Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Vet Comp Oncol ; 16(1): E76-E82, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28891260

ABSTRACT

Rabacfosadine (RAB), a novel double prodrug of the acyclic nucleotide phosphonate PMEG, preferentially targets neoplastic lymphocytes with reduced off target toxicity. Historical studies have suggested that every 21-day dosing is effective with acceptable toxicity. The purpose of this study was to evaluate RAB's safety and efficacy at 2 different doses every 21 days in dogs with relapsed B-cell lymphoma. Dogs that had failed 1 doxorubicin-based chemotherapy protocol were eligible for inclusion in this prospective trial. Once enrolled, dogs were randomized to receive RAB at either 0.82 mg/kg or 1.0 mg/kg as a 30-minute IV infusion every 21 days for up to 5 treatments. Response assessment and adverse event (AE) evaluation were performed every 21 days via VCOG criteria. Fifty dogs were enrolled, with 16 treated at 0.82 mg/kg and 34 treated at 1.0 mg/kg. The overall response rate was 74%, with 45% of dogs experiencing a complete response (CR). The median progression free intervals (PFIs) were 108 days, 172 days and 203 days for all dogs, all responders, and all CRs, respectively. Response rates and PFIs were similar in both treatment groups. The incidence of AEs, dose delays, dose reductions and withdrawals were not statistically different between the 2 groups. The AEs observed were similar to those previously reported and included hematologic, gastrointestinal, dermatologic and pulmonary AEs. One dog had grade 5 pulmonary fibrosis; otherwise, AEs resolved with supportive treatment. Rabacfosadine is a generally well tolerated, effective chemotherapy option for dogs with relapsed B-cell lymphoma.


Subject(s)
Alanine/analogs & derivatives , Antineoplastic Agents/therapeutic use , Dog Diseases/drug therapy , Lymphoma, B-Cell/veterinary , Purines/therapeutic use , Alanine/administration & dosage , Alanine/adverse effects , Alanine/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Dogs , Dose-Response Relationship, Drug , Lymphoma, B-Cell/drug therapy , Purines/administration & dosage , Purines/adverse effects , Recurrence , Treatment Outcome
2.
J Vet Intern Med ; 31(3): 872-878, 2017 May.
Article in English | MEDLINE | ID: mdl-28370378

ABSTRACT

BACKGROUND: Standard of care treatment for multicentric lymphoma in dogs remains doxorubicin (DOX)-based combination chemotherapy, but owners may hesitate to commit the time and financial resources to complete such a protocol, typically requiring 12-16 visits. Rabacfosadine (RAB), a double prodrug of the nucleotide analog 9-(2-phosphonylmethoxyethyl) guanine, has substantial single-agent activity in dogs with lymphoma, and a different mechanism of action than DOX. HYPOTHESIS/OBJECTIVES: Our objective was to evaluate the efficacy and adverse effect (AE) profile of alternating doses of RAB and DOX in dogs with naïve multicentric lymphoma. ANIMALS: Fifty-four dogs with previously untreated lymphoma. METHODS: Open-label, multicenter prospective clinical trial. Dogs received alternating RAB (1.0 mg/kg IV weeks 0, 6, 12) and DOX (30 mg/m2 IV weeks 3, 9, 15). Dogs that achieved complete response (CR) were followed by monthly evaluations. Complete clinicopathological evaluation and assessment of remission and AEs were performed every 21 days. RESULTS: The overall response rate was 84% (68%; CR; 16%; partial response [PR)]. The overall median progression-free interval (PFI) was 194 days (216 for CR and 63 for PR). Most AEs were mild and self-limiting: gastrointestinal and hematologic AEs were most common. Thirteen dogs experienced dermatologic AEs, and 2 dogs developed grade 5 pulmonary fibrosis. CONCLUSIONS AND CLINICAL IMPORTANCE: Alternating RAB/DOX generally was well tolerated and resulted in PFIs comparable to standard DOX-based multi-agent protocols, with fewer treatment visits. Most adverse events were mild or moderate and self-limiting. Further studies are warranted to explore long-term outcome and other RAB chemotherapy combinations.


Subject(s)
Alanine/analogs & derivatives , Antineoplastic Agents/therapeutic use , Dog Diseases/drug therapy , Doxorubicin/therapeutic use , Lymphoma/veterinary , Prodrugs/therapeutic use , Purines/therapeutic use , Alanine/administration & dosage , Alanine/adverse effects , Alanine/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Dogs , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Drug Administration Schedule/veterinary , Female , Lymphoma/drug therapy , Male , Prodrugs/administration & dosage , Prodrugs/adverse effects , Purines/administration & dosage , Purines/adverse effects , Treatment Outcome
4.
J Vet Intern Med ; 30(4): 1187-96, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27251585

ABSTRACT

BACKGROUND: The establishment and progression of metastases remains the life-limiting factor for dogs diagnosed with osteosarcoma (OS). The pattern of metastases is likely regulated through interactions between chemokine receptors and chemokines, and perturbations in these signaling cascades responsible for cytoskeletal organization and directional migration have the potential to alter metastatic cell trafficking behaviors. HYPOTHESIS: Zoledronate will impair directional migration of OS cells through downregulation of chemokine (C-X-C motif) receptor 4 (CXCR4) expression and functionality. SAMPLES: Nineteen archived tumor specimens and plasma from 20 dogs with OS. METHODS: Prospectively, the expressions of CXCR4 were studied in OS cell lines and spontaneous tumor samples. The effect of zoledronate on CXCR4 expression and functionality was investigated by characterizing responses in 3 OS cell lines. In 19 OS specimens and 20 dogs with OS, changes in CXCR4 expression and circulating CXCR4 concentrations were characterized in response to zoledronate therapy respectively. RESULTS: All canine OS cells express CXCR4, and zoledronate reduces CXCR4 expression and functionality by 27.7% (P < .0001), through augmented proteasome degradation and reduced prenylation of heterotrimeric G-proteins in 33% of tumor cell lines evaluated. In OS-bearing dogs, zoledronate reduces CXCR4 expressions by 40% within the primary tumor compared to untreated controls (P = .03) and also decreases the circulating concentrations of CXCR4 in 18 of 20 dogs with OS. CONCLUSIONS AND CLINICAL IMPORTANCE: Zoledronate can alter CXCR4 expression and functionality in OS cells, and consequent perturbations in CXCR4 intracellular signaling cascades might influence patterns of metastases.


Subject(s)
Bone Neoplasms/veterinary , Diphosphonates/pharmacology , Dog Diseases/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Imidazoles/pharmacology , Osteosarcoma/veterinary , Receptors, CXCR4/metabolism , Animals , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Cell Line, Tumor/drug effects , Diphosphonates/therapeutic use , Dog Diseases/blood , Dog Diseases/pathology , Dogs , Female , Imidazoles/therapeutic use , Male , Neoplasm Metastasis , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Zoledronic Acid
5.
Aust Vet J ; 94(5): 160-5, 2016 May.
Article in English | MEDLINE | ID: mdl-27113987

ABSTRACT

OBJECTIVES: The aims of this retrospective study were to identify clinical cases of dogs with appendicular osteosarcoma (OSA) in which hepatic metastasis was confirmed, to highlight the use of cytology for its diagnosis and to describe the radiographic and ultrasonographic appearances of the lesion. METHODS: Medical records were retrospectively reviewed for dogs with appendicular OSA and hepatic metastases between January 2005 and January 2013. Reviews of radiographs, ultrasounds and cytology were performed. RESULTS: Six dogs with appendicular OSA and hepatic metastases were identified. The ultrasonographic appearance of metastatic lesions varied, including hyperechoic with shadowing, hyperechoic without shadowing, hypoechoic and mixed echogenicity. In two cases, the hepatic metastases were also evident on thoracic radiographs. The mean survival time from diagnosis of appendicular OSA was 188 days (range 69-363 days) and from diagnosis of hepatic metastases was 35 days (range 2-69 days). Death was tumour-related in all cases. CONCLUSIONS: Hepatic metastasis varies widely in its ultrasonographic appearance. In three of six cases, hepatic metastasis was identified without concurrent pulmonary metastasis; therefore, abdominal ultrasound may be useful at regular intervals for patient evaluation, especially in clinical trials where accurate identification of the disease-free interval is crucial. Once hepatic metastasis is confirmed, survival times appear limited.


Subject(s)
Bone Neoplasms/veterinary , Dog Diseases/diagnosis , Liver Neoplasms/veterinary , Osteosarcoma/veterinary , Animals , Bone Neoplasms/diagnosis , Bone Neoplasms/pathology , Dog Diseases/diagnostic imaging , Dog Diseases/pathology , Dogs , Female , Liver Neoplasms/diagnosis , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/secondary , Male , Osteosarcoma/diagnosis , Osteosarcoma/diagnostic imaging , Radiography/veterinary , Retrospective Studies , Ultrasonography/veterinary
6.
J Vet Intern Med ; 29(6): 1584-94, 2015.
Article in English | MEDLINE | ID: mdl-26426813

ABSTRACT

BACKGROUND: Canine osteosarcoma (OS) is an aggressive sarcoma characterized by pathologic skeletal resorption and pulmonary metastases. A number of negative prognostic factors, including bone alkaline phosphatase, have been identified in dogs with OS, but the underlying biologic factors responsible for such observations have not been thoroughly investigated. Endothelin-1-mediated signaling is active during bone repair, and is responsible for osteoblast migration, survival, proliferation, and bone alkaline phosphatase expression. HYPOTHESIS: The endothelin-1 signaling axis is active in canine OS cells, and this pathway is utilized by malignant osteoblasts for promoting cellular migration, survival, proliferation, and bone alkaline phosphatase activities. ANIMALS: 45 dogs with appendicular OS. METHODS: The expressions of endothelin-1 and endothelin A receptor were studied in OS cell lines and in samples from spontaneously occurring tumors. Activities mediated by endothelin-1 signaling were investigated by characterizing responses in 3 OS cell lines. In 45 dogs with OS, bone alkaline phosphatase concentrations were correlated with primary tumor osteoproductivity. RESULTS: Canine OS cells express endothelin-1 and endothelin A receptor, and this signaling axis mediates OS migration, survival, proliferation, and bone alkaline phosphatase activities. In OS-bearing dogs, circulating bone alkaline phosphatase activities were positively correlated with primary tumor relative bone mineral densities. CONCLUSIONS AND CLINICAL IMPORTANCE: Canine OS cells express endothelin-1 and functional endothelin A receptors, with the potential for a protumorigenic signaling loop. Increases in bone alkaline phosphatase activity are associated with osteoblastic OS lesions, and might be an epiphenomenon of active endothelin-1 signaling or excessive osteoproduction within the localized bone microenvironment.


Subject(s)
Alkaline Phosphatase/metabolism , Dog Diseases/metabolism , Endothelin-1/metabolism , Osteosarcoma/veterinary , Signal Transduction/physiology , Alkaline Phosphatase/genetics , Animals , Bone Density , Bone Neoplasms/metabolism , Bone Neoplasms/veterinary , Cell Line, Tumor , Cell Migration Assays , Dogs , Endothelin-1/genetics , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Neoplastic/physiology , Osteosarcoma/metabolism , Receptor, Endothelin A/genetics , Receptor, Endothelin A/metabolism
7.
J Vet Intern Med ; 29(3): 759-69, 2015.
Article in English | MEDLINE | ID: mdl-25929293

ABSTRACT

Osteosarcoma is a malignant mesenchymal neoplasm that accounts for the majority of primary bone tumors in dogs and shares biological and clinical similarities with osteosarcoma in humans. Despite dose intensification with conventional cytotoxic therapies, survival times for dogs and humans diagnosed with high-grade osteosarcoma have not changed in the past 20 years, with the principal cause of mortality being the development of pulmonary metastases. Given the therapeutic plateau reached for delaying metastatic progression with cytotoxic agents, exploration of alterative adjuvant therapies for improving management of osteosarcoma micrometastases is clinically justified. Evidence suggests that osteosarcoma is an immunogenic tumor, and development of immunotherapies for the treatment of microscopic lung metastases might improve long-term outcomes. In this review, the history and foundational knowledge of immune interactions to canine osteosarcoma are highlighted. In parallel, immunotherapeutic strategies that have been explored for the treatment of canine osteosarcoma are summarized. With a greater understanding and awareness for how the immune system might be redirected toward combating osteosarcoma metastases, the rational development of diverse immune strategies for managing osteosarcoma holds substantial promise for transforming the therapeutic landscape and improving disease management in both dogs and human beings.


Subject(s)
Bone Neoplasms/veterinary , Dog Diseases/therapy , Immunotherapy/veterinary , Osteosarcoma/veterinary , Animals , Bone Neoplasms/immunology , Bone Neoplasms/therapy , Dog Diseases/immunology , Dogs , Immunity, Cellular , Immunity, Humoral , Immunotherapy/methods , Osteosarcoma/immunology , Osteosarcoma/therapy
8.
J Vet Intern Med ; 29(1): 268-75, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25572473

ABSTRACT

BACKGROUND: Canine osteosarcoma (OS) is associated with localized pain as a result of tissue injury from tumor infiltration and peritumoral inflammation. Malignant bone pain is caused by stimulation of peripheral pain receptors, termed nociceptors, which reside in the localized tumor microenvironment, including the periosteal and intramedullary bone cavities. Several nociceptive ligands have been determined to participate directly or indirectly in generating bone pain associated with diverse skeletal abnormalities. HYPOTHESIS: Canine OS cells actively produce nociceptive ligands with the capacity to directly or indirectly activate peripheral pain receptors residing in the bone tumor microenvironment. ANIMALS: Ten dogs with appendicular OS. METHODS: Expression of nerve growth factor, endothelin-1, and microsomal prostaglandin E synthase-1 was characterized in OS cell lines and naturally occurring OS samples. In 10 dogs with OS, circulating concentrations of nociceptive ligands were quantified and correlated with subjective pain scores and tumor volume in patients treated with standardized palliative therapies. RESULTS: Canine OS cells express and secrete nerve growth factor, endothelin-1, and prostaglandin E2. Naturally occurring OS samples uniformly express nociceptive ligands. In a subset of OS-bearing dogs, circulating nociceptive ligand concentrations were detectable but failed to correlate with pain status. Localized foci of nerve terminal proliferation were identified in a minority of primary bone tumor samples. CONCLUSIONS AND CLINICAL IMPORTANCE: Canine OS cells express nociceptive ligands, potentially permitting active participation of OS cells in the generation of malignant bone pain. Specific inhibitors of nociceptive ligand signaling pathways might improve pain control in dogs with OS.


Subject(s)
Dog Diseases/metabolism , Gene Expression Regulation, Neoplastic/physiology , Nociceptors/metabolism , Osteosarcoma/veterinary , Pain/veterinary , Animals , Cell Line, Tumor , Dogs , Endothelin-1/genetics , Endothelin-1/metabolism , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Ligands , Mice , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Osteosarcoma/metabolism , Osteosarcoma/pathology , Pain/metabolism , Prostaglandin-E Synthases , Rats
9.
Vet Comp Oncol ; 13(2): 89-97, 2015 Jun.
Article in English | MEDLINE | ID: mdl-23410097

ABSTRACT

The purpose of this retrospective study was to describe the biological behaviour of canine mandibular osteosarcoma (OSA) and to examine factors for their impact on metastasis-free interval (MFI) and survival time (ST). Records from dogs treated with mandibulectomy for OSA (1999-2007) were reviewed. Archived tumour samples were evaluated for mitotic index (MI) and tumour grade. Fifty dogs were included, 21 received chemotherapy. Twenty-nine dogs (58%) developed metastatic disease. The median MFI was 627 days, and median ST was 525 days. In univariate analysis MI > 40 was prognostic for decreased MFI and ST. Grade also influenced MFI and ST, with 5/21 (24%) dogs with grade II/III tumours metastasis-free at one year versus 16/22 (72%) dogs with grade I tumours (P = 0.002); and 5/21 (24%) dogs with grade II/III tumours alive versus 17/22 (77%) dogs with grade I tumours (P = 0.001). In multivariate analysis, histological grade and adjuvant chemotherapy were prognostic for MFI and ST.


Subject(s)
Dog Diseases/pathology , Mandibular Neoplasms/veterinary , Mitotic Index , Osteosarcoma/veterinary , Animals , Antineoplastic Agents/therapeutic use , Dog Diseases/drug therapy , Dog Diseases/surgery , Dogs , Mandibular Neoplasms/drug therapy , Mandibular Neoplasms/pathology , Mandibular Neoplasms/surgery , Neoplasm Grading , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Osteosarcoma/surgery , Prognosis , Retrospective Studies
10.
Vet Comp Oncol ; 13(3): 166-75, 2015 Sep.
Article in English | MEDLINE | ID: mdl-23489774

ABSTRACT

Osteosarcoma is an aggressive malignancy and represents the most frequent primary bone malignancy of dogs and humans. Prognostic factors reported for osteosarcoma include tumour size, presence of metastatic disease and serum alkaline phosphatase (ALP) concentration at the time of diagnosis. To date, there have been no studies to determine whether the behaviour of osteosarcoma cells differ based on serum ALP concentration. Here, we report on the generation of six canine osteosarcoma cell lines from osteosarcoma-bearing dogs with differences in serum ALP concentration. To determine whether in vitro behaviour differs between primary osteosarcoma cell lines generated from patients with normal or increased serum ALP, assays were performed to evaluate proliferation, migration, invasion and chemosensitivity. There were no significant differences in cell proliferation, migration, invasion or chemosensitivity between cell lines associated with normal or increased serum ALP concentration.


Subject(s)
Alkaline Phosphatase/blood , Bone Neoplasms/veterinary , Dog Diseases/blood , Dog Diseases/physiopathology , Osteosarcoma/veterinary , Analysis of Variance , Animals , Bone Neoplasms/blood , Bone Neoplasms/physiopathology , Cell Line, Tumor , Dogs , Female , In Vitro Techniques , Male , Osteosarcoma/blood , Osteosarcoma/physiopathology , Prognosis
11.
J Vet Intern Med ; 28(3): 894-904, 2014.
Article in English | MEDLINE | ID: mdl-24684686

ABSTRACT

BACKGROUND: Transforming growth factor beta 1 (TGFß1) is a pleiotropic cytokine that contributes to reparative skeletal remodeling by inducing osteoblast proliferation, migration, and angiogenesis. Organic bone matrix is the largest bodily reservoir for latent TGFß1, and active osteoblasts express cognate receptors for TGFß1 (TGFßRI and TGFßRII). During malignant osteolysis, TGFß1 is liberated from eroded bone matrix and promotes local progression of osteotropic solid tumors by its mitogenic and prosurvival activities. HYPOTHESIS: Canine osteosarcoma (OS) cells will possess TGFß1 signaling machinery. Blockade of TGFß1 signaling will attenuate pro-tumorigenic activities in OS cells. Naturally occurring primary OS samples will express cognate TGFß1 receptors; and in dogs with OS, focal malignant osteolysis will contribute to circulating TGFß1 concentrations. ANIMALS: Thirty-three dogs with appendicular OS. METHODS: Expression of TGFß1 and its cognate receptors, as well as the biologic effects of TGFß1 blockade, was characterized in OS cells. Ten spontaneous OS samples were characterized for TGFßRI/II expressions by immunohistochemistry. In 33 dogs with OS, plasma TGFß1 concentrations were quantified and correlated with bone resorption. RESULTS: Canine OS cells secrete TGFß1, express cognate receptors, and TGFß1 signaling blockade decreases proliferation, migration, and vascular endothelial growth factor secretion. Naturally occurring OS samples abundantly and uniformly express TGFßRI/II, and in OS-bearing dogs, circulating TGFß1 concentrations correlate with urine N-telopeptide excretion. CONCLUSIONS AND CLINICAL IMPORTANCE: Canine OS cells possess TGFß1 signaling machinery, potentially allowing for the establishment of an autocrine and paracrine pro-tumorigenic signaling loop. As such, TGFß1 inhibitors might impede localized OS progression in dogs.


Subject(s)
Bone Neoplasms/veterinary , Osteosarcoma/veterinary , Transforming Growth Factor beta1/physiology , Animals , Blotting, Western , Bone Neoplasms/physiopathology , Cell Line, Tumor , Dog Diseases , Dogs , Enzyme-Linked Immunosorbent Assay , Osteosarcoma/physiopathology , Pyrazoles/pharmacology , Pyrroles/pharmacology , Signal Transduction/drug effects , Transforming Growth Factor beta1/biosynthesis , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta2/biosynthesis , Transforming Growth Factor beta2/physiology , Vascular Endothelial Growth Factor A/physiology
12.
J Vet Intern Med ; 27(6): 1305-17, 2013.
Article in English | MEDLINE | ID: mdl-24128266

ABSTRACT

Noninferiority trials are clinical studies designed to demonstrate that an investigational drug is at least as effective as an established treatment within a predetermined margin. They are conducted, in part, because of ethical concerns of administering a placebo to veterinary patients when an established effective treatment exists. The use of noninferiority trial designs has become more common in veterinary medicine with the increasing number of established veterinary therapeutics and the desire to eliminate potential pain or distress in a placebo-controlled study. Selecting the appropriate active control and an a priori noninferiority margin between the investigational and active control drug are unique and critical design factors for noninferiority studies. Without reliable historical knowledge of the disease response in the absence of treatment and of the response to the selected active control drug, proper design and interpretation of a noninferiority trial is not possible. Despite the appeal of conducting noninferiority trials to eliminate ethical concerns of placebo-controlled studies, there are real limitations and possible ethical conundrums associated with noninferiority trials. The consequences of incorrect study conclusions because of poor noninferiority trial design need careful attention. Alternative trial designs to typical noninferiority studies exist, but these too have limitations and must also be carefully considered.


Subject(s)
Clinical Trials as Topic/veterinary , Evidence-Based Medicine/methods , Research Design/standards , Veterinary Medicine/methods , Animals , Cats , Clinical Trials as Topic/methods , Clinical Trials as Topic/standards , Dogs , Evidence-Based Medicine/standards , Veterinary Medicine/standards
13.
J Vet Intern Med ; 27(4): 955-63, 2013.
Article in English | MEDLINE | ID: mdl-23734720

ABSTRACT

BACKGROUND: In dogs with appendicular osteosarcoma (OSA), increased pretreatment serum bone-specific alkaline phosphatase (BALP) activity is a negative prognostic factor, associated with shorter disease-free intervals and survival times, but a biologic basis for observed differential serum BALP activities in canine OSA patients remains incompletely defined. OBJECTIVE: Serum BALP activity will correlate with absolute tumor burden in dogs with OSA. ANIMALS: This study included 96 client-owned dogs with appendicular OSA. METHODS: In canine OSA cell lines, the expression and membranous release of BALP was evaluated in vitro. The correlation between serum BALP activity and radiographic primary tumor size was evaluated in OSA-bearing dogs. In dogs developing visceral OSA metastases, serial changes in serum BALP activities were evaluated in relation to progression of macroscopic metastases, and visceral metastatic OSA cells were evaluated for BALP expression. RESULTS: In vitro, BALP expression was not associated with either tumorigenic or metastatic phenotype, rather the quantity of membranous BALP released was proportional with cell density. In dogs devoid of macroscopic metastases, there was a positive correlation between serum BALP activity and absolute primary tumor size. In dogs with progressive OSA metastases, serum BALP activity increased and coincided with the development of macroscopic metastases. OSA cells derived from visceral metastatic lesions retained BALP expression. CONCLUSIONS AND CLINICAL IMPORTANCE: Tumor burden is a determinant of serum BALP activity in dogs with appendicular OSA. The association between increased pretreatment BALP activity and negative clinical prognosis may simply be attributed to greater initial tumor burden, and consequently more advanced tumor stage.


Subject(s)
Alkaline Phosphatase/metabolism , Bone Neoplasms/metabolism , Dog Diseases/pathology , Osteosarcoma/veterinary , Alkaline Phosphatase/genetics , Animals , Cell Line, Tumor , Dog Diseases/enzymology , Dogs , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Male , Osteosarcoma/metabolism
14.
J Vet Intern Med ; 26(1): 126-34, 2012.
Article in English | MEDLINE | ID: mdl-22171552

ABSTRACT

BACKGROUND: Cathepsin K (CatK) is a lysosomal protease with collagenolytic activity, and its secretion by osteoclasts is responsible for degrading organic bone matrix. People with pathologic bone resorption have higher circulating CatK concentrations. HYPOTHESIS: Canine osteosarcoma (OS) cells will possess CatK, and its secretion will be cytokine inducible. Circulating CatK concentrations will be increased in dogs with OS, and will be a surrogate marker of bone resorption. ANIMALS: Fifty-one dogs with appendicular OS and 18 age- and weight-matched healthy control dogs. METHODS: In a prospective study, expressions of CatK mRNA and protein were investigated in OS cells. The inducible secretion and proteolytic activity of CatK from OS cells was assessed in vitro. Serum CatK concentrations were quantified in normal dogs and dogs with OS and its utility as a bone resorption marker was evaluated in dogs with OS treated with palliative radiation and antiresorptive agents. RESULTS: Canine OS cells contain preformed CatK within cytoplasmic vesicles. In OS cells, TGFß1 induced the secretion of CatK, which degraded bone-derived type I collagen in vitro. CatK concentrations were higher in dogs with OS than healthy dogs (11.3 ± 5.2 pmol/L versus 8.1 ± 5.0 pmol/L, P = .03). In a subset of dogs with OS, pretreatment CatK concentrations gradually decreased after palliative radiation and antiresorptive treatment, from 9.3 ± 3.2 pmol/L to 5.0 ± 3.1 pmol/L, P = .03. CONCLUSIONS AND CLINICAL IMPORTANCE: Canine OS is associated with pathologic bone resorption, and CatK inhibitors might aid in the management of canine OS-related malignant osteolysis.


Subject(s)
Bone Neoplasms/veterinary , Cathepsin K/biosynthesis , Dog Diseases/enzymology , Osteosarcoma/veterinary , Animals , Blotting, Western/veterinary , Bone Density Conservation Agents/pharmacology , Bone Neoplasms/enzymology , Cathepsin K/genetics , Cell Line, Tumor , Collagen Type I/metabolism , Dogs , Immunohistochemistry/veterinary , Osteosarcoma/enzymology , Prospective Studies , RNA/chemistry , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Transforming Growth Factor beta1/pharmacology
15.
J Vet Intern Med ; 24(5): 1131-7, 2010.
Article in English | MEDLINE | ID: mdl-20666981

ABSTRACT

BACKGROUND: Removal of leukocytes (LR) has been shown to eliminate or attenuate many of the adverse effects of transfusion in experimental animals and humans. HYPOTHESIS/OBJECTIVES: Transfusion of stored packed red blood cells (pRBCs) is associated with an inflammatory response in dogs and prestorage LR attenuates the inflammatory response. ANIMALS: Thirteen random-source, clinically healthy, medium and large breed dogs. METHODS: Experimental study. On day 0, animals were examined and baseline blood samples were collected for analysis. Whole blood was then collected for processing with and without LR, and stored as pRBC. Twenty-one days later, stored pRBCs were transfused back to the donor. Blood samples were collected before and 1 and 3 days after transfusion. RESULTS: In the dogs that received non-LR pRBCs (n = 6) there was a significant increase from baseline in white blood cell count from a mean (SD) of 8.20 (2.74) to 13.95 (4.60) × 10(3) cells/µL (P < .001) and in segmented neutrophil count from a mean (SD) of 5.76 (2.70) to 11.91 (4.71) × 10(3) cells/µL (P < .001). There were also significant increases in fibrinogen from a mean (SD) of 129.7 (24.2) to 268.6 (46.7) mg/dL (P < .001) and C-reactive protein from a mean (SD) of 1.9 (2.1) to 78.3 (39.3) µg/mL (P < .001). There was no significant increase from baseline in any of the markers in the dogs that received LR pRBC (n = 5). CONCLUSIONS AND CLINICAL IMPORTANCE: There is a profound inflammatory response to transfusion in normal dogs, which is eliminated by LR of the pRBC units.


Subject(s)
Blood Preservation/veterinary , Dog Diseases/etiology , Erythrocyte Transfusion/veterinary , Inflammation/veterinary , Leukocyte Reduction Procedures/veterinary , Animals , Dogs , Erythrocyte Transfusion/adverse effects , Female , Inflammation/etiology , Inflammation/prevention & control , Male
16.
J Vet Intern Med ; 23(1): 152-60, 2009.
Article in English | MEDLINE | ID: mdl-19175734

ABSTRACT

BACKGROUND: Canine osteosarcoma (OSA) causes focal malignant osteolysis leading to severe pain. Despite the documented efficacy of radiotherapy or IV aminobisphosphonates for managing cancer bone pain, their potential combined therapeutic value has not been reported in OSA-bearing dogs. HYPOTHESIS: Pamidronate combined with standardized palliative therapy will improve pain control and bone biologic effects in OSA-bearing dogs. ANIMALS: Fifty dogs with appendicular OSA treated with standardized palliative therapy and either pamidronate or sterile saline. METHODS: Randomized, prospective, double-blinded, placebo-controlled study. Treatment responses for dogs receiving standardized palliative therapy with (n = 26) or without (n = 24) adjuvant pamidronate were serially evaluated for changes in subjective pain scores, urine N-telopeptide (NTx) excretion, primary tumor relative bone mineral density (rBMD), and computerized pressure platform gait analysis. RESULTS: Median duration of subjective pain relief for dogs treated with adjuvant pamidronate or placebo was 76 and 75 days, respectively (P= .39). Forty percent (20/50; pamidronate [11/26] and placebo [9/24]) of dogs experienced durable analgesia, defined by pain alleviation > or =112 days. For patients achieving durable pain control, dogs treated with pamidronate achieved greater reductions in NTx excretion and larger increases in rBMD compared with placebo controls. Changes in peak vertical force assessed by computerized pressure platform gait analysis correlated with pain alleviation in OSA-bearing dogs. CONCLUSIONS AND CLINICAL IMPORTANCE: Combining pamidronate with standardized palliative therapy is safe, but does not clearly improve pain alleviation. However, in dogs achieving durable pain control, adjuvant pamidronate appears to decrease focal bone resorption in the local tumor microenvironment.


Subject(s)
Diphosphonates/administration & dosage , Diphosphonates/therapeutic use , Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Pain Management , Radiotherapy/veterinary , Analgesia/veterinary , Analgesics/administration & dosage , Analgesics/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Bone Density Conservation Agents/administration & dosage , Bone Density Conservation Agents/therapeutic use , Dog Diseases/drug therapy , Dog Diseases/radiotherapy , Dogs , Double-Blind Method , Extremities/pathology , Female , Male , Osteosarcoma/complications , Osteosarcoma/veterinary , Pamidronate
17.
J Vet Intern Med ; 22(5): 1181-8, 2008.
Article in English | MEDLINE | ID: mdl-18638015

ABSTRACT

BACKGROUND: The tropomyosin-related kinase A (TrkA) proto-oncogene encodes for a receptor that binds with high affinity to the neurotrophin ligand, nerve growth factor (NGF). Intracellular signaling mediated by the TrkA/NGF axis orchestrates neuronal cell differentiation, mitogenesis, and survival. Interestingly, TrkA also is expressed by bone forming cells, and its signaling promotes antiapoptotic effects in actively dividing osteoblasts. HYPOTHESIS: In canine immortalized cell lines and naturally occurring tumor samples, osteosarcoma (OSA) cells will express TrkA. In canine OSA cell lines, TrkA signaling will promote cell mitogenesis and survival. METHODS: In vitro, TrkA expression in canine OSA cell lines was assessed by reverse transcriptase-polymerase chain reaction, flow cytometry, and immunocytochemistry. In vitro, the involvement of TrkA-mediated signaling for cell mitogenesis and survival were investigated with commercially available assays. In vivo, TrkA expression was evaluated in primary tumors and pulmonary metastases with immunocytochemistry and immunohistochemistry, respectively. RESULTS: In vitro, canine OSA cells expressed TrkA mRNA and protein. Ligation of TrkA with exogenous NGF did not induce mitogenesis. Blockade of TrkA signaling with either a protein kinase inhibitor or NGF-neutralizing antibody induced apoptosis of canine OSA cell lines. In vivo, the majority (10/15) of canine OSA primary tumors and pulmonary metastases (9/12) expressed TrkA protein. CONCLUSIONS AND CLINICAL IMPORTANCE: Canine OSA cells express TrkA, and its signaling protects against apoptosis. Most dogs with spontaneously arising OSA express TrkA within their primary tumors and pulmonary metastatic lesions, warranting further investigations with TrkA antagonists as a novel treatment option for canine OSA.


Subject(s)
Dog Diseases/metabolism , Gene Expression Regulation, Neoplastic/physiology , Nerve Tissue Proteins/metabolism , Osteosarcoma/metabolism , Protein Kinases/metabolism , Animals , Apoptosis , Cell Line, Tumor , Dogs , Extremities/pathology , Lung Neoplasms/secondary , Nerve Tissue Proteins/genetics , Osteosarcoma/pathology , Protein Kinases/genetics , Signal Transduction
18.
J Vet Intern Med ; 22(4): 1008-13, 2008.
Article in English | MEDLINE | ID: mdl-18564220

ABSTRACT

BACKGROUND: Various bone resorption markers in humans are useful for supporting the diagnosis of malignant skeletal pathology, with certain bone resorption markers appearing to be more discriminatory for detecting cancer-induced osteolysis than others. Canine osteosarcoma (OSA) is characterized by focal bone destruction, but a systematic investigation for determining which bone resorption marker best supports the diagnosis of OSA in dogs has not been reported. HYPOTHESIS: Dogs with OSA will have increased concentrations of bone resorption markers compared with healthy dogs and dogs with orthopedic disorders. Differences will exist among various bone resorption markers for their ability to support the diagnosis of malignant osteolysis in dogs with OSA. ANIMALS: Single time point, cross-sectional, cohort study including dogs with OSA (n = 20) or orthopedic disorders (n = 20) and healthy dogs (n = 22). METHODS: Basal concentrations of urine and serum N-telopeptide (NTx), urine and serum C-telopeptide (CTx), and urine deoxypyridinoline (DPD) were compared among all 3 groups. RESULTS: Compared with healthy dogs and dogs with orthopedic disorders, urine NTx, serum NTx, and serum CTx concentrations were significantly increased in dogs with OSA. For urine NTx and serum NTx, the calculated lower and upper 95% confidence limits in dogs with OSA did not overlap with dogs diagnosed with orthopedic disorders or healthy dogs. CONCLUSIONS AND CLINICAL IMPORTANCE: Of the markers evaluated in this study, urine NTx and serum NTx appear to be the most discriminatory resorption markers supporting the diagnosis of focal malignant osteolysis in dogs with OSA.


Subject(s)
Biomarkers/blood , Biomarkers/urine , Bone Resorption/metabolism , Dog Diseases/metabolism , Osteoarthritis/veterinary , Osteosarcoma/veterinary , Animals , Dogs , Female , Male , Osteoarthritis/metabolism , Osteosarcoma/metabolism
19.
J Vet Intern Med ; 22(3): 602-8, 2008.
Article in English | MEDLINE | ID: mdl-18466248

ABSTRACT

BACKGROUND: Chemokine receptors (CXCRs) are transmembrane proteins classically studied for their participation in leukocyte homing. By their binding of cognate ligands, CXCRs orchestrate key cellular processes, including directional migration. Several different CXCRs are expressed on cancer cells and dictate tissue-specific metastases. In pediatric osteosarcoma (OSA), CXCR4 expression by tumor cells may participate in metastasis to tissues containing CXCL12, the partnering ligand for CXCR4. Canine and pediatric OSA share many biological similarities, including preferential metastasis to lung, bone, and lymph node. HYPOTHESIS: In canine immortalized cell lines and naturally occurring tumor samples, OSA cells will express CXCR4. In canine OSA cell lines, CXCR4 will participate in directional cell migration. METHODS: In vitro, CXCR4 expression in canine OSA cell lines was assessed by reverse-transcriptase polymerase chain reaction, Western blot analysis, flow cytometry, and immunocytochemistry. In vitro, involvement of CXCR4-mediated signaling for directional migration was investigated with a commercial assay. In vivo, CXCR4 expressions were evaluated in primary tumors and pulmonary metastases with immunocytochemistry and immunohistochemistry, respectively. RESULTS: In vitro, canine OSA cells express CXCR4 mRNA and protein. Ligation of CXCR4 with exogenous CXCL12 results in directional migration of canine OSA cell lines. In vivo, majority (8/11) of the canine OSA primary tumors, but minority (2/8) of the pulmonary metastases express CXCR4 protein. CONCLUSIONS AND CLINICAL IMPORTANCE: Canine OSA cells express CXCR4, and its signaling participates in directional migration. Most dogs with spontaneously arising OSA express CXCR4 within their primary tumors.


Subject(s)
Bone Neoplasms/veterinary , Dog Diseases/metabolism , Osteosarcoma/veterinary , Receptors, CXCR4/metabolism , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Chemokine CXCL12/metabolism , Dog Diseases/genetics , Dogs , Extremities , Gene Expression Regulation, Neoplastic , Osteosarcoma/genetics , Osteosarcoma/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, CXCR4/genetics , Signal Transduction
20.
J Vet Intern Med ; 22(2): 380-7, 2008.
Article in English | MEDLINE | ID: mdl-18346146

ABSTRACT

BACKGROUND: Malignant osteolysis is a process whereby cancer cells in concert with osteoclasts erode bone matrix. Aminobisphosphonates (NBPs) such as zoledronate induce osteoclast apoptosis and thereby decrease malignant skeletal destruction, severity of bone pain, and frequency of pathologic fracture. HYPOTHESIS: IV-administered zoledronate will reduce homeostatic bone turnover in healthy dogs and pathologic bone resorption in dogs diagnosed with primary and secondary bone tumors. ANIMALS: Six healthy dogs and 20 dogs with naturally occurring primary or metastatic bone tumors were administered zoledronate IV. METHODS: Prospective study: In all dogs, healthy (n = 6) and with malignant osteolysis (n = 20), the bone biologic effects of zoledronate were evaluated by quantifying changes in serum C-telopeptide (CTx) or urine N-telopeptide (NTx) concentrations or both. In dogs with osteosarcoma (OSA) (n = 10), serial changes in tumor relative bone mineral density (rBMD) assessed by dual-energy x-ray absorptiometry were used to characterize zoledronate's antiresorptive effects within the immediate tumor microenvironment. Additionally, the biochemical tolerability of zoledronate was assessed in 9 dogs receiving multiple (> or =2) consecutive treatments. RESULTS: All dogs had significant reductions in serum CTx or urine NTx concentrations or both after zoledronate administration. In a subset of dogs with appendicular OSA, reduced urine NTx concentrations and increased primary tumor rBMD coincided with improved limb usage as reported by pet owners in dogs treated with zoledronate and concurrent oral analgesics. Multiple zoledronate infusions were not associated with biochemical evidence of toxicosis. CONCLUSIONS AND CLINICAL IMPORTANCE: In dogs with skeletal neoplasms, IV-administered zoledronate exerts bone biologic effects, appears safe, and can provide pain relief.


Subject(s)
Bone Density Conservation Agents/pharmacology , Bone and Bones/drug effects , Diphosphonates/pharmacology , Dog Diseases/drug therapy , Imidazoles/pharmacology , Osteolysis/veterinary , Animals , Bone Density/drug effects , Bone Density Conservation Agents/therapeutic use , Cohort Studies , Diphosphonates/therapeutic use , Dog Diseases/etiology , Dogs , Female , Imidazoles/therapeutic use , Male , Neoplasms/complications , Neoplasms/veterinary , Osteolysis/drug therapy , Osteolysis/etiology , Prospective Studies , Zoledronic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...