Safety of administering canine melanoma DNA vaccine to cats with malignant melanoma

2017 ◽  
Vol 30 (12) ◽  
pp. 6-7
2016 ◽  
Vol 19 (2) ◽  
pp. 224-230 ◽  
Author(s):  
Luminita Sarbu ◽  
Barbara E Kitchell ◽  
Philip J Bergman

Objectives A xenogeneic human tyrosinase DNA vaccine was developed for treatment of dogs with oral malignant melanoma (Oncept; Merial). No studies have evaluated the safety or efficacy of this vaccine in cats. The purpose of this study was to evaluate the safety of the canine melanoma vaccine in cats diagnosed with melanoma. Methods Medical records were reviewed from cats diagnosed with malignant melanoma and treated with the canine melanoma DNA vaccine (Oncept). Data regarding signalment, melanoma location, treatments received, vaccine adverse effects and cause of death were collected. Results A total of 114 melanoma vaccines were administered to 24 cats. Seven cats (11.4%) had clinical adverse effects from a total of 13 vaccines classified as grade 1 or 2 based on the Veterinary Cooperative Oncology Group’s common terminology criteria for adverse events v1.1. These included pain on vaccine administration, brief muscle fasciculation, transient inappetence, depression, nausea and mild increase in pigmentation at the injection site. Nineteen cats were deceased at study close. The most common cause of death was melanoma (14 cats). Hematological and biochemical changes were observed in six cats, five of which had concurrent disease or treatments that likely caused or greatly contributed to the laboratory abnormalities found. Therefore, these adverse events were considered unlikely to be caused by the melanoma vaccine. One cat had transient grade 1 hypoalbuminemia, which was possibly caused by the vaccination but not thoroughly evaluated. Conclusions and relevance The canine melanoma DNA vaccine can be safely administered to cats, with minimal risk of adverse effects.


Author(s):  
M. Nawrath ◽  
J. Heinrich ◽  
B. Strack ◽  
J. Pavlovic ◽  
Karin Moelling

Author(s):  
Barbara J. Mangold ◽  
Jennifer E. Flower ◽  
Kristine E. Burgess ◽  
Elizabeth A. McNiel ◽  
Jeffrey C. Phillips ◽  
...  

Abstract CASE DESCRIPTION A 25-year-old 4.4-kg male aquarium-hatched African penguin (Spheniscus demersus) was evaluated because of a raised 1.5 × 0.5-cm pigmented mass extending from within the right naris noted 2 days earlier. CLINICAL FINDINGS The penguin had a raised pigmented mass extending out from the right naris and onto the upper beak. Histologic examination of excisional biopsy specimens confirmed a diagnosis of malignant melanoma. A treatment plan including administration of meloxicam, radiation therapy, and immunotherapy was initiated. TREATMENT AND OUTCOME Treatment with meloxicam (0.2 mg/kg, PO, q 24 h) was initiated and continued for a total of 45 weeks; however, the medication was discontinued for a period of 6 weeks because of the risk of toxic effects in the chick that the penguin was feeding at that time. The penguin underwent local hypofractionated radiation therapy and received 4 once weekly 8-Gy fractions of radiation (total radiation dose, 32 Gy). The penguin was administered a canine melanoma vaccine transdermally every other week for 4 doses, with a booster injection given 7 months after the first dose. Treatment with the vaccine appeared to have no adverse effects. The penguin’s pre- and postvaccination tyrosinase-specific antibody titers were measured with an anti–human tyrosinase-specific ELISA, and a 3-fold titer increase indicated a positive humoral immune response to the canine melanoma vaccination. The penguin died of unrelated causes 54 weeks after initial diagnosis, and there was no evidence of metastasis on necropsy. CLINICAL RELEVANCE These case findings suggested that vaccination with a canine melanoma vaccine may be a safe and useful adjunct treatment for management of malignant melanoma in penguins.


2021 ◽  
Vol 17 (1) ◽  
Author(s):  
Sarah Bernard ◽  
Andrew C. Poon ◽  
Peyton M. Tam ◽  
Anthony J. Mutsaers

Abstract Background Malignant melanoma in dogs is considered to be largely resistant to conventional chemotherapy, although responses to carboplatin have been documented. Invasion and early metastasis are common features of certain melanoma subtypes that contribute to tumour progression despite aggressive local and systemic therapy. Upregulation of the PI3K/AKT/mTOR pathway has been observed in canine malignant melanoma and may represent a potential target for therapy. Rapamycin (sirolimus) and everolimus are commercially available small molecule inhibitors that target mTOR and therefore may have anticancer activity in canine melanoma. It was hypothesized that there is synergism between rapamycin or everolimus and platinum chemotherapy, and that combination drug treatment would inhibit target/downstream proteins involved in cell viability/proliferation and increase cell death in canine melanoma cells. It was further hypothesized that rapamycin or everolimus would impact metabolism by reducing glycolysis in these cells. Four canine melanoma cell lines were treated in vitro with rapamycin and everolimus as sole treatment or combined with carboplatin. Cell viability, apoptosis, target modulation, and glycolytic metabolism were evaluated by crystal violet colourimetric assay, Annexin V/PI flow cytometry, western blotting, and Seahorse bioanalyzer, respectively. Results When combined with carboplatin chemotherapy, rapamycin or everolimus treatment was overall synergistic in reducing cell viability. Carboplatin-induced apoptosis was noted at 72 h after treatment compared to the vehicle control. Levels of phosphorylated mTOR were reduced by rapamycin and everolimus in all four cell lines, but activation of the downstream protein p70S6K was not consistently reduced by treatment in two of the cell lines. Both mTOR inhibitors decreased the extracellular acidification rate of canine melanoma cells, indicating reduced cancer cell glycolytic activity. Conclusions Inhibition of mTOR by rapalogs, such as rapamycin and everolimus combined with carboplatin chemotherapy may have activity in canine melanoma. Future mechanistic investigation is warranted, including in vivo assessment of this combination therapy.


Vaccine ◽  
2006 ◽  
Vol 24 (21) ◽  
pp. 4582-4585 ◽  
Author(s):  
P.J. Bergman ◽  
M.A. Camps-Palau ◽  
J.A. McKnight ◽  
N.F. Leibman ◽  
D.M. Craft ◽  
...  

2013 ◽  
Vol 1 (1) ◽  
pp. 20 ◽  
Author(s):  
Jianda Yuan ◽  
Geoffrey Y Ku ◽  
Matthew Adamow ◽  
Zhenyu Mu ◽  
Sapna Tandon ◽  
...  

2021 ◽  
Vol 8 ◽  
Author(s):  
Hiroto Takeuchi ◽  
Satoru Konnai ◽  
Naoya Maekawa ◽  
Satoshi Takagi ◽  
Hiroshi Ohta ◽  
...  

Cancer cells can evade host immune systems via multiple mechanisms. Transforming growth factor beta 1 (TGF-β1) is an immunosuppressive cytokine that induces regulatory T cell (Tregs) differentiation and is involved in immune evasion mechanisms in cancer. The inhibition of the TGF-β1 signaling pathway can suppress cancer progression and metastasis through the modulation of anticancer immune responses. However, to best of our knowledge, no implementation of treatments targeting TGF-β1 has been reported in dog cancers. This study aimed to examine whether TGF-β1 is upregulated in canine cancers. We measured TGF-β1 concentrations in culture supernatants of canine melanoma cell lines and in serum samples from dogs with oral malignant melanoma. TGF-β1 production was observed in several cell lines, and serum TGF-β1 levels were elevated in dogs with oral malignant melanoma. Interestingly, the addition of recombinant TGF-β1 to canine peripheral blood mononuclear cell cultures decreased Th1 cytokine production and increased differentiation of CD4+CD25+Foxp3+ lymphocytes, suggesting that TGF-β1 is immunosuppressive in canine immune systems. We developed a decoy receptor for TGF-β, namely TGF-βRII-Ig, by identifying an open reading frame of the canine TGFBR2 gene. TGF-βRII-Ig was prepared as a recombinant fusion protein of the extracellular region of canine TGF-βRII and the Fc region of canine IgG-B. As expected, TGF-βRII-Ig bound to TGF-β1. In the presence of TGF-β1, the treatment with TGF-βRII-Ig increased Th1 cytokine production and decreased the differentiation of CD4+CD25+Foxp3+ lymphocytes. Our results suggest that TGF-βRII-Ig competitively inhibits the immunosuppressive effects of TGF-β1 and thereby activates immune responses. This study demonstrated the potential of TGF-βRII-Ig as a novel biologic for canine melanoma.


2002 ◽  
Vol 5 (3) ◽  
pp. 291-299 ◽  
Author(s):  
Maki Tanaka ◽  
Yasufumi Kaneda ◽  
Shigeyuki Fujii ◽  
Tomoki Yamano ◽  
Kahoko Hashimoto ◽  
...  

2010 ◽  
Vol 25 (1) ◽  
pp. 94-99 ◽  
Author(s):  
C.A. Manley ◽  
N.F. Leibman ◽  
J.D. Wolchok ◽  
I.C. Rivière ◽  
S. Bartido ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document