Download Analysis of isocitrate dehydrogenase mutations in gliomas

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts
Transcript
Analysis of isocitrate dehydrogenase1/2 gene mutations in gliomas
YU Lei, QI Song-tao, OU Yang-hui and LI Zhi-yong
Keywords: IDH mutation; Glioma;α-Ketoglutarate; 2-Hydroxyglutatarate; Hypoxia-inducible
factor
Objective
Gliomas are the most frequent, lethal, and rapidly infiltrating form of brain cancer
that is resistant to surgery, radiotherapy and chemotherapy. The molecular profiling of gliomas
genomes is making enormous progress these days, but until 2008 the discovery of somatic
mutations in the gene encoding isocitrate dehydrogenase-1 , which has been not previously
identified with any known oncogenic pathway, bring great advantages to deeper
understanding of this disease. A better understanding of the functional role of isocitrate
dehydrogenase-1/2 mutations in the pathogenesis of gliomas would propel the development
of more effective treatments. The objective in this review is to highlight these recent
researches that may show promise for histomolecular classification and new treatments for
gliomas.
Data sources
All articles cited in this review were mainly searched from PubMed, which were
published in English from 1996 to 2010.
Study selection
Original articles and critical reviews selected were relevant to the isocitrate
dehydrogenase-1/2 mutation in gliomas and other tumors.
Results
Extraordinary high rates of somatic mutations in isocitrate dehydrogenase-1/2 occur
in the majority of World Health Organization grade II and grade III gliomas as well as grade Ⅳ
secondary glioblastomas. Isocitrate dehydrogenase-1/2 mutations are associated with younger
age at diagnosis and a better prognosis in patients with mutated tumors. The functional role of
isocitrate dehydrogenase-1/2 mutations in the pathogenesis of gliomas is still unclear.
Conclusion
Isocitrate dehydrogenase-1/2 mutations define a specific subtype of gliomas
and may have great significance in the diagnosis, prognosis, and treatment of patients with
these tumors.
G
liomas are the most frequent and lethal tumors of the central nervous system(CNS) and show
wide diversity with location, morphology, genetic status, and response to therapy. These tumors
have been classified as grade I to grade IV based on histopathological and clinical criteria established
by the World Health Organization (WHO).1 Despite intensive therapies, including surgery, radiotherapy,
and chemotherapy, the outcome of glioma patients remains depressing. 2, 3 Current study focused on
identifying genetic alterations implicated in gliomas has become increasingly hot. Then in 2008 by way
of an unbiased, genome-wide analysis of the somatic mutations occurring in 22 human glioblastomas,
researchers found that five of these tumors harbored mutations in the isocitrate dehydrogenase-1 gene
(IDH1), which as a metabolic enzyme gene, had never been implicated in cancer before.4 Subsequently
a genome-wide study declared that mutations that affected amino acid (R132) of IDH1 occurred in
more than 70% of WHO grade II and III astrocytomas and oligodendrogliomas and in secondary
glioblastomas.5 Tumors without mutations in IDH1 often had mutations affecting the analogous amino
acid (R172) of the IDH2,5 but never affecting IDH3. These results raise numerous questions. What are
the normal functions of the IDH1/2-encoded enzymes? What are the functional consequences of
IDH1/2 mutations in the pathogenesis of gliomas? What is the relationship between IDH1/2 mutation
and clinical diagnosis and treatments? Here we review these new findings.
1
Normal biochemical function of IDH
Xu et al. reported the crystal structure of human IDH1, which, encoded by IDH1 gene on chromosomal
2q33.3, often functioned as homodimers.6 Each homolog comprises a large domain, a clasp domain,
and a small domain. IDH1 homodimers contain two asymmetric active sites, with each active site made
up of a cleft formed by the large and small domains of one IDH1 molecule and the small domain of the
other IDH1 molecule in the dimer. The active sites are exposed to solvent and are accessible to the
substrate and cofactor. The clasp functions to hold the two subunits together to form this active site.
Human IDH1 transforms between an inactive open, an inactive semi-open, and a catalytically active
closed conformation.6 Located atop a β-sheet in the relatively rigid small domain, R132 acts as a
gate-keeper residue and appears to coordinate the hinge movement between the open and closed
conformations. Besides the side chain of R132 residue in the active site of the enzyme, uniquely forms
three hydrogen bonds with the substrate isocitrate but other residues involved in isocitrate binding form
no more than two hydrogen bonds.7
The human genome possess five IDH genes, coding for three distinct IDH enzymes whose coenzymes
are either nicotinamide adenine dinucleotide phosphate(NADP+,IDH1/2) or nicotinamide adenine
dinucleotide(NAD+,IDH3). IDH1 is located in the cytoplasm and peroxisomes, whereas IDH2 and
IDH3 in the mitochondria. IDHs catalyze the oxidative decarboxylation of isocitrate toα-ketoglutarate
(α-KG) and reduce NAD(P)+ to NAD(P)H. IDHs play significant yet distinctive roles in cellular
metabolism, with IDH1 involved in lipid metabolism and glucose sensing, IDH2 in the regulation of
oxidative respiration and IDH3 in aerobic energy production in the tricarboxylic acid (TCA) cycle. In
addition to their roles in normal cellular metabolism, 8-10 they also play a role in the cellular response to
oxidative insults.11, 12 Park and colleagues
13
have confirmed the role of IDH1 and IDH2 as protectors
against various insults.
Frequency and characteristic of IDH1/2 mutation
In order to determine the frequencies and different types of mutations in glioma development, IDH1/2
mutations have been assessed in a large number of gliomas of various types, and the results are striking:
mutations in IDH1 R132 are in fact common(50%–94%)in grades II and III gliomas and secondary
glioblastomas and also occur less frequently in primary glioblastomas(table1).5, 14-20 IDH1 mutaition
were also identified in 3/9(33%)14 and 2/34(6%)21 supratentorial primitive neuroectodermal tumors
(sPNET). Seiz, M., et al. examined the frequency of IDH1 mutations in 35 Gliomatosis cerebri (GC)
samples by direct sequencing and identified IDH1 mutations in 10/24(42%) cases22. In addition,
analysis of the closely related IDH2 has revealed recurrent somatic mutations of IDH2 R172, with most
mutations occurring in tumors lacking IDH1 mutations. 5, 17 Exceptionally one study examined all their
series for both IDH1 and IDH2 mutations and found four anaplastic gliomas with both mutations. 17 The
difference of IDH mutation frequencies in published literatures may partly be attributed to various
numbers of particular subtypes of gliomas; however, the use of different thresholds in scoring weaker
signals as sufficient for a mutation may also play a role. 17
Most recently, IDH1 R132 mutation have also been found in acute myeloid leukemia(AML),23,
B-acute lymphoblastic leukemia,
25
26
colorectal cancer, and prostate carcinoma.
human cancers besides glial tumors were almost absent,
and R140 in AMLs.
24, 28
27
25
24
IDH2 mutations in
except for rare cases of mutations at R172
In the latest study, scientists identified two novel homozygous IDH1 mutations
in thyroid cancer and follicular thyroid cancer.29 There were no IDH1 mutation reported in a broad
2
range of non-CNS tumors,4,
20
or in other CNS tumors including brain metastases of colorectal
30
cancers or melanoma metastases.31
Interestingly, all of the IDH1 mutations identified to date produce a single amino acid substitution at
R132 and all of the IDH2 mutations at R172 except for few cases at R140 in AMLs. 24, 28 In gliomas,
most IDH1 mutations were G395A (R132H) with Amino acid Arg changed to His, whereas IDH2
mutations were G515A(R172K) with Amino acid Arg changed to Lys(table2) 5,
14-20, 32, 33
. IDH1
mutations form the lion’s share of IDH mutations found in cancer, with IDH2 mutation being much less
common. Noticeably, IDH1 mutation of the R132C type was strongly associated with astrocytoma,
while IDH2 mutations predominantly occured in oligodendroglial tumors. 17
Association with other genetic changes(table1)
Tumor protein p53(TP53) encodes the p53 tumor suppressor that regulates cell cycle, apoptosis, DNA
repair and genome stability, and mutations in this pathway often lead to cancer development and poor
outcome.34 TP53 mutations, characteristic of early alterations in the development of an astrocytoma ,
are common in grades II and III astrocytomas and in secondary glioblastomas. In some studies, IDH
mutations have been found to be associated with TP53 mutations,5, 18, 19 although other studies did not
find a statistically significant association.14,
16
1p19q codeltion, which is commonly observed in
oligodendroglial tumors, is frequently observed in IDH-mutated oligodendroglial tumors, 5, 18 and even
in a study ,based on a large series, all tumors with complete 1p19q codeletion(n=128) were found
mutated in the IDH1(n=118) or IDH2(n=10)gene, suggesting that IDH1/2 mutation is a constant feature
in gliomas with complete 1p19q codeletion.33 EGFR amplification are characteristic of higher-grade
tumors,35-37 whereas BRAF fusion gene(at chromosomal band 7q34) is detected as a typical lesion for
PA I.38 O-6-methylguanine-DNA methyltransferase (MGMT) is a DNA repair enzyme that removes
alkylating lesions induced by chemotherapeutic agents, and as a result its methylation induces low
expression of MGMT protein resulting in decreasing DNA repair activity and increasing sensitivity to
alkylating agents such as nitrosourea and temozolomide.39, 40 Sanson et al.16 in a series of 404 gliomas
found that the IDH1 mutation was tightly related to the 1p/19q-codeleted group and MGMT
methylation but mutually exclusive with EGFR amplification. IDH1/2 mutations and the BRAF fusion
gene are mutually exclusive ; among IDH1/2-mutated gliomas, 1p/19q codeletion are mutually
exclusive with TP53 mutations and EGFR amplification.41 Besides above-mentioned genetic changes,
IDH1 mutations frequently accompanied alternative lengthening of telomeres (ALT) 42 and existence of
a glioma-CpG island methylator phenotype(G-CIMP). 43
These genetic changes often occur in a staged order during the progression to a high- grade tumor.
Watanabe and colleagues dissected multiple biopsies from the same patients and found that IDH1
mutations always preceded the acquisition of TP53 mutation or 1p/19q codeletion,suggesting that
IDH1 mutations were very early events in gliomagenesis and might affect a common glial precursor
cell population.19 In addition, patients with germline TP53 mutations, which predispose to grade II - IV
astrocytomas, had tumors that harbored somatic IDH1R132C mutations.44
Association with age
Intriguingly, mutations in IDH1/2 predominantly occurred in younger patients except for children.
Parsons, D.W., et al. reported that mutations in IDH1 preferentially occurred in younger GBM patients,
with a mean age of 33 years for IDH1-mutated patients, as opposed to 53 years for patients with
wild-type IDH1(P<0.001).4 Balss, J., et al. reported that In prGBM, AIII and OAIII, the mean age of
3
patients with IDH1 mutations versus patients without was respectively 40.3 years vs 52.6 years, 35.0
years vs 44.4 years, and 44.3years vs 63.5 years.14 Ichimura, K., et al. reported that the mean age of
GBM patients with IDH1 mutations was 41 years, compared with 56 years for GBM patients without
(p =0.002).18 Yan, H., et al. reported that patients with anaplastic astrocytomas or glioblastomas with
IDH1/2 mutations were significantly younger than were patients without (median age,34 years vs.56
years for patients with anaplastic astrocytomas and 32 years vs.59 years for those with glioblastomas;
P<0.001 for both comparisons).5 The association of IDH1 mutations with age seems mostly in the
highly malignant entities,but whether it is true or not will require further studies. However, rare IDH
1/2mutations have been identified in pediatric glioblastomas, and children with IDH1/2-mutated
gliomas are older than the others (median age 16 versus 7 years by De Carli et al. and 17 years vs.5
years by Yan et al.).45, 46
Association with prognosis
To date, only two molecular aberrations have been demonstrated to be of clinical significance in
prospective clinical trials. 1p19q codeltion are associated with a favorable prognosis in patients with
oligodendroglial and oligoastrocytic anaplastic gliomas treated with radiotherapy or radiotherapy plus
chemotherapy.47-50 MGMT promoter hypermethylation is associated with prolonged progression-free
survival (PFS)and overall survival(OS)in patients with glioblastoma treated with alkylating agents such
as nitrosoureas or temozolomide.39,
51-53
High throughput analyses have recently resulted in the
identification of IDH1 mutations as a novel prognostic marker in gliomas.4, 5, 14 Glioma patients with
IDH1/2 mutations survive longer than patients without,4, 5, 18, 54 and multivariate analysis has confirmed
that IDH1 mutation is an independent favorable prognostic marker in GBMs and anaplastic gliomas
after adjustment for other genomic profiles and treatment modality 16, 32, 49 except for the results reported
by Ichimura, K., et al.,18 who indicated that multivariate analysis failed to identify IDH1 status as a
prognostic factor independent of tumor type, grade, or age. Interestingly, those few primary
glioblastomas with IDH1 mutations also have a significantly better prognosis.55, 56
Functional properties in IDH1/2 mutations
Since the discovery of IDH mutations, the functional significance of IDH1/2 mutations in human
cancer remains a mystery. Zhao, S., et al.7 reported that the mutation caused a loss of function of IDH1
and higher levels of hypoxia-inducible factor subunit (HIF-1α) in tumors with mutation than tumors
without, so they concluded that it was a tumor suppressor gene. But Dang, L., et al.57 found high levels
of a single metabolite, 2-hydroxyglutatarate (2-HG), in IDH1-mutated tumors .Their data suggested
that the excess 2-HG which accumulated in vivo contributed to the formation and malignant
progression of gliomas, so later they announced that IDH1 was an oncogene, not a tumor suppressor.
Indeed,while the R132H mutation leads to a loss of enzymatic function for oxidative decarboxylation
of isocitrate, it also results in a gain of enzyme function for the NADPH-dependent reduction of α-KG.
Whether mutant IDH1 is a tumor suppressor gene or oncogene remains unsolved, yet we may discuss
from correlatively IDH-mutated metabolites such as α-KG, NADPH, 2-HG and HIF-1α.
α-KG
IDH1 mutation impairs the enzyme’s affinity for its substrate and dominantly inhibits the normal
function of wild-type IDH1 to convert isocitrate to α-KG through the formation of catalytically inactive
heterodimers, so, as a consequence, α-KG levels were reduced.7 Further study revealed that mutated
IDH1/2 reduce α-KG to D-2-HG while converting NADPH to NADP+.24, 57, 58 Experiments suggested
4
that α-KG levels decreased in cells transfected with mutant IDH1. 7 Xiong’s group also observed a
decrease of α-KG in cells with mutant IDH1.59 But Dang, L., et al. observed that the difference in
average α-KG between mutated-IDH1 tumors and wild-type IDH1 tumors was not statistically
significant, so it was for other proximal TCA cycle metabolites. 57 If α-KG don’t decrease, why would
α-KG levels stay the same since the mutations in IDH1 impair its ability to produce α-KG and the new
enzyme function for the NADPH-dependent reduction of α-KG consume it? The question remains
unsettled.
NADPH
The production of NADPH has been linked to the suppression of apoptosis and to enhanced cell
survival and growth.60-62 NADPH is required for the synthesis of glutathione, which protects cells from
redox stress and promotes resistance to apoptosis. 60 Moreover, cytosolic NADPH is the substrate for
the membrane-associated NADPH oxidases, whose production of hydrogen peroxide inhibits protein
tyrosine phosphatases, thereby promoting sustained activation of kinases that promote cell-autonomous
survival and mitogenic signaling.61 IDH1 and, to a lesser extent, IDH2 provide a significant fraction of
cellular NADPH for these processes.
62
The total NADPH production capacity in glioblastoma was
provided for 65% by IDH activity and the occurrence of R132 IDH1 mutation reduced this capacity by
38%.63 That is to say, IDH1 mutation in glioblastoma hinders NADPH production. In a sense, it is in
accordance with the acquired function converting α-KG, and NADPH into D-2-HG and NADP +.
Although NADPH is required for the synthesis of glutathione, which protects cells from redox stress,
there is still lack of biological data to support the hypothesis that decreased production of NADPH
caused by IDH mutations may impair a survival advantage and even the increase or decrease of
NADPH is controversial. Glial cells may have a higher-than-normal level of α-KG and a high level of
feedback inhibition of their IDH activity. Therefore, mutations that affect end-product inhibition might
facilitate increased production of NADPH in glial cells. 64 The question of NADPH level in
IDH-mutated gliomas awaits further confirmation.
2-HG
The normal metabolic role of 2-HG is not completely understood but 2-HG is not unnatural to cells. It
can be generated by specific α-KG reductase enzymes65 and oxidized back toα-KG by 2-HG
dehydrogenases(2-HGD). The novel enzymatic activity associated with R132 mutations in IDH1
results in the production of 2-HG in gliomas that harbor these mutations and 2-HG is the direct product
of NADPH-dependent α-KG reduction.57 Experiments showed that cancer tissue samples with IDH1/2
mutations had more than 100-fold higher concentrations of 2-HG than cancers without.24, 57, 58 The
accumulating metabolite, 2-HG, exists in the form of two enantiomers, l-2-HG and D-2-HG (each the
mirror image of the other), both of which accumulate whenever the relevant converting enzyme is
defective. D-2-HGD or L-2-HGD for inactivating mutations was analyzed in patients with sporadic
brain tumors without IDH1/2 mutations and there was no evidence for mutations in the genes
D-2-HGD and L-2-HGD as an alternative mechanism for raised 2-HG levels in brain tumors and for
distinct alleles of these genes conferring an increased risk for tumorigenesis.66 The clinical features of
pathological accumulation of 2-HG differ markedly according to the type of enantiomer involved.
Pathological accumulation of the L-2-HG is characterized by progressive neuronal defects and recently
linked to increased risk of brain tumors including gliomas, 67 but the D-2-HG generated by mutant
IDH1 protein(demonstrated by Dang et al.) is not known to have an increased risk for developing brain
tumors.
If 2-HG is associated with cancer initiation and progression, following hypotheses may be acceptable.
5
Elevated brain levels of 2-HG result in increased ROS levels,68, 69 potentially contributing to an
increased risk of cancer. 2-HG may also be toxic to cells by competitively inhibiting glutamate and/or
α-KG utilizing enzymes, such as transaminases which allow utilization of glutamate nitrogen for amino
and nucleic acid biosynthesis, and α-KG-dependent prolyl hydroxylases(PHDs) which regulate HIF-1α
levels.57 2-HG also acts on the family of dioxygenases known as histone demethylases, which regulate
gene expression among other processes by removing methyl groups from histones (proteins around
which DNA is coiled). 2-HG can also act on the tumor microenvironment and disable mitochondria,
thus leading to the reprogramming of the cells and basically pushing the cells toward anerobic
glycolysis(so basically promote the so-called Warburg effect).59 Otto Warburg observed that in most
cancer cells, energy is produced predominantly by aerobic glycolysis in the cytosol, rather than by
oxidation of pyruvate in mitochondria, as in most normal cells. He postulated that this change in
metabolism is the fundamental cause of cancer.70
HIF
HIF, is a master regulator of genes that are activated by low oxygen levels and regulates the expression
of genes implicated in glucose metabolism, angiogenesis, cell motility and invasion, and other
signaling pathways that are critical to tumor growth.71 PHDs use α-KG as a substrate for a reaction that
normally targets HIF-1α for degradation. The loss of activity of two other TCA cycle enzymes
mentioned earlier, succinate dehydrogenase (SDH) or fumarate hydratase (FH), supports
tumorigenesis by increasing succinate or fumarate. These substrates inhibit the PHDs by competing
with their cosubstrate α-KG,72 causing the activation of the HIF transcription factor and high levels of
HIF-1α.73 Experiments showed that HIF-1α levels were higher in human gliomas with IDH1 mutations
than without mutations,7 and high levels of HIF-1α can be transported into the nucleus for
transcriptional activity.64, 71, 74 IDH mutants could lead to lower cellular α-KG levels by consuming this
compound, which may lead to PHDs inactivation. Alternatively, 2-HG produced by IDH mutants has
been thought to competitively inhibit PHDs by occupying the α-KG-binding site on these enzymes.75
However, grades II and III gliomas do not demonstrate angiogenesis, as would be expected for tumors
that activate this hypoxia signaling pathway. 73 Finally, increased expression of HIF-1 target genes is not
found in AMLs,23 which calls into question the idea that HIF-1α stabilization is a major function of the
IDH mutations. Furthermore, it is still be unknown whether the level of α-KG in IDH-mutated tumors
is definitely decreased.
Utility of research and clinical application
IDH mutations seem to play a central role in the pathogenesis of gliomas and define a specific subtype
of gliomas. IDH mutations generally associate with specific gene expression signatures, 76 and
determination of the gene expression environment in mutated and nonmutated tumors of specific tumor
types may shed light on the mechanism of IDH-mutated gliomagenesis. Determining whether IDH
status is an independent prognostic factor for any of the tumor types that contain these mutations may
guide clinical management of a lethal group of cancers. Therefore, the IDH1 status should be
considered in the design of preclinical and clinical studies in future.
Although the IDH-mutated studies are incomplete and still controversial, they are paving the way for
diagnostic and therapeutic application. One study has concluded that combined molecular analysis of
BRAF and IDH1 is a sensitive and highly specific approach to separate pilocytic astrocytoma from
diffuse astrocytoma.38 IDH1 mutation is a strong predictor of a more favorable prognosis and the most
specific biomarker of secondary glioblastomas that complements clinical criteria for distinguishing
6
them from primary glioblastomas.32 This could be useful in cases especially when scant material is
available for histopathological analysis.
The localization of IDH1 /2 mutations to a single amino acid (R132 and R172, respectively)should
simplify the use of this genetic alteration for diagnostic purposes, needless to sequence DNA for
assessing IDH1 status. The current procedure not requiring sequencing but relying on polymerase chain
reaction (PCR) and restriction endonucleases recognizing a site composed of mismatched primer and
IDH1 mutation has recently been adopted widely.77 The real-time PCR/fluorescence melting curve
analysis assay,78 which has been used to evaluate the methylation status of the MGMT gene,79 and
rapid detection based on pyrosequencing 80 provide prompt and sensitive detection of IDH mutations in
formalin-fixed, paraffin-embedded tissues. Two mouse monoclonal antibodies targeting the IDH1
R132H mutation IMab-181 and mIDH1R132H,82 shows high specificity and sensitivity in the detection
of mutations .Especially, mIDH1R132H can differentiate reactive gliosis from neoplastic glial cells in
grade II and III gliomas and allow identifying tumor cells in post-therapy specimens with extensive
reactive changes.83 Measurement of 2-HG production may also simplify the diagnosis and management
of glioma patients, especially if this compound is also elevated in cerebrospinal fluid, serum, or urine
of patients.84
Future direction
Why are IDH genes mutated in the majority of gliomas? What are the cellular consequences of IDH1
mutations in gliomas? Should IDH1 status be used as a stratification factor in clinical trials? What role
does IDH1 play in response to chemotherapy and/or radiotherapy? All these tissues remain unsettled
and the solutions are certain to be relevant for basic and clinical cancer research.
These recent studies have partially shown the relationship of α-KG, NADPH, D- 2-HG, HIF-1α and
mutated IDH1 in gliomas. Long-term research may require the use of mammalian tissue for model. Pet
dogs that develop spontaneous gliomas have been proposed as a model to test human glioma
therapies,85 but now we should pay attention to the genetic differences between human and canine
tumors for studies using this model because IDH1 /2 hotspot mutations are not found in canine
gliomas.86
Further studies are needed to determine whether IDH1 status could influence radiotherapy and
chemotherapy. Dubbink, H.J., et al. reported that presence of IDH1 mutations significantly improved
overall survival but did not affect outcome of temozolomide treatment. 87 In patients with anaplastic
oligodendroglial tumours treated with radiotherapy alone or radiotherapy with adjuvant PCV
(procarbazine, lomustine, and vincristine), the presence of IDH1 mutations had no predictive value for
outcome to PCV.88
It is very likely that in the near future, large-scale genomic expression studies based on IDH mutations
are expected to refine the molecular classification of gliomas to replace the histological classification
and should help establish these mutant genes and the related metabolic pathways as attractive targets
for guiding glioma management. For example, it is vital to take into account the mutant genes present
in each patient’s tumor before chemotherapeutic agents that target molecular pathways can ever be
expected to achieve maximal clinical efficacy. Given the tumor specificity of metabolic enzyme
mutations and the striking difference between the cellular metabolism of cancer and normal cells,
IDH-mutated enzymes as prevalent and specific alterations in gliomas may prove to be fruitful targets
for anticancer therapies.64 Several dozen anticancer agents directly targeting mutant or wild-type IDH
are under development or being tested.59, 89 However, any such improvements in the treatment for
7
patients with IDH-mutated gliomas will hinge on a better understanding of the functional role of the
mutant IDH in the pathogenesis of these tumors.
8
Table1. Frequency of IDH1/2 mutations and other common genetic changes in gliomas
Gliomas*
IDH1 mutation†
(range of percentages)
IDH2 mutation
TP53 mutation
1p19q codeletion
EGFRamp
PA I
1/41,0/21, 0/3,
(0-9.7)
0/3,0/21
0/21
0/38
0/21
(33.3-88.2)
0/9,0/2,2/227
13/25,11/22
6/34,3/22, 0/30
0/17, 0/30
2/30
22/30
1/20,0/9,6/128
3/31,2/34, 8/51
30/50,23/54,26/34
0/11, 0/51
0/38,3/31
A II
34/46,3/9 10/12
165/227,13/22,
60/68, 25/30
O II
36/51,16/20,6/9
(66.6-82.0)
41/54,105/128,
2/51
31/51
23/34,31/39,41/51
OA II
36/46,1/1,26/34,
(50.0-100)
0/1,1/76
6/26,8/20, 1/3
24/45,2/34,1/20
0/10,0/3
(44.4-77.8)
0/9,1/21,2/228
13/30,42/62
5/43,3/62, 5/52
1/22,1/52
2/52
34/52
0/9,9/174, 3/36
4/31,3/20, 4/36
35/53,19/49,12/20
0/4, 0/36
62/76,10/20,16/17
3/3
A III
29/47,4/9,12/21
9/18,146/228,
32/62,21/27,36/52
O III
36/54,8/9,24/49
(49.0-88.9)
121/174,12/20
30/36
6/8,31/36
OA III
29/37, 3/4,34/54
(63.0-100)
0/4,11/177
6/22,11/23, 5/7
117/177,18/23
26/36,6/54,6/23
0/8
3/7
10/14, 7/7
prGBM
7/99,11/183,6/173
(3.5-7.1)
0/123
15/88,59/173
3/59,6/123,11/94
secGBM
7/8,10/13,5/10
6/73,3/173, 5/123
26/71, 47/123
28/123
(50.0-87.5)
0/13
7/8,6/10,8/13
1/8,0/10
0/8,0/13
0/15
2/3,5/15
0/3
0/2
28/34,11/13,11/15
pedGBM
1/14,0/15
*PA I: Pilocytic astrocytoma WHO grade I, A II: Diffuse astrocytoma WHO grade II, O II: Oligodendroglioma WHO grade II, OA II:
Oligoastrocytoma WHO grade II, A III: Anaplastic astrocytoma WHO grade III, O III: Anaplastic oligodendroglioma WHO grade III, OA
III: Anaplastic oligoastrocytoma WHO grade III, prGBM: Primary Glioblastoma WHO grade IV, secGBM: Secondary glioblastoma
WHO grade IV, pedGBM: Pediatric Glioblastoma WHO grade IV
†Number of mutated sample/total number of samples of neuropathological type analyzed for each report of the special type.
9
Table2. different reports of IDH1/2 mutation types
Amino acid substitution
Different reports
Percentage(%) in total IDH1 mutation
R132H
R132C
R132S
R132L
R132G
Balss, J., et al.
92.7
3.6
1.8
0.5
0.9
Horbinski, C., et al.
78.4
2.7
5.4
2.7
2.7
Nobusawa, S., et al.
83
2.8
2.8
Sanson, M., et al.
89
3.2
1.9
1.3
4.5
Hartmann, C., et al.
92.7
4.2
1.5
0.2
1.4
Ichimura, K., et al.
92.4
3.4
0.8
3.4
Watanabe, T., et al.
91
4.6
0.8
3.8
Yan, H., et al.
88.2
4.3
2.5
4.3
0.6
Bleeker, F.E., et al.
73.9
13.0
4.3
4.3
4.3
Labussiere, M., et al.
88.6
3.8
1.6
1.6
4.4
Number in total IDH2 mutation
R172G
R172K
R172M
R172W
2.7
11.1
2/9
20/31
6/31
4/9
3/9
12/16
1/16
5/31
1/16
R132H: Amino acid change Arg→His with Nucleotide change G395A(CGT→CAT) ; R132C: Amino acid change Arg→Cys with Nucleotide change
C394T(CGT→TGT); R132S: Amino acid change Arg→Ser with Nucleotide change C394A(CGT→AGT); R132L: Amino acid change Arg→Leu with
Nucleotide change G395T(CGT→CTT); R132G: Amino acid change Arg→Gly with Nucleotide change C394G(CGT→GGT); R172G : Amino acid change
Arg→Gly with Nucleotide change A514G(AGG→GGG) R172K: Amino acid change Arg→Lys with Nucleotide change G515A(AGG→AAG); R172M:
Amino acid change Arg→Met with Nucleotide change G515T(AGG→ATG); R172W: Amino acid change Arg→Try with Nucleotide change
A514T(AGG→TGG).
References:
1. Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, et al. The 2007 WHO
classification
of
tumours
of
the
central
nervous
system.
Acta
Neuropathol
2007;114:97-109.17618441
2. Ohgaki H. Epidemiology of brain tumors. Methods Mol Biol 2009;472:323-342.19107440
3. Zhu YJ, Zhu XD, Wang SH, Shen F, Shen H, Liu WG. A multivariate analysis of the prognostic
factors of grade III gliomas. Chin Med J (Engl) 2008;121:1072-1075.18706219
4. Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt P, et al. An integrated genomic
analysis of human glioblastoma multiforme. Science 2008;321:1807-1812.18772396
5. Yan H, Parsons DW, Jin G, McLendon R, Rasheed BA, Yuan W, et al. IDH1 and IDH2 mutations
in gliomas. N Engl J Med 2009;360:765-773.19228619
6. Xu X, Zhao J, Xu Z, Peng B, Huang Q, Arnold E, et al. Structures of human cytosolic
NADP-dependent isocitrate dehydrogenase reveal a novel self-regulatory mechanism of activity. J
Biol Chem 2004;279:33946-33957.15173171
7. Zhao S, Lin Y, Xu W, Jiang W, Zha Z, Wang P, et al. Glioma-derived mutations in IDH1
dominantly
inhibit
IDH1
catalytic
activity
2009;324:261-265.19359588
10
and
induce
HIF-1alpha.
Science
8. Minard KI, McAlister-Henn L. Dependence of peroxisomal beta-oxidation on cytosolic sources of
NADPH. J Biol Chem 1999;274:3402-3406.9920883
9. Shechter I, Dai P, Huo L, Guan G. IDH1 gene transcription is sterol regulated and activated by
SREBP-1a and SREBP-2 in human hepatoma HepG2 cells: evidence that IDH1 may regulate
lipogenesis in hepatic cells. J Lipid Res 2003;44:2169-2180.12923220
10. Ronnebaum SM, Ilkayeva O, Burgess SC, Joseph JW, Lu D, Stevens RD, et al. A pyruvate
cycling pathway involving cytosolic NADP-dependent isocitrate dehydrogenase regulates
glucose-stimulated insulin secretion. J Biol Chem 2006;281:30593-30602.16912049
11. Mailloux RJ, Beriault R, Lemire J, Singh R, Chenier DR, Hamel RD, et al. The tricarboxylic acid
cycle, an ancient metabolic network with a novel twist. PLoS One 2007;2:e690.17668068
12. Nakamura H. Thioredoxin and its related molecules: update 2005. Antioxid Redox Signal
2005;7:823-828.15890030
13. Lee SM, Koh HJ, Park DC, Song BJ, Huh TL, Park JW. Cytosolic NADP(+)-dependent isocitrate
dehydrogenase status modulates oxidative damage to cells. Free Radic Biol Med
2002;32:1185-1196.12031902
14. Balss J, Meyer J, Mueller W, Korshunov A, Hartmann C, von Deimling A. Analysis of the IDH1
codon 132 mutation in brain tumors. Acta Neuropathol 2008;116:597-602.18985363
15. Horbinski C, Kofler J, Kelly LM, Murdoch GH, Nikiforova MN. Diagnostic use of IDH1/2
mutation analysis in routine clinical testing of formalin-fixed, paraffin-embedded glioma tissues. J
Neuropathol Exp Neurol 2009;68:1319-1325.19915484
16. Sanson M, Marie Y, Paris S, Idbaih A, Laffaire J, Ducray F, et al. Isocitrate dehydrogenase 1
codon 132 mutation is an important prognostic biomarker in gliomas. J Clin Oncol
2009;27:4150-4154.19636000
17. Hartmann C, Meyer J, Balss J, Capper D, Mueller W, Christians A, et al. Type and frequency of
IDH1 and IDH2 mutations are related to astrocytic and oligodendroglial differentiation and age: a
study of 1,010 diffuse gliomas. Acta Neuropathol 2009;118:469-474.19554337
18. Ichimura K, Pearson DM, Kocialkowski S, Backlund LM, Chan R, Jones DT, et al. IDH1
mutations are present in the majority of common adult gliomas but rare in primary glioblastomas.
Neuro Oncol 2009;11:341-347.19435942
19. Watanabe T, Nobusawa S, Kleihues P, Ohgaki H. IDH1 mutations are early events in the
development
of
astrocytomas
and
oligodendrogliomas.
Am
J
Pathol
2009;174:1149-1153.19246647
20. Bleeker FE, Lamba S, Leenstra S, Troost D, Hulsebos T, Vandertop WP, et al. IDH1 mutations at
residue p.R132 (IDH1(R132)) occur frequently in high-grade gliomas but not in other solid
tumors. Hum Mutat 2009;30:7-11.19117336
21. Hayden JT, Fruhwald MC, Hasselblatt M, Ellison DW, Bailey S, Clifford SC. Frequent IDH1
mutations in supratentorial primitive neuroectodermal tumors (sPNET) of adults but not children.
Cell Cycle 2009;8:1806-1807.19411854
22. Seiz M, Tuettenberg J, Meyer J, Essig M, Schmieder K, Mawrin C, et al. Detection of IDH1
mutations in gliomatosis cerebri, but only in tumors with additional solid component: evidence for
molecular subtypes. Acta Neuropathol 2010;120:261-267.20514489
23. Mardis ER, Ding L, Dooling DJ, Larson DE, McLellan MD, Chen K, et al. Recurring mutations
found
by
sequencing
an
acute
myeloid
2009;361:1058-1066.19657110
11
leukemia
genome.
N
Engl
J
Med
24. Ward PS, Patel J, Wise DR, Abdel-Wahab O, Bennett BD, Coller HA, et al. The common feature
of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting
alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell 2010;17:225-234.20171147
25. Kang MR, Kim MS, Oh JE, Kim YR, Song SY, Seo SI, et al. Mutational analysis of IDH1 codon
132 in glioblastomas and other common cancers. Int J Cancer 2009;125:353-355.19378339
26. Sjoblom T, Jones S, Wood LD, Parsons DW, Lin J, Barber TD, et al. The consensus coding
sequences of human breast and colorectal cancers. Science 2006;314:268-274.16959974
27. Park SW, Chung NG, Han JY, Eom HS, Lee JY, Yoo NJ, et al. Absence of IDH2 codon 172
mutation in common human cancers. Int J Cancer 2009;125:2485-2486.19530255
28. Green A, Beer P. Somatic mutations of IDH1 and IDH2 in the leukemic transformation of
myeloproliferative neoplasms. N Engl J Med 2010;362:369-370.20107228
29. Murugan AK, Bojdani E, Xing M. Identification and functional characterization of isocitrate
dehydrogenase 1 (IDH1) mutations in thyroid cancer. Biochem Biophys Res Commun
2010;393:555-559.20171178
30. Holdhoff M, Parsons DW, Jr Diaz LA. Mutations of IDH1 and IDH2 are not detected in brain
metastases of colorectal cancer. J Neurooncol 2009;94:297.19350208
31. Lopez GY, Reitman ZJ, Solomon D, Waldman T, Bigner DD, McLendon RE, et al. IDH1(R132)
mutation identified in one human melanoma metastasis, but not correlated with metastases to the
brain. Biochem Biophys Res Commun 2010;398:585-587.20603105
32. Nobusawa S, Watanabe T, Kleihues P, Ohgaki H. IDH1 mutations as molecular signature and
predictive factor of secondary glioblastomas. Clin Cancer Res 2009;15:6002-6007.19755387
33. Labussiere M, Idbaih A, Wang XW, Marie Y, Boisselier B, Falet C, et al. All the 1p19q codeleted
gliomas are mutated on IDH1 or IDH2. Neurology 2010;74:1886-1890.20427748
34. Levine AJ. p53, the cellular gatekeeper for growth and division. Cell 1997;88:323-331.9039259
35. Ohgaki H, Kleihues P. Genetic pathways to primary and secondary glioblastoma. Am J Pathol
2007;170:1445-1453.17456751
36. Furnari FB, Fenton T, Bachoo RM, Mukasa A, Stommel JM, Stegh A, et al. Malignant astrocytic
glioma: genetics, biology, and paths to treatment. Genes Dev 2007;21:2683-2710.17974913
37. Weber RG, Sabel M, Reifenberger J, Sommer C, Oberstrass J, Reifenberger G, et al.
Characterization of genomic alterations associated with glioma progression by comparative
genomic hybridization. Oncogene 1996;13:983-994.8806688
38. Korshunov A, Meyer J, Capper D, Christians A, Remke M, Witt H, et al. Combined molecular
analysis of BRAF and IDH1 distinguishes pilocytic astrocytoma
from diffuse astrocytoma. Acta
Neuropathol 2009;118:401-405.19543740
39. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, et al. MGMT gene
silencing
and
benefit
from
temozolomide
in
glioblastoma.
N
Engl
J
Med
2005;352:997-1003.15758010
40. Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, et al. Effects of
radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival
in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial.
Lancet Oncol 2009;10:459-466.19269895
41. Ducray F, El HS, Idbaih A. Diagnostic and prognostic markers in gliomas. Curr Opin Oncol
2009;21:537-542.19667985
42. McDonald KL, McDonnell J, Muntoni A, Henson JD, Hegi ME, von Deimling A, et al. Presence
12
of alternative lengthening of telomeres mechanism in patients with glioblastoma identifies a less
aggressive
tumor
type
with
longer
survival.
J
Neuropathol
Exp
Neurol
2010;69:729-736.20535033
43. Noushmehr H, Weisenberger DJ, Diefes K, Phillips HS, Pujara K, Berman BP, et al.
Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma.
Cancer Cell 2010;17:510-522.20399149
44. Watanabe T, Vital A, Nobusawa S, Kleihues P, Ohgaki H. Selective acquisition of IDH1 R132C
mutations in astrocytomas associated with Li-Fraumeni syndrome. Acta Neuropathol
2009;117:653-656.19340432
45. De Carli E, Wang X, Puget S. IDH1 and IDH2 mutations in gliomas. N Engl J Med
2009;360:2248, 2249.19458374
46. Pollack IF, Hamilton RL, Sobol RW, Nikiforova MN, Lyons-Weiler MA, Laframboise WA, et al.
IDH1 mutations are common in malignant gliomas arising in adolescents: a report from the
Children's Oncology Group. Childs Nerv Syst 2010.20725730
47. Cairncross G, Berkey B, Shaw E, Jenkins R, Scheithauer B, Brachman D, et al. Phase III trial of
chemotherapy plus radiotherapy compared with radiotherapy alone for pure and mixed anaplastic
oligodendroglioma: Intergroup Radiation Therapy Oncology Group Trial 9402. J Clin Oncol
2006;24:2707-2714.16782910
48. van den Bent MJ, Carpentier AF, Brandes AA, Sanson M, Taphoorn MJ, Bernsen HJ, et al.
Adjuvant procarbazine, lomustine, and vincristine improves progression-free survival but not
overall survival in newly diagnosed anaplastic oligodendrogliomas and oligoastrocytomas: a
randomized European Organisation for
Research and Treatment of Cancer phase III trial. J Clin
Oncol 2006;24:2715-2722.16782911
49. Wick W, Hartmann C, Engel C, Stoffels M, Felsberg J, Stockhammer F, et al. NOA-04
randomized phase III trial of sequential radiochemotherapy of anaplastic glioma with
procarbazine,
lomustine,
and
vincristine
or
temozolomide.
J
Clin
Oncol
2009;27:5874-5880.19901110
50. Felsberg J, Erkwoh A, Sabel MC, Kirsch L, Fimmers R, Blaschke B, et al. Oligodendroglial
tumors: refinement of candidate regions on chromosome arm 1p and correlation of 1p/19q status
with survival. Brain Pathol 2004;14:121-130.15193024
51. Esteller M, Garcia-Foncillas J, Andion E, Goodman SN, Hidalgo OF, Vanaclocha V, et al.
Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to
alkylating
agents. N Engl J Med 2000;343:1350-1354.11070098
52. Herrlinger U, Rieger J, Koch D, Loeser S, Blaschke B, Kortmann RD, et al. Phase II trial of
lomustine plus temozolomide chemotherapy in addition to radiotherapy in newly diagnosed
glioblastoma: UKT-03. J Clin Oncol 2006;24:4412-4417.16983109
53. Glas M, Happold C, Rieger J, Wiewrodt D, Bahr O, Steinbach JP, et al. Long-term survival of
patients with glioblastoma treated with radiotherapy and lomustine plus temozolomide. J Clin
Oncol 2009;27:1257-1261.19188676
54. Bujko M, Kober P, Matyja E, Nauman P, Dyttus-Cebulok K, Czeremszynska B, et al. Prognostic
value of IDH1 mutations identified with PCR-RFLP assay in glioblastoma patients. Mol Diagn
Ther 2010;14:163-169.20560678
55. Weller M, Felsberg J, Hartmann C, Berger H, Steinbach JP, Schramm J, et al. Molecular
predictors of progression-free and overall survival in patients with newly diagnosed glioblastoma:
13
a prospective translational study of the German Glioma Network. J Clin Oncol
2009;27:5743-5750.19805672
56. Toedt G, Barbus S, Wolter M, Felsberg J, Tews B, Blond F, et al. Molecular signatures classify
astrocytic gliomas by IDH1 mutation status. Int J Cancer 2010.20473936
57. Dang L, White DW, Gross S, Bennett BD, Bittinger MA, Driggers EM, et al. Cancer-associated
IDH1 mutations produce 2-hydroxyglutarate. Nature 2009;462:739-744.19935646
58. Gross S, Cairns RA, Minden MD, Driggers EM, Bittinger MA, Jang HG, et al. Cancer-associated
metabolite 2-hydroxyglutarate accumulates in acute myelogenous
leukemia with isocitrate
dehydrogenase 1 and 2 mutations. J Exp Med 2010;207:339-344.20142433
59. Garber K. Oncometabolite? IDH1 discoveries raise possibility of new metabolism targets in brain
cancers and leukemia. J Natl Cancer Inst 2010;102:926-928.20576929
60. Kil IS, Kim SY, Lee SJ, Park JW. Small interfering RNA-mediated silencing of mitochondrial
NADP+-dependent isocitrate dehydrogenase enhances the sensitivity of HeLa cells toward tumor
necrosis factor-alpha and anticancer drugs. Free Radic Biol Med 2007;43:1197-1207.17854715
61. Lee JK, Edderkaoui M, Truong P, Ohno I, Jang KT, Berti A, et al. NADPH oxidase promotes
pancreatic cancer cell survival via inhibiting JAK2 dephosphorylation by tyrosine phosphatases.
Gastroenterology 2007;133:1637-1648.17983808
62. Kil IS, Huh TL, Lee YS, Lee YM, Park JW. Regulation of replicative senescence by NADP+
-dependent isocitrate dehydrogenase. Free Radic Biol Med 2006;40:110-119.16337884
63. Bleeker FE, Atai NA, Lamba S, Jonker A, Rijkeboer D, Bosch KS, et al. The prognostic
IDH1( R132 ) mutation is associated with reduced NADP+-dependent IDH activity in
glioblastoma. Acta Neuropathol 2010;119:487-494.20127344
64. Thompson CB. Metabolic enzymes as oncogenes or tumor suppressors. N Engl J Med
2009;360:813-815.19228626
65. Struys EA. D-2-Hydroxyglutaric aciduria: unravelling the biochemical pathway and the genetic
defect. J Inherit Metab Dis 2006;29:21-29.16601864
66. Brehmer S, Pusch S, Schmieder K, von Deimling A, Hartmann C. Mutational analysis of
D2HGDH and L2HGDH in brain tumours without IDH1 or IDH2 mutations. Neuropathol Appl
Neurobiol 2010.20727073
67. Aghili M, Zahedi F, Rafiee E. Hydroxyglutaric aciduria and malignant brain tumor: a case report
and literature
review. J Neurooncol 2009;91:233-236.18931888
68. Kolker S, Pawlak V, Ahlemeyer B, Okun JG, Horster F, Mayatepek E, et al. NMDA receptor
activation and respiratory chain complex V inhibition contribute to neurodegeneration in
d-2-hydroxyglutaric aciduria. Eur J Neurosci 2002;16:21-28.12153528
69. Latini A, Scussiato K, Rosa RB, Llesuy S, Bello-Klein A, Dutra-Filho CS, et al.
D-2-hydroxyglutaric acid induces oxidative stress in cerebral cortex of young rats. Eur J Neurosci
2003;17:2017-2022.12786967
70. Bayley JP, Devilee P. Warburg tumours and the mechanisms of mitochondrial tumour suppressor
genes. Barking up the right tree? Curr Opin Genet Dev 2010;20:324-329.20304625
71. Hughes JM, Groot AJ, van der Groep P, Sersansie R, Vooijs M, van Diest PJ, et al. Active HIF-1
in the normal human retina. J Histochem Cytochem 2010;58:247-254.19901273
72. MacKenzie ED, Selak MA, Tennant DA, Payne LJ, Crosby S, Frederiksen CM, et al.
Cell-permeating
alpha-ketoglutarate
derivatives
alleviate
pseudohypoxia
dehydrogenase-deficient cells. Mol Cell Biol 2007;27:3282-3289.17325041
14
in
succinate
73. King A, Selak MA, Gottlieb E. Succinate dehydrogenase and fumarate hydratase: linking
mitochondrial dysfunction and cancer. Oncogene 2006;25:4675-4682.16892081
74. Pollard
PJ,
Ratcliffe
PJ.
Cancer.
Puzzling
patterns
of
predisposition.
Science
2009;324:192-194.19359573
75. Frezza C, Tennant DA, Gottlieb E. IDH1 mutations in gliomas: when an enzyme loses its grip.
Cancer Cell 2010;17:7-9.20129244
76. Ducray F, Marie Y, Sanson M. IDH1 and IDH2 mutations in gliomas. N Engl J Med
2009;360:2248-2249, 2249.19469031
77. Meyer J, Pusch S, Balss J, Capper D, Mueller W, Christians A, et al. PCR- and restriction
endonuclease-based detection of IDH1 mutations. Brain Pathol 2010;20:298-300.19744125
78. Horbinski C, Kelly L, Nikiforov YE, Durso MB, Nikiforova MN. Detection of IDH1 and IDH2
mutations by fluorescence melting curve analysis as a
diagnostic tool for brain biopsies. J Mol
Diagn 2010;12:487-492.20431032
79. Chen G, Wu X, Yao Y, Zhou LF, Mao Y. Direct, real-time PCR (MethyLight) assay for
methylation of O6-methylguanine-DNA
methyltransferase promoter in glioma. Chin Med J
(Engl) 2009;122:1342-1345.19567148
80. Felsberg J, Wolter M, Seul H, Friedensdorf B, Goppert M, Sabel MC, et al. Rapid and sensitive
assessment of the IDH1 and IDH2 mutation status in cerebral gliomas based on DNA
pyrosequencing. Acta Neuropathol 2010;119:501-507.20131059
81. Kato Y, Jin G, Kuan CT, McLendon RE, Yan H, Bigner DD. A monoclonal antibody IMab-1
specifically recognizes IDH1R132H, the most common glioma-derived mutation. Biochem
Biophys Res Commun 2009;390:547-551.19818334
82. Capper D, Zentgraf H, Balss J, Hartmann C, von Deimling A. Monoclonal antibody specific for
IDH1 R132H mutation. Acta Neuropathol 2009;118:599-601.19798509
83. Capper D, Sahm F, Hartmann C, Meyermann R, von Deimling A, Schittenhelm J. Application of
mutant IDH1 antibody to differentiate diffuse glioma from nonneoplastic central nervous system
lesions and therapy-induced changes. Am J Surg Pathol 2010;34:1199-1204.20661018
84. Reitman ZJ, Parsons DW, Yan H. IDH1 and IDH2: not your typical oncogenes. Cancer Cell
2010;17:215-216.20227034
85. Kimmelman J, Nalbantoglu J. Faithful companions: a proposal for neurooncology trials in pet
dogs. Cancer Res 2007;67:4541-4544.17510377
86. Reitman ZJ, Olby NJ, Mariani CL, Thomas R, Breen M, Bigner DD, et al. IDH1 and IDH2
hotspot mutations are not found in canine glioma. Int J Cancer 2010;127:245-246.19877121
87. Dubbink HJ, Taal W, van Marion R, Kros JM, van Heuvel I, Bromberg JE, et al. IDH1 mutations
in low-grade astrocytomas predict survival but not response to temozolomide. Neurology
2009;73:1792-1795.19933982
88. van den Bent MJ, Dubbink HJ, Marie Y, Brandes AA, Taphoorn MJ, Wesseling P, et al. IDH1
and IDH2 mutations are prognostic but not predictive for outcome in anaplastic oligodendroglial
tumors: a report of the European Organization for Research and Treatment of Cancer Brain
Tumor Group. Clin Cancer Res 2010;16:1597-1604.20160062
89. Semenza GL. Evaluation of HIF-1 inhibitors as anticancer agents. Drug Discov Today
2007;12:853-859.17933687
15