Editorial Type: RETROSPECTIVE STUDIES
 | 
Online Publication Date: 01 Jan 2024

Retrospective Evaluation of Melphalan, Vincristine, and Cytarabine Chemotherapy for the Treatment of Relapsed Canine Lymphoma

DVM, DACVIM (Oncology),
DVM, MS, DACVIM (Oncology),
DVM, MS, DACVIM (Oncology), and
DVM, DACVIM (Oncology)
Article Category: Research Article
Page Range: 7 – 14
DOI: 10.5326/JAAHA-MS-7372
Save
Download PDF

ABSTRACT

Dogs diagnosed with multicentric lymphoma often relapse following induction therapy within the first year of treatment. The primary aim of this study was to evaluate the tolerability of a novel drug combination using melphalan, vincristine, and cytarabine (MOC) for the treatment of relapsed lymphoma. On day 1, dogs were treated with vincristine (0.5–0.6 mg/m2 IV) and cytarabine (300 mg/m2 IV over 4–6 hr or subcutaneously over 2 days). On day 7, dogs were treated with melphalan (20 mg/m2per os). This 2 wk protocol was repeated for at least three cycles or until treatment failure. Twenty-six dogs were treated with MOC and met the inclusion criteria. Twenty-three dogs had toxicity data, and all experienced adverse events with the majority graded as mild. The overall response rate was 38%, which included 19% of dogs who achieved a complete response. The median progression-free survival was 29 days (range 1–280 days). The overall clinical benefit was 65% for a median of 37 days (range 33–280 days). MOC is a safe treatment option for relapsed lymphoma in dogs.

Introduction

Canine high-grade lymphoma initially responds well to multiagent chemotherapy, with 80–90% of dogs experiencing a complete response.1,2 These responses are unfortunately short-lived, and relapse often occurs within the first year. Although second remissions may be attained with rescue chemotherapy, the response rates and durations are generally inferior compared with first-line therapy. Numerous rescue protocols have been investigated previously with reported response rates ranging from 7 to 74% and response durations ranging from 1 to 5 mo.318 Chemotherapy-resistant lymphoma remains clinically challenging to treat, and thus, continued investigation into novel protocols is warranted.

Combination chemotherapy, especially for the treatment of lymphoma, may be considered advantageous compared with single-agent protocols.1921 This is likely due to combination protocols using multiple mechanisms to target cancer cells while slowing the development of cellular drug resistance. Ideal multiagent protocols combine drugs with known single-agent efficacy and minimal overlapping toxicities.22

Vincristine and melphalan have demonstrated efficacy both as single agents and as part of combination protocols for the treatment of canine lymphoma.27,23 Although cytarabine has not been found to be efficacious as a single agent for the treatment of canine lymphoma, it has been reported as efficacious as part of combination protocols.2,3,6,10,24,25 Additionally, a prospective study of dogs with treatment-naive stage V lymphoma reported the addition of cytarabine into a vincristine, cyclophosphamide, doxorubicin, and prednisone (CHOP)–based protocol generated significantly higher response rates and longer survivals.26

Furthermore, these chemotherapeutics have disparate mechanisms of action. Vincristine is an antimicrotubule agent that binds tubulin, inhibiting microtubule assembly resulting in metaphase arrest and cytotoxicity.27 Melphalan is a cell-cycle nonspecific, bifunctional alkylating agent within the nitrogen mustard subclass.28 Cytarabine is an antimetabolite that acts as an analog of deoxycytidine. Intracellular phosphorylation generates arabinosylcytosine triphosphate, which competitively inhibits DNA polymerase α.29 Incorporation of cytarabine into DNA leads to cytotoxicity during the S phase of the cell cycle.30 The dose-limiting toxicity for cytarabine and melphalan is thrombocytopenia, whereas the dose-limiting toxicity of vincristine is neutropenia or gastrointestinal toxicity.3,6,7,23,24 Chemotherapeutics frequently have overlapping toxicities, and when used in combination, the dosing schedule may be altered to minimize additive effects.31

The primary aim of this study was to retrospectively evaluate the adverse event (AE) profile of combination chemotherapy with melphalan, vincristine, and cytarabine (MOC) for the treatment of relapsed canine multicentric lymphoma. A secondary aim was to evaluate the efficacy of this novel drug combination.

Materials and Methods

Data were obtained retrospectively from dogs with relapsed or refractory multicentric lymphoma intending to treat with MOC at Oregon State University from March 2013 and March 2020. Informed consent was obtained from all owners, and all dogs were clinically managed according to contemporary standards of care. Dogs were included if they had a cytologic or histologic diagnosis of high-grade or large cell lymphoma by a board-certified veterinary pathologist, had previously been treated with chemotherapy for lymphoma, and had measurable disease at the time of treatment initiation. Data collected included signalment, clinical stage at diagnosis, substage at diagnosis, immunophenotype, previous chemotherapy treatment protocols, and response, date of relapse(s), method of determining relapse, total cycles of MOC received, adverse events, date of progression after MOC, additional treatment protocols and response, date of death, and cause of death.

Dogs who lacked complete disease staging (thoracic radiographs, abdominal ultrasound, cytology of other organs/bone marrow) were included in the study. When performed, stage and substage were assigned at initial diagnosis and relapse according to the modified World Health Organization classification for canine lymphoma.32 Similarly, although immunophenotype was not required for inclusion in this study, when performed via immunocytochemistry, flow cytometry, or polymerase chain reaction for antigen receptor rearrangement, results were recorded.

Treatment Protocol

The dosages used in the treatment protocol were determined based on previously published multiagent chemotherapy protocols for the treatment of canine lymphoma.2,3,6,7,10,24 The treatment protocol is summarized in Table 1. Briefly, dogs were treated with vincristine and cytarabine on day 1. Cytarabine was administered either as a continuous IV infusion over 4–6 hr immediately following IV vincristine or subcutaneously over 2 days. On day 7, dogs were treated with melphalan. Treatment order could be altered depending on clinician or owner preference, e.g., melphalan administered during the first week and vincristine and cytarabine administered during week 2. Dogs were variably prescribed a steroid (prednisone or dexamethasone) throughout the treatment protocol depending on clinician preference. The cycle was repeated for at least three cycles or until treatment failure was documented. Antiemetics and antidiarrheals were prescribed prophylactically or symptomatically depending on clinician preference.

TABLE 1 Chemotherapy Drug Schedule for Melphalan, Vincristine, and Cytarabine Protocol
TABLE 1

Response to the therapy was evaluated through physical examinations weekly or biweekly depending on owner and clinician preference. Dogs with inadequate follow-up were excluded from the response assessment. Adequate follow-up was defined as a physical examination at least 1 day after treatment. Response was determined using the Veterinary Cooperative Oncology Group (VCOG) response evaluation criteria for lymphoma.33 A complete response (CR) was defined as resolution of all measurable peripheral lymphadenopathy. A partial response (PR) was defined as ≥30% reduction in the sum of the longest diameters of measurable peripheral lymph nodes. Stable disease (SD) was defined as <30% reduction or <20% increase in sum of the longest diameters of measurable peripheral lymph nodes. Progressive disease (PD) was defined as >20% increase in the sum of the longest diameter of measurable peripheral lymph nodes. Clinical benefit was defined as CR, PR, or SD. All responses, including SD, had to be maintained for ≥14 days to be classified as such. Evidence of gastrointestinal and other toxicities was assessed by evaluation of the medical records, including owner-reported history and physical examination findings. Hematological toxicities were evaluated every 7–10 days, and physical examinations were performed weekly or biweekly depending on owner preference. Dogs with inadequate follow-up were excluded from toxicity assessment. A complete blood count 1 wk after treatment and a physical examination 1–2 wk after treatment was defined as adequate follow-up to assess toxicity. Adverse events were graded at the time of patient assessment and retrospectively according to the VCOG Common Terminology Criteria for Adverse Events v2.34 Dose adjustments and treatment delays were permitted based on owner and clinician discretion, commonly if VCOG grade >2 toxicity occurred. If dogs had pre-existing cytopenias, hematological toxicity was attributed to MOC and included in analysis only if the grade was further increased from pretreatment levels.

Statistics

Continuous data were expressed as medians and ranges, and categorical data were expressed as frequencies and percentages. The primary endpoint was progression-free survival (PFS), which was defined as the date of MOC treatment initiation to the date of PD or date of death from any cause. If PD was not documented by a veterinarian, dogs were censored from PFS analysis from the date of the last physical examination. Data were tested for normality using Pearson’s correlation coefficient. Nonparametric data were compared using a Wilcoxon rank-sum test. Overall survival was not evaluated due to the variable pursuit of other rescue protocols. The Kaplan-Meier method was used to estimate and display the distribution of PFS. Differences between groups were compared using log-rank analysis. Variables with values of P ≤ .05 were considered significant. All statistical analyses were performed using standard statistical softwarea.

Results

Case Demographics

Twenty-six dogs met the inclusion criteria and were treated with MOC at Oregon State University between March 2013 and March 2020. Three dogs were excluded from the toxicity analysis due to inadequate follow-up (Figure 1). Two dogs were excluded from PFS analysis. Both dogs had experienced a clinical benefit (PR and SD) after one cycle but were switched to a different protocol because of gastrointestinal AEs and owner preference. Figure 1 outlines selection criteria and Table 2 details the patient population, which consisted mostly of large-breed, neutered dogs.

FIGURE 1FIGURE 1FIGURE 1
FIGURE 1 Flow chart provides details of patient screening and inclusion and exclusion from study. AE, adverse event; GI, gastrointestinal; MOC, melphalan, vincristine, and cytarabine; PFS, progression-free survival.

Citation: Journal of the American Animal Hospital Association 60, 1; 10.5326/JAAHA-MS-7372

TABLE 2 Characteristics of 26 Dogs Treated with Melphalan, Vincristine, and Cytarabine
TABLE 2

All dogs were initially diagnosed with large cell lymphoma through cytology. Histopathology was performed in 1 dog and confirmed high-grade lymphoma. Immunophenotype was determined via immunocytochemistry in 15 dogs and flow cytometry in 6 dogs. Immunogenotype was determined via polymerase chain reaction for antigen receptor rearrangement Pin 5 dogs. Three dogs had multiple diagnostics performed to determine phenotype; however, immunophenotype was unknown in 5 dogs. The majority (n = 16, 62%) of dogs were not fully staged and determined to be at least stage III based on generalized lymphadenopathy appreciated on physical examination. Abdominal ultrasound was performed on 4 dogs and thoracic radiographs were performed on 5 dogs. Three dogs were determined to be stage IV based on generalized lymphadenopathy and cytologic evidence of hepatic or splenic involvement. Eight dogs (31%) were determined to be stage V based on the presence of lymphocytosis and circulating atypical lymphocytes. The majority of dogs (n = 16, 62%) were substage b at the time of MOC treatment initiation.

Previous Treatment

Twenty-one dogs (81%) were initially treated with the 19 wk CHOP protocol. Two dogs (8%) were treated with single-agent doxorubicin, two dogs (8%) were treated with lomustine, and one dog (4%) was treated with vincristine, cyclophosphamide, and prednisone. Sixteen dogs (62%) achieved a CR, eight dogs (31%) achieved a PR, one dog (4%) achieved SD, and one dog (4%) achieved PD as their best response to initial therapy. The median duration of initial best response was 42 days (range 0–454 days). Cytology confirmed first relapse in 14 dogs (54%), and PD was noted via lymph node measurements in the remaining dogs. The median duration between initial diagnosis and initiation of the MOC protocol was 152 days (range 34–764 days). Two dogs (8%) were treated with MOC as first-line rescue therapy. Twelve (46%) dogs were treated with lomustine as first-line rescue treatment either with (n = 1) or without (n = 11) L-asparaginase. Various other rescue protocols (CHOP-based, single-agent doxorubicin, mitoxantrone, rabacfosadine, L-asparaginase) were used before MOC therapy. Two dogs received L-asparaginase within 1 wk of MOC. The median number of chemotherapy protocols prior to MOC was 2 (range 1–8). Twenty-four dogs (92%) had been treated with vincristine as part of first-line or rescue therapy before MOC initiation.

MOC Protocol

Eleven dogs (42%) were not treated with a steroid concurrently, whereas 8 (31%) were treated with prednisone (0.8–2 mg/kg per os [PO] q 24 hr), and 7 (27%) were treated with dexamethasone (0.10–0.17 mg/kg PO q 24 hr) concurrently depending on clinician preference. All dogs were prophylactically treated with maropitant (1 mg/kg IV) at the time of chemotherapy and for 4 days (2 mg/kg PO q 24 hr to nearest tablet size) following chemotherapy. Sixteen dogs were treated with multiple antiemetics concurrently (n = 13 dogs treated with dolasetron [0.6 mg/kg PO q 24 hr to nearest tablet size] and n = 13 dogs treated with ondansetron [0.5–1 mg/kg PO q 12 hr to nearest tablet size]). Four dogs were treated with an antidiarrhea (metronidazole 10 mg/kg PO q 12 hr to nearest tablet size). The median initial dose of vincristine during the first cycle of MOC was 0.5 mg/m2 (range 0.5–0.6 mg/m2). The dose of vincristine was escalated in 8 of 13 dogs treated with a second cycle of MOC. Three dogs were given cytarabine subcutaneously and the order of treatment was switched in three dogs (melphalan given week 1). The median number of MOC cycles received was 2 (range 0.5–3). Four dogs (15%) completed the planned three cycles. At the time of disease progression and before starting the MOC protocol, nine dogs (35%) were anemic (seven grade 1 and two grade 2) and nine dogs (35%) were thrombocytopenic (five grade 1 and four grade 2).

Toxicity

Twenty-three dogs had adequate follow-up to be included in the toxicity analysis. All 23 dogs experienced AEs, the majority of which were mild and did not require hospitalization. There were no VCOG Common Terminology Criteria for Adverse Events grade 4 or 5 AEs. There was a total of 98 AEs, and 13/23 dogs (56%) experienced both gastrointestinal and hematological AE. AEs are summarized in Table 3. The most common AEs were anemia, neutropenia, and diarrhea. Six dogs experienced multiple instances of anemia, accounting for 70% of anemia AEs. Only one of these dogs was anemic (grade 2) at the start of the protocol. This dog was treated with a blood transfusion during week 2 of treatment and completed a second cycle of MOC before developing PD. The same dog experienced a seizure (VCOG grade 2) 3 days after subcutaneous administration of cytarabine. The dog had no prior history of seizures, and antiepileptic therapy was not initiated. The dog subsequently tolerated an additional dose of cytarabine without experiencing seizures. None of the other dogs required a blood transfusion. Of the seven dogs that were anemic at the time of MOC initiation and had adequate follow-up for toxicity analysis, anemia progressed following MOC treatment in three dogs, remained stable in two dogs, and improved in two dogs.

TABLE 3 Adverse Events and Associated Veterinary Cooperative Oncology Group Grades Observed in 23 Dogs Treated with Melphalan, Vincristine, and Cytarabine
TABLE 3

There were seven dose delays and two dose reductions due to hematologic toxicities. Four dogs experienced multiple instances of neutropenia, accounting for 62% of the neutropenic events. Similarly, four dogs experienced multiple instances of diarrhea, accounting for 47% of diarrhea AEs. In two dogs, MOC was discontinued due to AEs and owner-perceived reduced quality of life. One dog experienced lethargy (grade 1) and diarrhea (grade 3) despite treatment with metronidazole. The other dog experienced anorexia (grade 2) despite treatment with ondansetron and maropitant, leading to discontinuation of the protocol. An appetite stimulant was not prescribed.

Patient Outcomes

The overall response rate for 26 evaluable dogs was 38%, which included 19% CR (n = 5 dogs) and 19% PR (n = 5 dogs). The overall clinical benefit (CR, PR, and SD) was 65%, which included 7 dogs with SD (27%). Two dogs with stage Vb T-cell lymphoma died less than three days after treatment and were assumed to have progressive disease. A physical examination was not performed prior to death, and therefore, these dogs were excluded from toxicity analysis. The other two dogs with T-cell lymphoma met inclusion criteria for response and experienced clinical benefit (CR and SD). The influence of individual factors on response could not be assessed due to low incidence in each subgroup.

The two dogs in which MOC was discontinued due to owner preference were excluded from PFS analysis. The median PFS for evaluable dogs (n = 24) was 29 days (range 1–280 days). The median PFS for dogs (n = 6) experiencing SD was 36 days (range 33–42 days). The median PFS for responders (CR and PR) was significantly longer compared with dogs who did not respond to MOC (47 days versus 14 days; P = .002). Similarly, the median PFS for dogs experiencing clinical benefit (CR, PR, and SD) was significantly longer than that for dogs experiencing PD (37 days versus 14 days; P = .005; Figure 2). Overall survival was not evaluated due to variable pursuit of additional rescue protocols. However, 21 dogs went on to receive additional chemotherapeutics.

FIGURE 2FIGURE 2FIGURE 2
FIGURE 2 Kaplan-Meier curve depicting effects of clinical benefit and progressive disease on progression-free survival. CB, clinical benefit; PD, progressive disease.

Citation: Journal of the American Animal Hospital Association 60, 1; 10.5326/JAAHA-MS-7372

Discussion

The primary objective of this study was to determine the tolerability of the combination of melphalan, vincristine, and cytarabine in a population of dogs with relapsed or refractory multicentric lymphoma. Our results demonstrate that the combination was well tolerated. Hematologic toxicity, specifically anemia and neutropenia, were the most common AEs observed in dogs treated with MOC, and AEs were generally mild.

The frequency of anemia in our study was unexpected. Myelosuppression is a common AE of chemotherapy; however, anemia is a less common AE due to the long lifespan of erythrocytes compared with platelets and neutrophils. Previous studies of protocols using cytarabine have not reported anemia as a side effect. However, the variable use of granulocyte colony-stimulating factor (G-CSF) and/or erythropoietin within several of these studies may have decreased the incidence of hematologic toxicity.26,35 Gillem et al. evaluated dogs with relapsed lymphoma treated with cytarabine and carboplatin without the use of G-CSF and reported anemia in 31% of dogs.10 Ruslander et al. evaluated single-agent cytarabine in treatment-naive dogs with lymphoma without the use of G-CSF.24 Although anemia was not a reported side effect, all dogs’ packed cell volume (PCV) decreased (median 5%, range 2–11%) following treatment. In the population described by Ruslander et al., the median initial PCV was 47% (range 23–57%), whereas in our population, the median initial PCV was 37% (range 24–54%).24 Subsequently, in our study, a mild decrease in PCV was often sufficient to cause a mild anemia. Importantly, the majority of anemia AEs in our study occurred in a subset of dogs (n = 6 of 23) and the risk of significant complications related to anemia was low. The only dog that required a blood transfusion had been anemic at the time of relapse and before initiating MOC (grade 2) and completed an additional cycle of MOC following the transfusion. The frequency of anemia seen in our study may be a consequence of this novel drug combination; however, anemia may also be related to undocumented bone marrow involvement, cumulative bone marrow suppression from previous chemotherapy, or both.

The frequency and severity of neutropenia was reduced in our study compared with previous studies.4,7,8,10,14,15,17,18 Dose delays were uncommon, and the dose of vincristine was escalated in 8 of 13 dogs treated with a second cycle of MOC. The only instance of grade 3 neutropenia was observed in a dog during the second cycle when the dose of vincristine had not been escalated. As the incidence of neutropenia was infrequent and mild, a higher initial dose of vincristine could be considered, which may improve the response rate and duration of response to the MOC protocol.

The third most common AEs were diarrhea and lethargy. Gastrointestinal and constitutional toxicity was relatively uncommon in the MOC protocol compared with other multiagent rescue protocols.6,1315,17 As this study was retrospective, gastrointestinal toxicity may have been underestimated, particularly if low grade, as it may not have been reported by the owners or noted in the clinical records. Although gastrointestinal AEs were generally mild, the MOC protocol was discontinued in two dogs by owners as a result of an AE or a perceived decrease in quality of life. One of these dogs experienced anorexia, and the other experienced lethargy and diarrhea. These AEs could have been addressed through dose reductions and/or prophylactic medications rather than discontinuation of chemotherapy. Balancing chemotherapy toxicity, tumor response, and patient quality of life remains challenging. A recent retrospective study reported multiagent chemotherapy protocols and hematopoietic tumors are reported risk factors for severe AEs.36 Severe AEs were defined as AEs that cause chemotherapy discontinuation, hospitalization, death, grade 4 hematological AEs, and symptomatic grade 3 hematological AEs. Chavelle et al. reported that severe AEs were seen in 38% of dogs treated with multiagent protocols, which is higher than observed in dogs treated with MOC (8%).36

Although response to treatment was not the primary objective of this study, the overall response rate of 38P% for a median duration of 29 days reported in our study is comparable to other second-line rescue protocols for the treatment of canine relapsed or refractory lymphoma.7,8,17,18 An additional seven dogs experienced SD for a median of 36 days, resulting in clinical benefit in 65% of MOC-treated dogs. In veterinary oncology, where a cure is often unattainable or clinically challenging, clinical benefit should be considered an acceptable measure of treatment outcome. Although a benefit was seen in a majority of patients, the duration of benefit was limited. Our population had a short median duration of response to first-line treatment. Thus, it is possible a longer duration of benefit to MOC may be seen in populations of dogs with more typical responses to induction treatment. Two dogs experienced durable remissions, experiencing a response for >100 days. Both of these dogs had been treated with MOC as first-line rescue. Only four dogs completed the planned three cycles of MOC; the median PFS for these dogs was 81 days (range 42–280 days). It is possible additional cycles of MOC would result in a more durable response for a subset of patients.

In this study, dogs were excluded from toxicity analysis if they did not have adequate follow-up. Three dogs died within 5 days of receiving cytarabine and vincristine. All three dogs had stage Vb lymphoma, had been refractory to CHOP, and were systemically ill before receiving vincristine/cytarabine. These dogs were suspected to have died of lymphoma; however, necropsy was not performed. The exclusion of these dogs may have introduced bias toward a more favorable toxicity profile in dogs treated with MOC.

Limitations of this study include the retrospective nature, small sample size, and heterogeneity of the population. Although all dogs had multicentric lymphoma, full staging was not required at the time of diagnosis or relapse. Additional limitations include variability in drug dosages, order of treatment, and route of drug administration. In this study, a subset of dogs (n = 3) received cytarabine subcutaneously in four doses over 2 days (target total dose 300 mg/m2). This route was chosen depending on clinician and owner preference. Alvarez et al. found no significant difference in response between subcutaneous or continuous rate infusion administration as part of the dexamethasone, melphalan, actinomycin D, and cytosine arabinoside protocol in dogs.3 However, a pharmacokinetic study of cytarabine in healthy dogs showed that subcutaneous administration has a limited ability to maintain steady-state concentrations compared with continuous rate infusions.37 A follow-up study in dogs with meningoencephalitis treated with subcutaneous cytarabine revealed similar plasma concentrations as previously reported.38 Although the plasma concentration of cytarabine necessary to produce a clinical response in dogs is unknown, rapid elimination may result in the drug being less efficacious when administered subcutaneously.37,38 In the present study, it was not possible to compare responses of dogs treated with either route of administration due to the small sample size.

Conclusion

Our findings suggest MOC combination chemotherapy is well tolerated in dogs with relapsed or refractory lymphoma. Additional studies are warranted to validate the potential benefit of the MOC protocol in a larger cohort and explore optimal dosing.

AE

(adverse event);

CHOP

(vincristine, cyclophosphamide, doxorubicin, and prednisone);

CR

(complete response);

G-CSF

(granulocyte colony-stimulating factor);

MOC

(melphalan, vincristine, and cytarabine);

PCV

(packed cell volume);

PD

(progressive disease);

PFS

(progression-free survival);

PO

(per os);

PR

(partial response);

SD

(stable disease);

VCOG

(Veterinary Cooperative Oncology Group)

Footnote

  1. Prism v9, GraphPad Software; La Jolla, California

REFERENCES

  • 1.
    Regan RC, Kaplan MS, Bailey DB. Diagnostic evaluation and treatment recommendations for dogs with substage-a high-grade multicentric lymphoma: results of a survey of veterinarians. Vet Comp Oncol2012;11:28795.
  • 2.
    Hosoya K, Kisseberth WC, Lord LK, et al. Comparison of COAP and UW-19 protocols for dogs with multicentric lymphoma. J Vet Intern Med2007;21(
    6
    )135563.
  • 3.
    Alvarez FJ, Kisseberth WC, Gallant SL, et al. Dexamethasone, melphalan, actinomycin D, cytosine arabinoside (DMAC) protocol for relapsed lymphoma. J Vet Intern Med2006;20:117883.
  • 4.
    Back AR, Schleis SE, Smrkovski OA, et al. Mechlorethamine, vincristine, melphalan, and prednisone (MOMP) for the treatment of relapsed lymphoma in dogs. Vet Comp Oncol2015;13:398408.
  • 5.
    Fahey CE, Milner RJ, Barabas K, et al. Evaluation of the University of Florida lomustine, vincristine, procarbazine, and prednisone chemotherapy for the treatment of relapsed lymphoma in dogs: 33 cases (2003-2009). J Am Vet Med Assoc2011;239:20915.
  • 6.
    Parsons-Doherty M, Poirier VJ, Monteith G. The efficacy and adverse event profile of dexamethasone, melphalan, actinomycin-D, and cytosine arabinoside (DMAC) chemotherapy in relapsed canine lymphoma. Can Vet J2014;55:17580.
  • 7.
    Mastromauro ML, Suter SE, Hauck ML, et al. Oral melphalan for the treatment of relapsed canine lymphoma. Vet Comp Oncol2018:16:E1239.
  • 8.
    Intile JL, Rassick KM, Al-Sarraf R et al. Evaluation of the tolerability of combination chemotherapy with mitoxantrone and dacarbazine in dogs with lymphoma. J Am Hosp Assoc2019;55:1019.
  • 9.
    Northrup NC, Gieger TL, Kosarek CE, et al. Mechlorethamine procarbazine and prednisone for the treatment of resistant lymphoma in dogs. Vet Comp Oncol2009;7:3844.
  • 10.
    Gillem J, Giuffrida M, Krick E. Efficacy of and toxicity of carboplatin and cytarabine chemotherapy for dogs with relapsed or refractory lymphoma (2000-2013). Vet Comp Oncol2017;15:40010.
  • 11.
    Rassick KM, Mauldin GE, Al-Sarraf R, et al. MOPP chemotherapy for the treatment of resistant lymphoma in dogs: a retrospective study of 117 cases (1989-2000). J Vet Intern Med2002;16:57680.
  • 12.
    Saba CF, Thamm DH, Vail DM, et al. Combination chemotherapy with L-asparaginase, lomustine, and prednisone for relapsed or refractory canine lymphoma. J Vet Intern Med2007;21:12732.
  • 13.
    Saba CF, Vickery KR, Clifford CA, et al. Rabacfosadine for relapsed canine B-cell lymphoma: efficacy and adverse event profiles of 2 different doses. Vet Comp Oncol2018;16:E7682.
  • 14.
    Tanis JB, Mason SL, Maddox TW, et al. Evaluation of a multicentric chemotherapy protocol combining lomustine, procarbazine, and prednisone (LPP) for the treatment of relapsed canine non-Hodgkin high-grade lymphomas. Vet Comp Oncol2018;16(
    3
    ):3619.
  • 15.
    Flory AB, Rassick KM, Al-Sarraf R, et al. Combination of CCNU and DTIC chemotherapy for the treatment of resistant lymphoma in dogs. J Am Vet Med Assoc2007;231:5639.
  • 16.
    Treggiari E, Elliot JW, Baines SJ, et al. Temozolamide alone or in combination with doxorubicin as a rescue agent in 37 cases of canine multicentric lymphoma. Vet Comp Oncol2018;16:194201.
  • 17.
    Lenz JA, Robat CS, Stein TJ. Vinblastine as a second rescue for treatment of canine multicentric lymphoma in 39 cases (2005 to 2014). J Small Anim Pract2016;57(
    8
    ):42934.
  • 18.
    Moore AS, London CA, Wood CA, et al. Lomustine (CCNU) for the treatment of resistant lymphoma in dogs. J Vet Intern Med1999;13:3958.
  • 19.
    Chun R, Garret LD, Vail DM. Evaluation of a high-dose chemotherapy protocol with no maintenance therapy for dogs with lymphoma. J Vet Intern Med2000;14:1204.
  • 20.
    Baskin CR, Couto GG, Wittum TE. Factors influencing first remission and survival in 145 dogs with lymphoma: A retrospective study. J Anim Hosp Assoc1987;23:58796.
  • 21.
    Stone MS, Goldstein MA, Cotter SM. Comparison of two protocols for induction of remission in dogs with lymphoma. J Am Anim Hosp Assoc1991;27:31521.
  • 22.
    Chu E, DeVita VT. Principles of medical oncology. In: DeVitaVT,HallmanS,RosenburgSA, eds. Cancer, Principles & Practice of Oncology.
    7th ed
    .
    Philadelphia
    :
    Lippincott Williams & Wilkins
    ; 2005:295306.
  • 23.
    Golden DL, Langston VC. Uses of vincristine and vinblastine in dogs and cats. J Am Vet Med Assoc1988;9:11147.
  • 24.
    Ruslander D, Moore AS, Gliatto JM et al. Cytosine arabinoside as a single agent for the induction of remission in canine lymphoma. J Vet Intern Med1994;8:299301.
  • 25.
    Weller RE, Theilen GH, Madewell BR. Chemotherapeutic responses in dogs with lymphosarcoma and hypercalcemia. J Am Vet Med Assoc1982;181(
    9
    )8913.
  • 26.
    Marconato L, Bonfanti U, Stefanello D, et al. Cytosine arabinoside in addition to VCAA-based protocols for the treatment of canine lymphoma with bone marrow involvement: does it make a difference? Vet Comp Oncol 2008;6(
    2
    ):809.
  • 27.
    Chau CH, Figg WD, Chabner BA. Antimitotic drugs. In: Cancer Chemotherapy, Immunotherapy and Biotherapy: Principles and Practice.
    6th ed
    .
    Philadelphia
    :
    Wolters Kluwer
    ; 2019:175200.
  • 28.
    Gerson SL, Weeks LD, Chabner BA. Alkylating and methylating agents. In: Cancer Chemotherapy, Immunotherapy and Biotherapy: Principles and Practice.
    6th Ed
    .
    Philadelphia
    :
    Wolters Klower
    ; 2019:20034.
  • 29.
    Furth JJ, Cohen SS. Inhibition of mammalian DNA polymerase by the 5′triphosphate of 1-beta-d-arabinofuranosylcytosine and the 5′triphosphate of 9-beta-arabinofuranoxyladenine. Cancer Res1968;28:20617.
  • 30.
    Kufe DW, Major PP, Egan EM, et al. Correlation of cytotoxicity with incorporation of Ara-C into DNA. J Biol Chem1980;255:8997900.
  • 31.
    Supko JG, Collins JM. Principles of pharmacokinetics. In: Cancer Chemotherapy, Immunotherapy and Biotherapy: Principles and Practice.
    6th ed
    .
    Philadelphia
    :
    Wolters Kluwer
    ; 2019:3747.
  • 32.
    Owen LN. TMN Classification of Tumors in Domestic Animals.
    1st ed
    .
    Geneva
    :
    World Health Organization
    ; 1989.
  • 33.
    Vail DM, Michels GM, Khanna C et al. Response evaluation criteria for peripheral nodal lymphoma in dogs (v1.0)- a Veterinary Cooperative Oncology Group (VCOG) consensus document. Vet Comp Oncol2010;8(
    1
    ):2837.
  • 34.
    LeBlanc AK, Atherton M, Bentley TR, et al. Veterinary Cooperative Oncology Group- Common Terminology Criteria for Adverse Events (VCOG-CTCAE v2) following investigational therapy in dogs and cats. Vet Comp Oncol2021;19:31152.
  • 35.
    Guillen A, Finotello R, Harper A, et al. Toxicity of cytarabine constant rate infusion in dogs with high-grade non-Hodgkin lymphoma with bone marrow or central nervous system involvement. Aust Vet J2020;98:6978.
  • 36.
    Chavelle T, Chamel G, Deneux P, et al. Are severe adverse events commonly observed in dogs during cancer chemotherapy? A retrospective study on 155 dogs. Vet Comp Oncol2022;20(
    2
    ):393403.
  • 37.
    Crook KI, Early PJ, Messenger KM, et al. The pharmacokinetics of cytarabine in dogs when administered via subcutaneous and continuous intravenous infusion routes. J Vet Pharmacol Ther2013;36:40811.
  • 38.
    Pastina B, Early PJ, Bergman RL, et al. The pharmacokinetics of cytarabine administered subcutaneously, combined with prednisone in dogs with meningoencephalitis of unknown origin. J Vet Pharmacol Ther2018;41:63843.
Copyright: © 2024 by American Animal Hospital Association 2024
FIGURE 1
FIGURE 1

Flow chart provides details of patient screening and inclusion and exclusion from study. AE, adverse event; GI, gastrointestinal; MOC, melphalan, vincristine, and cytarabine; PFS, progression-free survival.


FIGURE 2
FIGURE 2

Kaplan-Meier curve depicting effects of clinical benefit and progressive disease on progression-free survival. CB, clinical benefit; PD, progressive disease.


Contributor Notes

Correspondence: Margaret.Duckett1@nva.com (M.D.)
Accepted: 06 Nov 2023
  • Download PDF