2003 3 MAY Advances in medical oncology

background image

Preface

Advances in medical oncology

Guest Editor

The specialty of medical oncology has traditionally been characterized by

steady, incremental gains in the efficacy of therapy and only rarely by star-
tling breakthroughs. This is as true in veterinary medical oncology as it is in
the human specialty. The past decade in veterinary medical oncology has
produced modest advances in the treatment of several cancers. We have
added new compounds to our therapeutic arsenal and refined the use of
standard anticancer chemotherapy drugs through improvements in dose and
scheduling of combination therapy. Early intervention has contributed to
this increased treatment success. Improved diagnostics, better understand-
ing of the biologic behavior of individual tumors provided by new prog-
nostic and predictive markers, and an attitudinal shift in the minds of
veterinary practitioners and the pet owning public have all contributed to
increased success in medical oncology. Patient outcomes have also been
improved through refinements in multimodality therapy, combining the
expertise of specialists in medical, radiation, and surgical oncology. This
issue of Veterinary Clinics of North America: Small Animal Practice presents
updated information on the treatment of the diseases commonly encoun-
tered in veterinary oncology. I hope that the information provided will
prove useful to students, specialists in training, and practitioners.

Barbara E. Kitchell, DVM, PhD

Vet Clin Small Anim

33 (2003) xi–xiii

0195-5616/03/$ - see front matter

Ó 2003, Elsevier Inc. All rights reserved.

doi:10.1016/S0195-5616(03)00022-6

background image

The commonly encountered tumors that impair the longevity and quality

of life of our patients are reviewed in the chapters by Drs. Fan, London,
Chun and de Lorimier, Rassnick, Smith, Hauck, Sorenmo, Henry, and
Klein. A great deal of the information in their reviews of the literature was
garnered from individual case reports, small case series, and retrospective
studies. I am sure these authors would agree that there is an urgent need for
prospective, appropriately blinded and randomized trials in veterinary med-
ical oncology. In the past few years, the first large industry sponsored multi-
center trial with intent to achieve veterinary drug licensing was conducted,
and more such trials are ongoing or planned. The organization of multi-
center trial groups in the US, as well as the move into prospective work by
the venerable Veterinary Cooperative Oncology Group, promises to speed
progress in our specialty. The conduct of well-designed and ethically con-
ducted prospective trials is critical to our future.

Recent publications and abstracts describing the use of several agents

new to veterinary oncology are summarized here. The reader is advised to
consult veterinary medical oncology specialists for new information and
guidance regarding the use of these compounds. Clients should be informed
of the off-label status of all anticancer chemotherapy agents used in vet-
erinary medicine, and appropriate biosafety precautions should be
emphasized for all who come in contact with these drugs. Improved un-
derstanding of the mechanisms of drug resistance provided by Dr. Phil
Bergman’s chapter may prove helpful in explaining individual case out-
comes and in defining future directions for therapy.

The past decade has seen molecularly targeted therapeutic approaches

advance as an intense focus in human medicine, with new compounds de-
veloped through research in traditional pharmacology, recombinant biolog-
ics, and immunotherapy. Some of these novel agents have changed the
standard of care for human patients with disseminated cancers, and more
hold promise in the near future. The exquisitely selective nature of molecular
targeting permits enhanced efficacy while limiting adverse effects. Unfortu-
nately, it is this precise selectivity in molecular targeting that makes the
translational use of these agents in clinical companion animal practice dif-
ficult, if not impossible. The coming decade will be a challenging and
exciting one for veterinary medical oncology as we begin to move

en-

thusiastically into this new field. Clearly, we have work to do.

I wish to thank all of the contributors for the their thorough and thought-

ful reviews. The residents in medical oncology at the University of Illinois,
especially Louis-Philippe de Lorimier and Amy Wiedemann, were in-
valuable for editorial input regarding both content and style. Finally, John
Vassallo at W.B. Saunders is a man of formidable patience, good humor, and

xii

B.E. Kitchell / Vet Clin Small Anim 33 (2003) xi–xiii

background image

editorial skill. I owe him abundant thanks, a drink, and a case of Maalox for
putting up with me.

Barbara E. Kitchell, DVM, PhD

College of Veterinary Medicine

1008 West Hazelwood Drive

University of Illinois

Urbana, IL 61802, USA

E-mail address: kitchell@uiuc.edu

xiii

B.E. Kitchell / Vet Clin Small Anim 33 (2003) xi–xiii

background image

Lymphoma updates

Timothy M. Fan, DVM

Department of Veterinary Clinical Medicine, Veterinary Teaching Hospital,

College of Veterinary Medicine, University of Illinois,

100 West Hazelwood Drive, Urbana, IL 61802, USA

Lymphoma arises from the malignant clonal expansion of lymphoretic-

ular cells. Primary lymphoid organs, such as the bone marrow and thymus,
as well as secondary lymphoid structures, including the lymph nodes, spleen,
and gut-associated lymphoid tissue (GALT), are potential sites of neoplastic
transformation. As a result of continuous lymphocyte trafficking, the origin
of lymphoma is not restricted solely to primary and secondary lymphoid
organs. Malignant transformation of lymphocytes can occur virtually
anywhere. Common extranodal sites of lymphoma include the skin, eye,
central nervous system, testis, and bone [1].

Lymphoma is reported to be the most common hematopoietic neoplasm

affecting the dog and cat. In dogs, the incidence of lymphoma has been
reported to approach 0.1% in susceptible older individuals, with an annual
incidence rate of 84 per 100,000 dogs at risk [2]. The precise etiology of
lymphoma in the dog has not been identified. Several hypotheses have been
investigated but have not been definitely proven. Hypothesized etiologies for
canine lymphoma include retroviral infection, environmental contamination
with phenoxyacetic acid herbicides, magnetic field exposure, chromosomal
abnormalities, and immune dysfunction [3–8].

In cats, feline leukemia virus (FeLV) has been identified as a biologic car-

cinogen resulting in malignant lymphocyte transformation [9,10]. Historical
epidemiologic investigations before the wide use of preventative FeLV vac-
cines estimated the annual incidence of feline lymphoma to be 200 per
100,000 cats at risk [11]. With the development of efficacious FeLV vaccines
in conjunction with the practice of early detection and removal of viremic
cats from the general population, the incidence of FeLV-induced lymphoma
has been dramatically reduced [12].

Vet Clin Small Anim

33 (2003) 455–471

E-mail address:

t-fan@uiuc.edu

0195-5616

/03/$ - see front matter

Ó 2003, Elsevier Inc. All rights reserved.

doi:10.1016/S0195-5616(03)00005-6

background image

Although lymphoma has been reported to occur in both dogs and cats,

response to therapy, disease-free intervals (DFIs), and survival times differ
not only between species but among individuals of the same species. The
nonuniform response to treatment among dogs and cats with lymphoma is
caused, in part, by individual variances in host immunity and constitutional
status. A more significant determinant of therapeutic response is dictated
by the variable biologic behavior of lymphoma. Some differences in bio-
logic behavior have been attributed to factors like anatomic location
and histologic grade [13]. An additional variable that seems to influence
the biologic behavior of lymphoma is immunophenotype [14,15]. Immuno-
phenotyping identifies selected surface glycoproteins known as cluster of
differentiation (CD) antigens on lymphocytes and accessory immune cells.
Identifying CD antigens requires the application of antibodies specific for
differentiation antigens. Lymphocyte CD antigens serve multiple functions,
including intracellular signaling, cell-to-cell communication, and lympho-
cyte trafficking [16].

Diagnostically important CD antigens for the characterization of

malignant lymphoma include CD3 and CD79. CD3 is a complex of five
polypeptides associated with the T-cell receptor (TCR). High-affinity binding
of a TCR with its cognate major histocompatibility complex containing
processed peptide allows intracellular signaling to be mediated through the
CD3 complex [16]. Demonstration of the CD3 antigen on malignant
lymphocytes identifies the lymphoma as being of T-lymphocyte origin.
Similarly, CD79 exists as a heterodimer associated with the B-cell receptor
(BCR) and is required for B-lymphocyte intracellular signal transduction
[16]. Demonstration of the CD79 antigen on malignant lymphocytes delin-
eates the malignant lymphoid population as being of B-lymphocyte origin.
T-cell lymphomas tend to be more biologically aggressive than B-cell
lymphomas, resulting in shorter remission and survival times [14,15]. Rarely,
malignant lymphocytes fail to demonstrate either CD3 or CD79 antigens. In
such instances, the cell of origin cannot be determined, and these lymphomas
are classified as being derived from null cells (nonreactive with any specific
lymphocyte antibody).

Treatment of lymphoma in the dog and cat is often rewarding. With con-

ventional chemotherapeutic protocols, most patients respond favorably.
Dogs treated with cyclophosphamide, doxorubicin, vincristine, and predni-
sone (CHOP) style protocols generally achieve a complete response rate of
80% to 90%, with expected median survival times of 1 year [17,18]. Although
chemotherapy is reasonably effective in the treatment of feline lymphoma,
response rates and survival times tend to be worse in cats than in dogs. Cats
treated with chemotherapy typically achieve response rates and survival
times of 50% to 70% and 4 to 6 months, respectively [19–21].

In conjunction with systemic chemotherapy, the implementation of

additional therapeutics, including surgery, radiation therapy, and biologic
response modifiers, may augment treatment success. Although a cure for

456

T.M. Fan

/ Vet Clin Small Anim 33 (2003) 455–471

background image

lymphoma remains elusive for most veterinary patients, the rapid accumu-
lation of knowledge elucidating the fundamentals of cancer biology inevi-
tably translates into the development of better diagnostic techniques, more
accurate prognostic indicators, and superior treatment options. The focus of
this article is to highlight some new developments in the diagnosis, prognosis,
and therapeutic management of lymphoma in the dog and cat.

New developments in canine lymphoma

New diagnostic tools

Immunoglobulin rearrangement

Nonpainful generalized peripheral lymphadenopathy is the most common

clinical manifestation of canine lymphoma [1]. Traditionally, the definitive
diagnosis of lymphoma has been made by either cytologic or histologic
assessment of affected sites. Fine-needle aspiration and cytologic evaluation
of enlarged peripheral lymph nodes usually provide a sufficient quantity of
cells to identify a malignant population. Tissue biopsy of affected lymph
nodes by incisional or excisional techniques not only provides adequate
tissue sample size but preserves nodal architecture. Although cytologic and
histologic diagnosis is reliable and practical, in rare instances, these tech-
niques fail. Being able to identify a population of malignantly transformed
lymphocytes definitively would be valuable when cytologic or histologic
assessment is inconclusive.

Based on the monoclonality theory of cancer, all tumors originally arise

from a common progenitor transformed cell [22,23]. This theory also applies
to lymphoid malignancies; therefore, lymphoma should be considered a
clonal expansion of one malignant lymphocyte. Lymphocytes serve a critical
immune function by protecting the host from a diverse spectrum of
pathogens. For lymphocytes to execute their programmed effector functions,
they must recognize their cognate antigens through specialized antigen-
binding sites, which are unique to each individual lymphocyte [24]. In the
case of a T lymphocyte, the unique antigen-binding site is referred to as the
TCR. Likewise, in the case of a B lymphocyte, the antigen-binding site,
referred to as the BCR, is also distinctive. Because each TCR or BCR is
unique, isolating the DNA sequences coding these protein structures allows
for the differentiation of lymphocytes based on these DNA sequences [24].

The use of polymerase chain reaction (PCR) technology allows for the

amplification of DNA sequences coding for either the TCR or BCR in
lymphocyte populations. In tissue containing lymphocytes derived from
multiple clones, such as is found in lymphoid hyperplasia, PCR amplification
of the DNA encoding the antigen-binding region produces amplicons of
varying size. In lymphoid malignancies, however, PCR amplification of the
DNA encoding the antigen-binding region produces a PCR product of
predominantly one size, indicating the presence of a clonally expanded

457

T.M. Fan

/ Vet Clin Small Anim 33 (2003) 455–471

background image

lymphocyte population [25]. Although this PCR technique is highly sensitive,
the methodology is relatively laborious and should only be used when the
clinician has a high index of suspicion for lymphoma and where conventional
cytologic and histologic diagnostic techniques have failed to be definitively
diagnostic.

Telomerase activity

Telomeres are long stretches of tandem hexanucleotide repeats

(TTAGGG) found at the 39 end of chromosomes. Functionally, telomeres
serve as protective ‘‘caps,’’ preventing deleterious chromosomal degradation
and recombination [26], Telomere length dictates the proliferative capacity of
normal cells. Subsequent to each replicative cycle, telomere length is reduced.
When terminal telomere length is reached, cellular replication is halted and
the cell enters into a state of cellular senescence [27]. One mechanism by
which malignantly transformed cells may circumvent reductions in telomere
length, thereby achieving cellular immortality, is by the activity of an enzyme
called telomerase. Telomerase is a ribonucleoprotein DNA polymerase, and
its RNA component provides the primer for telomere repeat synthesis. Most
normal somatic cells do not express telomerase, but this enzyme activity has
been detected in up to 90% of human cancers [28].

The utility of detecting telomerase activity as a diagnostic tool for canine

lymphoma has been investigated. Two studies in dogs measuring telomerase
activity by either enzyme-linked immunosorbent assay (ELISA) or a telomere
repeat amplification protocol (TRAP) methodology failed to identify any
statistical difference in enzymatic activity between normal and malignantly
transformed lymph nodes [29,30]. Results from these two studies suggest that
telomerase activity may not be a valuable diagnostic marker for the discrimi-
nation of malignant lymphocytes from normal lymphoid cells. In a separate
investigation using a TRAP assay, however, strong telomerase activity was
identified in all malignant lymphoid tissues evaluated (n = 28), whereas only
weak telomerase activity could be demonstrated in few normal lymph nodes
(3 of 12 lymph nodes) [31]. Because of this divergence in results, additional
research is required to fully explore the potential for telomerase activity
as a diagnostic tool for detection of malignant transformation of canine
lymphocytes.

New prognostic markers

Because different types of lymphocytes may undergo malignant trans-

formation, lymphomas manifest a wide spectrum of clinical and biologic
behaviors. Given the emotional, time, and financial commitments required in
the course of lymphoma treatment, providing reliable information address-
ing response rates, DFIs, and survival times is important for owners in their
decision-making process. A client’s decision to treat a pet with traditional
chemotherapy may depend on the likelihood of achieving a favorable

458

T.M. Fan

/ Vet Clin Small Anim 33 (2003) 455–471

background image

response. Advances in our understanding of cancer biology have led to the
discovery of new predictive markers that facilitate more accurate prognos-
tication of canine lymphoma.

Serum alpha 1-acid glycoprotein concentrations

The presence of infectious, neoplastic, or immune-mediated diseases

results in the liberation of inflammatory cytokines from immune cells.
Cytokines, such as interleukins, interferons, and tumor necrosis factor syn-
thesized and released by the host organism, can directly and indirectly
augment host immunity. Subsequent to cytokine release, hepatocytes
synthesize a variety of acute-phase proteins that enhance antigen-presenting
cell phagocytosis [32]. One acute-phase protein, serum alpha 1-acid
glycoprotein (AGP), has been demonstrated to have value in predicting
lymphoma relapse in the dog. In dogs with lymphoma, baseline serum AGP
levels are significantly elevated when compared with those of healthy control
dogs. After complete remission induced by doxorubicin therapy, dogs with
lymphoma have serum AGP concentrations similar to those of healthy dogs.
Mean serum AGP concentrations 3 weeks before and at the time of relapse
are again significantly elevated, achieving the concentration levels measured
before treatment [33]. Although the investigation of AGP has proven to be
a statistically significant factor in predicting lymphoma relapse in dogs,
interpretative limitations exist. With any nonspecific inflammatory marker
like AGP, documented elevations warrant investigation for any systemic
inflammatory nidus, whether neoplastic or nonneoplastic.

Matrix metalloproteinase 2 and 9 expression

Multiple events are required for the successful distant spread of

a neoplasm. Being able to migrate through extracellular matrix (ECM) is
one prerequisite in the acquisition of a metastatic phenotype. Matrix
metalloproteinases (MMPs) are a family of zinc-dependent proteases capable
of degrading type IV collagen, resulting in ECM degradation [34]. Although
more than 20 distinct MMPs have been identified, MMP 2 and 9 have been
investigated most intensively for their potential roles in cancer metastasis. In
human cancer patients, MMP 2 and 9 expression is elevated in metastatic
neoplastic cells [34,35,36]. Similarly, alterations in MMP 2 and 9 levels have
been documented in canine neoplasms, including lymphoma, osteosarcoma,
and mast cell tumors [37–39]. In dogs with lymphoma, MMP 2 and 9 con-
centrations decreased significantly after treatment with chemotherapy and
remained low until 6 to 12 weeks before recurrence of lymphoma. Higher
concentrations of pro-MMP 2 and pro-MMP 9 as well as active MMP 9 in
tumor tissue correlated with shorter remission times after doxorubicin
therapy [38]. Measurement of MMP activity by gelatin zymography or MMP
gene expression by microarray analysis is not commercially available,
thereby restricting MMP analysis as a predicative marker to research
settings.

459

T.M. Fan

/ Vet Clin Small Anim 33 (2003) 455–471

background image

Monoclonal antibody C219 immunohistochemistry against P-glycoprotein

Although treatment with chemotherapy is intended to eradicate all cancer

cells, this is rarely the outcome in veterinary cancer patients. In part, failure to
cure dogs routinely with lymphoma stems from owner economic constraints
as well as from a lack of necessary support systems (ie, bone marrow
transplantation) required for managing the critically ill patient subsequent
to dose-intense chemotherapy. In addition to these exogenous limitations,
malignantly transformed lymphocytes may evade the cytotoxic effects of
chemotherapy by the upregulation of resistance mechanisms.

Several resistance mechanisms have been documented in neoplastic cells,

including defective apoptotic pathways, enhanced free radical scavenging
capabilities, and upregulated DNA repair machinery [40]. Multidrug-
resistant (MDR) phenotype is used to describe neoplastic cells that are
resistant to the cytotoxic effects of certain antineoplastic agents. The
overexpression of a drug efflux pump called P-glycoprotein (P-gp) has been
examined in dogs with lymphoma [41,42]. Functionally, P-gp prevents the
retention of cytotoxic agents within neoplastic cells, allowing for escape
from lethal DNA damage.

The expression of P-gp on canine malignant lymphocytes has been dem-

onstrated by immunohistochemistry using the C219 monoclonal antibody.
The frequency of positively staining malignant lymphocytes at disease relapse
was significantly higher than that found before the initiation of chemother-
apy. Furthermore, greater positive staining with C219 before initial
chemotherapy was inversely proportional to remission and survival times.
The frequency of positively staining malignant lymphocytes at relapse was
also inversely related to the time from relapse to death [43]. Several logical
deductions may be concluded from this investigation. First, P-gp expression
may be upregulated subsequent to chemotherapy exposure. Second, drug-
naive lymphomas with inherently greater expression of P-gp may be more
resistant to the cytotoxic effects of traditional chemotherapeutic agents,
especially doxorubicin and vincristine. Finally, overexpression of P-gp at
disease relapse may result in a poor response to rescue chemotherapeutic
agents and therefore shorter survival times. Although the expression of P-gp
maybe used as a surrogate marker for predicting response to therapy, DFIs,
and survival times, it should be emphasized that acquisition of an MDR
phenotype does not necessarily require overexpression of P-pg. Additional
mechanisms contributing to chemotherapeutic resistance, such as the
overexpression of glutathione-S-transferase, have also been demonstrated
in dogs with lymphoma. Functionally, glutathione-S-transferase catalyzes
the conjugation of glutathione with free radicals, thereby inactivating the
cytotoxic effects of several anticancer drugs, including doxorubicin,
cyclophosphamide, and platinum-based agents [40]. In dogs with lymphoma,
serum glutathione-S-transferase levels have been evaluated before disease
treatment, during treatment, and at disease relapse. Significant elevations of
serum glutathione-S-transferase were present at the time of disease relapse,

460

T.M. Fan

/ Vet Clin Small Anim 33 (2003) 455–471

background image

supporting the notion that additional mechanisms contribute to the
development of drug resistance in canine lymphoma [44].

Immunophenotyping: B5 antigen expression

Glycoproteins found on the surface of cells perform distinct functions,

including the mediation of cell-to-cell communication and the propagation of
intracellular signals. In lymphocytes, the identification of specific surface
glycoproteins known as CD antigens allows cells to be classified into discrete
subsets. CD antigens can be identified by monoclonal antibodies; this process
is termed immunophenotyping. CD antigens identified on malignant lympho-
cytes that have prognostic importance are CD3 and CD79. Lymphomas
expressing CD3 but not CD79 are derived from T-cell lineage. Lymphomas
positive for CD79 but not CD3 are of B-cell origin. Multiple investigations
have consistently demonstrated dogs diagnosed with lymphoma of T-cell
lineage have shorter DFIs and survival times than dogs with B-cell
lymphoma [14,15,45]. In addition to CD3 and CD79, a nonimmunoglobulin
B-cell marker called B5 also seems to have prognostic value. The B5 antigen
is normally expressed on nonneoplastic B lymphocytes. Dogs with B-cell
lymphomas expressing lower than normal levels of the B5 antigen experience
shorter DFIs and survival times [15]. The exact function of the B5 antigen is
unknown. Investigating the functional properties of the B5 antigen in normal
B lymphocytes may provide an explanation for correlating reduced
expression with poorer clinical outcome in canine lymphoma.

New therapeutic modalities

In dogs, systemic chemotherapy for the management of multicentric

lymphoma is rewarding. With the use of multiagent doxorubicin-based
protocols, response rates are reported to exceed 90% [45,46]. Traditional
chemotherapeutic protocols are composed of two distinct treatment phases
referred to as ‘‘induction’’ and ‘‘maintenance.’’ During the induction phase,
aggressive weekly treatments are instituted with the intent of rapidly reducing
the burden of cancer cells. After induction therapy, maintenance chemo-
therapy may be used to thwart the rapid regrowth of residual cancer cells.
The necessity of maintenance chemotherapy in the successful management of
canine lymphoma has been challenged. Several investigations demonstrate
favorable DFIs and survival times in dogs treated for lymphoma by
aggressive induction protocols or radiation therapy. The use of either dose-
intense chemotherapy or adjunctive radiation therapy may offer a reasonable
alternative for the treatment of lymphoma without the use of chronic long-
term maintenance chemotherapy.

Chemotherapy treatment options that exclude maintenance therapy

In one study, dogs with multicentric lymphoma were treated with a dose-

intense modified version of the University of Wisconsin (UW)–Madison

461

T.M. Fan

/ Vet Clin Small Anim 33 (2003) 455–471

background image

induction protocol without maintenance (Table 1). Compared with the
traditional UW-Madison regimen, the dose-intense protocol used a greater
dose and number of treatments of doxorubicin (37.5 mg

/m

2

 4 versus 30

mg

/m

2

 3) and cyclophosphamide (250 mg/m

2

 4 versus 200 mg/m

2

 2)

during a 25-week induction period. Interestingly, dogs treated with the dose-
intense induction protocol with no maintenance achieved statistically similar
DFIs and survival times as a historical control population of dogs treated
with the standard UW-Madison protocol (induction with maintenance).
Death because of treatment-related toxicity was significantly higher in dogs
treated with the dose-intense version of the UW-Madison induction protocol
(odds ratio = 8.8) [45]. Results from this study support the notion that
aggressive induction therapy without maintenance can provide equivalent
DFIs and survival times when compared with more traditional chemother-
apeutic regimens that include maintenance therapy. Unfortunately, the high
incidence of treatment-induced morbidity and mortality makes this specific
dose-intense protocol unattractive to many veterinary oncologists.

In lieu of the unacceptable toxicity observed with this protocol, a second

investigation using a slightly different dose-intense UW-Madison induction
protocol was evaluated. In this second study, the only difference between the
experimental dose-intense protocol and the traditional UW-Madison
regimen was the dose and number of treatments of cyclophosphamide
(250 mg

/m

2

 4 versus 200 mg/m

2

 2) and the number of treatments of

doxorubicin (30 mg

/m

2

 4 versus 30 mg/m

2

 3) during a 25-week induc-

tion period. Results demonstrated that dogs treated with the dose-intense
induction protocol without maintenance achieved statistically similar
DFIs and survival times as a historical control population of dogs treated
with the standard UW-Madison protocol (induction with maintenance).

Table 1
Comparison of University of Wisconsin–Madison canine maintenance-free lymphoma protocols

Week

1 2 3 4 5 6 7 8 9 11 13 15 17 19 21 23 25

Chun et al (2000)

L

-asparaginase (400 IU

/kg SC)

d

Vincristine (0.7 mg

/m

2

IV)

d

d

d

d

d

d

d

d

Cyclophosphamide (250 mg

/m

2

IV)

d

d

d

d

Doxorubicin (37.5 mg

/m

2

IV)

d

d

d

d

Prednisone (2 mg

/kg PO q 24 h

 7 d, then 1.5 mg/kg PO q 24 h  7 d, then 1.0 mg/kg PO q

24 h

 7 d, then 0.5 mg/kg PO q 24 h  7 d, then stop)

Garrett et al (2002)

L

-asparaginase (400 IU

/kg SC)

d

Vincristine (0.7 mg

/m

2

IV)

d

d

d

d

d

d

d

d

Cyclophosphamide (250 mg

/m

2

IV)

d

d

d

d

Doxorubicin (30.0 mg

/m

2

IV)

d

d

d

d

Prednisone (2 mg

/kg PO q 24 h

 7 d, then 1.5 mg/kg PO q 24 h  7 d, then 1.0 mg/kg PO q

24 h

 7 d then 0.5 mg/kg PO q 24 h  7 d, then stop)

Abbreviations:

IV, intravenous; PO, by month; q, every day; SC, subcutaneous.

462

T.M. Fan

/ Vet Clin Small Anim 33 (2003) 455–471

background image

Furthermore, there was no statistical difference in treatment-associated
morbidity or mortality between the experimental and historical control
populations [46]. The results from this study suggest that moderately dose-
intense induction protocols without maintenance may be well tolerated and
can provide equivalent DFIs and survival times as traditional induction

/

maintenance protocols.

Half-body radiation therapy in lieu of maintenance chemotherapy

Malignant lymphocytes may undergo apoptosis after irreparable DNA

damage caused by ionizing radiation [47,48]. Despite the exquisite sensitivity
of lymphocytes to the cytotoxic effects of radiation, its use in veterinary
oncology has been traditionally reserved for ‘‘crisis’’ situations, where the
rapid reduction of localized tumor volume is necessary. In treating diffuse
lymphoid malignancies, such as canine multicentric lymphoma, delivering
radiation to the entire body would be required to maximize the therapeutic
response. Unfortunately, without autologous bone marrow transplanta-
tion (AuBMT) or other hematologic support, dogs treated with total body
irradiation (TBI) experience profound bone marrow suppression and high
mortality rates. Even with extremely low-dose TBI (total dose of 1 Gy at
10 cGy

/min), dogs with lymphoma experience grade III and IV thrombo-

cytopenia [49]. To derive the benefits of TBI without the complications,
sequential half-body irradiation (HBI) of dogs with lymphoma in remission
has been investigated. In one study, 29 dogs were treated with HBI after
successful induction chemotherapy. HBI was delivered to the cranial half of
the body on week 8 and to the caudal half of the body on week 12. Side effects
of HBI included self-limiting alopecia, fever, and anorexia. Conclusions from
this study indicated that 800-cGy HBI is well tolerated and is as effective as
maintenance chemotherapy for dogs in remission with lymphoma [50]. In
a second study, 38 dogs with lymphoma were treated with HBI after
successful chemotherapy induction. After an 11-week induction protocol,
radiation was administered at a dose of 800 cGy per half body given in two
consecutive 400-cGy fractions. Cranial and caudal HBI was separated by a 3-
week interval and delivered on weeks 13 and 16, respectively. HBI resulted in
mild reductions in neutrophil and platelet numbers. Survival times of dogs
treated with HBI were comparable to those of dogs treated with traditional
maintenance chemotherapy [51].

New rescue therapies

Rescue chemotherapeutics

Although lymphoid neoplasms initially respond well to systemic chemo-

therapy, the development of resistant clones with eventual disease relapse is
common. Additional remissions after administration of rescue chemother-
apeutic agents are frequently achievable and may substantially prolong the
survival times of dogs and cats with lymphoma. The development of new

463

T.M. Fan

/ Vet Clin Small Anim 33 (2003) 455–471

background image

rescue chemotherapeutics has been limited; however, the use of an oral and
inexpensive alkylating agent called lomustine has been demonstrated to be an
effective rescue agent in dogs.

The safety and efficacy of single-agent lomustine have been evaluated in

the treatment of canine lymphoma. Dogs with lymphoma relapse receiving
lomustine dosed at 90 mg

/m

2

achieved a response rate of 28% and median

duration of remission of 86 days. The acute dose-limiting toxicity in this
study was neutropenia, and cumulative thrombocytopenia occurred in dogs
receiving continued long-term lomustine therapy [52]. Although lomustine
has not been formally reported to be an effective rescue agent for feline
lymphoma, anecdotal reports suggest lomustine’s efficacy for the treatment
of various round cell neoplasms in the cat [53]. Cats may be safely dosed at
50 to 60 mg

/m

2

every 6 weeks [54]. Alternatively, cats may be given a single

10-mg capsule every 21 days with a low incidence of hematologic side effects
[53].

Rescue radiation therapy

Malignant lymphocytes expressing MDR phenotypes are afforded a

survival advantage when exposed to cytotoxic agents. Attempting to treat
resistant lymphocyte clones, even with novel chemotherapeutic agents, still
may result in disease progression. Because of the limitations of chemical
cytotoxic agents in conjunction with the inherent radiosensitivity of malig-
nant lymphocytes, the use of total lymphoid irradiation (TLI) for confirmed
chemoresistant lymphoma has been investigated. Eleven dogs with confirmed
MDR lymphoma were treated with total nodal irradiation. A total dose of
200 cGy given in six fractions over 2 weeks was administered to all affected
peripheral lymph nodes. Complete remission was achieved in all treated
lymph nodes by the fourth radiation fraction. In the 9 dogs available for
follow up, the median survival time was 143 days [55].

Future therapies

Although aggressive chemotherapy and radiation therapy serve as excel-

lent treatment modalities for the management of lymphoma, newer treat-
ment options are continually being researched and developed. One type of
innovative therapy that demonstrates great promise is the selective targeting
of specific molecular proteins expressed on malignant lymphocytes. Trans-
lational use of these innovative therapies in dogs and cats may provide better
DFIs and survival times than what is currently achievable with existing
chemotherapy.

Recombinant immunotoxins

Lymphocytes express surface glycoproteins that serve as receptors for

soluble or membrane-bound cytokines. Binding of cytokines to their cognate
receptors elicits diverse cellular responses, including proliferation, differen-

464

T.M. Fan

/ Vet Clin Small Anim 33 (2003) 455–471

background image

tiation, and apoptosis [56]. Interleukin-2 (IL-2) is a cytokine essential for the
initiation of a robust immune response. Both B and T lymphocytes express
the IL-2 receptor (IL-2R) on their plasma membrane surface. The receptor is
a heterotrimer consisting of three distinct subunits (a, b, and c). In the
inactive state, the IL-2R is composed of constitutively expressed b and c
subunits. After cellular activation, however, gene transcription with
subsequent protein translation of the a subunit is induced. Concurrent
expression of all three subunits confers high-affinity IL-2 binding [57].

The expression of IL-2R has been identified on malignant T lymphocytes

and can be used as a molecular target for recombinant immunotoxins [58].
Denileukin diftitox (Ontak) is one such molecule, and consists of recom-
binant human IL-2 (rhIL-2) fused to a truncated diphtheria toxin [59]. This
provides a specific delivery system through which diphtheria toxin is delivered
to those malignant lymphocytes expressing the high-affinity IL-2R. Both
intermediate and high-affinity IL-2Rs have been demonstrated in canine T-
cell lymphomas and natural killer cell leukemias [60,61]. The translational
use of recombinant immunotoxins for the management of canine T-cell
malignancies remains to be investigated, and premature use of denileukin
diftitox should be discouraged. Without adequate pharmacokinetic studies
of denileukin diftitox in healthy and tumor-bearing dogs, the safe use of
denileukin diftitox cannot be supported.

Advances in immunotherapy

Monoclonal antibodies provide a method for the accurate delineation of

cell types. In addition to their diagnostic utility, monoclonal antibodies can
be used for the treatment of many cancers, including lymphoid malignancies
[62]. In human cancer patients, rituximab (Rituxan), a monoclonal antibody
recognizing the CD20 surface antigen found on B lymphocytes, is effective
for the treatment of B-lymphocyte malignancies [63]. Rituximab works
by means of several mechanisms, including enhancement of antibody-de-
pendent cellular cytotoxicity and intracellular calcium dysregulation [64,65].
Approximately 25% to 50% of human patients with B-cell malignancies
achieve a therapeutic response to rituximab [66]. Because rituximab’s
cytotoxic properties are partially dependent on host immunity, newer and
more potent monoclonal antibodies conjugated to radionuclides have been
developed. Currently, tositumomab (Bexxar) and ibritumomab tiuxetan
(Zevalin), two recombinant monoclonal antibodies against CD20 conjugated
to radioisotopes, are being evaluated for the treatment of B-lymphocytic
neoplasms in people [62].

Despite the effectiveness of monoclonal-based therapies in people, several

pitfalls prevent their immediate translational use in dogs with B-lymphocyte
malignancies. One question that remains is the level of CD20 in the dog and
cat. Although some investigations support the identification of CD20 in
canine B lymphocytes, the extracellular portion of this surface antigen that

465

T.M. Fan

/ Vet Clin Small Anim 33 (2003) 455–471

background image

is responsible for monoclonal antibody binding has not been identified on
malignant canine B lymphocytes [67]. Even if CD20 were expressed on
canine B-cell lymphomas, subtle conformational differences between the
human and canine CD20 recognition epitope could have a negative impact
on monoclonal antibody binding.

In addition to questionable CD20 expression, several immunologic

limitations may stand in the way of using commercially available mono-
clonal antibodies. A portion of rituximab’s tumoricidal effect is mediated
by antibody-dependent cellular cytotoxicity (ADCC). The favorable binding
kinetics of human phagocyte F

c

receptors and the F

c

fragment of rituximab

ensure optimal conditions for ADCC. Binding kinetics of canine phagocyte
F

c

receptors for rituximab may be of lower affinity, resulting in an at-

tenuated ADCC response. Furthermore, because rituximab, tositumomab,
and ibritumomab tiuxetan are either humanized or chimeric proteins, their
prolonged use in the dog may result in the formation of neutralizing canine
antihuman antibodies. The presence of these neutralizing titers would limit
the use of humanized or chimeric monoclonal antibodies for the long-term
management of canine lymphoma.

New developments in feline lymphoma

Although significant advances have been made in canine lymphoma, the

amount of information available for the diagnosis, prognosis, and treatment
of feline lymphoma remains disappointing. Even with aggressive chemo-
therapy, response rates and survival times in cats with lymphoma continue
to be inferior to results observed in the dog. In part, the lack of new
discoveries could be attributed to the decreased incidence of virally induced
lymphomas that resulted from the wide use of preventive FeLV vaccines.
Regardless, feline lymphoma remains a common and fatal hematopoietic
cancer. Continued research and investigation into the molecular under-
pinnings of feline lymphoma are required to achieve improved response
rates and survival times in affected cats.

Newly identified risk factors

Chemical carcinogens present in cigarette smoke have been causatively

linked to the development of various cancers in people and domestic animals
[68–70]. In addition, an increased incidence of lung and nasal sinus cancers in
dogs has been weakly correlated with exposure to passive cigarette smoke
[71,72]. Recently, an epidemiologic study has demonstrated that cats exposed
to household environmental tobacco smoke have an increased relative risk of
developing malignant lymphoma. The relative risk of developing malignant
lymphoma increased with duration and quantity of environmental tobacco
smoke exposure [73].

466

T.M. Fan

/ Vet Clin Small Anim 33 (2003) 455–471

background image

Inhibitor molecule p27Kip1 dysregulation

The cell cycle consists of four discrete phases G

1

, S, G

2

, and M. The

assembly of specific cyclins and their respective cyclin-dependent kinases
(Cdks) regulates orderly progression through each phase of the cell cycle [73].
Advancement of cells from G

1

to S requires G

1

cyclin

/Cdk complex for-

mation as well as passage through a restriction point called the G

1

check-

point. The G

1

checkpoint ensures that favorable conditions are available for

DNA replication. In normal cells, DNA damage, nutrient deficiency, and
other environmental stressors may prohibit cells from passing through the
G

1

checkpoint, resulting in cell cycle arrest [74]. The induction of cell cycle

arrest serves a protective function, allowing damaged DNA to be repaired
before DNA synthesis. The expression of an inhibitory protein class called
the Cip1

/Kip1 family promotes G

1

cell cycle arrest. Members of the

Cip1

/Kip1 family include p21, p27, and p57 [74]. Decreased expression of the

Cip1

/Kip1 inhibitory family may allow damaged cells to bypass the G

1

checkpoint, thus favoring the propagation of malignantly transformed cells.
In feline lymphoma, the expression of p27Kip1 has been investigated. The
expression of p27Kip1 was demonstrated by an immunohistochemical
method in normal lymphocytes of the follicular mantle zone as well as in
interfollicular small lymphocytes. Actively replicating lymphoblasts within
germinal centers did not stain for p27Kip1. Interestingly, feline T- and B-cell
lymphomas also failed to be immunolabeled. The results of this investigation
suggest that feline malignant lymphocytes may be able to bypass regulatory
checkpoints within the cell cycle [75].

Summary

Canine and feline lymphoma is a common hematopoietic malignancy that

generally responds well to systemic chemotherapy. In dogs, several recent
investigations have underscored the beneficial effects of adjunctive radiation
therapy for the treatment of multicentric lymphoma. With the emergence of
effective immunotherapeutic agents against non-Hodgkin’s lymphoma in
people, some of these specific targeted immunotherapeutics may soon be
a viable option for treating lymphoid malignancies in dogs. Although the
effective and durable treatment of feline lymphoma remains disappointing,
the identification of environmental etiologic factors may help to shape future
recommendations for disease prevention. It is only reasonable to assume that
as our fundamental understanding of lymphoid malignancies grows, better
diagnostic tools, predictive markers, and therapeutic options will also emerge.

References

[1] Vail DM, MacEwen GE, Young KM. Canine lymphoma and lymphoid leukemias. In:

Withrow S, MacEwen GE, editors. Small animal clinical oncology. 3rd edition.
Philadelphia: WB Saunders; 2001. p. 558–90.

467

T.M. Fan

/ Vet Clin Small Anim 33 (2003) 455–471

background image

[2] Dorn CR, Taylor D, Schneider R. The epidemiology of canine leukemia and lymphoma.

Bibl Haematol 1970:403–15.

[3] Chapman AL, Bopp WJ, Brightwell AS, et al. Preliminary report on virus-like particles in

canine leukemia and derived cell cultures. Cancer Res 1967;27:18–25.

[4] Hahn KA, Richardson RC, Hahn EA, et al. Diagnostic and prognostic importance of

chromosomal aberrations identified in 61 dogs with lymphosarcoma. Vet Pathol 1994;31:
528–40.

[5] Hayes HM, Tarone RE, Cantor KP. On the association between canine malignant

lymphoma and opportunity for exposure to 2,4-dichlorophenoxyacetic acid. Environ Res
1995;70:119–25.

[6] Hayes HM, Tarone RE, Cantor KP, Jessen CR, et al. Case-control study of canine

malignant lymphoma: positive association with dog owner’s use of 2,4-dichlorophenoxy-
acetic acid herbicides. J Natl Cancer Inst 1991;3:1226–31.

[7] Owen LN, Bostock DE, Halliwell REW. Cell-mediated and humoral immunity in dogs

with spontaneous lymphosarcoma. Eur J Cancer 1975;11:187–91.

[8] Reif JS, Lower KS, Ogilvie GK. Residential exposure to magnetic fields and risk of canine

lymphoma. Am J Epidemiol 1995;141:352–9.

[9] Francis DP, Cotter SM, Hardy WD, et al. Comparison of virus-positive and virus-negative

cases of feline leukemia and lymphoma. Cancer Res 1979;39:3866–70.

[10] Hardy ED Jr, McClelland AJ, Zuckerman EE, et al. Development of virus non-producer

lymphosarcomas in pet cats exposed to FeLV. Nature 1980;288:90–2.

[11] Essex M, Francis DP. The risk to humans from malignant diseases of their pets: an

unsettled issue. J Am Anim Hosp Assoc 1976;12:386–90.

[12] Vail DM, Moore AS, Ogilvie GK, et al. Feline lymphoma (145 cases): proliferation indices,

cluster of differentiation 3 immunoreactivity, and their association with prognosis in 90
cats. J Vet Intern Med 1998;12:349–54.

[13] Teske E, van Heerde P, Rutteman GR, et al. Prognostic factors for treatment of malignant

lymphoma in dogs. JAVMA 1994;205:1722–8.

[14] Greenlee PG, Filippa DA, Quimby FW, et al. Lymphomas in dogs. A morphologic,

immunologic, and clinical study. Cancer 1990;66:480–90.

[15] Ruslander DA, Gebhard DH, Tompkins MB, et al. Immunophenotypic characterization of

canine lymphoproliferative disorders. In Vivo 1997;11:169–72.

[16] Morrison S, Neuberger MS. Antigen recognition by B-cell and T-cell receptors. In:

Janeway CA, Traver P, Walport M, Shlomchik M, editors. Immunobiology: the immune
system in health and disease. 5th edition. New York: Garland Publishing; 2001. p. 92–184.

[17] Keller ET, MacEwen EG, Rosenthal RC, et al. Evaluation of prognostic factors and

sequential combination chemotherapy with doxorubicin for canine lymphoma. J Vet Intern
Med 1993;7:289–95.

[18] Khanna C, Lund EM, Redic KA, et al. Randomized controlled trial of doxorubicin versus

dactinomycin in a multiagent protocol for treatment of dogs with malignant lymphoma.
JAVMA 1998;213:985–90.

[19] Jeglum KA, Whereat A, Young K. Chemotherapy of lymphoma in 75 cats. JAVMA 1987;

190:174–8.

[20] Mooney SC, Hayes AA, MacEwen EG, et al. Treatment and prognostic factors in

lymphoma in cats: 103 cases (1977–1981). JAVMA 1989;194:696–702.

[21] Moore AS, Cotter SM, Frimberger AE, et al. A comparison of doxorubicin and COP for

maintenance of remission in cats with lymphoma. J Vet Intern Med 1996;10:372–5.

[22] Fearon ER, Hamilton SR, Vogelstein B. Clonal analysis of human colorectal tumors.

Science 1987;238:193–7.

[23] Najfeld V. Clonal origin of leukemia—revisited. A tribute to Philip J Fialkow, MD.

Leukemia 1998;12:106–7.

[24] Wilson IA, Davis MM, Allen PM, Trowsdale J. The generation of lymphocyte antigen

receptors. In: Janeway CA, Traver P, Walport M, Shlomchik M, editors. Immunobiology:

468

T.M. Fan

/ Vet Clin Small Anim 33 (2003) 455–471

background image

the immune system in health and disease. 5th edition. New York: Garland Publishing;
2001. p. 92–184.

[25] Burnett RC, Avery AC. Molecular aspects of canine lymphoma: PCR for antigen receptor

rearrangements (PARR). In: Proceedings of the 19th Annual Veterinary Medical Forum,
Chicago. Lakewood (CO): American College of Veterinary Internal Medicine; 2001.
p. 447–8.

[26] van Steensel B, Smogorzewska A, deLange T. TRF2 protects human telomeres from end-

to-end fusion. Cell 1998;92:401–3.

[27] Shay JW, Brasiskyte D, Ouelette M. Analysis of telomerase and telomeres. Methods Mol

Gen 1994;5:263–80.

[28] Mu J, Wei LX. Telomere and telomerase in oncology. Cell Res 2002;12:1–7.
[29] Carioto LM, Kruth SA, Betts DH, et al. Telomerase activity in clinically normal dogs and

dogs with malignant lymphoma. Am J Vet Res 2001;62:1442–6.

[30] Hipple AK, Colitz CMH, Mauldin GN, et al. Telomerase activity, telomere lengths and

related immunohistochemical properties of normal canine lymph node and canine lym-
phoma. In: Proceedings of the 21st Annual Veterinary Cancer Society Conference, Baton
Rouge. Spring Valley (CA): Veterinary Cancer Society; 2001. p. 23.

[31] Balkin RG, Kitchell BE, Cadile C, et al. Telomerase expression and telomere length in

canine lymphoma. In: Proceedings of the 21st Annual Veterinary Cancer Society
Conference, Baton Rouge. Spring Valley (CA): Veterinary Cancer Society; 2001. p. 65.

[32] Lefkovits I, Vella AT. Innate immunity. In: Janeway CA, Traver P, Walport M, Shlomchik

M, editors. Immunobiology: the immune system in health and disease. 5th edition. New
York: Garland Publishing; 2001. p. 35–92.

[33] Hahn KA, Freeman KP, Barnhill MA, et al. Serum alpha 1-acid glycoprotein concen-

trations before and after relapse in dogs with lymphoma treated with doxorubicin. JAVMA
1999;214:1023–5.

[34] Hanemaaijer R, Verheijen JH, Maquire TM, et al. Increased gelatinase-A and gelatinase-B

activities in malignant versus benign tumors. Int J Cancer 2000;86:204–7.

[35] Bianco FJ, Gervasi DC, Tiguert R, et al. Matrix metalloproteinase-9 expression in bladder

washes from bladder cancer patients predicts pathological stage and grade. Clin Cancer
Res 1998;4:3011–6.

[36] Sehgal G, Hua J, Bernhard EF, et al. Requirement for matrix metalloproteinase-9

(gelatinase B) expression in metastasis by murine prostate carcinoma. Am J Pathol
1998;152:591–6.

[37] Lana SE, Ogilvie GK, Hansen RA, et al. Identification of matrix metalloproteinases in

canine neoplastic tissue. Am J Vet Res 2000;61:111–4.

[38] Lana SE, Oglivie GK, Hansen RA, et al. Matrix metalloproteinase levels as a predicator of

early treatment failure in dogs with lymphoma. In: Proceedings of the 19th Annual
Veterinary Cancer Society Conference, Wood’s Hole. Spring Valley (CA): Veterinary
Cancer Society; 1999. p. 47

[39] Liebman N, Lana SE, Hansen RA, et al. Identification of matrix metalloproteinases in

canine mast cell tumors. J Vet Intern Med 2000;14:583–6.

[40] Scotto KW, Bertino JR. Molecular basis of cancer therapy: natural and acquired resistance

to chemotherapeutic agents. Cancer. In: Mendelsohn J, Howley PM, Israel MA, Liotta
LA, editors. The molecular basis of cancer. 2nd edition. Philadelphia: WB Saunders; 2001.
p. 407–19.

[41] Lee JJ, Hughes CS, Fine RL, et al. P-glycoprotein expression in canine lymphoma:

a relevant, intermediate model of multidrug resistance. Cancer 1996;77:1892–8.

[42] Moore AS, Leveille CR, Reimann KA, et al. The expression of P-glycoprotein in canine

lymphoma and its association with multidrug resistance. Cancer Invest 1995;13:475–9.

[43] Bergman PJ, Ogilvie GK, Powers BE. Monoclonal antibody C219 immunohistochemistry

against P-glycoprotein: sequential analysis and predictive ability in dogs with lymphoma.
J Vet Intern Med 1996;10:354–9.

469

T.M. Fan

/ Vet Clin Small Anim 33 (2003) 455–471

background image

[44] Hahn KA, Barnhill MA, Freeman KP, et al. Detection and clinical significance of plasma

glutathione-S-transferases in dogs with lymphoma. In Vivo 1999;13:173–5.

[45] Chun R, Garrett LD, Vail DM. Evaluation of a high-dose chemotherapy protocol with no

maintenance therapy for dogs with lymphoma. J Vet Intern Med 2000;14:120–4.

[46] Garrett LD, Thamm DH, Chun R, et al. Evaluation of a six month protocol with no

maintenance therapy for dogs with lymphoma. J Vet Intern Med 2002;16:704–9.

[47] Baxter GD, Lavin MF. Specific protein dephosphorylation in apoptosis induced by ioniz-

ing radiation and heat shock in human lymphoid tumor lines. J Immunol 1992;148:
1949–54.

[48] Fornace AJ, Fuks XZ, Weichselbaum RR, et al. Molecular basis of cancer therapy:

radiation therapy. Cancer. In: Mendelsohn J, Howley PM, Israel MA, Liotta LA,
editors. The molecular basis of cancer. 2nd edition. Philadelphia: WB Saunders; 2001.
p. 423–54.

[49] Frimberger AE, Ruslander DM, Moore AS, et al. Low-dose whole body irradiation

for dogs with chemoresistant lymphoma relapse. In: Proceedings of the 20th Annual
Veterinary Cancer Society Conference, Asilomar. Spring Valley (CA): Veterinary Cancer
Society; 2000. p. 28.

[50] Abrams-Ogg ACG, Norris AM, Woods JP, et al. Half-body radiation therapy versus

maintenance chemotherapy for the treatment of dogs with multicentric lymphoma in
remission. In: 19th Annual Veterinary Cancer Society Conference, WoodÕs Hole. Spring
Valley (CA): Veterinary Cancer Society; 1999. p. 8.

[51] Williams LE, Hauck ML, Ruslander DM, et al. Half-body radiation therapy following

induction chemotherapy for the treatment of canine lymphoma. In: Proceedings of the
Second Annual Mid-Year Veterinary Cancer Society Conference, Galena. Spring Valley
(CA): Veterinary Cancer Society; 2002. p. 10.

[52] Moore AS, London CA, Wood CA, et al. Lomustine (CCNU) for the treatment of

resistant lymphoma in dogs. J Vet Intern Med 1999;13:395–8.

[53] Fan TM, Kitchell BE, Dhaliwal RS, et al. Hematological toxicity and therapeutic efficacy

of lomustine in 20 tumor-bearing cats: critical assessment of a practical dosing regimen.
J Am Anim Hosp Assoc 2002;38:357–63.

[54] Rassnick KM, Gieger TL, Williams LE, et al. Phase I evaluation of CCNU (lomustine) in

tumor-bearing cats. J Vet Intern Med 2001;15:196–9.

[55] Hahn KA. Radiation therapy for drug resistant lymphoma. In: Proceedings of the 20th

Annual Veterinary Medical Forum, Dallas. Lakewood (CO): American College of Vet-
erinary Internal Medicine; 2002. p. 417–8.

[56] Hardy RR, Cooper MD, Nemazee D, et al. Signaling through immune system receptors.

In: Janeway CA, Traver P, Walport M, Shlomchik M, editors. Immunobiology: the
immune system in health and disease. 5th edition. New York: Garland Publishing; 2001.
p. 187–220.

[57] Paulnock D, Springer T, Jenkins MK, et al. T cell-mediated immunity: the production of

armed effector T cells. In: Janeway CA, Traver P, Walport M, Shlomchik M, editors.
Immunobiology: the immune system in health and disease. 5th edition. New York: Garland
Publishing; 2001. p. 295–340.

[58] Kreitman RJ. Immunotoxins. Expert Opin Pharmacother 2000;1:1117–29.
[59] Foss FM. DAB(389)IL-2 (denileukin diftitox, ONTAK): a new fusion protein technology.

Clin Lymphoma Suppl 2000;1:S27–31.

[60] Helfand SC, Modiano JF, Moore PF, et al. Functional interleukin-2 receptors are ex-

pressed on natural killer-like leukemic cells from a dog with cutaneous lymphoma. Blood
1995;86:636–45.

[61] Helfand SC, Dickerson EB, Modiano JF, et al. The interleukin-2 receptor as a potential

target in canine lymphoid malignancy. In: Proceedings of the 19th Annual Veterinary
Cancer Society Conference, WoodÕs Hole. Spring Valley (CA): Veterinary Cancer Society;
1999. p. 8.

470

T.M. Fan

/ Vet Clin Small Anim 33 (2003) 455–471

background image

[62] Maloney DG. Advances in immunotherapy of hematologic malignancies. Curr Opin

Hematol 1998;5:237–43.

[63] Grillo-Lopez AJ. AntiCD20 mAbs: modifying therapeutic strategies and outcomes in the

treatment of lymphoma patients. Expert Rev Anticancer Ther 2002;2:323–9.

[64] Shan D, Ledbetter JA, Press OW. Apoptosis of malignant human B cells by ligation of

CD20 with monoclonal antibodies. Blood 1998;91:1644–52.

[65] Shan D, Ledbetter JA, Press OW. Signaling events involved in anti-CD20-induced

apoptosis of malignant human B cells. Cancer Immunol Immunother 2000;48:673–83.

[66] Maloney DG, Liles TM, Czerwinski DK, et al. Phase I clinical trial using escalating single-

dose infusion of chimeric anti-CD20 monoclonal antibody (IDEC-C2B8) in patients with
recurrent B-cell lymphoma. Blood 1994;84:2457–66.

[67] Darbes J, Majzoub M, Hermanns W. Evaluation of the cross-reactivity between human

and feline or canine leucocyte antigens using commercially available antibodies. J Vet
Diagn Invest 1997;9:94–7.

[68] Hoffmann D, Brunnemann KD, Rivenson A, et al. N-nitrosodiethanolamine: analysis,

formation in tobacco products and carcinogenicity in Syrian golden hamsters. IARC Sci
Publ 1982;41:299–308.

[69] Stuart BO, Willard DH, Howard EB. Studies of inhaled radon daughters, uranium ore

dust, diesel exhaust, and cigarette smoke in dogs and hamsters. Inhaled Particles 1970;
1:543–60.

[70] Weisburger JH, Wynder EL. The role of genotoxic carcinogens and of promoters in carcino-

genesis and in human cancer causation. Acta Pharmacol Toxicol (Copenh) 1984;
55(Suppl 2):53–68.

[71] Reif JS, Dunn K, Ogilvie GK, et al. Passive smoking and canine lung cancer risk. Am J

Epidemiol 1992;135:234–9.

[72] Reif JS, Bruns C, Lower KS. Cancer of the nasal cavity and paranasal sinuses and exposure

to environmental tobacco smoke in pet dogs. Am J Epidemiol 1998;147:488–92.

[73] Bertone ER, Snyder LA, Moore AS. Environmental tobacco smoke and risk of malignant

lymphoma in pet cats. Am J Epidemiol 2002;156:268–73.

[74] Murakami MS, Vande Woude GF. Malignant transformation: regulation of the cell cycle.

In: Mendelsohn J, Howley PM, Israel MA, Liotta LA, editors. The molecular basis of
cancer. 2nd edition. Philadelphia: WB Saunders; 2001. p. 10–7.

[75] Madewell B, Griffey S, Walls J, et al. Reduced expression of cyclin-dependent kinase

inhibitor p27Kip1 in feline lymphoma. Vet Pathol 2001;38:698–702.

471

T.M. Fan

/ Vet Clin Small Anim 33 (2003) 455–471

background image

Mast cell tumors in the dog

Cheryl A. London, DVM, PhD*,

Bernard Seguin, DVM, MS

Department of Surgical and Radiological Sciences, 2112 Tupper Hall, One Shields Avenue,

University of California at Davis, Davis, CA 95616, USA

In most species, neoplastic processes involving mast cells are relatively

uncommon. In contrast, mast cell tumors (MCTs) occur frequently in the pet
population, representing up to 20% of all cutaneous canine tumors [1–3]. As
such, a detailed comprehension of the diagnosis and treatment of mast cell
neoplasia in the dog is extremely important. Before discussing the specifics of
MCTs, however, it is first necessary to understand normal mast cell biology.

Mast cell biology

Morphologic characteristics of mast cells

Mast cells are discrete round cells roughly one to three times the size of

a neutrophil. They possess a round to oval nucleus and distinct cytoplasmic
granules that stain with dyes, such as toluidine blue, Giemsa, and methylene
blue. Such staining is a result of the affinity of these basic dyes for the acidic
proteoglycans contained in the mast cell granules [4–7]. Some of these dyes
assume a different color when bound by the granules than they do when
staining nuclear DNA, so that the granules are often called metachromatic.
Although mast cells may be visualized on hematoxylin and eosin–stained
sections, the dyes described previously are used to characterize mast cells
definitively. Moreover, mast cell granules may not be visualized with stains
like Diff-Quick (Dade Behring, Deerfield, IL), precluding identification on
some cytologic specimens [8].

Development of mast cells

Mast cells are derived from bone marrow precursor cells. They leave

the bone marrow in a presumed immature state and migrate to many

Vet Clin Small Anim

33 (2003) 473–489

* Correspondence.
E-mail address:

calondon@ucdavis.edu (C.A. London).

0195-5616

/03/$ - see front matter

Ó 2003, Elsevier Inc. All rights reserved.

doi:10.1016/S0195-5616(03)00003-2

background image

tissues, particularly those having primary contact with foreign antigens
(eg, skin, respiratory and gastrointestinal tracts), where they then mature
into tissue mast cells [5]. Cytokines important in the development and
maturation of mast cells include interleukin (IL)-3, IL-6, IL-4, and stem
cell factor (SCF) among others [5]. The local tissue microenvironment
in which mast cells mature determines the subsequent functional ca-
pacities of these cells. For example, mast cells in the mucosa of the
gastrointestinal tract (labeled mucosal mast cells) have chondroitin sul-
fate as their major granule proteoglycan and contain little histamine. In
contrast, mast cells in the lung and serosa of body cavities (labeled tissue
mast cells) contain heparin as their major granule proteoglycan and pro-
duce large amounts of histamine [4]. Experiments in mice suggest that the
functional characteristics of mast cells are not fixed, because granule
content can change if mast cells are moved from one environment to
another. In summary, the precise nature of the mast cell and the mediators
it can produce varies with its anatomic location and is probably regulated
by the local cytokine environment.

Function of mast cells

Mature mast cells bind IgE on their cell surface through expression of the

high-affinity IgE receptor (FceRI). Mast cells also express receptors for
complement components (particularly C5a). The primary manner in which
mast cells are activated is by cross-linking of the FceRI-bound IgE on their
cell surface, leading to the release and production of various mediators
including the following [4,5]:

a. Contents of granules, such as histamine, heparin, chondroitin sulfate,

mast cell proteases, and others

b. Lipid mediators, such as prostaglandins, leukotrienes, and platelet-

activating factor

c. Cytokines, such as tumor necrosis factor-a, IL-3, IL-4, IL-5, and IL-6

The mediators described here lead to several reactions, including in-

creased vascular permeability, vasodilation, smooth muscle spasm, pruritus,
anticoagulation, and activation of eosinophils and neutrophils. Collectively,
these effects can lead to local hypersensitivity reactions, or more seriously,
to systemic hypersensitivity (anaphylactic shock) [4]. As such, mast cells
have primarily been associated with allergic reactions

/disorders. It is now

evident that mast cells seem to play an important role in the initiation of
innate immune responses. Experiments in mice demonstrated that mast cells
were important in initiating and sustaining neutrophil migration and ac-
tivation in response to bacteria [6]. Indeed, mast cells are often seen at sites
of inflammation as well as in reactive lymph nodes. Therefore, in addi-
tion to playing a role in the induction of pathologic allergic responses, mast
cells are critical players in protective immune responses.

474

C.A. London, B. Seguin

/ Vet Clin Small Anim 33 (2003) 473–489

background image

Canine mast cell tumors

Incidence and signalment

As previously mentioned, the MCT is the most common skin tumor of the

dog and one of the most common malignant tumors noted in the canine
population. Whereas MCTs are usually found in older dogs (mean age of
approximately 8–9 years), they have also been reported in younger dogs [9–
12]. Several breeds seem to be at increased risk for the development of MCTs,
including dogs of bulldog descent (Boxer, Boston Terrier, and English
Bulldog), Labrador and Golden Retrievers, Cocker Spaniels, Schnauzers, and
Chinese Sharpeis [9,12–14]. No sex predilection has been reported.

Etiology

The etiopathogenesis of MCTs in the dog is unknown, as is the reason for

the extremely high incidence in this species. Although some studies have
suggested the possibility of a viral cause and MCTs have been transmitted
from dogs with solid tumors to susceptible laboratory dogs using tumor
tissue or extracts, there is no epidemiologic evidence to indicate horizontal
transmission of tumors [15]. Because most of the tumors arise in the skin, it
has been suggested that topical carcinogens may play a role in this disease.
No reports exist to imply such a cause, however, and there seems to be no
particular regional distribution of these tumors. The increased incidence of
MCTs in certain breeds suggests the possibility of an underlying genetic
cause [13]. Interestingly, although dogs of bulldog ancestry are at higher risk
for MCT development, it is generally accepted that MCTs in these dogs are
more likely to be benign [12].

As previously mentioned, SCF is an important growth factor for mast cells

[7]. The receptor for SCF is Kit, encoded by the proto-oncogene c-kit; SCF-Kit
interactions are required for the differentiation, survival, and function of mast
cells [7,16–21]. Mutations in c-kit leading to activation of Kit in the absence of
SCF binding have been demonstrated to occur in systemic mastocytosis in
people [22–28]. Additionally, Kit dysfunction has now been recognized in
a variety of other human cancers, including gastrointestinal stromal tumors,
small cell lung cancer, ovarian carcinoma, and gliomas [29–36]. Several
authors have recently identified the presence of activating mutations in the
proto-oncogene c-kit in canine MCTs. These mutations consist of internal
tandem duplications in the negative regulatory juxtamembrane domain of Kit
[37–40]. The high frequency of mutations in a gene known to play a role in
tumorigenesis suggests that aberrations in c-kit may be involved in the
development or progression of MCTs in dogs.

History and clinical signs

Although normal mast cells are found in abundance in the lungs and

gastrointestinal tract, the overwhelming majority of MCTs in the dog occur

475

C.A. London, B. Seguin

/ Vet Clin Small Anim 33 (2003) 473–489

background image

in the dermis and subcutaneous tissue [9,41]. Rarely, primary MCTs may
present in other sites, such as the oral cavity, nasopharynx, larynx, and
gastrointestinal tract [42,43]. Visceral MCT involving the spleen, liver, or
bone marrow (often referred to as disseminated mastocytosis) is usually
the result of systemic spread of an aggressive primary cutaneous MCT,
although it can occur as an independent syndrome [44,45]. Primary mast cell
leukemia in the dog is extremely rare.

Cutaneous MCTs usually occur as solitary nodules, although roughly

10% to 15% of dogs present with multiple tumors [46]. Approximately
50% of cutaneous MCTs occur on the trunk and perineal region, 40%
on the limbs, and 10% on the head and neck [2,46,47]. Perhaps most
importantly, the clinical appearance of MCTs can vary widely. Dermal
MCT may be well circumscribed, raised, and firm, or the surface maybe
erythematous and ulcerated; invasion into the subcutaneous tissue may be
present. MCTs arising in the subcutaneous tissue are often poorly cir-
cumscribed and may resemble lipomas. It is therefore not possible to
identify a cutaneous lesion as an MCT simply by its appearance.
Cutaneous MCTs may also be present for various lengths of time. In
general, MCTs that are slow growing and present for at least 6 months are
more likely to behave in a benign manner, whereas those that are rapidly
growing large tumors are more likely to behave in a malignant manner
[41]. The duration of the lesion does not always predict the subsequent
biologic behavior, however.

Clinical signs of MCTs are caused by the release of histamine, heparin,

and other vasoactive amines. Mechanical manipulation of the tumor during
physical examination can induce degranulation leading to erythema and
wheal formation (termed Darrier’s sign), and an owner occasionally reports
that the tumor seems to change in size over short periods [47]. Gastro-
intestinal ulceration is also a potential complication of MCTs; between 35%
and 83% of dogs with MCTs that underwent necropsy had evidence of
gastric ulcers, and plasma histamine concentrations were found to be ele-
vated in dogs with MCT [48,49]. Elevated histamine levels presumably lead
to stimulation of H2 receptors on parietal cells, excessive gastric acid pro-
duction, and the development of ulcers. Consequently, signs like vomiting,
anorexia, melena, and abdominal pain may be present. In one study, the
levels of histamine in dogs with MCTs were not related to clinical stage, his-
tologic grade, or tumor size [48].

Diagnosis

Cytologic evaluation of fine-needle aspirates is probably the easiest

method to diagnose the presence of an MCT. The mast cells appear as
discrete round cells with a round to oval centrally placed nucleus that may
be difficult to visualize because of the presence of granules. As mentioned
previously, mast cell granules may not stain with Diff-Quik, leading to

476

C.A. London, B. Seguin

/ Vet Clin Small Anim 33 (2003) 473–489

background image

difficulty in making a definitive diagnosis by cytology. Additionally, poorly
differentiated malignant mast cells may contain few, if any, granules; in this
case, special stains (eg, toluidine blue, Giemsa) may be required. Other
round cell tumors that should be included in the differential diagnosis in-
clude lymphoma, plasmacytoma, histiocytoma, amelanotic melanoma, and
transmissible venereal tumor. Whereas the diagnosis of MCT can almost
always be made by fine-needle aspiration cytology, excisional biopsy is
required for histologic grading of the tumor. It is important to note that
because wide surgical excision is the treatment most likely to cure most
MCTs, every effort should be made to obtain a definitive diagnosis via
cytology before surgical intervention. Moreover, because any cutaneous
tumor may potentially be an MCT, fine-needle aspiration should be
performed on all masses before removal. If cytologic diagnosis proves
difficult, a needle or punch biopsy of the tumor can be obtained before
surgery. This may be preferable to a larger incisional biopsy, because local
release of mast cell mediators will significantly inhibit healing and may result
in excessive bleeding.

Staging

Because any MCT is capable of metastasis, all dogs with MCTs should be

staged to determine the extent of their disease and overall health. This is
particularly important for dogs in which radiation therapy may be used in
the course of treatment. Some of or all the following procedures may be
indicated.

Complete blood cell count, biochemistry profile, urinalysis

These tests are part of a minimum database and should be included in the

workup of any animal suspected to have cancer. Dogs with MCTs may have
eosinophilia because of chemotactic factors and IL-5 produced by the mast
cells [4]. Anemia may be present secondary to hypersplenism or gastroin-
testinal bleeding. Occasionally, mast cells may be seen on a routine complete
blood cell count.

Buffy coat smear

A buffy coat smear is made by spinning peripheral blood in a micro-

hematocrit tube, breaking the tube at the buffy coat layer, and smearing the
concentrated leukocytes on a slide. The buffy coat is then carefully examined
for the presence of mast cells. For many years, examination of the buffy
coat for circulating mast cells has been used to screen dogs with MCTs for
the presence of systemic

/metastatic disease. It was originally believed that al-

though the buffy coat smear was not an extremely sensitive test, it was fairly
specific for mast cell neoplasia. It is now clear that this may not be the case,

477

C.A. London, B. Seguin

/ Vet Clin Small Anim 33 (2003) 473–489

background image

however, because several studies have demonstrated that dogs with many
different kinds of disease, including pneumonia, parvovirus, pancreatitis,
skin disease, and gastrointestinal diseases, may have mast cells circulating
in the periphery [50–52]. In summary, the buffy coat smear is a relatively
easy test to perform; the finding of circulating mast cells in a patient
with cutaneous MCT warrants further investigation.

Bone marrow aspiration

In the normal bone marrow, mast cells are found infrequently. In one

study, of 51 bone marrow samples examined, only 2 contained a single mast
cell [52]. It should be pointed out that these marrow samples were obtained
from normal dogs, and it is possible that dogs with inflammatory disorders
may exhibit higher numbers of mast cells in the bone marrow [50,51]. In gen-
eral, bone marrow aspiration is considered to be a more sensitive indica-
tor of systemic involvement than the buffy coat smear [45].

Lymph node aspiration

All regional lymph nodes should be carefully examined for signs of en-

largement, and any suspicious nodes should be aspirated for cytologic exam-
ination. Additionally, because metastatic nodes may palpate within normal
size, it is recommended by some that all accessible regional lymph nodes be
examined by aspiration cytology. Mast cells may be present in normal
lymph nodes and are often found in reactive lymph nodes; thus, it may be
difficult to determine if mast cells found on cytologic examination are neo-
plastic or part of the normal immunologic cellular repertoire. Indeed, 24% of
lymph node aspirates from normal Beagles contained mast cells [52]. There-
fore, lymph node biopsy with histopathology may be a better tech-
nique for diagnosis of lymph node metastasis.

Evaluation of the abdominal and thoracic cavities

Thoracic radiographs are always indicated as part of any staging pro-

cedure, although pulmonary involvement is uncommon in dogs with MCTs.
Abnormalities reported include lymphadenopathy (sternal and hilar), pleural
effusion, and anterior mediastinal masses [45]. Evaluation of the abdomi-
nal cavity is important in dogs with MCTs, because spread to the liver and
spleen as well as other abdominal structures may be noted. Ultrasound is a
more sensitive diagnostic technique for evaluation of abdominal organs. Any
suspicious organ should be aspirated and examined by cytology. Some have
recommended that splenic and hepatic aspiration be performed as part of the
routine staging procedure, regardless of the presence of splenomegaly or
hepatomegaly. Because mast cells may be found normally in these organs,
however, the utility of this procedure is not clear. Evidence suggests that fine-
needle aspiration of the liver and spleen should be performed if abnormalities
are detected in these organs during ultrasound examination or if the dog is
known to have a grade III MCT [53–55].

478

C.A. London, B. Seguin

/ Vet Clin Small Anim 33 (2003) 473–489

background image

Clinical staging system for canine mast cell tumors

The following is the current clinical staging system for canine mast cell

tumors:

0: single tumor, incompletely excised from dermis

I: single tumor confined to dermis without regional lymph node

involvement

II: single tumor confined to the dermis with regional lymph node

involvement

III: multiple dermal tumors or large infiltrating tumors, with or without

regional lymph node involvement

IV: any tumor with distant metastases or recurrence with metastases

(including blood or bone marrow involvement
substage a: no signs of systemic disease
substage b: signs of systemic disease

Prognostic factors

Canine MCTs possess a wide range of biologic behaviors ranging from

benign to extremely aggressive leading to metastasis and eventual death from
disease. Several prognostic factors have been identified that help to predict
the biologic behavior of an MCT as well as to direct the course of therapy.

Histologic grade

The histologic grade of an MCT is determined after incisional or

excisional biopsy of the tumor and cannot be assessed simply by cytologic
evaluation of fine-needle aspirates. The grade of an MCT is determined by
the characteristics of the neoplastic cells (eg, degree of granulation, cytologic
and nuclear pleomorphism), number of mitotic figures, and extent of tumor
invasion into the underlying tissues (Table 1). Histologic grade is the most
consistent prognostic factor and correlates significantly with survival, but it
does not predict the behavior of every tumor. The most commonly used
grading system is the one described by Patnaik et al [14]:

i. Well-differentiated tumors (grade I) are considered to behave in benign

manner, and complete surgical excision is usually curative. These
represent between 30% and 55% of all MCTs reported [14,41,46,56].
Several retrospective studies have demonstrated that 75% to 90% of
dogs do not die of their disease after definitive therapy [14,41,56,57].

ii. Intermediately differentiated tumors (grade II) represent between

25% and 45% of all MCTs reported, and their biologic behavior is
more difficult to predict [14,41,46,56]. On histopathology, these
tumors exhibit invasion into the underlying subcutaneous tissue. As
a result, they may be more challenging to remove by surgical excision.
Historically, dogs with grade II MCT have a reported mean survival

time of 28 weeks after surgical removal, although the completeness of

479

C.A. London, B. Seguin

/ Vet Clin Small Anim 33 (2003) 473–489

background image

excision could not be assessed in this study [41]. More recently, it has
been shown that radiation therapy after incomplete excision of
solitary grade II MCTs can cure greater than 80% of affected
patients, indicating that adjuvant radiation therapy clearly improves
the survival times of dogs with these tumors [57,58]. It is important to
note that grade II MCTs have the ability to spread to local lymph
nodes as well as distant sites and that a proportion of dogs undergoing
definitive therapy (surgery and radiation) may go on to develop
metastatic disease. Furthermore, some dogs that present with grade II
MCTs already have evidence of metastatic disease, making appropri-
ate staging imperative for these dogs.

iii. Poorly differentiated tumors (grade III) represent between 20% and

40% of all MCTs reported [14,41,46,56]. They often behave in a
biologically aggressive manner, exhibiting metastasis early on in the
course of disease. The mean survival time of dogs with grade III MCT
has been reported as 18 weeks when treated with surgery alone [41]. In
one study, the percentage of dogs with grade III MCTs surviving at
1500 days was reported as 6%, and in another study, the percentage
of dogs surviving at 24 months was 7%, indicating that these tumors
are particularly malignant [14,59].

Table 1
Patnaik scheme for grading of canine mast cell tumors

Grade

Patnaik grade

Microscopic

Well differentiated

I

Well-differentiated mast cells with

clearly defined cytoplasmic borders
with regular spherical, or ovoid
nuclei; mitotic figures are rare or
absent; granules are large, deep
staining, and plentiful; cells confined
to the dermis and interfollicular
spaces

Intermediately differentiated

II

Cells closely packed with indistinct

cytoplasmic boundaries; nuclear

/

cytoplasmic ratio lower than
anaplastic; mitotic figures
infrequent; more granules than
anaplastic; neoplastic cells infiltrate
or replace the lower dermal and
subcutaneous tissue

Anaplastic undifferentiated

III

Highly cellular, undifferentiated

cytoplasmic boundaries, with
irregular size and shape of the
nuclei; frequent mitotic figures; low
number of cytoplasmic granules;
neoplastic tissue replaces the
subcutaneous and deep tissues

Data from

Patnaik AK, Ehler WJ, MacEwen EG. Canine cutaneous mast cell tumors:

morphologic grading and survival time in 83 dogs. Vet Pathol 1984;21:469–74.

480

C.A. London, B. Seguin

/ Vet Clin Small Anim 33 (2003) 473–489

background image

Clinical stage

Although the clinical staging scheme has been developed for prognostic

purposes, each increase in stage (0–IV) has not necessarily been proven to
worsen the prognosis. It has been demonstrated that when dogs are treated
with radiation therapy, those with stage 0 MCTs survived significantly
longer than did dogs with stage I through III MCTs. In that study, no dog
had stage IV MCT; therefore, stage IV MCT could not be compared for
prognostic purposes [57]. In two other studies, the presence of mast cells in
the regional lymph node was a negative prognostic factor for survival and
disease-free interval on univariate analysis, suggesting that stage II has
a worse prognosis than stage I [60,61]. Dogs with stage III MCT with the
presence of multiple dermal masses may not necessarily have a worse prog-
nosis than dogs with stage I or II MCT. Indeed, in one study, dogs with mul-
tiple MCTs (stage III) did not have a worse prognosis than dogs with a
single MCT when treated with chemotherapy [60].

Anatomic location

MCTs that develop in the oral cavity, nail bed, or inguinal, preputial, and

perineal regions have been reported to behave in a more malignant fashion
regardless of histologic grade [57]. Definitive evidence for this in the liter-
ature is lacking, however. MCTs that originate in the viscera (eg, gastro-
intestinal tract, liver, spleen), bone marrow, or peripheral blood carry a grave
prognosis [45,62].

Growth rate

Tumors present for long periods (months to years) may be more likely to

behave in a benign manner. In one study, 83% of dogs with tumors present
for longer than 28 weeks before surgery survived for at least 30 weeks com-
pared with only 25% of dogs with tumors present for less than 28 weeks [41].
The same study demonstrated that most dogs surviving for longer than 30
weeks after surgery appeared to be cured.

Breed

Boxers have a high incidence of MCTs, but these tend to be more well

differentiated and carry a better prognosis [12,41]. Nevertheless, every MCT
should be treated as potentially malignant, regardless of breed.

Argyrophilic nucleolar straining organizing regions

Argyrophilic nucleolar staining organizing regions (AgNORs) are areas

present in the nucleus that take up silver stain and provide an indirect
measure of cell proliferation. This stain can be performed on formalin-fixed
specimens or on cytologic samples. It has been demonstrated that the rel-
ative frequency of AgNORs correlates with histologic grade and is pre-
dictive of postsurgical outcome. The higher the AgNOR count, the poorer
is the prognosis [56,63,64].

481

C.A. London, B. Seguin

/ Vet Clin Small Anim 33 (2003) 473–489

background image

DNA ploidy

MCTs possessing an abnormal DNA content (aneuploid) exhibited

a trend toward shorter survival times; however, a significant difference was
found between aneuploid and diploid tumors when comparing stage I and
non-stage I disease [65].

Proliferating cell nuclear antigen

Proliferating cell nuclear antigen (PCNA) is a protein required for DNA

synthesis; expression is associated with cell proliferation. In one study,
PCNA was significantly higher in recurrent versus nonrecurrent tumors and
in metastatic versus nonmetastatic tumors, and it was a good predictor for
tumor recurrence at 6 months [56].

Ki-67

Ki-67 is composed of two protein subunits that are present in cells during

the active phases of the cell cycle but absent during rest. Levels of Ki-67 in
the nucleus seem to correlate with cell proliferation. In one study, the mean
number of Ki-67-positive nuclei per 1000 tumor nuclei was significantly
higher for dogs that died of MCTs than for those that survived. For dogs
with grade II tumors, the number of Ki-67-positive nuclei per 1000 tumor
nuclei (<93 versus

93) was significantly associated with outcome [59].

Other factors

Several other factors are currently under investigation to determine if

they may play a role in the aggressiveness of canine MCTs, including the
levels of active matrix metalloproteinase 2 and 9, tryptase, and chymase
[66,67]. Two recent studies evaluated the potential role of p53 in canine
MCTs; both found that although p53 overexpression was noted in some
MCTs, there was no obvious association with tumor behavior [68,69].

Treatment

The choice of treatment modalities used for a particular canine MCT

depends heavily on the prognostic indicators discussed previously, especially
the histologic grade and clinical stage.

Surgery

Wide surgical excision is indicated for all canine MCTs; although they

may feel like discrete masses, microscopically, most extend well beyond the
palpable borders. It is generally accepted that the lateral margins of excision
need to be at least 3 cm in each direction. Deep margins are as important as
the lateral margins and follow the concept of a quality margin as opposed to
a quantity margin. Collagen-dense and vascular-poor tissues tend to behave
as biologic barriers against cancer [70]. Therefore, the deep margin should
include a fascial plane that has not been invaded by the tumor and is
removed en bloc with the tumor. All the excised tissue should be submitted;

482

C.A. London, B. Seguin

/ Vet Clin Small Anim 33 (2003) 473–489

background image

the lateral and deep margins should be accurately labeled so that the
pathologist is able to identify specifically any areas of incomplete excision.
Careful examination of the histologic margins is imperative; however, even
histologically clean margins do not guarantee that a tumor will not recur.
This is particularly relevant for grade II and III tumors in which the
underlying tissues may be involved. Part of the difficulty in evaluating tumor
margins is that normal mast cells are present in all tissues, and it may be
difficult for the pathologist to determine if a mast cell present at the tissue
margin represents a malignant cell or a normal cell. In one study, 83% of
dogs with grade I MCT, 44% of dogs with grade II MCT, and 6% of dogs
with grade III MCT were alive 1500 days after surgical excision [14]. In
another study, 100% of dogs with grade I MCT, 44% of dogs with grade II
MCT, and 7% of dogs with grade III MCT were alive 2 years after surgical
excision [59].

Complete surgical excision is likely to be curative for dogs with grade I

MCT. The need to perform adjuvant therapy for local control of the tumor
in dogs with completely excised grade II MCT seems to be unnecessary. Two
studies have reported a rate of local recurrence of 5% and 11% after
complete excision of grade II MCT [71,72]. Postsurgical treatment rec-
ommendations are as follows:

Grade I complete excision: no further therapy
Grade I incomplete excision: wider excision or radiation therapy if
surgery is not possible
Grade II complete excision: consider radiation therapy only if margins
are close
Grade II incomplete excision: wider excision or radiation therapy if
surgery is not possible
Grade III complete excision: chemotherapy
Grade III incomplete excision: chemotherapy with or without radiation
therapy

It has been advocated that the injection of deionized water in the surgical

site can decrease the likelihood of local recurrence after incomplete MCT
removal [73]. A more recent study found no benefit in this procedure; as
such, the use of deionized water is currently not recommended [74].

Radiation therapy

Evidence suggests that radiation therapy is extremely effective at elim-

inating remaining microscopic disease after incomplete excision of grade I
and II MCT ([90% 3-year control rate) [58,75]. Protocols that deliver the
total dose of radiation in a shorter period by administering more fractions
per week seem to provide better control of the MCTs [61]. Unfortu-
nately, dogs with grade III tumors do not fare as well; whereas the
radiation may be effective at preventing local recurrence of tumor, most dogs
eventually develop metastasis.

483

C.A. London, B. Seguin

/ Vet Clin Small Anim 33 (2003) 473–489

background image

Radiation therapy has been used to treat solid MCTs (macroscopic

disease) when surgery was not an option. Varying degrees of success have
been found; in one study, a 50% 1-year control rate was obtained using total
doses of 45 to 57 Gy [61]. Radiation therapy should not be used as the
primary therapeutic modality if surgical intervention is an option, however.
Palliative radiation has also been used to treat dogs with nonresectable high-
grade MCT. This may result in an improvement in quality of life but is
unlikely to significantly increase survival time. Moreover, systemic effects of
mast cell degranulation after radiation may lead to vomiting, hypotension,
and gastrointestinal ulceration.

Chemotherapy

The use of adjuvant chemotherapy is indicated after excision of grade III

MCTs and metastatic MCTs as well as for nonresectable high-grade tumors.
It is important to note that radiation therapy (not chemotherapy) is the
treatment of choice for incompletely excised grade I and II MCTs. In
general, chemotherapy for macroscopic MCT has been unrewarding, and
long-term responses have not been demonstrated in well-controlled clinical
trials.

i. Corticosteroids: the exact mechanism of how corticosteroids kill

malignant mast cells is not known. The reported response rate of
canine MCT to prednisone is 20%, with reported remission times of
10 to 20 weeks [76]. Partial remissions are more common than
complete remissions, and at least some of the observed response may
be a result of a decrease in tumor-associated edema. Intralesional
corticosteroids have also been shown to be of some benefit.

ii. CCNU (lomustine): a nitrosourea alkylating agent that has been used

to treat lymphoma and brain tumors in dogs, CCNU, was recently
found to have activity against canine MCT. A response rate of ap-
proximately 42% was noted when dogs with grade II and III MCTs
that had failed all other therapies were treated with CCNU [77].
As with prednisone, most of these were partial responses, and the
duration of response was only 79 days. Nevertheless, it does appear
that this drug has some efficacy in treating MCT, and clinical trials
are currently underway to evaluate its efficacy.

iii. Vinblastine: vinblastine has been reported to have efficacy against

canine MCT in two separate studies. In the first study, dogs with
grade II or III MCT with metastasis to the regional lymph node
underwent surgical removal of the primary tumor and involved lymph
node. They were then treated with a combination of vinblastine,
prednisone, and cyclophosphamide, resulting in a median survival
time of 18 months [78]. Interestingly, dogs with grade II or III MCT
and metastasis to the regional lymph node that did not undergo
surgery but were treated with the combination chemotherapy protocol

484

C.A. London, B. Seguin

/ Vet Clin Small Anim 33 (2003) 473–489

background image

lived for a median of 5 months. This would seem to indicate that
vinblastine is more useful in the face of microscopic rather than bulky
MCT. The second study was a retrospective analysis of dogs with
various forms of MCT treated with a combination of vinblastine and
prednisone [60]. This study revealed a 47% response rate, although
the retrospective nature of the analysis makes the data difficult to
interpret. Interestingly, vincristine, a close relative of vinblastine,
appears to have little efficacy in the treatment of canine MCT [79].
Studies are currently underway to further evaluate the utility of
vinblastine in the treatment of canine MCT.

iv. Miscellaneous drugs: in limited clinical reports, both

L

-asparaginase

and chlorambucil have been found to have activity against MCT.

Supportive care

Animals with large primary MCT, evidence of metastatic disease, or

systemic signs should be treated with medications to block some of or all the
effects of histamine release.

a. H2 antagonists: because histamine stimulates gastric acid production

by parietal cells, MCT may cause gastrointestinal ulceration. To pre-
vent this, any of the standard H2 antagonists may be used, including
cimetidine, ranitidine, or famotidine. Alternatively, omeprazole may
be used; this is not a direct H2 antagonist but works by inhibiting
the proton pump on parietal cells necessary for the generation of gas-
tric acid.

b. H1 antagonists: massive mast cell degranulation can lead to hypo-

tensive shock and death. Therefore, patients with bulky mast cell dis-
ease should be placed on an H1 antagonist like diphenhydramine.

c. Miscellaneous: treatment with sucralfate is indicated for dogs with

evidence of gastrointestinal ulceration. Cyproheptadine and antihis-
taminic drugs with antiserotonin activity stabilize mast cells and may be
useful in the treatment of dogs with bulky mast cell disease.

References

[1] Dorn CR, Taylor DO, Schneider R, Hibbard HH, Klauber MR. Survey of animal neo-

plasms in Alameda and Contra Costa Counties, California. II. Cancer morbidity in dogs
and cats from Alameda County. J Natl Cancer Inst 1968;40:307–18.

[2] Cohen D, Reif JS, Brodey RS, Keiser M. Epidemiological analysis of the most prevalent

sites and types of canine neoplasia observed in a veterinary hospital. Cancer Res
1974;34:2859–68.

[3] Priester WA. Skin tumors in domestic animals. Data from 12 US and Canadian colleges of

veterinary medicine. J Natl Cancer Inst 1973;50:457–66.

[4] Abbas AK, Lichtman AH, Pober JS. Cellular and molecular immunology. 4th edition.

Philadelphia: WB Saunders; 2000. p. 235–67.

485

C.A. London, B. Seguin

/ Vet Clin Small Anim 33 (2003) 473–489

background image

[5] Austen KF, Boyce JA. Mast cell lineage development and phenotypic regulation. Leuk Res

2001;25:511–8.

[6] Galli SJ, Maurer M, Lantz CS. Mast cells as sentinels of innate immunity. Curr Opin

Immunol 1999;11:53–9.

[7] Galli SJ, Zsebo KM, Geissler EN. The kit ligand, stem cell factor. Adv Immunol

1994;55:1–96.

[8] Rebar AH, Boon GD, DeNicola DB. A cytologic comparison of Romanowsky stains and

Papanicolaou type stains. II. Cytology of inflammatory and neoplastic lesions. Vet Clin
Pathol 1982;11:16–25.

[9] Rothwell TL, Howlett CR, Middleton DJ, Griffiths DA, Duff BC. Skin neoplasms of dogs

in Sydney. Aust Vet J 1987;64:161–4.

[10] Finnie JW, Bostock DE. Skin neoplasia in dogs. Aust Vet J 1979;55:602–4.
[11] Brodey RS. Canine and feline neoplasia. Adv Vet Sci Comp Med 1970;14:309–54.
[12] Bostock DE. Neoplasms of the skin and subcutaneous tissues in dogs and cats. Br Vet J

1986;142:1–19.

[13] Peters JA. Canine mastocytoma: excess risk as related to ancestry. J Natl Cancer Inst

1969;42:435–43.

[14] Patnaik AK, Ehler WJ, MacEwen EG. Canine cutaneous mast cell tumors: morphologic

grading and survival time in 83 dogs. Vet Pathol 1984;21:469–74.

[15] Lombard LS, Moloney JB. Experimental transmission of mast cell sarcoma in dogs. Fed

Proc 1959;18:490–5.

[16] Ashman LK. The biology of stem cell factor and its receptor C-kit. Int J Biochem Cell Biol

1999;31:1037–51.

[17] Galli SJ, Kitamura Y. Genetically mast cell deficient w

/w

v

and sl

/sl

d

mice. Am J Pathol

1987;127:191–8.

[18] Zsebo KM, Williams DA, Geissler EN, et al. Stem cell factor is encoded at the Sl locus

of the mouse and is the ligand for the c-kit tyrosine kinase receptor. Cell 1990;633:
213–24.

[19] Zsebo KM, Wypych J, McNiece IK, et al. Identification, purification, and biological

characterization of hematopoietic stem cell factor from buffalo rat liver-conditioned
medium. Cell 1990;63:195–201.

[20] Tsai M, Takeishi T, Thompson H, et al. Induction of mast cell proliferation, maturation,

and heparin synthesis by the rat c-kit ligand, stem cell factor. Proc Natl Acad Sci USA
1991;88:6382–6.

[21] Taylor ML, Metcalfe DD. Kit signal transduction. Hematol Oncol Clin North Am

2000;14:517–35.

[22] Sperr WR, Walchshofer S, Horny HP, et al. Systemic mastocytosis associated with acute

myeloid leukaemia: report of two cases and detection of the c-kit mutation Asp-816 to Val.
Br J Haematol 1998;103:740–9.

[23] Pignon J-M, Giraudier S, Duquesnoy P, et al. A new c-kit mutation in a case of aggressive

mast cell disease. Br J Hematol 1997;96:374–6.

[24] Nagata H, Okada T, Worobec AS, Semere T, Metcalfe DD. c-Kit mutation in a population

of patients with mastocytosis. Int Arch Allergy Immunol 1997;113:184–6.

[25] Longley BJ Jr, Metcalfe DD, Tharp M, et al. Activating and dominant inactivating c-KIT

catalytic domain mutations in distinct clinical forms of human mastocytosis. Proc Natl
Acad Sci USA 1999;96:1609–14.

[26] Longley BJ, Reguera MJ, Ma Y. Classes of c-KIT activating mutations: proposed

mechanisms of action and implications for disease classification and therapy. Leuk Res
2001;25:571–6.

[27] Buttner C, Henz BM, Welker P, Sepp NT, Grabbe J. Identification of activating

c-kit mutations in adult-, but not in childhood-onset indolent mastocytosis: a
possible explanation for divergent clinical behavior. J Invest Dermatol 1998;111:
1227–31.

486

C.A. London, B. Seguin

/ Vet Clin Small Anim 33 (2003) 473–489

background image

[28] Worobec AS, Semere T, Nagata H, Metcalfe DD. Clinical correlates of the presence of the

Asp816Val c-kit mutation in the peripheral blood mononuclear cells of patients with
mastocytosis. Cancer 1998;83:2120–9.

[29] Hirota S, Isozaki K, Moriyama Y, et al. Gain-of-function mutations of c-kit in human

gastrointestinal stromal tumors. Science 1998;279:577–80.

[30] Ernst SI, Hubbs AE, Przygodzki RM, Emory TS, Sobin LH, O’Leary TJ. KIT mutation

portends poor prognosis in gastrointestinal stromal

/smooth muscle tumors. Lab Invest

1998;78:1633–6.

[31] Lux ML, Rubin BP, Biase TL, et al. KIT extracellular and kinase domain mutations in

gastrointestinal stromal tumors. Am J Pathol 2000;156:791–5.

[32] Miettinen M, Sarlomo-Rikala M, Lasota J. Gastrointestinal stromal tumors: recent

advances in understanding of their biology. Hum Pathol 1999;30:1213–20.

[33] Natali PG, Nicotra MR, Sures I, Santoro E, Bigotti A, Ullrich A. Expression of c-kit receptor

in normal and transformed human non-lymphoid tissues. Cancer Res 1992;52:6139–43.

[34] Inoue M, Kyo S, Fujita M, Enomoto T, Kondoh G. Co-expression of the c-kit receptor

and the stem cell factor in gynecological tumors. Cancer Res 1994;54:3049–53.

[35] Stanulla M, Welte K, Hadam MR, Pietsch T. Coexpression of stem cell factor and its

receptor c-kit in human malignant glioma cell lines. Acta Neuropathol (Berl) 1995;89:
158–65.

[36] Turner AM, Zsebo KM, Martin F, Jacobsen FW, Bennett LG, Broudy VC. Non-

hematopoietic tumor cell lines express stem cell factor and display c-kit receptors. Blood
1992;80:374–81.

[37] Zemke D, Yamini B, Yuzbasiyan-Gurkan V. Characterization of an undifferentiated

malignancy as a mast cell tumor using mutation analysis in the proto-oncogene c-KIT.
J Vet Diagn Invest 2001;13:341–5.

[38] London CA, Galli SJ, Yuuki T, Hu Z-Q, Helfand SC, Geissler EN. Spontaneous canine

mast cell tumors express tandem duplications in the proto-oncogene c-kit. Exp Hematol
1999;27:689–97.

[39] Ma Y, Longley BJ, Wang X, Blount JL, Langley K, Caughey GH. Clustering of activating

mutations in c-KIT’s juxtamembrane coding region of canine mast cell neoplasms. J Invest
Dermatol 1999;112:165–70.

[40] Downing S, Chien MB, Kass PH, Moore PE, London CA. Prevalence and significance of

internal tandem duplications in exons 11 and 12 of C-kit in mast cell tumors of dogs. Am J
Vet Res 2002;63:1718–23.

[41] Bostock DE. The prognosis following surgical removal of mastocytomas in dogs. J Small

Anim Pract 1973;14:27–40.

[42] Crowe DT, Goodwin MA, Greene CE. Total laryngectomy for laryngeal mast cell tumor in

a dog. J Am Anim Hosp Assoc 1986;22:809–16.

[43] Patnaik AK, MacEwen EG, Black AP, Luckow S. Extracutaneous mast-cell tumor in the

dog. Vet Pathol 1982;19:608–15.

[44] Pollack MJ, Flanders JA, Johnson RC. Disseminated malignant mastocytoma in a dog.

J Am Anim Hosp Assoc 1991;27:435–40.

[45] O’Keefe DA, Couto CG, Burke-Schwartz C, Jacobs RM. Systemic mastocytosis in 16

dogs. J Vet Intern Med 1987;1:75–80.

[46] Hottendorf GH, Nielsen SW. Pathologic survey of 300 extirpated canine mastocytomas.

Zentralbl Veterinarmed A 1967;14:272–81.

[47] Thamm DH, Vail DM. Mast cell tumors. In: Withrow SJ, MacEwen EG, editors. Small

animal clinical oncology. 3rd edition. Philadelphia: WB Saunders; 2001. p. 261–82.

[48] 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;4:242–6.

[49] Howard EB, Sawa TR, Nielsen SW, Kenyon AJ. Mastocytoma and gastroduodenal ulceration.

Gastric and duodenal ulcers in dogs with mastocytoma. Pathol Vet 1969;6:146–58.

487

C.A. London, B. Seguin

/ Vet Clin Small Anim 33 (2003) 473–489

background image

[50] McManus PM. Frequency and severity of mastocytemia in dogs with and without mast cell

tumors: 120 cases (1995–1997). JAVMA 1999;215:355–7.

[51] Cayatte SM, McManus PM, Miller WH Jr, Scott DW. Identification of mast cells in buffy

coat preparations from dogs with inflammatory skin diseases. JAVMA 1995;206:325–6.

[52] Bookbinder PF, Butt MT, Harvey HJ. Determination of the number of mast cells in lymph

node, bone marrow, and buffy coat cytologic specimens from dogs. JAVMA 1992;
200:1648–50.

[53] Hahn KA, King GK, Harris FD, et al. The usefulness of hepatic and splenic fine needle

aspiration cytology in the clinical staging of canine cutaneous mast cell tumors. An
evaluation of 88 dogs (1987–1998). Proceedings of the Mid-Year Conference of the
Veterinary Cancer Society. 2000. p. 23.

[54] Leibman NF, Guilpin VO, Fettman MJ, Powers BE, Schochet RA, Withrow SJ. Cytologic

comparison of mast cell numbers in liver and spleen of normal dogs and dogs with mast cell
tumors. Proceedings of the 19th Annual Veterinary Cancer Society Conference. 1999. p. 28.

[55] Siegel S, Cronin KL, Philibert JC, Beck K, Schelling S. Evaluation of a staging protocol for

cutaneous mast cell tumors. Proceedings of the Mid-Year Conference of the Veterinary
Cancer Society. 2000. p. 22.

[56] Simoes JP, Schoning P, Butine M. Prognosis of canine mast cell tumors: a comparison of

three methods. Vet Pathol 1994;31:637–47.

[57] Turrel JM, Kitchell BE, Miller LM, Theon A. Prognostic factors for radiation treatment of

mast cell tumor in 85 dogs. JAVMA 1988;193:936–40.

[58] Frimberger AE, Moore AS, LaRue SM, Gliatto JM, Bengtson AE. Radiotherapy of

incompletely resected, moderately differentiated mast cell tumors in the dog: 37 cases
(1989–1993). J Am Anim Hosp Assoc 1997;33:320–4.

[59] Abadie JJ, Amardeilh MA, Delverdier ME. Immunohistochemical detection of proliferating

cell nuclear antigen and Ki-67 in mast cell tumors from dogs. JAVMA 1999;215:1629–34.

[60] Thamm DH, Mauldin EA, Vail DM. Prednisone and vinblastine chemotherapy for canine

mast cell tumor–41 cases (1992–1997). J Vet Intern Med 1999;13:491–7.

[61] LaDue T, Price GS, Dodge R, Page RL, Thrall DE. Radiation therapy for incompletely

resected canine mast cell tumors. Vet Radiol Ultrasound 1998;39:57–62.

[62] Takahashi T, Kadosawa T, Nagase M, et al. Visceral mast cell tumors in dogs: 10 cases

(1982–1997). JAVMA 2000;216:222–6.

[63] Kravis LD, Vail DM, Kisseberth WC, Ogilvie GK, Volk LM. Frequency of argyrophilic

nucleolar organizer regions in fine-needle aspirates and biopsy specimens from mast cell
tumors in dogs. JAVMA 1996;209:1418–20.

[64] Bostock DE, Crocker J, Harris K, Smith P. Nucleolar organiser regions as indicators of post-

surgical prognosis in canine spontaneous mast cell tumors. Br J Cancer 1989;59:915–8.

[65] Ayl RD, Couto CG, Hammer AS, Weisbrode S, Ericson JG, Mathes L. Correlation of

DNA ploidy to tumor histologic grade, clinical variables, and survival in dogs with mast
cell tumors. Vet Pathol 1992;29:386–90.

[66] Fang KC, Wolters PJ, Steinhoff M, Bidgol A, Blount JL, Caughey GH. Mast cell expression

of gelatinases A and B is regulated by kit ligand and TGF-beta. J Immunol 1999;162:5528–35.

[67] Leibman NF, Lana SE, Hansen RA, et al. Identification of matrix metalloproteinases in

canine cutaneous mast cell tumors. J Vet Intern Med 2000;14:583–6.

[68] Ginn PE, Fox LE, Brower JC, Gaskin A, Kurzman ID, Kubilis PS. Immunohistochemical

detection of p53 tumor-suppressor protein is a poor indicator of prognosis for canine
cutaneous mast cell tumors. Vet Pathol 2000;37:33–9.

[69] Jaffe MH, Hosgood G, Taylor HW, et al. Immunohistochemical and clinical evaluation of

p53 in canine cutaneous mast cell tumors. Vet Pathol 2000;37:40–6.

[70] Gilson SD, Stone EA. Principles of oncologic surgery. Compend Contin Educ Pract Vet

1990;12:827–39.

[71] Seguin B, Leibman NF, Bregazzi VS, et al. Clinical outcome of dogs with grade-II mast cell

tumors treated with surgery alone: 55 cases (1996–1999). JAVMA 2001;218:1120–3.

488

C.A. London, B. Seguin

/ Vet Clin Small Anim 33 (2003) 473–489

background image

[72] Weisse C, Shofer FS, Sorenmo K. Recurrence rates and sites for grade II canine cutaneous

mast cell tumors following complete surgical excision. J Am Anim Hosp Assoc 2002;38:71–3.

[73] Grier RL, Di Guardo G, Schaffer CB, et al. Mast cell tumor destruction by deionized

water. Am J Vet Res 1990;51:1116–20.

[74] Jaffe MH, Hosgood G, Kerwin SC, Hedlund CS, Taylor HW. Deionised water as an

adjunct to surgery for the treatment of canine cutaneous mast cell tumours. J Small Anim
Pract 2000;41:7–11.

[75] al-Sarraf R, Mauldin GN, Patnaik AK, Meleo KA. A prospective study of radiation therapy

for the treatment of grade 2 mast cell tumors in 32 dogs. J Vet Intern Med 1996;10:376–8.

[76] McCaw DL, Miller MA, Ogilvie GK, et al. Response of canine mast cell tumors to

treatment with oral prednisone. J Vet Intern Med 1994;8:406–8.

[77] Rassnick KM, Moore AS, Williams LE, et al. Treatment of canine mast cell tumors with

CCNU (lomustine). J Vet Intern Med 1999;13:601–5.

[78] Elmslie R. Combination chemotherapy with and without surgery for dogs with high grade

mast cell tumors with regional lymph node metastases. Vet Cancer Society Newsletter
1996;6–7.

[79] McCaw DL, Miller MA, Bergman PJ, et al. Vincristine therapy for mast cell tumors in

dogs. J Vet Intern Med 1997;11:375–8.

489

C.A. London, B. Seguin

/ Vet Clin Small Anim 33 (2003) 473–489

background image

Update on the biology and management

of canine osteosarcoma

Ruthanne Chun, DVM

a,

*,

Louis-Philippe de Lorimier, DVM

b

a

Kansas State University College of Veterinary Medicine, Mosier Hall,

1800 Denison Avenue, Manhattan, KS 66506-5606, USA

b

Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign,

1008 W. Hazelwood Dr., Urbana, IL 61802, USA

Though the likelihood of developing osteosarcoma (OSA) remains

greatest in large to giant breed dogs, specific reasons for increased risk in
this population remain ill-defined. This article will first discuss the physical
and molecular changes that have been associated with the development of
osteosarcoma in dogs.

Risk factors and associations for the development of osteosarcoma

Ionizing radiation

Ionizing radiation, in both experimental and therapeutic settings, has

long been known to cause OSA in dogs [1–6]. One study described the
development of OSA in the radiation therapy (RT) field 1.7 to 5 years after
completing therapy in 3

/87 dogs (3.4%) treated with megavoltage ir-

radiation for soft tissue sarcomas [2]. The fractionation scheme described in
this article was coarse, consisting of 10 fractions of 3.5 to 5 Gy over 3 weeks.
Late responding tissues, such as bone, are more sensitive to coarse fractions.
The mutagenic effects of ionizing radiation are the likely cause of OSA as
a late complication. The same investigators evaluated an additional 67
beagle dogs randomized to receive either external beam RT (EBRT,
n

¼ 14), intraoperative RT (IORT, n ¼ 26) or EBRT þ IORT (n ¼ 27).

The fractionation schemes used in this study ranged widely, but all IORT
treatments were given as a single large dose. One dog in the IORT group

Vet Clin Small Anim

33 (2003) 491–516

* Corresponding author.
E-mail address:

chun@vet.k-state.edu (R. Chun).

0195-5616

/03/$ - see front matter

Ó 2003, Elsevier Inc. All rights reserved.

doi:10.1016

/S0195-5616(03)00021-4

background image

and 7 dogs in the EBRT

þ IORT group developed OSA 4 or more years

after RT, supporting development of OSA as a late complication of coarse
fraction radiation therapy. Osteosarcoma in the RT field has also been
described in two dogs treated for acanthomatous epulis [6,7]. These dogs
were treated with 250 kVp orthovoltage radiation; orthovoltage radiation
results in a bone-absorbed dose approximately 2 to 3 times the dose received
by adjacent soft tissues [8]. Because it delivers a more homogenous bone and
soft-tissue radiation dose, megavoltage irradiation used in a finer fraction
scheme may minimize the development of secondary malignancies as a late
complication.

Bone infarcts and fatigue microdamage

Bone infarcts, whether spontaneous or associated with orthopedic

procedures such as arthroplasty, are rarely implicated in the development
of canine OSA [9–12]. The miniature Schnauzer breed is over-represented in
the literature for the development of OSA secondary to spontaneous bone
infarcts [10–13]. In humans, neoplasia associated with bone infarcts is
believed to be secondary to a defect in the chronic reparative process oc-
curring at the periphery of the infarct [9]. The pathogenesis of OSA in dogs
with bone infarcts remains unknown.

Fatigue microdamage of the metaphyses has been proposed as a potential

risk factor for OSA in large-breed dogs [14]. A study compared bone
microdamage of the distal radial metaphyseal plates of 10 small (<15 kg)
and 10 large (>25 kg) normal dogs, and it failed to detect a significant
difference between the two groups. These authors concluded that such
microdamage might not be an important risk factor for OSA in large breeds
of dogs [14].

Gonadal hormone exposure

A recent report of 683 Rottweilers demonstrated that dogs subjected to

early gonadectomy (before 1 year of age) had a higher risk for bone sarcoma
[15]. This risk factor was found to be independent of adult height or body
weight. Early spay and neutering is still more beneficial than detrimental
in terms of overall patient health, and the results of this study should
not unduly influence the decision to spay or neuter a canine patient of any
breed.

Genetic alterations

Alterations in the tumor-suppressor genes Rb and p53 are commonly

identified in human OSA patients, and they have been evaluated in dogs
with OSA [16–19]. The Rb gene family consists of Rb, p107, and p130. These
genes code for proteins with inhibitory functions in the regulation of
cell proliferation and differentiation. Human cancers frequently harbor

492

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

mutations inactivating the Rb pathway. In human OSA, 60% or more have
Rb

-associated abnormalities [18]. By comparison, mutations in the related

genes p107 and p130 are rare. Normally, Rb protein regulates cell cycle
transit by controlling the availability and activity of specific members of the
E2F family of transcription factors. Inactivation of Rb protein through
phosphorylation results in release, and thereby activation, of E2F, which in
turn allows transcription of a variety of genes associated with cell repli-
cation. Normal Rb phosphorylation is controlled during mid-G1 by cyclins
and cyclin-dependent kinases (CDK). The CDK inhibitor p16

INK4A

re-

sponds to growth-inhibiting signals by blocking Rb phosphorylation, thus
preventing DNA synthesis [16,17]. Ultimately, aberrant Rb activity results in
unregulated activation of E2F-related gene transcription and, thus, un-
controlled cellular proliferation.

The p53 gene is a tumor suppressor that acts as a master regulator of cell

replication. It also plays a critical role in the cellular response to DNA
damage. Loss of functional p53 confers a selective growth advantage to
transformed cells. Under normal circumstances, damage to the DNA
template induces signaling through the interaction of p53 with the p21

WAF1

/

CIP1

pathway, which in turn causes cell cycle arrest at the G1 restriction

point by preventing hyperphosphorylation of Rb. This cell-cycle arrest
allows time for DNA repair, which is also partly mediated by the p53
protein. If DNA damage is too extensive for repairs to be completed within
a set time period, normal p53 triggers a cascade of events ultimately leading
to cellular apoptosis. Normal p53 protein is labile, and therefore rarely
detected in tissue sections by routine immunohistochemistry [17]. Accumu-
lation in malignant cells is generally caused by mutations in p53, which do
not allow triggering of downregulatory mechanisms such as those provided
by the mdm-2 gene. Increased expression of p53 may not be linked to
increased activity; in fact, excessive amounts of a nonfunctional protein are
usually produced. The p21

WAF1

/CIP1

gene codes for a CDK inhibitor that

blocks the G1-S transition phase of the cell cycle. This gene is one of a group
that depends on p53 for activation. When p53 activity is defective owing to
a mutation, affected cells have unregulated replicative capability from lack
of activation of p21

WAF1

/CIP1

.

In a study investigating tumor suppressor gene expression in canine OSA,

5 cell lines were evaluated for abnormalities in Rb, p107, p130, mdm2,
p21

WAF1

/CIP1

, p53, and p16

INK4A

[17]. All 5 cell lines had defective p53

function, as evidenced by lack of induction of p21

WAF1

/CIP1

and mdm2. The

p53

mRNA and protein levels were elevated in 3 cell lines, and decreased in

the other 2. One cell line had increased p16 mRNA and protein levels, and it
significantly reduced levels of Rb, p107, and p130 proteins. Another study
reported that 47% of 15 canine OSA samples analyzed had mutations
within the p53 gene at sites similar to those seen in human OSA [16]. In
a third study of 21 canine OSA samples, no gross gene alterations (ie,
translocations, amplification, or deletions) in the structure of p53, Rb, or

493

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

mdm2

were identified by Southern blotting; however, p53 mutations were

detected by single-strand conformation polymorphism following amplifica-
tion by polymerase chain reaction (PCR) in 8 of these samples [18]. Further
evaluation of Rb protein using Western blotting did not identify abnor-
malities in this protein. Thus, though both canine and human OSA patients
have p53 mutations at similar frequencies, the Rb gene does not appear to
play as important a role in canine OSA as it does in the human disease.

Another important tumor suppressor gene, PTEN, was evaluated in

a recent study [20]. Four of five canine OSA cell lines studied constitutively
expressed high levels of the phosphorylated form of Akt, an indirect
indicator of aberrant PTEN expression. Deletions, mutations, and down-
regulation of PTEN were observed in most of the cell lines, and in 10 of
15 tumors studied by immunohistochemistry. These findings suggest that
PTEN

mutations may play a role in the pathogenesis of canine OSA, as in

many human cancers.

A study evaluating the proto-oncogenes c-sis, c-myc, N-myc, and cH-ras

in 9 canine OSA and 17 normal canine tissue samples revealed significant
amplifications of the c-sis and c-myc genes in tumor tissues [21]. This study
also showed similar levels of expression of the sis gene product, PDGF-b, in
human and canine OSA when analyzed by immunostaining. A more recent
study described overexpression of the sis oncogene in a canine OSA cell line,
and it found that all of the canine OSA cell lines tested in that study
contained PDGF receptors [22]. This suggests the possibility of an autocrine
growth factor loop participating in the pathogenesis of a subset of canine
OSAs.

The MET proto-oncogene was also investigated for mutation in canine

OSA by a group in Italy [23]. This oncogene is known to affect metastatic
potential and invasiveness of certain cancers in humans, including OSA.
Five of seven samples of canine OSA examined by Northern blot expressed
high levels of the MET oncogene, and a lung metastasis from one of the
dogs expressed MET at higher levels than did the primary tumor.

A recent report described increased gene expression of growth hormone

(GH) in areas of active osteoid formation [24]. Expression of GH mRNA
was demonstrated in the metaphyseal area of the normal canine growth
plate, and in 25% of the canine OSA specimens studied. The authors of this
study concluded that locally produced GH is involved in osteoid formation
and could play a role in the growth of canine OSA.

Canine appendicular osteosarcoma

Appendicular OSA is the most common form of OSA seen in dogs. This

section will cover the typical incidence and signalment, history and physical
examination, prognostic factors, and treatment options for dogs with
appendicular OSA.

494

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

Incidence and signalment

Appendicular OSA is the most common of all primary bone tumors in

dogs, accounting for up to 75% to 85% of these lesions [25]. OSA tends to
occur in middle-aged to older dogs, with some reports showing another
incidence peak at 2 years of age (bimodal distribution) [25]. Though male
dogs are often reported to be over-represented, with a male to female ratio
of 1.5:1, this finding is not consistent among all publications. [26–31]. Large-
and giant-breed dogs including Irish setters, St. Bernards, Rottweilers, and
Doberman pinschers are at greater risk [15,25,26,28–30].

History and physical examination

Dogs with appendicular OSA most commonly are presented for acute to

chronic onset of lameness and limb swelling. Owners often report some
‘‘traumatic incident’’ associated with the onset of lameness. Dogs are usually
eating and drinking normally.

Weight-bearing or nonweight-bearing lameness may be observed, with

limb swelling typically localized away from the elbow or toward the knee.
Osteosarcoma occurs with equal frequency at the distal radius, proximal
humerus, and proximal tibia. The mass is usually firm and often painful on
palpation.

Prognostic factors

Though breed and sex have not been recognized to have prognostic

importance, young dogs with OSA appear to have shorter survival times
and biologically more aggressive disease [31]. The presence of detectable
metastatic lesions at the time of diagnosis is a recognized poor prognostic
factor, standard chemotherapy being ineffective at improving survival
in that setting [26,32,33]. A study found that, in addition to detectable
metastasis at presentation, a telangiectatic (vascular) subtype, rib or
scapular location, a higher body weight, and incompleteness of excision
were also negative prognostic factors for OSA of flat or irregular bones
[32].

Elevated alkaline phosphatase (ALP) levels (total, bone or liver iso-

enzymes) are also negative prognostic factors [34,35]. Animals with elevated
levels have shorter survival times by approximately 50% (

170d versus

>

400d), even when treated aggressively with surgery and chemotherapy

[34,35]. One study reported that tumor located within the proximal humerus
and body weight of 40 or more kg were associated with a shorter disease free
interval (DFI) and survival [36]. Cited explanations for these factors were that
proximal humeral location potentially allows for more advanced local growth

495

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

before detection, and larger dogs may receive a proportionately smaller
chemotherapy dose than small dogs, when treated on a meter-square body
surface area basis.

Percent tumor necrosis, induced by RT or chemotherapy before tumor

excision, is predictive of local tumor control when compared with dogs
treated with amputation alone [37]. In this study, 200 dogs were treated with
either nothing beyond surgery (n

¼ 100) or preoperative RT, IV cisplatin,

or intra-arterial (IA) cisplatin followed by a limb-sparing procedure in
a variety of combinations of these treatments. In spite of the varied treat-
ment protocols within each group, RT was concluded to be the best way to
induce tumor necrosis (82%); IA delivery (49%) reportedly increased tumor
necrosis over that seen with IV delivery (24%) of cisplatin. Overall, tumor
necrosis was strongly correlated with local tumor control, but there was no
correlation between tumor necrosis and time to metastasis. Another study
found a significant direct correlation between survival time and percent
necrosis following 2 or 3 doses of doxorubicin [38]. In that study, percent
necrosis ranged from 0 to 87% (mean, 24.9%). Tumor necrosis is a well-
recognized prognostic factor in human OSA (cutoff value for significance is
90% necrosis post-therapy). Percent necrosis obtained after primary chemo-
therapy is used to modify postoperative chemotherapy drug protocols [38].

The use of scintigraphy to help predict time to metastasis in dogs with

OSA has been described [39]. Quantitative bone scintigraphy of the primary
tumor was performed before treatment in 25 dogs and following neo-
adjuvant therapy in 22 of these dogs. A significant relationship was
identified between time to metastasis and (1) radiographic tumor area; (2)
pretreatment ratio of mean counts per pixel in tumor compared with the
counts in adjacent nontumor bone; and (3) the pre- versus post-treatment
ratio. A larger radiographic tumor area and high pretreatment tumor
activity were associated with earlier metastasis, as were tumors that had
a large decrease in uptake after treatment [39].

A recently described histologic grading system for OSA appears to

provide prognostic information [40,41]. A total of 166 dogs provided 166
primary and 34 metastatic OSA tumor samples. These were categorized into
one of three grades based on cellular pleomorphism, mitotic figures, tumor
matrix, tumor cellularity, and percent necrosis. The 25% of tumors graded
as 1 or 2 were associated with a significantly longer disease-free and overall
survival than the 75% categorized as grade 3 tumors [40,41]. An earlier
study of 52 canine OSA samples showed that patients presenting with
detectable pulmonary metastases had increased microvascular density in the
primary tumor; it recognized this histopathologic finding to be a negative
prognostic indicator [42].

Finally, it has been reported that dogs with infections of their allografts

after limb-sparing surgery lived twice as long as dogs without infection
[43,44]. Nonspecific activation of the immune system has been advanced as
a potential explanation for the prolonged survival in infected patients.

496

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

Diagnosis

A minimum database consisting of a complete blood count, serum

chemistry profile, and urinalysis is recommended to screen for concurrent
disease, and to aid in establishing a prognosis by quantitating the serum
alkaline phosphatase concentration. Certain therapeutic decisions might
also be altered based on the results of such tests. For example, a patient with
marginal renal function might not be an ideal candidate for cisplatin
therapy, a known nephrotoxic agent, or could require a dose reduction of
a renally excreted drug such as carboplatin.

Diagnostic imaging plays an important role in staging dogs with OSA.

Radiographs of the primary lesion typically reveal bony proliferation and
lysis in the metaphyseal region of long bones. Thoracic radiographs reveal
gross metastatic disease in less than 10% of dogs at presentation. It is
well established, however, that more than 90% of these dogs will go on
to develop pulmonary metastatic disease if treated by surgery alone
[26,28,31,45]. Pulmonary metastatic lesions appear as discrete soft-tissue
nodules, and multiple lesions are common [46].

Nuclear scintigraphy is more sensitive than radiography for imaging

skeletal metastases because scintigraphy reveals changes in the rate of bone
remodeling that develop before structural changes can be detected ra-
diographically [47,48]. Thus, scintigraphy in OSA patients may identify
occult metastatic lesions in other bone sites or metachronous primary
lesions. Reports of using scintigraphy to stage OSA patients are contra-
dictory, however [47–50]. One study reported that 14 of 25 dogs with
appendicular or axial OSA showed suspicious lesions, of which 7 were
proven to be OSA by biopsy [49]. The remaining seven lesions were not
biopsied but were radiographically interpreted to be unrelated to the pri-
mary tumor. A larger study of 70 dogs with bone tumors (62 with ap-
pendicular OSA, 6 with fibrosarcoma, and 2 with chondrosarcoma) found 1
soft tissue metastatic lesion (lymph node) in a dog with OSA, and 1 osseous
metastasis in a dog with an ulnar chondrosarcoma lesion [47]. The dis-
crepancy between these studies may be caused by the small numbers of
patients evaluated in the first study. Also, animals in the first study might
have been evaluated at later stages of disease. A third article commented on
the use of sequential scans to stage and monitor dogs with OSA [48].
Although no metastases were detected before treatment, 25

/26 dogs

eventually developed metastatic lesions. Of dogs with metastasis, 7

/12 with

bone lesions had radiographically occult metastases, supporting the use of
bone scans for monitoring OSA patients. Because bony radiographic
changes can lag behind scintigraphic changes, the use of scintigraphy to
screen OSA patients for occult disease is very sensitive [48]. The specificity of
this imaging modality is relatively low, however, and any scintigraphic
lesions beyond the primary tumor should ideally be verified by radiographs
or biopsy.

497

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

Three recent studies evaluated different imaging modalities for accuracy

regarding local extent of OSA, using histopathology as the gold standard for
comparison [51–53]. In a study of 20 dogs with distal radial OSA evaluated
before a limb-sparing procedure, nuclear scintigraphy was found to
overestimate the tumor length significantly more than did radiography
[52]. Although this might provide a larger margin of safety when de-
termining the site of proximal osteotomy, it could also inappropriately
eliminate limb sparing as a treatment option for some patients. A second
study compared the use of MRI and CT to radiographs and histologic
evaluation in eight dogs with OSA [53]. In that study, MRI provided the
most accurate tumor estimate and was recognized as the best modality for
preoperative assessment of appendicular OSA, especially in the context of
a limb-sparing procedure. The last study compared MRI, CT, and radio-
graphs in 10 dogs with appendicular OSA, and failed to identify a superior
modality [51]. That study was performed on samples obtained post-
amputation, making it impossible to use injectable contrast agents for
enhancement of soft-tissue components.

Though biopsy remains the gold standard for diagnosis of OSA, fine

needle aspirate via a 19-gauge needle may provide a diagnosis by less
invasive means [54]. Cells from lytic OSA lesions will readily exfoliate.
Aspirates often reveal large, immature mesenchymal cells that may have
intracytoplasmic or extracellular osteoid, which appears as an amorphous
pink matrix. In addition to malignant osteoblasts, benign osteoblasts and
osteoclasts may also be present. Fine needle aspiration of lytic bone lesions
may help to rule out fungal or bacterial osteomyelitis, where inflammatory
cells or organisms may be observed. A tissue biopsy can also be used to
provide preliminary cytologic information by touching or rolling the sample
on a slide. An ongoing study evaluates the usefulness of an ALP stain on
cytology samples to differentiate OSA from other sarcomas affecting bones.
Preliminary results show that only OSA lesions, as verified by biopsy,
consistently stain positive for ALP, whereas other sarcomas do not retain
the stain (Dr. Anne M. Barger, personal communication).

Important risks associated with bone biopsy in OSA patients include

the potential for infection, nondiagnostic biopsy and uncommonly iatro-
genic fracture. At minimum, many patients will have increased swelling
and lameness following the biopsy. Biopsy is a painful procedure, and the
patients must be heavily sedated or anesthetized. Core biopsies may be
obtained using a Jamshidi bone core biopsy needle (Baxter Health Care
Corporation, Valencia, CA) or Michel’s trephine; larger samples can be
procured by open surgical biopsy. Several histologic subtypes (telangiectatic,
osteoblastic, chondroblastic, fibroblastic) of osteosarcoma exist, but subtype
has yet to provide prognostic information. OSA arising from the periosteum
differ in prognosis from those of medullary origin, however [43]. Periosteal
OSA is a high-grade, invasive, and metastatic disease that arises from the
periosteal surface of bone. Parosteal OSA also arises from the periosteal

498

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

surface, but this lesion behaves in a manner more like that of a low-grade
soft-tissue sarcoma and is unlikely to invade bone locally or to metastasize
[43]. Histologically, paraosteal OSA lesions have a more benign appearance
than do those of intraosseous or periosteal OSA.

Biologic behavior

Appendicular OSA is a locally invasive, highly metastatic disease. The

pulmonary parenchyma is the most common metastatic site [29]. Lymph
nodes appear to be an uncommon site of metastases, with a reported rate
of 6.1% to 37% [26,29,31,55]. With the now common use of adjuvant
chemotherapy that provides excellent drug levels to pulmonary parenchyma,
bones and soft tissues are increasingly the site of OSA metastasis [55].

Treatment

Standard of care, defined as the treatment option that results in the

longest median survival times, is surgical resection of the primary tumor
followed by 3 to 6 cycles of either a platinum- or doxorubicin-based
chemotherapy protocol [27,30,36,38,45,56,57]. Surgery alone, in dogs with
no evidence of metastatic disease, is associated with a median survival time
of 19 weeks [31].

Surgical options for appendicular OSA include amputation or a limb-

sparing procedure, in which the affected bone is typically resected and
replaced by a normal bone allograft. The site that is most amenable to this
limb-sparing procedure is the distal radius [56,58,59]. Though it is tech-
nically possible to perform limb-sparing at other sites, patients typically
have poor limb function because the procedure requires arthrodesis of the
joint closest to the tumor [58,59]. Criteria for determining a patient’s
eligibility for limb-sparing surgery include tumor involving 50% or less of
the length of the bone, tumor not extending across the joint, and a patient
that is free from metastatic or concurrent disease [59]. Though the bone
allograft is the best described and most widely used technique, alternative
limb-sparing techniques have recently been reported and include ipsilateral
vascularized ulnar transposition autograft, pasteurized tumoral autograft,
bone transport osteogenesis, and intraoperative radiation techniques [43,
60–63].

There are also multiple reports of the use of combination chemother-

apy using doxorubicin and platinum-based chemotherapy drugs for the
treatment of canine OSA, and new protocols have been reported in abstract
form [28,44,64–67]. One report describes alternating doses of cisplatin and
doxorubicin as adjuvant to surgery in 19 dogs with OSA [28]. Survival times
in this study were similar to those reported for dogs treated with single agent
therapy (Table 1). A study evaluating 16 dogs treated with surgery and

499

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

Ta

ble

1

An

overvie

w

o

f

appe

ndicu

lar

osteosa

rcoma

therap

y

over

the

past

15

years

Su

rgery

and

ch

emothera

py

protoco

l

Autho

r,

year

M

edian

survival

(1-

yr,

2-yr

survival)

Am

p

alone

(n

¼

19)

Mauldin

,

1988

[28]

175d

(21%

,

0%)

Am

p

þ

DO

X

(30

mg

/m

2

dl)

þ

cD

DP

(60

mg

/m

2

d21

)

2

doses

(n

¼

19;

36

appe

ndicular,

2

axial)

300d

(37%

,

26%

)

Am

p

þ

cDDP

50

mg

/m

2

q28d



2–6

doses

(n

¼

19)

Shapiro

,

1988

[30]

301d

(unkn

own,

unkn

own)

Am

p

þ

cDDP

50

mg

/m

2

q28d



6

doses

or

until

me

tastasis

not

ed

(n

¼

16)

Kraegel,

1991

[27]

413d

(62%

,



18%

)

Am

p

alone

(n

¼

17)

Straw,

1991

[57]

119d

(11%

,

4%)

Am

p

þ

cDDP

70

mg

/m

2

q21d



2

doses

(n

¼

19)

262d

(38%

,

18%

)

Am

p

þ

cDDP

70

mg

/m

2

21d

preop

and

imm

ediately

postop

(n

¼

35)

282d

(43%

,

16%

)

Am

p

alone

(n

¼

15)

Thomp

son,

1992

[45]

168d

(20%

,

unkno

wn)

Am

p

þ

cDDP

60

mg

/m

2



2

doses

at

2

and

7

w

ks

po

stop

(n

¼

15)

290d

(33%

,

unkno

wn)

Am

p

alone

(n

¼

162)

Spodn

ick,

1992

[31]

134d

(11.5%

,

2

%)

Am

p

/limb

spare

þ

cDDP

60

mg

/m

2

q21d



1–6

doses

(n

¼

22;

17

¼

amp,

5

¼

limb

spa

re).

Surviv

al

data

not

divi

ded

for

sx

type

s

Berg,

1992

[56]

325d

(45.5%

,

20.9%)

Am

p

/limb

spare

þ

DO

X

3

0

m

g

/m

2

q2

wks



5

doses

(n

¼

35;

33

¼

amp,

2

¼

limb

spa

re).

Beca

use

2–3

doses

were

given

neoa

djuvant

to

surgery

,

only

patie

nts

with

minimal

clinical

sign

s

were

inc

luded;

surv

ival

date

not

divi

ded

for

sx

types

Berg,

1995

[38]

366d

(50.5%

,

9.7%

)

Am

p

þ

carbo

300

mg

/m

2

q21d



up

to

4

(n

¼

48)

Bergman

1996

[36]

321d

(35.4%

,

unkno

wn)

Am

p

þ

DO

X

(15–2

0

m

g

/m

2

1–2

hour

s

before

cDDP)

þ

cDDP

(60

mg

/m

2

)

3

doses

startin

g

2

(n

¼

53)

or

10

(n

¼

49)

days

postop;

12

dogs

died

after

first

cycle.

Berg,

1997

[65]

2d

postop

:

345

d

(48%,

28.3%,

3–yr

:

15.3%

)

10d

postop

:

330

d

(4

6.2%,

27.5%

,

3–yr

:

18%

)

Am

p

þ

cDDP

(5

0

m

g

/m

2

dl)

þ

DO

X

(15

mg

/m

2

d2)



4(

n

¼

16).

Also

tx

w

/empty

lipo

somes

Chun,

2000

[67]

540d

(68.7%

,

25%

)

A

bbrevia

tions:

Am

p,

amp

utation;

carbo

,

carbo

platin;

cDDP,

cisp

latin;

d,

day

;

DOX,

doxo

rubicin;

postop,

po

stopera

tive;

preop,

preop

erative;

sx,

surgery

;

tx,

treated.

500

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

4 cycles of cisplatin

/doxorubicin, each given within a 24-hour period,

reported significantly prolonged survival times (18.5-month median) [67]. A
report of 102 dogs treated with 3 cycles of the same protocol, however,
beginning either 2 or 10 days after surgery, documented median survivals of
11.5 months (beginning therapy 2 days postop) and 11 months (beginning
therapy 10 days postop) [65]. Potential reasons for the discrepancy in sur-
vival times between these studies include small numbers of dogs in the first
study, and the fact that the dogs in the first study also received empty
liposomes as part of the control arm of an immunotherapy study. Studies
have documented a greater combined immunomodulatory effect of doxo-
rubicin and L-MTP-PE than that seen with either agent alone [68–71].
Though no studies have reported the immunostimulatory effect of doxo-
rubicin with empty liposomes, it is possible that this combination could have
resulted in enhanced monocyte

/macrophage activity and, therefore, longer

survival time. In spite of some conflicting results, it remains clear that the
addition of chemotherapy increases survival times from a 4-month median
to an 18-month median at best (Table 1) [27,28,30,31,36,38,45,56,57,65,67].
Alternative routes for delivering chemotherapy and investigated innovative
therapies are briefly described in Tables 2 and 3 [37,86–89,110–114,116].

The most recent reports discussing novel therapies for adjuvant medical

management of canine OSA failed to show improvement in survival times
[41,72,73]. A liposome-encapsulated form of cisplatin, while permitting a
higher dose of cisplatin to be safely administered to 20 dogs with OSA, did
not improve survival time when compared with the carboplatin-treated
control group [73]. A phase 2 clinical trial evaluation with the new third-
generation platinum compound lobaplatin in 28 dogs with OSA showed the
drug to be well tolerated, but the one year survival of 31.8% did not differ
significantly from the results obtained with accepted standard-of-care
therapy [41]. Finally, a randomized controlled trial combining a long-acting
insulin-like growth factor (IGF) suppressant (octreotide) with carboplatin in
23 dogs did not improve survival when compared with the carboplatin-
treated control group of 21 dogs, despite a significant measurable decrease
in IGF levels [72].

It has been reported that interstitial fluid pressure is greatly increased and

blood flow decreased in OSA tissue, when compared with adjacent un-
affected tissue [74]. This could partly explain poor drug delivery or response
to radiation in a subset of patients, and strategies to bypass this problem
should be evaluated.

Once metastatic disease is clinically detectable, chemotherapy is usually

ineffective [33]. Pulmonary metastatectomy is a well-described procedure
that can contribute to significantly prolonged survival in dogs. Clinical
guidelines for determining patient eligibility for metastatectomy include:
primary tumor in complete and prolonged remission (ideally for >300 days);
1 to 2 radiographically detectable nodules only; no evidence of distant
metastatic disease at nonpulmonary sites; no evidence of life-limiting

501

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

Table

2

Alternative

chem

othera

py

de

livery

rout

es

Treatm

ent

Autho

r,

yea

r

Com

ments

Intra-a

rterial

cDDP

70

mg

/m

2



2

doses

þ

limb

spare

(n

¼

16)

Powe

rs,

1991

[37]

IA

on

ly

¼

49.1%

mean

tumor

necrosis

(3

/14

¼

80%)

Intra-a

rterial

cDDP

70

mg

/m

2



2

doses

w

/20–4

0

G

y

RT

þ

limb

spa

re

(n

¼

47)

IA

þ

RT

¼

83.7%

me

an

tum

or

necrosis

(31

/47

¼

80%)

Goal

was

to

determ

ine

pre

dictive

valu

e

o

f

p

ercent

tumor

necrosis

on

DFI

and

ove

rall

survival

Intrame

dullary

cDDP

60

mg

/m

2

(via

Jam-Sh

idi

bo

ne

core

biops

y

instrum

ent)



1–6

doses

(n

¼

4)

Hahn

,

1996

[111]

Case

1:

7-mo

survival

Case

2:

4.5-m

o

survival

(tum

or

present



6-mo

be

fore

tx)

Case

3:

3-mo

survival

Case

4:

<

1-mo

nth

surv

ival

Subcuta

neous

cDDP

50–7

0

m

g

/m

2

in

saline



1

dose

(n

¼

6)

Dernel

l,

1997

[110]

Unac

ceptab

le

toxicity

Inhala

tion

ch

emothera

py

using

paclitaxel

and

/or

DOX

q14

d,

with

a

spe

cially

de

signed

aerosol

device

Hersh

ey,

1999

[112]

10

cases

of

metastati

c

OSA

,

3

respo

nses

(2

PR,

1

C

R)

Abbrevia

tions:

CR,

co

mplete

remissio

n;

d,

day;

DO

X,

do

xorubicin

;

IA,

intra

arter

ial;

mo,

month;

PR,

par

tial

remission;

R

T

,

rad

iation

therap

y;

tx,

treatmen

t.

502

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

Ta

ble

3

Othe

r

repor

ted

tr

eatment

s

Treat

ment

Auth

or,

year

Comments

Samar

ium

-153

ethylene-d

iam

ine-tetram

ethylen

e

phosph

onic

acid

(Sm

153

-EDTMP)

37

MBq

(1.0

mCi)

/kg

IV



1–2

doses,

separa

ted

by

1

week

La

ttimer

,

1990

[87]

Axia

l

skeleto

n

(n

¼

8):

12-m

o

mean

survival.

Appe

ndicular

skel

eton

(n

¼

20):

me

an

surv

ival

by

site—

radi

us

/ulna

¼

7

m

o

,

humeru

s

¼

5m

o

,

tibia

¼

13

mo,

fem

ur

¼

7

m

o

Some

dogs

had

me

tastatic

lesions

that

respo

nded

to

therap

y.

43

and

45

M

B

q

IV



1

d

o

se

eac

h,

15

w

eeks

apart

(n

¼

1)

M

oe,

1996

[89]

Maxilla

ry

OSA

tx

with

Sm

and

maxillectomy,

surv

ival

>

21

mo.

37

MBq

/kg

(1

mCi

/kg)

IV



1

dose

(n

¼

9,

4

w

ith

biopsy-

proven

appe

ndicular

OSA)

M

ilner,

1998

[88]

Of

the

4

with

OSA

,

survival

w

a

s

3

,

3

,

4

,

and

6

m

o

.

No

response

s

were

noted.

36–5

7

M

Bq

/kg

IV



1–4

doses

(n

¼

15,

12

¼

appe

ndicular,

3

¼

axial

)

Aas,

1999

[86]

3

also

tx

with

surg

ery.

M

edian

survival

5

mo.

Lipo

some

encapsu

lated

muramyl-

tripeptid

ephosp

hatidyl-

ethanolam

ine

(L-M

TP-PE

)

116

dogs

in

mult

iple

stud

ies

ove

r

several

years

M

acEwe

n,

1996

[114]

Longest

survivo

rs

tx

with

amp

þ

cDDP

þ

liposo

mes

(af

ter

cDDP):

14.4

mo

media

n.

Onl

y

dogs

NED

aft

er

cD

DP

received

L-MT

P-PE.

Nebu

lized

IL-2

liposo

me

inhalatio

n

therap

y:

2



daily



15d

,

then

3



dail

y



15d

(n

¼

4),

2



daily



30d

(n

¼

5)

Kh

anna,

1997

[113]

Study

of

dogs

wi

th

metastati

c

o

r

primary

lung

cance

r,

2

/4

w

ith

OSA

had

CR



12

and

20

mo.

Am

p

þ

cDDP

(60

mg

/m

2



1–6)

þ

huma

n

cytot

oxic

T-cell

line

TA

LL-104

(c

-ir

radiated

T-cells

10

8

/kg

IV



5d,

then

2d

boosters

q30

d



9

mo)

(n

¼

23)

V

isonneau

,

1999

[116]

11.5

mo,

52.2%

1

year

survival,

8.7%

2

year

survival.

Only

dogs

NED

aft

er

cD

DP

received

T-c

ell

therap

y

(CD3

þ

,

CD8

þ

,

CD5

,

CD1

6

),

MHC

n

o

nrestricted

killer

activ

ity.

A

bbrevia

tions:

amp,

amp

utation;

cDDP,

cisp

latin;

CR,

comp

lete

rem

ission;

d,

day;

M

Bq,

mega

Be

cquerel;

mCi,

milliCurie;

mo

,

month

;

NED

,

n

o

eviden

ce

of

disease

;

OSA

,

osteosa

rcoma

;

tx,

tr

eated.

503

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

intercurrent disease; and a long tumor doubling time (>30 days) with no
new lesions observed during this time period [46]. Though this last point
remains to be of proven importance for veterinary patients, it is a prudent
guideline to follow.

Palliative therapy

Reported survival times for canine patients treated with palliative intent

therapy range from 3 to 10 months [26,31,75–78]. Amputation alone or
administration of medications such as nonsteroidal anti-inflammatory drugs
or opioids may significantly improve quality of life. Dogs treated with
amputation alone are most often euthanized because of pulmonary
metastatic disease, with a reported median survival time of 19 weeks [31].

The bisphosphonate drug alendronate has been anecdotally reported as

a palliative treatment in 2 dogs with OSA [78]. One dog had tibial disease
and survived 12 months after diagnosis; the other dog had maxillary disease
and survived 10 months after diagnosis. Both dogs were treated with 10 mg
alendronate orally, once daily. Bisphosphonates are antiresorptive agents
acting mainly by causing osteoclast apoptosis (cell death) and are used in
conditions such as osteoporosis and Paget’s disease in humans. The newer
and more potent aminobisphosphonates, such as pamidronate and zo-
ledronate, have been used to control hypercalcemia of malignancy and to
palliate for osteolytic bone pain in humans. Interestingly, recent in vitro
investigation of rat and human OSA cell lines, as well as of normal human
osteoblasts, show encouraging results for using aminobisphosphonates
as cytotoxic, cytostatic, or differentiating agents for OSA [79–83]. Ongoing
studies on canine OSA cell lines show similar preliminary findings, with
time- and dose-dependent cytotoxic and antiproliferative effects (de
Lorimier, unpublished data). Canine in vivo studies will address safety
and potential efficacy of such adjuvant therapy in the near future.

Palliative RT typically involves administering coarse fractions of 8 to 10

Gy of megavoltage irradiation, in 3 treatments at 0, 7, and 21 days [76,
77,84]. Palliative RT reportedly improves limb function and quality of life in
about 75% of patients for a median of 2 months duration [76,77,84]. A
recent report describing the use of 4 8-Gy fractions on days 0, 7, 14, and 21
showed response in 92% of sites treated, with a median duration of response
for appendicular sites of 94.5 days [75]. The median survival of these
patients was 313 days. Administration of radiation results in tumor cell
necrosis, replacement by fibrous tissue, and formation and calcification of
woven bone, which is gradually replaced by lamellar bone. Immediate pain
relief has been attributed to a cytotoxic process affecting normal bone cells
that release chemical mediators (prostaglandins, notably PGE

2

) in response

to the neoplastic process. These chemical mediators modulate pain per-
ception by stimulating nociceptors located in the periosteum. The later onset

504

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

of pain relief is felt to coincide with recalcification and repair of osteolytic
lesions [85].

The use of samarium has been described for both appendicular and axial

OSA in dogs (Table 3) [86–90]. Whether this treatment is definitive or
palliative remains to be fully determined. The radionuclide

153

Sm emits

both b particles, which provide the therapeutic effect, as well as c particles,
which allow for visualization of lesions on bone scans. When bound to
ethylene-diamino-tetramethylene-phosphate (EDTMP) and administered
intravenously (IV), this agent is rapidly and preferentially taken up by
metabolically active bone. While the use of

153

Sm-EDTMP as an alternative

treatment for bone tumors has been well described [86–89], a standardized
protocol for veterinary use is not well established. In general, the agent is
given as an IV bolus at 37 MBq

/kg, and the dose may be repeated at 1- to

19-week intervals. The main toxicity is myelosuppression, which may
continue for up to 4 weeks post-therapy [87,88].

Canine axial osteosarcoma

Signalment

Medium- to large-breed dogs are most commonly affected by axial OSA,

with a reported range in weights of less than 5 kg to 55 kg [25]. Middle-aged
dogs are most often reported to be affected, with the main exception being
dogs with rib tumors. Dogs with rib OSA tend to be younger, although
reported mean and median ages range from 5 to 9 years [91–93]. Female
dogs are more commonly affected than males (2.1:1) [91]. Golden and
Labrador retrievers, German Shepherds, Doberman Pinschers, and Irish
Setters are all over-represented, but the majority of reported dogs are mixed
breeds [91]. Up to 43% of all bone tumors in small (<15 kg) dogs are
osteosarcoma [94]. Unlike the disease in large-breed dogs, OSA in small
dogs tends to affect the axial skeleton more often than the appendicular
skeleton, with 9

/16 (56%) cases being located on the axial skeleton in one

study [94].

History and physical examination

The history varies depending on affected site. The tumor does not usually

grow rapidly, and clinical signs may be insidious. One article cited a range in
duration of signs before presentation extending from several days to 2 years
[91].

The most common sites of axial OSA are the mandible and maxilla; less

commonly affected sites include spine, ribs, nasal cavity, and cranium
[84,91]. One study reported that 4

/37 primary spinal tumors were OSA,

whereas another described vertebral OSA in 14

/20 dogs with primary or

505

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

metastatic tumors [55,95]. A total of 34 of 54 (63%) reported primary rib
tumors were OSA in one study, and 24 of 40 (60%) reported primary rib
tumors were OSA in another [92,93]. Thus, though primary bone tumors not
arising in the appendicular skeleton, mandible, or maxilla are rare, they are
likely to be diagnosed as OSA. Clinical signs will vary according to loca-
tion of the primary lesion. A visible or palpable mass, decreased appetite or
dysphagia, pain on opening the mouth, exophthalmia, ptyalism, epistaxis,
paraparesis, and dyspnea are among the observed findings.

Diagnosis

Diagnostic evaluation of dogs with axial skeletal OSA is similar to that

for dogs with appendicular OSA. A minimum database is advised to rule
out concurrent disease; the significance of elevations of serum alkaline
phosphatase in dogs with axial disease is unknown. Diagnostic imaging of
the primary site and thorax is also recommended. Only 11% of axial skeletal
OSA patients have visible pulmonary metastatic disease at the time of
diagnosis (6

/54); however, this metastatic incidence may be site-dependent

[91]. One study of 116 dogs reported thoracic radiographs for 54 cases [91].
None of the dogs with maxillary (n

¼ 11), nasal cavity (n ¼ 4), or pelvic

(n

¼ 2) OSA had lung metastases; however, lung metastases were observed

in dogs with mandibular (1

/19), spinal (1/6), rib (2/7), and cranium (2/5)

OSA. Myelography may be indicated to further delineate spinal tumors.
Vertebral OSA is typically an extradural lesion [95]. Advanced imaging
techniques, such as nuclear scintigraphy, CT, or MRI, can be extremely
valuable in the diagnostic evaluation of axial OSA and will help to plan
therapy. Abdominal radiographs or ultrasound to assess local extent of
disease or metastasis may be appropriate depending on the primary site.
Lymph node enlargement with histologic confirmation of metastasis is
identified in 7% or fewer cases [91,96]. As with appendicular OSA, biopsy or
fine needle aspirate is required for a diagnosis. Fluoroscopy or CT guidance
might prove valuable, especially for lesions of the spine.

Biologic behavior, prognosis, treatment

The biologic behavior of axial OSA has been postulated to be site-

dependent, with mandibular OSA being less likely to metastasize than OSA
originating in other sites [90]. Univariate analysis identified tumor loca-
tion on the mandible, complete surgical excision, and lower body weight
(<30 kg) as favorable prognostic factors in one study of 45 dogs [32]. In the
same article, surgical margins and body weight were the most significant
prognostic factors based on multivariate analysis. A large retrospective
study described 1- and 2-year overall survival times of 26.3% and 18.4%,
respectively, for animals treated with surgery alone (38/116) [91]. Local

506

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

recurrence was the cause of euthanasia in approximately 80% of cases,
whereas only 7% were euthanized because of metastatic disease. Because
local recurrence is the cause of euthanasia for the majority of cases,
metastases may not have time to manifest or to affect quality of life in axial
OSA patients.

General treatment recommendations for axial OSA are surgical resection

followed by chemotherapy with a platinum- or doxorubicin-based protocol.
The exception is mandibular OSA, for which surgery alone may provide up
to 71% 1–year survivals, and for which chemotherapy has not yet
demonstrated any survival advantage [90].

An unfortunate tendency in the veterinary literature is to retrospectively

report large numbers of dogs treated by a variety of therapies. Because so
few dogs are treated with the same protocol, the reported disease-free and
overall survival times are difficult to interpret. The following section at-
tempts to summarize usable data from reports studying bone tumors at
various anatomic sites.

Mandibular OSA has been described as having a lower metastatic rate

and better prognosis than appendicular OSA. One report of 12 dogs with
oral OSA (4 mandibular and 8 maxillary) treated with surgery alone
obtained a median 1–year survival of 42% [97]. Another article described 81
dogs with mandibular tumors, 9 of which were OSA [115]. In this study,
necropsies were performed in 5 dogs with OSA. Local recurrence was
documented in two of them, lymph node metastases were found in two, lung
metastases in four, and other sites of metastases in two. A third article
described 142 dogs treated surgically for mandibular tumors, 20 of which
were OSA [99]. These investigators reported that two dogs died because of
local recurrence, six dogs developed metastatic disease, and one dog died
from both local and distant disease. Straw and others documented 51 dogs
with mandibular OSA treated by surgery alone (n

¼ 32); surgery and

chemotherapy (n

¼ 10); surgery and radiation therapy (n ¼ 3); surgery,

radiation, and chemotherapy (n

¼ 4); or samarium (n ¼ 2) [90]. The dogs

treated with surgery alone had a 71% 1–year survival rate; there was no
apparent effect on survival of various other treatment modalities. Histologic
grade was significantly correlated with survival. Parameters measured for
grade included nuclear pleomorphism (0–3), mitotic rate (1–4), and percent
necrosis (0–3). Grade 1 tumors have a score of less than 6, grade 2 tumors
have a score of 6 to 8, and grade 3 tumors have a score more than 8.

Several studies have been published regarding dogs with maxillary OSA

[91,97,98,100]. A group reported 61 dogs with maxillary cancer. The 11 dogs
with OSA in this study were treated by surgery, with or without che-
motherapy, or radiation therapy [98]. Necropsy performed on 8 of 11 of the
dogs with OSA identified 3 with local recurrence and 4 with metastases (2 to
lung, and 2 elsewhere). All the dogs in the study, regardless of histologic
diagnosis, were divided into categories based on tumor location. Patients
with rostral lesions had better disease-free and overall survival times than

507

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

those with caudal tumor locations. Respectively, 1- and 2-year survival rates
for lesions rostral to the canine tooth, from the canine to premolar 3, and
caudal to the third premolar were 81% and 68%, 35% and 24%, and 40%
and 20%, respectively. Two cited explanations for this site-associated re-
sponse include a difference in biologic behavior based on tumor location,
and earlier recognition of rostral tumors. Another explanation is improved
surgical access for complete excision in rostral tumors. In this same article,
there was no observed difference in survival times among patients with the
different malignant tumors identified, and recurrence rates after ‘‘incom-
plete’’ and ‘‘complete’’ surgery were more than 50% and 40%, respectively.
Potential reasons cited for the high recurrence rate despite complete excision
included: (1) multifocal tumor; (2) development of new, unrelated tumor;
and (3) inadequate pathologic assessment. Overall median survival time for
dogs with maxillary OSA was 5 months, but these dogs received a variety of
treatment modalities [98]. Another study described 69 dogs treated with
hemimaxillectomy alone; 6 of these dogs had OSA and the median survival
time for them was 4.6 months [100].

The spine is an uncommonly reported site of primary OSA [55,91,95,101].

In 5 dogs treated by surgery alone, only one dog survived more than 3 days
[91]. This dog had a tail amputation and survived more than 2.5 years after
surgery. Another retrospective study of spinal tumors in dogs reported 2

/4

dogs treated by surgery alone as having survival times of 21 and 300 days
[95]. A larger retrospective study identified 14 dogs with primary or met-
astatic vertebral OSA treated by surgery alone, radiation therapy and
chemotherapy, surgery and chemotherapy, or surgery, radiation, and che-
motherapy combined [55]. Although a number of factors varied greatly in
this study, the median survival time for dogs with OSA was 135 days.

Primary rib OSA accounted for 10% of axial OSA cases in one study [91].

In another study, 27% of dogs with rib OSA had metastases at diagnosis,
whereas 45% had metastases identified at necropsy [102]. Thus, rib OSA
may have a more aggressive behavior than those from other sites on the
axial skeleton. Reported sites of metastases included lungs (5), bone (3),
myocardium and lymph node (1), and lung and mediastinum (1) [96]. In
another study evaluating survival times in 34 dogs with primary rib tumors,
9 dogs with OSA received surgery and chemotherapy (either cisplatin alone
or alternating doses of cisplatin and doxorubicin), and 20 dogs received
surgery alone. Median DFI and survival were 225 days and 240 days for
surgery combined with chemotherapy, but only 60 and 90 days for surgery
alone [93]. A report of 40 dogs treated with en bloc rib resection alone for
primary rib tumors identified 24 dogs with OSA [92]. Twenty of the dogs
with OSA were followed up beyond 2 weeks postoperatively; 14 died or were
euthanized because of pulmonary metastases, and 4 were euthanized
because of both pulmonary metastases and local recurrence. The remaining
2 dogs were euthanized because of vertebral metastases. Overall median
survival time for the dogs with rib OSA in this study was 3.3 months.

508

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

In a recent report on the use of 4 coarse fractions (8 Gy) of palliative

radiotherapy for OSA in 24 dogs, 9 tumors were of axial location [75]. All 9
dogs had a positive response to RT, with a median onset to diminution of
signs of 14 days, and median duration of 81.5 days. The survival times
ranged from 51 to 391 days, with a median of 162 days [75]. The ra-
dioisotope

153

Sm has been used for dogs with axial OSA, and, although

responses varied, good treatment response and pain control were obtained
in more than 50% of dogs treated [86,87,89]. A mean survival time of 12
months was reported for axial OSA in 8

153

Sm-treated dogs in one report

[87]. Because criteria establishing good candidates for

153

Sm therapy have

changed over time, future reports might provide improved response rates
and survival times.

Canine extraskeletal osteosarcoma and mammary gland osteosarcoma

Incidence and signalment

Canine extraskeletal osteosarcoma (ESOSA) is a rare tumor, accounting

for 0.13% of biopsy submissions (169

/130,754) and 12.6% of all OSA over

a 10-year period in 1 study [103]. In this retrospective study, mammary
gland OSA (MGOSA) was the most common site of ESOSA (108 of 169
cases) [103]. Of the remaining 61 cases, 10 were splenic, 13 originated within
the GI tract, 7 were in the urogenital tract, 6 were hepatic, 6 were cutaneous,
and 13 were in the subcutaneous tissues. In dogs, mixed malignant mam-
mary neoplasms (carcinosarcomas) that produce osteoid are fairly common,
but most arise in conjunction with adenocarcinomas [104]. Twenty of the
108 MGOSA patients had concurrent mammary gland tumors; 13 were
mixed adenomas, 6 were carcinomas, and 1 was a cystadenoma [103].
Although MGOSA comprised the majority of ESOSA in the aforemen-
tioned study, others have observed the spleen to be a more common site
[105–108]. Whereas 1 large retrospective study identified only 1 ESOSA in
1480 cases of splenic disease in dogs, 2 more recent studies of non-
angiomatous and nonlymphomatous neoplasms of the canine spleen
reported a higher incidence of this neoplasm [106–108]. Of these studies, 1
identified 8 cases of splenic ESOSA among 87 cases of primary splenic
mesenchymal tumors [106]. Another group reported 10 ESOSA out of 57
cases of splenic neoplasia [108]. In addition to the spleen and mammary
glands, two smaller retrospective studies of ESOSA in dogs described lung,
skin, axilla, mesenteric root, adrenal gland, eye, testicle, vagina, kidney,
intestine, gastric ligament, and liver as other primary sites [104,105]. Older
dogs are usually affected, with mean and median ages of about 10 and 11
years, respectively [103,105]. Although smaller studies reported more
females than males, this has not been borne out in larger studies noting
that ESOSA has no sex predilection [103]. All reported cases of MGOSA
were in females (75% intact, 25% neutered) [103,105]. Though Beagles and

509

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

Rottweilers were at higher risk for ESOSA, Miniature Poodles and German
Shepherds were at higher risk specifically for MGOSA [103].

History and physical examination

Reported clinical signs associated with ESOSA vary depending on the

location of the tumor and ranged from depression and lethargy to a distended
abdomen, mammary masses, difficulty breathing, weight loss, anorexia,
pyrexia, polyuria and polydipsia, vomiting, constipation, and adipsia [103,
105]. Duration of signs extended from 1 day to months before presentation.
Physical examination changes depend upon the site of the primary tumor.

Risk factors and associations

Beagles and Rottweilers have a 3.8 and 3.6 odds ratio, respectively, for

developing ESOSA [103]. Miniature Poodle and German Shepherd dogs
have a 2.7 and 2.2 odds ratio, respectively, for developing MGOSA [103].
Rare cases of esophageal OSA have been associated with Spirocerca lupi
infection [109]. Granulomas arise because of the migration and persistent
presence of larvae and adults within the esophagus; malignant trans-
formation of these lesions may then occur. Metastatic disease at the time
of initial presentation is a poor prognostic factor with ESOSA. The
significance of elevated serum alkaline phosphatase levels in ESOSA is
unknown.

Diagnosis

Although no pathognomonic features will be revealed, a minimum

database is recommended to assess overall health status before recommend-
ing and initiating treatment. Radiographs of the thorax are indicated to
screen for metastatic disease. Thoracic radiographs made in 32

/61 cases of

ESOSA revealed lesions in 3

/32 at the time of diagnosis; 38/108 cases of

MGOSA had thoracic radiographs made, and 3

/38 had lesions at diagnosis

[103]. Abdominal radiographs and ultrasonography are indicated depending
on the site of the primary tumor. Many other neoplasms may be differentials
for MGOSA and ESOSA, making biopsy or fine needle aspiration cytology
essential for diagnosis.

Biologic behavior

Similar to the more common forms of OSA, ESOSA, and MGOSA are

locally invasive and metastatic to lungs and other sites [103–106,108].
Lymph node involvement appears to be more common in ESOSA than in
primary bone OSA, being identified in 5

/11 dogs with ESOSA in a variety

510

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

of locations and in 2

/7 dogs with MGOSA [103,104]. Liver metastases,

diagnosed in 7

/14 dogs with visceral or pulmonary primary sites of OSA, are

also more commonly reported for ESOSA than for disease of appendicular
or axial locations [105].

Treatment and prognosis

Only 83 of 169 cases had follow-up to death in 1 retrospective study [103].

A total of 72 out of 83 were euthanized or died because of the tumor. Dogs
with ESOSA had a median survival of 26 days, with death from suspected
local recurrence in 92%. Dogs with MGOSA had a median survival of 90
days with death from metastatic disease in 62.5% [103]. Because all of the
reports of ESOSA and MGOSA are retrospective, patients received a variety
of treatments. Based on the known biologic behavior of this tumor, we
recommend surgical resection, if possible, followed by a platinum- or
doxorubicin-based chemotherapy protocol, although improved survival
times with such adjuvant therapy remain to be reported.

References

[1] Gillett NA, Pool RR, Taylor GN, et al. Strontium-90 induced bone tumors in beagle dogs:

Effects of route of exposure and dose rate. Int J Radiat Biol 1992;61:821–31.

[2] Gillette SM, Gillette EL, Powers BE, et al. Radiation-induced osteosarcoma in dogs after

external beam or intraoperative radiation therapy. Cancer Res 1990;50:54–7.

[3] Pool RR, Williams RJ, Goldman M. Induction of tumors involving bone in beagles fed

toxic levels of strontium 90. Am J Roentgenol Radium Ther Nucl Med 1973;118:900–8.

[4] Shyr LJ, Muggenburg BA. A comparison of the predicted risks of developing osteo-

sarcoma for dogs exposed to 238PuO2 based on average bone dose or endosteal cell dose.
Radiat Res 1992;132:13–8.

[5] Thurman GB, Mays CW, Taylor GN, et al. Growth dynamics of beagle osteosarcomas.

Growth 1971;35:119–25.

[6] White RA, Jefferies AR, Gorman NT. Sarcoma development following irradiation of

acanthomatous epulis in two dogs. Vet Rec 1986;118:668.

[7] Thrall DE. Orthovoltage radiotherapy of acanthomatous epulides in 39 dogs. J Am Vet

Med Assoc 1984;184:826–9.

[8] LaRue S, Wrigley R, Powers B. A review of the effects of radiation therapy on bone. Vet

Radiol Ultrasound 1987;28:17–22.

[9] Ansari MM. Bone infarcts associated with malignant sarcomas. The Compend

Continuing Ed Small Anim Pract 1991;13:367–70.

[10] Dubielzig RR, Biery DN, Brodey RS. Bone sarcomas associated with multifocal

medullary bone infarction in dogs. J Am Vet Med Assoc 1981;179:64–8.

[11] Marcellin-Little DJ, DeYoung DJ, Thrall DE, et al. Osteosarcoma at the site of bone

infarction associated with total hip arthroplasty in a dog. Vet Surg 1999;28:54–60.

[12] Riser WH, Brodey RS, Biery DN. Bone infarctions associated with malignant bone

tumors in dogs. J Am Vet Med Assoc 1972;160:414–21.

[13] Sebestyen P, Marcellin-Little DJ, DeYoung BA. Femoral medullary infarction secondary

to canine total hip arthroplasty. Vet Surg 2000;29:227–36.

[14] Gellasch KL, Kalscheur VL, Clayton MK, et al. Fatigue microdamage in the radial

predilection site for osteosarcoma in dogs. Am J Vet Res 2002;63:896–9.

511

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

[15] Cooley DM, Beranek BC, Schlittler DL, et al. Endogenous gonadal hormone exposure

and bone sarcoma risk. Cancer Epidemiol Biomarkers Prev 2002;11:1434–40.

[16] Johnson AS, Couto CG, Weghorst CM. Mutation of the p53 tumor suppressor gene in

spontaneously occurring osteosarcomas of the dog. Carcinogenesis 1998;19:213–7.

[17] Levine RA, Fleischli MA. Inactivation of p53 and retinoblastoma family pathways in

canine osteosarcoma cell lines. Vet Pathol 2000;37:54–61.

[18] Mendoza S, Konishi T, Dernell WS, et al. Status of the p53, Rb and MDM2 genes in

canine osteosarcoma. Anticancer Res 1998;18:4449–54.

[19] Setoguchi A, Sakai T, Okuda M, et al. Aberrations of the p53 tumor suppressor gene in

various tumors in dogs. Am J Vet Res 2001;62:433–9.

[20] Levine RA, Forest T, Smith C. Tumor suppressor PTEN is mutated in canine

osteosarcoma cell lines and tumors. Vet Pathol 2002;39:372–8.

[21] Kochevar DT, Kochevar J, Garrett L. Low level amplification of c-sis and c-myc in

a spontaneous osteosarcoma model. Cancer Lett 1990;53:213–22.

[22] Levine RA. Overexpression of the sis oncogene in a canine osteosarcoma model. Vet

Pathol 2002;39:411–2.

[23] Ferracini R, Angelini P, Cagliero E, et al. MET oncogene aberrant expression in canine

osteosarcoma. J Orthop Res 2000;18:253–6.

[24] Kirpensteijn J, Timmermans-Sprang EP, van Garderen E, et al. Growth hormone gene

expression in canine normal growth plates and spontaneous osteosarcoma. Mol Cell
Endocrinol 2002;197:179–85.

[25] Jongeward SJ. Primary bone tumors. Vet Clin North Am Small Anim Pract 1985;

15:609–41.

[26] Brodey RS, Abt DA. Results of surgical treatment in 65 dogs with osteosarcoma. J Am

Vet Med Assoc 1976;168:1032–5.

[27] Kraegel SA, Madewell BR, Simonson E, et al. Osteogenic sarcoma and cisplatin

chemotherapy in dogs: 16 cases (1986–1989). J Am Vet Med Assoc 1991;199:1057–9.

[28] Mauldin GN, Matus RE, Withrow SJ, et al. Canine osteosarcoma. Treatment by

amputation versus amputation and adjuvant chemotherapy using doxorubicin and
cisplatin. J Vet Intern Med 1988;2:177–80.

[29] Misdorp W, Hart AA. Some prognostic and epidemiologic factors in canine osteo-

sarcoma. J Natl Cancer Inst 1979;62:537–45.

[30] Shapiro W, Fossum TW, Kitchell BE, et al. Use of cisplatin for treatment of appendicular

osteosarcoma in dogs. J Am Vet Med Assoc 1988;192:507–11.

[31] Spodnick GJ, Berg J, Rand WM, et al. Prognosis for dogs with appendicular osteo-

sarcoma treated by amputation alone: 162 cases (1978–1988). J Am Vet Med Assoc
1992;200:995–9.

[32] Hammer AS, Weeren FR, Weisbrode SE, et al. Prognostic factors in dogs with

osteosarcomas of the flat or irregular bones. J Am Anim Hosp Assoc 1995;31:321–6.

[33] Ogilvie GK, Straw RC, Jameson VJ, et al. Evaluation of single-agent chemotherapy for

treatment of clinically evident osteosarcoma metastases in dogs: 45 cases (1987–1991).
J Am Vet Med Assoc 1993;202:304–6.

[34] Ehrhart N, Dernell WS, Hoffmann WE, et al. Prognostic importance of alkaline

phosphatase activity in serum from dogs with appendicular osteosarcoma: 75 cases (1990–
1996). J Am Vet Med Assoc 1998;213:1002–6.

[35] Garzotto CK, Berg J, Hoffmann WE, et al. Prognostic significance of serum alkaline

phosphatase activity in canine appendicular osteosarcoma. J Vet Intern Med 2000;
14:587–92.

[36] Bergman PJ, MacEwen EG, Kurzman ID, et al. Amputation and carboplatin for

treatment of dogs with osteosarcoma: 48 cases (1991 to 1993). J Vet Intern Med
1996;10:76–81.

[37] Powers BE, Withrow SJ, Thrall DE, et al. Percent tumor necrosis as a predictor of

treatment response in canine osteosarcoma. Cancer 1991;67:126–34.

512

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

[38] Berg J, Weinstein MJ, Springfield DS, et al. Results of surgery and doxorubicin

chemotherapy in dogs with osteosarcoma. J Am Vet Med Assoc 1995;206:1555–60.

[39] Forrest LJ, Dodge RK, Page RL, et al. Relationship between quantitative tumor

scintigraphy and time to metastasis in dogs with osteosarcoma. J Nucl Med 1992;
33:1542–7.

[40] Kirpensteijn J, Kik M, Rutteman G, et al. Prognostic significance of a new histologic

grading system for canine osteosarcoma. Vet Path 2002;39:240–6.

[41] Kirpensteijn J, Teske E, Kik M, et al. Lobaplatin as an adjuvant chemotherapy to surgery

in canine appendicular osteosarcoma: a phase II evaluation. Anticancer Res 2002;
22:2765–70.

[42] Coomber BL, Denton J, Sylvestre A, et al. Blood vessel density in canine osteosarcoma.

Can J Vet Res 1998;62:199–204.

[43] Dernell W, Straw R, Withrow S. Tumors of the skeletal system. In: MacEwen E, editor.

Small animal clinical oncology, 3rd edition. Philadelphia: W.B. Saunders; 2001. p. 378–417.

[44] Liptak J, Dernell W, Lascelles B, et al. Survival analysis of dogs with appendicular

osteosarcoma treated with limb sparing surgery and adjuvant carboplatin or carboplatin
and doxorubicin. Presented at Veterinary Cancer Society 21st Annual Conference. Baton
Rouge, LA, 2001.

[45] Thompson JP, Fugent MJ. Evaluation of survival times after limb amputation, with and

without subsequent administration of cisplatin, for treatment of appendicular osteosar-
coma in dogs: 30 cases (1979–1990). J Am Vet Med Assoc 1992;200:531–3.

[46] O’Brien MG, Straw RC, Withrow SJ, et al. Resection of pulmonary metastases in canine

osteosarcoma: 36 cases (1983–1992). Vet Surg 1993;22:105–9.

[47] Berg J, Lamb CR, O’Callaghan MW. Bone scintigraphy in the initial evaluation of dogs

with primary bone tumors. J Am Vet Med Assoc 1990;196:917–20.

[48] Forrest LJ, Thrall DE. Bone scintigraphy for metastasis detection in canine osteosarcoma.

Vet Radiol Ultrasound 1994;35:124–30.

[49] Hahn KA, Hurd C, Cantwell HD. Single-phase methylene diphosphate bone scintigraphy

in the diagnostic evaluation of dogs with osteosarcoma. J Am Vet Med Assoc
1990;196:1483–6.

[50] Jankowski M, Steyn P, Lana S, et al. Clinical sensitivity and specificity of nuclear

scanning with 99mTc-HDP for osseous metastasis in dogs with osteosarcoma. Presented
at Veterinary Cancer Society 21st Annual Conference. Baton Rouge, LA, 2001.

[51] Davis GJ, Kapatkin AS, Craig LE, et al. Comparison of radiography, computed

tomography, and magnetic resonance imaging for evaluation of appendicular osteosar-
coma in dogs. J Am Vet Med Assoc 2002;220:1171–6.

[52] Leibman NF, Kuntz CA, Steyn PF, et al. Accuracy of radiography, nuclear scintigraphy,

and histopathology for determining the proximal extent of distal radius osteosarcoma in
dogs. Vet Surg 2001;30:240–5.

[53] Wallack ST, Wisner ER, Werner JA, et al. Accuracy of magnetic resonance imaging for

estimating intramedullary osteosarcoma extent in pre-operative planning of canine limb-
salvage procedures. Vet Radiol Ultrasound 2002;43:432–41.

[54] Baker R, Lumsden J. The musculoskeletal system. In: Duncan L, editor. Color atlas of

cytology of the dog and cat. St. Louis, MO: Mosby; 1999. p. 199–207.

[55] Dernell WS, Van Vechten BJ, Straw RC, et al. Outcome following treatment of vertebral

tumors in 20 dogs (1986–1995). J Am Anim Hosp Assoc 2000;36:245–51.

[56] Berg J, Weinstein MJ, Schelling SH, et al. Treatment of dogs with osteosarcoma by

administration of cisplatin after amputation or limb-sparing surgery: 22 cases (1987–
1990). J Am Vet Med Assoc 1992;200:2005–8.

[57] Straw RC, Withrow SJ, Richter SL, et al. Amputation and cisplatin for treatment of

canine osteosarcoma. J Vet Intern Med 1991;5:205–10.

[58] Kuntz CA, Asselin TL, Dernell WS, et al. Limb salvage surgery for osteosarcoma of the

proximal humerus: outcome in 17 dogs. Vet Surg 1998;27:417–22.

513

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

[59] LaRue SM, Withrow SJ, Powers BE, et al. Limb-sparing treatment for osteosarcoma in

dogs. J Am Vet Med Assoc 1989;195:1734–44.

[60] Buracco P, Morello E, Martano M, et al. Pasteurized tumoral autograft as a novel

procedure for limb sparing in the dog: a clinical report. Vet Surg 2002;31:525–32.

[61] Degna MT, Ehrhart N, Feretti A, et al. Bone transport osteogenesis for limb salvage. Vet

Comp Orthop Traumatol 2000;13:18–22.

[62] Rovesti GL, Bascucci M, Schmidt K, et al. Limb sparing using a double bone-

transport technique for treatment of a distal osteosarcoma in a dog. Vet Surg 2002;31:
70–7.

[63] Seguin B, Walsh PJ, Mason DR, et al. Use of an ipsilateral vascularized ulnar

transposition autograft for limb-sparing surgery of the distal radius in dogs: an anatomic
and clinical study. Vet Surg 2003;32:69–79.

[64] Bailey D, Erb H, Williams L, et al. Preliminary evaluation of carboplatin and doxorubicin

combination chemotherapy for treatment of appendicular osteosarcoma. Presented at
Veterinary Cancer Society 21st Annual Conference. Baton Rouge, LA, 2001.

[65] Berg J, Gebhardt MC, Rand WM. Effect of timing of postoperative chemotherapy on

survival of dogs with osteosarcoma. Cancer 1997;79:1343–50.

[66] Chun R, Garrett L, Henry C, et al. Cisplatin and doxorubicin combination chemotherapy

for the treatment of canine osteosarcoma. Presented at Veterinary Cancer Society 21st
Annual Conference. Baton Rouge, LA, 2001.

[67] Chun R, Kurzman ID, Couto CG, et al. Cisplatin and doxorubicin combination

chemotherapy for the treatment of canine osteosarcoma: a pilot study. J Vet Intern Med
2000;14:495–8.

[68] Hudson MM, Snyder JS, Jaffe N, et al. In vitro and in vivo effect of adriamycin therapy

on monocyte activation by liposome-encapsulated immunomodulators. Cancer Res
1988;48:5256–63.

[69] Kleinerman ES, Snyder JS, Jaffe N. Influence of chemotherapy administration on

monocyte activation by liposomal muramyl tripeptide phosphatidylethanolamine in
children with osteosarcoma. J Clin Oncol 1991;9:259–67.

[70] Kurzman ID, MacEwen EG, Rosenthal RC, et al. Adjuvant therapy for osteosarcoma in

dogs: Results of randomized clinical trials using combined liposome-encapsulated
muramyl tripeptide and cisplatin. Clin Cancer Res 1995;1:1595–601.

[71] Shi F, MacEwen EG, Kurzman ID. In vitro and in vivo effect of doxorubicin combined

with liposome-encapsulated muramyl tripeptide on canine monocyte activation. Cancer
Res 1993;53:3986–91.

[72] Khanna C, Prehn J, Hayden D, et al. A randomized controlled trial of octreotide pamoate

long-acting release and carboplatin versus carboplatin alone in dogs with naturally oc-
curring osteosarcoma: evaluation of insulin-like growth factor suppression and che-
motherapy. Clin Cancer Res 2002;8:2406–12.

[73] Vail DM, Kurzman ID, Glawe PC, et al. STEALTH liposome-encapsulated cisplatin

(SPI-77) versus carboplatin as adjuvant therapy for spontaneously arising osteosarcoma
(OSA) in the dog: a randomized multicenter clinical trial. Cancer Chemother Pharmacol
2002;50:131–6.

[74] Zachos TA, Aiken SW, DiResta GR, et al. Interstitial fluid pressure and blood flow in

canine osteosarcoma and other tumors. Clin Orthop 2001;385:230–6.

[75] Green EM, Adams WM, Forrest LJ. Four fraction palliative radiotherapy for osteo-

sarcoma in 24 dogs. J Am Anim Hosp Assoc 2002;38:445–51.

[76] McEntee MC, Page RL, Novotney CA, et al. Palliative radiotherapy for canine

appendicular osteosarcoma. Vet Radiol Ultrasound 1993;34:367–70.

[77] Ramirez 3rd O, Dodge RK, Page RL, et al. Palliative radiotherapy of appendicular

osteosarcoma in 95 dogs. Vet Radiol Ultrasound 1999;40:517–22.

[78] Tomlin JL, Sturgeon C, Pead MJ, et al. Use of the bisphosphonate drug alendronate for

palliative management of osteosarcoma in two dogs. Vet Rec 2000;147:129–32.

514

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

[79] Fromigue´ O, Body JJ. Bisphosphonates influence the proliferation and the maturation of

normal human osteoblasts. J Endocrinol Invest 2002;25:539–46.

[80] Mackie PS, Fisher JL, Zhou H, et al. Bisphosphonates regulate cell growth and gene

expression in the UMR 106–01 clonal rat osteosarcoma cell line. Br J Cancer 2001;84:951–8.

[81] Reinholz GG, Getz B, Pederson L, et al. Bisphosphonates directly regulate cell pro-

liferation, differentiation, and gene expression in human osteoblasts. Cancer Res 2000;
60:6001–7.

[82] Sonnemann J, Eckervogt V, Truckenbrod B, et al. The bisphosphonate pamidronate is

a potent inhibitor of human osteosarcoma cell growth in vitro. Anticancer Drugs
2001;12:459–65.

[83] Viereck V, Emons G, Lauck V, et al. Bisphosphonates pamidronate and zoledronic acid

stimulate osteoprotegerin production by primary human osteoblasts. Biochem Biophys
Res Commun 2002;291:680–6.

[84] Bateman KE, Catton PA, Pennock PW, et al. 0–7-21 radiation therapy for the palliation

of advanced cancer in dogs. J Vet Intern Med 1994;8:394–9.

[85] McEntee MC. Radiation therapy in the management of bone tumors. Vet Clin North Am

Small Anim Pract 1997;27:131–8.

[86] Aas M, Moe L, Gamlem H, et al. Internal radionuclide therapy of primary osteosarcoma

in dogs, using 153Sm-ethylene-diamino-tetramethylene-phosphonate (EDTMP). Clin
Cancer Res 1999;5:3148s–52s.

[87] Lattimer JC, Corwin LA, Jr, Stapleton J, et al. Clinical and clinicopathologic response of

canine bone tumor patients to treatment with Samarium-153-EDTMP. J Nucl Med
1990;31:1316–25.

[88] Milner RJ, Dormehl I, Louw WKA, et al. Targeted radiotherapy with Sm-153-EDTMP in

nine cases of canine primary bone tumors. J South African Vet Assoc 1998;69:12–7.

[89] Moe L, Boysen M, Aas M, et al. Maxillectomy and targeted radionuclide therapy with

153Sm-EDTMP in a recurrent canine osteosarcoma. J Small Anim Pract 1996;37:241–6.

[90] Straw RC, Powers BE, Klausner J, et al. Canine mandibular osteosarcoma: 51 cases

(1980–1992). J Am Anim Hosp Assoc 1996;32:257–62.

[91] Heyman SJ, Diefenderfer DL, Goldschmidt MH, et al. Canine axial skeletal osteo-

sarcoma: a retrospective study of 116 cases (1986–1989). Vet Surg 1992;21:304–10.

[92] Matthiesen DT, Clark GN, Orsher RJ, et al. En bloc resection of primary rib tumors in 40

dogs. Vet Surg 1992;21:201–4.

[93] Pirkey-Ehrhart N, Withrow SJ, Straw RC, et al. Primary rib tumors in 54 dogs. J Am

Anim Hosp Assoc 1995;31:65–9.

[94] Cooley DM, Waters DJ. Skeletal neoplasms of small dogs: a retrospective study and

literature review. J Am Anim Hosp Assoc 1997;33:11–23.

[95] Levy MS, Kapatkin AS, Patnaik AK, et al. Spinal tumors in 37 dogs: clinical outcome and

long-term survival (1987–1994). J Am Anim Hosp Assoc 1997;33:307–12.

[96] Dickerson ME, Page RL, LaDue TA, et al. Retrospective analysis of axial skeleton

osteosarcoma in 22 large-breed dogs. J Vet Intern Med 2001;15:120–4.

[97] White R. Mandibulectomy and maxillectomy in the dog: long term survival in 100 cases.

J Small Anim Pract 1991;32:69–74.

[98] Schwarz P, Withrow S, Curtis C, et al. Partial maxillary resection as a treatment for oral

cancer in 61 dogs. J Am Anim Hosp Assoc 1991;27:617–24.

[99] Kosovsky JK, Matthiesen DT, Marretta SM, et al. Results of partial mandibulectomy for

the treatment of oral tumors in 142 dogs. Vet Surg 1991;20:397–401.

[100] Wallace J, Matthiesen DT, Patnaik AK. Hemimaxillectomy for the treatment of oral

tumors in 69 dogs. Vet Surg 1992;21:337–41.

[101] Moore GE, Mathey WS, Eggers JS, et al. Osteosarcoma in adjacent lumbar vertebrae in

a dog. J Am Vet Med Assoc 2000;217:1038–40, 1008.

[102] Feeney DA, Johnston GR, Grindem CB, et al. Malignant neoplasia of canine ribs:

clinical, radiographic, and pathologic findings. J Am Vet Med Assoc 1982;180:927–33.

515

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

[103] Langenbach A, Anderson MA, Dambach DM, et al. Extraskeletal osteosarcomas in dogs:

a retrospective study of 169 cases (1986–1996). J Am Anim Hosp Assoc 1998;34:113–20.

[104] Patnaik AK. Canine extraskeletal osteosarcoma and chondrosarcoma: a clinicopathologic

study of 14 cases. Vet Pathol 1990;27:46–55.

[105] Kuntz CA, Dernell WS, Powers BE, et al. Extraskeletal osteosarcomas in dogs: 14 cases.

J Am Anim Hosp Assoc 1998;34:26–30.

[106] Spangler W, Culbertson M, Kass P. Primary mesenchymal (nonangiomatous

/non-

lymphomatous) neoplasms occurring in the canine spleen: anatomic classification, im-
munohistochemistry, and mitotic activity correlated with patient survival. Vet Pathol
1994;31:37–47.

[107] Spangler WL, Culbertson MR. Prevalence, type, and importance of splenic diseases in

dogs: 1,480 cases (1985–1989). J Am Vet Med Assoc 1992;200:829–34.

[108] Weinstein MJ, Carpenter JL, Mehlhaff Schunk CJ. Nonangiogenic and nonlymphoma-

tous sarcomas of the canine spleen: 57 cases (1975–1987). J Am Vet Med Assoc
1989;195:784–8.

[109] Mazaki-Tovi M, Baneth G, Aroch I, et al. Canine spirocercosis: clinical, diagnostic,

pathologic, and epidemiologic characteristics. Vet Parasitol 2002;107:235–50.

[110] Dernell WS, Withrow SJ, Straw RC, et al. Adjuvant chemotherapy using cisplatin by

subcutaneous administration. In Vivo 1997;11:345–50.

[111] Hahn KA, Richardson RC, Blevins WE, et al. Intramedullary cisplatin chemotherapy:

experience in four dogs with osteosarcoma. J Small Anim Pract 1996;37:187–92.

[112] Hershey AE, Kurzman ID, Forrest LJ, et al. Inhalation chemotherapy for macroscopic

primary or metastatic lung tumors: proof of principle using dogs with spontaneously
occurring tumors as a model. Clin Cancer Res 1999;5:2653–9.

[113] Khanna C, Anderson PM, Hasz DE, et al. Interleukin-2 liposome inhalation therapy is safe

and effective for dogs with spontaneous pulmonary metastases. Cancer 1997;79:1409–21.

[114] MacEwen EG, Kurzman ID. Canine osteosarcoma: amputation and chemoimmunother-

apy. Vet Clin North Am Small Anim Pract 1996;26:123–33.

[115] Schwarz P, Withrow S, Curtis C, et al. Mandibular resection as a treatment for oral cancer

in 81 dogs. J Am Anim Hosp Assoc 1991;27:601–10.

[116] Visonneau S, Cesano A, Jeglum KA, et al. Adjuvant treatment of canine osteosarcoma

with the human cytotoxic T-cell line TALL-104. Clin Cancer Res 1999;5:1868–75.

516

R. Chun, L.-P. de Lorimier

/ Vet Clin Small Anim 33 (2003) 491–516

background image

Medical management of soft tissue

sarcomas

Kenneth M. Rassnick, DVM

Comparative Cancer Program, Cornell University, College of Veterinary Medicine,

Box 31, Ithaca, NY 14853, USA

Soft tissue sarcomas (STS) represent a heterogeneous family of

malignancies that arise from mesenchymal tissues. The most common
histological types in veterinary patients include fibrosarcoma, peripheral
nerve sheath tumor (schwannoma and neurofibrosarcoma), malignant
fibrous histiocytoma, and hemangiopericytoma.

Soft tissue sarcomas are locally invasive and infiltrative along fascial

planes by way of tendril-like microextensions. Because conservative surgical
resection rarely removes the entire tumor, local recurrence is common. Dogs
with STS having incomplete surgical margins are more than 10 times more
likely to have local recurrence than dogs with complete tumor-free surgical
margins [1].

Until recently, the metastatic potential of STS was difficult to define. Many

older studies relied on conservative surgical excision alone to control these
tumors, and many patients died as a result of local recurrence soon after
surgery. Based on current information, the overall metastatic potential of
STS is fairly low. In dogs, reports of the metastatic rate of STS range from 8%
to 17% [1–3]. Table 1 summarizes the results of the metastatic rate of canine
STS in various studies. In one study of 75 dogs, 13 developed metastases to
lungs (8), lymph nodes (1), or lungs and lymph nodes (4). Median time to
detection of metastases was 365 days (range, 0 to 1444 days) [1].

Histopathologic grade is predictive of metastases and prognosis in dogs

with STS (Table 2). In the study by Kuntz et al [1], 4 of 31 (13%) dogs with
grade 1 tumors developed metastases, and only 2 of 27 (7%) with grade 2
tumors developed metastases, whereas 7 of 17 (41%) with grade 3 tumors
developed metastases. Overall median survival for dogs with STS treated by
surgery alone was 1416 days. Median survival for dogs with more than 19
mitotic figures per 10 high-power fields (HPF; n

¼ 11) was 236 days

Vet Clin Small Anim

33 (2003) 517–531

E-mail address:

kmr32@cornell.edu

0195-5616

/03/$ - see front matter

Ó 2003, Elsevier Inc. All rights reserved.

doi:10.1016

/S0195-5616(03)00019-6

background image

compared with 1444 days (n

¼ 46) for dogs with less than 10 mitotic figures

per 10 HPF and 532 days for dogs with 10 to 19 mitotic figures per 10 HPF
(n

¼ 18). Percentage of histologic necrosis was also prognostic for survival

time. Dogs with greater than 10% necrosis were 2.78 times more likely to die
of tumor-related causes than dogs with less than 10% necrosis [1].

The true metastatic potential of STS in cats is controversial. Reported

metastatic rates have ranged from 0% up to 25% [4–7]. Table 1 summarizes
the results of the metastatic rate of feline STS in various studies. Recent
studies of cats with vaccine-associated sarcomas have included cats with
radical surgical procedures and often adjunctive treatment such as radiation
therapy. Local tumor control in these recent studies has improved, which
may in turn increase survival sufficiently for occurrence and detection of

Table 1
Results of the metastatic rate of canine and feline soft tissue sarcomas in various studies

Investigator

Number evaluated

Patients
Metastases

Canine

Kuntz [1]

75 (all grades)

17%

31 (grade 1)

13%

27 (grade 2)

7%

17 (grade 3)

41%

Forrest [2]

35

14%

McKnight [3]

48

8%

Feline

Bregazzi [4]

25

0%

Cohen [5]

78

12%

Hershey [7]

61

23%

Davidson [6]

45

26%

Cronin [8]

33

27%

Table 2
Variables used to determine histologic grade of canine soft tissue sarcomas

Score

Degree of
differentiation

Mitotic figures
per 10 HPF

Necrosis

1

Resemble normal adult

mesenchymal tissue

<

10

None

2

Specific histologic type

10–19

<

50%

3

Undifferentiated

>

19

>

50%

Grade

Description

1

Cumulative score of <5

2

Cumulative score of 5 or 6

3

Cumulative score of >6

Abbreviation:

HPF, high-powered field.

Data from

Kuntz CA, Dernell WS, Powers BE, et al. Prognostic factors for surgical

treatment of soft tissue sarcomas in dogs: 75 cases (1986–1996). J Am Vet Med Assoc 1997;211:
1147–51.

518

K.M. Rassnick

/ Vet Clin Small Anim 33 (2003) 517–531

background image

metastases. Although metastases have been reported as early as 5 weeks
after surgery [8], feline STS metastases usually occur much later. Whether
histopathologic features predict prognosis for cats with STS is unclear.
Some investigators believe that the metastatic rate for vaccine-site sarcomas
may be higher than for sarcomas affecting other sites. This has not been
confirmed, however. When compared with nonvaccine sites, vaccine-
associated sarcomas typically have increased amounts of necrosis and
cavitations, peritumoral lymphoid aggregates, pleomorphism, multinucle-
ated giant cells, and increased mitotic activity [9,10]. Davidson et al
evaluated a histopathologic grading system based on mitotic activity and
differentiation [6]. When cats were treated by surgical excision alone, tumor
grade did not influence tumor-free interval or survival time significantly.

Fortunately, most dogs and cats with STS will present without clinically

evident metastases. For this reason, adequate surgical resection with or
without adjuvant external beam irradiation remains the primary therapy for
STS. The greatest survival rates have been achieved in studies involving
radical resection [1,6,7]. If tumors are removed incompletely, however,
survival rates attained by use of radiation therapy adjunct to incomplete
surgical excision can equal or exceed those attained by wide surgical excision
[2–5].

Which patients are candidates for chemotherapy?

The overall metastatic rate of STS in dogs and cats is fairly low (Table 1).

Aggressive surgical resection or the combination of surgery and radiation
therapy represents the best chance for control. Nevertheless, many patients
with STS have a poorer prognosis and fail to be cured. For example, despite
adequate local control, the tumor may have metastasized early in the course
of disease. Also, several histologic types are included in the broad category
of STS but differ from the most common types, either in location of
occurrence, or metastatic potential. Chemotherapy can be considered in the
following clinical situations:

1. Patients presenting with distant metastases.
2. Patients presenting with tumors that are inoperable because of tumor

size or location. In some patients, a measurable response to chemo-
therapy might allow for function-preserving surgical resection.

3. Patients with tumors that recur despite radical excision or radiation

therapy and are not amenable to retreatment.

4. Dogs presenting for treatment following surgical removal of a high-

grade (grade 3) STS.

5. Cats presenting for treatment following surgical removal or radiation

therapy for an injection-site sarcoma.

6. Dogs with hemangiosarcoma, synovial cell sarcoma, osteosarcoma, rhab-

domyosarcoma, lymphangiosarcoma, or liposarcoma. These histologic

519

K.M. Rassnick

/ Vet Clin Small Anim 33 (2003) 517–531

background image

types are included in the broad classification of STS but differ in their
metastatic potential. Readers should refer to appropriate literature to
determine specific features of histologic grade or clinical presentation that
may predict biological behavior.

7. Dogs and cats with STS of internal organs. Prognosis for patients with

STS of internal organs such as the spleen is often very poor. For
example, in one study of 65 dogs with splenic sarcomas, overall median
survival was 2 months [11]. Again, readers should refer to appropriate
literature to determine specific features of grade or location that may
correlate with prognosis.

8. Dogs with oral sarcomas. When treated with radiation therapy, median

survival for dogs with oral sarcomas is significantly shorter than that of
dogs with sarcomas at other sites (1.5 years versus 6.2 years) [2]. Also,
histologically low-grade, yet biologically high-grade sarcomas occur in
the oral cavity of dogs. Often they are misdiagnosed as fibromas or even
chronic inflammation. Despite deceptively benign histologic features,
these tumors tend to grow rapidly and invade deeper structures, includ-
ing bone. In one study of 25 dogs, 20% developed metastases to the
lungs or lymph nodes [12]. The metastatic rate in another report of dogs
with oral sarcomas was 54% (7 of 13) [13].

Single-agent chemotherapy

There are reports evaluating various chemotherapy agents to treat STS.

Most of these have been phase I and II studies or case reports and small case
series. There is a relative paucity of randomized trials, and many of these
have included only small numbers of patients. A further complicating factor
is the histologic heterogeneity of these tumors. With few exceptions, chemo-
therapy agents have not been evaluated against single histologic subtypes,
and studies enrolling all histologic types have not been large enough to allow
valid conclusions to be drawn from subgroup analysis. Although there are
relatively few agents with very impressive antitumor efficacy against STS in
dogs and cats, it is important not to overlook the potential clinical utility of
certain commonly available drugs (Table 3).

Doxorubicin

Doxorubicin is considered to be the most active single agent to treat STS.

In people, doxorubicin has been reported to induce objective response rates
varying from 15% to 34% against STS, the majority of which are partial
responses [14]. In vitro sensitivity to doxorubicin has been examined for
various canine cell lines, including sarcomas [15]. In the study by Macy et al,
specimens from 34 canine tumors were grown in agar gel and exposed to
doxorubicin. Of the seven sarcoma cell lines examined, one was susceptible
to doxorubicin [15].

520

K.M. Rassnick

/ Vet Clin Small Anim 33 (2003) 517–531

background image

The clinical antitumor effect of two intravenous (IV) doses of doxorubicin

(30 mg

/m

2

body surface area) 3 weeks apart has been documented in a multi-

institutional phase II study [16]. In that study, 157 dogs with nonlymphoid
malignant neoplasms were evaluated. Remissions were documented in 23%
(11 of 48) dogs with sarcomas. Complete responses (100% reduction in
tumor size) were observed in dogs with synovial cell sarcoma, undifferen-
tiated sarcoma, liposarcoma, and neurofibrosarcoma. Partial responses (at
least 50% reduction in tumor size) occurred in dogs with hemangiosarcoma,
osteosarcoma, synovial cell sarcoma, undifferentiated sarcoma, fibrosar-
coma, and one dog with an infiltrating lipoma [16].

Several investigators have evaluated the use of doxorubicin combined

with cyclophosphamide to treat malignancies [17–20]. Sorenmo et al treated
dogs with hemangiosarcoma every 3 weeks with doxorubicin (30 mg

/m

2

IV,

day 0) and cyclophosphamide (50 to 75 mg

/m

2

per os, days 2 to 5). Nine

dogs had measurable subcutaneous disease, and complete responses
occurred in two [18].

Further reports have documented the use of the doxorubicin

/cyclophos-

phamide combination. For example, the combination has been used to treat
dogs with synovial cell sarcoma. Tilmant treated a dog with synovial cell
sarcoma of the tarsus and noted a durable complete remission for greater than
3 years [19]. Vail treated two dogs with the combination, and neither tumor
responded [20].

The supplemental activity of cyclophosphamide in addition to doxorubi-

cin in the management of STS is controversial. For dogs with hemangio-
sarcoma, data suggest that single-agent doxorubicin may be equally effective
as protocols containing doxorubicin and cyclophosphamide or doxorubicin
with vincristine and cyclophosphamide [21]. Most studies continue to be
conducted with the premise that doxorubicin represents the most active agent
and is the basis upon which combinations they should be built.

Doxorubicin also is considered to be the most effective single antitumor

agent to treat STS in cats. Williams demonstrated significant in vitro dose-
dependent effects of doxorubicin on the viability of two vaccine-associated
feline sarcoma cell lines [22].

Table 3
Single-agent chemotherapy for canine and peline soft tissue sarcomas

Drug

Dosage

Route

Interval (days)

Canine

Doxorubicin

30 mg

/m

2

IV

14–21

a

Mitoxantrone

6 mg

/m

2

IV

21

Ifosfamide

375 mg

/m

2

IV

14–21

a,b

Feline

Doxorubicin

25 mg

/m

2

IV

21

Mitoxantrone

6.5 mg

/m

2

IV

21

Carboplatin

210 mg

/m

2

IV

21–28

a

Abbreviation:

IV, intravenous.

a

Dosing interval depends on adequate neutrophil recovery after treatment.

b

In conjunction with a specific protocol for diuresis and mesna administration.

521

K.M. Rassnick

/ Vet Clin Small Anim 33 (2003) 517–531

background image

Phase II evaluations of single-agent doxorubicin in cats are lacking. The

combination of doxorubicin and cyclophosphamide has been examined,
however. Mauldin studied the use of doxorubicin (25 mg

/m

2

IV day 0) and

cyclophosphamide (50 mg

/m

2

per os, days 2 to 5) in 23 cats with malignant

nonhematopoietic tumors. Two of three cats with nonresectable oral fibro-
sarcoma responded to treatment for 60 and 150 days, respectively [23].

Barber examined 12 cats, all with vaccine-associated sarcomas, treated

with the same chemotherapy combination. Overall response rate was 50% (6
of 12). Two of the 12 cats had major responses for 153 and 190 days,
respectively. Partial responses (at least 50%) occurred in four cats (33%).
Duration of partial response ranged from 41 to 144 days, with a median of
103 days. Of the seven cats in the study treated only with chemotherapy,
there was a significant improvement in survival for those cats that responded.
Median survival of three cats responding to doxorubicin

/cyclophosphamide

combination therapy was 242 days, compared with only 83 days for the four
cats failing to respond [24].

Mitoxantrone

Mitoxantrone is an anthracenedione compound related to anthracyclines

such as doxorubicin. In people, mitoxantrone is minimally active against
STS. In a study of 115 patients with STS, response rate was only 1% [14]. A
dose escalation study of mitoxantrone in tumor-bearing dogs showed
activity against STS. Ogilvie et al used mitoxantrone to treat 126 dogs with
measurable tumors. Dosages ranged from 2.5 to 5 mg

/m

2

IV. Further data

support that the maximally tolerated dosage of mitoxantrone in dogs is 6
mg

/m

2

. Four of the 12 dogs with sarcomas responded in this study when

treated with mitoxantrone at 5 mg

/m

2

. Dogs with fibrosarcoma, chondro-

sarcoma, and hemangiopericytoma had measurable tumor reductions for 21
to 167 days [25].

Further evaluation of mitoxantrone for STS has been conducted by

Henry et al. In their study, dogs were treated with mitoxantrone (5 mg

/m

2

IV, day 0) combined with cyclophosphamide (150 mg

/m

2

IV, day 0) and

granulocyte colony stimulating factor (G-CSF). All six dogs with measur-
able sarcomas treated with the combination failed to respond. Tumor tissues
from dogs enrolled in this study were tested in vitro for sensitivity to the
mitoxantrone

/cyclophosphamide combination. Although the assay seemed

to predict response to the drugs, there was no correlation to clinical re-
sponse, since no dogs had measurable reduction of tumor [26].

There have been limited evaluations of mitoxantrone to treat STS in cats.

As with doxorubicin, mitoxantrone had significant dose-dependent effects
on the in vitro viability of two vaccine-associated feline sarcoma cell lines
[22]. In a study to evaluate in vivo toxicity and efficacy of mitoxantrone, 7 of
87 cats had sarcomas [27]. Two of the seven cats had complete remissions
when treated with mitoxantrone. One cat with rhabdomyosarcoma had

522

K.M. Rassnick

/ Vet Clin Small Anim 33 (2003) 517–531

background image

a durable complete remission for 120 days, and one cat with fibrosarcoma
had a complete remission for 90 days [27].

Ifosfamide

Ifosfamide is an oxazophosphorine nitrogen mustard developed as an

isomer of the alkylating agent cyclophosphamide. As a result of minor
structural differences, ifosfamide has a different spectrum of clinical anti-
tumor activity and toxicity than cyclophosphamide. Numerous clinical trials
in people have demonstrated that ifosfamide has major antitumor activity
against STS. Several phase II trials of ifosfamide in people with STS have
demonstrated response rates of 25% to 30% in untreated patients and 15% to
18% in patients previously treated with doxorubicin [28]. There has only been
one study to evaluate ifosfamide in tumor-bearing dogs. The efficacy and
toxicity of ifosfamide were evaluated in 72 dogs with spontaneously occurring
tumors [29]. A dosage of 375 mg

/m

2

was found to be safe in dogs. Treatments

can be given intravenously every 2 to 3 weeks, depending on hematopoietic
recovery. A specific protocol for saline fluid diuresis to prevent renal toxicity
and mesna injections to prevent urothelial toxicity was used. In that study, 13
dogs had measurable sarcomas. Two of the 13 dogs had durable complete
remissions. One dog with metastatic cutaneous hemangiosarcoma responded
for more than 445 days after three consecutive treatments. The other dog had
metastatic leiomyosarcoma of the urinary bladder. Complete regression of
metastatic lymph nodes occurred after treatment with ifosfamide, and the dog
died of other causes more than 500 days later [29].

Phase I and II studies are underway to evaluate ifosfamide in cats

(Rassnick, unpublished data). Preliminary results indicate ifosfamide is safe
in cats, although the dosage and spectrum of toxicity are much different
than that seen in dogs. As with dogs and people, ifosfamide appears active
against STS in cats. Although studies are still ongoing, complete and partial
responses have occurred in cats with injection-site sarcomas treated with
ifosfamide (Rassnick, unpublished data).

Other single-agent chemotherapy options

Other cytotoxic drugs may be useful to treat STS in dogs and cats.

Vincristine has been demonstrated to be effective in 15% of people with STS
[14]. In vitro, vincristine has significant dose-dependent effects on the viability
of four vaccine-associated feline sarcoma cell lines [30]. There have been a few
reports of STS-bearing dogs and cats treated with vincristine. Hahn reported
a complete remission for 210 days in a cat with an oral fibrosarcoma treated
solely with vincristine [31]. Vincristine was not successful in the treatment of
metastatic hemangiosarcoma in another cat, however [32].

Dacarbazine (DTIC) is an antitumor alkylating agent. In people, it often

is used with doxorubicin as first-line therapy against sarcomas [14]. There

523

K.M. Rassnick

/ Vet Clin Small Anim 33 (2003) 517–531

background image

have been anecdotal suggestions that dacarbazine may have efficacy against
sarcomas in dogs.

Actinomycin-D is an important component of chemotherapy in the

treatment of pediatric rhabdomyosarcoma [14]. Information on actinomy-
cin-D as a single-agent in veterinary patients is limited, however. Hammer
evaluated actinomycin-D in 34 dogs. Two dogs in that study had measur-
able sarcomas, and neither dog had a positive response to treatment with
actinomycin-D [33].

Docetaxel is occasionally effective against sarcomas in people. Response

rate is generally less than 15%, so use of the drug is limited [14]. Docetaxel
caused significant inhibition of four vaccine-associated feline sarcoma cell
lines when tested in vitro [30]. There are no published reports of the use of
systemic docetaxel in dogs and cats. Polysorbate-80 is an inorganic solvent
necessary for IV docetaxel administration, and when given to dogs, severe
acute hypersensitivity reactions occur.

Finally, platinum-derived agents including cisplatin and carboplatin hold

some promise of clinical activity for the treatment of STS. In dogs, cisplatin
has significant activity against bone sarcomas. The single-agent response
rate against other histologic subtypes has been poorly evaluated, however
[34]. Preliminary evidence to evaluate carboplatin in cats with STS suggests
activity; however, controlled studies are not available. Two cats that failed
treatment with doxorubicin subsequently were treated with carboplatin, and
no objective responses occurred [24].

New chemotherapy agents and approaches

Because of the relative lack of therapeutic efficacy and significant toxicity

of existing chemotherapy protocols, new agents, delivery methods, and
supportive measures to improve chemotherapy for STS are always areas
of research interest. For example, the most serious adverse effects of
doxorubicin are myelosuppression and cardiotoxicity, and several strategies
have been used to limit these toxicities. Doxil, a pegylated liposomal form of
doxorubicin, is one such novel formulation. Liposomes containing small
fractions of polyethylene-glycol (PEG)-phospholipid (so-called ‘‘stealth’’
liposomes) avoid mononuclear phagocyte sequestration and result in
improved circulation time and a different spectrum of toxicities. In a study
by Vail et al, 16 dogs with sarcomas were treated with Doxil. Responses were
observed in five dogs (31%), including complete remissions in dogs with
hemangiosarcoma and malignant fibrous histiocytoma and partial remissions
in dogs with anaplastic sarcoma, fibrosarcoma, and neurofibrosarcoma [35].

FCE 23762-methoxymorpholino-doxorubicin is another new doxorubi-

cin analog. The drug was evaluated in 15 dogs with various STS including
osteosarcoma, undifferentiated sarcoma, and lymphangiosarcoma. Three
dogs in this study achieved remission in response to treatment [36].

524

K.M. Rassnick

/ Vet Clin Small Anim 33 (2003) 517–531

background image

Inhalational delivery of chemotherapy offers several potential advantages

over oral and parenteral administration. These include locoregional delivery
to the lungs, avoidance of first pass metabolic degradation by the liver, and
possibly fewer systemic adverse effects. Hershey et al used an aerosol devise
designed to deliver paclitaxel and a new formation of doxorubicin to dogs
with primary and metastatic lung tumors. Two of three dogs with sarcomas
had measurable responses when treated with inhalational chemotherapy [37].

Another approach to limit systemic toxicities of drugs is through the use

of intratumoral or intracavitary implantation. OPLA (open cell polylactic
acid) is a porous biodegradable solid polymer that is impregnated with
cisplatin (OPLA-Pt). This local form of therapy can be placed into the wound
bed following resection of a tumor. Cisplatin concentrations within the
wound far exceed those attainable by IV administration, while obviating the
need for high systemic concentrations, thus avoiding systemic toxicities.
Dernell et al placed OPLA-Pt into the wound following marginal (histo-
logically incomplete) resection of STS in dogs. Local recurrence developed in
10 of 32 (31%) sites, possibly similar to the rate of recurrence after radiation
therapy. High tumor grade was associated with local tumor recurrence.
Unfortunately, OPLA-Pt had to be removed from 9 of 32 (28%) sites because
of local wound complications. This complication rate indicates the need for
further refinement of the polymer

/cisplatin system before the full therapeutic

potential of this agent can be evaluated [38].

Combination and sequential chemotherapy

Several cytotoxic drugs have been identified to have single-agent clinical

activity against STS (Table 4). Clinical oncology has a long tradition of
mixing and matching individual chemotherapy drugs with documented
single-agent activities to construct chemotherapy protocols combining or
alternating drugs (sequential chemotherapy) on a biweekly or triweekly
schedule. Most studies have been undertaken with the premise that doxo-
rubicin represents the most active drug and is therefore the basis upon which
combination or sequential therapy should be built.

The combination of doxorubicin and cyclophosphamide for treatment of

STS has been reported frequently. The supplemental activity of cyclophos-
phamide in addition to doxorubicin in the management of STS remains
controversial. For example, in canine hemangiosarcoma, data suggest that
single-agent doxorubicin may be equally effective as protocols containing
doxorubicin with cyclophosphamide. Definitive determination is not possible
in the absence of appropriately designed prospective randomized studies,
however [21].

Doxorubicin, cyclophosphamide, and vincristine (VAC protocol) have

been used extensively for adjunctive therapy of canine hemangiosarcoma
after surgery. Dogs are treated with doxorubicin (30 mg

/m

2

IV, day 0),

cyclophosphamide (100 to 150 mg

/m

2

per os, day 0), and with vincristine

525

K.M. Rassnick

/ Vet Clin Small Anim 33 (2003) 517–531

background image

(0.75 mg

/m

2

, days 7 and 14). The entire cycle is repeated every 21 days. In five

dogs with measurable hemangiosarcoma, no objective responses occurred;
however, the authors reported a median survival time of 172 days for the
15 dogs treated with the protocol [39]. Objective reductions in STS tumor
size when treated with VAC have been reported by other investigators.
For example, an atrial hemangiosarcoma responded for 16 weeks, and a
rhabdomyosarcoma of the bladder transiently responded for three weeks
[40,41]. Neutropenia, sepsis, and enterocolitis are significant adverse effects
of this protocol. Strict monitoring, appropriate supportive care, and overall
clinical caution are advised [17].

Because ifosfamide has been identified as an agent with antitumor

activity against STS, the author analyzed a biweekly, alternating protocol of
ifosfamide and doxorubicin in dogs. Dogs received ifosfamide (375 mg

/m

2

IV) with saline diuresis and mesna on day 0 and doxorubicin (30 mg

/m

2

IV)

on day 14. This cycle was repeated twice for a total of three doses of each
drug. The protocol was tolerated by dogs, but efficacy against STS has not
been determined (Rassnick, unpublished data).

Ifosfamide has activity against feline STS (Rassnick, unpublished data),

but protocols to combine or alternate ifosfamide with other agents have not
been evaluated in cats.

A protocol alternating doxorubicin and carboplatin can be given to cats

safely. Cats can receive doxorubicin at 25 mg

/m

2

IV on day 0 and carboplatin

at 190 mg

/m

2

IV on day 21. The cycle can be repeated twice for a total of

three doses of each drug. Careful hematologic monitoring is warranted,
especially after treatment with carboplatin because of the potential for
prolonged neutropenia. Treatment may need to be delayed if neutrophil
recovery is not adequate before the next scheduled treatment. Investigation

Table 4
Combination and sequential chemotherapy options for canine and feline soft tissue sarcomas

Protocol

Drug

Dosage

Route Day

Cycle
interval

Number
of cycles

Canine AC

Doxorubicin

30 mg

/m

2

IV

0

21 days

5

Cyclophosphamide

75 mg

/m

2

PO

2–5

VAC

Doxorubicin

30 mg

/m

2

IV

0

21 days

5

Cyclophosphamide

100–150

mg

/m

2

PO

0

Vincristine

0.75 mg

/m

2

IV

7, 14

Adria

/Ifos

Doxorubicin

30 mg

/m

2

IV

0

28 days

3

Ifosfamide

a

375 mg

/m

2

IV

14

Feline

AC

Doxorubicin

20 mg

/m

2

IV

0

21 days

5

Cyclophosphamide

100 mg

/m

2

IV

0

Adria

/Carbo Doxorubicin

25 mg

/m

2

IV

0

42 days

3

Carboplatin

190 mg

/m

2

IV

21

Abbreviations:

IV, intravenous; PO, by mouth.

a

In conjunction with a specific protocol for diuresis and mesna administration.

526

K.M. Rassnick

/ Vet Clin Small Anim 33 (2003) 517–531

background image

of the utility of this protocol to treat cats with STS is underway (Rassnick,
unpublished data).

Practical use of chemotherapy

Patients with measurable disease

Adequate surgical excision with or without radiation therapy remains the

principle treatment modality for dogs and cats with STS. Patients that may
qualify for chemotherapy, however, include those that present with dis-
tant metastatic disease, develop local recurrence despite aggressive local
therapies, or have nonresectable tumors.

Neoadjuvant or primary chemotherapy (chemotherapy before treatment

of the primary tumor with surgery or radiation therapy) increasingly has
been accepted as a modality that may assist in limiting the degree of radical
surgery. For patients that achieve a measurable response to preoperative
chemotherapy, a function-preserving surgical excision may be feasible.
Although neoadjuvant chemotherapy appears attractive in some situations,
caution remains appropriate, because one might do more harm to patients
by excessively decreasing local interventions aimed at achieving optimal
rates of local control. For example, if a tumor shrinks significantly in
response to neoadjuvant chemotherapy, what should the surgical oncologist
plan as the volume of resection? Additionally, what volume of tissue should
be irradiated to obtain local control postoperatively? Additional experience
from clinical trials studying neoadjuvant chemotherapy and multimodality
approaches will be necessary before answers to these questions are clear.

In the measurable disease setting, generally two consecutive cycles of

single-agent chemotherapy are administered. If no response is observed, then
a different chemotherapeutic should be considered. Response to therapy is
determined by measurable reduction (at least 50%) in size or extent of
disease. Tumors that do not show evidence of growth beyond 25% of the
original tumor volume, or volume reduction beyond 50%, are considered to
be in a state of ‘‘stable disease.’’ Because of the variable biologic behavior of
STS with regard to patterns of growth, it is difficult to determine whether
disease stabilization in a given case is the result of treatment, or rather caused
by the natural course of tumor progression. Clinicians faced with stable
disease in the face of therapy must decide on a case-by-case basis whether the
chemotherapy should be continued. Randomized, prospective trials are
needed to establish the clinical significance and patient benefit achieved by
disease stabilization in light of toxicity risks and therapy costs.

Adjuvant chemotherapy

The principle objective of adjuvant chemotherapy following sur-

gical removal of the primary tumor or radiation therapy is to destroy

527

K.M. Rassnick

/ Vet Clin Small Anim 33 (2003) 517–531

background image

micrometastatic disease and improve overall survival. The incorporation of
adjuvant chemotherapy in the management of canine appendicular osteo-
sarcoma and hemangiosarcoma is accepted. The clinically significant benefits
of adjuvant chemotherapy in the treatment of STS have not been clearly
established, however.

Although the response rates of most drugs in the advanced disease setting

is mild to moderate, adjuvant chemotherapy is based on the premise that
a reduced tumor burden might be eradicated more easily. In dogs, available
data suggest that low-grade STS is best managed with local therapies alone,
and that mortality caused by tumor in this population is low. In contrast, the
prognosis for patients with high-grade lesions is considerably worse, and
most of these patients ultimately die from disease. The biologic behavior
of oral sarcomas and of histologic STS subtypes such as osteosarcoma,
hemangiosarcoma, synovial cell sarcoma, and rhabdomyosarcoma may be
more aggressive. Early institution of chemotherapy may be indicated for
these patients, but continued investigation of systemic adjuvant therapies
is necessary. Commonly used adjuvant chemotherapy protocols for dogs
with STS include single-agent doxorubicin (five treatments), doxorubicin
combined with cyclophosphamide, VAC, or doxorubicin alternating with
ifosfamide (see Tables 1 and 2).

Options for adjuvant chemotherapy in cats with STS include single-agent

doxorubicin (five treatments), doxorubicin combined with cyclophospha-
mide, and doxorubicin alternating with carboplatin (see Tables 1 and 2). As
is the case with dogs, the use of adjuvant chemotherapy to treat cats with
STS remains controversial. In one study of 25 cats with vaccine-associated
sarcomas, 18 were treated with surgery and radiation therapy (58 Gy)
followed by doxorubicin (1 mg

/kg IV every 3 weeks for five treatments), and

seven cats were treated with surgery and radiation therapy only. The median
survival for the chemotherapy group was 674 days versus 842 days for the
group without chemotherapy. There was no significant difference between the
groups. The power of the study (5%) was low, however, and more cats would
be needed in each treatment group to detect a difference, should one exist [4].

In another study of 76 cats with vaccine-associated sarcomas, 26 received

adjuvant chemotherapy of doxorubicin (20 mg

/m

2

IV) and cyclophos-

phamide (100 mg

/m

2

IV) every 3 weeks. As with the Bregazzi study [4], no

advantage for the chemotherapy group was detected. Recurrence rate
(41%), rate of metastasis (12%), and survival (730 days) for cats that
received chemotherapy were not significantly different from values of cats
that did not receive chemotherapy [5]. Also, as in the Bregazzi study,
analysis was limited. Although 26 cats received chemotherapy, many re-
ceived treatment after a recurrence or development of metastases. Power to
detect a difference and sample size was very small [5].

The role of adjuvant chemotherapy to treat STS in dogs and cats remains

undetermined. Further study is necessary to provide objective support for
the use of adjuvant chemotherapy in patients with resected sarcomas and to

528

K.M. Rassnick

/ Vet Clin Small Anim 33 (2003) 517–531

background image

determine which regimens are most appropriate. The magnitude of any
expected benefit and the costs, both financial and in terms of toxicity, also
must be weighed before global recommendations can be made.

Summary

Local therapies for STS, including aggressive surgical resection and

radiation therapy, have improved dramatically over the last 10 years. This
has resulted in improvement of important clinical outcomes such as en-
hanced rates of local control and higher quality of life for patients. Systemic
therapy for STS, although efficacious to a certain degree, remains
investigational. A major task for oncologists will be to measure accurately
the single-agent activity of drugs and to integrate promising agents into
multiagent regimens. This approach may allow significant progress in the
management of STS in canine and feline patients.

References

[1] Kuntz CA, Dernell WS, Powers BE, et al. Prognostic factors for surgical treatment of soft

tissue sarcomas in dogs: 75 cases (1986–1996). J Am Vet Med Assoc 1997;211:1147–51.

[2] Forrest LJ, Chun R, Adams WM, et al. Postoperative radiotherapy for canine soft tissue

sarcoma. J Vet Intern Med 2000;14:578–82.

[3] McKnight JA, Mauldin GN, McEntee MC, et al. Radiation treatment for incompletely

resected soft tissue sarcomas in dogs. J Am Vet Med Assoc 2000;217:205–10.

[4] Bregazzi VS, LaRue SM, McNiel E, et al. Treatment with a combination of doxorubicin,

surgery, and radiation versus surgery and radiation alone for cats with vaccine-associated
sarcomas: 25 cases (1995–2000). J Am Vet Med Assoc 2001;218:547–50.

[5] Cohen M, Wright JC, Brawner WR, et al. Use of surgery and electron beam irradiation,

with or without chemotherapy, for treatment of vaccine-associated sarcomas in cats: 78
cases (1996–2000). J Am Vet Med Assoc 2001;219:1582–9.

[6] Davidson EB, Gregory CR, Kass PH. Surgical excision of soft tissue fibrosarcomas in cats.

Vet Surg 1997;26:265–9.

[7] Hershey AE, Sorenmo KU, Hendrick MJ, et al. Prognosis for presumed feline vaccine-

associated sarcoma after excision: 61 cases (1986–1996). J Am Vet Med Assoc 2000;
216:58–61.

[8] Cronin K, Page RL, Spodnick G, et al. Radiation therapy and surgery for fibrosarcoma in

33 cats. Vet Radiol Ultrasound 1998;39:51–6.

[9] Couto SS, Griffey SM, Duarte PC, et al. Feline vaccine-associated fibrosarcoma:

morphologic distinctions. Vet Pathol 2002;39:33–41.

[10] Doddy FD, Glickman LT, Glickman NW, et al. Feline fibrosarcomas at vaccination sites

and nonvaccination sites. J Comp Pathol 1996;114:165–74.

[11] Spangler WL, Culbertson MR, Kass PH. Primary mesenchymal (nonangiomatous

/

nonlymphomatous) neoplasms occurring in the canine spleen: anatomic classification,
immunohistochemistry, and mitotic activity correlated with patient survival. Vet Pathol
1994;31:37–47.

[12] Ciekot PA, Powers BE, Withrow SJ, et al. Histologically low-grade yet biologically high-

grade fibrosarcomas of the mandible and maxilla of 25 dogs (1982 to 1991). J Am Vet Med
Assoc 1994;204:610–5.

[13] Schwarz PD, Withrow SJ, Curtis CR, et al. Mandibular resection as a treatment for oral

cancer in 81 dogs. J Am Anim Hosp Assoc 1991;27:601–10.

529

K.M. Rassnick

/ Vet Clin Small Anim 33 (2003) 517–531

background image

[14] Demetri GD, Elias AD. Results of single-agent and combination chemotherapy for

advanced soft tissue sarcomas. Hematol Oncol Clin North Am 1995;9:765–86.

[15] Macy DW, Ensley BA, Gillette EL. In vitro susceptibility of canine tumor stem cells to

doxorubicin. Am J Vet Res 1988;49:1903–5.

[16] Ogilvie GK, Reynolds HA, Richardson RC, et al. Phase II evaluation of doxorubicin for

treatment of various canine neoplasms. J Am Vet Med Assoc 1989;195:1580–3.

[17] Ahaus EA, Couto CG, Valerius KD. Hematological toxicity of doxorubicin-containing

protocols in dogs with spontaneously occurring malignant tumors. J Am Anim Hosp Assoc
2000;36:422–6.

[18] Sorenmo KU, Jeglum KA, Helfand SC. Chemotherapy of canine hemangiosarcoma with

doxorubicin and cyclophosphamide. J Vet Intern Med 1993;7:370–6.

[19] Tilmant LL, Gorman NT, Ackerman N, et al. Chemotherapy of synovial cell sarcoma in

a dog. J Am Vet Med Assoc 1986;188:530–2.

[20] Vail DM, Powers BE, Getzy DM, et al. Evaluation of prognostic factors for dogs with

synovial sarcoma: 36 cases (1986–1991). J Am Vet Med Assoc 1994;205:1300–7.

[21] Ogilvie GK, Powers BE, Mallinckrodt CH, et al. Surgery and doxorubicin in dogs with

hemangiosarcoma. J Vet Intern Med 1996;10:379–84.

[22] Williams LE, Banerji N, Klausner JS, et al. Establishment of two vaccine-associated feline

sarcoma cell lines and determination of in vitro chemosensitivity to doxorubicin and
mitoxantrone. Am J Vet Res 2001;62:1354–7.

[23] Mauldin GN, Matus RE, Patnaik AK, et al. Efficacy and toxicity of doxorubicin and

cyclophosphamide used in the treatment of selected malignant tumors in 23 cats. J Vet
Intern Med 1988;2:60–5.

[24] Barber LG, Sorenmo KU, Cronin KL, et al. Combined doxorubicin and cyclophospha-

mide chemotherapy for nonresectable feline fibrosarcoma. J Am Anim Hosp Assoc 2000;
36:416–21.

[25] Ogilvie GK, Obradovich JE, Elmslie RE, et al. Efficacy of mitoxantrone against various

neoplasms in dogs. J Am Vet Med Assoc 1991;198:1618–21.

[26] Henry CJ, Buss MS, Potter KA, et al. Mitoxantrone and cyclophosphamide combination

chemotherapy for the treatment of various canine malignancies. J Am Anim Hosp Assoc
1999;35:236–9.

[27] Ogilvie GK, Moore AS, Obradovich JE, et al. Toxicoses and efficacy associated with

administration of mitoxantrone to cats with malignant tumors. J Am Vet Med Assoc
1993;202:1839–44.

[28] Connelly EF, Budd GT. Ifosfamide in the treatment of soft tissue sarcomas. Semin Oncol

1996;23:16–21.

[29] Rassnick KM, Frimberger AE, Wood CA, et al. Evaluation of ifosfamide for treatment of

various canine neoplasms. J Vet Intern Med 2000;14:271–6.

[30] Banerji N, Li X, Klausner JS, et al. Evaluation of in vitro chemosensitivity of vaccine-

associated feline sarcoma cell lines to vincristine and paclitaxel. Am J Vet Res 2002;
63:728–32.

[31] Hahn KA. Vincristine sulfate as single-agent chemotherapy in a dog and a cat with

malignant neoplasms. J Am Vet Med Assoc 1990;197:796–8.

[32] Kraje AC, Mears EA, Hahn KA, et al. Unusual metastatic behavior and clinicopathologic

findings in eight cats with cutaneous or visceral hemangiosarcoma. J Am Vet Med Assoc
1999;214:670–2.

[33] Hammer AS, Couto CG, Ayl RD, et al. Treatment of tumor-bearing dogs and cats with

Actinomycin D. J Vet Intern Med 1994;8:236–9.

[34] Knapp DW, Richardson RC, Bonney PL, et al. Cisplatin therapy in 41 dogs with

malignant tumors. J Vet Intern Med 1988;2:41–6.

[35] Vail DM, Kravis LD, Cooley AJ, et al. Preclinical trial of doxorubicin entrapped in

sterically stabilized liposomes in dogs with spontaneously arising malignant tumors.
Cancer Chemother Pharmacol 1997;39:410–6.

530

K.M. Rassnick

/ Vet Clin Small Anim 33 (2003) 517–531

background image

[36] Sheafor SE, Couto CG, Ward H, et al. Clinical evaluation of methoximorpholino-

doxorubicin (FCE 23762) in dogs with spontaneous malignancies. J Vet Intern Med
2000;14:86–9.

[37] Hershey AE, Kurzman ID, Forrest LJ, et al. Inhalation chemotherapy for macroscopic

primary or metastatic lung tumors: proof of principle using dogs with spontaneously
occurring tumors as a model. Clin Cancer Res 1999;5:2653–9.

[38] Dernell WS, Withrow SJ, Straw RC, et al. Intracavitary treatment of soft tissue sarcomas

in dogs using cisplatin in a biodegradable polymer. Anticancer Res 1997;17:4499–505.

[39] Hammer AS, Couto CG, Filppi J, et al. Efficacy and toxicity of VAC chemotherapy

(vincristine, doxorubicin, and cyclophosphamide) in dogs with hemangiosarcoma. J Vet
Intern Med 1991;5:160–6.

[40] deMadrone E, Helfand SC, Stebbins KE. Use of chemotherapy for treatment of cardiac

hemangiosarcoma in a dog. J Am Vet Med Assoc 1987;190:887–91.

[41] Van Vechten M, Goldschmidt M, Wortman JA. Embryonal rhabdomyosarcoma of the

urinary bladder in dogs. Compendium on Continuing Education for the Practicing
Veterinarian 1990;12:783–93.

531

K.M. Rassnick

/ Vet Clin Small Anim 33 (2003) 517–531

background image

Hemangiosarcoma in dogs and cats

Annette N. Smith, DVM, MS

Department of Clinical Sciences, Veterinary Teaching Hospital,

Wire Road, Auburn University, AL 36849, USA

Incidence and patient factors

Hemangiosarcoma (HSA, including angiosarcoma and malignant he-

mangioendothelioma) is a highly malignant tumor derived from the
endothelial cell line and is characterized by early and aggressive metastasis
[1–15]. HSA is a common tumor type in dogs, comprising up to 5% to 7%
of noncutaneous primary malignant neoplasms [4,7–12,16–18]. In other
species, the incidence is much lower; in cats, it is reported as 0.5% to 2%
[19–22].

HSA is usually seen in older animals, although there are sporadic reports

of the tumor occurring in younger dogs and cats (C.M. Johannes et al,
unpublished data) [8,9,11,23–25]. The mean age of occurrence ranges from 8
to 13 years in the dog [1,8,9,11,26] and from 8 to 10.5 years in the cat (C.M.
Johannes et al, unpublished data) [19,21,25,27].

The breed that appears to be predisposed is the German Shepherd Dog,

also known as the Alsatian [9–12,26], although this breed was also the most
popular dog at the time of these studies. Other commonly reported breeds
include the Golden Retriever, Pointer, Boxer, Labrador Retriever, English
Setter, Great Dane, Poodle, and Siberian Husky [1,4,28]. Any large-breed
dog appears to be at increased risk [26,29,30]. Lightly pigmented, sparsely
haired dogs (Beagles, Bloodhounds, White Bulldogs, English Pointers,
Salukis, Dalmatians, and Whippets) have also been reported as predisposed
to the development of cutaneous and subcutaneous HSAs [24]. No specific
breeds have been identified in the cat as having a predisposition for HSA
development; most tumors are seen in the domestic shorthair cat
(C.M. Johannes et al, unpublished data) [19,21,25,27,31]. Cats with unpig-
mented skin may be predisposed to cutaneous tumor development in those
areas [31].

Vet Clin Small Anim

33 (2003) 533–552

E-mail address:

smith30@auburn.edu

0195-5616

/03/$ - see front matter

Ó 2003, Elsevier Inc. All rights reserved.

doi:10.1016/S0195-5616(03)00002-0

background image

Sex predilection has been reported for male dogs [8,12], but some studies

have indicated no difference in occurrence between male and female dogs
[4,17], and in two studies, spayed female dogs were reported to be at
increased risk [11,32]. Because of the conflicting reports, some oncologists
have concluded that there is no true sex predilection [33,34]. One study
suggested a female predilection for splenic HSA in the cat [21], although the
male-to-female ratio was only 1:1.25 in 18 cats. Conversely, another study of
31 cats with HSA had more male cats than female cats affected, with a ratio
of 1.9:1 [25]. In a study of cutaneous vascular tumors, 12 of 15 cats were
male, and the authors suggested a male predilection [31].

Etiology

Cutaneous HSAs have been associated with ultraviolet light exposure in

dogs [24,35]. These tumors often arise on the ventral abdomen, where the
hair coat is sparse [24,36]. Breeds at risk are those with a light hair coat and
poor pigmentation, and the usual sites of occurrence are the prepuce and
ventral abdomen on glabrous skin [24]. Subcutaneous or hypodermal HSAs
do not seem to be associated with ultraviolet light exposure [24].

Other radiation sources can cause visceral HSA development. In dogs

exposed to intraoperative radiation therapy, dogs that received greater than
20 Gy seemed to be at more risk for sarcoma development [37]. One dog
developed an HSA of the bladder after 30 Gy had been administered to the
area 3.5 years previously [37]. Dogs that were exposed to aerosolized

144

Ce

or

90

SrC

2

developed primary pulmonary, bone, and liver HSAs at a high

rate and usually died of their tumor [38].

A breed predisposition suggests a genetic etiology. German Shepherd

Dogs are the most commonly reported breed with HSA [1,4,8,9,
12,15,18,26,39–42]. Other large-breed dogs are also overrepresented, espe-
cially Golden and Labrador Retrievers [5,18,41,42]. No specific genetic
mutations have been associated with HSA, although few genetic abnormal-
ities have been studied. In one study [43], the tumor suppressor protein p53
was investigated in feline tumors. Only one of seven HSAs demonstrated
p53 nuclear staining, with no evidence of cytoplasmic signals, suggesting
mutation of this gene is uncommon in feline HSA [43]. A progression of
cutaneous disease from hemangioma to HSA may occur, [24] reflecting
phenotypic development of more malignant tumor as a result of repeated
genetic damage. Subcutaneous or hypodermal HSAs do not seem to have
this progression [13,24].

The role of reproductive status and, therefore, hormonal status in dogs

and cats remains unclear at this time, although in one study of atrial HSA
[32], spayed female dogs had a greater than five times risk of developing this
tumor, and most cutaneous vascular tumors in cats have been reported in
male cats [31].

534

A.N. Smith

/ Vet Clin Small Anim 33 (2003) 533–552

background image

Currently, few causes of feline cutaneous or visceral HSA have been

suggested. Sunlight exposure may be important in tumor development in
hypopigmented areas, especially of the head and pinnae [31]. Feline
leukemia virus infection has not been associated with this tumor’s devel-
opment [6].

Other carcinogens that have been linked to the development of HSAs in

people include thorium dioxide, arsenicals, and vinyl chloride [44,45], and
methylnitrosamine has been reported to cause HSA development in mink
[8]. These compounds could possibly be extrapolated to cause tumorigenesis
in companion animals.

Biologic behavior

HSA can arise in any tissue with blood vessels, but the most common

sites in dogs are the spleen (50%–65%), right atrium (3%–25%), sub-
cutaneous tissues (13%–17%), and liver (5%–6%) in the dog [1,4,9,16,46].
In the cat, the liver, spleen, mesentery, omentum, and subcutaneous tissues
are the most frequent primary sites (C.M. Johannes et al, unpublished data)
[19,21,25,47]. Other primary sites reported include the skin, lung, aorta,
kidney, oral cavity, muscle, bone, urinary bladder, intestine, tongue, pro-
state, vulva

/vagina, conjunctiva, and peritoneum in dogs [1,8,10,36,

48–52] and the nasal cavity, intestine, muscle, diaphragm, pancreas, lung,
kidney, bone, eye, and thoracic cavity in cats (C.M. Johannes et al,
unpublished data) [13,21,25,53–55]. HSA is the most common canine
primary cardiac tumor [4,16,32,56]. HSA is reported to comprise 2% to 3%
of all canine bone tumors [57] and 4% to 10.5% of primary bone tumors
in the vertebral column [58]. Some have suggested that HSA may be
overdiagnosed in bone tumors, however, because it can be difficult to dif-
ferentiate a highly vascular area in another primary bone tumor from
HSA [48].

Overt metastasis is present in more than 80% of canine patients at clinical

presentation [5]. Splenic and atrial masses coexist in 25% of dogs with HSA
[15], although whether one site is primary or whether multicentric HSAs
have developed [3,8,9] is usually undetermined. Tumor spread to the liver,
omentum, mesentery, and lung [1,4,6,13,15,16,32] is most common in dogs,
although metastases have also been found in the kidney, muscle, peri-
toneum, lymph nodes, bone, adrenal glands, eye, prostate, and brain [1,5,6,
9,13,14,32,48,59]. HSA is the most common metastatic sarcoma in the
canine brain and is usually found in a cerebral location; 14% of patients
were reported to have brain metastasis in a study of 85 dogs with HSA [59].
Other metastatic sites were usually also present in these patients with brain
metastases [59]. In cats, metastatic disease can be found in the liver, intra-
abdominal lymph nodes, and lung (C.M. Johannes et al, unpublished data)
[25,27].

535

A.N. Smith

/ Vet Clin Small Anim 33 (2003) 533–552

background image

Death from treated visceral HSA is usually a result of metastatic disease

rather than local tumor recurrence [5]. Rupture of untreated atrial or
visceral organ tumors can also cause death as a result of cardiac tamponade
or intracavitary hemorrhage [4,5].

In cats, subcutaneous HSAs have a high recurrence rate after surgical

excision (C.M. Johannes et al, unpublished data) [19,25,31]. Cutaneous
HSAs (confined to the dermis) may be easier to excise completely, and
recurrence is much less frequent (C.M. Johannes et al, unpublished data).
The incidence of metastasis is reported to be low in cutaneous HSA in dogs
and cats (C.M. Johannes et al, unpublished data) [24,25,31], but some
patients develop metastatic disease [24,27,60]. Conjunctival HSA also seems
to be more locally invasive than metastatic, because six dogs with follow-up
information ranging from 6 to 17 months had no evidence of recurrence or
metastasis after surgical intervention [61]. HSAs with hypodermal or
muscular involvement in dogs and cats seem to have a high recurrence rate
and incidence of metastasis (C.M. Johannes et al, unpublished data)
[25,31,60].

Clinical presentation

Clinical signs can be subtle in dogs with HSA, usually related to episodic

weakness or acute collapse associated with hemorrhage from a visceral
mass, followed by recovery when the blood is reabsorbed from the body
cavity [2,4,8,46]. Other signs associated with hypovolemia caused by
hemorrhage might include tachycardia, tachypnea, and mucous membrane
pallor [15,46]. Abdominal distention, anorexia, and weight loss may also
be noted [5]. If the bleeding tumor is associated with the heart, signs of
right heart failure caused by cardiac tamponade might include abdominal
distention, jugular pulses, muffled heart sounds, and dyspnea [4,16,56].
Syncope, ataxia, and cyanosis can be seen with severe arrhythmias [4,62].
Subcutaneous masses can cause lameness or may be a primary complaint,
depending on tumor location and possibly the presence of active hemor-
rhage [3]. Primary osseous HSAs in the long bones can induce lameness
secondary to pathologic fractures or bone pain [29,48], and vertebral HSAs
can cause pain or paresis [58]. Active bleeding from tumor sites in locations
such as the urinary tract or nasal cavity may cause an owner to seek
veterinary attention [25,49,52]. Seizures can occur with HSA metastasis to
the brain [4,59]. Hemorrhage or petechiae formation on mucous membranes
in the presence of disseminated intravascular coagulation may also occur in
rare instances [7,63]. Finally, HSA that ruptures acutely and causes sudden
death may be an incidental finding at postmortem examination [5,46].

Cats with HSA also have a subtle clinical presentation, although the

onset of signs is generally acute (C.M. Johannes et al, unpublished data)
[21]. Subcutaneous or cutaneous masses with or without hemorrhage are

536

A.N. Smith

/ Vet Clin Small Anim 33 (2003) 533–552

background image

a common complaint (C.M. Johannes et al, unpublished data) [25,27].
Visceral HSAs can cause lethargy, anorexia, vomiting, abdominal disten-
tion, weight loss, and possibly dyspnea [25,27,64]. A palpable abdominal
mass and pallor can be found on physical examination [25]. Mesenteric root
HSAs can cause chylous ascites as a result of obstruction

/disruption of the

lymphatic vessels [64].

Diagnosis and staging

A clinical staging system for canine HSA and tests for diagnosing

/staging

HSA are found in Boxes 1 and 2. Complete blood cell count (CBC)
abnormalities in dogs most commonly include evidence of a regenerative
anemia, including anisocytosis, polychromasia, increased red blood cell
(RBC) distribution width, and reticulocytosis [4,7]. Acanthocytosis has been
seen in up to 50% of dogs with HSA [40,65]. With microangiopathic
changes, schistocytes can also be seen. Nucleated red blood cells (nRBCs)
are often not removed from circulation by an abnormal spleen, elevating
their numbers in peripheral blood [4]. Other causes of increased circulating
nRBCs include hypoxemia, bone marrow infiltration by tumor, and extra-
medullary hematopoiesis [4]. A neutrophilic leukocytosis can also be found

Box 1. Clinical staging system for canine hemangiosarcoma

Primary tumor (T)
T0: no tumor evident
T1: tumor less than 5 cm in diameter, confined to organ, and

does not invade beyond dermis (cutaneous HSA)

T2: tumor greater than or equal to 5 cm in diameter or ruptured

or invasive into subcutaneous tissues (cutaneous HSA)

T3: tumor invades adjacent structures

Regional lymph nodes (N)
N0: no regional node involvement
N1: regional node involvement
N2: distant node involvement

Distant metastasis (M)
M0: no distant metastasis found
M1: distant metastasis confirmed

Stage I: T0 or T1, N0, M0

Stage II: T1 or T2, N0 or N1, M0

Stage III: T2 or T3, N0, N1, or N2, M1

537

A.N. Smith

/ Vet Clin Small Anim 33 (2003) 533–552

background image

secondary to stress, tumor necrosis, or nonspecific bone marrow response
[1,4,7,14]. Thrombocytopenia is a common finding in dogs, occurring in
30% to 60% of dogs with HSA, secondary to immune-mediated processes,
sequestration, severe hemorrhage, or disseminated intravascular coagula-
tion (DIC) [66].

CBC abnormalities in cats usually include anemias and neutrophilic

leukocytosis (C.M. Johannes et al, unpublished data) [21,25,27]. Thrombo-
cytopenia can also occur (C.M. Johannes et al, unpublished data) [21,25,27].
The presence of nRBCs is rare [25], and fragmentation of RBCs has not
been reported in the cat [5].

Spontaneous hemorrhage may occur secondary to severe thrombocyto-

penias and DIC, which is often seen with canine HSA [63,66–68]. DIC is
associated with thrombocytopenia, elevations in prothrombin and partial
thromboplastin times, decreases in fibrinogen and antithrombin III levels,
and elevations in fibrin degradation products. DIC can be systemic or
localized to the tumor area [69]. DIC may resolve with therapy. The most
important treatment for DIC is tumor removal, but combinations of
heparin, plasma, or corticosteroids may prove beneficial [63,68,69]. Cats
with visceral HSA have also been reported to be predisposed to DIC [27].

Serum chemistry abnormalities are usually not specific for HSA but can

reflect organ system involvement. For example, serum alkaline phosphatase
and alanine transferase elevations may reflect infiltration of the liver.
Hypoglycemia has been reported as a paraneoplastic syndrome [70].

Radiographs of the abdomen and thorax can demonstrate pleural or

peritoneal effusion secondary to hemorrhage or right heart failure asso-
ciated with cardiac tamponade [4]. In 47% of dogs with cardiac HSA, an
abnormal cardiac silhouette can be seen [41]. Radiographic abnormalities
can include a globoid cardiac appearance secondary to pericardial effusion
or a soft tissue mass at the heart base [4,41].

Three-view thoracic radiographs (right and left lateral views plus

a ventrodorsal or dorsoventral view) are also necessary to evaluate the

Box 2. Tests for diagnosis/staging of hemangiosarcoma

Complete blood cell count
Serum chemistry panel
Urinalysis
Coagulation profile (prothrombin time, partial thromboplastin

time, fibrinogen, and fibrin degradation products (FDPs), with
or without antithrombin III)

Three-view thoracic radiographs
Abdominal ultrasound
Echocardiogram
Electrocardiography

538

A.N. Smith

/ Vet Clin Small Anim 33 (2003) 533–552

background image

lung fields for evidence of metastatic disease for the highest detection
sensitivity [41]. Both a nodular pattern and a diffuse miliary pattern are
consistent with metastatic HSA, and the nodules can be ill defined (most
frequently) or well circumscribed (less frequently) [71]. Occasionally, an
alveolar pattern consistent with intrapulmonary hemorrhage is seen [71].
Thoracic radiography has a sensitivity of 80% in detecting pulmonary
metastatic disease [41,71].

Abdominal radiographs may demonstrate splenomegaly, hepatomegaly,

or other intra-abdominal masses consistent with primary or metastatic HSA.
Generalized loss of detail often indicates hemoperitoneum [34]. Other
imaging should be performed as needed based on clinical signs, including
contrast studies or cystoscopy for the detection of renal or bladder masses
[49,52]. Osseous HSA usually has a lytic rather than proliferative radio-
graphic appearance, is often confined to the medullary cavity, and may
involve the entire shaft of the bone [29]. MRI is preferred, and CT scans may
also be used to confirm the presence of mass lesions within the cerebrum as
the cause of seizures in dogs with HSA. These advanced imaging modalities
are also useful for the diagnosis of subcutaneous masses and for planning
surgical and radiation therapy.

Pneumopericardiography and angiocardiography (selective and nonselec-

tive) have been used in the past to document space-occupying cardiac masses
[72], but these invasive procedures have largely been replaced by echocardi-
ography [73,74]. Echocardiography is considered more accurate than
radiography in cardiac HSA diagnosis and has been reported to have
a positive predictive value of 92% (11 of 12 cases) and a negative predictive
value of 64% (9 of 14 cases) in dogs with right atrial

/auricular cardiac masses

[73]. Some have suggested that nonselective angiography is a relatively simple,
safe, and quick procedure useful for imaging the right heart and delineating
masses, especially if echocardiography is equivocal or unavailable [75].
Endomyocardial biopsy has been used in a single case to diagnose a left
ventricular HSA [76], but this site is rare, and this invasive technique is not
used routinely to diagnose right auricular

/atrial masses.

Electrocardiographic abnormalities usually include nonlethal ventricular

arrhythmias associated with splenic disease rather than cardiac tumor
involvement [77,78]. Nevertheless, one report of right bundle branch block
caused by invasion of HSA locally into the right ventricle illustrates the
possibility of arrhythmias as a result of local tumor involvement of the
cardiac conduction system [62]. Pericardial effusion can result in decreased
complex amplitude or electric alternans [28]. Up to 40% of dogs with splenic
HSA have arrhythmias, including ventricular premature contractions
and ventricular tachycardia [78]. Arrhythmias may not be noted until
after splenectomy [78]. Pre-, peri-, and postsurgical arrhythmias have also
been reported in dogs with cardiac HSA, including atrial tachycardias [16].
Antiarrhythmic drug therapy may be necessary in some cases, and
anesthetic regimens should be chosen accordingly [16,78].

539

A.N. Smith

/ Vet Clin Small Anim 33 (2003) 533–552

background image

Splenic HSA cannot be definitively diagnosed on the basis of clinical

signs. Splenic hematomas and hemangiomas as well as other benign diseases
have a similar clinical presentation as well as gross and ultrasonographic
appearance (Table 1) [11,18,42]. The presence of splenic rupture, nRBCs,
abnormal red blood cell morphology, or anemia is consistently found more
frequently in the presence of neoplasia [26]. One study suggested that the
presence of anemia and splenic rupture may be up to 69% accurate in
predicting the presence of splenic neoplasia [26]. Up to two thirds of dogs
with splenic masses have a malignant tumor, and of these dogs with
a malignancy, up to two thirds have HSA. The remaining patients have
benign disease that can be treated with a splenectomy and have an excellent
prognosis [11,18,26,39,42]. Ultrasonography may be helpful in detecting
metastatic disease in other organs [79].

Abdomino- or pericardiocentesis can be performed to obtain a fluid

sample for analysis. Most effusions are characteristic of intracavitary hemor-
rhage, with a high packed cell volume and a nonclotting sample [28,34]. On
cytologic examination, evidence of fresh hemorrhage (peripheral blood
components) or previous hemorrhage may be seen (hemosiderin-laden
macrophages). Idiopathic pericardial effusions can also be hemorrhagic, so
differentiation of neoplastic versus nonneoplastic processes is difficult [80].
Protein concentrations and cell counts are also not different between ef-
fusions [80], although a pH greater than or equal to 7.0 has been reported to
be indicative of malignancy [81]. Sarcoma cells can also occasionally be

Table 1
Differential diagnoses for splenomegaly

/splenic nodules in the dog

Nonneoplastic disease

Neoplastic disease

Hyperplastic nodular enlargement

Benign

Hematoma

Fibroma

Vascular stasis

/congestion

Hemangioma

Thrombosis

/infarction

Lipoma

/myclolipoma

Torsion

Malignant

Traumatic rupture

Hemangiosarcoma

Abscessation

Fibrosarcoma

Extramedullary hematopoiesis

Leiomyosarcoma

Inflammation

/necrosis

Lymphosarcoma
Myxosarcoma
Osteosarcoma
Liposarcoma
Rhabdomyosarcoma
Chondrosarcoma
Mesenchymoma
Undifferentiated

/anaplastic sarcoma

Histiocytosis

/fibrohistiocytic nodules

Mast cell tumor
Metastatic neoplasia

Data from

Refs. [18,39,82,108,109,111].

540

A.N. Smith

/ Vet Clin Small Anim 33 (2003) 533–552

background image

apparent, indicating a presumptive diagnosis of HSA in about 25% of cases
[56], although false-positive results for malignancy have been reported in up
to 13% of cases [80]. False-negative results are quite common [16].

Aspiration cytology of masses is often obfuscated by hemorrhage, so this

technique is not considered to be sensitive in the diagnosis of HSA
[34,82,83]. Other causes of splenic masses or splenomegaly can be detected,
however, such as extramedullary hematopoiesis, primary hematopoietic
tumors (eg, lymphosarcoma, plasmacytoma), or metastatic disease (eg,
carcinoma, mast cell tumor) and may help to establish a diagnosis
[26,34,83,84]. The risk of splenic rupture or tumor seeding must be
considered when a decision is made to aspirate a splenic mass [83].

Histopathologic examination of tissues is required to diagnose HSA

definitively. An excisional biopsy is preferred, because it is both a diagnostic
and therapeutic procedure [5,33]. Multiple tissue samples or the entire mass
should be submitted, because most of a mass is usually composed of
hematomas, which can lead to an incorrect diagnosis in the absence of
representative tissues. Some authors advise slicing the spleen like a loaf
of bread to find multiple samples of different textures and appearance,
although submission of the entire organ, if possible, is preferred [5]. At
exploratory surgery, not only the primary mass but any other suspicious
lesions within the spleen, liver, or omentum should be biopsied and
submitted for analysis. Pathologists can confirm the origin of the tumor with
immunohistochemical staining for factor VIII-related antigen

/von Wille-

brand’s factor, which is highly specific for endothelial cells [85]. CD31 or
monoclonal antibody 3B5 may be even more useful in confirming a vascular
endothelial cell origin, because up to 10% to 20% of HSAs are negative for
factor VIII [85–87].

Therapy

Published median survival times for dogs with HSA are listed in Table 2.

Surgery

Surgical removal of a bleeding mass provides relief of clinical signs for

a period and is the initial treatment of choice for HSA [5]. In some animals,
this cessation of clinical signs of hemorrhage lasts for several months, but,
ultimately, dogs die of metastatic disease [5]. Median survival for dogs with
splenic HSA treated with surgery alone is short, ranging from 19 to 86 days
[1,11,26,88].

Surgical resection of right auricular masses is possible in some cases

to palliate a bleeding tumor [28,89]. Pericardectomy has been suggested
to provide relief from signs of cardiac tamponade [56] but did not affect
recurrence of signs or survival in one study [90]. All these dogs had de-
tectable metastasis at the time of surgery, however. Thoracoscopic removal

541

A.N. Smith

/ Vet Clin Small Anim 33 (2003) 533–552

background image

Table 2
Median survival times for dogs with hemangiosarcoma

Tumor site

Treatment

Number of
patients

Survival
(days)

Reference

Spleen

Surgery alone

59

19

Prymak et al [11]

Surgery alone

21

65

Brown et al [1]

Surgery alone

19

56

Johnson et al [26]

Stage I

4

91

Johnson et al [26]

Stage II

1

168

Johnson et al [26]

Stage III

14

56

Johnson et al [26]

Surgery alone

32

86

Wood et al [87]

Surgery + MBV

10

91

Brown et al [1]

Surgery + MBV

+ VMC

10

117

Brown et al [1]

Surgery + VAC

 ChM

6

145

Hammer et al [95]

Surgery + VAC

3

140

Johnson et al [26]

Surgery + AC

6

180

Sorenmo et al [30]

Surgery + AC

16

141

Vail et al [101]

Stage I disease

7

166

Vail et al [101]

Stage II disease

9

96

Vail et al [101]

Surgery + AC

+ L-MTP-PE

16

273

Vail et al [101]

Right atrium

VAC alone

1

140

deMadron

et al [110]

Incisional biopsy

+ AC

1

113

Sorenmo et al [30]

Surgery

9

120

a

Aronsohn [16]

Surgery

3

49

Berg and

Wingfield [56]

Cutaneous

/dermis

Surgery (stage I)

10

780

Ward et al [60]

Subcutaneous

VAC

 ChM

6

425

Hammer et al [95]

Surgery (stage II)

10

172

Ward et al [60]

Surgery (stage III)

5

307

Ward et al [60]

Surgery + AC

5

211

Sorenmo et al [30]

Various sites

Surgery + AC

16

202

Sorenmo et al [30]

Stage I

6

250

Sorenmo et al [30]

Stage II

4

186

Sorenmo et al [30]

Stage III

6

87

Sorenmo et al [30]

Surgery + AC

+ minocycline

17

170

Sorenmo et al [102]

Various nonskin

sites

Surgery

(complete excision)
+ A

27

172

Ogilvie et al [94]

Surgery (incomplete

excision) + A

19

60

Ogilvie et al [94]

Abbreviations:

A, doxorubicin alone; AC, doxorubicin, cyclophosphamide; ChM, chloram-

bucil, methotrexate; L-MTP-PE, liposome-encapsulated muramyl tripeptide-phosphatidyletha-
nolamine; MBV, mixed bacterial vaccine; VAC, vincristine, doxorubicin, cyclophosphamide;
VMC, vincristine, methotrexate, cyclophosphamide.

a

Mean.

542

A.N. Smith

/ Vet Clin Small Anim 33 (2003) 533–552

background image

of the pericardium has been described as an alternative to thoracotomy [91].
Patch grafts have been used to reconstruct the right atrium after aggressive
resection of large masses to obtain tumor-free margins [89]. Most dogs with
right atrial masses survive less than 4 months after surgery [23,89,90].

Cutaneous and subcutaneous HSAs can be removed surgically with wide

margins. Other primary or secondary HSA sites can also be surgically re-
moved depending on location (eg, bone tumors with amputation, renal tu-
mors with nephrectomy, bladder tumors with partial cystectomy). All
primary locations except superficial cutaneous tumors carry a poor long-
term prognosis [60], but surgery can be useful for palliation.

Radiation

The use of radiation therapy has not been extensively studied in dogs and

cats with HSA, because the tumor is generally considered a systemic rather
than localized disease. Palliative radiation has been used at our institution
and others with good short-term results to control superficial bleeding
masses not amenable to surgery and to relieve pain associated with tumors
within bone. Survival times of 12 to 42 weeks after 3 to 4 fractions up to
24 Gy total have been reported in four dogs with stage III disease, resulting
in stable disease (2 dogs), partial remission (1 dog), and complete (1 dog)
remission in one study [92]. Other investigators have reported poor
responses in two cats with large and unresectable cutaneous HSAs [93],
although the use of radiation therapy for control of cutaneous HSAs with
incomplete surgical resection, given their high recurrence rate, has been
suggested [5,33,93].

Chemotherapy

Chemotherapy for dogs with HSA is outlined in Table 3. Doxorubicin

has greatest chemotherapeutic efficacy against canine HSA. Doxorubicin
has been used both as a single agent [94] and in combination with
cyclophosphamide [30], with or without the addition of vincristine [94].
Toxicities were frequent when the three-drug combination (vincristine,
doxorubicin, and cyclophosphamide [referred to as VAC]) using a high dose
of vincristine (0.75 mg

/m

2

) was administered, necessitating hospitalization

for neutropenia or gastrointestinal toxicity in 7 of 15 patients [95]. Hos-
pitalization was only necessary in 3 of 16 dogs with the doxorubicin

/

cyclophosphamide protocol [30]. Survival times seem to be similar when
comparing doxorubicin alone versus combination chemotherapy [33,94],
although prospective trials to compare toxicity and efficacy have not been
performed. As with all cancer treatment, balancing the risk of side effects
and the benefits of therapy should be considered. In 1 dog, vincristine as
a single agent caused complete regression of presumed HSA metastatic lung
lesions after 12 weekly injections, which supports its use in combination

543

A.N. Smith

/ Vet Clin Small Anim 33 (2003) 533–552

background image

protocols [96]. Ifosfamide as a single agent [97] and a combination of vin-
cristine, cyclophosphamide, and methotrexate (with a mixed bacterial
vaccine) [1] have also demonstrated some response (Table 4).

Chemotherapy delivery by novel routes has shown activity against HSA.

Inhalation chemotherapy with a special formulation of doxorubicin resulted
in partial remission of metastatic lung lesions with minimal side effects in one
of three dogs with HSA [98]. The availability of the drug and specialized
administration equipment makes this therapy impractical currently but may
provide areas of future treatment of metastatic disease. Encapsulation of
doxorubicin in liposomes (Doxil; Sequus Pharmaceutical, Menlo Park, CA)
has been used to prevent clearance of the drug by the monophagocytic system,
thereby sustaining drug blood levels while decreasing systemic toxicity; this
therapy has been reported to have activity against canine HSA [99].

Table 3
Chemotherapy for dogs with hemangiosarcoma

Treatment day

Drug

Dose

Day 1

Doxorubicin (dilute to 0.5

mg

/mL in saline)

30 mg

/m

2

IV through

well-placed catheter slowly



Cyclophosphamide

100–200 mg

/m

2

IV or

Days 8 and 15

50 mg

/m

2

PO days 3–6



Vincristine

0.5–0.75 mg

/m

2

IV

Repeat cycle every 3 weeks for five to eight treatments.
Consider pretreatment with an antiemetic and antihistamine or corticosteroids to prevent

nausea and mast cell degranulation 20 minutes before doxorubicin administration.

Complete blood cell count with differential count before each chemotherapy injection.

Delay drug administration if neutrophil count is <2500–3000.

Can consider prophylactic antibiotic administration on days 1–8 after doxorubicin.
Dogs weighing <10 kg can be dosed with doxorubicin at 1 mg

/kg.

Consider echocardiogram periodically to monitor for possible myocardial necrosis as

evidenced by decreased fractional shortening or ejection fraction, especially at cumulative
doxorubicin doses >150–180 mg

/m

2

.

Cats can be given 20–25 mg

/m

2

or 1 mg

/kg of doxorubicin

 cyclophosphamide at 100

mg

/m

2

IV every 3 weeks. Pretreatment, antibiotic, and blood monitoring as above. Monitor

renal function at cumulative doxorubicin doses of >80 mg

/m

2

.

Abbreviations:

IV, intravenously; PO, per os.

Table 4
Vincristine, methotrexate, cyclophosphamide chemotherapy for dogs with hemangiosarcoma

Treatment day

Drug

Dose

Days 1, 8, 15, 22, 29, 36, 43,

50, 57, 64, 71, 78

Vincristine

0.0125 mg

/kg IV

Methotrexate

0.4–0.6 mg

/kg IV

Days 1–84

Cyclophosphamide

1 mg

/kg PO

Abbreviations:

IV, intravenously; PO, per os.

Data from

Brown NO, Patnaik AR, MacEwen EG. Canine hemangiosarcoma: retrospective

analysis of 104 cases. JAVMA 1985;186:56–8.

544

A.N. Smith

/ Vet Clin Small Anim 33 (2003) 533–552

background image

Chemotherapy has not been extensively studied in cats with HSA, but

doxorubicin-based protocols have been recommended in this species,
especially for cases with visceral disease (C.M. Johannes et al, unpublished
data) [5,33]. Its use may also be necessary in cats with subcutaneous HSAs,
because recent studies have demonstrated a possibly higher metastatic
potential than previously believed (C.M. Johannes, unpublished data) [27].
No controlled studies have been conducted to evaluate therapy in cats with
HSA; however, several individual case reports are available in the literature.
In a single case report, a combination of doxorubicin, cyclophosphamide,
and vincristine resulted in survival of longer than 1 year, although the cat
eventually succumbed to metastatic disease [100]. Single-agent vincristine
therapy was ineffective in another cat that died within 2.5 months of diag-
nosis [27]. Other therapies used include VAC in one cat with an incompletely
resected cutaneous tumor; this cat survived for longer than 3.5 years. A cat
with an incompletely excised subcutaneous mass survived 440 days and was
euthanized for unrelated disease after being treated with mitoxantrone and
cyclophosphamide. Another cat with an incompletely resected subcutaneous
mass HSA received doxorubicin at a dose of 25 mg

/m

2

intravenously every

3 weeks for four treatments and survived 290 days, ultimately dying of
probable metastatic thoracic disease. One cat with completely resected
colonic HSA received doxorubicin as described in the previous case and died
150 days later with suspected abdominal metastatic disease. Finally, one cat
died from progressive disease 60 days after incomplete surgical resection of
mesenteric HSA and carboplatin chemotherapy (C.M. Johannes et al,
unpublished data). Further controlled studies are necessary to determine the
utility of chemotherapy in cats with HSA.

Biologic therapy

Immunomodulators seem to be useful in prolonging survival times,

especially in combination with chemotherapy. A mixed bacterial vaccine [1]
and liposome-encapsulated muramyl tripeptide (L-MTP) [101] have both
been used to treat HSAs and have been associated with increased me-
dian survival times compared with those achieved by surgery alone. The
addition of chemotherapy (VMC) to the mixed bacterial vaccine increased
median survival further, to 117 days, although this increase was not statis-
tically different from splenectomy alone [1]. Splenectomy, L-MTP, and
doxorubicin

/cyclophosphamide chemotherapy resulted in 9.1-month median

survival times, which were significantly prolonged (P

¼ 0.03) compared

with those achieved by splenectomy and chemotherapy alone (median

¼ 5.7

months) [101].

L-MTP is not currently used except on an experimental basis because of

lack of availability and high cost, but it or a similar formulation may be
commercially available in the future.

545

A.N. Smith

/ Vet Clin Small Anim 33 (2003) 533–552

background image

Prognosis

Despite aggressive surgical, drug, or radiation therapy, median survival

times are short for almost all forms of primary HSA in cats and dogs. One-
year survival for dogs is less than 10% [2]. Stage of splenic disease at
diagnosis does not seem to affect survival according to several studies
[1,26,88], although in one study, dogs with hemoperitoneum associated with
splenic HSA (stage II–III) had a shorter median survival (17 days) after
splenectomy as opposed to dogs with stage I disease (median survival

¼

121 days) [11]. Another study also suggested an improved length of survival
with lower disease stage (stage I disease

¼ 250 days, stage II disease ¼ 186

days, and stage III disease

¼ 87 days), although the differences were not

statistically significant [30]. There seems to be no difference in survival
expectation between visceral and subcutaneous HSAs [30]. Only canine and
feline superficial dermal HSAs (stage I) treated with surgery have a
consistent low metastatic potential and prolonged survival times of longer
than 1 year (median: 780 days in dogs,

36 months in cats). Occasionally,

metastasis is seen even with these more favorably located tumors (C.M.
Johannes et al, unpublished data) [19,24,21,25,27,60].

Future directions

Angiogenesis modifiers have garnered much media and scientific

attention in the last several years. Vascular development is essential for
primary and metastatic tumor growth beyond 1 mm

3

, so blockade of this

process would seem to be a viable antitumor therapy. Advantages of anti-
angiogenic agents include a purported lack of the development of drug
resistance and few systemic side effects. Most experts agree that angiogenic
inhibitors will not replace primary tumor therapy but will be useful in an
adjunctive setting. Antiangiogenic therapy will likely need to be maintained
throughout the life of the patient . As a tumor composed of blood vessels,
HSA would theoretically be the ideal candidate for this type of therapy. In
a single clinical trial, the antiangiogenic antibiotic minocycline did not show
improved patient survival when added to a standard surgery and che-
motherapy protocol [102]. The ideal antiangiogenic dose of this drug has not
been determined, however, and it is likely that multiple agents in a ‘‘cocktail’’
will be necessary to target multiple pathways for blood vessel development.
Other agents being investigated as anticancer therapy include the
antiangiogenic agents thalidomide, interferon, matrix metalloproteinases,
recombinant endogenous angiogenesis inhibitors, and antibodies directed
against receptors or factors involved in angiogenesis [103].

Diagnosis of HSA before surgery would be useful for veterinarians so as

to assist owners in making informed decisions before embarking on
extensive procedures in the face of a disease with a guarded to grave prog-
nosis. Vascular endothelial growth factor (VEGF), a proangiogenic agent

546

A.N. Smith

/ Vet Clin Small Anim 33 (2003) 533–552

background image

related to endothelial cell proliferation, has been found to be increased in
tumor tissues in dogs [104]. In unpublished studies conducted by the author,
serum levels of VEGF seem to have some diagnostic utility for differentiating
HSA from hematoma or other benign disease processes. In another study,
plasma VEGF concentrations in dog with HSA were increased compared with
those of normal dogs, and in one dog, levels increased with disease progression
[105]. VEGF concentration was not useful as a predictor of tumor presence
when measured in body cavity effusions [106]. The role of platelet-derived
VEGF in this disease should be further investigated to determine the
usefulness of this factor in predicting tumor presence or as a monitoring device
for the presence of metastasis or recurrence.

Dietary therapy has been suggested to increase survival times in dogs

with lymphosarcoma, and patients with other tumor types may benefit as
well [107]. Adequate caloric intake (1.1 [30 (patient weight in kilograms)+
70]

¼ kilocalories needed per day) with a highly bioavailable and palatable

food is likely most important, but suggested alterations in diet to combat
cancer cachexia might include low simple carbohydrate levels, moderate
good-quality protein levels, moderate fat levels, and adequate fiber levels
(both soluble and insoluble) [107]. N-3 fatty acids seem to be especially
important in managing the metabolic alterations seen in cancer patients
[107]. Vitamin E, arginine, cystine, and glutamine have also been suggested
as supplements to enhance immune and gastrointestinal function [107].
Further studies in this arena are necessary to elucidate the role of diets and
nutrition in the management of patients with HSA.

References

[1] Brown NO, Patnaik AR, MacEwen EG. Canine hemangiosarcoma: retrospective

analysis of 104 cases. JAVMA 1985;186:56–8.

[2] Fees DL, Withrow SJ. Canine hemangiosarcoma. Compend Contin Educ Pract Vet

1981;3:1047–52.

[3] Hammer AS, Couto CG. Diagnosing and treating canine hemangiosarcoma. Veterinary

Medicine 1992;87:188–201.

[4] Kleine LJ, Zook BC, Munson TO, et al. Primary cardiac hemangiosarcomas in dogs.

JAVMA 1970;157:326–37.

[5] MacEwen EG. Miscellaneous tumors: hemangiosarcoma. In: Withrow SJ, MacEwen

EG, editors. Small animal clinical oncology. 3rd edition. Philadelphia: WB Saunders;
2001. p. 639–46.

[6] Madewell BR, Theilen GH. Skin tumors of mesenchymal origin. In: Theilen GH,

Madewell BR, editors. Veterinary cancer medicine. 2nd edition. Philadelphia: Lea &
Febiger; 1987. p. 295–7.

[7] Ng CY, Mills JN. Clinical and haematological features of haemangiosarcoma in dogs.

Aust Vet J 1985;62:1–4.

[8] Oksanen A. Haemangiosarcoma in dogs. J Comp Pathol 1978;88:585–95.
[9] Pearson GR, Head KW. Malignant hemangioendothelioma (angiosarcoma) in the dog.

J Small Anim Pract 1976;17:737–45.

547

A.N. Smith

/ Vet Clin Small Anim 33 (2003) 533–552

background image

[10] Priester WA, McKay FW. The occurrence of tumors in domestic animals. National

Cancer Institute Monograph 54, NIH publication no. 80–2046. 1980. p. 167.

[11] Prymak C, McKee L, Goldschmidt MH, Glickman LT. Epidemiologic, clinical,

pathologic, and prognostic characteristics of splenic hemangiosarcoma and splenic
hematoma in dogs: 217 cases. JAVMA 1985;193:706–12.

[12] Srebernik N, Appleby E. Breed prevalence and sites of haemangioma and haemangio-

sarcoma in dogs. Vet Rec 1991;129:408–9.

[13] Standard AA, Pulley T. Skin and soft tissues. In: Moulton JE, editor. Tumors

in domestic animals. 2nd edition. Berkeley: University of California Press; 1978.
p. 35–6.

[14] Waller T, Rubarth S. Hemangioendothelioma in domestic animals. Acta Vet Scand

1967;8:234–61.

[15] Waters DJ, Caywood DD, Hayden DW, Klausner JS. Metastatic pattern in dogs

with splenic hemangiosarcomas: clinical implications. J Small Anim Pract 1988;29:
805–14.

[16] Aronsohn M. Cardiac hemangiosarcoma in the dog: a review of 38 cases. JAVMA

1985;187:922–6.

[17] Priester WA. Brief communication: hepatic angiosarcomas in dogs: an excessive

frequency as compared with man. J Natl Cancer Inst 1976;57:451–4.

[18] Spangler WL, Culbertson MR. Prevalence, type, and importance of splenic diseases in

dogs: 1,480 cases (1985–1989). JAVMA 1992;200:829–34.

[19] Carpenter JL, Andrews LK, Holzworth J. Tumors and tumor-like lesions. In: Holzworth

J, editor. Diseases of the cat, vol. 1. Philadelphia: WB Saunders; 1987. p. 480–3.

[20] Engle GC, Brodey RS. A retrospective study of 395 feline neoplasms. J Am Anim Hosp

Assoc 1969;5:21–31.

[21] Patnaik AK, Liu SK. Angiosarcoma in cats. J Small Anim Pract 1977;18:191–8.
[22] Schmidt R, Langhan R. A survey of feline neoplasms. JAVMA 1967;151:1325–8.
[23] Arp LH, Grier RL. Disseminated cutaneous hemangiosarcoma in a young dog. JAVMA

1984;185:671–3.

[24] Hargis AM, Ihrke PJ, Spangler WL, Stannard AA. A retrospective clinicopathological

study of 212 dogs with cutaneous hemangiomas and hemangiosarcomas. Vet Pathol
1992;29:316–28.

[25] Scavelli TD, Patnaik AK, Mehlhaff CJ, Hayes AA. Hemangiosarcoma in the cat:

retrospective evaluation of 31 surgical cases. JAVMA 1985;187:817–9.

[26] Johnson KA, Powers BE, Withrow SJ, et al. Splenomegaly in dogs: predictors of

neoplasia and survival after splenectomy. J Vet Intern Med 1989;3:160–6.

[27] Kraje AC, Mears EA, Hahn KA, et al. Unusual metastatic behavior and clinicopath-

ologic findings in eight cats with cutaneous or visceral hemangiosarcoma. JAVMA
1999;214:670–2.

[28] Wykes P, Rouse G, Orton E. Removal of five canine cardiac tumors using a stapling

instrument. Vet Surg 1986;15:103–6.

[29] Ling GV, Morgan JP, Pool RR. Primary bone tumors in the dog: a combined clinical,

radiographic, and histologic approach to early diagnosis. JAVMA 1974;165:55–67.

[30] Sorenmo KU, Jeglum KA, Helfand SC. Chemotherapy of canine hemangiosarcoma with

doxorubicin and cyclophosphamide. J Vet Intern Med 1993;7:370–6.

[31] Miller MA, Ramos JA, Kreeger JM. Cutaneous vascular neoplasia in 15 cats: clinical,

morphologic, and immunohistochemical studies. Vet Pathol 1992;29:329–36.

[32] Ware WA, Hopper DL. Cardiac tumors in dogs: 1982–1995. J Vet Intern Med 1999;13:

95–103.

[33] Chun R. Feline and canine hemangiosarcoma. Compend Contin Educ Pract Vet

1999;21:622–9.

[34] Hosgood G. Canine hemangiosarcoma. Compend Contin Educ Pract Vet 1991;13:

1065–75.

548

A.N. Smith

/ Vet Clin Small Anim 33 (2003) 533–552

background image

[35] Nikula K, Benjamin S, Angleton G, et al. Ultraviolet radiation, solar dermatosis, and

cutaneous neoplasia in beagle dogs. Radiat Res 1992;129:11–8.

[36] Culbertson M. Hemangiosarcoma of the canine skin and tongue. Vet Pathol 1982;19:

556–8.

[37] Hoekstra HJ, Sindelar WF, Kinsella TJ, Mehta DM. Intraoperative radiation therapy-

induced sarcomas in dogs. Radiat Res 1989;120:508–15.

[38] Benjamin SA, Hahn FF, Chieffelle TL, et al. Occurrence of hemangiosarcomas in beagles

with internally deposited radionuclides. Cancer Res 1975;35:1745–55.

[39] Day MJ, Lucke VM, Pearson H. A review of pathological diagnoses made from 87

canine splenic biopsies. J Small Anim Pract 1995;36:426–33.

[40] Hirsch V, Jacobsen J, Mills J. A retrospective study of canine hemangiosarcoma and its

association with acanthocytosis. Can Vet J 1981;22:152–5.

[41] Holt D, Van Winkle T, Schelling C, Prymak C. Correlation between thoracic radiographs

and post-mortem findings in dogs with hemangiosarcoma: 77 cases (1984–1989). JAVMA
1992;200:1535–9.

[42] Spangler WL, Kass PH. Pathologic factors affecting postsplenectomy survival in dogs.

J Vet Intern Med 1997;11:166–71.

[43] Nasir L, Krasner H, Argyle DJ, Williams A. Immunocytochemical analysis of the

tumour suppressor protein (p53) in feline neoplasia. Cancer Lett 2000;155:1–7.

[44] Adam Y, Huvos A, Hajdu S. Malignant vascular tumors in the liver. Ann Surg

1972;175:375–83.

[45] Shin M, Carpenter J, Ho K. Epithelioid hemangioendothelioma: CT manifestations and

possible linkage to vinyl chloride exposure. J Comput Assist Tomogr 1991;15:505–7.

[46] Brown NO. Hemangiosarcoma. Vet Clin North Am Small Anim Pract 1985;15:

569–5.

[47] Spangler WL, Culbertson MR. Prevalence and type of splenic diseases in cats: 455 cases

(1985–1991). JAVMA 1992;201:773–6.

[48] Bingel SA, Brodey RS, Allen HL, Riser WH. Haemangiosarcoma of bone in the dog.

J Small Anim Pract 1974;15:303–22.

[49] Crow SE, Bell TG, Wortman JA. Hematuria associated with renal hemangiosarcoma in

a dog. JAVMA 1980;176:531–3.

[50] Dueland R, Dahlin D. Hemangioendothelioma of canine bone. J Am Anim Hosp Assoc

1972;8:81–5.

[51] Hill TP, Lobette RG, Schulman ML. Vulvovaginectomy and neo-urethrostomy

for treatment of hemangiosarcoma of the vulva and vagina. J S Afr Vet Assoc 2000;71:
256–9.

[52] Martinez SA, Schulman AJ. Hemangiosarcoma of the urinary bladder in a dog. JAVMA

1988;192:655–6.

[53] Gilger BC, McLaughlin SA, Whitley RD, Wright JC. Orbital neoplasms in cats: 21 cases

(1974–1990). JAVMA 1992;201:1083–6.

[54] Quigley PJ, Leedale AH. Tumours involving bone in the domestic cat: a review of fifty-

eight cases. Vet Pathol 1983;20:670–86.

[55] Sharpe A, Cannon MJ, Lucke VM, Day MJ. Intestinal haemangiosarcoma in the cat:

clinical and pathological features of four cases. J Small Anim Pract 2000;41:411–5.

[56] Berg R, Wingfield W. Pericardial effusion in the dog: a review of 42 cases. J Am Anim

Hosp Assoc 1984;20:721–30.

[57] Brodey RS, Riser WH. Canine osteosarcoma. A clinicopathologic study of 194 cases.

Clin Orthop 1969;62:54–64.

[58] Morgan JP, Ackerman N, Bailey CS, et al. Vertebral tumors in the dog: a clinical,

radiologic, and pathologic study of 61 primary and secondary lesions. Vet Radiol
1980;21:197–212.

[59] Waters DJ, Hayden DW, Walter PA. Intracranial lesions in dogs with hemangiosarcoma.

J Vet Intern Med 1989;3:222–30.

549

A.N. Smith

/ Vet Clin Small Anim 33 (2003) 533–552

background image

[60] Ward H, Fox LE, Calderwood-Mays MB, et al. Cutaneous hemangiosarcoma in 25 dogs:

a retrospective study. J Vet Intern Med 1994;8:345–8.

[61] Mughannam AJ, Hacker DV, Spangler WL. Conjunctival vascular tumors in six dogs.

Vet Comp Ophthalmol 1997;7:56–9.

[62] Chastain CB, Riedesel DH, Graham DL. Ventricular septal hemangiosarcoma associated

with right bundle branch block in a dog. JAVMA 1974;165:177–9.

[63] Legendre A, Krehbiel J. Disseminated intravascular coagulation in a dog with

hemothorax and hemangiosarcoma. J Vet Intern Med 1977;3:1070–1.

[64] Gores BR, Berg J, Carpenter JL, Ullman SL. Chylous ascites in cats: nine cases (1978–

1993). JAVMA 1994;205:1161–4.

[65] Gelberg H, Stackhouse LL. Three cases of canine acanthocytosis associated with splenic

neoplasia. Vet Med Small Anim Clin 1977;72:1183–4.

[66] Grindem CB, Breitschwerdt EB, Corbett WT, et al. Thrombocytopenia associated with

neoplasia in dogs. J Vet Intern Med 1994;8:400–5.

[67] Hammer AS, Couto CG, Swardson C, et al. Hemostatic abnormalities in dogs with

hemangiosarcoma. J Vet Intern Med 1991;5:11–4.

[68] Hargis AM, Feldman BF. Evaluation of hemostatic defects secondary to vascular tumors

in dogs: 11 cases (1983–1988). JAVMA 1991;198:891–4.

[69] Rishniw M, Lewis DC. Localized consumptive coagulopathy associated with cutaneous

hemangiosarcoma in a dog. J Am Anim Hosp Assoc 1994;30:261–4.

[70] Leifer CE, Peterson ME, Matus RE, Patnaik AK. Hypoglycemia associated with nonislet

cell tumor in 13 dogs. JAVMA 1985;186:53–5.

[71] Hammer AS, Bailey MQ, Sagartz JE. Retrospective assessment of thoracic radio-

graphic findings in metastatic canine hemangiosarcoma. Vet Radiol Ultrasound 1993;
34:235–8.

[72] Fox PR, Bond BR. Nonselective and selective angiocardiography. Vet Clin North Am

Small Anim Pract 1983;13:259–72.

[73] Fruchter A, Miller C, O’Grady M. Echocardiographic results and clinical considerations

in dogs with right atrial

/auricular masses. Can Vet J 1992;33:171–4.

[74] Thomas WP, Sisson DD, Bauer T, Reed J. Detection of cardiac masses in two-

dimensional echocardiography. Vet Radiol 1984;25:65–71.

[75] Smith KA, Miller LM, Biller DS. Detection of right atrial hemangiosarcoma using

nonselective angiocardiography in a dog. Can Vet J 1992;33:673–5.

[76] Keene BW, Rush JE, Cooley AJ, Subramanian R. Primary left ventricular hemangio-

sarcoma diagnosed by endomyocardial biopsy in a dog. JAVMA 1990;197:1501–3.

[77] Keyes M, Rush J. Ventricular arrhythmias in dogs with splenic masses. Vet Emerg Crit Care

1994;3:33–8.

[78] Knapp DW, Aronsohn MG, Harpster NK. Cardiac arrhythmias associated with mass

lesions of the canine spleen. J Am Anim Hosp Assoc 1993;29:122–8.

[79] Wrigley RH, Park RD, Konde LJ, Lebel JL. Ultrasonographic features of splenic

hemangiosarcoma in dogs: 18 cases (1980–1986). JAVMA 1988;192:1113–7.

[80] Sisson DD, Thomas WP, Ruehl WW, Zinkl JG. Diagnostic value of pericardial fluid

analysis in the dog. JAVMA 1984;184:51–5.

[81] Edwards NJ. The diagnostic value of pericardial fluid pH determination. J Am Anim

Hosp Assoc 1996;32:63–7.

[82] Neer TM. Clinical approach to splenomegaly in dogs and cats. Compend Contin Educ

Pract Vet 1996;18:35–49.

[83] O’Keefe DA, Couto CG. Fine-needle aspiration of the spleen as an aid in the diagnosis of

splenomegaly. J Vet Intern Med 1987;1:102–9.

[84] Frey AJ, Betts CW. A retrospective survey of splenectomy in the dog. J Am Anim Hosp

Assoc 1977;13:730–4.

[85] von Beust BR, Suter MM, Summers BA. Factor VIII related antigen in canine

endothelial neoplasms: an immunohistochemical study. Vet Pathol 1988;25:251–5.

550

A.N. Smith

/ Vet Clin Small Anim 33 (2003) 533–552

background image

[86] Ferrer L, Fondevila D, Rabanal RM, Vilafranca M. Immunohistochemical detection of

CD31 antigen in normal and neoplastic canine endothelial cells. J Comp Pathol
1995;112:319–26.

[87] Liu KX, Church-Bird AE, Lenz SD, et al. Antigen expression in normal and neoplastic

canine tissues defined by a monoclonal antibody generated against canine mesothelial
cells. Vet Pathol 1994;31:663–73.

[88] Wood CA, Moore AS, Gliatto JM, et al. Prognosis for dogs with stage I or II splenic

hemangiosarcoma treated by splenectomy alone: 32 cases (1991–1993). J Am Anim Hosp
Assoc 1998;34:417–21.

[89] Brisson BA, Holmberg DL. Use of pericardial patch graft reconstruction of the right

atrium for treatment of hemangiosarcoma in the dog. JAVMA 2001;218:723–5.

[90] Dunning D, Monnet E, Orton EC, Salman MD. Analysis of prognostic indicators for

dogs with pericardial effusion: 46 cases (1985–1996). JAVMA 1998;212:1276–80.

[91] Walsh P, Remedios A, Ferguson J, et al. Thoracoscopic versus open partial pericardectomy

in dogs: comparison of postoperative pain and morbidity. Vet Surg 1999;28:472–9.

[92] McEntee MC. Radiation therapy in the management of bone tumors. Vet Clin North Am

Small Anim Pract 1997;27:131–8.

[93] Meleo KA. Tumors of the skin and associated structures. Vet Clin North Am Small

Anim Pract 1997;27:73–94.

[94] Ogilvie GK, Powers BE, Mallinckrodt CH, Withrow SJ. Surgery and doxorubicin in dogs

with hemangiosarcoma. J Vet Intern Med 1996;10:379–84.

[95] Hammer AS, Couto CG, Filppi J, et al. Efficacy and toxicity of VAC chemotherapy

(vincristine, doxorubicin, and cyclophosphamide) in dogs with hemangiosarcoma. J Vet
Intern Med 1991;5:160–6.

[96] Hahn K. Vincristine sulfate as single-agent chemotherapy in a dog and cat with

malignant neoplasms. JAVMA 1990;197:504–6.

[97] Rassnick KM, Frimberger AE, Wood CA, et al. Evaluation of ifosfamide for treatment

of various canine neoplasms. J Vet Intern Med 2000;14:271–6.

[98] Hershey AE, Kurzman ID, Forrest LJ, et al. Inhalation chemotherapy for macroscopic

primary or metastatic lung tumors: proof of principle using dogs with spontaneously
occurring tumors as a model. Clin Cancer Res 1999;5:2653–9.

[99] Vail DM, Kravis LD, Cooley AJ, et al. Preclinical trial of doxorubicin entrapped in

sterically stabilized liposomes in dogs with spontaneously arising malignant tumors.
Cancer Chemother Pharmacol 1997;39:410–6.

[100] Tudor K, Greenlee P. Cerebral metastatic hemangiosarcoma in the cat. Feline Pract

1994;22:20–1.

[101] Vail DM, MacEwen EG, Kurzman ID, et al. Liposome-encapsulated muramyl tripeptide

phosphatidylethanolamine adjuvant immunotherapy for splenic hemangiosarcoma in the
dog: a randomized multi-institutional clinical trial. Clin Cancer Res 1995;1:1165–70.

[102] Sorenmo KU, Duda LE, Barber L, et al. Canine hemangiosarcoma treated with standard

chemotherapy and minocycline. J Vet Intern Med 2000;14:395–8.

[103] Clifford CA, Mackin AJ, Henry CJ. Treatment of canine hemangiosarcoma: 2000 and

beyond. J Vet Intern Med 2000;14:479–85.

[104] Scheidegger P, Weiglhofer W, Suarez S, et al. Vascular endothelial growth factor (VEGF)

and its receptors in tumor-bearing dogs. Biol Chem 1999;380:1449–54.

[105] Clifford CA, Hughes D, Beal MW, et al. Plasma vascular endothelial growth factor

concentrations in healthy dogs and dogs with hemangiosarcoma. J Vet Intern Med
2001;15:131–5.

[106] Clifford CA, Hughes D, Beal MW, et al. Vascular endothelial growth factor

concentrations in body cavity effusions in dogs. J Vet Intern Med 2002;16:164–8.

[107] Ogilvie GK. Metabolic alterations and nutritional therapy. In: Withrow SJ, MacEwen

EG, editors. Small animal clinical oncology. 3rd edition. Philadelphia: WB Saunders;
2001. p. 169–82.

551

A.N. Smith

/ Vet Clin Small Anim 33 (2003) 533–552

background image

[108] Hendrick M, Brooks J, Bruce E. Six cases of malignant fibrous histiocytoma of the canine

spleen. Vet Pathol 1992;29:351–4.

[109] Weinstein MJ, Carpenter JL, Melhoff-Schunk CJ. Nonangiogenic and nonlymphoid

sarcomas of the canine spleen: 57 cases (1975–1987). JAVMA 1989;195:784–8.

[110] de Madron E, Helfand SC, Stebbins KE. Use of chemotherapy for treatment of cardiac

hemangiosarcoma in a dog. JAVMA 1987;190:887–91.

[111] Hardie EM, Vaden SL, Spaulding K, Malarkey DE. Splenic infarction in 16 dogs:

a retrospective study. J Vet Intern Med 1995;9:141–8.

552

A.N. Smith

/ Vet Clin Small Anim 33 (2003) 533–552

background image

Feline injection site sarcomas

Marlene Hauck, DVM, PhD

Department of Clinical Sciences, College of Veterinary Medicine,

North Carolina State University,

4700 Hillsborough Street, Raleigh, NC 27606, USA

Drs. Mattie Hendrick and Michael Goldschmidt and their colleagues at

the Laboratory of Pathology of the University of Pennsylvania School of
Veterinary Medicine deserve credit for initially recognizing the increasing
incidence of both reactions at the sites of rabies vaccinations and the for-
mation of sarcomas at sites commonly used for injection and vaccination
in cats [1,2]. Their letter to the editor in the Journal of the American
Veterinary Medical Association

in October 1991 alerted the profession to ‘‘the

possibility that fibrosarcomas may arise at injection sites in cats.’’ They
suggested that this change was related to the enactment of a state law in
Pennsylvania requiring the rabies vaccination of cats, and they tracked an
increase in tumors arising at common vaccination sites (interscapular
region) versus tumors arising at nonvaccination sites (eg, head). Theirs was
the initial histopathologic description of the fibrosarcomas as tumors with
a marked inflammatory component, including macrophages containing a
gray-brown foreign substance.

This letter and other commentary resulted in mixed responses among

veterinarians, with some contesting the proposed association of sarcoma
development with vaccinations [3]. A follow-up study of these sarcomas by
Hendrick and colleagues [4] demonstrated the foreign substance in the
macrophages to be aluminum by electron probe microanalysis. Because
aluminum is used as a component of vaccine adjuvants, the identification
of aluminum within the macrophages was believed to be consistent with
residual adjuvant. Aluminum adjuvants have been reported to cause granu-
lomatous reactions in people [5].

Two changes occurred within the vaccine industry in the mid-1980s as well:

the first killed rabies vaccine licensed for subcutaneous administration

Vet Clin Small Anim

33 (2003) 553–571

E-mail address:

marlene_hauck@ncsu.edu

0195-5616

/03/$ - see front matter

Ó 2003, Elsevier Inc. All rights reserved.

doi:10.1016/S0195-5616(03)00006-8

background image

became available, as did a killed vaccine for feline leukemia virus (FeLV). To
evaluate further the suggested relation between sarcoma development and
vaccination, Dr. Philip Kass and others [6] undertook an epidemiologic study
using information about cats diagnosed with sarcomas at a large private
diagnostic laboratory. Sarcomas from sites typically used for vaccination
were compared with sarcomas developing at other sites (head, digit, and
forelimb). A total of 345 cats (185 sarcomas located at vaccination sites, 160
sarcomas located at nonvaccination sites) were included in an analysis of the
relation between vaccination status and tumor development. The findings
included an increased risk of sarcoma development, particularly in cats
vaccinated against FeLV but also with the administration of rabies vaccine.
The odds ratio for development of a sarcoma at a vaccination site after
vaccination with FeLV was 5.49 (95% confidence interval [CI]: 1.98–15.24),
and the odds ratio after rabies vaccination was 1.99 (95% CI: 0.72–5.54). A
single vaccination given in the interscapular region resulted in a 50% higher
risk of developing a sarcoma at that site compared with cats that were not
vaccinated at that site, two vaccinations resulted in a 127% increased risk, and
three or four vaccinations given simultaneously at this site resulted in an
approximately 175% risk increase. The overall estimated risk for cats in this
population to develop fibrosarcomas was relatively low, with roughly 20 cats
in 100,000 developing a sarcoma. Another interesting finding in this study was
a significant increase in the percentage of cats diagnosed with fibrosarcomas
at vaccination sites at the University of California–Davis Veterinary Medical
Teaching Hospital. The authors concluded that there was epidemiologic
support for a relation between vaccination and tumor development.

A similar retrospective study was undertaken at the University of

Pennsylvania and Tufts University under the direction of Dr. Mattie
Hendrick [7]. This group also compared cats with sarcomas arising at vac-
cination sites (n

¼ 181) with those arising at other sites (n ¼ 58). Cases were

drawn from the surgical pathology services at these two veterinary schools,
which provide services for approximately 2600 practices in the northeast. Of
the 181 tumors at sites used for vaccination, only 69 cats had a history of
receiving a vaccination at those sites. This study found that the cats with
sarcomas arising at sites of previous vaccination were significantly younger
and their tumors were significantly larger than those of cats with sarcomas
not associated with a previous vaccination. There were no differences
in the manufacturers of the vaccines used in the two groups. The authors
also noted that the tumors developing at the sites of vaccination recurred
more rapidly and more frequently than sarcomas at other sites and that the
interval from vaccination to sarcoma development ranged from 3 months to
3 years. Cats in the vaccine site sarcoma group were more likely to have
been vaccinated with FeLV than cats in the nonvaccine-associated sarcoma
group, although the reverse was true for rabies vaccination.

A commentary from a third large group of pathologists reported similar

findings [8]. The Animal Reference Pathology Division of the Associated

554

M. Hauck

/ Vet Clin Small Anim 33 (2003) 553–571

background image

Regional and University Pathologists reported identifying 176 sarcomas
arising at common vaccination sites in cats with features similar to those
described in the initial reports: anaplastic sarcomas with bluish material in
the inflammatory macrophages associated with the tumors. In these cases,
the interval reported between vaccination and sarcoma development ranged
from several months to more than 3 years.

In an attempt to define better the actual prevalence of injection site

sarcomas, a study was undertaken with members of the American
Association of Feline Practitioners. Coyne and others [9] surveyed these
veterinarians concerning their feline patients in 1992. This year was chosen
because it was the first year after the description of vaccine-associated
sarcomas and before the vaccine protocol recommendations designed to
decrease the incidence of injection site sarcomas. The overall incidence of
injection site sarcomas was estimated at 3.6 per 10,000 cats. If records only
from those practices using computerized medical records are considered
(19% of those responding), the prevalence of injection site sarcomas was
calculated as 2.7 per 10,000 cats. This is quite similar to the estimated
prevalence (based on polling 29 hospitals) of 1 to 2 per 10,000 cats [6]. One
of the criticisms of this study is that confirmation of the diagnosis was
not required, so there may have been overreporting of this disease. In addi-
tion, only 21% of the practices contacted participated in the study, and if
veterinarians did not participate because they did not diagnosis injection site
sarcomas, the true prevalence may have been overestimated.

Although, initially, only FeLV and rabies vaccines were implicated in an

increased risk of sarcoma development at the site of vaccination, an
additional study from Canada suggested a role for killed vaccines including
panleukopenia and respiratory viruses [10]. The authors report 14 cases
from their practice with a documented history of receiving a killed
panleukopenia

/respiratory virus vaccination in the interscapular space.

These same cats had not been vaccinated for FeLV, and their rabies
vaccinations had been given in the caudal thigh. The incidence of injection
site sarcomas in this clinic was reported as 13 per 10,000 cats, which is
higher than those of the two studies discussed previously. In this practice,
changing the panleukopenia

/respiratory vaccine to a modified live product

decreased the number of interscapular sarcomas diagnosed. Although this
report is limited to a single practice, it does raise a question regarding the
ability of vaccines aside from FeLV and rabies to be tumorigenic. In closer
agreement with the higher incidence reported by Lester et al [10] is a project
using prospective monitoring of 2000 cats at a feline practice in
Philadelphia. Dr. Hendrick [11] has reported the diagnosis of five sarcomas
at the site of a previous rabies vaccination. The average interval between
vaccination and tumor development was 26 months. Dr. Macy at Colorado
State University’s Veterinary Teaching Hospital, in association with
Dr. Hendrick, reported that 2 cats developed injection site sarcomas from
a population of cats that received 548 doses of either FeLV or rabies vaccine

555

M. Hauck

/ Vet Clin Small Anim 33 (2003) 553–571

background image

[12] One proposed reason for the wide variation in reported incidence is
possible regional differences, although there are currently no scientific data
to support this theory [13].

In an attempt to clarify this issue, a prospective study involving 40

veterinary practices in the Unites States and Canada was undertaken
[14]. These practices submitted monthly updates via the worldwide web on
the number cats vaccinated, the vaccination protocols followed, and the
development of reactions and sarcomas at the documented sites of vac-
cination. These 40 practices vaccinated more than 30,000 cats in the initial
2-year period of this study. Additional follow-up information was collected
for 1 year. In this group of cats, the incidence of injection site sarcomas was
0.63 per 10,000 cats vaccinated (95% CI: 0.081–2.3 per 10,000 cats).
Although additional sarcomas thought to be related to a previous
vaccination were diagnosed in these clinics over the 3-year study, they were
not included if they occurred as a result of a vaccine given before the
beginning of the study or if the information on the case was incomplete.

The incidence of vaccine site reactions was 11.8 per 10,000 vaccine doses

(95% CI: 9.3–14.9 per 10,000 cats). Of those cases with information on
the time to resolution of the reaction, most had resolved within 3 months
(>95%). Reactions were seen with all types of vaccinations administered
(with and without adjuvant). The frequency of vaccine site reactions was
higher in adjuvant vaccines compared with nonadjuvant vaccinations. Vac-
cines with multiple antigens, particularly FeLV, were also more likely to
cause a reaction at the site of injection.

It is worthwhile to note that none of these studies found any association

with gender or breed of cat and sarcoma development. There are several
potential explanations for the differing estimates of prevalence of injection site
sarcomas. The highest estimates were derived from the results at one or two
practices and could reflect the incidence of sarcoma development with a single
vaccine manufacturer. No manufacturer-related effect has been documented
in the larger epidemiologic studies; however, the low incidence of sarcoma
development may make differences between vaccine manufacturers difficult to
detect. Alternatively, the higher incidence rates may be the result of statistical
chance, and these higher rates would be reduced if a larger number of cats were
followed. Other possible interactions that have not been addressed by the
studies discussed previously include the effect of multiple vaccinations over the
lifetime of the cat, the potential for cats to develop sarcomas years after
a vaccination (which would be missed in studies of shorter duration, ie, 3
years), and the fact that some cats in these studies were followed for relatively
short periods [14]. These are not trivial issues to address, and they may require
extensive epidemiologic prospective studies to evaluate.

The United States Pharmacopeia (USP) is a private not-for-profit organi-

zation that maintains a database of adverse reactions to veterinary products.
At the 1998 Veterinary Cancer Society meeting, Dr. Meyer presented a
description of the first 169 cases of injection site sarcomas as collected by the

556

M. Hauck

/ Vet Clin Small Anim 33 (2003) 553–571

background image

USP (E. Kathryn Meyer, VMD, Rockville, MD, personal communication,
1998). Of the 243 vaccines potentially associated with these tumors, 41% (100)
were rabies; 33% (79) were feline rhinotracheitis virus, calicivirus, and
panleukopenia virus (FRVCP) plus or minus C 20% (49) were FeLV; and 6%
(15) were FRVCP plus or minus chlamydia (C)

/FeLV combinations. In 17

cats, the FRVCP vaccination was the last administered at the location of the
tumor, and in 3 cats, it was the only vaccination ever administered at that site.
Most cats (93%) developed their tumors within 4 years of vaccination. Of
these, 59% were diagnosed within 1 year of vaccination: 11 of these tumors
developed within 1 month of vaccination, 14 developed within 1 to 2 months,
and 11 cats developed tumors within 2 to 3 months.

It is of interest to note that this disease is not a problem exclusive to

North America. A recent survey in the United Kingdom documented 64
sarcomas with histologic characteristics of injection site sarcomas at the sites
of previous vaccinations [15]. These tumors were diagnosed over a 1-year
period through several commercial and university histopathology labora-
tories. The Australian Veterinary Journal has published a series of cases that
were consistent with injection site sarcomas as well [16].

An important part of the history of veterinarians’ response to this

problem is the formation of the Vaccine-Associated Feline Sarcoma Task
Force (VAFSTF) [17,18]. The American Association of Feline Practitioners,
American Animal Hospital Association, American Veterinary Medical
Association, and Veterinary Cancer Society joined resources to found the
VAFSTF. This task force was charged with the formation of a coordinated
mission of research and education with regard to this problem. In addition
to funding multiple research projects in the areas of epidemiology, etiology,
and treatment, the VAFSTF has issued vaccination protocol recommenda-
tions as well as recommendations on an appropriate response to a presumed
vaccine reaction granuloma. The VAFSTF has also played a large role in
the education of the public regarding injection site sarcomas.

Etiology

The first step in understanding a new problem is to describe it in complete

detail. A thorough understanding of the histopathology of injection site
sarcomas also may suggest possible mechanisms of disease development.
The first portion of this section discussing the pathogenesis of injection site
sarcomas is devoted to the histopathologic and cytologic descriptions of
these lesions and the questions that are raised on this basis. The second
portion discusses in greater depth the different avenues of causation that
have been investigated.

Histopathology and cytology

Although not diagnostic of an injection site sarcoma, the characteristic

histopathologic appearance coupled with the appropriate history can suggest

557

M. Hauck

/ Vet Clin Small Anim 33 (2003) 553–571

background image

a high likelihood of this disease syndrome. Doddy and colleagues [19] re-
viewed cats diagnosed with sarcomas (including fibrosarcoma, osteosarcoma,
malignant fibrous histiocytoma, giant cell tumor of soft parts, myofibro-
blastic sarcoma, rhabdomyosarcoma, leiomyosarcoma, chondrosarcoma,
sarcoma, and undifferentiated sarcoma) at the Indiana Animal Disease
Diagnostic Laboratory over a 6-year period. Of the 165 tumors analyzed, 104
arose at common vaccination sites, whereas 61 arose at other sites. The
histopathology of the sarcomas arising at vaccination sites differed from that
of sarcomas at other sites by their subcutaneous location, with necrosis being
more common, as well as an inflammatory infiltrate, increased mitotic index,
pleomorphism, and variable density of the extracellular matrix. The
inflammatory infiltrate was composed predominantly of lymphocytes, but
macrophages were also present. In a multivariate logistic regression analysis,
the vaccine site sarcomas were more likely to be subcutaneous and to
demonstrate an inflammatory component. Younger cats were found to be
more likely to develop a vaccine-associated sarcoma in this study as well.

These findings were consistent with the histopathologic appearance of

vaccine site sarcomas as reported by Hendrick et al [4] in 1992. In this report
and others, the sarcomas at vaccination sites are described as ‘‘surrounded
and partially infiltrated’’ by an inflammatory response composed of lym-
phocytes and macrophages [20]. The macrophages sometimes contained
a gray-brown material that was demonstrated to be aluminum. In addition,
this foreign material was described in postvaccinal granulomatous reactions
in cats and dogs [1]. Whereas inflammation was seen in 15% of sarcomas at
nonvaccination sites, 51% of the vaccine site sarcomas had an inflammatory
element.

On immunohistochemical marker staining, most tumors stained posi-

tively for vimentin, and reactivity to muscle markers was positive in most
cases as well [20]. A chondrosarcoma was positive for the S100 protein,
consistent with the diagnosis. Cytokeratin, CD34, factor VIII–related
antigen, epithelial membrane antigen, and factor 13a were negative. These
results are consistent with the surmised cell of origin for these tumors, the
fibroblast

/myofibroblast.

To characterize these tumors further, ultrastructural evaluation with

transmission electron microscopy was performed [21]. These studies
confirmed the presence of myofibroblasts, giant cells, fibroblastoid cells,
and histiocyte-like cells within the tumors as well as intracellular particulate
material confirmed by dispersive X-ray spectroscopy to be aluminum based.
Immunohistochemistry performed on a subset of these tumors was con-
sistent with the results discussed previously, with seven of seven staining posi-
tive for vimentin and one of seven staining positive for desmin.

Of particular interest are the similarities in the histopathologic ap-

pearance of injection site sarcomas and posttraumatic ocular sarcomas in
cats. Dubielzig and colleagues [22] described 13 cases of intraocular sar-
coma, and 11 of these cats had a history of ocular disease before tumor

558

M. Hauck

/ Vet Clin Small Anim 33 (2003) 553–571

background image

development. Diagnoses included fibrosarcoma (8 cases), osteosarcoma (2
cases), and anaplastic sarcoma (3 cases). The breakdown of the lenticular
capsule was present in all 12 cats in which the entire globe was available.
The latent period to tumor diagnosis was often several years. These tumors
were anaplastic, with variable degrees of differentiation and regions of
granulation tissue and metaplasia [23]. A similar pathogenesis has been
suggested for these two phenomena.

The inflammatory component of injection site sarcomas led several

investigators to study the inflammation present at the site of vaccine reac-
tions in rats, cats, mink, and ferrets. It is interesting to recall that an
increased incidence of vaccine-site reactions noted by the pathologists at the
University of Pennsylvania was responsible, in part, for their recognition of
the association between vaccinations and sarcoma development [1,2]. The
histologic description of the injection site reactions was of a ‘‘focal
necrotizing granulomatous panniculitis’’ in the subcutaneous tissues.

Rats given one of five modified live vaccines with a variety of feline

pathogens failed to produce any injection site reactions [24]. Cats, however,
demonstrated a more vigorous inflammatory response to vaccination.
Schultze and colleagues [25] evaluated three standard feline vaccines in cats
for their ability to produce cytologic evidence of inflammation and to
characterize further that inflammation. Cats were vaccinated at four sites with
saline, FVRCP, FeLV (killed), and rabies virus (killed). Vaccination sites
were aspirated weekly, and the smears were evaluated cytologically. The site
of the saline injection yielded few cells or blood on aspirate. The smears from
the vaccination sites were more cellular, with the cells consisting of
lymphocytes, neutrophils, and macrophages, and they were similar at all
sites at week 1. By the second week, the cellularity at the sites of the rabies and
FeLV vaccinations was higher than at the site of the FVRCP vaccination,
with increased lymphocytes at the both the rabies and FeLV injection sites
compared with the FVRCP site and increased macrophages at the rabies site.
By the third week, the inflammation at the sites of the FeLV and FVRCP
vaccinations had resolved, although the numbers of white blood cells
continued to increase at the site of the rabies vaccination. A similar pattern
was seen at the fourth week. The most common cell type detected throughout
the evaluation period was lymphocytes. All cats developed palpable nodules
at the site of the rabies vaccination, whereas only one other vaccine site
reaction was noted, at the site of an FVRCP vaccination.

A comparison between early vaccine reactions of cats, mink, and ferrets

was performed, in part, because of a case report of an injection site sarcoma in
a ferret [26]. The investigators evaluated two killed rabies vaccines and one
modified live rabies vaccine (with or without the addition of neuraminidase
type V), two killed FeLV vaccines (one with aluminum in the adjuvant), 10%
aluminum ammonium sulfate (alum), and saline [27]. The injection sites were
examined histologically at times from 1 to 21 days. In this study, cats differed
from mink and ferrets in their response to the rabies vaccines, with increased

559

M. Hauck

/ Vet Clin Small Anim 33 (2003) 553–571

background image

collagen and lymphocytes in response to both and increased fibroblasts as well
in response to one type of rabies vaccine. All three species responded similarly
to the FeLV vaccines. Granulomatous inflammation was seen in all species.
The authors conclude that the different response to vaccination may reflect
a subtle difference between these species. This difference may predispose cats’
inflammatory reactions to undergo neoplastic transformation.

A case report on a fibrosarcoma arising at the site of a lufenuron injection

prompted investigation into injection reactions seen with this agent (Dennis
Macy, DVM, Fort Collins, CO, personal communication, 2000) [28]. Six
rats were injected with lufenuron subcutaneously, and the injection sites
were evaluated histologically 21 days later. All six rats developed injection
site reactions, but the nature of these reactions was different from those seen
in previous studies with vaccines. These reactive infiltrates were composed
predominantly of macrophages, without the increased numbers of neu-
trophils, lymphocytes, and plasma cells.

Current research on injection site sarcomas

The presence of the inflammatory capsule or infiltrate of injection site

sarcomas, which is seen much more rarely in noninjection site sarcomas,
suggested to some investigators that inflammation may play a role in on-
cogenesis. This suspicion also resulted in the studies of vaccine reactions
discussed previously. This hypothesis was first proposed in 1992, with the
observation that some cases of injection site sarcoma were ‘‘transitional’’
foci of neoplastic cells within granulomatous inflammation [4]. This model
would be consistent with the development of ocular sarcomas at sites of
previous trauma or inflammation. There are several experimental models
demonstrating the role of inflammation in tumor promotion as well (for
a review, see the article by Macy [29]). It is hypothesized that the growth
factors elucidated at the site of inflammation interact with cells to result in
oncogenesis. For example, chickens infected with the Rous sarcoma virus
develop tumors at the site of injection. If a wound is made away from the
injection site, a tumor develops at the wounding site [30]. If, however,
inflammation at the wound is suppressed with methylprednisolone, no tumor
develops. The growth factors identified as sufficient to cause this effect are
tumor growth factor-b (TGFb) and acidic and basic fibroblastic growth
factors (aFGF and bFGF). This work is supportive of the concept that
inflammation is a necessary component of injection site tumor development.
It is also consistent with the observation that these tumors are seen more
commonly with killed vaccines (that contain more adjuvant) than with
modified live vaccines. To evaluate this hypothesis further, immunohisto-
chemistry was used to determine the expression of epidermal growth factor
(EGF) and its receptor, FGF and its receptor, platelet-derived growth factor
(PDGF) and its receptor, c-jun (AP-1), and TGFb in normal feline skin,
wounded skin, vaccine reactions, early malignant lesions, and injection site

560

M. Hauck

/ Vet Clin Small Anim 33 (2003) 553–571

background image

sarcomas as well as in sarcomas at other sites [31]. Sarcomas arising at
injection sites demonstrated consistent staining for EGF, PDGF, TGFb and
their receptors as well as for c-jun. Noninjection site sarcomas stained weakly
for these growth factors. The lymphocytes in the injection site tumors and
vaccine site reactions also stained positively for PDGF, although the lympho-
cytes of the noninjection site sarcomas and nonvaccine inflammation did
not stain for PDGF [32,33]. The strongest expression of the PDGF receptor
was in the adjacent sarcoma cells. Recently, the lymphoid follicles typically
seen along the periphery of the tumor have been shown on immunohisto-
chemistry to be composed of a high proportion of T cells (19%–87%) [34].

Additional studies of growth factor expression have been performed on

injection site sarcoma cell lines (J. Carew, Madison, WI, personal communi-
cation, 1999). The pattern of expression of PDGFa, PDGFb, PDGF
receptor, hepatocyte growth factor, c-met, EGF receptor, and insulin-like
growth factor-1 is being determined in 13 injection site sarcoma cell lines as
well as in 5 noninjection site sarcoma cell lines. In an extension of this de-
scriptive work, these investigators have also evaluated the ability of tyrosine
kinase inhibitor STI-571 to repress the proliferation of these cell lines in
vitro and to suppress receptor autophosphorylation (Ilene Kurzman, PhD,
Madison, WI, personal communication, 2001). STI-571 inhibited cell prolif-
eration and PDGF receptor autophosphorylation even in the presence of
PDGF.

In general, chronic inflammation is not thought to be capable of inducing

neoplastic transformation alone, and host factors are considered to be
essential in the transformation of cells. These factors could take the form of
infection with oncogenic viruses or genetic mutations present within host
cells that play a role in cancer initiation. This hypothesis is consistent with
the low incidence of tumor formation in the population of cats at risk.

The role of concurrent viral infection has been evaluated in cats with

injection site sarcomas. The transformative role of viruses in cats is well
known, particularly in the instance of FeLV. Another oncogenic virus, feline
sarcoma virus (FeSV), is a replication deficient variant of FeLV that
requires coinfection with FeLV to become pathogenic. Previous retrospec-
tive studies failed to demonstrate an association between serologic evidence
of infection with FeLV or feline immunodeficiency virus (FIV) and injection
site sarcoma development [7]. Further elucidation of the potential role of
viral infection has been performed using direct analysis of tumor cells with
immunohistochemistry, polymerase chain reaction (PCR) and reverse
transcriptase PCR evaluation, and in situ hybridization of the tumor cell
DNA [35–40]. A number of potentially oncogenic viruses were investigated,
including FeLV, FIV, polyomavirus, papillomavirus, endogenous FeLV,
and feline foamy virus. Immunohistochemistry and PCR failed to detect the
presence of FeLV

/FeSV in 130 suspected injection site sarcomas [35]. When

testing for endogenous FeLV and FIV, 50 sarcomas from injection sites
were compared with 50 sarcomas from noninjection sites. The remaining

561

M. Hauck

/ Vet Clin Small Anim 33 (2003) 553–571

background image

three viruses were evaluated only on sarcomas of injection sites. When
comparing the presence of endogenous FeLV RNA, there was no difference
between injection site and noninjection site sarcomas [36]. No evidence of
infection with FIV, polyomavirus, papillomavirus, or feline foamy virus was
detected in any of the tumor samples [37–40]. Given these results, it is
unlikely that any of these viruses have a role in the development of injection
site sarcomas.

Other groups have investigated the role of mutations in tumor suppressor

genes, such as p53 (Elizabeth Hershey, DVM, Madison, WI, personal
communication, 1999; Sagarika Kanjilal, PhD, St. Paul, MN, personal
communication, 1999) [41,42]. In human tumors, p53 mutations have been
associated with initiation and progression of neoplasia. Mutations in p53
result in a longer half-life of p53 protein in the cell, which can be detected
using immunohistochemistry. In a series of 40 injection site sarcomas, 17
stained darkly for p53 expression in the nucleus, the nuclei of 8 tumors
stained palely, and 15 tumors did not stain [42]. Sequence analysis of the p53
gene in 8 tumors in which the nuclei stained darkly demonstrated single mis-
sense mutations in 5 tumors [41]. These mutations were not present in
the normal tissues, suggesting that although the abnormal p53 protein may
have contributed to cancer progression, it is unlikely that p53 mutations
were a factor in tumor development. In another study, 1 of 20 injection
site sarcomas had a point mutation in p53, and 7 of 18 had loss of
heterozygosity at this gene (Sagarika Kanjilal, PhD, St. Paul, MN, personal
communication, 1999).

The tools of genomics have only just been brought to the study of

injection site sarcomas. Identification of chromosomal alterations in in-
jection site sarcomas may allow identification of cats at risk for tumor
development and better understanding of the pathogenesis. The production
of feline chromosome-specific probes was the first step for evaluating
the karyotype of injection site sarcomas [43]. The karyotypes of five
injection site sarcomas were evaluated using these probes, and all five were
‘‘highly aberrant’’ [44]. These abnormal chromosomal regions require
further evaluation before their significance is known. Evaluation of early
evidence of DNA damage in vaccination reactions is also under current
investigation (J. Mac Law, DVM, PhD, Raleigh, NC, personal communi-
cation, 2002). The unique nature of this inducible tumor offers many
opportunities to study tumorigenesis that may be relevant to human cancer
as well [45].

The in vitro chemosensitivity of injection site sarcoma cell lines also has

been evaluated. Two cell lines were established from injection site sarcomas
and exposed to various levels of five chemotherapeutic agents [46]. The
drugs included carboplatin (Paraplatin), doxorubicin (Adriamycin), mitox-
antrone (Novantrone), methotrexate (Folex, Mexate), and vincristine (On-
covin). Of these agents, mitoxantrone and doxorubicin demonstrated the
highest degree of cytotoxicity. Recently, four injection site sarcoma cell lines

562

M. Hauck

/ Vet Clin Small Anim 33 (2003) 553–571

background image

were grown in the presence of vincristine and paclitaxel. Both drugs
showed significant cytotoxicity at concentrations that may be achievable
in patients [47].

Clinical presentation and treatment

As mentioned previously, injection site sarcomas are typically diagnosed

in cats at a younger age than noninjection site sarcomas, and the tumors are
frequently larger at the time of diagnosis [7,19]. As veterinarians are alerted
to the possibility of sarcoma development at the site of a vaccination
reaction, the delay to diagnosis has hopefully decreased and the size of tu-
mors at initial presentation no longer may be as dramatic. It is important to
note that these tumors may develop after the injection of substances other
than vaccinations [28,48]. On presentation, these masses might have a cystic
cavity as a result of the large necrotic center that is a common histo-
pathologic aspect of injection site sarcomas. Often, strands of tumor tissue
extending to the underlying musculature or dorsal spinal processes can
be palpated.

A biopsy is necessary for definitive diagnosis. The degree of inflammation

associated with these tumors makes interpretation of a cytology sample
difficult. Biopsies should be taken with the final treatment protocol in mind,
with particular emphasis on placing the biopsy tract within tissue that is to
be removed at the time of surgical excision of the tumor. The current
recommendations of the VAFSTF for management of a mass that develops
at the site of a vaccine are to record the size and location and to perform
a biopsy if the mass (1) persists for more than 3 months, (2) is greater than
2 cm in diameter, or (3) is increasing in size 1 month after injection [17].

Currently, there are no known histopathologic prognostic indicators for

feline injection site sarcomas. Argyrophilic nucleolar organizer region
(AgNOR) staining has been shown to be a useful prognostic factor in several
canine tumors and was evaluated in 21 cats with injection site sarcomas
(Laura Garrett, DVM, Manhattan KS, personal communication, 2001). No
correlation was found between cats with fewer or greater numbers of
AgNORs than the median AgNOR count and the incidence of metastasis or
duration of survival. A grading scheme has been applied to injection site
sarcomas in cats, but no correlation with outcome has been documented [34].

The evaluation of a tumor for surgical excision or radiation therapy

requires more definitive evaluation than simple physical palpation of the
tumor. When contrast-enhanced CT images are used to determine the
tumor volume and are compared with tumor volumes based on physical
examination measurements, the inadequacy of the physical examination to
determine tumor extent becomes apparent. A study compared the two
methods of determining tumor volume [49]. Thirty-five cats with injection
site sarcomas had their tumor volumes measured by physical examination
and with CT (with and without contrast). The tumor volumes of 33 cats

563

M. Hauck

/ Vet Clin Small Anim 33 (2003) 553–571

background image

were larger when calculated using the postcontrast CT images. The median
tumor volume as measured on physical examination was 23.4 cm

3

, and

the median tumor volume as measured on postcontrast CT images was
57.2 cm

3

. The authors found that treatment recommendations were often

altered based on the CT images, further emphasizing the need for accurate
imaging before formulating a treatment plan.

In addition to careful evaluation of the extent of the primary tumor,

standard staging and health assessment are performed in feline patients with
injection site sarcomas. The most common sites of metastasis are the lungs,
followed by the lymph nodes and other internal organs [50]. The percentage
of cats with metastasis at the time of presentation is relatively low, with
studies reporting from 5% to 5.3% (Tetsuya Kobayashi, DVM, Raleigh,
NC, personal communication, 1999) [51]. The overall metastatic rate of cats
treated for injection site sarcomas has been reported to range from 0% to
28% [51,52]. In a retrospective study of 92 cats treated with multimodality
therapy at North Carolina State University, 21.7% (20 of 92 cats) developed
detectable metastasis after treatment. The median time to metastasis in these
20 cats was 155 days (range: 66–1022 days, unpublished data). The absolute
rate of metastasis may be higher, because not all cats were dead at the time
of analysis and not all cats were necropsied [50].

Surgical excision as the sole modality of treatment has been shown to be

inadequate in a number of studies, although the importance of surgery in
a multimodality approach is unquestionable. A retrospective analysis of 61
cats from two veterinary teaching hospitals treated with surgical excision
reported a median overall time to recurrence of 94 days, with only 11% of
cats remaining tumor-free for at least 1 year [51]. Surgery performed at the
referral institutions resulted in an increased median time to recurrence of
274 days. Cats with appendicular tumors (most commonly treated with
amputation) had a longer time to recurrence than cats with tumors at other
sites. Of the 5 cats that were long-term survivors (>1300 days), 4 were
treated with amputation. Interestingly, the median survival time for these 61
cats was 576 days, and many cats had multiple surgical procedures before
succumbing to their disease. Similar results with surgical excision were
reported from a third veterinary teaching hospital, with an overall median
disease-free interval (DFI) for cats treated with surgery alone of 124 days
(Carrie Wood. DVM, St. Paul, MN, personal communication, 1999). For
cats with complete excision, the median DFI was 221 days. A third study
had an overall median DFI of 10 months, and cats with complete tumor
excisions had a DFI of longer than 16 months [53].

There have been a few smaller studies evaluating more radical excision of

injection site sarcomas with promising results. Lidbetter et al [54] described
6 cats that underwent a lateral body wall resection for this tumor. Three cats
had preoperative radiation therapy. At a mean follow-up of 17.2 months, no
cats had evidence of recurrence. Kuntz [55] reported on a modified wide
local excision technique, in which 5-cm margins in all directions and two

564

M. Hauck

/ Vet Clin Small Anim 33 (2003) 553–571

background image

muscle planes deep were taken at surgery. Histopathologically, 23 of 24 cats
had complete excisions, and follow-up of these patients is continuing to
determine the effectiveness of this approach. At North Carolina State
University, surgeons currently excise 3- to 5-cm margins around injection
site sarcomas, removing as much of the surrounding tissue as possible while
still leaving the cats able to function relatively normally (Elizabeth Hardie,
DVM, PhD, Raleigh, NC, personal communication, 2002).

Limited evaluation has been done on the use of radiation therapy alone for

the treatment of injection site sarcomas, because radiation would not typically
be used as a single modality for palpable disease. A small study evaluating
hypofractionated radiation therapy (four weekly fractions of 8 Gray given
with palliative intent) with or without chemotherapy in cats with gross disease
demonstrated a median progression-free interval (PFI) of 210 days (Janean
Fidel, DVM, Zurich, Switzerland, personal communication, 2001).

The most promising outcomes achieved in treating cats with injection site

sarcomas have been the result of multimodality therapy, including radiation
therapy and surgery. Several types of radiation therapy have been evaluated in
combination with surgery, including orthovoltage (administered after
surgery, median DFI of 313 days) and brachytherapy (administered after
surgery, median DFI not yet achieved) as well as megavoltage (including
electron beam therapy) (Janet Burke, VMD, Philadelphia, PA, personal
communication, 1999) [56]. A recent retrospective study of 25 cats treated at
Colorado State University with surgery and megavoltage radiation, with or
without doxorubicin chemotherapy, reported an overall median survival of
701 days [52]. Seven cats received surgery, followed by 57-Gy radiation
therapy (19 daily fractions of 3 Gy). The remaining 18 cats were treated with
surgery, radiation therapy, and doxorubicin chemotherapy in the adjuvant
setting. Doxorubicin did not appear to improve median DFI, but the small size
of the groups and retrospective nature of this study make it difficult to
determine the true impact of chemotherapy in this setting. These results
contrast with those presented by King and colleagues [57]. In this study, 61 cats
were treated with preoperative radiation therapy (60–62 Gy on a Monday

/

Wednesday

/Friday schedule over 7 weeks) and surgical excision, either with

or without five doxorubicin treatments. The DFI in the cats receiving
doxorubicin was prolonged as compared with the cats not receiving che-
motherapy (median of 360 days versus 162 days); however, there was no
difference in length of survival.

A retrospective study evaluating the effect of the extent of surgery was

performed in 76 cats treated at Auburn University [58]. Nineteen of these
cats were treated with conservative resection of the palpable disease. All cats
received postoperative radiation therapy of 52 Gy (13 fractions of 4 Gy
given on a Monday

/Wednesday/Friday schedule) with 4- to 8-MeV elec-

trons. Median DFI for these cats was 405 days, with a median survival of
469 days from the start of radiation therapy. No difference in recurrence
rate was detected between cats treated with a conservative versus wide first

565

M. Hauck

/ Vet Clin Small Anim 33 (2003) 553–571

background image

excision, although the statistical power of this comparison was low. Com-
pleteness of excision did not affect DFI in this group of cats. Cats that
had undergone multiple surgeries before radiation therapy had a decreased
DFI compared with cats with only a single sarcoma excision.

At the North Carolina Animal Cancer Program, we recently evaluated

outcome in 92 cats treated since 1985, with definitive preoperative radiation
therapy (16 daily fractions of 3 Gy) and wide surgical excision [50]. Thirty-
three of these cats were reported in a previous publication [59]. Of these 92
patients, 33 received chemotherapy: 19 received only carboplatin and 14
received other chemotherapy agents (doxorubicin, vincristine, cyclophos-
phamide [Cytoxan], with or without carboplatin). In this study, first-event
analysis was used, which is a conservative statistical method where the
only animals censored are those that are alive and disease-free at the time
of analysis. The overall median time to first event (death, metastasis, or
local recurrence) was 1.6 years. The completeness of surgical excision was
the only variable predictive of time to first event. For cats with complete
surgical excision of their sarcoma, time to first event was 2.7 years. Median
time to first event was 0.8 years for cats with incomplete tumor resection.

The impact of chemotherapy on the treatment of injection site sarcomas

has yet to be clearly determined. Multiple chemotherapy agents have been
shown to have activity against injection site sarcomas, including carboplatin,
doxorubicin, and cyclophosphamide. Two phase I trials of carboplatin in cats
reported at least partial responses (>50% reduction in size) in cats with
injection site sarcomas, including 6 of 16 cats (37.5%) in one trial (William
Kisseberth, DVM, Madison, WI, personal communication, 1996; Carrie
Wood, DVM, North Grafton, MA, personal communication, 1996). An
additional 12 cats whose tumors were deemed unresectable were treated with
a combination of doxorubicin and cyclophosphamide in another study [60].
Six of the 12 cats had at least a partial response to this combination protocol,
although the responses were not durable (median of 125 days). The two
longest responses were in cats that achieved clinical complete remission on
this protocol. A case report from Europe also supports the assessment that
doxorubicin has activity against injection site sarcomas [61]. In this report,
a cat with an injection site sarcoma was presented to the Veterinary Teaching
Hospital in Vienna after the third excision of its tumor and was treated with
adjuvant doxorubicin. Recurrence of the tumor was delayed until more than
12 months after surgery and adjuvant doxorubicin, whereas the first two
relapses had occurred by 2 and 3 weeks after surgery, respectively (Miriam
Kleiter, DVM, Raleigh, NC, personal communication, 2002).

Doxil is a liposome-encapsulated form of doxorubicin. The efficacy of

Doxil in comparison to unencapsulated doxorubicin was evaluated in a
randomized trial in which these two formulations were given either to cats
with palpable disease or to cats that had incomplete surgical excisions of
their tumors [62]. The PFI in cats receiving chemotherapy was compared
with that in a historical control group of cats that were treated with surgery

566

M. Hauck

/ Vet Clin Small Anim 33 (2003) 553–571

background image

alone. The cats with microscopic disease that received either formulation of
doxorubicin had a significant increase in their PFI (median of 388 days)
compared with the surgical control group’s PFI (median of 93 days).
No difference in PFI was seen between the two treatment groups. Some
cats receiving Doxil, however, developed a delayed nephrotoxicity, leading
the investigators to conclude that free doxorubicin was the preferred
formulation.

In the retrospective study at North Carolina State University, chemo-

therapy did not result in an increased time to first event, although the 10 of
19 cats receiving carboplatin had not yet failed therapy, with a median
follow-up time in these 10 cats of 3.5 years [50]. The small sample size
necessitates the evaluation of this combination in a larger number of cats
before conclusions can be drawn; however, the initial results warrant further
investigation.

Additional prospective studies are required to address adequately the

role that chemotherapy, used in the adjuvant or neoadjuvant setting, should
have in the treatment of injection site sarcomas. The high rate of response,
even in phase I trials, suggests that chemotherapy may be a useful addition
to multimodality treatment.

The role of immunotherapy and novel approaches in the treatment of this

disease is unclear. Acemannan has been used to treat sarcomas in cats, but
no controlled studies have been performed to date [63]. The use of tyrosine
kinase inhibitors to block PDGF receptor signaling is an interesting ap-
proach to treatment and, possibly, prevention of injection site sarcomas,
but this modality of treatment is not yet being tested in the clinic. At North
Carolina State University, we have evaluated the use of local hyperthermia
to enhance liposome uptake in injection site sarcomas, and we were able to
demonstrate a significant increase in liposome accumulation ranging from
2- to 13-fold in the heated tumors [64]. This approach may be used to im-
prove drug delivery to tumors. In conjunction with investigators at Duke
University, North Carolina State University and Colorado State University
are also evaluating interleukin-12 gene therapy in injection site sarcomas.
Much remains to be learned regarding the role of these novel approaches in
the treatment of this tumor.

Prevention

Injection site sarcomas are a challenge to the veterinary profession on

a number of levels: they are iatrogenic, difficult to treat, and potentially
preventable. Thoughtful vaccination protocols to avoid the vaccination of
cats against diseases for which they are not at risk represent an often
recommended and relatively easy approach to integrate into routine well cat
care. The use of antibody titers to determine the requirement for booster
vaccinations is currently under evaluation, and initial results are promising
[65]. Although we can decrease the number of cats vaccinated against FeLV

567

M. Hauck

/ Vet Clin Small Anim 33 (2003) 553–571

background image

and use modified live FVRCP vaccines, vaccinating cats against rabies is
a matter of law in most states as well as being a significant public health
concern. For the years 1998 through 2000, more than twice as many cats
as dogs tested positive for the rabies virus in the United States [66–68].
In a population survey from Pennsylvania during 1995, although 75% of
the bites were from dogs, people bitten by cats were six times more likely
to receive rabies postexposure prophylaxis (PEP) [69]. Dog bites accounted
for 30% of the PEP treatments, whereas cat bites accounted for 44% of
PEP—the largest group. When evaluating the percentage of owned animals
that were up to date on their rabies vaccination, 59% of dogs were vac-
cinated but only 41% of cats were current on their rabies vaccination.
Clearly, there is still room for improvement in the rabies vaccination of pet
cats in terms of the public health arena.

Following the recommendations of the VAFSTF on standardization of

vaccination protocols will help veterinarians to better correlate a given
vaccination and subsequent tumor development. These recommendations
include administering any vaccine containing rabies as distally as possible
in the right hind leg, administering FeLV vaccinations as distally as
possible in the left hind leg, and injecting FVRCP (with or without
chlamydia) vaccines in the right shoulder. Mixing of vaccinations is dis-
couraged. Documentation of the vaccination site as well as the manu-
facturer and lot or serial number is important if an adverse event develops.
All adverse reactions (including sarcoma development) would ideally be
reported to the USP (1-800-4-USP-PRN) for better population surveillance.
This problem is unlikely to go away on its own, and it is important
for veterinarians to play an active role in the prevention, monitoring,
and development of treatments for injection site sarcomas.

References

[1] Hendrick MJ, Dunagan CA. Focal necrotizing granulomatous panniculitis associated with

subcutaneous injection of rabies vaccine in cats and dogs: 10 cases (1988–1989). JAVMA
1991;198:304–5.

[2] Hendrick MJ, Goldschmidt MH. Do injection site reactions induce fibrosarcomas in cats?

JAVMA 1991;199:968.

[3] Thornburg LP. Postvaccination sarcomas in cats. JAVMA 1993;203:193–4.
[4] Hendrick MJ, Goldschmidt MH, Shofer FS, et al. Postvaccinal sarcomas in the cat:

epidemiology and electron probe microanalytical identification of aluminum. Cancer Res
1992;52:5391–4.

[5] Slater DN, Underwood JCN, Durrant TE, et al. Aluminum hydroxide granulomas: light

and electron microscopic studies and x-ray microanalysis. Br J Dermatol 1982;107:103–8.

[6] Kass PH, Barnes WG, Spangler WL, et al. Epidemiologic evidence for a causal relation

between vaccination and fibrosarcoma tumorigenesis in cats. JAVMA 1993;203:396–405.

[7] Hendrick MJ, Shofer FS, Goldschmidt MH, et al. Comparison of fibrosarcomas that

developed at vaccination sites and at nonvaccination sites in cats—239 cases (1991–1992).
JAVMA 1994;205:1425–9.

568

M. Hauck

/ Vet Clin Small Anim 33 (2003) 553–571

background image

[8] Esplin DG, McGill LD, Meininger AC, et al. Postvaccination sarcomas in cats. JAVMA

1993;202:1245–7.

[9] Coyne MJ, Reeves NCP, Rosen DK. Estimated prevalence of injection-site sarcomas in

cats during 1992. JAVMA 1997;210:249–51.

[10] Lester S, Clemett T, Burt A. Vaccine site-associated sarcomas in cats—clinical experience

and a laboratory review (1982–1993). J Am Anim Hosp Assoc 1996;32:91–5.

[11] Hendrick MJ. Historical review and current knowledge of risk factors involved in feline

vaccine-associated sarcomas. JAVMA 1998;213:1422–3.

[12] Macy DW, Hendrick MJ. The potential role of inflammation in the development of

postvaccinal sarcomas in cats. Vet Clin North Am Small Anim Pract 1996;26:103–9.

[13] Bergman P, Hendrick MJ, Macy D, et al. Feline sarcoma and vaccination. Vet Forum

1999;March:40–7.

[14] Gobar GM, Kass PH. World wide web-based survey of vaccination practices, postvaccinal

reactions and vaccine site-associated sarcomas in cats. JAVMA 2002;220:1477–82.

[15] Tennant B. Feline injection-site fibrosarcomas: results of a BSAVA survey. J Small Anim

Pract 2000;41:181–2.

[16] Burton G, Mason KV. Do postvaccinal sarcomas occur in Australian cats? Aust Vet J

1997;75:102–6.

[17] Morrison WB, Starr RM. Vaccine-associated feline sarcomas. JAVMA 2001;218:697–702.
[18] Starr RM. Vaccine-Associated Feline Sarcoma Task Force: a new model for problem

solving in veterinary medicine. JAVMA 1998;213:1428–9.

[19] Doddy FD, Glickman LT, Glickman NW, et al. Feline fibrosarcomas at vaccination sites

and non-vaccination sites. J Comp Pathol 1996;114:165–74.

[20] Hendrick MJ, Brooks JJ. Postvaccinal sarcomas in the cat—histology and immunohisto-

chemistry. Vet Pathol 1994;31:126–9.

[21] Madewell BR, Griffey SM, McEntee MC, et al. Feline vaccine associated fibrosarcoma: an

ultrastructural study of 20 tumors (1996–1999). Vet Pathol 2001;38:196–202.

[22] Dubielzig RR, Everitt J, Shadduck JA, et al. Clinical and morphologic features of post-

traumatic ocular sarcomas in cats. Vet Pathol 1990;27:62–5.

[23] Dubielzig RR. Ocular sarcoma following trauma in three cats. JAVMA 1984;184:578–81.
[24] Macy DW, Chretin J. Local postvaccinal reactions of a recombinant rabies vaccine. Vet

Forum 1999;August:44–9.

[25] Schultze AE, Frank LA, Hahn KA. Repeated physical and cytologic characterizations of

subcutaneous postvaccinal reactions in cats. Am J Vet Res 1997;58:719–24.

[26] Murray J. Vaccine injection-site sarcoma in a ferret. JAVMA 1998;213:955.
[27] Carroll EE, Dubielzig RR, Schultz RD. Cats differ from mink and ferrets in their response

to commercial vaccines: a histologic comparison of early vaccine reactions. Vet Pathol
2002;39:216–27.

[28] Esplin DG, Bigelow M, McGill LD, et al. Fibrosarcoma at the site of a lufenuron injection

in a cat. Vet Cancer Soc Newsletter 1999;23:8–9.

[29] Macy DW. The potential role and mechanisms of FeLV vaccine-induced neoplasms. Semin

Vet Med Surg 1995;10:234–7.

[30] Martins-Green M, Boudreau N, Bissell MJ. Inflammation is responsible for the

development of wound-induced tumors in chickens infected with Rous sarcoma virus.
Cancer Res 1994;54:4334–41.

[31] Dambach D, Carlson J, Riddle D, et al. Immunohistochemical identification and

localization of growth factors in feline postvaccinal lesions [abstract 150]. Vet Pathol
1996;33:607.

[32] Hendrick MJ. Feline vaccine-associated sarcomas: current studies on pathogenesis.

JAVMA 1998;213:1425–6.

[33] Hendrick MJ. Feline vaccine-associated sarcomas. Cancer Invest 1999;17:273–7.
[34] Couto SS, Griffey SM, Duarte PC, et al. Feline vaccine-associated fibrosarcoma:

morphologic distinctions. Vet Pathol 2002;39:33–41.

569

M. Hauck

/ Vet Clin Small Anim 33 (2003) 553–571

background image

[35] Ellis JA, Jackson ML, Bartsch RC, et al. Use of immunohistochemistry and polymerase

chain reaction for detection of oncornaviruses in formalin-fixed, paraffin-embedded
fibrosarcomas from cats. JAVMA 1996;209:767–71.

[36] Kidney BA, Ellis JA, Haines DM, et al. Comparison of endogenous feline leukemia virus

RNA content in feline vaccine and nonvaccine site-associated sarcomas. Am J Vet Res
2001c;62:1990–4.

[37] Kidney BA, Ellis JA, Haines DM, et al. Evaluation of formalin-fixed paraffin-embedded

tissues obtained from vaccine site-associated sarcomas of cats for DNA of feline
immunodeficiency virus. Am J Vet Res 2000;61:1037–41.

[38] Kidney BA, Haines DM, Ellis JA, et al. Evaluation of formalin-fixed paraffin-embedded

tissues from feline vaccine site-associated sarcomas for feline foamy virus DNA. Am J Vet
Res 2002;63:60–3.

[39] Kidney BA, Haines DM, Ellis JA, et al. Evaluation of formalin-fixed paraffin-embedded

tissues from vaccine site-associated sarcomas of cats for papillomavirus DNA and antigen.
Am J Vet Res 2001b;62:833–9.

[40] Kidney BA, Haines DM, Ellis JA, et al. Evaluation of formalin-fixed paraffin-embedded

tissues from vaccine site-associated sarcomas of cats for polyomavirus DNA and antigen.
Am J Vet Res 2001a;62:828–32.

[41] Nambiar PR, Haines DM, Ellis JA, et al. Mutational analysis of tumor suppressor gene

p53 in feline vaccine site-associated sarcomas. Am J Vet Res 2000;61:1277–81.

[42] Nambiar PR, Jackson ML, Ellis JA, et al. Immunohistochemical detection of tumor

suppressor gene p53 protein in feline injection site-associated sarcomas. Vet Pathol
2001;38:236–8.

[43] Hoots EA, McNeil EA, LaRue SM. Characterization of genetic alterations in feline

vaccine-associated sarcoma using whole chromosome painting probes [abstract 83]. J Vet
Intern Med 2001;15:292.

[44] McNeil EA, Hoots EA, LaRue SM. Characterization of chromosomal aberrations in feline

vaccine-associated sarcoma using comparative genomic hybridization [abstract 84]. J Vet
Intern Med 2001;292.

[45] McNeil EA. Vaccine-associated sarcomas in cats—a unique cancer model. Clin Orthop

2001;382:21–7.

[46] Williams LE, Banerji N, Klausner JS, et al. Establishment of two vaccine-associated feline

sarcoma cell lines and determination of in vitro chemosensitivity to doxorubicin and
mitoxantrone. Am J Vet Res 2001;62:1354–7.

[47] Banerji N, Li X, Klausner JS, et al. Evaluation of in vitro chemosensitivity of vaccine-

associated feline sarcoma cell lines to vincristine and paclitaxel. Am J Vet Res 2002;63:
728–32.

[48] Gagnon AC. Drug injection-associated fibrosarcoma in a cat. Feline Pract 2000;28:18–21.
[49] McEntee MC, Samii VF. The utility of contrast enhanced computed tomography in feline

vaccine associated sarcomas: 35 cases [abstract]. Vet Radiol Ultrasound 2000;41:575.

[50] Kobayashi T, Hauck ML, Dodge R, et al. Preoperative radiotherapy for vaccine site

sarcoma in 92 cats. Vet Radiol Ultrasound 2002;43:473–9.

[51] Hershey AE, Sorenmo KU, Hendrick MJ, et al. Prognosis for presumed feline vaccine-

associated sarcoma after excision: 61 cases (1986–1996). JAVMA 2000;216:58–61.

[52] Bregazzi VS, LaRue SM, McNiel E, et al. Treatment with a combination of doxorubicin,

surgery, and radiation versus surgery and radiation alone for cats with vaccine-associated
sarcomas: 25 cases (1995–2000). JAVMA 2001;218:547–50.

[53] Davidson EB, Gregory CR, Kass PH. Surgical excision of soft tissue fibrosarcomas in cats.

Vet Surg 1997;26:265–9.

[54] Lidbetter DA, Williams FA, Krahwinkel DJ, et al. Radical lateral body-wall resection for

fibrosarcoma with reconstruction using polypropylene mesh and a caudal superficial
epigastric axial pattern flap: a prospective clinical study of the technique and results in 6
cats. Vet Surg 2002;31:57–64.

570

M. Hauck

/ Vet Clin Small Anim 33 (2003) 553–571

background image

[55] Kuntz CA. Modified wide local excision for vaccine associated soft tissue sarcomas in cats

[abstract 7]. Vet Surg 2000;29:481.

[56] Rogers K, Walker MA, Barton CL, et al. Post-operative brachytherapy for vaccine-

associated sarcoma in cats [abstract]. Vet Radiol Ultrasound 2000;41:577.

[57] King GK, Harris DF, Hottinger H, et al. Prolonged remission and survival time in cats

with injection-site sarcomas using pre-operative radiation and doxorubicin protocol: 61
cases (1998–2000) [abstract]. Vet Radiol Ultrasound 2001;42:585.

[58] Cohen M, Wright JC, Brawner WR, et al. Use of surgery and electron beam irradiation,

with or without chemotherapy, for treatment of vaccine-associated sarcomas in cats: 78
cases (1996–2000). JAVMA 2001;219:1582–9.

[59] Cronin K, Page RL, Spodnick G, et al. Radiation therapy and surgery for fibrosarcoma in

33 Cats. Vet Radiol Ultrasound 1998;39:51–6.

[60] Barber LG, Sorenmo KU, Cronin KL, et al. Combined doxorubicin and cyclophospha-

mide chemotherapy for nonresectable feline fibrosarcoma. J Am Anim Hosp Assoc 2000;
36:416–21.

[61] Kleiter M, Leschnik M. Postoperative chemotherapie zur behandlung eines zweifach

rezidivierten vakzine-assoziierten fibrosarkoms. Kleintierpraxis 1998;43:295–302.

[62] Poirier VJ, Thamm DH, Kurzman ID, et al. Liposome-encapsulated doxorubicin (Doxil)

and doxorubicin in the treatment of vaccine-associated sarcoma in cats. J Vet Intern Med
2002;16:726–31.

[63] Harris C, Pierce K, King G, et al. Efficacy of acemannan in treatment of canine and feline

spontaneous neoplasms. Mol Biother 1991;3:207–13.

[64] Matteucci ML, Anyarambhatla G, Rosner G, et al. Hyperthermia increases accumulation

of technetium-99m-labeled liposomes in feline sarcomas. Clin Cancer Res 2000;6:3748–55.

[65] Lappin MR, Andrews J, Simpson D, et al. Use of serologic tests to predict resistance to

feline herpesvirus 1, feline calicivirus, and feline parvovirus infection in cats. JAVMA
2002;220:38–42.

[66] Krebs JW, Mondul AM, Rupprecht CE, et al. Rabies surveillance in the United States

during 2000. JAVMA 2001;219:1687–99.

[67] Krebs JW, Smith JS, Rupprecht CE, et al. Rabies surveillance in the United States during

1998. JAVMA 1999;215:1786–98.

[68] Krebs JW, Rupprecht CE, Childs JE. Rabies surveillance in the United States during 1999.

JAVMA 2000;217:1799–811.

[69] Moore DA, Sischo WM, Hunter A, et al. Animal bite epidemiology and surveillance for

rabies postexposure prophylaxis. JAVMA 2000;217:190–4.

571

M. Hauck

/ Vet Clin Small Anim 33 (2003) 553–571

background image

Canine mammary gland tumors

Karin Sorenmo, DVM

University of Pennsylvania, School of Veterinary Medicine,

Department of Clinical Studies, 3900 Delancey Street,

Philadelphia, PA 19104, USA

Mammary tumors are the most common type of tumors in intact female

dogs. Several epidemiologic studies have been conducted over the years and
provide an estimate of the incidence of canine mammary gland tumors. A
survey from Alameda and Contra Costa counties in California from 1963
through 1966 found that mammary gland tumors comprised 13.4% of all
tumors in dogs and 41.7% of all tumors in intact female dogs [1]. The
incidence of canine mammary gland tumors in the United States, however,
has been reduced significantly since that time because of the common
practice of performing ovariohysterectomy (OHE) at an early age.
Mammary gland tumors are much more common in many European
countries where ovariohysterectomy is not routinely performed. Recent data
from The Norwegian Canine Cancer Register reported a crude incidence of
malignant mammary gland tumors of 53.3% in female dogs of any breed [2].
This number appears high but may be influenced by the common use of
progestins to prevent estrus in this particular population. A previous study
from the same group found an increased risk, with an odds ratio of 2.32, of
mammary gland tumor development in dogs treated with progestins [3].
Estrogens and progesterones play a major role in normal mammary gland
development, but these hormones have also been implicated in tumor
development [4–7]. Estrogens are promoters of initiated cells in addition to
regulating the transcription of several nuclear proto-oncogenes [8–11].

Mammary gland tumors, both benign and malignant, express estrogen

receptors (ERs) [12,13]. In addition to being involved in the initial malignant
transformation, the ER may also represent a rationale therapeutic target in
canine mammary gland tumors, as in breast cancer in women. A woman’s
lifetime risk of developing breast cancer is influenced by the cumulative dose
of bioavailable estrogen exposure [100]. Endocrine therapy, specifically with

Vet Clin Small Anim 33

(2003) 573–596

E-mail address:

karins@vet.upenn.edu

0195-5616

/03/$ - see front matter

Ó 2003, Elsevier Inc. All rights reserved.

doi:10.1016

/S0195-5616(03)00020-2

background image

anti-estrogens, is one of the most commonly prescribed treatments for
women with ER-positive breast cancer [9,12,14–17]. The situation is similar
in the dog. The duration of exposure to ovarian hormones early in life
determines the overall mammary cancer risk. The risk of developing
mammary gland tumors increases from 0.5% to 8%, and to 26%, depending
on whether the ovariohysterectomy is performed before the first, second, or
any estrus thereafter, respectively [6]. Progestin treatment also increases the
risk of tumor development [3,5]. There is conflicting information in the
literature regarding whether the progesterone-induced tumors are benign,
malignant, or both. Earlier studies reported that dogs treated with
progestins developed mostly benign tumors or had a slightly higher risk
of all types of tumors [5,18–20]. A more recent study found, however, that
91% of dogs with mammary gland tumors that had been treated with
progestins had malignant tumors [3]. Mechanisms involved in pro-
gesterone-induced mammary gland tumors include an upregulation of
growth hormone (GH) production within the mammary gland [21,22]. GH
has direct growth stimulatory effects on mammary tissue but also has
indirect effects via insulin-like growth factor I (IGF-I) [7,21]. The insulin
growth factors play a crucial role both in normal cell proliferation and
malignant transformation; IGF-I has been shown to be involved in
tumorigenesis in many different types of tumors, including mammary
gland tumors [23,24]. Women with low-serum IGF-I have a reduced risk
of developing breast cancer [25,26]. Transgenic mice, which overexpress
IGF-I, have abnormal mammary gland development and an increased
incidence of tumors; IGF-I interferes with apoptosis and is also a potent
mitogen for several breast cancer cell lines [27,28].

In addition to the hormonal influence on the risk for developing

mammary gland tumors, there are also other factors, such as genetic
predisposition and diet, which have been shown to contribute to tumor
development. Certain breeds are at increased risk of developing mammary
gland tumors. The breeds at increased mammary cancer risk vary somewhat
according to study and geographic location, but Toy and Miniature
Poodles, English Springer Spaniels, Brittany Spaniels, Cocker Spaniels,
English Setters, Pointers, German Shepherds, Maltese, Yorkshire Terriers,
and Dachshunds have been reported to have increased incidence of
mammary gland tumors in various studies [29–31]. The fact that certain
breeds have an increased risk of developing mammary gland tumors sug-
gests a genetic component. A common genetic mutation has not been
identified in dogs with mammary gland tumors, however. The tumor
suppression gene p53 is the most frequently mutated gene in human tumors,
and it has also been evaluated in canine mammary gland tumors. Several
studies have evaluated p53 in canine mammary gland tumors and have
reported variable results regarding overexpression and the frequency of
mutations. A recent study reported that 15

/20 mammary gland tumors

overexpressed mutant p53 protein [32]. Other studies have found a lower

574

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

incidence of p53 mutations ranging from 15% to 63% of cases [33–35]. In
addition, the oncogene c-erbB2 has also been found to be overexpressed in
the majority of canine malignant mammary gland tumors evaluated, and
mutations in BCRA1 have also been documented in a few cases [36,37].
Germline mutations in BRCA1 and BRCA2 are thought to account for 5%
to 10% of all breast cancer cases in women, and they may also be involved
in the selected cases of mammary gland tumors in dogs [38]. Many different
genetic alterations are probably involved in mammary gland tumorigenesis,
even though these various mutations may lead to convergent phenotypes
and result in clinically indistinguishable tumors. The importance of studying
these alterations lies in the hope that understanding and identifying the
mechanisms involved in malignant transformation may provide opportuni-
ties for prevention and treatment of cancer. An excellent example is the use
of trastuzumab (Herceptin

ä

) in the treatment of breast cancer in women.

Trastuzumab is a humanized monoclonal antibody that binds to the
extracellular domain of the HER2

/neu (c-erbB2) receptor, which is a member

of the EGFr family of growth factor receptors. Trastuzumab has direct
antitumor efficacy, improves survival in breast cancer patients with HER2

/

neu

overexpression, and is the first successful example of molecularly

targeted therapy in the management of breast cancer in women [39,40].

Several studies have evaluated the effect of various dietary factors and

obesity on the risk of developing mammary gland tumors in dogs. One study
reported that the risk of developing mammary gland tumors is significantly
decreased in spayed dogs that were thin at 9 to 12 months of age, but it did
not find an increased tumor risk in dogs that were obese 1 year before
mammary gland tumor development, or were fed a high-fat diet [41].
Another study by the same group evaluated the effects of several dietary
factors on prognosis in dogs with mammary gland tumors and found that
dogs fed a low-fat, high-protein diet had a significantly prolonged survival
compared with dogs fed a low-fat, low-protein diet. Survival was not
influenced by protein intake in dogs fed a high-fat diet [42]. A more recent
study confirmed that obesity at 1 year of age and a high red-meat diet were
independent risk factors for developing mammary tumors and dysplasia
[43].

Obesity and a high-fat diet have also been linked to an increased breast

cancer risk in women. Obesity is associated with increased risk of breast
cancer in postmenopausal women. This may be caused by various factors
including hyperinsulinemia, increased levels of IGF-I, and decreased levels
of serum sex hormone–binding globulin (SHBG), which results in increased
free serum estrogen levels and, subsequently, an increased breast cancer risk
[44–46]. Immigrants from countries with low breast cancer rates, such as
Asian countries, often increase their breast cancer risk when adopting the
dietary habits of Western countries, which typically consists of high-fat diets
compared with traditional Eastern diets [5,47]. A woman’s risk of
developing breast cancer is influenced by the cumulative dose of estrogen

575

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

exposure, and high-fat diets may contribute to an increased breast cancer
risk by increasing serum levels of bioavailable estrogens [9,10]. Women on
a low-fat diet have significantly lower serum estrogen levels and decreased
breast cancer risk when compared with women on a high-fat diet. Several
independent retrospective studies as well as meta-analyses have confirmed
that there is a direct association between dietary fat intake, serum estrogen
levels, and breast cancer risk [44,48,49].

Tumor types, histopathologic classification, and biologic behavior

Though canine mammary gland tumors may be benign or malignant,

approximately 40% to 50% of these tumors are malignant [50,51]. Further
classification may be performed according to tissue of origin (epithelial,
myoepithelial, or mesenchymal tissue), descriptive morphologic features,
and prognosis. The World Health Organization International Histological
Classification of Mammary Tumors of the dog and the cat combines
histogenic and descriptive morphologic classification, incorporating histo-
logic prognostic features that have been associated with increasing ma-
lignancy [52]. Most mammary gland tumors are of epithelial origin. Some,
however, can have mixed histology consisting of both epithelial and
myoepithelial tissue, with areas of cartilage and bone, and a few tumors are
of purely mesenchymal origin.

Epithelial tumors are often classified according to histopathologic

borders and differentiation. A carcinoma in situ is an epithelial tumor with
malignant features that has not invaded the basement membrane. These
lesions are often multicentric and can grow in pre-existing ducts or lobules.
Tubular carcinomas (adenocarcinoma) are the most common type of mam-
mary gland tumors in the dog; these tumors have retained some of the
original ductal or tubular morphology of the normal mammary gland. The
solid carcinomas are less differentiated; these tumors have lost the tubular

/

ductal structures and form solid sheets. Anaplastic mammary gland car-
cinomas are undifferentiated, pleomorphic, and infiltrative epithelial tumors
that are not classifiable in any of the other categories of carcinomas [52].
Inflammatory mammary gland carcinomas are anaplastic carcinomas with
characteristic clinical and histopathologic features such as involvement of
the overlying skin with edema and pain, extensive inflammatory cell
infiltrate, malignant epithelial cells in the dermal lymphatics, and a rapid
clinical progression [53]. The histopathologic differentiation of epithelial
mammary gland tumors has an impact on prognosis, with a worsening of
prognosis associated with loss of differentiation. Carcinoma in situ and
adenocarcinomas have the best prognosis, and anaplastic and inflammatory
carcinomas have the worst prognosis [52].

Malignant myoeptheliomas, or spindle cell carcinomas, are malignant

tumors arising from the myoepithelial cells of mammary tissue, and they are

576

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

quite rare in dogs. Differentiating between malignant myoepitheliomas and
fibrosarcomas often requires immunohistochemical stains [29].

Primary mammary gland sarcomas are not common and are thought to

arise from pre-existing benign mixed tumors by malignant transformation, or
to arise from the interlobular stroma. The most common primary mammary
gland sarcomas include osteosarcomas and fibrosarcomas. Other mammary
gland sarcomas occasionally encountered are chondrosarcomas and lipo-
sarcomas [29]. The mammary gland is the most common site of extraskeletal
soft tissue osteosarcoma according to a recent study [54]. The mammary gland
osteosarcomas behave as the appendicular osteosarcomas do, and they are
associated with early hematogenous metastasis and a short survival.

Mixed mammary gland tumors consist of both ductal and myoepithelial

cells with areas of cartilage and bone. The origin of the cartilage or bone in
these mixed tumors is controversial and may include metaplastic changes in
the epithelial cells, myoepithelial cells, or interstitial stromal cells. Malignant
mixed tumors, also called carcinosarcomas, are uncommon tumors in the
dog and are composed of both malignant epithelial cells and malignant
connective tissue elements. These tumors are most often a combination
of a carcinoma and an osteosarcoma [29]. The prognosis for dogs with
malignant mixed tumors is poor, and most dogs develop metastasis within
the first year [55].

All malignant mammary gland tumors have the potential to metastasize.

The metastatic risk and pattern are influenced by the tumor type, histologic
differentiation, and several clinical prognostic factors. In general, malignant
epithelial tumors metastasize via the lymphatics to the regional lymph nodes
and the lungs, whereas the mesenchymal tumors typically metastasize by the
hematogenous route directly to the lungs [29]. Dogs with malignant mammary
gland tumors have a significantly shorter survival time than dogs with benign
tumors. The overall 2-year survival has been reported to be between 25% and
40% with a mean survival time ranging from 4 to 17 months, but the survival is
influenced by multiple factors, and it can vary significantly depending on
histologic type and differentiation, stage of disease, and type of treatment [52].
Dogs with small, well-differentiated malignant epithelial tumors may have an
excellent prognosis with surgical resection alone, and dogs with more un-
differentiated, advanced tumors have a guarded prognosis and may require
adjuvant therapy. There are currently no accepted guidelines or recommen-
dations for dogs in the latter group, however, and there are few reports
regarding the effectiveness of adjuvant therapy in such dogs.

Clinical signs and physical exam

Dogs with mammary gland tumors are typically older, approximately 9

to 11 years old, sexually intact, or spayed later in life [5,29,30]. Most dogs
with mammary gland tumors are clinically healthy when they initially
present for evaluation of their tumors. The duration of the clinical signs also

577

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

varies greatly from just a few days to many months. Several studies have
found that dogs with shorter duration of clinical signs have more aggressive
tumors and a worse prognosis than dogs with longer clinical histories
[19,53,56]. The tumor(s) may have been found by the owner or may be an
incidental finding during routine physical exam. Depending on the tumor
type and how soon it is detected, the tumors may be small, large, ulcerated,
fixed, well circumscribed, or involving only one or multiple glands. The
caudal 4th and 5th mammary glands are more commonly involved than the
more cranial glands, but location does not appear to affect prognosis [57,58].
It is not uncommon to find more than one tumor in different glands; more
than 60% of the cases have more than one tumor [55,56]. All of the
individual tumors should be biopsied because they may be of different
histopathologic types [56]. Multiple tumors do not necessary imply a worse
prognosis. Rather, the prognosis is influenced by the size, type, and dif-
ferentiation of the individual tumors [52]. The regional lymph nodes may be
normal or palpably enlarged. Previous reports have found that 10% to
50% of dogs with mammary gland tumors have enlarged lymph nodes [31,52,
53]. Dogs with advanced metastatic disease or inflammatory mam-
mary carcinomas typically have systemic signs of illness when they are
diagnosed. Dogs with metastatic disease may present with nonspecific signs
such as fatigue, lethargy, and weight loss. The severity of these signs depends
on the extent and location of the metastases. Dogs with metastases may or
may not have obvious mammary gland tumors, depending on whether they
have had previous surgical resection of primary tumors. Most mammary
gland tumor metastases occur within 1 year of the initial surgery [6,59].
Dogs with inflammatory mammary gland carcinoma present with more
dramatic clinical signs. Typical clinical signs include extensive inflammation
of the involved mammary glands with edema and pain. These dogs therefore
may be misdiagnosed as having mastitis. They are often in poor clini-
cal condition and have generalized weakness, weight loss, polyuria and
polydipsia, and a high incidence of metastatic disease, both to the regional
lymph nodes and lungs. Prognosis is extremely poor with a mean survival of
25 days from diagnosis [53].

Diagnosis and staging

A surgical biopsy, typically an excisional biopsy, is recommended as the

initial diagnostic approach to dogs with mammary gland tumors. This
biopsy will provide tissue for histopathologic diagnosis and be therapeutic
for dogs with benign tumors. Dogs with small, well-differentiated malignant
tumors may be cured by excisional biopsy if the surgical borders are
complete. Fine needle aspirates may not always accurately differentiate
between malignant and benign epithelial tumors.

Complete staging requires blood work including complete blood count

(CBC), serum chemistry profile, and urinalysis. Evaluation of the primary

578

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

tumor, including size, type, and histologic differentiation; assessment of the
regional lymph nodes; and three-view thoracic radiographs, generally
completes the staging evaluation. The purpose of the staging is to evaluate
general health and to determine the extent of the tumor. Results from
staging assessment provide important prognostic information, which may
affect the owner’s decision to treat. In addition, staging is also necessary for
treatment planning.

Blood work is normal in most dogs with mammary gland tumors, unless

they have other concurrent medical problems or nonspecific age-related
changes. A recent study evaluated hemostatic changes in dogs with ma-
lignant mammary gland tumors and found that two thirds of the dogs
had one or more hemostatic abnormalities, with an increased incidence in
dogs with stage III and IV disease. Of these, dogs with distant metastasis,
invasive or fixed tumors, severe tumor necrosis, and inflammatory car-
cinomas were more likely to have coagulopathies. The clinical sig-
nificance of these abnormalities is not clear, however [60]. Hypercalcemia
of malignancy is a not-infrequent complication of breast cancer in women.
This paraneoplastic manifestation may be caused by osteolytic bone
metastases or the production of PTHrP by the tumor cells. This finding is
rare in dogs with mammary carcinoma, however.

The status of the regional lymph node has a strong impact on survival in

dogs with mammary gland tumors [30,31,57]. Therefore, the regional lymph
nodes should be evaluated in all dogs with malignant tumors, so that systemic
treatment may be initiated in cases with regional lymph node metastasis. The
methods for assessing the regional lymph nodes include palpation, fine needle
aspirates, Tru-cut biopsy

Ò

(Allegiance Healthcare Corporation, McGaw

Park, IL), or whole lymph node excision. Studies in human breast cancer
patients have found that physical exam alone is notoriously inaccurate in
determining axillary lymph node involvement; patients with palpably
enlarged lymph nodes may have reactive inflammatory changes, and
normal-appearing lymph nodes may harbor metastatic disease [61].

A study in veterinary medicine compared the sensitivity and specificity of

these four methods of evaluating lymph nodes in patients with various types
of solid tumors and found similar results. Palpation was inaccurate in
predicting lymph node metastasis, whereas cytology provided an accurate
method of assessing the draining lymph nodes, with a sensitivity of 100%
and a specificity of 96% [62]. This study included dogs with many different
types of tumors, but it seems reasonable to assume that cytology would have
similar accuracy in dogs with mammary gland tumors. Fine needle aspirates
are usually easy to perform, are noninvasive, do not require sedation of the
patient, and provide quick and reliable results. Cytologic evaluation of the re-
gional lymph nodes should therefore be performed as the initial screening in
all dogs with malignant tumors. If the cytology is positive or questionable,
a complete excision of the involved lymph node may be considered. It is
controversial whether removing metastatic lymph nodes in cancer patients

579

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

significantly improves survival, but node resection may improve regional
tumor control and prevent signs associated with progressive lymph node
enlargement [63]. Effective loco-regional control may also translate into
improvements in overall survival; results from human breast cancer studies
show significant improvement in tumor-specific survival in women with
four or more positive axillary lymph nodes treated with both radiation
therapy and chemotherapy [64]. The 1st and 2nd cranial mammary glands
drain to the axillary lymph node on the ipsilateral side, and the 4th and
5th glands drain to their respective superficial inguinal lymph nodes. The
lymphatic drainage of the 3rd mammary gland is most commonly to the
axillary lymph node, but this gland may also drain to the inguinal lymph
node. Both sites should be aspirated, therefore, in dogs with tumors in-
volving the 3rd gland [65].

All dogs with malignant mammary gland tumors should have three-view

thoracic radiographs taken. Radiography is still the standard diagnostic
method for evaluating veterinary patients for metastatic lung disease.
Conventional radiography may detect lung lesions ranging from 6 to 8 mm
in diameter. The ability to detect early metastasis can be improved by using
CT for metastasis as small as 4 mm in diameter. Early detection and
treatment of metastatic disease may have impact on response and outcome
in human patients, and CT has become the standard method of evaluat-
ing human cancer patients for lung metastasis [66]. The occurrence of
synchronous pulmonary metastatic disease at the time of initial diagnosis
was 35% according to an older study; however, a more recent study re-
ported a synchronous metastasis rate of only 6%. [31,59]. Changes in
veterinary medicine, as well as client commitment and awareness about
cancer in pets, may have led to earlier detection and, therefore, diagnosis of
dogs with early-stage disease. Dogs with clinically aggressive tumors should
have thoracic radiographs taken before surgical resection of the primary
tumors because the presence of pulmonary metastasis may influence the
decision to perform an extensive local resection. All dogs with malignant
mammary gland tumors should undergo thoracic imaging because distant
metastases are associated with a grave prognosis. The lungs are the most
common site for distant metastasis in dogs with malignant mammary gland
tumors, but additional diagnostic tests may be indicated, and other
anatomic sites may need to be assessed according to the specific clinical
signs of an individual patient. Metastasis to abdominal viscera or lymph
nodes may occasionally be observed. Abdominal ultrasonography or ra-
diography may be helpful to detect abdominal metastasis.

Mammary gland carcinomas are staged according to the World Health

Organization (WHO) TNM system or a modification of this system [67,68].
The T designation describes the primary tumor, specifically the size of
the tumor, as follows: T0 (no evidence of tumor); T1 (less than 3 cm in
diameter); T2 (tumor between 3 and 5 cm); T3 (tumor larger than 5 cm);
and T4 (inflammatory carcinoma, any size). The N category describes

580

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

involvement of the regional lymph nodes, where N0 is a histologic normal
lymph node, N1 is a positive ipsilateral lymph node, and N2 is bilateral
lymph node involvement. The M category represents distant metastasis; M0
indicates no metastasis, and M1 indicates the presence of distant metastasis.
The TNM system can be further organized as stage grouping (Table 1). Both
the TNM system by itself, or the modified stage grouping system, provide
valuable prognostic information that should be incorporated into the
treatment plan for dogs with mammary gland tumors.

Prognostic factors

Several prognostic factors have been identified in dogs with mammary

gland tumors including age, tumor size and stage, clinical behavior of the
tumor, tumor histopathologic type, tumor grade, estrogen receptor status,
microvessel density, and molecular genetic alterations.

Advanced age at diagnosis is reported to be a negative prognostic factor

in several veterinary studies [6,57]. Age has also been found to be a negative
prognostic factor in women with breast cancer, and older women are
reported to have shorter remissions and survival than younger women
[69,70]. However, age is usually not associated with a more malignant

Table 1
Canine mammary tumor staging modified (excluding inflammatory carcinoma)

T: Primary tumor

T

1

\3 cm maximum diameter

T

2

3–5 cm maximum diameter

T

3

[5 cm maximum diameter

N: Regional lymph node status

N

0

histologic or cytologic no metastasis

N

1

histologic or cytologic metastasis

M: Distant metastasis

M

0

No distant metastasis

M

1

Distant metastasis detected

Stage grouping

Stage
I

T

1

N

0

M

0

II

T

2

N

0

M

0

III

T

3

N

0

M

0

IV

Any T

N

1

M

0

V

Any T

Any N

M

1

Modified from

The World Health Organization (WHO) Clinical Stage Classification (Owen

LN. Classification of tumors in domestic animals, 1st edition. Geneva: World Health
Organization; 1980); and Rutteman GR, Withrow SJ, MacEwen EG. Tumors of the mammary
gland. In SJ Withrow, EG MacEwen, editors. Small animal clinical oncology, 3rd edition.
Philadelphia: WB Saunders, 2001.

581

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

phenotype and may therefore not be a negative prognostic factor in itself.
Rather, advanced age and concerns about toxicity may influence the type of
adjuvant therapy administered to older patients and therefore result in
suboptimal, less effective treatments. This trend is not unique to breast
cancer patients and has been recognized in older cancer patients in general
[71]. Most of the veterinary studies showing that older dogs had a shorter
survival than younger dogs did not perform multi-variant analysis, did not
correlate outcome with tumor stage and treatment, or did not report on
cancer-specific deaths. Older dogs may be more likely to die from other
noncancer causes than younger dogs.

Tumor size has been found to be an independent prognostic factor in

many different studies [30,31,58,72]. Tumors smaller than 3 cm in diameter
are associated with a significantly better prognosis than tumors larger than
3 cm (Fig. 1). Information about tumor size should be readily available,
does not require additional diagnostic testing, and provides important
prognostic information that should be considered when deciding whether
additional therapy is needed.

Stage of disease using the WHO staging system, or a modified version of

this system, provides important prognostic information according to several
different studies. As with many other solid tumors, dogs with mammary
gland tumors have a worsening prognosis with advancing stage [30,31].
Dogs with lymph node metastasis (stage IV disease) have a significantly
shorter survival expectation than dogs with negative node staging, and dogs

Fig. 1. Size as a prognostic factor in canine mammary gland tumors. Kaplan-Meier disease-
free-interval curve according to tumor size in dogs with locally invasive malignant mammary
gland tumors. (From Kurzman ID, Gilbertson SR. Prognostic factors in canine mammary
tumors. Semin Vet Med Surg 1986;1:25–32; with permission.)

582

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

with distant metastasis (stage V disease) have a worse prognosis than dogs
with negative staging or only regional lymph node metastasis (Figs. 2 and 3).

Tumor type, histologic differentiation, and grade have all been found to

have prognostic importance. In general, dogs with mammary gland sarcoma
have a worse prognosis than dogs with malignant epithelial tumors, except
for dogs with inflammatory carcionomas [54,57,58]. Primary mammary
gland sarcomas behave as high-grade sarcomas and are associated with
early hematogenous metastasis and high incidence of local recurrence.
According to the WHO Histological Classification System, the biologic
behavior of the tumor corresponds with the histologic differentiation with
increasing malignancy from noninfiltrating carcinoma (carcinoma in situ) to
complex carcinoma (two cell types), to simple carcinoma (one cell type),
tubulopapillary type, solid type, and to anaplastic carcinoma [52]. The
updated WHO staging system does not subclassify mammary gland car-
cinoma into ductal carcinomas, which arise from the small interlobular
or intralobular ductules, and alveolar carcinomas, which arise from the
alveolar epithelium. A study on lifetime morbidity and mortality of mam-
mary gland tumors in Beagles using a modified CRHL (Collaborative
Radiological Health Laboratory) classification system found, however, that
the ductal carcinomas accounted for the majority of the fatalities in dogs
with malignant mammary gland carcinomas, even though they represented
only 19% of all carcinomas [55]. In addition to the histologic classification
and differentiation of the tumors, there are other pathologic features

Fig. 2. Lymph node status as a prognostic factor in canine mammary gland tumors. Kaplan-
Meier disease-free-interval curve comparing disease-free- interval in dogs with lymph node
metastasis to disease-free interval in dogs without lymph metastasis. (From Kurzman ID,
Gilbertson SR. Prognostic factors in canine mammary tumors. Semin Vet Med Surg 1986;1:25–
32; with permission.)

583

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

including vascular invasion, stromal invasion, absence of an inflammatory
response, the agyrophilic nucleolar organizer regions (AgNOR) index, and
a high histologic grade that may be associated with a guarded prognosis
[31,42,56–58,73–75].

The development of most mammary gland carcinomas is estrogen-

dependent, and the majority of canine mammary gland carcinomas express
ERs [6,8,9]. There is an inverse relationship between ER expression and
histologic differentiation. Benign tumors and well-differentiated tumors are
more likely to be ER-positive, whereas undifferentiated, anaplastic tumors
are more likely to be ER-negative [8,9,76]. The association between
histologic grade and ER expression is further underscored by the fact that
there is an inverse correlation between ER expression and nuclear pro-
liferation indices as measured by proliferating cell nuclear antigen and
Ki-67 analysis. Tumors with high-proliferation indices had a more ag-
gressive clinical behavior with increased risk of metastasis [76] Estrogen
receptor expression has also been found to be associated with the hormonal
status of the dog. Younger, intact dogs were more likely to have ER-positive
tumors than older ovariohysterectomized dogs [8,76]. In addition to pro-
viding prognostic information, ER expression may also predict response
to hormonal therapy. Patients with ER-positive tumors are likely to respond
to estrogen ablation or ER blockage, and the decision whether to use
hormonal therapy in women with breast cancer is influenced by the tumor’s
receptor status. Estrogen receptor expression has traditionally been
evaluated using a standard dextran-coated charcoal biochemical assay but
can also be evaluated using immunohistochemistry on formalin-fixed
tissues. There have been several recent studies successfully identifying ER
using immunohistochemistry for canine mammary gland tumors [77,78].

Fig. 3. WHO stage and prognosis in dogs with malignant mammary gland tumors. Survival
percentage according to TNM classification and stage grouping. (From Yamagami T,
Kobayashi T, Takahashi K. Prognosis for canine malignant mammary tumors based on the
TNM and histologic classification. J Vet Med Sci 1996;58(7):1079–83; with permission.)

584

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

Angiogenesis is crucial for the growth and metastasis of most solid

tumors. Elevated serum levels of various tumor angiogenesis factors and
histopathologic evidence of increased neovascularization in the tumor,
measured as microvessel density, have been reported to have prognostic
importance in human cancer patients with various types of solid tumors [79–
81]. Microvessel density has been reported to be an important independent
prognostic factor in women with node-negative breast cancer [82,83].
Microvessel density has also been found to correlate with malignancy in
canine mammary gland tumors; malignant tumors have increased neo-
vascularization compared with benign tumors. Increased microvessel
density also correlates with risk of local recurrence, presence of lymph
node metastasis, and histologic differentiation in several studies [74,84,85].
In addition to the prognostic importance of the various angiogenesis assays,
results from these studies may also provide a rationale for the use of
antiangiogenic therapy in patients with increased microvessel density.

Molecular alterations, including p53 mutations and overexpression of

HER2

/neu (also called c-erbB2) have been associated with a more malignant

phenotype and a poor prognosis in women with breast cancer. Over-
expression and mutation of p53 have also been evaluated in many different
veterinary studies. Results from these studies are discordant. In a few
studies, there was no significant difference in outcome between dogs with
or without mutations, but other studies reported an increased risk of
recurrence and death in addition to a correlation between p53 mutations and
a high histologic grade of the tumors [34,35]. Overexpression of HER2

/neu

has been associated with resistance to endocrine therapy, shorter time
to relapse, and a low survival rate in women with breast cancer [35,39].
According to one study, this particular alteration is common in the dog, and
most malignant tumors overexpressed this oncogene, but there was no
correlation between expression and local invasion or metastasis [36].

Treatment of canine mammary gland tumors

Spontaneous tumors in dogs and cats are excellent models for human

tumors, and we often advocate using them to evaluate the efficacy of new
and investigational therapies. The practice of clinical oncology in human
medicine is decades ahead of veterinary oncology, however, and we often
look to human oncology for guidance regarding therapy. The modalities
used in the treatment of women with breast cancer include surgery, ra-
diation therapy, hormonal therapy, or chemotherapy. The choice of
modality depends on results from many large prospective studies evaluating
the efficacy of various treatments in patient groups stratified according to
stage and well-established prognostic factors. In general, the treatments
intensify with advancing clinical stage and increasing seriousness of the
prognostic factors. The Consensus Group on the treatment of women with

585

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

breast cancer has provided specific recommendations regarding the use of
radiation therapy, chemotherapy, and hormonal therapy in women with
various stages of disease, which serve as guidelines to practicing oncologists
around the country [17].

This type of consensus recommendation does not exist in veterinary

medicine. Currently, surgery is the only accepted treatment for dogs with
mammary gland tumors, and there are no established guidelines for
treatment beyond surgery. Surgery is the mainstay of treatment for canine
mammary gland tumors and is the single most effective modality for local
tumor control. The type of surgery does not appear to influence survival as
long as the entire tumor is removed with clean histologic margins [86,87].
The recommended surgical approach in dogs with mammary gland tumors
is to perform an excisional biopsy of the tumor; the size of the surgery,
ie, lumpectomy versus simple mastectomy, versus regional mastectomy,
depends on the size of the primary tumor [101]. An excisional biopsy will
provide tissues for histopathologic evaluation and may also provide local
control if the tumor margins are clean. Surgical excision can be curative
in dogs with stage I disease, and those with small, noninvasive, well-
differentiated carcinomas. Dogs with high-grade or larger tumors are likely
to develop metastatic disease, however, and may benefit from additional
therapy [30,31].

Breast sparing surgery, followed by radiation therapy, is often used as an

alternative to radical mastectomy, or in addition to radical mastectomy in
women with large primary tumors or more than four positive axillary lymph
nodes. Studies have found significant improvements in loco-regional control
and tumor-specific survival in women who received postmastectomy ra-
diation [17,64]. Radiation therapy has not been evaluated in treatment of
mammary gland tumors in dogs but may be a reasonable adjuvant to
surgery in dogs with primary mammary gland sarcomas because these
tumors often recur locally as well as at distant sites.

Both endocrine therapy and chemotherapy are used in women with high-

risk breast cancer. The use of endocrine therapy in the treatment of breast
cancer in women is empirical. Oophorectomy was first used successfully to
induce regression of breast tumors 100 years ago [88]. Since that time,
endocrine therapy has evolved to include various types of medical and
surgical hormonal ablation. Today, medical hormonal ablation of various
types is commonly used to treat human breast cancer. The purpose of
hormonal therapy is to prevent further estrogen stimulation of breast cancer
cells. This can be achieved by several different strategies, including blocking
receptors with specific estrogen receptor modulators (SERMS), which are
receptor antagonists such as tamoxifen. Suppression of estrogen synthesis
can be accomplished by administration of aromatase inhibitors, or by use of
luteinizing hormone-releasing hormone (LHRH) agonists. Tamoxifen is
currently the most commonly prescribed treatment for breast cancer in
postmenopausal women and is recommended as the first antiestrogen

586

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

approach by the National Consensus Group [17]. The newer agents, such as
the aromatase inhibitors and the LHRH agonists, are usually second-line
agents in women who have failed on tamoxifen. The decision whether to use
hormonal therapy in women with breast cancer depends on results from ER
analysis because only patients with ER-positive tumors are likely to benefit
significantly from such treatment. The development of mammary gland
tumors in the dog is also estrogen-dependent, and studies have found that
50% to 80% of malignant epithelial tumors express ER receptors [8,9].
These facts suggest that hormonal therapy may also be effective in dogs with
mammary gland carcinomas, as it is in women. Most of the veterinary
literature evaluating the effect of estrogen ablation in the form of OHE
has not found a survival benefit in dogs with mammary gland tumors
[6,7,86,89]. A recent large retrospective study reported, however, that dogs
that underwent OHE concurrent with their tumor removal lived signifi-
cantly longer than dogs that were treated with tumor removal alone. The
timing of the OHE in relation to the tumor surgery was important, and dogs
that were ovariohysterectomized concurrent with or less than 2 years before
their tumor surgery had the greatest benefit from the OHE (Fig. 4) [90]. The
authors hypothesized that the sexual status of the dogs might affect the
tumor receptor status, and dogs that were intact until their tumor surgery

Fig. 4. Survival in canine mammary gland tumors according to spay status and spay time.
Survival in intact dogs, dogs spayed more than 2 years prior to their tumor surgery, and dogs
spayed less than 2 years or concurrently with their tumor surgery. (From Sorenmo KU, Shofer
FS, Goldschmidt MH. Effect of spaying and timing of spaying on survival of dogs with
mammary carcinoma. J Vet Intern Med 2000;14:266–70; with permission.)

587

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

were more likely to have ER-positive tumors and, therefore, to benefit the
most from hormonal modulation. Thus, more than 100 years after the initial
report in the Lancet, the benefit of ovarian hormonal ablation may also have
been confirmed in dogs with mammary gland tumors. The relationship
between the sexual status of the dog and the tumor ER status has been
confirmed by other studies, and intact dogs have been demonstrated to be
more likely to have ER-positive tumors [8,76]. Information about ER status
is not routinely available in veterinary medicine and may therefore not be
available for inclusion in the treatment decisions. Ovariohysterectomy
should still be considered for intact dogs with malignant epithelial tumors.
Most of these tumors are ER-positive according to previous studies, and the
OHE may have other potential health benefits in an older female dog. The
benefit of OHE and tumor removal in dogs with mammary glad tumors
should ideally be evaluated by a prospective randomized clinical trial; The
Veterinary Hospital of the University of Pennsylvania is currently con-
ducting such a study in collaboration with The College of Veterinary
Medicine, University of Helsinki, Finland.

Chemotherapy is offered to all women with breast cancer who are at risk

of failure from distant metastasis. Optimal selection of patients for adjuvant
chemotherapy requires accurate identification of all patients at risk for
failing without including patients who do not need further treatment, thus
avoiding undertreatment as well as overtreatment. Recommendations re-
garding adjuvant treatment of early breast cancer have experienced major
changes in the past 25 years in human medicine. Since the mid-1970s, when
cyclophosphamide, methotrexate, and 5-fluorouracil (CMF) resulted in
statistically significant and clinically meaningful improvements in disease-
free and overall survival, the use of adjuvant chemotherapy has become
common practice in human breast cancer therapy [91,92]. Anthracyclines
such as doxorubicin and epirubicin have long been considered to be among
the most active available agents to treat breast cancer, and they have
become a core component of adjuvant regimens in the past decade.
Adjuvant chemotherapy regimens containing an anthracycline result in
statistically significant improvements in survival compared with regimens
without an anthracycline [17,92,93].

Chemotherapy is also used in dogs with malignant mammary gland

tumors, both in the adjuvant setting as well as in dogs with gross metastatic
disease. There is, however, limited information regarding efficacy of ad-
juvant chemotherapy in dogs with high-risk mammary gland tumors,
and only single case reports showing efficacy of chemotherapy in dogs with
gross metastases [94]. It is encouraging to see objective regression of
pulmonary nodules in a dog with mammary gland tumor metastasis treated
with chemotherapy. Single case reports do not predict response rates in
a larger group of patients, however. Cases with positive responses to
chemotherapy, furthermore, may be more likely to be published than cases
with negative responses, and such reports may therefore lead to a general

588

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

practice of treating patients with high-risk tumors with chemotherapy,
without further evaluating the benefits and risks through randomized
prospective trials. There are no randomized studies on the effect of adjuvant
chemotherapy in dogs with high-risk mammary gland tumors, but a recent
prospective study on 16 dogs with mammary gland carcinomas reported
significant improvement in survival for dogs treated with a combination of
5-fluorouracil and cyclophosphamide compared with dogs treated with
surgery alone (Fig. 5) [95]. Both groups of dogs underwent surgery for the
primary tumor and had stage III

/IV disease with similar tumor character-

istics. Eight of the sixteen dogs in this study were given chemotherapy based
on the owners’ decision, and the other eight dogs were treated with surgery
alone. All 16 of the dogs had high-risk tumors. The median survival was
only 6 months in the group treated with surgery alone compared with
24 months in the group treated with chemotherapy. This difference was
statistically significant and provides encouraging data regarding the use of
adjuvant chemotherapy in dogs with mammary gland tumors. This is a small
study, the dogs were not randomized to treatment groups, and the results
may represent a type II error. Nevertheless, this study provides the basis for
additional larger prospective studies evaluating the benefit of adjuvant
chemotherapy in dogs with negative prognostic factors. The drugs used in
this study were 5-fluorouracil and cyclophosphamide, which were among
the first chemotherapeutic drugs shown to have activity in breast cancer in

Fig. 5. Survival in dogs with high-risk mammary gland tumors treated with surgery alone
versus surgery and postoperative chemotherapy. (From Karayannopoulo M, Kaldrymidou E,
Constantinidis TC, et al. Adjuvant postoperative chemotherapy in bitches with mammary
cancer. J Vet Med Series A 2001;48(2):85–96; with permission.)

589

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

women. Anthracycline-containing protocols are commonly used as first-line
therapy in women with breast cancer today, and the use of anthracyclines,
specifically doxorubicin, may also be reasonable to evaluate further in the
adjuvant setting for canine mammary gland tumors. Doxorubicin was found
to be effective in dogs with pulmonary metastasis, and a preliminary report
showed improved survival in dogs with high-risk mammary gland tumors
receiving adjuvant doxorubicin compared with dogs treated with surgery
alone [96,97]. Doxorubicin has also been found to have antitumor activity in
clonogenic assays, using established cell lines from canine mammary gland
tumors [98]. Primary mammary gland sarcomas are rare, and most studies
report that dogs with such tumors have a poor prognosis. A recent large
retrospective study reported a median survival of 90 days in dogs with
mammary gland osteosarcoma when treated with surgery alone [54].
Another study published around the same time evaluated the effect of
chemotherapy in dogs with soft tissue osteosarcoma, including mammary
gland osteosarcomas, and found a significant difference in survival between
dogs treated with chemotherapy versus the dogs treated with surgery alone.

Table 2
Proposed treatment guidelines for malignant canine mammary gland tumors based on tumor
size, stage, histopathological type, and differentiation

Tumor
size

Tumor
stage

Tumor
type

Histological
differentiation

Treatment
recommendations

\3 cm

Stage 1

Carcinoma

Well differentiated

Complete excision

Tubular

/papillary

OHE if intact

\3 cm

Stage 1

Carcinoma

Undifferentiated

Complete excision
OHE if intact
Chemotherapy or clinical trial

>3 cm

Stages 2–3

Carcinoma

Any

Complete excision
OHE if intact
Chemotherapy or clinical trial

Any size

Stage 4

Carcinoma

Any

Complete excision

(including LN)

OHE if intact
Chemotherapy or clinical trial

Any size

Stage 5

Carcinoma

Any

+

/

ÿ surgery (if needed

for palliation)

Palliative chemotherapy

Any size

Any stage

Carcinoma

Inflammatory

+

/

ÿ surgery

Palliative treatments
Analgesics, anti-inflammatory
Chemotherapy or clinical trial

Any size

Stages 1–3

Sarcomas

Any

Complete (wide) excision

Carcinosarcomas

Radiation therapy if

incomplete excision

Chemotherapy or clinical trial

Abbreviation:

LN, lymph node; OHE, ovariohysterectomy.

590

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

The drugs used in this study were those traditionally used to treat dogs with
appendicular osteosarcoma, such as cisplatin and doxorubicin [99].

Summary

The National Consensus Group recommends that all women with tumors

larger than 1 cm be offered chemotherapy regardless of tumor histology of
lymph node status. This recommendation is to ensure that everyone at risk
for failing, even though the risk may be low in women with relatively small
tumors and favorable histology, has a choice and receives the benefit of
adjuvant chemotherapy. This type of treatment recommendation may also
be made in dogs based on recognized, well-accepted prognostic factors such
as tumor size, stage, type, and histologic differentiation. Based on the
limited clinical information available in veterinary medicine, the drugs that
are effective in human breast cancer, such as cyclophosphamide, 5-fluo-
rouracil, and doxorubicin, may also have a role in the treatment of ma-
lignant mammary gland tumors in dogs. Randomized prospective studies
are needed, however, to evaluate the efficacy of chemotherapy in dogs with
high-risk mammary gland tumors and to determine which drugs and
protocols are the most efficacious. Until such studies are performed, the
treatment of canine mammary gland tumors will be based on the individual
oncologist’s understanding of tumor biology, experience, interpretation of
the available studies, and a little bit of gut-feeling. Table 2 is a proposal
for treatment guidelines for malignant canine mammary gland tumors
according to established prognostic factors, results from published vet-
erinary studies, and current recommendations for breast cancer treatment in
women.

Acknowledgments

The author would like to thank Katherine A. Kruger for her editorial

assistance.

References

[1] Dorn CR, Taylor DON, Schneider R, et al. Survey of animal neoplasms in Alameda and

Contra Costa Counties, California. II. Cancer morbidity in dogs and cats from Alameda
County. J Natl Cancer Inst 1968;40:307–18.

[2] Moe L. Population-based incidence of mammary tumors in some dog breeds. J Reprod

Fertil-Suppl 2001;57:439–43.

[3] Stovring M, Moe L, Glattre E. A population based case-control study of canine

mammary

tumors

and

clinical

use

of

medroxyprogesterone

acetate.

APMIS

1997;105(8):590–6.

[4] Key TJA, Pike MC. The role of oestrogens and prostagens in the epidemiology and

prevention of breast cancer. Eur J Cancer Clin Oncol 1982;24(1):29–43.

[5] Miller BA, Kolonel LN, Bernstein L, et al. Racial

/ethnic patterns of cancer in the United

States 1988–1992. SEER Monograph, National Cancer Institute. Bethesda, MD; 1996.
US Department of Health, Education, and Welfare, Publication #NIH96–4104.

591

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

[6] Schneider R, Dorn CR, Taylor DON. Factors influencing canine mammary cancer

development and postsurgical survival. J Natl Cancer Inst 1969;43(6):1249–61.

[7] Selman PJ, Mol JA, Rutteman GR, et al. Progestin-induced growth hormone excess in the

dog originates in the mammary gland. Endocrinology 1994;134:287–92.

[8] Dubik D, Dembinski TC, Shiu RPC. Stimulation of C-myc oncogene expression

associated with estrogen-induced proliferation of human breast cancer. Cancer Res
1987;47:6517–21.

[9] Henderson BE, Ross R, Bernstein L. Estrogens as a cause of human cancer: the Richard

and Hinda Rosenthal Foundation Award lecture. Cancer Res 1988;48:246–53.

[10] Hulka BS, Edison TL, Lininger RA. Steroid hormones and risk of breast cancer. Cancer

Suppl 1994;74(3):1111–24.

[11] MacMahon B, Cole P, Brown J. Etiology of human breast cancer: a review. J Natl Cancer

Inst 1973;50(1):21–41.

[12] Donney I, Rauis J, Devleeschouwer N, Wouters-Ballman P, et al. Comparison of estrogen

and progesterone receptor expression in normal and tumor mammary tissues from dogs.
Am J Vet Res 1995;56(9):1188–94.

[13] MacEwen EG, Patnaik AK, Harvey HJ, et al. Estrogen receptors in canine mammary

tumors. Cancer Res 1982;42:2255–9.

[14] Fisher B, Constantino J, Redmond C, et al. A randomized trial evaluating tamoxifen in

the treatment of patients with node-negative breast cancer who have estrogen-receptor-
positive tumors. N Engl J Med 1989;320(8):479–84.

[15] Harris JR, Hellman S, Canellos GP, Fisher B. Cancer of the breast. In: DeVita VT,

Hellman S, Rosenberg SA, editors. Cancer principles and practice of oncology, 2nd
edition. Philadelphia: JB Lippincott Company; 1985. p. 1119–79.

[16] Henderson CI, Canellos GP. Medical progress: cancer of the breast, the past decade.

N Engl J Med 1980;302(1):78–90.

[17] National Institutes of Health Consensus Development Panel. National Institutes of

Health Consensus Statement: adjuvant therapy for breast cancer. J Natl Cancer Inst
Monogr 2001;30:5–15.

[18] Briggs MH. Progestins and mammary tumors in the beagle bitch. Res Vet Sci 1980;

281:199–292.

[19] Concannon PW, Spraker TR, Casey HW, et al. Gross and histopathologic effects of

medroxyprogesterone acetate and progesterone on the mammary gland of adult beagle
bitches. Fertil Steril 1981;36:373.

[20] Giles RC, Kwapien RP, Geil RG, et al. Mammary nodules in beagle dogs administered

investigational oral contraceptive steroids. J Natl Cancer Inst 1978;60(6):1351–64.

[21] Mol JA, Lantinga-van Leeuwen IS, van Garderen E, et al. Mammary growth hormone

and tumorigenesis—lessons from the dog. Vet Q 1999;21(4):111–5.

[22] Mol JA, Selman PJ, Sprang EPM. The role of progestins, insulin-like growth factors

(IGF) and IGF-binding proteins in the normal and neoplastic mammary gland of the
bitch: a review. J Reprod Fertil Suppl 1997;51:339–44.

[23] Khandwala HM, McCutcheon IE, Flyvbjerg A, et al. The effects of insulin-like growth

factor on tumorigenesis and neoplastic growth. Endocr Rev 2000;21(3):215–44.

[24] Zumkeller W. IGFs and IGFBPs: surrogate markers for diagnosis and surveillance of

tumor growth? J Clin Pathol: Mol Pathol 2001;54:285–8.

[25] Cohen P, Clemmons DR, Rosenfeld RG. Does the GH-IGF axis play a role in cancer

pathogenesis? Growth Horm IGF Res 2000;10:297–305.

[26] Innes KE, Bryer TE. Preeclampsia and breast cancer risk. Epidemiology 1999;10:722–32.
[27] Hasdell DL, Bonnette SG. IGF and insulin action in the mammary gland: lessons from

transgenic and knockout models. J Mammary Gland Biol Neoplasia 2000;5(1):19–30.

[28] Hasdell DL, Murphy KL, Bonnette SG, et al. Cooperative interaction between mutant

p53 and des(1–3)IGF accelerates mammary tumorigenesis. Oncogene 2000;19(7):
889–98.

592

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

[29] Goldschmidt MH, Shofer FS, Smelstoys JA. Neoplastic lesions of the mammary gland.

In: Mohr U, editor. Pathobiology of the aging dog. Ames, IA: Iowa State University
Press; 2001. p. 168–78.

[30] Kurzman ID, Gilbertson SR. Prognostic factors in canine mammary tumors. Semin Vet

Med Surg 1986;1(1):25–32.

[31] Yamagami T, Kobayashi T, Takahashi K. Prognosis for canine malignant mammary

tumors based on the TNM and histologic classification. J Vet Med Sci 1996;58(7):
1079–83.

[32] Haga S, Nakayama M, Tatsumi K. Overexpression of the p53 gene product in canine

mammary gland tumors. Oncol Rep 2001;8(6):1215–9.

[33] Chu LL, Rutteman GR, Kong JM, et al. Genomic organization of the canine p53 gene

and its mutational status in canine mammary neoplasia. Breast Cancer Res Treat
1998;50(1):11–25.

[34] Fernando SS, Wu X, McKenzie P, et al. Immunohistochemical detection pf p53 protein

expression in mammary carcinoma: a study of 80 cases. Pathology 1995;27(4):365–9.

[35] Wakui S, Muto T, Yokoo K. Prognostic status of p53 mutations in canine mammary

carcinoma. Anticancer Res 2001;21(1B):611–6.

[36] Ahern TE, Bird RC, Bird AE, et al. Expression of the onco-gene c-erbB-2 in canine

mammary cancers and tumor-derived cell lines. Am J Vet Res 1996;57(5):693–6.

[37] Ochiai K, Morimatsu M, Tomizawa N, et al. Cloning and sequencing full length of canine

Brca2 and Rad51 CDNA. J Vet Med Sci 2001;63(10):1103–8.

[38] Martin AM, Weber BL. Genetic and hormonal risk factors in breast cancer. J Natl

Cancer Inst 2000;92(14):1126–35.

[39] Hortobagyi GN. Overview of treatment results with trastuzumab (herceptin) in metastatic

breast cancer. Semin Oncol 2001;28:43–7.

[40] Wang SC, Zhang L, Hortobagyi GN, et al. Targeting HER2: recent developments and

future directions for breast cancer patients. Semin in Oncol 2001;28:21–9.

[41] Sonnenschein EG, Glickman LT, Goldschmidt MH, et al. Body conformation, diet,

and risk of breast cancer in pet dogs: a case-control study. Am J Epidemiol 1991;133(7):
694–703.

[42] Shofer FS, Sonnenschein EG, Goldschmidt MH, et al. Histopathologic and dietary

prognostic factors for canine mammary carcinoma. Breast Cancer Res Treat 1989;13:
49–60.

[43] Perez Alenza D, Rutteman GR, Pena L, et al. Relation between habitual diet and canine

mammary tumors in a case-control study. J Vet Intern Med 1998;12:132–9.

[44] La Guardia M, Giammanco M. Breast cancer and obesity. Panminerva Med

2001;43(2):123–33.

[45] Stoll BA. Adiposity as a risk determinant for postmenopausal breast cancer. Int J Obes

Relat Metab Disord 2000;24(5):527–33.

[46] Tymchuk CN, Tessler SB, Barnard RJ. Changes in sex hormone-binding globulin, insulin,

and serum lipids in postmenopausal women on a low-fat high-fiber diet combined with
exercise. Nutr Cancer 2001;38(2):158–62.

[47] Nagata C, Kawakami N, Shimizu H. Trends in the incidence rate and risk factors for

breast cancer in Japan. Breast Cancer Res Treat 1997;44:75–82.

[48] Hankinson SE, Wilett WC, Manson JE, et al. Plasma sex steroid hormone levels and risk

of breast cancer in postmenopausal women. J Natl Cancer Inst 1998;90:1292–9.

[49] Wu AH, Pike MC, Stram DO. Meta-analysis: dietary fat intake, serum estrogen levels,

and the risk of breast cancer. J Natl Cancer Inst 1999;91:529–34.

[50] Hampe JF, Misdorp W. Tumors and dysplasias of the mammary gland. Bull WHO

1973;50:111–3.

[51] Priester WA, Mantel N. Occurrence of tumors in domestic animals. Data from 12

United States and Canadian colleges in veterinary medicine. J Natl Cancer Inst
1971;43:1333–44.

593

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

[52] Misdorp W, Else RW, Hellmen E, et al. Histological classification of mammary tumors of

the dog and cat. WHO International Histological Classification of Tumors of Domestic
Animals. 2nd Series, vol VII. Washington DC: Armed Forces Institute of Pathology,
American Registry of Pathology; 2001.

[53] Perez Alenza MD, Tabanera E, Pena L. Inflammatory mammary carcinoma in dogs: 33

cases (1995–1999). J Am Vet Med Assoc 2001;219(8):1110–4.

[54] Langenbach A, Anderson M, Dambach DM. Extraskeletal osteosarcoma in dogs:

a retrospective study of 169 cases (1986–1996). J Am Anim Hosp Assoc 1998;34:
113–20.

[55] Benjamin SA, Lee AC, Saunders WJ. Classification and behavior of canine mammary

epithelial neoplasms based on life-span observations in beagles. Vet Pathol 1999;36(5):
423–36.

[56] Fowler EH, Wilson GP, Koester A. Biologic behavior of canine mammary neoplasms

based on a histogenic classification. Vet Pathol 1974;11:212–29.

[57] Hellemen E, Bergstrom R, Holmberg L. Prognostic factors in canine mammary gland

tumors: a multivariate study of 202 consecutive cases. Vet Pathol 1993;30:20–7.

[58] Misdorp W, Hart AAM. Prognostic factors in canine mammary cancer. J Natl Cancer

Inst 1976;56:779–86.

[59] Fidler IJ, Abt DA, Brody RS. The biological behavior of canine mammary neoplasms.

J Am Vet Med Assoc 1967;151(10):1311–8.

[60] Stockhaus C, Kohn B, Rudolph R, et al. Correlation of hemostatic abnormalities with

tumor stage and characteristics in dogs with mammary carcinoma. J Small Anim Pract
1999;40:326–31.

[61] de Freitas R Jr, Costa MV, Schneider SV, et al. Accuracy of ultrasound and clinical

examination in the diagnosis of axillary lymph node metastases in breast cancer. Eur J
Surg Oncol 1991;17:240–4.

[62] Langenbach A, McManus P, Hendrick M, et al. Sensitivity and specificity of methods of

assessing the regional methods for evidence of metastasis in dogs and cats with solid
tumors. J Am Vet Med Assoc 2001;218(9):1424–8.

[63] White RR, Stanley WE, Johnson JL, et al. Long-term survival in 2,505 patients with

melanoma with regional lymph node metastasis. Ann Surg 2002;235(6):879–87.

[64] Whelan TJ, Julian J, Wright J, et al. Does loco-regional radiation therapy improve

survival in breast cancer? A meta-analysis. J Clin Oncol 2000;18:1220–9.

[65] Christensen GC. The mammae. In: Evans HE, Christensen GC, editors. Miller’s anatomy

of the dog. 2nd edition. Philadelphia: WB Saunders; 1979. p. 101–6.

[66] Glasspool RM, Evans TRJ. Clinical imaging of cancer metastasis. Eur J Cancer

2000;36(13):1661–70.

[67] Owens LN. Classification of tumors in domestic animals, 1st edition. Geneva: World

Health Organization; 1980.

[68] Rutteman GR, Withrow SJ, MacEwen EG. Tumors of the mammary gland. In: Withrow

SJ, MacEwen EG, editors. Small animal clinical oncology. 3rd edition. Philadelphia: WB
Saunders; 2001. p. 445–77.

[69] Greenfield S, Blanco DM, Elashoff RM, Ganz PA. Patterns of care related to age of

breast cancer patients. JAMA 1987;257:2766–70.

[70] Winchester DP, Osteen RT, Menck HR. The National Cancer Data Base report on

breast carcinoma characteristics and outcome in relation to age. Cancer 1996;78:
1838–43.

[71] Goodwin JS, Samet JS, Hunt WC. Determinants of survival in older cancer patients.

J Natl Cancer Inst 1996;88(15):1031–8.

[72] Bostock DE. Canine and feline mammary neoplasms. Br Vet J 1986;142:506–15.
[73] Gilbertson SR, Kurzman ID, Zachrau RE, et al. Canine mammary epithelial neoplasms:

biologic implications of morphologic characteristics assessed in 232 dogs. Vet Pathol
1983;20(2):127–42.

594

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

[74] Graham JC, Myers RK. The prognostic significance of angiogenesis in canine mammary

tumors. J Vet Intern Med 2000;14(3):248–9.

[75] Sarli G, Preziosi R, Benazzi C. Prognostic value of histological stage and pro-

liferative activity in canine malignant mammary tumors. J Vet Diagn Invest 2002;
14:25–34.

[76] Nieto A, Pena L, Perez-Alenza MD. Immunohistologic detection of estrogen receptor

alpha in canine mammary tumors: clinical and pathological associations and prognostic
significance. Vet Pathol 2000;37(3):239–47.

[77] Graham JC, O’Keefe DA, Gelberg HB. Immunohistochemical assay for detecting

estrogen receptors in canine mammary tumors. Am J Vet Res 1999;60(5):627–30.

[78] Sobczak-Filipiak M, Malicka E. Estrogen receptors in canine mammary gland tumors.

Polish J of Vet Sciences 2002;5(1):1–5.

[79] Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med 1971;285:

1182–1186.

[80] Folkman J, Klagsburn N. Angiogenic factors. Science 1987;235:444–7.
[81] Folkman J. Clinical implications of research on angiogenesis. N Engl J Med 1995;

3333:1757–63.

[82] Leek RD. The prognostic role of angiogenesis in breast cancer. Anticancer Res

2001;21(6B):4325–31.

[83] Weidner N, Folkman J, Pozza F, et al. Tumor angiogenesis: a new significant and

independent prognostic indicator in early-stage breast carcinoma. J Natl Cancer Inst
1992;84(24):1875–87.

[84] Griffey SM, Verstraete FJ, Kraegel SA, et al. Computer-assisted image analysis of

intratumoral vessel density in mammary tumors from dogs. Am J Vet Res
1998;59(10):1238–42.

[85] Restucci B, De Vico G, Maiolino P. Evaluation of angiogenesis in canine mammary

tumors by quantitative platelet endothelial cell adhesion molecule immunohistochemistry.
Vet Pathol 2000;37(4):297–301.

[86] Allen SW, Mahaffey EA. Canine mammary neoplasia: prognostic indicators and response

to surgical therapy. J Am Anim Hosp Assoc 1989;25:540–6.

[87] MacEwen EG, Harvey HJ, Patnaik AK, et al. Evaluation of the effect of levamizole and

surgery on canine mammary cancer. J Biol Resp Mod 1985;4:418–26.

[88] Beatson GW. On the treatment of inoperable cases of carcinoma of the mamma:

suggestions for a new method of treatment with illustrative cases. Lancet 1896;2:104–7.

[89] Yamagami T, Kobayashi T, Takahashi, et al. Influence of ovariectomy at the time of

mastectomy on the prognosis for canine malignant mammary tumors. J Small Anim Pract
1996;37:462–4.

[90] Sorenmo KU, Shofer FS, Goldschmidt MH. Effect of spaying and timing of spaying on

survival of dogs with mammary carcinoma. J Vet Intern Med 2000;14:266–70.

[91] Bonadonna G, Brusamolino E, Valagussa P, et al. Combination chemotherapy as an

adjuvant treatment in operable breast cancer. N Engl J Med 1976;294:405–10.

[92] Hortobagyi GN. Progress in systemic chemotherapy of primary breast cancer: an

overview. J Natl Cancer Inst Monogr 2001;30:72–9.

[93] Piccart MJ, Lohrisch C, Duchateau L, et al. Taxanes in the adjuvant treatment of breast

cancer: why not yet? J Natl Cancer Inst Monogr 2001;30:88–95.

[94] Hahn KA, Richardson RC, Kapp DW. Canine malignant mammary neoplasia: biological

behavior, diagnosis, and treatment alternatives. J Am Anim Hosp Assoc 1992;28:
251–6.

[95] Karayannopoulo M, Kaldrymidou E, Constantinidis TC, et al. Adjuvant post-operative

chemotherapy in bitches with mammary cancer. J Vet Med Series A 2001;48(2):85–96.

[96] Hershey AE, Kurzman ID, Forrest LJ. Inhalation chemotherapy for macroscopic

primary or metastatic lung tumors: proof of principle using dogs with spontaneously
occurring tumors as a model. Clin Cancer Res 1999;5(9):2653–9.

595

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

[97] Schoenrock SD, Ueberschaer S, Nolte I. Adjuvant chemotherapy with docetaxel and

doxorubicin in canine invasive mammary gland tumors: first results. Presented at the 19th
Annual Veterinary Cancer Society Conference, Wood’s Hole, MA, November 13–16,
1999. Spring Valley (CA): Veterinary Cancer Society; 1999.

[98] Simon DK, Knebel JW, Baumgartner W, et al. In vitro efficacy of chemotherapeutics as

determined by 50% inhibitory concentration in cell cultures of mammary gland tumors
obtained from dogs. Am J Vet Res 2001;62(11):1825–30.

[99] Kuntz CA, Dernell WD, Powers BE, et al. Extraskeletal osteosarcomas in dogs: 14 cases.

J Am Anim Hosp Assoc 1998;34:26–30.

[100] Bernstein L, Ross RK. Endogenous hormones and breast cancer risk. Epidemiol Rev

1993;15:48–65.

[101] Johnston S. Reproductive systems. In: Slatter D, editor. Textbook of small animal

surgery, 2nd edition. (vol 2). Philadelphia: WB Saunders; 1999. p. 2177–200.

596

K. Sorenmo

/ Vet Clin Small Anim 33 (2003) 573–596

background image

Management of transitional

cell carcinoma

Carolyn J. Henry, DVM, MS

a,b,

*

a

Department of Veterinary Clinical Sciences, College of Veterinary Medicine,

University of Missouri-Columbia, 379 East Campus Drive,

Columbia, MO 65211, USA

b

Division of Hematology and Oncology, Department of Medicine, School of Medicine,

University of Missouri-Columbia, Columbia, MO 65211, USA

Primary malignancy of the bladder is rare in dogs, comprising only 0.5%

to 1% of all canine cancers in published reports [1–3]. No large reviews of
canine bladder cancer incidence have been reported within the past decade.
Of the malignancies occurring in the canine bladder, transitional cell car-
cinoma (TCC) is the most common, accounting for approximately 50% to
75% of all reported cases [1–7].

Etiology and risk factors

The etiology of bladder cancer in dogs, as with other malignancies,

is likely multifactorial. Identified risk factors include obesity, exposure
to topical flea and tick insecticides, exposure to marshes that have been
sprayed with mosquito control products, and possibly treatment with
cyclophosphamide [8–10]. Although early surveys indicated no gender pre-
disposition [2,11], subsequent reports document a female predisposition
[1,3,5,9,12,13]. One proposed explanation for the increased risk in females
is that male dogs urinate more frequently as a marking behavior, thus limit-
ing the contact time of bladder mucosa to carcinogens in the urine [5,14].
Further support for this theory, as opposed to a hormone-related cause of
carcinogenesis, came from laboratory studies indicating an ability to induce
bladder cancer in dogs, even when sex hormone levels were altered [15].
Certain breeds, including Shetland sheepdogs, beagles, collies, and various

Vet Clin Small Anim

33 (2003) 597–613

* Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University

of Missouri-Columbia, 379 East Campus Drive, Columbia, MO 65211, USA.

E-mail address:

henryc@missouri.edu

0195-5616

/03/$ - see front matter

Ó 2003, Elsevier Inc. All rights reserved.

doi:10.1016

/S0195-5616(03)00032-9

background image

terriers including Scottish, Airedale, and West Highland white and
Wirehaired fox terriers, appear to be predisposed to development of TCC
[1,5,9,13]. Of these, Scottish terriers and Shetland sheepdogs are the breeds
affected most often [5,9,13].

Clinical presentation

Canine TCC occurs most commonly in older dogs, with a median age of

11 years [2,12,16,17]. The tumor may be seen in young dogs, however.
Therefore, TCC should not be excluded from a differential diagnosis simply
on the basis of patient age.

The bladder trigone is the most common site for TCC in dogs, often

leading to partial or complete urinary tract obstruction. The urethra and
prostate also are infiltrated with neoplastic cells often, either as advanced-
stage tumors infiltrating surrounding tissues or by way of metastatic foci. In
one report of 102 dogs with bladder TCC, 56% had concurrent urethral
involvement, and 29% of the male dogs had prostatic infiltration [9,13].
Some dogs diagnosed with primary prostatic carcinoma may, in fact,
represent misdiagnosed cases of TCC involving the prostatic urethra [18–20].

Dogs with TCC typically present with pollakiuria, stranguria, hematuria,

or tenesmus and may have a history of improvement with antibiotics. Severe
cases may present with bladder rupture, having associated signs including
acute abdominal pain and distension [4]. Occasionally, dogs are presented
for signs related to metastasis, rather than urinary signs. Hypertrophic
osteopathy (HPO) has been reported as a paraneoplastic syndrome in
association with TCC [21]. Although considered rare with TCC, the pre-
senting complaint of lameness and typical osteoproliferative lesions of HPO
on radiographs always warrants a search for underlying neoplastic or pri-
mary thoracic disease.

Physical examination should include abdominal palpation and rectal

examination. The bladder may be distended or have a palpable mass and wall
thickening. Urethral and trigonal thickening may be detectable upon pal-
pation in some cases. Physical exam may reveal no abnormalities, however, as
was noted for 30% of dogs with bladder and urethral tumors in one report [1].

Diagnosis and staging

Urinalysis

Urinalysis (UA) is often the first test used to diagnose bladder TCC. One

must be cautious when obtaining urine for analysis, as TCC transplantation
has been reported in dogs following tumor manipulation with cystocentesis
or at the time of surgery [21–24]. Accordingly, the author cautions against
cystocentesis when TCC is suspected. Urinalysis results for dogs with TCC
may be indistinct from those noted with cystitis, including white blood cells,

598

C.J. Henry

/ Vet Clin Small Anim 33 (2003) 597–613

background image

red blood cells, and bacteria. Although urine sediment exam may reveal
tumor cells in at least 30% of cases [1,25], reactive, nonneoplastic tran-
sitional cells can look similar to TCC cells [26,27]. Cytocentrifugation of
freshly collected urine samples may be helpful in improving accuracy of
cytologic diagnosis. Cytologic evaluation should be supported by other
findings and confirmed with histopathology when possible.

A veterinary bladder tumor antigen test (V-BTA Test; Alidex, Inc.,

Redmond, WA) is available and may serve as a quick screening test for
TCC. The V-BTA is a rapid latex agglutination dipstick test that uses anti-
bodies to a bladder tumor-associated glycoprotein complex detectable in
urine. The V-BTA test requires 0.5 mL of test urine, which should be spun
before testing. Additionally, the test should be run within 48 hours of
sample collection. The author and others evaluated the V-BTA in a pro-
spective, multisite study in which urine samples were collected from healthy
dogs and dogs with TCC, non-TCC urologic disease, and nonurologic
disease. A total of 229 specimens were analyzed, including 48 samples from
dogs with suspected (n

¼ 3) or confirmed (n ¼ 45) TCC. Calculated

sensitivities were 88%, 87%, and 85% for all TCC cases, confirmed TCC
cases, and confirmed bladder TCC cases, respectively. Calculated specific-
ities were 84%, 41%, and 86% using samples from healthy dogs, dogs with
non-TCC urinary tract disease, and dogs with nonurinary disease,
respectively. Hematuria and proteinuria were associated with some false-
positive test results. These causes of false-positive test results have been
reported previously, as has glucosuria [28,29]. Two previous reports have
documented a 90% sensitivity for the bladder tumor antigen test [28,29].
The high sensitivity of the V-BTA test indicates that negative test results
correlate with absence of TCC. Thus, the clinical value of this test lies in its
ability to determine which dogs do not warrant further workup for TCC. As
with any screening test, positive results should prompt further evaluation to
confirm a diagnosis.

Other urine screening tests that have been investigated for human bladder

cancer include the nuclear mitotic apparatus test (NMP22; Matritech,
Newton, MA), the Hemastix hematuria test (Bayer Corporation, Tarry-
town, NY), assays of telomerase, and assays (reverse transcriptase PCR or
immunoassay) of survivin, an inhibitor of apoptosis detected in 100% of
patients with new or recurrent bladder cancer. [30–32] Although these tests
(especially the latter) show great promise in human oncology, they remain
largely unexplored for the detection of canine bladder TCC.

Imaging and confirmatory testing

Definitive diagnosis of TCC should be based on demonstration of

a bladder mass, along with cytological or, preferably, histopathological
demonstration of neoplastic cells from the mass. Demonstration of the mass
may be accomplished with imaging techniques such as radiography and

599

C.J. Henry

/ Vet Clin Small Anim 33 (2003) 597–613

background image

ultrasonography or by direct tumor visualization by way of cystoscopy or
laparotomy. Double-contrast cystography is considered to be a reliable
method for detection of bladder masses in more than 95% of cases [1].
Ultrasonography of the bladder (cystosonography) is performed best when
the bladder is distended with urine or infused saline. Cystosonography offers
the advantages of enhanced detection of intra-abdominal metastasis and the
ability to obtain biopsy samples by way of urinary catheterization under
ultrasound guidance. Visualization of the biopsy site by way of imaging may
improve yield, although one limitation of obtaining biopsy specimens by
catheterization techniques is the small sample size [33]. Alternatively, bi-
opsies may be obtained by cystoscopy or laparotomy.

In addition to verifying a diagnosis of TCC, tissue specimens may be

assigned a tumor grade using a modified World Health Organization (WHO)
system [34]. Tumor grade is based upon growth patterns, cell type, degree of
differentiation, and depth of invasion. In a published review applying this
grading system to tissues from bladder and urethral tumors of 110 dogs,
there were 58 transitional cell carcinomas and 42 tumors classified as TCC
with squamous or glandular metaplasia. Tumors were classified as grade 1, 2,
or 3, with grade 3 tumors being the most anaplastic. The authors concluded
that tumor grade and peritumoral desmoplasia were associated with survival.
A focal peritumoral lymphocytic infiltration was characteristic of transi-
tional cell carcinomas that metastasized. Considerable variability was noted
within single tissue samples, suggesting that obtaining biopsy specimens from
multiple sites is ideal for complete characterization of tumor tissue.

Tumor staging

Complete staging of bladder cancer in dogs requires evaluation of the

primary tumor and assessment for lymph node and distant metastasis. The
WHO clinical staging system (Table 1) uses degree of local tumor invasion
and metastasis to assign a clinical stage [35]. One recent abstract indicated
a correlation between bladder tumor stage and prognosis [36]. Median
survival times were 118 days for dogs with T3 tumors, compared with 218
days for dogs with T1 or T2 tumors. Those with N0 tumors had a median
survival time of 234 days, while those with N1 tumors had a median survival
time of 70 days. Distant metastasis was associated with a median survival
time of 105 days, compared with 203 days for dogs without distant
metastasis at the time of diagnosis. Complete tumor staging may provide
useful information for determining prognosis, developing a treatment plan,
and monitoring response to therapy.

Abdominal ultrasound is useful for staging, not only to assess tumor size

and degree of invasion within the bladder, but also for detecting intra-
abdominal metastasis. The metastatic rate for bladder TCC in dogs has been
reported to exceed 50% at the time of necropsy [1]. Reported metastatic sites
are listed in Table 2.

600

C.J. Henry

/ Vet Clin Small Anim 33 (2003) 597–613

background image

Three-view thoracic radiographs are recommended to detect pulmonary

metastasis. Pulmonary metastatic disease from TCC may be misinterpreted,
unless one is familiar with the radiographic appearance of metastases with
this disease. Several distinct radiographic patterns have been described for
thoracic metastases of TCC and include nodular interstitial opacity, diffuse
unstructured interstitial opacity, cavitating pulmonary lesions, lobar in-
terstitial or alveolar infiltrates, multiple nodules, and normal pulmonary
opacity [37,38]. Hilar lymphadenopathy may be noted also. Dogs with
unstructured interstitial opacity have lesions described as a lacelike haze of
semidense diffuse opacity. This pattern can be easily misinterpreted as age-
related change. In one report, three of eight dogs with pulmonary metastasis
were incorrectly diagnosed as having normal age-related changes on
thoracic radiographs [38]. The cavitating pattern described with TCC is
likely caused by central necrosis of the metastatic lesions. This pattern also
has been noted uncommonly with metastatic transitional cell carcinoma in
people [39].

Treatment options for bladder transitional cell carcinoma

Surgery

The suitability of surgery for treatment of TCC depends upon the tumor

location and invasiveness and the client’s goals. In general, surgery is con-
sidered a palliative procedure, because of the high metastatic rate of canine
TCC, and because even grossly normal tissue may contain neoplastic or
preneoplastic tissue. Surgical options include partial cystectomy, permanent

Table 1
World Health Organization Clinical staging system (TNM) for canine bladder cancer

T: primary tumor

T

is

Carcinoma in situ

T

0

No evidence of primary tumor

T

1

Superficial papillary tumor

T

2

Tumor invading bladder wall, with induration

T

3

Tumor invading neighboring organs (prostate, uterus, vagina, and

pelvic canal)

N: regional lymph nodes (internal and external iliac lymph nodes)

N

0

No regional lymph node involvement

N

1

Regional lymph node involvement

N

2

Regional lymph node and juxta-regional lymph node involvement

M: distant metastases

M

0

No evidence of distant metastasis

M

1

Distant metastasis present

Data from

Owen LN. TNM classification of tumors in domestic animals. 1st edition.

Geneva: World Health Organization; 1980. p. 34.

601

C.J. Henry

/ Vet Clin Small Anim 33 (2003) 597–613

background image

cystostomy catheter placement, or urinary diversion techniques such as total
cystectomy with ureterocolonic anastomosis [40,41]. Often, owners consid-
ering surgery choose the least invasive of these techniques, based on quality-
of-life and convenience issues.

Partial cystectomy

Partial cystectomy may be considered for dogs with localized bladder

tumors in areas amenable to resection with 1-cm to 2-cm margins of grossly
normal tissue. Potential postoperative complications include pollakiuria
caused by reduced bladder capacity and dehiscence of cystectomy suture
lines. Pollakiuria may resolve with time as the bladder size increases post-
surgery. Urinary catheters may be used in the acute postoperative period to
reduce tension on the cystectomy suture line and decrease the likelihood of
wound dehiscence. In one report of partial cystectomy for various urinary

Table 2
Metastatic sites reported for transitional cell carcinoma of the urinary bladder in dogs

Organ system

Site

References

Alimentary

Salivary glands

[2]

Esophagus

[2]

Liver

[2,19,71]

Intestines

[2]

Cardiovascular

Aorta

[2]

Heart

/pericardium

[2,19,82]

Central nervous system

Brain

[2,19]

Spinal cord

[19]

Eye

[82]

Endocrine

Thyroid

[82]

Adrenal

[2,21,82]

Pancreas

[82]

Genitourinary

Uterus

[2,3]

Prostate

[2,19,82]

Urethra

[2,3,19]

Ureter

[2,3,19]

Ovary

[2,19]

Vagina

[2]

Mammary gland

[19]

Musculoskeletal

Rib

[2,21]

Vertebrae

[2,19,71]

Humerus

[2]

Femur

[83]

Skull

[81]

Respiratory

Lungs

[2,3,19,21,71,80,82]

Reticuloendothelial

Sublumbar lymph nodes

[2,3,19,80]

Other lymph nodes

[2,19,71,82]

Spleen

[2,3,71,80]

602

C.J. Henry

/ Vet Clin Small Anim 33 (2003) 597–613

background image

bladder tumors in 11 dogs, survival times ranged from 2 to 48 months and
the 1-year survival rate was 54.5% (6 of 11 dogs) [42]. The dog with the
longest survival had TCC treated with partial cystectomy on two different
occasions 2 years apart and eventually was euthanized elsewhere for a pre-
sumed gastric torsion. One important finding in the study was that visual
assessment at the time of surgery was inaccurate for determining tumor-free
margins. Five dogs thought to have tumor-free margins based on gross
appearance of the tissue at the time of surgery subsequently were found to
have had incomplete excision when the tissue was examined by histopa-
thology. Therefore, if intraoperative evaluation of surgical margins by way
cytology or frozen section is not available, margins should be taken as
generously as possible.

Even with complete tumor excision, TCC recurrence is likely in dogs, as it

is in people following partial cystectomy. A review of the Purdue Compar-
ative Oncology Program Tumor Registry indicated a 106-day median
survival time for 42 dogs treated with surgery alone for localized TCC [43].
Recurrence rates of 30% to nearly 50% have been reported for people
undergoing partial or radical cystectomy, and the recurrence usually occurs
within 1 year [44,45]. This may be caused by microscopic tumors at the
surgical margins, or development of de novo tumors because of field car-
cinogenesis. Because the etiology of bladder tumors is likely to involve
exposure of the bladder mucosa to carcinogenic substances, it has been
hypothesized that de novo tumors develop in the remaining nonexcised
bladder mucosa as a result of the same carcinogenic mechanisms that led to
initial tumor development, rather than a failure of surgical technique [42].

Permanent cystostomy catheter

Urinary outflow obstruction associated with bladder TCC may be

managed by placement of a permanent cystostomy catheter. This palliative
procedure is intended to relieve stranguria and to prevent secondary com-
plications associated with urinary outflow obstruction. Because ureteral
obstruction is not relieved by this procedure, excretory urography is recom-
mended before cystostomy catheter placement to ensure that palliation is
likely. In one report of cystostomy tube placement in seven dogs with known
or suspected TCC, six dogs had resolution of stranguria, and owners
reported satisfaction with the procedure [46]. Median survival time for these
dogs was 106 days. Cystostomy catheter placement predisposes dogs to
urinary tract infections, a complication noted in four of the seven dogs.
Therefore, periodic urinalysis and culture may be needed.

Ureterocolonic anastomosis

Although ureterocolonic anastomosis is dismissed by many clinicians as

being a labor-intensive procedure and impractical for most owners to
manage, it is possible to achieve excellent palliation of urinary symptoms
with this technique [47]. The procedure entails complete bladder resection,

603

C.J. Henry

/ Vet Clin Small Anim 33 (2003) 597–613

background image

followed by end-to-side anastomosis of the ureters to the colonic mucosa.
Postoperative complications may include hyperchloremic acidosis and
related blood gas and electrolyte abnormalities, hydronephrosis, hydro-
ureter, obstruction at the anastomosis site, and pyelonephritis [40]. Owners
must be prepared to administer long-term antibiotics, adhere to recommen-
dations for dietary modifications for their pet including low-salt, low protein
diets with sodium bicarbonate supplementation, allow for frequent blood gas
and electrolyte monitoring, and be able to allow frequent access (every
4 hours) to the outdoors for urination. Despite these requirements, good
quality of life, urinary and fecal continence, and survival times up to 7 months
have been reported for dogs appropriately chosen for this surgery [40,47].

Bladder reconstruction

One relatively unexplored surgical option for TCC of the bladder in dogs

is radical cystectomy followed by bladder reconstruction. The author has
managed one case treated with surgical resection and use of a small in-
testinal submucosa (SIS) graft (Vet BioSISt; Cook Veterinary Products,
Bloomington, IN) to repair the bladder defect. Small intestinal submucosa
is an acellular biodegradable collagen-based material that is derived from
swine small intestines. The material has the ability to regenerate multi-
ple tissues, including ligaments, tendons, abdominal wall, skin, aorta, vena
cava, bladder transitional epithelium, smooth muscle, and peripheral nerves
[48–51]. The dog in the aforementioned case was treated in the adjuvant
setting with chemotherapy and a nonsteroidal anti-inflammatory agent and
had a survival time of 414 days. The ultrasonographic appearance of the
graft changed from hyperechoic to hypoechoic with time and eventually
resembled normal bladder wall thickness. The utility of SIS for regenerative
urinary bladder augmentation has been demonstrated in the preclinical
setting in normal dogs [48–50]. The ability of SIS to sustain or even en-
courage tumor growth at sites of incomplete excision, however, is a potential
risk that must be evaluated before the routine use of this product for
reconstruction of bladders with residual tumor cells after resection.

Medical therapy or chemotherapy for transitional cell carcinoma

Given its high rates of recurrence and metastasis, TCC of the canine

bladder is likely to require systemic chemotherapy from the outset or as
adjuvant therapy after surgery if one is to achieve long-term remissions or
cures. Several drugs and combination protocols have been evaluated for the
treatment of canine TCC.

Doxorubicin in combination therapy

Doxorubicin has shown significant survival benefit when used in

combination protocols to treat human bladder cancer in the neoadjuvant
setting [52]. It has not been widely used in veterinary medicine for this

604

C.J. Henry

/ Vet Clin Small Anim 33 (2003) 597–613

background image

application, however. In one retrospective study of dogs with TCC,
doxorubicin and cyclophosphamide combination therapy was compared
with intravesicular thiotepa or surgery alone [53]. The doxorubicin and
cyclophosphamide combination provided a median survival time of 259
days, compared with 86 days with surgery alone and 57 days with intra-
vesicular thiotepa. Although no other reports have evaluated this protocol
critically for treatment of canine TCC, the overall survival times in the study
compare favorably to those obtained using other protocols. A retrospective
review of 25 dogs with unresectable urinary bladder carcinoma suggested
a survival advantage might exist when dogs receive an anthracycline drug
(doxorubicin or mitoxantrone) in addition to a platinum-based (cisplatin or
carboplatin) chemotherapy protocol, compared with those receiving only
a platinum compound [7]. Although this was not a randomized, prospective
trial, median survival for dogs in the platinum-only group was 132 days,
compared with 358 days for the anthracycline and platinum combination
group. In light of these preliminary results and the favorable responses in
human muscle-invasive bladder cancer to protocols containing methotrex-
ate, vinblastine, doxorubicin, and cisplatin (M-VAC), similar combination
protocols may warrant prospective investigation in dogs with TCC of the
bladder [52].

Piroxicam

The nonsteroidal anti-inflammatory (NSAID) piroxicam has shown

promise for the treatment of TCC, as monotherapy and in combination
protocols. Expression of cyclooxygenase-2 (COX-2) in canine bladder TCC
tissue, but not in normal bladder epithelium, suggests that COX-2 inhibition
by piroxicam may play a key role as a therapeutic adjuvant [54]. A recent
report, however, showed no association between tumor COX-2 expression
and response to piroxicam [55]. In vitro work has not shown evidence for
a direct cytotoxic effect of piroxicam against canine TCC cell lines [56].
Despite the fact that its mechanism of action is not entirely clear, in a pro-
spective clinical trial, piroxicam provided responses in 6 of 34 (17%) dogs
with TCC for a median of 7 months [57]. Reports in the human oncology
literature have documented the chemopreventative and antitumor activity of
COX-2 inhibitors against bladder cancer, colorectal cancer, and other
carcinomas [58–67]. Piroxicam is administered at 0.3 mg

/kg daily by mouth.

As with other NSAIDs, adverse effects may include gastrointestinal
irritation and nephrotoxicity. The prostaglandin analog misoprostol may
be used concurrently to protect against gastric ulceration.

Cisplatin

Despite its use to treat invasive bladder cancer in people and its apparent

cytotoxicity against canine transitional cell carcinoma in vitro [56], cisplatin
has been a disappointing treatment for canine TCC. Response rates have
been less than 25%, with median survival times of 6 months or less

605

C.J. Henry

/ Vet Clin Small Anim 33 (2003) 597–613

background image

[16,17,43,68]. In a trial comparing single-agent cisplatin with the combina-
tion of cisplatin and piroxicam, none of the dogs that received cisplatin
alone experienced remission [43]. The combination resulted in a better
response rate than that seen with cisplatin monotherapy, but renal toxicity
was frequent (12 of 14 dogs, 87%) and dose limiting. Although the 71%
response rate attained with cisplatin and piroxicam therapy is promising,
unless dosage modifications permit less toxicity while maintaining efficacy,
the author does not recommend use of these drugs together in clinical
practice.

Carboplatin

Despite its expense, carboplatin is considered an attractive alternative to

cisplatin for chemotherapy in dogs at risk of renal dysfunction and when the
6-hour diuresis protocol used with cisplatin is impractical. Two initial phase
II clinical trials of carboplatin in dogs with various tumor types reported
remissions in two of three dogs with TCC and 2 of 12 dogs with various
tumors types [69,70]. A prospective trial of 14 dogs with histologically-
proven bladder TCC treated with 300 mg

/m

2

carboplatin every 3 weeks

resulted in no remissions [71]. A combination protocol using carboplatin
and piroxicam was evaluated subsequently and provided five partial re-
missions in 13 dogs [72]. Nephrotoxicity was not noted as a treatment
complication with this protocol. The 38% remission rate was an im-
provement over the lack of remissions reported with carboplatin single-
agent therapy [71]. Researchers noted that the median survival time of the
dogs treated with the combination protocol (93 days) was not better than
that achieved with either carboplatin (132 days) or piroxicam (180 days)
alone [72]. Most of the dogs (10 of 14) that failed carboplatin single-agent
therapy in the initial report went on to receive additional therapy
(piroxicam, mitoxantrone, or actinomycin D). For the dogs that received
additional therapy, median survival time was 204 days, compared with 25
days for those that did not. Accordingly, it is difficult to compare results
beyond the initial response rates between the two studies.

Mitoxantrone

The anthracenedione mitoxantrone has known antitumor activity against

bladder cancer in people and dogs [7,73,74]. Because the toxicity profile of
mitoxantrone does not include nephrotoxicity, one would anticipate that its
use in combination protocols with NSAIDs would be less likely to pre-
cipitate renal disease in dogs than would protocols featuring platinum
compounds. Results of a prospective study evaluating the tolerance and
efficacy of mitoxantrone and piroxicam combination therapy for treatment
of canine bladder TCC have been determined subsequently [12]. Fifty-
five dogs were enrolled in the trial. The protocol included mitoxantrone
(5 mg

/m

2

intravenously every 21 days) for four treatments and piroxicam

(0.3 mg

/kg per day) for the study duration. Tumor staging was performed

606

C.J. Henry

/ Vet Clin Small Anim 33 (2003) 597–613

background image

at baseline, on day 42, and every 3 months after protocol completion.
Endpoints included time to treatment failure (TTF) and survival time (ST).
Data regarding response to therapy were available for 48 dogs and included
one complete response, 16 partial responses, 22 with disease stabilization
and nine with progressive disease, for an overall 35.4% measurable response
rate. Subjective clinical improvement as assessed by owners occurred in 75%
of treated dogs. Gastrointestinal toxicity and azotemia were the most com-
mon treatment complications, occurring in 18% and 10% of treated dogs,
respectively. The median TTF was 194 days, and the median ST was
350 days. Overall, the protocol was tolerated well and induced remission
more frequently than either drug as a single agent. This is the protocol used
by the author for initial therapy of canine TCC.

Radiation therapy

Radiation therapy, both intraoperative and postoperative, has been

described for the treatment of bladder TCC in dogs [1,75,76,84]. In one
report of 16 dogs treated with intraoperative radiation therapy, seven had
TCC of the bladder and were treated with total intraoperative electron doses
of 1000 cGy to 3160 cGy using a linear accelerator with 6 mV electrons [76].
Five of the seven dogs subsequently underwent postoperative radiation.
Survival times ranged from 1 month to 13 months. When ureters were included
in the intraoperative radiation fields, they tended to stenose and become
fibrotic, leading to secondary hydroureter and hydronephrosis. Likewise,
fields encompassing most of the urinary bladder led to bladder fibrosis, non-
distensible bladder tissue by 1 to 2 months, and urinary incontinence. These
results were inferior to an earlier report of intraoperative radiotherapy using
a

137

Cs teletherapy unit for 13 dogs with bladder neoplasia [75]. Although the

report included various bladder tumors, 11 of the 13 were TCC. The median
survival for dogs with TCC was 15 months and their 1-year, 18-month, and 2-
year survival rates were 54%, 27%, and 9%, respectively. A 46% recurrence
rate was noted, as were treatment complications including incontinence (46%
of cases), cystitis (38% of cases), and stranguria (15% of cases). None of the
dogs in the early report received external beam radiation after intraoperative
therapy, but the surgical excisions were more complete than those of the
subsequent report by Withrow et al. In a 1992 review, nine dogs with TCC
were treated with radiation and surgery [1]. Seven of the dogs had TCC of the
bladder; one had bladder and urethral TCC, and one had urethral
involvement only. Total radiation doses ranged from 0 cGy to 3000 cGy
intraoperatively and from 0 cGy to 4800 cGy as subsequent external beam
radiation. When all nine dogs with TCC were evaluated, the overall survival
times ranged from 30 days to 630 days, with a median survival of 105 days. All
dogs died of tumor-related causes. Treatment complications included in-
continence and bladder fibrosis in six dogs. Based on these three reports,
a clear benefit of external beam radiation over intraoperative radiation alone

607

C.J. Henry

/ Vet Clin Small Anim 33 (2003) 597–613

background image

has not been demonstrated. Experimental bladder irradiation in normal dogs
and reported clinical experiences suggest that the total radiation dose
delivered to the canine ureters should be less than 3000 cGy to minimize the
likelihood of ureteral fibrosis [75–77].

Combined radiation and cisplatin chemotherapy has been reported for

two dogs with bladder TCC. Both dogs were treated with Cobalt-60
teletherapy, which was fractionated using 400 cGy per fraction for total
doses of 4400 cGy and 4800 cGy. Intra-arterial cisplatin was administered at
a dose of 50 mg

/m

2

, divided equally into three doses given 6 to 8 hours

before the first three radiation fractions. Cisplatin was administered by the
same dose schedule again with the last three fractions, but was infused
intravenously, rather than by intra-arterial injection. Both dogs had
subjective clinical improvement and documented reduction in tumor size.
Survival times were 6 months and 7 months, respectively [68]. More re-
cently, the combination of mitoxantrone, piroxicam, and coarsely fraction-
ated radiation was evaluated in 10 dogs with TCC of the bladder [78]. The
protocol included six weekly 575 cGy fractions of radiation using a Cobalt-
60 unit, piroxicam (0.3 mg

/kg per day orally), and mitoxantrone (5 mg/m

2

intravenously every 21 days) until evidence of disease progression. Adverse
effects were considered mild and consisted of dermatitis (n

¼ 1), hyper-

pigmentation (n

¼ 1), and mild urinary incontinence (n ¼ 4). No complete

or partial remissions were noted, but seven dogs had disease stabilization
and clinical improvement for 47 days to 320 days (median of 90 days). The
authors concluded that the protocol was tolerated well, and the severe
adverse effects of bladder irradiation described in earlier reports were not
noted for these 10 dogs.

Photodynamic therapy

Photodynamic therapy (PDT) has been employed for treatment of bladder

cancer in people with variable results. Although an effective protocol for PDT
of canine bladder cancer has not been established, investigations are ongoing.
A preclinical evaluation of bladder PDT using the prophotosensitizing agent
5-aminolevulinic acid, which is metabolized to protoporphyrin IX (PpIX)
after oral administration, demonstrated that PpIX fluorescence was confined
to the mucosa and spared the muscularis and serosa [79]. This is considered
a favorable finding, in that it will limit normal bladder tissue toxicity. Further
investigation of this treatment modality is warranted.

Summary

Canine TCC of the bladder is a disease for which early detection and

multimodality therapy are likely to produce the most favorable results.
Urine screening tests are being investigated as tools to permit earlier
detection. The possibility of tumor seeding must be considered when

608

C.J. Henry

/ Vet Clin Small Anim 33 (2003) 597–613

background image

obtaining urine for analysis and when performing surgery. Because these
tumors tend to be very locally invasive at the time of diagnosis and are likely
to metastasize, cures are unlikely. Currently, combination protocols using
chemotherapy and the nonsteroidal anti-inflammatory agent piroxicam
show the most promise in producing tumor responses. Surgery and radi-
ation therapy are useful treatment modalities in select cases. Despite
advances in treatment of canine TCC, median survival times reported for
prospective clinical trials have never exceeded 1 year, regardless of the
treatment modality. Development of accurate tests for early tumor detection
could have a significant impact on the success of treatment of this tumor in
canine patients.

Acknowledgments

The author would like to thank Ms. Allison Critchlow for her assistance

in preparing this manuscript.

References

[1] Norris AM, Laing EJ, Valli VEO, et al. Canine bladder and urethral tumors: a retrospective

study of 115 cases (1980–1985). J Vet Intern Med 1992;6(3):145–53.

[2] Osborne CA, Low DG, Perman V, et al. Neoplasms of the canine and feline urinary

bladder: incidence, etiologic factors, occurrence and pathologic features. Am J Vet Res
1968;29(10):2041–55.

[3] Strafuss AC, Dean MJ. Neoplasms of the urinary bladder. J Am Vet Med Assoc

1975;166(12):1161–3.

[4] Grognet J. Transitional cell carcinoma and subsequent rupture of the canine bladder: a case

report and review of the literature. Can Vet J 1983;24:338–40.

[5] Hayes Jr. HM. Canine bladder cancer: epidemiologic features. Am J Epidemiol

1976;104(6):673–7.

[6] Krawiec DR. Canine bladder tumors: the incidence, diagnosis, therapy, and prognosis.

Veterinary Medicine 1991;86:47–54.

[7] Rocha TA, Mauldin GN, Patnaik AK, et al. Prognostic factors in dogs with urinary

bladder carcinoma. J Vet Intern Med 2000;14(5):486–90.

[8] Glickman LT, Schofer FS, McKee LF, et al: Epidemiologic study of insecticide exposures,

obesity, and risk of bladder cancer in household dogs. J Toxicol Environ Health
1989;28(4):407–14.

[9] Knapp DW, Glickman NW, DeNicola DB, et al. Naturally occurring canine transitional

cell carcinoma of the urinary bladder. A relevant model of human invasive bladder cancer.
Urol Oncol 2000;5:47–59.

[10] Macy DW, Withrow SJ, Hoopes J. Transitional cell carcinoma of the bladder associated

with cyclophosphamide administration. J Am Anim Hosp Assoc 1983;19:965–9.

[11] Pamukcu AM. Tumours of the urinary bladder. Bull World Health Organ 1974;50:43–52.
[12] Henry CJ, McCaw DL, Turnquist SE, et al. Clinical evaluation of mitoxantrone and

piroxicam in a canine model of human invasive urinary bladder carcinoma (a Veterinary
Cooperative Oncology Group Study). Clin Cancer Res 2003;9:906–11.

[13] Knapp DW. Tumors of the urinary system. In: Withrow SJ, MacEwen EG, editors. Small

Animal Clinical Oncology. 3rd edition. Philadelphia: WB Saunders; 2001. p. 490–9.

609

C.J. Henry

/ Vet Clin Small Anim 33 (2003) 597–613

background image

[14] Sprague RH, Anisko JJ. Elimination patterns in the laboratory beagle. Behaviour

1973;47(3):257–67.

[15] Bonser GM. How valuable the dog in the routine testing of suspected carcinogens? J Natl

Cancer Inst 1969;43(1):271–4.

[16] Chun R, Knapp DW, Widmer WR, et al. Cisplatin treatment of transitional cell carcinoma of

the urinary bladder in dogs: 18 cases (1983–1993). J Am Vet Med Assoc 1996;209(9):1588–91.

[17] Moore AS, Cardona A, Shapiro W, et al. Cisplatin (cisdiaminedichloroplatinum) for

treatment of transitional cell carcinoma of the urinary bladder or urethra. A retrospective
study of 15 dogs. J Vet Intern Med 1990;4(3):148–52.

[18] Gorman NT, Calderwood Mays MB, Spencer C. A case of transitional cell carcinoma of

the canine urethra. J Am Anim Hosp Assoc 1984;20:817–20.

[19] Nikula KJ, Benjamin SA, Angleton GM, et al. Transitional cell carcinomas of the urinary

tract in a colony of beagle dogs. Vet Path 1989;26(6):455–61.

[20] O’Shea JD. Studies on the canine prostate gland. II. Prostatic neoplasms. J Comp Pathol

1963;73:244–52.

[21] Brodey RS, Riser WH, Allen H. Hypertrophic pulmonary osteoarthropathy in a dog with

carcinoma of the urinary bladder. J Am Vet Med Assoc 1973;162(6):474–8.

[22] Anderson WI, Dunham BM, King JM, et al. Presumptive subcutaneous surgical

transplantation of a urinary bladder transitional cell carcinoma in a dog. Cornell Vet
1989;79(3):263–6.

[23] Gilson SD, Stone EA. Surgically induced tumor-seeding in eight dogs and two cats. J Am

Vet Med Assoc 1990;196(11):1811–5.

[24] Nyland TG, Wallack ST, Wisner ER. Needle tract implantation following US-guided fine-

needle aspiration biopsy of transitional cell carcinoma of the bladder, urethra, and
prostate. Vet Radiol Ultrasound 2002;43(1):50–3.

[25] Burnie AG, Weaver AD. Urinary bladder neoplasia in the dog: a review of 70 cases.

J Small Anim Pract 1983;24:129–43.

[26] Crow SE. Urinary tract neoplasms in dogs and cats. Compendium on Continuing

Education for the Practicing Veterinarian 1985;7:607–18.

[27] Crow SE, Klausner JS. Management of transitional cell carcinoma of the urinary bladder.

In: Kirk RW, editor. Current veterinary therapy VIII. Philadelphia: WB Saunders; 1983.
p. 1119–21.

[28] Billet JPHG, Moore AH, Holt PE. Evaluation of a bladder tumor antigen test for the

diagnosis of lower urinary tract malignancies in dogs. Am J Vet Res 2002;63(3):370–3.

[29] Borjesson DL, Christopher MM, Ling GV. Detection of canine transitional cell carcinoma

using a bladder tumor antigen urine dipstick test. Vet Clin Pathol 1999;28(1):33–8.

[30] Lokeshwar VB, Soloway MS. Current bladder tumor tests: does their projected utility

fulfill clinical necessity? J Urol 2001;165(4):1067–77.

[31] Sanchez-Carbayo M, Urrutia M, Silva JM, et al. Comparative predictive values of urinary

bladder cancer antigen Cyfra 21–1 and NMP22 for evaluating symptomatic patients at risk
for bladder cancer. J Urol 2001;165(5):1462–7.

[32] Smith SD, Wheeler MA, Plescia J, et al. Urine detection of survivin and diagnosis of

bladder cancer. JAMA 2001;285(3):324–8.

[33] Lamb CR, Trower ND, Gregory SP. Ultrasound-guided catheter biopsy of the lower

urinary tract: technique and results in 12 dogs. J Small Anim Pract 1996;37(9):413–6.

[34] Valli VE, Norris A, Jacobs RM, et al. Pathology of canine bladder and urethral cancer and

correlation with tumour progression and survival. J Comp Path 1995;113:113–30.

[35] Owen LN. TNM classification of tumors in domestic animals. 1st edition. Geneva: World

Health Organization; 1980. p. 34.

[36] Knapp DW, Glickman NW, Bonney PL, et al. Risk and prognostic factors for canine

transitional cell carcinoma of the urinary bladder [abstract]. In: Proceedings of the 19th
Annual Veterinary Cancer Society Conference, Wood’s Hole (MA). Spring Valley (CA):
Veterinary Cancer Society; 1999. p. 9.

610

C.J. Henry

/ Vet Clin Small Anim 33 (2003) 597–613

background image

[37] Mitten RW, Riedesel DH, Flatt RE. Cavitating pulmonary metastases in a dog. J Am Vet

Med Assoc 1978;173(2):203–5.

[38] Walter PA, Haynes JS, Feeney DA, et al. Radiographic appearance of pulmonary

metastases from transitional cell carcinoma of the bladder and urethra of the dog. J Am
Vet Med Assoc 1984;185(4):411–8.

[39] Koh KB, Rogawski K, Smith PH. Cavitating pulmonary metastases from superficial

transitional cell carcinoma of urinary bladder. Case report. Scand J Urol Nephrol 1994;
28(2):201–2.

[40] Stone EA, Withrow SJ, Page RL, et al. Ureterocolonic anastomosis in ten dogs with

transitional cell carcinoma. Vet Surg 1988;17(3):147–53.

[41] Stone EA. Urogenital tumors. Vet Clin North Am Small Anim Pract 1985;15(3):

597–608.

[42] Stone EA, George TF, Gilson SE, et al. Partial cystectomy for urinary bladder neoplasia:

surgical technique and outcome in 11 dogs. J Small Anim Pract 1996;37(10):480–5.

[43] Knapp DW, Glickman NW, Widmer WR, et al. Cisplatin versus cisplatin combined with

piroxicam in a canine model of human invasive urinary bladder cancer. Cancer Chemother
Pharmacol 2000;46(3):221–6.

[44] Cummings KB, Mason JT, Correa RG, et al. Segmental resection in the management of

bladder carcinoma. J Urol 1978;119(3):56–8.

[45] Stein JP, Lieskovsky G, Cote R, et al. Radical cystectomy in the treatment of invasive

bladder cancer: long-term results in 1054 patients. J Clin Oncol 2001;19(3):666–75.

[46] Smith JD, Stone EA, Gilson SD. Placement of a permanent cystostomy catheter to relieve

urine outflow obstruction in dogs with transitional cell carcinoma. J Am Vet Med Assoc
1995;206(4):495–9.

[47] Montgomery RD, Hankes GH. Ureterocolonic anastomosis in a dog with transitional cell

carcinoma of the urinary bladder. J Am Vet Med Assoc 1987;190(11):1427–9.

[48] Cheng EY, Kropp BP. Urologic tissue engineering with small-intestinal submucosa:

potential clinical applications. World J Urol 2000;18(1):26–30.

[49] Kropp BP. Small intestinal submucosa for bladder augmentation: a review of preclinical

studies. World J Urol 1998;16(4):262–7.

[50] Kropp BP, Rippy MK, Badylak SF, et al. Regenerative urinary bladder augmentation

using small intestinal submucosa: urodynamic and histopathologic assessment in long-term
canine bladder augmentations. J Urol 1996;155(6):2098–104.

[51] Pope JC, Davis MM, Smith ER, et al. The ontogeny of canine small intestinal submucosa

regenerated bladder. J Urol 1997;158:1105–10.

[52] Natale R, Grossman H, Blumenstein B, et al. SWOG 8710(INT-0080): randomized phase

II trial of neoadjuvant MVAC plus cystectomy versus cystectomy alone in patients with
locally advanced bladder cancer [abstract]. Proceedings of the American Society of Clinical
Oncology, 2001:202a.

[53] Helfand SC, Hamilton TA, Hungerford LL, et al. Comparison of three treatments for

transitional cell carcinoma of the bladder in the dog. J Am Anim Hosp Assoc 1994;30:
270–5.

[54] Khan KN, Knapp DW, DeNicola DB, et al. Expression of cyclooxygenase-2 in transitional

cell carcinoma of the urinary bladder in dogs. Am J Vet Res 2000;61(5):478–81.

[55] Mutsaers AJ, Mohammed SI, DeNicola DB, et al. Cyclooxygenase-2 expression,

prostaglandin E2 production, and response to piroxicam in canine transitional cell
carcinoma of the urinary bladder [abstract]. In: Proceedings of the Veterinary Cancer
Society 20th Annual Conference, Pacific Grove, CA. Spring Valley (CA): Veterinary
Cancer Society; 2000. p. 57.

[56] Knapp DW, Chan TC, Kuczek T, et al. Evaluation of in vitro cytotoxicity of nonsteroidal

anti-inflammatory drugs against canine tumor cells. Am J Vet Res 1995;56(6):801–5.

[57] Knapp DW, Richardson RC, Chan TCK, et al. Piroxicam therapy in 34 dogs with

transitional cell carcinoma of the urinary bladder. J Vet Intern Med 1994;8(4):273–8.

611

C.J. Henry

/ Vet Clin Small Anim 33 (2003) 597–613

background image

[58] Castelao JE, Yuan JM, Gago-Domingues M, et al. Nonsteroidal anti-inflammatory drugs

and bladder cancer prevention. Br J Cancer 2000;82(7):1364–9.

[59] Dempke W, Rie C, Grothey A, et al. Cyclooxygenase-2: a novel target for cancer

chemotherapy? J Cancer Res Clin Oncol 2001;127(7):411–7.

[60] Farrow DC, Vaughan TL, Hansten PD, et al. Use of aspirin and other nonsteroidal anti-

inflammatory drugs and risk of esophageal and gastric cancer. Cancer Epidemiol
Biomarkers Prev 1998;7(2):97–102.

[61] Gupta RA, DuBois RN. Colorectal cancer prevention and treatment by inhibition of

cyclooxygenase-2. Nat Rev Cancer 2001;1(1):11–21.

[62] Hida T, Kozaki K, Muramatsu H, et al. Cyclooxygenase-2 inhibitor induces apoptosis and

enhances cytotoxicity of various anticancer agents in nonsmall cell lung cancer cell lines.
Clin Cancer Res 2000;6(5):2006–11.

[63] Nishimura G, Yanoma S, Mizuno H, et al. A selective cyclooxygenase-2 inhibitor

suppresses tumor growth in nude mouse xenografted with head and neck squamous
carcinoma cells. Jpn J Cancer Res 1999;90(10):1152–62.

[64] Norrish AE, Jackson RT, McRae CU. Nonsteroidal anti-inflammatory drugs and prostate

cancer progression. Int J Cancer 1998;77(4):511–5.

[65] Rao KVN, Detrisac CJ, Steele VE, et al. Differential activity of aspirin, ketoprofen, and

sulindac as cancer chemopreventative agents in the mouse urinary bladder. Carcinogenesis
1996;17(7):1435–8.

[66] Smalley W, DuBois RN. Colorectal cancer and nonsteroidal anti-inflammatory drugs. Adv

Pharmacol 1997;39:1–20.

[67] Thun MJ, Nambooodiri MM, Heath CW. Aspirin use and reduced risk of fatal colon

cancer. N Engl J Med 1991;325(23):1593–6.

[68] Shapiro W, Kitchell BE, Fossum TW, et al. Cisplatin for the treatment of transitional

cell and squamous cell carcinoma in dogs. J Am Vet Med Assoc 1988;193(12):
1530–3.

[69] Hutson CA, Degen LA, Rackear DG. Preliminary results of carboplatin efficacy in 12

canines and four felines [abstract]. In: Proceedings of the Veterinary Cancer Society 10th
Annual Conference, Auburn, AL. Spring Valley (CA): Veterinary Cancer Society; 1990.
p. 87–8.

[70] Rodriguez CO, Kraegel SA, Peaston AE, et al. A phase II trial of carboplatin in canine

neoplasms [abstract]. In: Proceeding of the Veterinary Cancer Society 12th Annual
Conference, Asilomar, CA. Spring Valley (CA): Veterinary Cancer Society; 1992. p. 59.

[71] Chun R, Knapp DW, Widmer WR, et al. Phase II clinical trial of carboplatin in canine

transitional cell carcinoma of the urinary bladder. J Vet Intern Med 1997;11(5):279–83.

[72] Knapp DW, Schmidt BR, Widmer WR, et al. Preliminary results of carboplatin

/piroxicam

therapy in canine transitional cell carcinoma [abstract]. In: Veterinary Cancer Society–
American College of Veterinary Radiology Combined Conference Proceedings. Chicago;
1997. p. 89.

[73] Ogilvie GK, Obradovich JE, Elmslie RE, et al. Efficacy of mitoxantrone against various

neoplasms in dogs. J Am Vet Med Assoc 1991;198(9):1618–21.

[74] Yaman LS, Yudakul T, Zissis NP, et al. Intravesicular mitoxantrone for superficial bladder

tumors. Anticancer Drugs 1994;5(1):95–8.

[75] Walker M, Breider M. Intraoperative radiotherapy of canine bladder cancer. Vet Radiol

Ultrasound 1987;28:200–4.

[76] Withrow SJ, Gillette EL, Hoopes PJ, et al. Intraoperative irradiation of 16 spontaneously

occurring canine neoplasms. Vet Surg 1989;18(1):7–11.

[77] Sindelar WF, Kinsella T, Tepper J, et al. Experimental and clinical studies with

intraoperative radiotherapy. Surgery. Gynecology & Obstetrics 1983;157(3):205–19.

[78] Poirier VJ, Vail DM, Forrest LJ. Pilot study evaluating palliative radiotherapy (RT) in

combination with mitoxantrone

/piroxicam in the treatment of transitional cell carcinoma

(10 cases) [abstract]. In: Proceedings of the 19th Annual Veterinary Cancer Society

612

C.J. Henry

/ Vet Clin Small Anim 33 (2003) 597–613

background image

Conference, Wood’s Hole, MA. Spring Valley (CA): Veterinary Cancer Society; 1999.
p. 18.

[79] Lucroy MD, Peavy GM, Healey TA. Endogenous photosensitization for photodynamic

therapy of canine transitional cell carcinoma: preclinical evaluation [abstract]. In:
Proceedings of the 21st Annual Conference of the Veterinary Cancer Society, Baton
Rouge, LA. Spring Valley (CA): Veterinary Cancer Society; 2001. p. 54.

[80] Henry CJ, Tyler JW, McEntee MC, et al. Evaluation of the V-BTA urine test for detection

of canine transitional cell carcinoma [abstract 162]. In: Proceedings of the American
College of Veterinary Internal Medicine Annual Forum. Dallas; 2002. p. 801.

[81] McCaw DL, Hogan PM, Shaw DP. Canine urinary bladder transitional cell carcinoma

with skull metastasis and unusual pulmonary metastases. Can Vet J 1988;29:386–8.

[82] Schmidt RE. Transitional cell carcinoma metastatic to the eye of a dog. Vet Pathol

1981;18(6):832–4.

[83] Schwarz PD, Willer RL. Urinary bladder neoplasia in the dog and cat. Probl Vet Med

1989;1(1):128–40.

[84] McCaw DL, Lattimer JC. Radiation and cisplatin for treatment of canine urinary bladder

carcinoma: a report of two case histories. Vet Radiol Ultrasound 1988;29:264–8.

613

C.J. Henry

/ Vet Clin Small Anim 33 (2003) 597–613

background image

Multimodality therapy for head and neck

cancer

Mary K. Klein, DVM, MS

Southwest Veterinary Oncology, 141 East Fort Lowell Road, Tucson, AZ 85705, USA

For the purposes of this article, head and neck tumors include nasal

tumors, oral tumors, and tumors of the salivary glands, thyroid glands,
and ear canals. As a group, these tumors remain a treatment challenge in
both human and veterinary medicine. Generally, surgery is considered the
mainstay of treatment for head and neck tumors. Tumors that cannot be
completely resected and those associated with significant metastatic poten-
tial are considered appropriate candidates for multimodality therapy.
Although there are now years of anecdotal experience in veterinary medicine
to indicate that multimodality approaches to these tumors increase control
rates, there are few studies to date that have accumulated enough cases to
make strong recommendations. The rate of development of distant metas-
tases can be reduced with systemic chemotherapy, but an overall effect
on survival remains to be definitively shown. Those studies that are avail-
able are included in this review. In addition, several decades of informa-
tion accumulated through human clinical trials are summarized briefly.

Nasal tumors

With the possible exception of extremely small well-defined tumors,

surgery alone is not indicated in the treatment of nasal tumors. Most nasal
tumors are carcinomas or soft tissue sarcomas and are treated with radiation
therapy. Radiation therapy alone has increased reported median survival
times from the 3 to 7 months reported for surgery, chemotherapy, or im-
munotherapy [1–5] to 8 to 31 months [6–11]. Significant variation exists
between protocols instituted and degree of staging employed. Elective
treatment of regional lymph nodes does not seem to be indicated, because

Vet Clin Small Anim

33 (2003) 615–628

E-mail address:

mkkl@mindspring.com

0195-5616

/03/$ - see front matter

Ó 2003, Elsevier Inc. All rights reserved.

doi:10.1016/S0195-5616(03)00018-4

background image

regional spread of nasal tumors is unusual. All agree that there is un-
avoidable morbidity associated with the administration of conventional
external beam radiation therapy.

Side effects of radiation therapy are usually separated into acute and late

effects. Acute effects are generally related to inflammatory reactions in the
tissues within the radiation field. The temporary or permanent nature of
these effects is usually related to dose and site. Late effects develop over
several months to years but are irreversible and more likely to affect the
patient’s quality of life. For example, ocular effects that can be expected to
develop in the treatment of nasal tumors include cataracts and keratocon-
junctivitis sicca [12]. Although these side effects are quite acceptable for
most owners as the cost of controlling the tumor, they are discouraging
to deal with in the face of recurrent tumor within months of finishing
treatment. Improvements in survival times might make these side effects
more acceptable to owners, and minimizing side effects while increasing
expected survival duration would induce more owners to treat nasal tumors.

Modern imaging techniques, including CT and MRI, have dramatically

increased the accuracy of radiation therapy treatment planning for both
human and veterinary patients. Before the routine use of CT and MRI for
diagnosis and treatment planning, clinicians grossly underestimated the
extent of disease present in nasal tumor cases. It has been demonstrated that
using external landmarks without the benefit of CT scans for radiation
planning would result in geographic misses in 90% of veterinary patients
[11,13]. Although the information gained from using these imaging
modalities usually increases the amount of tissue included in the treatment
field, in general, the use of computerized treatment planning allows for
a decrease in the amount of normal tissue irradiated. Advanced imaging also
allows for more accurate evaluation of responses and their duration. The
greatest extent of regression seen on CT scans seems to occur 3 to 6 months
after the completion of treatment [14].

For external beam treatments, three-dimensional conformal radiation

and stereotactic radiotherapy are particularly exciting new areas. The
development of multileaf collimators and on-line portal imaging techniques
should make the delivery of three-dimensional radiation therapy more
efficient. Intensity-modulated radiation is also being developed. All these
improvements decrease the amount of normal tissues irradiated and
improve the distribution of dose across tumor tissue. This should decrease
side effects, but it remains to be seen whether these advances in radiation
planning and delivery translate into increased survival times with radiation
used as a single treatment modality. All these improvements have been
instituted in the treatment of most human patients. Unfortunately, only
small increases in tumor control have been noted, without a concomitant
positive impact on survival. Not until multimodality approaches were
instituted in human oncology were increases in survival documented [15].
It is important to note that only by improving the therapeutic ratio of

616

M.K. Klein

/ Vet Clin Small Anim 33 (2003) 615–628

background image

radiation therapy with the technologic improvements listed previously were
radiation oncologists able to decrease morbidity sufficiently that the ad-
dition of other treatment modalities could be considered.

Most canine nasal tumor patients relapse in the mid- to caudal nasal

cavity. Boosting the dose there with conventional external beam radiother-
apy via shrinking fields added to morbidity but not to increased tumor
control in a previous study [16]. Although numbers were small in this study,
it is unlikely, given the human experience, that a significant difference would
have been detected regardless of cohort size. Implementation of improve-
ments in treatment planning should allow for increased dose intensity to the
tumor and safe administration of multimodality approaches, including the
addition of surgery, radioprotectors, radiosensitizers, concurrent chemo-
therapy, immunotherapy, and targeted biologic therapies. The challenge
remains to find a multimodality approach that successfully increases
survival times with acceptable morbidity. The radioprotector amifostine
has been shown to reduce the incidence of acute and chronic side effects in
human head and neck cancer patients treated with radiation and to allow
more patients to complete treatment without interruption [17]. The use of
radioprotectors, such as amifostine, has yet to be fully explored in veterinary
patients. Although surgery performed before megavoltage external beam
radiation therapy does not seem to influence outcome, the impact of surgery
after radiation therapy has not yet been assessed. Brachytherapy techniques
have also been successful in treating human head and neck tumors [15,84].
Because of the noncompliant nature of our patients, these techniques can be
difficult to apply to veterinary head and neck cases. Several brachytherapy
techniques have been developed and successfully applied, however [18,19].

Rationale for chemoradiation therapy

The purpose of administering chemotherapy and radiotherapy together is

to take advantage of the radiosensitizing capability of many active
chemotherapeutic drugs for various tumor types and thereby increase
regional control rates as well as survival. Protracted radiation therapy as
a single modality treatment results in decreased local control rates, pre-
sumably because of accelerated repopulation of tumor cells surviving the
initial treatment [20]. The failure of induction chemotherapy to provide any
survival benefit when compared with surgery or radiation alone in ran-
domized trials may have a similar cause. Significant benefits were not dem-
onstrated until chemotherapy was administered concurrently with the
radiation therapy. Administering cytotoxic drugs concurrent with radiation
has the potential to increase toxicity substantially, however, and necessitates
frequent interruptions in radiotherapy.

Mechanisms behind the synergy of chemoradiation therapy have been

postulated to include interference with sublethal damage repair, tumor cell
cycle synchronization, and prevention of the emergence of radioresistant or

617

M.K. Klein

/ Vet Clin Small Anim 33 (2003) 615–628

background image

drug-resistant cells [21]. Cisplatin, carboplatin, paclitaxel, docetaxel, and
gemcitabine all have radiation-enhancing properties [15,22–24]. Cisplatin is
perhaps the most important chemotherapeutic agent for treating squamous
cell carcinoma of the head and neck in human patients [25] and is also the
only chemotherapeutic drug with significant activity documented against
canine nasal carcinomas [26]. Carboplatin has significantly less toxicity but
also lower response rates [27]. In human patients, fluorouracil (5-FU) has
demonstrated synergy with cisplatin, leading to the establishment of the now
standard human treatment combination regimen of cisplatin plus 5-FU [28].
This combination has not been evaluated in canine patients. The optimal
schedule for radiosensitization has not been determined in human or
veterinary clinical trials. Nevertheless, the greatest survival benefit observed
in most human studies is seen in the patient group receiving concurrent
cisplatin chemoradiation [29,30]. Because these trials have resulted in an
improvement in regional control that is profound enough to affect survival
by 20% to 30% on average, concurrent chemoradiation with cisplatin
is now considered the standard of care in human medicine [15]. Other
treatment modifications, such as altered fractionation with concomitant
boost or hyperfractionation with or without the addition of pre- or post-
radiation chemotherapy, only provided modest increases in regional control.
Interestingly, the longest survival times reported to date have resulted
from treating canine nasal tumors with external beam megavoltage radio-
therapy combined with the use of cisplatin as a slow-release formulation
likely to result in radiosensitizing doses [31]. This study yielded 1-year
survival rates of 81%, and the 2-year survival rate was 39%. These results
have not been duplicated using carboplatin as a radiosensitizer [32]. Con-
current chemotherapy with 5-FU

/cyclophosphamide or mitoxantrone or

preoperative surgery has not affected outcome in previous veterinary series,
but none of these compounds are documented to be good radiosensitizers
[6,7,9,33]. Phase I and II studies are in progress using radiation combined
with gemcitabine chemotherapy [35,36], but response rates are not yet
available.

It is important to note that increased toxicity, especially to mucous

membranes, was also documented in all these multimodality studies. Ag-
gressive support in the form of analgesics, oral care, and, on occasion,
gastrostomy tube placement is required, ideally at a treatment center
familiar with the expected severity of toxicity and potential complications.
Until we make improvements in limiting the morbidity associated with
chemoradiation, the biggest advantage is to patients with excellent per-
formance status and minimal tumor burden. Durable complete responses
and prolonged survival are probably possible in this small subset of patients,
based on the information gathered in human clinical trials [15]. In the
interim, new treatment modalities, such as immunotherapy, gene therapy
and biologically targeted compounds, should continue to be evaluated for
dogs with nasal sinus tumors.

618

M.K. Klein

/ Vet Clin Small Anim 33 (2003) 615–628

background image

Oral tumors

Aggressive surgical techniques are well described in the literature and

allow for complete resection of a significant percentage of oral tumors
[37,85,86]. The most common oral tumors in veterinary medicine include
dental tumors, fibrosarcomas, melanomas, and squamous cell carcinomas.

Dental tumors

Common dental tumors in veterinary patients include epulides,

ameloblastoma, and other odontogenic tumors. Most dental tumors are
well controlled with surgery or radiation therapy even if they achieve fairly
large dimensions. Median survival times of 2 to 3 years are reported for both
surgery and radiation used as single modalities [38–43]. Except in the case of
extremely large tumors, single modality therapy is usually adequate.
Combination therapy should be reserved for large ameloblastomas that
are incompletely resected. These tumors should also be treated with radi-
ation therapy to minimize risk of recurrence.

Fibrosarcomas

Because of the low metastatic potential of oral fibrosarcomas, most are

best treated by surgery wherever possible. Unresectable or incompletely
resected fibrosarcomas require the application of multiple treatment mo-
dalities. Unfortunately, this situation occurs in a significant percentage of
oral fibrosarcoma cases.

In a summary of various papers on mandibulectomy or maxillectomy,

fibrosarcomas were found to recur in greater than half of the cases, with
median survival times of approximately 11 months and 35% of the patients
alive at 1 year [37]. The administration of radiation therapy to those patients
with microscopic disease after resection seems to improve outcome, pro-
viding a median survival time of 540 days [44]. This is in contrast to
radiation alone, where control rates without surgical cytoreduction are
approximately 50% at 1 year [45]. Oral fibrosarcomas had a statistically
significant lower median survival time (540 days) when compared with
fibrosarcomas located in other tumor sites (2270 days), indicating the
difficulty of effectively treating large portions of the oral cavity with high
doses of radiation therapy while avoiding unacceptable normal tissue
complications [44].

Radiation is usually delivered after surgery to dogs with oral fibro-

sarcoma. There is evidence of a dose response. The human literature
indicates that patients who began radiation more than 6 weeks after surgery
and whose total therapy time extended beyond 12 to 13 weeks have worse
outcomes [15].

619

M.K. Klein

/ Vet Clin Small Anim 33 (2003) 615–628

background image

Melanomas

Two important prognostic factors have been identified repeatedly in

studies of canine malignant melanoma: the size of the primary tumor
and the ability of the first treatment intervention to control the disease
effectively. Thus, any melanoma with a diameter larger than 2 cm is
considered to have significant metastatic potential, and recurrence after
surgery is inevitably associated with aggressive behavior. These findings
should be interpreted as a caveat to treat aggressively the first time. Patients
with small lesions (< 2 cm in diameter) that are completely resected have
a median survival time of 511 days in comparison to median survival times
of only 164 days for those patients with a tumor greater than 2 cm in diam-
eter or positive lymph node status [46,47].

Melanomas are also responsive to coarsely fractionated radiation therapy

[48–51]. Complete response rates ranging from 53% to 69% have been
reported, with overall median survival times ranging from 5 to 9.5 months.
Again, the size of the primary tumor is found to influence the survival time.
Dogs with less than stage II disease (primary tumor <2 cm in diameter) have
reported survival times ranging from 14.9 to 20 months in comparison to 5 to
6 months for cases with higher stage disease. Distant disease is the usual cause
of death in malignant melanoma, particularly when large tumors or early
metastasis is present. This highlights the need for multimodality treatment in
melanoma. Early studies with hyperthermia added to radiation therapy
demonstrated extremely high response rates; however, these response rates did
not translate into increased survival times, and hyperthermia is not routinely
available [52,53]. The addition of radiation therapy to surgery for malignant
melanomas is unlikely to increase overall survival times, because distant
disease is the most common cause of death in these cases [49,50].

Based on early studies indicating objective responses in measurable

melanomas, carboplatin has been added to the treatment regimen of ma-
lignant melanomas at many institutions across the country [54]. Results
from those studies are just beginning to be presented and must be cautiously
interpreted, because the data are not yet mature and there is substantial
variability in the protocols employed. One early study indicates that survival
time increased for all stages of malignant melanoma when chemotherapy
was added to the treatment regimen but only achieved statistical significance
for dogs with stage III disease [50]. Large multi-institutional trials are re-
quired to elucidate fully any benefit gained through the addition of chemo-
therapy to the treatment of canine oral melanoma.

Immunotherapy also shows promise in the treatment of oral melanomas. A

randomized study of 98 dogs treated surgically or by surgery in con-
junction with liposome muramyl tripeptide immunotherapy (L-MTP) showed
that those dogs with tumors less than 2 cm in diameter and lymph node
positivity had an 80% survival rate 2 years later in comparison to only
25% in the surgery alone arm. Unfortunately, L-MTP did not positively

620

M.K. Klein

/ Vet Clin Small Anim 33 (2003) 615–628

background image

alter survival durations in those cases with larger tumors or metastatic
disease [55]. In vivo transfections of established tumors with immunostim-
ulatory genes can elicit antitumor activity and have been demonstrated
to induce complete and sustained local regression of large tumor burdens
in some canine melanoma patients [56]. Studies are currently underway to
combine immunotherapy with surgical resection. Further indication of the
role of immunotherapy in the manipulation of oral melanomas was pro-
vided by a phase I trial of human tyrosinase DNA vaccination [57]. As
these studies mature, adjuvant immunotherapy may replace the use of che-
motherapy in these tumors.

Osteosarcomas

Oral osteosarcoma is another tumor that challenges both local control

and the control of distant disease. With the possible exception of man-
dibular osteosarcoma, where surgery alone may prove curative [58], oral
osteosarcomas require both surgery to control the primary site and chemo-
therapy to address distant disease. In those animals in which complete
resection is not possible, it is logical to add definitive radiation therapy
to increase control rates. There are limited published data available to
provide information on the impact of adding adjunctive radiation on
local control rates or survival times other than evidence of activity in
vertebral tumors, however [59]. As for dogs with melanoma, the survival of
most of these animals is limited by the development of distant disease and
would not be expected to improve unless systemic therapy is combined with
effective local control.

Canine oral squamous cell carcinomas

The prognosis for these tumors seems to be quite site specific, with

tumors in the rostral oral cavity curable by surgery [37,60] or radiation
therapy alone [61]. Those of the caudal oral cavity, including the tonsil and
base of the tongue, are highly metastatic, and a multimodality approach is
indicated. A radiation dose response has been documented for these tumors,
with 1-year control rates of 46% in those cases receiving greater than 40 Gy
[61]. Median disease-free intervals of approximately 1 year are recorded
in response to radiation alone, with doses ranging from 38.5 to 57 Gy in
a variety of schema. Negative prognostic factors for survival include ad-
vanced age of the patient, caudal oral location, larger radiation portal size
requirement, and recurrent disease [62,63]. The addition of hyperthermia
increases control rates significantly, but this modality is not routinely
available [53,64]. The addition of chemotherapy to treatment regimens for
canine oral squamous cell carcinoma of the caudal mandible or maxilla,
whether as an induction agent or concurrent radiosensitizer or in the

621

M.K. Klein

/ Vet Clin Small Anim 33 (2003) 615–628

background image

adjuvant setting, awaits further study. Tonsillar squamous cell carcinomas
seem to be favorably affected by a multimodality approach. In a small
series of eight cases treated with radiation alone, median survival times of
110 days were reported [65]. Local recurrence was noted in only two of
the eight cases, but distant disease developed in five of the eight cases.
When surgery, radiation therapy, and chemotherapy were applied to a
similar patient population, the median survival time was increased to 270
days [66].

Feline oral squamous cell carcinomas have poor outcomes and remain

a therapeutic challenge. Patients left untreated or treated with any single
treatment modality have survival expectations of less than 3 months [67].
In those few cats with mandibular tumors that are amenable to surgical
resection followed by radiation, a median disease-free interval of 11 months
has been reported. Most of these cats suffered local recurrence [68]. The use
of etanidazole as a radiosensitizer resulted in a median survival time of 116
days [69]. Clearly, treatment of feline oral squamous cell carcinoma is an
area warranting further investigation.

Salivary gland tumors

Primary salivary gland neoplasias are rare in the cat and dog. Many

patients present with extracapsular extension of tumor, and the numerous
vital structures in close proximity to the salivary glands make aggressive
surgical removal difficult. Incomplete removal invariably results in local
recurrence [70]. The addition of radiation therapy to surgical excision seems
to increase control rates and survival times [71,72]. Median survival times of
550 days for dogs and 516 days for cats have been reported with the addition
of radiation therapy. Radiation significantly affected outcome in these cases,
but the role of chemotherapy remains to be defined. Over half of the cats
presented with more advanced stages of primary tumor had metastatic
disease at the time of diagnosis [72].

Ear canal tumors

Many ceruminous gland adenocarcinomas and squamous cell carcinomas

of the ear are amenable to surgical resection via total ear canal ablation with
or without bulla osteotomy [73–75]. Most dogs live longer than 2 years when
treated with surgery alone, and most cats live longer than 1 year when
treated with surgery alone. In animals with incomplete tumor resection, the
addition of adjuvant radiation therapy seems to be of potential benefit [76].
Because the metastatic potential of aural tumors is generally low, ranging
from 5% to 15%, chemotherapy is unlikely to have a major role in the
treatment of these cases [73].

622

M.K. Klein

/ Vet Clin Small Anim 33 (2003) 615–628

background image

Thyroid tumors

Eighty to ninety percent of canine thyroid tumors can be expected to be

malignant. Although smaller and freely moveable tumors are amenable to
long-term control by surgery alone [77], larger nonresectable tumors have
been shown to be responsive to external beam radiation therapy [78].
Progression-free survival rates were 80% at 1 year and 72% at 3 years in one
radiation study. It often took many months for responses to be evident.
Twenty-eight percent of these dogs developed distant disease. Dogs with
bilateral disease were found to be at increased risk for metastatic disease
[78]. Responses to doxorubicin and cisplatin chemotherapy have been doc-
umented in canine thyroid carcinoma cases [79,80]. Thus, chemotherapy
may also contribute to the multimodality treatment of canine thyroid
carcinomas in combination with radiation therapy or surgery for those
animals at increased risk of developing metastatic disease [81]. Multi-
modality approaches have been shown to benefit human anaplastic thyroid
carcinoma patients [34].

Biologically targeted therapies

The molecular and cellular pathways involved in the unregulated cell

growth that leads to head and neck tumors are complex. As clinical
researchers learn more, we can expect the development of biologically
targeted therapies. Three targeted therapies in human head and neck tumors
show early promising results. These include treatment with epidermal
growth factor receptor (EGFr) antagonists and cyclin-dependent kinase
(cdk) inhibitors and the administration of replication competent adeno-
viruses. The EGFr is a transmembrane glycoprotein that is a member of
the tyrosine kinase growth factor receptor family. Activation of this proto-
oncogene results in overexpression of the receptor and has been dem-
onstrated to occur in more than 90% of human squamous cell head
and neck tumors. Several monoclonal antibodies directed against epitopes
on the EGFr are in clinical development, including the chimeric antibody
C225 [82]. Enhanced toxicity is noted when this chimeric antibody is com-
bined with a number of chemotherapy agents, including cisplatin, as well
as when it is combined with radiotherapy. Phase I clinical trials are under-
way in human squamous cell carcinoma patients [83].

Summary

The refinement of radiation therapy techniques should result in a decrease

in morbidity in canine and feline nasal carcinoma patients and should
further allow for the addition of adjuvant therapies. Patients with large oral
tumors that are incompletely excised should have radiation therapy added
to their treatment regimen. Tumors with significant metastatic potential,

623

M.K. Klein

/ Vet Clin Small Anim 33 (2003) 615–628

background image

such as melanoma, should be considered for addition of chemotherapy.
Carboplatin has activity in melanomas and is being added at several
institutions, but trial results are not yet available. Chemoradiation has
become the treatment of choice for human head and neck squamous cell car-
cinomas but remains largely unexplored in veterinary medicine. Hopefully,
development of chemoradiation will benefit feline squamous cell car-
cinoma patients, because current treatment regimens are largely ineffective.
Immunotherapy agents and targeted biologic therapeutics seem to hold
promise for the future.

References

[1] Holmberg DL, Fries C, Cockshutt J, et al. Ventral rhinotomy in the dog and cat. Vet Surg

1989;18:446–9.

[2] Laing EJ, Binnington AG. Surgical therapy of canine nasal tumors. A retrospective study

(1982–1986). Canine Vet 1988;29:809–13.

[3] MacEwen EG, Withrow SJ, Patnaik AK. Nasal tumors in the dog: retrospective evaluation

of diagnosis, prognosis and treatment. JAVMA 1977;170:45–8.

[4] Madewell BR, Priester WA, Gillette EL, et al. Neoplasms of the nasal passages and

paranasal sinuses in domestic animals as reported by 13 veterinary colleges. Am J Vet Res
1976;37:851–6.

[5] Norris AM. Intranasal neoplasms in the dog. J Am Anim Hosp Assoc 1979;15:231–6.
[6] Adams WM, Withrow SJ, Walshaw R, et al. Radiotherapy of malignant nasal tumors in 67

dogs. JAVMA 1987;191:311–5.

[7] Adams WM, Miller PE, Vail DM, et al. An accelerated technique for irradiation of malig-

nant canine nasal and paranasal sinus tumors. Vet Radiol Ultrasound 1998;39:475–81.

[8] Evans SM, Goldschmidt M, McKee LF, et al. Prognostic factors and survival after

radiotherapy for intranasal neoplasm in dogs: 70 cases (1974–1985). JAVMA 1989;194:
1460–63.

[9] McEntee MC, Page RL, Heidner GL, et al. A retrospective study of 27 dogs with intranasal

neoplasms treated with cobalt radiation. Vet Radiol Ultrasound 1991;32:135–9.

[10] Theon AP, Madewell BR, Harb MF, et al. Megavoltage irradiation of neoplasms of the

nasal and paranasal cavities in 77 dogs. JAVMA 1993;202:1469–75.

[11] Thrall DE, Robertson ID, McLeod DA, et al. A comparison of radiographic and

computed tomographic findings in 31 dogs with malignant nasal cavity tumors. Vet Radiol
Ultrasound 1989;30:59–66.

[12] Roberts SM, Lavach JD, Severin GA, et al. Ophthalmic complications following

megavoltage irradiation of the nasal and paranasal cavities in dogs. JAVMA 1987;
100:43–7.

[13] Park RD, Beck ER, LeCouteur RA. Comparison of computed tomography and

radiography for detecting changes induced by malignant nasal neoplasia in dogs. JAVMA
1992;201:1720–4.

[14] Thrall DE, Heidner GL, Novotney CA, et al. Failure patterns following cobalt irradiation

in dogs with nasal carcinoma. Vet Radiol Ultrasound 1993;34:295–300.

[15] Schantz SP, Harrison LB, Forastiere AA. Cancer of the head and neck: tumors of the nasal

cavity and paranasal sinuses, nasopharynx, oral cavity, and oropharynx. In: DeVita VT Jr,
Hellman S, Rosenberg SA, editors. Cancer: principles and practice of oncology. 6th
edition. Philadelphia: Lippincott Williams & Wilkins; 2001. p. 797–860.

[16] LaDue TA, Dodge R, Page RL, et al. Factors influencing survival after radiotherapy of

nasal tumors in 130 dogs. Vet Radiol Ultrasound 1999;40:312–7.

624

M.K. Klein

/ Vet Clin Small Anim 33 (2003) 615–628

background image

[17] Antonadou D, Pepelassi M, Synodinou M, et al. Prophylactic use of amifostine to prevent

radiochemotherapy-induced mucositis and xerostomia in head-and-neck cancer. Int J
Radiat Oncol Biol Phys 2002;52:739–47.

[18] Thompson JP, Ackerman N, Bellah JR, et al.

192

Iridium brachytherapy, using an

intracavitary afterload device, for treatment of intranasal neoplasms in dogs. Am J Vet Res
1992;53:617–22.

[19] White R, Walker M, Legendre AM, et al. Development of brachytherapy technique for

nasal tumors in dogs. Am J Vet Res 1990;51:1250–6.

[20] Pajak TF, Laramore GE, Marcial VA, et al. Elapsed treatment days—a critical item for

radiotherapy quality control review in head and neck trials: RTOG report. Int J Radiat
Oncol Biol Phys 1991;20:13–20.

[21] Fu KK, Phillips TL. Biologic rationale of combined radiotherapy and chemotherapy.

Hematol Oncol Clin North Am 1991;5:737–51.

[22] Choy H, Rodriguez FF, Loester S, et al. Investigation of Taxol as a potential radiation

sensitizer. Cancer 1993;71(Suppl 11):3774–8.

[23] Douple EB, Richmond RC, O’Hara JA, et al. Carboplatin as a potentiator or radiation

therapy. Cancer Treat Rev 1985;12(Suppl A):111–24.

[24] Mason KA, Milas L, Hunter NR, et al. Maximizing therapeutic gain with gemcitabine and

fractionated radiation. Int J Radiat Oncol Biol Phys 1999;44:1125–35.

[25] Havlin KA, Huhn JG, Myers JW, et al. High-dose cisplatin for locally advanced or

metastatic head and neck cancer: a phase II pilot study. Cancer 1989;63:423–7.

[26] Hahn KA, Knapp DW, Richardson RC, et al. Clinical response of nasal adenocarcinoma

to cisplatin chemotherapy in 11 dogs. JAVMA 1992;200:355–7.

[27] Al-Sarraf M. Management strategies in head and neck cancer: the role of carboplatin. In:

Bunn PA, Jr, Canetta R, Ozols PF, et al, editors. Carboplatin: current perspectives and
future directions. Philadelphia: WB Saunders; 1990. p. 221–31.

[28] Browman GP, Cronin L. Standard chemotherapy in squamous cell head and neck cancer:

what we have learned from randomized trials. Semin Oncol 1994;21:311–9.

[29] Al-Sarraf M, LeBlanc M, Shanker Giri PG, et al. Chemoradiotherapy versus radiotherapy

in patients with advanced nasopharyngeal cancer. Phase III Randomized Intergroup Study
0099. J Clin Oncol 1998;16:1310–7.

[30] Cooper JS, Lee H, Torrey M, et al. Improved outcome secondary to concurrent

chemoradiotherapy for advanced carcinoma of the nasopharynx. Preliminary corrobora-
tion of the Intergroup experience. Int J Radiat Oncol Biol Phys 2000;47:861–6.

[31] Lana SE, Dernell WS, LaRue SM, et al. Slow release cisplatin combined with radiation for

the treatment of canine nasal tumors. Vet Radiol Ultrasound 1997;38:474–8.

[32] Mauldin GN, Meleo KA. Combination carboplatin and radiotherapy for nasal tumors

in dogs [abstract]. In: Proceedings of the 14th Annual Conference of the Veterinary
Cancer Society. Spring Valley (CA): Veterinary Cancer Society; 1994. p. 129.

[33] Henry CJ, Brewer WG, Tyler JW, et al. Survival in dogs with nasal adenocarcinoma: 64

cases (1981–1995). J Vet Intern Med 1998;12:436–9.

[34] Tennvall J, Lundell G, Wahlberg P, et al. Anaplastic thyroid carcinoma: three protocols

combining doxorubicin, hyperfractionated radiotherapy and surgery. Br J Cancer 2002;
86:1848–53.

[35] Jones PD, Kitchell BE, Losonsky JM. Gemcitabine as a radiosensitizer for non-resectable

feline oral squamous cell carcinoma [abstract]. In: Proceedings of the 21st Annual
Conference of the Veterinary Cancer Society, Baton Rouge. Spring Valley (CA):
Veterinary Cancer Society; 2001. p. 36.

[36] LaDue TA. In: Proceedings of the American College of Veterinary Radiology, Chicago;

2002 [abstract].

[37] Withrow SJ. Cancer of the gastrointestinal tract A. Cancer of the oral cavity. In: Withrow

SJ, MacEwen EG, editors. Small animal clinical oncology. 3rd edition. Philadelphia: WB
Saunders; 2001. p. 305–18.

625

M.K. Klein

/ Vet Clin Small Anim 33 (2003) 615–628

background image

[38] Bradley RL, MacEwen EG, Loar AS. Mandibular resection for removal of oral tumors in

30 dogs and 6 cats. JAVMA 1984;184:460–3.

[39] Langham RF, Mostosky UV, Schirmer RG. X-ray therapy of selected odontogenic

neoplasms in the dog. JAVMA 1977;170:820–2.

[40] Salisbury SK, Richardson DC, Lantz GC. Partial maxillectomy and premaxillectomy in

the treatment of oral neoplasia in the dog and cat. Vet Surg 1986;15:16–26.

[41] Salisbury SK, Lantz GC. Long-term results of partial mandibulectomy for treatment of

oral tumors in 30 dogs. J Am Anim Hosp Assoc 1988;24:285–94.

[42] Thrall DE. Orthovoltage radiotherapy of acanthomatous epulides in 39 dogs. JAVMA

1984;184:826–9.

[43] Wallace J, Matthiesen DT, Patnaik AK. Hemimaxillectomy for the treatment of oral

tumors in 69 dogs. Vet Surg 1992;21:337–41.

[44] Forrest LJ, Chun R, Adams WM, Cooley AJ, Vail DM. Postoperative radiotherapy for

canine soft tissue sarcoma. J Vet Intern Med 2000;14:578–82.

[45] McChesney S, Withrow SJ, Gillette E, et al. Radiotherapy of soft tissue sarcomas in dogs.

JAVMA 1989;194:60–3.

[46] Harvey HJ, MacEwen EG, Braun D, et al. Prognostic criteria for dogs with oral mel-

anoma. JAVMA 1981;178:580–2.

[47] MacEwen EG, Patnaik AK, Harvey HJ, et al. Canine oral melanoma: comparison of

surgery versus surgery plus Corynebacterium parvum. Cancer Invest 1986;4:397–402.

[48] Bateman KE, Catton PA, Pennock PW, et al. Radiation therapy for the treatment of

canine oral melanoma. J Vet Intern Med 1994;8:267–72.

[49] Blackwood L, Dobson JM. Radiotherapy of oral malignant melanomas in dogs. JAVMA

1996;209:98–102.

[50] Overly B, Goldschmidt M, Shofer F, et al. Canine oral melanoma: a retrospective study

[abstract]. In: Proceedings of the 21st Annual Conference of the Veterinary Cancer Society,
Baton Rouge. Spring Valley (CA): Veterinary Cancer Society; 2001. p. 43.

[51] Proulx DR, Horn B, Ruslander DM, et al. Canine oral malignant melanoma:

a retrospective analysis of 140 dogs treated with external beam radiation therapy
(1984–2001) [abstract]. In: Proceedings of the 21st Annual Conference of the Veterinary
Cancer Society, Baton Rouge. Spring Valley (CA): Veterinary Cancer Society; 2001.
p. 45.

[52] Dewhirst MW, Sim DA, Forsyth K, et al. Local control and distant metastases in primary

canine malignant melanomas treated with hyperthermia and

/or radiotherapy. Int J

Hyperthermia 1985;1:219–34.

[53] Thompson JM, Dhoodhat YA, Bleehen NM, et al. Microwave hyperthermia in the

treatment of spontaneous canine tumours: an analysis of treatment parameters and tumour
response. Int J Hyperthermia 1988;4:383–99.

[54] Rassnick KM, Ruslander DM, Cotter SM, et al. Use of carboplatin for treatment of dogs

with malignant melanoma: 27 cases (1989–2000). JAVMA 2001;218:1444–8.

[55] MacEwen EG, Kurzman ID, Vail DM, et al. Adjuvant therapy for melanoma in dogs:

results of randomized clinical trials using surgery, liposome-encapsulated muramyl
tripeptide and granulocyte-macrophage colony-stimulating factor. Clin Cancer Res
1999;5:4249–58.

[56] Dow SW, Elmslie RE, Willson AP, et al. In vivo transfection with superantigen plus

cytokine genes induces tumor regression and prolongs survival in dogs with malignant
melanoma. J Clin Invest 1998;101:2406–14.

[57] Bergman PJ, McKnight JA, Novosad CA, et al. Phase I trial of human tyrosinase DNA

vaccination in dogs with advanced malignant melanoma [abstract]. In: Proceedings of the
21st Annual Conference of the Veterinary Cancer Society, Baton Rouge. Spring Valley
(CA): Veterinary Cancer Society; 2001. p. 47.

[58] Straw RC, Powers BE, Klausner J, et al. Canine mandibular osteosarcoma: 51 cases

(1980–1992). J Am Anim Hosp Assoc 1996;32:257–62.

626

M.K. Klein

/ Vet Clin Small Anim 33 (2003) 615–628

background image

[59] Dernell WS, Van Vechten BJ, Straw RC, et al. Outcome following treatment for vertebral

tumors in 20 dogs (1986–1995). J Am Anim Hosp Assoc 2000;36:245–51.

[60] Theon AP, Rodriguez C, Madewell BR. Analysis of prognostic factors and patterns of

failure in dogs with malignant oral tumors treated with megavoltage irradiation. JAVMA
1997;210:778–84.

[61] Gillette EL. Radiation therapy of canine and feline tumors. J Am Anim Hosp Assoc

1976;12:359–62.

[62] Evans SM, Shofer F. Canine oral nontonsillar squamous cell carcinoma. Prognostic factors for

recurrence and survival following orthovoltage radiation therapy. Vet Radiol Ultrasound
1988;29:133–7.

[63] LaDue-Miller TA, Price GS, Page RL, et al. Radiotherapy of canine non-tonsillar

squamous cell carcinoma. Vet Radiol Ultrasound 1996;37:74–7.

[64] Gillette EL, McChesney SL, Dewhirst MW, et al. Response of canine oral carcinomas to

heat and radiation. Int J Radiat Oncol Biol Phys 1987;13:1861–7.

[65] MacMillan R, Withrow SJ, Gillette EL. Surgery and regional irradiation for treatment of

canine tonsillar squamous cell carcinoma: retrospective review of eight cases. J Am Anim
Hosp Assoc 1982;18:311–4.

[66] Brooks MB, Matus RE, Leifer CE, et al. Chemotherapy versus chemotherapy plus

radiotherapy in the treatment of tonsillar squamous cell carcinoma in the dog. J Vet Intern
Med 1988;2:206–11.

[67] Postorino-Reeves NC, Turrell JM, Withrow SJ. Oral squamous cell carcinoma in the cat.

J Am Anim Hosp Assoc 1993;29:438–41.

[68] Hutson CA, Willauer CC, Walder EJ, et al. Treatment of mandibular squamous cell

carcinoma in cats by use of mandibulectomy and radiotherapy: seven cases (1987–1989).
JAVMA 1992;201:777–81.

[69] Evans SM, LaCreta F, Helfand S, et al. Technique, pharmacokinetics, toxicity, and efficacy

of intratumoral etanidazole and radiotherapy for treatment of spontaneous feline oral
squamous cell carcinoma. Int J Radiat Oncol Biol Phys 1991;20:703–8.

[70] Carberry CA, Glanders JA, Harvey HJ, et al. Salivary gland tumors in dogs and cats:

a literature and case review. J Am Anim Hosp Assoc 1988;24:561–7.

[71] Evans SM, Thrall DE. Postoperative orthovoltage radiation therapy of parotid salivary

gland adenocarcinoma in three dogs. JAVMA 1983;182:993–4.

[72] Hammer A, Getzy D, Ogilvie G, et al. Salivary gland neoplasia in the dog and cat: survival

times and prognostic factors. J Am Anim Hosp Assoc 2001;37:478–82.

[73] London CA, Dubilzeig RR, Vail DM, et al. Evaluation of dogs and cats with tumors of the

ear canal: 145 cases (1978–1992). JAVMA 1996;208:1413–8.

[74] Marino DJ, MacDonald JM, Matthiesen DT, et al. Results of surgery and long-term

follow-up in dogs with ceruminous gland adenocarcinoma. J Am Anim Hosp Assoc
1993;29:560–3.

[75] Marino DJ, MacDonald JM, Matthiesen DT, et al. Results of surgery in cats with

ceruminous gland adenocarcinoma. J Am Anim Hosp Assoc 1994;30:54–8.

[76] Theon AP, Barthez PY, Madewell BR, et al. Radiation therapy of ceruminous gland

carcinomas in dogs and cats. JAVMA 1994;205:566–9.

[77] Klein MK, Powers BE, Withrow SJ, et al. Treatment of thyroid carcinoma in dogs by

surgical resection alone: 20 cases (1981–1989). JAVMA 1995;206:1007–9.

[78] Theon AP, Marks SL, Feldman ES, et al. Prognostic factors and patterns of treatment

failure in dogs with unresectable differentiated thyroid carcinomas treated with mega-
voltage irradiation. JAVMA 2000;217:466–7.

[79] Fineman LS, Hamilton TA, de Gortari A. Cisplatin chemotherapy for treatment of thyroid

carcinoma in dogs: 13 cases. J Am Anim Hosp Assoc 1998;34:109–12.

[80] Jeglum KA, Whereat A. Chemotherapy of canine thyroid carcinoma. Compend Contin

Educ Pract Vet 1983;5:96–8.

627

M.K. Klein

/ Vet Clin Small Anim 33 (2003) 615–628

background image

[81] Post GS, Mauldin GN. Radiation and adjuvant chemotherapy for the treatment of thyroid

adenocarcinoma in dogs [abstract]. In: Proceedings of the 12th Annual Conference of the
Veterinary Cancer Society. Spring Valley (CA): Veterinary Cancer Society; 1992. p. 43–4.

[82] Mendelsohn J, Shin DM, Donato N, et al. The epidermal growth factor receptor as a target

for cancer therapy. Endocr Relat Cancer 2001;8:3–9.

[83] Shin DM, Donato NJ, Perez-Soler R, et al. Epidermal growth factor receptor-targeted

therapy with C225 and cisplatin in patients with head and neck cancer. Clin Cancer Res
2001;7:1204–13.

[84] Mazeron JJ, Noel G, Simon JM. Head and neck brachytherapy. Semin Radiat Oncol

2002;12:95–108.

[85] Schwarz PD, Withrow SJ, Curtis CR, et al. Mandibular resection as a treatment of oral

cancer in 81 dogs. J Am Anim Hosp Assoc 1991;27:601–10.

[86] Schwarz PD, Withrow SJ, Curtis CR, et al. Partial maxillary resection as a treatment for

oral cancer in 81 dogs. J Am Anim Hosp Assoc 1991;27:617–24.

628

M.K. Klein

/ Vet Clin Small Anim 33 (2003) 615–628

background image

New chemotherapy agents in

veterinary medicine

Antony S. Moore, BVSc, MVSc

a,

*,

Barbara E. Kitchell, DVM, PhD

b

a

Section of Oncology and Harrington Oncology Program, Tufts University School of

Veterinary Medicine, 200 Westboro Road, North Grafton, MA 01536, USA

b

Veterinary Cancer Care Clinic, University of Illinois College of Veterinary Medicine,

1008 West Hazelwood Drive, Urbana, IL 61802, USA

Several agents have been developed in recent years for anticancer therapy.

This article discusses experience with older agents lomustine and strepto-
zocin and newer agents ifosfamide and gemcitabine.

Lomustine

Mechanisms of action

Lomustine (cyclohexylchloroethylnitrosourea, CCNU, Ceenu) is a nitro-

sourea that is hydroxylated intracellularly to form an alkylating molecule
that creates DNA–DNA and DNA to protein cross-linkages. The key site
for DNA alkylation appears to be the O-6 methyl group of guanine. In
addition, an isocyanate compound is generated that can inhibit DNA
polymerase, DNA ligase, glutathione reductase, and enzymes involved in
RNA synthesis and processing. Hepatic microsomal metabolic transfor-
mation is responsible for deactivation of lomustine, and enhanced micro-
somal activity, such as would be seen with concurrent phenobarbital
administration, leads to a decrease in therapeutic efficacy. Hepatic deg-
radation occurs rapidly and completely in most species, but this has not
been explored in cats. The plasma half life in dogs after intravenous (IV)
injection is approximately 15 minutes, and lomustine is detectable in the

Vet Clin Small Anim

33 (2003) 629–649

* Corresponding author.
E-mail address:

antony.moore@tufts.edu

0195-5616

/03/$ - see front matter

Ó 2003, Elsevier Inc. All rights reserved.

doi:10.1016

/S0195-5616(03)00033-0

background image

cerebrospinal fluid (CSF) within 10 minutes, reaching a plasma: CSF ratio
of 1:3 within 30 minutes [1].

Lomustine is highly lipophilic, and it is completely absorbed by the oral

route of administration. Lipid solubility is responsible for distribution
across biologic membranes including the blood-brain barrier. In contrast
to other alkylating agents, lomustine enters cells through passive diffusion.
Because cellular transport is passive, reduced drug entry into cells is not
a mechanism of resistance. Enhanced repair of alkylation at the O-6 methyl
group of guanine by the enzyme guanine-O-6-transferase is associated with
drug resistance to lomustine. Increased levels of intracellular thiol may
function to inactivate alkylating agents including lomustine. Glutathione
transferases are important in the denitrosation inactivation of CCNU [2].

Activity in people

The major indications for use of lomustine in people are treatment of

brain tumors, lymphoma (Hodgkin’s and non-Hodgkin’s), melanoma, and
renal and pulmonary carcinomas.

Preclinical studies

In preclinical studies using beagles, the most consistent toxicities involved

the bone marrow, liver, kidneys, and gastrointestinal (GI) tract. At high
doses, lomustine caused leukopenia and anemia during treatment. If the
animals did not die of sepsis or disseminated intravascular coagulation
(DIC), recovery of bone marrow was seen. Thrombocytopenia appeared to
be delayed, but reversible. Single doses of 200 mg

/m

2

and above were

associated with 50% mortality, while doses of 60 mg

/m

2

and below were

associated with only mild hematological toxicity when lomustine was given
by the oral or intravenous route [3,4]. Daily oral dosing of 25 to 200 mg

/m

2

per day for 14 days was associated with almost 100% mortality, but
13 mg

/m

2

per day for 14 days was associated with reversible leukopenia and

thrombocytopenia and reversible hepatic damage [3]. Studies of autologous
bone marrow support for high-dose chemotherapy allowed doses of 300 to
400 mg

/m

2

to be given without lethal bone marrow toxicity. At doses of 600

mg

/m

2

, however, gastrointestinal toxicity was dose-limiting [5].

Delayed hepatotoxicity resulted in elevations of serum transaminase and

alkaline phosphatase activity and increased bromsulfophthalein (BSP) re-
tention. There was an inverse relationship between the size of the lomustine
dosage and the time to development of hepatotoxicity as signaled by eleva-
tions in serum alanine aminotransferase (ALT) activity. Hepatotoxicity was
seen as late as 1 month after dosing was discontinued but appeared to be
reversible at the lowest dosages (13 mg

/m

2

per day for 14 days). Oral doses

of 6 mg

/m

2

or less for 14 days caused only mild hepatic fatty changes [3].

Further investigation using serial liver biopsies following a single oral

630

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

lomustine dose of approximately 100 mg

/m

2

showed glycogen depletion and

increased serum alkaline phosphatase (ALP) activity, noted 3 to 4 weeks
after dosing and continuing for more than 3 months. Histologic changes
were persistent, although serum alanine transferase (ALT) activity returned
to normal within 3 to 4 months [6].

Renal toxicity was uncommon in dogs but was seen at dosages of 150 to

200 mg

/m

2

per day. In monkeys, renal toxicity is unpredictable and ir-

reversible at high doses [3]. Gastrointestinal [GI] toxicity was only seen at
the very highest dosages [3].

There are no preclinical studies of lomustine in cats.

Clinical applications in dogs

Dosage

Several doses of lomustine have been reported in different settings. The

largest case series recommend clinical dosing of lomustine at 90 mg

/m

2

orally every 4 weeks, with a dose reduction to 70 mg

/m

2

every 4 weeks if

severe leukopenia (neutropenia; less than 500 cells per lL) occurs. The
neutrophil nadir is seen 6 to 7 days after dosing. It may be prudent to
prophylactically administer oral broad-spectrum antibiotics such as tri-
methoprim-sulfa combinations during the expected nadir from 5 until 10
days after dosing. The drug should be discontinued if thrombocytopenia or
increased serum creatinine or ALT activity is documented at the time
a dosage is due.

A lomustine dose of 60 to 80 mg

/m

2

given every 6 to 8 weeks also has

been promoted for treatment of brain tumors in dogs. Lomustine treatment
at a dose of 50 mg

/m

2

every 3 weeks has been reported in abstract form for

treatment of cutaneous lymphoma in dogs [10,11].

Toxicities

Myelosuppression is the major dose-limiting toxicity when lomustine is

administered to dogs in the clinical setting. When lomustine was given at
a dose of 100 mg

/m

2

, acute neutropenia was dose-limiting, and this dosage

was abandoned as too toxic [7]. Although neutropenia following lower doses
can often be severe (less than 500 neutrophils per lL), it is rarely associated
with sepsis, possibly because of the lack of GI toxicity seen at these clinical
dosages. As discussed previously, oral broad-spectrum antibiotics such as
trimethoprim-sulfa combinations may be added prophylactically during the
expected neutrophil nadir from 5 to 10 days after dosing.

Thrombocytopenia may be documented at the nadir (7 to 14 days), but

platelet counts usually return to normal by the time the next dosage is
scheduled. Thrombocytopenia that persists at the time of next dose
necessitates dose delay. Persistent thrombocytopenia may be cause to dis-
continue the drug. Continued administration of lomustine to even mildly
thrombocytopenic dogs may result in dramatic worsening, with delayed and

631

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

prolonged (months) thrombocytopenia that progresses even after the drug is
discontinued.

Idiopathic fevers that were not associated with neutropenia have been

observed in dogs receiving lomustine for relapsed lymphoma and mast cell
tumors, but this does not seem to be a common toxicity [7,8]. Hepatotoxicity
is uncommon at clinical doses, but this can be severe and progressive. In
a series of 180 dogs that were considered at true risk for developing
hepatotoxicity (more than one treatment, serum chemistry profile monitor-
ing performed, and no evidence of pretreatment liver disease), 12 dogs
developed hepatotoxicity. Toxicity occurred after 2 to 10 doses of lomustine,
and was associated with clinical signs in 11 of the 12 dogs. Clinical signs seen
most commonly were loss of appetite and weight loss. The average time to
developing clinical signs of liver disease was 10 weeks after the last dose was
administered. Seven of the 12 dogs died from liver disease at a median of 7
weeks after diagnosis, while four dogs improved clinically [9]. Therefore, it is
recommended that a serum chemistry profile be obtained before each
treatment with lomustine and that the drug be discontinued in the event of
elevated serum ALT activity. As in preclinical studies, it appears that the
liver changes seen in tumor-bearing dogs may be reversible if the drug is
discontinued at a low cumulative dosage.

Renal toxicity appears to be less common than hepatotoxicity at

recommended clinical doses in the dog (Kristal, personal communication,
2002). One dog developed acute fatal renal failure after receiving six doses of
CCNU. On necropsy, the renal lesion was membranoproliferative glomer-
ulonephritis [7]. Although nephrotoxicity is uncommon, dogs should be
monitored for evidence of renal damage during therapy with CCNU.

Efficacy

A preliminary report of lomustine given at a dosage of 60 to 80 mg

/m

2

every 6 to 8 weeks documented measurable brain tumor regressions in dogs,
and possibly prolonged remission in dogs that were treated adjunctively
following surgery [10]. Although this information has doubtless resulted in
treatment of other dogs, there have been no further reports on the efficacy of
lomustine for brain tumors.

Forty-three dogs received lomustine at doses between 90 and 100mg

/m

2

by mouth as treatment for relapsed and resistant lymphoma. These dogs
had received a median of five chemotherapy drugs for a median of 180 days
before rescue therapy with lomustine [7]. The overall response rate was 28%,
with a median remission duration of 86 days. The complete response rate
was low, however. Only 7% of dogs had a median complete remission of 110
days.

In a group of seven dogs with cutaneous lymphoma, the complete re-

sponse rate was 100% to lomustine treatment at a dose of 50 mg

/m

2

every 3

weeks, with remission lasting 2 to 15 months. Those with epitheliotropic T

632

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

cell lymphoma (mycosis fungoids) that had surgical excision followed by
chemotherapy with lomustine had the longest survival times [11].

Nineteen dogs with measurable mast cell tumors of various grades were

treated with lomustine at a dose of 90 mg

/m

2

every 3 weeks [8]. One dog had

a long complete response (14 months), and the tumors in seven dogs reduced
by more than 50% in size for a median of nearly 3 months. The acute dose
limiting toxicity was neutropenia; 41% of these dogs had a neutrophil count
of less than 1000 neutrophils per lL during lomustine therapy. Following
these studies, clinical experience with longer-term therapy in the adjuvant
setting revealed an apparent cumulative myelosuppression in dogs, which
primarily affected platelet production. It was reasoned that a 3-week inter-
treatment interval was too short to allow for full marrow recovery. As
a result, it is recommended that a dose of 90 mg

/m

2

be administered no more

frequently than every 4 weeks, and that a complete blood count (CBC),
including a platelet count, be performed before each treatment. If the
platelet count is below normal at the time the next dose is due, then CCNU
should be discontinued. Continued therapy can lead to a marked and pro-
gressive drop in platelet numbers that continues for months after the drug is
discontinued.

Similarly, the occurrence of hepatotoxicity, while uncommon in the

clinical setting, appears to be cumulative, and often irreversible if not
recognized early. A serum chemistry profile that includes serum alanine
transferase activity should be performed before each treatment, and CCNU
should be discontinued if the levels are elevated above normal.

Clinical applications in cats

Dosage

The recommended dose of lomustine for cats is 50 to 60 mg

/m

2

orally

every 5 to 6 weeks [12]. The drug is reformulated as 2.5 mg capsules and
administered to the nearest 2.5 mg based on body size. This difference in
recommended dosage and administration interval between cats and dogs
reflects more the more significant, prolonged, and severe neutropenia seen in
cats treated with lomustine. It is thought that cats are more similar to people
than to dogs with regard to lomustine toxicity. In people, hematologic
toxicities are delayed (neutrophil nadir at 3 to 4 weeks,) and complete
hematologic recovery is not seen until 6 to 7 weeks after therapy [13].

Toxicities

Lomustine at the recommended dosage reported previously resulted in

a median neutrophil count at the nadir of less than 1000 neutrophils per lL
[12]. In that study, more than half the cats experienced a neutrophil nadir
14 days after treatment, but nadirs also were seen from 7 to 28 days after
treatment. In addition, the neutrophil nadirs were often prolonged, with
a median of 7 days, but up to 14 days in individual cats. Information

633

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

regarding platelet counts was less reliable, but the nadir appeared to occur
14 to 21 days after treatment [12].

Because of the formulation of lomustine capsules at a minimum size of 10

mg, another investigator administered 10 mg every 3 weeks to each cat
regardless of body size. This approach to dosing resulted in a range of doses
from 32 to 59 mg

/m

2

in the series of cats studied [14]. In that study,

myelosuppression was rare, occurring in only 4% of patients, but there was
a trend to more severe neutropenia at higher cumulative doses. The low
incidence of toxicities in this study indicates that optimum therapeutic dose
intensity may not have been achieved by the 10 mg per cat approach. When
treating cats with lomustine, reformulation to achieve a uniform dose of 50
to 60 mg

/m

2

is recommended.

There are insufficient data to comment on the risk of hepatotoxicity,

renal damage, and thrombocytopenia in cats. It is recommended that pa-
tients be monitored as described for dogs, until further data become
available.

Efficacy

There have been no studies of the efficacy of CCNU in the treatment of

feline tumors. The toxicity studies previously documented responses in cats
with lymphoma [14,12], mast cell tumor [12], fibrosarcoma [14], and multiple
myeloma [14]. The relatively modest response rates seen in these pilot studies
mean that it is unlikely that lomustine will have a similar therapeutic profile
for feline malignancies as that documented in dogs.

Ifosfamide and mesna

Mechanisms of action

Ifosfamide (Ifex) is an oxazaphosphorine nitrogen mustard that, like its

isomeric parent compound cyclophosphamide, has no intrinsic alkylating
activity until it is metabolized hepatically to an active form.

The alkylating effect of the active metabolites causes DNA interstrand

cross linking that correlates to toxicity; areas of active transcription appear
to be the most vulnerable to damage. Ifosfamide requires hepatic P450
mixed function oxidase for metabolism to acrolein and an active compound.
It is therefore at least theoretically possible that hepatic dysfunction may
lead to reduced activation and therefore to reduced efficacy.

Without accompanying urothelial protection, the renally excreted

metabolites of ifosfamide cause urothelial toxicity (hemorrhagic cystitis) in
all patients. The metabolites of ifosfamide also can cause renal damage on
rare occasions. Identification of the urothelial damaging metabolites leads to
the subsequent discovery of the protective drug mesna (sodium 2-mercapto-
ethane sulfonate). When administered intravenously, mesna rapidly forms

634

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

a disulfide, which is eliminated rapidly and completely through glomerular
filtration. During excretion, the disulfide is reduced to mesna. Mesna
(Mesnex) acts as an active thiol in the urinary tract to scavenge the urothelial
metabolites of the alkylating agent, thereby protecting the urinary system
epithelium.

Activity in people

Ifosfamide is one of the most active chemotherapeutics in the treatment

of soft tissue sarcomas and osteosarcomas in humans. Additionally,
ifosfamide has significant activity in the treatment of urothelial transitional
cell carcinoma; germ cell tumors; pulmonary, ovarian, and breast car-
cinomas; and lymphoma. Ifosfamide has been demonstrated to have
superior antineoplastic activity when compared with cyclophosphamide in
many tumors in people.

Preclinical studies

Administration of ifosfamide at a dosage of 300 mg

/m

2

daily for 4 days

resulted in the septic death of one dog at the neutrophil nadir on day 8;
however, three dogs survived [15]. Administration of a higher dosage (450
mg

/m

2

daily for 4 days) to the remaining dogs resulted in sepsis in all

animals; however, all dogs recovered with supportive care. Continued
administration of the higher dosage at intervals of 3 weeks resulted in a 50%
mortality caused by sepsis. No urothelial toxicity was seen; all dogs received
mesna at a daily dose of 1.5 times the dose of ifosfamide and a further two
doses 8 and 16 hours after the last treatment. Ifosfamide was delivered in 1 L
of lactated Ringer’s solution over 90 minutes.

In separate dog studies, the highest nontoxic dose for ifosfamide given

daily for 5 days was 4.12 mg

/kg per day [16]. At a dose of 8.25 mg/kg per day

for 5 days, an intertreatment break of 9 days was insufficient to allow full
hematological recovery, as evidenced by worsening leukopenia with
subsequent cycles of therapy [16]. Cystitis was common in dogs treated
every 3 weeks at a dosage of 10 to 20 mg

/kg for 6 months. These dogs did

not receive mesna. Leukopenia was characterized as moderate, although
some dogs developed pneumonia. Ifosfamide had sufficient activity when
given orally at a dose of 4.64 mg

/kg per day, 6 days a week for 26 weeks, to

result in marked neutropenia in most treated dogs [16].

Preclinical studies of mesna in the dog showed that single doses of 200

mg

/kg and higher caused bradycardia and decreased blood pressure, and

death was seen at doses of 400 mg

/kg and above. When given IV daily for

6 weeks, intermittent vomiting and diarrhea was seen at doses between 100
and 300 mg

/kg. Oral dosing up to 2000 mg/kg had no effect on dogs. These

studies emphasize the safety of mesna at the much lower, clinically relevant

635

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

doses. The lack of interaction between mesna and ifosfamide activity (with
the exception of urothelial toxicity) has been confirmed in dogs [17].

In the cat, ifosfamide administered at 100 mg

/kg IV did not result in

anaphylaxis [16], and mesna caused no adverse effects up to the tested
dosage of 100 mg

/kg [17].

Clinical applications in dogs

Dosage

The recommended dosage of ifosfamide in dogs is 350 to 375 mg

/m

2

by

IV every 2 to 3 weeks. The drug must be given with IV diuresis and mesna.
Mesna is administered at 20% of the ifosfamide dose, before treatment.
Diuresis is initiated at 18.3 mL

/kg per hour for 30 minutes, and then the

ifosfamide is given at the same fluid rate over a further 30 minutes. Diuresis
is continued at the same rate for an additional 5 hours. Two additional
doses of mesna (20% of the ifosfamide dose each time) are given 2 and 5
hours after the ifosfamide infusion is completed during the post-treatment
diuresis phase.

Note: Mesna (Mesnex) is supplied with Ifex in the same package, and

does not have to be purchased as a separate drug.

Toxicities

A total of 134 doses of ifosfamide were given to 72 dogs in one study [18].

A dosage of 375 mg

/m

2

was given to 37 evaluable dogs. Six dogs (16%)

developed neutropenia of less than 1000 cells

/lL, and three dogs developed

sepsis. Renal toxicity does not appear to be a substantial risk with this
protocol. Anecdotally, attempts to increase the dosage of ifosfamide to 400
mg

/m

2

have failed because of severe neutropenia (Rassnick and Moore,

unpublished data).

When given according to the protocol described previously, no dog

reported in this study, nor any dogs treated subsequently by the author,
has developed hemorrhagic cystitis following ifosfamide administration.
A preclinical trial that did not use diuresis, but did administer mesna, also
did not result in hemorrhagic cystitis [15]. There have been no reported
GI toxicities reported in dogs treated with ifosfamide.

Efficacy

In one study, complete responses were documented in 2 of 13 dogs with

measurable sarcomas. In this report, a dog with leiomyosarcoma had
a response of longer than 549 days, and a dog with hemangiosarcoma had
a response of longer than 445 days. Only 1 of 40 dogs treated for resistant
lymphoma was noted to have a partial response lasting 112 days. Seven dogs
with stage II splenic hemangiosarcoma lived a median of 147 days, which

636

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

was not statistically different from the survival of 86 days noted with surgery
alone.

The antitumor activity of ifosfamide in dogs does not appear to be as

high as it is in people with cancer. The reasons for this lack of efficacy are
not clear.

Clinical applications in cats

Dosage

Clinical dosing of ifosfamide in cats is being investigated. From

preliminary work, it appears that cats tolerate higher ifosfamide dosages
than do dogs, possibly doses as high as 900 mg

/m

2

when given with diuresis

as outlined previously (Rassnick, personal communication). The reader is
encouraged to confirm this dosage before administering the drug to cats.

Toxicities

Myelosuppression appears to be the major dose-limiting toxicity in cats,

but this observation needs to be confirmed through further studies. Other
toxicities seen include GI toxicity, probable idiosyncratic hypersensitivity
(swollen face and puffy eyes) in one cat, and renal failure in two cats, one of
which had pre-existing chronic renal failure. Ifosfamide is a nephrotoxic drug,
and careful case selection and monitoring of renal function are mandatory
before administration to cats (Rassnick, personal communication).

Efficacy

Responses have been seen in cats with lymphoma and in cats treated for

soft tissue sarcomas. Therefore, ifosfamide and mesna may prove useful for
the treatment of sarcomas in cats (Rassnick, personal communication),
although further study is needed.

Streptozocin

Mechanisms of action

Streptozocin (streptozotocin, Zanosar) is a methylnitrosourea alkylating

agent that causes interstrand cross-linking in DNA. It is excreted primarily
in the urine. Streptozocin is directly cytotoxic to pancreatic beta cells. When
given by IV, streptozocin metabolism is rapid. Degradation occurs at an
estimated rate of 5 mg per minute in dogs [19], and the half-life in people is
15 minutes [20]. The drug is retained in the liver of dogs for many hours
after blood levels are undetectable [19].

Activity in people

Streptozocin is used for the treatment of unresectable or metastatic

insulinoma. Other diseases in people that have responded to streptozocin

637

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

treatment include carcinoid, Hodgkin’s disease, lymphocytic lymphoma,
acute lymphoblastic leukemia, and synovial cell sarcoma [21–23].

Preclinical studies

In preclinical studies in dogs, the LD

50

of streptozocin was found to be

1500 mg

/m

2

[24]. In acute toxicity studies in dogs, the major toxicoses were

diabetes mellitus, which developed after IV administration of a single dose
of 700 mg

/m

2

, renal tubular damage, weight loss, and reversible hepatic

injury [25,26]. Streptozocin was reported to be a powerful emetogen in dogs,
with vomiting occurring 45 to 90 minutes after administration [27]. Toxic
effects of streptozocin are dose-dependent, with vomiting and increases in
serum hepatic enzyme activities occurring at lower dosages (400 mg

/m

2

),

and acute renal tubular damage occurring at higher dosages (greater than
700 mg

/m

2

) [25,26].

There are no preclinical studies in cats.

Clinical applications in dogs

Dosage

Clinical use of streptozocin in two dogs was reported to result in acute

renal failure and death. These reports were flawed, in that the drug was
administered without appropriate renal support [28,29]. Induction of
diuresis through administration of 0.9% NaCl has been reported to
ameliorate the renal toxic effects of streptozocin in canine patients. This
effect is attributed to protection of renal tubular macromolecules from the
alkylating effects of streptozocin by reduced contact time between the drug
and the renal tubular epithelium [30].

Intravenous diuresis, using 0.9% NaCl at a rate of 18.3 mL

/kg per hour,

was administered for 3 hours before streptozocin administration. The dose
of streptozocin (500 mg

/m

2

) was diluted to the appropriate volume and

administered over the subsequent 2 hours at the same calculated fluid rate,
and 0.9% NaCl was administered for an additional 2 hours at the same rate
after streptozocin administration was completed. Butorphanol (0.4 mg

/kg)

was administered intramuscularly (IM) as an antiemetic immediately after
streptozocin administration was completed. Protection against vomiting
provided by butorphanol is less than complete. Treatments are repeated at
3-week intervals [30].

Toxicities

The major toxicity associated with streptozocin treatment in dogs is

proximal renal tubular necrosis, which is dose-related and cumulative.
Renal failure may develop. Less common toxicoses include nausea and
vomiting, which may be severe, and increases in serum hepatic enzyme
activities. Bone marrow toxicity is rare.

638

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

In one study, 58 treatments were administered to 17 dogs with pancreatic

islet cell tumors [30]. Two dogs developed diabetes mellitus after receiving
five doses of streptozotocin. Only one dog developed azotemia during
treatment. This dog had pre-existing renal disease. Serum ALT activity
increased in some dogs, but decreased again when treatment was
discontinued. Hematologic toxicoses were rare and mild. Vomiting during
administration was seen following approximately 30% of streptozocin
treatments, and one dog was withdrawn from treatment by its owners
because of severe vomiting after each treatment. The high incidence of
vomiting after streptozocin treatment occurred despite pretreatment with
butorphanol. Butorphanol acts centrally to reduce nausea and has been
shown to reduce the risk of vomiting after cisplatin chemotherapy in dogs
[31] but was ineffective in preventing streptozocin-associated vomiting. It is
possible that other antiemetics such as dolasetron or ondansetron may be
more effective in preventing vomiting in dogs receiving streptozocin. Acute
post-treatment hypoglycemia has been reported, presumably as a result of
streptozocin-induced beta cell degranulation. Clinicians should be aware
of this potential complication and be prepared to administer glucose if signs
of hypoglycemia occur.

Efficacy

There was no measurable response in one dog with a nonfunctional islet

cell tumor. Despite renal toxicity seen in early case reports, evidence of
antineoplastic activity was seen in two dogs with insulinoma [28,29]. In
a more recent study, median duration of normoglycemia for 14 dogs with
stage II or III insulinoma treated with streptozocin was 163 days. This
patient cohort included dogs that had shown signs of tumor recurrence after
surgery and medical management. The normoglycemic period seen in this
study was not significantly different from that for dogs treated with surgery
and medical management alone (90 days). Two dogs had rapid resolution of
paraneoplastic peripheral neuropathy within 2 weeks of starting treatment,
however, and two others had measurable reductions in tumor size [30].

Based on the findings of these two reports, it would appear that

streptozocin has a role in the treatment of metastatic insulinoma in dogs.
Additional studies are required to better define the therapeutic potential of
this agent.

Clinical applications in cats

Streptozotocin administration has not been reported in cats.

Gemcitabine

Gemcitabine (2,2-difluorodeoxycytidine, dFdC, Gemzar) is a difluori-

nated pyrimidine analog of deoxycytidine, synthesized as an analog of

639

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

cytarabine. This drug was the first compound licensed based on antitumor
efficacy and on improved quality-of-life scores in the pivotal study of people
with pancreatic carcinoma. The drug was licensed for human use in the 1996
[32,33].

Mechanism of action

The synthetic design used to create gemcitabine substitutes the hydrogens

of the 2-carbon position of the deoxyribose moiety with two fluorine atoms.
This confers on the drug its DNA-directed biological activities [34].
Gemcitabine, like other nucleoside analogs, is hydrophilic and cannot tra-
verse cell membranes by passive diffusion. The prodrug gemcitabine enters
cells through the activity of specialized transporter systems (facilitated
nucleoside transport). Phosphorylation of gemcitabine is essential to its
biologic activity. The prodrug is metabolized intracellularly to the 59-
monophosphate form (dFdCMP) by deoxycytidine kinase. This intermediate
compound subsequently is phosphorylated by nucleoside monophosphate
and diphosphate kinases to the active diphosphate (dFdCDP) and tri-
phosphate (dFdCTP) nucleosides, respectively [34-37]. Gemcitabine also
increases its own intracellular concentration by a positive feedback loop of
activation. The drug requires intracellular phosphorylation by deoxycytidine
kinases to achieve the activated triphosphate form. This deoxycytidine kinase
then blocks ribonucleotide reductase function, which depletes the normal
cytidine base. Thus, greater incorporation of the gemcitabine in the DNA
occurs, as fewer molecules of the normal base are available to compete for
insertion (Fig. 1) [34–37].

The cytotoxic effect of gemcitabine is attributed to a combination of two

actions of the diphosphate and triphosphate nucleosides, which results in
inhibition of DNA synthesis [34–37]. Following the incorporation of
dFdCTP into DNA, one additional nucleotide molecule is added to the
elongating DNA strands. After this normal base addition, DNA polymerase
e is unable to remove the dFdCTP and repair the growing DNA strands.
This ‘‘masked chain termination’’ results in inhibition of DNA synthesis and
induction of apoptosis. Gemcitabine exhibits cell phase specificity, primarily
killing cells undergoing DNA synthesis (S-phase) and also blocking the
progression of cells through the G1

/S phase boundary. Resistance appears

to be caused by reduced nucleoside transport into the cells and also to
decreased activity of the enzyme deoxycytidine kinase [38].

Activity in people

Gemcitabine is licensed in human medicine for treatment of pancreatic

carcinoma and nonsmall cell lung cancer [32,39], but its utility in com-
bination with other drugs has resulted in the expansion of the drug’s clinical
indication to include treatment of other GI [40], genitourinary [41,42], and

640

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

respiratory carcinomas [39]. Preclinical studies and phase II trials document
the potential utility of gemcitabine in the combination treatment of various
lymphomas and soft tissue and bone sarcomas [43–45]. The drug appears to
be synergistic with platinum agents [41,46], anthracyclines, and alkylating
drugs [46,47] and is a very potent radiosensitizer [48–50]. Significantly
enhanced radiation injury to normal tissues in the radiation field have
limited its use as a radiosensitizer in people, and substantial dose reductions
are required over what are used in systemic chemotherapy [48,49]. Human
dose de-escalation studies were required for the use of gemcitabine as
a radiosensitizer in head and neck cancer, because of deep ulceration and
esophageal stricture. The estimated maximum tolerated dose in combination
with radiation therapy in people is between 10 to 50 mg

/m

2

, which is less

than 5% of the human systemic cytotoxic dose [50].

Preclinical studies

A wide dose range of gemcitabine was administered to normal beagles in

published preclinical studies (3 to 10 mg

/kg, or 500 mg/m

2

once weekly for 3

weeks) [51,52]. Dogs metabolize gemcitabine by a two-compartment model.
The drug is deaminated to the uracil metabolite and subsequently cleared
through the kidneys. Plasma protein binding is negligible, and the plasma
half-life in the dogs is approximately 1.38 hours. Approximately 76% to
86% of the dose is detected as the uracil metabolite in the urine within the
first 24 hours of administration [50,51]. Administration of gemcitabine at

Fig. 1. Molecular structure olf gemcitabine and mechanism of activation (Courtesy of Dr L-P
de Lorimier, modified from Abbruzzese JL. New applications of gemcitabine and future
directions in the management of pancreatic cancer. Cancer 2002;95:941–5; with permission.)

641

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

a dosage of 350 mg

/m

2

administered as a 1-hour IV infusion to normal

healthy beagles resulted in minimal toxicity [53,54]. Administration of
1200 mg

/m

2

as an IV bolus was the estimated toxic dose (Horton, personal

communication).

There are no preclinical studies in cats.

General clinical applications

The first veterinary administration of gemcitabine occurred at the

University of Illinois shortly after the drug became commercially available.
Results of the initial pilot study were reported by Boyce [55]. Doses used in
this study were derived from the ‘‘no toxic effect’’ dose in preclinical studies
conducted at Lilly (Englehart, personal communication). Thus, the dose for
dogs was 60 mg

/m

2

IV once weekly for a maximum of five weekly treat-

ments; 45 mg

/m

2

was administered once weekly for five treatments to cats.

All doses were given as a 20-minute infusion. Most patients had previously
failed standard chemotherapy protocols and were assessed to have
progressive disease. If the drug was found to be efficacious for a given
patient after the first cycle, the cycle was repeated after a 2-week rest period.
Tumor types treated included hepatocellular carcinoma, biliary carcinoma,
lymphosarcoma, cholangiocarcinoma, pancreatic carcinoma, mammary
carcinoma, and bronchoalveolar carcinoma. Stable disease with prolonged
survival over that seen in historical controls was reported for most cases
treated. Partial responses and one complete response were seen in a dog with
multicentric hepatocellular carcinoma. In this initial study, toxicity was
minimal, and no animals died from treatment-related causes [55].

Walter et al reported on the use of gemcitabine as a rescue agent for

recurrent or refractory canine lymphoma. A dose of 275 mg

/m

2

was ad-

ministered to five dogs as a 30-minute infusion every 2 weeks for up to five
cycles. No complete or partial responses were observed [56].

Kisseberth et al reported the results of a dose escalation study of 17 dogs

conducted at The Ohio State University. Escalation was performed in
cohorts of three, with 50% escalation in the succeeding cohort if no
significant toxicities were observed. Doses administered ranged from 300 to
675 mg

/m

2

(109 doses in total) administered as a 30-minute IV infusion of

drug in saline at a rate of 1 mL

/kg. Thirteen of these dogs received four or

more treatments. GI toxicity was evaluated for the first four treatments,
with toxicity scores assigned as follows: 11 grade I; 5 grade II; 0 grade III;
and 1 grade IV (gastric carcinoma) in 61 evaluable treatments. Only one
grade I episode of neutropenia was reported. No complete or partial tumor
responses were observed during the first 8 weeks in this group of dogs, with
the best response being stable disease in nine dogs. One dog with thyroid
carcinoma suffered bilateral retinal hemorrhages, which resolved without
treatment, and dosing was continued. These authors concluded that gem-
citabine could be safely administered at 675 mg

/m

2

at 14-day intervals with

642

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

minimal gastrointestinal and hematological toxicity. Further escalations
were not performed because of the single incident of unexplained retinal
hemorrhage [57].

Marconato et al reported on 33 dogs treated for confirmed transitional

cell carcinoma of the urinary bladder. All dogs in this group were treated
concurrently with piroxicam. Dosing started at 800 mg

/m

2

IV once weekly,

with planned treatment to continue for at least seven cycles. Infusion du-
ration was not reported. Dose escalation was attempted in three dogs. The
median number of doses administered was eight, with a range of 1 to 27
doses. Nausea or vomiting occurred in 65% of the patients treated at the 800
mg

/m

2

dose, but GI toxicity was typically grade I to II. Neutropenia was the

most common hematological adverse effect (nadir 3 to 7 days), and no
febrile episodes or sepsis were observed. Thrombocytopenia and anemia
were rare. Clinical improvement of stranguria, pollakiuria, and hematuria
was reported for all treated dogs [58].

Gemcitabine is being investigated as a radiosensitizer for dogs and cats.

An abstract presented by Ladue and Klein summarized the interim report of
the Veterinary Radiation Therapy Oncology Group trial of gemcitabine as
a radiosensitizer in canine sinonasal carcinoma and feline oral squamous cell
carcinoma. Fifteen dogs were entered from two radiation facilities. Radi-
ation therapy was administered to an average dose of 50 Gy in Monday
through Friday fractions of 3 to 3.2 Gy per fraction. The average number of
gemcitabine doses was five per patient, with a mean dose of 40 mg

/m

2

.

Twelve of 15 dogs required dose delay or reduction because of myelo-
suppression or local tissue toxicity in the radiation field. Results for these
dogs were not encouraging, as the median local disease control interval was
only 8.6 months. Nine dogs died of progressive disease. Three dogs were
alive with no evidence of disease at the time of the report (2, 8, and
29 months). Two dogs died of unrelated disease, while one was disease-free
but lost to follow-up at 18 months [59].

Clinical applications in dogs

Dosage

The optimal dose regimen for use of gemcitabine in tumor-bearing dogs

has yet to be established. Despite the common clinical use of this agent in
oncology practice for the past decade, controversy still exists as to the ap-
propriate dose and schedule for people with cancer [60]. One problem
associated with establishing efficacy and toxicity of gemcitabine in tumor-
bearing dogs is that its effects are dose- and schedule-dependent [61]. Using
an identical dose, but prolonging the infusion of drug from an IV bolus
injection to a 30-minute or longer duration, commonly results in increased
myelosuppression (Kitchell, unpublished data). One could theorize that this
also could provide increased anticancer efficacy. There are several critical
limiting factors to be considered in gemcitabine administration, however. As

643

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

a relatively cell cycle phase-specific drug, with maximum effect seen during S
phase, it is likely that continuous IV infusion over a long period would result
in increased exposure of S phase cells to the toxic effects of the drug. This
efficacy is initially dependent upon the presence of appropriate nucleoside
transporters for intracellular uptake of the parent compound, however.
Subsequently, adequate activity of deoxycytidine kinase (rate-limiting step)
and other kinases in the tumor target cells is required to ensure appropriate
phosphorylation to the active forms. The steps of uptake and activation are
saturable; therefore, rapid infusion of high doses possibly could result in
rapid renal elimination, with no appreciable therapeutic impact [62,63]. The
intracellular accumulation of the triphosphate form of gemcitabine occurs
rapidly after infusion, which results in incorporation of the active agent into
the growing DNA chain during S phase and subsequent chain termination.
The added cytotoxic impact of accumulation of gemcitabine triphosphate
within the cell, with resultant inhibition of deoxycytidine kinase and
ribonucleotide reductase activity, occurs only with prolonged intracellular
concentration of the triphosphate form, which is achieved best by con-
tinuous long-term exposure [64]. The conceptual problem in designing dose-
finding studies for gemcitabine is that dose escalation and duration of
infusion (‘‘time-escalation’’) should be incorporated. Dose and duration of
infusion are under investigation in many clinical trials in people [40].

The main clinical utility of gemcitabine is likely not as a single agent, but

rather as a synergistic combination chemotherapy drug, by human medical
oncologists. There is no consensus for gemcitabine single-agent therapy or
as use to enhance the cytotoxic effect of radiation therapy. This is clearly the
direction taken by dosing regimes in veterinary oncology. It is even more
daunting to contemplate combination therapy. It is clear that this agent is
synergistic or supra-additive with platinum agents, anthracyclines and
alkylating drugs, however. There is limited experience with administration
of gemcitabine and cisplatin or carboplatin in patients with treatment-
refractory carcinomas.

Toxicities

Myelosuppression is dose-limiting. Nausea and vomiting are rare and

generally mild. Although rare, additional toxicities observed in people
include mucositis and alopecia, along with transient rises in transaminases.
Eighteen percent of 979 people treated with single-agent gemcitabine ex-
perienced transient influenza-like symptoms, and mild fever was observed in
37% of patients. Renal insufficiency of undetermined etiology was rarely
reported, and 20% of patients had mild peripheral edema in the absence of
cardiac, hepatic, or renal failure [65]. Although few adverse effects have been
reported in dogs, it is reasonable to assume that additional toxicities will be
added to the canine list as dose optimization is achieved.

644

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

Efficacy

Thus far, anecdotal responses of a partial nature have been the best

recorded for gemcitabine as a single agent. In most cases, disease stabi-
lization is achieved for variable periods of time. At the University of Illinois,
the authors have had brief complete responses in two dogs treated with
gemcitabine as third and fourth line rescue for high-grade refractory
lymphomas, which suggest the potential for addition of this agent into
combination protocols. Randomized control trials need to be designed to
evaluate this potential.

One case of biopsy confirmed that multicentric hepatocellular carcinoma

treated with gemcitabine as a single agent at relatively low doses had
apparent complete remission based on follow-up ultrasonography. This
remission was not biopsy confirmed and thus is suspect, however. Individual
cases have had prolonged survival over what might be predicted based on
historical reports of diseases such as pancreatic carcinoma and transitional
cell carcinoma.

Indications for use of gemcitabine in dogs would be expected to parallel

those in human oncology. Thus, randomized controlled trials of treatment
in combination protocols for advanced GI, genitourinary, and pulmonary
cancers seems the logical starting point for further clinical research.

Clinical applications in cats

Dosage

Most doses reported for gemcitabine use in cats are the radiosensitizing

doses.

Reported single agent doses range from 45 mg

/m

2

given as a 30-minute

saline infusion [55], to the current single agent dose in use at the University of
Illinois of 250 to 275 mg

/m

2

(Kitchell, unpublished data). The radiosensitizing

dose reported for cats ranges from 21 mg

/m

2

in conjunction with full course

radiotherapy [59], to 25 mg

/m

2

administered twice weekly in conjunction with

palliative radiation therapy of 6 Gy per fraction for six treatments [66].

Toxicities

Myelosuppression has been seen in cats treated with gemcitabine alone or

in combination with radiation therapy or carboplatinum.

Efficacy

Of five cats treated with single-agent gemcitabine at the University of

Illinois between 1996 and 1999, two had stable disease, two had progressive
disease, and one was nonevaluable. The dose range was 60 to 125 mg

/m

2

for

this group of cats, with one cat receiving a cumulative dose of 1907 mg
during the period of disease stabilization (Kitchell, unpublished data).

Ladue and Klein reported on a group of 10 cats with oral squamous cell

carcinoma treated with curative intent radiation therapy with gemcitabine

645

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

radiosensitization [59]. In this trial, radiation was delivered in Monday
through Friday fractions of 3 to 3.1 Gy per fraction. Gemcitabine was
administered at an average dose of 21 mg

/m

2

IV. Five cats required chemo-

therapy delay or dose reduction because of myelosuppression or unaccept-
able local tissue toxicity. Median local disease control was 3 months, with
a mean of 6 months. Six cats died of progressive local disease, while two died
of metastasis without local recurrence. Two cats were alive without evidence
of disease progression at the time of the report (1 and 32 months). Grades 2
to 3 local radiation toxicity was reported [59].

Eight cats with nonresectable squamous cell carcinoma of the oral cavity

were treated with palliative radiation therapy at 6 Gy fractions delivered
twice weekly for a total dose of 36 Gy. Low-dose gemcitabine (25 mg

/m

2

)

was administered twice weekly in conjunction with this megavoltage ir-
radiation. The mean number of doses of radiation was 4.9, and a mean of
five doses of gemcitabine was given. In this population of cats, two had
complete responses, three had partial responses, and three were non-
responsive. Median duration of remission was 42.5 days for the cats that
responded, and median survival time was 111.5 days (range 11 to 234 days).
These data suggest that short-term therapeutic benefit from radiation and
gemcitabine is possible in a palliative intent setting [66].

References

[1] Olivero VT. Pharmacology of the nitrosoureas: an overview. Cancer Treat Rep

1976;60:703–7.

[2] Tuvesson H, Gunnarsson PO, Seidegard J. Measurement and characterization of the

denitrosation of tauromustine and related nitrosoureas by glutathione transferases in liver
cytosol from various species. Carcinogenesis 1993;14:1143–7.

[3] Carter SK, Newman JW. Nitrosoureas: 1,3-bis(2-chloroethyl)–1-nitrosourea (NSC-409962;

BCNU) and 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (NSC-79037; CCNU)-clinical
brochure. Cancer Chemother Rep 1968;1:115–51.

[4] Schaeppi U, Fleischmann RW, Phelan RS, et al. CCNU 9NSC-79037: preclinical

toxicologic evaluation of a single intravenous infusion in dogs and monkeys. Cancer
Chemother Rep 1974;5:53–64.

[5] Abb J, Netzel B, Rodt HV, et al. Autologous bone marrow grafts in dogs treated with

lethal doses of 1(2-chloroethyl)-3-cyclohexyl-1-nitrosourea. Cancer Res 1978;38:2157–9.

[6] Henry MC, Davis RD, Schien PS. Hepatotoxicity of 1(2-chloroethyl)–3-cyclohexyl-1-

nitrosourea (CCNU) in dogs. Toxicol Appl Pharmacol 1973;25:410–7.

[7] Moore AS, London CA, Wood CA, et al. Lomustine (CCNU) for the treatment of relapsed

lymphoma in dogs. J Vet Intern Med 1999;13:395–8.

[8] Rassnick KM, Moore AS, Williams LE, et al. Treatment of canine mast cell tumors with

CCNU. J Vet Intern Med 1999;13:601–5.

[9] Kristal O, Rassnick KM, Gliatto JM, et al. Hepatotoxicity associated with CCNU

(lomustine) chemotherapy in dogs. Submitted.

[10] Fulton LM, Steinberg HS. Preliminary study of lomustine in the treatment of intracranial

masses in dogs following localization by imaging techniques. Semin Vet Med Surg (Small
Anim) 1990;5:241–5.

646

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

[11] Graham JC, Myers RK. Pilot study on the use of lomustine (CCNU) for the treatment of

cutaneous lymphoma in dogs [abstract]. J Vet Intern Medicine 1999;13:257.

[12] Rassnick KM, Geiger TL, Williams LE, et al. Phase I evaluation of CCNU (lomustine) in

tumor-bearing cats. J Vet Intern Med 2001;15:196–9.

[13] Weiss RB, Issell BF. The nitrosoureas: carmustine (BCNU) and lomustine (CCNU).

Cancer Treat Rev 1982;9:313–30.

[14] Fan TM, Kitchell BE, Dhaliwal RS, et al. Hematological toxicity and therapeutic efficacy

of lomustine in 20 tumor-bearing cats: a critical assessment of a practical dosing regimen.
J Am Anim Hosp Assoc 2002;38:357–63.

[15] Ikeda K, Inoue N, Frassica FJ, et al. Development of a canine chemotherapeutic model

with ifosfamide. Lab Anim Sci 1996;46:503–6.

[16] Barnett D. Preclinical toxicology of ifosfamide. Semin Oncol 1982;9(Suppl 1):8–13.
[17] Brock N, Pohl J, Stekar J, et al. Studies on the urotoxicity of oxazaphosphorine cytostatics

and its prevention-III. Profile of action of sodium 2-mercaptoethane sulfonate (mesna).
Eur J Cancer Clin Oncol 1982;18:1377–87.

[18] Rassnick KM, Frimberger AE, Wood CA, et al. Evaluation of ifosfamide for treatment of

various canine neoplasms. J Vet Intern Med 2000;14:271–6.

[19] White FR. Streptozotocin. Cancer Chemother Rep 1963;30:49–53.
[20] Tew KD, Colvin M, Chabner BA. Alkylating agents. In: Chabner BA, Longo DL,

editors. Cancer chemotherapy and biotherapy. Philadelphia: Lippincott-Raven; 1996.
p. 297–332.

[21] Stolinsky DC, Sadoff L, Braunwald J, et al. Streptozotocin in the treatment of cancer:

phase II study. Cancer 1972;30:61–6.

[22] Broder LE, Carter SK. Pancreatic islet cell carcinoma. II. Results of therapy with

streptozotocin in 52 patients. Ann Intern Med 1973;79:108–18.

[23] Schein PS, O’Connell MJ, Blom J, et al. Clinical antitumor activity and toxicity of

streptozotocin (NSC-85998). Cancer 1974;34:993–1000.

[24] Handelsman H, Broder LE, Slavik M, et al. Streptozotocin NSC-85998. Clinical brochure.

Investigational Drug Branch, Cancer Therapy Evaluation, Division of Cancer Treatment,
National Cancer Institute; 1974. p. 1–68.

[25] Levine BS, Henry MC, Port CD, et al. Toxicologic evaluation of streptozotocin (NSC

85998) in mice, dogs and monkeys. Drug Chem Toxicol 1980;3:201–12.

[26] Kaneko JJ, Mattheeuws D, Rottiers RP, et al. Renal function, insulin secretion, and

glucose tolerance in mild streptozotocin diabetes in the dog. Am J Vet Res 1978;39:
807–9.

[27] Gans JH, Cater MR. Hypercholesterolemia of streptozotocin-induced diabetes mellitus in

dogs. Life Sci 1971;10:301–8.

[28] Meyer DJ. Pancreatic islet cell carcinoma in a dog treated with streptozotocin. Am J Vet

Res 1976;37:1221–3.

[29] Meyer DJ. Temporary remission of hypoglycemia in a dog with an insulinoma after

treatment with streptozotocin. Am J Vet Res 1977;38:1201–4.

[30] Moore AS, Nelson RW, Henry CJ, et al. Streptozotocin for treatment of pancreatic islet

cell tumors in dogs: 17 cases (1989–1999). J Am Vet Med Assoc 2002;221:811–8.

[31] Moore AS, Rand WM, Berg J, et al. Evaluation of butorphanol and cyproheptadine for

prevention of cisplatin-induced vomiting in dogs. J Am Vet Med Assoc 1994;205:441–3.

[32] Burris HA, Moore MJ, Andersen J, et al. Improvements in survival and clinical benefit

with gemcitabine as first-line therapy for patients with advanced pancreas cancer:
a randomized trial. J Clin Oncol 1997;15:2403–13.

[33] Storniolo AM, Enas NH, Brown CA, et al. An investigational new drug treatment program

for patients with gemcitabine. Results for over 3000 patients with pancreatic carcinoma.
Cancer 1999;85:1261–8.

[34] Heinemann V, Xu YZ, Chubb S, et al. Cellular elimination of 29,29difluorodeoxycytidine

59-triphosphate: a mechanism of self-potentiation. Cancer Res 1992;52:533–9.

647

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

[35] Heinemann V, Hertel LW, Grindey GB, et al. Comparison of the cellular pharmacokinetics

and toxicity of 29,29-difluorodeoxycytidine and 1-beta-D-arabinofuranosylcytosine. Cancer
Res 1988;48:4024–31.

[36] Plunkett W, Huang P, Xu YZ, et al. Gemcitabine: metabolism, mechanisms of action, and

self-potentiation. Semin Oncol 1995;22:3–10.

[37] Xu YZ, Plunkett W. Modulation of deoxycytidylate deaminase in intact human leukemia

cells. Action of 29,29-difluorodeoxycytidine. Biochem Pharmacol 1992;44:1819–27.

[38] Galmarini CM, Mackey JR, Dumontet C. Nucleoside analogues: mechanisms of drug

resistance and reversal strategies. Leukemia 2001;15:875–90.

[39] Schiller JH, Harrington D, Belani CP, et al. The Eastern Cooperative Oncology Group.

Comparison of four chemotherapy regimens for advanced nonsmall cell lung cancer.
N Engl J Med 2002;346:92–8.

[40] Abbruzzese JL. New applications of gemcitabine and future directions in the management

of pancreatic cancer. Cancer 2002;95:941–5.

[41] Von der Maase H, Hansen SW, Roberts JT, et al. Gemcitabine and cisplatin versus

methotrexate, vinblastine, doxorubicin, and cisplatin in advanced or metastatic bladder
cancer: results of a large, randomized, multinational, multicenter, phase III study. J Clin
Oncol 2000;18:3068–77.

[42] Seidman AD. The evolving role of gemcitabine in the management of breast cancer.

Oncology 2001;60:189–98.

[43] Nabhan C, Krett N, Gandhi V, et al. Gemcitabine in hematologic malignancies. Curr Opin

Oncol 2001;13:514–21.

[44] Spath-Schwalbe E, Genvresse I, Koschuth A, et al. Phase II trial of gemcitabine in patients

with pretreated advanced soft tissue sarcomas. Anticancer Drugs2000;13(11):325–9.

[45] Merimsky O, Meller I, Flusser G, et al. Gemcitabine in soft tissue or bone sarcoma

resistant to standard chemotherapy: a phase II study. Cancer Chemother Pharmacol
2000;45:177–81.

[46] Plunkett W, Gandhi V. Nucleoside analogs: cellular pharmacology, mechanisms of action,

and strategies for combination therapy. In: Cheson B, Keating JM, Plunkett W, editors.
Nucleoside analogs in cancer therapy. New York: Marcel Dekker Inc.; 1997. p. 1–35.

[47] Millikan RE, Plunkett WK, Smith TL, et al. Gemcitabine modulation of alkylator therapy.

A phase I trial of escalating gemcitabine added to fixed doses of ifosfamide and
doxorubicin. Cancer 2001;92:194–9.

[48] Eisbruch A, Shewach DS, Bradford CR, et al. Radiation concurrent with gemcitabine for

locally advanced head and neck cancer: A phase I trial and intracellular drug incorporation
study. J Clin 2001;19(3):792–9.

[49] Lawrence TS, Eisbburch A, McGinn CJ, et al. Radiosensitization by gemcitabine.

Oncology 1999;13(Suppl 5):55–69.

[50] Doyle TH, Mornex F, McKenna WG. The clinical implications of gemcitabine

radiosensitization. Clin Cancer Res 2001;7:226–8.

[51] Shipley LA, Brown TJ, Cornpropst JD, et al. Metabolism and disposition of gemcitabine,

an oncolytic deoxycytidine analog, in mice, rats, and dogs. Drug Metab Dispos
1992;20:849–55.

[52] Esumi Y, Mitsugi K, Takao A, et al. Disposition of gemcitabine in rat and dog after single

and multiple dosings. Xenobiotica 1994;24:805–17.

[53] Cozzi PJ, Bajorin DF, Tong W, et al. Toxicology and pharmacokinetics of intravesical

gemcitabine: a preclinical study in dogs. Clin Cancer Res 1999;5:2629–37.

[54] Storniolo AM, Allerheiligen SR, Pearce HL. Preclinical, pharmacologic and phase I studies

of gemcitabine. Semin Oncol 1997;24(Suppl 7):2–7.

[55] Boyce KL, Kitchell BE. Gemcitabine single agent for the treatment of advanced cancer in

the dog and cat: a pilot study. Proc Vet Cancer Soc Ann Meeting 1998;18:24.

[56] Walter CU, Shaw NG, LaDue TA. Gemcitabine in the treatment of refractory or recurrent

canine lymphoma. Proc Vet Cancer Soc Ann Meeting 2001;21:46.

648

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

[57] Kisseberth WE, Kosarek CE, Couto CG. Phase I evaluation of gemcitabine (Gemzar) in

tumor-bearing dogs. Proc Vet Cancer Soc Ann Meeting 2002;22:7.

[58] Marconato L, Lindner DL, Suslak-Brown L, et al. A phase I clinical trial of high-dose

gemcitabine in 33 dogs with transitional cell carcinoma. Proc Vet Cancer Soc Ann Meeting
2002;22:9.

[59] LaDue TA, Klein MK. What is a radiosensitizer (?): update on gemcitabine for canine

sinonasal carcinoma and feline oral squamous cell carcinoma. Proc Am College of
Veterinary Radiologist Annual Scientific Meeting, 2002.

[60] Poplin E, Benson A III, Musanti R, et al. Pilot study of gemcitabine (10 mg

/m

2

per min)

and cisplatin. Cancer Chemother Pharmacol 2002;50:80–3.

[61] Kroep JR, Peters GJ, Van Moorsel CJ, et al. Gemcitabine – cisplatin: a schedule finding

study. Ann Oncol 1999;10:1503–10.

[62] Grunewald R, Abbruzzese JL, Tarassoff P, et al. Saturation of 29,29-difluorodeoxycytidine

59triphosphate accumulation by mononuclear cells during a phase I trial of gemcitabine.
Cancer Chemother Pharmacol 1991;27:258–62.

[63] Gandhi V, Plunkett W, Du M. Prolonged infusion of gemcitabine: clinical and

pharmacodynamic studies during a phase I trial in relapsed acute myelogenous leukemia.
J Clin Oncol 2002;20:665–73.

[64] Abbruzzese JL, Grunewald R, Weeks EA, et al. A phase I clinical, plasma and cellular

pharmacology study of gemcitabine. J Clin Oncol 1991;9:491–8.

[65] Aapro MS, Martin C, Hatty S. Gemcitabine—a safety review. Anticancer Drugs

1998;9:191–201.

[66] Jones PD, deLorimier LP, Kitchell BE, et al. Gemcitabine as a radiosensitizer for

nonresectable feline oral squamous cell carcinoma. J Am Anim Hosp Assoc, in press.

649

A.S. Moore, B.E. Kitchell

/ Vet Clin Small Anim 33 (2003) 629–649

background image

Mechanisms of anticancer drug resistance

Philip J. Bergman, DVM, MS, PhD

Donaldson-Atwood Cancer Clinic and Flaherty Comparative Oncology Laboratory,

The Animal Medical Center, 510 East 62nd Street, New York, NY 10021, USA

For veterinarians involved in the treatment of pets with cancer via

chemotherapeutics, drug resistance is a common phenomenon and the most
common cause of treatment failure. For example, many chemotherapeutic
agents are capable of inducing rapid remissions in dogs with lymphoma;
however, the ability to treat these patients effectively on relapse is routinely
significantly impaired [1–6]. This inability to treat patients effectively after
relapse is likely the result of a multitude of resistance factors, which have
been elucidated over the last three decades. Multiple mechanisms of re-
sistance that represent a level of redundancy for protection of the cell and
the organism are likely present in most normal cells. Unfortunately, many
cancers have derived methods of employing these resistance mechanisms for
their own protection.

Drug resistance is generally categorized into intrinsic and acquired

forms, with probable overlap in mechanisms across these two categories.
For example, mechanisms of acquired drug resistance would include re-
duced drug accumulation, increased repair, increased detoxification,
decreased apoptosis, and, lastly, alterations in drug targets, whereas cell
cycle–associated, cell adhesion–mediated, and other mechanisms would
represent intrinsic or de novo drug resistance mechanisms [7–15]. For
a complete list of mechanisms of classical acquired chemotherapy
resistance see Box 1.

In addition, resistance mechanisms can be categorized from organism

and cellular levels. Although there are many recognized processes re-
sponsible for overall resistance, there are many that we as clinicians can
easily circumvent, which include inappropriate underdosing of chemo-
therapeutics, improper scheduling of drugs, and inappropriate lengths of
time from diagnosis to the start of therapy. Others exist that we may not
presently have any control over, including poor absorption and erratic

Vet Clin Small Anim

33 (2003) 651–667

E-mail address:

philip.bergman@amcny.org

0195-5616

/03/$ - see front matter

Ó 2003, Elsevier Inc. All rights reserved.

doi:10.1016/S0195-5616(03)00004-4

background image

bioavailability of oral drugs, decreased drug activation, increased repair of
drug damage, increased drug extrusion, and poor penetration of the drug
into the tumor as a result of carrier-mediated processes. Over the past two
decades, bench research has found mechanisms to circumvent some of these
previously untouchable resistance mechanisms; in fact, some have become
clinically relevant resistance reversal strategies. This overview focuses on the
major known cellular and molecular mechanisms of resistance. Recom-
mendations for minimizing the development of anticancer drug resistance
include the following:

Treat animals as soon as reasonably possible after diagnosis is confirmed.
Use standardized and published chemotherapy protocols.
Adhere to the chemotherapy schedule whenever possible.

Box 1. Mechanisms of classical acquired chemotherapy
resistance

Reduced intracellular drug concentration
P-glycoprotein
Multidrug resistance–related protein
Lung resistance–related protein
Others

Drug target alterations
Topoisomerase II
Topoisomerase I

Drug detoxification
Glutathione
Glutathione-s-transferase
Dihydrofolate reductase
Thymidylate synthase
Aldehyde dehydrogenase
Others

Increased DNA damage repair
O6-alkylguanine-DNA alkyltransferase
Others (eg, mismatch repair, XPF and ERCC1)

Apoptosis/programmed cell death modulation
Bcl-2 family
p53, p21, p27, and others
Attenuated death receptor signaling (eg, tumor necrosis

factor-related apoptosis-inducing ligand [TRAIL])

Caspase based (eg, Caspase-8)
Others (eg, NF-jB, DNA repair)

652

P.J. Bergman

/ Vet Clin Small Anim 33 (2003) 651–667

background image

Use the recommended doses of chemotherapy in the protocol.
Use of proper mixing, handling, administration, and disposal techniques

is of paramount importance.

Whenever possible, do not use prednisone before a diagnosis is con-

firmed or before induction chemotherapy is instituted for dogs with
lymphoma.

Treat patients as soon as reasonably possible with the next appropriate

chemotherapy protocol once relapse has occurred.

P-glycoprotein

The most researched form of cellular drug resistance is the overexpression

of a plasma membrane protein given the name P-glycoprotein (Pgp
[P

¼ permeability]) or P-170 (protein molecular weight of 160–180 kd)

[16,17]. Resistance related to Pgp overexpression is one of the major factors
leading to the multidrug resistance (MDR) phenotype [9,18–20]. This is
the phenomenon whereby cancer cells become simultaneously resistant to
a variety of different drugs commonly used in veterinary medicine (eg,
doxorubicin, vincristine, actinomycin-

D

, mitoxantrone and others). Such

drugs are typically developed from natural sources and are hydrophobic
but otherwise have few similarities in their mechanisms of action or their
chemical structures [9]. Chemotherapeutics exhibiting cross-resistance in
MDR because of Pgp include the following:

Vincristine
Vinblastine
Doxorubicin
Daunorubicin
Mitoxantrone
Actinomycin-

D

Etoposide
Mitomycin-C
Taxol

/taxanes

Steroids (corticosteroids and sex steroids)
Others

Not only do cancer cells develop resistance to these chemotherapeutics,

but these same compounds induce the formation of the MDR phenotype.
Pgp genes have been found in bacteria, viruses, plants, insects, nematodes,
and mammals; such extreme evolutionary conservation signifies the extreme
level of importance of this gene and its protein.

Pgp acts as a plasma membrane drug efflux pump that actively extrudes

drugs from cancer cells, thereby limiting the cytotoxicity of the drug at its
cellular site of action. Pgp is an adenosine triphosphate (ATP)–dependent
efflux pump that is a member of a family of ATP-binding cassette (ABC)
transporters [16]. Almost 50 ABC transporters have been discovered in

653

P.J. Bergman

/ Vet Clin Small Anim 33 (2003) 651–667

background image

humans to date. The normal function of Pgp is not completely known, but it
has been found to be expressed in normal human or canine adrenal gland,
kidney, liver, intestine, placenta, blood–brain barrier, lung, peripheral
blood

/bone marrow cells, and multiple fetal cell lines [9,16–18,20,21]. It is

therefore hypothesized that Pgp normally functions as a drug efflux pump.
For example, Pgp is located on the luminal surface of the endothelial cells in
the brain, thereby preventing cytotoxins from penetrating the brain across
the endothelium [22]. Mice that have been genetically modified to not
express Pgp (MDR ‘‘knockouts’’) are normally deficient in Pgp or are given
Pgp inhibitors and are exquisitely sensitive to Pgp substrates, such as
ivermectin [23–28]. Interestingly, these mice and some Collies show similar
neurologic side effects on ivermectin administration [29–31]. Recent studies
have shown that a subpopulation of Collies sensitive to ivermectin ad-
ministration has a mutant Pgp and that 35% of Collies are homozygous for
the mutant allele of Pgp, which is consistent with previous reports sug-
gesting that 30% to 40% of Collies have sensitivity to ivermectin [30,32,33].
Studies are presently ongoing within this author’s laboratory to investigate
the immunohistochemical expression of Pgp in Collies compared with other
canine breeds.

In addition to numerous chemotherapy agents and ivermectin, there are

many other Pgp substrates that are commonly used therapeutic agents.
These include but are not limited to ondansetron, loperamide, itraconazole,
ketoconazole, cyclosporine, rifampin, phenobarbital, digoxin, doxycycline,
omeprazole, and many types of steroids, antibiotics, and antihistamines
[34–40]. Interestingly, Collies have been previously reported to be extremely
sensitive neurologically to loperamide, and this may be explained by loper-
amide being a known Pgp substrate [41,42]. Unfortunately, many of the
aforementioned agents are substrates as well as inducers of Pgp expression.
Therefore, the use of many of these agents may be inducing a drug-resistant
phenotype in our clinical patients, and additional research in this area is
strongly encouraged. Similarly, concomitant use of these agents at the time
of chemotherapy administration may change the pharmacokinetic and
toxicity profile of chemotherapy agents or other Pgp substrates.

Many tumors derived from the normal anatomic locations that express

large amounts of Pgp are commonly intrinsically resistant to chemother-
apeutic agents because of increased Pgp expression [43–45]. The clinical
importance of MDR is demonstrated in human oncology by the fact that
increasing levels of Pgp expression positively correlate with a lack of re-
sponse or remission with appropriate forms of chemotherapy for a variety of
human malignancies [16,46].

A potential explanation for why dogs with lymphoma treated with

the chemotherapy protocol containing cyclophosphamide, vincristine, and
prednisone (COP) have a shorter remission than those treated with doxo-
rubicin alone may lie within the realm of MDR [3,47,48]. Two of the
drugs in the COP protocol are known to induce the MDR phenotype,

654

P.J. Bergman

/ Vet Clin Small Anim 33 (2003) 651–667

background image

whereas doxorubicin induces the MDR phenotype in single fashion.
Similarly, the use of corticosteroids before the initiation of chemotherapy
for canine lymphoma is thought to be a negative prognostic factor; however,
this is controversial [49]. This is likely related to the induction of Pgp
expression via prednisone, because steroids are well-known potent inducers
of Pgp expression and activity [50–53].

Three studies in veterinary medicine published in 1995 and 1996 dem-

onstrated the importance of Pgp in dogs with lymphoma. The first study
used a Western blotting anti-Pgp methodology in membrane preparations
of lymphoma cells from 31 dogs with lymphoma [54]. These investigators
found positive expression of Pgp in 1 of 31 dogs and 3 of 8 dogs before the
initiation of chemotherapy and once resistant to chemotherapy, respectively.
Unfortunately, Western blotting methodologies are relatively insensitive
and laborious, and they generally require fresh cells. Similarly, Western
blotting probes for Pgp expression in the entire sample and some Pgp
antibodies may label Pgp in lymphoma cells as well as incorrectly label other
noncancerous tissues. For this reason and various others, a consensus
workshop on determination of Pgp expression in human cancers has
previously recommended the use of in situ technologies, such as im-
munohistochemistry, to allow for specific Pgp expression in the cancerous
tissue of choice [55].

In the second study, which was completed by this author and his

colleagues, the prevalence of positive staining of Pgp via immunohisto-
chemistry was ascertained in 58 dogs with lymphoma [56]. Dogs had
samples evaluated before the initiation of chemotherapy, at time of relapse,
at the time of necropsy, and at all three times in some dogs. Consistent with
previous findings in Pgp research in human oncology, Pgp expression levels
significantly increased at relapse and necropsy when compared with levels at
the initiation of chemotherapy. This study also demonstrated that the level
of Pgp staining before initiation of chemotherapy was inversely correlated to
both remission and survival times, whereas the level of Pgp staining at
relapse was inversely correlated to the time from relapse to death. Therefore,
those patients with high pretreatment levels of Pgp had significantly lower
remission times and survival times, and those patients with high Pgp levels
at relapse had significantly decreased times from relapse to death. An ad-
ditional striking observation was that Pgp staining at the initiation of
chemotherapy was the most prognostic factor of any of those examined in
the study (including stage, substage, presence of hypercalcemia, age, weight,
and gender).

In the third study, expression of Pgp was again ascertained by im-

munohistochemistry, and pretreatment Pgp expression was found to be an
independent negative predictor of overall survival [57]. Additionally, Pgp
expression was greater after relapse when compared with pretreatment
samples. More recent studies in the in vitro setting show that canine cancer
cell lines can be induced to overexpress Pgp and be modulated by known

655

P.J. Bergman

/ Vet Clin Small Anim 33 (2003) 651–667

background image

Pgp inhibitors no differently than is seen in human in vitro systems [58,59].
Taken in concert, it seems that canine Pgp behaves similarly to human Pgp
and that Pgp expression determination in a prospective fashion for dogs
with lymphoma represents a viable pretreatment and intratreatment diag-
nostic tool. In addition, these works suggest that canine lymphoma
represents an excellent comparative model for human Pgp research. Future
studies in canine drug resistance incorporating determination of Pgp expres-
sion and the other aforementioned mechanisms in Box 1 performed in
a prospective fashion are hereby encouraged for lymphoma and other
neoplasms. Unfortunately, few data have been published concerning feline
Pgp; one study using a feline lymphoma cell line (FT-1) and its doxorubicin-
resistant subline (FT-1

/ADM) has shown upregulation of Pgp in FT-

1

/ADM with an MDR phenotype [60]. Interestingly, polymerase chain

reaction (PCR) cDNA amplification of the feline Pgp showed 90% se-
quence identity to human Pgp, thereby continuing the theme of high
sequence identity across species for Pgp.

There are many methods for attempting reversal of Pgp-associated

MDR; however, to date, few have shown any significant clinical benefit
[16,61–63]. There are many compounds, such as verapamil, cyclosporine,
tamoxifen, and others, that competitively bind to Pgp to inhibit its efflux
actions [64]. Drugs known to modulate Pgp activity and

/or expression are

seen in Box 2.

Box 2. Drugs known to modulate P-glycoprotein
activity or expression

Verapamil
Quinine
Cyclosporine
Reserpine
Chloroquine
Trifluoperazine
Tamoxifen
Various surfactants (eg, Cremophor-EL)
Progesterone and other sex steroids
Corticosteroids
Loperamide
Ondansetron
Doxycycline
Itraconazole/ketoconazole
Digoxin
Omeprazole
Others (some antibiotics and antihistamines)

656

P.J. Bergman

/ Vet Clin Small Anim 33 (2003) 651–667

background image

A phase I trial of high-dose tamoxifen and chemotherapy in normal and

tumor-bearing dogs was published by the North Carolina State University
group and showed that plasma concentrations capable of reversing Pgp
could be safely achieved in dogs with mild to moderate toxicity [65].
Unfortunately, many of these compounds have either poor efficacy (Pgp
substrates themselves) or severe unexpected toxicities at the levels necessary
for Pgp reversal; therefore, the search for less toxic resistance reversal
strategies continues [7]. Pgp inhibitors that are not Pgp substrates but retain
significant sensitivity and specificity for Pgp, such as XR-9576, OC-1440935,
LY335979, and GF-120918, hold great promise and are presently being
evaluated in human clinical trials [16,66].

The dose-response curves of most cancer cells are extremely steep; thus,

one way to circumvent resistance is to use higher and higher doses of drugs.
Significant toxicity, especially myelosuppression, is usually the result,
however. Similarly, although it is common for cancer cells to develop an
MDR phenotype, normal tissues continue their sensitivity to chemotherapy.
Because the bone marrow is invariably the dose-limiting toxicity for most
chemotherapeutics, investigators are transferring the genes that encode
Pgp (and other drug resistance genes) to bone marrow cells to confer
myeloprotection [67]. Pilot studies by this author and his colleagues at
Memorial Sloan-Kettering Cancer Center have shown that normal canine
hematopoietic stem cells (HSCs) can undergo drug resistance gene transfer
with concomitant in vitro myeloprotection to various chemotherapy agents
(unpublished observations). Strategies by other human and veterinary in-
vestigators to confer myeloprotection include the use of autologous HSC
infusion with high-dose chemotherapy [68,69].

Multidrug resistance–associated protein

Multiple studies have shown that when Pgp is not present, an MDR

phenotype is still possible; therefore, other resistance mechanisms are likely
at work in MDR. The second most frequently examined form of drug
resistance is that involving multidrug resistance–related protein (MRP)
[70–74]. MRP is a 190-kd protein that is similar in structure to Pgp.
Interestingly, MRP localizes to the cytoplasm in most normal tissues,
whereas plasma membrane localization is more common in neoplastic
tissues, suggesting an excretory function in the cancer cell. The normal
function of MRP has been identified as the carrier of glutathione S-
conjugates and bilirubin glucuronides in the cell and then excretion of these
conjugates to outside the cell [73]. Normal MRP seems to be most strongly
expressed in various epithelia, macrophages, the heart, and tissues with an
excretory function, such as the liver, kidney, and others, similar to the
normal expression of Pgp [72]. Therefore, MRP has been referred to by
many as the ‘‘toxic waste manager’’ of the cell for those toxins conjugated

657

P.J. Bergman

/ Vet Clin Small Anim 33 (2003) 651–667

background image

by the glutathione system, which, unfortunately, is a common detoxification
system for many of the frequently used chemotherapy agents in veterinary
medicine (see section on glutathione). As a measure of MRP’s clinical
importance, rats and human beings with mutations in MRP have chronic
conjugated hyperbilirubinemia (Dubin-Johnson syndrome). Eight addi-
tional MRP-like proteins have been recently identified and are likely addi-
tional drug resistance mechanisms worthy of future investigation [74,75].

No studies have been published to date on MRP in veterinary oncology;

however, the prognostic significance of MRP in human oncology is strong.
Many tumor cell lines coexpress Pgp and MRP, whereas others with an
MDR phenotype without Pgp expression can have strong MRP expression.
Acute leukemias, transitional cell carcinomas, and squamous cell carcino-
mas seem to be human neoplasms with the strongest MRP expression,
suggesting normal cells that undergo malignant transformation have
retained their MRP expression [76,77].

Lung resistance–related protein

Lung resistance–related protein (LRP) is a 110-kd protein originally

isolated from a human lung cancer cell line that is a distant relative of Pgp
and MRP [78]. The normal function of this protein is that of a major
‘‘vault’’ ribonucleoprotein, which regulates transport of substances between
the nucleus and cytoplasm [79]. This suggests LRP may be involved in the
transport of cytotoxic agents by redistributing drugs away from intracellular
targets. LRP has been detected in numerous cancer cell lines and clinical
specimens of many tumors, including acute myeloid leukemia, ovarian car-
cinoma, myeloma, and fibrosarcoma [80]. No studies have been pub-
lished to date on the detection of LRP in veterinary patients, and no studies
have been published on reversal strategies to LRP-associated drug resistance.

Topoisomerases

DNA topoisomerases (Topo I and II) are essential enzymes that catalyze

various conformational changes in DNA necessary for normal steps in
DNA metabolism [81]. Topo II forms a transient DNA break followed by
DNA strand passage and then religation of the DNA. Inhibitors of Topo II
prevent the religation process by freezing the ‘‘cleavable complex’’ formed
between the enzyme and DNA, leading to cell death [82]. Drugs that tar-
get Topo II include actinomycin-

D

, epipodophyllotoxins (eg, etoposide),

anthracenediones (eg, mitoxantrone), anthracyclines (eg, doxorubicin,
daunorubicin), and other experimental agents. Chemotherapeutics involved
with Topo II–mediated MDR include all those in Pgp-associated MDR,
except the vinca alkaloids and taxanes. In contrast to Pgp-associated MDR,
chemoresistance associated with Topo II is the result of reduced expression
or activity of the enzyme [83,84].

658

P.J. Bergman

/ Vet Clin Small Anim 33 (2003) 651–667

background image

Glutathione system

The major role of glutathione S-transferase (GST) and glutathione

(GSH) is the detoxification of cytotoxic drugs [85]. Many cancer cell lines
and clinical cancer specimens have increased production of GST and GSH
when compared with normal tissues [86,87]. It seems that doxorubicin,
chlorambucil, cyclophosphamide, melphalan, nitrosoureas, cisplatin, and
others are involved with GST

/GSH-mediated MDR [88,89]. Attempts at

reversal of GST

/GSH-mediated MDR with ethacrynic acid as a GST in-

hibitor or buthionine sulfoximine as a GSH synthesis inhibitor have been
met to date with poor efficacy and significant toxicity in human clinical trials
[87]. Two reports show the potential importance of the GSH system in
canine osteosarcoma and mammary tumor drug resistance [90,91].

Dihydrofolate reductase and thymidylate synthase

Amplification of the dihydrofolate reductase (DHFR) gene leads to

subsequent overexpression of DHFR. DHFR is an enzyme responsible for
catalyzation of the reduction of dihydrofolate to tetrahydrofolate. DHFR
overexpression leads to a decreased concentration of the classic antifolate
chemotherapeutic methotrexate and represents one of the earliest forms of
drug resistance elucidated to date and the major cause of methotrexate
resistance [92]. Although other mechanisms of methotrexate resistance exist
(ie, antifolate transport modulation via reduced folate carriers, mutation of
target enzyme, polyglutamation modulation, thymidylate synthase), DHFR
overamplification remains the most important methodology of methotrexate
resistance investigated to date [93–96]. DHFR overamplification-based
methotrexate resistance may be superseded by massive doses of methotrex-
ate followed by leucovorin rescue. In addition, newer methotrexate
analogues, such as trimetrexate and edatrexate, may be useful, because
these compounds are believed to be more selectively inhibitory of DHFR
[97,98]. DHFR-based chemoresistance may have a limited role in veterinary
oncology, because methotrexate is not a commonly used veterinary che-
motherapy agent outside of some multiagent lymphoma protocols, and
there are no studies reporting the efficacy of single-agent methotrexate
against lymphoma to date [99].

Others

Many antineoplastic drugs interact with DNA at the O

6

-position of

guanine to form extremely potent cytotoxic DNA adducts [100,101].
O

6

-alkylguanine-DNA alkyltransferase (O

6

-AT) is a DNA repair enzyme

encoded by the gene MGMT that has been recently implicated as
a mechanism of chemoresistance [102]. High levels of O

6

-AT have been

659

P.J. Bergman

/ Vet Clin Small Anim 33 (2003) 651–667

background image

associated with resistance to dacarbazine, streptozotocin, and nitrosoureas
(eg, CCNU, BCNU), presumably because this enzyme repairs the
chemotherapy-induced DNA damage [100,103]. A relatively new ‘‘methyl-
ating’’ chemotherapy agent, temozolomide, primarily induces O

6

-methyl-

guanine adducts, and high levels of O

6

-AT strongly predict temozolomide

resistance [104]. Phase II human clinical trials are ongoing to evaluate the
use of the O

6

-AT inhibitor O

6

-benzylguanine in concert with chemotherapy

agents that promote O

6

-guanine DNA adducts [100]. No veterinary studies

have been published to date on O

6

-AT–based chemoresistance.

The induction of DNA interstrand (between complementary strands of

DNA) and intrastrand crosslinks seems to be an important mechanism of
cytotoxicity for clinically relevant chemotherapy agents in the platinum class
and others [105]. Although reduced drug uptake and increased inactivation
by glutathione appear to be important mechanisms of platinum class
chemotherapy agents, it seems that DNA repair mechanisms, such as
overexpression of XPF and ERCC1 proteins, are of even greater importance
[106,107]. Interestingly, human cells derived from patients with xeroderma
pigmentosum are extremely sensitive to various chemotherapy agents,
radiation, and ultraviolet (UV) light, because these cells are nucleotide
excision repair deficient [108]. The extreme sensitivity of these cells and
others like them is a result of myriad mutations in DNA repair genes,
including mismatch repair genes (eg, hMSH2, hMLH1); unfortunately,
cancer cells upregulate these same but nonmutated genes to confer che-
moresistance [106–110]. This has widespread implications for chemother-
apy agents that act on DNA and represents yet another mechanism for
cancer cells to escape the effects of various chemotherapy agents. No
veterinary studies have been published to date on DNA-repair–based
chemoresistance.

Aldehyde dehydrogenase (AD) is an additional mechanism of cellular

drug resistance to chemotherapy agents [111]. AD-based resistance seems
to be important for cyclophosphamide resistance in a variety of human
malignancies, including breast cancer, medulloblastomas, and leukemias
[112–114]. This method of chemoresistance is particularly intriguing,
because cyclophosphamide is a commonly used chemotherapy agent and
is not a Pgp substrate [16]. No veterinary studies have been published to
date on AD-based chemoresistance.

Apoptosis resistance–related multidrug resistance

Apoptosis, or programmed cell death, is an internally programmed

mechanism of cell death. Apoptosis is morphologically and biochemically
distinct from necrosis and permits noninflammatory single cell deletion
[115]. Most forms of chemotherapy and radiation kill cancer cells by in-
ducing them to undergo apoptosis [116,117]. Many normal endogenous

660

P.J. Bergman

/ Vet Clin Small Anim 33 (2003) 651–667

background image

activators and suppressors of apoptosis have recently been elucidated [13].
Unfortunately, cancer cells have developed mechanisms of upregulating or
activating these suppressors of apoptosis. Although much is left to be
learned about apoptosis and its relation to cancer biology, it seems that
apoptosis-resistant cancer cells are resistant to even the highest doses
possible of chemotherapeutics and radiation [8,118–121]. Therefore, based
on the fact that such apoptosis regulators lie profoundly downstream from
our therapeutic agents, a greater understanding of how apoptosis affects
resistance and cancer therapy in general is urgently needed. Such an
understanding should open new and likely extremely clinically relevant
therapeutic modalities for cancer as well as autoimmune, infectious, and
degenerative diseases.

Relatively little has been published to date on apoptosis and cancer in

veterinary medicine. The University of California–Davis group has found
that apoptotic and proliferation indexes as well as the proliferation

/

apoptotic ratio were predictive of relapse-free interval in dogs with lym-
phoma [122]. The Purdue University group has shown that urinary bladder
transitional cell carcinoma diagnosed in dogs that experience an antitumor
response to piroxicam was strongly associated with induction of apoptosis
and reduction in urine basic fibroblastic growth factor concentration
[123,124]. Other studies published to date have involved in vitro apoptosis
investigations using cancer cell lines from veterinary patients, and we ea-
gerly await additional in vivo investigations [125–130].

Summary

Chemotherapy agents are extremely important in the treatment of liquid

malignancies, such as lymphoma, myeloma, and chronic lymphocytic
leukemia. In addition, chemotherapy agents have proven effective in the
adjuvant treatment of solid tumors, such as osteosarcoma, hemangiosar-
coma, transitional cell carcinoma, and others. Unfortunately, chemotherapy
resistance in these situations is the most significant cause of treatment
failure. Therefore, the ability to predict, treat, or circumvent resistance is
extremely likely to improve clinical outcomes. This article has reviewed the
most widely investigated forms of chemotherapy resistance, such as reduced
drug accumulation, increased DNA damage repair, decreased apoptosis,
and others; however, new mechanisms are being found at an alarming pace.
In addition, investigations to date have routinely centered on single-cell
mechanisms of drug resistance, and cancer is truly a three-dimensional
disease. The elucidation of mechanisms surrounding (1) how tumors interact
with their normal microenvironment, (2) how tumors interact in a three-
dimensional environment, and (3) a better understanding of basic tumor
physiology and biology may supersede in importance those previously elu-
cidated single-cell mechanisms of chemoresistance.

661

P.J. Bergman

/ Vet Clin Small Anim 33 (2003) 651–667

background image

References

[1] Calvert CA, Leifer CE. Doxorubicin for treatment of canine lymphosarcoma after

development of resistance to combination chemotherapy. JAVMA 1981;179:1011–2.

[2] Greenlee PG, Filippa DA, Quimby FW, et al. Lymphomas in dogs: a morphologic,

immunologic, and clinical study. Cancer 1990;66:480–90.

[3] Postorino NC, Susaneck SJ, Withrow SJ, et al. Single agent therapy with adriamycin for

canine lymphosarcoma. J Am Anim Hosp Assoc 1989;25:221–5.

[4] Teske E. Canine malignant lymphoma: a review and comparison with human non-

Hodgkin’s lymphoma. Vet Q 1994;16:209–19.

[5] Van Vechten M, Helfand SC, Jeglum KA. Treatment of relapsed canine lymphoma with

doxorubicin and dacarbazine. J Vet Intern Med 1990;4:187–91.

[6] Weller RE, Theilen GH, Madewell BR. Chemotherapeutic responses in dogs with

lymphosarcoma and hypercalcemia. JAVMA 1982;181:891–3.

[7] Bergman PJ, Gravitt KR, O’Brian CA. An N-myristoylated protein kinase C-alpha

pseudosubstrate peptide that functions as a multidrug resistance reversal agent in human
breast cancer cells is not a P-glycoprotein substrate. Cancer Chemother Pharmacol
1997;40:453–6.

[8] Bergman PJ, Harris D. Radioresistance, chemoresistance, and apoptosis resistance. The

past, present, and future. Vet Clin North Am Small Anim Pract 1997;27:47–57.

[9] Broxterman HJ, Giaccone G, Lankelma J. Multidrug resistance proteins and other drug

transport-related resistance to natural product agents. Curr Opin Oncol 1995;7:532–40.

[10] Cabral F. Factors determining cellular mechanisms of resistance to antimitotic drugs.

Drug Resist Update 2001;4:3–8.

[11] Ferreira CG, Tolis C. Giaccone G. p53 and chemosensitivity. Ann Oncol 1999;10:

1011–21.

[12] Gottesman MM. Mechanisms of cancer drug resistance. Annu Rev Med 2002;53:615–27.
[13] Johnstone RW, Ruefli AA, Lowe SW. Apoptosis: a link between cancer genetics and

chemotherapy. Cell 2002;108:153–64.

[14] Schmitz JC, Liu J, Lin X, et al. Translational regulation as a novel mechanism for the

development of cellular drug resistance. Cancer Metastasis Rev 2001;20:33–41.

[15] Shah MA, Schwartz GK. Cell cycle-mediated drug resistance: an emerging concept in

cancer therapy. Clin Cancer Res 2001;7:2168–81.

[16] Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP-

dependent transporters. Nat Rev Cancer 2002;2:48–58.

[17] Tan B, Piwnica-Worms D, Ratner L. Multidrug resistance transporters and modulation.

Curr Opin Oncol 2000;12:450–8.

[18] Lehne G. P-glycoprotein as a drug target in the treatment of multidrug resistant cancer.

Curr Drug Targets 2000;1:85–99.

[19] Marie JP. Drug resistance in hematologic malignancies. Curr Opin Oncol 2001;13:463–9.
[20] Schneider E, Paul D, Ivy P, et al. Multidrug resistance. Cancer Chemother Biol Response

Modif 1999;18:152–77.

[21] Ginn PE. Immunohistochemical detection of P-glycoprotein in formalin-fixed and

paraffin-embedded normal and neoplastic canine tissues. Vet Pathol 1996;33:533–41.

[22] Bart J, Groen HJ, Hendrikse NH, et al. The blood-brain barrier and oncology: new

insights into function and modulation. Cancer Treat Rev 2000;26:449–62.

[23] Didier AD, Loor F. Decreased biotolerability for ivermectin and cyclosporin A in mice

exposed to potent P-glycoprotein inhibitors. Int J Cancer 1995;63:263–7.

[24] Kwei GY, Alvaro RF, Chen Q, et al. Disposition of ivermectin and cyclosporin A in CF-1

mice deficient in mdr1a P-glycoprotein. Drug Metab Dispos 1999;27:581–7.

[25] Lankas GR, Cartwright ME, Umbenhauer D. P-glycoprotein deficiency in a subpop-

ulation of CF-1 mice enhances avermectin-induced neurotoxicity. Toxicol Appl Phar-
macol 1997;143:357–65.

662

P.J. Bergman

/ Vet Clin Small Anim 33 (2003) 651–667

background image

[26] Schinkel AH, Smit JJ, van Tellingen O, et al. Disruption of the mouse mdr1a P-

glycoprotein gene leads to a deficiency in the blood-brain barrier and to increased
sensitivity to drugs. Cell 1994;77:491–502.

[27] Schinkel AH, Wagenaar E, Mol CA, et al. P-glycoprotein in the blood-brain barrier of

mice influences the brain penetration and pharmacological activity of many drugs. J Clin
Invest 1996;97:2517–24.

[28] Schinkel AH, Wagenaar E, van Deemter L, et al. Absence of the mdr1a P-glycoprotein in

mice affects tissue distribution and pharmacokinetics of dexamethasone digoxin and
cyclosporin A. J Clin Invest 1995;96:1698–705.

[29] Paul AJ, Tranquilli WJ, Seward RL, et al. Clinical observations in collies given ivermectin

orally. Am J Vet Res 1987;48:684–5.

[30] Ryan WG, Jones PH. Ivermectin use in collie dogs. Vet Rec 1989;125:425.
[31] Tranquilli WJ, Paul AJ, Seward RL, et al. Response to physostigmine adminis-

tration in collie dogs exhibiting ivermectin toxicosis. J Vet Pharmacol Ther 1987;10:
96–100.

[32] Mealey KL, Bentjen SA, Gay JM, et al. Ivermectin sensitivity in collies is associated with

a deletion mutation of the mdr1 gene. Pharmacogenetics 2001;11:727–33.

[33] Mealey KL, Bentjen SA, Waiting DK. Frequency of the mutant MDR1 allele associated

with ivermectin sensitivity in a sample population of collies from the northwestern United
States. Am J Vet Res 2002;63:479–81.

[34] Chishty M, Reichel A, Siva J, et al. Affinity for the P-glycoprotein efflux pump at the

blood-brain barrier may explain the lack of CNS side-effects of modern antihistamines.
J Drug Target 2001;9:223–8.

[35] Hochman JH, Yamazaki M, Ohe T, et al. Evaluation of drug interactions with P-

glycoprotein in drug discovery: in vitro assessment of the potential for drug-drug
interactions with P-glycoprotein. Curr Drug Metab 2002;3:257–73.

[36] Matheny CJ, Lamb MW, Brouwer KR, et al. Pharmacokinetic and pharmacodynamic

implications of P-glycoprotein modulation. Pharmacotherapy 2001;21:778–96.

[37] Mealey KL, Barhoumi R, Burghardt RC, et al. Doxycycline induces expression of P

glycoprotein in MCF-7 breast carcinoma cells. Antimicrob Agents Chemother 2002;
46:755–61.

[38] Pauli-Magnus C, Rekersbrink S, Klotz U, et al. Interaction of omeprazole, lansoprazole

and pantoprazole with P-glycoprotein. Naunyn Schmiedebergs Arch Pharmacol 2001;
364:551–7.

[39] Pea F, Furlanut M. Pharmacokinetic aspects of treating infections in the intensive care

unit: focus on drug interactions. Clin Pharmacokinet 2001;40:833–68.

[40] Wang EJ, Lew K, Casciano CN, et al. Interaction of common azole antifungals with P

glycoprotein. Antimicrob Agents Chemother 2002;46:160–5.

[41] Hugnet C, Cadore JL, Buronfosse F, et al. Loperamide poisoning in the dog. Vet Hum

Toxicol 1996;38:31–3.

[42] Wandel C, Kim R, Wood M, et al. Interaction of morphine, fentanyl, sufentanil,

alfentanil, and loperamide with the efflux drug transporter P-glycoprotein. Anesthesiology
2002;96:913–20.

[43] Ban T. Pleiotropic, multidrug-resistant phenotype and P-glycoprotein: a review. Chemo-

therapy 1992;38:191–6.

[44] Chenivesse X, Franco D, Bre´chot C. MDR1 (multidrug resistance) gene expression in

human primary liver cancer and cirrhosis. J Hepatol 1993;18:168–72.

[45] Kim WJ, Kakehi Y, Kinoshita H, et al. Expression patterns of multidrug-resistance

(MDR1), multidrug resistance-associated protein (MRP), glutathione-S-transferase-p
(GST-p) and DNA topoisomerase II (Topo II) genes in renal cell carcinomas and normal
kidney. J Urol 1996;156:506–11.

[46] Sikic BI. Modulation of multidrug resistance: at the threshold. J Clin Oncol 1993;11:

1629–35.

663

P.J. Bergman

/ Vet Clin Small Anim 33 (2003) 651–667

background image

[47] Bergman PJ, Ogilvie GK. Drug resistance and cancer therapy. Compend Contin Educ

Pract Vet 1995;17:549–56.

[48] Cotter SM. Treatment of lymphoma and leukemia with cyclophosphamide, vincristine,

and prednisone. I: treatment of dogs. J Am Anim Hosp Assoc 1983;19:159–65.

[49] Price GS, Page RL, Fischer BM, et al. Efficacy and toxicity of doxorubicin

/cyclo-

phosphamide maintenance therapy in dogs with multicentric lymphosarcoma. J Vet
Intern Med 1991;5:259–62.

[50] Altuvia S, Stein WD, Goldenberg S, et al. Targeted disruption of the mouse mdr1b

gene reveals that steroid hormones enhance mdr gene expression. J Biol Chem 1993;268:
27127–32.

[51] Becker KF, Allmeier H, Ho¨llt V. New mechanisms of hormone secretion: MDR-like gene

products as extrusion pumps for hormones. Horm Metab Res 1992;24:210–3.

[52] Kerb R, Aynacioglu AS, Brockmoller J, et al. The predictive value of MDR1, CYP2C9,

and CYP2C19 polymorphisms for phenytoin plasma levels. Pharmacogenom J 2001;
1:204–10.

[53] Pagnini U, Florio S, Lombardi P, et al. Modulation of anthracycline activity in canine

mammary tumour cells in vitro by medroxyprogesterone acetate. Res Vet Sci 2000;69:
255–62.

[54] Moore AS, Leveille CR, Reimann KA, et al. The expression of P-glycoprotein in canine

lymphoma and its association with multidrug resistance. Cancer Invest 1995;13:475–9.

[55] Beck WT, Grogan TM, Willman CL, et al. Methods to detect P-glycoprotein-associated

multidrug resistance in patients’ tumors: consensus recommendations. Cancer Res
1996;56:3010–20.

[56] Bergman PJ, Ogilvie GK, Powers BE. Monoclonal antibody C219 immunohistochemistry

against P-glycoprotein: sequential analysis and predictive ability in dogs with lymphoma.
J Vet Intern Med 1996;10:354–9.

[57] Lee JJ, Hughes CS, Fine RL, et al. P-glycoprotein expression in canine lymphoma—a

relevant, intermediate model of multidrug resistance. Cancer 1996;77:1892–8.

[58] Mealey KL, Barhoumi R, Rogers K, et al. Doxorubicin induced expression of P-

glycoprotein in a canine osteosarcoma cell line. Cancer Lett 1998;126:187–92.

[59] Page RL, Hughes CS, Huyan S, et al. Modulation of P-glycoprotein-mediated

doxorubicin resistance in canine cell lines. Anticancer Res 2000;20:3533–8.

[60] Okai Y, Nakamura N, Matsushiro H, et al. Molecular analysis of multidrug resistance in

feline lymphoma cells. Am J Vet Res 2000;61:1122–7.

[61] Ferry DR, Traunecker H, Kerr DJ. Clinical trials of P-glycoprotein reversal in solid

tumours. Eur J Cancer [A] 1996;32A:1070–81.

[62] Hegewisch-Becker S. MDR1 reversal: criteria for clinical trials designed to overcome the

multidrug resistance phenotype. Leukemia 1996;10(Suppl):S32–8.

[63] Malayeri R, Filipits M, Suchomel RW, et al. Multidrug resistance in leukemias and its

reversal. Leuk Lymphoma 1996;23:451–8.

[64] Leyland-Jones B, Dalton W, Fisher GA, et al. Reversal of multidrug resistance to cancer

chemotherapy. Cancer 1993;72(Suppl):3484–8.

[65] Waddle JR, Fine RL, Case BC, et al. Phase I and pharmacokinetic analysis of high-dose

tamoxifen and chemotherapy in normal and tumor-bearing dogs. Cancer Chemother
Pharmacol 1999;44:74–80.

[66] Dantzig AH, Law KL, Cao J, et al. Reversal of multidrug resistance by the P-glycoprotein

modulator, LY335979, from the bench to the clinic. Curr Med Chem 2001;8:39–50.

[67] Banerjee D, Bertino JR. Myeloprotection with drug-resistance genes. Lancet 2002;3:

154–8.

[68] Lefrere F, Delmer A, Suzan F, et al. Sequential chemotherapy by CHOP and DHAP

regimens followed by high-dose therapy with stem cell transplantation induces a high rate
of complete response and improves event-free survival in mantle cell lymphoma:
a prospective study. Leukemia 2002;16:587–93.

664

P.J. Bergman

/ Vet Clin Small Anim 33 (2003) 651–667

background image

[69] Quesenberry PJ, Stewart FM, Becker P, et al. Stem cell engraftment strategies. Ann NY

Acad Sci 2001;938:54–61.

[70] Krishnamachary N, Center MS. The MRP gene associated with a non-P-glycoprotein

multidrug resistance encodes a 190-kDa membrane bound glycoprotein. Cancer Res
1993;53:3658–61.

[71] Lautier D, Canitrot Y, Deeley RG, et al. Multidrug resistance mediated by the multidrug

resistance protein (MRP) gene. Biochem Pharmacol 1996;52:967–77.

[72] Lee DW, Deeley RG, Cole SPC. Biology of the multidrug resistance-associated protein.

MRP. Eur J Cancer [A] 1996;32A:945–57.

[73] Loe DW, Almquist KC, Deeley RG, et al. Multidrug resistance protein (MRP)-mediated

transport of leukotriene C4 and chemotherapeutic agents in membrane vesicles—
demonstration of glutathione-dependent vincristine transport. J Biol Chem 1996;271:
9675–82.

[74] Scheffer GL, Kool M, Heijn M, et al. Specific detection of multidrug resistance proteins

MRP1, MRP2, MRP3, MRP5, and MDR3 P-glycoprotein with a panel of monoclonal
antibodies. Cancer Res 2000;60:5269–77.

[75] Kartenbeck J, Leuschner U, Mayer R, et al. Absence of the canalicular isoform of the

MRP gene-encoded conjugate export pump from the hepatocytes in Dubin-Johnson
syndrome. Hepatology 1996;23:1061–6.

[76] Clifford SC, Neal DE, Lunec J. Alterations in expression of the multidrug resistance-

associated protein (MRP) gene in high-grade transitional cell carcinoma of the bladder. Br
J Cancer 1996;73:659–66.

[77] Zhou DC, Zittoun R, Marie JP. Expression of multidrug resistance-associated protein

MRP and multidrug resistance (MDR1) genes in acute myeloid leukemia. Leukemia
1995;9:1661–6.

[78] Scheper RJ, Scheffer GL, Flens MJ, et al. Transporter molecules in multidrug resistance.

Cytotechnology 1996;19:187–90.

[79] Slovak ML, Ho JP, Cole SPC, et al. The LRP gene encoding a major vault protein

associated with drug resistance maps proximal to MRP on chromosome 16: evidence that
chromosome breakage plays a key role in MRP or LRP gene amplification. Cancer Res
1995;55:4214–9.

[80] Izquierdo MA, Scheffer GL, Flens MJ, et al. Major vault protein LRP-related multidrug

resistance. Eur J Cancer [A] 1996;32A:979–84.

[81] Berger JM, Gamblin SJ, Harrison SC, et al. Structure and mechanism of DNA

topoisomerase II. Nature 1996;379:225–32.

[82] Capranico G, Giaccone G, D’Incalci M. DNA topoisomerase II poisons and inhibitors.

Cancer Chemother Biol Response Modif 1999;18:125–43.

[83] Alton PA, Harris AL. The role of DNA topoisomerases II in drug resistance. Br J

Haematol 1993;85:241–5.

[84] Beck WT, Danks MK, Wolverton JS, et al. Altered DNA topoisomerase II in multidrug

resistance. Cytotechnology 1993;11:115–9.

[85] Moscow JA, Dixon KH. Glutathione-related enzymes, glutathione and multidrug

resistance. Cytotechnology 1993;12:155–70.

[86] Green JA, Robertson LJ, Clark AH. Glutathione S-transferase expression in benign and

malignant ovarian tumours. Br J Cancer 1993;68:235–9.

[87] Hoban PR, Robson CN, Davies SM, et al. Reduced topoisomerase II and elevated alpha

class glutathione S-transferase expression in a multidrug resistant CHO cell line highly
cross-resistant to mitomycin C. Biochem Pharmacol 1992;43:685–93.

[88] Giaccone G. Clinical perspectives on platinum resistance. Drugs 2000;59(Suppl 4):

9–17.

[89] Ramachandran C, Kang Yuan Z, Ling Huang X, et al. Doxorubicin resistance in human

melanoma cells: MDR-1 and glutathione S-transferase p gene expression. Biochem
Pharmacol 1993;45:743–51.

665

P.J. Bergman

/ Vet Clin Small Anim 33 (2003) 651–667

background image

[90] Kuchan MJ, Milner JA. Influence of intracellular glutathione on selenite-mediated growth

inhibition of canine mammary tumor cells. Cancer Res 1992;52:1091–5.

[91] Shoieb AM, Hahn KA, Van Laack RL, et al. In vitro reversal of glutathione-S-

transferase-mediated resistance in canine osteosarcoma (COS31) cells. In Vivo 1998;12:
455–62.

[92] Bertino JR. Ode to methotrexate. J Clin Oncol 1993;11:5–14.
[93] Li X-K, Kobayashi H, Holland JF, et al. Expression of dihydrofolate reductase and

multidrug resistance genes in trimetrexate-resistant human leukemia cell lines. Leuk Res
1993;17:483–90.

[94] Rustum YM, Harstrick A, Cao S, et al. Thymidylate synthase inhibitors in cancer

therapy: direct and indirect inhibitors. J Clin Oncol 1997;15:389–400.

[95] Spears CP. Clinical resistance to antimetabolites. Hematol Oncol Clin North Am 1995;

9:397–413.

[96] Takemura Y, Kobayashi H, Miyachi H. Cellular and molecular mechanisms of resistance

to antifolate drugs: new analogues and approaches to overcome the resistance. Int J
Hematol 1997;66:459–77.

[97] Lee JS, Libshitz HI, Fossella FV, et al. Edatrexate improves the antitumor effects of

cyclophosphamide and cisplatin against non-small cell lung cancer. Cancer 1991;68:959–64.

[98] Lin JT, Bertino JR. Update on trimetrexate, a folate antagonist with antineoplastic and

antiprotozoal properties. Cancer Invest 1991;9:159–72.

[99] Keller ET, MacEwen EG, Rosenthal RC, et al. Evaluation of prognostic factors and

sequential combination chemotherapy with doxorubicin for canine lymphoma. J Vet
Intern Med 1993;7:289–95.

[100] Gerson SL. Clinical relevance of MGMT in the treatment of cancer. J Clin Oncol

2002;20:2388–99.

[101] Mitchell RB, Dolan ME. Effect of temozolomide and dacarbazine on O6-alkylguanine-

DNA alkyltransferase activity and sensitivity of human tumor cells and xenografts to
1,3-bis(2-chloroethyl)-1-nitrosourea. Cancer Chemother Pharmacol 1993;32:59–63.

[102] Joncourt F, Oberli A, Redmond SMS, et al. Cytostatic drug resistance: parallel assess-

ment of glutathione-based detoxifying enzymes, O6-alkylguanine-DNA-alkyltrans-
ferase and P-glycoprotein in adult patients with leukaemia. Br J Haematol 1993;85:
103–11.

[103] Baer JC, Freeman AA, Newlands ES, et al. Depletion of O6-alkylguanine-DNA

alkyltransferase correlates with potentiation of temozolomide and CCNU toxicity in
human tumour cells. Br J Cancer 1993;67:1299–302.

[104] Tentori L, Graziani G. Pharmacological strategies to increase the antitumor activity of

methylating agents. Curr Med Chem 2002;9:1285–301.

[105] Takahara PM, Rosenzweig AC, Frederick CA, et al. Crystal structure of double-stranded

DNA containing the major adduct of the anticancer drug cisplatin. Nature 1995;377:
649–52.

[106] Kartalou M, Essigmann JM. Mechanisms of resistance to cisplatin. Mutat Res 2001;

478:23–43.

[107] McHugh PJ, Spanswick VJ, Hartley JA. Repair of DNA interstrand crosslinks: molecular

mechanisms and clinical relevance. Lancet 2001;2:483–90.

[108] Murray D, Vallee-Lucic L, Rosenberg E, et al. Sensitivity of nucleotide excision repair-

deficient human cells to ionizing radiation and cyclophosphamide. Anticancer Res
2002;22:21–6.

[109] Jacob S, Aguado M, Fallik D, et al. The role of the DNA mismatch repair system in the

cytotoxicity of the topoisomerase inhibitors camptothecin and etoposide to human
colorectal cancer cells. Cancer Res 2001;61:6555–62.

[110] Stoehlmacher J, Ghaderi V, Iobal S, et al. A polymorphism of the XRCC1 gene predicts

for response to platinum based treatment in advanced colorectal cancer. Anticancer Res
2001;21:3075–9.

666

P.J. Bergman

/ Vet Clin Small Anim 33 (2003) 651–667

background image

[111] Magni M, Shammah S, Schiro R, et al. Induction of cyclophosphamide-resistance by

aldehyde-dehydrogenase gene transfer. Blood 1996;87:1097–103.

[112] Friedman HS, Colvin OM, Kaufmann SH, et al. Cyclophosphamide resistance in

medulloblastoma. Cancer Res 1992;52:5373–8.

[113] Rekha GK, Devaraj VR, Sreerama L, et al. Inhibition of human class 3 aldehyde

dehydrogenase, and sensitization of tumor cells that express significant amounts of this
enzyme to oxazaphosphorines, by chlorpropamide analogues. Biochem Pharmacol 1998;
55:465–74.

[114] Sladek NE, Kollander R, Sreerama L, et al. Cellular levels of aldehyde dehydrogenases

(ALDH1A1 and ALDH3A1) as predictors of therapeutic responses to cyclophosphamide-
based chemotherapy of breast cancer: a retrospective study. Rational individualization
of oxazaphosphorine-based cancer chemotherapeutic regimens. Cancer Chemother Phar-
macol 2002;49:309–21.

[115] Wyllie AH. Apoptosis and the regulation of cell numbers in normal and neoplastic tissues:

An overview. Cancer Metastasis Rev 1992;11:95–103.

[116] Dewey WC, Ling CC, Meyn RE. Radiation-induced apoptosis: relevance to radiotherapy.

Int J Radiat Oncol Biol Phys 1995;33:781–96.

[117] Makin G. Targeting apoptosis in cancer chemotherapy. Expert Opin Ther Targets

2002;6:73–84.

[118] Kim PK, Mahidhara R, Seol DW. The role of caspase-8 in resistance to cancer che-

motherapy. Drug Resist Update 2001;4:293–6.

[119] Makin G, Dive C. Apoptosis and cancer chemotherapy. Trends Cell Biol 2001;

11(Suppl):S22–6.

[120] Perego P, Corna E, De Cesare M, et al. Role of apoptosis and apoptosis-related genes in

cellular response and antitumor efficacy of anthracyclines. Curr Med Chem 2001;8:31–7.

[121] Satyamoorthy K, Bogenrieder T, Herlyn M. No longer a molecular black box—new clues

to apoptosis and drug resistance in melanoma. Trends Mol Med 2001;7:191–4.

[122] Phillips BS, Kass PH, Naydan DK, et al. Apoptotic and proliferation indexes in canine

lymphoma. J Vet Diagn Invest 2000;12:111–7.

[123] Knapp DW, Chan TCK, Kuczek T, et al. Evaluation of in vitro cytotoxicity of

nonsteroidal anti-inflammatory drugs against canine tumor cells. Am J Vet Res 1995;56:
801–5.

[124] Mohammed SI, Bennett PF, Craig BA, et al. Effects of the cyclooxygenase inhibitor,

piroxicam, on tumor response, apoptosis, and angiogenesis in a canine model of human
invasive urinary bladder cancer. Cancer Res 2002;62:356–8.

[125] Barroga EF, Kadosawa T, Asano K, et al. Apoptosis induction of POS canine

osteosarcoma cells by vitamin D and retinoids. J Vet Med Sci 1998;60:1269–72.

[126] Hong SH, Ohashi E, Kadosawa T, et al. Retinoid receptors and the induction of apoptosis

in canine osteosarcoma cells. J Vet Med Sci 2000;62:469–72.

[127] Ohashi E, Hong SH, Takahashi T, et al. Effect of retinoids on growth inhibition of two

canine melanoma cell lines. J Vet Med Sci 2001;63:83–6.

[128] Okano F, Yamada K. Canine interleukin-18 induces apoptosis and enhances Fas ligand

mRNA expression in a canine carcinoma cell line. Anticancer Res 2000;20:3411–5.

[129] Roels S, Tilmant K. Ducatelle R. p53 expression and apoptosis in melanomas of dogs

and cats. Res Vet Sci 2001;70:19–25.

[130] Takahashi T, Kadosawa T, Nagase M, et al. Inhibitory effects of glucocorticoids on

proliferation of canine mast cell tumor. J Vet Med Sci 1997;59:995–1001.

667

P.J. Bergman

/ Vet Clin Small Anim 33 (2003) 651–667

background image

Index

Note: Page numbers of article titles are in boldface type.

A

Abdominal cavity

evaluation of

in mast cell tumors in dogs

assessment, 478

Actinomycin-D

for soft tissue sarcomas, 524

Anastomosis(es)

uretercolonic

for bladder TCC, 603–604

Angiocardiography

for hemangiosarcoma in dogs

and cats assessment, 539

Anticancer drug resistance

dihydrofolate reductase in, 659
glutathione system in, 659
lung resistance–related, 659
mechanisms of, 651–667
multidrug apoptosis resistance,

660–661

multidrug resistance–associated

protein, 657–658

P-glycoprotein, 653–657
thymidylate synthase in, 659
topoisomerases in, 658

Apoptosis resistance–related

multidrug resistance, 660–661

Argyrophilic nucleolar straining

organizing regions

in mast cell tumors in dogs

assessment, 481

Aspiration

bone marrow

in mast cell tumors

in dogs assessment, 478

lymph node

in mast cell tumors in dogs

assessment, 478

Aspiration cytology

for hemangiosarcoma

in dogs and cats
assessment, 540

Axial canine osteosarcoma, 505–509. See

also Osteosarcoma, canine, axial.

B

B5 antigen expression

in canine lymphoma

prognosis, 461

Biochemistry profile

in mast cell tumors in dogs

assessment, 477

Biologic therapy

for hemangiosarcoma in dogs and

cats, 545

Bladder

transitional cell carcinoma of,

597–613. See also Transitional
cell carcinoma, bladder.

Bladder reconstruction

for bladder TCC, 604

Bone infarcts

canine osteosarcoma

due to, 492

Bone marrow aspiration

in mast cell tumors in dogs

assessment, 478

Buffy coat smear

in mast cell tumors in dogs

assessment, 477–478

C

Cancer

head and neck. See Head and neck

cancer.

Canine appendicular osteosarcoma,

494–505. See also Osteosarcoma,
canine, appendicular.

Canine extraskeletal osteosarcoma,

509–511. See also Osteosarcoma,
canine, extraskeletal.

Vet Clin Small Anim

33 (2003) 669–675

0195-5616/03/$ - see front matter

Ó 2003, Elsevier Inc. All rights reserved.

doi:10.1016/S0195-5616(03)00051-2

background image

Carboplatin

for bladder TCC, 606
for soft tissue sarcomas, 524

Cat(s)

hemangiosarcoma in, 533–552.

See also Hemangiosarcoma,
in dogs and cats.

injection site sarcomas in, 553–571.

See also Injection site sarcomas,
feline.

lymphoma in

inhibitor molecule p27Kip1

dysregulation in, 467

risk factors for, 466
updates related to, 466–467

Catheter(s)

permanent cystostomy

for bladder TCC, 603

Chemoradiation therapy

for head and neck cancer, 617–618

Chemotherapy

for canine lymphoma, 461–463
for hemangiosarcoma in dogs and

cats, 543–545

for mast cell tumors in dogs,

484–485

for soft tissue sarcomas

candidates for, 519–520
combination, 525–527
for patients with measurable

disease, 527

new agents, 524–525
practical uses, 527–529
sequential, 525–527
single-agent, 520–524

new agents, 629–649. See also

specific agents, e.g., Lomustine.

gemcitabine, 639–647
ifosfamide, 634–637
lomustine, 629–634
streptozocin, 637–639

rescue-related

for canine lymphoma,

463–464

Cisplatin

for bladder TCC, 605–606
for soft tissue sarcomas, 524

Complete blood count (CBC)

for hemangiosarcoma in dogs

and cats assessment, 538

in mast cell tumors in dogs

assessment, 477

Cystectomy

partial

for bladder TCC, 602–603

Cystostomy catheter

permanent

for bladder TCC, 603

Cytology

aspiration

for hemangiosarcoma in dogs

and cats assessment, 540

D

Dacarbazine

for soft tissue sarcomas, 523–524

Dental tumors

treatment of, 619

Dihydrofolate reductase

in anticancer drug resistance, 659

DNA ploidy

in mast cell tumors in dogs

assessment, 482

Docetaxel

for soft tissue sarcomas, 524

Dog(s)

hemangiosarcoma in, 533–552.

See also Hemangiosarcoma,
in dogs and cats.

lymphoma in

new developments in,

457–466. See also
Lymphoma(s), updates
related to, in dogs.

mammary gland tumors in,

573–596. See also Mammary
gland tumors, canine.

osteosarcoma in, 491–516.

See also Osteosarcoma, canine.

Doxorubicin

for bladder TCC, 604–605
for soft tissue sarcomas, 520–522

Drug resistance

anticancer

mechanisms of, 651–667

to chemotherapy

acquired

mechanisms of, 652

E

Ear canal tumors

treatment of, 622

ECG. See Electrocardiography (ECG).

Electrocardiography (ECG)

for hemangiosarcoma in dogs

and cats assessment, 539

670

Index / Vet Clin Small Anim 33 (2003) 669–675

background image

F

Fatigue microdamage

canine osteosarcoma due to, 492

FCE 23762-methoxymorpholino-

doxorubicin

for soft tissue sarcomas, 524

Feline leukemia virus (FeLV), 455–471

Fibrosarcoma(s)

treatment of, 619

G

Gemcitabine, 639–647

activity in people, 640
clinical applications of

general, 642–643
in cats, 645–646
in dogs, 643–645

dosage of, 643–644
efficacy of, 645
toxicities of, 644

described, 639–640
mechanism of action of, 640
preclinical studies of, 641–642

Gender

as factor in hemangiosarcoma

in dogs and cats, 534

Genetic alterations

canine osteosarcoma due to,

492–494

Glutathione system

in anticancer drug resistance, 659

Gonadal hormone exposure

canine osteosarcoma due to, 492

H

Head and neck cancer

ear canal tumors, 622
nasal tumors, 615–618
oral tumors, 619–622
salivary gland tumors, 622
thyroid tumors, 623
treatment of

biologically targeted

therapies in, 623

chemoradiation in, 617–618
multimodality, 615–628

Hemangiosarcoma

in dogs and cats, 533–552

biologic behavior associated

with, 535–536

causes of, 534–535

clinical presentation of,

536–537

diagnosis of, 537–541
future directions in, 546–547
gender predilection for, 534
incidence of, 533–534
patient factors in, 533–534
prognosis of, 546
staging of, 537–541
treatment of, 541–545

biologic therapy in, 545
chemotherapy in,

543–545

radiation in, 543
surgical, 541–543

Histopathology

for hemangiosarcoma in dogs and

cats assessment, 540–541

I

Ifosfamide, 634–637

activity in people, 635
clinical applications of

in cats, 637
in dogs, 636–637

for soft tissue sarcomas, 523
mechanisms of action of, 634–635
preclinical studies, 635–636

Immunoglobulin rearrangement

in canine lymphoma diagnosis,

457–458

Immunophenotyping

in canine lymphoma prognosis, 461

Immunotherapy

for canine lymphoma, 465–466

Immunotoxin(s)

recombinant

for canine lymphoma,

464–465

Inhibitor molecule p27Kip1

dysregulation

in cats, 467

Injection site sarcomas

feline, 553–571

causes of, 557–560
clinical presentation of,

563–567

epidemiology of, 553–557
historical background of,

553–557

prevention of, 567–568
research related to, 560–563
treatment of, 563–567

671

Index / Vet Clin Small Anim 33 (2003) 669–675

background image

Ionizing radiation

canine osteosarcoma due to,

491–492

K

Ki-67

in mast cell tumors in dogs

assessment, 482

L

Lomustine, 629–634

activity in people, 630
clinical applications of

in cats, 633–634

dosage of, 633
efficacy of, 634
toxicities of, 633–634

in dogs, 631–633

dosage of, 631
efficacy of, 632–633
toxicities of, 631–632

mechanisms of action of, 629–630
preclinical studies, 630–631

Lung resistance–related protein, 659

Lymph node aspiration

in mast cell tumors in dogs

assessment, 478

Lymphoma(s)

in cats

risk factors for, 466

in dogs

new developments in, 457–466
treatment of

chemotherapy in, 461–463
future directions in,

464–466

immunotherapy in,

465–466

radiation therapy in, 463
recombinant

immunotoxins in,
464–465

rescue-related, 463–464

updates related to, 455–471

in cats, 466–467
in dogs, 457–466

B5 antigen expression, 461
diagnostic tools, 457–458
immunoglobulin

rearrangement,
457–458

immunophenotyping, 461
matrix metalloproteinase

2 and 9 expression,
459

monoclonal antibody C219

immunohistochemistry
against
P-glycoprotein,
460–461

prognostic markers,

458–461

serum alpha 1–acid

glycoprotein
concentrations, 459

telomerase activity, 458
treatment-related, 461–466.

See also
Lymphoma(s),
in dogs, treatment of.

treatment of, 456–457

M

Mammary gland tumors

canine, 573–596

biologic behavior in, 576–577
causes of, 573–576
clinical signs of, 577–578
diagnosis of, 578–581
epidemiology of, 573–576
histopathologic classification of,

576–577

obesity and, 575
physical examination in,

577–578

prognostic factors in, 581–585
staging of, 578–581
treatment of, 585–591
tumor types, 576–577

Mast cell(s)

biology of, 473–474
development of, 473–474
function of, 474
morphologic characteristics of, 473

Mast cell tumors

in dogs, 473–489

abdominal cavity evaluation

in, 478

anatomic location of, 481
argyrophilic nucleolar straining

organizing regions in, 481

biochemistry profile in, 477
bone marrow aspiration in, 478
breed and, 481
buffy coat smear in, 477–478
causes of, 475
clinical signs of, 475–476
clinical staging of, 479, 481
complete blood count in, 477
diagnosis of, 476–477
DNA ploidy in, 482
growth rate of, 481

672

Index / Vet Clin Small Anim 33 (2003) 669–675

background image

histologic grade of, 479–480
incidence of, 475
Ki-67 in, 482
lymph node aspiration

in, 478

patient history in, 475–476
PCNA in, 482
prognostic factors in, 479–482
signalment of, 475
staging of, 477
thoracic cavity assessment in, 478
treatment of, 482–485

chemotherapy in, 484–485
radiation therapy in,

483–484

supportive care in, 486
surgical, 482–483

urinalysis in, 477

Matrix metalloproteinase

2 and 9 expression

in canine lymphoma prognosis, 459

Melanoma(s)

treatment of, 620–621

Mesna, 634–637

Mitoxantrone

for bladder TCC, 606–607
for soft tissue sarcomas, 522–523

Monoclonal antibody C219

immunohistochemistry against
P-glycoprotein

in canine lymphoma prognosis,

460–461

Multidrug resistance–associated protein,

657–658

N

Nasal tumors

treatment of, 615–618

Neck cancer

treatment of

multimodality, 615–628

O

Obesity

breast cancer due to, 575

Open cell polylactic acid (OPLA)

for soft tissue sarcomas, 525

OPLA (open cell polylactic acid)

for soft tissue sarcomas, 525

Oral tumors

dental tumors, 619
fibrosarcomas, 619

melanomas, 620–621
osteosarcomas, 621
squamous cell carcinoma, 621–622
treatment of, 619–622

Osteosarcoma

canine, 491–516

appendicular, 494–505

biologic behavior in, 499
diagnosis of, 497–499
incidence of, 495
patient history in, 495
physical examination in,

495

prognostic factors in,

495–496

signalment of, 495
treatment of, 499–504

palliative, 504–505

axial, 505–509

biologic behavior in,

506–509

diagnosis of, 506
patient history in, 505–506
physical examination in,

505–506

prognosis of, 506–509
signalment in, 505
treatment of, 506–509

bone infarcts and, 492
extraskeletal, 509–511

biologic behavior in,

510–511

diagnosis of, 510
incidence of, 509–510
patient history in, 510
physical examination in,

510

prognosis of, 511
risk factors for, 510
signalment in, 509–510
treatment of, 511

fatigue microdamage

due to, 492

genetic alterations and,

492–494

gonadal hormone exposure

and, 492

ionizing radiation and,

491–492

risk factors for, 491–494

treatment of, 621

P

Partial cystectomy

for bladder TCC, 602–603

PCNA. See Proliferating cell nuclear

antigen (PCNA).

673

Index / Vet Clin Small Anim 33 (2003) 669–675

background image

Pericardiocentesis

for hemangiosarcoma in dogs

and cats assessment, 540

Permanent cystostomy catheter

for bladder TCC, 603

P-glycoprotein

monoclonal antibody C219

immunohistochemistry against

in canine lymphoma prognosis,

460–461

resistance to, 653–657

Piroxicam

for bladder TCC, 605

Pneumopericardiography

for hemangiosarcoma in dogs

and cats assessment, 539

Proliferating cell nuclear antigen (PCNA)

in mast cell tumors in dogs

assessment, 482

Protein(s)

lung resistance–related, 659
multidrug resistance–associated,

657–658

R

Radiation

ionizing

canine osteosarcoma due to,

491–492

Radiation therapy

for bladder TCC, 607–608
for canine lymphoma, 463
for hemangiosarcoma in dogs and

cats, 543

for mast cell tumors in dogs,

483–484

rescue-related

for canine lymphoma, 464

Radiography

for hemangiosarcoma in dogs

and cats assessment, 538–539

Recombinant immunotoxins

for canine lymphoma, 464–465

S

Salivary gland tumors

treatment of, 622

Sarcoma(s)

soft tissue

treatment of, 517–531.

See also Soft tissue
sarcomas, treatment of.

Serum alpha 1–acid glycoprotein

concentrations

in canine lymphoma

prognosis, 459

Serum chemistry panel

for hemangiosarcoma in dogs and

cats assessment, 538

Soft tissue sarcomas

described, 517
histopathologic grade of,

517–518

metastatic potential of, 518–519
treatment of

actinomycin-D in, 524
carboplatin in, 524
chemotherapy in

adjuvant, 527–529
candidates for, 519–520
combination, 525–527
for patients with

measurable disease,
527

new agents, 524–525
practical uses, 527–529
sequential, 525–527
single-agent, 520–524

cisplatin in, 524
dacarbazine in, 523–524
docetaxel in, 524
doxorubicin in, 520–522
iosfamide in, 523
medical, 517–531
metoxantrone in, 522–523
OPLA in, 525
vincristine in, 523

Squamous cell carcinoma

oral

canine, 621–622

Streptozocin, 637–639

activity in people, 637–638
clinical applications of

in cats, 639
in dogs, 638–639

mechanisms of action of, 637
preclinical studies, 638

T

TCC. See Transitional cell carcinoma

(TCC).

Telomerase activity

in canine lymphoma diagnosis, 458

Thoracic cavity

evaluation of

in mast cell tumors in dogs

assessment, 478

674

Index / Vet Clin Small Anim 33 (2003) 669–675

background image

Thymidylate synthase

in anticancer drug resistance, 659

Thyroid tumors

treatment of, 623

Topoisomerase(s), 658

Transitional cell carcinoma (TCC)

bladder, 597–613

causes of, 597–598
clinical presentation of, 598
diagnosis of, 598–601
prevalence of, 597
risk factors for, 597–598
staging of, 598–601
treatment of, 601–608

medical, 604–607
photodynamic therapy,

608

radiation therapy, 607–608
surgical, 601–604

urinalysis in, 598–599

Tumor(s). See also specific types,

e.g., Mast cell tumors.

dental

treatment of, 619

ear canal

treatment of, 622

mast cell

in dogs, 473–489. See also

Mast cell tumors, in dogs.

nasal

treatment of, 615–618

oral. See also Oral tumors.
salivary gland

treatment of, 622

thyroid

treatment of, 623

U

Uretercolonic anastomosis

for bladder TCC, 603–604

Urinalysis

in mast cell tumors in dogs

assessment, 477

in transitional cell carcinoma,

598–599

V

Vincristine

for soft tissue sarcomas, 523

675

Index / Vet Clin Small Anim 33 (2003) 669–675


Document Outline


Wyszukiwarka

Podobne podstrony:
2002 3 MAY Lasers in Medicine and Surgery
Modern Advances in Chromatography Freitag R
Clinical Advances in Cognitive Psychotherapy Theory and Application R Leahy, E Dowd (Springer, 200
Modern Advances in Chromatography Springer
ADVANCE IN SCIENCE
Advances in the Detection and Diag of Oral Precancerous, Cancerous Lesions [jnl article] J Kalmar (
2002 3 MAY Lasers in Medicine and Surgery
David Suendermann Advances in Commercial Deployment of Spoken Dialog Systems
Advances in flavonoids research since 1992
S D Houston Into the Minds of Ancients Advances in Maya Glyph Studies
2003 08 trouble in paradise
2003 09 a blogger in their midst
Cheltenham Word 2003 Manual Advanced Level sample
Cheltenham Word 2003 Manual Advanced Level USA sample
Haisch et al Advances in the Proposed Electromagnetic Zero Point Field Theory of Inertia (1998)
Clinical Advances in Cognitive Psychotherapy Theory and Application R Leahy, E Dowd (Springer, 200
Margaret May Business Process Management Integration in a Web Enabled Environment 2003 (By Laxxu

więcej podobnych podstron