities and relative efficacy should be undertaken.
Collapse
|
2
|
Geiger R, Mankin J, Volk HA, de Decker S, van Asselt N, Randhawa K, Ehling T, Maitz CA, Naramor A, Coates JR, Stalin C, Johnstone L, Morris J, Plessas IN, Forward A, Garosi L, Scarpante E, Cherubini G, Harcourt‐Brown T, Carrera‐Justiz S, Gutti JR, Takada M, White J, Nagata K, Kent M, Barber R, Ito D, Nakayama T, Oshima A, Sekigucki N, Smith‐Oskrochi L, Jeffery N. Comparison of Survival After Treatment of Presumed Intracranial Meningioma by Radiotherapy or Surgery in 285 Dogs. J Vet Intern Med 2025; 39:e70011. [PMID: 39968764 PMCID: PMC11836666 DOI: 10.1111/jvim.70011] [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] [Received: 05/30/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND The comparative effectiveness of radiotherapy and surgery for treating intracranial meningioma is unknown. OBJECTIVES To compare survival after treatment of suspected intracranial meningioma by either surgery or radiotherapy. ANIMALS Two hundred eighty-five companion dogs with suspected intracranial meningiomas presenting to 11 specialty clinics in three countries. METHODS Parallel cohort comparison study on retrospective data. Dogs diagnosed with intracranial meningioma by board-certified veterinary neurologists or radiologists and treated by radiotherapy or surgery were identified through medical record searches and presenting and survival data extracted. Lesion site was classified as rostro- or caudotentorial and size was measured on contrast magnetic resonance images. Outcome was all-cause death. Analysis of survival by Cox proportional hazards, including selection for optimal multivariable model using lasso, counterfactual modeling including variables associated with treatment allocation and survival. RESULTS One hundred sixty-eight dogs received radiotherapy and 117 received surgery. All analyses indicated reduced survival associated with surgery compared to radiotherapy. There was a median survival after surgery of 297 (IQR: 99-768) days compared with 696 (IQR: 368-999) for dogs treated by radiation, associated with a univariable hazard ratio of 1.802 (95% CI: 1.357-2.394). Counterfactual modeling estimated a mean survival of 480 (95% CI: 395-564) days after surgery and 673 (95% CI: 565-782) days after radiotherapy, representing a decrease in survival of 29%. Location and size of the lesion were not associated with survival duration. CONCLUSIONS AND CLINICAL IMPORTANCE Dogs with suspected intracranial meningioma have substantially superior survival after radiotherapy compared to surgery.
Collapse
Affiliation(s)
- Rachel Geiger
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Joe Mankin
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Holger A. Volk
- Department of Clinical Science and ServicesRoyal Veterinary CollegeHatfieldUK
| | - Steven de Decker
- Department of Clinical Science and ServicesRoyal Veterinary CollegeHatfieldUK
| | - Nate van Asselt
- Department of Surgical SciencesUniversity of Wisconsin Veterinary CareMadisonWisconsinUSA
| | - Karanbir Randhawa
- Department of Surgical SciencesUniversity of Wisconsin Veterinary CareMadisonWisconsinUSA
| | - Tara Ehling
- Department of Veterinary Medicine and Surgery, College of Veterinary MedicineUniversity of MissouriColumbiaMissouriUSA
| | - Charles A. Maitz
- Department of Veterinary Medicine and Surgery, College of Veterinary MedicineUniversity of MissouriColumbiaMissouriUSA
| | - Ada Naramor
- Department of Veterinary Medicine and Surgery, College of Veterinary MedicineUniversity of MissouriColumbiaMissouriUSA
| | - Joan R. Coates
- Department of Veterinary Medicine and Surgery, College of Veterinary MedicineUniversity of MissouriColumbiaMissouriUSA
| | - Catherine Stalin
- Small Animal Clinical Division, School of Biodiversity, One Health and Veterinary MedicineUniversity of GlasgowGlasgowUK
| | - Lauren Johnstone
- Small Animal Clinical Division, School of Biodiversity, One Health and Veterinary MedicineUniversity of GlasgowGlasgowUK
| | - Joanna Morris
- Small Animal Clinical Division, School of Biodiversity, One Health and Veterinary MedicineUniversity of GlasgowGlasgowUK
| | | | | | | | | | | | | | - Sheila Carrera‐Justiz
- College of Veterinary Medicine, Department of Small Animal Clinical SciencesUniversity of FloridaGainesvilleFloridaUSA
| | - Jishnu Rao Gutti
- College of Veterinary Medicine, Department of Small Animal Clinical SciencesUniversity of FloridaGainesvilleFloridaUSA
| | - Marilia Takada
- College of Veterinary Medicine, Department of Small Animal Clinical SciencesUniversity of FloridaGainesvilleFloridaUSA
| | - Joel White
- Department of Small Animal Medicine & SurgeryUniversity of Georgia College of Veterinary MedicineAthensGeorgiaUSA
| | - Koichi Nagata
- Department of Small Animal Medicine & SurgeryUniversity of Georgia College of Veterinary MedicineAthensGeorgiaUSA
| | - Marc Kent
- Department of Small Animal Medicine & SurgeryUniversity of Georgia College of Veterinary MedicineAthensGeorgiaUSA
| | - Renee Barber
- Department of Small Animal Medicine & SurgeryUniversity of Georgia College of Veterinary MedicineAthensGeorgiaUSA
| | - Daisuke Ito
- Department of Veterinary Medicine, College of Bioresource SciencesNihon UniversityFujisawaJapan
| | - Tomohiro Nakayama
- Department of Veterinary Medicine, College of Bioresource SciencesNihon UniversityFujisawaJapan
| | - Ayaka Oshima
- Department of Veterinary Medicine, College of Bioresource SciencesNihon UniversityFujisawaJapan
| | - Naoki Sekigucki
- Department of Veterinary Medicine, College of Bioresource SciencesNihon UniversityFujisawaJapan
| | - Lauren Smith‐Oskrochi
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Nick Jeffery
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTexasUSA
| |
Collapse
|
3
|
Kent MS, Rosseel M. Updates on Radiation Therapy for Pituitary Tumors: Techniques and Prognosis. Vet Clin North Am Small Anim Pract 2025; 55:119-133. [PMID: 39227254 DOI: 10.1016/j.cvsm.2024.07.010] [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: 09/05/2024]
Abstract
Radiation therapy for the treatment of both functional and nonfunctional pituitary tumors for dogs and cats has been described in veterinary medicine with a recent shift in focus toward stereotactic techniques. While the technology required and normal tissue constraints for stereotactic procedures are more stringent, recent publications indicate that, while it helps alleviate clinical signs, the survival response may not be as durable as with conventionally fractionated radiation therapy in dogs, despite being seen in cats. Regardless of the protocol recommendation, potential benefit to the patient is excellent with manageable side effect profiles.
Collapse
Affiliation(s)
- Michael S Kent
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, USA.
| | - Matthias Rosseel
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| |
Collapse
|
4
|
Gieger TL. Radiation Therapy for Brain Tumors in Dogs and Cats. Vet Clin North Am Small Anim Pract 2025; 55:67-80. [PMID: 39393930 DOI: 10.1016/j.cvsm.2024.07.007] [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: 10/13/2024]
Abstract
External beam radiation therapy (RT) has become the standard of care for non-resectable or post-operative incompletely excised brain tumors in dogs and cats due to its relatively low side effect profile and increasing availability. This article reviews the indications for, expected outcomes of and possible toxicities associated with RT, follow-up care recommendations after RT, and publications about specific tumor types in dogs and cats with brain tumors.
Collapse
Affiliation(s)
- Tracy L Gieger
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, Raleigh, NC 27607, USA.
| |
Collapse
|
5
|
Strasberg JR, Rossmeisl JH, Kelsey KL, Yoshikawa H, Gieger TL, Nolan MW. A prospective evaluation of succinct prednisone tapering after brain tumor irradiation in dogs. J Vet Intern Med 2024; 38:2571-2577. [PMID: 39122668 PMCID: PMC11423478 DOI: 10.1111/jvim.17163] [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] [Received: 12/30/2023] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND To ameliorate anticipated or ongoing neurological deficits, dogs undergoing brain tumor irradiation often are prescribed lengthy courses of prednisone PO during and after radiotherapy (RT). This practice can contribute to unwanted corticosteroid-associated morbidity and may be unnecessary. OBJECTIVE Determine whether long-term corticosteroid dependency can be minimized by use of succinct prednisone tapering. ANIMALS Fifty-five pet dogs undergoing brain tumor irradiation. METHODS Nineteen dogs were treated using a "rapid-taper" protocol wherein corticosteroid dose reduction began 0 to 20 days after completing RT. Outcomes were compared with a retrospectively studied control group ("slow-taper"; N = 36 dogs) in which corticosteroids were tapered more slowly according to individual clinician recommendations. RESULTS Patient demographics were similar between groups. Mean time to lowest prednisone dose was 41 days postirradiation in the rapid-taper group and 117 days in the slow-taper group (P = .003). In the rapid-taper group, 15 of 19 dogs (84%) were completely tapered off prednisone, vs 18 of 36 (50%) in the slow-taper group (P = .04). Rates at which corticosteroids had to be reinstituted later were similar for the 2 groups (approximately 1 in 3 dogs). Adverse effect rates were similar for the 2 groups. Although no comparable questionnaire-derived data were available for the "slow-taper" group, overall and neurologic quality of life remained stable after RT in the rapid-taper group. CONCLUSIONS AND CLINICAL IMPORTANCE For many dogs, lengthy courses of PO prednisone are avoidable after intracranial RT. Future efforts should aim to identify which dogs benefit most from accelerated prednisone tapering.
Collapse
Affiliation(s)
- Jason R Strasberg
- College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - John H Rossmeisl
- Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, USA
| | - Krista L Kelsey
- Carolina Veterinary Specialists, Mathews, North Carolina, USA
| | - Hiroto Yoshikawa
- College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Tracy L Gieger
- College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Michael W Nolan
- College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
6
|
Panek WK, Toedebusch RG, Mclaughlin BE, Dickinson PJ, Van Dyke JE, Woolard KD, Berens ME, Lesniak MS, Sturges BK, Vernau KM, Li C, Miska J, Toedebusch CM. The CCL2-CCR4 axis promotes Regulatory T cell trafficking to canine glioma tissues. J Neurooncol 2024; 169:647-658. [PMID: 39046599 PMCID: PMC11341612 DOI: 10.1007/s11060-024-04766-4] [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: 05/24/2024] [Accepted: 07/02/2024] [Indexed: 07/25/2024]
Abstract
PURPOSE Spontaneously occurring glioma in pet dogs is increasingly recognized as a valuable translational model for human glioblastoma. Canine high-grade glioma and human glioblastomas share many molecular similarities, including the accumulation of immunosuppressive regulatory T cells (Tregs) that inhibit anti-tumor immune responses. Identifying in dog mechanisms responsible for Treg recruitment may afford to target the cellular population driving immunosuppression, the results providing a rationale for translational clinical studies in human patients. Our group has previously identified C-C motif chemokine 2 (CCL2) as a glioma-derived T-reg chemoattractant acting on chemokine receptor 4 (CCR4) in a murine orthotopic glioma model. Recently, we demonstrated a robust increase of CCL2 in the brain tissue of canine patients bearing high-grade glioma. METHODS We performed a series of in vitro experiments using canine Tregs and patient-derived canine glioma cell lines (GSC 1110, GSC 0514, J3T-Bg, G06A) to interrogate the CCL2-CCR4 signaling axis in the canine. RESULTS We established a flow cytometry gating strategy for identifying and isolating FOXP3+ Tregs in dogs. The canine CD4 + CD25high T-cell population was highly enriched in FOXP3 and CCR4 expression, indicating they are bona fide Tregs. Canine Treg migration was enhanced by CCL2 or by glioma cell line-derived supernatant. Blockade of the CCL2-CCR4 axis significantly reduced migration of canine Tregs. CCL2 mRNA was expressed in all glioma cell lines, and expression increased when exposed to Tregs but not CD4 + helper T-cells. CONCLUSION Our study validates CCL2-CCR4 as a bi-directional Treg-glioma immunosuppressive and tumor-promoting axis in canine high-grade glioma.
Collapse
Affiliation(s)
- W K Panek
- Department of Surgical and Radiological Sciences, University of California, Davis, One Shields Avenue, 2112 Tupper Hall, Davis, CA, 95616-5270, USA.
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, 380 South University Avenue, 419 Hill Pavilion, Philadelphia, PA, 19104, USA.
| | - R G Toedebusch
- Department of Surgical and Radiological Sciences, University of California, Davis, One Shields Avenue, 2112 Tupper Hall, Davis, CA, 95616-5270, USA
| | - B E Mclaughlin
- University of California Davis, Flow Cytometry Shared Resource, Davis, CA, USA
| | - P J Dickinson
- Department of Surgical and Radiological Sciences, University of California, Davis, One Shields Avenue, 2112 Tupper Hall, Davis, CA, 95616-5270, USA
| | - J E Van Dyke
- University of California Davis, Flow Cytometry Shared Resource, Davis, CA, USA
| | - K D Woolard
- Department of Pathology, Microbiology and Immunology, University of California, Davis, Davis, CA, USA
| | - M E Berens
- Cancer and Cell Biology Division, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - M S Lesniak
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - B K Sturges
- Department of Surgical and Radiological Sciences, University of California, Davis, One Shields Avenue, 2112 Tupper Hall, Davis, CA, 95616-5270, USA
| | - K M Vernau
- Department of Surgical and Radiological Sciences, University of California, Davis, One Shields Avenue, 2112 Tupper Hall, Davis, CA, 95616-5270, USA
| | - C Li
- Department of Surgical and Radiological Sciences, University of California, Davis, One Shields Avenue, 2112 Tupper Hall, Davis, CA, 95616-5270, USA
| | - J Miska
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Christine M Toedebusch
- Department of Surgical and Radiological Sciences, University of California, Davis, One Shields Avenue, 2112 Tupper Hall, Davis, CA, 95616-5270, USA.
| |
Collapse
|
7
|
Kundapur V, Torlakovic E, Auer RN. The Story Behind the First Mini-BEAM Photon Radiation Treatment: What is the Mini-Beam and Why is it Such an Advance? Semin Radiat Oncol 2024; 34:337-343. [PMID: 38880542 DOI: 10.1016/j.semradonc.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Radiation treatment has been the cornerstone in cancer management. However, long term treatment-related morbidity always accompanies tumor control which has significant impact on quality of life of the patient who has survived the cancer. Spatially fractionated radiation has the potential to achieve both cure and to avoid dreaded long term sequelae. The first ever randomized study of mini-beam radiation treatment (MBRT) of canine brain tumor has clearly shown the ability to achieve this goal. Dogs have gyrencephalic brains functionally akin to human brain. We here report long term follow-up and final outcome of the dogs, revealing both tumor control and side effects on normal brain. The results augur potential for conducting human studies with MBRT.
Collapse
Affiliation(s)
- Vijayananda Kundapur
- Saskatoon Cancer Centre, Clinical Professor, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 4H4 Canada.
| | - Emina Torlakovic
- Department of Pathology, University of Saskatchewan, Royal University Hospital, Saskatoon, SK S7N 0W8 Canada
| | - Roland N Auer
- Department of Pathology, University of Saskatchewan, Royal University Hospital, Saskatoon, SK S7N 0W8 Canada
| |
Collapse
|
8
|
Panek WK, Toedebusch RG, Mclaughlin BE, Dickinson PJ, Dyke JE, Woolard KD, Berens ME, Lesniak MS, Sturges BK, Vernau KM, Li C, Miska JM, Toedebusch CM. The CCL2-CCR4 Axis Promotes Regulatory T Cell Trafficking to Canine Glioma Tissues. RESEARCH SQUARE 2024:rs.3.rs-4474288. [PMID: 38947002 PMCID: PMC11213221 DOI: 10.21203/rs.3.rs-4474288/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Purpose Spontaneously occurring glioma in pet dogs is increasingly recognized as a valuable translational model for human glioblastoma. Canine high grade glioma and human glioblastomas share many molecular similarities, including accumulation of immunosuppressive regulatory T cells (Tregs) that inhibit anti-tumor immune responses. Identifying in dog mechanisms responsible for Treg recruitment may afford targeting the cellular population driving immunosuppression, the results providing a rationale for translational clinical studies in human patients. Our group has previously identified C-C motif chemokine 2 (CCL2) as a glioma-derived T-reg chemoattractant acting on chemokine receptor 4 (CCR4) in a murine orthotopic model of glioma. Recently, we demonstrated a robust increase of CCL2 in the brain tissue of canine patients bearing high-grade glioma. Methods We performed a series of in vitro experiments using canine Tregs and patient-derived canine glioma cell lines (GSC 1110, GSC 0514, J3T-Bg, G06A) to interrogate the CCL2-CCR4 signaling axis in the canine. Results We established a flow cytometry gating strategy for identification and isolation of FOXP3+ Tregs in dogs. The canine CD4 + CD25high T-cell population was highly enriched in FOXP3 and CCR4 expression, indicating they are bona fide Tregs. Canine Treg migration was enhanced by CCL2 or by glioma cell line-derived supernatant. Blockade of the CCL2-CCR4 axis significantly reduced migration of canine Tregs. CCL2 mRNA was expressed in all glioma cell lines and expression increased when exposed to Tregs but not to CD4 + helper T-cells. Conclusion Our study validates CCL2-CCR4 as a bi-directional Treg-glioma immunosuppressive and tumor-promoting axis in canine high-grade glioma.
Collapse
Affiliation(s)
| | | | - B E Mclaughlin
- University of California Davis, Flow Cytometry Shared Resource
| | | | - J E Dyke
- University of California Davis, Flow Cytometry Shared Resource
| | | | - M E Berens
- The Translational Genomics Research Institute
| | | | | | | | - C Li
- University of California, Davis
| | | | | |
Collapse
|
9
|
Cooper-Khan RS, Frankovich AN, Thompson CA, Thomovsky SA, Lewis MJ. Clinical Findings and Outcome in 30 Dogs with Presumptive or Confirmed Nerve Sheath Tumors. Vet Sci 2024; 11:192. [PMID: 38787164 PMCID: PMC11125868 DOI: 10.3390/vetsci11050192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/28/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024] Open
Abstract
Nerve sheath tumors (NSTs) are well-recognized primary nervous system tumors, but there is relatively limited information in dogs including comparison of NSTs in different anatomical locations. This retrospective study describes the clinical features and outcomes in a group of dogs with NSTs affecting the cranial nerves or spinal nerves. Thirty dogs were included, 25 with a presumptive diagnosis and five confirmed by histopathologic analysis. Seven dogs also had cytology of tumor samples, which were supportive of the NST diagnosis in four. Eight dogs had cranial nerve-associated NSTs, with six involving the trigeminal nerve. Twenty-two dogs had spinal nerve-associated NSTs including 13 invading the spinal canal and nine peripheral to the spinal canal, with the majority affecting nerves or nerve roots of the brachial plexus. The prognosis was poor, with dogs being euthanized eventually because of disease progression. Among dogs alive 1 week after diagnosis, the median survival time was 4 months but ranged from 2 weeks to >2 years. While there was a broad overlap between NST locations, survival was generally longer for dogs without spinal canal or intracranial involvement. The results expand available information on NSTs in dogs but should be interpreted with caution given the small number of dogs with a definitive diagnosis. Further investigation is warranted to determine how tumor location, invasiveness, and treatments pursued impact outcome.
Collapse
Affiliation(s)
- Rachel S. Cooper-Khan
- Department of Veterinary Clinical Sciences, Purdue University College of Veterinary Medicine, 625 Harrison Street, West Lafayette, IN 47907, USA; (R.S.C.-K.); (S.A.T.)
| | - Alexandra N. Frankovich
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, 625 Harrison Street, West Lafayette, IN 47907, USA; (A.N.F.); (C.A.T.)
| | - Craig A. Thompson
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, 625 Harrison Street, West Lafayette, IN 47907, USA; (A.N.F.); (C.A.T.)
| | - Stephanie A. Thomovsky
- Department of Veterinary Clinical Sciences, Purdue University College of Veterinary Medicine, 625 Harrison Street, West Lafayette, IN 47907, USA; (R.S.C.-K.); (S.A.T.)
| | - Melissa J. Lewis
- Department of Veterinary Clinical Sciences, Purdue University College of Veterinary Medicine, 625 Harrison Street, West Lafayette, IN 47907, USA; (R.S.C.-K.); (S.A.T.)
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, Raleigh, NC 27606, USA
| |
Collapse
|
10
|
Hansen KS, Li CF, Théon AP, Kent MS. Stereotactic radiotherapy outcomes for intraventricular brain tumours in 11 dogs. Vet Comp Oncol 2023; 21:665-672. [PMID: 37604645 DOI: 10.1111/vco.12929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/23/2023]
Abstract
Published radiotherapy data for canine intraventricular tumours are limited. In this retrospective, longitudinal study (9/2011-2018), 11 dogs with intraventricular masses were treated with stereotactic radiotherapy (SRT). Pathologic diagnosis was available from surgery or necropsy in 6/11 cases, revealing choroid plexus papilloma (3) or carcinoma (2), and ependymoma (1). The remainder were magnetic resonance imaging (MRI)-diagnosed as suspected choroid tumours or ependymomas. Tumours were located in the third or lateral ventricle (8), fourth ventricle (2), and cerebellopontine angle (1). Surgery was performed in three dogs prior to radiotherapy, and all showed gross residual/recurrent disease at treatment. Dogs received 8 Gray × 3 fractions (7), or 15 Gray × 1 fraction (4). Ten dogs were deceased at analysis, and one was living. The estimated median overall survival time (OS) from first SRT treatment was 16.9 months (515 days, 95% CI 33-1593 days). The survival time for two pathology-diagnosed carcinoma dogs were 24 and 133 days, respectively, and survival time for dogs with moderate to marked ventriculomegaly (4/11) ranged from 24 to 113 days. A total of 10/11 showed clinical improvement per owner or clinician, but two had short-lived benefits and were euthanized within 6 weeks of SRT. Limited conclusions on radiation-specific complications are possible due to the small dataset and limited follow-up imaging. This study provides preliminary evidence that radiotherapy outcomes are variable with intraventricular tumours, and some long-term survivors are noted.
Collapse
Affiliation(s)
- Katherine Sarah Hansen
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Chai Fei Li
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Alain Pierre Théon
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Michael Sean Kent
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| |
Collapse
|
11
|
Toedebusch RG, Wei NW, Simafranca KT, Furth-Jacobus JA, Brust-Mascher I, Stewart SL, Dickinson PJ, Woolard KD, Li CF, Vernau KM, Meyers FJ, Toedebusch CM. Intra- and Intertumoral Microglia/Macrophage Infiltration and Their Associated Molecular Signature Is Highly Variable in Canine Oligodendroglioma: A Preliminary Evaluation. Vet Sci 2023; 10:403. [PMID: 37368789 PMCID: PMC10303632 DOI: 10.3390/vetsci10060403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
The goal of this study was to define the glioma-associated microglia/macrophage (GAM) response and associated molecular landscape in canine oligodendrogliomas. Here, we quantified the intratumoral GAM density of low- and high-grade oligodendrogliomas compared to that of a normal brain, as well as the intratumoral concentration of several known GAM-derived pro-tumorigenic molecules in high-grade oligodendrogliomas compared to that in a normal brain. Our analysis demonstrated marked intra- and intertumoral heterogeneity of GAM infiltration. Correspondingly, we observed significant variability in the intratumoral concentrations of several GAM-associated molecules, unlike what we previously observed in high-grade astrocytomas. However, high-grade oligodendroglioma tumor homogenates (n = 6) exhibited an increase in the pro-tumorigenic molecules hepatocyte growth factor receptor (HGFR) and vascular endothelial growth factor (VEGF), as we observed in high-grade astrocytomas. Moreover, neoplastic oligodendrocytes displayed robust expression of GAL-3, a chimeric galectin implicated in driving immunosuppression in human glioblastoma. While this work identifies shared putative therapeutic targets across canine glioma subtypes (HGFR, GAL-3), it highlights several key differences in the immune landscape. Therefore, a continued effort to develop a comprehensive understanding of the immune microenvironment within each subtype is necessary to inform therapeutic strategies going forward.
Collapse
Affiliation(s)
- Ryan G. Toedebusch
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (R.G.T.); (N.-W.W.); (K.T.S.); (J.A.F.-J.); (P.J.D.); (C.-F.L.); (K.M.V.)
| | - Ning-Wei Wei
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (R.G.T.); (N.-W.W.); (K.T.S.); (J.A.F.-J.); (P.J.D.); (C.-F.L.); (K.M.V.)
| | - Kulani T. Simafranca
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (R.G.T.); (N.-W.W.); (K.T.S.); (J.A.F.-J.); (P.J.D.); (C.-F.L.); (K.M.V.)
| | - Jennie A. Furth-Jacobus
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (R.G.T.); (N.-W.W.); (K.T.S.); (J.A.F.-J.); (P.J.D.); (C.-F.L.); (K.M.V.)
| | - Ingrid Brust-Mascher
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA;
| | - Susan L. Stewart
- Division of Biostatistics, School of Medicine, University of California, Davis, CA 95616, USA;
- UC Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA;
| | - Peter J. Dickinson
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (R.G.T.); (N.-W.W.); (K.T.S.); (J.A.F.-J.); (P.J.D.); (C.-F.L.); (K.M.V.)
- UC Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA;
| | - Kevin D. Woolard
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA;
| | - Chai-Fei Li
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (R.G.T.); (N.-W.W.); (K.T.S.); (J.A.F.-J.); (P.J.D.); (C.-F.L.); (K.M.V.)
| | - Karen M. Vernau
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (R.G.T.); (N.-W.W.); (K.T.S.); (J.A.F.-J.); (P.J.D.); (C.-F.L.); (K.M.V.)
| | - Frederick J. Meyers
- UC Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA;
- Department of Internal Medicine, Division of Hematology and Oncology, Center for Precision Medicine, Microbiology, and Immunology, School of Medicine, University of California, Sacramento, CA 95817, USA
| | - Christine M. Toedebusch
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (R.G.T.); (N.-W.W.); (K.T.S.); (J.A.F.-J.); (P.J.D.); (C.-F.L.); (K.M.V.)
- UC Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA;
| |
Collapse
|
12
|
Schneider N, Blutke A, Matiasek K, Parzefall B. Intracranial Subdural Fluid Accumulation Associated with a Choroid Plexus Carcinoma in a Dog. Vet Sci 2022; 10:vetsci10010024. [PMID: 36669025 PMCID: PMC9867285 DOI: 10.3390/vetsci10010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/24/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
Choroid plexus tumors are commonly described as intraventricular mass lesions and account for 7-10% of intracranial, primary tumors in dogs. A 3-year-old Shetland sheepdog was presented with a history of slowly progressive lethargy, vision impairment and cognitive deficits. On magnetic resonance imaging, a subdural fluid accumulation (SFA) overlying and compressing the left parietotemporal lobe as well as multifocal changes consisting of cyst-like lesions, supposed intra-axial brain lesions and mild, multifocal meningeal thickening and generalized contrast enhancement were identified. Cerebrospinal fluid (CSF) analysis showed a mononuclear pleocytosis with negative results for infectious agents. The dog was treated with prednisolone followed by burr hole craniotomy with puncture of the SFA, which macroscopically appeared to be CSF-like fluid. After initial improvement, the dog deteriorated despite continuation of prednisolone and cytarabine therapy and was euthanized four weeks after surgery. Histopathology was consistent with a disseminated, neuroinvasive choroid plexus carcinoma (CPC) that involved the entire neuroaxis including the meninges of the brain and spinal cord. Immunohistochemical examination showed a strong Kir7.1 and a heterogenous cytokeratin-immunoreactivity in neoplastic cells. In conclusion, a CPC should be considered as a possible cause of a SFA even in the absence of an intraventricular mass lesion.
Collapse
Affiliation(s)
- Nina Schneider
- Small Animal Clinic Oberhaching, Bajuwarenring 10, 82041 Oberhaching, Germany
| | - Andreas Blutke
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, Ludwig-Maximilians Universität München, Veterinärstr. 13, 80539 Munich, Germany
| | - Kaspar Matiasek
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, Ludwig-Maximilians Universität München, Veterinärstr. 13, 80539 Munich, Germany
| | - Birgit Parzefall
- Small Animal Clinic Oberhaching, Bajuwarenring 10, 82041 Oberhaching, Germany
- Correspondence: ; Tel.: +49-89-63893020
| |
Collapse
|
13
|
Beckmann K, Kowalska M, Meier V. Solitary intraventricular tumors in dogs and cats treated with radiotherapy alone or combined with ventriculoperitoneal shunts: A retrospective descriptive case series. Vet Med (Auckl) 2022; 37:204-215. [PMID: 36382395 PMCID: PMC9889607 DOI: 10.1111/jvim.16583] [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: 06/23/2022] [Accepted: 10/31/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Intraventricular tumors are rare, optimal treatment is not defined. Symptomatic patients often exhibit life-threatening hydrocephalus. With several months time-to-effect after radiotherapy (RT), increased intracranial pressure is concerning. This increase in pressure can be overcome by ventriculoperitoneal shunting (VPS). OBJECTIVES Retrospective evaluation of outcome and complications in dogs and cats with intracranial tumors treated with either RT or VPS/RT. ANIMALS Twelve client-owned cats and dogs. METHODS Dogs and cats with symptomatic intraventricular tumors treated with definitive-intent RT or VPS/RT were included in a retrospective, descriptive case series. Complications, tumor volume evolution, time-to-progression, and survival time were determined. RESULTS Twelve animals were included: 1 cat and 5 dogs treated with single-modality RT and 4 cats and 2 dogs treated with VPS/RT. Neurological worsening seen in 4/6 animals during single-modality RT and 2/6 died during RT (suspected brain herniation). All dogs with VPS normalized clinically by the end of RT or earlier. Complications occurred in 4/6 animals, all but 1 were successfully managed surgically. Imaging follow-up in 8 animals surviving RT showed a marked decrease in tumor volume. Median survival time was 162 days (95% confidence interval [CI]: 16; infinity) for animals treated with RT and 1103 days (95%CI: 752; infinity) for animals treated with VPS/RT. Median time-to-progression was 71 days (95%CI: 7; infinity) and 895 days (95%CI: 704; infinity) for each group, respectively. Two dogs died because of intraventricular metastasis 427 and 461 days after single-modality RT. CONCLUSIONS AND CLINICAL IMPORTANCE Ventriculoperitoneal shunting led to rapid normalization of neurological signs and RT had a measurable effect on tumor volume. Combination of VPS/RT seems to be beneficial.
Collapse
Affiliation(s)
- Katrin Beckmann
- Department for Small Animals, Division of Surgery, Section of NeurologyVetsuisse Faculty, University of ZurichZurichSwitzerland
| | - Malwina Kowalska
- Section of Epidemiology, Vetsuisse FacultyUniversity of ZurichZurichSwitzerland,Ophthalmology Section, Equine DepartmentVetsuisse Faculty, University of ZurichZurichSwitzerland
| | - Valeria Meier
- Department for Small Animals, Division of Radiation OncologyVetsuisse Faculty, University of ZurichZurichSwitzerland
| |
Collapse
|
14
|
Kundapur V, Mayer M, Auer RN, Alexander A, Weibe S, Pushie MJ, Cranmer-Sargison G. Is Mini Beam Ready for Human Trials? Results of Randomized Study of Treating De-Novo Brain Tumors in Canines Using Linear Accelerator Generated Mini Beams. Radiat Res 2022; 198:162-171. [PMID: 35536992 DOI: 10.1667/rade-21-00093.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 04/22/2022] [Indexed: 11/03/2022]
Abstract
The main challenge in treating malignant brain neoplasms lies in eradicating the tumor while minimizing treatment-related damage. Conventional radiation treatments are associated with considerable side effects. Synchrotron generated micro-beam radiation (SMBRT) has shown to preserve brain architecture while killing tumor cells, however physical characteristics and limited facility access restrict its use. We have created a new clinical device which produces mini beams on a linear accelerator, to provide a new type of treatment called mini-beam radiation therapy (MBRT). The objective of this study is to compare the treatment outcomes of linear accelerator based MBRT versus standard radiation treatment (SRT), to evaluate the tumor response and the treatment-related changes in the normal brain with respect to each treatment type. Pet dogs with de-novo brain tumors were accrued for treatment. Dogs were randomized between standard fractionated stereotactic (9 Gy in 3 fractions) radiation treatment vs. a single fraction of MBRT (26 Gy mean dose). Dogs were monitored after treatment for clinical assessment and imaging. When the dogs were euthanized, a veterinary pathologist assessed the radiation changes and tumor response. We accrued 16 dogs, 8 dogs in each treatment arm. In the MBRT arm, 71% dogs achieved complete pathological remission. The radiation-related changes were all confined to the target region. Structural damage was not observed in the beam path outside of the target region. In contrast, none of the dogs in control group achieved remission and the treatment related damage was more extensive. Therapeutic superiority was observed with MBRT, including both tumor control and the normal structural preservation. The MBRT findings are suggestive of an immune related mechanism which is absent in standard treatment. These findings together with the widespread availability of clinical linear accelerators make MBRT a promising research topic to explore further treatment and clinical trial opportunities.
Collapse
Affiliation(s)
- V Kundapur
- Radiation Oncology, Saskatchewan Cancer Agency, Saskatoon Cancer Centre, Saskatoon, SK Canada S7N4H4
| | - M Mayer
- Veterinary Radiation Oncology, Department of Small Animal clinical Sciences, University of Saskatchewan, Saskatoon, SK Canada S7N 0W8
| | - R N Auer
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK Canada S7N 0W8
| | - A Alexander
- Radiation Physics, Saskatchewan Cancer Agency, Saskatoon Cancer Centre, Saskatoon, SK Canada S7N4H4
| | - S Weibe
- Department of Clinical Imaging, University of Saskatchewan, Saskatoon, SK Canada S7N 0W8
| | - M J Pushie
- Department of Surgery, University of Saskatchewan, Saskatoon, SK Canada S7N 0W8
| | - G Cranmer-Sargison
- Radiation Physics, Saskatchewan Cancer Agency, Saskatoon Cancer Centre, Saskatoon, SK Canada S7N4H4
| |
Collapse
|
15
|
Mahon E, Eiras-Diaz A, Mason S, Stabile F, Uriarte A. Case Report: Ventriculoperitoneal Shunting and Radiation Therapy Treatment in a Cat With a Suspected Choroid Plexus Tumor and Hypertensive Hydrocephalus. Front Vet Sci 2022; 9:828083. [PMID: 35400094 PMCID: PMC8989464 DOI: 10.3389/fvets.2022.828083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
A 14-year-old male neutered domestic short-hair cat was presented for a history of behavioral changes and episodes of urinary retention. Neurological examination was consistent with a multifocal intracranial neuroanatomical localization, with suspected right sided lateralisation and suspected raised intracranial pressure (ICP). Brain magnetic resonance imaging (MRI) revealed an intraventricular multilobulated well-defined T2W-hyperintense and T1W-isointense, markedly contrast enhancing mass lesion within the dorsal aspect of the III ventricle extending into the left lateral ventricle, causing hypertensive obstructive hydrocephalus. A ventriculoperitoneal shunt (VPS) was placed within the left lateral ventricle, followed by a radiation therapy (RT) course of 45 Gy total dose in 18 daily fractions. Six-months post-RT, computed tomography revealed mild reduction in mass size and resolution of the hydrocephalus. The patient was neurologically normal with no medical treatment. Raised ICP causes severe clinical signs, can lead to brain ischaemia and herniation, and significantly increases anesthetic risk during RT. Placement of a VPS in cats with hypertensive obstructive hydrocephalus may allow improvement of neurological signs due to raised ICP, and therefore making the patient a more stable candidate for anesthesia and radiation therapy.
Collapse
Affiliation(s)
- Elizabeth Mahon
- Department of Neurology and Neurosurgery, Southfields Veterinary Specialists, Essex, United Kingdom
| | - Aldara Eiras-Diaz
- Department of Internal Medicine, Southfields Veterinary Specialists, Essex, United Kingdom
| | - Sarah Mason
- Department of Oncology/Radiation Therapy, Southfields Veterinary Specialists, Essex, United Kingdom
| | - Fabio Stabile
- Department of Neurology and Neurosurgery, Southfields Veterinary Specialists, Essex, United Kingdom
| | - Ane Uriarte
- Department of Neurology and Neurosurgery, Southfields Veterinary Specialists, Essex, United Kingdom
| |
Collapse
|
16
|
Takahashi T, Kagawa Y, Ito D. Successful use of prednisolone and radiation therapy in a dog with intracranial histiocytic sarcoma. J Vet Med Sci 2021; 83:1782-1785. [PMID: 34556593 PMCID: PMC8636892 DOI: 10.1292/jvms.21-0206] [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] [Indexed: 11/22/2022] Open
Abstract
The ideal treatment for intracranial histiocytic sarcoma (HS) remains unclear. Herein, we report a case of intracranial HS that was successfully treated using prednisolone and radiation therapy. The patient was a 9-year-old spayed female Pembroke Welsh Corgi that presented with epileptic seizures. Magnetic resonance imaging revealed a contrast-enhancing mass adjacent to the right piriform lobe. Prednisolone administration (1 mg/kg/day) decreased the lesion size. Additional palliative radiation therapy (total dose, 37 Gy) resulted in complete disappearance of the lesion. However, on day 164, the dog’s neurological signs deteriorated, and she was euthanized. Necropsy revealed an intracranial metastasis of HS via the cerebrospinal fluid without any extracranial metastasis. Nonetheless, combined prednisolone and radiation therapy might be effective in treating intracranial HS.
Collapse
Affiliation(s)
- Tomoko Takahashi
- Laboratory of Veterinary Radiology, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | - Yumiko Kagawa
- North Lab, 8-35 Kita, 2 Hondori, Shiroishi-ku, Sapporo, Hokkaido 003-0027, Japan
| | - Daisuke Ito
- Laboratory of Veterinary Neurology, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| |
Collapse
|
17
|
Carter GL, Ogilvie GK, Mohammadian LA, Bergman PJ, Lee RP, Proulx DR. CyberKnife stereotactic radiotherapy for treatment of primary intracranial tumors in dogs. J Vet Intern Med 2021; 35:1480-1486. [PMID: 33755255 PMCID: PMC8163137 DOI: 10.1111/jvim.16086] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/05/2021] [Accepted: 02/17/2021] [Indexed: 12/19/2022] Open
Abstract
Background Limited data exist about the use, efficacy, and prognostic factors influencing outcome when CyberKnife is used to treat dogs with intracranial neoplasia. Objectives To determine the prognosis and associated prognostic factors for dogs that were imaged, determined to have primary intracranial tumors, and treated with CyberKnife radiotherapy. Animals Fifty‐nine dogs treated with CyberKnife radiotherapy for primary intracranial tumors. Methods Retrospective medical record review of cases from January 2010 to June 2016. Data extracted from medical records included signalment, weight, seizure history, tumor location, tumor type (based on imaging), gross tumor volume, planned tumor volume, treatment dates, radiation dose, recurrence, date of death, and cause of death. Results The median progression‐free interval (PFI) was 347 days (range 47 to 1529 days), and the median survival time (MST) was 738 days (range 4 to 2079 days). Tumor location was significantly associated with PFI when comparing cerebrum (median PFI 357 days; range 47‐1529 days) versus cerebellum (median PFI 97 days; range 97‐168 days) versus brainstem (median PFI 266 days; range 30‐1484 days), P = .03. Additionally, the presumed tumor type was significantly associated with MST (P < .001). Conclusions and Clinical Importance Use of Cyberknife and SRT might improve MST, compared with RT, in dogs with intracranial neoplasia.
Collapse
Affiliation(s)
| | - Gregory K Ogilvie
- VCA California Veterinary Specialists, Carlsbad, California, USA.,Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, USA
| | | | | | - Rachel P Lee
- VCA California Veterinary Specialists, Carlsbad, California, USA
| | - David R Proulx
- VCA California Veterinary Specialists, Carlsbad, California, USA
| |
Collapse
|
18
|
Toedebusch R, Grodzki AC, Dickinson PJ, Woolard K, Vinson N, Sturges B, Snyder J, Li CF, Nagasaka O, Consales B, Vernau K, Knipe M, Murthy V, Lein PJ, Toedebusch CM. Glioma-associated microglia/macrophages augment tumorigenicity in canine astrocytoma, a naturally occurring model of human glioma. Neurooncol Adv 2021; 3:vdab062. [PMID: 34131649 PMCID: PMC8193901 DOI: 10.1093/noajnl/vdab062] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Glioma-associated microglia/macrophages (GAMs) markedly influence glioma progression. Under the influence of transforming growth factor beta (TGFB), GAMs are polarized toward a tumor-supportive phenotype. However, neither therapeutic targeting of GAM recruitment nor TGFB signaling demonstrated efficacy in glioma patients despite efficacy in preclinical models, underscoring the need for a comprehensive understanding of the TGFB/GAM axis. Spontaneously occurring canine gliomas share many features with human glioma and provide a complementary translational animal model for further study. Given the importance of GAM and TGFB in human glioma, the aims of this study were to further define the GAM-associated molecular profile and the relevance of TGFB signaling in canine glioma that may serve as the basis for future translational studies. METHODS GAM morphometry, levels of GAM-associated molecules, and the canonical TGFB signaling axis were compared in archived samples of canine astrocytomas versus normal canine brain. Furthermore, the effect of TGFB on the malignant phenotype of canine astrocytoma cells was evaluated. RESULTS GAMs diffusely infiltrated canine astrocytomas. GAM density was increased in high-grade tumors that correlated with a pro-tumorigenic molecular signature and upregulation of the canonical TGFB signaling axis. Moreover, TGFB1 enhanced the migration of canine astrocytoma cells in vitro. CONCLUSIONS Canine astrocytomas share a similar GAM-associated immune landscape with human adult glioma. Our data also support a contributing role for TGFB1 signaling in the malignant phenotype of canine astrocytoma. These data further support naturally occurring canine glioma as a valid model for the investigation of GAM-associated therapeutic strategies for human malignant glioma.
Collapse
Affiliation(s)
- Ryan Toedebusch
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Ana Cristina Grodzki
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Peter J Dickinson
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Kevin Woolard
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Nicole Vinson
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Beverly Sturges
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - John Snyder
- Riemann Computing, LLC, St. Louis, Missouri, USA
| | - Chai-Fei Li
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Ori Nagasaka
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Blaire Consales
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Karen Vernau
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Marguerite Knipe
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Vishal Murthy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Christine M Toedebusch
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| |
Collapse
|
19
|
Definitive-intent uniform megavoltage fractioned radiotherapy protocol for presumed canine intracranial gliomas: retrospective analysis of survival and prognostic factors in 38 cases (2013-2019). BMC Vet Res 2020; 16:412. [PMID: 33129320 PMCID: PMC7603708 DOI: 10.1186/s12917-020-02614-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/07/2020] [Indexed: 12/15/2022] Open
Abstract
Background Radiotherapy (RT) is currently considered the treatment of choice for presumed canine intracranial gliomas. However, variable therapeutic responses are described, due to heterogeneous populations and different radiation methods or protocols. Only one study dedicated to intracranial suspected glioma highlighted prognostic criteria. Determination or confirmation of specific clinical and imaging prognostic factors may guide the therapeutic management of these tumours. The objectives were to provide data on long-term clinical outcome (including quality of life, QoL) and to determine specific prognostic factors associated with survival time. We report a single-institution retrospective study, including all dogs with suspected symptomatic primary solitary intracranial glioma, treated with a complete uniform fractionated megavoltage radiation protocol of 15x3Gy over 5 weeks, between January 2013 and February 2019. Thirty-eight client-owned dogs were included. Medical records were retrospectively evaluated for median overall survival time (MST), clinical and imaging responses. Prognostic factors on survival were researched in terms of signalment, clinical presentation, tumour imaging characteristics and response following RT. Finally, the RT’s impact on the dogs’ clinical signs and Qol were evaluated by the owners. Results The disease-specific MST was 698 days (95% CI: 598–1135). Survival at 1 and 2 years were respectively 74.2 ± 7.4% and 49.0 ± 9.8%. Initial clinical signs were related to survival, as well as tumour characteristics such as cystic-pattern, mass effect and Tumour/Brain volume ratio. No significant adverse effect or radiotoxicity was observed. Conclusions RT appears as a safe and effective treatment for canine intracranial gliomas, allowing long-term tumour control, improvement of life’s quality and management of associated clinical signs. The initial clinical signs and MRI characteristics (Tumour/Brain volume ratio, cyst-like lesion and mass effect) may help predict the prognosis.
Collapse
|
20
|
Morimoto CY, Waldner CL, Fan V, Sidhu N, Matthews Q, Randall E, Griffin L, Keyerleber M, Rancilio N, Vanhaezebrouck I, Zwueste D, Mayer MN. Use of MRI increases interobserver agreement on gross tumor volume for imaging-diagnosed canine intracranial meningioma. Vet Radiol Ultrasound 2020; 61:726-737. [PMID: 33090601 DOI: 10.1111/vru.12915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/21/2020] [Accepted: 07/21/2020] [Indexed: 01/01/2023] Open
Abstract
There is a lack of information regarding interobserver agreement on canine meningioma gross tumor volume (GTV) delineation, and on the impact of MRI on this agreement. The objectives of this retrospective, secondary analysis, observer agreement study were to describe agreement between veterinary radiation oncologists on GTV for canine intracranial meningioma, and to compare interobserver agreement between delineation based on CT alone and delineation based on fused CT-MRI. Eighteen radiation oncologists delineated GTV for 13 dogs with an imaging diagnosis of meningioma on pre- and postcontrast CT, pre- and postcontrast T1-weighted magnetic resonance, and T2-weighted magnetic resonance images. Dice similarity coefficient (DSC), concordance index (CI), and center of volume (COV) were used to quantify interobserver agreement. Multilevel mixed models were used to examine the difference in volume, DSC, CI and COV 3D distance between CT and CT-MR imaging. The mean volume for GTV contours delineated using fused CT-MRI was larger than when CT alone was used for delineation (mean difference CT-MR - CT = 0.89 cm3, 95% CI 0.66 to 1.12, P < .001). Interobserver agreement on GTV was improved when MRI was used; the mean DSC and CI were higher, and the mean COV 3D distance was lower, when fused CT-MRI was used than when CT alone was used (P < .001 for all differences). Based on our results, fused CT-MRI is recommended for radiation therapy planning of canine intracranial meningioma.
Collapse
Affiliation(s)
- Celina Y Morimoto
- Departments of Small Animal Clinical Sciences, University of Saskatchewan, Saskatoon, Canada
| | - Cheryl L Waldner
- Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Vivian Fan
- Departments of Small Animal Clinical Sciences, University of Saskatchewan, Saskatoon, Canada
| | | | | | - Elissa Randall
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Lynn Griffin
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Michele Keyerleber
- Department of Clinical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | - Nicholas Rancilio
- Department of Small Animal Clinical Sciences & Animal Cancer Care & Research Center, Virginia Maryland College of Veterinary Medicine, University of Maryland, Roanoke, Virginia, USA
| | | | - Danielle Zwueste
- Departments of Small Animal Clinical Sciences, University of Saskatchewan, Saskatoon, Canada
| | - Monique N Mayer
- Departments of Small Animal Clinical Sciences, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
21
|
Gieger TL, Nolan MW. Treatment outcomes and target delineation utilizing CT and MRI in 13 dogs treated with a uniform stereotactic radiation therapy protocol (16 Gy single fraction) for pituitary masses: (2014-2017). Vet Comp Oncol 2020; 19:17-24. [PMID: 32548944 DOI: 10.1111/vco.12627] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/18/2020] [Accepted: 06/06/2020] [Indexed: 12/31/2022]
Abstract
Canine pituitary tumours are increasingly treated with stereotactic radiotherapy (SRT). Here, we report clinical outcomes in dogs treated with single-fraction SRT; we also explore technical aspects of SRT treatment planning. A single-institution retrospective study was performed, including any dog with a pituitary mass (PM) that was treated using a standardized single-fraction (16 Gy) SRT protocol between 2014 and 2017. Via medical records review, 13 cases were identified. Nine dogs neurologically improved after SRT. Four dogs experienced MRI-documented tumour volume reduction. Nine dogs experienced neurologic decline in 1.5 to 18 months after SRT and were euthanized. The median overall survival time was 357 days, with 15% alive 18 months after SRT. To better understand whether SRT target delineation is predictably altered by use of magnetic resonance imaging (MRI) in addition to computed tomography (CT), two radiation oncologists (RO) retrospectively re-evaluated all imaging studies used for SRT planning in these 13 cases. Gross tumour volume (GTV) was contoured on co-registered CT and MRIs for each case. In seven cases, CT alone was deemed inadequate for GTV contouring by at least one RO. T1 post-contrast MRI was considered the ideal image for GTV contouring in 11 cases. Contouring on MRI yielded larger GTV than CT for 11 cases. Inter-observer variability existed in each case and was greater for MRI. In summary, use of co-registered CT and MRI images is generally considered advantageous for PM delineation when using SRT. Notably, survival times reported herein are shorter than what has previously been reported for PM treated with finely fractionated full-course RT protocols.
Collapse
Affiliation(s)
- Tracy L Gieger
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Michael W Nolan
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA.,Duke Cancer Institute, Duke University, Durham, North Carolina, USA
| |
Collapse
|
22
|
Van Asselt N, Christensen N, Meier V, Bley CR, Laliberte S, Poirier VJ, Desai N, Chen Y, Turek M. Definitive-intent intensity-modulated radiation therapy provides similar outcomes to those previously published for definitive-intent three-dimensional conformal radiation therapy in dogs with primary brain tumors: A multi-institutional retrospective study. Vet Radiol Ultrasound 2020; 61:481-489. [PMID: 32356907 PMCID: PMC7953428 DOI: 10.1111/vru.12868] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 02/26/2020] [Accepted: 03/09/2020] [Indexed: 12/14/2022] Open
Abstract
Radiotherapy with or without surgery is a common choice for brain tumors in dogs. Although numerous studies have evaluated use of three-dimensional conformal radiotherapy, reports of definitive-intent, IMRT for canine intracranial tumors are lacking. Intensity-modulated radiation therapy has the benefit of decreasing dose to nearby organs at risk and may aid in reducing toxicity. However, increasing dose conformity with IMRT calls for accurate target delineation and daily patient positioning, in order to decrease the risk of a geographic miss. To determine survival outcome and toxicity, we performed a multi-institutional retrospective observational study evaluating dogs with brain tumors treated with IMRT. Fifty-two dogs treated with fractionated, definitive-intent IMRT at four academic radiotherapy facilities were included. All dogs presented with neurologic signs and were diagnosed via MRI. Presumed radiological diagnoses included 37 meningiomas, 12 gliomas, and one peripheral nerve sheath tumor. One dog had two presumed meningiomas and one dog had either a glioma or meningioma. All dogs were treated in the macroscopic disease setting and were prescribed a total dose of 45-50 Gy (2.25-2.5 Gy per fraction in 18-20 daily fractions). Median survival time for all patients, including seven cases treated with a second course of therapy was 18.1 months (95% confidence of interval 12.3-26.6 months). As previously described for brain tumors, increasing severity of neurologic signs at diagnosis was associated with a worse outcome. Intensity-modulated radiation therapy was well tolerated with few reported acute, acute delayed, or late side effects.
Collapse
Affiliation(s)
- Nathaniel Van Asselt
- University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, USA
| | - Neil Christensen
- University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, USA
| | - Valeria Meier
- Division of Radiation Oncology, Vetsuisse Faculty, University of Zurich, Switzerland
- Department of Physics, University of Zurich, Switzerland
| | - Carla Rohrer Bley
- Division of Radiation Oncology, Vetsuisse Faculty, University of Zurich, Switzerland
| | - Sarah Laliberte
- University of Guelph Ontario Veterinary College, Guelph, Ontario, Canada
| | - Valerie J. Poirier
- University of Guelph Ontario Veterinary College, Guelph, Ontario, Canada
| | - Noopur Desai
- The Ohio State University College of Veterinary Medicine, Columbus, Ohio, USA
| | - Yi Chen
- University of Wisconsin-Madison Department of Biostatistics and Medical Informatics, Madison, Wisconsin, USA
| | - Michelle Turek
- University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, USA
| |
Collapse
|
23
|
Orlandi R, Vasilache CG, Mateo I. Palliative ventriculoperitoneal shunting in dogs with obstructive hydrocephalus caused by tumors affecting the third ventricle. J Vet Intern Med 2020; 34:1556-1562. [PMID: 32472726 PMCID: PMC7379019 DOI: 10.1111/jvim.15818] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Hypertensive or obstructive hydrocephalus is a common complication in dogs with tumors affecting the third ventricle for which few therapeutic options are available. OBJECTIVES To describe signalment, neurological status, and pre- and postsurgical findings, complications and survival time in 4 dogs with obstructive hydrocephalus caused by third ventricle tumors that were palliatively treated using ventriculoperitoneal shunting (VPS). ANIMALS Four client-owned dogs with obstructive hydrocephalus caused by tumors affecting the third ventricle. METHODS Medical records were reviewed for dogs diagnosed with third ventricular tumors. Inclusion criteria were complete medical record, advanced diagnostic imaging for review, and VPS as sole surgical treatment. RESULTS At the time of diagnosis, all patients displayed acute onset and rapidly progressive diffuse intracranial clinical signs. On advanced imaging, all dogs had a homogeneously enhancing mass occupying or collapsing the third ventricle as well as obstructive hydrocephalus. All of the dogs underwent VPS of the most dilated lateral ventricle. In 2 of the patients, intracranial hypertension followed by normotension after VPS placement was confirmed intraoperatively by means of direct intracranial pressure monitoring. Excellent clinical improvement was observed in all dogs immediately after surgery. Three patients required a second VPS in the contralateral lateral ventricle 3, 7 and 11 months after the first surgery, all of them with renewed improvement in clinical signs. CONCLUSION AND CLINICAL IMPORTANCE Ventriculoperitoneal shunting is a rapid and effective treatment for patients with obstructive (hypertensive) hydrocephalus caused by tumors located within the third ventricle.
Collapse
Affiliation(s)
- Rocio Orlandi
- Servicio de Neurología, Hospital Veterinario VETSIA, Madrid, Spain
| | | | - Isidro Mateo
- Servicio de Neurología, Hospital Veterinario VETSIA, Madrid, Spain.,Servicio de Neurología, Hospital Clínico Veterinario, Universidad Alfonso X el Sabio, Madrid, Spain
| |
Collapse
|
24
|
Moirano SJ, Dewey CW, Haney S, Yang J. Efficacy of frameless stereotactic radiotherapy for the treatment of presumptive canine intracranial gliomas: A retrospective analysis (2014-2017). Vet Comp Oncol 2020; 18:528-537. [PMID: 32011065 DOI: 10.1111/vco.12573] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 01/27/2020] [Accepted: 01/28/2020] [Indexed: 01/21/2023]
Abstract
The use of conventional multi-fractionated radiotherapy for the treatment of glial tumours is well documented in the literature. Recently, stereotactic radiotherapy (SRT) has become more widely available allowing for hypo-fractionated protocols; however, its usefulness in the treatment of canine intracranial gliomas is largely undetermined. We conducted a retrospective analysis, including 21 dogs diagnosed with presumptive intracranial gliomas treated with one or more courses of three fractions of 8 to 10 Gy CyberKnife SRT. The objective of this study was to evaluate the efficacy, safety and prognostic factors associated with the use of SRT for the treatment of canine intracranial gliomas. Overall MST for all dogs was 636 days (d). Dogs treated with one course of the described SRT protocol had a MST of 258 days while those treated with >1 course had a MST of 865 days (P = .0077 log rank, 0.0139 Wilcoxon). Dogs treated with one course of SRT who received adjuvant chemotherapy had a MST of >658 days and lived significantly longer than those who did not receive chemotherapy (MST, 230 days) (P = .0414 log rank, 0.0453 Wilcoxon). The most common adverse event included presumptive transient demyelination in 3/21 dogs, which was treated successfully with corticosteroids in all patients. This study provides evidence that SRT is effective in prolonging survival in dogs with intracranial gliomas, and may provide similar results to conventional fractionated protocols, while decreasing the number of hospital visits and anaesthetic episodes. Additionally, it appears that patients can be safely treated with multiple rounds of SRT resulting in improved survival times.
Collapse
Affiliation(s)
- Steven J Moirano
- General Medicine and Surgery, The Animal Medical Center, New York City, New York
| | - Curtis W Dewey
- Department of Clinical Sciences, Cornell University, College of Veterinary Medicine, Ithaca, New York.,Department of Neurology/Neurosurgery, Long Island Veterinary Specialists, Plainview, New York.,Department of Neurology, Veterinary Specialists and Emergency Services of Rochester, Rochester, New York
| | - Siobhan Haney
- Department of Radiation Oncology and Cyberknife, Hope Veterinary Specialists and the Veterinary CyberKnife Cancer Center, Malvern, Pennsylvania
| | - Jun Yang
- Department of Radiation Oncology, Drexel University, Philadelphia, Pennsylvania
| |
Collapse
|
25
|
Fox-Alvarez S, Shiomitsu K, Lejeune AT, Szivek A, Kubicek L. Outcome of intensity-modulated radiation therapy-based stereotactic radiation therapy for treatment of canine nasal carcinomas. Vet Radiol Ultrasound 2020; 61:370-378. [PMID: 32189433 DOI: 10.1111/vru.12854] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 11/09/2019] [Accepted: 12/23/2019] [Indexed: 12/31/2022] Open
Abstract
Stereotactic radiation therapy (SRT) has emerged as a convenient definitive treatment modality in veterinary medicine, but few studies exist evaluating outcome with treatment for canine nasal tumors, and no studies report the treatment of one single tumor histotype. This retrospective, observational study evaluates toxicity, response, and survival in 17 dogs with nasal carcinomas treated with SRT. Dogs received a median of 3000 centigray in three fractions via 6-MV linear accelerator. Eighty-eight percent of patients (n = 15) demonstrated clinical benefit. Of dogs with repeated CT imaging (n = 10), 60% (n = 6) achieved a partial response and 10% (n = 1) achieved a complete response. Median progression-free survival (PFS) was 359 days. Median survival time (MST) was 563 days. Among dogs evaluable for acute toxicity, 50% (n = 10) developed low grade toxicity (grade 1, n = 4; grade 2, n = 1). No patients developed grade 3 toxicity. 16 dogs (87%) evaluable over the long term developed signs consistent with possible late toxicity. The majority of late toxicities were mild (alopecia, hyperpigmentation, and leukotrichia n = 10; ocular discharge and keratoconjunctivitis sicca n = 5). Thirty-seven percent of patients (n = 6) developed seven possible grade 3 late toxicities (blindness, n = 3; fistula, n = 1; seizures, n = 3), which were difficult to distinguish from progressive disease in most patients. Of the prognostic factors evaluated (demographics, tumor stage, dosimetric data, epistaxis, facial deformity, clinical response, image-based response, nonsteroidal anti-inflammatory drugs, and chemotherapy), only clinical response was a positive prognostic factor on MST (P < .00). No factors were found to be significantly associated with PFS.
Collapse
Affiliation(s)
- Stacey Fox-Alvarez
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Keijiro Shiomitsu
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Amandine T Lejeune
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Anna Szivek
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Lyndsay Kubicek
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida.,Angell Animal Medical Center, Boston, Massachusetts
| |
Collapse
|
26
|
Partridge B, Rossmeisl JH. Companion animal models of neurological disease. J Neurosci Methods 2020; 331:108484. [PMID: 31733285 PMCID: PMC6942211 DOI: 10.1016/j.jneumeth.2019.108484] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 10/28/2019] [Accepted: 10/28/2019] [Indexed: 02/07/2023]
Abstract
Clinical translation of novel therapeutics that improve the survival and quality of life of patients with neurological disease remains a challenge, with many investigational drug and device candidates failing in advanced stage clinical trials. Naturally occurring inherited and acquired neurological diseases, such as epilepsy, inborn errors of metabolism, brain tumors, spinal cord injury, and stroke occur frequently in companion animals, and many of these share epidemiologic, pathophysiologic and clinical features with their human counterparts. As companion animals have a relatively abbreviated lifespan and genetic background, are immunocompetent, share their environment with human caregivers, and can be clinically managed using techniques and tools similar to those used in humans, they have tremendous potential for increasing the predictive value of preclinical drug and device studies. Here, we review comparative features of spontaneous neurological diseases in companion animals with an emphasis on neuroimaging methods and features, illustrate their historical use in translational studies, and discuss inherent limitations associated with each disease model. Integration of companion animals with naturally occurring disease into preclinical studies can complement and expand the knowledge gained from studies in other animal models, accelerate or improve the manner in which research is translated to the human clinic, and ultimately generate discoveries that will benefit the health of humans and animals.
Collapse
Affiliation(s)
- Brittanie Partridge
- Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA; Brain Tumor Center of Excellence, Wake Forest University Comprehensive Cancer Center, Medical Center Blvd, NRC 405, Winston Salem, NC, 27157, USA
| | - John H Rossmeisl
- Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA; Brain Tumor Center of Excellence, Wake Forest University Comprehensive Cancer Center, Medical Center Blvd, NRC 405, Winston Salem, NC, 27157, USA.
| |
Collapse
|
27
|
Miller AD, Miller CR, Rossmeisl JH. Canine Primary Intracranial Cancer: A Clinicopathologic and Comparative Review of Glioma, Meningioma, and Choroid Plexus Tumors. Front Oncol 2019; 9:1151. [PMID: 31788444 PMCID: PMC6856054 DOI: 10.3389/fonc.2019.01151] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/16/2019] [Indexed: 12/22/2022] Open
Abstract
In the dog, primary intracranial neoplasia represents ~2-5% of all cancers and is especially common in certain breeds including English and French bulldogs and Boxers. The most common types of primary intracranial cancer in the dog are meningioma, glioma, and choroid plexus tumors, generally occurring in middle aged to older dogs. Much work has recently been done to understand the characteristic imaging and clinicopathologic features of these tumors. The gross and histologic landscape of these tumors in the dog compare favorably to their human counterparts with many similarities noted in histologic patterns, subtype, and grades. Data informing the underlying molecular abnormalities in the canine tumors have only begun to be unraveled, but reveal similar pathways are mutated between canine and human primary intracranial neoplasia. This review will provide an overview of the clinicopathologic features of the three most common forms of primary intracranial cancer in the dog, delve into the comparative aspects between the dog and human neoplasms, and provide an introduction to current standard of care while also highlighting novel, experimental treatments that may help bridge the gap between canine and human cancer therapies.
Collapse
Affiliation(s)
- Andrew D. Miller
- Section of Anatomic Pathology, Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY, United States
| | - C. Ryan Miller
- Division of Neuropathology, Department of Pathology, O'Neal Comprehensive Cancer Center and Comprehensive Neuroscience Center, University of Alabama School of Medicine, Birmingham, AL, United States
| | - John H. Rossmeisl
- Section of Neurology and Neurosurgery, Veterinary and Comparative Neuro-Oncology Laboratory, Department of Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, United States
| |
Collapse
|
28
|
Morimoto CY, Mayer MN, Sidhu N, Bloomfield R, Waldner CL. Residual setup error in the canine intracranial region after megavoltage, kilovoltage, or cone-beam computed tomographic image guidance for radiation therapy. Vet Comp Oncol 2019; 18:199-205. [PMID: 31433554 DOI: 10.1111/vco.12532] [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: 04/23/2019] [Revised: 07/22/2019] [Accepted: 08/16/2019] [Indexed: 11/27/2022]
Abstract
Sources of residual setup error after image guidance include image localization accuracy, errors associated with image registration, and inability of some treatment couches to correct submillimeter translational errors and/or pitch and roll errors. The purpose of this experimental study was to measure setup error after image-guided correction of the canine intracranial region, using a four degrees-of-freedom couch capable of 1 mm translational moves. Six cadaver dogs were positioned 45 times as for clinical treatment using a vacuum deformable body cushion, a customizable head cushion, a thermoplastic mask and an indexed maxillary plate with a dental mould. The location of five fiducial markers in the skull bones was compared between the reference position and after megavoltage (MV), kilovoltage (kV) and cone-beam computed tomography (CBCT)-guided correction using orthogonal kV images. The mean three-dimensional distance vectors (3DDV) after MV, kV and CBCT-guided correction were 1.7, 1.5 and 2.2 mm, respectively. All values were significantly different (P < .01). The 95th percentiles of the 3DDV after online MV, kV and CBCT-guided correction were 2.8, 2.6 and 3.6 mm, respectively. Residual setup error in the clinical scenario examined was on the order of millimetres and should be considered when choosing PTV margins for image-guided radiation therapy of the canine intracranial region.
Collapse
Affiliation(s)
- Celina Y Morimoto
- Department of Small Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Monique N Mayer
- Department of Small Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Narinder Sidhu
- British Columbia Cancer, Prince George, British Columbia, Canada
| | - Rachel Bloomfield
- Department of Small Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Cheryl L Waldner
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
29
|
Schlein LJ, Fadl-Alla B, Pondenis HC, Lezmi S, Eberhart CG, LeBlanc AK, Dickinson PJ, Hergenrother PJ, Fan TM. Immunohistochemical Characterization of Procaspase-3 Overexpression as a Druggable Target With PAC-1, a Procaspase-3 Activator, in Canine and Human Brain Cancers. Front Oncol 2019; 9:96. [PMID: 30859090 PMCID: PMC6397847 DOI: 10.3389/fonc.2019.00096] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 02/04/2019] [Indexed: 11/24/2022] Open
Abstract
Gliomas and meningiomas are the most common brain neoplasms affecting both humans and canines, and identifying druggable targets conserved across multiple brain cancer histologies and comparative species could broadly improve treatment outcomes. While satisfactory cure rates for low grade, non-invasive brain cancers are achievable with conventional therapies including surgery and radiation, the management of non-resectable or recurrent brain tumors remains problematic and necessitates the discovery of novel therapies that could be accelerated through a comparative approach, such as the inclusion of pet dogs with naturally-occurring brain cancers. Evidence supports procaspase-3 as a druggable brain cancer target with PAC-1, a pro-apoptotic, small molecule activator of procaspase-3 that crosses the blood-brain barrier. Procaspase-3 is frequently overexpressed in malignantly transformed tissues and provides a preferential target for inducing cancer cell apoptosis. While preliminary evidence supports procaspase-3 as a viable target in preclinical models, with PAC-1 demonstrating activity in rodent models and dogs with spontaneous brain tumors, the broader applicability of procaspase-3 as a target in human brain cancers, as well as the comparability of procaspase-3 expressions between differing species, requires further investigation. As such, a large-scale validation of procaspase-3 as a druggable target was undertaken across 651 human and canine brain tumors. Relative to normal brain tissues, procaspase-3 was overexpressed in histologically diverse cancerous brain tissues, supporting procaspase-3 as a broad and conserved therapeutic target. Additionally, procaspase-3 expressing glioma and meningioma cell lines were sensitive to the apoptotic effects of PAC-1 at biologically relevant exposures achievable in cancer patients. Importantly, the clinical relevance of procaspase-3 as a potential prognostic variable was demonstrated in human astrocytomas of variable histologic grades and associated clinical outcomes, whereby tumoral procaspase-3 expression was negatively correlated with survival; findings which suggest that PAC-1 might provide the greatest benefit for patients with the most guarded prognoses.
Collapse
Affiliation(s)
- Lisa J. Schlein
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Bahaa Fadl-Alla
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Holly C. Pondenis
- Department of Veterinary Clinical Medicine and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Stéphane Lezmi
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Charles G. Eberhart
- Department of Neuropathology and Ophthalmic Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Amy K. LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Peter J. Dickinson
- Department of Surgical and Radiological Sciences, University of California, Davis, Davis, CA, United States
| | - Paul J. Hergenrother
- Department of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Timothy M. Fan
- Department of Veterinary Clinical Medicine and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
30
|
Yu S, Morrow D, Moutrie V, Lurie D. The Elekta Fraxion™ system is not suitable for maxillary fixation in canine conformal radiation therapy techniques. Vet Radiol Ultrasound 2019; 60:233-240. [PMID: 30656773 DOI: 10.1111/vru.12710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/29/2018] [Accepted: 10/03/2018] [Indexed: 11/28/2022] Open
Abstract
In this prospective, exploratory study, we evaluated the positioning accuracy in a group of 15 dogs undergoing fractionated stereotactic radiotherapy for tumors affecting the head, using a modified human maxillary fixation device (Elekta Fraxion™ system). Positioning was assessed using on-board volumetric imaging, with a six-degrees-of-freedom image registration technique. Prior to treatment delivery, CBCT images were obtained and patient alignment was corrected, in both translational and rotational planes, using a six-degrees-of-freedom robotic patient positioning system (HexaPOD Evo RT System). The maximum angular inter-fraction motions observed were 6.1° (yaw), 10.9° (pitch), and 4.5° (roll). The mean systematic translational errors were 4.7, 2.6, and 2.3 mm, mean random translational errors were 3.0, 2.2, and 2.5 mm, and mean overall translational errors were 2.4, 0.7, and 2.3 mm in the cranial-caudal, lateral, and dorsal-ventral directions, respectively. The mean systematic rotational errors were 1.17°, 0.77°, and 1.43°, the mean rotational random errors were 1.65°, 1.46°, and 1.34° and the mean overall rotational errors were 0.56°, 0.22°, and 0.29° in the yaw, pitch, and roll directions, respectively. The mean error of the three-dimensional vector was 6.9 mm with a standard deviation of 3.8 mm. Ninety-five percent of the three-dimensional vectors were <14.8 mm. This study demonstrates that this maxillary fixation device relies on six-degrees-of-freedom registration and an ability to apply corrections using a six-degrees-of-freedom couch for accurate patient positioning and tumor targeting. Its use in conformal radiation therapy in dogs is not recommended.
Collapse
Affiliation(s)
- Sonya Yu
- Oncology Department, Animal Referral Hospital, Homebush West, New South Wales, Australia
| | - Deanna Morrow
- Waratah Private Hospital, Hurstville, New South Wales, Australia
| | - Vaughan Moutrie
- Genesis Cancer Care, Macquarie University Hospital, Sydney, New South Wales, Australia
| | - David Lurie
- Oncology Department, Animal Referral Hospital, Homebush West, New South Wales, Australia
| |
Collapse
|
31
|
Hansen KS, Zwingenberger AL, Théon AP, Kent MS. Long-term survival with stereotactic radiotherapy for imaging-diagnosed pituitary tumors in dogs. Vet Radiol Ultrasound 2018; 60:219-232. [PMID: 30575174 DOI: 10.1111/vru.12708] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 09/20/2018] [Accepted: 09/23/2018] [Indexed: 12/16/2022] Open
Abstract
Published studies on the use of stereotactic radiotherapy for dogs with pituitary tumors are limited. This retrospective observational study describes results of stereotactic radiotherapy for 45 dogs with imaging-diagnosed pituitary tumors. All dogs were treated at a single hospital during the period of December 2009-2015. The stereotactic radiotherapy was delivered in one 15 Gray (Gy) fraction or in three 8 Gy fractions. At the time of analysis, 41 dogs were deceased. Four were alive and censored from all survival analyses; one dog received 8 Gy every other day and was removed from protocol analyses. The median overall survival from first treatment was 311 days (95% confidence interval 226-410 days [range 1-2134 days]). Thirty-two dogs received 15 Gy (median overall survival 311 days; 95% confidence interval [range 221-427 days]), and 12 received 24 Gy on three consecutive days (median overall survival 245 days, 95% confidence interval [range 2-626 days]). Twenty-nine dogs had hyperadrenocorticism (median overall survival 245 days), while 16 had nonfunctional masses (median overall survival 626 days). Clinical improvement was reported in 37/45 cases. Presumptive signs of acute adverse effects within 4 months of stereotactic radiotherapy were noted in 10/45, and most had improvement spontaneously or with steroids. Late effects versus tumor progression were not discernable, but posttreatment blindness (2), hypernatremia (2), and progressive neurological signs (31) were reported. There was no statistical difference in median overall survival for different protocols. Patients with nonfunctional masses had longer median overall survival than those with hyperadrenocorticism (P = 0.0003). Survival outcomes with stereotactic radiotherapy were shorter than those previously reported with definitive radiation, especially for dogs with hyperadrenocorticism.
Collapse
Affiliation(s)
- Katherine S Hansen
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, 95616
| | - Allison L Zwingenberger
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, 95616
| | - Alain P Théon
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, 95616
| | - Michael S Kent
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, 95616
| |
Collapse
|
32
|
Sanders K, Kooistra HS, Galac S. Treating canine Cushing's syndrome: Current options and future prospects. Vet J 2018; 241:42-51. [PMID: 30340659 DOI: 10.1016/j.tvjl.2018.09.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/17/2018] [Accepted: 09/25/2018] [Indexed: 12/17/2022]
Abstract
Naturally occurring hypercortisolism, also known as Cushing's syndrome, is a common endocrine disorder in dogs that can be caused by an adenocorticotrophic hormone (ACTH)-producing pituitary adenoma (pituitary-dependent hypercortisolism, PDH; 80-85% of cases), or by an adrenocortical tumor (ACT; 15-20% of cases). To determine the optimal treatment strategy, differentiating between these two main causes is essential. Good treatment options are surgical removal of the causal tumor, i.e. hypophysectomy for PDH and adrenalectomy for an ACT, or radiotherapy in cases with PDH. Because these options are not without risks, not widely available and not suitable for every patient, pharmacotherapy is often used. In cases with PDH, the steroidogenesis inhibitor trilostane is most often used. In cases with an ACT, either trilostane or the adrenocorticolytic drug mitotane can be used. Although mostly effective, both treatments have disadvantages. This review discusses the current treatment options for canine hypercortisolism, and considers their mechanism of action, efficacy, adverse effects, and effect on survival. In addition, developments in both adrenal-targeting and pituitary-targeting drugs that have the potential to become future treatment options are discussed, as a more selective and preferably also tumor-targeted approach could have many advantages for both PDH and ACTs.
Collapse
Affiliation(s)
- K Sanders
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 108, 3584 CM Utrecht, The Netherlands
| | - H S Kooistra
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 108, 3584 CM Utrecht, The Netherlands
| | - S Galac
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 108, 3584 CM Utrecht, The Netherlands.
| |
Collapse
|
33
|
Dunfield EM, Turek MM, Buhr KA, Christensen NI. A survey of stereotactic radiation therapy in veterinary medicine. Vet Radiol Ultrasound 2018; 59:786-795. [PMID: 30062728 DOI: 10.1111/vru.12671] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 05/14/2018] [Accepted: 05/22/2018] [Indexed: 12/31/2022] Open
Abstract
Radiotherapy plays an important role in curative and palliative cancer treatment. As a novel radiation delivery technique, stereotactic radiotherapy utilizes three-dimensional-conformal treatment planning, high-precision beam delivery technology, and patient specific position verification to target tumors, often in one to five high-dose fractions. Currently, there is no consensus about best stereotactic radiotherapy practices in veterinary radiotherapy. The objective of this study was to document the breadth of perspectives, techniques, and applications of stereotactic radiotherapy in veterinary medicine. We conducted an online survey of American College of Veterinary Radiology members specializing in radiation oncology to assess how, when, and why stereotactic radiotherapy is being used. Both stereotactic radiotherapy users and nonusers completed the survey. The overall response and survey completion rates were 54% (67/123) and 87% (58/67), respectively. Overall, 55% of respondents reported providing stereotactic radiotherapy at their facility, with a median of 4.5 canine cases and one feline case per month. Delivery methods included C-arm linear accelerator with multi-leaf collimator, helical tomotherapy, and CyberKnife. Nonpituitary intracranial tumors, pituitary tumors, and sinonasal tumors were the most common cancers treated using stereotactic radiotherapy in both species. The most common fractionation scheme was three fractions of 10 Gy/fraction. The results of this survey suggest common availability of stereotactic radiotherapy in veterinary radiation facilities. These results provide valuable information regarding current stereotactic radiotherapy practices in veterinary medicine, and may provide an initial step toward standardizing methods and establishing consensus guidelines.
Collapse
Affiliation(s)
- Elizabeth M Dunfield
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, 53706
| | - Michelle M Turek
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, 53706
| | - Kevin A Buhr
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706
| | - Neil I Christensen
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, 53706
| |
Collapse
|
34
|
Berlato D, Zwingenberger AL, Ruiz-Drebing M, Pradel J, Clark N, Kent MS. Canine meningiomas treated with three-dimensional conformal radiation therapy require magnetic resonance imaging to avoid a geographic miss. Vet Radiol Ultrasound 2018; 59:777-785. [DOI: 10.1111/vru.12653] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/11/2018] [Accepted: 04/12/2018] [Indexed: 12/31/2022] Open
Affiliation(s)
- Davide Berlato
- Animal Health Trust; Centre for Small Animal Studies; Suffolk CB87UU UK
| | - Allison L Zwingenberger
- Department of Surgical and Radiological Sciences; School of Veterinary Medicine; University of California, Davis; Davis CA 95616
| | | | - Julie Pradel
- Animal Health Trust; Centre for Small Animal Studies; Suffolk CB87UU UK
| | - Nicola Clark
- Animal Health Trust; Centre for Small Animal Studies; Suffolk CB87UU UK
| | - Michael S Kent
- Department of Surgical and Radiological Sciences; School of Veterinary Medicine; University of California, Davis; Davis CA 95616
| |
Collapse
|
35
|
Dolera M, Malfassi L, Marcarini S, Mazza G, Carrara N, Pavesi S, Sala M, Finesso S, Urso G. High dose hypofractionated frameless volumetric modulated arc radiotherapy is a feasible method for treating canine trigeminal nerve sheath tumors. Vet Radiol Ultrasound 2018; 59:624-631. [PMID: 29885013 DOI: 10.1111/vru.12637] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 10/18/2017] [Accepted: 10/19/2017] [Indexed: 12/31/2022] Open
Abstract
The aim of this prospective pilot study was to evaluate the feasibility and effectiveness of curative intent high dose hypofractionated frameless volumetric modulated arc radiotherapy for treatment of canine trigeminal peripheral nerve sheath tumors. Client-owned dogs with a presumptive imaging-based diagnosis of trigeminal peripheral nerve sheath tumor were recruited for the study during the period of February 2010 to December 2013. Seven dogs were enrolled and treated with high dose hypofractionated volumetric modulated arc radiotherapy delivered by a 6 MV linear accelerator equipped with a micro-multileaf beam collimator. The plans were computed using a Monte Carlo algorithm with a prescription dose of 37 Gy delivered in five fractions on alternate days. Overall survival was estimated using a Kaplan-Meier curve analysis. Magnetic resonance imaging (MRI) follow-up examinations revealed complete response in one dog, partial response in four dogs, and stable disease in two dogs. Median overall survival was 952 days with a 95% confidence interval of 543-1361 days. Volumetric modulated arc radiotherapy was demonstrated to be feasible and effective for trigeminal peripheral nerve sheath tumor treatment in this sample of dogs. The technique required few sedations and spared organs at risk. Even though larger studies are required, these preliminary results supported the use of high dose hypofractionated volumetric modulated arc radiotherapy as an alternative to other treatment modalities.
Collapse
Affiliation(s)
- Mario Dolera
- La Cittadina Fondazione Studi e Ricerche Veterinarie, Lodi, Italy
| | - Luca Malfassi
- La Cittadina Fondazione Studi e Ricerche Veterinarie, Lodi, Italy
| | - Silvia Marcarini
- La Cittadina Fondazione Studi e Ricerche Veterinarie, Lodi, Italy
| | - Giovanni Mazza
- La Cittadina Fondazione Studi e Ricerche Veterinarie, Lodi, Italy
| | - Nancy Carrara
- La Cittadina Fondazione Studi e Ricerche Veterinarie, Lodi, Italy
| | - Simone Pavesi
- La Cittadina Fondazione Studi e Ricerche Veterinarie, Lodi, Italy
| | - Massimo Sala
- La Cittadina Fondazione Studi e Ricerche Veterinarie, Lodi, Italy
| | - Sara Finesso
- La Cittadina Fondazione Studi e Ricerche Veterinarie, Lodi, Italy
| | - Gaetano Urso
- Azienda Ospedaliera della Provincia di Lodi, Lodi, Italy
| |
Collapse
|
36
|
Kelsey KL, Gieger TL, Nolan MW. Single fraction stereotactic radiation therapy (stereotactic radiosurgery) is a feasible method for treating intracranial meningiomas in dogs. Vet Radiol Ultrasound 2018; 59:632-638. [PMID: 29873144 DOI: 10.1111/vru.12636] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 12/19/2017] [Accepted: 03/08/2018] [Indexed: 02/06/2023] Open
Abstract
The aim of this retrospective, pilot study was to evaluate stereotactic radiosurgery as a method for treating intracranial meningiomas in dogs. Included dogs had an imaging diagnosis of presumed intracranial meningioma, were treated using a standardized stereotactic radiosurgery protocol, and had a follow-up time of >6 months after stereotactic radiosurgery. A single fraction of 16 Gy stereotactic radiosurgery was delivered to the tumor, with an internal simultaneously integrated boost to a total dose of 20-24 Gy to the central portion of the tumor. Thirty-two dogs were sampled. One dog was euthanized in the periprocedural period, and 10 of the remaining 31 dogs (31%) experienced an acute adverse event (defined as declining neurologic function due to tumor progression or treatment-associated complication within the first 6 months after stereotactic radiosurgery), three of which were fatal. Too few subjects (n = 6) had cross-sectional imaging after stereotactic radiosurgery to determine an objective response rate; however, 17/30 (57%) dogs assessed for response had a perceived clinical benefit from treatment. The overall median survival time was 519 days (95% confidence interval: 330-708 days); 64% and 24% of dogs were alive at 1 and 2 years after stereotactic radiosurgery, respectively. Dogs with infratentorial tumor location and high gradient indices had shorter survival. There were no factors identified which were predictive of acute adverse event. Survival times reported herein are similar to what has previously been reported for other stereotactic and traditional fractionated radiotherapy protocols. Findings therefore supported the use of stereotactic radiosurgery as an alternative method for treating dogs with presumed intracranial meningiomas.
Collapse
Affiliation(s)
- Krista L Kelsey
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC, 27607
| | - Tracy L Gieger
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC, 27607.,Comparative Medicine Institute, North Carolina State University College of Veterinary Medicine, Raleigh, NC, 27607
| | - Michael W Nolan
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC, 27607.,Comparative Medicine Institute, North Carolina State University College of Veterinary Medicine, Raleigh, NC, 27607
| |
Collapse
|
37
|
Packer RA, Rossmeisl JH, Kent MS, Griffin JF, Mazcko C, LeBlanc AK. Consensus recommendations on standardized magnetic resonance imaging protocols for multicenter canine brain tumor clinical trials. Vet Radiol Ultrasound 2018. [PMID: 29522650 DOI: 10.1111/vru.12608] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The National Cancer Institute Comparative Brain Tumor Consortium, Patient Outcomes Working Group, propose a consensus document in support of standardized magnetic resonance imaging protocols for canine brain tumor clinical trials. The intent of this manuscript is to address the widely acknowledged need to ensure canine brain tumor imaging protocols are relevant and have sufficient equivalency to translate to human studies such that: (1) multi-institutional studies can be performed with minimal inter-institutional variation, and (2) imaging protocols are consistent with human consensus recommendations to permit reliable translation of imaging data to human clinical trials. Consensus recommendations include pre- and postcontrast three-dimensional T1-weighted images, T2-weighted turbo spin echo in all three planes, T2*-weighted gradient recalled echo, T2-weighted fluid attenuated inversion recovery, and diffusion weighted imaging/diffusion tensor imaging in transverse plane; field of view of ≤150 mm; slice thickness of ≤2 mm, matrix ≥ 256 for two-dimensional images, and 150 or 256 for three-dimensional images.
Collapse
Affiliation(s)
- Rebecca A Packer
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523-1678
| | - John H Rossmeisl
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, 24061
| | - Michael S Kent
- Department of Surgical and Radiological Sciences, University of California Davis, School of Veterinary Medicine, Davis, CA, 95616
| | - John F Griffin
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843
| | - Christina Mazcko
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892
| | - Amy K LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892
| |
Collapse
|
38
|
Takada M, Parys M, Gregory-Bryson E, Vilar Saavedra P, Kiupel M, Yuzbasiyan-Gurkan V. A novel canine histiocytic sarcoma cell line: initial characterization and utilization for drug screening studies. BMC Cancer 2018; 18:237. [PMID: 29490634 PMCID: PMC5831740 DOI: 10.1186/s12885-018-4132-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 02/14/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Histiocytic sarcoma is a rare disorder in humans, however it is seen with appreciable frequency in certain breeds of dogs, such as Bernese mountain dog. The purpose of this study was to fully characterize a novel canine histiocytic sarcoma cell line, and utilize it as a tool to screen for potential therapeutic drugs. METHODS The histiocytic sarcoma cell line was characterized by expression of cellular markers as determined by immunohistochemistry and flow cytometry techniques. The neoplastic cells were also evaluated for their capability of phagocytizing beads particles, and their potential to grow as xenograft in an immunodeficient mouse. We investigated the in vitro cytotoxic activity of a panel of thirteen compounds using the MTS proliferation assay. Inhibitory effects of different drugs were compared using one-way ANOVA, and multiple means were compared using Tukey's test. RESULTS Neoplastic cells expressed CD11c, CD14, CD18, CD45, CD172a, CD204, MHC I, and vimentin. Expression of MHC II was upregulated after exposure to LPS. Furthermore, the established cell line clearly demonstrated phagocytic activity similar to positive controls of macrophage cell line. The xenograft mouse developed a palpable subcutaneous soft tissue mass after 29 days of inoculation, which histologically resembled the primary neoplasm. Dasatinib, a tyrosine kinase pan-inhibitor, significantly inhibited the growth of the cells in vitro within a clinically achievable and tolerable plasma concentration. The inhibitory response to dasatinib was augmented when combined with doxorubicin. CONCLUSIONS In the present study we demonstrated that a novel canine histiocytic sarcoma cell line presents a valuable tool to evaluate novel treatment approaches. The neoplastic cell line favorably responded to dasatinib, which represents a promising anticancer strategy for the treatment of this malignancy in dogs and similar disorders in humans.
Collapse
Affiliation(s)
- Marilia Takada
- Comparative Medicine and Integrative Biology, Michigan State University, East Lansing, MI 48824 USA
| | - Maciej Parys
- Comparative Medicine and Integrative Biology, Michigan State University, East Lansing, MI 48824 USA
- Present address: Royal (Dick) School of Veterinary Studies and the Roslin Institute, Roslin, Midlothian, UK
| | - Emmalena Gregory-Bryson
- Comparative Medicine and Integrative Biology, Michigan State University, East Lansing, MI 48824 USA
| | - Paulo Vilar Saavedra
- Small Animal Clinical Science, Michigan State University, East Lansing, 48824 MI USA
| | - Matti Kiupel
- Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, MI 48824 USA
| | - Vilma Yuzbasiyan-Gurkan
- Comparative Medicine and Integrative Biology, Michigan State University, East Lansing, MI 48824 USA
| |
Collapse
|
39
|
Dolera M, Malfassi L, Pavesi S, Marcarini S, Sala M, Mazza G, Carrara N, Finesso S, Urso G. Stereotactic Volume Modulated Arc Radiotherapy in Canine Meningiomas: Imaging-Based and Clinical Neurological Posttreatment Evaluation. J Am Anim Hosp Assoc 2018; 54:77-84. [PMID: 29372870 DOI: 10.5326/jaaha-ms-6488] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A prospective study to assess high-dose hypofractionated volume modulated arc radiotherapy feasibility and efficacy in canine meningiomas was conducted. Thirty-nine patients with encephalic and spinal meningiomas assumed from MRI findings were recruited and received high-dose hypofractionated volumetric modulated arc radiotherapy by a linear accelerator equipped with an external beam modulator micro-multileaf collimator and an XVI cone beam computed tomography system. The prescribed mean dose was 33 Gy in five fractions. The treatment feasibility was tested through planned and delivered dose agreement checks. Regular clinical examinations were performed during and after irradiation time, with regard to mentation, deambulation, cranial nerve dysfunction, and seizures. Serial MRI exams were done 60 days after irradiation and after 4, 6, 12, 18, and 24 mo. Volumetric disease reduction criteria implemented with clinical neurological systematic evaluation were adopted to assess the course and to categorize patients' responses. Complete and partial responses were observed on the whole in 65.5% of alive patients 24 mo after irradiation. Two-yr overall and disease-specific survival rates were 74.3% and 97.4%, respectively, and the putative radiotoxic effects were found to be few and slight.
Collapse
Affiliation(s)
- Mario Dolera
- From the La Cittadina Fondazione Studi e Ricerche Veterinarie, Romanengo, Italy (M.D., L.M., S.P., S.M., M.S., G.M., N.C., S.F.); and Azienda Ospedaliera della Provincia di Lodi, Lodi, Italy (G.U.)
| | - Luca Malfassi
- From the La Cittadina Fondazione Studi e Ricerche Veterinarie, Romanengo, Italy (M.D., L.M., S.P., S.M., M.S., G.M., N.C., S.F.); and Azienda Ospedaliera della Provincia di Lodi, Lodi, Italy (G.U.)
| | - Simone Pavesi
- From the La Cittadina Fondazione Studi e Ricerche Veterinarie, Romanengo, Italy (M.D., L.M., S.P., S.M., M.S., G.M., N.C., S.F.); and Azienda Ospedaliera della Provincia di Lodi, Lodi, Italy (G.U.)
| | - Silvia Marcarini
- From the La Cittadina Fondazione Studi e Ricerche Veterinarie, Romanengo, Italy (M.D., L.M., S.P., S.M., M.S., G.M., N.C., S.F.); and Azienda Ospedaliera della Provincia di Lodi, Lodi, Italy (G.U.)
| | - Massimo Sala
- From the La Cittadina Fondazione Studi e Ricerche Veterinarie, Romanengo, Italy (M.D., L.M., S.P., S.M., M.S., G.M., N.C., S.F.); and Azienda Ospedaliera della Provincia di Lodi, Lodi, Italy (G.U.)
| | - Giovanni Mazza
- From the La Cittadina Fondazione Studi e Ricerche Veterinarie, Romanengo, Italy (M.D., L.M., S.P., S.M., M.S., G.M., N.C., S.F.); and Azienda Ospedaliera della Provincia di Lodi, Lodi, Italy (G.U.)
| | - Nancy Carrara
- From the La Cittadina Fondazione Studi e Ricerche Veterinarie, Romanengo, Italy (M.D., L.M., S.P., S.M., M.S., G.M., N.C., S.F.); and Azienda Ospedaliera della Provincia di Lodi, Lodi, Italy (G.U.)
| | - Sara Finesso
- From the La Cittadina Fondazione Studi e Ricerche Veterinarie, Romanengo, Italy (M.D., L.M., S.P., S.M., M.S., G.M., N.C., S.F.); and Azienda Ospedaliera della Provincia di Lodi, Lodi, Italy (G.U.)
| | - Gaetano Urso
- From the La Cittadina Fondazione Studi e Ricerche Veterinarie, Romanengo, Italy (M.D., L.M., S.P., S.M., M.S., G.M., N.C., S.F.); and Azienda Ospedaliera della Provincia di Lodi, Lodi, Italy (G.U.)
| |
Collapse
|
40
|
Dolera M, Malfassi L, Carrara N, Finesso S, Marcarini S, Mazza G, Pavesi S, Sala M, Urso G. Volumetric Modulated Arc (Radio) Therapy in Pets Treatment: The "La Cittadina Fondazione" Experience. Cancers (Basel) 2018; 10:E30. [PMID: 29364837 PMCID: PMC5836062 DOI: 10.3390/cancers10020030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/29/2017] [Accepted: 01/16/2018] [Indexed: 12/31/2022] Open
Abstract
Volumetric Modulated Arc Therapy (VMAT) is a modern technique, widely used in human radiotherapy, which allows a high dose to be delivered to tumor volumes and low doses to the surrounding organs at risk (OAR). Veterinary clinics takes advantage of this feature due to the small target volumes and distances between the target and the OAR. Sparing the OAR permits dose escalation, and hypofractionation regimens reduce the number of treatment sessions with a simpler manageability in the veterinary field. Multimodal volumes definition is mandatory for the small volumes involved and a positioning device precisely reproducible with a setup confirmation is needed before each session for avoiding missing the target. Additionally, the elaborate treatment plan must pursue hard constraints and objectives, and its feasibility must be evaluated with a per patient quality control. The aim of this work is to report results with regard to brain meningiomas and gliomas, trigeminal nerve tumors, brachial plexus tumors, adrenal tumors with vascular invasion and rabbit thymomas, in comparison with literature to determine if VMAT is a safe and viable alternative to surgery or chemotherapy alone, or as an adjuvant therapy in pets.
Collapse
Affiliation(s)
- Mario Dolera
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy.
| | - Luca Malfassi
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy.
| | - Nancy Carrara
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy.
| | - Sara Finesso
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy.
| | - Silvia Marcarini
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy.
| | - Giovanni Mazza
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy.
| | - Simone Pavesi
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy.
| | - Massimo Sala
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy.
| | - Gaetano Urso
- Azienda Socio Sanitaria Territoriale della provincia di Lodi, 26841 Casalpusterlengo, Italy.
| |
Collapse
|
41
|
Kent MS, Turek MM, Farrelly J. Recent advances in veterinary radiation oncology. Vet Comp Oncol 2018; 16:167-169. [PMID: 29333737 DOI: 10.1111/vco.12366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 10/12/2017] [Indexed: 12/28/2022]
Affiliation(s)
- M S Kent
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, California
| | - M M Turek
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - J Farrelly
- Blue Pearl Veterinary Partners, New York, New York
| |
Collapse
|
42
|
Kent MS, Turek MM, Farrelly J. Recent advances in veterinary radiation oncology. Vet Radiol Ultrasound 2018; 59:10-12. [DOI: 10.1111/vru.12573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 10/04/2017] [Indexed: 11/30/2022] Open
Affiliation(s)
- Michael S. Kent
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine; University of California; Davis USA
| | - Michelle M. Turek
- Department of Surgical Sciences, School of Veterinary Medicine; University of Wisconsin-Madison; Madison USA
| | | |
Collapse
|
43
|
Rancilio NJ, Bentley RT, Plantenga JP, Parys MM, Crespo BG, Moore GE. Safety and feasibility of stereotactic radiotherapy using computed portal radiography for canine intracranial tumors. Vet Radiol Ultrasound 2017; 59:212-220. [PMID: 29205634 DOI: 10.1111/vru.12579] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 08/07/2017] [Accepted: 09/08/2017] [Indexed: 11/29/2022] Open
Abstract
Stereotactic radiotherapy is a highly conformal treatment option for intracranial and extracranial malignancies. Stereotactic radiotherapy utilizes specialized equipment specifically designed to avoid normal tissue while delivering ablative treatments with submillimeter precision and accuracy. Linear accelerator based stereotactic radiotherapy incorporates on-board image guidance utilizing cone beam computed tomography (CT). Many institutions lack the ability to provide image guidance with cone beam CT but delivery of highly conformal treatments with submillimeter precision and accuracy is still feasible. The purpose of this retrospective, pilot study was to describe clinical outcomes for a group of dogs with neurological disease that were treated with an stereotactic radiotherapy technique utilizing intensity modulated radiation therapy, megavoltage computed portal radiography, a bite plate, thermoplastic mold, and mask based positioning system. Twelve dogs with neurological clinical signs were included. The diagnosis of intracranial tumor was made based on advanced imaging (12/12) and confirmed via histopathology (3/12). Twelve courses of stereotactic radiotherapy, utilizing three fractions of 8.0 Gy, were delivered on alternating days. Self-resolving neurological deterioration was observed in two patients during stereotactic radiotherapy. Neurological progression free interval and median survival time were 273 days (range: 16-692 days) and 361 days (range: 25-862 days). Stereotactic radiotherapy using computed portal radiography may be a safe treatment option for dogs with intracranial tumors.
Collapse
Affiliation(s)
- Nicholas J Rancilio
- Departments of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - R Timothy Bentley
- Departments of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Jeannie Poulson Plantenga
- Departments of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Magdalena M Parys
- Departments of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Beatriz G Crespo
- Departments of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - George E Moore
- Veterinary Administration, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
44
|
Owen TJ, Martin LG, Chen AV. Transsphenoidal Surgery for Pituitary Tumors and Other Sellar Masses. Vet Clin North Am Small Anim Pract 2017; 48:129-151. [PMID: 29056398 DOI: 10.1016/j.cvsm.2017.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Transsphenoidal surgery is an option for dogs and cats with functional and nonfunctional pituitary masses or other sellar and parasellar masses. An adrenocorticotropic hormone-secreting tumor causing Cushing disease is the most common clinically relevant pituitary tumor in dogs, and the most common pituitary tumor seen in cats is a growth hormone-secreting tumor causing acromegaly. Transsphenoidal surgery can lead to rapid resolution of clinical signs and provide a cure for these patients. Because of the risks associated with this surgery, it should only be attempted by a cohesive pituitary surgery group with a sophisticated medical and surgical team.
Collapse
Affiliation(s)
- Tina J Owen
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, PO Box 647060, Pullman, WA 99164-7060, USA.
| | - Linda G Martin
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, PO Box 646610, Pullman, WA 99164-6610, USA
| | - Annie V Chen
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, PO Box 646610, Pullman, WA 99164-6610, USA
| |
Collapse
|
45
|
Bentley RT, Ahmed AU, Yanke AB, Cohen-Gadol AA, Dey M. Dogs are man's best friend: in sickness and in health. Neuro Oncol 2017; 19:312-322. [PMID: 27298310 DOI: 10.1093/neuonc/now109] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/08/2016] [Indexed: 01/17/2023] Open
Abstract
With the median survival of 14.6 months following best available standard of care, malignant gliomas (MGs) remain one of the biggest therapeutic challenges of the modern time. Although the last several decades have witnessed tremendous advancement in our understanding of MG and evolution of many successful preclinical therapeutic strategies, even the most successful preclinical therapeutic strategies often fail to cross the phase I/II clinical trial threshold. One of the significant, but less commonly discussed, barriers in developing effective glioma therapy is the lack of a robust preclinical model. For the last 30 years, rodent orthotopic xenograft models have been extensively used in the preclinical setting. Although they provide a good basic model for understanding tumor biology, their value in successfully translating preclinical therapeutic triumph into clinical success is extremely poor. Companion dogs, which share the same environmental stress as their human counterparts, also spontaneously develop MGs. Dog gliomas that develop spontaneously in an immunocompetent host are very similar to human gliomas and potentially provide a stronger platform for validating the efficacy of therapeutic strategies proven successful in preclinical mouse models. Integrating this model can accelerate development of effective therapeutic options that will benefit both human subjects and pet dogs.
Collapse
Affiliation(s)
- R Timothy Bentley
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, Indiana, USA
| | | | - Amy B Yanke
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, Indiana, USA
| | | | - Mahua Dey
- Department of Neurosurgery, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|
46
|
Swift KE, McGrath S, Nolan MW, Young M, Reese M, Rao S, Randall E, Leary D, LaRue S. Clinical and imaging findings, treatments, and outcomes in 27 dogs with imaging diagnosed trigeminal nerve sheath tumors: A multi-center study. Vet Radiol Ultrasound 2017; 58:679-689. [DOI: 10.1111/vru.12535] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 04/18/2017] [Accepted: 04/24/2017] [Indexed: 11/26/2022] Open
Affiliation(s)
- Katie E. Swift
- Department of Environmental and Radiological Health Sciences; Colorado State University; Fort Collins CO 80523
| | - Stephanie McGrath
- Department of Clinical Sciences; Colorado State University; Fort Collins CO 80523
| | - Michael W. Nolan
- Department of Clinical Sciences and Comparative Medicine Institute; North Carolina State University; Raleigh NC 27607
| | - Martin Young
- Bush Veterinary Neurology Service; Richmond VA 23230
| | - Michael Reese
- Department of Veterinary Clinical Sciences; Purdue University; West Lafayette IN 47907
| | - Sangeeta Rao
- Department of Clinical Sciences; Colorado State University; Fort Collins CO 80523
| | - Elissa Randall
- Department of Environmental and Radiological Health Sciences; Colorado State University; Fort Collins CO 80523
| | - Del Leary
- Department of Environmental and Radiological Health Sciences; Colorado State University; Fort Collins CO 80523
| | - Susan LaRue
- Department of Environmental and Radiological Health Sciences; Colorado State University; Fort Collins CO 80523
| |
Collapse
|
47
|
Dolera M, Malfassi L, Bianchi C, Carrara N, Finesso S, Marcarini S, Mazza G, Pavesi S, Sala M, Urso G. Frameless stereotactic radiotherapy alone and combined with temozolomide for presumed canine gliomas. Vet Comp Oncol 2017. [DOI: 10.1111/vco.12316] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- M. Dolera
- La Cittadina Fondazione Studi e Ricerche Veterinarie; Romanengo Italy
| | - L. Malfassi
- La Cittadina Fondazione Studi e Ricerche Veterinarie; Romanengo Italy
| | - C. Bianchi
- La Cittadina Fondazione Studi e Ricerche Veterinarie; Romanengo Italy
| | - N. Carrara
- La Cittadina Fondazione Studi e Ricerche Veterinarie; Romanengo Italy
| | - S. Finesso
- La Cittadina Fondazione Studi e Ricerche Veterinarie; Romanengo Italy
| | - S. Marcarini
- La Cittadina Fondazione Studi e Ricerche Veterinarie; Romanengo Italy
| | - G. Mazza
- La Cittadina Fondazione Studi e Ricerche Veterinarie; Romanengo Italy
| | - S. Pavesi
- La Cittadina Fondazione Studi e Ricerche Veterinarie; Romanengo Italy
| | - M. Sala
- La Cittadina Fondazione Studi e Ricerche Veterinarie; Romanengo Italy
| | - G. Urso
- La Cittadina Fondazione Studi e Ricerche Veterinarie; Romanengo Italy
- Azienda Socio Sanitaria Territoriale di Lodi; Lodi Italy
| |
Collapse
|
48
|
LeBlanc AK, Mazcko C, Brown DE, Koehler JW, Miller AD, Miller CR, Bentley RT, Packer RA, Breen M, Boudreau CE, Levine JM, Simpson RM, Halsey C, Kisseberth W, Rossmeisl JH, Dickinson PJ, Fan TM, Corps K, Aldape K, Puduvalli V, Pluhar GE, Gilbert MR. Creation of an NCI comparative brain tumor consortium: informing the translation of new knowledge from canine to human brain tumor patients. Neuro Oncol 2016; 18:1209-18. [PMID: 27179361 PMCID: PMC4999002 DOI: 10.1093/neuonc/now051] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/27/2016] [Indexed: 12/14/2022] Open
Abstract
On September 14-15, 2015, a meeting of clinicians and investigators in the fields of veterinary and human neuro-oncology, clinical trials, neuropathology, and drug development was convened at the National Institutes of Health campus in Bethesda, Maryland. This meeting served as the inaugural event launching a new consortium focused on improving the knowledge, development of, and access to naturally occurring canine brain cancer, specifically glioma, as a model for human disease. Within the meeting, a SWOT (strengths, weaknesses, opportunities, and threats) assessment was undertaken to critically evaluate the role that naturally occurring canine brain tumors could have in advancing this aspect of comparative oncology aimed at improving outcomes for dogs and human beings. A summary of this meeting and subsequent discussion are provided to inform the scientific and clinical community of the potential for this initiative. Canine and human comparisons represent an unprecedented opportunity to complement conventional brain tumor research paradigms, addressing a devastating disease for which innovative diagnostic and treatment strategies are clearly needed.
Collapse
Affiliation(s)
- Amy K LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Christina Mazcko
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Diane E Brown
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Jennifer W Koehler
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Andrew D Miller
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - C Ryan Miller
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - R Timothy Bentley
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Rebecca A Packer
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Matthew Breen
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - C Elizabeth Boudreau
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Jonathan M Levine
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - R Mark Simpson
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Charles Halsey
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - William Kisseberth
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - John H Rossmeisl
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Peter J Dickinson
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Timothy M Fan
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Kara Corps
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Kenneth Aldape
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Vinay Puduvalli
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - G Elizabeth Pluhar
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| | - Mark R Gilbert
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (A.K.L, C.M.); American Kennel Club Canine Health Foundation, Raleigh, North Carolina (D.E.B); Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (J.W.K); Department of Biomedical Sciences, Section of Anatomic Pathology, Cornell University College of Veterinary Medicine, Ithaca, New York (A.D.M); Departments of Pathology and Laboratory Medicine and Neurology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina (C.R.M); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana (R.T.B); Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado , (R.A.P); Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina (M.B.); Department of Small Animal Clinical Sciences, Texas A&M University, College Station, Texas (J.M.L, C.E.B); Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (R.M.S, C.H.); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio , (W.K.); Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia (J.H.R); Department of Surgery and Radiology, School of Veterinary Medicine, University of California, Davis, California (P.J.D); Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois (T.M.F); National Institute of Neurological Disorders and Stroke and National Cancer Institute, Bethesda, Maryland (K.C., M.R.G); De
| |
Collapse
|
49
|
Christensen NI, Forrest LJ, White PJ, Henzler M, Turek MM. SINGLE INSTITUTION VARIABILITY IN INTENSITY MODULATED RADIATION TARGET DELINEATION FOR CANINE NASAL NEOPLASIA. Vet Radiol Ultrasound 2016; 57:639-645. [PMID: 27465316 DOI: 10.1111/vru.12398] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 06/09/2016] [Accepted: 06/12/2016] [Indexed: 11/29/2022] Open
Abstract
Contouring variability is a significant barrier to the accurate delivery and reporting of radiation therapy. The aim of this descriptive study was to determine the variation in contouring radiation targets and organs at risk by participants within our institution. Further, we also aimed to determine if all individuals contoured the same normal tissues. Two canine nasal tumor datasets were selected and contoured by two ACVR-certified radiation oncologists and two radiation oncology residents from the same institution. Eight structures were consistently contoured including the right and left eye, the right and left lens, brain, the gross tumor volume (GTV), clinical target volume (CTV), and planning target volume (PTV). Spinal cord, hard and soft palate, and bulla were contoured on 50% of datasets. Variation in contouring occurred in both targets and normal tissues at risk and was particularly significant for the GTV, CTV, and PTV. The mean metric score and dice similarity coefficient were below the threshold criteria in 37.5-50% and 12.5-50% of structures, respectively, quantitatively indicating contouring variation. This study refutes our hypothesis that minimal variation in target and normal tissue delineation occurs. The variation in contouring may contribute to different tumor response and toxicity for any given patient. Our results also highlight the difficulty associated with replication of published radiation protocols or treatments, as even with replete contouring description the outcome of treatment is still fundamentally influenced by the individual contouring the patient.
Collapse
Affiliation(s)
- Neil I Christensen
- Department of Surgical Sciences, School of Veterinary, Medicine, University of Wisconsin - Madison, Madison, WI.
| | - Lisa J Forrest
- Department of Surgical Sciences, School of Veterinary, Medicine, University of Wisconsin - Madison, Madison, WI
| | | | - Margaret Henzler
- Department of Surgical Sciences, School of Veterinary, Medicine, University of Wisconsin - Madison, Madison, WI
| | - Michelle M Turek
- Department of Medical Sciences, School of Veterinary, Medicine, University of Wisconsin - Madison, Madison, WI
| |
Collapse
|
50
|
Awan MJ, Dorth J, Mani A, Kim H, Zheng Y, Mislmani M, Welford S, Yuan J, Wessels BW, Lo SS, Letterio J, Machtay M, Sloan A, Sohn JW. Development and Validation of a Small Animal Immobilizer and Positioning System for the Study of Delivery of Intracranial and Extracranial Radiotherapy Using the Gamma Knife System. Technol Cancer Res Treat 2016; 16:203-210. [PMID: 27444980 DOI: 10.1177/1533034616658394] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The purpose of this research is to establish a process of irradiating mice using the Gamma Knife as a versatile system for small animal irradiation and to validate accurate intracranial and extracranial dose delivery using this system. A stereotactic immobilization device was developed for small animals for the Gamma Knife head frame allowing for isocentric dose delivery. Intercranial positional reproducibility of a reference point from a primary reference animal was verified on an additional mouse. Extracranial positional reproducibility of the mouse aorta was verified using 3 mice. Accurate dose delivery was validated using film and thermoluminescent dosimeter measurements with a solid water phantom. Gamma Knife plans were developed to irradiate intracranial and extracranial targets. Mice were irradiated validating successful targeted radiation dose delivery. Intramouse positional variability of the right mandible reference point across 10 micro-computed tomography scans was 0.65 ± 0.48 mm. Intermouse positional reproducibility across 2 mice at the same reference point was 0.76 ± 0.46 mm. The accuracy of dose delivery was 0.67 ± 0.29 mm and 1.01 ± 0.43 mm in the coronal and sagittal planes, respectively. The planned dose delivered to a mouse phantom was 2 Gy at the 50% isodose with a measured thermoluminescent dosimeter dose of 2.9 ± 0.3 Gy. The phosphorylated form of member X of histone family H2A (γH2AX) staining of irradiated mouse brain and mouse aorta demonstrated adjacent tissue sparing. In conclusion, our system for preclinical studies of small animal irradiation using the Gamma Knife is able to accurately deliver intracranial and extracranial targeted focal radiation allowing for preclinical experiments studying focal radiation.
Collapse
Affiliation(s)
- Musaddiq J Awan
- 1 Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH, USA
| | - Jennifer Dorth
- 1 Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH, USA
| | - Arvind Mani
- 2 Department of Computer Science and Electrical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Haksoo Kim
- 1 Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH, USA
| | - Yiran Zheng
- 1 Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH, USA
| | - Mazen Mislmani
- 1 Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH, USA
| | - Scott Welford
- 1 Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH, USA
| | - Jiankui Yuan
- 1 Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH, USA
| | - Barry W Wessels
- 1 Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH, USA
| | - Simon S Lo
- 1 Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH, USA
| | - John Letterio
- 3 Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Mitchell Machtay
- 1 Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH, USA
| | - Andrew Sloan
- 4 Department of Neurosurgery, Case Western Reserve University, Cleveland, OH, USA
| | - Jason W Sohn
- 1 Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|