Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
2.
Sci Rep ; 10(1): 11581, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32665562

ABSTRACT

Insulinomas (INS) are the most common human and canine functioning pancreatic neuroendocrine tumours. The long-term prognosis for malignant INS is poor, because micrometastases are frequently missed during surgery. As human and canine malignant INS share clinical and histopathological features, dogs have been proposed as models for INS research. Using RNA-sequencing, we conducted a pilot study to better understand the underlying molecular mechanisms of canine INS. Normal canine pancreas and lymph node control tissues were compared with primary INS and INS-metastatic lymph nodes, revealing more than 3,000 genes differentially expressed in normal pancreas compared to primary INS. Only 164 genes were differentially expressed between primary INS and INS-metastatic lymph nodes. Hierarchical clustering analysis demonstrated similar genetic profiles in normal pancreas and early clinical stage primary INS, whereas late clinical stage primary INS resembled the genetic profile of INS-metastatic lymph nodes. These findings suggest that markers of malignant behaviour could be identified at the primary site of the disease. Finally, using the REACTOME pathways database, we revealed that an active collagen metabolism, extracellular matrix remodelling, beta-cell differentiation and non-beta-cell trans-differentiation might cause disease progression and hyperinsulinism in INS, identifying major pathways worthy of future research in this currently poorly controlled disease.


Subject(s)
Dog Diseases/genetics , Insulinoma/genetics , Neoplasm Proteins/genetics , Transcriptome/genetics , Animals , Disease Progression , Dog Diseases/pathology , Dogs , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Insulinoma/pathology , Insulinoma/veterinary , Neoplasm Metastasis , Sequence Analysis, RNA
3.
Vet Comp Oncol ; 16(3): 301-310, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29892997

ABSTRACT

Bacterial infection following cancer chemotherapy-induced neutropenia is a serious cause of morbidity and mortality in human and veterinary patients. Antimicrobial prophylaxis is controversial in the human oncology field, as any decreased incidence in bacterial infections is countered by patient adverse effects and increased antimicrobial resistance. Comprehensive guidelines exist to aid human oncologists in prescribing antimicrobial prophylaxis but similar recommendations are not available in veterinary literature. As the veterinarian's role in antimicrobial stewardship is increasingly emphasized, it is vital that veterinary oncologists implement appropriate antimicrobial use. By considering the available human and veterinary literature we present an overview of current clinical practices and are able to suggest recommendations for prophylactic antimicrobial use in veterinary cancer chemotherapy patients.


Subject(s)
Antibiotic Prophylaxis/veterinary , Antineoplastic Agents/therapeutic use , Neoplasms/veterinary , Animals , Antibiotic Prophylaxis/adverse effects , Antibiotic Prophylaxis/methods , Antineoplastic Agents/adverse effects , Bacterial Infections/prevention & control , Bacterial Infections/veterinary , Neoplasms/drug therapy
4.
J Comp Pathol ; 160: 23-33, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29729718

ABSTRACT

Myxomatous mitral valve disease is the most common cardiac disease of the dog, but examination of the associated cellular and molecular events has relied on the use of cadaveric valve tissue, in which functional studies cannot be undertaken. The aim of this study was to develop a three-dimensional (3D) cell co-culture model as an experimental platform to examine disease pathogenesis. Mitral valve interstitial (VIC) and endothelial (VEC) cells were cultured from normal and diseased canine (VIC only) valves. VICs were embedded in a fibrin-based hydrogel matrix and one surface was lined with VECs. The 3D static cultures (constructs) were examined qualitatively and semiquantitatively by light microscopy, immunofluorescence microscopy and protein immunoblotting. Some constructs were manipulated and the endothelium damaged, and the response examined. The construct gross morphology and histology demonstrated native tissue-like features and comparable expression patterns of cellular (α-smooth muscle actin [SMA] and embryonic smooth muscle myosin heavy chain [SMemb]) and extracellular matrix associated markers (matrix metalloproteinase [MMP]-1 and MMP-3), reminiscent of diseased valves. There were no differences between constructs containing normal valve VICs and VECs (type 1) and those containing diseased valve VICs and normal valve VECs (type 2). Mechanical manipulation and endothelial damage (type 3) tended to decrease α-SMA and SMemb expression, suggesting reversal of VIC activation, but with retention of SMemb+ cells adjacent to the wounded endothelium consistent with response to injury. Fibrin-based 3D mitral valve constructs can be produced using primary cell cultures derived from canine mitral valves, and show a phenotype reminiscent of diseased valves. The constructs demonstrate a response to endothelial damage indicating their utility as experimental platforms.


Subject(s)
Cell Culture Techniques , Dog Diseases , Mitral Valve , Tissue Engineering/methods , Animals , Coculture Techniques , Dogs
5.
Endocr Relat Cancer ; 25(2): 131-144, 2018 02.
Article in English | MEDLINE | ID: mdl-29175872

ABSTRACT

Insulinomas (INS) are the most common neuroendocrine pancreatic tumours in humans and dogs. The long-term prognosis for malignant INS is still poor due to a low success rate of the current treatment modalities, particularly chemotherapy. A better understanding of the molecular processes underlying the development and progression of INS is required to develop novel targeted therapies. Cancer stem cells (CSCs) are thought to be critical for the engraftment and chemoresistance of many tumours, including INS. This study was aimed to characterise and target INS CSCs in order to develop novel targeted therapies. Highly invasive and tumourigenic human and canine INS CSC-like cells were successfully isolated. These cells expressed stem cell markers (OCT4, SOX9, SOX2, CD133 and CD34), exhibited greater resistance to 5-fluorouracil (5-FU) and demonstrated a more invasive and tumourigenic phenotype in vivo compared to bulk INS cells. Here, we demonstrated that Notch-signalling-related genes (NOTCH2 and HES1) were overexpressed in INS CSC-like cells. Protein analysis showed an active NOTCH2-HES1 signalling in INS cell lines, especially in cells resistant to 5-FU. Inhibition of the Notch pathway, using a gamma secretase inhibitor (GSI), enhanced the sensitivity of INS CSC-like cells to 5-FU. When used in combination GSI and 5-FU, the clonogenicity in vitro and the tumourigenicity in vivo of INS CSC-like cells were significantly reduced. These findings suggested that the combined strategy of Notch signalling inhibition and 5-FU synergistically attenuated enriched INS CSC populations, providing a rationale for future therapeutic exploitation.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Drug Resistance, Neoplasm/drug effects , Fluorouracil/pharmacology , Insulinoma , Neoplastic Stem Cells/drug effects , Pancreatic Neoplasms , Receptors, Notch/metabolism , Animals , Cell Line, Tumor , Dogs , Humans , Neoplastic Stem Cells/metabolism , Signal Transduction/drug effects
6.
Vet J ; 224: 1-6, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28697868

ABSTRACT

Mast cell tumours (MCTs) in dogs can present in a variety of forms. Non-resectable, recurrent or metastatic MCTs usually carry a poor prognosis and present a therapeutic challenge. Both toceranib and lomustine have shown single agent activity against MCTs in dogs. In this study, 10 dogs with advanced MCTs were enrolled prospectively and treated with toceranib (median dose 2.7mg/kg orally every other day), lomustine (median dose 60mg/m2 orally every 3 weeks) and prednisolone (1mg/kg orally every other day, alternating with toceranib). Severe adverse events (SAEs), requiring alterations in the protocol, occurred in all dogs. The objective response rate was 50%. Three dogs died or were euthanased due to SAEs and therefore enrolment of new dogs was discontinued prematurely. A long term response (>1year) was observed in two dogs. Modifications of the protocol are required for future prospective studies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dog Diseases/drug therapy , Indoles/administration & dosage , Lomustine/administration & dosage , Mast Cells/pathology , Pyrroles/administration & dosage , Skin Neoplasms/veterinary , Animals , Antineoplastic Agents, Alkylating , Dog Diseases/pathology , Dogs , Europe , Indoles/adverse effects , Lomustine/adverse effects , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/veterinary , Prednisolone/administration & dosage , Prospective Studies , Pyrroles/adverse effects , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology
7.
Vet Comp Oncol ; 15(2): 655-666, 2017 Jun.
Article in English | MEDLINE | ID: mdl-26778136

ABSTRACT

Human inflammatory breast cancer (IBC) and canine inflammatory mammary cancer (CIMC) are the most aggressive forms of mammary cancer. Current research aims to identify new therapeutic targets. Here, we investigated gene expression levels of biomarkers associated with the inflammatory microenvironment. A total of 32 formalin-fixed paraffin-embedded samples of canine mammary carcinoma (CIMC = 26; non-CIMC = 6) were used and their cDNA subjected to quantitative polymerase chain reaction (qPCR) to establish gene expression levels for mediators commonly implicated in linking carcinogenesis with inflammation. Gene expression differences between CIMC and non-CIMC types were obtained for cyclooxygenase 2 (COX-2) (P = 0.004), synuclein gamma (SNCG) (P = 0.006), tribbles 1 (P = 0.025), vascular endothelial growth factor (VEGF) (P = 0.017) and CSF1R (P = 0.045). Among these biomarkers correlations were found, particularly between SNCG and tribbles 1 (r = 0.512, P = 0.001). The efficient metastasis of CIMC is intimately linked to components in the tumour microenvironment. This study suggests that upregulation and correlation of SNCG and tribbles 1 deserves to be further explored.


Subject(s)
Dog Diseases/metabolism , Mammary Neoplasms, Animal/chemistry , Animals , Biomarkers/analysis , Cyclooxygenase 2/metabolism , Dog Diseases/pathology , Dogs , Female , Inflammation/metabolism , Inflammation/veterinary , Intracellular Signaling Peptides and Proteins/metabolism , Mammary Glands, Animal/chemistry , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/pathology , Polymerase Chain Reaction/veterinary , Synucleins/metabolism , Tumor Microenvironment , Vascular Endothelial Growth Factor A/metabolism
8.
Vet Comp Oncol ; 15(3): 890-909, 2017 Sep.
Article in English | MEDLINE | ID: mdl-27229930

ABSTRACT

Members of the epidermal growth factor receptor (EGFR/ERBB) gene family are frequently dysregulated in a range of human cancers, and therapeutics targeting these proteins are in clinical use. We hypothesized that similar pathways are involved in feline and canine tumours and that the same drugs may be of clinical use in veterinary patients. We investigated EGFR and ERBB2 targeting using a panel of feline and canine cell lines. EGFR and ERBB2 were targeted with siRNAs or tyrosine kinase inhibitors (TKIs) and their effect on cellular proliferation, colony formation and migration was investigated in vitro. Here we report that EGFR and ERBB2 combined siRNA targeting produced synergistic effects in feline and canine cell lines similar to that reported in human cell lines. We conclude that dual EGFR and ERBB2 targeting using TKIs should be further evaluated as a potential new therapeutic strategy in feline head and neck and mammary tumours and canine mammary tumours.


Subject(s)
ErbB Receptors/drug effects , Neoplasms/veterinary , Receptor, ErbB-2/drug effects , Signal Transduction/drug effects , Animals , Antineoplastic Agents/therapeutic use , Cat Diseases/drug therapy , Cat Diseases/metabolism , Cat Diseases/pathology , Cats , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Dog Diseases/drug therapy , Dog Diseases/metabolism , Dog Diseases/pathology , Dogs , Drug Synergism , ErbB Receptors/genetics , ErbB Receptors/physiology , In Vitro Techniques , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA, Small Interfering/therapeutic use , Real-Time Polymerase Chain Reaction , Receptor, ErbB-2/genetics , Receptor, ErbB-2/physiology , Signal Transduction/physiology
12.
Vet J ; 205(2): 161-74, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25981934

ABSTRACT

Advances in biotechnology have enabled the collection of an immeasurable amount of information from genomic, transcriptomic, metabolomic and proteomic studies of tumours within their microenvironments. The dissection of cytokine and chemokine networks has provided new clues to the interactions between cancer cells and their surrounding inflammatory landscape. To bridge the gap between chronic inflammation and cancer, dynamic participants in the tumour microenvironment have been identified, including tumour-associated macrophages (TAMs) and regulatory T cells (Tregs). Both of these cell types are notable for their ability to cause immunosuppressive conditions and support the evasion of tumour immune surveillance. It is clear now that the tumour-promoting inflammatory environment has to be included as one of the major cancer hallmarks. This review explores the recent advances in the understanding of cancer-related inflammation and how this is being applied to comparative oncology studies in humans and domestic species, such as the dog.


Subject(s)
Inflammation/veterinary , Neoplasms/veterinary , Animals , Biomarkers, Tumor , Carcinogenesis , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Neoplasm Invasiveness/physiopathology , Neoplasm Metastasis/physiopathology , Neoplasms/metabolism , Neoplasms/pathology
13.
Vet J ; 204(1): 23-31, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25841900

ABSTRACT

Myxomatous mitral valve disease (MMVD) is the single most common acquired heart disease of the dog and is particularly common in small pedigree breed dogs such as the Cavalier King Charles spaniel (CKCS). There are limited data on the mitral valve transcriptome and the aim of this study was to use the microarray technology in conjunction with bioinformatics platforms to analyse transcript changes in MMVD in CKCS compared to normal dogs (non-CKCS). Differentially expressed genes (n = 5397) were identified using cut-off settings of fold change, false discovery rate (FDR) and P <0.05. In total, 4002 genes were annotated to a specific transcript in the Affymetrix canine database, and after further filtering, 591 annotated canine genes were identified: 322 (55%) were up-regulated and 269 (45%) were down-regulated. Canine microRNAs (cfa-miR; n = 59) were also identified. Gene ontology and network analysis platforms identified between six and 10 significantly different biological function clusters from which the following were selected as relevant to MMVD: inflammation, cell movement, cardiovascular development, extracellular matrix organisation and epithelial-to-mesenchymal (EMT) transition. Ingenuity Pathway Analysis identified three canonical pathways relevant to MMVD: caveolar-mediated endocytosis, remodelling of epithelial adherens junctions, and endothelin-1 signalling. Considering the biological relevance to MMVD, the gene families of importance with significant difference between groups included collagens, ADAMTS peptidases, proteoglycans, matrix metalloproteinases (MMPs) and their inhibitors, basement membrane components, cathepsin S, integrins, tight junction cell adhesion proteins, cadherins, other matrix-associated proteins, and members of the serotonin (5-HT)/transforming growth factor -ß signalling pathway.


Subject(s)
Dog Diseases/metabolism , Mitral Valve Insufficiency/veterinary , Oligonucleotide Array Sequence Analysis/veterinary , RNA/genetics , Animals , Computer Simulation , Dog Diseases/genetics , Dogs , Mitral Valve Insufficiency/genetics , Mitral Valve Insufficiency/metabolism , Models, Genetic , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Transcriptome
14.
Vet J ; 204(1): 32-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25747697

ABSTRACT

Valve interstitial cells (VICs) have an important role in the aetiopathogenesis of myxomatous mitral valve disease (MMVD) in the dog. Furthermore, there is evidence that valve endothelial cells (VECs) also contribute to disease development. In addition to examining native valve tissue to understand MMVD, another strategy is to separately examine VIC and VEC biology under in vitro culture conditions. The aim of this study was to isolate and characterise canine mitral VICs and VECs from normal dog valves using a combination of morphology, immunohistochemistry and reverse transcription PCR (RT-PCR). Canine mitral VECs and VICs were isolated and cultured in vitro. The two cell populations exhibited different morphologies and growth patterns. VECs, but not VICs, expressed the endothelial markers, platelet endothelial cell adhesion molecule (PECAM-1 or CD31) and acetylated low density lipoprotein (Dil-Ac-LDL). Both VECs and VICs expressed vimentin and embryonic non-smooth muscle myosin heavy chain (SMemb), an activated mesenchymal cell marker. The myofibroblast marker, alpha smooth muscle actin (α-SMA), was detected at the mRNA level in both VEC and VIC cultures, but only at the protein level in VIC cultures. The morphological heterogeneity and expression of non-endothelial phenotypic markers in VEC cultures suggested that a mixture of cell types was present, which might be due to cell contamination and/or endothelial-mesenchymal transition (EndoMT). The use of a specific endothelial culture medium for primary VEC cultures enhanced the endothelial properties of the cells and reduced α-SMA and SMemb expression.


Subject(s)
Dogs , Endothelial Cells/physiology , Mitral Valve/cytology , Animals , Cells, Cultured , Culture Media , Female , Male
15.
Vet Comp Oncol ; 13(4): 464-74, 2015 Dec.
Article in English | MEDLINE | ID: mdl-24119241

ABSTRACT

Tumour-associated macrophages (TAMs) have been implicated in carcinogenesis including an important role in angiogenesis. In this study, we describe the relationship between TAMs and angiogenesis in canine mammary tumours (CMT). Formalin-fixed paraffin-embedded CMT samples [(n = 128: malignant (n = 97) and benign (n = 31)] were submitted to immunohistochemical staining to detect MAC387, vascular endothelial growth factor VEGF and CD31 expression. A statistical analysis was carried out to assess possible associations with clinicopathological variables and biological markers of tumour angiogenesis. TAMs, detected by MAC387 expression, were significantly associated with malignant CMT (P < 0.001) and VEGF positive tumours (P = 0.002) and also associated with VEGF expression within malignant CMT (P = 0.043). Associations with clinicopathological variables were found between TAMs and the presence of infiltrative growth (P = 0.031), low tubule formation (P = 0.040) and lymph node metastasis (P = 0.016). The results support the hypothesis that TAMs influence angiogenesis in CMT suggesting TAMs may represent a therapeutic target in this disease.


Subject(s)
Dog Diseases/metabolism , Macrophages/physiology , Mammary Neoplasms, Animal/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Dog Diseases/immunology , Dog Diseases/pathology , Dogs , Female , Gene Expression Regulation, Neoplastic/physiology , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/veterinary , Platelet Endothelial Cell Adhesion Molecule-1/metabolism
16.
J Comp Pathol ; 150(4): 366-72, 2014 May.
Article in English | MEDLINE | ID: mdl-24529510

ABSTRACT

NOTCH-1 is a transmembrane receptor protein. Ligand proteins expressed on the surface of neighbouring cells bind to the NOTCH-1 extracellular domain by juxtacrine signalling and release the NOTCH intracellular domain (NICD) to alter gene expression. Forty feline mammary lesions (34 malignant and six hyperplastic) were submitted for immunohistochemical analysis of NICD expression using an anti-feline NICD monoclonal antibody. Associations between NICD expression in carcinomas and morphological parameters, as well as overall survival (OS), were investigated. NICD nuclear expression was observed in hyperplastic lesions (100%) while cytoplasmic localization was evident in carcinomas (0% nuclear positive; 87.5% cytoplasmic positive; 12.5% negative). Cytoplasmic NICD localization was statistically associated with carcinomas, while nuclear labelling was associated with hyperplasia. No significant correlation between positive or negative NICD expression and OS or morphological parameters was detected. NOTCH-1 activation, immunohistochemically identified by the NICD active form, appears to play a role in feline mammary carcinoma biology as the majority of tumours express this protein. Nuclear localization is consistent with the established NICD metabolic intranuclear pathway while cytoplasmic accumulation suggests aberrant NOTCH-1 signalling typical of malignant tumour progression.


Subject(s)
Carcinoma/veterinary , Cat Diseases/metabolism , Mammary Neoplasms, Animal/metabolism , Receptor, Notch1/metabolism , Animals , Carcinoma/metabolism , Carcinoma/pathology , Cat Diseases/pathology , Cats , Female , Hyperplasia/metabolism , Hyperplasia/pathology , Hyperplasia/veterinary , Immunohistochemistry , Mammary Neoplasms, Animal/pathology
17.
Vet Comp Oncol ; 11(3): 169-84, 2013 Sep.
Article in English | MEDLINE | ID: mdl-22404947

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a complex process involved in embryonic development, wound healing and carcinogenesis. During this process, epithelial cells lose their defining characteristics and acquire mesenchymal properties: loss of cell-cell adhesion; increased motility and invasiveness; resistance to apoptosis and changes in cellular morphology. EMT has been implicated as a driver of metastasis and tumour invasion, as this process allows cells to detach from their niche and migrate through blood and lymphatic vessels to invade different organs. This transition involves a diverse range of transcription factors, including Twist, Snail and ZEB1, and downstream transcriptional targets, including E-cadherin, ß-catenin, fibronectin and vimentin. Recent evidence indicates that cancer stem cells are required for metastatic tumours to become established at a distant site, and that cancer cells undergoing EMT may develop stem-cell characteristics as well as increased invasive potential. The role of EMT in cancer biology is newly emerging in the human field, and to date very little has been done in veterinary medicine. EMT may therefore be an important molecular determinant of tumour metastasis, and further understanding of this process may lead to novel drug targets to be exploited in both veterinary and human medicine.


Subject(s)
Carcinogenesis , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic/physiology , Neoplasms/metabolism , Animals
18.
Vet J ; 196(3): 414-23, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23219486

ABSTRACT

Cancer stem cells were identified in a feline mammary carcinoma cell line by demonstrating expression of CD133 and utilising the tumour sphere assay. A population of cells was identified that had an invasive, mesenchymal phenotype, expressed markers of pluripotency and enhanced tumour formation in the NOD-SCID mouse and chick embryo models. This population of feline mammary carcinoma stem cells was resistant to chemotherapy and radiation, possibly due to aberrant activation of the ATM/p53 DNA damage pathway. Epithelial-mesenchymal transition was a feature of the invasive phenotype. These data demonstrate that cancer stem cells are a feature of mammary cancer in cats.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Cat Diseases/pathology , Drug Resistance, Neoplasm , Mammary Neoplasms, Animal/pathology , Neoplastic Stem Cells/physiology , Tumor Suppressor Protein p53/metabolism , AC133 Antigen , Adenosine Deaminase/deficiency , Agammaglobulinemia , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins/genetics , Carcinoma/veterinary , Cats , DNA Damage , Female , Gene Expression Regulation, Neoplastic/physiology , Glycoproteins/genetics , Glycoproteins/metabolism , Mice , Neoplasms, Experimental , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Peptides/genetics , Peptides/metabolism , Severe Combined Immunodeficiency , Tumor Suppressor Protein p53/genetics
19.
Vet Comp Oncol ; 10(3): 163-73, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22882485

ABSTRACT

Tyrosine kinases (TKs) are key mediators of signalling pathways in cells. Located at the surface of the cell, within the cytoplasm or in the nucleous, TKs have been showed to be involved in regulation of normal cellular processes and also play an important role in the development, progression and spread of several types of cancer. Seventy percent of TK's are Receptor Tyrosine Kinases and these have become key targets for cancer therapy. This short synopsis is intended to give the reader an introduction to this area of targeted therapy for cancer as a prelude to this special edition of Veterinary and Comparative Oncology.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/veterinary , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Benzamides , Drug Therapy, Combination , Humans , Indoles/pharmacology , Indoles/therapeutic use , Neoplasms/drug therapy , Neoplasms/enzymology , Piperidines , Pyridines , Pyrroles/pharmacology , Pyrroles/therapeutic use , Receptor Protein-Tyrosine Kinases/drug effects , Receptor Protein-Tyrosine Kinases/physiology , Thiazoles/pharmacology , Thiazoles/therapeutic use
20.
Vet Comp Oncol ; 10(3): e1-e29, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22882486

ABSTRACT

In preparing this document the Authors aimed to pool current information on canine and feline mast cell disease. The information was gathered from international studies and a emphasis was placed on material and opinion with a strong evidence base. We intend it to form the basis of our understanding in this disease at the current time and we anticipate that it will be particularly useful for the general practitioner. It should be emphasized that the authors are presenting this work from a European perspective.


Subject(s)
Cat Diseases/diagnosis , Dog Diseases/diagnosis , Mastocytosis/veterinary , Animals , Cat Diseases/pathology , Cat Diseases/therapy , Cats , Dog Diseases/pathology , Dog Diseases/therapy , Dogs , Mast Cells/pathology , Mastocytosis/diagnosis , Mastocytosis/pathology , Mastocytosis/therapy , Paraneoplastic Syndromes/diagnosis , Paraneoplastic Syndromes/pathology , Paraneoplastic Syndromes/therapy , Paraneoplastic Syndromes/veterinary
SELECTION OF CITATIONS
SEARCH DETAIL