1
|
Ruzzi F, Riccardo F, Conti L, Tarone L, Semprini MS, Bolli E, Barutello G, Quaglino E, Lollini PL, Cavallo F. Cancer vaccines: Target antigens, vaccine platforms and preclinical models. Mol Aspects Med 2024; 101:101324. [PMID: 39631227 DOI: 10.1016/j.mam.2024.101324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
This review provides a comprehensive overview of the evolving landscape of cancer vaccines, highlighting their potential to revolutionize tumor prevention. Building on the success of vaccines against virus-related cancers, such as HPV- and HBV-associated cervical and liver cancers, the current challenge is to extend these achievements to the prevention of non-viral tumors and the treatment of preneoplastic or early neoplastic lesions. This review analyzes the critical aspects of preventive anti-cancer vaccination, focusing on the choice of target antigens, the development of effective vaccine platforms and technologies, and the use of various model systems for preclinical testing, from laboratory rodents to companion animals.
Collapse
Affiliation(s)
- Francesca Ruzzi
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126, Bologna, Italy
| | - Federica Riccardo
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Laura Conti
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Lidia Tarone
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Maria Sofia Semprini
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126, Bologna, Italy
| | - Elisabetta Bolli
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Giuseppina Barutello
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Elena Quaglino
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Pier-Luigi Lollini
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126, Bologna, Italy; IRCCS Azienda Ospedaliera Universitaria di Bologna, 40138, Bologna, Italy.
| | - Federica Cavallo
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy.
| |
Collapse
|
2
|
Chirullo B, Fruscione F, Del Zotto G, Dell'Anno F, Tarantino M, Porcellato I, Petrucci P, De Ciucis CG, Bufalari A, Guardone L, Cappelli K, Moretti G, Mecocci S, Monti E, De Paolis L, Razzuoli E. Evaluation of attenuated Salmonella Typhimurium (STMΔznuABC) anticancer activity on canine mammary cancer-associated fibroblasts. Res Vet Sci 2024; 180:105438. [PMID: 39447298 DOI: 10.1016/j.rvsc.2024.105438] [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: 06/10/2024] [Revised: 09/23/2024] [Accepted: 10/13/2024] [Indexed: 10/26/2024]
Abstract
Bacteria-mediated treatments gained increasing attention as alternative therapies against tumors. An attenuated mutant strain of Salmonella enterica serovar Typhimurium (STMΔznuABC) has recently been considered as a potential new anti-cancer strategy. However, it is unclear whether this activity is tumor-induced or species-specific, and no data are available regarding STMΔznuABC on canine mammary tumors (CMTs). This study aimed to investigate the ability of STMΔznuABC in modulating the response of CMTs, focusing on cancer-associated fibroblasts. Four CMT cell lines (CF33, TM51, TM52 TM53) were treated with STMΔznuABC. Then, antiproliferative activity (MTT assay), bacterial invasion, and CMT cell lines gene expression analysis (RT-qPCR) of genes involved in immune response and cancer aggressiveness were evaluated. STMΔznuABC penetrated in TM51, TM52, TM53, and CF33 cell lines, causing a significant reduction of cell viability. Moreover, the expression of several genes was significantly modulated in all CMT cell lines: STMΔznuABC infection determined a significant up-regulation of CXCL8, IL18, IL10, TLR4 and RAD51, while CD14, IL6, CXCR4, P53, PTEN, STAT5, TLR5 and TGFB1 were downregulated in TM53. In CF33, CXCL8 and P53 were upregulated, while MYD88, MD2, IL18, TLR4,5, TGFB1 were downregulated. In TM52, CXCL8, CD44 and MD2 were upregulated and PTEN was downregulated, while in TM51 CXCL8, CD44 and ErbB2 were downregulated. We demonstrated the anti-proliferative and immuno-modulatory activity of STMΔznuABC in CMTs, paving the way for potential new anti-cancer treatments.
Collapse
Affiliation(s)
- Barbara Chirullo
- Unit of Emerging Zoonoses, Department of Food Safety, Nutrition and Veterinary Public Health Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Floriana Fruscione
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy.
| | | | - Filippo Dell'Anno
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy.
| | - Michela Tarantino
- Unit of Emerging Zoonoses, Department of Food Safety, Nutrition and Veterinary Public Health Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Ilaria Porcellato
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Paola Petrucci
- Unit of Emerging Zoonoses, Department of Food Safety, Nutrition and Veterinary Public Health Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Chiara Grazia De Ciucis
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy; Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Via Forlanini 2, 27100 Pavia, Italy.
| | - Antonello Bufalari
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Lisa Guardone
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy; Department of Veterinary Sciences, University of Pisa, Viale delle Piagge 2, 56124 Pisa, Italy.
| | - Katia Cappelli
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Giulia Moretti
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Samanta Mecocci
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Eleonora Monti
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Livia De Paolis
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy; Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Elisabetta Razzuoli
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, Piazza Borgo Pila 39/24, 16129 Genoa, Italy.
| |
Collapse
|
3
|
Lo Giudice A, Porcellato I, Giglia G, Sforna M, Lepri E, Mandara MT, Leonardi L, Mechelli L, Brachelente C. Exploring the Epidemiology of Melanocytic Tumors in Canine and Feline Populations: A Comprehensive Analysis of Diagnostic Records from a Single Pathology Institution in Italy. Vet Sci 2024; 11:435. [PMID: 39330814 PMCID: PMC11436034 DOI: 10.3390/vetsci11090435] [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: 07/18/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024] Open
Abstract
MTs are prevalent in dogs, representing the most frequent oral malignancy, compared to cats, in which ocular melanomas predominate. This study investigates the canine and feline MT epidemiology (2005-2024) of cases submitted to the Veterinary Pathology Service (University of Perugia). Among the canine neoplasms, 845 (4%) were melanocytic: 329 (39%) melanocytomas; 512 (61%) melanomas. Of these, 485 (57%) were cutaneous (4% of canine cutaneous neoplasms), 193 (23%) were oral (50% of oral canine neoplasms), and 104 (12%) were mucocutaneous. The average age of affected dogs was 10 years. Older dogs were more likely to have melanomas compared to melanocytomas (p < 0.001). There were 60 (1%) feline MTs: 6 (10%) melanocytomas; 53 (88%) melanomas. Of these, 29 (48%) were cutaneous (1% of feline cutaneous tumors), 18 (30%) were ocular, and 9 (15%) were oral (22% of feline oral tumors). The average age of affected cats was 11 years. In dogs, mucocutaneous melanomas were more common compared to cutaneous ones (p < 0.05); oral melanomas were more common compared to all other sites (p < 0.001). In cats, ocular melanomas were more common compared to cutaneous ones (p < 0.05). Our study provides the MT prevalence in a selected canine and feline population, revealing MT epidemiological patterns, highlighting species-specific differences in the tumor prevalence, localization, and age distribution.
Collapse
Affiliation(s)
| | - Ilaria Porcellato
- Department of Veterinary Medicine, University of Perugia, 06126 Perugia, Italy; (A.L.G.); (G.G.); (M.S.); (E.L.); (M.T.M.); (L.L.); (L.M.); (C.B.)
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Gola C, Maniscalco L, Iussich S, Morello E, Olimpo M, Martignani E, Accornero P, Giacobino D, Mazzone E, Modesto P, Varello K, Aresu L, De Maria R. Hypoxia-associated markers in the prognosis of oral canine melanoma. Vet Pathol 2024; 61:721-731. [PMID: 38613423 DOI: 10.1177/03009858241244853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2024]
Abstract
Canine oral malignant melanoma (COMM) is the most common neoplasm in the oral cavity characterized by local invasiveness and high metastatic potential. Hypoxia represents a crucial feature of the solid tumor microenvironment promoting cancer progression and drug resistance. Hypoxia-inducible factor-1α (HIF-1α) and its downstream effectors, vascular endothelial growth factor A (VEGF-A), glucose transporter isoform 1 (GLUT1), C-X-C chemokine receptor type 4 (CXCR4), and carbonic anhydrase IX (CAIX), are the main regulators of the adaptive response to low oxygen availability. The prognostic value of these markers was evaluated in 36 COMMs using immunohistochemistry. In addition, the effects of cobalt chloride-mediated hypoxia were evaluated in 1 primary COMM cell line. HIF-1α expression was observed in the nucleus, and this localization correlated with the presence or enhanced expression of HIF-1α-regulated genes at the protein level. Multivariate analysis revealed that in dogs given chondroitin sulfate proteoglycan-4 (CSPG4) DNA vaccine, COMMs expressing HIF-1α, VEGF-A, and CXCR4 were associated with shorter disease-free intervals (DFI) compared with tumors that were negative for these markers (P = .03), suggesting hypoxia can influence immunotherapy response. Western blotting showed that, under chemically induced hypoxia, COMM cells accumulate HIF-1α and smaller amounts of CAIX. HIF-1α induction and stabilization triggered by hypoxia was corroborated by immunofluorescence, showing its nuclear translocation. These findings reinforce the role of an hypoxic microenvironment in tumor progression and patient outcome in COMM, as previously established in several human and canine cancers. In addition, hypoxic markers may represent promising prognostic markers, highlighting opportunities for their use in therapeutic strategies for COMMs.
Collapse
Affiliation(s)
- Cecilia Gola
- University of Surrey, Guildford, UK
- University of Turin, Grugliasco, Turin, Italy
| | | | | | | | | | | | | | | | | | - Paola Modesto
- Istituto Zooprofilattico Sperimentale del Piemonte Liguria e Valle d'Aosta, Turin, Italy
| | - Katia Varello
- Istituto Zooprofilattico Sperimentale del Piemonte Liguria e Valle d'Aosta, Turin, Italy
| | - Luca Aresu
- University of Turin, Grugliasco, Turin, Italy
| | | |
Collapse
|
5
|
Wise C, Breen M, Stapleton HM. Canine on the Couch: The New Canary in the Coal Mine for Environmental Health Research. ENVIRONMENT & HEALTH (WASHINGTON, D.C.) 2024; 2:517-529. [PMID: 39170948 PMCID: PMC11334179 DOI: 10.1021/envhealth.4c00029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/10/2024] [Accepted: 05/12/2024] [Indexed: 08/23/2024]
Abstract
Human health is intimately connected and tied to the health of our environment and ecosystem, with only a very small fraction of the risk for chronic diseases explained by genetics alone. Companion animals are prone to disease types that are shared with people, including cancers and endocrine disorders, reinforcing the thought that environmental factors contribute to the risks for chronic diseases. These factors include air and water pollution and the built environment. As such, there is increasing interest in pursuing research with companion animals, and specifically dogs, as sentinel species to inform comparative health assessments and identify risk factors for disease. Of the canine diseases for which environmental exposure research has been published, cancers have received the most attention. This review summarizes two main aspects of this comparative approach: (1) cancers that occur in dogs and which are similar to humans and (2) research investigating environmental exposures and health outcomes in dogs. The goal of this review is to highlight the diverse conditions in which pet dogs may provide unique perspectives and advantages to examine relationships between environmental exposures and health outcomes, with an emphasis on chemical pollution and cancer. Furthermore, this review seeks to raise awareness and stimulate discussion around the best practices for the use of companion animals as environmental health sentinels.
Collapse
Affiliation(s)
- Catherine
F. Wise
- Nicholas
School of the Environment, Duke University, Durham, North Carolina 27708, United States
- Duke
Cancer Institute, Durham, North Carolina 27710, United States
| | - Matthew Breen
- Department
of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, Raleigh, North Carolina 27607, United States
- Comparative
Medicine Institute, North Carolina State
University, Raleigh, North Carolina 27607, United States
- Center
for Human Health and the Environment, North
Carolina State University, Raleigh, North Carolina 27607, United States
| | - Heather M. Stapleton
- Nicholas
School of the Environment, Duke University, Durham, North Carolina 27708, United States
- Duke
Cancer Institute, Durham, North Carolina 27710, United States
| |
Collapse
|
6
|
Lo Giudice A, Porcellato I, Pellegrini M, Rottenberg S, He C, Dentini A, Moretti G, Cagiola M, Mechelli L, Chiaradia E, Brachelente C. Establishment of Primary Cell Cultures from Canine Oral Melanomas via Fine-Needle Aspiration: A Novel Tool for Tumorigenesis and Cancer Progression Studies. Animals (Basel) 2024; 14:1948. [PMID: 38998060 PMCID: PMC11240394 DOI: 10.3390/ani14131948] [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: 04/05/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024] Open
Abstract
Oral melanomas are the most common oral malignancies in dogs and are characterized by an aggressive nature, invasiveness, and poor prognosis. With biological and genetic similarities to human oral melanomas, they serve as a valuable spontaneous comparative model. Primary cell cultures are widely used in human medicine and, more recently, in veterinary medicine to study tumorigenesis, cancer progression, and innovative therapeutic approaches. This study aims to establish two- and three-dimensional primary cell lines from oral canine melanomas using fine-needle aspiration as a minimally invasive sampling method. For this study, samples were collected from six dogs, represented by four primary oral melanomas and five lymph nodal metastases. The cells were digested to obtain single-cell suspensions, seeded in flasks, or processed with Matrigel® to form organoids. The cell cultures were characterized through flow cytometry using antibodies against Melan-A, PNL2, and Sox-10. This technique offers a minimally invasive means to obtain cell samples, particularly beneficial for patients that are ineligible for surgical procedures, and enables the establishment of in vitro models crucial for comparative studies in mucosal melanoma oncology. To the best of our knowledge, this is the first work establishing neoplastic primary cell cultures via fine-needle aspiration in dogs.
Collapse
Affiliation(s)
- Adriana Lo Giudice
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy; (A.L.G.); (G.M.); (L.M.); (E.C.); (C.B.)
| | - Ilaria Porcellato
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy; (A.L.G.); (G.M.); (L.M.); (E.C.); (C.B.)
| | - Martina Pellegrini
- Istituto Zooprofilattico Sperimentale dell’Umbria e delle Marche “Togo Rosati”, Via G. Salvemini 1, 06126 Perugia, Italy; (M.P.); (M.C.)
| | - Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Länggassstrasse 120, 3012 Bern, Switzerland; (S.R.); (C.H.)
| | - Chang He
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Länggassstrasse 120, 3012 Bern, Switzerland; (S.R.); (C.H.)
| | - Alfredo Dentini
- Clinica Veterinaria Tyrus, Strada delle Campore 30L, 05100 Terni, Italy;
| | - Giulia Moretti
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy; (A.L.G.); (G.M.); (L.M.); (E.C.); (C.B.)
| | - Monica Cagiola
- Istituto Zooprofilattico Sperimentale dell’Umbria e delle Marche “Togo Rosati”, Via G. Salvemini 1, 06126 Perugia, Italy; (M.P.); (M.C.)
| | - Luca Mechelli
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy; (A.L.G.); (G.M.); (L.M.); (E.C.); (C.B.)
| | - Elisabetta Chiaradia
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy; (A.L.G.); (G.M.); (L.M.); (E.C.); (C.B.)
| | - Chiara Brachelente
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy; (A.L.G.); (G.M.); (L.M.); (E.C.); (C.B.)
| |
Collapse
|
7
|
Ammons DT, Hopkins LS, Cronise KE, Kurihara J, Regan DP, Dow S. Single-cell RNA sequencing reveals the cellular and molecular heterogeneity of treatment-naïve primary osteosarcoma in dogs. Commun Biol 2024; 7:496. [PMID: 38658617 PMCID: PMC11043452 DOI: 10.1038/s42003-024-06182-w] [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: 11/06/2023] [Accepted: 04/10/2024] [Indexed: 04/26/2024] Open
Abstract
Osteosarcoma (OS) is a heterogeneous, aggressive malignancy of the bone that disproportionally affects children and adolescents. Therapeutic interventions for OS are limited, which is in part due to the complex tumor microenvironment (TME). As such, we used single-cell RNA sequencing (scRNA-seq) to describe the cellular and molecular composition of the TME in 6 treatment-naïve dogs with spontaneously occurring primary OS. Through analysis of 35,310 cells, we identified 41 transcriptomically distinct cell types including the characterization of follicular helper T cells, mature regulatory dendritic cells (mregDCs), and 8 tumor-associated macrophage (TAM) populations. Cell-cell interaction analysis predicted that mregDCs and TAMs play key roles in modulating T cell mediated immunity. Furthermore, we completed cross-species cell type gene signature homology analysis and found a high degree of similarity between human and canine OS. The data presented here act as a roadmap of canine OS which can be applied to advance translational immuno-oncology research.
Collapse
Affiliation(s)
- Dylan T Ammons
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.
| | - Leone S Hopkins
- Flint Animal Cancer Center, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Kathryn E Cronise
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Jade Kurihara
- Flint Animal Cancer Center, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Daniel P Regan
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
- Flint Animal Cancer Center, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Steven Dow
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.
- Flint Animal Cancer Center, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
8
|
Béguin J, Laloy E, Cochin S, Gantzer M, Farine I, Pichon C, Moreau B, Foloppe J, Balloul JM, Machon C, Guitton J, Tierny D, Klonjkowski B, Quéméneur E, Maurey C, Erbs P. Oncolytic virotherapy with intratumoral injection of vaccinia virus TG6002 and 5-fluorocytosine administration in dogs with malignant tumors. Mol Ther Oncolytics 2023; 30:103-116. [PMID: 37635744 PMCID: PMC10448017 DOI: 10.1016/j.omto.2023.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 07/17/2023] [Indexed: 08/29/2023] Open
Abstract
TG6002 is an oncolytic vaccinia virus expressing FCU1 protein, which converts 5-fluorocytosine into 5-fluorouracil. The study objectives were to assess tolerance, viral replication, 5-fluorouracil synthesis, and tumor microenvironment modifications to treatment in dogs with spontaneous malignant tumors. Thirteen dogs received one to three weekly intratumoral injections of TG6002 and 5-fluorocytosine. The viral genome was assessed in blood and tumor biopsies by qPCR. 5-Fluorouracil concentrations were measured in serum and tumor biopsies by liquid chromatography or high-resolution mass spectrometry. Histological and immunohistochemical analyses were performed. The viral genome was detected in blood (7/13) and tumor biopsies (4/11). Viral replication was suspected in 6/13 dogs. The median intratumoral concentration of 5-fluorouracil was 314 pg/mg. 5-Fluorouracil was not detected in the blood. An increase in necrosis (6/9) and a downregulation of intratumoral regulatory T lymphocytes (6/6) were observed. Viral replication, 5-fluorouracil synthesis, and tumor microenvironment changes were more frequently observed with higher TG6002 doses. This study confirmed the replicative properties, targeted chemotherapy synthesis, and reversion of the immunosuppressive tumor microenvironment in dogs with spontaneous malignant tumors treated with TG6002 and 5-fluorocytosine.
Collapse
Affiliation(s)
- Jérémy Béguin
- Transgene, 67405 Illkirch-Graffenstaden, France
- UMR Virologie, INRAE, École Nationale Vétérinaire d’Alfort, ANSES, Université Paris-Est, 94700 Maisons-Alfort, France
- Department of Internal Medicine, École Nationale Vétérinaire d’Alfort, Université Paris-Est, 94700 Maisons-Alfort, France
| | - Eve Laloy
- UMR Virologie, INRAE, École Nationale Vétérinaire d’Alfort, ANSES, Université Paris-Est, 94700 Maisons-Alfort, France
- Anatomical Pathology Unit, Biopôle, École Nationale Vétérinaire d’Alfort, Université Paris-Est, 94700 Maisons-Alfort, France
| | | | | | | | | | | | | | | | - Christelle Machon
- Service de Biochimie et pharmacotoxicologie, Hôpital Lyon-Sud, Hospices Civils de Lyon, 69310, France
| | - Jérôme Guitton
- Service de Biochimie et pharmacotoxicologie, Hôpital Lyon-Sud, Hospices Civils de Lyon, 69310, France
| | | | - Bernard Klonjkowski
- UMR Virologie, INRAE, École Nationale Vétérinaire d’Alfort, ANSES, Université Paris-Est, 94700 Maisons-Alfort, France
| | | | - Christelle Maurey
- Department of Internal Medicine, École Nationale Vétérinaire d’Alfort, Université Paris-Est, 94700 Maisons-Alfort, France
| | | |
Collapse
|
9
|
Marconato L, Tiraboschi L, Aralla M, Sabattini S, Melacarne A, Agnoli C, Balboni A, Salvi M, Foglia A, Punzi S, Romagnoli N, Rescigno M. A Phase 2, Single-Arm, Open-Label Clinical Trial on Adjuvant Peptide-Based Vaccination in Dogs with Aggressive Hemangiosarcoma Undergoing Surgery and Chemotherapy. Cancers (Basel) 2023; 15:4209. [PMID: 37686485 PMCID: PMC10486958 DOI: 10.3390/cancers15174209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/05/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
To test the antitumor effect and safety of peptide-based anticancer vaccination in dogs with hemangiosarcoma undergoing the standard of care (SOC; surgery and doxorubicin), canine hemangiosarcoma cells were infected with Salmonella typhi Ty21a to release immunogenic endoplasmic reticulum stress-related peptides into the extracellular milieu via CX43 hemichannels opening. The infected tumor cell secretome constituted the vaccine. Following the SOC, dogs with biologically aggressive hemangiosarcoma were vaccinated a total of five times, once every 3 weeks, and were followed up with serial imaging. A retrospective population of dogs undergoing the SOC alone served as controls. The primary endpoints were the time to progression (TTP) and overall survival (OS), and the secondary endpoints were toxicity and immune responses. A total of 28 dogs were vaccinated along with the SOC, and 32 received only the SOC. A tumor-specific humoral response along with a vaccine-specific T-cell response was observed. Toxicity did not occur. The TTP and OS were significantly longer in vaccinated versus unvaccinated dogs (TTP: 195 vs. 160 days, respectively; p = 0.001; OS: 276 vs. 175 days, respectively; p = 0.002). One-year survival rates were 35.7% and 6.3% for vaccinated and unvaccinated dogs, respectively. In dogs with hemangiosarcoma undergoing the SOC, the addition of a peptide-based vaccine increased the TTP and OS, while maintaining a safe profile. Moreover, vaccinated dogs developed a tumor-specific response, supporting the feasibility of future phase three studies.
Collapse
Affiliation(s)
- Laura Marconato
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Ozzano dell’Emilia, 40064 Bologna, Italy; (S.S.); (C.A.); (A.B.); (A.F.); (S.P.); (N.R.)
| | - Luca Tiraboschi
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (L.T.); (A.M.); (M.S.); (M.R.)
| | - Marina Aralla
- Pronto Soccorso Veterinario Laudense, 26900 Lodi, Italy;
| | - Silvia Sabattini
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Ozzano dell’Emilia, 40064 Bologna, Italy; (S.S.); (C.A.); (A.B.); (A.F.); (S.P.); (N.R.)
| | - Alessia Melacarne
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (L.T.); (A.M.); (M.S.); (M.R.)
| | - Chiara Agnoli
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Ozzano dell’Emilia, 40064 Bologna, Italy; (S.S.); (C.A.); (A.B.); (A.F.); (S.P.); (N.R.)
| | - Andrea Balboni
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Ozzano dell’Emilia, 40064 Bologna, Italy; (S.S.); (C.A.); (A.B.); (A.F.); (S.P.); (N.R.)
| | - Marta Salvi
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (L.T.); (A.M.); (M.S.); (M.R.)
| | - Armando Foglia
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Ozzano dell’Emilia, 40064 Bologna, Italy; (S.S.); (C.A.); (A.B.); (A.F.); (S.P.); (N.R.)
| | - Sofia Punzi
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Ozzano dell’Emilia, 40064 Bologna, Italy; (S.S.); (C.A.); (A.B.); (A.F.); (S.P.); (N.R.)
| | - Noemi Romagnoli
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Ozzano dell’Emilia, 40064 Bologna, Italy; (S.S.); (C.A.); (A.B.); (A.F.); (S.P.); (N.R.)
| | - Maria Rescigno
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (L.T.); (A.M.); (M.S.); (M.R.)
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Italy
| |
Collapse
|
10
|
Mannheimer JD, Tawa G, Gerhold D, Braisted J, Sayers CM, McEachron TA, Meltzer P, Mazcko C, Beck JA, LeBlanc AK. Transcriptional profiling of canine osteosarcoma identifies prognostic gene expression signatures with translational value for humans. Commun Biol 2023; 6:856. [PMID: 37591946 PMCID: PMC10435536 DOI: 10.1038/s42003-023-05208-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 08/03/2023] [Indexed: 08/19/2023] Open
Abstract
Canine osteosarcoma is increasingly recognized as an informative model for human osteosarcoma. Here we show in one of the largest clinically annotated canine osteosarcoma transcriptional datasets that two previously reported, as well as de novo gene signatures devised through single sample Gene Set Enrichment Analysis (ssGSEA), have prognostic utility in both human and canine patients. Shared molecular pathway alterations are seen in immune cell signaling and activation including TH1 and TH2 signaling, interferon signaling, and inflammatory responses. Virtual cell sorting to estimate immune cell populations within canine and human tumors showed similar trends, predominantly for macrophages and CD8+ T cells. Immunohistochemical staining verified the increased presence of immune cells in tumors exhibiting immune gene enrichment. Collectively these findings further validate naturally occurring osteosarcoma of the pet dog as a translationally relevant patient model for humans and improve our understanding of the immunologic and genomic landscape of the disease in both species.
Collapse
Affiliation(s)
- Joshua D Mannheimer
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Gregory Tawa
- Division of Preclinical Innovation, Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - David Gerhold
- Division of Preclinical Innovation, Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - John Braisted
- Division of Preclinical Innovation, Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Carly M Sayers
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Troy A McEachron
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Paul Meltzer
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christina Mazcko
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jessica A Beck
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amy K LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
11
|
Ammons D, Hopkins L, Cronise K, Kurihara J, Regan D, Dow S. Single-cell RNA sequencing reveals the cellular and molecular heterogeneity of treatment-naïve primary osteosarcoma in dogs. RESEARCH SQUARE 2023:rs.3.rs-3232360. [PMID: 37609233 PMCID: PMC10441479 DOI: 10.21203/rs.3.rs-3232360/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Osteosarcoma (OS) is a heterogeneous, aggressive malignancy of the bone that disproportionally affects children and adolescents. Therapeutic interventions for OS are limited, which is in part due to the complex tumor microenvironment (TME) that has proven to be refractory to immunotherapies. Thus, there is a need to better define the complexity of the OS TME. To address this need, we used single-cell RNA sequencing (scRNA-seq) to describe the cellular and molecular composition of the TME in 6 treatment-naïve dogs with spontaneously occurring primary OS. Through analysis of 35,310 cells, we identified 30 distinct immune cell types, 9 unique tumor populations, 1 cluster of fibroblasts, and 1 cluster of endothelial cells. Independent reclustering of major cell types revealed the presence of follicular helper T cells, mature regulatory dendritic cells (mregDCs), and 8 transcriptomically distinct macrophage/monocyte populations. Cell-cell interaction inference analysis predicted that mregDCs and tumor-associated macrophages (TAMs) play key roles in modulating T cell mediate immunity. Furthermore, we used publicly available human OS scRNA-seq data to complete a cross-species cell type gene signature homology analysis. The analysis revealed a high degree of cell type gene signature homology between species, suggesting the cellular composition of OS is largely conserved between humans and dogs. Our findings provide key new insights into the biology of canine OS and highlight the conserved features of OS across species. Generally, the data presented here acts as a cellular and molecular roadmap of canine OS which can be applied to advance the translational immuno-oncology research field.
Collapse
|
12
|
Lundstrom K. Viral vectors engineered for gene therapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 379:1-41. [PMID: 37541721 DOI: 10.1016/bs.ircmb.2023.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/06/2023]
Abstract
Gene therapy has seen major progress in recent years. Viral vectors have made a significant contribution through efficient engineering for improved delivery and safety. A large variety of indications such as cancer, cardiovascular, metabolic, hematological, neurological, muscular, ophthalmological, infectious diseases, and immunodeficiency have been targeted. Viral vectors based on adenoviruses, adeno-associated viruses, herpes simplex viruses, retroviruses including lentiviruses, alphaviruses, flaviviruses, measles viruses, rhabdoviruses, Newcastle disease virus, poxviruses, picornaviruses, reoviruses, and polyomaviruses have been used. Proof-of-concept has been demonstrated for different indications in animal models. Therapeutic efficacy has also been achieved in clinical trials. Several viral vector-based drugs have been approved for the treatment of cancer, and hematological, metabolic, and neurological diseases. Moreover, viral vector-based vaccines have been approved against COVID-19 and Ebola virus disease.
Collapse
|
13
|
Lundstrom K. Alphaviruses in cancer immunotherapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 379:143-168. [PMID: 37541722 DOI: 10.1016/bs.ircmb.2023.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/06/2023]
Abstract
Alphaviruses have frequently been engineered for cancer therapy, cancer immunotherapy, and cancer vaccine development. As members of self-replicating RNA viruses, alphaviruses provide high levels of transgene expression through efficient self-amplifying of their RNA genome in host cells. Alphavirus vectors can be used as recombinant viral particles or oncolytic viruses. Alternatively, either naked or nanoparticle-encapsulated RNA and DNA replicons can be utilized. In the context of cancer prevention and treatment, antitumor, cytotoxic and suicide genes have been expressed from alphavirus vectors to provide tumor regression and tumor eradication. Moreover, immunostimulatory genes such as cytokines and chemokines have been used for cancer immunotherapy approaches. Expression of tumor antigens has been applied for cancer vaccine development. Alphavirus vectors has demonstrated tumor regression and even cure in various preclinical animal models. Immunization has elicited strong immune responses and showed protection against challenges with tumor cells in animal models. Several clinical trials have confirmed good safety and tolerability of alphaviruses in cancer patients although therapeutic efficacy will still require optimization.
Collapse
|
14
|
Yang Y, Mei C, Xian H, Zhang X, Li J, Liang ZX, Zhi Y, Ma Y, Wang HJ. Toosendanin-induced apoptosis of CMT-U27 is mediated through the mitochondrial apoptotic pathway. Vet Comp Oncol 2023; 21:315-326. [PMID: 36809669 DOI: 10.1111/vco.12889] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 02/08/2023] [Accepted: 02/08/2023] [Indexed: 02/24/2023]
Abstract
Toosendanin (TSN) is an active compound from the fruit of Melia toosendan Sieb et Zucc. TSN has been shown to have broad-spectrum anti-tumour activities in human cancers. However, there are still many gaps in the knowledge of TSN on canine mammary tumours (CMT). CMT-U27 cells were used to select the optimal acting time and best concentration of TSN to initiate apoptosis. Cell proliferation, cell colony formation, cell migration and cell invasion were analysed. The expression of apoptosis-related genes and proteins were also detected to explore the mechanism of action of TSN. A murine tumour model was established to detect the effect of TSN treatments. The results showed that TSN decreased cell viability of migration and invasion, altered CMT-U27 cell morphology, and inhibited DNA synthesis. TSN-induced cell apoptosis by upregulating BAX, cleaved caspase-3, cleaved caspase-9, p53 and cytochrome C (cytosolic) protein expression, and downregulating Bcl-2 and cytochrome C (mitochondrial) expression. In addition, TSN increased the mRNA transcription levels of cytochrome C, p53 and BAX, and decreased the mRNA expression of Bcl-2. Furthermore, TSN inhibited the growth of CMT xenografts by regulating the expression of genes and proteins activated by the mitochondrial apoptotic pathway. In conclusion, TSN effectively inhibited cell proliferation, migration and invasion activity, as well as induced CMT-U27 cell apoptosis. The study provides a molecular basis for the development of clinical drugs and other therapeutic options.
Collapse
Affiliation(s)
- Yin Yang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Municipal Academy of Agriculture and Forestry, Beijing, China
- School of Veterinary Medicine, Southwest University, Rongchang Chongqing, China
| | - Chen Mei
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Municipal Academy of Agriculture and Forestry, Beijing, China
| | - Hong Xian
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Municipal Academy of Agriculture and Forestry, Beijing, China
| | - Xue Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Municipal Academy of Agriculture and Forestry, Beijing, China
| | - Jun Li
- School of Veterinary Medicine, Southwest University, Rongchang Chongqing, China
| | - Zhi-Xuan Liang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Municipal Academy of Agriculture and Forestry, Beijing, China
| | - Yan Zhi
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Municipal Academy of Agriculture and Forestry, Beijing, China
| | - Yue Ma
- School of Veterinary Medicine, Southwest University, Rongchang Chongqing, China
| | - Hong-Jun Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Municipal Academy of Agriculture and Forestry, Beijing, China
| |
Collapse
|
15
|
Dall'Ara P, Filipe J, Pilastro C, Turin L, Lauzi S, Gariboldi EM, Stefanello D. Can Chemotherapy Negatively Affect the Specific Antibody Response toward Core Vaccines in Canine Cancer Patients? Vet Sci 2023; 10:vetsci10040303. [PMID: 37104458 PMCID: PMC10143758 DOI: 10.3390/vetsci10040303] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/10/2023] [Accepted: 04/18/2023] [Indexed: 04/28/2023] Open
Abstract
The life expectancy of our pets has been getting longer in recent years due to new therapeutic opportunities, better nutrition, and better diagnostic approaches. This positive effect, however, has been accompanied by a concomitant increase in neoplasms, particularly in canine patients. Therefore, veterinarians inevitably face new issues related to these diseases, poorly or never investigated in the past, such as the possible side effects resulting from chemotherapy. The aim of this study was to investigate whether and how chemotherapy influences the antibody response against CPV-2, CDV, and CAdV-1 in dogs vaccinated before starting chemotherapy. Twenty-one canine patients with different types of malignancies were sampled before, during, and after different chemotherapy protocols to determine their actual levels of seroprotection against CPV-2, CDV, and CadV-1 by using the in-practice test VacciCheck. Differences related to sex, breed size, type of tumor, and chemotherapy protocol were evaluated. No statistically significant changes in antibody protection emerged for any of the chemotherapy protocol used, suggesting that, contrary to expectation, chemotherapy does not have a marked immunosuppressive effect on the post-vaccine antibody response. These results, although preliminary, may be useful in improving the clinical approach to the canine cancer patient, helping veterinarians fully manage their patients, and enabling owners to feel more confident about their pets' quality of life.
Collapse
Affiliation(s)
- Paola Dall'Ara
- Department of Veterinary Medicine and Animal Sciences (DIVAS), University of Milan, Via Dell'Università 6, 26900 Lodi, Italy
| | - Joel Filipe
- Department of Veterinary Medicine and Animal Sciences (DIVAS), University of Milan, Via Dell'Università 6, 26900 Lodi, Italy
| | - Chiara Pilastro
- Department of Veterinary Medicine and Animal Sciences (DIVAS), University of Milan, Via Dell'Università 6, 26900 Lodi, Italy
| | - Lauretta Turin
- Department of Veterinary Medicine and Animal Sciences (DIVAS), University of Milan, Via Dell'Università 6, 26900 Lodi, Italy
| | - Stefania Lauzi
- Department of Veterinary Medicine and Animal Sciences (DIVAS), University of Milan, Via Dell'Università 6, 26900 Lodi, Italy
| | - Elisa Maria Gariboldi
- Department of Veterinary Medicine and Animal Sciences (DIVAS), University of Milan, Via Dell'Università 6, 26900 Lodi, Italy
| | - Damiano Stefanello
- Department of Veterinary Medicine and Animal Sciences (DIVAS), University of Milan, Via Dell'Università 6, 26900 Lodi, Italy
| |
Collapse
|
16
|
Tarone L, Mareschi K, Tirtei E, Giacobino D, Camerino M, Buracco P, Morello E, Cavallo F, Riccardo F. Improving Osteosarcoma Treatment: Comparative Oncology in Action. LIFE (BASEL, SWITZERLAND) 2022; 12:life12122099. [PMID: 36556464 PMCID: PMC9783386 DOI: 10.3390/life12122099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
Osteosarcoma (OSA) is the most common pediatric malignant bone tumor. Although surgery together with neoadjuvant/adjuvant chemotherapy has improved survival for localized OSA, most patients develop recurrent/metastatic disease with a dismally poor outcome. Therapeutic options have not improved for these OSA patients in recent decades. As OSA is a rare and "orphan" tumor, with no distinct targetable driver antigens, the development of new efficient therapies is still an unmet and challenging clinical need. Appropriate animal models are therefore critical for advancement in the field. Despite the undoubted relevance of pre-clinical mouse models in cancer research, they present some intrinsic limitations that may be responsible for the low translational success of novel therapies from the pre-clinical setting to the clinic. From this context emerges the concept of comparative oncology, which has spurred the study of pet dogs as a uniquely valuable model of spontaneous OSA that develops in an immune-competent system with high biological and clinical similarities to corresponding human tumors, including in its metastatic behavior and resistance to conventional therapies. For these reasons, the translational power of studies conducted on OSA-bearing dogs has seen increasing recognition. The most recent and relevant veterinary investigations of novel combinatorial approaches, with a focus on immune-based strategies, that can most likely benefit both canine and human OSA patients have been summarized in this commentary.
Collapse
Affiliation(s)
- Lidia Tarone
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - Katia Mareschi
- Department of Public Health and Paediatrics, University of Torino, Piazza Polonia 94, 10126 Torino, Italy
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Department, Regina Margherita Children’s Hospital, City of Health and Science of Torino, 10126 Torino, Italy
| | - Elisa Tirtei
- Department of Public Health and Paediatrics, University of Torino, Piazza Polonia 94, 10126 Torino, Italy
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Department, Regina Margherita Children’s Hospital, City of Health and Science of Torino, 10126 Torino, Italy
| | - Davide Giacobino
- Department of Veterinary Sciences, University of Torino, Largo Paolo Braccini 2, Grugliasco, 10095 Torino, Italy
| | - Mariateresa Camerino
- Department of Veterinary Sciences, University of Torino, Largo Paolo Braccini 2, Grugliasco, 10095 Torino, Italy
| | - Paolo Buracco
- Department of Veterinary Sciences, University of Torino, Largo Paolo Braccini 2, Grugliasco, 10095 Torino, Italy
| | - Emanuela Morello
- Department of Veterinary Sciences, University of Torino, Largo Paolo Braccini 2, Grugliasco, 10095 Torino, Italy
| | - Federica Cavallo
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
- Correspondence: (F.C.); (F.R.)
| | - Federica Riccardo
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
- Correspondence: (F.C.); (F.R.)
| |
Collapse
|
17
|
Dhawan D, Ramos-Vara JA, Utturkar SM, Ruple A, Tersey SA, Nelson JB, Cooper B, Heng HG, Ostrander EA, Parker HG, Hahn NM, Adams LG, Fulkerson CM, Childress MO, Bonney P, Royce C, Fourez LM, Enstrom AW, Ambrosius LA, Knapp DW. Identification of a naturally-occurring canine model for early detection and intervention research in high grade urothelial carcinoma. Front Oncol 2022; 12:1011969. [PMID: 36439482 PMCID: PMC9692095 DOI: 10.3389/fonc.2022.1011969] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/24/2022] [Indexed: 09/23/2023] Open
Abstract
Background Early detection and intervention research is expected to improve the outcomes for patients with high grade muscle invasive urothelial carcinoma (InvUC). With limited patients in suitable high-risk study cohorts, relevant animal model research is critical. Experimental animal models often fail to adequately represent human cancer. The purpose of this study was to determine the suitability of dogs with high breed-associated risk for naturally-occurring InvUC to serve as relevant models for early detection and intervention research. The feasibility of screening and early intervention, and similarities and differences between canine and human tumors, and early and later canine tumors were determined. Methods STs (n=120) ≥ 6 years old with no outward evidence of urinary disease were screened at 6-month intervals for 3 years with physical exam, ultrasonography, and urinalysis with sediment exam. Cystoscopic biopsy was performed in dogs with positive screening tests. The pathological, clinical, and molecular characteristics of the "early" cancer detected by screening were determined. Transcriptomic signatures were compared between the early tumors and published findings in human InvUC, and to more advanced "later" canine tumors from STs who had the typical presentation of hematuria and urinary dysfunction. An early intervention trial of an oral cyclooxygenase inhibitor, deracoxib, was conducted in dogs with cancer detected through screening. Results Biopsy-confirmed bladder cancer was detected in 32 (27%) of 120 STs including InvUC (n=29, three starting as dysplasia), grade 1 noninvasive cancer (n=2), and carcinoma in situ (n=1). Transcriptomic signatures including druggable targets such as EGFR and the PI3K-AKT-mTOR pathway, were very similar between canine and human InvUC, especially within luminal and basal molecular subtypes. Marked transcriptomic differences were noted between early and later canine tumors, particularly within luminal subtype tumors. The deracoxib remission rate (42% CR+PR) compared very favorably to that with single-agent cyclooxygenase inhibitors in more advanced canine InvUC (17-25%), supporting the value of early intervention. Conclusions The study defined a novel naturally-occurring animal model to complement experimental models for early detection and intervention research in InvUC. Research incorporating the canine model is expected to lead to improved outcomes for humans, as well as pet dogs, facing bladder cancer.
Collapse
Affiliation(s)
- Deepika Dhawan
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - José A. Ramos-Vara
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
- Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | - Sagar M. Utturkar
- Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | - Audrey Ruple
- Purdue University Center for Cancer Research, West Lafayette, IN, United States
- Department of Public Health, College of Health and Human Sciences, Purdue University, West Lafayette, IN, United States
| | - Sarah A. Tersey
- Department of Medicine, Section of Endocrinology, Metabolism, and Diabetes, University of Chicago, Chicago, IL, United States
| | - Jennifer B. Nelson
- Department of Medicine, Section of Endocrinology, Metabolism, and Diabetes, University of Chicago, Chicago, IL, United States
| | - Bruce R. Cooper
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, United States
| | - Hock Gan Heng
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Elaine A. Ostrander
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Heidi G. Parker
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Noah M. Hahn
- Department of Oncology and Urology, Johns Hopkins University School of Medicine, and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, United States
| | - Larry G. Adams
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Christopher M. Fulkerson
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Michael O. Childress
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Patty L. Bonney
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Christine Royce
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Lindsey M. Fourez
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Alexander W. Enstrom
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Lisbeth A. Ambrosius
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Deborah W. Knapp
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
- Purdue University Center for Cancer Research, West Lafayette, IN, United States
| |
Collapse
|
18
|
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: 5] [Impact Index Per Article: 2.5] [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
|
19
|
Riccardo F, Tarone L, Camerino M, Giacobino D, Iussich S, Barutello G, Arigoni M, Conti L, Bolli E, Quaglino E, Merighi IF, Morello E, Dentini A, Ferrone S, Buracco P, Cavallo F. Antigen mimicry as an effective strategy to induce CSPG4-targeted immunity in dogs with oral melanoma: a veterinary trial. J Immunother Cancer 2022; 10:e004007. [PMID: 35580930 PMCID: PMC9114861 DOI: 10.1136/jitc-2021-004007] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Melanoma is the most lethal form of skin cancer in humans. Conventional therapies have limited efficacy, and overall response is still unsatisfactory considering that immune checkpoint inhibitors induce lasting clinical responses only in a low percentage of patients. This has prompted us to develop a vaccination strategy employing the tumor antigen chondroitin sulfate proteoglycan (CSPG)4 as a target. METHODS To overcome the host's unresponsiveness to the self-antigen CSPG4, we have taken advantage of the conservation of CSPG4 sequence through phylogenetic evolution, so we have used a vaccine, based on a chimeric DNA molecule encompassing both human (Hu) and dog (Do) portions of CSPG4 (HuDo-CSPG4). We have tested its safety and immunogenicity (primary objectives), along with its therapeutic efficacy (secondary outcome), in a prospective, non-randomized, veterinary clinical trial enrolling 80 client-owned dogs with surgically resected, CSPG4-positive, stage II-IV oral melanoma. RESULTS Vaccinated dogs developed anti-Do-CSPG4 and Hu-CSPG4 immune response. Interestingly, the antibody titer in vaccinated dogs was significantly associated with the overall survival. Our data suggest that there may be a contribution of the HuDo-CSPG4 vaccination to the improvement of survival of vaccinated dogs as compared with controls treated with conventional therapies alone. CONCLUSIONS HuDo-CSPG4 adjuvant vaccination was safe and immunogenic in dogs with oral melanoma, with potential beneficial effects on the course of the disease. Thanks to the power of naturally occurring canine tumors as predictive models for cancer immunotherapy response, these data may represent a basis for the translation of this approach to the treatment of human patients with CSPG4-positive melanoma subtypes.
Collapse
Affiliation(s)
- Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Lidia Tarone
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | | | - Davide Giacobino
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Selina Iussich
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Maddalena Arigoni
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Elisabetta Bolli
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Irene Fiore Merighi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Emanuela Morello
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | | | - Soldano Ferrone
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Paolo Buracco
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| |
Collapse
|
20
|
Lenz JA, Assenmacher CA, Costa V, Louka K, Rau S, Keuler NS, Zhang PJ, Maki RG, Durham AC, Radaelli E, Atherton MJ. Increased tumor-infiltrating lymphocyte density is associated with favorable outcomes in a comparative study of canine histiocytic sarcoma. Cancer Immunol Immunother 2022; 71:807-818. [PMID: 34415404 PMCID: PMC8858331 DOI: 10.1007/s00262-021-03033-z] [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: 06/28/2020] [Accepted: 08/12/2021] [Indexed: 12/18/2022]
Abstract
Histiocytic sarcoma (HS) is a rare and aggressive tumor in humans with no universally agreed standard of care therapy. Spontaneous canine HS exhibits increased prevalence in specific breeds, shares key genetic and biologic similarities with the human disease, and occurs in an immunocompetent setting. Previous data allude to the immunogenicity of this disease in both species, highlighting the potential for their successful treatment with immunotherapy. Quantification of CD3 tumor-infiltrating lymphocytes (TIL) in five cases of human HS revealed variable intra-tumoral T cell infiltration. Due to the paucity of human cases and lack of current model systems in which to appraise associations between anti-tumor immunity and treatment-outcome in HS, we analyzed clinical data and quantified TIL in 18 dogs that were previously diagnosed with localized HS and treated with curative-intent tumor resection with or without adjuvant chemotherapy. As in humans, assessment of TIL in biopsy tissues taken at diagnosis reveal a spectrum of immunologically "cold" to "hot" tumors. Importantly, we show that increased CD3 and granzyme B TIL are positively associated with favorable outcomes in dogs following surgical resection. NanoString transcriptional analyses revealed increased T cell and antigen presentation transcripts associated with prolonged survival in canine pulmonary HS and a decreased tumor immunogenicity profile associated with shorter survivals in splenic HS. Based on these findings, we propose that spontaneous canine HS is an accessible and powerful novel model to study tumor immunology and will provide a unique platform to preclinically appraise the efficacy and tolerability of anti-cancer immunotherapies for HS.
Collapse
Affiliation(s)
- Jennifer A Lenz
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Charles-Antoine Assenmacher
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Victoria Costa
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Katie Louka
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Suzanne Rau
- Metropolitan Veterinary Associates, Norristown, PA, USA
| | - Nicholas S Keuler
- Department of Statistics, University of Wisconsin-Madison, Madison, WI, USA
| | - Paul J Zhang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert G Maki
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy C Durham
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Enrico Radaelli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew J Atherton
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
21
|
A Target Animal Effectiveness Study on Adjuvant Peptide-Based Vaccination in Dogs with Non-Metastatic Appendicular Osteosarcoma Undergoing Amputation and Chemotherapy. Cancers (Basel) 2022; 14:cancers14051347. [PMID: 35267655 PMCID: PMC8909565 DOI: 10.3390/cancers14051347] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/27/2022] [Accepted: 03/04/2022] [Indexed: 01/12/2023] Open
Abstract
Simple Summary Despite efforts to develop novel treatment strategies, human and canine osteosarcomas continue to have limited overall survival. Spontaneous canine osteosarcoma shares many molecular similarities with humans, and shows the same aggressive disease course, thereby rendering the dog an effective model for the human disease equivalent. In both species, surgery followed by chemotherapy represents the gold standard treatment. Immunotherapy represents a promising treatment modality. A peptide-based anticancer vaccine was administered to 20 dogs with non-metastatic osteosarcoma as an add-on therapy to standard treatment consisting of limb amputation and adjuvant chemotherapy. Endpoints were to evaluate the efficacy and safety of this combined therapeutic approach. By using a bacterial-based strategy for vaccine development, we report an efficacious induction of an immune response, ultimately translating in improved outcome compared with historical controls receiving standard-of-care treatment. The results of this clinical trial provide promising potential for future management in both humans and dogs with osteosarcoma. Abstract Despite efforts to develop novel treatment strategies, human and canine osteosarcomas continue to have poor prognosis and limited overall survival. The aim of this clinical trial was to test the antitumor effect and safety of multiple dermal administrations of a peptide-based anticancer vaccine in dogs with non-metastatic appendicular osteosarcoma undergoing standard of care (SOC), consisting of limb amputation and adjuvant chemotherapy. Salmonella-infected canine osteosarcoma cells were induced to release immunogenic peptides in the extracellular space via Cx43 hemichannels opening; the secretome was collected and constituted the vaccine. Dogs with non-metastatic appendicular osteosarcoma were eligible for recruitment. Following limb amputation and adjuvant carboplatin, dogs were vaccinated on a monthly basis for six times and followed up with serial thoracic radiographs. A population of dogs undergoing SOC treatment (amputation and adjuvant carboplatin) before the vaccine was available served as controls. Primary endpoints were time to metastasis (TTM) and tumor-specific survival (TSS). Secondary endpoints were feasibility, toxicity, T-cell and humoral immune responses. A total of 20 dogs were vaccinated along with SOC and 34 received SOC only. Vaccine-specific humoral and T-cell responses were observed; their amplitude correlated with TSS. Vaccine-associated toxicity was not recorded. TTM and TSS were significantly longer in vaccinated versus unvaccinated dogs (TTM: 308 vs. 240 days, respectively; p = 0.010; TSS: 621 vs. 278 days, respectively; p = 0.002). In dogs with non-metastatic osteosarcoma undergoing SOC, the addition of a bacteria-based vaccination strategy increased TTM, thereby prolonging survival, while maintaining a safe profile. Additionally, vaccinated dogs developed a long-term tumor-specific response, as documented by the immunomonitoring of these patients over time. These results hold promise for future management of canine osteosarcoma.
Collapse
|
22
|
Camerino M, Giacobino D, Manassero L, Iussich S, Riccardo F, Cavallo F, Tarone L, Olimpo M, Lardone E, Martano M, Del Magno S, Buracco P, Morello E. Prognostic impact of bone invasion in canine oral malignant melanoma treated by surgery and anti-CSPG4 vaccination: A retrospective study on 68 cases (2010-2020). Vet Comp Oncol 2022; 20:189-197. [PMID: 34392602 PMCID: PMC9290081 DOI: 10.1111/vco.12761] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 05/17/2021] [Accepted: 08/12/2021] [Indexed: 12/15/2022]
Abstract
Prognosis of canine oral malignant melanoma encompasses clinical, histological and immunohistochemical parameters. The aim of this study was to evaluate the prognostic impact of bone invasion in oral canine melanoma. Sixty-eight dogs bearing oral melanoma staged II and III that underwent surgery and anti-CSPG4 electrovaccination, with available histological data and a minimum follow up of minimum 1 year, were retrospectively selected. Bone invasion was detected on imaging and/or histology. Median survival time of dogs with evidence of bone invasion (group 1) was 397 days and significantly shorter compared with dogs with oral melanomas not invading the bone (group 2, 1063 days). Dogs with tumours localised at the level of the cheek, lip, tongue and soft palate (soft tissue - group 3) lived significantly longer compared with dogs having tumours within the gingiva of the maxilla or mandible (hard tissue - group 4) with a median survival time of 1063 and 470 days, respectively. Within group 4, the subgroup of dogs with tumours not invading the bone (group 5) showed a significant prolonged survival time (972 days) in comparison with dogs of group 1 (bone invasion group). Similar results were obtained for the disease-free intervals amongst the different groups. Statistical analysis showed that Ki67 and mitotic count were correlated with shorter survival in patients of group 1 (with bone invasion). Bone invasion should always be assessed since it appears to be a negative prognostic factor.
Collapse
Affiliation(s)
| | - Davide Giacobino
- Department of Veterinary SciencesUniversity of TorinoTorinoItaly
| | - Luca Manassero
- Department of Veterinary SciencesUniversity of TorinoTorinoItaly
| | - Selina Iussich
- Department of Veterinary SciencesUniversity of TorinoTorinoItaly
| | - Federica Riccardo
- Department of Molecular Biotechnology and Health SciencesMolecular Biotechnology Center, University of TorinoTorinoItaly
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health SciencesMolecular Biotechnology Center, University of TorinoTorinoItaly
| | - Lidia Tarone
- Department of Molecular Biotechnology and Health SciencesMolecular Biotechnology Center, University of TorinoTorinoItaly
| | - Matteo Olimpo
- Department of Veterinary SciencesUniversity of TorinoTorinoItaly
| | - Elena Lardone
- Department of Veterinary SciencesUniversity of TorinoTorinoItaly
| | - Marina Martano
- Department of Medical Veterinary ScienceUniversity of ParmaParmaItaly
| | - Sara Del Magno
- Department of Veterinary Medical SciencesUniversity of BolognaOzzano dell'EmiliaItaly
| | - Paolo Buracco
- Department of Veterinary SciencesUniversity of TorinoTorinoItaly
| | - Emanuela Morello
- Department of Veterinary SciencesUniversity of TorinoTorinoItaly
| |
Collapse
|
23
|
Tarone L, Giacobino D, Camerino M, Ferrone S, Buracco P, Cavallo F, Riccardo F. Canine Melanoma Immunology and Immunotherapy: Relevance of Translational Research. Front Vet Sci 2022; 9:803093. [PMID: 35224082 PMCID: PMC8873926 DOI: 10.3389/fvets.2022.803093] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/10/2022] [Indexed: 11/17/2022] Open
Abstract
In veterinary oncology, canine melanoma is still a fatal disease for which innovative and long-lasting curative treatments are urgently required. Considering the similarities between canine and human melanoma and the clinical revolution that immunotherapy has instigated in the treatment of human melanoma patients, special attention must be paid to advancements in tumor immunology research in the veterinary field. Herein, we aim to discuss the most relevant knowledge on the immune landscape of canine melanoma and the most promising immunotherapeutic approaches under investigation. Particular attention will be dedicated to anti-cancer vaccination, and, especially, to the encouraging clinical results that we have obtained with DNA vaccines directed against chondroitin sulfate proteoglycan 4 (CSPG4), which is an appealing tumor-associated antigen with a key oncogenic role in both canine and human melanoma. In parallel with advances in therapeutic options, progress in the identification of easily accessible biomarkers to improve the diagnosis and the prognosis of melanoma should be sought, with circulating small extracellular vesicles emerging as strategically relevant players. Translational advances in melanoma management, whether achieved in the human or veterinary fields, may drive improvements with mutual clinical benefits for both human and canine patients; this is where the strength of comparative oncology lies.
Collapse
Affiliation(s)
- Lidia Tarone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Davide Giacobino
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | | | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Paolo Buracco
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| |
Collapse
|
24
|
Razmara AM, Judge SJ, Gingrich AA, Cruz SM, Culp WTN, Kent MS, Rebhun RB, Canter RJ. Natural Killer and T Cell Infiltration in Canine Osteosarcoma: Clinical Implications and Translational Relevance. Front Vet Sci 2021; 8:771737. [PMID: 34869744 PMCID: PMC8635198 DOI: 10.3389/fvets.2021.771737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/08/2021] [Indexed: 11/29/2022] Open
Abstract
Metastatic osteosarcoma has a bleak prognosis in both humans and dogs, and there have been minimal therapeutic advances in recent decades to improve outcomes. Naturally occurring osteosarcoma in dogs is shown to be a highly suitable model for human osteosarcoma, and limited data suggest the similarities between species extend into immune responses to cancer. Studies show that immune infiltrates in canine osteosarcoma resemble those of human osteosarcoma, and the analysis of tumor immune constituents as predictors of therapeutic response is a promising direction for future research. Additionally, clinical studies in dogs have piloted the use of NK transfer to treat osteosarcoma and can serve as valuable precursors to clinical trials in humans. Cytotoxic lymphocytes in dogs and humans with osteosarcoma have increased activation and exhaustion markers within tumors compared with blood. Accordingly, NK and T cells have complex interactions among cancer cells and other immune cells, which can lead to changes in pathways that work both for and against the tumor. Studies focused on NK and T cell interactions within the tumor microenvironment can open the door to targeted therapies, such as checkpoint inhibitors. Specifically, PD-1/PD-L1 checkpoint expression is conserved across tumors in both species, but further characterization of PD-L1 in canine osteosarcoma is needed to assess its prognostic significance compared with humans. Ultimately, a comparative understanding of T and NK cells in the osteosarcoma tumor microenvironment in both dogs and humans can be a platform for translational studies that improve outcomes in both dogs and humans with this frequently aggressive disease.
Collapse
Affiliation(s)
- Aryana M Razmara
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Sean J Judge
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Alicia A Gingrich
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Sylvia M Cruz
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
| | - William T N Culp
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Michael S Kent
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Robert B Rebhun
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Robert J Canter
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
25
|
Giacobino D, Camerino M, Riccardo F, Cavallo F, Tarone L, Martano M, Dentini A, Iussich S, Lardone E, Franci P, Valazza A, Manassero L, Del Magno S, De Maria R, Morello E, Buracco P. Difference in outcome between curative intent vs marginal excision as a first treatment in dogs with oral malignant melanoma and the impact of adjuvant CSPG4-DNA electrovaccination: A retrospective study on 155 cases. Vet Comp Oncol 2021; 19:651-660. [PMID: 33751759 PMCID: PMC9290641 DOI: 10.1111/vco.12690] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/26/2021] [Accepted: 03/05/2021] [Indexed: 12/28/2022]
Abstract
Canine oral malignant melanoma is locally invasive and highly metastatic. At present, the best option for local control is en bloc excision followed by radiation if excision margins are incomplete. Adjuvantly, the role of chemotherapy is dubious while immunotherapy appears encouraging. This retrospective study evaluated 155 dogs with oral malignant melanomas (24 stage I, 54 stage II, 66 stage III and 11 stage IV) managed in a single institution. The aim was to evaluate the differences in median survival time (MST) and disease-free interval (DFI) between dogs which, at presentation, were treated surgically with a curative intent (group 1) vs those marginally excised only (group 2). MST in group 1 was longer than in group 2 (594 vs 458 days), but no significant difference was found (P = .57); a statistical difference was, however, found for DFI (232 vs 183 days, P = .008). In the subpopulation of vaccinated dogs, the impact of adjuvant anti-CSPG4 DNA electrovaccination was then evaluated (curative intent, group 3, vs marginal, group 4); a significant difference for both MST (1333 vs 470 days, respectively, P = .03) and DFI (324 vs 184 days, respectively, P = .008) was found. Progressive disease was significantly more common in dogs undergoing marginal excision than curative intent excision for both the overall population (P = .03) and the vaccinated dogs (P = .02). This study pointed out that, after staging, wide excision together with adjuvant immunotherapy was an effective approach for canine oral malignant melanoma.
Collapse
Affiliation(s)
- Davide Giacobino
- Department of Veterinary SciencesUniversity of TorinoTorinoItaly
| | | | - Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoTorinoItaly
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoTorinoItaly
| | - Lidia Tarone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoTorinoItaly
| | - Marina Martano
- Department of Medical Veterinary ScienceUniversity of ParmaItaly
| | | | - Selina Iussich
- Department of Veterinary SciencesUniversity of TorinoTorinoItaly
| | - Elena Lardone
- Department of Veterinary SciencesUniversity of TorinoTorinoItaly
| | - Paolo Franci
- Department of Veterinary SciencesUniversity of TorinoTorinoItaly
| | - Alberto Valazza
- Department of Veterinary SciencesUniversity of TorinoTorinoItaly
| | - Luca Manassero
- Department of Veterinary SciencesUniversity of TorinoTorinoItaly
| | - Sara Del Magno
- Department of Veterinary Medical ScienceUniversity of BolognaItaly
| | | | - Emanuela Morello
- Department of Veterinary SciencesUniversity of TorinoTorinoItaly
| | - Paolo Buracco
- Department of Veterinary SciencesUniversity of TorinoTorinoItaly
| |
Collapse
|
26
|
Nanobody-based CTLA4 inhibitors for immune checkpoint blockade therapy of canine cancer patients. Sci Rep 2021; 11:20763. [PMID: 34675296 PMCID: PMC8531395 DOI: 10.1038/s41598-021-00325-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/06/2021] [Indexed: 12/16/2022] Open
Abstract
Cancer is the leading cause of death in the geriatric dog population. Currently, the use of immune checkpoint inhibitors (ICIs) such as anti-CTLA4 antibodies has markedly improved the prognosis of several cancers in their advanced stages. However, ICIs targeting CTLA4 blockade to treat canine cancer patients are yet to define. In this study, we sought to develop, characterize and assess whether chimeric heavy chain only antibodies (cHcAbs) against CTLA4 are viable therapeutic candidates for the treatment of canine cancers. Anti-CTLA4 nanobodies (Nbs) were identified from a yeast nanobody (Nb) library using magnetic-assisted cell sorting (MACS) and flow cytometry. cHcAbs were engineered by genetically fusing the DNA sequences coding for anti-CTLA4 Nbs with the Fc domain of the subclass B of canine IgG. Recombinant cHcAbs were purified from ExpiCHO-S cells. Stable cell lines expressing canine CTLA4 and FcγRI were used to elucidate the binding ability and specificity of cHcAbs. PBMCs isolated from healthy dogs were used to evaluate the ability of cHcAbs to activate canine PBMCs (cPBMCs). Novel Nbs were identified using the extracellular domain of canine CTLA4 protein to screen a fully synthetic yeast nanobody library. Purified Nbs bind specifically to natïve canine CTLA4. We report that chimeric HcAbs, which were engineered by fusing the anti-CTLA4 Nbs and Fc region of subclass B of canine IgG, were half the size of a conventional mAb and formed dimers. The chimeric HcAbs specifically binds both with canine CTLA4 and Fcγ receptors. As the binding of Nbs overlapped with the MYPPPY motif of canine CTLA4, these Nbs were expected to sterically disrupt the interaction of canine CTLA4 to B-7s. Like their human counterpart, canine CTLA4 was expressed on helper T cells and a small subset of cytotoxic T cells. Canine Tregs also constitutively expressed CTLA4, and stimulation with PMA/Ionomycin dramatically increased expression of CTLA4 on the cell surface. Stimulation of cPBMCs in the presence of agonistic anti-CD3 Ab and cHcAb6 significantly increased the expression of IFN-γ as compared to the isotype control. This study identifies a novel nanobody-based CTLA4 inhibitor for the treatment of canine cancer patients.
Collapse
|
27
|
Li Y, Wang G, Griffin L, Banda NK, Saba LM, Groman EV, Scheinman R, Moghimi SM, Simberg D. Complement opsonization of nanoparticles: Differences between humans and preclinical species. J Control Release 2021; 338:548-556. [PMID: 34481928 DOI: 10.1016/j.jconrel.2021.08.048] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/25/2021] [Accepted: 08/30/2021] [Indexed: 12/19/2022]
Abstract
The complement system plays a key role in opsonization and immune clearance of engineered nanoparticles. Understanding the efficiency, inter-subject, and inter-strain differences of complement opsonization in preclinical species can help with translational nanomedicine development and improve our ability to model complement response in humans. Dextran-coated superparamagnetic iron oxide (SPIO) nanoparticles and a wide range of non-magnetic iron oxide nanoparticle formulations are widely used in magnetic resonance imaging and as clinically approved iron supplements. Previously we found that opsonization of SPIO nanoworms (NW) with the third complement protein (C3) proceeds mostly via the alternative pathway in humans, and via the lectin pathway in mice. Here, we studied the pathway and efficiency of opsonization of 106 nm SPIO NW with C3 in different preclinical species and commonly used laboratory strains. In sera of healthy human donors (n = 6), C3 opsonization proceeded exclusively through the alternative pathway. On the other hand, the C3 opsonization in dogs (6 breeds), rats (4 strains) and mice (5 strains) sera was either partially or completely dependent on the complement Ca2+-sensitive pathways (lectin and/or classical). Specifically, C3 opsonization in sera of Long Evans rat strain, and mouse strains widely used in nanomedicine research (BALB/c, C57BL/6 J, and A/J) was only through the Ca2+-dependent pathways. Dogs and humans had the highest between-subject variability in C3 opsonization levels, while rat and mouse sera showed the lowest between-strain variability. Furthermore, using a panel of SPIO nanoparticles of different sizes and dextran coatings, we found that the level of C3 opsonization (C3 molecules per milligram Fe) in human sera was lower than in animal sera. At the same time, there was a strong predictive value of complement opsonization in dog and rat sera; nanoparticles with higher C3 deposition in animals showed higher deposition in humans, and vice versa. Notably, the opsonization decreased with decreasing size in all sera. The studies highlight the importance of the consideration of species and strains for predicting human complement responses (opsonization) towards nanomedicines.
Collapse
Affiliation(s)
- Yue Li
- Translational Bio-Nanosciences Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Guankui Wang
- Translational Bio-Nanosciences Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lynn Griffin
- Department of Environmental and Radiological Health Sciences, Veterinary Teaching Hospital, Colorado State University, Fort Collins, CO, USA
| | - Nirmal K Banda
- Division of Rheumatology, School of Medicine, University of Colorado Anschutz Medical Campus, 1775 Aurora Court, Aurora, CO, USA
| | - Laura M Saba
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ernest V Groman
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Robert Scheinman
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - S Moein Moghimi
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; School of Pharmacy, King George VI Building, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; Translational and Clinical Research Institute, Framlington Place, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Dmitri Simberg
- Translational Bio-Nanosciences Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
28
|
Lin Z, Wu Z, Luo W. Chimeric Antigen Receptor T-Cell Therapy: The Light of Day for Osteosarcoma. Cancers (Basel) 2021; 13:cancers13174469. [PMID: 34503279 PMCID: PMC8431424 DOI: 10.3390/cancers13174469] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/24/2021] [Accepted: 08/28/2021] [Indexed: 01/14/2023] Open
Abstract
Simple Summary As a novel immunotherapy, chimeric antigen receptor (CAR) T-cell therapy has achieved encouraging results in leukemia and lymphoma. Furthermore, CAR-T cells have been explored in the treatment of osteosarcoma (OS). However, there is no strong comprehensive evidence to support their efficacy. Therefore, we reviewed the current evidence on CAR-T cells for OS to demonstrate their feasibility and provide new options for the treatment of OS. Abstract Osteosarcoma (OS) is the most common malignant bone tumor, arising mainly in children and adolescents. With the introduction of multiagent chemotherapy, the treatments of OS have remarkably improved, but the prognosis for patients with metastases is still poor, with a five-year survival rate of 20%. In addition, adverse effects brought by traditional treatments, including radical surgery and systemic chemotherapy, may seriously affect the survival quality of patients. Therefore, new treatments for OS await exploitation. As a novel immunotherapy, chimeric antigen receptor (CAR) T-cell therapy has achieved encouraging results in treating cancer in recent years, especially in leukemia and lymphoma. Furthermore, researchers have recently focused on CAR-T therapy in solid tumors, including OS. In this review, we summarize the safety, specificity, and clinical transformation of the targets in treating OS and point out the direction for further research.
Collapse
|
29
|
Mestrinho LA, Santos RR. Translational oncotargets for immunotherapy: From pet dogs to humans. Adv Drug Deliv Rev 2021; 172:296-313. [PMID: 33705879 DOI: 10.1016/j.addr.2021.02.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/10/2021] [Accepted: 02/27/2021] [Indexed: 12/21/2022]
Abstract
Preclinical studies in rodent models have been a pivotal role in human clinical research, but many of them fail in the translational process. Spontaneous tumors in pet dogs have the potential to bridge the gap between preclinical models and human clinical trials. Their natural occurrence in an immunocompetent system overcome the limitations of preclinical rodent models. Due to its reasonable cellular, molecular, and genetic homology to humans, the pet dog represents a valuable model to accelerate the translation of preclinical studies to clinical trials in humans, actually with benefits for both species. Moreover, their unique genetic features of breeding and breed-related mutations have contributed to assess and optimize therapeutics in individuals with different genetic backgrounds. This review aims to outline four main immunotherapy approaches - cancer vaccines, adaptive T-cell transfer, antibodies, and cytokines -, under research in veterinary medicine and how they can serve the clinical application crosstalk with humans.
Collapse
|
30
|
Characterization of Primary Cultures of Normal and Neoplastic Canine Melanocytes. Animals (Basel) 2021; 11:ani11030768. [PMID: 33802040 PMCID: PMC7998744 DOI: 10.3390/ani11030768] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Melanoma is one of the most aggressive cancers in humans, with high rates of metastasis and a poor prognosis. Because of its environmental, biological and genetic features, numerous studies indicate the dog as a good comparative model for human melanoma. Primary cell cultures of healthy and neoplastic melanocytes derived from skin and oral mucosa of dogs with spontaneous tumors are established in this study. This model could represent a suitable tool to compare biological and molecular features of normal and neoplastic melanocytes from the same patient, to investigate the pathways underlying the oncogenic transformation, and to apply a more personalized therapeutic strategy. The cell cultures also meet international guidelines that encourage the use of alternative models to animal ones for the study of oncological diseases. Abstract Although numerous animal models, especially mouse models, have been established for the study of melanoma, they often fail to accurately describe the mechanisms of human disease because of their anatomic, physiological, and immune differences. The dog, as a spontaneous model of melanoma, is nowadays considered one of the most valid alternatives due to the heterogeneity of clinical presentations and of histological and genetic similarities of canine melanoma with the human counterpart. The aim of the study was to optimize a protocol for the isolation and cultivation of healthy and neoplastic canine melanocytes derived from the same animal and obtained from cutaneous and mucosal (oral) sites. We obtained five primary tumor cell cultures (from 2 cutaneous melanoma, 2 mucosal melanoma and 1 lymph node metastasis) and primary normal melanocyte cell cultures (from normal skin and mucosa) from the same dogs. Immunocytochemical characterization with Melan A, PNL2 and S100 antibodies confirmed the melanocytic origin of the cells. This work contributes to expanding the case record of studies on canine melanoma cell cultures as suitable model to study human and canine melanoma. To the authors’ knowledge, this is the first report of isolation of normal skin and mucosal canine melanocytes.
Collapse
|
31
|
Serkova NJ, Glunde K, Haney CR, Farhoud M, De Lille A, Redente EF, Simberg D, Westerly DC, Griffin L, Mason RP. Preclinical Applications of Multi-Platform Imaging in Animal Models of Cancer. Cancer Res 2021; 81:1189-1200. [PMID: 33262127 PMCID: PMC8026542 DOI: 10.1158/0008-5472.can-20-0373] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/10/2020] [Accepted: 11/25/2020] [Indexed: 11/16/2022]
Abstract
In animal models of cancer, oncologic imaging has evolved from a simple assessment of tumor location and size to sophisticated multimodality exploration of molecular, physiologic, genetic, immunologic, and biochemical events at microscopic to macroscopic levels, performed noninvasively and sometimes in real time. Here, we briefly review animal imaging technology and molecular imaging probes together with selected applications from recent literature. Fast and sensitive optical imaging is primarily used to track luciferase-expressing tumor cells, image molecular targets with fluorescence probes, and to report on metabolic and physiologic phenotypes using smart switchable luminescent probes. MicroPET/single-photon emission CT have proven to be two of the most translational modalities for molecular and metabolic imaging of cancers: immuno-PET is a promising and rapidly evolving area of imaging research. Sophisticated MRI techniques provide high-resolution images of small metastases, tumor inflammation, perfusion, oxygenation, and acidity. Disseminated tumors to the bone and lung are easily detected by microCT, while ultrasound provides real-time visualization of tumor vasculature and perfusion. Recently available photoacoustic imaging provides real-time evaluation of vascular patency, oxygenation, and nanoparticle distributions. New hybrid instruments, such as PET-MRI, promise more convenient combination of the capabilities of each modality, enabling enhanced research efficacy and throughput.
Collapse
Affiliation(s)
- Natalie J Serkova
- Department of Radiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
- Animal Imaging Shared Resource, University of Colorado Cancer Center, Aurora, Colorado
| | - Kristine Glunde
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology, and the Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Chad R Haney
- Center for Advanced Molecular Imaging, Northwestern University, Evanston, Illinois
| | | | | | | | - Dmitri Simberg
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - David C Westerly
- Animal Imaging Shared Resource, University of Colorado Cancer Center, Aurora, Colorado
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Lynn Griffin
- Department of Radiology, Veterinary Teaching Hospital, Colorado State University, Fort Collins, Colorado
| | - Ralph P Mason
- Department of Radiology, University of Texas Southwestern, Dallas, Texas
| |
Collapse
|
32
|
Nemec PS, Holmes JC, Hess PR. Dog leukocyte antigen-88*034:01 presents nonamer peptides from canine distemper virus hemagglutinin, large polymerase, and matrix proteins. HLA 2021; 97:428-434. [PMID: 33527745 DOI: 10.1111/tan.14197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 12/30/2022]
Abstract
Canine spontaneous cancers may offer greater fidelity than rodent models in advancing clinical immunotherapies. Boxers in particular are distinguished as study subjects by their popularity, and high incidence of human-relevant cancers. Further, the MHC class I allele DLA-88*034:01, with a known motif, dominates the breed, facilitating discovery of shared CTL responses against mutation-origin neoepitopes by standard prediction methods. We experimentally confirmed the allomorph's binding motif by developing an MHC surface stabilization assay. The assay validated four DLA-88*034:01-presented peptides from canine distemper virus, ubiquitously administered in routine vaccines, for positive controls in future CTL studies. In turn, these viral peptides substantiated motif-based prediction for DLA-88*034:01. The study adds new tools for studying neoepitope-specific CTL in Boxers to foster canine comparative oncology.
Collapse
Affiliation(s)
- Paige S Nemec
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA.,Precision Biosciences, Durham, North Carolina, USA
| | - Jennifer C Holmes
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
| | - Paul R Hess
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
| |
Collapse
|
33
|
Valdivia G, Alonso-Diez Á, Pérez-Alenza D, Peña L. From Conventional to Precision Therapy in Canine Mammary Cancer: A Comprehensive Review. Front Vet Sci 2021; 8:623800. [PMID: 33681329 PMCID: PMC7925635 DOI: 10.3389/fvets.2021.623800] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/11/2021] [Indexed: 12/16/2022] Open
Abstract
Canine mammary tumors (CMTs) are the most common neoplasm in intact female dogs. Canine mammary cancer (CMC) represents 50% of CMTs, and besides surgery, which is the elective treatment, additional targeted and non-targeted therapies could offer benefits in terms of survival to these patients. Also, CMC is considered a good spontaneous intermediate animal model for the research of human breast cancer (HBC), and therefore, the study of new treatments for CMC is a promising field in comparative oncology. Dogs with CMC have a comparable disease, an intact immune system, and a much shorter life span, which allows the achievement of results in a relatively short time. Besides conventional chemotherapy, innovative therapies have a large niche of opportunities. In this article, a comprehensive review of the current research in adjuvant therapies for CMC is conducted to gather available information and evaluate the perspectives. Firstly, updates are provided on the clinical-pathological approach and the use of conventional therapies, to delve later into precision therapies against therapeutic targets such as hormone receptors, tyrosine kinase receptors, p53 tumor suppressor gene, cyclooxygenases, the signaling pathways involved in epithelial-mesenchymal transition, and immunotherapy in different approaches. A comparison of the different investigations on targeted therapies in HBC is also carried out. In the last years, the increasing number of basic research studies of new promising therapeutic agents on CMC cell lines and CMC mouse xenografts is outstanding. As the main conclusion of this review, the lack of effort to bring the in vitro studies into the field of applied clinical research emerges. There is a great need for well-planned large prospective randomized clinical trials in dogs with CMC to obtain valid results for both species, humans and dogs, on the use of new therapies. Following the One Health concept, human and veterinary oncology will have to join forces to take advantage of both the economic and technological resources that are invested in HBC research, together with the innumerable advantages of dogs with CMC as a spontaneous animal model.
Collapse
Affiliation(s)
- Guillermo Valdivia
- Department Animal Medicine, Surgery and Pathology, Veterinary School, Complutense University of Madrid, Madrid, Spain
- Mammary Oncology Unit, Complutense Veterinary Teaching Hospital, Complutense University of Madrid, Madrid, Spain
| | - Ángela Alonso-Diez
- Department Animal Medicine, Surgery and Pathology, Veterinary School, Complutense University of Madrid, Madrid, Spain
- Mammary Oncology Unit, Complutense Veterinary Teaching Hospital, Complutense University of Madrid, Madrid, Spain
| | - Dolores Pérez-Alenza
- Department Animal Medicine, Surgery and Pathology, Veterinary School, Complutense University of Madrid, Madrid, Spain
- Mammary Oncology Unit, Complutense Veterinary Teaching Hospital, Complutense University of Madrid, Madrid, Spain
| | - Laura Peña
- Department Animal Medicine, Surgery and Pathology, Veterinary School, Complutense University of Madrid, Madrid, Spain
- Mammary Oncology Unit, Complutense Veterinary Teaching Hospital, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
34
|
Abstract
Comparative oncology clinical trials play an important and growing role in cancer research and drug development efforts. These trials, typically conducted in companion (pet) dogs, allow assessment of novel anticancer agents and combination therapies in a veterinary clinical setting that supports serial biologic sample collections and exploration of dose, schedule and corresponding pharmacokinetic/pharmacodynamic relationships. Further, an intact immune system and natural co-evolution of tumour and microenvironment support exploration of novel immunotherapeutic strategies. Substantial improvements in our collective understanding of the molecular landscape of canine cancers have occurred in the past 10 years, facilitating translational research and supporting the inclusion of comparative studies in drug development. The value of the approach is demonstrated in various clinical trial settings, including single-agent or combination response rates, inhibition of metastatic progression and randomized comparison of multiple agents in a head-to-head fashion. Such comparative oncology studies have been purposefully included in the developmental plan for several US FDA-approved and up-and-coming anticancer drugs. Challenges for this field include keeping pace with technology and data dissemination/harmonization, improving annotation of the canine genome and immune system, and generation of canine-specific validated reagents to support integration of correlative biology within clinical trial efforts.
Collapse
Affiliation(s)
- Amy K LeBlanc
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Christina N Mazcko
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
35
|
Harman RM, Das SP, Bartlett AP, Rauner G, Donahue LR, Van de Walle GR. Beyond tradition and convention: benefits of non-traditional model organisms in cancer research. Cancer Metastasis Rev 2020; 40:47-69. [PMID: 33111160 DOI: 10.1007/s10555-020-09930-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023]
Abstract
Traditional laboratory model organisms are indispensable for cancer research and have provided insight into numerous mechanisms that contribute to cancer development and progression in humans. However, these models do have some limitations, most notably related to successful drug translation, because traditional model organisms are often short-lived, small-bodied, genetically homogeneous, often immunocompromised, are not exposed to natural environments shared with humans, and usually do not develop cancer spontaneously. We propose that assimilating information from a variety of long-lived, large, genetically diverse, and immunocompetent species that live in natural environments and do develop cancer spontaneously (or do not develop cancer at all) will lead to a more comprehensive understanding of human cancers. These non-traditional model organisms can also serve as sentinels for environmental risk factors that contribute to human cancers. Ultimately, expanding the range of animal models that can be used to study cancer will lead to improved insights into cancer development, progression and metastasis, tumor microenvironment, as well as improved therapies and diagnostics, and will consequently reduce the negative impacts of the wide variety of cancers afflicting humans overall.
Collapse
Affiliation(s)
- Rebecca M Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Sanjna P Das
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Arianna P Bartlett
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Gat Rauner
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Leanne R Donahue
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
36
|
Dow S. A Role for Dogs in Advancing Cancer Immunotherapy Research. Front Immunol 2020; 10:2935. [PMID: 32010120 PMCID: PMC6979257 DOI: 10.3389/fimmu.2019.02935] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 11/29/2019] [Indexed: 12/27/2022] Open
Abstract
While rodent cancer models are essential for early proof-of-concept and mechanistic studies for immune therapies, these models have limitations with regards to predicting the ultimate effectiveness of new immunotherapies in humans. As a unique spontaneous, large animal model of cancer, the value of conducting studies in pet dogs with cancer has been increasingly recognized by the research community. This review will therefore summarize key aspects of the dog cancer immunotherapy model and the role that these studies may play in the overall immunotherapy drug research effort. We will focus on cancer types and settings in which the dog model is most likely to impact clinical immuno-oncology research and drug development. Immunological reagent availability is discussed, along with some unique opportunities and challenges associated with the dog immunotherapy model. Overall it is hoped that this review will increase awareness of the dog cancer immunotherapy model and stimulate additional collaborative studies to benefit both man and man's best friend.
Collapse
Affiliation(s)
- Steven Dow
- Flint Animal Cancer Center, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
37
|
Porcellato I, Silvestri S, Menchetti L, Recupero F, Mechelli L, Sforna M, Iussich S, Bongiovanni L, Lepri E, Brachelente C. Tumour-infiltrating lymphocytes in canine melanocytic tumours: An investigation on the prognostic role of CD3 + and CD20 + lymphocytic populations. Vet Comp Oncol 2019; 18:370-380. [PMID: 31750993 DOI: 10.1111/vco.12556] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 12/16/2022]
Abstract
The study of the immune response in several types of tumours has been rapidly increasing in recent years with the dual aim of understanding the interactions between neoplastic and immune cells and their importance in cancer pathogenesis and progression, as well as identifying targets for cancer immunotherapy. Despite being considered one of the most immunogenic tumour types, melanoma can progress in the presence of abundant lymphocytic infiltration, therefore suggesting that the immune response is not able to efficiently control tumour growth. The purpose of this study was to investigate whether the density, distribution and grade of tumour-infiltrating lymphocytes (TILs) in 97 canine melanocytic tumours is associated with histologic indicators of malignancy and can be considered a prognostic factor in the dog. As a further step in the characterization of the immune response in melanocytic tumours, an immunohistochemical investigation was performed to evaluate the two main populations of TILs, T-lymphocytes (CD3+ ) and B-lymphocytes (CD20+ ). The results of our study show that TILs are present in a large proportion of canine melanocytic tumours, especially in oral melanomas, and that the infiltrate is usually mild. The quantity of CD20+ TILs was significantly associated with some histologic prognostic factors, such as the mitotic count, the cellular pleomorphism and the percentage of pigmented cells. Remarkably, a high infiltration of CD20+ TILs was associated with tumour-related death, presence of metastasis/recurrence, shorter overall and disease-free survival, increased hazard of death and of developing recurrence/metastasis, hence representing a potential new negative prognostic factor in canine melanocytic tumours.
Collapse
Affiliation(s)
- Ilaria Porcellato
- Department of Veterinary Medicine, Università degli Studi di Perugia, Perugia, Italy
| | - Serenella Silvestri
- Department of Veterinary Medicine, Università degli Studi di Perugia, Perugia, Italy
| | - Laura Menchetti
- Department of Veterinary Medicine, Università degli Studi di Perugia, Perugia, Italy
| | - Francesca Recupero
- Department of Veterinary Medicine, Università degli Studi di Perugia, Perugia, Italy
| | - Luca Mechelli
- Department of Veterinary Medicine, Università degli Studi di Perugia, Perugia, Italy
| | - Monica Sforna
- Department of Veterinary Medicine, Università degli Studi di Perugia, Perugia, Italy
| | - Selina Iussich
- Department of Veterinary Science, Università degli Studi di Torino, Turin, Italy
| | - Laura Bongiovanni
- Faculty of Veterinary Medicine, Università degli Studi di Teramo, Teramo, Italy
| | - Elvio Lepri
- Department of Veterinary Medicine, Università degli Studi di Perugia, Perugia, Italy
| | - Chiara Brachelente
- Department of Veterinary Medicine, Università degli Studi di Perugia, Perugia, Italy
| |
Collapse
|
38
|
Castillo-Tandazo W, Mutsaers AJ, Walkley CR. Osteosarcoma in the Post Genome Era: Preclinical Models and Approaches to Identify Tractable Therapeutic Targets. Curr Osteoporos Rep 2019; 17:343-352. [PMID: 31529263 DOI: 10.1007/s11914-019-00534-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE OF REVIEW Osteosarcoma (OS) is the most common cancer of bone, yet is classified as a rare cancer. Treatment and outcomes for OS have not substantively changed in several decades. While the decoding of the OS genome greatly advanced the understanding of the mutational landscape of OS, immediately actionable therapeutic targets were not apparent. Here we describe recent preclinical models that can be leveraged to identify, test, and prioritize therapeutic candidates. RECENT FINDINGS The generation of multiple high fidelity murine models of OS, the spontaneous disease that arises in pet dogs, and the establishment of a diverse collection of patient-derived OS xenografts provide a robust preclinical platform for OS. These models enable evidence to be accumulated across multiple stages of preclinical evaluation. Chemical and genetic screening has identified therapeutic targets, often demonstrating cross species activity. Clinical trials in both PDX models and in canine OS have effectively tested new therapies for prioritization. Improving clinical outcomes in OS has proven elusive. The integrated target discovery and testing possible through a cross species platform provides validation of a putative target and may enable the rigorous evaluation of new therapies in models where endpoints can be rapidly assessed.
Collapse
Affiliation(s)
- Wilson Castillo-Tandazo
- St. Vincent's Institute, 9 Princes St, Fitzroy, VIC, 3065, Australia
- Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, VIC, 3065, Australia
| | - Anthony J Mutsaers
- Department of Biomedical Sciences, Ontario Veterinary College, Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Canada.
| | - Carl R Walkley
- St. Vincent's Institute, 9 Princes St, Fitzroy, VIC, 3065, Australia.
- Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, VIC, 3065, Australia.
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, 3000, Australia.
| |
Collapse
|