Skip to main content
Erschienen in: Journal of Neuro-Oncology 3/2017

Open Access 20.10.2017 | Topic Review

Current trends in mouse models of glioblastoma

verfasst von: Masafumi Miyai, Hiroyuki Tomita, Akio Soeda, Hirohito Yano, Toru Iwama, Akira Hara

Erschienen in: Journal of Neuro-Oncology | Ausgabe 3/2017

Abstract

Glioblastoma is the most deadly brain tumor type and is characterized by a severe and high rate of angiogenesis, remaining an incurable disease in the majority of cases. Mechanistic understanding of glioblastoma initiation and progression is complicated by the complexity of genetic and/or environmental initiating events and lack of clarity regarding the cell or tissue of origin. To determine these mechanisms, mouse models that recapitulate the molecular and histological characteristics of glioblastoma are required. Unlike in other malignancies, viral-mediated mouse models of glioblastoma rather than chemically induced mouse models have been developed because of its sensitivity to viruses. Based on recent molecular analyses reported for human glioblastoma, this review critically evaluates genetically engineered, xenograft, allograft, viral-mediated, and chemically induced mouse models of glioblastoma. Further, we focus on the clinical value of these models by examining their contributions to studies of glioblastoma prevention, tumorigenesis, and chemoresistance.

Introduction

Glioblastoma is the most common and deadly primary brain tumor [1], and the most aggressive type of glioma, a collection of tumors arising from glial cells. It is also termed glioblastoma multiforme because of its complex phenotype. The current standard of care is based on maximal safe surgical resection and concurrent chemoradiation with temozolomide, followed by maintenance chemotherapy, resulting in median survival rates of approximately 15 months [2].
Over the past few years, genomic and proteomic characterization along with robust animal models of glioblastoma have provided invaluable data. In addition, pre-clinical models and a better understanding of the core pathways disrupted in glioblastoma are providing renewed optimism for novel strategies targeting these devastating tumors. Here we discuss the current advances in rodent models, particularly mouse models, of glioblastoma and other gliomas, and how these developments have influenced strategies for therapeutic intervention.

Pathological features of glioblastoma

The 2016 World Health Organization Classification of Tumors of the Central Nervous System uses molecular parameters in addition to histology to define many tumor entities [3]. In this new classification, diffuse gliomas include WHO grade II and grade III astrocytic tumors, grade II and III oligodendrogliomas, and grade IV glioblastomas.
Glioblastomas are divided into three categories in the 2016 CNS WHO classification according to a key genetic prognostic marker, isocitrate dehydrogenase (IDH): glioblastoma, IDH-wildtype; glioblastoma, IDH-mutant; glioblastoma, NOS. IDH-wildtype (about 90% of cases) corresponds most frequently with clinically defined primary or de novo glioblastoma and predominates in patients over 55 years of age [4]. IDH-mutant glioblastoma (about 10% of cases) corresponds closely to so-called secondary glioblastoma with a history of lower-grade diffuse glioma, and preferentially arises in younger patients [4]. Glioblastoma NOS is a diagnosis that is reserved for tumors for which full IDH evaluation cannot be performed or for which test results remain inconclusive.
Pathological characteristics of glioblastoma is typically a highly cellular glioma, usually comprised of poorly differentiated, sometimes pleomorphic tumor cells with nuclear atypia and vigorous mitotic activity. Conspicuous microvascular proliferation and/or necrosis is an essential diagnosis characterization.
Molecular characteristics typical of IDH-wild type glioblastoma include TERT promorter mutations (present in ~ 80% of cases), homozygous deletion of CDKN2A/CDKN2B (~ 60%), loss of chromosomes 10p (~ 50%) and 10q (~ 70%), EGFR alterations (i.e. mutation, rearrangement, altered splicing, and/or amplification; ~ 50%), PTEN mutations(25–30%), and PI3K mutations (~ 25%) [4, 5].

Mouse models of glioblastoma

The use of mice to create suitable models for the study of specific tumors or to investigate the role of candidate genes has obvious advantages. Firstly, manipulation of the mouse genome to create specific genetic changes by microinjection of DNA into fertilized eggs or by homologous recombination in embryonic stem cells is relatively easy compared with other mammalian species such as rats. Another advantage is the availability of inbred strains of mice that are genetically identical, obtained by breeding sibling mice over 20 generations. Since these animals present the same genetic background, they can be compared for their response to a treatment or a genetic modification between different laboratories. The laboratory mouse shares extensive molecular and physiological similarities to humans and is a powerful tool for studying cancer.
Transgenic mouse models offer an opportunity to develop and utilize an easily replenished, reproducible, spontaneously manipulated, and more accurate pre-clinical model of human cancers, which we can use to enhance our molecular knowledge and to test promising therapies. Therefore, mouse models that recapitulate human glioblastoma may be an invaluable tool. However, these conventional genetic approaches, such as transgenics and knockouts, are limited by the time and costs associated with extensive intercrossing of mouse lines. Several new viral vector-mediated genetic approaches offer the ability to directly modify the genome of somatic cells in mouse tissues and these have recently been applied to the rapid generation of complex mouse tumor models that harbor multiple genetic changes. On the other hand, xenograft and allograft models can be used to measure therapeutic responses to drugs more rapidly than genetically engineered or viral vector-mediated models. Chemical carcinogen-induced models are usually generated in rats, and only a small number of instances of chemical carcinogen-induced models are currently known.

Genetically engineered models

The molecular progression of gliomas, like many tumors, involves the accumulation of genetic and epigenetic alterations that result in the loss of tumor suppressor gene function (PTEN, TP53, CDKN2A, RB) or the activation of oncogenic pathways (p21–RAS, PI3K, EGFR, CDK4, MDM2) [68].
There are several examples of aberrant expression of relevant downstream signaling pathways in mouse glioma modeling. These include astrocytomas of varying grades resulting from glial fibrillary acidic protein (GFAP)-regulated expression of v-src [9]. Weissenberger et al. [9] generated a transgenic mouse model for low-grade astrocytoma (early) and high-grade astrocytoma (later) by expressing v-src kinase under the control of GFAP gene regulatory elements in astrocytes.
Ding et al. [10, 11] utilized our initial observation of aberrant activation of the p21–RAS signaling pathway in astrocytomas to develop glioma models using ES transgenesis [11]. Since wild-type EGFR and mutant EGFRvIII are the most common gain-of-function alterations in malignant human astrocytomas, they reported the generation of mice expressing these proteins under the regulation of the GFAP promoter [10, 11].
When the glioblastoma-like tumors are examined in these mice, additional genetic alterations such as those found in human glioblastomas (overexpression of EGFR, CDK4, MDM2; decreased expression of CDKN2A, TP53, PTEN) are present [12].
Zhu et al. [13] reported that loss of TP53 and activation of the RAS pathway via NF1 inactivation in CNS cells is sufficient to cause malignant astrocytoma formation with 100% penetrance.
Overexpression of relevant oncogenic receptors or downstream signaling pathways has also been employed in the development of mouse glioma models. These have been either alone or in combination with mice harboring specific knockouts of relevant cell cycle regulatory proteins. For example, S100 glial precursor promoter-regulated v-ERBB (an activated member of the EGFR family) transgenic mice develop oligodendrogliomas, which are potentiated in terms of shorter latency and increased malignancy when initiated in mice deficient for both p16 and p19 (Cdkn2a-null mice) [14].
Briefly, Bardella et al. [15] have established small nodules like glioma in the subventricular zone. Nes-CreER(T2); Idh1fl(R132H)/+ developed small nodules (up to 1 mm diameter) originating from the subventricular zone at 2–6 weeks after tamoxifen injection.
The nodules expressed proliferation markers, such as Ki67, and retained BrdU label, suggesting that they exhibited a variety of proliferative behaviors. Further, many nodule cells also expressed the astrocytic and NSC marker GFAP and in some lesions, a few cells expressed the neuroblast marker Doublecortin (Dcx).
Table 1 summarizes the currently used and relevant glioma mouse models that recapitulate the hallmarks of human glioblastoma.
Table 1
Genetically engineered and viral vector-mediated transduction mouse models of human glioma
Tumor classification
Transgene
Knockout, knockin
Grade
Incidences
Study
Small nodule like glioma
NES-CreER(T2)
IDH1 R132H knockin
NA*
100% by 2–6 weeks
Bardella et al. [15]
Low-grade astrocytoma
Src transgene
 
II
14% by 2.5–65 weeks
Weissenberger et al. [9]
GFAP-HRAS V12
 
II
95% by 16–24 weeks
Ding et al. [10]
High-grade astrocytoma
Src transgene
 
III
10–20% at later
Weissenberger et al. [9]
GFAP-HRAS V12
Floxed NF1 + Trp53 knockout
III–IV
100% by 2–16 weeks
Ding et al. [10]
GFAP-T121 transgene
PTEN +/−
II–III
100% by 4–32 weeks
Xiao et al. [55]
GFAP-Cre
NF1 + Trp53 cis
II–IV
30–75% by 15–55 weeks
Reilley et al. [56]
HRAS V12 and AKT
 
III–IV
40% by 16–20 weeks
Marumoto et al. [23]
Glioblastoma
GFAP-Cre
Floxed NF1 + Trp53 knockout
II–IV
100% by 10–45 weeks
Zhu et al. [13]
Kras and AKT (RCAS virus)
Cdkn2a knockout
IV
42–49% by 12 weeks
Uhrbom et al. [18]
PDGFB (RCAS virus)
Cdkn2a knockout, Trp53 knockout
IV
100% by 4–7 weeks
Hambardzumyan et al. [19]
EGFRvIII(Ad-Cre virus)
PTEN F/F
II–IV
93% by 6–15 weeks
Wei et al. [21]
HRAS V12 and AKT
Trp53 knockout
IV
100% by 10–13 weeks
Marumoto et al. [23]
NES-CreER
Floxed NF1, Floxed PTEN, Floxed Trp53
III–IV
100% by 24–56 weeks
Alcantara Llaguno et al. [24]
EGFRvIII(Ad-Cre virus)
Cdkn2a, PTEN F/F
IV
100% by 5–13 weeks
Zhu et al. [57]
Low-grade oligodendroglioma
S100b-vERBB transgene
 
II
75% by 52 weeks
Weiss et al. [14]
PDGFB (RCAS virus)
 
II
60% by 12 weeks
Dai et al. [20]
High-grade oligodendroglioma
S100b-vERBB transgene
Cdkn2a knockout
III
90% by 4–24 weeks
Weiss et al. [14]
GFAP-HRAS V12 + GFAP-EGFRvIII
 
III
100% by 2–13 weeks
Ding et al. [11]
Diffuse intrinsic pontine glioma
PDGFB + H3.3K27M (RCAS virus)
Pax3-Tv-a
Trp53 knockout
II–IV
73% by 5–12 weeks
Misuraca et al. [25]
*not applicable

Viral vector-mediated transduction model

In recent years, viral vectors have been extensively used for the generation of mouse models of interest in the study of brain tumors. Several routes for viral vector delivery to the brain are available: intracerebral stereotaxic injection, intrathecal and intraventricular injection, and intravascular infusion with or without modification of the blood–brain barrier. The choice of route for viral vector administration needs to be carefully considered since it affects neuronal cell transduction efficiency and spatial distribution, as well as the level of transgene expression in the infected cells [16]. Intracerebral injection offers the advantages of low toxicity, high local vector concentrations, and localized transgene delivery, but it does not allow wide viral vector distribution and requires invasive surgical intervention. Ubiquitous distribution of viral vectors in the CNS could be achieved by intrathecal or intraventricular injection but these methods do not permit spatial selectivity of delivery and require a large amount of vectors. Finally, intravascular viral vector applications do not require invasive surgical intervention but necessitate the use of high vector concentrations because of losses in peripheral organs such as the liver.
Virally transduced expression of relevant gain-of-function alterations, in combination with transgenic mouse technology, allows one to model such somatic alterations at a later stage in life, though it does not lead to germline colonies. Although the link between a viral etiology and human gliomas is weak, retroviruses that have been engineered to express relevant gain-of-function genes have been used to create glioma models in mice and other mammals [17, 18]. This includes members of the Rous sarcoma virus family and simian sarcoma virus, whose transforming properties result from overexpression of the viral oncogene v-sis, the cellular counterpart of which is c-sis or PDGF-B.
Retroviruses carrying v-sis (PDGFB) injected into normal mice have yielded astrocytic tumors, with varying glioma types generated when injected in Cdkn2a-null mice. One of the best examples of coupling retroviruses to express somatically defined gain-of-function genes in varying cell lineages and genetic backgrounds to model gliomas is the RCAS-tva system [19]. This system results in focal gliomas, the subtype and grade of which varies with the injected retrovirally transduced gene (i.e. PDGFB, EGFRvIII, activated p21–RAS, activated AKT), the lineage of the cell expressing the tva receptor (GFAP, NES) and underlying genetic cell cycle alterations in the mice (null for Cdkn2a, Trp53 etc.). For example, retrovirally transduced expression of v-sis or PDGFB in GFAP-tva mice resulted in oligodendrogliomas or mixed oligoastrocytomas in 40% of the mice, with 60% of NES-tva mice developing similar gliomas [10]. When these experiments were undertaken in Cdkn2a-null mice, the gliomas formed with a shorter latency and were of higher grade [20]. On the other hand, injection of adenovirus containing the EGFRvIII mutant into mice harboring activated RAS led to efficient formation of glioblastoma [21].
Lentiviruses expressing oncogenes such as HRAS or AKT were efficiently introduced into mice expressing Cre-recombinase under varying promoters such as GFAP. Glioblastoma tumors were efficiently formed when lentiviruses harboring activated RAS and AKT were injected into mice expressing GFAP-Cre on a Trp53 heterozygous background [22, 23]. Current studies have allowed the determination that the gliomagenesis potential of mice is greater at a younger age with excision of glioblastoma-relevant genes such as PTEN, NF1, and TP53 [24]. Misuraca et al. [25] have established low- and high-grade glioma, which phenocopies diffuse intrinsic pontine glioma, resulting from injection of Pax3-Tv-a;Trp53 fl/fl mice with RCAS-PDGFB and RCAS-Cre, with or without RCAS-H3.3K27M. In the RCAS/Tv-a glioma mouse model, avian retroviruses (produced from RCAS plasmids) infect mouse cells expressing Tv-a (the receptor for RCAS viruses) [26].

Xenograft and allograft models

For many years, immunodeficient rodents have been an important tool in modeling human glioblastoma. Propagation and testing of glioblastoma in such animals is most commonly accomplished in the subcutaneous flank location (heterotopic), although recent years have seen increased use of orthotopic (intracranial) xenograft models. For both heterotopic and orthotopic studies, xenograft and allograft tumors are usually established from permanent human glioblastoma cell lines.
Invasive orthotopic xenografts have also been established from surgical specimens that were first maintained as tissue spheroids in short-term culture [27, 28]. Finally invasive intracranial tumors have been established from heterotopic xenografts generated by direct transplantation of surgical specimens and subsequently sustained by serial passaging in the flanks of nude mice [29, 30].
Glioblastomas that have been continuously propagated as flank tumors recapitulate this very important and characteristic feature of human glioblastoma following intracranial transfer. Different from the heterotopic transplantation, the direct orthotopic transplantation denies the influences of in vitro culture, provides a proper microenvironment, and preserves the integrity of tumor-initiating cells [31, 32].
Many human and mouse cell lines have been used in xenograft and allograft models (Table 2). Tateishi et al. [33] used the SCID mouse to study the vulnerability of IDH1-mutant cancers to NAD + depletion. Ashizawa et al. [34] used NOD-SCID mice and NOG mice to study the effect of the STAT3 inhibitor STX-0119 on the proliferation of cancer stem-like cells derived from recurrent glioblastoma. Other mouse strains such as athymic nude (Nu/Nu) mice [3537], CD1 nude mice [38], and athymic nude Foxn1-nu mice have also been used [3941].
Table 2
Trends in xenograft and allograft mouse models of glioma
No
Study
Type
Cell line
Tumor histrogy
Genetic change
Animal model
Therapy
Drug administration method
Injection point of cells
Time of initiating the therapy
Duration of treatment
Observation period
1
Tateishi et al. [33]
Xenograft
MGG152(TIC*), HT1080(Human cell line)
Recurrent glioblastoma fibrosarcoma
IDH1 mutant(MGG152) IDH1 R132C mutant (HT1080)
SCID mice 7–10-week old female
NAMPT inhibitor
Oral administration (MGG152) intraperitoneal injection (HT1080)
Right striatum (MGG152) right flank (HT1080)
one week (MGG152) tumor diameters reached 5 mm (HT1080)
1 × /week (MGG152) 4 days/week (HT1080)
About 30 days (MGG152) 17 days (HT1080)
2
Ashizawa et al. [34]
Xenograft
GB-SCC010 GB-SCC026 (primary glioblastoma stem cell lines from patients)
Primary glioblastoma
NA*****
NOD-SCID** mice, NOG*** mice 5–6 week old
STAT3 inhibitor
Oral administration
Subcutaneous
Bearing tumor of > 35mm3
Daily/three weeks
28 days
3
Wykosky et al. [37]
Allograft
Primary ink4a/arf-/- astrocyte (mouse cell line)
Similar to glioblastoma
ΔEGFR-expressing and PTEN wild-type
Athymic mice 6-8 week old female
EGFR inhibitor(gefitinib)
Oral gavage
Cerebral (2 mm lateral and 1 mm anterior to the bregma)
20 days
5 days per week
About 50 days
4
Plowman et al. [35]
Xenograft
SF-295(Human cell line) U251(Human cell line)
Glioblastoma
NA*****
Athymic mice
Temozolomide BCNU
Oral gavage(TMZ)
tail vein injection(BCNU)
Right cerebral hemisphere
1 day
day 1, day 5, day 9
90 days
5
Szabo et al. (2016) [39]
Xenograft
LNT-229 (Human cell line) LN-308(Human cell line)
Glioblastoma
Silencing microRNA-adapted shRNA
CD1-Foxn1nu nude mice 6-12 week old
Neutralizing antibodies to VEGF or PIGF
Intraperitoneal injection
Right striatum
NA*****
Twice weekly
60 days
6
Sharpe et al. [41]
Xenograft
BT111(TIC*) BT116(TIC*)
Primary glioblastoma
Unmethylated MGMT(BT111) NA*****(BT116)
NU-Foxn1nu nude mice
Monoamine oxidase B-activated pro-drug
Tail vein injection
Flank postglenoid foramen
4 weeks (flank model) 90 days (intracranial model)
day 0, day 12, day 23 (flank model) day 115, day 123, day 131
(intracranial model)
36 days (flank model) 307 days(intracranial model)
7
Zeng et al. [58]
Allograft
GL261-Luc(mouse cell line)
Glioblastoma
NA*****
C57BL/6J mice 6-8 week old
Radiation plus anti-PD-1 antibody
Intraperitoneal injection
left striatum(1 mm lateral and
1 mm anterior
to the bregma, 3 mm deep from the cortical surface)
10 days
day 10, day 12, day 14
90 days
8
Zhang et al. [59]
Xenograft and allograft
LN-319(Human cell line) GL261(mouse cell line) [51]
Glioblastoma
NA***** (LN-319) ErbB2 expression(GL261)
NSG**** mice (LN-319) C57BL/6 mice(GL261)
ErbB2/HER2 -Specific NK Cells
Intratumoral injection
Subcutaneous right striatum (depth of 3 mm)
7 days
Weekly for 11 weeks (LN-319) weekly for 3 weeks(GL261)
303 days (LN-319) 200 days (GL261)
9
Parrish et al. [60]
Xenograft
GBM12(TIC*) [67]
Primary glioblastoma
MGMT hypermethylated
NA*****
PARP inhibitor (rucaparib) Temozolomide
Intraperitoneal injection oral gavage
Flank cerebral
NA*****
Days 1–5 every 28 days for 3 cycles
121 days (flank model) 81 days (intracranial model)
10
Gupta et al. [61]
Xenograft
GBM12(TIC*) [67]
Primary glioblastoma
TMZ-mgmt High TMZ-mgmt Low
Athymic mice
PARP inhibitor(veliparib) Temozolomide
 
Flank
Tumor of ~ 100 ± 15mm3
5 days every 28days for 3cycles
About 50 days
11
Garros-Regulez et al. [62]
Xenograft
U251(Human cell line)
Glioblastoma
NA*****
Foxn1nu-Foxn1nu nude mice 8 week old
mTOR inhibition(rapamycin) Temozolomide
Intraperitoneal injection
Flank
1 week
Twice weekly for 12 weeks
About 60 days
12
Hashizume et al. [63]
Xenograft
SF7761(TIC*) SF8628(TIC*) GBM43(TIC*) [68]
Primary pediatric human glioma adult glioblastoma
H3F3A k27M mutation H3F3A k27M mutation MGMT unmethylated
Athymic mice 6 week old female
Demethylase inhibitor
Intraperitoneal injection
Flank brain stem
SF7761: about 50 days SF8628: 56 days GBM43: 5 days
SF7761: daily /10 days SF8628: daily /10 days GBM43: daily/7 days
SF7761: 160 days SF8628: 77 days GBM43: 18 days
13
Mathieu et al. [64]
Xenograft
Hs683 (Human cell line) U373 (Human cell line)
Glioma (Hs683) glioblastoma (U373)
NA*****
Nude mice (immunocompromised mice) 6 week old female
Bevacizumab Temozolomide
Tail vein injection oral administration
Cerebral
5 days
3 times per week for 3 consecutive weeks
80 days
14
Cho et al. [65]
Xenograft
LN443 (Human cell line)
Glioblastoma
Expressing EGFRvIII CT Del1 mutant (by retroviral infection)
SCID mice
Cetuximab erlotinib
Intraperitoneal injection oral administration
Right striatum
1 week
3 times per week
100 days
15
Yoshida et al. [66]
Xenograft
GBM39(TIC*) [69] U87(Human cell line) GBM12(TIC*) [69]
glioblastoma
EGFRvIII amplified (GBM39) expressing EGFRvIII (by retroviral infection:U87) wild-type EGFR(GBM12)
Athymic mice 5 week old female
Pan-ERBB inhibitor
Oral administration
Right caudate putamen
14 days (GBM39) 11 days (U87) 6 days (GBM12)
2 week (GBM39) 21 days (U87) day 10, day 13, day16,day 20, day 23 (GBM12)
66 days (GBM39) 32 days (U87) 70 days(GBM12)
16
Joo et al. [42]
Xenograft
Surgical specimens from glioblastoma patients
Glioblastoma
Depending on specimens
NOG*** mice
NA*****
NA*****
cerebral
With in 12 months
NA*****
About 200 weeks
*Patient-derived tumor initiating cell
**NOD/Shi-Parkdcscid
***NOD/Shi-scid IL-2Rγ-null
****NOD-SCID IL2Rγnull
*****not applicable
A library of orthotopic glioblastoma xenograft models using surgical samples of glioblastoma patients has been established. These patient-derived glioblastoma xenograft (PDX) tumors recapitulated histopathological properties and maintained genomic characteristics of parental glioblastoma in situ [42, 43]. Soeda et al. [44] reported a glioblastoma xenograft model containing heterogeneous subclones derived from a single tumor of a patient. This model may be useful for evaluating cell- and patient-specific drug responses. Patient-derived primary glioma cells might be a good solution but they are sometimes unable to maintain for long in culture and finding an accessible cell type for gliomas might be problematic.
Patient-derived stem cells are used to identify cell functions that are altered by disease, such as Alzheimer’s and Parkinson’s disease and thereby provide a target for drug discovery. Patient-derived glioblastoma stem cells have been generated from xenograft tumors of the glioblastoma surgically resected [45, 46]. Patient-derived glioblastoma stem cells are in nature, formed much larger neurospheres in a short period of time rather than patient-derived glioblastoma cells (not stem cells) [47]. Further, Sancho-Martinez et al. [48] have recently established human induced pluripotent stem (iPS) cells based glioma models in vivo.
Recent progress and expansion in next-generation sequencing (NGS) technologies enable to characterize the cancer genome in a time frame that is corresponding to treatment decisions, providing the chance to potentially increase the therapeutic effect by targeting the genomic alterations driving tumor behavior [49]. To challenge proposed therapy strategies on the patient of gliomas, the “Avatar mouse models”, which are based on the NGS data and generated as the individualized mouse xenografts by transplanting patient-derived tumor cells, have been investigated [50]. The development of PDX and patient-derived neurosphere/stem cell based xenograft models may approve bench testing of treatment strategies derived from the innovative genomic analysis.
There are important caveats to this approach that still need to be addressed in xenograft models. Firstly, the mice do not have an intact immune system. Inflammatory cells may be a critical component to the biology of the tumor and its response to certain drugs, particularly immunotherapy. Secondly, the surrounding stroma and microenvironment is of mouse origin, not human, and may interfere with drug response.

Chemical carcinogen-induced models

Previously, only a chemical carcinogen-induced mouse model, the GL261 model, have been derived from an intracranially-induced methylcholanthrene tumor in C57BL/6 mice [51]. Recently, Johanns et al. [52] have reported that, by the assessment of the ability of the epitopes predicted in silico to be the highest affinity binders to activate tumor-infiltrating T cells harvested from GL261, they have found the mechanisms of the T cell–activating immune-directed therapy, presumably due to its hypermutator phenotype. The ability of gliomas to induce local and systemic immunosuppression restrict the innate defense against tumor growth and the efficacy of adaptive immunotherapy and thus presents a significant challenge to the development of innovative therapies [53].

Spontaneous models

In 1971, H. Fraser has described the first incidence of a spontaneous glioma within the SMA-560 mouse strain. Initially, these tumors, which resembled human anaplastic astrocytomas, were restricted to in vivo studies only, as tumorigenicity was lost with repeated in vitro passaging of tumor explant cultures [54].

Conclusions

Glioblastoma is one of the most problematic cancers to treat. Despite advances in molecular profiling of the disease, information is still lacking, particularly regarding treatment.
As genome-wide sequencing efforts continue in humans, mouse glioma models that better recapitulate the complex genomic landscape of human glioma will be generated. These models will provide increasingly powerful tools for the validation of hypotheses engendered by human genomic data, such as confirming the driver mutations that are causal to oncogenesis, as well as for preclinical testing of personalized therapy.

Acknowledgements

We thank all members of the Department of Tumor Pathology and Neurosurgery at Gifu University Graduate School of Medicine. This work was supported by grants from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (Grant Numbers 26430111 (H. T.) and 26670639 (A. S.)).
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

e.Med Neurologie

Kombi-Abonnement

Mit e.Med Neurologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes, den Premium-Inhalten der neurologischen Fachzeitschriften, inklusive einer gedruckten Neurologie-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Committee of Brain Tumor Registry of Japan: Report of Brain Tumor Registry of Japan (2001–2004). Neurol Med Chir (Tokyo) : 1-102, 2014. Neurol Med Chir (Tokyo) 13 (1):12 Committee of Brain Tumor Registry of Japan: Report of Brain Tumor Registry of Japan (2001–2004). Neurol Med Chir (Tokyo) : 1-102, 2014. Neurol Med Chir (Tokyo) 13 (1):12
2.
Zurück zum Zitat Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, Curschmann J, Janzer RC, Ludwin SK, Gorlia T, Allgeier A, Lacombe D, Cairncross JG, Eisenhauer E, Mirimanoff RO, European Organisation for R, Treatment of Cancer Brain T., Radiotherapy G, National Cancer Institute of Canada Clinical Trials G (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352 (10):987–996. doi:10.1056/NEJMoa043330 CrossRefPubMed Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, Curschmann J, Janzer RC, Ludwin SK, Gorlia T, Allgeier A, Lacombe D, Cairncross JG, Eisenhauer E, Mirimanoff RO, European Organisation for R, Treatment of Cancer Brain T., Radiotherapy G, National Cancer Institute of Canada Clinical Trials G (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352 (10):987–996. doi:10.​1056/​NEJMoa043330 CrossRefPubMed
3.
Zurück zum Zitat Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P, Ellison DW (2016) The 2016 World Health Organization Classification of tumors of the central nervous system: a summary. Acta Neuropathol 131(6):803–820. doi:10.1007/s00401-016-1545-1 CrossRefPubMed Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P, Ellison DW (2016) The 2016 World Health Organization Classification of tumors of the central nervous system: a summary. Acta Neuropathol 131(6):803–820. doi:10.​1007/​s00401-016-1545-1 CrossRefPubMed
5.
Zurück zum Zitat Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR, Zheng S, Chakravarty D, Sanborn JZ, Berman SH, Beroukhim R, Bernard B, Wu CJ, Genovese G, Shmulevich I, Barnholtz-Sloan J, Zou L, Vegesna R, Shukla SA, Ciriello G, Yung WK, Zhang W, Sougnez C, Mikkelsen T, Aldape K, Bigner DD, Van Meir EG, Prados M, Sloan A, Black KL, Eschbacher J, Finocchiaro G, Friedman W, Andrews DW, Guha A, Iacocca M, O’Neill BP, Foltz G, Myers J, Weisenberger DJ, Penny R, Kucherlapati R, Perou CM, Hayes DN, Gibbs R, Marra M, Mills GB, Lander E, Spellman P, Wilson R, Sander C, Weinstein J, Meyerson M, Gabriel S, Laird PW, Haussler D, Getz G, Chin L, Network TR (2013) The somatic genomic landscape of glioblastoma. Cell 155(2):462–477. doi:10.1016/j.cell.2013.09.034 CrossRefPubMedPubMedCentral Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR, Zheng S, Chakravarty D, Sanborn JZ, Berman SH, Beroukhim R, Bernard B, Wu CJ, Genovese G, Shmulevich I, Barnholtz-Sloan J, Zou L, Vegesna R, Shukla SA, Ciriello G, Yung WK, Zhang W, Sougnez C, Mikkelsen T, Aldape K, Bigner DD, Van Meir EG, Prados M, Sloan A, Black KL, Eschbacher J, Finocchiaro G, Friedman W, Andrews DW, Guha A, Iacocca M, O’Neill BP, Foltz G, Myers J, Weisenberger DJ, Penny R, Kucherlapati R, Perou CM, Hayes DN, Gibbs R, Marra M, Mills GB, Lander E, Spellman P, Wilson R, Sander C, Weinstein J, Meyerson M, Gabriel S, Laird PW, Haussler D, Getz G, Chin L, Network TR (2013) The somatic genomic landscape of glioblastoma. Cell 155(2):462–477. doi:10.​1016/​j.​cell.​2013.​09.​034 CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Cancer Genome Atlas Research N (2008) Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455 (7216):1061–1068. doi:10.1038/nature07385 CrossRef Cancer Genome Atlas Research N (2008) Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455 (7216):1061–1068. doi:10.​1038/​nature07385 CrossRef
7.
Zurück zum Zitat Furnari FB, Fenton T, Bachoo RM, Mukasa A, Stommel JM, Stegh A, Hahn WC, Ligon KL, Louis DN, Brennan C, Chin L, DePinho RA, Cavenee WK (2007) Malignant astrocytic glioma: genetics, biology, and paths to treatment. Genes Dev 21(21):2683–2710. doi:10.1101/gad.1596707 CrossRefPubMed Furnari FB, Fenton T, Bachoo RM, Mukasa A, Stommel JM, Stegh A, Hahn WC, Ligon KL, Louis DN, Brennan C, Chin L, DePinho RA, Cavenee WK (2007) Malignant astrocytic glioma: genetics, biology, and paths to treatment. Genes Dev 21(21):2683–2710. doi:10.​1101/​gad.​1596707 CrossRefPubMed
8.
Zurück zum Zitat Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Siu IM, Gallia GL, Olivi A, McLendon R, Rasheed BA, Keir S, Nikolskaya T, Nikolsky Y, Busam DA, Tekleab H, Diaz LA Jr, Hartigan J, Smith DR, Strausberg RL, Marie SK, Shinjo SM, Yan H, Riggins GJ, Bigner DD, Karchin R, Papadopoulos N, Parmigiani G, Vogelstein B, Velculescu VE, Kinzler KW (2008) An integrated genomic analysis of human glioblastoma multiforme. Science 321(5897):1807–1812. doi:10.1126/science.1164382 CrossRefPubMedPubMedCentral Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Siu IM, Gallia GL, Olivi A, McLendon R, Rasheed BA, Keir S, Nikolskaya T, Nikolsky Y, Busam DA, Tekleab H, Diaz LA Jr, Hartigan J, Smith DR, Strausberg RL, Marie SK, Shinjo SM, Yan H, Riggins GJ, Bigner DD, Karchin R, Papadopoulos N, Parmigiani G, Vogelstein B, Velculescu VE, Kinzler KW (2008) An integrated genomic analysis of human glioblastoma multiforme. Science 321(5897):1807–1812. doi:10.​1126/​science.​1164382 CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Ding H, Roncari L, Shannon P, Wu X, Lau N, Karaskova J, Gutmann DH, Squire JA, Nagy A, Guha A (2001) Astrocyte-specific expression of activated p21-ras results in malignant astrocytoma formation in a transgenic mouse model of human gliomas. Cancer Res 61(9):3826–3836PubMed Ding H, Roncari L, Shannon P, Wu X, Lau N, Karaskova J, Gutmann DH, Squire JA, Nagy A, Guha A (2001) Astrocyte-specific expression of activated p21-ras results in malignant astrocytoma formation in a transgenic mouse model of human gliomas. Cancer Res 61(9):3826–3836PubMed
11.
Zurück zum Zitat Ding H, Shannon P, Lau N, Wu X, Roncari L, Baldwin RL, Takebayashi H, Nagy A, Gutmann DH, Guha A (2003) Oligodendrogliomas result from the expression of an activated mutant epidermal growth factor receptor in a RAS transgenic mouse astrocytoma model. Cancer Res 63(5):1106–1113PubMed Ding H, Shannon P, Lau N, Wu X, Roncari L, Baldwin RL, Takebayashi H, Nagy A, Gutmann DH, Guha A (2003) Oligodendrogliomas result from the expression of an activated mutant epidermal growth factor receptor in a RAS transgenic mouse astrocytoma model. Cancer Res 63(5):1106–1113PubMed
14.
Zurück zum Zitat Weiss WA, Burns MJ, Hackett C, Aldape K, Hill JR, Kuriyama H, Kuriyama N, Milshteyn N, Roberts T, Wendland MF, DePinho R, Israel MA (2003) Genetic determinants of malignancy in a mouse model for oligodendroglioma. Cancer Res 63(7):1589–1595PubMed Weiss WA, Burns MJ, Hackett C, Aldape K, Hill JR, Kuriyama H, Kuriyama N, Milshteyn N, Roberts T, Wendland MF, DePinho R, Israel MA (2003) Genetic determinants of malignancy in a mouse model for oligodendroglioma. Cancer Res 63(7):1589–1595PubMed
15.
Zurück zum Zitat Bardella C, Al-Dalahmah O, Krell D, Brazauskas P, Al-Qahtani K, Tomkova M, Adam J, Serres S, Lockstone H, Freeman-Mills L, Pfeffer I, Sibson N, Goldin R, Schuster-Boeckler B, Pollard PJ, Soga T, McCullagh JS, Schofield CJ, Mulholland P, Ansorge O, Kriaucionis S, Ratcliffe PJ, Szele FG, Tomlinson I (2016) Expression of Idh1R132H in the murine subventricular zone stem cell niche recapitulates features of early gliomagenesis. Cancer Cell 30(4):578–594. doi:10.1016/j.ccell.2016.08.017 CrossRefPubMedPubMedCentral Bardella C, Al-Dalahmah O, Krell D, Brazauskas P, Al-Qahtani K, Tomkova M, Adam J, Serres S, Lockstone H, Freeman-Mills L, Pfeffer I, Sibson N, Goldin R, Schuster-Boeckler B, Pollard PJ, Soga T, McCullagh JS, Schofield CJ, Mulholland P, Ansorge O, Kriaucionis S, Ratcliffe PJ, Szele FG, Tomlinson I (2016) Expression of Idh1R132H in the murine subventricular zone stem cell niche recapitulates features of early gliomagenesis. Cancer Cell 30(4):578–594. doi:10.​1016/​j.​ccell.​2016.​08.​017 CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat von Jonquieres G, Mersmann N, Klugmann CB, Harasta AE, Lutz B, Teahan O, Housley GD, Frohlich D, Kramer-Albers EM, Klugmann M (2013) Glial promoter selectivity following AAV-delivery to the immature brain. PLoS ONE 8(6):e65646. doi:10.1371/journal.pone.0065646 CrossRef von Jonquieres G, Mersmann N, Klugmann CB, Harasta AE, Lutz B, Teahan O, Housley GD, Frohlich D, Kramer-Albers EM, Klugmann M (2013) Glial promoter selectivity following AAV-delivery to the immature brain. PLoS ONE 8(6):e65646. doi:10.​1371/​journal.​pone.​0065646 CrossRef
17.
Zurück zum Zitat Holland EC, Celestino J, Dai C, Schaefer L, Sawaya RE, Fuller GN (2000) Combined activation of Ras and Akt in neural progenitors induces glioblastoma formation in mice. Nat Genet 25(1):55–57. doi:10.1038/75596 CrossRefPubMed Holland EC, Celestino J, Dai C, Schaefer L, Sawaya RE, Fuller GN (2000) Combined activation of Ras and Akt in neural progenitors induces glioblastoma formation in mice. Nat Genet 25(1):55–57. doi:10.​1038/​75596 CrossRefPubMed
18.
Zurück zum Zitat Uhrbom L, Dai C, Celestino JC, Rosenblum MK, Fuller GN, Holland EC (2002) Ink4a-Arf loss cooperates with KRas activation in astrocytes and neural progenitors to generate glioblastomas of various morphologies depending on activated Akt. Cancer Res 62(19):5551–5558PubMed Uhrbom L, Dai C, Celestino JC, Rosenblum MK, Fuller GN, Holland EC (2002) Ink4a-Arf loss cooperates with KRas activation in astrocytes and neural progenitors to generate glioblastomas of various morphologies depending on activated Akt. Cancer Res 62(19):5551–5558PubMed
19.
20.
Zurück zum Zitat Dai C, Celestino JC, Okada Y, Louis DN, Fuller GN, Holland EC (2001) PDGF autocrine stimulation dedifferentiates cultured astrocytes and induces oligodendrogliomas and oligoastrocytomas from neural progenitors and astrocytes in vivo. Genes Dev 15(15):1913–1925. doi:10.1101/gad.903001 CrossRefPubMedPubMedCentral Dai C, Celestino JC, Okada Y, Louis DN, Fuller GN, Holland EC (2001) PDGF autocrine stimulation dedifferentiates cultured astrocytes and induces oligodendrogliomas and oligoastrocytomas from neural progenitors and astrocytes in vivo. Genes Dev 15(15):1913–1925. doi:10.​1101/​gad.​903001 CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Wei Q, Clarke L, Scheidenhelm DK, Qian B, Tong A, Sabha N, Karim Z, Bock NA, Reti R, Swoboda R, Purev E, Lavoie JF, Bajenaru ML, Shannon P, Herlyn D, Kaplan D, Henkelman RM, Gutmann DH, Guha A (2006) High-grade glioma formation results from postnatal pten loss or mutant epidermal growth factor receptor expression in a transgenic mouse glioma model. Cancer Res 66(15):7429–7437. doi:10.1158/0008-5472.CAN-06-0712 CrossRefPubMed Wei Q, Clarke L, Scheidenhelm DK, Qian B, Tong A, Sabha N, Karim Z, Bock NA, Reti R, Swoboda R, Purev E, Lavoie JF, Bajenaru ML, Shannon P, Herlyn D, Kaplan D, Henkelman RM, Gutmann DH, Guha A (2006) High-grade glioma formation results from postnatal pten loss or mutant epidermal growth factor receptor expression in a transgenic mouse glioma model. Cancer Res 66(15):7429–7437. doi:10.​1158/​0008-5472.​CAN-06-0712 CrossRefPubMed
28.
Zurück zum Zitat Mahesparan R, Read TA, Lund-Johansen M, Skaftnesmo KO, Bjerkvig R, Engebraaten O (2003) Expression of extracellular matrix components in a highly infiltrative in vivo glioma model. Acta Neuropathol 105(1):49–57. doi:10.1007/s00401-002-0610-0 PubMed Mahesparan R, Read TA, Lund-Johansen M, Skaftnesmo KO, Bjerkvig R, Engebraaten O (2003) Expression of extracellular matrix components in a highly infiltrative in vivo glioma model. Acta Neuropathol 105(1):49–57. doi:10.​1007/​s00401-002-0610-0 PubMed
29.
Zurück zum Zitat Antunes L, Angioi-Duprez KS, Bracard SR, Klein-Monhoven NA, Le Faou AE, Duprez AM, Plenat FM (2000) Analysis of tissue chimerism in nude mouse brain and abdominal xenograft models of human glioblastoma multiforme: what does it tell us about the models and about glioblastoma biology and therapy? J Histochem Cytochem 48(6):847–858. doi:10.1177/002215540004800613 CrossRefPubMed Antunes L, Angioi-Duprez KS, Bracard SR, Klein-Monhoven NA, Le Faou AE, Duprez AM, Plenat FM (2000) Analysis of tissue chimerism in nude mouse brain and abdominal xenograft models of human glioblastoma multiforme: what does it tell us about the models and about glioblastoma biology and therapy? J Histochem Cytochem 48(6):847–858. doi:10.​1177/​0022155400048006​13 CrossRefPubMed
30.
31.
Zurück zum Zitat Shu Q, Wong KK, Su JM, Adesina AM, Yu LT, Tsang YT, Antalffy BC, Baxter P, Perlaky L, Yang J, Dauser RC, Chintagumpala M, Blaney SM, Lau CC, Li XN (2008) Direct orthotopic transplantation of fresh surgical specimen preserves CD133 + tumor cells in clinically relevant mouse models of medulloblastoma and glioma. Stem Cells 26(6):1414–1424. doi:10.1634/stemcells.2007-1009 CrossRefPubMed Shu Q, Wong KK, Su JM, Adesina AM, Yu LT, Tsang YT, Antalffy BC, Baxter P, Perlaky L, Yang J, Dauser RC, Chintagumpala M, Blaney SM, Lau CC, Li XN (2008) Direct orthotopic transplantation of fresh surgical specimen preserves CD133 + tumor cells in clinically relevant mouse models of medulloblastoma and glioma. Stem Cells 26(6):1414–1424. doi:10.​1634/​stemcells.​2007-1009 CrossRefPubMed
32.
Zurück zum Zitat Suggitt M, Bibby MC (2005) 50 years of preclinical anticancer drug screening: empirical to target-driven approaches. Clin Cancer Res 11(3):971–981PubMed Suggitt M, Bibby MC (2005) 50 years of preclinical anticancer drug screening: empirical to target-driven approaches. Clin Cancer Res 11(3):971–981PubMed
33.
Zurück zum Zitat Tateishi K, Wakimoto H, Iafrate AJ, Tanaka S, Loebel F, Lelic N, Wiederschain D, Bedel O, Deng G, Zhang B, He T, Shi X, Gerszten RE, Zhang Y, Yeh JR, Curry WT, Zhao D, Sundaram S, Nigim F, Koerner MV, Ho Q, Fisher DE, Roider EM, Kemeny LV, Samuels Y, Flaherty KT, Batchelor TT, Chi AS, Cahill DP (2015) Extreme vulnerability of IDH1 mutant cancers to NAD + depletion. Cancer Cell 28(6):773–784. doi:10.1016/j.ccell.2015.11.006 CrossRefPubMedPubMedCentral Tateishi K, Wakimoto H, Iafrate AJ, Tanaka S, Loebel F, Lelic N, Wiederschain D, Bedel O, Deng G, Zhang B, He T, Shi X, Gerszten RE, Zhang Y, Yeh JR, Curry WT, Zhao D, Sundaram S, Nigim F, Koerner MV, Ho Q, Fisher DE, Roider EM, Kemeny LV, Samuels Y, Flaherty KT, Batchelor TT, Chi AS, Cahill DP (2015) Extreme vulnerability of IDH1 mutant cancers to NAD + depletion. Cancer Cell 28(6):773–784. doi:10.​1016/​j.​ccell.​2015.​11.​006 CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Ashizawa T, Miyata H, Iizuka A, Komiyama M, Oshita C, Kume A, Nogami M, Yagoto M, Ito I, Oishi T, Watanabe R, Mitsuya K, Matsuno K, Furuya T, Okawara T, Otsuka M, Ogo N, Asai A, Nakasu Y, Yamaguchi K, Akiyama Y (2013) Effect of the STAT3 inhibitor STX-0119 on the proliferation of cancer stem-like cells derived from recurrent glioblastoma. Int J Oncol 43(1):219–227. doi:10.3892/ijo.2013.1916 CrossRefPubMed Ashizawa T, Miyata H, Iizuka A, Komiyama M, Oshita C, Kume A, Nogami M, Yagoto M, Ito I, Oishi T, Watanabe R, Mitsuya K, Matsuno K, Furuya T, Okawara T, Otsuka M, Ogo N, Asai A, Nakasu Y, Yamaguchi K, Akiyama Y (2013) Effect of the STAT3 inhibitor STX-0119 on the proliferation of cancer stem-like cells derived from recurrent glioblastoma. Int J Oncol 43(1):219–227. doi:10.​3892/​ijo.​2013.​1916 CrossRefPubMed
35.
Zurück zum Zitat Plowman J, Waud WR, Koutsoukos AD, Rubinstein LV, Moore TD, Grever MR (1994) Preclinical antitumor activity of temozolomide in mice: efficacy against human brain tumor xenografts and synergism with 1,3-bis(2-chloroethyl)-1-nitrosourea. Cancer Res 54(14):3793–3799PubMed Plowman J, Waud WR, Koutsoukos AD, Rubinstein LV, Moore TD, Grever MR (1994) Preclinical antitumor activity of temozolomide in mice: efficacy against human brain tumor xenografts and synergism with 1,3-bis(2-chloroethyl)-1-nitrosourea. Cancer Res 54(14):3793–3799PubMed
36.
Zurück zum Zitat Singh D, Chan JM, Zoppoli P, Niola F, Sullivan R, Castano A, Liu EM, Reichel J, Porrati P, Pellegatta S, Qiu K, Gao Z, Ceccarelli M, Riccardi R, Brat DJ, Guha A, Aldape K, Golfinos JG, Zagzag D, Mikkelsen T, Finocchiaro G, Lasorella A, Rabadan R, Iavarone A (2012) Transforming fusions of FGFR and TACC genes in human glioblastoma. Science 337(6099):1231–1235. doi:10.1126/science.1220834 CrossRefPubMedPubMedCentral Singh D, Chan JM, Zoppoli P, Niola F, Sullivan R, Castano A, Liu EM, Reichel J, Porrati P, Pellegatta S, Qiu K, Gao Z, Ceccarelli M, Riccardi R, Brat DJ, Guha A, Aldape K, Golfinos JG, Zagzag D, Mikkelsen T, Finocchiaro G, Lasorella A, Rabadan R, Iavarone A (2012) Transforming fusions of FGFR and TACC genes in human glioblastoma. Science 337(6099):1231–1235. doi:10.​1126/​science.​1220834 CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Wykosky J, Hu J, Gomez GG, Taylor T, Villa GR, Pizzo D, VandenBerg SR, Thorne AH, Chen CC, Mischel PS, Gonias SL, Cavenee WK, Furnari FB (2015) A urokinase receptor-Bim signaling axis emerges during EGFR inhibitor resistance in mutant EGFR glioblastoma. Cancer Res 75(2):394–404. doi:10.1158/0008-5472.CAN-14-2004 CrossRefPubMed Wykosky J, Hu J, Gomez GG, Taylor T, Villa GR, Pizzo D, VandenBerg SR, Thorne AH, Chen CC, Mischel PS, Gonias SL, Cavenee WK, Furnari FB (2015) A urokinase receptor-Bim signaling axis emerges during EGFR inhibitor resistance in mutant EGFR glioblastoma. Cancer Res 75(2):394–404. doi:10.​1158/​0008-5472.​CAN-14-2004 CrossRefPubMed
38.
Zurück zum Zitat Mercurio L, Ajmone-Cat MA, Cecchetti S, Ricci A, Bozzuto G, Molinari A, Manni I, Pollo B, Scala S, Carpinelli G, Minghetti L (2016) Targeting CXCR4 by a selective peptide antagonist modulates tumor microenvironment and microglia reactivity in a human glioblastoma model. J Exp Clin Cancer Res 35:55. doi:10.1186/s13046-016-0326-y CrossRefPubMedPubMedCentral Mercurio L, Ajmone-Cat MA, Cecchetti S, Ricci A, Bozzuto G, Molinari A, Manni I, Pollo B, Scala S, Carpinelli G, Minghetti L (2016) Targeting CXCR4 by a selective peptide antagonist modulates tumor microenvironment and microglia reactivity in a human glioblastoma model. J Exp Clin Cancer Res 35:55. doi:10.​1186/​s13046-016-0326-y CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Kessler JD, Kahle KT, Sun T, Meerbrey KL, Schlabach MR, Schmitt EM, Skinner SO, Xu Q, Li MZ, Hartman ZC, Rao M, Yu P, Dominguez-Vidana R, Liang AC, Solimini NL, Bernardi RJ, Yu B, Hsu T, Golding I, Luo J, Osborne CK, Creighton CJ, Hilsenbeck SG, Schiff R, Shaw CA, Elledge SJ, Westbrook TF (2012) A SUMOylation-dependent transcriptional subprogram is required for Myc-driven tumorigenesis. Science 335(6066):348–353. doi:10.1126/science.1212728 CrossRefPubMed Kessler JD, Kahle KT, Sun T, Meerbrey KL, Schlabach MR, Schmitt EM, Skinner SO, Xu Q, Li MZ, Hartman ZC, Rao M, Yu P, Dominguez-Vidana R, Liang AC, Solimini NL, Bernardi RJ, Yu B, Hsu T, Golding I, Luo J, Osborne CK, Creighton CJ, Hilsenbeck SG, Schiff R, Shaw CA, Elledge SJ, Westbrook TF (2012) A SUMOylation-dependent transcriptional subprogram is required for Myc-driven tumorigenesis. Science 335(6066):348–353. doi:10.​1126/​science.​1212728 CrossRefPubMed
42.
Zurück zum Zitat Joo KM, Kim J, Jin J, Kim M, Seol HJ, Muradov J, Yang H, Choi YL, Park WY, Kong DS, Lee JI, Ko YH, Woo HG, Lee J, Kim S, Nam DH (2013) Patient-specific orthotopic glioblastoma xenograft models recapitulate the histopathology and biology of human glioblastomas in situ. Cell Rep 3(1):260–273. doi:10.1016/j.celrep.2012.12.013 CrossRefPubMed Joo KM, Kim J, Jin J, Kim M, Seol HJ, Muradov J, Yang H, Choi YL, Park WY, Kong DS, Lee JI, Ko YH, Woo HG, Lee J, Kim S, Nam DH (2013) Patient-specific orthotopic glioblastoma xenograft models recapitulate the histopathology and biology of human glioblastomas in situ. Cell Rep 3(1):260–273. doi:10.​1016/​j.​celrep.​2012.​12.​013 CrossRefPubMed
45.
Zurück zum Zitat Teng J, Carla da Hora C, Kantar RS, Nakano I, Wakimoto H, Batchelor TT, Chiocca EA, Badr CE, Tannous BA (2017) Dissecting inherent intratumor heterogeneity in patient-derived glioblastoma culture models. Neuro Oncol 19(6):820–832. doi:10.1093/neuonc/now253 PubMed Teng J, Carla da Hora C, Kantar RS, Nakano I, Wakimoto H, Batchelor TT, Chiocca EA, Badr CE, Tannous BA (2017) Dissecting inherent intratumor heterogeneity in patient-derived glioblastoma culture models. Neuro Oncol 19(6):820–832. doi:10.​1093/​neuonc/​now253 PubMed
46.
Zurück zum Zitat Jijiwa M, Demir H, Gupta S, Leung C, Joshi K, Orozco N, Huang T, Yildiz VO, Shibahara I, de Jesus JA, Yong WH, Mischel PS, Fernandez S, Kornblum HI, Nakano I (2011) CD44v6 regulates growth of brain tumor stem cells partially through the AKT-mediated pathway. PLoS ONE 6(9):e24217. doi:10.1371/journal.pone.0024217 CrossRefPubMedPubMedCentral Jijiwa M, Demir H, Gupta S, Leung C, Joshi K, Orozco N, Huang T, Yildiz VO, Shibahara I, de Jesus JA, Yong WH, Mischel PS, Fernandez S, Kornblum HI, Nakano I (2011) CD44v6 regulates growth of brain tumor stem cells partially through the AKT-mediated pathway. PLoS ONE 6(9):e24217. doi:10.​1371/​journal.​pone.​0024217 CrossRefPubMedPubMedCentral
47.
48.
Zurück zum Zitat Sancho-Martinez I, Nivet E, Xia Y, Hishida T, Aguirre A, Ocampo A, Ma L, Morey R, Krause MN, Zembrzycki A, Ansorge O, Vazquez-Ferrer E, Dubova I, Reddy P, Lam D, Hishida Y, Wu MZ, Esteban CR, O’Leary D, Wahl GM, Verma IM, Laurent LC, Izpisua Belmonte JC (2016) Establishment of human iPSC-based models for the study and targeting of glioma initiating cells. Nat Commun 7:10743. doi:10.1038/ncomms10743 CrossRefPubMedPubMedCentral Sancho-Martinez I, Nivet E, Xia Y, Hishida T, Aguirre A, Ocampo A, Ma L, Morey R, Krause MN, Zembrzycki A, Ansorge O, Vazquez-Ferrer E, Dubova I, Reddy P, Lam D, Hishida Y, Wu MZ, Esteban CR, O’Leary D, Wahl GM, Verma IM, Laurent LC, Izpisua Belmonte JC (2016) Establishment of human iPSC-based models for the study and targeting of glioma initiating cells. Nat Commun 7:10743. doi:10.​1038/​ncomms10743 CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Garralda E, Paz K, Lopez-Casas PP, Jones S, Katz A, Kann LM, Lopez-Rios F, Sarno F, Al-Shahrour F, Vasquez D, Bruckheimer E, Angiuoli SV, Calles A, Diaz LA, Velculescu VE, Valencia A, Sidransky D, Hidalgo M (2014) Integrated next-generation sequencing and avatar mouse models for personalized cancer treatment. Clin Cancer Res 20(9):2476–2484. doi:10.1158/1078-0432.CCR-13-3047 CrossRefPubMedPubMedCentral Garralda E, Paz K, Lopez-Casas PP, Jones S, Katz A, Kann LM, Lopez-Rios F, Sarno F, Al-Shahrour F, Vasquez D, Bruckheimer E, Angiuoli SV, Calles A, Diaz LA, Velculescu VE, Valencia A, Sidransky D, Hidalgo M (2014) Integrated next-generation sequencing and avatar mouse models for personalized cancer treatment. Clin Cancer Res 20(9):2476–2484. doi:10.​1158/​1078-0432.​CCR-13-3047 CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Ausman JI, Shapiro WR, Rall DP (1970) Studies on the chemotherapy of experimental brain tumors: development of an experimental model. Cancer Res 30(9):2394–2400PubMed Ausman JI, Shapiro WR, Rall DP (1970) Studies on the chemotherapy of experimental brain tumors: development of an experimental model. Cancer Res 30(9):2394–2400PubMed
52.
Zurück zum Zitat Johanns TM, Ward JP, Miller CA, Wilson C, Kobayashi DK, Bender D, Fu Y, Alexandrov A, Mardis ER, Artyomov MN, Schreiber RD, Dunn GP (2016) Endogenous neoantigen-specific CD8 T Cells identified in two glioblastoma models using a cancer immunogenomics approach. Cancer Immunol Res 4(12):1007–1015. doi:10.1158/2326-6066.CIR-16-0156 CrossRefPubMed Johanns TM, Ward JP, Miller CA, Wilson C, Kobayashi DK, Bender D, Fu Y, Alexandrov A, Mardis ER, Artyomov MN, Schreiber RD, Dunn GP (2016) Endogenous neoantigen-specific CD8 T Cells identified in two glioblastoma models using a cancer immunogenomics approach. Cancer Immunol Res 4(12):1007–1015. doi:10.​1158/​2326-6066.​CIR-16-0156 CrossRefPubMed
53.
Zurück zum Zitat Gousias K, Markou M, Arzoglou V, Voulgaris S, Vartholomatos G, Kostoula A, Voulgari P, Polyzoidis K, Kyritsis AP (2010) Frequent abnormalities of the immune system in gliomas and correlation with the WHO grading system of malignancy. J Neuroimmunol 226(1–2):136–142. doi:10.1016/j.jneuroim.2010.05.027 CrossRefPubMed Gousias K, Markou M, Arzoglou V, Voulgaris S, Vartholomatos G, Kostoula A, Voulgari P, Polyzoidis K, Kyritsis AP (2010) Frequent abnormalities of the immune system in gliomas and correlation with the WHO grading system of malignancy. J Neuroimmunol 226(1–2):136–142. doi:10.​1016/​j.​jneuroim.​2010.​05.​027 CrossRefPubMed
54.
55.
Zurück zum Zitat Xiao A, Wu H, Pandolfi PP, Louis DN, Van Dyke T (2002) Astrocyte inactivation of the pRb pathway predisposes mice to malignant astrocytoma development that is accelerated by PTEN mutation. Cancer Cell 1(2):157–168CrossRefPubMed Xiao A, Wu H, Pandolfi PP, Louis DN, Van Dyke T (2002) Astrocyte inactivation of the pRb pathway predisposes mice to malignant astrocytoma development that is accelerated by PTEN mutation. Cancer Cell 1(2):157–168CrossRefPubMed
56.
Zurück zum Zitat Reilly KM, Loisel DA, Bronson RT, McLaughlin ME, Jacks T (2000) Nf1;Trp53 mutant mice develop glioblastoma with evidence of strain-specific effects. Nat Genet 26(1):109–113. doi:10.1038/79075 CrossRefPubMed Reilly KM, Loisel DA, Bronson RT, McLaughlin ME, Jacks T (2000) Nf1;Trp53 mutant mice develop glioblastoma with evidence of strain-specific effects. Nat Genet 26(1):109–113. doi:10.​1038/​79075 CrossRefPubMed
57.
Zurück zum Zitat Zhu H, Acquaviva J, Ramachandran P, Boskovitz A, Woolfenden S, Pfannl R, Bronson RT, Chen JW, Weissleder R, Housman DE, Charest A (2009) Oncogenic EGFR signaling cooperates with loss of tumor suppressor gene functions in gliomagenesis. Proc Natl Acad Sci USA 106(8):2712–2716. doi:10.1073/pnas.0813314106 CrossRefPubMedPubMedCentral Zhu H, Acquaviva J, Ramachandran P, Boskovitz A, Woolfenden S, Pfannl R, Bronson RT, Chen JW, Weissleder R, Housman DE, Charest A (2009) Oncogenic EGFR signaling cooperates with loss of tumor suppressor gene functions in gliomagenesis. Proc Natl Acad Sci USA 106(8):2712–2716. doi:10.​1073/​pnas.​0813314106 CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Zeng J, See AP, Phallen J, Jackson CM, Belcaid Z, Ruzevick J, Durham N, Meyer C, Harris TJ, Albesiano E, Pradilla G, Ford E, Wong J, Hammers HJ, Mathios D, Tyler B, Brem H, Tran PT, Pardoll D, Drake CG, Lim M (2013) Anti-PD-1 blockade and stereotactic radiation produce long-term survival in mice with intracranial gliomas. Int J Radiat Oncol Biol Phys 86(2):343–349. doi:10.1016/j.ijrobp.2012.12.025 CrossRefPubMedPubMedCentral Zeng J, See AP, Phallen J, Jackson CM, Belcaid Z, Ruzevick J, Durham N, Meyer C, Harris TJ, Albesiano E, Pradilla G, Ford E, Wong J, Hammers HJ, Mathios D, Tyler B, Brem H, Tran PT, Pardoll D, Drake CG, Lim M (2013) Anti-PD-1 blockade and stereotactic radiation produce long-term survival in mice with intracranial gliomas. Int J Radiat Oncol Biol Phys 86(2):343–349. doi:10.​1016/​j.​ijrobp.​2012.​12.​025 CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Zhang C, Burger MC, Jennewein L, Genssler S, Schonfeld K, Zeiner P, Hattingen E, Harter PN, Mittelbronn M, Tonn T, Steinbach JP, Wels WS (2016) ErbB2/HER2-Specific NK Cells for Targeted Therapy of Glioblastoma. J Natl Cancer Inst 108(5):djv375. doi:10.1093/jnci/djv375 CrossRef Zhang C, Burger MC, Jennewein L, Genssler S, Schonfeld K, Zeiner P, Hattingen E, Harter PN, Mittelbronn M, Tonn T, Steinbach JP, Wels WS (2016) ErbB2/HER2-Specific NK Cells for Targeted Therapy of Glioblastoma. J Natl Cancer Inst 108(5):djv375. doi:10.​1093/​jnci/​djv375 CrossRef
60.
Zurück zum Zitat Parrish KE, Cen L, Murray J, Calligaris D, Kizilbash S, Mittapalli RK, Carlson BL, Schroeder MA, Sludden J, Boddy AV, Agar NY, Curtin NJ, Elmquist WF, Sarkaria JN (2015) Efficacy of PARP inhibitor rucaparib in orthotopic glioblastoma xenografts is limited by ineffective drug penetration into the central nervous system. Mol Cancer Ther 14(12):2735–2743. doi:10.1158/1535-7163.MCT-15-0553 CrossRefPubMedPubMedCentral Parrish KE, Cen L, Murray J, Calligaris D, Kizilbash S, Mittapalli RK, Carlson BL, Schroeder MA, Sludden J, Boddy AV, Agar NY, Curtin NJ, Elmquist WF, Sarkaria JN (2015) Efficacy of PARP inhibitor rucaparib in orthotopic glioblastoma xenografts is limited by ineffective drug penetration into the central nervous system. Mol Cancer Ther 14(12):2735–2743. doi:10.​1158/​1535-7163.​MCT-15-0553 CrossRefPubMedPubMedCentral
61.
Zurück zum Zitat Gupta SK, Mladek AC, Carlson BL, Boakye-Agyeman F, Bakken KK, Kizilbash SH, Schroeder MA, Reid J, Sarkaria JN (2014) Discordant in vitro and in vivo chemopotentiating effects of the PARP inhibitor veliparib in temozolomide-sensitive versus -resistant glioblastoma multiforme xenografts. Clin Cancer Res 20(14):3730–3741. doi:10.1158/1078-0432.CCR-13-3446 CrossRefPubMedPubMedCentral Gupta SK, Mladek AC, Carlson BL, Boakye-Agyeman F, Bakken KK, Kizilbash SH, Schroeder MA, Reid J, Sarkaria JN (2014) Discordant in vitro and in vivo chemopotentiating effects of the PARP inhibitor veliparib in temozolomide-sensitive versus -resistant glioblastoma multiforme xenografts. Clin Cancer Res 20(14):3730–3741. doi:10.​1158/​1078-0432.​CCR-13-3446 CrossRefPubMedPubMedCentral
62.
Zurück zum Zitat Garros-Regulez L, Aldaz P, Arrizabalaga O, Moncho-Amor V, Carrasco-Garcia E, Manterola L, Moreno-Cugnon L, Barrena C, Villanua J, Ruiz I, Pollard S, Lovell-Badge R, Sampron N, Garcia I, Matheu A (2016) mTOR inhibition decreases SOX2-SOX9 mediated glioma stem cell activity and temozolomide resistance. Expert Opin Ther Targets 20(4):393–405. doi:10.1517/14728222.2016.1151002 CrossRefPubMedPubMedCentral Garros-Regulez L, Aldaz P, Arrizabalaga O, Moncho-Amor V, Carrasco-Garcia E, Manterola L, Moreno-Cugnon L, Barrena C, Villanua J, Ruiz I, Pollard S, Lovell-Badge R, Sampron N, Garcia I, Matheu A (2016) mTOR inhibition decreases SOX2-SOX9 mediated glioma stem cell activity and temozolomide resistance. Expert Opin Ther Targets 20(4):393–405. doi:10.​1517/​14728222.​2016.​1151002 CrossRefPubMedPubMedCentral
63.
Zurück zum Zitat Hashizume R, Andor N, Ihara Y, Lerner R, Gan H, Chen X, Fang D, Huang X, Tom MW, Ngo V, Solomon D, Mueller S, Paris PL, Zhang Z, Petritsch C, Gupta N, Waldman TA, James CD (2014) Pharmacologic inhibition of histone demethylation as a therapy for pediatric brainstem glioma. Nat Med 20(12):1394–1396. doi:10.1038/nm.3716 CrossRefPubMedPubMedCentral Hashizume R, Andor N, Ihara Y, Lerner R, Gan H, Chen X, Fang D, Huang X, Tom MW, Ngo V, Solomon D, Mueller S, Paris PL, Zhang Z, Petritsch C, Gupta N, Waldman TA, James CD (2014) Pharmacologic inhibition of histone demethylation as a therapy for pediatric brainstem glioma. Nat Med 20(12):1394–1396. doi:10.​1038/​nm.​3716 CrossRefPubMedPubMedCentral
64.
Zurück zum Zitat Mathieu V, De Nève N, Le Mercier M, Dewelle J, Gaussin J-F, Dehoux M, Kiss R, Lefranc F (2008) Combining bevacizumab with temozolomide increases the antitumor efficacy of temozolomide in a human glioblastoma orthotopic xenograft model. Neoplasia 10(12):1383–1392. doi:10.1593/neo.08928 CrossRefPubMedPubMedCentral Mathieu V, De Nève N, Le Mercier M, Dewelle J, Gaussin J-F, Dehoux M, Kiss R, Lefranc F (2008) Combining bevacizumab with temozolomide increases the antitumor efficacy of temozolomide in a human glioblastoma orthotopic xenograft model. Neoplasia 10(12):1383–1392. doi:10.​1593/​neo.​08928 CrossRefPubMedPubMedCentral
65.
Zurück zum Zitat Cho J, Pastorino S, Zeng Q, Xu X, Johnson W, Vandenberg S, Verhaak R, Cherniack AD, Watanabe H, Dutt A, Kwon J, Chao YS, Onofrio RC, Chiang D, Yuza Y, Kesari S, Meyerson M (2011) Glioblastoma-derived epidermal growth factor receptor carboxyl-terminal deletion mutants are transforming and are sensitive to EGFR-directed therapies. Cancer Res 71(24):7587–7596. doi:10.1158/0008-5472.CAN-11-0821 CrossRefPubMedPubMedCentral Cho J, Pastorino S, Zeng Q, Xu X, Johnson W, Vandenberg S, Verhaak R, Cherniack AD, Watanabe H, Dutt A, Kwon J, Chao YS, Onofrio RC, Chiang D, Yuza Y, Kesari S, Meyerson M (2011) Glioblastoma-derived epidermal growth factor receptor carboxyl-terminal deletion mutants are transforming and are sensitive to EGFR-directed therapies. Cancer Res 71(24):7587–7596. doi:10.​1158/​0008-5472.​CAN-11-0821 CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat Yoshida Y, Ozawa T, Yao TW, Shen W, Brown D, Parsa AT, Raizer JJ, Cheng SY, Stegh AH, Mazar AP, Giles FJ, Sarkaria JN, Butowski N, Nicolaides T, James CD (2014) NT113, a pan-ERBB inhibitor with high brain penetrance, inhibits the growth of glioblastoma xenografts with EGFR amplification. Mol Cancer Ther 13(12):2919–2929. doi:10.1158/1535-7163.MCT-14-0306 CrossRefPubMedPubMedCentral Yoshida Y, Ozawa T, Yao TW, Shen W, Brown D, Parsa AT, Raizer JJ, Cheng SY, Stegh AH, Mazar AP, Giles FJ, Sarkaria JN, Butowski N, Nicolaides T, James CD (2014) NT113, a pan-ERBB inhibitor with high brain penetrance, inhibits the growth of glioblastoma xenografts with EGFR amplification. Mol Cancer Ther 13(12):2919–2929. doi:10.​1158/​1535-7163.​MCT-14-0306 CrossRefPubMedPubMedCentral
67.
68.
Zurück zum Zitat Sarkaria JN, Carlson BL, Schroeder MA, Grogan P, Brown PD, Giannini C, Ballman KV, Kitange GJ, Guha A, Pandita A, James CD (2006) Use of an orthotopic xenograft model for assessing the effect of epidermal growth factor receptor amplification on glioblastoma radiation response. Clin Cancer Res 12(7 Pt 1):2264–2271. doi:10.1158/1078-0432.CCR-05-2510 CrossRefPubMed Sarkaria JN, Carlson BL, Schroeder MA, Grogan P, Brown PD, Giannini C, Ballman KV, Kitange GJ, Guha A, Pandita A, James CD (2006) Use of an orthotopic xenograft model for assessing the effect of epidermal growth factor receptor amplification on glioblastoma radiation response. Clin Cancer Res 12(7 Pt 1):2264–2271. doi:10.​1158/​1078-0432.​CCR-05-2510 CrossRefPubMed
69.
Zurück zum Zitat Pandita A, Aldape KD, Zadeh G, Guha A, James CD (2004) Contrasting in vivo and in vitro fates of glioblastoma cell subpopulations with amplified EGFR. Genes Chromosomes Cancer 39(1):29–36. doi:10.1002/gcc.10300 CrossRefPubMed Pandita A, Aldape KD, Zadeh G, Guha A, James CD (2004) Contrasting in vivo and in vitro fates of glioblastoma cell subpopulations with amplified EGFR. Genes Chromosomes Cancer 39(1):29–36. doi:10.​1002/​gcc.​10300 CrossRefPubMed
Metadaten
Titel
Current trends in mouse models of glioblastoma
verfasst von
Masafumi Miyai
Hiroyuki Tomita
Akio Soeda
Hirohito Yano
Toru Iwama
Akira Hara
Publikationsdatum
20.10.2017
Verlag
Springer US
Erschienen in
Journal of Neuro-Oncology / Ausgabe 3/2017
Print ISSN: 0167-594X
Elektronische ISSN: 1573-7373
DOI
https://doi.org/10.1007/s11060-017-2626-2

Weitere Artikel der Ausgabe 3/2017

Journal of Neuro-Oncology 3/2017 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.