Anda di halaman 1dari 10

Cancer Investigation

ISSN: 0735-7907 (Print) 1532-4192 (Online) Journal homepage: http://www.tandfonline.com/loi/icnv20

Naturally Occurring Canine Glioma as a Model for


Novel Therapeutics

Molly E. Hubbard, Susan Arnold, Abdullah Bin Zahid, Matthew McPheeters,


M. Gerard O’Sullivan, Alexandru-Flaviu Tabaran, Matthew A. Hunt & G.
Elizabeth Pluhar

To cite this article: Molly E. Hubbard, Susan Arnold, Abdullah Bin Zahid, Matthew McPheeters,
M. Gerard O’Sullivan, Alexandru-Flaviu Tabaran, Matthew A. Hunt & G. Elizabeth Pluhar (2018):
Naturally Occurring Canine Glioma as a Model for Novel Therapeutics, Cancer Investigation, DOI:
10.1080/07357907.2018.1514622

To link to this article: https://doi.org/10.1080/07357907.2018.1514622

Published online: 20 Sep 2018.

Submit your article to this journal

Article views: 5

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=icnv20
CANCER INVESTIGATION
https://doi.org/10.1080/07357907.2018.1514622

REVIEW

Naturally Occurring Canine Glioma as a Model for Novel Therapeutics


Molly E. Hubbarda, Susan Arnoldb, Abdullah Bin Zahidc, Matthew McPheetersd, M. Gerard O’Sullivanb,e,
Alexandru-Flaviu Tabaranb,e, Matthew A. Hunta and G. Elizabeth Pluharb
a
Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA; bCollege of Veterinary Medicine, University of
Minnesota, Minneapolis, MN, USA; cDivision of Neurosurgery, Hennepin County Medical Center, Minneapolis, MN, USA; dCollege of
Medicine, University of Minnesota, Minneapolis, MN, USA; eComparitive Pathology Shared Resource at Masonic Cancer Center,
University of Minnesota, Minneapolis, MN, USA

ABSTRACT ARTICLE HISTORY


Background: Current animal models of glioma are limited to small animal models, which Received 12 March 2018
are less predictive of treatment of human disease. Canines often develop gliomas de novo, Revised 5 July 2018
but the natural history of the disease is not well described. Accepted 19 August 2018
Objective: We provide data for naturally occurring canine gliomas; evaluating medical and
KEYWORDS
surgical therapies. Canine glioma model;
Methods: We reviewed medical records of pet dogs with a presumptive diagnosis of glioma natural glioma
from MRI imaging that underwent surgery as part of the Canine Brain Tumor Clinical Trials
Program. Breed, age, sex, median progression-free, and overall survival times and cause of
death were recorded for multivariate analysis.
Results: Ninety five dogs (56 male; mean age ¼ 8.3 years) were included, but nine were
excluded as final pathology was non-neoplastic. Gross total resection was reported in 81
cases based on postoperative MRI. Seventy had high-grade tumors (grade III or IV). Eighty
three dogs presented with seizures, being the most common presenting clinical sign.
Median survival after surgery was 723 days (95% CI 343–1103) for grade II tumors, 301 days
(197–404) for grade III and 200 days (126–274) for grade IV (p ¼ .009 Kaplan–Meier survival
analysis; Log Rank test). Age (cox regression, p ¼ .14) or sex (Kaplan–Meier test, p ¼ .22) did
not predict survival.
Conclusions: This study establishes normative data for a model exploiting dogs with natur-
ally occurring glioma, which can be used to test novel therapies prior to translation to
human trials. Further work will focus on the effects of different therapies, including chemo-
therapy, radiation therapy, and immunotherapy.

Introduction The earliest reported surgical resection of


Primary intracranial neoplasms are a relatively com- intracranial tumors in dogs was published by
mon disorder in dogs, with many studies based on Oliver in the 1960s (3, 4). The surgical
necropsy reports having a prevalence range between approaches and techniques for gaining access to
0.014–4.5%. While some breeds are reported to be intracranial tumors were adapted based on tech-
predisposed to developing brain tumors, most breeds niques from human neurosurgical procedures.
are represented (1). Meningiomas are the most fre- From this, the feasibility and success of canine
quently encountered canine primary intracranial neurosurgery was established (5). Procedures
neoplasm, accounting for 45–50% of tumors, with continue to be adapted as technology advances.
treatment well described in the literature (1, 2). Over the past 50 years, surgical resection com-
Gliomas and choroid plexus tumors are estimated to bined with radiation has become the standard of
account for approximately 31–36% and 5–7% of care for canine intracranial meningiomas (6, 7).
tumors, respectively. Surgical resection improves outcomes when com-
pared to radiation alone, with low risk of

CONTACT Molly Hubbard hubba166@umn.edu University of Minnesota, Mayo Building, MMC 96 Room D-429 420 Delaware St SE, Minneapolis,
MN 55455, USA.
Color versions of one or more of the figures in the article can be found online at www.tandfonline.com/icnv.
Supplemental data for this article can be accessed on the publisher’s website
ß 2018 Taylor & Francis Group, LLC
2 M. HUBBARD ET AL.

perioperative morbidity or mortality (8–10). In Methods


the early 1990s, Motta et al. reported a median A database consisting of pet dogs treated in the
survival of 214 days after surgery (7), which was Canine Brain Tumor Treatment Clinical Trials
substantially improved in a later study to 1254
Program between August 2008 and December
days for transitional and meningothelial grade I
2015 was used for subject selection. All dogs
tumors after resection with an ultrasonic aspir-
underwent surgical treatment as a part of the
ator (10). Potential perioperative complications of
Canine Brain Tumor Treatment Clinical Trials
surgical treatment include cerebral edema, seiz-
program. All dogs had a presumptive diagnosis
ures, bradycardia, hyperthermia, and altered level
of glioma based on previous workup and MRI
of consciousness (8, 10).
(GE Signa HDxt3.0 T MRI, Wabasha, Wisconsin)
Despite extensive evidence in favor of surgical
characteristics of an intracranial mass by the
resection for treatment of intracranial meningio-
referring veterinary neurologist prior to surgical
mas, there is limited data to support or refute a
intervention (see the list of MRI sequences in
similar treatment for canine gliomas. In 1991, a
Supplementary material). All owners gave written
small study by Niebauer et al. evaluating craniot-
informed consent for the therapies. Data were
omy in pet dogs and cats found a survival rate
obtained by the operative veterinarian or trained
for non-meningiomas to be greater than meningi-
omas with 40% and 30% one-year canine survival surgical resident. Surgery was performed at the
rates, respectively (11). However, only two of the College of Veterinary Medicine Veterinary
11 non-meningioma lesions were astrocytoma/gli- Medical Center by a veterinary and a human
oma; and survival times for individual animals neurosurgeon. The surgical approaches used were
were not given. Another retrospective study on planned as based on the tumor location (3, 4).
survival of dogs with brain tumors had a median The intent in all cases was to perform gross total
survival of 0.9 months for dogs treated by surgery resection of the lesion when possible. Follow up
þ/ 125I implants (12). In addition, there have MRIs were performed at 2, 6, 12 months, or 4, 8,
been no studies published regarding the out- and 12 months, pending on subject availability;
comes of surgical resection of gliomas. A recent unless tumor recurrence was suspected prior to a
systematic review of brain tumor treatment in schedule recheck time. Initial tumor volume,
dogs gave a median survival time of 226 days for residual tumor volume and recurrent tumor vol-
all dogs with presumptive intra-axial, extra-axial, ume was measured on all MRIs.
and pituitary tumors (12). There were no specific Samples of each mass were collected and
results for surgery of intra-axial tumors listed. reviewed by veterinary pathologists with consult-
Given the paucity of published research, the pur- ation from a neuropathologist at the medical
pose of this study is to describe the techniques school. After the confirmation of the diagnosis
and outcomes for a series of canine gli- on histopathology, adjuvant treatment with
oma resections. chemotherapy, immunotherapy, gene therapy, or
The Canine Brain Tumor Treatment Clinical a combination, was offered. Extent of resection
Trials program aims to offer state–of-the-art (EOR) was evaluated with a complete brain MRI
treatment to the dogs suffering from intracranial study that among other sequences includes T2W,
lesions, and also use the information gained from T2 FLAIR, T1W with and without contrast in 3
treating dogs to design similar treatments for planes immediately after the craniotomy. EOR
human brain tumors. As part of the program, was defined as: gross total resection (GTR) if no
animals are randomized to various treatments residual contrast-enhancement was seen for
after surgical debulking including vaccine-based tumors that were gadolinium enhancing or no T2
immunotherapy with or without a checkpoint fluid attenuating inversion recovery (FLAIR) sig-
blockade inhibitor, chemotherapy with temozolo- nal abnormality was seen for non-contrast
mide, or combination thymidine kinase/Flt3L or enhancing tumors; near total resection (NTR) if
IFN-c gene therapy. This study reports the base- 20% of the preoperative lesion remained; and
line features of the animals enrolled. subtotal resection (STR) if >20% of the
CANCER INVESTIGATION 3

Table 1. Demographics of all animals referred to the program with suspicion for glioma.
Parameter Canines with tumor Canine with non-tumor
N 86 9
Age 8.0 (range: 9 months–14 years) 9.6 (range: 3–14 years)
Sex Female 35 4
Male 51 5
Skull shape Brachycephalic 63 2
Dolichocephalic 4 3
Mesocephalic 19 4
Diagnosis Astrocytoma: 42 (48.8%) Infarct: 4
Ganglioglioma: 14 (16.3%) GME: 2
Meningioma: 2 (2.3%) Histiocytic Sarcoma/tumor: 2
Oligoastrocytoma: 1 (1.2%) Cavernoma: 1
Oligodendroglioma: 23 (26.7%)
PNET: 4 (4.7%)
Tumor grade II 7 n/a
III 51 n/a
IV 28 n/a

preoperative lesion remained. Finally, the dogs Based on the postoperative MRI imaging, gross
were followed in outpatient clinic until death. or near total resection was reported in 80 out of
The progression of disease or recurrence of the 87 tumor cases. Three of the remaining dogs did
tumor was determined either on repeat MRI, not have postoperative imaging, six dogs had sub-
subsequent surgical intervention or at necropsy. total tumor resection as it was not deemed safe to
Descriptive statistics were performed for the remove the entire lesion. Eighty were found to
demographics. The dogs with final pathology as have high-grade tumors (defined as grade III or
non-tumor were excluded from the analyses and grade IV). These included anaplastic astrocytoma,
described separately. Survival analysis was per- glioblastoma, anaplastic oligodendroglioma, and
formed to calculate all-cause mortality and cancer primitive neuroectodermal tumor-like neoplasms
specific mortality rates. Kaplan–Meir and cox (called PNET in this report).
regression was done to analyze the effect of age Few dogs required second resection of their
and gender on survival. For the dogs which under- tumor after recurrence. In general, their surgeries
went multiple resections, only first instance was were slightly more complex due to scar tissue
used in the analyses. p-Values were considered sig- from the previous surgery. All four animals who
nificant at <0.05. All statistics were done using underwent a second surgical resection had high-
SPSS statistics 24 (IBM crop, Armonk, NY, USA). grade tumors initially. Two were diagnosed ini-
tially with GBM, which recurred eight months
Results later in both cases. One was euthanized due to
A total of 99 surgeries were performed on 95 pet symptom progression two months after the
dogs, 39 female and 56 male; four dogs under- second surgical intervention, however, no tumor
went two surgical interventions. Eight dogs were was found on pathologic examination. An MRI
excluded from analyses as the final pathology was documented recurrence of the cerebellar tumor
not glioma. The age ranged from 0.75 to 14.04 before the second surgery and the immediate
with a mean age of 8.3 years. There was an postoperative MRI after the second surgery
increased incidence in certain brachycephalic showed subtotal resection due to scar tissue
breeds, specifically Boxer, Boston, and adhering to the brain stem. The postmortem
Staffordshire terriers and French and English examination of the brain did show residual GBM;
bulldogs. Demographic and histologic data are the volume of tumor on PM was the same as
described in Table 1. Seizure was the most com- that measured on the postoperative MRI. The
mon presenting symptom, being present in 83 other animal was euthanized one month after the
(87%) dogs at the time of presentation for sur- second surgical intervention due to progressive
gery. Followup rate till death was 100%, however, symptoms and tumor recurrence was later con-
not all animals had necropsy. firmed on the necropsy. A third animal was
4 M. HUBBARD ET AL.

diagnosed with an anaplastic astrocytoma (grade


III); and on the second resection three months
later, pathology was unchanged. However, the
animal died 27 days after the second surgery
from medical complications of treatment. The
dog was receiving a series of autologous tumor
lysate vaccines with OX40L after subtotal resec-
tion of the recurrent temporal lobe GBM. Four
days after a vaccine, she suddenly became
obtunded and rapidly progressed to stupor/coma.
The owner declined medical intervention and
opted for humane euthanasia, given the poor
long-term prognosis. The postmortem examin-
ation of the brain showed recurrence of the GBM
and spread to the lateral, third, and fourth ven-
tricles. The last dog underwent gross total resec-
tion of a PNET, and a second resection was done
six months later with unchanged pathology. This Figure 1. MRI of the brain with and without contrast demon-
strating a typical low grade lesion seen in our study of
animal was euthanized six months after the
pet dogs
second resection due to tumor recurrence, which
was confirmed on necropsy. All recurrences were
within the tumor bed and not at remote sites.
MRI findings demonstrating a typical low- and
high-grade lesion are seen in Figures 1 and 2.
Recently, a study by Bentley et al. concluded that
tumor type and grade are difficult to diagnose
using radiological characteristics typified in
humans. They found that intra-axial tumors that
have mild or no contrast enhancement, no cystic
or necrotic center and are superficial to the
internal capsule are more likely to be low grade.
Astrocytomas tend to cause less ventricular dis-
tortion and tend to have a greater amount of
peri-tumoral edema compared to oligodendro-
gliomas (13).
Astrocytoma was the most common diagnosis,
including both low and high-grade tumors, which Figure 2. MRI of the brain with and without contrast demon-
strating a typical high grade lesion seen in our study of
was found in 59 canines. This was followed by pet dogs
grade II or anaplastic oligodendroglioma in 22
animals. High-grade neoplasms were more com-
mon than low grade, as only 7 dogs were diag- The median survival after surgery was 723
nosed with grade II tumors. The most common days (95% CI 343–1103 days) for grade II
location for tumors were the frontal (27.9%) or tumors, 301 days (197–404) for grade III, and
temporal (26.7%) lobes, which dictated surgical 200 days (126–274) for grade IV (p-value for
approach (3, 4). There was no predilection for decreasing survival with higher grade ¼ 0.009
specific lobes based on grade (Figure 3). Kaplan–Meier survival analysis; Log Rank test
Representative histological images of grade II, III, (Figure 5). Age (cox regression, p-value ¼ .14)
and IV tumors are shown in Figure 4 (A–C). and sex (Kaplan–Meier test, p-value .22) did not
CANCER INVESTIGATION 5

predict survival. However, no dogs with grade II treatment groups are low at this time (n ¼ 4–10
tumors were alive at the time of the data analysis dogs). There were significant differences in
(Figure 6). Overall survival was highest for grade response and OS between low-grade (II) and
II tumors, at 986 days (95% CI 517–1454 days) high-grade (III–IV) tumors but none between
followed by grade III tumors at 453 days (95% CI various types of high-grade tumors (i.e., anaplas-
257–649 days), and grade IV tumors at 274 days tic astrocytoma, anaplastic oligodendroglioma,
(95% CI 178–370 days). Younger age at diagnosis GBM, PNET, etc.). Animals with higher grade
was associated with decreased survival when eval- tumors were more likely to die from recurrence
uating tumor specific survival (odds ratio 0.82; than those with lower grade (Figure 7)
CI 0.735–0.922). Adjuvant treatments will be dis- When evaluating at tumor specific mortality
cussed in future manuscripts, however, there was as an endpoint, the median survival was
no difference seen in tumor response and overall 853 days (767–938) for grade II tumors, 274 days
survival among the various adjuvant therapies. It (188–359) for grade III tumors, and 211 days
is unknown if this is a real finding or secondary (12–409) for grade IV tumors (p-value for
to type II error as patient numbers in the decreased survival with higher grade ¼ 0.031
Kaplan–Meier survival analysis; Log-Rank test).
Four dogs were still alive at the time of analysis;
430, 531, 1147, and 2054 days after surgery.
No dogs were lost to follow up, and mortality
data was available for each subject. However, the
cause of death was unknown for six dogs. The
most common cause of death was tumor recur-
rence (50 canines, 58.1%); followed by elective
euthanasia (15 animals, 17.4%) for reasons unre-
lated to tumor recurrence (Table 2). Animals with
grade III and IV tumors were more likely to die
due to recurrence (62.7% of grade III and 53.6%
of grade IV) compared to GRADE II (42.8%).

Discussion
Herein, we present data in a canine population
with spontaneous glioma that is a relevant model
Figure 3. Bar graph showing the location of tumors.
for the human disease. There have been many

Figure 4. (A–C) Representative histological images of canine gliomas. Panel A. Grade II astrocytoma, arrow indicates a gemistocytic
astrocyte. Panel B. Grade III anaplastic oligodendroglioma, arrow indicates microvascular proliferation. Panel C. Grade IV glioblast-
oma, asterisk indicates serpentine necrosis with palisading of tumor cells (arrows). All bars are 100 mm.
6 M. HUBBARD ET AL.

Figure 7. Tumor specific cause of death.

Figure 5. Kaplan–Meier estimate showing overall survival in


grade II, III, and IV glioma. Table 2. List of cause of death in dogs included in the study.
Natural death was concluded after necropsy found no signs of
recurrent tumor.
Cause of death Number (%)
Alive 4 (5.8)
Recurrence 55 (58.1)
Euthanized-non-tumor 16 (7.0)
Natural 6 (17.4)
Other 6 (11.6)
Total 87 (100)

novel treatments, which can later be used in


human trials (15–18). The National Cancer
Institute has recently endorsed the utility of
canine models in the Comparative Brain Tumor
Consortium as well (19).
Herranz et al have described cell culture and
immunohistochemical characteristics of de novo
gliomas in dogs that suggest they have similar
phenotypic features and are an adequate model with
Figure 6. Kaplan–Meier estimate when cause of death is from similar genetic markings for human gliomas (20).
the tumor.
Other studies have also demonstrated the molecular
similarities between human and canine gliomas,
studies outlining the surgical outcomes after which adds support for using a canine model for
meningioma resection in dogs (8–10, 14), but novel treatment of gliomas (21). We also found that
this is the first report to our knowledge describ- tumors occur in similar locations in the canine
ing outcomes after surgery followed by adjuvant model and in human glioma (22).Our current work
medical treatment of gliomas. Reports that have is in the early stages of accumulating genomic data
been published include any intracranial neoplasia to continue to improve the understanding of the
in dogs, and rarely cats, and have very low num- similarities between human and canine disease.
bers for glioma (1, 2, 11). The use of a naturally Further efforts to classify tumors based on genetic
occurring model for cancer research and therapy markers is out of the scope of this article, but is
provides an important approach for developing underway in other studies.
CANCER INVESTIGATION 7

this will be examined further in future studies


evaluating genetic markers.
There are limitations to this report, most obvi-
ous being that the animals underwent treatment in
addition to surgical intervention, which may alter
the survival in dogs with gliomas. Not all dogs
underwent necropsy, thus some dogs may have
been misclassified as to their cause of death being
from tumor progression. Unfortunately, when
working with pet dogs, the outcome data are
skewed as few dogs die a natural death. Most dogs
undergo elective euthanasia due to clinical deterior-
ation, uncontrollable epilepsy, or progression of the
disease. Furthermore, with or without necropsy,
the cause of death may not be accurately defined,
Figure 8. Kaplan–Meier estimate evaluating survival based on thus altering the results of treatment. In addition,
extent of resection (EOR). while we understand that canine models may not
be a perfect model, a spontaneously occurring
Hu et al. (12) published a review of treatments tumor may be more relevant than induced models;
for any canine brain tumor, and reported several including patient-derived xenograft, multiple muta-
limitations of their literature search; many of tion, or humanized mouse models.
which we have addressed here. This was a single There is virtually no information regarding
center study, all the animals were recruited by response to any therapy in dogs with high-grade
the same veterinarian and final histologic diagno- glioma, and there certainly is no accepted stand-
sis was done in every case. Furthermore, survival ard of care. The median survival time reported
times and adverse events were known for all can- for surgical resection alone in dogs with supra-
ines enrolled, and cause of death was available tentorial glioma (6 low-grade, 8 high-grade) was
for 96% of canines in our study. We were able to 66 days, similar to that of palliative care (69
identify a “cause specific” mortality for the ani- days) (24). Therefore, in our study, no dog was
mals based on the grade of their tumor. treated with surgical resection alone. When ana-
Interestingly, in our cohort, there was a predom- lyzing our cohort, we found no significant differ-
inance of male animals, similar to what is seen in ences in the overall survival among the groups
some subtypes of human glioblastomas (23). (Mantel–Cox log-rank test (p ¼ .4054) and
While this has been partially explained by sexual Gehan–Breslow–Wilcoxon test (p ¼ .4484)).
dimorphism in expression of tumor suppressor In addition, molecular and genetic markers were
genes, it is not clear if this is the case in the not assessed in this study, which represents a rapidly
canine model as well, however, the model lends emerging field in neuro-oncology. The diagnoses
itself to further investigation in this area as well. were made on histopathologic and immunohisto-
However, this has not been previously reported chemical criteria from the surgical biopsies, and pro-
in a canine model. Furthermore, any influence of gression was determined either after a subsequent
hormones is unlikely as the majority of dogs had surgical intervention or at necropsy. Nevertheless,
undergone castration. EOR was associated with this report remains useful as a benchmark to meas-
improved survival, whereas dogs with GTR had ure further treatment improvements and survival
improved survival when compared to NTR or gains in the setting of canine glioma.
STR (p ¼ .026), Figure 8. Interestingly, in our
cohort, older dogs had an increased overall sur-
Conclusions
vival when compared to younger dogs. We were
not able to compare between grades as the num- This is descriptive data of a naturally occurring gli-
bers were not high enough in each group, but oma in pet dogs, which can be used to test
8 M. HUBBARD ET AL.

different therapies prior to human trials, and is the of brain meningioma. J Vet Med Sci/Jpn Soc Vet Sci.
largest study of its type. By understanding the 2014;76(3):331–8.
9. Motta L, Mandara MT, Skerritt GC. Canine and feline
course of disease in a canine model, even with
intracranial meningiomas: an updated review. Vet J
immunotherapy and chemotherapy treatments, we (London, England: 1997). 2012;192(2):153–65.
can assess the efficacy of novel treatments. Using a 10. Greco JJ, Aiken SA, Berg JM, Monette S, Bergman PJ.
naturally occurring model of glioma may decrease Evaluation of intracranial meningioma resection with
the complications of using genetic knock-out mod- a surgical aspirator in dogs: 17 cases (1996–2004).
J Am Vet Med Assoc. 2006;229(3):394–400.
els (21), and further advancement of treatment reg-
11. Niebauer GW, Dayrell-Hart BL, Speciale J. Evaluation
imens for human tumors. This naturally occurring of craniotomy in dogs and cats. J Am Vet Med Assoc.
model also allows to test multimodality treatment 1991;198(1):89–95.
regimes, otherwise not possible. Further work is 12. Hu H, Barker A, Harcourt-Brown T, Jeffery N.
needed to understand the effects of different Systematic review of brain tumor treatment in dogs.
therapies, (chemotherapy, radiotherapy, and/or J Vet Int Med. 2015;29(6):1456–63.
13. Bentley RT, Ober CP, Anderson KL, Feeney DA,
immunotherapy) alone and combined. Naughton JF, Ohlfest JR, et al. Canine intracranial
gliomas: relationship between magnetic resonance
Disclosure statement imaging criteria and tumor type and grade. Vet J
(London, England: 1997). 2013;198(2):463–71.
The authors have no conflict of interest with the data presented. 14. Heidner GL, Kornegay JN, Page RL, Dodge RK,
Thrall DE. Analysis of survival in a retrospective
study of 86 dogs with brain tumors. J Vet Int Med/
Funding
Am College Vet Int Med. 1991;5(4):219–26.
This work was supported by American Cancer Society, NIH 15. Schiffman JD, Breen M. Comparative oncology: what
Clinical Center, and Humor to Fight the Tumor dogs and other species can teach us about humans
Foundation This work was supported by American Cancer with cancer. Philos Trans R Soc Lond B Biol Sci.
Society, NIH Clinical Center, and Humor to Fight the 2015;370(1673): 20140231–20140233.
Tumor Foundation 16. Grenier JK, Foureman PA, Sloma EA, Miller AD.
RNA-seq transcriptome analysis of formalin fixed,
paraffin-embedded canine meningioma. PloS one.
References 2017;12(10):e0187150.
1. Song RB, Vite CH, Bradley CW, Cross JR. 17. Thomas R, Duke SE, Wang HJ, Breen TE, Higgins RJ,
Postmortem evaluation of 435 cases of intracranial Linder KE, et al. ’Putting our heads together’: insights
neoplasia in dogs and relationship of neoplasm with into genomic conservation between human and canine
intracranial tumors. J Neurooncol. 2009;94(3):333–49.
breed, age, and body weight. J Vet Intern Med/Am
18. Hicks J, Platt S, Kent M, Haley A. Canine brain
College Vet Intern Med. 2013;27(5):1143–52.
tumours: a model for the human disease? Vet Comp
2. Snyder JM, Shofer FS, Van Winkle TJ, Massicotte C.
Oncol. 2017;15(1):252–72.
Canine intracranial primary neoplasia: 173 cases
19. LeBlanc AK, Mazcko C, Brown DE, Koehler JW,
(1986–2003). J Vet Intern Med/Am College Vet
Miller AD, Miller CR, et al. Creation of an NCI com-
Intern Med. 2006;20(3):669–75. parative brain tumor consortium: informing the trans-
3. Oliver J. Surgical approaches to the canine brain. Am lation of new knowledge from canine to human brain
J Vet Res. 1968;29(2):353–78. tumor patients. Neuro-oncology. 2016;18(9):1209–18.
4. Oliver J. Principles of Canine Brain Surgery. J Am 20. Herranz C, Fernandez F, Martin-Ibanez R, Blasco E,
Anim Hosp Assoc. 1966;2:73–88. Crespo E, De la Fuente C, et al. Spontaneously arising
5. Hoerlein BF, Few AB, Petty MF. Brain surgery in the canine glioma as a potential model for human glioma.
dog–preliminary studies. J Am Vet Med Assoc. 1963; J Comp Pathol. 2016;154(2–3):169–79.
143:21–9. 21. Candolfi M, Curtin JF, Nichols WS, Muhammad AG,
6. Kube SA, Bruyette DS, Hanson SM. Astrocytomas in King GD, Pluhar GE, et al. Intracranial glioblastoma
young dogs. J Am Anim Hosp Assoc. 2003;39(3): models in preclinical neuro-oncology: neuropatho-
288–93. logical characterization and tumor progression. J
7. LeCouteur RA. Current concepts in the diagnosis and Neurooncol. 2007;85(2):133–48.
treatment of brain tumours in dogs and cats. J Small 22. Larjavaara S, M€antyl€a R, Salminen T, Haapasalo H,
Anim Pract. 1999;40(9):411–6. Raitanen J, J€a€askel€ainen J, et al. Incidence of gliomas
8. Ijiri A, Yoshiki K, Tsuboi S, Shimazaki H, Akiyoshi by anatomic location. Neuro-oncology. 2007;9(3):
H, Nakade T. Surgical resection of twenty-three cases 319–25.
CANCER INVESTIGATION 9

23. Sun T, Warrington NM, Luo J, Brooks MD, 24. Sunol A, Mascort J, Font C, Bastante AR, Pumarola
Dahiya S, Snyder SC, et al. Sexually dimorphic RB M, Feliu-Pascual AL. Long-term follow-up of surgical
inactivation underlies mesenchymal glioblastoma resection alone for primary intracranial rostrotentorial
prevalence in males. J Clin Invest. 2014;124(9): tumors in dogs: 29 cases (2002–2013). Open Vet J.
4123–33. 2017;7(4):375–83.

Anda mungkin juga menyukai