Lauren Alfino
DVM
Dr. Alfino is a medical oncology intern at Michigan State University. She received her DVM degree from Colorado State University in 2022 and completed a small animal rotating internship at a private practice in South Carolina. Her previous research has focused on small molecule inhibitors in canine and human osteosarcoma. She has a special interest in comparative oncology, with a focus on helping veterinary patients while concurrently developing treatment options with translational value.
Read Articles Written by Lauren AlfinoAlison Masyr
DVM, MS, DACVIM (Oncology)
Dr. Masyr received her DVM degree from the University of Illinois College of Veterinary Medicine. She then completed a small animal rotating internship at the University of Minnesota College of Veterinary Medicine and a medical oncology and clinical trials specialty internship, a master of science degree, and a medical oncology residency at the University of Illinois College of Veterinary Medicine. Dr. Masyr is currently an assistant clinical track professor in the small animal clinical sciences department at the Michigan State University College of Veterinary Medicine. Her research interests focus on tumor metabolomics, novel therapeutics, and clinical trials.
Read Articles Written by Alison MasyrMast cell tumors (MCTs) are common cutaneous neoplasms in dogs. This article discusses recent updates in the management of mast cell disease, specifically the use of the MCT prognostic panel; evaluation of regional and sentinel lymph nodes for the presence of metastasis; and local and systemic treatments, including novel therapeutics such as tigilanol tiglate injection and gilvetmab. Ideal scenarios for prognostic panel use and an overview of the implications of lymph node metastasis for survival and systemic therapy are also provided.
Take-Home Points
- The mast cell tumor (MCT) prognostic panel is useful in guiding treatment recommendations for histologically low-grade MCTs with equivocal histologic features or clinical history concerning for aggressive behavior.
- While detecting metastatic lymph nodes can be challenging, addressing metastatic lymph nodes through surgery or radiation can improve survival. In certain cases, addressing the metastatic lymph node may eliminate the recommendation for systemic therapy.
- Local therapy for MCTs includes surgery, radiation therapy, electrochemotherapy, and intralesional agents. Tigilanol tiglate injection is an intralesional treatment that causes wound formation, which, in rare cases, can be extensive.
- Proportional surgical margin recommendations appear adequate for achieving local control of low-grade MCTs up to 2 cm in diameter.
- Some options for systemic therapy for MCTs are cytotoxic chemotherapy, tyrosine kinase inhibitors, a new checkpoint inhibitor (gilvetmab), and/or corticosteroids. Additional supportive care most often includes histamine antagonist and/or proton pump inhibitor medications.
Mast cell tumors (MCTs) are common cutaneous neoplasms in dogs. As treatment recommendations and prognostic factors for MCTs continually change, this article aims to demystify the clinical approach to MCTs by reviewing indications for use of the MCT prognostic panel, the clinical relevance of regional lymph node metastasis, and various local and systemic therapies.
Use of the MCT Prognostic Panel
An MCT prognostic panel is available through IDEXX and the Michigan State University Veterinary Diagnostic Laboratory. This multifaceted panel is a valuable tool for further characterizing cutaneous and subcutaneous MCTs with equivocal histopathology. The panel includes proliferative indices, c-kit mutation status (exons 8 and 11), and KIT localization pattern. These findings are most informative when considered together rather than individually.1,2
For histologically high-grade MCTs, the information gained from the prognostic panel may not change treatment recommendations. The authors feel this tool is best employed when the clinician has reason to suspect that an initial interpretation of low grade is incorrect.
Possible indications for MCT prognostic panel use include:
- Low-grade tumors in breeds predisposed to high-grade tumors (e.g., Chinese Shar-Pei; FIGURE 1)3
- Low-grade tumors with conflicting history (e.g., rapid growth, ulceration, edema, systemic illness consistent with degranulation)2
- Low-grade tumors in concerning anatomic locations (e.g., perioral, ear base, nail bed, other mucocutaneous sites, inguinal/perineal, preputial/scrotal)3-5
- Low-grade tumors with contradictory histopathologic features (e.g., borderline mitotic count [4 to 8/10 high-power fields], low mitotic count with concurrent marked multinucleation/nuclear atypia)6
Figure 1. A female Chinese Shar-Pei with diffuse mast cell disease 1 month after presenting for a solitary mast cell tumor.
Importance of Lymph Nodes
Detecting metastatic lymph nodes presents a challenge, as not all lymph nodes can be palpated or identified as abnormal via physical examination or imaging. For example, 50% of palpably normal lymph nodes were found to contain metastasis in a population consisting of 90% low-grade tumors.7 Furthermore, the regional (nearest) lymph node is not always the sentinel (draining) lymph node. A recent study using lymph node mapping found that regional and sentinel lymph nodes differed in nearly 60% of MCT cases.8
While the presence of lymph node involvement is used to stage MCTs (TABLE 1), MCT lymph node metastasis is not histologically classified as a “yes/no” binary. Instead, Weishaar’s histologic classification system assesses the number, distribution, and architectural disruption of mast cells within a lymph node (BOX 1).10 This classification describes the extent of nodal metastasis and is increasingly provided on biopsy reports. One study found a difference in survival between patients with HN0 + HN1 lymph nodes (60 months) and those with HN2 + HN3 lymph nodes (27 months).10 This study included all tumor grades. A more recent study that focused on grade II/low-grade MCTs with HN2 metastasis found that these patients experienced prolonged survival with surgery alone, with median survival time not reached (median follow-up time, 750 days), suggesting early lymph node metastasis (HN2) is insufficient to warrant systemic treatment.11
HN1: Premetastatic
HN2: Early metastasis
HN3: Overt metastasis
Histologic tumor grade is more predictive of patient outcome than lymph node status; however, a common thread has emerged over the past decade: Treatment of metastatic lymph nodes has a more significant impact on patient survival than the presence of lymph node metastasis.12-14 Treatment can include extirpation, chemotherapy, and/or radiation therapy.
Patients with lymph node involvement (stage II) that do not undergo extirpation have a 5-fold increased risk of tumor progression and a 4-fold increased risk of tumor-related death.13 Among patients with grade II (intermediate) MCTs, survival is unaffected by lymph node metastasis (stage I versus stage II), assuming the lymph node is removed. Extirpation permitted patients a median survival time of “not reached” (median follow-up time, 409 days) compared with a median survival time of 360 days in patients in which the metastatic lymph node was not removed (median follow-up time, 620 days) in 1 study.13
The value of treating affected lymph nodes is not exclusive to intermediate-grade MCTs. In dogs with high-grade MCTs, prophylactic irradiation of regional lymph nodes resulted in significantly increased progression-free survival compared with those that did not have lymph node extirpation or irradiation.14
Local MCT Therapy
Surgery
Surgery is considered the gold standard for MCT treatment and is often curative. While recommended surgical margins of 3 cm laterally and 1 fascial plane deep were previously applied to all MCTs, recent studies have suggested this may be more tissue than needed to achieve adequate local control of many MCTs.15-17 Since most MCTs are low grade and dogs experience low recurrence rates, even with incomplete histologic margins, a smaller surgical dose may be sufficient.17
Several updated recommendations have been proposed. Most prominent is the use of proportional margins. In this approach, a 0.5-cm mass would be removed with 0.5-cm lateral margins, a 1-cm mass with 1-cm margins, and so on up to 2 cm. The recommendation of removing 1 fascial plane deep remains. With this approach, 95% of MCTs were completely excised (all margins histologically clear of mast cell infiltration), regardless of grade, with an overall recurrence rate of 3% in 1 study.17 Despite the active components of mast cell granules, these patients do not experience increased incision site complications compared to those with soft tissue sarcomas.18
With recurrence rates for incompletely excised, low-grade tumors at 10% to 30%, several follow-up options exist.15,17,19 These may include active surveillance for tumor regrowth, scar revision, or radiation therapy. Scar revision can improve long-term survival (from 24 to 98 months) and decrease recurrence rates (from 38% to 13%).20
Tumor grade and stage may alter the decision to proceed with surgery and associated surgical dose. Cases with negative prognostic factors (see Use of the MCT Prognostic Panel) should undergo complete staging preoperatively. This includes cytologic evaluation of regional (ideally sentinel) lymph nodes and the spleen (plus or minus the liver).8,21 Since ultrasonography has poor sensitivity and specificity in identifying MCT metastasis in the liver and spleen, percutaneous fine-needle aspiration with cytologic evaluation is recommended regardless of the ultrasonographic appearance of these organs. While uncommon, viscerally disseminated disease has a considerable impact on expected survival and may alter the decision to proceed with surgery.21
Radiation Therapy
Radiation therapy has been used in the adjuvant setting following incomplete resection or in the gross tumor setting for nonresectable tumors.14,20 The long-term benefits of radiation therapy for high-grade tumors remain unclear, although radiation therapy is an effective means for reducing the recurrence of low-grade MCTs. A recent study of predominantly low-grade MCTs treated with radiation therapy following incomplete excision demonstrated a recurrence rate of less than 7% and tumor-specific survival time that was not reached.22
Acute toxicity (adverse effects seen during radiation therapy or within 2 weeks of completion) can be seen in MCT patients with gross or microscopic disease. These effects include erythema, desquamation, or other adverse events outlined in the modified Veterinary Radiation Therapy Oncology Group scoring criteria.22,23 Concurrent use of supportive medications, particularly prednisone, histamine H1 and H2 antagonists, or proton pump inhibitors, can help decrease the severity of adverse events.22
Intralesional Therapy
Triamcinolone
Triamcinolone is a long-acting steroid that can be administered intralesionally to decrease or maintain MCT size. In 1 study of 23 dogs, the overall response rate (complete and partial responses) was 67%, including patients receiving concurrent therapies. For patients treated with triamcinolone with or without oral corticosteroids, the median time to progression was 28 days and 114 days, respectively. The most common adverse effects were gastrointestinal (GI) upset and bleeding at the injection site, which was suspected to be related to degranulation. This therapy is economical and does not require special equipment or training, although the duration of response appears limited.24
Tigilanol Tiglate Injection
Tigilanol tiglate injection (Stelfonta; Virbac, stelfonta.com) is an intralesional treatment labeled for canine cutaneous and subcutaneous MCTs that received full U.S. Food and Drug Administration approval in 2020. Tigilanol tiglate injection uses a multimodal mechanism of action, including causing tumor cell oncolysis, activating an acute inflammatory response, and increasing tumor vascular permeability. Ultimately, tigilanol tiglate injection results in tumor necrosis and tissue sloughing, resulting in wound formation at the tumor site (FIGURE 2).25
Corticosteroids and H1 and H2 antagonists are initiated in advance and at the start of treatment. Tigilanol tiglate injection should be handled using personal protective equipment. Dose calculation is based on tumor volume, which is labeled for tumors less than 10 cm3. The dose is labeled not to exceed 0.25 mL/kg or 5 mL/dog. Using a 23-g needle, a single insertion site is made at the base of the tumor and the drug is administered in a fanning motion to maximize drug delivery. It is recommended to sedate patients to improve drug delivery and decrease the risk of accidental needlesticks to the administrator.25 The authors use Kevlar gloves to increase administrator protection.
Proper case selection and client education are crucial. Tigilanol tiglate injection is labeled for use in nonmetastatic MCTs. Cutaneous MCTs can be located anywhere, but subcutaneous MCTs must be distal to the elbow or hock to reduce peritumoral tissue damage. Clients should be prepared for wound formation and associated healing times. Tumor size is often similar to wound size, which correlates with healing time. While most patients experience expected wounds, rare instances (2%) experience extensive wound formation. Wound formation peaks at 7 days and typically heals by weeks 4 through 6. Unless purulent material is noted or a patient is traumatizing the area, wound care, bandaging, antibiotics, and Elizabethan collars are unnecessary. Local pain is usually well controlled with analgesics for about 1 week in 63% of patients.25
Tigilanol tiglate injection is an exciting new tool, as most responders have significantly smaller wounds than the 2- or 3-cm margins that were previously recommended with surgery. Approximately 75% of patients (predominantly with cytologically low-grade tumors) had a complete response after 1 treatment. This increases to nearly 90% with inclusion of dogs that received a second treatment. Cytologically high-grade tumors have a lower complete response rate: 44% after 1 treatment and 67% after 2 treatments.26,27 Of patients with cytologically high-grade tumors that respond to tigilanol tiglate injection, approximately 70% remain disease-free at 1 year.25 It is important to note that cytologic grading is not widely accepted, and a limitation of tigilanol tiglate injection treatment is the inability to obtain a histopathologic grade to better estimate prognosis.26
Systemic MCT Therapy
Chemotherapy
Chemotherapy can be used as a neoadjuvant, adjuvant, or sole therapy for MCTs. In the neoadjuvant setting, chemotherapy and/or steroids can be used to reduce tumor size and facilitate surgery (also referred to as downstaging). Care should be taken when interpreting margins after neoadjuvant therapy since this approach is used to allow for marginal excision in challenging anatomic locations.28 No studies have evaluated the effects of neoadjuvant chemotherapy on histologic grade or proliferation indices; however, neoadjuvant steroid therapy is unlikely to alter these values.29
As an adjuvant treatment, chemotherapy has demonstrated substantial benefit in delaying metastasis in patients with high-grade MCTs.30 Without systemic treatment, most dogs succumb to progressive or metastatic disease within 1 year. Chemotherapy can improve this time to more than 2 to 3 years.4 Fortunately, adjuvant chemotherapy is unnecessary for most patients with low-grade MCTs based on low metastatic and recurrence rates.13
The most common chemotherapy drugs used for MCTs are vinblastine and lomustine (CCNU [cyclohexylchloroethylnitrosourea]). Chemotherapy can be used in single-agent or multiagent protocols or in combination with prednisone.4,30-32
Toceranib Phosphate and Other Tyrosine Kinase Inhibitors
Toceranib phosphate (Palladia; Zoetis, zoetisus.com) and other tyrosine kinase inhibitors (TKIs), such as masitinib mesylate (Masivet; AB Science, masivet.co.uk) and imatinib mesylate (Gleevec; Novartis, gleevec.com), have been used in MCT treatment.33-35 Responsiveness to TKIs is independent of c-kit mutation status. However, higher response rates occur in patients with c-kit mutations. Ultimately, c-kit mutation status should not limit treatment with TKIs if clinically warranted.36
TKIs can be combined with radiation therapy and/or traditional chemotherapy in gross or microscopic disease settings, although this may increase adverse events. Toceranib phosphate is currently the only TKI labeled for canine MCT treatment in the United States. Imatinib mesylate is predominantly used to treat chronic myelogenous leukemia in humans and may be more affordable than toceranib phosphate for large dogs. Overall response rates for TKIs range from 30% to 60% with variable progression-free intervals.33-35
Patients receiving TKIs should be monitored closely for adverse effects and disease progression. The most common adverse events include GI upset, anorexia, and weight loss.33-35 Additional side effects are unique to each TKI. Serious adverse events seen with toceranib phosphate may also include increased liver enzymes, proteinuria, hypertension, and GI bleeding.33,36,37 Due to these risks, the authors recommend consultation with a veterinary oncologist before prescribing TKIs.
Checkpoint Inhibitors
Gilvetmab (Merck, merck-animal-health-usa.com) is a checkpoint inhibitor that received U.S. Department of Agriculture conditional approval for use in canine MCTs (stage I, II, or III) and melanoma (greater than 2 cm with or without lymph node involvement) in October 2023. Numerous canine tumors express programmed cell death ligand 1, which binds to programmed cell death 1 (PD-1) on tumor-infiltrating lymphocytes to evade destruction.38 Gilvetmab is a caninized PD-1 monoclonal antibody that binds PD-1 on lymphocytes, thus enabling T lymphocytes to recognize tumor cells and promote cell killing.39
Gilvetmab is given as an intravenous infusion repeated every 2 weeks for up to 10 treatments. It has an overall response rate of 46% in canine patients with MCTs. As with many immunotherapies, patients should be monitored for pseudoprogression, a phenomenon whereby tumor size increases due to resurgent inflammation. This local growth can be misinterpreted as treatment failure and may result in premature treatment discontinuation. Pseudoprogression can postpone tumor shrinkage by 8 to 12 weeks.39
The manufacturer recommends pretreatment with diphenhydramine and reports the most common adverse effects as lethargy, hyporexia, and increased alkaline phosphatase. Approximately 11% of patients with MCTs have severe to life-threatening adverse events. The most concerning events include anaphylaxis, hypotension, and tumor hemorrhage. As gilvetmab is an immunotherapeutic treatment, it is not recommended for patients receiving chronic glucocorticoids or other immunosuppressive drugs. Clinical trials are ongoing.39
Corticosteroids
Prednisone and other steroids have been shown to have inhibitory effects on mast cell viability and proliferation.40 They can also decrease clinical signs associated with degranulation, such as edema, pruritus, and pain. Like chemotherapy, steroids can be used as a neoadjuvant, adjuvant, or sole therapy. While single-agent corticosteroids are economical and accessible, response is often short-lived at a few weeks. Anecdotally, neoadjuvant steroids achieve maximal response within 7 to 14 days; therefore, surgical excision should be scheduled during this timeframe.29
Supportive Care
Dogs with MCTs are at risk for degranulation and increased plasma histamine concentrations, which lead to local and systemic effects such as changes in tumor size, wheal formation, GI upset/ulceration, anaphylaxis, and even death. As such, many veterinarians utilize over-the-counter histamine antagonist drugs to decrease the signs associated with mast cell degranulation and hyperhistaminemia.41 While H1 antagonists (e.g., diphenhydramine) can limit cutaneous inflammation, H2 antagonists (e.g., famotidine) are more important as about 30% of dogs with MCTs have evidence of GI ulceration at necropsy.41 The authors rely on H2 antagonists for most cases, although for dogs with high disease burden (e.g., large, weeping tumors; disseminated metastasis), the authors switch to proton pump inhibitors (e.g., omeprazole) for more targeted blockade and long-term efficacy.42 Once microscopic disease is achieved, histamine antagonists and/or proton pump inhibitors can be discontinued.
Summary
The diagnostic and therapeutic approach to canine MCTs has undergone several recent advances. The MCT prognostic panel is useful in guiding treatment recommendations, particularly in patients with histologically low-grade tumors with conflicting information. There is no singular test that can perfectly predict the behavior of a tumor; rather, results should always be interpreted in the context of clinical and diagnostic findings. Furthermore, the removal of metastatic lymph nodes significantly improves survival, even if systemic treatment is not warranted. MCTs can be managed with local and/or systemic treatment. With the broad range of MCT behavior and as treatment options evolve, veterinarians should reach out to veterinary oncologists as needed for support.
References
1. Thompson JJ, Yager JA, Best SJ, et al. Canine subcutaneous mast cell tumors: cellular proliferation and KIT expression as prognostic indices. Vet Pathol. 2011;48(1):169-181. doi:10.1177/0300985810390716
2. dos Santos Costa Poggiani S, Maria Terra E, Torres Neto R, Tinucci Costa M, Laufer Amorim R. Canine cutaneous mast cell tumor: biologic behavior and its correlation with prognostic indicators. Open J Vet Med. 2012;2(4):255-261. doi:10.4236/ojvm.2012.24041
3. Reynolds BD, Thomson MJ, O’Connell K, Morgan EJ, Gummow B. Patient and tumour factors influencing canine mast cell tumour histological grade and mitotic index. Vet Comp Oncol. 2019;17(3):338-344. doi:10.1111/vco.12477
4. Thamm DH, Turek MM, Vail DM. Outcome and prognostic factors following adjuvant prednisone/vinblastine chemotherapy for high-risk canine mast cell tumour: 61 cases. J Vet Med Sci. 2006;68(6):581-587. doi:10.1292/jvms.68.581
5. Chalfon C, Finotello R, Sabattini S, et al. Patterns of nodal metastases, biological behaviour and prognosis of canine mast cell tumours of the pinna: a multi-institutional retrospective study. Vet Comp Oncol. 2023;21(2):332-338. doi:10.1111/vco.12893
6. Kiupel M, Webster JD, Bailey KL, et al. Proposal of a 2-tier histologic grading system for canine cutaneous mast cell tumors to more accurately predict biological behavior. Vet Pathol. 2011;48(1):147-155. doi:10.1177/0300985810386469
7. Ferrari R, Marconato L, Buracco P, et al. The impact of extirpation of non-palpable/normal-sized regional lymph nodes on staging of canine cutaneous mast cell tumours: a multicentric retrospective study. Vet Comp Oncol. 2018;16(4):505-510. doi:10.1111/vco.12408
8. Annoni M, Borgonovo S, Aralla M. Sentinel lymph node mapping in canine mast cell tumours using a preoperative radiographic indirect lymphography: technique description and results in 138 cases. Vet Comp Oncol. 2023;21(3):469-481. doi:10.1111/vco.12906
9. Owens LN. Skin: mastocytoma. In: Owens LN, ed. TNM Classification of Tumors in Domestic Animals. Geneva: World Health Organization; 1980:15.
10. Weishaar KM, Thamm DH, Worley DR, Kamstock DA. Correlation of nodal mast cells with clinical outcome in dogs with mast cell tumour and a proposed classification system for the evaluation of node metastasis. J Comp Pathol. 2014;151(4):329-338. doi:10.1016/j.jcpa.2014.07.004
11. Cino M, Gariboldi EM, Stefanello D, et al. Ki67 Index in Patnaik grade 2/Kiupel low-grade canine cutaneous mast cell tumors with early lymph node metastasis: a descriptive study. Vet Sci. 2023;10(7):436. doi:10.3390/vetsci10070436
12. Guerra D, Faroni E, Sabattini S, et al. Histologic grade has a higher-weighted value than nodal status as predictor of outcome in dogs with cutaneous mast cell tumours and overtly metastatic sentinel lymph nodes. Vet Comp Oncol. 2022;20(3):551-558. doi:10.1111/vco.12806
13. Marconato L, Polton G, Stefanello D, et al. Therapeutic impact of regional lymphadenectomy in canine stage II cutaneous mast cell tumours. Vet Comp Oncol. 2018;16(4):580-589. doi:10.1111/vco.12425
14. Mendez SE, Drobatz KJ, Duda LE, White P, Kubicek L, Sorenmo KU. Treating the locoregional lymph nodes with radiation and/or surgery significantly improves outcome in dogs with high-grade mast cell tumours. Vet Comp Oncol. 2020;18(2):239-246. doi:10.1111/vco.12541
15. Donnelly L, Mullin C, Balko J, et al. Evaluation of histological grade and histologically tumour-free margins as predictors of local recurrence in completely excised canine mast cell tumours. Vet Comp Oncol. 2015;13(1):70-76. doi:10.1111/vco.12021
16. Simpson AM, Ludwig LL, Newman SJ, Bergman PJ, Hottinger HA, Patnaik AK. Evaluation of surgical margins required for complete excision of cutaneous mast cell tumors in dogs. JAVMA. 2004;224(2):236-240. doi:10.2460/javma.2004.224.236
17. Saunders H, Thomson MJ, O’Connell K, Bridges JP, Chau L. Evaluation of a modified proportional margin approach for complete surgical excision of canine cutaneous mast cell tumours and its association with clinical outcome. Vet Comp Oncol. 2021;19(4):604-615. doi:10.1111/vco.12630
18. Iodence AE, Wallace ML, Grimes JA, Schmiedt CW. Dogs undergoing surgical excision of mast cell tumors are not at increased risk of incisional complications. JAVMA 2022;260(S1):S88-S95. doi:10.2460/javma.20.09.0488
19. Karbe GT, Davis E, Runge JJ, Brown DC, Holt DE. Evaluation of scar revision after inadequate primary excision of cutaneous mast cell tumors in 85 dogs (2000–2013). Vet Surg. 2021;50(4):807-815. doi:10.1111/vsu.13619
20. Kry KL, Boston SE. Additional local therapy with primary re-excision or radiation therapy improves survival and local control after incomplete or close surgical excision of mast cell tumors in dogs. Vet Surg. 2014;43(2):182-189. doi:10.1111/j.1532-950X.2014.12099.x
21. Pecceu E, Serra Varela JC, Handel I, Piccinelli C, Milne E, Lawrence J. Ultrasound is a poor predictor of early or overt liver or spleen metastasis in dogs with high-risk mast cell tumours. Vet Comp Oncol. 2020;18(3):389-401. doi:10.1111/vco.12563
22. Mason SL, Pittaway C, Gil BP, et al. Outcomes of adjunctive radiation therapy for the treatment of mast cell tumors in dogs and assessment of toxicity: a multicenter observational study of 300 dogs. J Vet Intern Med. 2021;35(6):2853-2864. doi:10.1111/jvim.16264
23. Poirier VJ, Keyerleber M, Gordon IK, et al. ACVR and ECVDI consensus statement: reporting elements for toxicity criteria of the veterinary radiation therapy oncology group v2.0. Vet Radiol Ultrasound. 2023;64(5):789-797. doi:10.1111/vru.13291
24. Case A, Burgess K. Safety and efficacy of intralesional triamcinolone administration for treatment of mast cell tumors in dogs: 23 cases (2005–2011). JAVMA. 2018;252(1):84-91. doi:10.2460/javma.252.1.84
25. Stelfonta (tigilanol tiglate injection) 1 mg/mL client information sheet. Virbac. October 22, 2020. Accessed March 4, 2024. https://vet-us.virbac.com/files/live/sites/virbac-b2b-usa/files/client%20leaflet/Products/Stelfonta/Stelfonta%20Insert_23OCT2020%20(1).PDF
26. Brown GK, Campbell JE, Jones PD, De Ridder TR, Reddell P, Johannes CM. Intratumoural treatment of 18 cytologically diagnosed canine high-grade mast cell tumours with tigilanol tiglate. Front Vet Sci. 2021;8:675804. doi:10.3389/fvets.2021.675804
27. Jones PD, Campbell JE, Brown G, Johannes CM, Reddell P. Recurrence-free interval 12 months after local treatment of mast cell tumors in dogs using intratumoral injection of tigilanol tiglate. J Vet Intern Med. 2021;35(1):451-455. doi:10.1111/jvim.16018
28. Ossowska M, Picornell JA, Finotello R, Amores-Fuster I, Tanis JB. Pre-operative neoadjuvant vinblastine-prednisolone in canine mast cell tumours: a single-centre retrospective cohort study. Vet Comp Oncol. 2023;21(3):447-459. doi:10.1111/vco.12904
29. Klahn S, Dervisis N, Lahmers K, Benitez M. Evaluation of tumor grade and proliferation indices before and after short-course anti-inflammatory prednisone therapy in canine cutaneous mast cell tumors: a pilot study. Vet Sci. 2022;9(6):277. doi:10.3390/vetsci9060277
30. Hayes A, Adams V, Smith K, Maglennon G, Murphy S. Vinblastine and prednisolone chemotherapy for surgically excised grade III canine cutaneous mast cell tumours. Vet Comp Oncol. 2007;5(3):168-176. doi:10.1111/j.1476-5829.2007.00135.x
31. Hay JK, Larson VS. Lomustine (CCNU) and prednisone chemotherapy for high-grade completely excised canine mast cell tumors. Can Vet J. 2019;60(12):1326-1330.
32. Todd JE, Nguyen SM, White J, Langova V, Thomas PM, Tzannis S. Combination vinblastine and palladia for high-grade and metastatic mast cell tumors in dogs. Can Vet J. 2021;62(12):1335-1340.
33. London CA, Malpas PB, Wood-Follis SL, et al. Multi-center, placebo-controlled, double-blind, randomized study of oral toceranib phosphate (SU11654), a receptor tyrosine kinase inhibitor, for the treatment of dogs with recurrent (either local or distant) mast cell tumor following surgical excision. Clin Cancer Res. 2009;15(11):3856-3865. doi:10.1158/1078-0432.CCR-08-1860
34. Smrkovski OA, Essick L, Rohrbach BW, Legendre AM. Masitinib mesylate for metastatic and non-resectable canine cutaneous mast cell tumours. Vet Comp Oncol. 2015;13(3):314-321. doi:10.1111/vco.12053
35. Macedo TR, de Queiroz GF, Casagrande TAC, et al. Imatinib mesylate for the treatment of canine mast cell tumors: assessment of the response and adverse events in comparison with the conventional therapy with vinblastine and prednisone. Cells. 2022;11(3):571. doi:10.3390/cells11030571
36. Weishaar KM, Ehrhart EJ, Avery AC, et al. c-Kit mutation and localization status as response predictors in mast cell tumors in dogs treated with prednisone and toceranib or vinblastine. J Vet Intern Med. 2018;32(1):394-405. doi:10.1111/jvim.14889
37. Tjostheim SS, Stepien Rl, Markovic LE, Stein TJ. Effects of toceranib phosphate on systolic blood pressure and proteinuria in dogs. J Vet Intern Med. 2016;30(4):951-957. doi:10.1111/jvim.13951
38. Muscatello LV, Gobbo F, Avallone G, et al. PDL1 immunohistochemistry in canine neoplasms: validation of commercial antibodies, standardization of evaluation, and scoring systems. Vet Pathol. 2023:3009858231209410. doi:10.1177/03009858231209410
39. Gilvetmab (Merck Animal Health). In: Compendium of Veterinary Products. Intervet/Merck Animal Health; 2023. Accessed November 12, 2023. Available at: https://merckusa.cvpservice.com/product/basic/view/1047586
40. Matsuda A, Tanaka A, Amagai Y, et al. Glucocorticoid sensitivity depends on expression levels of glucocorticoid receptors in canine neoplastic mast cells. Vet Immunol Immunopathol. 2011;144(3-4):321-328. doi:10.1016/j.vetimm.2011.08.013
41. Fox LE, Rosenthal RC, Twedt DC, Dubielzig RR, MacEwen EG, Grauer GF. Plasma histamine and gastrin concentrations in 17 dogs with mast cell tumors. J Vet Intern Med. 1990;4(5):242-246. doi:10.1111/j.1939-1676.1990.tb03116.x
42. Tolbert MK, Graham A, Odunayo A, et al. Repeated famotidine administration results in a diminished effect on intragastric pH in dogs. J Vet Intern Med. 2017;31(1):117-123. doi:10.1111/jvim.14615