You are on page 1of 8

Biochimica et Biophysica Acta 1836 (2013) 158–165

Contents lists available at SciVerse ScienceDirect

Biochimica et Biophysica Acta


journal homepage: www.elsevier.com/locate/bbacan

Review

Vertebrate animal models of glioma: Understanding the mechanisms


and developing new therapies
Leon Chen a, Yuqing Zhang b, Jingxuan Yang a, John P. Hagan a, Min Li a,⁎
a
The Vivian L. Smith Department of Neurosurgery, the University of Texas Medical School at Houston, Houston, TX 77030, USA
b
Department of Cancer Biology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA

a r t i c l e i n f o a b s t r a c t

Article history: Glioblastoma Multiforme (GBM) is recognized as one of the most deadly cancers characterized by cellular
Received 10 March 2013 atypia, severe necrosis, and high rate of angiogenesis. In this review, we discuss a diversified group of GBM
Received in revised form 14 April 2013 xenograft models and compare them with the genetically engineered mouse (GEM) model systems. Next,
Accepted 15 April 2013
we describe common genetic defects observed in GBM and numerous GEM models that recapitulate these ab-
Available online 22 April 2013
normalities. Finally, we focus on the clinical value of other vertebrate animal models such as the canine
Keywords:
model by examining their contributions to GBM research.
Brain tumor © 2013 Elsevier B.V. All rights reserved.
Animal model
Tumorigenesis
Cancer therapy

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 158
2. Mouse models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 159
2.1. Xenograft mouse models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 159
2.2. Genetically engineered mouse models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 160
3. Common genetic mutations in GBM . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 161
3.1. Epidermal growth factor receptor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 161
3.2. PTEN . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 161
3.3. INK4a/ARF . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 162
3.4. TP53 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 162
4. The canine model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 162
5. Other animal models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 163
6. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 163
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 163
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 163
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 163

1. Introduction (oligodendrogliomas), ependymal cells (ependymomas), or mixtures of


glial cells. As the most common primary tumors in adult CNS, an estimat-
Glioma covers various primary tumor types with histological features ed of 30,000 patients are diagnosed with glioma in the United States
similar to glia in the central nervous system (CNS). These gliomas are every year [1]. Glioblastoma multiforme (GBM), a grade IV glioma classi-
composed predominantly of astrocytes (astrocytomas), oligodendrocytes fied by the World Health Organization (WHO), is considered the most
malignant and invasive subtype with a median survival of 14.6 months
⁎ Corresponding author at: The Vivian L. Smith Department of Neurosurgery, Department even after radiotherapy and concomitant chemotherapy [2]. GBM is char-
of Integrative Biology & Pharmacology, The University of Texas Medical School at
acterized by histopathologic features of cellular atypia, severe necrosis as
Houston, 6431 Fannin Street, MSE R266, Houston, TX 77030, USA. Tel.: +1 713 500 6491;
fax: +1 713 500 6493. well as high rate of angiogenesis [3]. There is no cure for this deadly dis-
E-mail address: Min.Li@uth.tmc.edu (M. Li). ease and the multimodal treatment for GBM typically consists of surgical

0304-419X/$ – see front matter © 2013 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.bbcan.2013.04.003
L. Chen et al. / Biochimica et Biophysica Acta 1836 (2013) 158–165 159

resection followed by concomitant chemotherapy and focal radiotherapy. and tumor suppressor genes in GBM pathogenesis (Table 1). We will
Temozolomide, an oral alkylating agent, is considered the standard care also discuss xenograft models as well as the clinical significance of
for GBM treatment [2]. other animal models including the canine, zebrafish, and fruit fly
GBM itself is divided into two categories. Primary GBM mainly af- models in glioma investigations.
fects older patient population with a mean age of 62 years old and
has a predilection to affect men. This type of GBM is referred to as 2. Mouse models
the “de novo” glioblastoma because it is a fully developed tumor
resulted from accumulation of multiple genetic aberrations instead One of the first documented mouse model for cancer study can be
of arising from low-grade precursors. In contrast, the secondary traced back to 1916 when Lathrop and Loeb published the effects of hor-
GBM results from gradual progression from low-grade astrocytoma mones on the development of tumors in mice [6]. Since then, mice have
(WHO grade II) or anaplastic astrocytoma (WHO grade III) over a pe- remained a favorite animal model to study human diseases due to their
riod of 5–10 years. Secondary GBM is often diagnosed in younger evolutionary similarities and fast generation time. Mouse models can
population with a median age of 45 years old and strikes women help to probe the underlying etiological cause and the detailed mecha-
with a higher proportion [4]. Primary GBM accounts for up to 95% of nism of tumorigenesis. To date, there are four predominant strategies to
GBM cases [5]. model malignancies in mice: chemical mutagen-induced, xenograft
Several crucial GBM-contributing genetic aberrations such as transplantation, germline genetic modification, and somatic genetic mod-
EGFR amplification, loss of tumor suppressor genes p53, INK4a/ARF, ification mouse models [7]. In this review, we will focus on the most wide-
and PTEN have been well documented (Fig. 1). The impact of these ly employed xenograft model and the genetic engineered mouse model.
mutations in tumor initiation and progression has been investigated
both in vitro and in vivo. Like many other cancer types, the genetically 2.1. Xenograft mouse models
engineered mouse (GEM) models allow manipulation of the GBM-
contributing genes at the molecular level. The ex vivo xenograft Mouse brain tumor models have been employed for research since
mouse models also offer clinical values in drug screening and gene the mid-1970s [8]. Xenograft mouse models are typically used in pre-
function investigations. Interestingly, other animal models such as ca- clinical trials to test the efficacy of novel therapeutic agents as well as
nine models have been used extensively in GBM studies due to their in studying the gene functions in glioma pathogenesis. In these
high incidence rate of spontaneous intracranial neoplasia and the re- mouse models, human or mice glioma cell lines are injected under
semblance to human tumors. the skin or into the brain of immunocompromised mice. Kaye and
In this review, we will focus on various genetically engineered colleagues [9] injected 10 6 cells of rat C6 glioma both subcutaneously
mouse models that have assisted unraveling the role of the oncogenes and intracranially and found exclusively visible tumor growth in mice

Normal ependymal cell ependymomas


EGFR amplification
P16INK4a deletion
TP53 TP53 mutation
Low-grade astrocytoma mutation Anaplastic astrocytoma PTEN mutation Secondary glioblastoma
(WHO grade II) (WHO grade III) (WHO grade IV)

TP53
mutation

Normal astrocyte
↑ hypercellularity ↑ hypercellularity
Atypical nuclei Atypical nuclei ↑ hypercellularity
High mitotic activity Atypical nuclei
High mitotic activity
EGFR amplification Angiogenesis
P16INK4a deletion Necrosis
TP53 mutation
PTEN mutation Primary glioblastoma
(WHO grade IV)
“de novo”

Normal oligodendrocyte oligodendrocytoma

Fig. 1. The key genetic alterations involved in primary and secondary GBM initiation and progression. Primary GBM, responsible for up to 95% of GBM cases, resulted from accu-
mulation of multiple genetic aberrations instead of arising from low-grade precursor seen in secondary GBM. Both primary and secondary GBM involves EGFR amplification,
TP53 deletion as well as mutations of INK4/ARF and PTEN.
160 L. Chen et al. / Biochimica et Biophysica Acta 1836 (2013) 158–165

Table 1
Transgenic and genetically engineered mouse models for glioblastoma multiforme.

Mouse model Significant findings References

RCAS-EGFR; tva-INK4a-ARF−/− EGFR mutation alone is insufficient to induce glioma Holland [34]
RCAS-EGFR; tva-INK4a-ARF+/− unless the loss of INK4a-ARF occurs concomitantly
GFAP-V12Ha-ras; GFAP-EGFRVIII Aberrant EGFR level is involved in glioma progression Wei et al. [50]
rather than initiation and it can give rise to both
astrocytoma & oligodendrocytoma
S100β-V-erb(EGFR); S100β-INK4a-ARF−/− Rapid tumor development from low-grade to Weiss et al. [36]
S100β-V-erb(EGFR); S100β-INK4a-ARF+/− high-grade with shorter latency and increased penetration
CAGGS-Cre; EGFRVIII; INK4a−/−; PTEN−/− EGFRVIII alone is insufficient to induce cell transformation Zhu et al. [37]
and only when concomitant loss of tumor suppresser gene
INK4a along with PTEN can promote a fully-penetrant,
rapid onset malignant glioma that resembles to human GBM
TgG(ΔZ)T121; PTEN−/− (pRB inactivated through T21 expression) PTEN inactivation in pRB-inactivated model gives rise to Xiao et al. [46]
focal hypercellularity, enhancing astrocytoma invasiveness
as well as promoting angiogenesis
RCAS-KRas; PTEN−/− The mice display similar histopathological characteristics Hu et al. [47]
such as increased cell density, pseudopalisading necrosis,
microvascular proliferation
p53−/−; Nf1−/−; PTEN+/− Heterozygosities of PTEN, Nf1 and p53 cause accelerated Kwon et al. [51]
glioma formation similar to primary “de novo” GBM
+/−
Ntva-INK4a-ARF ; RCAS-KRas; RCAS-AKT The deletion of INK4a-ARF locus in addition to the activation Uhrbom et al. [49]
of KRas and AKT permits GBM formation from differentiated
astrocytes as well as enhances gliomageneis from neural progenitors.
Nf1+/−; p53+/− The mouse model displays different stages of glioma including GBM. Reilly et al. [59]
P53−/−; Nf1−/−; GFAP-Cre TP53 loss prior to or concomitant to Nf1 loss is required for Zhu et al. [62]
P53+/−; Nf1−/−; GFAP-Cre malignant astrocytoma formation when early loss of Nf1 fails
P53+/−; Nf1+/−; GFAP-Cre to induce gliomagenesis

with intracranial injection. When the cells are introduced into the these flank xenografts and cultured them before injecting into the
frontal lobe of the brain, localized tumor growths were observed in brains of the nude mice. They found this heterotopic-to-orthotopic ap-
both neonatal and adult mice in a reproducible manner. proach for establishing intracranial tumor has a 100% success rate
One of the most well-known glioma xenograft models is the U251 with minimal (b1%) mortality rate. This short-term flank xenograft
glioma model established by Ponten et al. [10], and over time this cell model retains some of the genetic characterization involved in GBM tu-
line has been used both in subcutaneous and intracranial mouse morigenesis such as aberrant level of amplification and overexpression
models. Due to the difference in gene expression profile as well as the of EGFR gene. The retention of genetic abnormalities along with the
microenvironment, the intracranial mouse model injected with U251 highly mitotic and invasive features observed in this model contributes
cell lines better recapitulate the histopathological feature of GBM com- to the versatility of their use in neuro-oncological research. Although
pared to the subcutaneous model [11]. Not only did it demonstrate in- this method successfully preserved the invasive nature of tumor cells,
filtrative invasion into brain parenchyma and significant foci of necrosis and endothelial proliferation were not observed. Lee and col-
palisading necrosis microscopically, it also reveals striking resemblance leagues [19] cultured the GBM cells under NBE growth conditions
to human GBMs under immunohistochemical analysis with positive consisting of serum-free Neurobasal media supplemented with basic
GFAP, S100B, and Vimentin staining [12]. Moreover, the losses of FGF and EGF and found that these glioma tumor cells more closely
tumor suppressor genes p53 and PTEN along with the deletion of mimic the phenotype and genotype of primary tumors. Due to glioma's
INK4a/ARF are observed [13]. Another notable cell line established by highly proliferative nature and high recurrence rate, Bello et al. [20] cre-
Ponten and colleagues [10] is the U87 glioma model. Unlike the U251 ated xenograft mouse models with human cell lines U87 and D566 in
glioma model, U87 displays a non-diffusely infiltrative growth pattern order to assess the post-surgical treatment efficacy for recurring glioma.
with a well-demarcated tumor border that is rarely accompanied by ne- Twenty days after tumor cell implantation, the primary tumors were
crotic foci [13,14]. However, the more homogenous tumor vasculature surgically resected followed by administration of endogenous inhibitors
and leakier vessels in the U87 model allows greater entry by systemic PEX and a fragment of platelet factor 4 (PF-4/CTF). They found that the
therapeutic drugs, making it a superior model for evaluating tumor an- systemic administration of PEX or PF-4/CTF after tumor resection signif-
giogenesis and anti-angiogenic therapeutic approaches [15]. icantly improved the survival and delayed glioma reoccurrence in the
Although these models display well-defined tumor with predict- xenograft mouse models. Although some xenograft models can partially
able proliferation rates, the histopathological feature of the tumors recapitulate human GBM histological features and maintain key molec-
do not always recapitulate what is observed in their human counter- ular characteristics, the differences in tumor microenvironment as well
part [16]. Several factors limit the utility of xenograft models, as there as lack of endogenous spontaneous tumor initiation diminish the clini-
are significant differences in selective pressures occurring during cell cal value of xenograft models.
culture in comparison to the natural brain environment [1]. In addi-
tion, these xenograft gliomas in immunocompromised mice grow in 2.2. Genetically engineered mouse models
the absence of a natural tumor-harboring environment. Other xeno-
graft mouse models include the U1242 MG intracranial model created Genetically Engineered Mouse (GEM) models are manipulated at
by Zhao et al. [17]. This intracranial xenograft mouse model expresses the molecular level by either germline modification or somatic cell
high level of matrix metalloproteinase (MMP-9) and shows extensive gene transfer. In germline-modification models, DNA is incorporated
infiltration and hypervascularity, making it a valuable model to study into totipotent cells prior to the developmental stage at which the
GBM invasion and angiogenesis. germline forms and usually involves introducing DNA into gametes
To overcome many shortcomings in recapitulating GBM invasive- such as oocyte, egg or early embryos, or embryonic stem (ES) cells.
ness in most established GBM cell lines, Giannini and colleagues [18] The first germline GEM models for brain tumors were described by
injected the tumor cells subcutaneously into the flank of mice, excised Brinster et al. in 1984 [21] as they microinjected eggs with simian
L. Chen et al. / Biochimica et Biophysica Acta 1836 (2013) 158–165 161

virus SV40 gene containing the metallothionein fusion gene. This ran- we will discuss each mutation as well as the mouse models that help
dom integration of viral oncogene significantly lowers the threshold to provide some insights on the pathogenesis of GBM.
for other mutation and increases the likelihood of a secondary muta-
tion to occur that is typically seen during tumor development. 3.1. Epidermal growth factor receptor
Germline modifications typically include gain-of function, loss-of-
function, and chromosome engineering [22]. The epidermal growth factor receptor (EGFR) is a transmembrane
Another strategy employed in GEM is the somatic gene transfer, glycoprotein that regulates the growth of epithelial cells. Aberrant
which is not heritable and targets only a specific subset of cell popula- EGFR expression is the most frequently detected genetic defects ob-
tion. Fisher and colleagues [23] expressed the avian retroviral receptor, served in GBM and consists of three major mechanisms for oncogenic
TVA, under various mammalian promoters in transgenic mice and signaling. The most common mechanism involves overexpression or
infected the mice with avian leukosis virus derived gene vectors. This amplification of the wild-type receptor. Alternatively, the increase
RCAS/tv-a system for oncogene delivery is more specific and allows a in oncogenic signaling may be due to increased autocrine secretion
single transgenic mouse line to be evaluated for multiple lesions. Unlike of EGF-family ligands, leading to activation of the wild-type EGF sig-
germline gene transfer that requires extensive breeding to combine naling. Lastly, oncogenic signaling is a direct result of constitutive re-
multiple mutations, this system offers a much faster oncogene delivery. ceptor activation in a mutant variant EGFRvIII [29], which is the most
However, secondary events are less likely to occur in this strategy and common variant of EGFR resulted from deletion of exons 2 to 7 of the
thus tumor formation is the direct result of introduced oncogene with- EGFR gene. This critical variant is observed in approximately 40% of pri-
out other genetic events in tumorigenesis. Due to the direct injection of mary GBM and results in an in-frame deletion of 801 amino acids
oncogene-carrying retrovirus into the brain tissues, local injury and in- [30–32]. EGFRvIII's inability to bind ligand renders it constitutively ac-
flammatory response may alter the gene expression. tive, conferring growth advantage for glioma cells [33]. EGFR activation
The glial fibrillary acidic protein (GFAP), an intermediate filament has also been found to contribute to drug resistance in cancer cells [29].
protein found exclusively in astrocytes, is often employed as a pro- Holland and colleagues [34] initially developed a transgenic system
moter in transgenic mice. The very first transgenic mouse model for expressing the ALV subgroup A receptor TVA from the Nestin promoter
astrocytomas was reported by Danks et al. [24]. They expressed so that the constitutively active mutant form of EGFR can be transferred
SV40 T large T antigen, which was found to bind and inactivate p53 via a RCAS vector. The resulting transgenic mice with elevated EGFR failed
and Rb under the control of GFAP promoter [25]. These GFAP/T Ag display any glioma-like lesions; however, in conjunction with INK4a/ARF
mice displayed rapid transformation of T Ag-expressing astrocytic loss, glioma-like lesions with high cell density and vascular proliferation
cells with astrocytoma-like secondary structures; however, all mice are induced in these mice. This work demonstrates EGFR alone does not
died within 30 days postnatally. Weissenberger and colleagues [26] lead to gliomagenesis unless another genetic event such as concomitant
created a transgenic mouse model that constitutively expresses active loss of INK4a/ARF. The presence of EGFR mutation alone is insufficient
Src kinase in astrocytes under the control of GFAP gene regulatory ele- to induce gliomagenesis was further confirmed by Ding et al. [35]. They
ments. Src kinase is a tyrosine kinase that binds and activated EGF recep- used ES cell-mediated transgenesis to establish mouse models that
tor, however, integration of the constitutively active v-src gene alone did expressed either wild-type EGFR or mutant EGFRvIII in astrocytes under
not result in tumor formation. When the GFAP promoter was used to ex- the control of GFAP promoter. The authors found both models had
press an oncogenic mutant form of H-Ras (V12H), the resulting astrocyto- an increased number of astrocytes in the brain without glioma
mas demonstrated histopathological similarities to the human disease formation. They further created a construct that combined both mutant
including high mitotic activity, infiltration, necrosis, and increased vascu- EGFRvIII and oncogenic RAS overexpression, and this GFAP-V12Ha-ras;
larity, and the tumors also exhibited other common mutations associated GFAP-EGFRvIII double transgenic mouse led to accelerated glioma
with astrocytoma [27]. In order to exam the role pRB plays in astrocytoma formation with histopathological feature of oligodendrogliomas and
formation under the GFAP promoter, Xiao et al. [28] used a truncated ver- mixed oligoastrocytoma. They came into conclusion that aberrant
sion of SV40 T antigen mutant-T121, and found astrocyte-specific pRB in- EGFR level is involved in glioma progression rather than initiation, and
activation caused widespread brain abnormalities. dysregulated EGFR expression can give rise to both astrocytomas and
Since xenograft mouse model requires immunocompromised mice to oligodendrocytomas. In a separate study, Weiss and colleague [36] creat-
prevent rejection of human tumor cells, the study of GBM tumorigenesis ed a mouse model that expressed v-erbB, a transforming homologue of
is limited by the modified tumor immunology of these mice. In the con- EGFR, under the control of S100β promoter with a goal to recapitulate
trary, GEM models can be established in immunocompetent animals, high levels of EGFR expression during glioma formation. Most tumors
allowing a more realistic microenvironment for spontaneous and endog- found in this S100-v-erbB model shared histopathological features of
enous tumor growth. The manipulation at the genetic level of GEM low-grade oligodendroglioma with less than 10% displayed resemblance
models allows thorough investigation of key mutations in a spatial- and to that of astrocytomas. The authors suggested that overexpression of
temporal-specific fashion. Appropriate interaction between tumor and EGFR occurs early in oligodendrocytoma pathogenesis but late in astrocy-
stromal tissues is also observed in GEM models. With the readily available toma formation. To further evaluate the significance of EGFR overex-
advanced imaging techniques, non-invasive monitoring of the tumor pression, they knocked out either tumor suppressor gene INK4a/ARF or
growth is no longer an issue for the GEM models. However, the GEM p13 in addition to the S100-v-erbB construct and found rapid developed
model has its own weakness in relatively poor prediction of drug thera- tumors from low-grade to higher-grade with shorter latency and higher
peutic response, high cost, time consuming and slow tumor development. penetrance in both models. A more recent work by Zhu et al. [37] also
The xenograft model still holds significant clinical value in initial drug demonstrated the constitutively active mutant EGFRvIII alone is not
screening and studying gene functions. In the next section, we discuss adequate to promote cell transformation in vivo in their Cre-induced
the most frequently observed mutations in gliomagenesis and the corre- conditional transgenic model. However, concomitant loss of the tumor
sponding GEM models. suppressor gene INK4a/ARF along with PTEN resulted in glioma with
characteristics resembling to GBM.

3. Common genetic mutations in GBM 3.2. PTEN

Many crucial GBM-contributing genetic aberrations such as epidermal PTEN (Phosphatase and tensin homologue) acts as a tumor suppressor
growth factor receptor (EGFR) amplification, loss of tumor suppressor gene through its phosphatase activity keeping cell proliferation in check
genes p53, INK4a/ARF and PTEN have been identified. In this section, [38,39]. PTEN is thought to antagonize the function of PI3K and induce
162 L. Chen et al. / Biochimica et Biophysica Acta 1836 (2013) 158–165

cell cycle arrest and apoptosis in glioma cells [40]. In addition to its role as does not bind to CDKs, instead, it inhibits MDM2, an ubiquitin ligase
a cell proliferation inhibitor, inactivation of PTEN is found to promote an that targets p53 for degradation.
undifferentiated state with high self-renewal and tumorigenic potential The first transgenic mouse was created by Serrano and colleagues who
[41]. PTEN mutation is found in approximately 30% primary GBMs, usu- deleted this locus in mouse, resulting in the loss of both P16INK4a and
ally by complete loss of its locus on chromosome 10q, however, this P19ARF and the development of tumors at an early age [56]. This particular
mutation is rarely detected in secondary GBMs [42,43]. PTEN mutation study demonstrates that INK4a/ARF locus plays a key role in suppressing
in patients with Cowden disease also showed higher incidence of glio- tumor growth. Interestingly, Kamijo and colleagues [57] used a conven-
mas development compared to normal individuals [44]. tional targeting vector to replace ARF exon 1β with a neomycin resistance
It has been shown that PTEN-deficient human glioma cell lines are gene in mouse embryonic stem cells so that the mice expressed only the
highly invasive and when PTEN activity is restored, the cell invasion is transcript encoding p16INK4a but not P19ARF. Surprisingly, these ARF-/-
significantly decreased [45]. To study the astrocytoma suppression mice developed tumors that are virtually identical to that of INK4a/ARF
mechanisms of PTEN, Xiao et al. [46] conditionally inactivated PTEN null mice early in life, suggesting loss of ARF plays a dominant role in
with localized somatic retroviral (MSCV)-Cre in a GEM astrocytoma glioma pathogenesis. The genetic effect of INK4a/ARF is often studied in
model with inactivated pRB. Inactivation of pRB family proteins in- conjunction with other mutations such as constitutively active EGFR
duced aberrant proliferation and apoptosis in astrocytes. The local- overexpression [34,36,37]. Uhrbom and colleagues also combined
ized PTEN inactivation in these mice alleviated apoptosis induced INK4a/ARF loss in addition to one of their models in which K-Ras
from pRB inactivation and gave rise to focal hypercellularity, enhanc- and AKT signaling are activated [49]. They used the RCAS/tv-a system to
ing astrocytoma invasiveness and a more pronounced angiogenesis. combine gene transfer of activated forms of Akt and KRas into nestin-
The protein kinase AKT typically promotes cell survival and prolifer- expressing neural progenitor cells and observed glioma formation with
ation by activating a downstream effector mammalian Target of histopathological features resembling human GBM from neural progeni-
Rapamycin (mTOR) that has been found to be overexpressed without tor cells [48]. However, these induced GBMs were only derived from neu-
identified mutation in most GBMs. To investigate the relationship be- ral progenitor cells but not from differentiated astrocytes.
tween AKT and PTEN, Hu and coworkers [47] injected a combination
of cells producing RCAS-KRas and RCAS-Cre intracranially to 37 3.4. TP53
PTEN−/− mice and these mice displayed similar histopathological char-
acteristics such as increased cell density, pseudopalisading necrosis and The tumor suppressor gene TP53 regulates the cell cycle and is com-
microvascular proliferation that resemble to the KRas+AKT-induced monly inactivated in various types of cancer. TP53 mutation creates geno-
GBM in their previous study [48,49]. These findings suggested the asso- mic instability and inhibits apoptosis. Unlike other genetic alterations,
ciation of an oncogenic effect of AKT activity in the context of PTEN loss. TP53 mutations occur more frequently in secondary GBMs (>60%) than
They also found loss of PTEN alone was insufficient to induce GBMs as in primary GBMs (~10%) [58]. In order to explore the role of TP53 in glio-
none of the 32 PTEN-deficient mice injected with RCAS-Cre displayed ma, Reilly and colleagues constructed the Nf1+/−p53+/− model [59].
tumor formation. Although PTEN mutation or deletion is frequently Since Nf1 mutation is associated with increased risk of optic gliomas, as-
found in GBM, it requires other genetic event to give rise to GBM forma- trocytomas and gliobasltoams as well as its close proximity with TP53
tion. Wei and colleagues generated a conditional PTEN-/- model by on chromosome 11 [60,61], they hypothesized that Nf1 and TP53 are
crossing mice with null PTEN alleles to mice that express Cre rarely separated by recombination and can be treated as a single genetic
recombinase under the control of the GFAP promoter [50]. The condi- event in gliomagenesis. As they predicted, the combination of Nf1 and
tional PTEN knockout mice showed characteristics of generalized sei- TP53 mutations gave rise to malignant tumors. In a similar study, Zhu et
zures and all died by 6 weeks of age. The brains of these mice weighed al. [62] manipulated Nf1 and TP53 under the control of human GFAP pro-
approximately 30% more than that of control with a 3-fold increase in moter in three different mouse strains P53−/−Nf1−/−GFAP-CreP53+/−;
the number of astrocytes, demonstrating an increase in brain mass cor- Nf1−/−GFAP-Cre, and P53+/−Nf1+/−GFAP-Cre. Not only did they find
relating with increased astroglial cell proliferation. However, no glioma TP53 inactivation along with Nf1 loss can lead to the development of
formation was observed in these conditional PTEN mice possibly due to human malignant astrocytomas, they came to the conclusion that TP53
their early lethality before glioma formation could take place. This sug- loss prior to or concomitant to Nf1 loss is essential for malignant astrocy-
gests PTEN inactivation by itself only contributes to glioma progression toma formation. This finding is consistent with previously studies regard-
instead of initiating gliomagenesis. The role of PTEN in tumor progres- ing TP53 mutation as an early genetic event in glioma formation [1].
sion rather than tumor initiation was further confirmed by Kwon et al.
[51]. The authors introduced a loxP-PTEN allele in the Nf1- and 4. The canine model
p53-deficient astrocytoma model that was previously found to develop
malignant astrocytoma. The presence of somatic heterozygosities of Employing dogs as model in cancer research has been documented
PTEN, Nf1 and p53 cause accelerated glioma formation similar to prima- for over 40 years [63]. The canine genome exhibits closer evolutionary
ry “de novo” GBM and significantly shortens the survival of mice. All relationship to human compared to the mouse counterpart. The infiltra-
GEM mouse models mentioned above consistently demonstrate the tive nature of spontaneous GBM in dogs as well as their larger brain size
role of PTEN in promoting tumorigenesis rather than glioma initiation. makes them an appealing model for testing novel therapeutic treat-
ment for GBM [64]. Viral-induced non-spontaneous brain tumor has
3.3. INK4a/ARF been used in canine models [65,66]; however, spontaneous brain
tumor represents the true nature of tumor initiation and progression
The INK4a/ARF locus on chromosome 9p21 by differential splicing in humans. Intracranial neoplasm in dogs has an incidence rate of ap-
and alternate reading frame encodes two distinct proteins, p16 INK4a proximately 20 per 100,000 according to a study in United Kingdom
and P19ARF, that ultimately links the retinoblastoma and p53 tumor [67]. Among brain tumor types, meningiomas and gliomas are the most
suppressor pathways, respectively [52]. P16 INK4a is a cyclin-dependent frequently reported in canine models with 43% and 32% of incidence, re-
kinase inhibitor that induces cell cycle arrest in the G1 phase by binding spectively. Dogs that are seven years old or older have the highest inci-
to and inhibiting CDK4/6, as sequential phosphorylation of Rb by CDK4/ dence of brain tumors among domestic animals [68]. The average age of
6 is required for cell cycle progression. Homozygous deletions at the dogs at the time of onset of astrocytomas is 8.6 years [69]. Two common
INK4a/ARF locus are identified in 57% of GBMs [53], while loss of hetero- mutations associated with human astrocytomas, TP53 and EGFR, are
zygosity (LOH) on chromosome 13q where Rb gene is located is ob- expressed in an aberrant level in 35% and 23% of canine astrocytomas
served in 45% of glioblastomas [54,55]. In contrast to P16 INK4a, ARF models respectively. Interestingly, intracranial neoplasia occurs far more
L. Chen et al. / Biochimica et Biophysica Acta 1836 (2013) 158–165 163

frequently in dogs than human counterpart with almost a 4-fold increase 4 months, large brood size, transparent embryos for better visual ob-
in incidence rate with 14.5 per 100,000 canines every year [70,71]. servation and many other advantages make zebrafish an excellent
Dog model has been used to investigate the cell-mediated immunity model for medical research. Feitsma et al. [80] investigated the role
against spontaneous gliomas [72]. Ingram and colleagues [72] studied of defective mismatch repair (MMR) in cancer and found brain neo-
the immunotherapy for recurrent malignant glioma in dogs by plasms the MMR-deficient zebrafish models. Both TP53 mutant and
implanting stimulated autologous lymphocytes into the tumor bed fol- PTEN mutant zebrafish have also been created and found to develop
lowing surgical debulking, and they observed clinical improvement and cancer, however, the cancer grown from these zebrafish models are
decrease in tumor size in all dogs. The therapeutic value of brachyther- not glioma-specific.
apy was investigated by Stubbs et al. [73]. The authors implanted the Using the drosophila model, Read and colleagues [81] created the
brachytherapy applicator, an inflatable balloon catheter, in the resec- Gal4-UAS system with the repo-Gal driver to co-activate EGFR-Ras and
tion cavity in dog brain and inflated the balloon catheter with Iotrex PI3K in a glial-specific manner. The glial precursors in this drosophila
containing iodine-125 as a reliable radiation delivery method to target model developed tumor resembling to human glioma. Witte and col-
canine brain tumors. Chauvet et al. [74] injected the recombinant ade- leagues [82] also created similar glioma models using the Gal4-UAS
novirus vector bearing the Escherichia coli beta-galactosidase reporter system to overexpress homolog of human tyrosine kinase receptors
gene into meningioma of canine models as a gene therapy for spontane- under control of the glia-specific promoter reversed polarity (repo) in
ous CNS neoplasia. Xiong et al. [46] developed a conditional cytotoxic/ Drosophila. The authors found the overexpression of activated EGFR
immunotherapeutic approach employing adenoviral vector (ads) that and PI3K both resulted in enhanced proliferation and migration of larval
encodes human soluble fms-like tyrosine kinase 3 ligand (hsFlt3L) in glial cells, promoting their tumor-like overgrowth. Efforts in using
dogs bearing spontaneous GBM and found that dogs received subcuta- zebrafish as well as Drosophilas have facilitated many significant dis-
neous injections of human recombinant Flt3L displayed circulating den- coveries and will continue to shed new lights in cancer research.
dritic cells, demonstrating hsFlt3L's ability to induce the proliferation of
canine dendritic cells. The resulting induced dendritic cells are similar 6. Conclusion
to that induced by canine cytokine cocktail consisting of IL-4 and
GM-CSF. Canine models were also adopted to study the convection- Glioma has plagued approximately 30,000 patients in the United
enhanced delivery (CED) that utilized pressure gradient on the tip to States every year. Despite vigorous research efforts, the overall survival
allow an increase volume of drugs past the blood-brain barrier with rate has struggled to improve, lingering at approximately a year after
minimal toxicity [75]. Stoica and colleague [76] also identified cancer initial diagnosis. Animal models have provided a glimpse into various
stem cell (CSC) in dog GBMs and examined the self-renewal ability of aspects of brain tumor biology, becoming an indispensible tool for can-
these glioblastoma cells with single colony formation and found both cer research. Mouse models, both xenograft and genetically engineered,
clone formation rate and subclone formation rate to be 100%. These are used most commonly for cancer research due to their similar mam-
dog GBM cells expressed CD133, the CSC surface marker reported in malian genetic makeup and relatively fast generation time. Many genes
human gliomas. To elucidate the ability of CSC's ability to initiate tumor- that contribute to gliomagenesis have been manipulated in genetically
igenesis upon xenograft transplantation, they injected the tumor cells engineered mouse models to elucidate the genetic contribution as
intracranially into nude mice, which all develop tumors characterized well as the intricate mechanisms of glioma development. Interestingly,
with high cellular heterogeneity, neovascularization and necrosis. the canine model is of emerging significance due to its even closer evo-
Higgins and colleague [77] identified overexpression of EGFR, PDGFRα lutionary relationship to humans. The infiltrative nature and high spon-
and IGFBP2 in spontaneous canine glioma, which is consistent with taneous rate of canine GBM make them an appealing model to assess
their human counterpart. novel therapeutic GBM interventions. Zebrafish and the fruit fly are
The dogs have become a valuable model to study brain tumor because among other models used in brain cancer research, offering many ad-
of their closer revolutionary relationship to human as well as their larger vantages such as lower cost and even shorter generation time although
brain size compared to the murine models [78]. Due to the complexity of they exhibit less human cancer histopathological features than those of
human tumor environment and host immune interactions, the majority the mouse and canine models. Despite the dismal prognosis of GBM,
of successful cancer therapies administered in mice fails to show the these animal models could potentially lead to groundbreaking discover-
same efficacy in their human counterpart while some drug toxicities ob- ies in cancer research.
served later in human were not first identified in mice. Since there's no
treatment regimen deemed the “gold standard”, trials of various veteri- Conflict of interest
nary treatment in canine cancer are advantageous of early testing of
novel therapies [79]. Therefore, the canine model could be a more suitable The authors declare no conflict of interest.
candidate for preclinical drug screening as well as clinical trials for other
therapeutic strategies. Yet, the consistency of experimental studies relies
Acknowledgements
heavily on pet owners' compliance and consent, which can limit the ac-
cessibility of these canine models by clinicians. The number of canine
This work was supported in part by the National Institutes of
cases available for study is still much lower compared to that of murine
Health (NIH) grants R21CA133604, R01CA138701, Dr. Marnie Rose
models despite the higher rate of spontaneous brain tumor formation in
Foundation, and the William and Ella Owens Medical Research Foun-
dogs. Nowadays the canine model for brain cancer research employs
dation (M. Li).
only the spontaneous model. Unlike the murine models in which the
tumor can be induced by genetic alteration, the same progress has not
been made in canine model due to technical challenges, higher cost and References
ethical dilemmas [79]. Nevertheless, the canine model holds great poten- [1] E.C. Holland, Gliomagenesis: genetic alterations and mouse models, Nat. Rev.
tial for human cancer research due to its phenotypic diversity as well as its Genet. 2 (2001) 120–129.
[2] R. Stupp, W.P. Mason, M.J. van den Bent, M. Weller, B. Fisher, M.J. Taphoorn, K.
closer cancer pathogenesis resemblance to humans.
Belanger, A.A. Brandes, C. Marosi, U. Bogdahn, J. Curschmann, R.C. Janzer, S.K.
Ludwin, T. Gorlia, A. Allgeier, D. Lacombe, J.G. Cairncross, E. Eisenhauer, R.O.
5. Other animal models Mirimanoff, Radiotherapy plus concomitant and adjuvant temozolomide for glio-
blastoma, N. Engl. J. Med. 352 (2005) 987–996.
[3] D.J. Brat, E.G. Van Meir, Vaso-occlusive and prothrombotic mechanisms associat-
Other animal models employed in brain tumor include zebrafish ed with tumor hypoxia, necrosis, and accelerated growth in glioblastoma, Lab.
and Drosophila melanogaster (fruit fly). Shorter gestation time of 3– Invest. 84 (2004) 397–405.
164 L. Chen et al. / Biochimica et Biophysica Acta 1836 (2013) 158–165

[4] E.A. Maher, F.B. Furnari, R.M. Bachoo, D.H. Rowitch, D.N. Louis, W.K. Cavenee, R.A. [30] K.J. Hatanpaa, S. Burma, D. Zhao, A.A. Habib, Epidermal growth factor receptor in
DePinho, Malignant glioma: genetics and biology of a grave matter, Genes Dev. 15 glioma: signal transduction, neuropathology, imaging, and radioresistance, Neo-
(2001) 1311–1333. plasia 12 (2010) 675–684.
[5] H. Ohgaki, P. Dessen, B. Jourde, S. Horstmann, T. Nishikawa, P.L. Di Patre, C. [31] N. Sugawa, A.J. Ekstrand, C.D. James, V.P. Collins, Identical splicing of aberrant epi-
Burkhard, D. Schuler, N.M. Probst-Hensch, P.C. Maiorka, N. Baeza, P. Pisani, Y. dermal growth factor receptor transcripts from amplified rearranged genes in
Yonekawa, M.G. Yasargil, U.M. Lutolf, P. Kleihues, Genetic pathways to glioblasto- human glioblastomas, Proc Natl Acad Sci USA 87 (1990) 8602–8606.
ma: a population-based study, Cancer Res. 64 (2004) 6892–6899. [32] J. Schlegel, A. Merdes, G. Stumm, F.K. Albert, M. Forsting, N. Hynes, M. Kiessling,
[6] A.E. Lathrop, L. Loeb, Further investigations on the origin of tumors in mice. III. On Amplification of the epidermal-growth-factor-receptor gene correlates with dif-
the part played by internal secretion in the spontaneous development of tumors, ferent growth behaviour in human glioblastoma, Int. J. Cancer 56 (1994) 72–77.
The Journal of cancer research 1 (1916) 1–19. [33] Y. Zhu, L.F. Parada, The molecular and genetic basis of neurological tumours, Nat.
[7] C. Dai, E.C. Holland, Glioma models, Biochim. Biophys. Acta 1551 (2001) M19–M27. Rev. Cancer 2 (2002) 616–626.
[8] M. Salvati, E. Ramundo Orlando, R. Causo, G. Innocenzi, R. Capone, F. Cosentino Jr., [34] E.C. Holland, A mouse model for glioma: biology, pathology, and therapeutic op-
F. Cosentino, Tumors of the central nervous system induced by ionizing radiation. portunities, Toxicol. Pathol. 28 (2000) 171–177.
Update on their pathology and presentation of a case, G. Ital. Oncol. 10 (1990) [35] H. Ding, P. Shannon, N. Lau, X. Wu, L. Roncari, R.L. Baldwin, H. Takebayashi, A.
15–18. Nagy, D.H. Gutmann, A. Guha, Oligodendrogliomas result from the expression of
[9] A.H. Kaye, G. Morstyn, I. Gardner, K. Pyke, Development of a xenograft glioma an activated mutant epidermal growth factor receptor in a RAS transgenic
model in mouse brain, Cancer Res. 46 (1986) 1367–1373. mouse astrocytoma model, Cancer Res. 63 (2003) 1106–1113.
[10] J. Ponten, Neoplastic human glia cells in culture, In Human Tumor Cells, in Vitro [36] W.A. Weiss, M.J. Burns, C. Hackett, K. Aldape, J.R. Hill, H. Kuriyama, N. Kuriyama,
(1975) 175–185. N. Milshteyn, T. Roberts, M.F. Wendland, R. DePinho, M.A. Israel, Genetic determi-
[11] K. Camphausen, B. Purow, M. Sproull, T. Scott, T. Ozawa, D.F. Deen, P.J. Tofilon, nants of malignancy in a mouse model for oligodendroglioma, Cancer Res. 63
Orthotopic growth of human glioma cells quantitatively and qualitatively influ- (2003) 1589–1595.
ences radiation-induced changes in gene expression, Cancer Res. 65 (2005) [37] H. Zhu, J. Acquaviva, P. Ramachandran, A. Boskovitz, S. Woolfenden, R. Pfannl, R.T.
10389–10393. Bronson, J.W. Chen, R. Weissleder, D.E. Housman, A. Charest, Oncogenic EGFR sig-
[12] V.L. Jacobs, P.A. Valdes, W.F. Hickey, J.A. De Leo, Current review of in vivo GBM ro- naling cooperates with loss of tumor suppressor gene functions in gliomagenesis,
dent models: emphasis on the CNS-1 tumour model, ASN Neuro 3 (2011) e00063. Proc. Natl. Acad. Sci. U.S.A. 106 (2009) 2712–2716.
[13] E. Radaelli, R. Ceruti, V. Patton, M. Russo, A. Degrassi, V. Croci, F. Caprera, G. [38] E.C. Chu, A.S. Tarnawski, PTEN regulatory functions in tumor suppression and cell
Stortini, E. Scanziani, E. Pesenti, R. Alzani, Immunohistopathological and neuro- biology, Med. Sci. Monit. 10 (2004) RA235–RA241.
imaging characterization of murine orthotopic xenograft models of glioblastoma [39] M. Cully, H. You, A.J. Levine, T.W. Mak, Beyond PTEN mutations: the PI3K pathway
multiforme recapitulating the most salient features of human disease, Histol. as an integrator of multiple inputs during tumorigenesis, Nat. Rev. Cancer 6
Histopathol. 24 (2009) 879–891. (2006) 184–192.
[14] M. Candolfi, J.F. Curtin, W.S. Nichols, A.G. Muhammad, G.D. King, G.E. Pluhar, E.A. [40] L. Simpson, R. Parsons, PTEN: life as a tumor suppressor, Exp. Cell Res. 264 (2001)
McNiel, J.R. Ohlfest, A.B. Freese, P.F. Moore, J. Lerner, P.R. Lowenstein, M.G. Castro, 29–41.
Intracranial glioblastoma models in preclinical neuro-oncology: neuropathologi- [41] H. Zheng, H. Ying, H. Yan, A.C. Kimmelman, D.J. Hiller, A.J. Chen, S.R. Perry, G.
cal characterization and tumor progression, J. Neurooncol 85 (2007) 133–148. Tonon, G.C. Chu, Z. Ding, J.M. Stommel, K.L. Dunn, R. Wiedemeyer, M.J. You, C.
[15] N.A. de Vries, J.H. Beijnen, O. van Tellingen, High-grade glioma mouse models and Brennan, Y.A. Wang, K.L. Ligon, W.H. Wong, L. Chin, R.A. DePinho, p53 and Pten
their applicability for preclinical testing, Cancer Treat. Rev. 35 (2009) 714–723. control neural and glioma stem/progenitor cell renewal and differentiation, Na-
[16] S.D. Finkelstein, P. Black, T.P. Nowak, C.M. Hand, S. Christensen, P.W. Finch, Histo- ture 455 (2008) 1129–1133.
logical characteristics and expression of acidic and basic fibroblast growth factor [42] T.C.G.A.R. Network, Comprehensive genomic characterization defines human
genes in intracerebral xenogeneic transplants of human glioma cells, Neurosur- glioblastoma genes and core pathways, Nature 455 (2008) 1061–1068.
gery 34 (1994) 136–143. [43] Y. Tohma, C. Gratas, W. Biernat, A. Peraud, M. Fukuda, Y. Yonekawa, P. Kleihues, H.
[17] Y. Zhao, A. Xiao, C.G. diPierro, J.E. Carpenter, R. Abdel-Fattah, G.T. Redpath, M.B. Ohgaki, PTEN (MMAC1) mutations are frequent in primary glioblastomas (de
Lopes, I.M. Hussaini, An extensive invasive intracranial human glioblastoma xe- novo) but not in secondary glioblastomas, J. Neuropathol. Exp. Neurol. 57
nograft model: role of high level matrix metalloproteinase 9, Am. J. Pathol. 176 (1998) 684–689.
(2010) 3032–3049. [44] K. Ichimura, H. Ohgaki, P. Kleihues, V.P. Collins, Molecular pathogenesis of astro-
[18] C. Giannini, J.N. Sarkaria, A. Saito, J.H. Uhm, E. Galanis, B.L. Carlson, M.A. cytic tumours, J. Neurooncol 70 (2004) 137–160.
Schroeder, C.D. James, Patient tumor EGFR and PDGFRA gene amplifications [45] D. Koul, R. Parthasarathy, R. Shen, M.A. Davies, S.A. Jasser, S.K. Chintala, J.S. Rao,
retained in an invasive intracranial xenograft model of glioblastoma multiforme, Y. Sun, E.N. Benvenisite, T.J. Liu, W.K. Yung, Suppression of matrix
Neuro Oncol. 7 (2005) 164–176. metalloproteinase-2 gene expression and invasion in human glioma cells by
[19] J. Lee, S. Kotliarova, Y. Kotliarov, A. Li, Q. Su, N.M. Donin, S. Pastorino, B.W. Purow, MMAC/PTEN, Oncogene 20 (2001) 6669–6678.
N. Christopher, W. Zhang, J.K. Park, H.A. Fine, Tumor stem cells derived from glio- [46] A. Xiao, C. Yin, C. Yang, A. Di Cristofano, P.P. Pandolfi, T. Van Dyke, Somatic induc-
blastomas cultured in bFGF and EGF more closely mirror the phenotype and ge- tion of Pten loss in a preclinical astrocytoma model reveals major roles in disease
notype of primary tumors than do serum-cultured cell lines, Cancer Cell 9 progression and avenues for target discovery and validation, Cancer Res. 65
(2006) 391–403. (2005) 5172–5180.
[20] L. Bello, C. Giussani, G. Carrabba, M. Pluderi, V. Lucini, M. Pannacci, D. Caronzolo, [47] X. Hu, P.P. Pandolfi, Y. Li, J.A. Koutcher, M. Rosenblum, E.C. Holland, mTOR pro-
G. Tomei, R. Villani, F. Scaglione, R.S. Carroll, A. Bikfalvi, Suppression of malignant motes survival and astrocytic characteristics induced by Pten/AKT signaling in
glioma recurrence in a newly developed animal model by endogenous inhibitors, glioblastoma, Neoplasia 7 (2005) 356–368.
Clinical Cancer Research: An Official Journal of the American Association for Can- [48] E.C. Holland, J. Celestino, C. Dai, L. Schaefer, R.E. Sawaya, G.N. Fuller, Combined ac-
cer Research 8 (2002) 3539–3548. tivation of Ras and Akt in neural progenitors induces glioblastoma formation in
[21] R.L. Brinster, H.Y. Chen, A. Messing, T. van Dyke, A.J. Levine, R.D. Palmiter, Trans- mice, Nat. Genet. 25 (2000) 55–57.
genic mice harboring SV40 T-antigen genes develop characteristic brain tumors, [49] L. Uhrbom, C. Dai, J.C. Celestino, M.K. Rosenblum, G.N. Fuller, E.C. Holland,
Cell 37 (1984) 367–379. Ink4a-Arf loss cooperates with KRas activation in astrocytes and neural progeni-
[22] E.I. Fomchenko, E.C. Holland, Mouse models of brain tumors and their applica- tors to generate glioblastomas of various morphologies depending on activated
tions in preclinical trials, Clinical Cancer Research: An Official Journal of the Akt, Cancer Res. 62 (2002) 5551–5558.
American Association for Cancer Research 12 (2006) 5288–5297. [50] Q. Wei, L. Clarke, D.K. Scheidenhelm, B. Qian, A. Tong, N. Sabha, Z. Karim, N.A.
[23] G.H. Fisher, S. Orsulic, E. Holland, W.P. Hively, Y. Li, B.C. Lewis, B.O. Williams, H.E. Bock, R. Reti, R. Swoboda, E. Purev, J.F. Lavoie, M.L. Bajenaru, P. Shannon, D.
Varmus, Development of a flexible and specific gene delivery system for produc- Herlyn, D. Kaplan, R.M. Henkelman, D.H. Gutmann, A. Guha, High-grade glioma
tion of murine tumor models, Oncogene 18 (1999) 5253–5260. formation results from postnatal pten loss or mutant epidermal growth factor re-
[24] R.A. Danks, J.M. Orian, M.F. Gonzales, S.S. Tan, B. Alexander, K. Mikoshiba, A.H. ceptor expression in a transgenic mouse glioma model, Cancer Res. 66 (2006)
Kaye, Transformation of astrocytes in transgenic mice expressing SV40 T antigen 7429–7437.
under the transcriptional control of the glial fibrillary acidic protein promoter, [51] C.H. Kwon, D. Zhao, J. Chen, S. Alcantara, Y. Li, D.K. Burns, R.P. Mason, E.Y. Lee, H.
Cancer Res. 55 (1995) 4302–4310. Wu, L.F. Parada, Pten haploinsufficiency accelerates formation of high-grade as-
[25] J. Chen, G.J. Tobin, J.M. Pipas, T. Van Dyke, T-antigen mutant activities in vivo: trocytomas, Cancer Res. 68 (2008) 3286–3294.
roles of p53 and pRB binding in tumorigenesis of the choroid plexus, Oncogene [52] S.M. Ivanchuk, S. Mondal, P.B. Dirks, J.T. Rutka, The INK4A/ARF locus: role in cell
7 (1992) 1167–1175. cycle control and apoptosis and implications for glioma growth, J. Neurooncol
[26] J. Weissenberger, J.P. Steinbach, G. Malin, S. Spada, T. Rulicke, A. Aguzzi, Develop- 51 (2001) 219–229.
ment and malignant progression of astrocytomas in GFAP-v-src transgenic mice, [53] S.M. Ranuncolo, M. Varela, A. Morandi, J. Lastiri, S. Christiansen, E. Bal de Kier
Oncogene 14 (1997) 2005–2013. Joffe, M.G. Pallotta, L. Puricelli, Prognostic value of Mdm2, p53 and p16 in patients
[27] H. Ding, L. Roncari, P. Shannon, X. Wu, N. Lau, J. Karaskova, D.H. Gutmann, J.A. with astrocytomas, J. Neurooncol 68 (2004) 113–121.
Squire, A. Nagy, A. Guha, Astrocyte-specific expression of activated p21-ras re- [54] R. Nishikawa, F.B. Furnari, H. Lin, W. Arap, M.S. Berger, W.K. Cavenee, H.J.Su.
sults in malignant astrocytoma formation in a transgenic mouse model of Huang, Loss of P16INK4 expression is frequent in high grade gliomas, Cancer
human gliomas, Cancer Res. 61 (2001) 3826–3836. Res. 55 (1995) 1941–1945.
[28] A. Xiao, H. Wu, P.P. Pandolfi, D.N. Louis, T. Van Dyke, Astrocyte inactivation of the [55] K. Ichimura, E.E. Schmidt, H.M. Goike, V.P. Collins, Human glioblastomas with no
pRb pathway predisposes mice to malignant astrocytoma development that is ac- alterations of the CDKN2A (p16INK4A, MTS1) and CDK4 genes have frequent mu-
celerated by PTEN mutation, Cancer Cell 1 (2002) 157–168. tations of the retinoblastoma gene, Oncogene 13 (1996) 1065–1072.
[29] P.H. Huang, A.M. Xu, F.M. White, Oncogenic EGFR signaling networks in glioma, [56] M. Serrano, H. Lee, L. Chin, C. Cordon-Cardo, D. Beach, R.A. DePinho, Role of the
Sci. Signal. 2 (2009) re6. INK4a locus in tumor suppression and cell mortality, Cell 85 (1996) 27–37.
L. Chen et al. / Biochimica et Biophysica Acta 1836 (2013) 158–165 165

[57] T. Kamijo, F. Zindy, M.F. Roussel, D.E. Quelle, J.R. Downing, R.A. Ashmun, G. [71] P.R. Gavin, J.R. Fike, P.J. Hoopes, Central nervous system tumors, Semin. Vet. Med.
Grosveld, C.J. Sherr, Tumor suppression at the mouse INK4a locus mediated by Surg. (Small Anim.) 10 (1995) 180–189.
the alternative reading frame product p19ARF, Cell 91 (1997) 649–659. [72] M. Ingram, J.G. Buckwalter, D.B. Jacques, D.B. Freshwater, R.M. Abts, G.B. Techy,
[58] F.F. Lang, D.C. Miller, M. Koslow, E.W. Newcomb, Pathways leading to glioblasto- K. Miyagi, C.H. Shelden, R.W. Rand, L.W. English, Immunotherapy for recurrent
ma multiforme: a molecular analysis of genetic alterations in 65 astrocytic tu- malignant glioma: an interim report on survival, Neurol. Res. 12 (1990)
mors, J. Neurosurg. 81 (1994) 427–436. 265–273.
[59] K.M. Reilly, D.A. Loisel, R.T. Bronson, M.E. McLaughlin, T. Jacks, Nf1;Trp53 mutant [73] J.B. Stubbs, R.H. Frankel, K. Schultz, I. Crocker, D. Dillehay, J.J. Olson, Preclinical
mice develop glioblastoma with evidence of strain-specific effects, Nat. Genet. 26 evaluation of a novel device for delivering brachytherapy to the margins of
(2000) 109–113. resected brain tumor cavities, J. Neurosurg. 96 (2002) 335–343.
[60] S.A. Sorensen, J.J. Mulvihill, A. Nielsen, Long-term follow-up of von Recklinghau- [74] A.E. Chauvet, P.P. Kesava, C.S. Goh, B. Badie, Selective intraarterial gene delivery
sen neurofibromatosis: survival and malignant neoplasms, N. Engl. J. Med. 314 into a canine meningioma, J. Neurosurg. 88 (1998) 870–873.
(1986) 1010–1015. [75] P.J. Dickinson, R.A. LeCouteur, R.J. Higgins, J.R. Bringas, B. Roberts, R.F. Larson, Y.
[61] D.H. Gutmann, New insights into the neurofibromatoses, Curr. Opin. Neurol. 7 Yamashita, M. Krauze, C.O. Noble, D. Drummond, D.B. Kirpotin, J.W. Park, M.S.
(1994) 166–171. Berger, K.S. Bankiewicz, Canine model of convection-enhanced delivery of lipo-
[62] Y. Zhu, F. Guignard, D. Zhao, L. Liu, D.K. Burns, R.P. Mason, A. Messing, L.F. Parada, somes containing CPT-11 monitored with real-time magnetic resonance imaging:
Early inactivation of p53 tumor suppressor gene cooperating with NF1 loss in- laboratory investigation, J. Neurosurg. 108 (2008) 989–998.
duces malignant astrocytoma, Cancer Cell 8 (2005) 119–130. [76] G. Stoica, G. Lungu, H. Martini-Stoica, S. Waghela, J. Levine, R. Smith 3rd, Identi-
[63] M. Paoloni, C. Khanna, Translation of new cancer treatments from pet dogs to fication of cancer stem cells in dog glioblastoma, Vet. Pathol. 46 (2009)
humans, Nat. Rev. Cancer 8 (2008) 147–156. 391–406.
[64] D.N. Louis, Molecular pathology of malignant gliomas, Annu. Rev. Pathol. 1 (2006) [77] R.J. Higgins, P.J. Dickinson, R.A. LeCouteur, A.W. Bollen, H. Wang, L.J. Corely, L.M.
97–117. Moore, W. Zang, G.N. Fuller, Spontaneous canine gliomas: overexpression of EGFR,
[65] P.C. Warnke, P. Molnar, G.D. Lapin, A. Kuruvilla, D.R. Groothuis, The effects of PDGFRalpha and IGFBP2 demonstrated by tissue microarray immunophenotyping,
dexamethasone on transcapillary transport in experimental brain tumors: II. Ca- J. Neurooncol 98 (2010) 49–55.
nine brain tumors, J. Neurooncol 25 (1995) 29–38. [78] E.F. Kirkness, V. Bafna, A.L. Halpern, S. Levy, K. Remington, D.B. Rusch, A.L.
[66] H.T. Whelan, J.A. Clanton, R.E. Wilson, N.B. Tulipan, Comparison of CT and MRI brain Delcher, M. Pop, W. Wang, C.M. Fraser, J.C. Venter, The dog genome: survey se-
tumor imaging using a canine glioma model, Pediatr. Neurol. 4 (1988) 279–283. quencing and comparative analysis, Science 301 (2003) 1898–1903.
[67] J.M. Dobson, S. Samuel, H. Milstein, K. Rogers, J.L. Wood, Canine neoplasia in the [79] K. Hansen, C. Khanna, Spontaneous and genetically engineered animal models;
UK: estimates of incidence rates from a population of insured dogs, J. Small use in preclinical cancer drug development, Eur. J. Cancer 40 (2004) 858–880.
Anim. Pract. 43 (2002) 240–246. [80] H. Feitsma, R.V. Kuiper, J. Korving, I.J. Nijman, E. Cuppen, Zebrafish with muta-
[68] S.A. Snyder, M.W. Dewhirst, M.L. Hauck, The role of hypoxia in canine cancer, Vet. tions in mismatch repair genes develop neurofibromas and other tumors, Cancer
Comp. Oncol. 6 (2008) 213–223. Res. 68 (2008) 5059–5066.
[69] E.T. Keller, B.R. Madewell, Locations and types of neoplasms in immature dogs: 69 [81] R.D. Read, W.K. Cavenee, F.B. Furnari, J.B. Thomas, A drosophila model for
cases (1964–1989), J. Am. Vet. Med. Assoc. 200 (1992) 1530–1532. EGFR-Ras and PI3K-dependent human glioma, PLoS Genet. 5 (2009) e1000374.
[70] G.L. Heidner, J.N. Kornegay, R.L. Page, R.K. Dodge, D.E. Thrall, Analysis of survival in a ret- [82] H.T. Witte, A. Jeibmann, C. Klambt, W. Paulus, Modeling glioma growth and inva-
rospective study of 86 dogs with brain tumors, J. Vet. Intern. Med. 5 (1991) 219–226. sion in Drosophila melanogaster, Neoplasia 11 (2009) 882–888.

You might also like