Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Vet Clin Pathol ; 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38923556

ABSTRACT

A 7-year-old female spayed Bernese Mountain dog was presented for evaluation of hematuria. Incidentally, a right stifle sarcoma was diagnosed via cytology, which raised concern for histiocytic sarcoma (given the patient's signalment) versus another joint-associated sarcoma. Histopathology and immunohistochemistry revealed a CD18-negative, non-histiocytic origin cell population. Findings were consistent with a joint-associated grade II soft tissue sarcoma (STS). The patient's hematuria was progressive over 5 months, and urinary bladder transitional cell carcinoma (TCC) was diagnosed via cystoscopy and histopathology. An enlarged right medial iliac lymph node was identified on routine restaging via abdominal ultrasound 3 months later. Cytology of the lymph node revealed a markedly pleomorphic cell population, again raising concern for histiocytic sarcoma (HS). Other differentials included an anaplastic metastatic population from the joint-associated STS or the TCC. Immunocytochemistry revealed a cytokeratin-positive, CD18-, CD204-, and vimentin-negative cell population, consistent with a carcinoma. DNA was extracted from cytology slides to sequence cells for BRAF mutation status. Sequencing revealed a homozygous V596E (transcript ENSCAFT00845055173.1) BRAF mutation, consistent with the known biology of TCC. In neither case was HS truly present in this patient, but immunocytochemistry provided information that helped to optimize the patient's chemotherapy recommendations.

2.
Vet Pathol ; 61(1): 20-31, 2024 01.
Article in English | MEDLINE | ID: mdl-37357953

ABSTRACT

Increased or constitutive activation of nuclear factor kappa B (NF-kB) is a feature of many chronic disease processes, including cancer. While NF-kB overactivation has been documented extensively in human oncology, there is a relative paucity of data documenting the same phenomenon in veterinary medicine. To assess NF-kB activity, antibodies to p65 and p100/p52, which are components of NF-kB heterodimers, were first validated for specificity and canine cross-reactivity via Western blot and labeling of immortalized cell pellets. Then, nuclear labeling for these antibodies was assessed via QuPath software in over 200 tumor tissue samples (10 hemangiosarcomas, 94 histiocytic sarcomas, 71 lymphomas, and 28 mast cell tumors) and compared to immunolabeling in appropriate normal tissue counterparts. Greater than 70% of spontaneous canine tumors evaluated in this study had more nuclear p65 and p100/p52 immunoreactivity than was observed in comparable normal cell populations. Specifically, 144/204 (70.58%) of tumors evaluated had positive p65 nuclear labeling and 179/195 (91.79%) had positive p100/p52 nuclear labeling. Surprisingly, greater nuclear p100/p52 reactivity was associated with a longer progression-free survival (PFS) and overall survival (OS) in canine lymphomas. These results provide support and preliminary data to investigate the role of NF-kB signaling in different types of canine cancer.


Subject(s)
Dog Diseases , Hemangiosarcoma , Histiocytic Sarcoma , Lymphoma , Animals , Dogs , Humans , NF-kappa B/metabolism , Histiocytic Sarcoma/veterinary , Hemangiosarcoma/veterinary , Mast Cells , NF-kappa B p52 Subunit/metabolism , Lymphoma/veterinary
3.
J Pharmacol Exp Ther ; 388(3): 774-787, 2024 02 15.
Article in English | MEDLINE | ID: mdl-38135509

ABSTRACT

This study provides a unique translational research opportunity to help both humans and dogs diagnosed with diseases that carry dismal prognoses in both species: histiocytic sarcoma (HS), hemangiosarcoma (HSA), and disseminated mastocytosis/mast cell tumor (MCT). Although exceedingly rare in humans, these so called "orphan diseases" are relatively more common in dogs. For these and other more commonplace cancers like lymphoma (Lym), dogs are an excellent translational model for human disease due to remarkably similar disease biology. In this study, assays were performed to assess the therapeutic potential of parthenolide (PTL), a known canonical nuclear factor kappa B (NF-κB) signaling inhibitor with additional mechanisms of antineoplastic activity, including alteration of cellular reduction-oxidation balance. Canine cell lines and primary cells are sensitive to PTL and undergo dose-dependent apoptosis after exposure to drug. PTL exposure also leads to glutathione depletion, reactive oxygen species generation, and NF-κB inhibition in canine cells. Standard-of-care therapeutics broadly synergize with PTL. In two canine HS cell lines, expression of NF-κB pathway signaling partners is downregulated with PTL therapy. Preliminary data suggest that PTL inhibits NF-κB activity of cells and extends survival time in a mouse model of disseminated canine HS. These data support further investigation of compounds that can antagonize canonical NF-κB pathway signaling in these cancers and pave the way for clinical trials of PTL in affected dogs. As dogs are an excellent natural disease model for these cancers, these data will ultimately improve our understanding of their human disease counterparts and hopefully improve care for both species. SIGNIFICANCE STATEMENT: Disseminated neoplasms in human and canine cancers are challenging to treat, and novel therapeutic approaches are needed to improve outcomes. Parthenolide is a promising treatment for histiocytic sarcoma, hemangiosarcoma, and mast cell neoplasia.


Subject(s)
Hemangiosarcoma , Histiocytic Sarcoma , Sesquiterpenes , Mice , Humans , Animals , Dogs , NF-kappa B/metabolism , Cell Line, Tumor , Histiocytic Sarcoma/drug therapy , Hemangiosarcoma/drug therapy , Sesquiterpenes/pharmacology , Sesquiterpenes/therapeutic use , Apoptosis
5.
Vet Pathol ; 59(5): 724-732, 2022 09.
Article in English | MEDLINE | ID: mdl-35499088

ABSTRACT

Spontaneous tumors in dogs share several environmental, epidemiologic, biologic, clinical and molecular features with a wide variety of human cancers, making this companion animal an attractive model. Nuclear factor kappa B (NF-kB) transcription factor overactivation is common in several human cancers, and there is evidence that similar signaling aberrations also occur in canine cancers including lymphoma, leukemia, hemangiosarcoma, mammary cancer, melanoma, glioma, and prostate cancer. This review provides an overview of NF-kB signaling biology, both in health and in cancer development. It also summarizes available evidence of aberrant NF-kB signaling in canine cancer, and reviews antineoplastic compounds that have been shown to inhibit NF-kB activity used in various types of canine cancers. Available data suggest that dogs may be an excellent model for human cancers that have overactivation of NF-kB.


Subject(s)
Antineoplastic Agents , Dog Diseases , NF-kappa B , Neoplasms , Animals , Dog Diseases/metabolism , Dog Diseases/pathology , Dogs , NF-kappa B/metabolism , Neoplasms/metabolism , Neoplasms/veterinary , Signal Transduction
6.
Can J Vet Res ; 84(2): 115-123, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32255906

ABSTRACT

The objective of this in-vitro study was to evaluate taurolidine as a therapy for transitional cell carcinomas in canine patients. Transitional cell carcinoma (TCC) is the most common cancer of the urinary bladder in dogs and accounts for approximately 2% of reported malignancies in this species. There is no cure for this neoplasm and most dogs are lost from complications associated with progression of the local disease. Taurolidine has been shown to have anti-tumor and antiangiogenic effects against a variety of neoplasms in human and animal models. Four canine TCC cell lines were treated with various concentrations of taurolidine, mitoxantrone, and piroxicam alone. In addition, combinations of taurolidine/mitoxantrone, taurolidine/piroxicam, mitoxantrone/piroxicam, and taurolidine/mitoxantrone/piroxicam were assessed. Susceptibility of the TCC cell lines was based on a 72-hour growth inhibition assay using resazurin with absorbance measured at λ530/590. The ability of taurolidine to induce apoptosis was evaluated on 2 of the cell lines with an Annexin-V/propidium iodide assay. All cell lines were susceptible to treatment with taurolidine, mitoxantrone, and piroxicam alone. The results of the combination therapies of the 3 drugs were dependent on cell line and concentration and revealed no change in cell growth inhibition, a subadditive relationship, or a synergistic relationship. Taurolidine induced apoptosis in a concentration- and time-dependent fashion. Taurolidine alone showed significant effects on cell viability in vitro in canine TCC cell lines and these effects can be potentially enhanced with the addition of mitoxantrone and/or piroxicam.


Le carcinome à cellules transitionnelles est le cancer le plus fréquent de la vessie urinaire du chien et compte pour approximativement 2 % des tumeurs malignes rapportées chez cette espèce. Il n'existe présentement pas de cure pour cette tumeur et la plupart des chiens succombent des complications associées à la progression de la tumeur au niveau de la vessie. La taurolidine est une substance avec des propriétés anticancéreuses et anti-angiogéniques contre plusieurs cancers chez l'humain et différents modèles animaux. Cette étude in vitro a évalué la taurolidine comme thérapie pour le carcinome à cellules transitionnelles chez le chien. Quatre lignées cellulaires de carcinome à cellules transitionnelles canins ont été traitées avec différentes concentrations de taurolidine, mitoxantrone, et piroxicam, seul et en combinaisons (taurolidine-mitoxantrone, taurolidine-piroxicam, mitoxantrone-piroxicam et taurolidine-mitoxantrone-piroxicam). La susceptibilité des lignées cellulaires a été déterminée par l'inhibition de croissance en 72 heures et la viabilité cellulaire a été évaluée par l'absorbance de la résazurine mesurée à λ530/590. La capacité de la taurolidine à induire l'apoptose a été évaluée avec l'essai à l'Annexin-V/Iodate de Propidium. Toutes les lignées cellulaires étaient sensibles au traitements avec la taurolidine, le mitoxantrone, et le piroxicam seul. Les résultats des traitements de combinaisons de médicaments étaient dépendants des lignées cellulaires et des concentrations des médicaments. Les combinaisons avaient soit aucun effect comparé au médicaments seuls, ou un effet sous-additif ou synergique. La taurolidine a induite l'apoptose de façon dépendante à sa concentration et temps d'exposition. La taurolidine seule a démontré des effets significatifs sur la viabilité in vitro chez chacune des quatre lignées cellulaires de carcinome à cellules transitionnelles canins. Ces effets peuvent potentiellement être améliorés par l'ajout du mitoxantrone ou piroxicam.(Traduit par les auteurs).


Subject(s)
Carcinoma, Transitional Cell/veterinary , Dog Diseases/drug therapy , Mitoxantrone/pharmacology , Piroxicam/pharmacology , Taurine/analogs & derivatives , Thiadiazines/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Transitional Cell/drug therapy , Cell Line , Cell Survival , Dogs , Drug Synergism , Mitoxantrone/administration & dosage , Piroxicam/administration & dosage , Taurine/administration & dosage , Taurine/pharmacology , Thiadiazines/administration & dosage
8.
Front Oncol ; 9: 96, 2019.
Article in English | MEDLINE | ID: mdl-30859090

ABSTRACT

Gliomas and meningiomas are the most common brain neoplasms affecting both humans and canines, and identifying druggable targets conserved across multiple brain cancer histologies and comparative species could broadly improve treatment outcomes. While satisfactory cure rates for low grade, non-invasive brain cancers are achievable with conventional therapies including surgery and radiation, the management of non-resectable or recurrent brain tumors remains problematic and necessitates the discovery of novel therapies that could be accelerated through a comparative approach, such as the inclusion of pet dogs with naturally-occurring brain cancers. Evidence supports procaspase-3 as a druggable brain cancer target with PAC-1, a pro-apoptotic, small molecule activator of procaspase-3 that crosses the blood-brain barrier. Procaspase-3 is frequently overexpressed in malignantly transformed tissues and provides a preferential target for inducing cancer cell apoptosis. While preliminary evidence supports procaspase-3 as a viable target in preclinical models, with PAC-1 demonstrating activity in rodent models and dogs with spontaneous brain tumors, the broader applicability of procaspase-3 as a target in human brain cancers, as well as the comparability of procaspase-3 expressions between differing species, requires further investigation. As such, a large-scale validation of procaspase-3 as a druggable target was undertaken across 651 human and canine brain tumors. Relative to normal brain tissues, procaspase-3 was overexpressed in histologically diverse cancerous brain tissues, supporting procaspase-3 as a broad and conserved therapeutic target. Additionally, procaspase-3 expressing glioma and meningioma cell lines were sensitive to the apoptotic effects of PAC-1 at biologically relevant exposures achievable in cancer patients. Importantly, the clinical relevance of procaspase-3 as a potential prognostic variable was demonstrated in human astrocytomas of variable histologic grades and associated clinical outcomes, whereby tumoral procaspase-3 expression was negatively correlated with survival; findings which suggest that PAC-1 might provide the greatest benefit for patients with the most guarded prognoses.

11.
Oncotarget ; 8(46): 80124-80138, 2017 Oct 06.
Article in English | MEDLINE | ID: mdl-29113289

ABSTRACT

PURPOSE: Glioblastoma is a deadly brain cancer with a median survival time of ∼15 months. Ionizing radiation plus the DNA alkylator temozolomide (TMZ) is the current standard therapy. PAC-1, a procaspase-3 activating small molecule, is blood-brain barrier penetrant and has previously demonstrated ability to synergize with diverse pro-apoptotic chemotherapeutics. We studied if PAC-1 could enhance the activity of TMZ, and whether addition of PAC-1 to standard treatment would be feasible in spontaneous canine malignant gliomas. EXPERIMENTAL DESIGN: Using cell lines and online gene expression data, we identified procaspase-3 as a potential molecular target for most glioblastomas. We investigated PAC-1 as a single agent and in combination with TMZ against glioma cells in culture and in orthotopic rodent models of glioma. Three dogs with spontaneous gliomas were treated with an analogous human glioblastoma treatment protocol, with concurrent PAC-1. RESULTS: Procaspase-3 is expressed in gliomas, with higher gene expression correlating with increased tumor grade and decreased prognosis. PAC-1 is cytotoxic to glioma cells in culture and active in orthotopic rodent glioma models. PAC-1 added to TMZ treatments in cell culture increases apoptotic death, and the combination significantly increases survival in orthotopic glioma models. Addition of PAC-1 to TMZ and radiation was well-tolerated in 3 out of 3 pet dogs with spontaneous glioma, and partial to complete tumor reductions were observed. CONCLUSIONS: Procaspase-3 is a clinically relevant target for treatment of glioblastoma. Synergistic activity of PAC-1/TMZ in rodent models and the demonstration of feasibility of the combined regime in canine patients suggest potential for PAC-1 in the treatment of glioblastoma.

SELECTION OF CITATIONS
SEARCH DETAIL
...