1
|
Giantin M, Montanucci L, Lopparelli RM, Tolosi R, Dentini A, Grieco V, Stefanello D, Sabattini S, Marconato L, Pauletto M, Dacasto M. Expression Profile of Twelve Transcripts as a Supporting Tool for the Molecular Characterization of Canine Cutaneous Mast Cell Tumors at Diagnosis: Association with Histological Grading and Clinical Staging. Genes (Basel) 2025; 16:340. [PMID: 40149490 PMCID: PMC11942052 DOI: 10.3390/genes16030340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 02/27/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Mast cell tumors (MCTs) are the second most common malignant neoplasms in dogs. Histopathological grading and clinical staging are the main tools for estimating biological behavior and disease extent; thus, both are essential for therapeutic decision-making and prognostication. However, the biological behavior of MCTs in dogs is variable, and it sometimes deviates from expectations. In a previous study, we identified 12 transcripts whose expression profile allowed a clear distinction between Kiupel low-grade and high-grade cutaneous MCTs (cMCTs) and was associated with prognosis. Building on these findings, this study evaluated the predictive potential of these transcripts' expression profiles in classifying cMCTs into low-grade and high-grade. METHODS A logistic regression classifier based on the expression profiles of the identified transcripts and able to classify cMCTs as low- or high-grade was developed and subsequently tested on a novel dataset of 50 cMCTs whose expression profiles have been determined in this study through qPCR. RESULTS The developed logistic regression classifier reaches an accuracy of 67% and an area under the receiver operating characteristic curve (AUC) of 0.76. Interestingly, the molecular classification clearly identifies stage-IV disease (90% true positive rate). CONCLUSIONS qPCR analysis of these biomarkers combined with the machine learning-based classifier might serve as a tool to support cMCT clinical management at diagnosis.
Collapse
Affiliation(s)
- Mery Giantin
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, I-35020 Legnaro, PD, Italy; (R.M.L.); (R.T.); (M.P.); (M.D.)
| | - Ludovica Montanucci
- Department of Neurology, Mc Govern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 44106, USA;
| | - Rosa Maria Lopparelli
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, I-35020 Legnaro, PD, Italy; (R.M.L.); (R.T.); (M.P.); (M.D.)
| | - Roberta Tolosi
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, I-35020 Legnaro, PD, Italy; (R.M.L.); (R.T.); (M.P.); (M.D.)
| | - Alfredo Dentini
- Clinica Veterinaria Tyrus, Via Aldo Bartocci 1G, I-05100 Terni, TR, Italy;
| | - Valeria Grieco
- Department of Veterinary Medicine and Animal Science, University of Milan, Via dell’Università 6, I-26900 Lodi, MI, Italy; (V.G.); (D.S.)
| | - Damiano Stefanello
- Department of Veterinary Medicine and Animal Science, University of Milan, Via dell’Università 6, I-26900 Lodi, MI, Italy; (V.G.); (D.S.)
| | - Silvia Sabattini
- Department of Veterinary Medical Sciences, Alma Mater Studiorum, University of Bologna, Via Tolara di Sopra 50, I-40064 Ozzano dell’Emilia, BO, Italy; (S.S.); (L.M.)
| | - Laura Marconato
- Department of Veterinary Medical Sciences, Alma Mater Studiorum, University of Bologna, Via Tolara di Sopra 50, I-40064 Ozzano dell’Emilia, BO, Italy; (S.S.); (L.M.)
| | - Marianna Pauletto
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, I-35020 Legnaro, PD, Italy; (R.M.L.); (R.T.); (M.P.); (M.D.)
| | - Mauro Dacasto
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, I-35020 Legnaro, PD, Italy; (R.M.L.); (R.T.); (M.P.); (M.D.)
| |
Collapse
|
2
|
Lee D, Kozurek EC, Abdullah M, Wong EJ, Li R, Liu ZS, Nguyen HD, Dickerson EB, Kim JH. PIK3CA mutation fortifies molecular determinants for immune signaling in vascular cancers. Cancer Gene Ther 2025; 32:254-267. [PMID: 39709507 PMCID: PMC11839470 DOI: 10.1038/s41417-024-00867-4] [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: 08/28/2024] [Revised: 11/23/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024]
Abstract
Angiosarcomas are a group of vascular cancers that form malignant blood vessels. These malignancies are seemingly inflamed primarily due to their pathognomonic nature, which consists of irregular endothelium and tortuous blood channels. PIK3CA mutations are oncogenic and disrupt the PI3K pathway. In this study, we aimed to define the molecular and functional consequences of oncogenic PIK3CA mutations in angiosarcoma. We first generated two isogenic hemangiosarcoma cell lines harboring the H1047R hotspot mutations in PIK3CA gene using CRISPR/Cas9. We found PIK3CA-mutant cells established distinct molecular signatures in global gene expression and chromatin accessibility, which were associated with enrichment of immune cytokine signaling, including IL-6, IL-8, and MCP-1. These molecular processes were disrupted by the PI3K-α specific inhibitor, alpelisib. We also observed that the molecular distinctions in PIK3CA-mutant cells were linked to metabolic reprogramming in glycolytic activity and mitochondrial respiration. Our multi-omics analysis revealed that activating PIK3CA mutations regulate molecular machinery that contributes to phenotypic alterations and resistance to alpelisib. Furthermore, we identified potential therapeutic vulnerabilities of PIK3CA mutations in response to PI3K-α inhibition mediated by MAPK signaling. In summary, we demonstrate that PIK3CA mutations perpetuate PI3K activation and reinforce immune enrichment to promote drug resistance in vascular cancers.
Collapse
Affiliation(s)
- Donghee Lee
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Emma C Kozurek
- 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
| | - Md Abdullah
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Ethan J Wong
- 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
| | - Rong Li
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Zhiyan Silvia Liu
- Department of Pharmacology, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Hai Dang Nguyen
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Department of Pharmacology, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Erin B Dickerson
- 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
| | - Jong Hyuk Kim
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA.
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA.
- Artificial Intelligence Academic Initiative (AI2) Center, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
3
|
Furukawa T, Shiotsuki A, Okada Y, Nibe K, Tei M, Anazawa T, Yoshikawa M, Ono K, Hirao H. Prognostic value of tumour-related factors associated with canine retroperitoneal hemangiosarcoma in comparison with other anatomic presentations: A retrospective observational study. Vet Med Sci 2024; 10:e1495. [PMID: 38889089 PMCID: PMC11185220 DOI: 10.1002/vms3.1495] [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: 04/22/2023] [Revised: 04/23/2024] [Accepted: 05/17/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Dogs with retroperitoneal hemangiosarcoma (HSA) exhibit variable postoperative median survival times (MST). OBJECTIVE To retrospectively evaluate the prognostic value of selected tumour-related factors, such as tumour size, rupture, invasion into adjacent tissue, involvement of lymph node and distant metastasis, they were analysed in dogs with retroperitoneal HSA. METHODS Ten dogs with retroperitoneal HSA managed solely with surgical excision were reviewed and compared with spleen (71) and liver (9) HSA. The Kaplan-Meier method and log-rank analysis were used compare MSTs between factors. Multivariable Cox proportional-hazard analysis was used to compare differences between arising sites. RESULTS Retroperitoneal HSA showed comparatively longer postoperative MST compared with that of spleen and liver HSA and demonstrated significantly longer MST (p = 0.003) for tumours ≥5 cm (195 days) than <5 cm (70 days). Spleen HSA revealed significantly shorter MSTs in involvement of distant lymph nodes (23 days) and distant metastasis (39 days) than those in negative (83 days, p = 0.002 and 110 days, p < 0.001, respectively). Liver HSA also revealed significantly shorter MST (16.5 days compared with 98 days, p = 0.003) for distant metastasis. Additionally, hazard ratios (HRs) and their forest plot for overall HSA revealed as poor prognostic factors, arising sites (spleen; HR 2.78, p = 0.016 and liver; HR 3.62, p = 0.019), involvement of distant lymph nodes (HR 2.43, p = 0.014), and distant metastasis (HR 2.86, p < 0.001), and as better prognostic factor of tumour size ≥5 cm (HR 0.53, p = 0.037). CONCLUSION In combination with overall HSA, retroperitoneal HSA shows comparatively longer postoperative MST compared to spleen and liver HSA, associated with tumour size ≥5 cm suggesting better prognostic factor.
Collapse
Affiliation(s)
| | - Akiko Shiotsuki
- Japan Animal Referral Medical Center (Nagoya)Nagoya‐shiAichiJapan
| | - Yusami Okada
- Japan Animal Referral Medical Center (Nagoya)Nagoya‐shiAichiJapan
| | - Kazumi Nibe
- Japan Animal Referral Medical CenterKawasaki‐shiKanagawaJapan
| | - Meina Tei
- Japan Animal Referral Medical CenterKawasaki‐shiKanagawaJapan
| | - Tetsuya Anazawa
- Japan Animal Referral Medical Center (Nagoya)Nagoya‐shiAichiJapan
| | | | - Kenichiro Ono
- Japan Animal Referral Medical CenterKawasaki‐shiKanagawaJapan
| | - Hidehiro Hirao
- Japan Animal Referral Medical CenterKawasaki‐shiKanagawaJapan
| |
Collapse
|
4
|
Kim JH, Schulte AJ, Sarver AL, Lee D, Angelos MG, Frantz AM, Forster CL, O'Brien TD, Cornax I, O'Sullivan MG, Cheng N, Lewellen M, Oseth L, Kumar S, Bullman S, Pedamallu CS, Goyal SM, Meyerson M, Lund TC, Breen M, Lindblad-Toh K, Dickerson EB, Kaufman DS, Modiano JF. Hemangiosarcoma Cells Promote Conserved Host-derived Hematopoietic Expansion. CANCER RESEARCH COMMUNICATIONS 2024; 4:1467-1480. [PMID: 38757809 PMCID: PMC11166094 DOI: 10.1158/2767-9764.crc-23-0441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/29/2024] [Accepted: 05/10/2024] [Indexed: 05/18/2024]
Abstract
Hemangiosarcoma and angiosarcoma are soft-tissue sarcomas of blood vessel-forming cells in dogs and humans, respectively. These vasoformative sarcomas are aggressive and highly metastatic, with disorganized, irregular blood-filled vascular spaces. Our objective was to define molecular programs which support the niche that enables progression of canine hemangiosarcoma and human angiosarcoma. Dog-in-mouse hemangiosarcoma xenografts recapitulated the vasoformative and highly angiogenic morphology and molecular characteristics of primary tumors. Blood vessels in the tumors were complex and disorganized, and they were lined by both donor and host cells. In a series of xenografts, we observed that the transplanted hemangiosarcoma cells created exuberant myeloid hyperplasia and gave rise to lymphoproliferative tumors of mouse origin. Our functional analyses indicate that hemangiosarcoma cells generate a microenvironment that supports expansion and differentiation of hematopoietic progenitor populations. Furthermore, gene expression profiling data revealed hemangiosarcoma cells expressed a repertoire of hematopoietic cytokines capable of regulating the surrounding stromal cells. We conclude that canine hemangiosarcomas, and possibly human angiosarcomas, maintain molecular properties that provide hematopoietic support and facilitate stromal reactions, suggesting their potential involvement in promoting the growth of hematopoietic tumors. SIGNIFICANCE We demonstrate that hemangiosarcomas regulate molecular programs supporting hematopoietic expansion and differentiation, providing insights into their potential roles in creating a permissive stromal-immune environment for tumor progression.
Collapse
Affiliation(s)
- Jong Hyuk Kim
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
- University of Florida Health Cancer Center, University of Florida, Gainesville, Florida
- Intelligent Critical Care Center, University of Florida, Gainesville, Florida
- Artificial Intelligence Academic Initiative (AI) Center, University of Florida, Gainesville, Florida
| | - Ashley J. Schulte
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Aaron L. Sarver
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Institute for Health Informatics, University of Minnesota, Minneapolis, Minnesota
| | - Donghee Lee
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Mathew G. Angelos
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
- Department of Medicine (Division of Hematology, Oncology, and Transplantation), Medical School, University of Minnesota, Minneapolis, Minnesota
- Microbiology, Immunology and Cancer Biology (MICaB) Graduate Program, University of Minnesota, Minneapolis, Minnesota
- Department of Medicine, Division of Hematology and Oncology, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Aric M. Frantz
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Capstan Therapeutics, San Diego, California
| | - Colleen L. Forster
- The University of Minnesota Biological Materials Procurement Network (BioNet), University of Minnesota, Minneapolis, Minnesota
| | - Timothy D. O'Brien
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
| | - Ingrid Cornax
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Janssen Research and Development, LLC
| | - M. Gerard O'Sullivan
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
| | - Nuojin Cheng
- School of Mathematics, College of Science and Engineering, University of Minnesota, Minneapolis, Minnesota
- Applied Mathematics, University of Colorado Boulder, Boulder, Colorado
| | - Mitzi Lewellen
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - LeAnn Oseth
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Sunil Kumar
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
| | - Susan Bullman
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Chandra Sekhar Pedamallu
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Sagar M. Goyal
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
| | - Matthew Meyerson
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Troy C. Lund
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Matthew Breen
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
- Cancer Genetics Program, University of North Carolina Lineberger Comprehensive Cancer Center, Raleigh, North Carolina
| | - Kerstin Lindblad-Toh
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Science of Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Erin B. Dickerson
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Dan S. Kaufman
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
- Department of Medicine (Division of Hematology, Oncology, and Transplantation), Medical School, University of Minnesota, Minneapolis, Minnesota
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota
- Division of Regenerative Medicine, Department of Medicine, University of California-San Diego, La Jolla, California
| | - Jaime F. Modiano
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, Minnesota
- Center for Engineering in Medicine, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
5
|
Suzuki T, Henshaw MJ, Yanagi T, Aoshima K. Current understanding of comparative pathology and prospective research approaches for canine hemangiosarcoma. Res Vet Sci 2024; 167:105120. [PMID: 38150941 DOI: 10.1016/j.rvsc.2023.105120] [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: 06/19/2023] [Revised: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 12/29/2023]
Abstract
Hemangiosarcoma (HSA) is a malignant tumor originating from endothelial cells. HSA typically develops in dogs, but is rare in other animals, including humans. Although surgery and chemotherapy are conventional treatments for HSA, neither treatment can significantly improve patient prognosis. To develop novel and effective therapeutics, a deeper understanding of HSA pathogenesis must be acquired. However, the limited research tools for HSA have been unable to make a breakthrough; therefore, it is crucial to widely utilize or establish novel research tools such as patient-derived xenograft models, organoids, and chicken embryo xenograft models. The pathogenesis of the human counterpart of HSA, angiosarcoma (AS), also remains incompletely understood, preventing the extrapolation of findings from humans to dogs, unlike other diseases. In this review, we summarize the clinicopathological and morphological features of HSA, and then we discuss the current understanding of the molecular pathology of HSA. Finally, we highlight promising research tools that may accelerate HSA basic research toward developing novel therapeutics. We also briefly summarize AS to help researchers comprehend HSA from the perspective of comparative pathology.
Collapse
Affiliation(s)
- Tamami Suzuki
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - Michael James Henshaw
- English Education Section, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - Teruki Yanagi
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido 060-8638, Japan
| | - Keisuke Aoshima
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan; Cancer Research Unit, One Health Research Center, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan.
| |
Collapse
|
6
|
Kerboeuf M, Haugeberg DA, Olsen T, Sørling LK, Koppang EO, Moe L, Haaland AH. Tumor-associated macrophages in canine visceral hemangiosarcoma. Vet Pathol 2024; 61:32-45. [PMID: 37341055 PMCID: PMC10687809 DOI: 10.1177/03009858231179947] [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] [Indexed: 06/22/2023]
Abstract
Canine hemangiosarcoma (HSA) is a highly malignant tumor derived from hematopoietic stem cells and commonly occurs in visceral organs or skin. Visceral HSAs are particularly aggressive and progress rapidly despite multimodal treatment. Tumor-associated macrophages (TAMs) play a central role in carcinogenesis, tumor progression, and metastasis in humans and murine models. In this retrospective study, we investigated the prevalence and phenotype of TAMs in privately owned, treatment-naïve dogs with naturally occurring HSA. We used CD204 as a general macrophage marker and CD206 as a marker for M2-polarized macrophages. Formalin-fixed paraffin-embedded tissues from HSAs in the spleen (n = 9), heart (n = 6), and other locations (n = 12) from 17 dogs were sectioned and immunohistochemically labeled with CD204 and CD206 antibodies. The mean number of log(CD204)- and log(CD206)-positive cells and the ratio of log(CD206/CD204)-positive cells were compared with normal surrounding tissues and between tumor locations. There were significantly more macrophages and M2 macrophages, and a higher ratio of M2 macrophages to total macrophages in tumor hot spots (P = .0002, P < .0001, and P = .0002, respectively) and in tumor tissues outside of hot spots (P = .009, P = .002, and P = .007, respectively) than in normal surrounding tissues. There were no significant differences between tumor locations, but there was a trend toward higher numbers of CD204-positive macrophages within the splenic tumors. There was no association between histological parameters or clinical stage and TAM numbers or phenotype. As in humans, TAMs in dogs with HSA have a predominantly M2-skewed phenotype. Dogs with HSA could serve as excellent models to evaluate new TAM-reprogramming therapies.
Collapse
Affiliation(s)
| | | | - Tobias Olsen
- Norwegian University of Life Sciences, Ås, Norway
| | | | | | - Lars Moe
- Norwegian University of Life Sciences, Ås, Norway
| | | |
Collapse
|
7
|
Heishima K, Aketa N, Heishima M, Kawachi A. Hemangiosarcoma in dogs as a potential non-rodent animal model for drug discovery research of angiosarcoma in humans. Front Oncol 2023; 13:1250766. [PMID: 38130992 PMCID: PMC10733437 DOI: 10.3389/fonc.2023.1250766] [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: 06/30/2023] [Accepted: 11/06/2023] [Indexed: 12/23/2023] Open
Abstract
Since the domestication of dogs 10,000 years ago, they have shared their living environment with humans and have co-evolved. The breeding process that dogs have undergone in only a few centuries has led to a significant accumulation of specific genetic alterations that could induce particular diseases in certain breeds. These canine diseases are similar to what is found in humans with several differences; therefore, comparing such diseases occurring in humans and dogs can help discover novel disease mechanisms, pathways, and causal genetic factors. Human angiosarcoma (AS) and canine hemangiosarcoma (HSA), which are sarcomas originating from endothelium, are examples of diseases shared between humans and dogs. They exhibit similar characteristics and clinical behaviors, although with some critical differences resulting from evolution. In this review, we will describe the similarities and differences in terms of clinical and molecular characteristics between human AS and canine HSA, and discuss how these similarities and differences can be applied to advance the treatment of these diseases.
Collapse
Affiliation(s)
- Kazuki Heishima
- Institute for Advanced Study (GUiAS), Gifu University, Gifu, Japan
- Center for One Medicine Innovative Translational Research (COMIT), Gifu University, Gifu, Japan
| | - Naohiko Aketa
- Clinical and Translational Research Center, Keio University Hospital, Tokyo, Japan
| | | | - Asuka Kawachi
- Division of Cancer RNA Research, National Cancer Center, Tokyo, Japan
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
8
|
Kapturska KM, Pawlak A. New molecular targets in canine hemangiosarcoma-Comparative review and future of the precision medicine. Vet Comp Oncol 2023; 21:357-377. [PMID: 37308243 DOI: 10.1111/vco.12917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 05/10/2023] [Accepted: 05/30/2023] [Indexed: 06/14/2023]
Abstract
Human angiosarcoma and canine hemangiosarcoma reveal similarities not only in their aggressive clinical behaviour, but especially in molecular landscape and genetic alterations involved in tumorigenesis and metastasis formation. Currently, no satisfying treatment that allows for achieving long overall survival or even prolonged time to progression does not exist. Due to the progress that has been made in targeted therapies and precision medicine the basis for a new treatment design is to uncover mutations and their functions as possible targets to provide tailored drugs for individual cases. Whole exome or genome sequencing studies and immunohistochemistry brought in the last few years important discoveries and identified the most common mutations with probably crucial role in this tumour development. Also, despite a lack of mutation in some of the culprit genes, the cancerogenesis cause may be buried in main cellular pathways connected with proteins encoded by those genes and involving, for example, pathological angiogenesis. The aim of this review is to highlight the most promising molecular targets for precision oncology treatment from the veterinary perspective aided by the principles of comparative science. Some of the drugs are only undergoing laboratory in vitro studies and others entered the clinic in the management of other cancer types in humans, but those used in dogs with promising responses have been mentioned as priorities.
Collapse
Affiliation(s)
- Karolina Małgorzata Kapturska
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
- Veterinary Clinic NEOVET s.c. Hildebrand, Jelonek, Michalek-Salt, Wroclaw, Poland
| | - Aleksandra Pawlak
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| |
Collapse
|
9
|
Pimentel PAB, Giuliano A, Bęczkowski PM, Horta RDS. Molecular Profile of Canine Hemangiosarcoma and Potential Novel Therapeutic Targets. Vet Sci 2023; 10:387. [PMID: 37368773 DOI: 10.3390/vetsci10060387] [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: 05/14/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Canine hemangiosarcoma (HSA) is a relatively common neoplasia, occurring mainly in the skin, spleen, liver and right atrium. Despite the numerous studies investigating the treatment of canine HSA, no significant improvement in survival has been achieved in the last 20 years. Advancements in genetic and molecular profiling presented molecular similarities between canine HSA and human angiosarcoma. It could therefore serve as a valuable model for investigating new and more effective treatments in people and dogs. The most common genetic abnormalities in canine HSA have been found in the phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) and neuroblastoma RAS viral oncogene homolog (NRAS) pathways. Mutations are also found in tumor protein p53 (TP53), phosphatase and tensin homolog (PTEN) and cyclin dependent kinase inhibitor 2A (CDKN2A). Known abnormal protein expression could be exploited to trial new target treatments that could be beneficial for both canine and human patients. Despite the high expression of vascular endothelial growth factor (VEGF) and its receptor (VEGFR), no correlation with overall survival time has ever been found. In this review, we explore the most recent developments in molecular profiling in canine HSA and discuss their possible applications in the prognosis and treatment of this fatal disease.
Collapse
Affiliation(s)
| | - Antonio Giuliano
- Department of Veterinary Clinical Science, Jockey Club College of Veterinary Medicine, City University of Hong Kong, Hong Kong, China
- Veterinary Medical Centre, City University of Hong Kong, Hong Kong, China
| | - Paweł Marek Bęczkowski
- Department of Veterinary Clinical Science, Jockey Club College of Veterinary Medicine, City University of Hong Kong, Hong Kong, China
| | - Rodrigo Dos Santos Horta
- Department of Veterinary Clinic and Surgery, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| |
Collapse
|
10
|
Klosowski M, Haines L, Alfino L, McMellen A, Leibowitz M, Regan D. Naturally occurring canine sarcomas: Bridging the gap from mouse models to human patients through cross-disciplinary research partnerships. Front Oncol 2023; 13:1130215. [PMID: 37035209 PMCID: PMC10076632 DOI: 10.3389/fonc.2023.1130215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/20/2023] [Indexed: 04/11/2023] Open
Abstract
Fueled by support from the National Cancer Institute's "Cancer Moonshot" program, the past few years have witnessed a renewed interest in the canine spontaneous cancer model as an invaluable resource in translational oncology research. Increasingly, there is awareness that pet dogs with cancer provide an accessible bridge to improving the efficiency of cancer drug discovery and clinical therapeutic development. Canine tumors share many biological, genetic, and histologic features with their human tumor counterparts, and most importantly, retain the complexities of naturally occurring drug resistance, metastasis, and tumor-host immune interactions, all of which are difficult to recapitulate in induced or genetically engineered murine tumor models. The utility of canine models has been particularly apparent in sarcoma research, where the increased incidence of sarcomas in dogs as compared to people has facilitated comparative research resulting in treatment advances benefitting both species. Although there is an increasing awareness of the advantages in using spontaneous canine sarcoma models for research, these models remain underutilized, in part due to a lack of more permanent institutional and cross-institutional infrastructure to support partnerships between veterinary and human clinician-scientists. In this review, we provide an updated overview of historical and current applications of spontaneously occurring canine tumor models in sarcoma research, with particular attention to knowledge gaps, limitations, and growth opportunities within these applications. Furthermore, we propose considerations for working within existing veterinary translational and comparative oncology research infrastructures to maximize the benefit of partnerships between veterinary and human biomedical researchers within and across institutions to improve the utility and application of spontaneous canine sarcomas in translational oncology research.
Collapse
Affiliation(s)
- Marika Klosowski
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Laurel Haines
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Lauren Alfino
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Alexandra McMellen
- Center for Cancer and Blood Disorders, Children’s Hospital Colorado, Aurora, CO, United States
| | - Michael Leibowitz
- Center for Cancer and Blood Disorders, Children’s Hospital Colorado, Aurora, CO, United States
| | - Daniel Regan
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
11
|
Brewer DJ, Marwood R, Leeming G, Ressel L, Dukes‐McEwan J. Concurrent right atrial myxoma with visceral haemangiosarcoma in a dog. VETERINARY RECORD CASE REPORTS 2023. [DOI: 10.1002/vrc2.552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- David John Brewer
- Small Animal Teaching Hospital, Department of Small Animal Clinical Sciences Institute of Infection, Veterinary and Ecological Sciences, Leahurst University of Liverpool Neston UK
| | - Rachel Marwood
- Small Animal Teaching Hospital, Department of Small Animal Clinical Sciences Institute of Infection, Veterinary and Ecological Sciences, Leahurst University of Liverpool Neston UK
| | - Gail Leeming
- Department of Veterinary Anatomy, Physiology and Pathology Institute of Infection, Veterinary and Ecological Sciences, Leahurst University of Liverpool Neston UK
| | - Lorenzo Ressel
- Department of Veterinary Anatomy, Physiology and Pathology Institute of Infection, Veterinary and Ecological Sciences, Leahurst University of Liverpool Neston UK
| | - Joanna Dukes‐McEwan
- Small Animal Teaching Hospital, Department of Small Animal Clinical Sciences Institute of Infection, Veterinary and Ecological Sciences, Leahurst University of Liverpool Neston UK
| |
Collapse
|
12
|
Lam L, Tien T, Wildung M, White L, Sellon RK, Fidel JL, Shelden EA. Comparative whole transcriptome analysis of gene expression in three canine soft tissue sarcoma types. PLoS One 2022; 17:e0273705. [PMID: 36099287 PMCID: PMC9469979 DOI: 10.1371/journal.pone.0273705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/11/2022] [Indexed: 11/29/2022] Open
Abstract
Soft tissue sarcomas are pleiotropic tumors of mesenchymal cell origin. These tumors are rare in humans but common in veterinary practice, where they comprise up to 15% of canine skin and subcutaneous cancers. Because they present similar morphologies, primary sites, and growth characteristics, they are treated similarly, generally by surgical resection followed by radiation therapy. Previous studies have examined a variety of genetic changes as potential drivers of tumorigenesis and progression in soft tissue sarcomas as well as their use as markers for soft tissue sarcoma subtypes. However, few studies employing next generation sequencing approaches have been published. Here, we have examined gene expression patterns in canine soft tissue sarcomas using RNA-seq analysis of samples obtained from archived formalin-fixed and paraffin-embedded tumors. We provide a computational framework for using resulting data to categorize tumors, perform cross species comparisons and identify genetic changes associated with tumorigenesis. Functional overrepresentation analysis of differentially expressed genes further implicate both common and tumor-type specific transcription factors as potential mediators of tumorigenesis and aggression. Implications for tumor-type specific therapies are discussed. Our results illustrate the potential utility of this approach for the discovery of new therapeutic approaches to the management of canine soft tissue sarcomas and support the view that both common and tumor-type specific mechanisms drive the development of these tumors.
Collapse
Affiliation(s)
- Lydia Lam
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| | - Tien Tien
- Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| | - Mark Wildung
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| | - Laura White
- Washington Animal Disease Diagnostic Laboratory, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| | - Rance K. Sellon
- Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| | - Janean L. Fidel
- Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| | - Eric A. Shelden
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| |
Collapse
|
13
|
Wong S, Ehrhart EJ, Stewart S, Zismann V, Cawley J, Halperin R, Briones N, Richter K, Sivaprakasam K, Perdigones N, Contente-Cuomo T, Facista S, Trent JM, Murtaza M, Khanna C, Hendricks WPD. Genomic landscapes of canine splenic angiosarcoma (hemangiosarcoma) contain extensive heterogeneity within and between patients. PLoS One 2022; 17:e0264986. [PMID: 35867969 PMCID: PMC9307279 DOI: 10.1371/journal.pone.0264986] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 02/21/2022] [Indexed: 11/18/2022] Open
Abstract
Cancer genomic heterogeneity presents significant challenges for understanding oncogenic processes and for cancer’s clinical management. Variation in driver mutation frequency between patients with the same tumor type as well as within an individual patients’ cancer can shape the use of mutations as diagnostic, prognostic, and predictive biomarkers. We have characterized genomic heterogeneity between and within canine splenic hemangiosarcoma (HSA), a common naturally occurring cancer in pet dogs that is similar to human angiosarcoma (AS). HSA is a clinically, physiologically, and genomically complex canine cancer that may serve as a valuable model for understanding the origin and clinical impact of cancer heterogeneity. We conducted a prospective collection of 52 splenic masses from 43 dogs (27 HSA, 15 benign masses, and 1 stromal sarcoma) presenting for emergency care with hemoperitoneum secondary to a ruptured splenic mass. Multi-platform genomic analysis included matched tumor/normal targeted sequencing panel and exome sequencing. We found candidate somatic cancer driver mutations in 14/27 (52%) HSAs. Among recurrent candidate driver mutations, TP53 was most commonly mutated (30%) followed by PIK3CA (15%), AKT1 (11%), and CDKN2AIP (11%). We also identified significant intratumoral genomic heterogeneity, consistent with a branched evolution model, through multi-region exome sequencing of three distinct tumor regions from selected primary splenic tumors. These data provide new perspectives on the genomic landscape of this veterinary cancer and suggest a cross-species value for using HSA in pet dogs as a naturally occurring model of intratumoral heterogeneity.
Collapse
Affiliation(s)
- Shukmei Wong
- Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - E. J. Ehrhart
- Charles River Laboratories, Wilmington, MA, United States of America
| | - Samuel Stewart
- Ethos Discovery, San Diego, CA, United States of America
- Ethos Veterinary Health, Woburn, MA, United States of America
| | - Victoria Zismann
- Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Jacob Cawley
- Charles River Laboratories, Wilmington, MA, United States of America
- Ethos Discovery, San Diego, CA, United States of America
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Rebecca Halperin
- Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Natalia Briones
- Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Keith Richter
- Ethos Discovery, San Diego, CA, United States of America
- Ethos Veterinary Health, Woburn, MA, United States of America
| | | | - Nieves Perdigones
- Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Tania Contente-Cuomo
- Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Salvatore Facista
- Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Jeffrey M. Trent
- Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Muhammed Murtaza
- Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Chand Khanna
- Ethos Discovery, San Diego, CA, United States of America
- Ethos Veterinary Health, Woburn, MA, United States of America
| | - William P. D. Hendricks
- Translational Genomics Research Institute, Phoenix, Arizona, United States of America
- * E-mail:
| |
Collapse
|
14
|
Nance RL, Sajib AM, Smith BF. Canine models of human cancer: Bridging the gap to improve precision medicine. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 189:67-99. [PMID: 35595353 DOI: 10.1016/bs.pmbts.2021.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Dogs are remarkable, adaptable, and dependable creatures that have evolved alongside humans while contributing tremendously to our survival. Our canine companions share many similarities to human disease, particularly cancer. With the advancement of next-generation sequencing technology, we are beginning to unravel the complexity of cancer and the vast intra- and intertumoral heterogeneity that makes treatment difficult. Consequently, precision medicine has emerged as a therapeutic approach to improve patient survival by evaluating and classifying an individual tumor's molecular profile. Many canine and human cancers share striking similarities in terms of genotypic, phenotypic, clinical, and histological presentations. Dogs are superior to rodent models of cancer because they are a naturally heterogeneous population in which tumors occur spontaneously, are exposed to similar environmental conditions, and show more similarities in key modulators of tumorigenesis and clinical response, including the immune system, drug metabolism, and gut microbiome. In this chapter, we will explore various canine models of human cancers and emphasize the dog's critical role in advancing precision medicine and improving the survival of both man and man's best friend.
Collapse
Affiliation(s)
- Rebecca L Nance
- Scott-Ritchey Research Center, Auburn University College of Veterinary Medicine, Auburn, AL, United States; Department of Pathobiology, Auburn University College of Veterinary Medicine, Auburn, AL, United States
| | - Abdul Mohin Sajib
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Bruce F Smith
- Scott-Ritchey Research Center, Auburn University College of Veterinary Medicine, Auburn, AL, United States; Department of Pathobiology, Auburn University College of Veterinary Medicine, Auburn, AL, United States.
| |
Collapse
|
15
|
Sato T, Hamai A, Kadonosono T, Kizaka-Kondoh S, Omata T. Droplet-based valveless microfluidic system for phage-display screening against spheroids. BIOMICROFLUIDICS 2022; 16:024107. [PMID: 35464138 PMCID: PMC9010049 DOI: 10.1063/5.0085459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/31/2022] [Indexed: 06/14/2023]
Abstract
In this study, we proposed a droplet-based valveless microfluidic system that has the necessary functions to perform the binding, washing, eluting, and collecting processes of phage-display screening against spheroids, which can be expected to present a similar repertoire and number of membrane proteins as in vivo. Although spheroids have much larger sizes than single cells, spheroids are difficult to manipulate through manual operation. The proposed microfluidic system actively controls the position and velocity of droplets using a camera, three air pumps, and three liquid pumps to perform the processes for phage-display screening. The cross section of the microchannel is large in width and height for the passage of spheroids. Valves that can close such a large cross-sectional microchannel are not readily available. Thus, we proposed valveless flow control using liquid pumps. In addition, the proposed microfluidic system involves complex flow channels with airflow subchannels to perform phage-display screening. For washing, nonspecific-binding phages remaining in the flow channels must be minimized. The proposed microfluidic system can perform selective blocking and flush washing. Selective blocking can prevent the airflow channels from becoming hydrophilic with blocking liquid, and flush washing can flush phages remaining in the flow channel. We experimentally verified the functions of the developed microfluidic device based on the proposed system.
Collapse
Affiliation(s)
- Tsuyohi Sato
- Department of Mechanical Engineering, Tokyo Institute of Technology, 226-8503 Kanagawa, Japan
| | - Akira Hamai
- Department of Mechanical Engineering, Tokyo Institute of Technology, 226-8503 Kanagawa, Japan
| | - Tetsuya Kadonosono
- Department of Life Science and Technology, Tokyo Institute of Technology, 226-8503 Kanagawa, Japan
| | - Shinae Kizaka-Kondoh
- Department of Life Science and Technology, Tokyo Institute of Technology, 226-8503 Kanagawa, Japan
| | - Toru Omata
- Department of Mechanical Engineering, Tokyo Institute of Technology, 226-8503 Kanagawa, Japan
| |
Collapse
|
16
|
Li L, Zhong L, Tang C, Gan L, Mo T, Na J, He J, Huang Y. CD105: tumor diagnosis, prognostic marker and future tumor therapeutic target. Clin Transl Oncol 2022; 24:1447-1458. [PMID: 35165838 DOI: 10.1007/s12094-022-02792-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/21/2022] [Indexed: 02/06/2023]
Abstract
Cancer is one of the diseases with the highest morbidity and mortality rates worldwide, and its therapeutic options are inadequate. The endothelial glycoprotein, also known as CD105, is a type I transmembrane glycoprotein located on the surface of the cell membranes and it is one of the transforming growth factor-β (TGF-β) receptor complexes. It regulates the responses associated with binding to transforming growth factor β1 egg (Activin-A), bone morphogenetic protein 2 (BMP-2), and bone morphogenetic protein 7 (BMP-7). Additionally, it is involved in the regulation of angiogenesis. This glycoprotein is indispensable in the treatment of tumor angiogenesis, and it also plays a leading role in tumor angiogenesis therapy. Therefore, CD105 is considered to be a novel therapeutic target. In this study, we explored the significance of CD105 in the diagnosis, treatment and prognosis of various tumors, and provided evidence for the effect and mechanism of CD105 on tumors.
Collapse
Affiliation(s)
- Lan Li
- National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Liping Zhong
- National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Chao Tang
- National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Lu Gan
- National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Tong Mo
- National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jintong Na
- National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jian He
- National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yong Huang
- National Center for International Research of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
17
|
Hanna JA, Langdon CG, Garcia MR, Benton A, Lanman NA, Finkelstein D, Rehg JE, Hatley ME. Genetic context of oncogenic drivers dictates vascular sarcoma development in
aP2‐Cre
mice. J Pathol 2022; 257:109-124. [PMID: 35066877 PMCID: PMC9007915 DOI: 10.1002/path.5873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 01/06/2022] [Accepted: 01/20/2022] [Indexed: 12/02/2022]
Abstract
Angiosarcomas are aggressive vascular sarcomas that arise from endothelial cells and have an extremely poor prognosis. Because of the rarity of angiosarcomas, knowledge of molecular drivers and optimized treatment strategies is lacking, highlighting the need for in vivo models to study the disease. Previously, we generated genetically engineered mouse models of angiosarcoma driven by aP2‐Cre‐mediated biallelic loss of Dicer1 or conditional activation of KrasG12D with Cdkn2a loss that histologically and genetically resemble human tumors. In the present study, we found that DICER1 functions as a potent tumor suppressor and its deletion, in combination with either KRASG12D expression or Cdkn2a loss, is associated with angiosarcoma development. Independent of the genetic driver, the mTOR pathway was activated in all murine angiosarcoma models. Direct activation of the mTOR pathway by conditional deletion of Tsc1 with aP2‐Cre resulted in tumors that resemble intermediate grade human kaposiform hemangioendotheliomas, indicating that mTOR activation was not sufficient to drive the malignant angiosarcoma phenotype. Genetic dissection of the spectrum of vascular tumors identified genes specifically regulated in the aggressive murine angiosarcomas that are also enriched in human angiosarcoma. The genetic dissection driving the transition across the malignant spectrum of endothelial sarcomas provides an opportunity to identify key determinants of the malignant phenotype, novel therapies for angiosarcoma, and novel in vivo models to further explore angiosarcoma pathogenesis. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jason A. Hanna
- Department of Oncology St. Jude Children's Research Hospital 262 Danny Thomas Place Memphis TN 38105 USA
- Department of Biological Sciences Purdue University 201 S. University Street West Lafayette IN 47906 USA
- Purdue University Center for Cancer Research Purdue University West Lafayette, IN, 47907 USA
| | - Casey G. Langdon
- Department of Oncology St. Jude Children's Research Hospital 262 Danny Thomas Place Memphis TN 38105 USA
| | - Matthew R. Garcia
- Department of Oncology St. Jude Children's Research Hospital 262 Danny Thomas Place Memphis TN 38105 USA
| | - Annaleigh Benton
- Department of Biological Sciences Purdue University 201 S. University Street West Lafayette IN 47906 USA
- Purdue University Center for Cancer Research Purdue University West Lafayette, IN, 47907 USA
| | - Nadia A. Lanman
- Department of Comparative Pathobiology Purdue University 201 S. University Street West Lafayette IN 47906 USA
- Purdue University Center for Cancer Research Purdue University West Lafayette, IN, 47907 USA
| | - David Finkelstein
- Department of Computational Biology St. Jude Children's Research Hospital 262 Danny Thomas Place Memphis TN 38105 USA
| | - Jerold E. Rehg
- Department of Pathology St. Jude Children's Research Hospital 262 Danny Thomas Place Memphis TN 38105 USA
| | - Mark E. Hatley
- Department of Oncology St. Jude Children's Research Hospital 262 Danny Thomas Place Memphis TN 38105 USA
| |
Collapse
|
18
|
Massimini M, Romanucci M, De Maria R, Della Salda L. An Update on Molecular Pathways Regulating Vasculogenic Mimicry in Human Osteosarcoma and Their Role in Canine Oncology. Front Vet Sci 2021; 8:722432. [PMID: 34631854 PMCID: PMC8494780 DOI: 10.3389/fvets.2021.722432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/23/2021] [Indexed: 01/16/2023] Open
Abstract
Canine tumors are valuable comparative models for human counterparts, especially to explore novel biomarkers and to understand pathways and processes involved in metastasis. Vasculogenic mimicry (VM) is a unique property of malignant cancer cells which promote metastasis. Thus, it represents an opportunity to investigate both the molecular mechanisms and the therapeutic targets of a crucial phenotypic malignant switch. Although this biological process has been largely investigated in different human cancer types, including osteosarcoma, it is still largely unknown in veterinary pathology, where it has been mainly explored in canine mammary tumors. The presence of VM in human osteosarcoma is associated with poor clinical outcome, reduced patient survival, and increased risk of metastasis and it shares the main pathways involved in other type of human tumors. This review illustrates the main findings concerning the VM process in human osteosarcoma, search for the related current knowledge in canine pathology and oncology, and potential involvement of multiple pathways in VM formation, in order to provide a basis for future investigations on VM in canine tumors.
Collapse
|
19
|
Griffin MA, Culp WTN, Rebhun RB. Canine and feline haemangiosarcoma. Vet Rec 2021; 189:e585. [PMID: 34213807 DOI: 10.1002/vetr.585] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/12/2021] [Accepted: 05/04/2021] [Indexed: 11/07/2022]
Abstract
INTRODUCTION Haemangiosarcoma (HSA) is a malignant neoplasm of dogs and cats that is suspected to originate from a pluripotent bone marrow progenitor with a complex and multifactorial pathogenesis. APPROACH Pertinent literature was identified, reviewed, and summarized for inclusion in the manuscript. RESULTS/INTERPRETATION Dogs are more frequently diagnosed with HSA than cats, and primary sites of this disease include dermal, subcutaneous/intramuscular, and visceral (most commonly the spleen). Dogs and cats with HSA generally have a poor prognosis owing to the rapid and widespread metastasis typically associated with this disease. However, some forms such as cutaneous HSA behave in a less aggressive fashion with improved outcomes. Surgical excision and anthracycline-based chemotherapy remain the mainstays of treatment, although novel treatment modalities are currently under investigation for potential roles in treatment of this disease. CONCLUSION This review aims to describe the clinical presentation and progression of the various forms of HSA in dogs and cats as well as to provide a systematic review of the veterinary literature with a focus on the various published treatment options and associated outcomes.
Collapse
Affiliation(s)
- Maureen A Griffin
- School of Veterinary Medicine, University of California-Davis, Davis, California, USA
| | - William T N Culp
- School of Veterinary Medicine, University of California-Davis, Davis, California, USA
| | - Robert B Rebhun
- School of Veterinary Medicine, University of California-Davis, Davis, California, USA
| |
Collapse
|
20
|
Kim JH, Megquier K, Thomas R, Sarver AL, Song JM, Kim YT, Cheng N, Schulte AJ, Linden MA, Murugan P, Oseth L, Forster CL, Elvers I, Swofford R, Turner-Maier J, Karlsson EK, Breen M, Lindblad-Toh K, Modiano JF. Genomically Complex Human Angiosarcoma and Canine Hemangiosarcoma Establish Convergent Angiogenic Transcriptional Programs Driven by Novel Gene Fusions. Mol Cancer Res 2021; 19:847-861. [PMID: 33649193 DOI: 10.1158/1541-7786.mcr-20-0937] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/21/2020] [Accepted: 02/10/2021] [Indexed: 11/16/2022]
Abstract
Sporadic angiosarcomas are aggressive vascular sarcomas whose rarity and genomic complexity present significant obstacles in deciphering the pathogenic significance of individual genetic alterations. Numerous fusion genes have been identified across multiple types of cancers, but their existence and significance remain unclear in sporadic angiosarcomas. In this study, we leveraged RNA-sequencing data from 13 human angiosarcomas and 76 spontaneous canine hemangiosarcomas to identify fusion genes associated with spontaneous vascular malignancies. Ten novel protein-coding fusion genes, including TEX2-PECAM1 and ATP8A2-FLT1, were identified in seven of the 13 human tumors, with two tumors showing mutations of TP53. HRAS and NRAS mutations were found in angiosarcomas without fusions or TP53 mutations. We found 15 novel protein-coding fusion genes including MYO16-PTK2, GABRA3-FLT1, and AKT3-XPNPEP1 in 11 of the 76 canine hemangiosarcomas; these fusion genes were seen exclusively in tumors of the angiogenic molecular subtype that contained recurrent mutations in TP53, PIK3CA, PIK3R1, and NRAS. In particular, fusion genes and mutations of TP53 cooccurred in tumors with higher frequency than expected by random chance, and they enriched gene signatures predicting activation of angiogenic pathways. Comparative transcriptomic analysis of human angiosarcomas and canine hemangiosarcomas identified shared molecular signatures associated with activation of PI3K/AKT/mTOR pathways. Our data suggest that genome instability induced by TP53 mutations might create a predisposition for fusion events that may contribute to tumor progression by promoting selection and/or enhancing fitness through activation of convergent angiogenic pathways in this vascular malignancy. IMPLICATIONS: This study shows that, while drive events of malignant vasoformative tumors of humans and dogs include diverse mutations and stochastic rearrangements that create novel fusion genes, convergent transcriptional programs govern the highly conserved morphologic organization and biological behavior of these tumors in both species.
Collapse
Affiliation(s)
- Jong Hyuk Kim
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota. .,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Institute for Engineering in Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Kate Megquier
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Rachael Thomas
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine & Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | - Aaron L Sarver
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Institute for Health Informatics, University of Minnesota, Minneapolis, Minnesota
| | - Jung Min Song
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Yoon Tae Kim
- Department of Electrical Engineering and Computer Science, York University, Toronto, Ontario, Canada
| | - Nuojin Cheng
- School of Mathematics, College of Science and Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Ashley J Schulte
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Michael A Linden
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Paari Murugan
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - LeAnn Oseth
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Colleen L Forster
- The University of Minnesota Biological Materials Procurement Network (BioNet), University of Minnesota, Minneapolis, Minnesota
| | - Ingegerd Elvers
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Ross Swofford
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | | | - Elinor K Karlsson
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,University of Massachusetts Medical School, Worcester, Massachusetts
| | - Matthew Breen
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine & Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina.,Cancer Genetics Program, University of North Carolina Lineberger Comprehensive Cancer Center, Raleigh, North Carolina
| | - Kerstin Lindblad-Toh
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Jaime F Modiano
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Institute for Engineering in Medicine, University of Minnesota, Minneapolis, Minnesota.,Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, Minnesota.,Center for Immunology, University of Minnesota, Minneapolis, Minnesota.,Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
21
|
Cheng N, Schulte AJ, Santosa F, Kim JH. Machine learning application identifies novel gene signatures from transcriptomic data of spontaneous canine hemangiosarcoma. Brief Bioinform 2020; 22:5930848. [PMID: 33078825 DOI: 10.1093/bib/bbaa252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/02/2020] [Accepted: 09/08/2020] [Indexed: 01/20/2023] Open
Abstract
Angiosarcomas are soft-tissue sarcomas that form malignant vascular tissues. Angiosarcomas are very rare, and due to their aggressive behavior and high metastatic propensity, they have poor clinical outcomes. Hemangiosarcomas commonly occur in domestic dogs, and share pathological and clinical features with human angiosarcomas. Typical pathognomonic features of this tumor are irregular vascular channels that are filled with blood and are lined by a mixture of malignant and nonmalignant endothelial cells. The current gold standard is the histological diagnosis of angiosarcoma; however, microscopic evaluation may be complicated, particularly when tumor cells are undetectable due to the presence of excessive amounts of nontumor cells or when tissue specimens have insufficient tumor content. In this study, we implemented machine learning applications from next-generation transcriptomic data of canine hemangiosarcoma tumor samples (n = 76) and nonmalignant tissues (n = 10) to evaluate their training performance for diagnostic utility. The 10-fold cross-validation test and multiple feature selection methods were applied. We found that extra trees and random forest learning models were the best classifiers for hemangiosarcoma in our testing datasets. We also identified novel gene signatures using the mutual information and Monte Carlo feature selection method. The extra trees model revealed high classification accuracy for hemangiosarcoma in validation sets. We demonstrate that high-throughput sequencing data of canine hemangiosarcoma are trainable for machine learning applications. Furthermore, our approach enables us to identify novel gene signatures as reliable determinants of hemangiosarcoma, providing significant insights into the development of potential applications for this vascular malignancy.
Collapse
Affiliation(s)
- Nuojin Cheng
- School of Mathematics, College of Science and Engineering at the University of Minnesota, Minneapolis, MN, USA
| | - Ashley J Schulte
- Animal Cancer Care and Research Program, Department of Veterinary Clinical Sciences, College of Veterinary Medicine at the University of Minnesota, St Paul, MN, USA
| | - Fadil Santosa
- Department of Applied Mathematics & Statistics, Whiting School of Engineering at the Johns Hopkins University, Baltimore, MD, USA
| | - Jong Hyuk Kim
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine at the University of Minnesota, St Paul, MN, USA
| |
Collapse
|
22
|
Mukai C, Choi E, Sams KL, Klampen EZ, Anguish L, Marks BA, Rice EJ, Wang Z, Choate LA, Chou SP, Kato Y, Miller AD, Danko CG, Coonrod SA. Chromatin run-on sequencing analysis finds that ECM remodeling plays an important role in canine hemangiosarcoma pathogenesis. BMC Vet Res 2020; 16:206. [PMID: 32571313 PMCID: PMC7310061 DOI: 10.1186/s12917-020-02395-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 05/29/2020] [Indexed: 01/20/2023] Open
Abstract
Background Canine visceral hemangiosarcoma (HSA) is a highly aggressive cancer of endothelial origin that closely resembles visceral angiosarcoma in humans, both clinically and histopathologically. Currently there is an unmet need for new diagnostics and therapies for both forms of this disease. The goal of this study was to utilize Chromatin run-on sequencing (ChRO-seq) and immunohistochemistry (IHC) to identify gene and protein expression signatures that may be important drivers of HSA progression. Results ChRO-seq was performed on tissue isolated from 17 HSA samples and 4 normal splenic samples. Computational analysis was then used to identify differentially expressed genes and these factors were subjected to gene ontology analysis. ChRO-seq analysis revealed over a thousand differentially expressed genes in HSA tissue compared with normal splenic tissue (FDR < 0.005). Interestingly, the majority of genes overexpressed in HSA tumor tissue were associated with extracellular matrix (ECM) remodeling. This observation correlated well with our histological analysis, which found that HSA tumors contain a rich and complex collagen network. Additionally, we characterized the protein expression patterns of two highly overexpressed molecules identified in ChRO-seq analysis, podoplanin (PDPN) and laminin alpha 4 (LAMA4). We found that the expression of these two ECM-associated factors appeared to be largely limited to transformed endothelial cells within the HSA lesions. Conclusion Outcomes from this study suggest that ECM remodeling plays an important role in HSA progression. Additionally, our study identified two potential novel biomarkers of HSA, PDPN and LAMA4. Interestingly, given that function-blocking anti-PDPN antibodies have shown anti-tumor effects in mouse models of canine melanoma, our studies raise the possibility that these types of therapeutic strategies could potentially be developed for treating canine HSA.
Collapse
Affiliation(s)
- Chinatsu Mukai
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| | - Eunju Choi
- Department of Biomedical Sciences, Section of Anatomic Pathology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Kelly L Sams
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Elena Zu Klampen
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Lynne Anguish
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Brooke A Marks
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Edward J Rice
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Zhong Wang
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Lauren A Choate
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Shao-Pei Chou
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan.,New Industry Creation Hatchery Center, Tohoku University, Sendai, Japan
| | - Andrew D Miller
- Department of Biomedical Sciences, Section of Anatomic Pathology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Charles G Danko
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Scott A Coonrod
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| |
Collapse
|
23
|
Atherton MJ, Lenz JA, Mason NJ. Sarcomas-A barren immunological wasteland or field of opportunity for immunotherapy? Vet Comp Oncol 2020; 18:447-470. [PMID: 32246517 DOI: 10.1111/vco.12595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/13/2020] [Accepted: 03/24/2020] [Indexed: 12/11/2022]
Abstract
Key advances in our understanding of immunobiology and the immunosuppressive mechanisms of the tumour microenvironment have led to significant breakthroughs in manipulating the immune system to successfully treat cancer. Remarkable therapeutic responses have occurred with tumours that carry a high mutational burden. In these cases, pre-existing tumour-specific T cells can be rejuvenated via checkpoint inhibition to eliminate tumours. Furthermore, durable remissions have been achieved in haematological malignancies following adoptive transfer of T cells that specifically target cell surface proteins where expression is restricted to the malignancy's cell of origin. Soft tissue sarcomas and bone sarcomas have a paucity of non-synonymous somatic mutations and do not commonly express known, targetable, tumour-specific antigens. Historically, soft tissue sarcomas have been considered immunologically 'cold' and as such, unlikely candidates for immune therapy. Here, we review the immune landscape of canine and feline sarcomas and the immunotherapeutic strategies that have been employed in veterinary clinical trials to improve patient outcome. We also provide insight into immunotherapeutic approaches being used to treat human sarcomas. Together, current data indicates that, rather than a barren immunological wasteland, sarcomas represent a field of opportunities for immunotherapies. Furthermore, we and others would suggest that strategic combinations of immunotherapeutic approaches may hold promise for more effective treatments for high grade soft tissue sarcomas and bone sarcomas.
Collapse
Affiliation(s)
- Matthew J Atherton
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jennifer A Lenz
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nicola J Mason
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
24
|
Borgatti A, Fieberg A, Winter AL, Stuebner K, Taras E, Todhunter D, Masyr A, Rendhal A, Vallera DA, Koopmeiners JS, Modiano JF. Impact of repeated cycles of EGF bispecific angiotoxin (eBAT) administered at a reduced interval from doxorubicin chemotherapy in dogs with splenic haemangiosarcoma. Vet Comp Oncol 2020; 18:664-674. [PMID: 32187827 DOI: 10.1111/vco.12590] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 12/13/2022]
Abstract
We previously reported that eBAT, an EGF-targeted angiotoxin, was safe and it improved the overall survival for dogs with splenic haemangiosarcoma when added to the standard of care in a single cycle of three administrations in the minimal residual disease setting. Our objective for the SRCBST-2 trial was to assess whether increased dosing through multiple cycles of eBAT would be well tolerated and would further enhance the benefits of eBAT. Eligibility was expanded to dogs with stage 3 haemangiosarcoma, provided that gross lesions could be surgically excised. The interval between eBAT and the start of chemotherapy was reduced, and the experimental therapy was expanded to three cycles, each administered at the biologically active dose (50 μg/kg) on a Monday/Wednesday/Friday schedule following splenectomy, and scheduled 1 week prior to the first, second and fifth doxorubicin chemotherapy. Twenty-five dogs were enrolled; six experienced acute hypotension with two requiring hospitalization. Self-limiting elevation of ALT was observed in one dog. A statistically significant survival benefit was not seen in this study in eBAT-treated dogs compared with a Contemporary comparison group of dogs with stages 1-3 haemangiosarcoma treated with standard of care alone. Our results indicate that repeated dosing cycles of eBAT starting 1 week prior to doxorubicin chemotherapy led to greater toxicity and reduced efficacy compared with a single cycle given between surgery and a delayed start of chemotherapy. Further work is needed to understand the precise mechanisms of action of eBAT in order to optimize its clinical benefits in the treatment of canine haemangiosarcoma and other tumours. IACUC Protocols 1110A06186 and 1507-32804A.
Collapse
Affiliation(s)
- Antonella Borgatti
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota, USA.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.,Clinical Investigation Center, College of Veterinary Medicine, St. Paul, Minnesota, USA
| | - Ann Fieberg
- Coordinating Center for Biometric Research, University of Minnesota, Minneapolis, Minnesota, USA
| | - Amber L Winter
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota, USA.,Clinical Investigation Center, College of Veterinary Medicine, St. Paul, Minnesota, USA
| | - Kathleen Stuebner
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota, USA.,Clinical Investigation Center, College of Veterinary Medicine, St. Paul, Minnesota, USA
| | - Elizabeth Taras
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Radiation Oncology, School of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Deborah Todhunter
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Radiation Oncology, School of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Alison Masyr
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Aaron Rendhal
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota, USA.,Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Daniel A Vallera
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Radiation Oncology, School of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Joseph S Koopmeiners
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.,Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jaime F Modiano
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota, USA.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.,Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA.,Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
25
|
Wang G, Wu M, Durham AC, Radaelli E, Mason NJ, Xu X, Roth DB. Molecular subtypes in canine hemangiosarcoma reveal similarities with human angiosarcoma. PLoS One 2020; 15:e0229728. [PMID: 32210430 PMCID: PMC7094861 DOI: 10.1371/journal.pone.0229728] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/12/2020] [Indexed: 12/12/2022] Open
Abstract
Angiosarcoma (AS) is a rare neoplasm with limited treatment options and a poor survival rate. Development of effective therapies is hindered by the rarity of this disease. Dogs spontaneously develop hemangiosarcoma (HSA), a common, histologically similar neoplasm. Metastatic disease occurs rapidly and despite chemotherapy, most dogs die several months after diagnosis. These features suggest that HSA might provide a tractable model to test experimental therapies in clinical trials. We previously reported whole exome sequencing of 20 HSA cases. Here we report development of a NGS targeted resequencing panel to detect driver mutations in HSA and other canine tumors. We validated the panel by resequencing the original 20 cases and sequenced 30 additional cases. Overall, we identified potential driver mutations in over 90% of the cases, including well-documented (in human cancers) oncogenic mutations in PIK3CA (46%), PTEN (6%), PLCG1(4%), and TP53 (66%), as well as previously undetected recurrent activating mutations in NRAS (24%). The driver role of these mutations is further demonstrated by augmented downstream signaling crucial to tumor growth. The recurrent, mutually exclusive mutation patterns suggest distinct molecular subtypes of HSA. Driver mutations in some subtypes closely resemble those seen in some AS cases, including NRAS, PLCG1, PIK3CA and TP53. Furthermore, activation of the MAPK and PI3K pathways appear to be key oncogenic mechanisms in both species. Together, these observations suggest that dogs with spontaneous HSA could serve as a useful model for testing the efficacy of targeted therapies, some of which could potentially be of therapeutic value in AS.
Collapse
Affiliation(s)
- Guannan Wang
- Department of Pathology and Laboratory Medicine, Raymond and Ruth Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- * E-mail: (GW); (DBR)
| | - Ming Wu
- Illumina, San Diego, CA, United States of America
| | - Amy C. Durham
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Enrico Radaelli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Nicola J. Mason
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - XiaoWei Xu
- Department of Pathology and Laboratory Medicine, Raymond and Ruth Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - David B. Roth
- Department of Pathology and Laboratory Medicine, Raymond and Ruth Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- * E-mail: (GW); (DBR)
| |
Collapse
|
26
|
Kakiuchi-Kiyota S, Obert LA, Crowell DM, Xia S, Roy MD, Coskran TM, Kreeger JM, Crabbs TA, Cohen SM, Cattley RC, Cook JC. Expression of Hematopoietic Stem and Endothelial Cell Markers in Canine Hemangiosarcoma. Toxicol Pathol 2020; 48:481-493. [PMID: 31918642 DOI: 10.1177/0192623319897539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Several chemicals and pharmaceuticals increase the incidence of hemangiosarcomas (HSAs) in mice, but the relevance to humans is uncertain. Recently, canine HSAs were identified as a powerful tool for investigating the pathogenesis of human HSAs. To characterize the cellular phenotype of canine HSAs, we evaluated immunoreactivity and/or messenger RNA (mRNA) expression of markers for hematopoietic stem cells (HSCs), endothelial cells (ECs), a tumor suppressor protein, and a myeloid marker in canine HSAs. Neoplastic canine cells expressed EC markers and a myeloid marker, but expressed HSC markers less consistently. The canine tumor expression results were then compared to previously published immunoreactivity results for these markers in human and mouse HSAs. There are 2 noteworthy differences across species: (1) most human HSAs had HSC marker expression, indicating that they were comprised of tumor cells that were less differentiated than those in canine and mouse tumors; and (2) human and canine HSAs expressed a late-stage EC maturation marker, whereas mouse HSAs were negative, suggesting that human and canine tumors may retain greater differentiation potential than mouse tumors. These results indicate that HSA development is variable across species and that caution is necessary when discussing translation of carcinogenic risk from animal models to humans.
Collapse
Affiliation(s)
| | - Leslie A Obert
- Drug Safety Research & Development, Pfizer Inc., Groton, CT, USA
| | | | - Shuhua Xia
- Drug Safety Research & Development, Pfizer Inc., Groton, CT, USA
| | - Marc D Roy
- Drug Safety Research & Development, Pfizer Inc., Groton, CT, USA
| | | | - John M Kreeger
- Drug Safety Research & Development, Pfizer Inc., Groton, CT, USA
| | - Torrie A Crabbs
- Experimental Pathology Laboratories, Inc., Research Triangle Park, NC, USA
| | - Samuel M Cohen
- Department of Pathology and Microbiology, University of Nebraska Medical Center, NE, USA
| | - Russell C Cattley
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, AL, USA. Kakiuchi-Kiyota is now with Safety Assessment, Genentech Inc., South San Francisco, CA, USA; Obert is now with Translational Medicine & Comparative Pathology, GlaxoSmithKline, Collegeville, PA, USA; Roy is now with Nonclinical Development, Sarepta Therapeutics, Cambridge, MA, USA
| | - Jon C Cook
- Drug Safety Research & Development, Pfizer Inc., Groton, CT, USA
| |
Collapse
|
27
|
Megquier K, Turner-Maier J, Swofford R, Kim JH, Sarver AL, Wang C, Sakthikumar S, Johnson J, Koltookian M, Lewellen M, Scott MC, Schulte AJ, Borst L, Tonomura N, Alfoldi J, Painter C, Thomas R, Karlsson EK, Breen M, Modiano JF, Elvers I, Lindblad-Toh K. Comparative Genomics Reveals Shared Mutational Landscape in Canine Hemangiosarcoma and Human Angiosarcoma. Mol Cancer Res 2019; 17:2410-2421. [PMID: 31570656 PMCID: PMC7067513 DOI: 10.1158/1541-7786.mcr-19-0221] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 07/12/2019] [Accepted: 09/25/2019] [Indexed: 12/23/2022]
Abstract
Angiosarcoma is a highly aggressive cancer of blood vessel-forming cells with few effective treatment options and high patient mortality. It is both rare and heterogenous, making large, well-powered genomic studies nearly impossible. Dogs commonly suffer from a similar cancer, called hemangiosarcoma, with breeds like the golden retriever carrying heritable genetic factors that put them at high risk. If the clinical similarity of canine hemangiosarcoma and human angiosarcoma reflects shared genomic etiology, dogs could be a critically needed model for advancing angiosarcoma research. We assessed the genomic landscape of canine hemangiosarcoma via whole-exome sequencing (47 golden retriever hemangiosarcomas) and RNA sequencing (74 hemangiosarcomas from multiple breeds). Somatic coding mutations occurred most frequently in the tumor suppressor TP53 (59.6% of cases) as well as two genes in the PI3K pathway: the oncogene PIK3CA (29.8%) and its regulatory subunit PIK3R1 (8.5%). The predominant mutational signature was the age-associated deamination of cytosine to thymine. As reported in human angiosarcoma, CDKN2A/B was recurrently deleted and VEGFA, KDR, and KIT recurrently gained. We compared the canine data to human data recently released by The Angiosarcoma Project, and found many of the same genes and pathways significantly enriched for somatic mutations, particularly in breast and visceral angiosarcomas. Canine hemangiosarcoma closely models the genomic landscape of human angiosarcoma of the breast and viscera, and is a powerful tool for investigating the pathogenesis of this devastating disease. IMPLICATIONS: We characterize the genomic landscape of canine hemangiosarcoma and demonstrate its similarity to human angiosarcoma.
Collapse
Affiliation(s)
- Kate Megquier
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | | | - Ross Swofford
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Jong-Hyuk Kim
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Aaron L Sarver
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Institute for Health Informatics, University of Minnesota, Minneapolis, Minnesota
| | - Chao Wang
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Sharadha Sakthikumar
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Jeremy Johnson
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | | | - Mitzi Lewellen
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Milcah C Scott
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Ashley J Schulte
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Luke Borst
- Department of Clinical Sciences, North Carolina State College of Veterinary Medicine, Raleigh, North Carolina
| | - Noriko Tonomura
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts
| | - Jessica Alfoldi
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Corrie Painter
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Count Me In, Cambridge, Massachusetts
| | - Rachael Thomas
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, and Comparative Medicine Institute, Raleigh, North Carolina
| | - Elinor K Karlsson
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Matthew Breen
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, and Comparative Medicine Institute, Raleigh, North Carolina
| | - Jaime F Modiano
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Center for Immunology, University of Minnesota, Minneapolis, Minneapolis
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
- Institute for Engineering in Medicine, University of Minnesota, Minneapolis, Minnesota
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Ingegerd Elvers
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Kerstin Lindblad-Toh
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
28
|
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
|
29
|
Pulz LH, Barra CN, Alexandre PA, Huete GC, Cadrobbi KG, Nishiya AT, de Freitas SH, Fukumasu H, Strefezzi RF. Identification of two molecular subtypes in canine mast cell tumours through gene expression profiling. PLoS One 2019; 14:e0217343. [PMID: 31216299 PMCID: PMC6583995 DOI: 10.1371/journal.pone.0217343] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 05/09/2019] [Indexed: 12/16/2022] Open
Abstract
Mast cell tumours (MCTs) are common neoplasms in dogs and are usually regarded as potentially malignant. Several studies have attempted to identify biomarkers to better predict biological behaviours for this tumour. The aim of this study was to identify pathways connected to clinical and histopathological malignancies, shorter survival times, and poor prognoses associated with MCTs. We performed genome-wide gene expression analyses on tissues obtained from 15 dogs with single MCTs, and identified two distinct tumour subtypes—high-risk and low-risk—associated with differences in histological grades, survival times, Ki67 indices, and occurrence of death due the disease. Comparative analyses of RNA sequence profiles revealed 71 genes that were differentially expressed between high- and low-risk MCTs. In addition to these analyses, we also examined gene co-expression networks to explore the biological functions of the identified genes. The network construction revealed 63 gene modules, of which 4 were significantly associated with the more aggressive tumour group. Two of the gene modules positively correlated with high-risk MCTs were also associated with cell proliferation and extracellular matrix-related terms. At the top of the extracellular matrix module category, genes with functions directly related to those of cancer-associated fibroblasts (CAFs) were identified. Immunohistochemical analyses also revealed a greater number of CAFs in high-risk MCTs. This study provides a method for the molecular characterisation of canine MCTs into two distinct subtypes. Our data indicate that proliferation pathways are significantly involved in malignant tumour behaviours, which are known to be relevant for the induction and maintenance of MCTs. Finally, animals presenting high-risk MCTs overexpress genes associated with the extracellular matrix that can be robustly linked to CAF functions. We suggest that CAFs in the MCT stroma contribute to cancer progression.
Collapse
Affiliation(s)
- Lidia H. Pulz
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
- Department of Pathology, Faculty of Veterinary Medicine and Animal Science- FMVZ, University of Sao Paulo, São Paulo, SP, Brazil
- * E-mail:
| | - Camila N. Barra
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
- Department of Pathology, Faculty of Veterinary Medicine and Animal Science- FMVZ, University of Sao Paulo, São Paulo, SP, Brazil
| | - Pamela A. Alexandre
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| | - Greice C. Huete
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| | - Karine G. Cadrobbi
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| | - Adriana T. Nishiya
- Veterinary Hospital Anhembi Morumbi, R. Conselheiro Lafaiete, São Paulo—SP, Anhembi Morumbi University, São Paulo, SP, Brazil
| | - Silvio Henrique de Freitas
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| | - Heidge Fukumasu
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| | - Ricardo F. Strefezzi
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| |
Collapse
|
30
|
Abstract
Histiocytic sarcoma (HS) and hemangiosarcoma (HSA) are uncommon and aggressive neoplasms that develop much more frequently in dogs than in cats. Breed-specific predispositions have been identified for both cancers. The development of novel diagnostics is underway and may aid in earlier diagnosis. Therapeutic approaches to HS and HSA depend on the stage of disease and may include surgery, radiation therapy, and chemotherapy. Such interventions improve outcome; however, aside from a small number of clinical circumstances, both diseases are considered largely incurable. Continued efforts toward the identification of driver mutations and subsequent druggable targets may lead to improvements in long-term prognosis.
Collapse
Affiliation(s)
- Christine Mullin
- Hope Veterinary Specialists, 40 Three Tun Road, Malvern, PA 19355, USA.
| | - Craig A Clifford
- Hope Veterinary Specialists, 40 Three Tun Road, Malvern, PA 19355, USA
| |
Collapse
|
31
|
Megquier K, Genereux DP, Hekman J, Swofford R, Turner-Maier J, Johnson J, Alonso J, Li X, Morrill K, Anguish LJ, Koltookian M, Logan B, Sharp CR, Ferrer L, Lindblad-Toh K, Meyers-Wallen VN, Hoffman A, Karlsson EK. BarkBase: Epigenomic Annotation of Canine Genomes. Genes (Basel) 2019; 10:E433. [PMID: 31181663 PMCID: PMC6627511 DOI: 10.3390/genes10060433] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 12/20/2022] Open
Abstract
Dogs are an unparalleled natural model for investigating the genetics of health and disease, particularly for complex diseases like cancer. Comprehensive genomic annotation of regulatory elements active in healthy canine tissues is crucial both for identifying candidate causal variants and for designing functional studies needed to translate genetic associations into disease insight. Currently, canine geneticists rely primarily on annotations of the human or mouse genome that have been remapped to dog, an approach that misses dog-specific features. Here, we describe BarkBase, a canine epigenomic resource available at barkbase.org. BarkBase hosts data for 27 adult tissue types, with biological replicates, and for one sample of up to five tissues sampled at each of four carefully staged embryonic time points. RNA sequencing is complemented with whole genome sequencing and with assay for transposase-accessible chromatin using sequencing (ATAC-seq), which identifies open chromatin regions. By including replicates, we can more confidently discern tissue-specific transcripts and assess differential gene expression between tissues and timepoints. By offering data in easy-to-use file formats, through a visual browser modeled on similar genomic resources for human, BarkBase introduces a powerful new resource to support comparative studies in dogs and humans.
Collapse
Affiliation(s)
- Kate Megquier
- Vertebrate Genomics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Diane P Genereux
- Vertebrate Genomics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Jessica Hekman
- Vertebrate Genomics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Ross Swofford
- Vertebrate Genomics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Jason Turner-Maier
- Vertebrate Genomics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Jeremy Johnson
- Vertebrate Genomics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Jacob Alonso
- Vertebrate Genomics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Xue Li
- Vertebrate Genomics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
- Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | - Kathleen Morrill
- Vertebrate Genomics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
- Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | - Lynne J Anguish
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| | - Michele Koltookian
- Vertebrate Genomics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Brittney Logan
- Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | - Claire R Sharp
- School of Veterinary and Life Sciences, College of Veterinary Medicine, Murdoch University, Perth, Murdoch, WA 6150, Australia.
| | - Lluis Ferrer
- Departament de Medicina i Cirurgia Animals Veterinary School, Universitat Autonoma de Barcelona, 08193 Barcelona, Spain.
| | - Kerstin Lindblad-Toh
- Vertebrate Genomics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
- Science for Life Laboratory, Department of Medical Biochemistry & Microbiology, Uppsala University, 751 23 Uppsala, Sweden.
| | - Vicki N Meyers-Wallen
- Baker Institute for Animal Health and Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14850, USA.
| | - Andrew Hoffman
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
- Cummings School of Veterinary Medicine, Tufts University, Grafton, MA 01536, USA.
| | - Elinor K Karlsson
- Vertebrate Genomics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
- Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| |
Collapse
|
32
|
Alfaleh MA, Arora N, Yeh M, de Bakker CJ, Howard CB, Macpherson P, Allavena RE, Chen X, Harkness L, Mahler SM, Jones ML. Canine CD117-Specific Antibodies with Diverse Binding Properties Isolated from a Phage Display Library Using Cell-Based Biopanning. Antibodies (Basel) 2019; 8:E15. [PMID: 31544821 PMCID: PMC6640692 DOI: 10.3390/antib8010015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 12/28/2018] [Accepted: 01/29/2019] [Indexed: 12/11/2022] Open
Abstract
CD117 (c-Kit) is a tyrosine kinase receptor that is overexpressed in multiple dog tumors. There is 100% homology between the juxtamembrane domain of human and canine CD117, and many cancer-causing mutations occur in this region in both species. Thus, CD117 is an important target for cancer treatment in dogs and for comparative oncology studies. Currently, there is no monoclonal antibody (mAb) specifically designed to target the exposed region of canine CD117, although there exist some with species cross-reactivity. We panned a naïve phage display library to isolate antibodies against recombinant CD117 on whole cells. Several mAbs were isolated and were shown to bind recombinant canine CD117 at low- to sub-nanomolar affinity. Additionally, binding to native canine CD117 was confirmed by immunohistochemistry and by flow cytometry. Competitive binding assays also identified mAbs that competed with the CD117 receptor-specific ligand, the stem cell factor (SCF). These results show the ability of our cell-based biopanning strategy to isolate a panel of antibodies that have varied characteristics when used in different binding assays. These in vitro/ex vivo assessments suggest that some of the isolated mAbs might be promising candidates for targeting overexpressed CD117 in canine cancers for different useful applications.
Collapse
Affiliation(s)
- Mohamed A Alfaleh
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia.
- Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center (KFMRC), King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Neetika Arora
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Michael Yeh
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Christopher J de Bakker
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Christopher B Howard
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia.
- Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD 4072, Australia.
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Philip Macpherson
- School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia.
| | - Rachel E Allavena
- School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia.
| | - Xiaoli Chen
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Linda Harkness
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Stephen M Mahler
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia.
- Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Martina L Jones
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia.
- Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
33
|
The Role of Tumor Microenvironment and Impact of Cancer Stem Cells on Breast Cancer Progression and Growth. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2019. [DOI: 10.2478/sjecr-2018-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Breast cancer is not only a mass of genetically abnormal tissue in the breast. This is a well-organized system of a complex heterogeneous tissue. Cancer cells produce regulatory signals that stimulate stromal cells to proliferate and migrate; then, stromal elements respond to these signals by releasing components necessary for tumor development that provide structural support, vasculature, and extracellular matrices. Developing tumors can mobilize a variety of cell types from both local and distant niches via secret chemical factors derived from cancer cells themselves or neighboring cells disrupted by growing neoplasm, such as fibroblasts, immune inflammatory cells, and endothelial cells. CSCs are a group of very few cells that are tumorigenic (able to form tumors) and are defined as those cells within a tumor that can self-renew and lead to tumorigenesis. BCSCs represent a small population of cells that have stem cell characteristics and are related to breast cancer. There are different theories about the origin of BCSCs. BCSCs are responsible for breast carcinoma metastasis. Usually, there is a metastatic spread to the bones, and rarely to the lungs and liver. A phenomenon that allows BCSCs to make the transition from epithelial to mesenchymal expression and thus avoid the effect of cytotoxic agents is the epithelial-mesenchymal transition (EMT). During this process, cells change their molecular characteristics in terms of loss of epithelial characteristics taking the mesenchymal phenotype. This process plays a key role in the progression, invasion, and metastasis of breast tumors.
Collapse
|
34
|
Dowling M, Samuelson J, Fadl-Alla B, Pondenis HC, Byrum M, Barger AM, Fan TM. Overexpression of prostate specific membrane antigen by canine hemangiosarcoma cells provides opportunity for the molecular detection of disease burdens within hemorrhagic body cavity effusions. PLoS One 2019; 14:e0210297. [PMID: 30601866 PMCID: PMC6314605 DOI: 10.1371/journal.pone.0210297] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 12/18/2018] [Indexed: 12/22/2022] Open
Abstract
Background Canine hemangiosarcoma (cHSA) is a highly metastatic mesenchymal cancer that disseminates by hematogenous and direct implantation routes. Therapies for cHSA are generally ineffective, in part due to advanced clinical disease stage at the time of diagnosis. The validation of conventional molecular methods for detecting novel biomarkers preferentially expressed by cHSA could lead to more timely diagnosis, earlier therapeutic interventions, and improved outcomes. In humans, prostate-specific membrane antigen (PSMA) is a transmembrane protein overexpressed by prostate carcinoma and tumor-associated endothelium of various solid cancer histologies. Importantly, the preferential overexpression of PSMA by certain cancers has been leveraged for the development of diagnostic molecular imaging reagents and targeted therapeutics. Recently, PSMA has been qualitatively demonstrated to be expressed in cHSA cell lines, however, quantitative PSMA expressions and the potential utility of PSMA transcript identification in biologic fluids to support the presence of microscopic cHSA burden has not been reported. Therefore, this study sought to characterize the differential quantitative expressions of PSMA between cHSA and non-malignant tissues, and to determine the potential diagnostic utility of PCR-generated PSMA amplicons as a surrogate of rare cHSA cells dwelling within peritoneal and pericardial cavities. Methods Quantitative gene and protein expressions for PSMA were compared between one normal endothelial and six cHSA cell lines by RT-PCR, western blot analysis, and fluorescent microscopy. Additionally, gene and protein expressions of PSMA in normal canine tissues were characterized. Graded expressions of PSMA were determined in spontaneously-arising cHSA tumor samples and the feasibility of qualitative PCR as a molecular diagnostic to detect PSMA transcripts in whole blood from healthy dogs and hemorrhagic effusions from cHSA-bearing dogs were evaluated. Results PSMA gene and protein expressions were elevated (up to 6-fold) in cHSA cells compared with non-malignant endothelium. By immunohistochemistry, protein expressions of PSMA were detectable in all cHSA tissue samples evaluated. As predicted by human protein atlas data, PSMA’s expression was comparably identified at substantial levels in select normal canine tissues including kidney, liver, and intestine. In young healthy pet dogs, PSMA amplicons could not be identified in circulating whole blood yet were detectable in hemorrhagic effusions collected from pet dogs with confirmed cHSA or PSMA-expressing cancer. Conclusions PSMA is quantitatively overexpressed in cHSA compared to normal endothelium, but its protein expression is not restricted to only cHSA tumor tissues, as specific visceral organs also substantively express PSMA. Optimized qualitative PCR methods failed to amplify PSMA amplicons sufficiently for visible detection in circulating whole blood derived from healthy young dogs, yet PSMA transcripts were readily identifiable in hemorrhagic effusions collected from pet dogs with histologically confirmed cHSA or PSMA-expressing cancer. While preliminary, findings derived from a limited cohort of normal and diseased pet dogs provocatively raise the potential value of PSMA amplicon detection as an ancillary molecular diagnostic test for supporting the presence of microscopic cHSA disease burden within hemorrhagic body cavity effusions.
Collapse
Affiliation(s)
- Matthew Dowling
- Department of Veterinary Clinical Medicine, University of Illinois, Urbana, IL, United States of America
| | - Jonathan Samuelson
- Department of Pathobiology, University of Illinois, Urbana, IL, United States of America
| | - Bahaa Fadl-Alla
- Department of Pathobiology, University of Illinois, Urbana, IL, United States of America
| | - Holly C. Pondenis
- Department of Veterinary Clinical Medicine, University of Illinois, Urbana, IL, United States of America
| | - Mark Byrum
- Department of Veterinary Clinical Medicine, University of Illinois, Urbana, IL, United States of America
| | - Anne M. Barger
- Department of Veterinary Clinical Medicine, University of Illinois, Urbana, IL, United States of America
| | - Timothy M. Fan
- Department of Veterinary Clinical Medicine, University of Illinois, Urbana, IL, United States of America
- * E-mail:
| |
Collapse
|
35
|
Hedgespeth BA, Gal A. Pathology in Practice. J Am Vet Med Assoc 2018; 253:1545-1548. [PMID: 30668259 DOI: 10.2460/javma.253.12.1545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
36
|
Li R, Miao J, Tabaran AF, O’Sullivan MG, Anderson KJ, Scott PM, Wang Z, Cormier RT. A novel cancer syndrome caused by KCNQ1-deficiency in the golden Syrian hamster. J Carcinog 2018; 17:6. [PMID: 30450013 PMCID: PMC6187935 DOI: 10.4103/jcar.jcar_5_18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/31/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The golden Syrian hamster is an emerging model organism. To optimize its use, our group has made the first genetically engineered hamsters. One of the first genes that we investigated is KCNQ1 which encodes for the KCNQ1 potassium channel and also has been implicated as a tumor suppressor gene. MATERIALS AND METHODS We generated KCNQ1 knockout (KO) hamsters by CRISPR/Cas9-mediated gene targeting and investigated the effects of KCNQ1-deficiency on tumorigenesis. RESULTS By 70 days of age seven of the eight homozygous KCNQ1 KOs used in this study began showing signs of distress, and on necropsy six of the seven ill hamsters had visible cancers, including T-cell lymphomas, plasma cell tumors, hemangiosarcomas, and suspect myeloid leukemias. CONCLUSIONS None of the hamsters in our colony that were wild-type or heterozygous for KCNQ1 mutations developed cancers indicating that the cancer phenotype is linked to KCNQ1-deficiency. This study is also the first evidence linking KCNQ1-deficiency to blood cancers.
Collapse
Affiliation(s)
- Rong Li
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, USA
| | - Jinxin Miao
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, USA
| | - Alexandru-Flaviu Tabaran
- Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Minnesota, Duluth, MN, USA
- Comparative Pathology Shared Resource, Masonic Cancer Center, University of Minnesota, Duluth, MN, USA
| | - M. Gerard O’Sullivan
- Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Minnesota, Duluth, MN, USA
- Comparative Pathology Shared Resource, Masonic Cancer Center, University of Minnesota, Duluth, MN, USA
| | - Kyle J. Anderson
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, USA
| | - Patricia M. Scott
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, USA
| | - Zhongde Wang
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, USA
| | - Robert T. Cormier
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, USA
| |
Collapse
|
37
|
Abstract
Pet dogs are becoming increasingly recognized as a population with the potential to inform medical research through their treatment for a variety of maladies by veterinary health professionals. This is the basis of the One Health initiative, supporting the idea of collaboration between human and animal health researchers and clinicians to study spontaneous disease processes and treatment in animals to inform human health. Cancer is a major health burden in pet dogs, accounting for approximately 30% of deaths across breeds. As such, pet dogs with cancer are becoming increasingly recognized as a resource for studying the pharmacology and therapeutic potential of anticancer drugs and therapies under development. This was recently highlighted by a National Academy of Medicine Workshop on Comparative Oncology that took place in mid-2015 (http://www.nap.edu/21830). One component of cancer burden in dogs is their significantly higher incidence of sarcomas as compared to humans. This increased incidence led to canine osteosarcoma being an important component in the development of surgical approaches for osteosarcoma in children. Included in this review of sarcomas in dogs is a description of the incidence, pathology, molecular characteristics and previous translational therapeutic studies associated with these tumors. An understanding of the patho-physiological and molecular characteristics of these naturally occurring canine sarcomas holds great promise for effective incorporation into drug development schemas, for evaluation of target modulation or other pharmacodynamic measures associated with therapeutic response. These data could serve to supplement other preclinical data and bolster clinical investigations in tumor types for which there is a paucity of human patients for clinical trials.
Collapse
Affiliation(s)
- Daniel L Gustafson
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA; Flint Animal Cancer Center, Colorado State University, Fort Collins, CO 80523, USA; University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Dawn L Duval
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA; Flint Animal Cancer Center, Colorado State University, Fort Collins, CO 80523, USA; University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Daniel P Regan
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA; Flint Animal Cancer Center, Colorado State University, Fort Collins, CO 80523, USA; University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Douglas H Thamm
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA; Flint Animal Cancer Center, Colorado State University, Fort Collins, CO 80523, USA; University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
38
|
The Notch Pathway in Breast Cancer Progression. ScientificWorldJournal 2018; 2018:2415489. [PMID: 30111989 PMCID: PMC6077551 DOI: 10.1155/2018/2415489] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 05/10/2018] [Accepted: 06/12/2018] [Indexed: 12/29/2022] Open
Abstract
Objective Notch signaling pathway is a vital parameter of the mammalian vascular system. In this review, the authors summarize the current knowledge about the impact of the Notch signaling pathway in breast cancer progression and the therapeutic role of Notch's inhibition. Methods The available literature in MEDLINE, PubMed, and Scopus, regarding the role of the Notch pathway in breast cancer progression was searched for related articles from about 1973 to 2017 including terms such as “Notch,” “Breast Cancer,” and “Angiogenesis.” Results. Notch signaling controls the differentiation of breast epithelial cells during normal development. Studies confirm that the Notch pathway has a major participation in breast cancer progression through overexpression and/or abnormal genetic type expression of the notch receptors and ligands that determine angiogenesis. The cross-talk of Notch and estrogens, the effect of Notch in breast cancer stem cells formation, and the dependable Notch overexpression during breast tumorigenesis have been studied enough and undoubtedly linked to breast cancer development. The already applied therapeutic inhibition of Notch for breast cancer can drastically change the course of the disease. Conclusion Current data prove that Notch pathway has a major participation and multiple roles during breast tumor progression. Inhibition of Notch receptors and ligands provides innovative therapeutic results and could become the therapy of choice in the next few years, even though further research is needed to reach safe conclusions.
Collapse
|
39
|
Kim JH, Frantz AM, Sarver AL, Gorden Klukas BH, Lewellen M, O’Brien TD, Dickerson EB, Modiano JF. Modulation of fatty acid metabolism and immune suppression are features of in vitro tumour sphere formation in ontogenetically distinct dog cancers. Vet Comp Oncol 2018; 16:E176-E184. [PMID: 29152836 PMCID: PMC5821546 DOI: 10.1111/vco.12368] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/18/2017] [Accepted: 10/13/2017] [Indexed: 12/19/2022]
Abstract
Non-adherent, 3-dimensional sphere formation is used as an in vitro surrogate to evaluate cellular potential for tumour initiation and self-renewal. To determine if a shared molecular program underlies the capacity for sphere formation by cells originating from diverse tumour types, we characterized molecular and functional properties of 10 independent cell lines derived from 3 ontogenetically distinct dog cancers: hemangiosarcoma, osteosarcoma and glial brain tumours. Genome-wide gene expression profiling identified tumour-of-origin-dependent patterns of adjustment to sphere formation in a uniform culture condition. However, expression of the stem/progenitor markers CD34 and CD117, resistance to cytotoxic drugs and dye efflux (side population assays) showed no association with these gene expression profiles. Instead, primary sphere-forming capacity was inversely correlated with the ability to reform secondary spheres, regardless of tumour ontogeny. Primary sphere formation seemed to be proportional to the number of pre-existing cells with sphere-forming capacity in the cell lines. Cell lines where secondary sphere formation was more proficient than primary sphere formation showed enrichment of genes involved in fatty acid synthesis and immunosuppressive cytokines. In contrast, cell lines where secondary sphere formation was approximately equivalent to or less proficient than primary sphere formation showed upregulation of CD40 and enrichment of genes involved in fatty acid oxidation. Our data suggest that in vitro sphere formation is associated with upregulation of gene clusters involved in metabolic and immunosuppressive functions, which might be necessary for self-renewal and for tumour initiation and/or tumour propagation in vivo.
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
| | - Aric M. Frantz
- 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
| | - Aaron L. Sarver
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Brandi H. Gorden Klukas
- 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
| | - Mitzi Lewellen
- 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
| | - Timothy D. O’Brien
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Erin B. Dickerson
- 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
| | - Jaime F. Modiano
- 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
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
40
|
Maharani A, Aoshima K, Onishi S, Gulay KCM, Kobayashi A, Kimura T. Cellular atypia is negatively correlated with immunohistochemical reactivity of CD31 and vWF expression levels in canine hemangiosarcoma. J Vet Med Sci 2018; 80:213-218. [PMID: 29311493 PMCID: PMC5836755 DOI: 10.1292/jvms.17-0561] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Canine hemangiosarcoma (HSA) is one of the most common mesenchymal tumors in dogs. Its
high metastatic and growth rates are usually associated with poor prognosis. Neoplastic
cells of HSA can show various levels of cellular atypia in the same mass and may consist
of various populations at different differentiated stages. Up to present, however, there
is no report analyzing their differentiation states by comparing cellular atypia with
differentiation-related protein expressions. To evaluate whether cellular atypia can be
used as a differentiation marker in HSA, we analyzed correlation between cellular atypia
and intensities of CD31 and von Willebrand Factor (vWF) staining in HSA cases. We also
compared cellular atypia and expression levels of CD31 and vWF in each growth patterns.
Our results show that cellular atypia was negatively correlated to CD31 and vWF expression
levels but no significant correlation was found between growth patterns and cellular
atypia or CD31 and vWF expression levels. Our study suggests that cellular atypia is
useful for identifying differentiation levels in HSA cases. This study also provides
useful information to determine differentiation levels of cell populations within HSA
based only on morphological analysis, which will aid further HSA research such as
identifying undifferentiation markers of endothelial cells or finding undifferentiated
cell population in tissue sections.
Collapse
Affiliation(s)
- Aprilia Maharani
- Laboratory of Comparative Pathology, Department of Clinical Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido 060-0818, Japan
| | - Keisuke Aoshima
- Laboratory of Comparative Pathology, Department of Clinical Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido 060-0818, Japan
| | - Shinichi Onishi
- Laboratory of Comparative Pathology, Department of Clinical Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido 060-0818, Japan
| | - Kevin Christian Montecillo Gulay
- Laboratory of Comparative Pathology, Department of Clinical Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido 060-0818, Japan
| | - Atsushi Kobayashi
- Laboratory of Comparative Pathology, Department of Clinical Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido 060-0818, Japan
| | - Takashi Kimura
- Laboratory of Comparative Pathology, Department of Clinical Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido 060-0818, Japan
| |
Collapse
|
41
|
Wang G, Wu M, Maloneyhuss MA, Wojcik J, Durham AC, Mason NJ, Roth DB. Actionable mutations in canine hemangiosarcoma. PLoS One 2017; 12:e0188667. [PMID: 29190660 PMCID: PMC5708669 DOI: 10.1371/journal.pone.0188667] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/10/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Angiosarcomas (AS) are rare in humans, but they are a deadly subtype of soft tissue sarcoma. Discovery sequencing in AS, especially the visceral form, is hampered by the rarity of cases. Most diagnostic material exists as archival formalin fixed, paraffin embedded tissue which serves as a poor source of high quality DNA for genome-wide sequencing. We approached this problem through comparative genomics. We hypothesized that exome sequencing a histologically similar tumor, hemangiosarcoma (HSA), that occurs in approximately 50,000 dogs per year, may lead to the identification of potential oncogenic drivers and druggable targets that could also occur in angiosarcoma. METHODS Splenic hemangiosarcomas are common in dogs, which allowed us to collect a cohort of archived matched tumor and normal tissue samples suitable for whole exome sequencing. Mapping of the reads to the latest canine reference genome (Canfam3) demonstrated that >99% of the targeted exomal regions were covered, with >80% at 20X coverage and >90% at 10X coverage. RESULTS AND CONCLUSIONS Sequence analysis of 20 samples identified somatic mutations in PIK3CA, TP53, PTEN, and PLCG1, all of which correspond to well-known tumor drivers in human cancer, in more than half of the cases. In one case, we identified a mutation in PLCG1 identical to a mutation observed previously in this gene in human visceral AS. Activating PIK3CA mutations present novel therapeutic targets, and clinical trials of targeted inhibitors are underway in human cancers. Our results lay a foundation for similar clinical trials in canine HSA, enabling a precision medicine approach to this disease.
Collapse
Affiliation(s)
- Guannan Wang
- Department of Pathology and Laboratory Medicine, Raymond and Ruth Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Ming Wu
- Illumina, San Diego, CA, United States of America
| | - Martha A. Maloneyhuss
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - John Wojcik
- Department of Pathology and Laboratory Medicine, Raymond and Ruth Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Amy C. Durham
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Nicola J. Mason
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - David B. Roth
- Department of Pathology and Laboratory Medicine, Raymond and Ruth Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| |
Collapse
|
42
|
Alfaleh MA, Jones ML, Howard CB, Mahler SM. Strategies for Selecting Membrane Protein-Specific Antibodies using Phage Display with Cell-Based Panning. Antibodies (Basel) 2017; 6:E10. [PMID: 31548525 PMCID: PMC6698842 DOI: 10.3390/antib6030010] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/05/2017] [Accepted: 07/07/2017] [Indexed: 12/14/2022] Open
Abstract
Membrane proteins are attractive targets for monoclonal antibody (mAb) discovery and development. Although several approved mAbs against membrane proteins have been isolated from phage antibody libraries, the process is challenging, as it requires the presentation of a correctly folded protein to screen the antibody library. Cell-based panning could represent the optimal method for antibody discovery against membrane proteins, since it allows for presentation in their natural conformation along with the appropriate post-translational modifications. Nevertheless, screening antibodies against a desired antigen, within a selected cell line, may be difficult due to the abundance of irrelevant organic molecules, which can potentially obscure the antigen of interest. This review will provide a comprehensive overview of the different cell-based phage panning strategies, with an emphasis placed on the optimisation of four critical panning conditions: cell surface antigen presentation, non-specific binding events, incubation time, and temperature and recovery of phage binders.
Collapse
Affiliation(s)
- Mohamed A Alfaleh
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia.
- Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Martina L Jones
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia.
- Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Christopher B Howard
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia.
- Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, Queensland 4072, Australia.
- Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Stephen M Mahler
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia.
- Australian Research Council Training Centre for Biopharmaceutical Innovation, The University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
43
|
Ceciliani F, Roccabianca P, Giudice C, Lecchi C. Application of post-genomic techniques in dog cancer research. MOLECULAR BIOSYSTEMS 2017; 12:2665-79. [PMID: 27345606 DOI: 10.1039/c6mb00227g] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Omics techniques have been widely applied to veterinary science, although mostly on farm animal productions and infectious diseases. In canine oncology, on the contrary, the use of omics methodologies is still far behind. This review presents the most recent achievement in the application of postgenomic techniques, such as transcriptomics, proteomics, and metabolomics, to canine cancer research. The protocols to recover material suitable for omics analyses from formalin-fixed, paraffin-embedded tissues are presented, and omics applications for biomarker discovery and their potential for cancer diagnostics in veterinary medicine are highlighted.
Collapse
Affiliation(s)
- F Ceciliani
- Department of Veterinary Medicine, Università di Milano, Via Celoria 02, 20133 Milano, Italy.
| | - P Roccabianca
- Department of Veterinary Medicine, Università di Milano, Via Celoria 02, 20133 Milano, Italy.
| | - C Giudice
- Department of Veterinary Medicine, Università di Milano, Via Celoria 02, 20133 Milano, Italy.
| | - C Lecchi
- Department of Veterinary Medicine, Università di Milano, Via Celoria 02, 20133 Milano, Italy.
| |
Collapse
|
44
|
Im K, Graef AJ, Breen M, Lindblad-Toh K, Modiano JF, Kim JH. Interactions between CXCR4 and CXCL12 promote cell migration and invasion of canine hemangiosarcoma. Vet Comp Oncol 2017; 15:315-327. [PMID: 26337509 PMCID: PMC7199805 DOI: 10.1111/vco.12165] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 06/14/2015] [Accepted: 07/07/2015] [Indexed: 12/14/2022]
Abstract
The CXCR4/CXCL12 axis plays an important role in cell locomotion and metastasis in many cancers. In this study, we hypothesized that the CXCR4/CXCL12 axis promotes migration and invasion of canine hemangiosarcoma (HSA) cells. Transcriptomic analysis across 12 HSA cell lines and 58 HSA whole tumour tissues identified heterogeneous expression of CXCR4 and CXCL12, which was associated with cell movement. In vitro, CXCL12 promoted calcium mobilization, cell migration and invasion that were directly proportional to surface expression of CXCR4; furthermore, these responses proved sensitive to the CXCR4 antagonist, AMD3100, in HSA cell lines. These results indicate that CXCL12 potentiates migration and invasion of canine HSA cells through CXCR4 signalling. The direct relationship between these responses in HSA cells suggests that the CXCR4/CXCL12 axis contributes to HSA progression.
Collapse
Affiliation(s)
- KeumSoon Im
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, MN
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, MN
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Ashley J. Graef
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, MN
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, MN
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Matthew Breen
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, & Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, NC, USA
- Cancer Genetics Program, University of North Carolina Lineberger Comprehensive Cancer Center, Raleigh, NC, USA
| | - Kerstin Lindblad-Toh
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Science for Life Laboratory, Dept. of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Jaime F. Modiano
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, MN
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, MN
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN
- Stem Cell Institute, University of Minnesota, Minneapolis, MN
- Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Jong-Hyuk Kim
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, MN
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, MN
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| |
Collapse
|
45
|
Subramani R, Nandy SB, Pedroza DA, Lakshmanaswamy R. Role of Growth Hormone in Breast Cancer. Endocrinology 2017; 158:1543-1555. [PMID: 28379395 DOI: 10.1210/en.2016-1928] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/28/2017] [Indexed: 12/19/2022]
Abstract
Breast cancer is one of the most common cancers diagnosed in women. Approximately two-thirds of all breast cancers diagnosed are classified as hormone dependent, which indicates that hormones are the key factors that drive the growth of these breast cancers. Ovarian and pituitary hormones play a major role in the growth and development of normal mammary glands and breast cancer. In particular, the effect of the ovarian hormone estrogen has received much attention in regard to breast cancer. Pituitary hormones prolactin and growth hormone have also been associated with breast cancer. Although the role of these pituitary hormones in breast cancers has been studied, it has not been investigated extensively. In this review, we attempt to compile basic information from most of the currently available literature to understand and demonstrate the significance of growth hormone in breast cancer. Based on the available literature, it is clear that growth hormone plays a significant role in the development, progression, and metastasis of breast cancer by influencing tumor angiogenesis, stemness, and chemoresistance.
Collapse
Affiliation(s)
- Ramadevi Subramani
- Center of Emphasis in Cancer Research, Department of Biomedical Sciences MSB1, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas 79905
| | - Sushmita B Nandy
- Center of Emphasis in Cancer Research, Department of Biomedical Sciences MSB1, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas 79905
| | - Diego A Pedroza
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, Texas 79905
| | - Rajkumar Lakshmanaswamy
- Center of Emphasis in Cancer Research, Department of Biomedical Sciences MSB1, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas 79905
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, Texas 79905
| |
Collapse
|
46
|
Borgatti A, Koopmeiners JS, Sarver AL, Winter AL, Stuebner K, Todhunter D, Rizzardi AE, Henriksen JC, Schmechel S, Forster CL, Kim JH, Froelich J, Walz J, Henson MS, Breen M, Lindblad-Toh K, Oh F, Pilbeam K, Modiano JF, Vallera DA. Safe and Effective Sarcoma Therapy through Bispecific Targeting of EGFR and uPAR. Mol Cancer Ther 2017; 16:956-965. [PMID: 28193671 PMCID: PMC5418099 DOI: 10.1158/1535-7163.mct-16-0637] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 01/19/2017] [Accepted: 01/23/2017] [Indexed: 01/12/2023]
Abstract
Sarcomas differ from carcinomas in their mesenchymal origin. Therapeutic advancements have come slowly, so alternative drugs and models are urgently needed. These studies report a new drug for sarcomas that simultaneously targets both tumor and tumor neovasculature. eBAT is a bispecific angiotoxin consisting of truncated, deimmunized Pseudomonas exotoxin fused to EGF and the amino terminal fragment of urokinase. Here, we study the drug in an in vivo "ontarget" companion dog trial as eBAT effectively kills canine hemangiosarcoma and human sarcoma cells in vitro We reasoned the model has value due to the common occurrence of spontaneous sarcomas in dogs and a limited lifespan allowing for rapid accrual and data collection. Splenectomized dogs with minimal residual disease were given one cycle of eBAT followed by adjuvant doxorubicin in an adaptive dose-finding, phase I-II study of 23 dogs with spontaneous, stage I-II, splenic hemangiosarcoma. eBAT improved 6-month survival from <40% in a comparison population to approximately 70% in dogs treated at a biologically active dose (50 μg/kg). Six dogs were long-term survivors, living >450 days. eBAT abated expected toxicity associated with EGFR targeting, a finding supported by mouse studies. Urokinase plasminogen activator receptor and EGFR are targets for human sarcomas, so thorough evaluation is crucial for validation of the dog model. Thus, we validated these markers for human sarcoma targeting in the study of 212 human and 97 canine sarcoma samples. Our results support further translation of eBAT for human patients with sarcomas and perhaps other EGFR-expressing malignancies. Mol Cancer Ther; 16(5); 956-65. ©2017 AACR.
Collapse
Affiliation(s)
- Antonella Borgatti
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota.
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Joseph S Koopmeiners
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Aaron L Sarver
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Amber L Winter
- Clinical Investigation Center, College of Veterinary Medicine, St. Paul, Minnesota
| | - Kathleen Stuebner
- Clinical Investigation Center, College of Veterinary Medicine, St. Paul, Minnesota
| | - Deborah Todhunter
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Radiation Oncology, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Anthony E Rizzardi
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Jonathan C Henriksen
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Stephen Schmechel
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Colleen L Forster
- BioNet Histology Research Laboratory, Academic Health Center, University of Minnesota, Minneapolis, Minnesota
| | - Jong-Hyuk Kim
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Jerry Froelich
- Department of Radiology, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Jillian Walz
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
| | - Michael S Henson
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Matthew Breen
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina
- Cancer Genetics Program, University of North Carolina Lineberger Comprehensive Cancer Center, Raleigh, North Carolina
| | - Kerstin Lindblad-Toh
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Felix Oh
- Department of Radiation Oncology, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Kristy Pilbeam
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Jaime F Modiano
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota
| | - Daniel A Vallera
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Radiation Oncology, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
47
|
Borgatti A, Winter AL, Stuebner K, Scott R, Ober CP, Anderson KL, Feeney DA, Vallera DA, Koopmeiners JS, Modiano JF, Froelich J. Evaluation of 18-F-fluoro-2-deoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) as a staging and monitoring tool for dogs with stage-2 splenic hemangiosarcoma - A pilot study. PLoS One 2017; 12:e0172651. [PMID: 28222142 PMCID: PMC5319762 DOI: 10.1371/journal.pone.0172651] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 02/07/2017] [Indexed: 02/04/2023] Open
Abstract
Positron Emission Tomography-Computed Tomography (PET-CT) is routinely used for staging and monitoring of human cancer patients and is becoming increasingly available in veterinary medicine. In this study, 18-fluorodeoxyglucose (18FDG)-PET-CT was used in dogs with naturally occurring splenic hemangiosarcoma (HSA) to assess its utility as a staging and monitoring modality as compared to standard radiography and ultrasonography. Nine dogs with stage-2 HSA underwent 18FDG-PET-CT following splenectomy and prior to commencement of chemotherapy. Routine staging (thoracic radiography and abdominal ultrasonography) was performed prior to 18FDG-PET-CT in all dogs. When abnormalities not identified on routine tests were noted on 18FDG-PET-CT, owners were given the option to repeat a PET-CT following treatment with eBAT. A PET-CT scan was repeated on Day 21 in three dogs. Abnormalities not observed on conventional staging tools, and most consistent with malignant disease based on location, appearance, and outcome, were detected in two dogs and included a right atrial mass and a hepatic nodule, respectively. These lesions were larger and had higher metabolic activity on the second scans. 18FDG-PET-CT has potential to provide important prognostic information and influence treatment recommendations for dogs with stage-2 HSA. Additional studies will be needed to precisely define the value of this imaging tool for staging and therapy monitoring in dogs with this and other cancers.
Collapse
Affiliation(s)
- Antonella Borgatti
- Animal Cancer Care and Research (ACCR) Program, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Amber L. Winter
- Animal Cancer Care and Research (ACCR) Program, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
- Clinical Investigation Center, College of Veterinary Medicine, St. Paul, Minnesota, United States of America
| | - Kathleen Stuebner
- Animal Cancer Care and Research (ACCR) Program, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
- Clinical Investigation Center, College of Veterinary Medicine, St. Paul, Minnesota, United States of America
| | - Ruth Scott
- Clinical Investigation Center, College of Veterinary Medicine, St. Paul, Minnesota, United States of America
| | - Christopher P. Ober
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Kari L. Anderson
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Daniel A. Feeney
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Daniel A. Vallera
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Therapeutic Radiology, School of Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Joseph S. Koopmeiners
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jaime F. Modiano
- Animal Cancer Care and Research (ACCR) Program, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jerry Froelich
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
- Center for Clinical Imaging Research (CCIR) in Diagnostic Radiology, School of Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
48
|
Shima H, Yamada A, Ishikawa T, Endo I. Are breast cancer stem cells the key to resolving clinical issues in breast cancer therapy? Gland Surg 2017; 6:82-88. [PMID: 28210556 DOI: 10.21037/gs.2016.08.03] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Despite the dramatic advances in breast cancer treatment over the past two decades, it is still the most common malignancies in women. One of the reasons patients succumb to breast cancer is treatment resistance leading to metastasis and recurrence. Recently, cancer stem cells (CSCs) have been suggested as a cause of metastasis and recurrence in several cancers because of their unique characteristics, including self-renewal, pluripotency, and high proliferative ability. Increasing evidence has implicated breast cancer stem cells (BCSCs) as essential for tumor development, progression, recurrence, and treatment resistance. BCSCs exhibit resistance to treatment owing to several inter-related factors, including overexpression of ATP-binding cassette (ABC) transporters and increased aldehyde dehydrogenase (ALDH) activity, DNA repair, and reactive oxygen species (ROS) scavenging. In addition, the Notch, Hedgehog, and Wnt signaling pathways have been suggested as the major pathways involved in the self-renewal and differentiation of BCSCs. Despite growing evidence suggesting the importance of BCSCs in progression and metastasis, clear criteria for the identification of BCSCs in clinical practice have yet to be established. Several potential markers have been suggested, including CD44+/CD24-/low, ALDH1, EpCAM/ESA, and nestin; however, there is no standard method to detect BCSCs. Triple-negative breast cancer, which shows initial chemosensitivity, demonstrates worsened prognosis due to therapy resistance, which might be related to the presence of BCSCs. Several clinical trials aimed at the identification of BCSCs or the development of BCSC-targeted therapy are in progress. Determining the clinical relevance of BCSCs may provide clues for overcoming therapy resistance in breast cancer.
Collapse
Affiliation(s)
- Hidetaka Shima
- Department of Gastroenterological Surgery, Yokohama City University, Yokohama, Kanagawa 236-0004, Japan
| | - Akimitsu Yamada
- Department of Gastroenterological Surgery, Yokohama City University, Yokohama, Kanagawa 236-0004, Japan
| | - Takashi Ishikawa
- Department of Breast disease, Tokyo Medical University Hospital, Tokyo 160-0023, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University, Yokohama, Kanagawa 236-0004, Japan
| |
Collapse
|
49
|
Pérez-Martínez C, Regueiro-Purriños M, Fernández-Martínez B, Altónaga JR, Gonzalo-Orden JM, García-Iglesias MJ. Magnetic resonance imaging and immunohistochemistry of primary vertebral hemangiosarcoma in a dog and implications for diagnosis and therapy. THE CANADIAN VETERINARY JOURNAL = LA REVUE VETERINAIRE CANADIENNE 2016; 57:1247-1250. [PMID: 27928170 PMCID: PMC5109625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
A vertebral mass in a dog with an acute onset paraparesis was identified by magnetic resonance imaging. A poorly differentiated hemangiosarcoma was diagnosed by histopathology and immunohistochemistry. Endothelial nitric oxide synthase could be a new differential marker for poorly differentiated hemangiosarcoma in dogs. Immunohistochemical detection of p53 phosphorylated at Serine392, p53, CD117, and CD44 suggest targets for design of therapeutic strategies.
Collapse
|
50
|
Thamm K, Graupner S, Werner C, Huttner WB, Corbeil D. Monoclonal Antibodies 13A4 and AC133 Do Not Recognize the Canine Ortholog of Mouse and Human Stem Cell Antigen Prominin-1 (CD133). PLoS One 2016; 11:e0164079. [PMID: 27701459 PMCID: PMC5049760 DOI: 10.1371/journal.pone.0164079] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/19/2016] [Indexed: 02/07/2023] Open
Abstract
The pentaspan membrane glycoprotein prominin-1 (CD133) is widely used in medicine as a cell surface marker of stem and cancer stem cells. It has opened new avenues in stem cell-based regenerative therapy and oncology. This molecule is largely used with human samples or the mouse model, and consequently most biological tools including antibodies are directed against human and murine prominin-1. Although the general structure of prominin-1 including its membrane topology is conserved throughout the animal kingdom, its primary sequence is poorly conserved. Thus, it is unclear if anti-human and -mouse prominin-1 antibodies cross-react with their orthologs in other species, especially dog. Answering this issue is imperative in light of the growing number of studies using canine prominin-1 as an antigenic marker. Here, we address this issue by cloning the canine prominin-1 and use its overexpression as a green fluorescent protein fusion protein in Madin-Darby canine kidney cells to determine its immunoreactivity with antibodies against human or mouse prominin-1. We used immunocytochemistry, flow cytometry and immunoblotting techniques and surprisingly found no cross-species immunoreactivity. These results raise some caution in data interpretation when anti-prominin-1 antibodies are used in interspecies studies.
Collapse
Affiliation(s)
- Kristina Thamm
- Tissue Engineering Laboratories, Biotechnology Center (BIOTEC), Technische Universität Dresden, Dresden, Germany
| | - Sylvi Graupner
- Tissue Engineering Laboratories, Biotechnology Center (BIOTEC), Technische Universität Dresden, Dresden, Germany
| | - Carsten Werner
- DFG-Research Center and Cluster of Excellence for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- Institute for Biofunctional Polymer Materials, Leibniz Institute of Polymer Research Dresden, Dresden, Germany
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Denis Corbeil
- Tissue Engineering Laboratories, Biotechnology Center (BIOTEC), Technische Universität Dresden, Dresden, Germany
- DFG-Research Center and Cluster of Excellence for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- * E-mail:
| |
Collapse
|