1
|
Bryan JN, Maitz CA. Translational History and Hope of Immunotherapy of Canine Tumors. Clin Cancer Res 2024; 30:4272-4285. [PMID: 39042399 PMCID: PMC11444889 DOI: 10.1158/1078-0432.ccr-23-2266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/13/2024] [Accepted: 07/01/2024] [Indexed: 07/24/2024]
Abstract
Companion dogs have served an important role in cancer immunotherapy research. Sharing similar environments and diets with humans, dogs naturally develop many of the same cancers. These shared exposures, coupled with dogs' diverse genetic makeup, make them ideal subjects for studying cancer therapies. Tumors like osteosarcoma, hemangiosarcoma, soft-tissue sarcoma, and non-Hodgkin lymphoma occur with greater frequency than their counterpart disease in humans. Canine brain tumors allow the study of therapy strategies with imaging, surgery, and radiotherapy equipment in veterinary patients with near-human geometry. Nonspecific immunostimulants, autologous and allogeneic vaccines, immune checkpoint inhibitors, and cellular therapies used in treating canine cancers have been tested in veterinary clinical trials. These treatments have not only improved outcomes for dogs but have also provided valuable insights for human cancer treatment. Advancements in radiation technology and the development of tools to characterize canine immune responses have further facilitated the ability to translate veterinary clinical trial results to human applications. Advancements in immunotherapy of canine tumors have directly supported translation to human clinical trials leading to approved therapies for patients with cancer around the world. The study of immunotherapy in dogs has been and will continue to be a promising avenue for advancing human cancer treatment.
Collapse
Affiliation(s)
- Jeffrey N. Bryan
- Comparative Oncology, Radiobiology, and Epigenetics Laboratory, Department of Veterinary Medicine and Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO
| | - Charles A. Maitz
- Comparative Oncology, Radiobiology, and Epigenetics Laboratory, Department of Veterinary Medicine and Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO
| |
Collapse
|
2
|
Molín J, José-López R, Ramírez GA, Pumarola M. Immunohistochemical Expression of PTEN in Canine Gliomas. Animals (Basel) 2024; 14:2115. [PMID: 39061577 PMCID: PMC11273977 DOI: 10.3390/ani14142115] [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: 06/23/2024] [Revised: 07/14/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Phosphatase and tensin homolog (PTEN) is a critical tumor suppressor gene with a vital role in regulating cell proliferation, migration, and survival. The loss of PTEN function, either by genetic alterations or decreased protein expression, is frequent in human gliomas and has been correlated with tumor progression, grade, therapeutic resistance, and decreased overall survival in patients with glioma. While different genetic mutations in PTEN gene have been occasionally reported in canine gliomas, no alterations in protein expression have been reported. This study investigates the immunohistochemical expression of PTEN in canine gliomas to evaluate possible alterations, as those reported in human gliomas. Immunohistochemical PTEN expression and pattern distribution were analyzed in 37 spontaneous canine gliomas. Among gliomas, 52.6% cases showed high PTEN expression and 48.6% displayed reduced (13.5%) or highly reduced (35.1%) immunopositivity. Most oligodendrogliomas showed high expression (73.7%), while the majority of astrocytomas (69.2%) showed a reduced or highly reduced expression. A reduced PTEN expression was mostly associated with a heterogeneous loss of PTEN immunopositivity. These observations are in line with those reported in human gliomas and provide a rationale for future studies regarding abnormalities in PTEN expression and PI3K/Akt/mTor pathway in canine gliomas, to evaluate its prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Jéssica Molín
- Departament Ciència Animal, Campus Agroalimentari, Forestal i Veterinari, Universitat de Lleida, 25198 Lleida, Spain;
| | - Roberto José-López
- Division of Small Animal Clinical Sciences, School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK;
- Neurology and Neurosurgery Service, Southfields Veterinary Specialists, Part of Linnaeus Veterinary Ltd., Basildon SS14 3AP, UK
| | - Gustavo A. Ramírez
- Departament Ciència Animal, Campus Agroalimentari, Forestal i Veterinari, Universitat de Lleida, 25198 Lleida, Spain;
| | - Martí Pumarola
- Unitat de Patologia Murina i Comparada, Departament de Medicina i Cirurgia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
| |
Collapse
|
3
|
Cartiaux B, Deviers A, Delmas C, Abadie J, Pumarola Battle M, Cohen-Jonathan Moyal E, Mogicato G. Evaluation of in vitro intrinsic radiosensitivity and characterization of five canine high-grade glioma cell lines. Front Vet Sci 2023; 10:1253074. [PMID: 38098992 PMCID: PMC10720585 DOI: 10.3389/fvets.2023.1253074] [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: 07/04/2023] [Accepted: 11/17/2023] [Indexed: 12/17/2023] Open
Abstract
Glioma is the most common primary brain tumor in dogs and predominantly affects brachycephalic breeds. Diagnosis relies on CT or MRI imaging, and the proposed treatments include surgical resection, chemotherapy, and radiotherapy depending on the tumor's location. Canine glioma from domestic dogs could be used as a more powerful model to study radiotherapy for human glioma than the murine model. Indeed, (i) contrary to mice, immunocompetent dogs develop spontaneous glioma, (ii) the canine brain structure is closer to human than mice, and (iii) domestic dogs are exposed to the same environmental factors than humans. Moreover, imaging techniques and radiation therapy used in human medicine can be applied to dogs, facilitating the direct transposition of results. The objective of this study is to fully characterize 5 canine glioma cell lines and to evaluate their intrinsic radiosensitivity. Canine cell lines present numerous analogies between the data obtained during this study on different glioma cell lines in dogs. Cell morphology is identical, such as doubling time, clonality test and karyotype. Immunohistochemical study of surface proteins, directly on cell lines and after stereotaxic injection in mice also reveals close similarity. Radiosensitivity profile of canine glial cells present high profile of radioresistance.
Collapse
Affiliation(s)
- Benjamin Cartiaux
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT), University Paul Sabatier Toulouse III, Toulouse, France
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, ENVT, Toulouse, France
| | - Alexandra Deviers
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, ENVT, Toulouse, France
| | - Caroline Delmas
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT), University Paul Sabatier Toulouse III, Toulouse, France
- IUCT-oncopole, Toulouse, France
| | - Jérôme Abadie
- Department of Biology, Pathology and Food Sciences, Laboniris, Nantes, France
| | - Martí Pumarola Battle
- Unit of Murine and Comparative Pathology, Department of Animal Medicine and Surgery, Veterinary Faculty, Autonomous University of Barcelona, Barcelona, Spain
| | - Elizabeth Cohen-Jonathan Moyal
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT), University Paul Sabatier Toulouse III, Toulouse, France
- IUCT-oncopole, Toulouse, France
| | - Giovanni Mogicato
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, ENVT, Toulouse, France
| |
Collapse
|
4
|
Wang J, Nan Y, Liu M, Hu K. The Role of CD4 + T Cells in the Immunotherapy of Brain Disease by Secreting Different Cytokines. J Neuroimmune Pharmacol 2022; 17:409-422. [PMID: 36443518 DOI: 10.1007/s11481-022-10056-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 11/17/2022] [Indexed: 11/30/2022]
Abstract
Upon different stimulation, naïve CD4+ T cells differentiate into various subsets of T helper (Th) cells, including Th1, Th2, Th17, and Tregs. They play both protective and pathogenic roles in the central nervous system (CNS) by secreting different cytokines. Failure of the homeostasis of the subgroups in the CNS can result in different brain diseases. Recently, immunotherapy has drawn more and more attention in the therapy of various brain diseases. Here, we describe the role of different CD4+ T cell subsets and their secreted cytokines in various brain diseases, as well as the ways in which by affecting CD4+ T cells in therapy of the CNS diseases. Understanding the role of CD4+ T cells and their secreted cytokines in the immunotherapy of brain disease will provide new targets and therapeutics for the treatment of brain disease. The role of CD4 + T cell subtypes in different diseases and their associated regulatory genes, proteins, and enzymes. CD4 + T cell subtypes play both protective (green) and pathogenic (red) roles in different brain diseases. The immune regulatory effects of CD4 + T cells and their subtypes are promoted or inhibited by different genes, proteins, and enzymes.
Collapse
Affiliation(s)
- Jing Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yunrong Nan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Mei Liu
- Industrial Development Center of Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Kaili Hu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
5
|
Boudreau CE, Najem H, Ott M, Horbinski C, Fang D, DeRay CM, Levine JM, Curran MA, Heimberger AB. Intratumoral Delivery of STING Agonist Results in Clinical Responses in Canine Glioblastoma. Clin Cancer Res 2021; 27:5528-5535. [PMID: 34433652 PMCID: PMC8989401 DOI: 10.1158/1078-0432.ccr-21-1914] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/19/2021] [Accepted: 08/16/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Activation of STING (stimulator of interferon genes) can trigger a robust, innate antitumor immune response in immunologically "cold" tumors such as glioblastoma. PATIENTS AND METHODS A small-molecule STING agonist, IACS-8779, was stereotactically administered using intraoperative navigation intratumorally in dogs with spontaneously arising glioblastoma. The phase I trial used an escalating dose design, ascending through four dose levels (5-20 μg). Treatment was repeated every 4-6 weeks for a minimum of two cycles. Radiographic response to treatment was determined by response assessment in neuro-oncology (RANO) criteria applied to isovoxel postcontrast T1-weighted MR images obtained on a single 3T magnet. RESULTS Six dogs were enrolled and completed ≥1 cycle of treatment. One dog was determined to have an abscess and was removed from further analysis. One procedure-related fatality was observed. Radiographic responses were dose dependent after the first cycle. The first subject had progressive disease, whereas there was 25% volumetric reduction in one subject and greater than 50% in the remaining surviving subjects. The median progression-free survival time was 14 weeks (range: 0-22 weeks), and the median overall survival time was 32 weeks (range: 11-39 weeks). CONCLUSIONS Intratumoral STING agonist (IACS-8779) administration was well tolerated in dogs with glioblastoma to a dose of 15 μg. Higher doses of IACS-8779 were associated with radiographic responses.
Collapse
Affiliation(s)
- C Elizabeth Boudreau
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas.
| | - Hinda Najem
- Department of Neurosurgery, Northwestern University, Chicago, Illinois
| | | | - Craig Horbinski
- Department of Neurosurgery, Northwestern University, Chicago, Illinois
- Department of Pathology, Northwestern University, Chicago, Illinois
| | - Dexing Fang
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chase M DeRay
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Jonathan M Levine
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Michael A Curran
- Department of Melanoma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amy B Heimberger
- Department of Neurosurgery, Northwestern University, Chicago, Illinois.
| |
Collapse
|
6
|
Krane GA, O'Dea CA, Malarkey DE, Miller AD, Miller CR, Tokarz DA, Jensen HL, Janardhan KS, Shockley KR, Flagler N, Rainess BA, Mariani CL. Immunohistochemical evaluation of immune cell infiltration in canine gliomas. Vet Pathol 2021; 58:952-963. [PMID: 34196247 PMCID: PMC11404454 DOI: 10.1177/03009858211023946] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Evasion of the immune response is an integral part of the pathogenesis of glioma. In humans, important mechanisms of immune evasion include recruitment of regulatory T cells (Tregs) and polarization of macrophages toward an M2 phenotype. Canine glioma has a robust immune cell infiltrate that has not been extensively characterized. The purpose of this study was to determine the distribution of immune cells infiltrating spontaneous intracranial canine gliomas. Seventy-three formalin-fixed, paraffin-embedded tumor samples were evaluated using immunohistochemistry for CD3, forkhead box 3 (FOXP3), CD20, Iba1, calprotectin (Mac387), CD163, and indoleamine 2,3-dioxygenase (IDO). Immune cell infiltration was present in all tumors. Low-grade and high-grade gliomas significantly differed in the numbers of FoxP3+ cells, Mac387+ cells, and CD163+ cells (P = .006, .01, and .01, respectively). Considering all tumors, there was a significant increase in tumor area fraction of CD163 compared to Mac387 (P < .0001), and this ratio was greater in high-grade tumors than in low-grade tumors (P = .005). These data warrant further exploration into the roles of macrophage repolarization or Treg interference therapy in canine glioma.
Collapse
Affiliation(s)
- Gregory A Krane
- 6857National Institute of Environmental Health Sciences, National Toxicology Program, Cellular and Molecular Pathology Branch, Research Triangle Park, NC, USA
- Department of Clinical Sciences, North Carolina State University, Raleigh, NC, USA
- Comparative Neuroimmunology and Neuro-Oncology Laboratory, North Carolina State University, Raleigh, NC, USA
| | | | - David E Malarkey
- 6857National Institute of Environmental Health Sciences, National Toxicology Program, Cellular and Molecular Pathology Branch, Research Triangle Park, NC, USA
| | | | | | - Debra A Tokarz
- Experimental Pathology Laboratories Inc, Research Triangle Park, NC, USA
| | - Heather L Jensen
- 6857National Institute of Environmental Health Sciences, National Toxicology Program, Cellular and Molecular Pathology Branch, Research Triangle Park, NC, USA
| | | | - Keith R Shockley
- Division of Intramural Research, Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Norris Flagler
- 6857National Institute of Environmental Health Sciences, National Toxicology Program, Cellular and Molecular Pathology Branch, Research Triangle Park, NC, USA
| | - Brittani A Rainess
- Comparative Neuroimmunology and Neuro-Oncology Laboratory, North Carolina State University, Raleigh, NC, USA
| | - Christopher L Mariani
- Comparative Neuroimmunology and Neuro-Oncology Laboratory, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
7
|
Rohrer Bley C, Staudinger C, Bley T, Marconato L, Sabattini S, Beckmann K. Canine presumed glial brain tumours treated with radiotherapy: Is there an inferior outcome in tumours contacting the subventricular zone? Vet Comp Oncol 2021; 20:29-37. [PMID: 33900018 DOI: 10.1111/vco.12703] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/21/2021] [Accepted: 04/21/2021] [Indexed: 12/21/2022]
Abstract
Post-treatment outcome in canine glial tumours is described with a broad range of survival times between 2 and 28 months. After surgery or radiation therapy, the tumours may progress locally or spread within the central nervous system. It is unknown if tumour- or patient-specific factors influence prognosis. In humans, glioblastoma involving the subventricular zone has been found to recur distantly, with shortened time to progression and overall survival. We included 32 dogs irradiated for a presumptive primary glial brain tumour in this retrospective cohort study. Tumours were grouped relative to subventricular zone contact and overt ventricular invasion assessing pre-treatment magnetic resonance images. Median time to progression (TTP) for all cases was 534 days (95%CI, 310-758), with a significantly shorter TTP in dogs with lesions at the subventricular zone (median TTP, 260 vs. 687 days; p = .049). Tumours at the subventricular zone progressed more often (p = .001), and more likely as CNS-metastasis (52.9% vs. 13.3%, p = .028). Median overall survival (OS) was 489 days (95%CI, 147-831) and median tumour-specific survival 609 days (95%CI, 382-835). Involvement of the subventricular zone was significantly associated with a shorter tumour-specific survival (median, 306 vs. 719 days; p = .044). Glial tumours contacting the subventricular zone in dogs have a shorter tumour-specific survival and a higher rate of progression and CNS-metastasis. Despite local tumour control, metastasis must be considered and should prompt further treatment approaches.
Collapse
Affiliation(s)
- Carla Rohrer Bley
- Division of Radiation Oncology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Chris Staudinger
- Division of Radiation Oncology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Tim Bley
- Neurology Service, Small Animal Clinic Aarau West, Oberentfelden, Switzerland
| | - Laura Marconato
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Silvia Sabattini
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Katrin Beckmann
- Neurology Service, Clinic of Small Animal Surgery, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
8
|
Omar NB, Bentley RT, Crossman DK, Foote JB, Koehler JW, Markert JM, Platt SR, Rissi DR, Shores A, Sorjonen D, Yanke AB, Gillespie GY, Chambers MR. Safety and interim survival data after intracranial administration of M032, a genetically engineered oncolytic HSV-1 expressing IL-12, in pet dogs with sporadic gliomas. Neurosurg Focus 2021; 50:E5. [PMID: 33524948 DOI: 10.3171/2020.11.focus20844] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/12/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The diagnosis of glioma remains disheartening in the clinical realm. While a multitude of studies and trials have shown promise, improvements in overall survival have been disappointing. Modeling these tumors in the laboratory setting has become increasingly challenging, given their complex in situ behavior and interactions for therapeutic evasion. Dogs, particularly brachycephalic breeds, are known to spontaneously develop gliomas that resemble human gliomas both clinically and pathophysiologically, making canines with sporadic tumors promising candidates for study. Typically, survival among these dogs is approximately 2 months with palliation alone. METHODS The authors have completed the first stage of a unique phase I dose-escalating canine clinical trial in which the safety and tolerability of M032, a nonneurovirulent oncolytic herpes simplex virus-1 vector genetically engineered to express interleukin-12, are being studied in pet dogs with gliomas undergoing maximum safe tumor resection and inoculation of the cavity with the viral infusate. RESULTS Twenty-five canine patients were enrolled between January 2018 and August 2020. One patient was electively withdrawn from the trial by its owner, and 3 did not receive the virus. For the 21 dogs that remained, 13 had high-grade gliomas, 5 had low-grade gliomas, and 3 were undetermined. According to histopathological analysis, 62% of the tumors were oligodendrogliomas. At the time of this report, the median overall survival from the date of treatment was 151 days (± 78 days). No significant adverse events attributable to M032 or dose-limiting toxicities have been observed to date. CONCLUSIONS In this largest study of oncolytic viral therapy for canine brain tumors to date, treatment with M032 did not cause harm and the combination of surgery and oncolytic viral therapy may have contributed to prolonged survival in pet dogs with spontaneous gliomas. Forthcoming in-depth radiographic, immunohistochemical, and genetic analyses will afford a more advanced understanding of how this treatment impacts these tumors and the immune system. Our goal is to utilize these findings bitranslationally to inform human studies and refine therapies that will improve outcomes in both humans and pet dogs with gliomas.
Collapse
Affiliation(s)
| | - R Timothy Bentley
- 4Purdue University College of Veterinary Medicine, West Lafayette, Indiana
| | | | - Jeremy B Foote
- 3Microbiology, The University of Alabama at Birmingham, Alabama
| | | | | | - Simon R Platt
- 5University of Georgia College of Veterinary Medicine, Athens, Georgia
| | - Daniel R Rissi
- 5University of Georgia College of Veterinary Medicine, Athens, Georgia
| | - Andy Shores
- 6Mississippi State University College of Veterinary Medicine, Mississippi State, Mississippi; and
| | - Donald Sorjonen
- 7Auburn University College of Veterinary Medicine, Auburn, Alabama
| | - Amy B Yanke
- 7Auburn University College of Veterinary Medicine, Auburn, Alabama
| | | | | |
Collapse
|
9
|
Chambers MR, Foote JB, Bentley RT, Botta D, Crossman DK, Della Manna DL, Estevez-Ordonez D, Koehler JW, Langford CP, Miller MA, Markert JM, Olivier AK, Omar NB, Platt SR, Rissi DR, Shores A, Sorjonen DC, Yang ES, Yanke AB, Gillespie GY. Evaluation of immunologic parameters in canine glioma patients treated with an oncolytic herpes virus. JOURNAL OF TRANSLATIONAL GENETICS AND GENOMICS 2021; 5:423-442. [PMID: 35342877 PMCID: PMC8955901 DOI: 10.20517/jtgg.2021.31] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
AIM To molecularly characterize the tumor microenvironment and evaluate immunologic parameters in canine glioma patients before and after treatment with oncolytic human IL-12-expressing herpes simplex virus (M032) and in treatment naïve canine gliomas. METHODS We assessed pet dogs with sporadically occurring gliomas enrolled in Stage 1 of a veterinary clinical trial that was designed to establish the safety of intratumoral oncoviral therapy with M032, a genetically modified oncolytic herpes simplex virus. Specimens from dogs in the trial and dogs not enrolled in the trial were evaluated with immunohistochemistry, NanoString, Luminex cytokine profiling, and multi-parameter flow cytometry. RESULTS Treatment-naive canine glioma microenvironment had enrichment of Iba1 positive macrophages and minimal numbers of T and B cells, consistent with previous studies identifying these tumors as immunologically "cold". NanoString mRNA profiling revealed enrichment for tumor intrinsic pathways consistent with suppression of tumor-specific immunity and support of tumor progression. Oncolytic viral treatment induced an intratumoral mRNA transcription signature of tumor-specific immune responses in 83% (5/6) of canine glioma patients. Changes included mRNA signatures corresponding with interferon signaling, lymphoid and myeloid cell activation, recruitment, and T and B cell immunity. Multiplexed protein analysis identified a subset of oligodendroglioma subjects with increased concentrations of IL-2, IL-7, IL-6, IL-10, IL-15, TNFα, GM-CSF between 14 and 28 days after treatment, with evidence of CD4+ T cell activation and modulation of IL-4 and IFNγ production in CD4+ and CD8+ T cells isolated from peripheral blood. CONCLUSION These findings indicate that M032 modulates the tumor-immune microenvironment in the canine glioma model.
Collapse
Affiliation(s)
- M R Chambers
- Department of Neurosurgery, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - J B Foote
- Department of Microbiology, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - R T Bentley
- Department of Neurosurgery, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| | - D Botta
- Department of Microbiology, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - D K Crossman
- Department of Genetics, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - D L Della Manna
- Department of Radiation Oncology, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - D Estevez-Ordonez
- Department of Neurosurgery, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - J W Koehler
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - C P Langford
- Department of Neurosurgery, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - M A Miller
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| | - J M Markert
- Department of Neurosurgery, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - A K Olivier
- Department of Pathology, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA
| | - N B Omar
- Department of Neurosurgery, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - S R Platt
- Department of Neurosurgery, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - D R Rissi
- Athens Veterinary Diagnostic Laboratory, Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - A Shores
- Department of Neurology & Neurosurgery, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA
| | - D C Sorjonen
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - E S Yang
- Department of Radiation Oncology, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| | - A B Yanke
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - G Y Gillespie
- Department of Neurosurgery, University of Alabama at Birmingham (UAB), Birmingham, AL 35294, USA
| |
Collapse
|
10
|
Abstract
Comparative oncology clinical trials play an important and growing role in cancer research and drug development efforts. These trials, typically conducted in companion (pet) dogs, allow assessment of novel anticancer agents and combination therapies in a veterinary clinical setting that supports serial biologic sample collections and exploration of dose, schedule and corresponding pharmacokinetic/pharmacodynamic relationships. Further, an intact immune system and natural co-evolution of tumour and microenvironment support exploration of novel immunotherapeutic strategies. Substantial improvements in our collective understanding of the molecular landscape of canine cancers have occurred in the past 10 years, facilitating translational research and supporting the inclusion of comparative studies in drug development. The value of the approach is demonstrated in various clinical trial settings, including single-agent or combination response rates, inhibition of metastatic progression and randomized comparison of multiple agents in a head-to-head fashion. Such comparative oncology studies have been purposefully included in the developmental plan for several US FDA-approved and up-and-coming anticancer drugs. Challenges for this field include keeping pace with technology and data dissemination/harmonization, improving annotation of the canine genome and immune system, and generation of canine-specific validated reagents to support integration of correlative biology within clinical trial efforts.
Collapse
Affiliation(s)
- Amy K LeBlanc
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Christina N Mazcko
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
11
|
Pi Castro D, José-López R, Fernández Flores F, Rabanal Prados RM, Mandara MT, Arús C, Pumarola Batlle M. Expression of FOXP3 in Canine Gliomas: Immunohistochemical Study of Tumor-Infiltrating Regulatory Lymphocytes. J Neuropathol Exp Neurol 2020; 79:184-193. [PMID: 31846038 DOI: 10.1093/jnen/nlz120] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 10/15/2019] [Accepted: 11/06/2019] [Indexed: 12/19/2022] Open
Abstract
Dogs develop gliomas with similar histopathological features to human gliomas and share with them the limited success of current therapeutic regimens such as surgery and radiation. The tumor microenvironment in gliomas is influenced by immune cell infiltrates. The present study aims to immunohistochemically characterize the tumor-infiltrating lymphocyte (TIL) population of naturally occurring canine gliomas, focusing on the expression of Forkhead box P3-positive (FOXP3+) regulatory T-cells (Tregs). Forty-three canine gliomas were evaluated immunohistochemically for the presence of CD3+, FOXP3+, and CD20+ TILs. In low-grade gliomas, CD3+ TILs were found exclusively within the tumor tissue. In high-grade gliomas, they were present in significantly higher numbers throughout the tumor and in the brain-tumor junction. CD20+ TILs were rarely found in comparison to CD3+ TILs. FOXP3+ TILs shared a similar distribution with CD3+ TILs. The accumulation of FOXP3+ Tregs within the tumor was more pronounced in astrocytic gliomas than in tumors of oligodendroglial lineage and the difference in expression was significant when comparing low-grade oligodendrogliomas and high-grade astrocytomas. Only high-grade astrocytomas presented FOXP3+ cells with tumoral morphology. In spontaneous canine gliomas, TILs display similar characteristics (density and distribution) as described for human gliomas, supporting the use of the dog as an animal model for translational immunotherapeutic studies.
Collapse
Affiliation(s)
- Dolors Pi Castro
- From the Unit of Murine and Comparative Pathology (UPMiC), Department of Animal Medicine and Surgery, Veterinary Faculty, Universitat Autónoma de Barcelona, Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Roberto José-López
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Francisco Fernández Flores
- Department of Veterinary Pathology and Public Health, Institute of Veterinary Science, University of Liverpool, UK
| | - Rosa M Rabanal Prados
- From the Unit of Murine and Comparative Pathology (UPMiC), Department of Animal Medicine and Surgery, Veterinary Faculty, Universitat Autónoma de Barcelona, Barcelona, Spain
| | | | - Carles Arús
- From the Unit of Murine and Comparative Pathology (UPMiC), Department of Animal Medicine and Surgery, Veterinary Faculty, Universitat Autónoma de Barcelona, Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Universitat Autònoma de Barcelona, Barcelona, Spain.,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain.,Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Martí Pumarola Batlle
- From the Unit of Murine and Comparative Pathology (UPMiC), Department of Animal Medicine and Surgery, Veterinary Faculty, Universitat Autónoma de Barcelona, Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
12
|
Kim JH. Interleukin-8 in the Tumor Immune Niche: Lessons from Comparative Oncology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1240:25-33. [PMID: 32060885 DOI: 10.1007/978-3-030-38315-2_2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Interleukin (IL)-8 is a chemokine that is essential for inflammation and angiogenesis. IL-8 expression is elevated in tumor cell lines and tissues, as well as in peripheral blood obtained from cancer patients. Primary works have attempted to determine the biological effect of IL-8 on tumor cells, including cell proliferation, survival, and migration. More recently, IL-8 has acquired considerable attention as an immune modulator in the context of certain tumor microenvironments (TME); specifically, it can support a niche that favors tumor progression and metastasis. Tumor-derived IL-8 stimulates inflammation by interacting with the microenvironmental constituents, including fibroblasts, endothelial cells, and immune cells. However, the tumor immune system is complex, and mechanisms that construct the immune phenotype remain incompletely characterized. Herein, we will (1) address a potential role of IL-8 in regulating gene expression to establish immune landscape in tumor. Then, we will (2) review IL-8 signaling in the maintenance of stem cells and regulation of hematopoietic progenitors. Finally, (3) IL-8 functions will be discussed in naturally occurring animal cancers that offer a clinically realistic model for translational research. This chapter will provide a new insight into the tumor immune niche and help us develop immunotherapies for cancers.
Collapse
Affiliation(s)
- Jong-Hyuk Kim
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, MN, USA. .,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, MN, USA. .,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
13
|
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: 16] [Impact Index Per Article: 3.2] [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
|
14
|
Knapp DW, Dhawan D, Ramos-Vara JA, Ratliff TL, Cresswell GM, Utturkar S, Sommer BC, Fulkerson CM, Hahn NM. Naturally-Occurring Invasive Urothelial Carcinoma in Dogs, a Unique Model to Drive Advances in Managing Muscle Invasive Bladder Cancer in Humans. Front Oncol 2020; 9:1493. [PMID: 32039002 PMCID: PMC6985458 DOI: 10.3389/fonc.2019.01493] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 12/11/2019] [Indexed: 12/11/2022] Open
Abstract
There is a great need to improve the outlook for people facing urinary bladder cancer, especially for patients with invasive urothelial carcinoma (InvUC) which is lethal in 50% of cases. Improved outcomes for patients with InvUC could come from advances on several fronts including emerging immunotherapies, targeted therapies, and new drug combinations; selection of patients most likely to respond to a given treatment based on molecular subtypes, immune signatures, and other characteristics; and prevention, early detection, and early intervention. Progress on all of these fronts will require clinically relevant animal models for translational research. The animal model(s) should possess key features that drive success or failure of cancer drugs in humans including tumor heterogeneity, genetic-epigenetic crosstalk, immune cell responsiveness, invasive and metastatic behavior, and molecular subtypes (e.g., luminal, basal). Experimental animal models, while essential in bladder cancer research, do not possess these collective features to accurately predict outcomes in humans. These key features, however, are present in naturally-occurring InvUC in pet dogs. Canine InvUC closely mimics muscle-invasive bladder cancer in humans in cellular and molecular features, molecular subtypes, immune response patterns, biological behavior (sites and frequency of metastasis), and response to therapy. Thus, dogs can offer a highly relevant animal model to complement other models in research for new therapies for bladder cancer. Clinical treatment trials in pet dogs with InvUC are considered a win-win-win scenario; the individual dog benefits from effective treatment, the results are expected to help other dogs, and the findings are expected to translate to better treatment outcomes in humans. In addition, the high breed-associated risk for InvUC in dogs (e.g., 20-fold increased risk in Scottish Terriers) offers an unparalleled opportunity to test new strategies in primary prevention, early detection, and early intervention. This review will provide an overview of canine InvUC, summarize the similarities (and differences) between canine and human InvUC, and provide evidence for the expanding value of this canine model in bladder cancer research.
Collapse
Affiliation(s)
- Deborah W Knapp
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN, United States.,Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Deepika Dhawan
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN, United States
| | - José A Ramos-Vara
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, United States.,Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, United States
| | - Timothy L Ratliff
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, United States.,Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, United States
| | - Gregory M Cresswell
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Sagar Utturkar
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Breann C Sommer
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN, United States
| | - Christopher M Fulkerson
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN, United States.,Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Noah M Hahn
- Department of Oncology and Urology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
15
|
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: 64] [Impact Index Per Article: 10.7] [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
|
16
|
Langsten KL, Kim JH, Sarver AL, Dewhirst M, Modiano JF. Comparative Approach to the Temporo-Spatial Organization of the Tumor Microenvironment. Front Oncol 2019; 9:1185. [PMID: 31788448 PMCID: PMC6854022 DOI: 10.3389/fonc.2019.01185] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 10/21/2019] [Indexed: 12/13/2022] Open
Abstract
The complex ecosystem in which tumor cells reside and interact, termed the tumor microenvironment (TME), encompasses all cells and components associated with a neoplasm that are not transformed cells. Interactions between tumor cells and the TME are complex and fluid, with each facet coercing the other, largely, into promoting tumor progression. While the TME in humans is relatively well-described, a compilation and comparison of the TME in our canine counterparts has not yet been described. As is the case in humans, dog tumors exhibit greater heterogeneity than what is appreciated in laboratory animal models, although the current level of knowledge on similarities and differences in the TME between dogs and humans, and the practical implications of that information, require further investigation. This review summarizes some of the complexities of the human and mouse TME and interjects with what is known in the dog, relaying the information in the context of the temporo-spatial organization of the TME. To the authors' knowledge, the development of the TME over space and time has not been widely discussed, and a comprehensive review of the canine TME has not been done. The specific topics covered in this review include cellular invasion and interactions within the TME, metabolic derangements in the TME and vascular invasion, and the involvement of the TME in tumor spread and metastasis.
Collapse
Affiliation(s)
- Kendall L Langsten
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN, United States
| | - Jong Hyuk Kim
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN, United States.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Aaron L Sarver
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN, United States.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States.,Institute for Health Informatics, University of Minnesota, Minneapolis, MN, United States
| | - Mark Dewhirst
- Radiation Oncology Department, Duke University Medical School, Durham, NC, United States
| | - Jaime F Modiano
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN, United States.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States.,Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, United States.,Center for Immunology, University of Minnesota, Minneapolis, MN, United States.,Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States.,Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
17
|
Koehler JW, Miller AD, Miller CR, Porter B, Aldape K, Beck J, Brat D, Cornax I, Corps K, Frank C, Giannini C, Horbinski C, Huse JT, O'Sullivan MG, Rissi DR, Mark Simpson R, Woolard K, Shih JH, Mazcko C, Gilbert MR, LeBlanc AK. A Revised Diagnostic Classification of Canine Glioma: Towards Validation of the Canine Glioma Patient as a Naturally Occurring Preclinical Model for Human Glioma. J Neuropathol Exp Neurol 2019; 77:1039-1054. [PMID: 30239918 DOI: 10.1093/jnen/nly085] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The National Cancer Institute-led multidisciplinary Comparative Brain Tumor Consortium (CBTC) convened a glioma pathology board, comprising both veterinarian and physician neuropathologists, and conducted a comprehensive review of 193 cases of canine glioma. The immediate goal was to improve existing glioma classification methods through creation of a histologic atlas of features, thus yielding greater harmonization of phenotypic characterization. The long-term goal was to support future incorporation of clinical outcomes and genomic data into proposed simplified diagnostic schema, so as to further bridge the worlds of veterinary and physician neuropathology and strengthen validity of the dog as a naturally occurring, translationally relevant animal model of human glioma. All cases were morphologically reclassified according to a new schema devised by the entire board, yielding a majority opinion diagnosis of astrocytoma (43, 22.3%), 19 of which were low-grade and 24 high-grade, and oligodendroglioma (134, 69.4%), 35 of which were low-grade and 99 were high-grade. Sixteen cases (8.3%) could not be classified as oligodendroglioma or astrocytoma based on morphology alone and were designated as undefined gliomas. The simplified classification scheme proposed herein provides a tractable means for future addition of molecular data, and also serves to highlight histologic similarities and differences between human and canine glioma.
Collapse
Affiliation(s)
- Jennifer W Koehler
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Andrew D Miller
- Department of Biomedical Sciences, Section of Anatomic Pathology, College of Veterinary Medicine, Cornell University, Ithaca, New York
| | - C Ryan Miller
- Department of Pathology and Laboratory Medicine.,Department of Neurology.,Department of Pharmacology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Brian Porter
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jessica Beck
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Daniel Brat
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Ingrid Cornax
- Department of Pediatrics, University of California-San Diego, San Diego California
| | - Kara Corps
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Viral Immunology and Intravital Imaging Section, Bethesda, Maryland
| | - Chad Frank
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, Colorado
| | - Caterina Giannini
- Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Craig Horbinski
- Department of Pathology.,Department of Neurosurgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jason T Huse
- Departments of Pathology and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - M Gerard O'Sullivan
- Masonic Cancer Center Comparative Pathology Shared Resource and Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
| | - Daniel R Rissi
- Department of Pathology and Athens Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Georgia, Athens, Georgia
| | - R Mark Simpson
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Molecular Pathology Unit, Laboratory of Cancer Biology and Genetics, Bethesda, Maryland
| | - Kevin Woolard
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California-Davis, Davis, California
| | - Joanna H Shih
- Biometrics Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Christina Mazcko
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mark R Gilbert
- National Institute of Neurological Disorders and Stroke and the Center for Cancer Research, National Cancer Institute, National Institutes of Health, NeuroOncology Branch, Bethesda, Maryland
| | - Amy K LeBlanc
- National Cancer Institute, National Institutes of Health, Comparative Oncology Program, Center for Cancer Research, Bethesda, Maryland
| |
Collapse
|
18
|
Mitchell D, Chintala S, Fetcko K, Henriquez M, Tewari BN, Ahmed A, Bentley RT, Dey M. Common Molecular Alterations in Canine Oligodendroglioma and Human Malignant Gliomas and Potential Novel Therapeutic Targets. Front Oncol 2019; 9:780. [PMID: 31475119 PMCID: PMC6702544 DOI: 10.3389/fonc.2019.00780] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/31/2019] [Indexed: 01/05/2023] Open
Abstract
Spontaneous canine (Canis lupus) oligodendroglioma (ODG) holds tremendous potential as an immunocompetent large animal model of human malignant gliomas (MG). However, the feasibility of utilizing this model in pre-clinical studies depends on a thorough understanding of the similarities and differences of the molecular pathways associated with gliomas between the two species. We have previously shown that canine ODG has an immune landscape and expression pattern of commonly described oncogenes similar to that of human MG. In the current study, we performed a comprehensive analysis of canine ODG RNAseq data from 4 dogs with ODG and 2 normal controls to identify highly dysregulated genes in canine tumors. We then evaluated the expression of these genes in human MG using Xena Browser, a publicly available database. STRING-database inquiry was used in order to determine the suggested protein associations of these differentially expressed genes as well as the dysregulated pathways commonly enriched by the protein products of these genes in both canine ODG and human MG. Our results revealed that 3,712 (23%) of the 15,895 differentially expressed genes demonstrated significant up- or downregulation (log2-fold change > 2.0). Of the 3,712 altered genes, ~50% were upregulated (n = 1858) and ~50% were downregulated (n = 1854). Most of these genes were also found to have altered expression in human MG. Protein association and pathway analysis revealed common pathways enriched by members of the up- and downregulated gene categories in both species. In summary, we demonstrate that a similar pattern of gene dysregulation characterizes both human MG and canine ODG and provide additional support for the use of the canine model in order to therapeutically target these common genes. The results of such therapeutic targeting in the canine model can serve to more accurately predict the efficacy of anti-glioma therapies in human patients.
Collapse
Affiliation(s)
- Dana Mitchell
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sreenivasulu Chintala
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Kaleigh Fetcko
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Mario Henriquez
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Brij N Tewari
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Atique Ahmed
- Department of Neurological Surgery, Northwestern University, Chicago, IL, United States
| | - R Timothy Bentley
- Department of Veterinary Clinical Sciences, Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Mahua Dey
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
19
|
Arami H, Patel CB, Madsen SJ, Dickinson PJ, Davis RM, Zeng Y, Sturges BK, Woolard KD, Habte FG, Akin D, Sinclair R, Gambhir SS. Nanomedicine for Spontaneous Brain Tumors: A Companion Clinical Trial. ACS NANO 2019; 13:2858-2869. [PMID: 30714717 PMCID: PMC6584029 DOI: 10.1021/acsnano.8b04406] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Nanoparticles' enhanced permeation and retention (EPR) variations due to tumor heterogeneity in naturally occurring brain tumors are commonly neglected in preclinical nanomedicine studies. Recent pathological studies have shown striking similarities between brain tumors in humans and dogs, indicating that canine brain tumors may be a valuable model to evaluate nanoparticles' EPR in this context. We recruited canine clinical cases with spontaneous brain tumors to investigate nanoparticles' EPR in different brain tumor pathologies using surface-enhanced Raman spectroscopy (SERS). We used gold nanoparticles due to their surface plasmon effect that enables their sensitive and microscopic resolution detection using the SERS technique. Raman microscopy of the resected tumors showed heterogeneous EPR of nanoparticles into oligodendrogliomas and meningiomas of different grades, without any detectable traces in necrotic parts of the tumors or normal brain. Raman observations were confirmed by scanning electron microscopy (SEM) and X-ray elemental analyses, which enabled localization of individual nanoparticles embedded in tumor tissues. Our results demonstrate nanoparticles' EPR and its variations in clinically relevant, spontaneous brain tumors. Such heterogeneities should be considered alongside routine preoperative imaging and histopathological analyses in order to accelerate clinical management of brain tumors using nanomedicine approaches.
Collapse
Affiliation(s)
- Hamed Arami
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, California 94305, United States
| | - Chirag B. Patel
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, California 94305, United States
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94304, United States
| | - Steven J. Madsen
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Peter J. Dickinson
- Department of Surgical and Radiological Sciences, University of California at Davis, Davis, California 95616, United States
| | - Ryan M. Davis
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, California 94305, United States
| | - Yitian Zeng
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Beverly K. Sturges
- Department of Surgical and Radiological Sciences, University of California at Davis, Davis, California 95616, United States
| | - Kevin D. Woolard
- Department of Pathology, Microbiology and Immunology, University of California, Davis, California 95616, United States
| | - Frezghi G. Habte
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, California 94305, United States
| | - Demir Akin
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, California 94305, United States
| | - Robert Sinclair
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Sanjiv S. Gambhir
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, California 94305, United States
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
- Department of Bioengineering, Stanford University, Stanford, California 94305, United States
- Stanford Neuroscience Institute, Stanford University School of Medicine, Stanford, California 94305, United States
- Corresponding Author (Sanjiv S. Gambhir).
| |
Collapse
|
20
|
Tagawa M, Kurashima C, Takagi S, Maekawa N, Konnai S, Shimbo G, Matsumoto K, Inokuma H, Kawamoto K, Miyahara K. Evaluation of costimulatory molecules in dogs with B cell high grade lymphoma. PLoS One 2018; 13:e0201222. [PMID: 30040869 PMCID: PMC6057677 DOI: 10.1371/journal.pone.0201222] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/11/2018] [Indexed: 12/21/2022] Open
Abstract
B cell high grade lymphoma is the most common hematopoietic malignancy in dogs. Although the immune checkpoint molecules, programmed death-1 (PD-1) and cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), and immune checkpoint inhibitors have been evaluated for the treatment of various human lymphoid malignancies, the expression of those molecules and their relationship with prognosis remain unknown in canine lymphoma. The objective of this study was to evaluate the expression of costimulatory molecules on peripheral blood lymphocytes and tumor infiltrating lymphocytes, in addition to associated ligand expression in the lymph nodes of patients with B cell multicentric high grade lymphoma. Eighteen patients diagnosed with B cell high grade lymphoma and nine healthy control dogs were enrolled. Flow cytometric analysis revealed that the expression of PD-1 on CD4+ peripheral and tumor infiltrating lymphocytes and CTLA-4 on CD4+ peripheral lymphocytes was significantly higher in the lymphoma group than in the control group. The expression level of CD80 mRNA was significantly lower in the lymphoma group than in the control group. In contrast, there were no significant differences in PD-L1, PD-L2, and CD86 expression between the groups. Dogs with CTLA-4 levels below the cutoff values, which were determined based on receiver operating characteristic curves, on peripheral CD4+, CD8+, and tumor infiltrating CD4+ lymphocytes had significantly longer survival than dogs with values above the cutoff. Although it is uncertain whether the expression of immune checkpoint molecules affect the biological behavior of canine lymphoma, one possible explanation is that PD-1 and CTLA-4 might be associated with the suppression of antitumor immunity in dogs with B cell high grade lymphoma, particularly through CD4+ T cells.
Collapse
Affiliation(s)
- Michihito Tagawa
- Veterinary Medical Center, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
- * E-mail:
| | - Chihiro Kurashima
- Veterinary Medical Center, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Satoshi Takagi
- Department of Veterinary Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Veterinary Teaching Hospital, Sapporo, Hokkaido, Japan
| | - Naoya Maekawa
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Satoru Konnai
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Genya Shimbo
- Veterinary Medical Center, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Kotaro Matsumoto
- Department of Clinical Veterinary Science, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Hisashi Inokuma
- Department of Clinical Veterinary Science, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Keiko Kawamoto
- Research Center for Global Agromedicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Kazuro Miyahara
- Veterinary Medical Center, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| |
Collapse
|
21
|
Bentley RT, Thomovsky SA, Miller MA, Knapp DW, Cohen-Gadol AA. Canine (Pet Dog) Tumor Microsurgery and Intratumoral Concentration and Safety of Metronomic Chlorambucil for Spontaneous Glioma: A Phase I Clinical Trial. World Neurosurg 2018; 116:e534-e542. [PMID: 29775768 DOI: 10.1016/j.wneu.2018.05.027] [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: 03/16/2018] [Revised: 05/03/2018] [Accepted: 05/04/2018] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Metronomic (daily low-dose) chlorambucil requires further study before use in human patients with glioma. The aim of this study was to investigate distribution and safety of metronomic chlorambucil in naturally occurring canine glioma. METHODS Eight client-owned (pet) dogs with newly diagnosed spontaneous glioma were prospectively enrolled. Chlorambucil was administered preoperatively at 4 mg/m2 every 24 hours for ≥3 days and continued postoperatively until death or dose-limiting adverse events. Chlorambucil concentrations in the surgical glioma specimen, cerebrospinal fluid, and serum were analyzed. Dogs additionally received lomustine postoperatively. Dogs were monitored for seizures, myoclonus, cytopenias, and tumor recurrence. RESULTS Complete microsurgical resection was achieved in 7 oligodendrogliomas and 1 astrocytoma (6 high grade, 2 low grade). Median surgical glioma specimen chlorambucil concentration was 0.52 ng/g (range, 0-2.62 ng/g), or 37% (range, 0%-178%) of serum concentration. Median cerebrospinal fluid concentration was 0.1 ng/mL (range, 0-0.3 ng/mL). Chlorambucil was not associated with increase in seizure activity. Six dogs displayed prolonged seizure-free intervals. There was no myoclonus. Three dogs developed asymptomatic thrombocytopenia after 8-12 months of chlorambucil. Median progression-free survival was 253 days (range, 63-860 days). Median overall survival was 257 days (range, 64-860 days). CONCLUSIONS The presence of intratumoral chlorambucil indicated an altered blood-brain barrier that varied from case to case. Despite sporadic previous reports of neurotoxicity, prolonged seizure-free intervals supported a high safety margin at this dose in this species. Metronomic chlorambucil was well tolerated. Spontaneous canine glioma offers a robust preclinical model.
Collapse
Affiliation(s)
- R Timothy Bentley
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, Indiana, USA; Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana, USA
| | - Stephanie A Thomovsky
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Margaret A Miller
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA
| | - Deborah W Knapp
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, Indiana, USA; Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana, USA
| | - Aaron A Cohen-Gadol
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, Indiana, USA; Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| |
Collapse
|