1
|
Berry D, Moldoveanu D, Rajkumar S, Lajoie M, Lin T, Tchelougou D, Sakthivel S, Sharon I, Bernard A, Pelletier S, Ripstein Y, Spatz A, Miller WH, Jamal R, Lapointe R, Mes-Masson AM, Petrecca K, Meguerditchian AN, Richardson K, Wang B, Chergui M, Guiot MC, Watters K, Stagg J, Schmeing TM, Rodier F, Turcotte S, Mihalcioiu C, Meterissian S, Watson IR. The NF1 tumor suppressor regulates PD-L1 and immune evasion in melanoma. Cell Rep 2025; 44:115365. [PMID: 40023845 DOI: 10.1016/j.celrep.2025.115365] [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: 10/20/2023] [Revised: 11/24/2024] [Accepted: 02/06/2025] [Indexed: 03/04/2025] Open
Abstract
Hotspot BRAF, hotspot NRAS, and NF1 loss-of-function mutations are found in approximately 50%, 25%, and 15% of cutaneous melanomas, respectively. Compared to mutant BRAF and NRAS, the role of NF1 loss in melanoma remains understudied. NF1 has a RAS GTPase-activating protein (GAP) function; however, studies also support NF1 RAS-independent tumor-suppressor functions. Recent reports indicate that patients with NF1 mutant melanoma have high response rates to anti-PD-1 immune checkpoint inhibitors (ICIs) for reasons that are not entirely clear. Here, we present data demonstrating that NF1 interacts with PD-L1. Furthermore, NF1 loss in melanoma lines increases PD-L1 cell surface expression through a RAS-GAP-independent mechanism. Co-culture experiments demonstrate that NF1 depletion in melanoma increases resistance to T cell killing, which can be abrogated with anti-PD-1/PD-L1 ICIs. These results support a model whereby NF1 loss leads to immune evasion through the PD-L1/PD-1 axis, providing support for the examination of anti-PD-1 therapies in other NF1 mutant cancers.
Collapse
Affiliation(s)
- Diana Berry
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Dan Moldoveanu
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - Shivshankari Rajkumar
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Mathieu Lajoie
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada
| | - Tiffany Lin
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Daméhan Tchelougou
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Samridhi Sakthivel
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Itai Sharon
- Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Antoine Bernard
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Sandy Pelletier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Yael Ripstein
- Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Alan Spatz
- McGill University Health Centre, Montréal, QC H4A 3J1, Canada; Lady Davis Institute, McGill University, Montréal, QC H3T 1E1, Canada
| | - Wilson H Miller
- Lady Davis Institute, McGill University, Montréal, QC H3T 1E1, Canada
| | - Rahima Jamal
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Réjean Lapointe
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada; Montreal Neurological Institute and Hospital, Montréal, QC H3A 2B4, Canada
| | - Anne-Marie Mes-Masson
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Kevin Petrecca
- Montreal Neurological Institute and Hospital, Montréal, QC H3A 2B4, Canada
| | | | | | - Beatrice Wang
- McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - May Chergui
- McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | | | - Kevin Watters
- McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - T Martin Schmeing
- Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Francis Rodier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Simon Turcotte
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | | | | | - Ian R Watson
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada; McGill University Health Centre, Montréal, QC H4A 3J1, Canada.
| |
Collapse
|
2
|
Barbosa MMP, Kamerer RL, Schmit J, Lopez AJ, Uyehara R, Tighe R, Battula S, Kaufman HL, Fan TM. Preclinical Evaluation of an Anchored Immunotherapy Strategy with Aluminum Hydroxide-Tethered IL-12 in Dogs with Advanced Malignant Melanoma. Mol Cancer Ther 2025; 24:406-418. [PMID: 39632727 PMCID: PMC11879767 DOI: 10.1158/1535-7163.mct-24-0317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 10/01/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
Melanoma is an aggressive cancer in dogs involving skin and mucosa similar to humans. Anchored immunotherapeutics offer a novel approach to increase intratumoral retention of therapeutic payloads while decreasing systemic exposure, and this strategy can be critically evaluated through a comparative oncology approach. JEN-101 is an anchored canine IL-12 tethered to aluminum hydroxide administered by local injection. A phase I study was conducted to determine the tolerability, activity, and immune responses of JEN-101 in dogs with advanced melanoma. A 3 + 3 dose-escalation design was used to evaluate intratumoral injection of JEN-101 at 1, 3, 10, or 20 μg/kg every 3 weeks for four cycles. A second course was allowable in the absence of disease progression or toxicity. Peripheral blood, serum, and tumor biopsies were collected at baseline and at prespecified timepoints for pharmacokinetic and immune analyses, which included serum cytokine assay, IHC, and gene expression assessment. JEN-101 was well tolerated with adverse events being fever, lethargy, and isolated elevated liver enzymes. Five dogs experienced grade 3 events, and no grade 4 events were observed. Pharmacokinetic analysis showed a trend toward dose-related maximum serum concentration within 8 hours of injection. Responding dogs demonstrated increased systemic IFN-γ and IL-10 AUC levels and local recruitment of CD3+ T cells. Increased proinflammatory and antigen-processing gene expression was identified in responding lesions. JEN-101 was well tolerated with evidence of biological and therapeutic activities. Anchored IL-12 immunotherapy merits further investigation in dogs with melanoma, and our approach represents an immunocompetent model to inform human clinical trials.
Collapse
Affiliation(s)
- Matheus Moreno Passos Barbosa
- Dept. of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL
- Dept. of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Rebecca L. Kamerer
- Dept. of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Joanna Schmit
- Dept. of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Angel J. Lopez
- Dept. of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Rachel Uyehara
- Dept. of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL
| | | | | | | | - Timothy M. Fan
- Dept. of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL
| |
Collapse
|
3
|
Beebe E, Krudewig C, Motamed Z, Malbon A, Markkanen E. Stromal Expression Profiling Reveals Immune-Driven Adaption to Malignancy in Canine Melanoma Subtypes. Vet Comp Oncol 2025; 23:20-29. [PMID: 39420530 PMCID: PMC11830460 DOI: 10.1111/vco.13021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024]
Abstract
Canine mucosal melanoma (CMM) is the most common oral malignancy in dogs and is significantly more aggressive than its cutaneous counterpart (CCM), yet the reasons for this disparity remain unclear. Cancer-associated stroma (CAS) plays a crucial role in tumour progression, but a detailed understanding of CAS in canine melanoma is missing. To assess stromal reprogramming, we analysed CAS from 21 CMM, 14 CCM and normal stroma from 10 skin and 9 oral mucosa samples by laser-capture microdissection followed by RNA sequencing. Results were assessed in relation to subtypes, prognostic factors including mitotic count (MC), ulceration, necrosis, pigmentation and immune cell infiltration (CD3, CD20 and CD68), scored using immunohistochemistry and RNA in situ hybridisation. Stromal reprogramming was evident in both subtypes but significantly more pronounced in CMM. Immune-excluded tumours exhibited higher MC than desert/cold ones. MC strongly correlated with genes associated with B-cells, T-helper cells and CTLA4 in CCM, suggesting CAS reprogramming to depend on tumour malignancy. Finally, we identify an immune-suppressive stromal signature in a subset of CMM characterised by the downregulation of key immune checkpoints and pathways. Together, these findings provide a solid foundation for understanding the role of CAS in canine melanoma, specific to cutaneous and mucosal subtypes.
Collapse
Affiliation(s)
- Erin Beebe
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse FacultyUniversity of ZurichZurichSwitzerland
| | - Christiane Krudewig
- Institute of Veterinary Pathology, Vetsuisse FacultyUniversity of ZurichZurichSwitzerland
| | - Zahra Motamed
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse FacultyUniversity of ZurichZurichSwitzerland
| | - Alexandra Malbon
- Institute of Veterinary Pathology, Vetsuisse FacultyUniversity of ZurichZurichSwitzerland
- The Royal (Dick) School of Veterinary Studies and the Roslin InstituteMidlothianUK
| | - Enni Markkanen
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse FacultyUniversity of ZurichZurichSwitzerland
| |
Collapse
|
4
|
Blacklock KLB, Donnelly K, Lu Y, del Pozo J, Glendinning L, Polton G, Selmic L, Tanis J, Killick D, Parys M, Morris JS, Breathnach I, Zago S, Gould SM, Shaw DJ, Tivers MS, Malucelli D, Marques A, Purzycka K, Cantatore M, Mathers ME, Stares M, Meynert A, Patton EE. Oronasal mucosal melanoma is defined by two transcriptional subtypes in humans and dogs with implications for diagnosis and therapy. J Pathol 2025; 265:245-259. [PMID: 39828982 PMCID: PMC11794980 DOI: 10.1002/path.6377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/31/2024] [Indexed: 01/22/2025]
Abstract
Mucosal melanoma is a rare melanoma subtype associated with a poor prognosis and limited existing therapeutic interventions, in part due to a lack of actionable targets and translational animal models for preclinical trials. Comprehensive data on this tumour type are scarce, and existing data often overlooks the importance of the anatomical site of origin. We evaluated human and canine oronasal mucosal melanoma (OMM) to determine whether the common canine disease could inform the rare human equivalent. Using a human and canine primary OMM cohort of treatment-naive archival tissue, alongside clinicopathological data, we obtained transcriptomic, immunohistochemical, and microbiome data from both species. We defined the transcriptomic landscape in both species and linked our findings to immunohistochemical, microbiome, and clinical data. Human and dog OMM stratified into two distinctive transcriptional groups, which we defined using a species-independent 41-gene signature. These two subgroups are termed CTLA4-high and MET-high and indicate actionable targets for OMM patients. To guide clinical decision-making, we developed immunohistochemical diagnostic tools that distinguish between transcriptomic subgroups. We found that OMM had conserved transcriptomic subtypes and biological similarity between human and canine OMM, with significant implications for patient classification, treatment, and clinical trial design. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Kelly L Bowlt Blacklock
- Royal (Dick) School of Veterinary Studies and the Roslin InstituteEdinburghUK
- MRC Human Genetics Unit, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
- Edinburgh Cancer Research, CRUK Scotland Centre, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| | - Kevin Donnelly
- MRC Human Genetics Unit, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| | - Yuting Lu
- MRC Human Genetics Unit, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
- Edinburgh Cancer Research, CRUK Scotland Centre, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| | - Jorge del Pozo
- Royal (Dick) School of Veterinary Studies and the Roslin InstituteEdinburghUK
| | - Laura Glendinning
- Royal (Dick) School of Veterinary Studies and the Roslin InstituteEdinburghUK
| | | | - Laura Selmic
- Department of Veterinary Clinical SciencesThe Ohio State UniversityColumbusOHUSA
| | - Jean‐Benoit Tanis
- Department of Small Animal Clinical Sciences, Institute of Infection, Veterinary and Ecological ScienceUniversity of LiverpoolNestonUK
| | - David Killick
- Department of Small Animal Clinical Sciences, Institute of Infection, Veterinary and Ecological ScienceUniversity of LiverpoolNestonUK
| | - Maciej Parys
- Royal (Dick) School of Veterinary Studies and the Roslin InstituteEdinburghUK
| | | | | | | | | | - Darren J Shaw
- Royal (Dick) School of Veterinary Studies and the Roslin InstituteEdinburghUK
| | - Michael S Tivers
- Paragon Veterinary Referrals, Paragon Point, Red Hall CrescentWakefieldUK
| | - Davide Malucelli
- Paragon Veterinary Referrals, Paragon Point, Red Hall CrescentWakefieldUK
| | | | - Katarzyna Purzycka
- Anderson Moores Veterinary Specialists, The Granary, Bunstead BarnsHampshireUK
| | - Matteo Cantatore
- Anderson Moores Veterinary Specialists, The Granary, Bunstead BarnsHampshireUK
| | | | - Mark Stares
- Edinburgh Cancer Research, CRUK Scotland Centre, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
- Edinburgh Cancer Centre, Western General Hospital, Crewe RoadEdinburghUK
| | - Alison Meynert
- MRC Human Genetics Unit, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| | - E Elizabeth Patton
- MRC Human Genetics Unit, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
- Edinburgh Cancer Research, CRUK Scotland Centre, Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| |
Collapse
|
5
|
Gualtieri P, Lee BI, Beeney A, Hart C, Leary D, Martin T, Boss MK. Response of Spontaneous Oral Tumors in Canine Cancer Patients Treated with Stereotactic Body Radiation Therapy (SBRT). Radiat Res 2024; 202:807-824. [PMID: 39478420 DOI: 10.1667/rade-24-00079.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 10/21/2024] [Indexed: 11/09/2024]
Abstract
The objective of this study is describe outcome and toxicity for dogs with oral tumors, specifically oral malignant melanoma (OMM), squamous cell carcinoma (SCC), and soft tissue sarcoma (STS) after stereotactic body radiation therapy (SBRT). A single institution retrospective study was conducted. Outcomes were analyzed using Kaplan-Meier analysis and Cox proportional hazard analysis. Treatment responses at different time points were evaluated with Pearson's Chi-squared test to identify prognostic factors. Acute and late toxicities were recorded according to VRTOG criteria and were analyzed to identify risk factors. Adverse events other than acute and late toxicities were recorded. A total of 98 patients met the inclusion criteria (OMM n = 37; SCC n = 18; STS n = 43). The SBRT prescription was 1-6 fractions, with a total dose range of 12-40 Gy. Local progression-free survival (PFS) for OMM, SCC, and STS was 187, 253, and 161 days, respectively. Overall PFS was 152 days and median survival time (MST) was 270 days, with no statistical difference between tumor types. The presence of lymph node metastasis and the use of elective nodal irradiation (ENI) were associated with shorted PFS and MST. Severe acute toxicities to organs at risk affected 10/85 (11.8%) of patients. Osteoradionecrosis and oronasal fistula formation occurred in 23/81 (28.4%) of patients and was significantly associated with tumor type (SCC, P = 0.006). SBRT can be offered as a treatment option for oral tumors in dogs. Toxicities were common and warrant risk factor considerations and adjustments to current SBRT protocols.
Collapse
Affiliation(s)
- Patricia Gualtieri
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado
| | - Ber-In Lee
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Amber Beeney
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Cullen Hart
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Del Leary
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado
| | - Tiffany Martin
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Mary-Keara Boss
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
6
|
Dagli MLZ, Nagamine MK, Ikeda TL, da Fonseca IIM, Kremer FS, Seixas FK, Hernandez CD, Leite JVP, Yasumaru CC, Massoco CO, Hsieh R, Lourenço SV, Collares TV. Identification of mutations in canine oral mucosal melanomas by exome sequencing and comparison with human melanomas. Sci Rep 2024; 14:24174. [PMID: 39406779 PMCID: PMC11480479 DOI: 10.1038/s41598-024-74748-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
Oral mucosal melanomas (OMMs) are aggressive neoplasms commonly found in dogs but rare in humans. Utilizing whole exome sequencing (WES), which focuses on protein-coding regions to reveal mutation profiles, we conducted a comparative analysis of canine OMM and human melanomas. This study involved DNA extraction, exome enrichment, and sequencing from three canine OMM cell lines (CMGD-2, CMGD-5, TLM-1), five canine OMM frozen samples, a human OMM cell line (MEMO), and a human commercial skin melanoma cell line (SK-MEL-28). The sequencing and subsequent analysis of FASTQ files yielded final variant files, leading to the identification of mutations. Our findings revealed a total of 500 mutated genes in canine OMM, including significant ones such as EP300, FAT4, JAK3, LRP1B, NCOR1, and NOTCH1. Notably, 82 shared mutations were identified between human melanomas and canine OMM genomes. These mutations were categorized based on the gene functions. The identification of these mutations provides critical insights that can pave the way for the development of novel therapeutic strategies for both canine and human OMM, offering hope for more effective treatments in the future.
Collapse
Affiliation(s)
- Maria Lucia Zaidan Dagli
- Laboratory of Experimental and Comparative Oncology, Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil.
| | - Márcia Kazumi Nagamine
- Laboratory of Experimental and Comparative Oncology, Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Tatícia Lieh Ikeda
- Laboratory of Experimental and Comparative Oncology, Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Ivone Izabel Mackowiak da Fonseca
- Laboratory of Experimental and Comparative Oncology, Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | | | | | | | - João Vitor Pereira Leite
- Laboratory of Experimental and Comparative Oncology, Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Cassia Correa Yasumaru
- Laboratory of Comparative Imuno-Oncology, Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Cristina Oliveira Massoco
- Laboratory of Comparative Imuno-Oncology, Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Ricardo Hsieh
- School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
| | | | - Tiago Veiras Collares
- Laboratory of Cancer Biotechnology, Federal University of Pelotas, Pelotas, RS, Brazil
| |
Collapse
|
7
|
Zhang W, Xiao Y, Zhou Q, Zhu X, Zhang Y, Xiang Q, Wu S, Song X, Zhao J, Yuan R, Xiao B, Li L. KNSTRN Is a Prognostic Biomarker That Is Correlated with Immune Infiltration in Breast Cancer and Promotes Cell Cycle and Proliferation. Biochem Genet 2024; 62:3709-3739. [PMID: 38198023 PMCID: PMC11427568 DOI: 10.1007/s10528-023-10615-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/29/2023] [Indexed: 01/11/2024]
Abstract
Kinetochore-localized astrin/SPAG5-binding protein (KNSTRN) promotes the progression of bladder cancer and lung adenocarcinoma. However, its expression and biological function in breast cancer remain largely unknown. Therefore, this study aimed to analyze KNSTRN expression, prognoses, correlation with immune infiltration, expression-associated genes, and regulated signaling pathways to characterize its role in regulating the cell cycle using both bioinformatics and in vitro functional experiments. Analyses of The Cancer Genome Atlas, Gene Expression Omnibus, TIMER, and The Human Protein Atlas databases revealed a significant upregulation of KNSTRN transcript and protein levels in breast cancer. Kaplan-Meier survival analyses demonstrated a significant association between high expression of KNSTRN and poor overall survival, relapse-free survival, post-progression survival, and distant metastases-free survival in patients with breast cancer. Furthermore, multivariate Cox regression analyses confirmed that KNSTRN is an independent prognostic factor for breast cancer. Immune infiltration analysis indicated a positive correlation between KNSTRN expression and T regulatory cell infiltration while showing a negative correlation with Tgd and natural killer cell infiltration. Gene set enrichment analysis along with single-cell transcriptome data analysis suggested that KNSTRN promoted cell cycle progression by regulating the expression of key cell cycle proteins. The overexpression and silencing of KNSTRN in vitro, respectively, promoted and inhibited the proliferation of breast cancer cells. The overexpression of KNSTRN enhanced the expression of key cell cycle regulators, including CDK4, CDK6, and cyclin D3, thereby accelerating the G1/S phase transition and leading to aberrant proliferation of breast cancer cells. In conclusion, our study demonstrates that KNSTRN functions as an oncogene in breast cancer by regulating immune response, promoting G1/S transition, and facilitating breast cancer cell proliferation. Moreover, KNSTRN has potential as a molecular biomarker for diagnostic and prognostic prediction in breast cancer.
Collapse
Affiliation(s)
- Wenwu Zhang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
- Department of Laboratory Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215008, China
| | - Yuhan Xiao
- School of Public Health, Dali University, Dali, 671000, China
| | - Quan Zhou
- Department of Laboratory Medicine, General Hospital of Southern Theater Command of People's Liberation Army (PLA), Guangzhou, 510010, China
| | - Xin Zhu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Yanxia Zhang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Qin Xiang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Shunhong Wu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Xiaoyu Song
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Junxiu Zhao
- School of Public Health, Dali University, Dali, 671000, China
| | - Ruanfei Yuan
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Bin Xiao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China.
| | - Linhai Li
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China.
- Department of Laboratory Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215008, China.
| |
Collapse
|
8
|
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
|
9
|
Mazzone E, Aresu L. Comprehensive Analysis of Microsatellite Instability in Canine Cancers: Implications for Comparative Oncology and Personalized Veterinary Medicine. Animals (Basel) 2024; 14:2484. [PMID: 39272269 PMCID: PMC11394029 DOI: 10.3390/ani14172484] [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: 07/01/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
Microsatellite instability (MSI) is a crucial feature in cancer biology, yet its prevalence and significance in canine cancers remain largely unexplored. This study conducted a comprehensive analysis of MSI across 10 distinct canine cancer histotypes using whole-exome sequencing data from 692 tumor-normal sample pairs. MSI was detected in 64% of tumors, with prevalence varying significantly among cancer types. B-cell lymphomas exhibited the highest MSI burden, contrasting with human studies. A novel "MSI-burden" score was developed, correlating significantly with tumor mutational burden. MSI-high (MSI-H) tumors showed elevated somatic mutation counts compared to MSI-low and microsatellite stable tumors. The study identified 3632 recurrent MSI-affected genomic regions across cancer types. Notably, seven of the ten cancer types exhibited MSI-H tumors, with prevalence ranging from 1.5% in melanomas to 37% in B-cell lymphomas. These findings highlight the potential importance of MSI in canine cancer biology and suggest opportunities for targeted therapies, particularly immunotherapies. The high prevalence of MSI in canine cancers, especially in B-cell lymphomas, warrants further investigation into its mechanistic role and potential as a biomarker for prognosis and treatment response.
Collapse
Affiliation(s)
- Eugenio Mazzone
- Department of Veterinary Sciences, University of Turin, 10095 Grugliasco, Italy
| | - Luca Aresu
- Department of Veterinary Sciences, University of Turin, 10095 Grugliasco, Italy
| |
Collapse
|
10
|
Li S, Liu Z, Lv J, Lv D, Xu H, Shi H, Liu G, Lin D, Jin Y. Establishment of Canine Oral Mucosal Melanoma Cell Lines and Their Xenogeneic Animal Models. Cells 2024; 13:992. [PMID: 38891124 PMCID: PMC11171988 DOI: 10.3390/cells13110992] [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: 03/30/2024] [Revised: 05/11/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Canine oral melanoma is the most prevalent malignant tumor in dogs and has a poor prognosis due to its high aggressiveness and high metastasis and recurrence rates. More research is needed into its treatment and to understand its pathogenic factors. In this study, we isolated a canine oral mucosal melanoma (COMM) cell line designated as COMM6605, which has now been stably passaged for more than 100 generations, with a successful monoclonal assay and a cell multiplication time of 22.2 h. G-banded karyotype analysis of the COMM6605 cell line revealed an abnormal chromosome count ranging from 45 to 74, with the identification of a double-armed chromosome as the characteristic marker chromosome of this cell line. The oral intralingual and dorsal subcutaneous implantation models of BALB/c-nu mice were successfully established; Melan-A (MLANA), S100 beta protein (S100β), PNL2, tyrosinase-related protein 1 (TRP1), and tyrosinase-related protein 2 (TRP2) were stably expressed positively in the canine oral tumor sections, tumor cell lines, and tumor sections of tumor-bearing mice. Sublines COMM6605-Luc-EGFP and COMM6605-Cherry were established through lentiviral transfection, with COMM6605-Luc-EGFP co-expressing firefly luciferase (Luc) and enhanced green fluorescent protein (EGFP) and COMM6605-Cherry expressing the Cherry fluorescent protein gene. The COMM6605-Luc-EGFP fluorescent cell subline was injected via the tail vein and caused lung and lymph node metastasis, as detected by mouse live imaging, which can be used as an animal model to simulate the latter steps of hematogenous spread during tumor metastasis. The canine oral melanoma cell line COMM6605 and two sublines isolated and characterized in this study can offer a valuable model for studying mucosal melanoma.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yipeng Jin
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Rd, Haidian District, Beijing 100193, China; (S.L.); (Z.L.); (J.L.); (D.L.); (H.X.); (H.S.); (G.L.); (D.L.)
| |
Collapse
|
11
|
Aupperle-Lellbach H, Kehl A, de Brot S, van der Weyden L. Clinical Use of Molecular Biomarkers in Canine and Feline Oncology: Current and Future. Vet Sci 2024; 11:199. [PMID: 38787171 PMCID: PMC11126050 DOI: 10.3390/vetsci11050199] [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: 03/28/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Molecular biomarkers are central to personalised medicine for human cancer patients. It is gaining traction as part of standard veterinary clinical practice for dogs and cats with cancer. Molecular biomarkers can be somatic or germline genomic alterations and can be ascertained from tissues or body fluids using various techniques. This review discusses how these genomic alterations can be determined and the findings used in clinical settings as diagnostic, prognostic, predictive, and screening biomarkers. We showcase the somatic and germline genomic alterations currently available to date for testing dogs and cats in a clinical setting, discussing their utility in each biomarker class. We also look at some emerging molecular biomarkers that are promising for clinical use. Finally, we discuss the hurdles that need to be overcome in going 'bench to bedside', i.e., the translation from discovery of genomic alterations to adoption by veterinary clinicians. As we understand more of the genomics underlying canine and feline tumours, molecular biomarkers will undoubtedly become a mainstay in delivering precision veterinary care to dogs and cats with cancer.
Collapse
Affiliation(s)
- Heike Aupperle-Lellbach
- Laboklin GmbH&Co.KG, Steubenstr. 4, 97688 Bad Kissingen, Germany; (H.A.-L.); (A.K.)
- School of Medicine, Institute of Pathology, Technical University of Munich, Trogerstr. 18, 80333 München, Germany
| | - Alexandra Kehl
- Laboklin GmbH&Co.KG, Steubenstr. 4, 97688 Bad Kissingen, Germany; (H.A.-L.); (A.K.)
- School of Medicine, Institute of Pathology, Technical University of Munich, Trogerstr. 18, 80333 München, Germany
| | - Simone de Brot
- Institute of Animal Pathology, COMPATH, University of Bern, 3012 Bern, Switzerland;
| | | |
Collapse
|
12
|
Meuten TK, Dean GA, Thamm DH. Review: The PI3K-AKT-mTOR signal transduction pathway in canine cancer. Vet Pathol 2024; 61:339-356. [PMID: 37905509 DOI: 10.1177/03009858231207021] [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: 11/02/2023]
Abstract
Tumors in dogs and humans share many similar molecular and genetic features, incentivizing a better understanding of canine neoplasms not only for the purpose of treating companion animals, but also to facilitate research of spontaneously developing tumors with similar biologic behavior and treatment approaches in an immunologically competent animal model. Multiple tumor types of both species have similar dysregulation of signal transduction through phosphatidylinositol 3-kinase (PI3K), protein kinase B (PKB; AKT), and mechanistic target of rapamycin (mTOR), collectively known as the PI3K-AKT-mTOR pathway. This review aims to delineate the pertinent aspects of the PI3K-AKT-mTOR signaling pathway in health and in tumor development. It will then present a synopsis of current understanding of PI3K-AKT-mTOR signaling in important canine cancers and advancements in targeted inhibitors of this pathway.
Collapse
|
13
|
Babu S, Chen J, Robitschek E, Baron CS, McConnell A, Wu C, Dedeilia A, Sade-Feldman M, Modhurima R, Manos MP, Chen KY, Cox AM, Ludwig CG, Yang J, Kellis M, Buchbinder EI, Hacohen N, Boland GM, Abraham BJ, Liu D, Zon LI, Insco ML. Specific oncogene activation of the cell of origin in mucosal melanoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590595. [PMID: 38712250 PMCID: PMC11071392 DOI: 10.1101/2024.04.22.590595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Mucosal melanoma (MM) is a deadly cancer derived from mucosal melanocytes. To test the consequences of MM genetics, we developed a zebrafish model in which all melanocytes experienced CCND1 expression and loss of PTEN and TP53. Surprisingly, melanoma only developed from melanocytes lining internal organs, analogous to the location of patient MM. We found that zebrafish MMs had a unique chromatin landscape from cutaneous melanoma. Internal melanocytes could be labeled using a MM-specific transcriptional enhancer. Normal zebrafish internal melanocytes shared a gene expression signature with MMs. Patient and zebrafish MMs have increased migratory neural crest gene and decreased antigen presentation gene expression, consistent with the increased metastatic behavior and decreased immunotherapy sensitivity of MM. Our work suggests the cell state of the originating melanocyte influences the behavior of derived melanomas. Our animal model phenotypically and transcriptionally mimics patient tumors, allowing this model to be used for MM therapeutic discovery.
Collapse
Affiliation(s)
- Swathy Babu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Jiajia Chen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Emily Robitschek
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Chloé S Baron
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Alicia McConnell
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Constance Wu
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | | | - Moshe Sade-Feldman
- Massachusetts General Hospital (MGH) Cancer Center, Boston, MA, 02114, USA
| | - Rodsy Modhurima
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Michael P Manos
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Kevin Y Chen
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Anna M Cox
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Calvin G Ludwig
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Jiekun Yang
- Broad Institute of Massachusetts Institute of Technology (MIT), Cambridge, MA, 02142, USA
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, 02139, USA
| | - Manolis Kellis
- Broad Institute of Massachusetts Institute of Technology (MIT), Cambridge, MA, 02142, USA
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, 02139, USA
| | | | - Nir Hacohen
- Massachusetts General Hospital (MGH) Cancer Center, Boston, MA, 02114, USA
- Broad Institute of Massachusetts Institute of Technology (MIT), Cambridge, MA, 02142, USA
- Harvard Medical School (HMS), Boston, MA, USA; Department of Immunology, HMS, Boston, MA, 02115, USA
| | - Genevieve M Boland
- Massachusetts General Hospital (MGH) Cancer Center, Boston, MA, 02114, USA
| | - Brian J Abraham
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - David Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Leonard I Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Megan L Insco
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| |
Collapse
|
14
|
Pimenta J, Prada J, Pires I, Cotovio M. The Impact of Excision Interval on Equine Melanoma Progression: Time Matters? Animals (Basel) 2024; 14:1244. [PMID: 38672392 PMCID: PMC11047369 DOI: 10.3390/ani14081244] [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: 03/19/2024] [Revised: 04/17/2024] [Accepted: 04/20/2024] [Indexed: 04/28/2024] Open
Abstract
Equine melanomas are a common neoplasm in gray horses. However, scientific knowledge about their progression over time is quite scarce. Some owners and veterinarians still believe that early intervention is not necessary, stating that tumors evolve very slowly and intervention could worsen the animal's condition. This work aims to identify clinical and histological differences that may exist between equine melanomas with different excision intervals (time between tumor detection and surgical excision). A total of 42 tumors (13 benign and 29 malignant) from 34 horses were included in this study. There was a statistically significant association between excision interval and tumor size (p = 0.038), with tumors excised later being significantly larger than the ones excised sooner. The excision interval was also statistically associated with the number of tumors (p = 0.011), since the horses that carried a tumor for longer seemed to be prone to have multiple tumors. Furthermore, there was an association between excision interval and malignancy (p = 0.035), with tumor excised later being fives times more likely to be malignant. This study provides evidence of delayed excision's effect on the progression of equine melanomas. Additionally, it reinforces the importance of the early excision of these tumors.
Collapse
Affiliation(s)
- José Pimenta
- CECAV-Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (M.C.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
- CIVG–Vasco da Gama Research Center, EUVG–Vasco da Gama University School, 3020-210 Coimbra, Portugal
| | - Justina Prada
- CECAV-Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (M.C.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
- Veterinary Sciences Department, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| | - Isabel Pires
- CECAV-Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (M.C.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
- Veterinary Sciences Department, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| | - Mário Cotovio
- CECAV-Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (M.C.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
- Veterinary Sciences Department, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
- Faculty of Veterinary Medicine, Lusófona University, Campo Grande 376, 1749-024 Lisbon, Portugal
| |
Collapse
|
15
|
Polton G, Borrego JF, Clemente-Vicario F, Clifford CA, Jagielski D, Kessler M, Kobayashi T, Lanore D, Queiroga FL, Rowe AT, Vajdovich P, Bergman PJ. Melanoma of the dog and cat: consensus and guidelines. Front Vet Sci 2024; 11:1359426. [PMID: 38645640 PMCID: PMC11026649 DOI: 10.3389/fvets.2024.1359426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/12/2024] [Indexed: 04/23/2024] Open
Abstract
Melanoma of the dog and cat poses a clinical challenge to veterinary practitioners across the globe. As knowledge evolves, so too do clinical practices. However, there remain uncertainties and controversies. There is value for the veterinary community at large in the generation of a contemporary wide-ranging guideline document. The aim of this project was therefore to assimilate the available published knowledge into a single accessible referenced resource and to provide expert clinical guidance to support professional colleagues as they navigate current melanoma challenges and controversies. Melanocytic tumors are common in dogs but rare in cats. The history and clinical signs relate to the anatomic site of the melanoma. Oral and subungual malignant melanomas are the most common malignant types in dogs. While many melanocytic tumors are heavily pigmented, making diagnosis relatively straightforward, melanin pigmentation is variable. A validated clinical stage scheme has been defined for canine oral melanoma. For all other locations and for feline melanoma, TNM-based staging applies. Certain histological characteristics have been shown to bear prognostic significance and can thus prove instructive in clinical decision making. Surgical resection using wide margins is currently the mainstay of therapy for the local control of melanomas, regardless of primary location. Radiotherapy forms an integral part of the management of canine oral melanomas, both as a primary and an adjuvant therapy. Adjuvant immunotherapy or chemotherapy is offered to patients at high risk of developing distant metastasis. Location is the major prognostic factor, although it is not completely predictive of local invasiveness and metastatic potential. There are no specific guidelines regarding referral considerations for dogs with melanoma, as this is likely based on a multitude of factors. The ultimate goal is to provide the best options for patients to extend quality of life and survival, either within the primary care or referral hospital setting.
Collapse
Affiliation(s)
- Gerry Polton
- North Downs Specialist Referrals, Bletchingley, United Kingdom
| | - Juan F. Borrego
- Hospital Aúna Especialidades Veterinarias IVC Evidensia, Paterna, Spain
| | | | | | - Dariusz Jagielski
- Veterinary Institute, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Toruń, Poland
| | - Martin Kessler
- Department of Clinical Oncology, Tierklinik Hofheim, Hofheim, Germany
| | | | | | | | | | - Péter Vajdovich
- Department of Physiology and Oncology, University of Veterinary Medicine, Budapest, Hungary
| | - Philip J. Bergman
- VCA Clinical Studies, Katonah-Bedford Veterinary Center, Bedford Hills, NY, United States
| |
Collapse
|
16
|
Sakthikumar S, Warrier M, Whitley D, Facista S, Adkins J, Aman S, Tsinajinnie D, Duran N, Siravegna G, Ahmed Z, Day K, Jenkins B, Patel N, Ryden K, Nadai J, Banovich K, Powers B, Edwards J, Steinberg J, Fielder S, Wong S, Byron SA, Izatt T, Zismann V, Boateng M, Zhu Z, Chuang HY, Trent JM, Haworth D, Chon E, Hendricks W, Wang G. Genomic analysis across 53 canine cancer types reveals novel mutations and high clinical actionability potential. Vet Comp Oncol 2024; 22:30-41. [PMID: 38053317 DOI: 10.1111/vco.12944] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/27/2023] [Accepted: 11/12/2023] [Indexed: 12/07/2023]
Abstract
A genomic understanding of the oncogenic processes and individual variability of human cancer has steadily fueled improvement in patient outcomes over the past 20 years. Mutations within tumour tissues are routinely assessed through clinical genomic diagnostic assays by academic and commercial laboratories to facilitate diagnosis, prognosis and effective treatment stratification. The application of genomics has unveiled a wealth of mutation-based biomarkers in canine cancers, suggesting that the transformative principles that have revolutionized human cancer medicine can be brought to bear in veterinary oncology. To advance clinical genomics and genomics-guided medicine in canine oncology, we have developed and validated a canine cancer next-generation sequencing gene panel for the identification of multiple mutation types in clinical specimens. With this panel, we examined the genomic landscapes of 828 tumours from 813 dogs, spanning 53 cancer types. We identified 7856 alterations, encompassing copy number variants, single nucleotide variants, indels and internal tandem duplications. Additionally, we evaluated the clinical utility of these alterations by incorporating a biomarker framework from comprehensive curation of primary canine literature and inferences from human cancer genomic biomarker literature and clinical diagnostics. Remarkably, nearly 90% of the cases exhibited mutations with diagnostic, prognostic or therapeutic implications. Our work represents a thorough assessment of genomic landscapes in a large cohort of canine cancers, the first of its kind for its comprehensive inclusion of multiple mutation types and structured annotation of biomarkers, demonstrating the clinical potential of leveraging mutation-based biomarkers in veterinary oncology.
Collapse
Affiliation(s)
| | | | | | | | | | - Sara Aman
- Vidium Animal Health, Phoenix, Arizona, USA
| | | | | | | | | | | | | | | | - Kirk Ryden
- Vidium Animal Health, Phoenix, Arizona, USA
| | - Joe Nadai
- Vidium Animal Health, Phoenix, Arizona, USA
| | | | | | | | - Jennifer Steinberg
- Center for Cancer Research at the National Cancer Institute, National Cancer Institute, Bethesda, Maryland, USA
| | | | - Shukmei Wong
- Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Sara A Byron
- Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Tyler Izatt
- Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Victoria Zismann
- Translational Genomics Research Institute, Phoenix, Arizona, USA
| | | | | | | | - Jeffrey M Trent
- Translational Genomics Research Institute, Phoenix, Arizona, USA
| | | | | | | | | |
Collapse
|
17
|
Pimenta J, Prada J, Pires I, Cotovio M. Cyclooxygenase-2 (COX-2) Expression in Equine Melanocytic Tumors. Vet Sci 2024; 11:77. [PMID: 38393095 PMCID: PMC10891553 DOI: 10.3390/vetsci11020077] [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: 01/08/2024] [Revised: 01/27/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Equine melanocytic tumors are common and have an unusual benign behavior with low invasiveness and metastatic rates. However, tumoral mass growth is usually a concern that can have life-threatening consequences. COX-2 is related to oncogenesis, promoting neoplastic cell proliferation, invasion, and metastasis. The aim of this study was to evaluate the immunohistochemical expression of COX-2 in equine melanocytic tumors. Through extension and intensity of labeling, 39 melanocytomas and 38 melanomas were evaluated. Of the malignant tumors, 13.2% were negative and 63.2% presented a low COX-2 expression. Only 6 malignant tumors presented >50% of labeled cells, 18 malignant and 8 benign had an expression between 21 and 50%, 8 malignant and 3 benign tumors had an expression between 6 and 20%, 1 malignant tumor had an expression between 1 and 5%, and 5 malignant and 28 benign tumors had no expression. Malignant tumors showed higher COX-2 expression than did benign tumors, with statistically significant differences. The low levels of COX-2 may be one of the molecular reasons for the presence of expansive mass growth instead of the invasive pattern of other species, which is related to high COX-2 levels.
Collapse
Affiliation(s)
- José Pimenta
- CECAV—Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (M.C.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
- CIVG—Vasco da Gama Research Center, EUVG—Vasco da Gama University School, 3020-210 Coimbra, Portugal
| | - Justina Prada
- CECAV—Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (M.C.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
- Veterinary Sciences Department, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| | - Isabel Pires
- CECAV—Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (M.C.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
- Veterinary Sciences Department, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| | - Mário Cotovio
- CECAV—Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (M.C.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
- Faculty of Veterinary Medicine, Lusófona University, Campo Grande 376, 1749-024 Lisbon, Portugal
| |
Collapse
|
18
|
Gao Y, Packeiser EM, Wendt S, Sekora A, Cavalleri JMV, Pratscher B, Alammar M, Hühns M, Brenig B, Junghanss C, Nolte I, Murua Escobar H. Cross-Species Comparison of the Pan-RAF Inhibitor LY3009120's Anti-Tumor Effects in Equine, Canine, and Human Malignant Melanoma Cell Lines. Genes (Basel) 2024; 15:202. [PMID: 38397192 PMCID: PMC10887541 DOI: 10.3390/genes15020202] [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: 12/20/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Malignant melanomas (MMs) are the abnormal proliferation of melanocytes and are one of the lethal skin cancers in humans, equines, and canines. Accordingly, MMs in companion animals can serve as naturally occurring animal models, completing conventional cancer models. The common constitutive activation of the MAPK and PI3K pathways in MMs has been described in all three species. Targeting the related pathways is considered a potential option in comparative oncologic approaches. Herein, we present a cross-species comparative analysis exposing a set of ten melanoma cell lines (one human, three equine, and six canine) derived from primary tumors or metastasis to a pan-RAF and RAF dimer inhibitor (LY3009120). Cellular response (proliferation, biomass, metabolism, early and late apoptosis/necrosis, and morphology) and the presence of pathogenic single-nucleotide variants (SNVs) within the mutational hotspot genes BRAF exon 11 and 15, NRAS exon 2 and 3, KRAS exon 2, and KIT exon 11 were analyzed. This study showed that equine malignant melanoma (EMM) cells (MelDuWi) harbor the KRAS p.Q61H mutation, while canine malignant melanoma (CMM) cells (cRGO1 and cRGO1.2) carry NRAS p.G13R. Except for EMM metastasis cells eRGO6 (wild type of the above-mentioned hotspot genes), all melanoma cell lines exhibited a decrease in dose dependence after 48 and 72 h of exposure to LY3009120, independent of the mutation hotspot landscape. Furthermore, LY3009120 caused significant early apoptosis and late apoptosis/necrosis in all melanoma cell lines except for eRGO6. The anti-tumor effects of LY3009120 were observed in nine melanoma cell lines, indicating the potential feasibility of experimental trials with LY3009120. The present study reveals that the irradiation-resistant canine metastasis cells (cRGO1.2) harboring the NRAS p.G13R mutation are significantly LY3009120-sensitive, while the equine metastases-derived eRGO6 cells show significant resistance to LY3009120, which make them both valuable tools for studying resistance mechanisms in comparative oncology.
Collapse
Affiliation(s)
- Yu Gao
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany
- Department of Medicine, Clinic III, Hematology, Oncology and Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany
| | - Eva-Maria Packeiser
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany
| | - Sophia Wendt
- Department of Medicine, Clinic III, Hematology, Oncology and Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany
| | - Anett Sekora
- Department of Medicine, Clinic III, Hematology, Oncology and Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany
| | - Jessika-Maximiliane V. Cavalleri
- Clinical Unit of Equine Internal Medicine, Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Barbara Pratscher
- Clinical Unit of Equine Internal Medicine, Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
- Clinical Unit of Small Animal Internal Medicine, Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Moosheer Alammar
- Department of Medicine, Clinic III, Hematology, Oncology and Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany
| | - Maja Hühns
- Institute of Pathology, University Medicine of Rostock, Strempelstrasse, 18055 Rostock, Germany
| | - Bertram Brenig
- Institute of Veterinary Medicine, Division of Molecular Biology of Livestock and Molecular Diagnostics, Georg-August-University of Göttingen, 37077 Göttingen, Germany
| | - Christian Junghanss
- Department of Medicine, Clinic III, Hematology, Oncology and Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany
| | - Ingo Nolte
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany
| | - Hugo Murua Escobar
- Department of Medicine, Clinic III, Hematology, Oncology and Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany
| |
Collapse
|
19
|
Pimenta J, Prada J, Pires I, Cotovio M. Programmed Cell Death-Ligand 1 (PD-L1) Immunohistochemical Expression in Equine Melanocytic Tumors. Animals (Basel) 2023; 14:48. [PMID: 38200779 PMCID: PMC10778310 DOI: 10.3390/ani14010048] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Currently available treatments for equine melanocytic tumors have limitations, mainly due to mass localization and dimension, or the presence of metastases. Therefore, a search for new therapies is necessary. Programmed cell death-ligand 1 (PD-L1) is expressed by several tumors, blocking T cell-mediated elimination of the tumor cells by binding to programmed cell death protein 1 (PD-1). A novel therapeutic approach using PD-1/PD-L1 blockade in human melanoma resulted in tumor regression and prolonged tumor-free survival. This study aimed to evaluate the immunohistochemical expression of PD-L1 in equine melanocytic tumors. A total of 77 melanocytic tumors were classified as benign or malignant and evaluated by extension of labeling. A total of 59.7% of the tumors showed >50% of immunolabeled cells. Regarding malignant tumors, 24/38 tumors presented >50% of labeled cells, 13 tumors presented between 25-50% and one tumor presented <10%. Regarding benign tumors, 22/39 tumors presented >50% of labeled cells, nine tumors presented 25-50%, three tumors presented 10-25%, two tumors presented <10% and three tumors did not present expression. Our results suggest that PD-L1 blockade may be a potential target for immunotherapy in equine melanocytic tumors and that future clinical research trials into the clinical efficacy of the anti-PD-L1 antibody are necessary.
Collapse
Affiliation(s)
- José Pimenta
- CECAV—Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (M.C.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
- CIVG—Vasco da Gama Research Center, EUVG—Vasco da Gama University School, 3020-210 Coimbra, Portugal
| | - Justina Prada
- CECAV—Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (M.C.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
- Veterinary Sciences Department, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| | - Isabel Pires
- CECAV—Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (M.C.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
- Veterinary Sciences Department, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| | - Mário Cotovio
- CECAV—Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (M.C.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
- Faculty of Veterinary Medicine, Lusófona University, Campo Grande 376, 1749-024 Lisbon, Portugal
| |
Collapse
|
20
|
Zhou BW, Wu QQ, Mauki DH, Wang X, Zhang SR, Yin TT, Chen FL, Li C, Liu YH, Wang GD, Zhang YP. Germline gene fusions across species reveal the chromosomal instability regions and cancer susceptibility. iScience 2023; 26:108431. [PMID: 38205119 PMCID: PMC10777377 DOI: 10.1016/j.isci.2023.108431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 06/24/2023] [Accepted: 11/08/2023] [Indexed: 01/12/2024] Open
Abstract
The canine transmissible venereal tumor (CTVT) is a clonal cell-mediated cancer with a long evolutionary history and extensive karyotype rearrangements in its genome. However, little is known about its genetic similarity to human tumors. Here, using multi-omics data we identified 11 germline gene fusions (GGFs) in CTVT, which showed higher genetic susceptibility than others. Additionally, we illustrate a mechanism of a complex gene fusion of three gene segments (HSD17B4-DMXL1-TNFAIP8) that we refer to "greedy fusion". Our findings also provided evidence that expressions of GGFs are downregulated during the tumor regressive phase, which is associated with DNA methylation level. This study presents a comprehensive landscape of gene fusions (GFs) in CTVT, which offers a valuable genetic resource for exploring potential genetic mechanisms underlying the development of cancers in both dogs and humans.
Collapse
Affiliation(s)
- Bo-Wen Zhou
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Qing-Qin Wu
- School of Ecology and Environmental Sciences, Yunnan University, Kunming, Yunnan 650500, China
| | - David H. Mauki
- Institute of Neurological Disease, National-Local Joint Engineering Research Center of Translational Medicine, State Key Lab of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xuan Wang
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Shu-Run Zhang
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Ting-Ting Yin
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Fang-Liang Chen
- Kunming Police Dog Base of the Ministry of Public Security, Kunming, Yunnan 650204, China
| | - Chao Li
- State Key Laboratory for Conservation and Utilization of Bio-Resource, Yunnan University, Kunming, Yunnan 650500, China
| | - Yan-Hu Liu
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Guo-Dong Wang
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Ya-Ping Zhang
- State Key Laboratory of Genetic Resources and Evolution & Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| |
Collapse
|
21
|
Jin L, Zhang X, Fan M, Li W, Zhang X. NamiRNA-mediated high expression of KNSTRN correlates with poor prognosis and immune infiltration in hepatocellular carcinoma. Contemp Oncol (Pozn) 2023; 27:163-175. [PMID: 38239867 PMCID: PMC10793618 DOI: 10.5114/wo.2023.133507] [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: 09/04/2023] [Accepted: 11/12/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction Mutations of kinetochore-localized astrin/sperm-associated antigen 5 (KNSTRN) can interfere with chromatid cohesion, increase aneuploidy in tumours, and enhance tumourigenesis. However, the role of the KNSTRN-binding protein in hepatocellular carcinoma (HCC) remains unclear. Material and methods Using The Cancer Genome Atlas databases, we investigated the potential oncogenic functions of KNSTRN in HCC along with R and various computational tools. Results Detailed results revealed that elevated expression of KNSTRN was considerably associated with poor overall survival (HR = 1.48, 95% CI: 1.05-2.09, p = 0.027) and progress-free interval (HR = 1.41, 95% CI: 1.05-1.89, p = 0.021) in HCC. Gene ontology/Kyoto Encyclopedia of Genes and Genomes functional enrichment analysis showed that KNSTRN is closely related to organelle fission, chromosomal region, tubulin binding, and cell cycle signalling pathway. TIMER database analysis showed the correlations between KNSTRN expression and tumour-infiltrating immune cells, biomarkers of immune cells, and immune checkpoint expression. Moreover, the KNSTRN level was significantly positively associated with immunosuppressive cells in the tumour microenvironment, including regulatory T-cells, myeloid-derived suppressor cells, and cancer-associated fibrocytes. Finally, a possible nuclear activating miRNA (NamiRNA)-enhancer network of hsa-miR-107, which activates the KNSTRN expression in liver hepatocellular carcinoma, was constructed by correlation analysis. Conclusions NamiRNA-mediated upregulation of KNSTRN correlated with poor prognosis and tumour immune infiltration in HCC. KNSTRN could serve as an effective biomarker for the diagnosis and prognosis of HCC and support the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Liang Jin
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Xiaojing Zhang
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Ming Fan
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Weimin Li
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Xuan Zhang
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
22
|
Pisamai S, Edwards SW, Cheng CW, Chaivichit P, Sooksiri M, Yanakam S, Maneewong S, Suriyaphol G. Tissue transcriptome profiling and pathway analyses revealed novel potential biomarkers in the tumor progression of canine oral melanoma. Res Vet Sci 2023; 165:105036. [PMID: 37856944 DOI: 10.1016/j.rvsc.2023.105036] [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: 09/06/2022] [Revised: 09/16/2023] [Accepted: 09/22/2023] [Indexed: 10/21/2023]
Abstract
Canine oral melanoma (COM) is an aggressive oral malignancy in dogs, mostly with metastasis. However, the understanding of total gene expression of oral melanoma (OM) at different clinical stages has been limited. The objective of this study was to identify novel mRNA biomarkers of early-stage OM (EOM) and late-stage OM (LOM). Transcriptome sequencing of 3 EOM, 5 LOM and 4 normal gingival tissues (controls) was performed. Selected transcriptome results were validated by quantitative reverse transcription-PCR (qRT-PCR) using 12 LOM and 10 controls. We found 534 differentially expressed in EOM compared with controls, whereas 696 genes in LOM were differentially expressed compared with controls (P < 0.05). Moreover, 27 genes were differentially expressed in LOM compared with EOM (P < 0.05). The genes expressed in COM were involved in the molecular mechanism of cancer and melanocyte development pathways, promoting melanoma progression. qRT-PCR confirmed an increased expression of genes encoding an important protein in chemotherapy resistance (dopachrome tautomerase, DCT) and tumor progression (forkhead box M1, FOXM1), and decreased expression of a tumor suppression gene (N-myc downstream-regulated gene 2, NDRG2) in LOM, concordant with transcriptome results. In conclusion, this study revealed the comprehensive transcriptome from COM tissues, and increased DCT and FOXM1 and decreased NDRG2 gene expression indicated the potential candidate biomarkers in COM progression.
Collapse
Affiliation(s)
- Sirinun Pisamai
- Department of Veterinary Surgery, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand; Center of Excellence for Companion Animal Cancer, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Steven W Edwards
- Institute of Infection, Veterinary and Ecological Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | - Chew Weng Cheng
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Phannita Chaivichit
- Biochemistry Unit, Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Mokhapoom Sooksiri
- Biochemistry Unit, Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Sujittra Yanakam
- Biochemistry Unit, Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Sattabongkoch Maneewong
- Biochemistry Unit, Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Gunnaporn Suriyaphol
- Center of Excellence for Companion Animal Cancer, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand; Biochemistry Unit, Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
23
|
Shinada M, Kato D, Motegi T, Tsuboi M, Ikeda N, Aoki S, Iguchi T, Li T, Kodera Y, Ota R, Hashimoto Y, Takahashi Y, Chambers J, Uchida K, Kato Y, Nishimura R, Nakagawa T. Podoplanin Drives Amoeboid Invasion in Canine and Human Mucosal Melanoma. Mol Cancer Res 2023; 21:1205-1219. [PMID: 37493578 DOI: 10.1158/1541-7786.mcr-22-0929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 04/09/2023] [Accepted: 07/13/2023] [Indexed: 07/27/2023]
Abstract
Mucosal melanoma metastasizes at an early stage of the disease in human and dog. We revealed that overexpression of podoplanin in tumor invasion fronts (IF) was related to poor prognosis of dogs with mucosal melanoma. Moreover, podoplanin expressed in canine mucosal melanoma cells promotes proliferation and aggressive amoeboid invasion by activating Rho-associated kinase (ROCK)-myosin light chain 2 (MLC2) signaling. PDPN-ROCK-MLC2 signaling plays a role in cell-cycle arrest and cellular senescence escape as a mechanism for regulating proliferation. Podoplanin induces amoeboid invasion in the IFs of mouse xenografted tumor tissues, similar to canine mucosal melanoma clinical samples. We further identified that podoplanin expression was related to poor prognosis of human patients with mucosal melanoma, and human mucosal melanoma with podoplanin-high expression enriched gene signatures related to amoeboid invasion, similar to canine mucosal melanoma. Overall, we propose that podoplanin promotes canine and human mucosal melanoma metastasis by inducing aggressive amoeboid invasion and naturally occurring canine mucosal melanoma can be a novel research model for podoplanin expressing human mucosal melanoma. IMPLICATIONS Podoplanin could be a new therapeutic target to restrict the metastatic dissemination of canine and human mucosal melanoma.
Collapse
Affiliation(s)
- Masahiro Shinada
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Daiki Kato
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Tomoki Motegi
- Veterinary Medical Center, The University of Tokyo, Tokyo, Japan
| | - Masaya Tsuboi
- Veterinary Medical Center, The University of Tokyo, Tokyo, Japan
| | - Namiko Ikeda
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Susumu Aoki
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Takaaki Iguchi
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Toshio Li
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuka Kodera
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Ryosuke Ota
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuko Hashimoto
- Veterinary Medical Center, The University of Tokyo, Tokyo, Japan
| | - Yosuke Takahashi
- Veterinary Medical Center, The University of Tokyo, Tokyo, Japan
| | - James Chambers
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuyuki Uchida
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Ryohei Nishimura
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Takayuki Nakagawa
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
24
|
Sakthikumar S, Facista S, Whitley D, Byron SA, Ahmed Z, Warrier M, Zhu Z, Chon E, Banovich K, Haworth D, Hendricks WPD, Wang G. Standing in the canine precision medicine knowledge gap: Improving annotation of canine cancer genomic biomarkers through systematic comparative analysis of human cancer mutations in COSMIC. Vet Comp Oncol 2023; 21:482-491. [PMID: 37248814 DOI: 10.1111/vco.12911] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 04/26/2023] [Accepted: 05/08/2023] [Indexed: 05/31/2023]
Abstract
The accrual of cancer mutation data and related functional and clinical associations have revolutionised human oncology, enabling the advancement of precision medicine and biomarker-guided clinical management. The catalogue of cancer mutations is also growing in canine cancers. However, without direct high-powered functional data in dogs, it remains challenging to interpret and utilise them in research and clinical settings. It is well-recognised that canine and human cancers share genetic, molecular and phenotypic similarities. Therefore, leveraging the massive wealth of human mutation data may help advance canine oncology. Here, we present a structured analysis of sequence conservation and conversion of human mutations to the canine genome through a 'caninisation' process. We applied this analysis to COSMIC, the Catalogue of Somatic Mutations in Cancer, the most prominent human cancer mutation database. For the project's initial phase, we focused on the subset of the COSMIC data corresponding to Cancer Gene Census (CGC) genes. A total of 670 canine orthologs were found for 721 CGC genes. In these genes, 365 K unique mutations across 160 tumour types were converted successfully to canine coordinates. We identified shared putative cancer-driving mutations, including pathogenic and hotspot mutations and mutations bearing similar biomarker associations with diagnostic, prognostic and therapeutic utility. Thus, this structured caninisation of human cancer mutations facilitates the interpretation and annotation of canine mutations and helps bridge the knowledge gap to enable canine precision medicine.
Collapse
Affiliation(s)
| | | | - Derick Whitley
- Vidium Animal Health, a TGen Subsidiary, Phoenix, Arizona, USA
| | - Sara A Byron
- Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Zeeshan Ahmed
- Vidium Animal Health, a TGen Subsidiary, Phoenix, Arizona, USA
| | - Manisha Warrier
- Vidium Animal Health, a TGen Subsidiary, Phoenix, Arizona, USA
| | - Zhanyang Zhu
- Vidium Animal Health, a TGen Subsidiary, Phoenix, Arizona, USA
| | - Esther Chon
- Vidium Animal Health, a TGen Subsidiary, Phoenix, Arizona, USA
| | | | - David Haworth
- Vidium Animal Health, a TGen Subsidiary, Phoenix, Arizona, USA
| | | | - Guannan Wang
- Vidium Animal Health, a TGen Subsidiary, Phoenix, Arizona, USA
| |
Collapse
|
25
|
Wicks MN, Glinka M, Hill B, Houghton D, Sharghi M, Ferreira I, Adams D, Din S, Papatheodorou I, Kirkwood K, Cheeseman M, Burger A, Baldock RA, Arends MJ. The Comparative Pathology Workbench: Interactive visual analytics for biomedical data. J Pathol Inform 2023; 14:100328. [PMID: 37693862 PMCID: PMC10491844 DOI: 10.1016/j.jpi.2023.100328] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/07/2023] [Accepted: 08/04/2023] [Indexed: 09/12/2023] Open
Abstract
Pathologists need to compare histopathological images of normal and diseased tissues between different samples, cases, and species. We have designed an interactive system, termed Comparative Pathology Workbench (CPW), which allows direct and dynamic comparison of images at a variety of magnifications, selected regions of interest, as well as the results of image analysis or other data analyses such as scRNA-seq. This allows pathologists to indicate key diagnostic features, with a mechanism to allow discussion threads amongst expert groups of pathologists and other disciplines. The data and associated discussions can be accessed online from anywhere in the world. The Comparative Pathology Workbench (CPW) is a web-browser-based visual analytics platform providing shared access to an interactive "spreadsheet" style presentation of image and associated analysis data. The CPW provides a grid layout of rows and columns so that images that correspond to matching data can be organised in the form of an image-enabled "spreadsheet". An individual workbench can be shared with other users with read-only or full edit access as required. In addition, each workbench element or the whole bench itself has an associated discussion thread to allow collaborative analysis and consensual interpretation of the data. The CPW is a Django-based web-application that hosts the workbench data, manages users, and user-preferences. All image data are hosted by other resource applications such as OMERO or the Digital Slide Archive. Further resources can be added as required. The discussion threads are managed using WordPress and include additional graphical and image data. The CPW has been developed to allow integration of image analysis outputs from systems such as QuPath or ImageJ. All software is open-source and available from a GitHub repository.
Collapse
Affiliation(s)
- Michael N. Wicks
- Edinburgh Pathology & Centre for Comparative Pathology, Institute of Genetics & Cancer, University of Edinburgh, Crewe Road, Edinburgh EH4 2XR, UK
| | - Michael Glinka
- Edinburgh Pathology & Centre for Comparative Pathology, Institute of Genetics & Cancer, University of Edinburgh, Crewe Road, Edinburgh EH4 2XR, UK
| | - Bill Hill
- Department of Computer Science, School of Mathematical and Computer Sciences, Heriot-Watt University, Edinburgh, UK
| | - Derek Houghton
- Department of Computer Science, School of Mathematical and Computer Sciences, Heriot-Watt University, Edinburgh, UK
| | - Mehran Sharghi
- Department of Computer Science, School of Mathematical and Computer Sciences, Heriot-Watt University, Edinburgh, UK
| | - Ingrid Ferreira
- Edinburgh Pathology & Centre for Comparative Pathology, Institute of Genetics & Cancer, University of Edinburgh, Crewe Road, Edinburgh EH4 2XR, UK
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - David Adams
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Shahida Din
- Edinburgh IBD Unit Western General Hospital, NHS Lothian, Edinburgh, UK
| | - Irene Papatheodorou
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridge, UK
| | - Kathryn Kirkwood
- Pathology Department, Western General Hospital, NHS Lothian, Edinburgh, UK
| | - Michael Cheeseman
- Edinburgh Pathology & Centre for Comparative Pathology, Institute of Genetics & Cancer, University of Edinburgh, Crewe Road, Edinburgh EH4 2XR, UK
| | - Albert Burger
- Department of Computer Science, School of Mathematical and Computer Sciences, Heriot-Watt University, Edinburgh, UK
| | - Richard A. Baldock
- Edinburgh Pathology & Centre for Comparative Pathology, Institute of Genetics & Cancer, University of Edinburgh, Crewe Road, Edinburgh EH4 2XR, UK
| | - Mark J. Arends
- Edinburgh Pathology & Centre for Comparative Pathology, Institute of Genetics & Cancer, University of Edinburgh, Crewe Road, Edinburgh EH4 2XR, UK
| |
Collapse
|
26
|
Rodrigues L, Watson J, Feng Y, Lewis B, Harvey G, Post G, Megquier K, White ME, Lambert L, Miller A, Lopes C, Zhao S. Shared hotspot mutations in oncogenes position dogs as an unparalleled comparative model for precision therapeutics. Sci Rep 2023; 13:10935. [PMID: 37414794 PMCID: PMC10325973 DOI: 10.1038/s41598-023-37505-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/22/2023] [Indexed: 07/08/2023] Open
Abstract
Naturally occurring canine cancers have remarkable similarities to their human counterparts. To better understand these similarities, we investigated 671 client-owned dogs from 96 breeds with 23 common tumor types, including those whose mutation profile are unknown (anal sac carcinoma and neuroendocrine carcinoma) or understudied (thyroid carcinoma, soft tissue sarcoma and hepatocellular carcinoma). We discovered mutations in 50 well-established oncogenes and tumor suppressors, and compared them to those reported in human cancers. As in human cancer, TP53 is the most commonly mutated gene, detected in 22.5% of canine tumors overall. Canine tumors share mutational hotspots with human tumors in oncogenes including PIK3CA, KRAS, NRAS, BRAF, KIT and EGFR. Hotspot mutations with significant association to tumor type include NRAS G61R and PIK3CA H1047R in hemangiosarcoma, ERBB2 V659E in pulmonary carcinoma, and BRAF V588E (equivalent of V600E in humans) in urothelial carcinoma. Our findings better position canines as a translational model of human cancer to investigate a wide spectrum of targeted therapies.
Collapse
Affiliation(s)
- Lucas Rodrigues
- One Health Company, Inc, 530 Lytton Ave, 2nd Floor, Palo Alto, CA, 94301, USA.
| | - Joshua Watson
- Department of Biochemistry and Molecular Biology, Institute of Bioinformatics, University of Georgia, B304B Life Sciences Building, 120 Green Street, Athens, GA, 30602-7229, USA
| | - Yuan Feng
- Department of Biochemistry and Molecular Biology, Institute of Bioinformatics, University of Georgia, B304B Life Sciences Building, 120 Green Street, Athens, GA, 30602-7229, USA
| | - Benjamin Lewis
- One Health Company, Inc, 530 Lytton Ave, 2nd Floor, Palo Alto, CA, 94301, USA
| | - Garrett Harvey
- One Health Company, Inc, 530 Lytton Ave, 2nd Floor, Palo Alto, CA, 94301, USA
| | - Gerald Post
- One Health Company, Inc, 530 Lytton Ave, 2nd Floor, Palo Alto, CA, 94301, USA
| | - Kate Megquier
- One Health Company, Inc, 530 Lytton Ave, 2nd Floor, Palo Alto, CA, 94301, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Michelle E White
- One Health Company, Inc, 530 Lytton Ave, 2nd Floor, Palo Alto, CA, 94301, USA
| | - Lindsay Lambert
- One Health Company, Inc, 530 Lytton Ave, 2nd Floor, Palo Alto, CA, 94301, USA
| | - Aubrey Miller
- One Health Company, Inc, 530 Lytton Ave, 2nd Floor, Palo Alto, CA, 94301, USA
| | - Christina Lopes
- One Health Company, Inc, 530 Lytton Ave, 2nd Floor, Palo Alto, CA, 94301, USA
| | - Shaying Zhao
- Department of Biochemistry and Molecular Biology, Institute of Bioinformatics, University of Georgia, B304B Life Sciences Building, 120 Green Street, Athens, GA, 30602-7229, USA.
| |
Collapse
|
27
|
Pimenta J, Pires I, Prada J, Cotovio M. E-Cadherin Immunostaining in Equine Melanocytic Tumors. Animals (Basel) 2023; 13:2216. [PMID: 37444014 DOI: 10.3390/ani13132216] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Melanocytic tumors are an important neoplastic disease in human and veterinary medicine, presenting large differences regarding tumor behavior between species. In horses, these tumors present a prolonged benign behavior, with rare invasiveness and metastases. In humans and small animals, invasion and metastasis have been associated with an Epithelial-Mesenchymal Transition, where the loss of E-cadherin expression plays a key role in tumor progression. This process and the role of E-cadherin have not yet been evaluated in equine melanocytic tumors. This study aimed to assess the immunolabeling of E-cadherin in equine melanocytic tumors and relate this with clinicopathological variables. A total of 72 equine melanocytic tumors were classified as benign and malignant and evaluated by immunohistochemistry for E-cadherin expression. A different pattern of immunostaining was found, contrasting with other species. A total of 69.4% of tumors presented raised immunolabeling of E-cadherin, with 70.7% of melanomas remaining with high expression. The typical loss of immunostaining was not seen in malignant melanomas and no differences were found between benign and malignant melanomas regarding E-cadherin immunostaining. The high immunolabeling of E-cadherin may contribute to the low invasiveness of these tumors, and it is in accordance with the benign behavior of equine melanoma and with the genetic factors associated with its development.
Collapse
Affiliation(s)
- José Pimenta
- Veterinary Sciences Department, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
- CECAV-Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
| | - Isabel Pires
- Veterinary Sciences Department, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
- CECAV-Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
| | - Justina Prada
- Veterinary Sciences Department, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
- CECAV-Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
| | - Mário Cotovio
- Veterinary Sciences Department, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
- CECAV-Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
| |
Collapse
|
28
|
Gniadecki R, O’Keefe S, Hennessey D, Iyer A. Is Cutaneous T-Cell Lymphoma Caused by Ultraviolet Radiation? A Comparison of UV Mutational Signatures in Malignant Melanoma and Mycosis Fungoides. Cells 2023; 12:1616. [PMID: 37371087 PMCID: PMC10297369 DOI: 10.3390/cells12121616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/05/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Ultraviolet (UV) radiation is a strong environmental carcinogen responsible for the pathogenesis of most skin cancers, including malignant melanoma (MM) and non-melanoma (keratinocyte) skin cancers. The carcinogenic role of UV was firmly established based on epidemiological evidence and molecular findings of the characteristic mutation signatures which occur during the excision repair of cyclobutane pyrimidine dimers and 6,4-photoproducts. The role of UV in the pathogenesis of mycosis fungoides (MF), the most common type of primary cutaneous T-cell lymphoma, remains controversial. Here, we performed whole-exome sequencing of 61 samples of MF cells microdissected from cutaneous lesions, and compared their mutational signatures to 340 MMs. The vast majority of MM mutations had a typical UV mutational signature (SBS 7, SBS 38, or DSB 1), underscoring the key role of ultraviolet as a mutagen. In contrast, the SBS 7 signature in MF comprised < 5% of all mutations. SBS 7 was higher in the intraepidermal MF cells (when compared to the dermal cells) and in the cells from tumors as compared to that in early-stage plaques. In conclusion, our data do not support the pathogenic role of UV in the pathogenesis of MF and suggest that the UV mutations are the result of the cumulative environmental ultraviolet exposure of cutaneous lesions rather than an early mutagenic event.
Collapse
Affiliation(s)
- Robert Gniadecki
- Division of Dermatology, University of Alberta, Edmonton, AB T6G 2G3, Canada; (S.O.); (D.H.); (A.I.)
| | | | | | | |
Collapse
|
29
|
AbdulJabbar K, Castillo SP, Hughes K, Davidson H, Boddy AM, Abegglen LM, Minoli L, Iussich S, Murchison EP, Graham TA, Spiro S, Maley CC, Aresu L, Palmieri C, Yuan Y. Bridging clinic and wildlife care with AI-powered pan-species computational pathology. Nat Commun 2023; 14:2408. [PMID: 37100774 PMCID: PMC10133243 DOI: 10.1038/s41467-023-37879-x] [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: 03/21/2022] [Accepted: 04/04/2023] [Indexed: 04/28/2023] Open
Abstract
Cancers occur across species. Understanding what is consistent and varies across species can provide new insights into cancer initiation and evolution, with significant implications for animal welfare and wildlife conservation. We build a pan-species cancer digital pathology atlas (panspecies.ai) and conduct a pan-species study of computational comparative pathology using a supervised convolutional neural network algorithm trained on human samples. The artificial intelligence algorithm achieves high accuracy in measuring immune response through single-cell classification for two transmissible cancers (canine transmissible venereal tumour, 0.94; Tasmanian devil facial tumour disease, 0.88). In 18 other vertebrate species (mammalia = 11, reptilia = 4, aves = 2, and amphibia = 1), accuracy (range 0.57-0.94) is influenced by cell morphological similarity preserved across different taxonomic groups, tumour sites, and variations in the immune compartment. Furthermore, a spatial immune score based on artificial intelligence and spatial statistics is associated with prognosis in canine melanoma and prostate tumours. A metric, named morphospace overlap, is developed to guide veterinary pathologists towards rational deployment of this technology on new samples. This study provides the foundation and guidelines for transferring artificial intelligence technologies to veterinary pathology based on understanding of morphological conservation, which could vastly accelerate developments in veterinary medicine and comparative oncology.
Collapse
Affiliation(s)
- Khalid AbdulJabbar
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Simon P Castillo
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Katherine Hughes
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, UK
| | - Hannah Davidson
- Zoological Society of London, London, UK
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Sq, London, UK
| | - Amy M Boddy
- Department of Anthropology, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Lisa M Abegglen
- Department of Pediatrics and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- PEEL Therapeutics, Inc., Salt Lake City, UT, USA
| | - Lucia Minoli
- Department of Veterinary Sciences, University of Turin, 10095, Grugliasco, Italy
| | - Selina Iussich
- Department of Veterinary Sciences, University of Turin, 10095, Grugliasco, Italy
| | - Elizabeth P Murchison
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, UK
| | - Trevor A Graham
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Sq, London, UK
| | | | - Carlo C Maley
- Arizona Cancer Evolution Center, Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Luca Aresu
- Department of Veterinary Sciences, University of Turin, 10095, Grugliasco, Italy
| | - Chiara Palmieri
- School of Veterinary Science, The University of Queensland, 4343, Gatton, QLD, Australia
| | - Yinyin Yuan
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK.
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
30
|
Pinto C, Aluai-Cunha C, Santos A. The human and animals' malignant melanoma: comparative tumor models and the role of microbiome in dogs and humans. Melanoma Res 2023; 33:87-103. [PMID: 36662668 DOI: 10.1097/cmr.0000000000000880] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Currently, the most progressively occurring incident cancer is melanoma. The mouse is the most popular model in human melanoma research given its various benefits as a laboratory animal. Nevertheless, unlike humans, mice do not develop melanoma spontaneously, so they need to be genetically manipulated. In opposition, there are several reports of other animals, ranging from wild to domesticated animals, that spontaneously develop melanoma and that have cancer pathways that are similar to those of humans. The influence of the gut microbiome on health and disease is being the aim of many recent studies. It has been proven that the microbiome is a determinant of the host's immune status and disease prevention. In human medicine, there is increasing evidence that changes in the microbiome influences malignant melanoma progression and response to therapy. There are several similarities between some animals and human melanoma, especially between canine and human oral malignant melanoma as well as between the gut microbiome of both species. However, microbiome studies are scarce in veterinary medicine, especially in the oncology field. Future studies need to address the relevance of gut and tissue microbiome for canine malignant melanoma development, which results will certainly benefit both species in the context of translational medicine.
Collapse
Affiliation(s)
- Catarina Pinto
- Department of Veterinary Clinics, Institute of Biomedical Sciences Abel Salazar of the University of Porto (ICBAS-UP)
| | - Catarina Aluai-Cunha
- Department of Veterinary Clinics, Institute of Biomedical Sciences Abel Salazar of the University of Porto (ICBAS-UP)
| | - Andreia Santos
- Department of Veterinary Clinics, Institute of Biomedical Sciences Abel Salazar of the University of Porto (ICBAS-UP)
- Animal Science and Study Centre (CECA), Food and Agragrian Sciences and Technologies Institute (ICETA), Apartado, Porto, Portugal
| |
Collapse
|
31
|
Oh JH, Cho JY. Comparative oncology: overcoming human cancer through companion animal studies. Exp Mol Med 2023; 55:725-734. [PMID: 37009802 PMCID: PMC10167357 DOI: 10.1038/s12276-023-00977-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/20/2023] [Accepted: 01/25/2023] [Indexed: 04/04/2023] Open
Abstract
Comparative oncology is a field of study that has been recently adopted for studying cancer and developing cancer therapies. Companion animals such as dogs can be used to evaluate novel biomarkers or anticancer targets before clinical translation. Thus, the value of canine models is increasing, and numerous studies have been conducted to analyze similarities and differences between many types of spontaneously occurring cancers in canines and humans. A growing number of canine cancer models as well as research-grade reagents for these models are becoming available, leading to substantial growth in comparative oncology research spanning from basic science to clinical trials. In this review, we summarize comparative oncology studies that have been conducted on the molecular landscape of various canine cancers and highlight the importance of the integration of comparative biology into cancer research.
Collapse
Affiliation(s)
- Ji Hoon Oh
- Department of Biochemistry, Brain Korea 21 Project and Research Institute for Veterinary Science, Seoul National University College of Veterinary Medicine, Seoul, 08826, Republic of Korea
- Comparative Medicine Disease Research Center, Seoul National University, Seoul, 08826, Republic of Korea
| | - Je-Yoel Cho
- Department of Biochemistry, Brain Korea 21 Project and Research Institute for Veterinary Science, Seoul National University College of Veterinary Medicine, Seoul, 08826, Republic of Korea.
- Comparative Medicine Disease Research Center, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
32
|
Koo K, Wuenschmann A, Rendahl A, Song KY, Forster C, Wolf-Ringwall A, Borgatti A, Giubellino A. Expression and Prognostic Evaluation of the Receptor Tyrosine Kinase MET in Canine Malignant Melanoma. Vet Sci 2023; 10:vetsci10040249. [PMID: 37104404 PMCID: PMC10144085 DOI: 10.3390/vetsci10040249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
The overexpression and activation of the MET receptor tyrosine kinase has been identified in many human malignancies, but its role in canine cancer has only been minimally investigated. In this study we evaluated the expression of MET in two canine malignant melanoma (CMM) cell lines as well as in 30 CMM tissue samples that were collected from the clinical service at our institution. We were able to confirm the expression of the MET protein in both melanoma cell lines, and we demonstrated MET activation by its ligand, HGF, through phosphorylation, in Western blot analysis. We were also able to demonstrate, by immunohistochemistry, the expression of MET in 63% of the tumor tissue samples analyzed, with the majority demonstrating a relatively low expression profile. We then evaluated the association of MET expression scores with histologic parameters, metastasis, and survival. While statistically significant associations were not found across these parameters, an inverse relationship between MET expression levels and time to lymph node versus distant metastasis was suggested in our cohort. These findings may require assessment in a larger group of specimens to further evaluate the role of MET expression in the homing of metastasis in lymph nodes versus that in distant organs.
Collapse
|
33
|
Feng Y, Hess PR, Tompkins SM, Hildebrand WH, Zhao S. A Kmer-based paired-end read de novo assembler and genotyper for canine MHC class I genotyping. iScience 2023; 26:105996. [PMID: 36798440 PMCID: PMC9926114 DOI: 10.1016/j.isci.2023.105996] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/28/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
The major histocompatibility complex class I (MHC-I) genes are highly polymorphic. MHC-I genotyping is required for determining the peptide epitopes available to an individual's T-cell repertoire. Current genotyping software tools do not work for the dog, due to very limited known canine alleles. To address this, we developed a Kmer-based paired-end read (KPR) de novo assembler and genotyper, which assemble paired-end RNA-seq reads from MHC-I regions into contigs, and then genotype each contig and estimate its expression level. KPR tools outperform other popular software examined in typing new alleles. We used KPR tools to successfully genotype152 dogs from a published dataset. The study discovers 33 putative new alleles, finds dominant alleles in 4 dog breeds, and builds allele diversity and expression landscapes among the 152 dogs. Our software meets a significant need in biomedical research.
Collapse
Affiliation(s)
- Yuan Feng
- Department of Biochemistry and Molecular Biology, Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA
| | - Paul R. Hess
- Department of Clinical Sciences, North Carolina State University, College of Veterinary Medicine, Raleigh, NC 27607, USA
| | - Stephen M. Tompkins
- Center for Vaccines and Immunology, University of Georgia, UGA, Athens, GA 30602, USA
| | - William H. Hildebrand
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Shaying Zhao
- Department of Biochemistry and Molecular Biology, Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
34
|
Forsberg EMV, Riise R, Saellström S, Karlsson J, Alsén S, Bucher V, Hemminki AE, Olofsson Bagge R, Ny L, Nilsson LM, Rönnberg H, Nilsson JA. Treatment with Anti-HER2 Chimeric Antigen Receptor Tumor-Infiltrating Lymphocytes (CAR-TILs) Is Safe and Associated with Antitumor Efficacy in Mice and Companion Dogs. Cancers (Basel) 2023; 15:cancers15030648. [PMID: 36765608 PMCID: PMC9913266 DOI: 10.3390/cancers15030648] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
Patients with metastatic melanoma have a historically poor prognosis, but recent advances in treatment options, including targeted therapy and immunotherapy, have drastically improved the outcomes for some of these patients. However, not all patients respond to available treatments, and around 50% of patients with metastatic cutaneous melanoma and almost all patients with metastases of uveal melanoma die of their disease. Thus, there is a need for novel treatment strategies for patients with melanoma that do not benefit from the available therapies. Chimeric antigen receptor-expressing T (CAR-T) cells are largely unexplored in melanoma. Traditionally, CAR-T cells have been produced by transducing blood-derived T cells with a virus expressing CAR. However, tumor-infiltrating lymphocytes (TILs) can also be engineered to express CAR, and such CAR-TILs could be dual-targeting. To this end, tumor samples and autologous TILs from metastasized human uveal and cutaneous melanoma were expanded in vitro and transduced with a lentiviral vector encoding an anti-HER2 CAR construct. When infused into patient-derived xenograft (PDX) mouse models carrying autologous tumors, CAR-TILs were able to eradicate melanoma, even in the absence of antigen presentation by HLA. To advance this concept to the clinic and assess its safety in an immune-competent and human-patient-like setting, we treated four companion dogs with autologous anti-HER2 CAR-TILs. We found that these cells were tolerable and showed signs of anti-tumor activity. Taken together, CAR-TIL therapy is a promising avenue for broadening the tumor-targeting capacity of TILs in patients with checkpoint immunotherapy-resistant melanoma.
Collapse
Affiliation(s)
- Elin M. V. Forsberg
- Sahlgrenska Translational Melanoma Group, Sahlgrenska Center for Cancer Research, Departments of Surgery and Oncology, Institute of Clinical Sciences, University of Gothenburg, Sahlgrenska University Hospital, 40530 Gothenburg, Sweden
| | - Rebecca Riise
- Sahlgrenska Translational Melanoma Group, Sahlgrenska Center for Cancer Research, Departments of Surgery and Oncology, Institute of Clinical Sciences, University of Gothenburg, Sahlgrenska University Hospital, 40530 Gothenburg, Sweden
| | - Sara Saellström
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, 75007 Uppsala, Sweden
| | - Joakim Karlsson
- Sahlgrenska Translational Melanoma Group, Sahlgrenska Center for Cancer Research, Departments of Surgery and Oncology, Institute of Clinical Sciences, University of Gothenburg, Sahlgrenska University Hospital, 40530 Gothenburg, Sweden
- Harry Perkins Institute of Medical Research, University of Western Australia, Perth, WA 6009, Australia
| | - Samuel Alsén
- Sahlgrenska Translational Melanoma Group, Sahlgrenska Center for Cancer Research, Departments of Surgery and Oncology, Institute of Clinical Sciences, University of Gothenburg, Sahlgrenska University Hospital, 40530 Gothenburg, Sweden
| | - Valentina Bucher
- Sahlgrenska Translational Melanoma Group, Sahlgrenska Center for Cancer Research, Departments of Surgery and Oncology, Institute of Clinical Sciences, University of Gothenburg, Sahlgrenska University Hospital, 40530 Gothenburg, Sweden
| | - Akseli E. Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
- Department of Oncology, Comprehensive Cancer Centre, Helsinki University Hospital, 00290 Helsinki, Finland
| | - Roger Olofsson Bagge
- Sahlgrenska Translational Melanoma Group, Sahlgrenska Center for Cancer Research, Departments of Surgery and Oncology, Institute of Clinical Sciences, University of Gothenburg, Sahlgrenska University Hospital, 40530 Gothenburg, Sweden
| | - Lars Ny
- Sahlgrenska Translational Melanoma Group, Sahlgrenska Center for Cancer Research, Departments of Surgery and Oncology, Institute of Clinical Sciences, University of Gothenburg, Sahlgrenska University Hospital, 40530 Gothenburg, Sweden
| | - Lisa M. Nilsson
- Sahlgrenska Translational Melanoma Group, Sahlgrenska Center for Cancer Research, Departments of Surgery and Oncology, Institute of Clinical Sciences, University of Gothenburg, Sahlgrenska University Hospital, 40530 Gothenburg, Sweden
- Harry Perkins Institute of Medical Research, University of Western Australia, Perth, WA 6009, Australia
| | - Henrik Rönnberg
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, 75007 Uppsala, Sweden
| | - Jonas A. Nilsson
- Sahlgrenska Translational Melanoma Group, Sahlgrenska Center for Cancer Research, Departments of Surgery and Oncology, Institute of Clinical Sciences, University of Gothenburg, Sahlgrenska University Hospital, 40530 Gothenburg, Sweden
- Harry Perkins Institute of Medical Research, University of Western Australia, Perth, WA 6009, Australia
- Correspondence: or ; Tel.: +61-08-6151-0979
| |
Collapse
|
35
|
Equine Melanocytic Tumors: A Narrative Review. Animals (Basel) 2023; 13:ani13020247. [PMID: 36670786 PMCID: PMC9855132 DOI: 10.3390/ani13020247] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/04/2023] [Accepted: 01/08/2023] [Indexed: 01/12/2023] Open
Abstract
Adult grey horses have a high incidence of melanocytic tumors. This article narratively reviews the role of some genetic features related to melanoma formation in horses, such as STX17 mutation, ASIP or MITF alterations, and the link between the graying process and the development of these tumors. A clear system of clinical and pathological classification of melanocytic tumors in naevus, dermal melanoma, dermal melanomatosis and anaplastic malignant melanoma is provided. Clinical and laboratorial methods of diagnosing are listed, with fine needle aspiration and histopathology being the most relevant. Relevance is given to immunohistochemistry, describing potentially important diagnostic biomarkers such as RACK1 and PNL2. Different therapeutical options available for equine practitioners are mentioned, with surgery, chemotherapy and electroporation being the most common. This article also elucidatesnew fields of research, perspectives, and new therapeutic targets, such as CD47, PD-1 and COX-2 biomarkers.
Collapse
|
36
|
Stevenson VB, Klahn S, LeRoith T, Huckle WR. Canine melanoma: A review of diagnostics and comparative mechanisms of disease and immunotolerance in the era of the immunotherapies. Front Vet Sci 2023; 9:1046636. [PMID: 36686160 PMCID: PMC9853198 DOI: 10.3389/fvets.2022.1046636] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/12/2022] [Indexed: 01/09/2023] Open
Abstract
Melanomas in humans and dogs are highly malignant and resistant to therapy. Since the first development of immunotherapies, interest in how the immune system interacts within the tumor microenvironment and plays a role in tumor development, progression, or remission has increased. Of major importance are tumor-infiltrating lymphocytes (TILs) where distribution and cell frequencies correlate with survival and therapeutic outcomes. Additionally, efforts have been made to identify subsets of TILs populations that can contribute to a tumor-promoting or tumor-inhibiting environment, such as the case with T regulatory cells versus CD8 T cells. Furthermore, cancerous cells have the capacity to express certain inhibitory checkpoint molecules, including CTLA-4, PD-L1, PD-L2, that can suppress the immune system, a property associated with poor prognosis, a high rate of recurrence, and metastasis. Comparative oncology brings insights to comprehend the mechanisms of tumorigenesis and immunotolerance in humans and dogs, contributing to the development of new therapeutic agents that can modulate the immune response against the tumor. Therapies that target signaling pathways such as mTOR and MEK/ERK that are upregulated in cancer, or immunotherapies with different approaches such as CAR-T cells engineered for specific tumor-associated antigens, DNA vaccines using human tyrosinase or CGSP-4 antigen, anti-PD-1 or -PD-L1 monoclonal antibodies that intercept their binding inhibiting the suppression of the T cells, and lymphokine-activated killer cells are already in development for treating canine tumors. This review provides concise and recent information about diagnosis, comparative mechanisms of tumor development and progression, and the current status of immunotherapies directed toward canine melanoma.
Collapse
Affiliation(s)
- Valentina B. Stevenson
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Shawna Klahn
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
- Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Tanya LeRoith
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - William R. Huckle
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
37
|
Birkeälv S, Harland M, Matsuyama LSAS, Rashid M, Mehta I, Laye JP, Haase K, Mell T, Iyer V, Robles‐Espinoza CD, McDermott U, van Loo P, Kuijjer ML, Possik PA, Maria Engler SS, Bishop DT, Newton‐Bishop J, Adams DJ. Mutually exclusive genetic interactions and gene essentiality shape the genomic landscape of primary melanoma. J Pathol 2023; 259:56-68. [PMID: 36219477 PMCID: PMC10098817 DOI: 10.1002/path.6019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/02/2022] [Accepted: 09/28/2022] [Indexed: 11/09/2022]
Abstract
Melanoma is a heterogenous malignancy with an unpredictable clinical course. Most patients who present in the clinic are diagnosed with primary melanoma, yet large-scale sequencing efforts have focused primarily on metastatic disease. In this study we sequence-profiled 524 American Joint Committee on Cancer Stage I-III primary tumours. Our analysis of these data reveals recurrent driver mutations, mutually exclusive genetic interactions, where two genes were never or rarely co-mutated, and an absence of co-occurring genetic events. Further, we intersected copy number calls from our primary melanoma data with whole-genome CRISPR screening data to identify the transcription factor interferon regulatory factor 4 (IRF4) as a melanoma-associated dependency. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Sofia Birkeälv
- Wellcome Sanger InstituteWellcome Trust Genome CampusCambridgeUK
| | - Mark Harland
- Division of Haematology and ImmunologyUniversity of Leeds School of MedicineLeedsUK
| | - Larissa Satiko Alcantara Sekimoto Matsuyama
- Wellcome Sanger InstituteWellcome Trust Genome CampusCambridgeUK
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical SciencesUniversity of Sao PauloSao PauloBrazil
| | - Mamun Rashid
- Wellcome Sanger InstituteWellcome Trust Genome CampusCambridgeUK
| | - Ishan Mehta
- Wellcome Sanger InstituteWellcome Trust Genome CampusCambridgeUK
| | - Jonathan P Laye
- Division of Haematology and ImmunologyUniversity of Leeds School of MedicineLeedsUK
| | | | - Tracey Mell
- Division of Haematology and ImmunologyUniversity of Leeds School of MedicineLeedsUK
| | - Vivek Iyer
- Wellcome Sanger InstituteWellcome Trust Genome CampusCambridgeUK
| | - Carla Daniela Robles‐Espinoza
- Wellcome Sanger InstituteWellcome Trust Genome CampusCambridgeUK
- Laboratorio Internacional de Investigación sobre el Genoma HumanoUniversidad Nacional Autónoma de México, Campus JuriquillaSantiago de QuerétaroMexico
| | - Ultan McDermott
- Wellcome Sanger InstituteWellcome Trust Genome CampusCambridgeUK
| | | | - Marieke L Kuijjer
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, Faculty of MedicineUniversity of OsloOsloNorway
- Department of Pathology and Leiden Center for Computational OncologyLeiden University Medical CenterLeidenthe Netherlands
| | - Patricia A Possik
- Division of Experimental and Translational ResearchBrazilian National Cancer InstituteRio de JaneiroBrazil
| | - Silvya Stuchi Maria Engler
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical SciencesUniversity of Sao PauloSao PauloBrazil
| | - D Timothy Bishop
- Division of Haematology and ImmunologyUniversity of Leeds School of MedicineLeedsUK
| | - Julia Newton‐Bishop
- Division of Haematology and ImmunologyUniversity of Leeds School of MedicineLeedsUK
| | - David J Adams
- Wellcome Sanger InstituteWellcome Trust Genome CampusCambridgeUK
| |
Collapse
|
38
|
Pan-cancer landscape of AID-related mutations, composite mutations, and their potential role in the ICI response. NPJ Precis Oncol 2022; 6:89. [PMID: 36456685 PMCID: PMC9715662 DOI: 10.1038/s41698-022-00331-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 11/02/2022] [Indexed: 12/03/2022] Open
Abstract
Activation-induced cytidine deaminase, AICDA or AID, is a driver of somatic hypermutation and class-switch recombination in immunoglobulins. In addition, this deaminase belonging to the APOBEC family may have off-target effects genome-wide, but its effects at pan-cancer level are not well elucidated. Here, we used different pan-cancer datasets, totaling more than 50,000 samples analyzed by whole-genome, whole-exome, or targeted sequencing. AID mutations are present at pan-cancer level with higher frequency in hematological cancers and higher presence at transcriptionally active TAD domains. AID synergizes initial hotspot mutations by a second composite mutation. AID mutational load was found to be independently associated with a favorable outcome in immune-checkpoint inhibitors (ICI) treated patients across cancers after analyzing 2000 samples. Finally, we found that AID-related neoepitopes, resulting from mutations at more frequent hotspots if compared to other mutational signatures, enhance CXCL13/CCR5 expression, immunogenicity, and T-cell exhaustion, which may increase ICI sensitivity.
Collapse
|
39
|
Yi Z, Gao Y, Yu F, Zhu Y, Liu H, Li J, Murua Escobar H. Interventions for treatment of cutaneous melanoma in horses: a structured literature review. Vet Res Commun 2022; 47:347-360. [PMID: 36329228 DOI: 10.1007/s11259-022-10023-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
Abstract
Several therapies have been developed to treat equine cutaneous melanoma, but formal comparisons among different treatment options are currently unavailable. It was our intent to assess the efficacy of different treatment protocols and the quality of the studies based on the original published data, and summarize the knowledge concerning the outcome after equine cutaneous melanoma management. This structured review followed PRISMA procedure to search for treatment protocols on equine cutaneous melanoma published from 1960 until June 2021. Studies were assessed for the risk of bias. A descriptive analysis was performed, considering the disease control rate, the recurrence rate of the tumor, comorbidities, need for anesthesia, and horses' welfare. Twenty-three studies were included, from which the treatment outcomes of 173 horses were assessed. The homogeneity of the included trials was low. The percentages of each treatment arm achieving a partial response and curative effects accounted for 93.1% (surgical intervention), 90% (medication), and 39.4% (immunotherapies), respectively. A variable efficacy of different therapies of equine cutaneous melanoma was observed. Surgical intervention performed the best from the perspective of local antitumor effects alone. This literature review and descriptive analysis can serve as a source to assist in designing quality therapy research and can potentially aid in providing a clinical treatment reference for equine cutaneous melanoma.
Collapse
Affiliation(s)
- Ziwen Yi
- Equine Clinical Diagnostic Center, College of Veterinary Medicine, China Agriculture University, No. 2 Yuanmingyuan West Road, 100094, Beijing, China
| | - Yu Gao
- University of Veterinary Medicine Hannover Foundation, Hannover, Germany
| | - Feng Yu
- College of Veterinary Medicine, China Agriculture University, Beijing, China
| | - Yiping Zhu
- Equine Clinical Diagnostic Center, College of Veterinary Medicine, China Agriculture University, No. 2 Yuanmingyuan West Road, 100094, Beijing, China
| | - Haoqian Liu
- Equine Clinical Diagnostic Center, College of Veterinary Medicine, China Agriculture University, No. 2 Yuanmingyuan West Road, 100094, Beijing, China
| | - Jing Li
- Equine Clinical Diagnostic Center, College of Veterinary Medicine, China Agriculture University, No. 2 Yuanmingyuan West Road, 100094, Beijing, China.
| | - Hugo Murua Escobar
- Department of Hematology, Oncology and Palliative Medicine, Department of Medicine III, Rostock University Medical Center, Ernst Heydemann Street No. 6, 18057, Rostock, Germany.
| |
Collapse
|
40
|
Gao Y, Jiang G, Yang W, Jin W, Gong J, Xu X, Niu X. Animal-SNPAtlas: a comprehensive SNP database for multiple animals. Nucleic Acids Res 2022; 51:D816-D826. [PMID: 36300636 PMCID: PMC9825464 DOI: 10.1093/nar/gkac954] [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: 08/11/2022] [Revised: 10/06/2022] [Accepted: 10/26/2022] [Indexed: 01/30/2023] Open
Abstract
Single-nucleotide polymorphisms (SNPs) as the most important type of genetic variation are widely used in describing population characteristics and play vital roles in animal genetics and breeding. Large amounts of population genetic variation resources and tools have been developed in human, which provided solid support for human genetic studies. However, compared with human, the development of animal genetic variation databases was relatively slow, which limits the genetic researches in these animals. To fill this gap, we systematically identified ∼ 499 million high-quality SNPs from 4784 samples of 20 types of animals. On that basis, we annotated the functions of SNPs, constructed high-density reference panels and calculated genome-wide linkage disequilibrium (LD) matrixes. We further developed Animal-SNPAtlas, a user-friendly database (http://gong_lab.hzau.edu.cn/Animal_SNPAtlas/) which includes high-quality SNP datasets and several support tools for multiple animals. In Animal-SNPAtlas, users can search the functional annotation of SNPs, perform online genotype imputation, explore and visualize LD information, browse variant information using the genome browser and download SNP datasets for each species. With the massive SNP datasets and useful tools, Animal-SNPAtlas will be an important fundamental resource for the animal genomics, genetics and breeding community.
Collapse
Affiliation(s)
| | | | - Wenqian Yang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, P. R. China
| | - Weiwei Jin
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, P. R. China
| | - Jing Gong
- To whom correspondence should be addressed. Tel: +86 27 87285085; Fax: +86 27 87285085;
| | - Xuewen Xu
- Correspondence may also be addressed to Xuewen Xu. Tel: +86 27 87285085; Fax: +86 27 87285085;
| | - Xiaohui Niu
- Correspondence may also be addressed to Xiaohui Niu. Tel: +86 27 87285085; Fax: +86 27 87285085;
| |
Collapse
|
41
|
Pinard CJ, Lagree A, Lu FI, Klein J, Oblak ML, Salgado R, Cardenas JCP, Brunetti B, Muscatello LV, Sarli G, Foschini MP, Hardas A, Castillo SP, AbdulJabbar K, Yuan Y, Moore DA, Tran WT. Comparative Evaluation of Tumor-Infiltrating Lymphocytes in Companion Animals: Immuno-Oncology as a Relevant Translational Model for Cancer Therapy. Cancers (Basel) 2022; 14:5008. [PMID: 36291791 PMCID: PMC9599753 DOI: 10.3390/cancers14205008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/04/2022] [Accepted: 10/08/2022] [Indexed: 11/16/2022] Open
Abstract
Despite the important role of preclinical experiments to characterize tumor biology and molecular pathways, there are ongoing challenges to model the tumor microenvironment, specifically the dynamic interactions between tumor cells and immune infiltrates. Comprehensive models of host-tumor immune interactions will enhance the development of emerging treatment strategies, such as immunotherapies. Although in vitro and murine models are important for the early modelling of cancer and treatment-response mechanisms, comparative research studies involving veterinary oncology may bridge the translational pathway to human studies. The natural progression of several malignancies in animals exhibits similar pathogenesis to human cancers, and previous studies have shown a relevant and evaluable immune system. Veterinary oncologists working alongside oncologists and cancer researchers have the potential to advance discovery. Understanding the host-tumor-immune interactions can accelerate drug and biomarker discovery in a clinically relevant setting. This review presents discoveries in comparative immuno-oncology and implications to cancer therapy.
Collapse
Affiliation(s)
- Christopher J. Pinard
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
- Odette Cancer Program, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Radiogenomics Laboratory, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
| | - Andrew Lagree
- Odette Cancer Program, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Radiogenomics Laboratory, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
| | - Fang-I Lu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jonathan Klein
- Department of Radiation Oncology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA
| | - Michelle L. Oblak
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Roberto Salgado
- Division of Research, Peter MacCallum Cancer Centre, Melbourne 3000, Australia
- Department of Pathology, GZA-ZNA Hospitals, 2610 Antwerp, Belgium
| | | | - Barbara Brunetti
- Department of Veterinary Medical Sciences, University of Bologna, 40064 Ozzano dell’Emilia, Italy
| | - Luisa Vera Muscatello
- Department of Veterinary Medical Sciences, University of Bologna, 40064 Ozzano dell’Emilia, Italy
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences, University of Bologna, 40064 Ozzano dell’Emilia, Italy
| | - Maria Pia Foschini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40127 Bologna, Italy
| | - Alexandros Hardas
- Department of Pathobiology & Population Sciences, The Royal Veterinary College, Hertfordshire AL9 7TA, UK
| | - Simon P. Castillo
- Centre for Evolution and Cancer, The Institute of Cancer Research, London SM2 5NG, UK
- Division of Molecular Pathology, The Institute of Cancer Research, London SM2 5NG, UK
| | - Khalid AbdulJabbar
- Centre for Evolution and Cancer, The Institute of Cancer Research, London SM2 5NG, UK
- Division of Molecular Pathology, The Institute of Cancer Research, London SM2 5NG, UK
| | - Yinyin Yuan
- Centre for Evolution and Cancer, The Institute of Cancer Research, London SM2 5NG, UK
- Division of Molecular Pathology, The Institute of Cancer Research, London SM2 5NG, UK
| | - David A. Moore
- Department of Pathology, UCL Cancer Institute, London WC1E 6DD, UK
- University College Hospitals NHS Trust, London NW1 2PG, UK
| | - William T. Tran
- Odette Cancer Program, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Radiogenomics Laboratory, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON M5T 1P5, Canada
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Temerty Centre for AI Research and Education in Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
42
|
Yu Y, Manders F, Grinwis GCM, Groenen MAM, Crooijmans RPMA. A recurrent somatic missense mutation in GNAS gene identified in familial thyroid follicular cell carcinomas in German longhaired pointer dogs. BMC Genomics 2022; 23:669. [PMID: 36151521 PMCID: PMC9508735 DOI: 10.1186/s12864-022-08885-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 08/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We previously reported a familial thyroid follicular cell carcinoma (FCC) in a large number of Dutch German longhaired pointers and identified two deleterious germline mutations in the TPO gene associated with disease predisposition. However, the somatic mutation profile of the FCC in dogs has not been investigated at a genome-wide scale. RESULTS Herein, we comprehensively investigated the somatic mutations that potentially contribute to the inherited tumor formation and progression using high depth whole-genome sequencing. A GNAS p.A204D missense mutation was identified in 4 out of 7 FCC tumors by whole-genome sequencing and in 20 out of 32 dogs' tumors by targeted sequencing. In contrast to this, in the human TC, mutations in GNAS gene have lower prevalence. Meanwhile, the homologous somatic mutation in humans has not been reported. These findings suggest a difference in the somatic mutation landscape between TC in these dogs and human TC. Moreover, tumors with the GNAS p.A204D mutation had a significantly lower somatic mutation burden in these dogs. Somatic structural variant and copy number alterations were also investigated, but no potential driver event was identified. CONCLUSION This study provides novel insight in the molecular mechanism of thyroid carcinoma development in dogs. German longhaired pointers carrying GNAS mutations in the tumor may be used as a disease model for the development and testing of novel therapies to kill the tumor with somatic mutations in the GNAS gene.
Collapse
Affiliation(s)
- Yun Yu
- Animal Breeding and Genomics, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB, Wageningen, The Netherlands
| | - Freek Manders
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584CS, Utrecht, The Netherlands
| | - Guy C M Grinwis
- Department of Biomolecular Health Sciences, Division of Pathology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, Utrecht, The Netherlands
| | - Martien A M Groenen
- Animal Breeding and Genomics, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB, Wageningen, The Netherlands
| | - Richard P M A Crooijmans
- Animal Breeding and Genomics, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB, Wageningen, The Netherlands.
| |
Collapse
|
43
|
Giannuzzi D, Marconato L, Fanelli A, Licenziato L, De Maria R, Rinaldi A, Rotta L, Rouquet N, Birolo G, Fariselli P, Mensah AA, Bertoni F, Aresu L. The genomic landscape of canine diffuse large B-cell lymphoma identifies distinct subtypes with clinical and therapeutic implications. Lab Anim (NY) 2022; 51:191-202. [PMID: 35726023 DOI: 10.1038/s41684-022-00998-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 05/13/2022] [Indexed: 12/13/2022]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoid neoplasm in dogs and in humans. It is characterized by a remarkable degree of clinical heterogeneity that is not completely elucidated by molecular data. This poses a major barrier to understanding the disease and its response to therapy, or when treating dogs with DLBCL within clinical trials. We performed an integrated analysis of exome (n = 77) and RNA sequencing (n = 43) data in a cohort of canine DLBCL to define the genetic landscape of this tumor. A wide range of signaling pathways and cellular processes were found in common with human DLBCL, but the frequencies of the most recurrently mutated genes (TRAF3, SETD2, POT1, TP53, MYC, FBXW7, DDX3X and TBL1XR1) differed. We developed a prognostic model integrating exonic variants and clinical and transcriptomic features to predict the outcome in dogs with DLBCL. These results comprehensively define the genetic drivers of canine DLBCL and can be prospectively utilized to identify new therapeutic opportunities.
Collapse
Affiliation(s)
- Diana Giannuzzi
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padua, Padua, Italy
| | - Laura Marconato
- Department of Veterinary Medical Science, University of Bologna, Ozzano dell'Emilia, Bologna, Italy
| | - Antonella Fanelli
- Department of Veterinary Sciences, University of Turin, Grugliasco, Turin, Italy
| | - Luca Licenziato
- Department of Veterinary Sciences, University of Turin, Grugliasco, Turin, Italy
| | - Raffaella De Maria
- Department of Veterinary Sciences, University of Turin, Grugliasco, Turin, Italy
| | - Andrea Rinaldi
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
| | - Luca Rotta
- Department of Experimental Oncology, European Institute of Oncology (IEO), Milan, Italy
| | | | - Giovanni Birolo
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Piero Fariselli
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Afua A Mensah
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland. .,Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.
| | - Luca Aresu
- Department of Veterinary Sciences, University of Turin, Grugliasco, Turin, Italy.
| |
Collapse
|
44
|
Wang M, Banik I, Shain AH, Yeh I, Bastian BC. Integrated genomic analyses of acral and mucosal melanomas nominate novel driver genes. Genome Med 2022; 14:65. [PMID: 35706047 PMCID: PMC9202124 DOI: 10.1186/s13073-022-01068-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 06/03/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Acral and mucosal melanomas are aggressive subtypes of melanoma, which have a significantly lower burden of somatic mutations than cutaneous melanomas, but more frequent copy number variations, focused gene amplifications, and structural alterations. The landscapes of their genomic alterations remain to be fully characterized. METHODS We compiled sequencing data of 240 human acral and mucosal melanoma samples from 11 previously published studies and applied a uniform pipeline to call tumor cell content, ploidy, somatic and germline mutations, as well as CNVs, LOH, and SVs. We identified genes that are significantly mutated or recurrently affected by CNVs and implicated in oncogenesis. We further examined the difference in the frequency of recurrent pathogenic alterations between the two melanoma subtypes, correlation between pathogenic alterations, and their association with clinical features. RESULTS We nominated PTPRJ, mutated and homozygously deleted in 3.8% (9/240) and 0.8% (2/240) of samples, respectively, as a probable tumor suppressor gene, and FER and SKP2, amplified in 3.8% and 11.7% of samples, respectively, as probable oncogenes. We further identified a long tail of infrequent pathogenic alterations, involving genes such as CIC and LZTR1. Pathogenic germline mutations were observed on MITF, PTEN, ATM, and PRKN. We found BRAF V600E mutations in acral melanomas with fewer structural variations, suggesting that they are distinct and related to cutaneous melanomas. Amplifications of PAK1 and GAB2 were more commonly observed in acral melanomas, whereas SF3B1 R625 codon mutations were unique to mucosal melanomas (12.9%). Amplifications at 11q13-14 were frequently accompanied by fusion to a region on chromosome 6q12, revealing a recurrent novel structural rearrangement whose role remains to be elucidated. CONCLUSIONS Our meta-analysis expands the catalog of driver mutations in acral and mucosal melanomas, sheds new light on their pathogenesis and broadens the catalog of therapeutic targets for these difficult-to-treat cancers.
Collapse
Affiliation(s)
- Meng Wang
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Ishani Banik
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - A Hunter Shain
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Iwei Yeh
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA.
- Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA.
| | - Boris C Bastian
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA.
- Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
45
|
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: 3.0] [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
|
46
|
Amudzi AA, Piedra-Mora C, Ma DJ, Wong NB, David CN, Robinson NA, Almela RM, Richmond JM. Using Gene Expression Analysis to Understand Complex Autoimmune Skin Disease Patients: A Series of Four Canine Cutaneous Lupus Erythematosus Cases. Front Vet Sci 2022; 9:778934. [PMID: 35280134 PMCID: PMC8907585 DOI: 10.3389/fvets.2022.778934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 01/21/2022] [Indexed: 11/13/2022] Open
Abstract
Cutaneous Lupus Erythematosus (CLE) is an autoimmune skin disease that occurs in almost two-thirds of people with Systemic Lupus Erythematosus (SLE) and can exist as its own entity. Despite its negative impact on the quality of life of patients, lupus pathogenesis is not fully understood. In recent years, the role of gene expression analysis has become important in understanding cellular functions and disease causation within and across species. Interestingly, dogs also develop CLE, providing a spontaneous animal model of disease. Here, we present a targeted transcriptomic analysis of skin biopsies from a case series of four dogs with complex autoimmunity with suspected CLE. We identified 92 differentially expressed genes (DEGs), including type 1 interferon, B cell, and T cell-related genes, in the four cases compared to healthy skin margin controls. Additionally, we compared our results with existing CLE datasets from humans and mice and found that humans and canines share 49 DEGs, whereas humans and mice shared only 25 DEGs in our gene set. Immunohistochemistry of IFNG and CXCL10, two of the most highly upregulated inflammatory mediators, confirmed protein-level expression and revealed immune cells as the primary source of CXCL10 in dogs with SLE, whereas keratinocytes stained strongly for CXCL10 in dogs without SLE. We propose that gene expression analysis may aid the diagnosis of complex autoimmune skin diseases and that dogs may provide important insights into CLE and SLE pathogeneses, or more broadly, skin manifestations during systemic autoimmunity.
Collapse
Affiliation(s)
- Alice A. Amudzi
- Dermatology Department, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Cesar Piedra-Mora
- Pathology Department, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, United States
| | - Diana Junyue Ma
- Dermatology Department, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Neil B. Wong
- Dermatology Department, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | | | - Nicholas A. Robinson
- Pathology Department, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, United States
| | - Ramón M. Almela
- Department of Clinical Sciences, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, United States
| | - Jillian M. Richmond
- Dermatology Department, University of Massachusetts Chan Medical School, Worcester, MA, United States
| |
Collapse
|
47
|
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: 8] [Impact Index Per Article: 2.7] [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
|
48
|
Conrad D, Kehl A, Beitzinger C, Metzler T, Steiger K, Pfarr N, Fischer K, Klopfleisch R, Aupperle-Lellbach H. Molecular Genetic Investigation of Digital Melanoma in Dogs. Vet Sci 2022; 9:vetsci9020056. [PMID: 35202309 PMCID: PMC8874500 DOI: 10.3390/vetsci9020056] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/10/2022] [Accepted: 01/26/2022] [Indexed: 01/27/2023] Open
Abstract
Canine digital melanoma, in contrast to canine oral melanoma, is still largely unexplored at the molecular genetic level. The aim of this study was to detect mutant genes in digital melanoma. Paraffin-embedded samples from 86 canine digital melanomas were examined for the BRAF V595E variant by digital droplet PCR (ddPCR), and for exon 11 mutations in c-kit. Furthermore, exons 2 and 3 of KRAS and NRAS were analysed by Sanger sequencing. Copy number variations (CNV) of KITLG in genomic DNA were analysed from nine dogs. The BRAF V595E variant was absent and in c-kit, a single nucleotide polymorphism was found in 16/70 tumours (23%). The number of copies of KITLG varied between 4 and 6. KRAS exon 2 codons 12 and 13 were mutated in 22/86 (25.6%) of the melanomas examined. Other mutually exclusive RAS mutations were found within the hotspot loci, i.e., KRAS exon 3 codon 61: 2/55 (3.6%); NRAS exon 2 codons 12 and 13: 2/83 (2.4%); and NRAS exon 3 codon 61: 9/86 (10.5%). However, no correlation could be established between histological malignancy criteria, survival times and the presence of RAS mutations. In summary, canine digital melanoma differs from molecular genetic data of canine oral melanoma and human melanoma, especially regarding the proportion of RAS mutations.
Collapse
Affiliation(s)
- David Conrad
- Department of Pathology, LABOKLIN GmbH & Co. KG, 97688 Bad Kissingen, Germany;
- Correspondence:
| | - Alexandra Kehl
- Department of Molecular Biology, LABOKLIN GmbH & Co. KG, 97688 Bad Kissingen, Germany; (A.K.); (C.B.)
| | - Christoph Beitzinger
- Department of Molecular Biology, LABOKLIN GmbH & Co. KG, 97688 Bad Kissingen, Germany; (A.K.); (C.B.)
| | - Thomas Metzler
- Institute of Pathology, School of Medicine, Technische Universität München, 81675 München, Germany; (T.M.); (K.S.); (N.P.)
| | - Katja Steiger
- Institute of Pathology, School of Medicine, Technische Universität München, 81675 München, Germany; (T.M.); (K.S.); (N.P.)
| | - Nicole Pfarr
- Institute of Pathology, School of Medicine, Technische Universität München, 81675 München, Germany; (T.M.); (K.S.); (N.P.)
| | - Konrad Fischer
- School of Life Sciences Weihenstephan, Technische Universität München, 85354 Freising, Germany;
| | - Robert Klopfleisch
- Department of Pathology, Freie Universität Berlin, 14163 Berlin, Germany;
| | | |
Collapse
|
49
|
Du L, Wang D, Wei X, Liu C, Xiao Z, Qian W, Song Y, Hou X. MS275 as Class I HDAC inhibitor displayed therapeutic potential on malignant ascites by iTRAQ-based quantitative proteomic analysis. BMC Gastroenterol 2022; 22:29. [PMID: 35062876 PMCID: PMC8783488 DOI: 10.1186/s12876-022-02101-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 01/07/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Malignant ascites is a manifestation of end stage events in a variety of cancers and is associated with significant morbidity. Epigenetic modulators play a key role in cancer initiation and progression, among which histone deacetylases (HDACs) are considered as one of the most important regulators for various cancer development, such as liver cancer, ovarian cancer, and pancreatic cancer et al. Thus, in this paper, we sought to explore the therapeutic effect of HDAC inhibitor on malignant ascites. METHODS In this report, we tested the therapeutic effect of different isoform selective HDAC inhibitors (Class I HDACI MS275, Class IIa HDACI MC1568, pan-HDAC inhibitors SAHA) on malignant ascites in vitro and in vivo. We further used proteome analysis to find the potential mechanisms for malignant ascites therapy. RESULTS Among the different isoform-selective HDAC inhibitors, the class I selective HDACI, MS275, exhibited preferential inhibition on various ascites cells. MS275 could induce cell cycle arrest in G0/G1 phase and promote apoptosis on ascites cells. Through proteome analysis, we found MS275 could downregulate proteins related to cell cycle progression, such as CDK4, CDC20, CCND1; MS275 could upregulate pro-apoptosis proteins such as PAPR1, LMNB2 and AIFM1; in addition, MS275 could change the expression of tumorigenic proteins related to the specific malignant ascites bearing tumors, such as TSP1 and CDK4 for bladder cancer. We then confirmed that abemaciclib (CDK4/6 selective inhibitor) could inhibit the proliferation of ascites cells, and the combination of abemaciclib and MS275 had synergistic anti-tumor effect. Finally, we found that MS275 could in vivo inhibit malignant ascites progression (ascites volume: 2.9 ± 1.0 mL vs 7.5 ± 1.2 mL, p < 0.01), tumor growth, and prolong 66% of the life-span when compared with the untreated group. CONCLUSION This present research revealed that the class I selective HDAC inhibitor, MS275, could effectively inhibit malignant ascites development and tumor growth via multiple pathways. These results indicated that HDACI could have great potential for clinical therapy of malignant ascites.
Collapse
Affiliation(s)
- Li Du
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Dongyuan Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Xiuqi Wei
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Chang Liu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Zhuanglong Xiao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Wei Qian
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Yuhu Song
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.
| |
Collapse
|
50
|
Prouteau A, Mottier S, Primot A, Cadieu E, Bachelot L, Botherel N, Cabillic F, Houel A, Cornevin L, Kergal C, Corre S, Abadie J, Hitte C, Gilot D, Lindblad-Toh K, André C, Derrien T, Hedan B. Canine Oral Melanoma Genomic and Transcriptomic Study Defines Two Molecular Subgroups with Different Therapeutical Targets. Cancers (Basel) 2022; 14:cancers14020276. [PMID: 35053440 PMCID: PMC8774001 DOI: 10.3390/cancers14020276] [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: 11/25/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 02/05/2023] Open
Abstract
Simple Summary In humans, mucosal melanoma (MM) is a rare and aggressive cancer. The canine model is frequently and spontaneously affected by MM, thus facilitating the collection of samples and the study of its genetic bases. Thanks to an integrative genomic and transcriptomic analysis of 32 canine MM samples, we identified two molecular subgroups of MM with a different microenvironment and structural variant (SV) content. We demonstrated that SVs are associated with recurrently amplified regions, and identified new candidate oncogenes (TRPM7, GABPB1, and SPPL2A) for MM. Our findings suggest the existence of two MM molecular subgroups that could benefit from dedicated therapies, such as immune checkpoint inhibitors or targeted therapies, for both human and veterinary medicine. Abstract Mucosal melanoma (MM) is a rare, aggressive clinical cancer. Despite recent advances in genetics and treatment, the prognosis of MM remains poor. Canine MM offers a relevant spontaneous and immunocompetent model to decipher the genetic bases and explore treatments for MM. We performed an integrative genomic and transcriptomic analysis of 32 canine MM samples, which identified two molecular subgroups with a different microenvironment and structural variant (SV) content. The overexpression of genes related to the microenvironment and T-cell response was associated with tumors harboring a lower content of SVs, whereas the overexpression of pigmentation-related pathways and oncogenes, such as TERT, was associated with a high SV burden. Using whole-genome sequencing, we showed that focal amplifications characterized complex chromosomal rearrangements targeting oncogenes, such as MDM2 or CDK4, and a recurrently amplified region on canine chromosome 30. We also demonstrated that the genes TRPM7, GABPB1, and SPPL2A, located in this CFA30 region, play a role in cell proliferation, and thus, may be considered as new candidate oncogenes for human MM. Our findings suggest the existence of two MM molecular subgroups that may benefit from dedicated therapies, such as immune checkpoint inhibitors or targeted therapies, for both human and veterinary medicine.
Collapse
Affiliation(s)
- Anais Prouteau
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Stephanie Mottier
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Aline Primot
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Edouard Cadieu
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Laura Bachelot
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Nadine Botherel
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Florian Cabillic
- Laboratoire de Cytogénétique et Biologie Cellulaire, CHU de Rennes, INSERM, INRA, University of Rennes 1, Nutrition Metabolisms and Cancer, 35000 Rennes, France; (F.C.); (L.C.)
| | - Armel Houel
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Laurence Cornevin
- Laboratoire de Cytogénétique et Biologie Cellulaire, CHU de Rennes, INSERM, INRA, University of Rennes 1, Nutrition Metabolisms and Cancer, 35000 Rennes, France; (F.C.); (L.C.)
| | - Camille Kergal
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Sébastien Corre
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Jérôme Abadie
- Laboniris, Department of Biology, Pathology and Food Sciences, Oniris, 44300 Nantes, France;
| | - Christophe Hitte
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - David Gilot
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Kerstin Lindblad-Toh
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA;
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Catherine André
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
| | - Thomas Derrien
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
- Correspondence: (T.D.); (B.H.); Tel.: +33-2-23-23-43-19 (B.H.)
| | - Benoit Hedan
- IGDR—UMR 6290, CNRS, University of Rennes 1, 35000 Rennes, France; (A.P.); (S.M.); (A.P.); (E.C.); (L.B.); (N.B.); (A.H.); (C.K.); (S.C.); (C.H.); (D.G.); (C.A.)
- Correspondence: (T.D.); (B.H.); Tel.: +33-2-23-23-43-19 (B.H.)
| |
Collapse
|