Glioblastoma is the most frequent and malignant brain tumor. The vast majority of glioblastomas (∼90%) develop rapidly de novo in elderly patients, without clinical or histologic evidence of a less malignant precursor lesion (primary glioblastomas). Secondary glioblastomas progress from low-grade diffuse astrocytoma or anaplastic astrocytoma. They manifest in younger patients, have a lesser degree of necrosis, are preferentially located in the frontal lobe, and carry a significantly better prognosis. Histologically, primary and secondary glioblastomas are largely indistinguishable, but they differ in their genetic and epigenetic profiles. Decisive genetic signposts of secondary glioblastoma are IDH1 mutations, which are absent in primary glioblastomas and which are associated with a hypermethylation phenotype. IDH1 mutations are the earliest detectable genetic alteration in precursor low-grade diffuse astrocytomas and in oligodendrogliomas, indicating that these tumors are derived from neural precursor cells that differ from those of primary glioblastomas. In this review, we summarize epidemiologic, clinical, histopathologic, genetic, and expression features of primary and secondary glioblastomas and the biologic consequences of IDH1 mutations. We conclude that this genetic alteration is a definitive diagnostic molecular marker of secondary glioblastomas and more reliable and objective than clinical criteria. Despite a similar histologic appearance, primary and secondary glioblastomas are distinct tumor entities that originate from different precursor cells and may require different therapeutic approaches. Clin Cancer Res; 19(4); 764–72. ©2012 AACR.

Historical perspective

From a biologic and clinical point of view, the secondary glioblastomas developing in astrocytomas must be distinguished from “primary” glioblastomas. They are probably responsible for most of the glioblastomas of long clinical duration.

H.-J. Scherer (1940; ref. 1).

In a series of publications on gliomas, Hans-Joachim Scherer (1906–1945), a young German neuropathologist working in exile at the Institut Bunge (Antwerp, Belgium) was decades ahead of his contemporaries in his insight into the pathology and biology of brain tumors (2). The distinction between primary and secondary glioblastomas was a remarkable observation at that time. In the first edition of the World Health Organization (WHO) Classification of Tumors of the Nervous System, published 40 years later, glioblastomas were not even recognized as astrocytic neoplasms but listed in a group of “poorly differentiated and embryonal tumors” (3). Only after the introduction of immunohistochemistry was their astrocytic origin unequivocally established (4). Although it was recognized in clinical practice that some glioblastomas developed after resection of low-grade or anaplastic astrocytomas, Scherer's distinction remained conceptual as histopathologically these subtypes could not reliably be distinguished.

In 1996, we reported evidence that primary and secondary glioblastomas carry distinct genetic alterations (5). TP53 mutations were found to be uncommon in primary glioblastomas but occurred with a high incidence in secondary glioblastomas; EGF receptor (EGFR) overexpression prevailed in primary glioblastomas but was rare in secondary glioblastomas. Only 1 of 49 glioblastomas showed TP53 mutation and EGFR overexpression, indicating that these alterations are mutually exclusive events defining 2 different genetic pathways in the evolution of glioblastoma (5). Subsequently, many studies provided evidence that primary and secondary glioblastomas develop through distinct genetic pathways (6, 7). Typical for primary glioblastomas are EGFR amplification, PTEN mutation, and entire loss of chromosome 10 (6–8). Genetic alterations more common in secondary glioblastomas include TP53 mutations and 19q loss (6, 7, 9). However, until the identification of IDH1 mutation as a molecular marker of secondary glioblastoma (10–13), the patterns of genetic alterations, though different, did not allow an unequivocal separation of the 2 subtypes.

IDH1/2 mutations in neural tumors

IDH1 mutations were first reported by Parsons and colleagues (14) in 2008, and in this first study the authors already pointed out that “mutations in IDH1 occurred in a large fraction of young patients and in most patients with secondary glioblastomas and were associated with an increase in overall survival.” Subsequent studies showed that these mutations are very frequent in secondary (>80%) but very rare in primary glioblastomas (<5%; refs. 10–13). IDH1 mutations as a genetic marker of secondary, but not primary, glioblastoma closely correspond to the respective clinical diagnosis in 385 of 407 (95%) of cases (13). It is now agreed that IDH1 mutation is a definitive diagnostic molecular marker of secondary glioblastomas and more reliable and objective than clinical and/or pathologic criteria.

IDH1 mutations are frequent (>80%) in diffuse astrocytoma WHO grade II and anaplastic astrocytoma WHO grade III, the precursor lesions of secondary glioblastomas, as well as in oligodendroglial tumors including oligodendroglioma WHO grade II, anaplastic oligodendroglioma WHO grade III, oligoastrocytoma WHO grade II, and anaplastic oligoastrocytoma WHO grade III (10–12, 15). In contrast, IDH1 mutations are very rare or absent in pilocytic astrocytomas, as well as in most other CNS neoplasms, including ependymomas, medulloblastomas, and meningiomas (10–12, 15). IDH2 mutations are less frequent and prevail in anaplastic oligodendrogliomas (∼5%) and oligoastrocytomas (∼6%; ref. 12).

All IDH1 mutations reported are located at the first or second base of codon 132 (10, 11, 14). Most frequent is R132H (CGT–>CAT), observed in 83% to 91% of IDH1 mutations in astrocytic and oligodendroglial gliomas (10–12). Other mutations are rare, including R132C (CGT–>TGT; 3.6%–4.6%; refs. 10–12), R132G (0.6%–3.8%; refs. 10–12), R132S (0.8%–2.5%; refs. 10–12), and R132L (0.5%–4.4%; refs. 10, 12). IDH2 mutations are located at codon 172 (12), with R172K being most frequent.

IDH1/2 mutations in non-neural tumors

IDH1/2 mutations are absent or very rare in most tumors at other organ sites, including bladder, breast, stomach, colorectum, lung, liver, ovary, and prostate (12, 15). Exceptions are central chondrosarcomas (∼55%; ref. 16), intrahepatic cholangiocarcinomas (23%; ref. 17), acute myelogenous leukemia (AML; 15%–20%; refs. 18–23), angioimmunoblastic T-cell lymphoma (AITL; ∼20%; ref. 24), malignant melanoma (∼10%; ref. 25), and anaplastic thyroid cancer (∼10%; ref. 26). This suggests that IDH1/2 mutations may confer a growth advantage in specific cell lineages at defined stages of development and differentiation.

Are there primary glioblastomas with IDH1 mutations?

In a population-based study, only 14 of 407 glioblastomas clinically diagnosed as primary (3.4%) carried an IDH1 mutation (13). These patients were 10 years younger and their genetic profiles were similar to those of secondary glioblastomas, including frequent TP53 mutations and absence of EGFR amplification (13). Similarly, several hospital-based studies showed that primary glioblastomas with IDH1 mutations were 13 to 27 years younger than those without IDH1 mutations (10, 12, 27, 28). Toedt and colleagues (27) showed that primary glioblastomas with IDH1 mutations have gene expression profiles similar to those of IDH1-mutated secondary glioblastomas. Glioblastomas with IDH1 mutations clinically diagnosed as primary may have rapidly progressed from precursor lesions that escaped clinical diagnosis and are likely to have been misclassified as primary glioblastoma.

Are there secondary glioblastomas that do not have IDH1 mutations?

Secondary glioblastomas lacking IDH1 mutations have infrequent TP53 mutations and patients have a shorter clinical history (13). Furthermore, most secondary glioblastomas lacking IDH1 mutations (7 of 8) had developed through progression from an anaplastic glioma (WHO grade III), whereas the majority of secondary glioblastomas with IDH1 mutations had progressed from a WHO grade II glioma (13). The possibility therefore exists that some tumors diagnosed as anaplastic astrocytoma were actually primary glioblastomas that were misdiagnosed because of a sampling error. In the absence of diagnostic hallmarks, that is, necrosis and/or microvascular proliferation, pathologists hesitate to make a diagnosis of glioblastoma even if MRI suggests this.

Timing of IDH1/2 mutations in the pathway to secondary glioblastoma

IDH1/2 mutations are an early event in gliomagenesis and persist during progression to secondary glioblastoma. In addition to frequent IDH1/2 mutations, about 65% of diffuse astrocytomas carry a TP53 mutation, whereas oligodendrogliomas show frequent 1p/19q loss (>75%; refs. 11, 29–33). IDH1/2 mutations are likely to occur before TP53 mutation or 1p/19q loss, as low-grade diffuse gliomas carrying only IDH1/2 mutations are more frequent (17%) than those carrying only a TP53 mutation (2%) or those showing only 1p/19q loss (3%; ref. 33). Furthermore, the analysis of multiple biopsies from the same patient revealed that there were no cases in which an IDH1 mutation occurred after the acquisition of a TP53 mutation or loss of 1p/19q (11, 33).

Acquisition of 1p/19q loss in cells with IDH1/2 mutations may be the driving force toward oligodendroglial differentiation in low-grade diffuse glioma (11, 31, 33). It has been shown that tumors with the typical histologic signature of oligodendroglioma (e.g., honeycomb appearance of most neoplastic cells) showed loss at 1p/19q in the vast majority of cases (>90%; ref. 31). Furthermore, exomic sequencing recently revealed that mutations in the CIC gene (homolog of the Drosophila gene capicua) at 19q13.2 and in the FUBP1 gene at 1p are frequent in oligodendrogliomas but rare or absent in diffuse astrocytomas (34–36).

Astrocytomas typically develop in cells with IDH1/2 mutations that subsequently acquire TP53 mutations. Recent studies have also described mutations in the ATRX (α-thalassemia/mental-retardation-syndrome-X-linked) gene that are often copresent with IDH1/2 mutations and TP53 mutations in diffuse astrocytomas WHO grades II/III and secondary glioblastomas (36, 37). Our current concept of the genetic pathways leading to astrocytic and oligodendroglial gliomas is summarized in Figure 1.

Figure 1.

Genetic pathways to primary and secondary glioblastomas. Note that only secondary glioblastomas share common origin of cells with oligodendrogliomas.

Figure 1.

Genetic pathways to primary and secondary glioblastomas. Note that only secondary glioblastomas share common origin of cells with oligodendrogliomas.

Close modal

Only 7% of WHO grade II diffuse gliomas had none of these genetic alterations (i.e., IDH1/2 mutations, TP53 mutations, and 1p/19q loss) and were termed “triple negative” (33). These cases are still poorly understood; a minor fraction shows loss of cell-cycle control regulated by the RB1 pathway (38). The possibility cannot be excluded that they are derived from a different precursor cell population.

IDH1 mutations in gliomas associated with the Li-Fraumeni syndrome

As indicated earlier, IDH1 mutations precede TP53 mutations in sporadic astrocytic tumors. Patients with Li-Fraumeni syndrome (LFS) carry a germline TP53 mutation that is present in every somatic cell. Thus, by definition TP53 mutations would be the first event in LFS-associated gliomas, which account for 12% to 13% of all tumors occurring in LFS families (39, 40). In patients from 3 families with LFS, we identified IDH1 mutations in 5 astrocytic gliomas that developed in carriers of a TP53 germline mutation. Without exception, all contained the R132C (CGT–>TGT) mutation (41), which in sporadic astrocytic tumors amounts to less than 5% of all IDH1 mutations (10–12). This remarkably selective occurrence suggests a preference for R132C mutations in neural precursor cells that already carry a germline TP53 mutation.

The mechanisms by which IDH1 mutations contribute to the development and malignant progression of astrocytic and oligodendroglial tumors are still not fully understood. Conditional IDH1 (R132H) knockin mice with expression in all hematopoietic cells or cells of the myeloid lineage caused an increased number of early hematopoietic progenitors with splenomegaly, anemia, and extramedullary hematopoiesis (42), whereas brain-specific IDH1 (R132H)-conditional knockin mice exhibited hemorrhage and perinatal lethality (43). Like EGFR amplification in primary glioblastomas, IDH1 mutations in secondary glioblastomas are typically lost during culture in vitro (44). This is enigmatic, since selective suppression of endogenous mutant IDH1 expression in a fibrosarcoma cell line with a native IDH1R132C heterozygous mutation significantly inhibits cell proliferation (45). Only recently, using a neurosphere culture method, it has been possible to establish a brain tumor stem cell line from an IDH1-mutant anaplastic oligoastrocytoma with an endogenous IDH1 mutation and detectable production of 2-hydroxyglutarate (2HG; ref. 46). This may suggest that IDH1-mutant glioma cells have stem cell–like features that confer a growth advantage under neurosphere culture conditions. Alternatively, there may be intratumoral heterogeneity of IDH1 mutations, and neoplastic cells lacking IDH1 mutations are positively selected during culture.

Impaired enzymatic activity and accumulation of 2-hydroxyglutarate

The IDH1 gene encodes isocitrate dehydrogenase 1, an enzyme participating in the citric acid (Krebs) cycle (47, 48), the metabolic pathway used by aerobic organisms to generate usable energy (49). IDH1/2 mutations reduce the wild-type activity of the enzyme, that is, the conversion of isocitrate to α-ketoglutarate (α-KG), and increase levels of hypoxia-inducible factor-1α (HIF-1α), a transcription factor, and its targets (e.g., GLUT1, VEGF, and PGK1; ref. 50). Importantly, IDH1/2 mutations are gain-of-function mutations that also produce the oncometabolite 2HG from α-KG (51, 52). The production of 2HG is a function shared by all the commonly occurring IDH1/2 mutants analyzed (51–53). Malignant IDH1mut gliomas contain an increased (up to 100-fold) concentration of 2HG (51). Cells from brain-specific IDH1 (R132H)-conditional knockin mice also show high levels of 2HG that are associated with inhibited prolyl-hydroxylation of HIF-1α and upregulation of its target genes (43). 2HG also blocks prolyl-hydroxylation of collagen, causing a defect in collagen protein maturation, leading to basement-membrane aberrations that may play a role in glioma progression (43).

Hypermethylation phenotype

Noushmehr and colleagues (54) first reported that IDH1/2mut glioblastomas displayed concerted CpG island methylation at a large number of loci. A similar hypermethylation phenotype was also observed in IDH1/2mut diffuse astrocytomas (55) and oligodendroglial tumors (55), as well as in IDH1/2mut AML (56). Turcan and colleagues (57) introduced mutant IDH1 into primary human astrocytes, causing alteration of specific histone markers and induction of extensive DNA hypermethylation, suggesting that the presence of an IDH1 mutation is sufficient to establish a hypermethylation phenotype in glioma. This is supported by the observation that the expression of IDH1 (R132H) in cells of myeloid lineage in knockin mice resulted in hypermethylated histones and changes in DNA methylation similar to those observed in human IDH1/2mut AML (42). Stable transfection of a 2HG-producing IDH mutant into immortalized astrocytes resulted in progressive accumulation of histone methylation, which was associated with repression of the inducible expression of lineage-specific differentiation genes and a block to differentiation, suggesting that 2HG-producing IDH1/2 mutants can prevent the histone demethylation that is required for lineage-specific progenitor cells to differentiate into terminally differentiated cells (58). Duncan and colleagues (59) knocked in a single copy of the IDH1 R132H into a human cancer cell line and profiled changes in DNA methylation in more than 27,000 CpG dinucleotides. Heterozygous expression of mutant IDH1 was sufficient to induce widespread alterations in DNA methylation, including hypermethylation of 2,010 and hypomethylation of 842 CpG loci, many of which were consistent with those observed in IDH1-mutant and glioma–CpG island methylator phenotype (G-CIMP)+ primary gliomas (59).

Glioblastomas have been classified on the basis of cDNA expression profiles, with distinct proneural, neural, classical, mesenchymal, and proliferative patterns (60, 61). Most IDH1mut glioblastomas (11 of 12, 92%) show a proneural expression signature; conversely, approximately 30% of glioblastomas with a proneural signature are IDH1mut (61). Glioblastomas lacking IDH1 mutations have all been identified as classical, mesenchymal, neural, or proneural (61).

These observations suggest that secondary glioblastomas are a rather homogeneous group of tumors characterized by a proneural expression pattern, whereas primary glioblastomas are heterogeneous, with several distinct expression profiles. Diffuse astrocytomas, oligodendrogliomas, and oligoastrocytomas all show the typical proneural signatures (62), again supporting the view that these neoplasms share common neural progenitor cells.

Until the discovery of IDH1 mutations as a molecular marker, the distinction between primary and secondary glioblastomas was based on clinical observations. Tumors were considered primary if the diagnosis of glioblastoma was made at the first biopsy, without radiologic or histologic evidence of a preexisting, less malignant precursor lesion. The diagnosis of secondary glioblastoma required neuroimaging and/or histologic evidence of a preceding low-grade or anaplastic astrocytoma (6, 7).

In developed countries, the annual incidence of glioblastomas is usually in the range of 3 to 4 cases per 100,000 persons per year (7, 63–65). In a population-based study in Switzerland, using clinical criteria and histopathologic evidence, only 5% of all glioblastomas diagnosed were secondary (7, 63). Similarly, a study by the University of Alabama (Tuscaloosa, AL) showed that 19 of 392 (5%) cases of glioblastoma had a histologically proven prior low-grade glioma (66). When IDH1 mutations are used as a genetic marker, secondary glioblastomas accounted for 9% of all glioblastomas at the population level (13) and for 6% to 13% in hospital-based studies (10, 12, 67, 68).

The combined incidence rates of low-grade and anaplastic astrocytomas are approximately twice as high as that of clinically diagnosed secondary glioblastoma or IDH1mut glioblastoma (30, 64, 69). This may be explained at least in part by the fact that some patients with low-grade or anaplastic astrocytoma succumb to the disease before progression to glioblastoma occurs. Furthermore, cases with rapid progression from low-grade or anaplastic astrocytoma may be misclassified as primary glioblastoma.

There is a striking difference in the age distribution of patients with primary and secondary glioblastoma. At a population level, the mean age of patients with glioblastoma clinically diagnosed as primary was 62 years, whereas secondary glioblastomas developed in younger patients (mean, 45 years; refs. 7, 63). Similarly, the mean ages of patients with or without IDH1 mutations were 61 and 48 years, respectively (Table 1; ref. 13). Several hospital-based studies showed that patients with IDH1mut glioblastoma are significantly younger (mean age, 32–41 years) than those without IDH1 mutations (mean age, 56–59 years; refs. 12, 67, 70).

Table 1.

Primary and secondary glioblastomas: comparison of clinical versus genetic diagnosis

Primary glioblastomaSecondary glioblastoma
Clinical criteriaaGenetic criteria (IDH1wt)Clinical criteriaaGenetic criteria (IDH1mut)References
Fraction in a population 94.7% 91.2% 5.3% 8.8% (7, 13) 
Mean age, y 59–62 56–61 33–45 32–48 (7, 12, 13, 67, 70) 
Male/female ratio 1.33–1.5 1.2–1.46 0.65–2.3 1.0–1.12 (7, 12, 13, 70) 
Mean clinical history, mo 6.3 3.9 16.8 15.2 (7, 13) 
Median overall survival, mo     (7, 12, 13) 
 Surgery + radiotherapy 4.7b 9.9 7.8b 24 (7, 13) 
 Surgery + radio/chemotherapy  15  31 (12) 
Histologic features 
 Oligodendroglial comp. 18% 20% 42% 54% (13, 80) 
 Necrosis 89% 90% 63% 50% (13, 80) 
Genetic alterations 
IDH1 mutations 4–7% 0% 73–88% 100% (10, 12, 13) 
TP53 mutations 17–35% 19–27% 60–88% 76–81% (7, 10, 12, 13, 67, 91) 
ATRX mutations 4–7%  57–80%  (36, 37) 
EGFR amplification 36–45% 35–39% 0–8% 0–6.5% (7, 10, 12, 13, 91) 
CDKN2A deletion 31–52% 30–45% 19–20% 7–22% (7, 12, 13, 91) 
PTEN mutations 23–25% 24–26% 4–12% 0–8% (7, 12, 13) 
 19q loss 6% 4% 54% 32% (9, 13) 
 1p/19q loss 2–8%  0–13%  (10, 12, 67) 
 10p loss 47%  8%  (8) 
 10q loss 70% 67% 63% 73% (7, 13) 
Primary glioblastomaSecondary glioblastoma
Clinical criteriaaGenetic criteria (IDH1wt)Clinical criteriaaGenetic criteria (IDH1mut)References
Fraction in a population 94.7% 91.2% 5.3% 8.8% (7, 13) 
Mean age, y 59–62 56–61 33–45 32–48 (7, 12, 13, 67, 70) 
Male/female ratio 1.33–1.5 1.2–1.46 0.65–2.3 1.0–1.12 (7, 12, 13, 70) 
Mean clinical history, mo 6.3 3.9 16.8 15.2 (7, 13) 
Median overall survival, mo     (7, 12, 13) 
 Surgery + radiotherapy 4.7b 9.9 7.8b 24 (7, 13) 
 Surgery + radio/chemotherapy  15  31 (12) 
Histologic features 
 Oligodendroglial comp. 18% 20% 42% 54% (13, 80) 
 Necrosis 89% 90% 63% 50% (13, 80) 
Genetic alterations 
IDH1 mutations 4–7% 0% 73–88% 100% (10, 12, 13) 
TP53 mutations 17–35% 19–27% 60–88% 76–81% (7, 10, 12, 13, 67, 91) 
ATRX mutations 4–7%  57–80%  (36, 37) 
EGFR amplification 36–45% 35–39% 0–8% 0–6.5% (7, 10, 12, 13, 91) 
CDKN2A deletion 31–52% 30–45% 19–20% 7–22% (7, 12, 13, 91) 
PTEN mutations 23–25% 24–26% 4–12% 0–8% (7, 12, 13) 
 19q loss 6% 4% 54% 32% (9, 13) 
 1p/19q loss 2–8%  0–13%  (10, 12, 67) 
 10p loss 47%  8%  (8) 
 10q loss 70% 67% 63% 73% (7, 13) 

aTumors were considered to be primary if the diagnosis of glioblastoma was made at the first biopsy, without clinical or histological evidence of a preexisting, less malignant precursor lesion, whereas the diagnosis of secondary glioblastoma required histological and/or clinical (neuroimaging) evidence of a preceding low-grade or anaplastic astrocytoma.

bData from population-based study: all the patients who were treated in different ways were included.

Glioblastomas predominantly affect males, with a population-based M/F ratio ranging from 1.28 (7) to 1.32 (64, 65). In contrast, diffuse astrocytomas (WHO grade II) have a less pronounced male predominance, with M/F ratios of approximately 1.17 (29, 65). Because secondary glioblastomas typically develop from diffuse astrocytomas, one would expect that they have a similar gender ratio. This is indeed the case. In a population-based study, IDH1mut secondary glioblastomas had an M/F ratio of 1.12, significantly lower than the ratio of 1.46 in patients with primary glioblastoma (7). Several hospital-based studies also showed a tendency toward a lower M/F ratio of patients with secondary glioblastomas (5, 71–74). In a recent multicenter study, 49 of 618 glioblastomas (7.9%) carried an IDH1 mutation and had an M/F ratio of 0.96, in contrast to a ratio of 1.63 for nonmutated (primary) glioblastomas (68).

At a population level, the majority of patients with primary glioblastoma (68%) had a clinical history of less than 3 months (6). The mean duration of the clinical history of patients with primary and secondary glioblastoma was 6.3 and 16.8 months, respectively (7, 63). Similarly, patients with glioblastomas lacking IDH1 mutations had a mean duration of preceding clinical symptoms of 3.9 months, significantly shorter than patients with IDH1mut glioblastoma (mean, 15.2 months; P = 0.0003; ref. 13). Glioblastomas clinically diagnosed as primary had a mean clinical history of about 4 (IDHwt) and 29 months (IDHmut), respectively (13). It remains to be analyzed why the precursor lesions of these IDHmut secondary glioblastomas escaped clinical detection despite their long clinical history.

Lai and colleagues (68) reported that glioblastomas lacking IDH1 mutations show widespread anatomic distribution, whereas IDH1mut glioblastomas have a striking predominance of frontal lobe involvement, in particular in the region surrounding the rostral extension of the lateral ventricles. Stockhammer and colleagues (75) showed that IDH1/2mut WHO grade II astrocytomas tend to develop in a frontal location, and that seizures were the initial symptom in approximately 70% of patients. According to Zlatescu and colleagues (76) oligodendroglial tumors with 1p/19q losses occur most frequently in the frontal lobe and have a tendency for widespread growth across the midline. Similarly, Laigle-Donadey and colleagues (77) showed that oligodendrogliomas with 1p/19q loss were located predominantly in the frontal lobe. These observations suggest that oligodendrogliomas, astrocytomas, and secondary glioblastomas derived thereof originate from precursor cells located in or migrating to the frontal lobe.

Already in his 1940 publication, Scherer noted “the absence of extensive necrosis and peritumoral brain swelling in secondary and their almost constant presence in primary glioblastomas may play a certain role in the clinical behavior of the two types” (1). He attributed this to the slower growth rate of secondary glioblastomas. It is now understood that a hypoxia-mediated activation of the coagulation system causes intravascular thrombosis, further increases intratumoral hypoxia, and leads to abnormal endothelial cell proliferation and tumor necrosis (78, 79). Microvascular proliferation is induced by VEGF, which shows a markedly higher expression in primary than in secondary glioblastomas (71).

Histopathologically, large areas of ischemic and/or pseudopalisading necrosis are more frequent in primary (89%) than secondary glioblastomas (63%; P = 0.0014; ref. 80) and glioblastomas without IDH1 mutations (90% vs. 50%; P < 0.0001; ref. 13; Table 1). This was confirmed in clinical MRI studies showing that necrosis was less frequent in IDH1mut glioblastomas, while exhibiting more frequent nonenhancing tumor components, larger size at diagnosis, lesser extent of edema, and increased prevalence of cystic and diffuse components (68).

According to the 2007 WHO classification, histologic criteria for the diagnosis of glioblastoma include nuclear atypia, cellular pleomorphism, mitotic activity, vascular thrombosis, microvascular proliferation, and necrosis (29). Glioblastomas may show significant intertumoral and intratumoral heterogeneity, both histologically and genetically (80–82) and this may also apply to glioma-initiating cells (82, 83). This heterogeneity reflects genomic instability and, occasionally, focal new clones arising as a result of additional genetic alterations can be seen histologically (84, 85). Areas with oligodendroglioma-like components are significantly more frequent in secondary than primary glioblastomas (42% vs. 18%; P = 0.0138; ref. 80) and, accordingly, more frequent in IDH1mut glioblastomas than in IDH1wt glioblastomas (54% vs. 20%; P < 0.0001; ref. 13; Table 1). An increase in the fraction of tumor cells with oligodendroglial morphology in IDH1mut glioblastomas was also reported in a large study of 618 cases (68). This is not surprising as secondary glioblastomas assumedly share IDH1mut precursor cells with oligodendrogliomas (Fig. 1).

In a population-based study, the median overall survival of clinically diagnosed secondary glioblastoma was 7.8 months, significantly longer than the survival of patients with primary glioblastoma (4.7 months; P = 0.003; ref. 7). Similarly, the analysis of patients with glioblastoma who were treated with surgery and radiotherapy showed that the mean overall survival time of patients with IDH1mut glioblastoma was 27.1 months, more than twice as long as that of patients with IDH1wt glioblastoma (11.3 months; P < 0.0001; ref. 13). Yan and colleagues (12) reported that IDH1mut glioblastomas treated with radio/chemotherapy had an overall survival time of 31 months, again twice as long as IDH1wt tumors. A different response to therapy is also supported by the observation that dose enhancement did not improve the outcome of glioblastomas with a proneural signature (containing IDH1mut cases), in contrast to glioblastomas with neural, classical, or mesenchymal signature, which all profited from more intensive therapy (61). If confirmed in prospective clinical trials, this may eventually allow for dose deescalation in patients with secondary glioblastoma.

Before the discovery of IDH1 mutations as a lineage marker, it was assumed that primary and secondary glioblastomas developed from the same precursor cell population but showed distinct clinical and biologic behavior due to the acquisition of different genetic alterations (6). There is now increasing evidence that despite their similar histologic features, primary and secondary glioblastomas develop from different cells of origin. The evidence supporting this hypothesis includes the observations that: (i) only secondary glioblastomas but not primary glioblastomas share common IDH1/2 mutations with oligodendrogliomas; (ii) primary and secondary glioblastomas develop in patients of different age groups and have a different sex distribution; (iii) primary and secondary glioblastomas are located in different brain regions; and (iv) primary and secondary glioblastomas have a significantly different clinical outcome. All these data suggest that primary and secondary glioblastomas are in fact different tumor entities that are derived from distinctly different neural precursor cells.

There is also evidence that cancer stem cells in primary and secondary glioblastomas may also be different. In one study, the relative content of CD133+ cells was significantly higher in primary than in secondary glioblastomas, and CD133+ expression was associated with neurosphere formation only in primary but not secondary glioblastomas (86).

Despite differences in clinical history and genetic, epigenetic, and expression profiles, primary and secondary glioblastomas are histologically largely indistinguishable, except that extensive necrosis is more frequent in primary glioblastomas and oligodendroglioma components are more frequent in secondary glioblastomas (80; Table 1). This similarity may be attributable to genetic alterations that are common to both primary and secondary glioblastomas.

The most frequent genetic alteration shared by both primary and secondary glioblastomas is LOH at 10q (∼60% of cases; refs. 6–8, 87–89), the most commonly deleted region being 10q25-qter, distal to D10S1683 (8). Because mutations of the PTEN gene located at 10q23.3 prevail in primary glioblastomas (∼25%), but are rare in secondary glioblastomas (<5%; refs. 6, 7, 90), loss of function of gene(s) other than PTEN is likely to be responsible for the common malignant phenotype. Circumscribed glioblastoma foci in low-grade diffuse astrocytoma or anaplastic astrocytoma show additional deletions at 10q25-qter, distal to D10S597, including the DMBT1 and FGFR2 loci (84). This suggests that the acquisition of a highly malignant glioblastoma phenotype is associated with loss of putative tumor suppressor gene(s) on 10q25-qter.

The ability to predict the pace of progression from low-grade diffuse astrocytoma to secondary glioblastoma would be clinically very important by giving oncologists a rational basis for deciding whether and at which stage to administer adjuvant radiotherapy. Histopathologically, this is not possible, as Scherer noted already in 1940: “Why certain astrocytomas become transformed into glioblastomas while others remain pure, is still obscure. No morphological sign explaining or announcing this tendency could be found” (1).

When commonly deleted genes at 10q25-qter in IDHwt and IDHmut glioblastomas were searched for in The Cancer Genome Atlas (TCGA; ref. 91), 10 genes were identified with log-ratio thresholds of −1.0, and, of these, DMBT1 at 10q26.13 was the only homozygously deleted gene in glioblastomas with or without IDH1 mutations (12.5% vs. 8.0%; ref. 92). DMBT1 homozygous deletion was detected at a similar frequency in an independent set of primary and secondary glioblastomas (20% vs. 21%; ref. 92). A small fraction (11.3%) of diffuse astrocytomas WHO grade II also showed a DMBT1 homozygous deletion, and this was significantly associated with shorter overall patient survival (92). A similar approach was used to search for commonly amplified genes in IDHwt and IDHmut glioblastomas in the TCGA database. A total of 25 genes were identified, of which 21 were located at 7q31-34 (93). Further analyses revealed gain of the MET gene at 7q31.2 in primary glioblastomas (47%) and secondary glioblastomas (44%). Interestingly, MET gain is also common in diffuse astrocytomas (38%), and was associated with shorter survival (93). These results indicate that several genetic alterations frequent in both primary and secondary glioblastomas may be responsible for the common histologic phenotype and, if already present in diffuse astrocytomas, may predict unfavorable clinical outcome (92, 93). Whole-genome DNA sequencing of diffuse astrocytomas (WHO grade II) and anaplastic astrocytomas (WHO grade III) in patients with favorable or poor outcome is likely to identify further genetic alterations that drive the malignant progression to secondary glioblastoma.

No potential conflicts of interest were disclosed.

Conception and design: H. Ohgaki, P. Kleihues

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): H. Ohgaki

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): H. Ohgaki, P. Kleihues

Writing, review, and/or revision of the manuscript: H. Ohgaki, P. Kleihues

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): H. Ohgaki, P. Kleihues

Study supervision: H. Ohgaki

1.
Scherer
HJ
. 
Cerebral astrocytomas and their derivatives
.
Am J Cancer
1940
;
40
:
159
98
.
2.
Peiffer
J
,
Kleihues
P
. 
Hans-Joachim Scherer (1906–1945), pioneer in glioma research
.
Brain Pathol
1999
;
9
:
241
5
.
3.
Zulch
KJ
. 
Histological typing of tumours of the central nervous system
.
Geneva
:
World Health Organization
; 
1979
.
4.
Kleihues
P
,
Burger
PC
,
Scheithauer
BW
. 
Histological typing of tumours of the central nervous system
. 2nd ed.
Springer-Verlag
; 
1993
.
5.
Watanabe
K
,
Tachibana
O
,
Sato
K
,
Yonekawa
Y
,
Kleihues
P
,
Ohgaki
H
. 
Overexpression of the EGF receptor and p53 mutations are mutually exclusive in the evolution of primary and secondary glioblastomas
.
Brain Pathol
1996
;
6
:
217
24
.
6.
Ohgaki
H
,
Kleihues
P
. 
Genetic pathways to primary and secondary glioblastoma
.
Am J Pathol
2007
;
170
:
1445
53
.
7.
Ohgaki
H
,
Dessen
P
,
Jourde
B
,
Horstmann
S
,
Nishikawa
T
,
Di Patre
PL
, et al
Genetic pathways to glioblastoma: a population-based study
.
Cancer Res
2004
;
64
:
6892
9
.
8.
Fujisawa
H
,
Reis
RM
,
Nakamura
M
,
Colella
S
,
Yonekawa
Y
,
Kleihues
P
, et al
Loss of heterozygosity on chromosome 10 is more extensive in primary (de novo) than in secondary glioblastomas
.
Lab Invest
2000
;
80
:
65
72
.
9.
Nakamura
M
,
Yang
F
,
Fujisawa
H
,
Yonekawa
Y
,
Kleihues
P
,
Ohgaki
H
. 
Loss of heterozygosity on chromosome 19 in secondary glioblastomas
.
J Neuropathol Exp Neurol
2000
;
59
:
539
43
.
10.
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
.
11.
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
53
.
12.
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
73
.
13.
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
7
.
14.
Parsons
DW
,
Jones
S
,
Zhang
X
,
Lin
JC-H
,
Leary
RJ
,
Angenendt
P
, et al
An integrated genomic analysis of human glioblastoma multiforme
.
Science
2008
;
321
:
1807
12
.
15.
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
.
16.
Amary
MF
,
Bacsi
K
,
Maggiani
F
,
Damato
S
,
Halai
D
,
Berisha
F
, et al
IDH1 and IDH2 mutations are frequent events in central chondrosarcoma and central and periosteal chondromas but not in other mesenchymal tumours
.
J Pathol
2011
;
224
:
334
43
.
17.
Borger
DR
,
Tanabe
KK
,
Fan
KC
,
Lopez
HU
,
Fantin
VR
,
Straley
KS
, et al
Frequent mutation of isocitrate dehydrogenase (IDH)1 and IDH2 in cholangiocarcinoma identified through broad-based tumor genotyping
.
Oncologist
2012
;
17
:
72
9
.
18.
Mardis
ER
,
Ding
L
,
Dooling
DJ
,
Larson
DE
,
McLellan
MD
,
Chen
K
, et al
Recurring mutations found by sequencing an acute myeloid leukemia genome
.
N Engl J Med
2009
;
361
:
1058
66
.
19.
Dang
L
,
Jin
S
,
Su
SM
. 
IDH mutations in glioma and acute myeloid leukemia
.
Trends Mol Med
2010
;
16
:
387
97
.
20.
Patel
KP
,
Ravandi
F
,
Ma
D
,
Paladugu
A
,
Barkoh
BA
,
Medeiros
LJ
, et al
Acute myeloid leukemia with IDH1 or IDH2 mutation: frequency and clinicopathologic features
.
Am J Clin Pathol
2011
;
135
:
35
45
.
21.
Paschka
P
,
Schlenk
RF
,
Gaidzik
VI
,
Habdank
M
,
Kronke
J
,
Bullinger
L
, et al
IDH1 and IDH2 mutations are frequent genetic alterations in acute myeloid leukemia and confer adverse prognosis in cytogenetically normal acute myeloid leukemia with NPM1 mutation without FLT3 internal tandem duplication
.
J Clin Oncol
2010
;
28
:
3636
43
.
22.
Abbas
S
,
Lugthart
S
,
Kavelaars
FG
,
Schelen
A
,
Koenders
JE
,
Zeilemaker
A
, et al
Acquired mutations in the genes encoding IDH1 and IDH2 both are recurrent aberrations in acute myeloid leukemia: prevalence and prognostic value
.
Blood
2010
;
116
:
2122
6
.
23.
Pardanani
A
,
Lasho
TL
,
Finke
CM
,
Mai
M
,
McClure
RF
,
Tefferi
A
. 
IDH1 and IDH2 mutation analysis in chronic- and blast-phase myeloproliferative neoplasms
.
Leukemia
2010
;
24
:
1146
51
.
24.
Cairns
RA
,
Iqbal
J
,
Lemonnier
F
,
Kucuk
C
,
de
LL
,
Jais
JP
, et al
IDH2 mutations are frequent in angioimmunoblastic T-cell lymphoma
.
Blood
2012
;
119
:
1901
3
.
25.
Shibata
T
,
Kokubu
A
,
Miyamoto
M
,
Sasajima
Y
,
Yamazaki
N
. 
Mutant IDH1 confers an in vivo growth in a melanoma cell line with BRAF mutation
.
Am J Pathol
2011
;
178
:
1395
402
.
26.
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
9
.
27.
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
2011
;
128
:
1095
103
.
28.
Ohgaki
H
,
Kleihues
P
. 
Genetic profile of astrocytic and oligodendroglial gliomas
.
Brain Tumor Pathol
2011
;
28
:
177
83
.
29.
Louis
DN
,
Ohgaki
H
,
Wiestler
OD
,
Cavenee
WK
,
editors
. 
WHO classification of tumours of the central nervous system
.
Lyon: IARC
; 
2007
.
p.
1
309
.
30.
Okamoto
Y
,
Di Patre
PL
,
Burkhard
C
,
Horstmann
S
,
Jourde
B
,
Fahey
M
, et al
Population-based study on incidence, survival rates, and genetic alterations of low-grade astrocytomas and oligodendrogliomas
.
Acta Neuropathol
2004
;
108
:
49
56
.
31.
Watanabe
T
,
Nakamura
M
,
Kros
JM
,
Burkhard
C
,
Yonekawa
Y
,
Kleihues
P
, et al
Phenotype versus genotype correlation in oligodendrogliomas and low-grade diffuse astrocytomas
.
Acta Neuropathol
2002
;
103
:
267
75
.
32.
Reifenberger
G
,
Louis
DN
. 
Oligodendroglioma: toward molecular definitions in diagnostic neuro-oncology
.
J Neuropathol Exp Neurol
2003
;
62
:
111
26
.
33.
Kim
YH
,
Nobusawa
S
,
Mittelbronn
M
,
Paulus
W
,
Brokinkel
B
,
Keyvani
K
, et al
Molecular classification of low-grade diffuse gliomas
.
Am J Pathol
2010
;
177
:
2708
14
.
34.
Bettegowda
C
,
Agrawal
N
,
Jiao
Y
,
Sausen
M
,
Wood
LD
,
Hruban
RH
, et al
Mutations in CIC and FUBP1 contribute to human oligodendroglioma
.
Science
2011
;
333
:
1453
5
.
35.
Yip
S
,
Butterfield
YS
,
Morozova
O
,
Chittaranjan
S
,
Blough
MD
,
An
J
, et al
Concurrent CIC mutations, IDH mutations, and 1p/19q loss distinguish oligodendrogliomas from other cancers
.
J Pathol
2012
;
226
:
7
16
.
36.
Jiao
Y
,
Killela
PJ
,
Reitman
ZJ
,
Rasheed
AB
,
Heaphy
CM
,
de Wilde
RF
, et al
Frequent ATRX, CIC, and FUBP1 mutations refine the classification of malignant gliomas
.
Oncotarget
2012
;
3
:
709
22
.
37.
Liu
XY
,
Gerges
N
,
Korshunov
A
,
Sabha
N
,
Khuong-Quang
DA
,
Fontebasso
AM
, et al
Frequent ATRX mutations and loss of expression in adult diffuse astrocytic tumors carrying IDH1/IDH2 and TP53 mutations
.
Acta Neuropathol
2012
;
124
:
615
25
.
38.
Kim
YH
,
Lachuer
J
,
Mittelbronn
M
,
Paulus
W
,
Brokinkel
B
,
Keyvani
K
, et al
Alterations in the RB1 pathway in low-grade diffuse gliomas lacking common genetic alterations
.
Brain Pathol
2011
;
21
:
645
51
.
39.
Kleihues
P
,
Schauble
B
,
zur Hausen
A
,
Esteve
J
,
Ohgaki
H
. 
Tumors associated with p53 germline mutations: a synopsis of 91 families
.
Am J Pathol
1997
;
150
:
1
13
.
40.
Olivier
M
,
Goldger
DE
,
Sodha
N
,
Ohgaki
H
,
Kleihues
P
,
Hainaut
P
, et al
Li-Fraumeni and related syndromes: correlation between tumor type, family structure and TP53 genotype
.
Cancer Res
2003
;
63
:
6643
50
.
41.
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
6
.
42.
Sasaki
M
,
Knobbe
CB
,
Munger
JC
,
Lind
EF
,
Brenner
D
,
Brustle
A
, et al
IDH1(R132H) mutation increases murine haematopoietic progenitors and alters epigenetics
.
Nature
2012
;
488
:
656
9
.
43.
Sasaki
M
,
Knobbe
CB
,
Itsumi
M
,
Elia
AJ
,
Harris
IS
,
Chio
II
, et al
D-2-hydroxyglutarate produced by mutant IDH1 perturbs collagen maturation and basement membrane function
.
Genes Dev
2012
;
26
:
2038
49
.
44.
Piaskowski
S
,
Bienkowski
M
,
Stoczynska-Fidelus
E
,
Stawski
R
,
Sieruta
M
,
Szybka
M
, et al
Glioma cells showing IDH1 mutation cannot be propagated in standard cell culture conditions
.
Br J Cancer
2011
;
104
:
968
70
.
45.
Jin
G
,
Pirozzi
CJ
,
Chen
LH
,
Lopez
GY
,
Duncan
CG
,
Feng
J
, et al
Mutant IDH1 is required for IDH1 mutated tumor cell growth
.
Oncotarget
2012
;
3
:
774
82
.
46.
Luchman
HA
,
Stechishin
OD
,
Dang
NH
,
Blough
MD
,
Chesnelong
C
,
Kelly
JJ
, et al
An in vivo patient-derived model of endogenous IDH1-mutant glioma
.
Neuro Oncol
2012
;
14
:
184
91
.
47.
Narahara
K
,
Kimura
S
,
Kikkawa
K
,
Takahashi
Y
,
Wakita
Y
,
Kasai
R
, et al
Probable assignment of soluble isocitrate dehydrogenase (IDH1) to 2q33.3
.
Hum Genet
1985
;
71
:
37
40
.
48.
Geisbrecht
BV
,
Gould
SJ
. 
The human PICD gene encodes a cytoplasmic and peroxisomal NADP(+)-dependent isocitrate dehydrogenase
.
J Biol Chem
1999
;
274
:
30527
33
.
49.
Devlin
TM
. 
Textbook of biochemistry with clinical correlations
.
Hoboken, NJ
:
Wiley-Liss, John Wiley & Sons
; 
2006
.
50.
Zhao
S
,
Lin
Y
,
Xu
W
,
Jiang
W
,
Zha
Z
,
Wang
P
, et al
Glioma-derived mutations in IDH1 dominantly inhibit IDH1 catalytic activity and induce HIF-1alpha
.
Science
2009
;
324
:
261
5
.
51.
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
44
.
52.
Ward
PS
,
Patel
J
,
Wise
DR
,
bdel-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
34
.
53.
Jin
G
,
Reitman
ZJ
,
Spasojevic
I
,
Batinic-Haberle
I
,
Yang
J
,
Schmidt-Kittler
O
, et al
2-Hydroxyglutarate production, but not dominant negative function, is conferred by glioma-derived NADP-dependent isocitrate dehydrogenase mutations
.
PLoS ONE
2011
;
6
:
e16812
.
54.
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
22
.
55.
Christensen
BC
,
Smith
AA
,
Zheng
S
,
Koestler
DC
,
Houseman
EA
,
Marsit
CJ
, et al
DNA methylation, isocitrate dehydrogenase mutation, and survival in glioma
.
J Natl Cancer Inst
2011
;
103
:
143
53
.
56.
Figueroa
ME
,
bdel-Wahab
O
,
Lu
C
,
Ward
PS
,
Patel
J
,
Shih
A
, et al
Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation
.
Cancer Cell
2010
;
18
:
553
67
.
57.
Turcan
S
,
Rohle
D
,
Goenka
A
,
Walsh
LA
,
Fang
F
,
Yilmaz
E
, et al
IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype
.
Nature
2012
;
483
:
479
83
.
58.
Lu
C
,
Ward
PS
,
Kapoor
GS
,
Rohle
D
,
Turcan
S
,
bdel-Wahab
O
, et al
IDH mutation impairs histone demethylation and results in a block to cell differentiation
.
Nature
2012
;
483
:
474
8
.
59.
Duncan
CG
,
Barwick
BG
,
Jin
G
,
Rago
C
,
Kapoor-Vazirani
P
,
Powell
DR
, et al
A heterozygous IDH1R132H/WT mutation induces genome-wide alterations in DNA methylation
.
Genome Res
2012
;
22
:
2339
55
.
60.
Phillips
HS
,
Kharbanda
S
,
Chen
R
,
Forrest
WF
,
Soriano
RH
,
Wu
TD
, et al
Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis
.
Cancer Cell
2006
;
9
:
157
73
.
61.
Verhaak
RG
,
Hoadley
KA
,
Purdom
E
,
Wang
V
,
Qi
Y
,
Wilkerson
MD
, et al
Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1
.
Cancer Cell
2010
;
17
:
98
110
.
62.
Cooper
LA
,
Gutman
DA
,
Long
Q
,
Johnson
BA
,
Cholleti
SR
,
Kurc
T
, et al
The proneural molecular signature is enriched in oligodendrogliomas and predicts improved survival among diffuse gliomas
.
PLoS ONE
2010
;
5
:
e12548
.
63.
Ohgaki
H
,
Kleihues
P
. 
Population-based studies on incidence, survival rates, and genetic alterations in astrocytic and oligodendroglial gliomas
.
J Neuropathol Exp Neurol
2005
;
64
:
479
89
.
64.
Central Brain Tumor Registry of the United States
. 
Central brain tumor registry of the United States (CBTRUS)
; 
2002
.
Available from
: http://www.cbtrus.org.
65.
Wohrer
A
,
Waldhor
T
,
Heinzl
H
,
Hackl
M
,
Feichtinger
J
,
Gruber-Mosenbacher
U
, et al
The Austrian brain tumour registry: a cooperative way to establish a population-based brain tumour registry
.
J Neurooncol
2009
;
95
:
401
11
.
66.
Dropcho
EJ
,
Soong
SJ
. 
The prognostic impact of prior low grade histology in patients with anaplastic gliomas: a case–control study
.
Neurology
1996
;
47
:
684
90
.
67.
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
7
.
68.
Lai
A
,
Kharbanda
S
,
Pope
WB
,
Tran
A
,
Solis
OE
,
Peale
F
, et al
Evidence for sequenced molecular evolution of IDH1 mutant glioblastoma from a distinct cell of origin
.
J Clin Oncol
2011
;
29
:
4482
90
.
69.
Ohgaki
H
,
Kleihues
P
. 
Epidemiology and etiology of gliomas
.
Acta Neuropathol
2005
;
109
:
93
108
.
70.
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
94
.
71.
Godard
S
,
Getz
G
,
Delorenzi
M
,
Farmer
P
,
Kobayashi
H
,
Desbaillets
I
, et al
Classification of human astrocytic gliomas on the basis of gene expression: a correlated group of genes with angiogenic activity emerges as a strong predictor of subtypes
.
Cancer Res
2003
;
63
:
6613
25
.
72.
Xie
D
,
Zeng
YX
,
Wang
HJ
,
Wen
JM
,
Tao
Y
,
Sham
JS
, et al
Expression of cytoplasmic and nuclear Survivin in primary and secondary human glioblastoma
.
Br J Cancer
2006
;
94
:
108
14
.
73.
Choe
G
,
Park
JK
,
Jouben-Steele
L
,
Kremen
TJ
,
Liau
LM
,
Vinters
HV
, et al
Active matrix metalloproteinase 9 expression is associated with primary glioblastoma subtype
.
Clin Cancer Res
2002
;
8
:
2894
901
.
74.
Karcher
S
,
Steiner
HH
,
Ahmadi
R
,
Zoubaa
S
,
Vasvari
G
,
Bauer
H
, et al
Different angiogenic phenotypes in primary and secondary glioblastomas
.
Int J Cancer
2006
;
118
:
2182
9
.
75.
Stockhammer
F
,
Misch
M
,
Helms
HJ
,
Lengler
U
,
Prall
F
,
von
DA
, et al
IDH1/2 mutations in WHO grade II astrocytomas associated with localization and seizure as the initial symptom
.
Seizure
2012
;
21
:
194
7
.
76.
Zlatescu
MC
,
TehraniYazdi
A
,
Sasaki
H
,
Megyesi
JF
,
Betensky
RA
,
Louis
DN
, et al
Tumor location and growth pattern correlate with genetic signature in oligodendroglial neoplasms
.
Cancer Res
2001
;
61
:
6713
5
.
77.
Laigle-Donadey
F
,
Martin-Duverneuil
N
,
Lejeune
J
,
Criniere
E
,
Capelle
L
,
Duffau
H
, et al
Correlations between molecular profile and radiologic pattern in oligodendroglial tumors
.
Neurology
2004
;
63
:
2360
2
.
78.
Rong
Y
,
Durden
DL
,
Van Meir
EG
,
Brat
DJ
. 
‘Pseudopalisading’ necrosis in glioblastoma: a familiar morphologic feature that links vascular pathology, hypoxia, and angiogenesis
.
J Neuropathol Exp Neurol
2006
;
65
:
529
39
.
79.
Svensson
KJ
,
Kucharzewska
P
,
Christianson
HC
,
Skold
S
,
Lofstedt
T
,
Johansson
MC
, et al
Hypoxia triggers a proangiogenic pathway involving cancer cell microvesicles and PAR-2-mediated heparin-binding EGF signaling in endothelial cells
.
Proc Natl Acad Sci U S A
2011
;
108
:
13147
52
.
80.
Homma
T
,
Fukushima
T
,
Vaccarella
S
,
Yonekawa
Y
,
Di Patre
PL
,
Franceschi
S
, et al
Correlation among pathology, genotype, and patient outcomes in glioblastoma
.
J Neuropathol Exp Neurol
2006
;
65
:
846
54
.
81.
Szerlip
NJ
,
Pedraza
A
,
Chakravarty
D
,
Azim
M
,
McGuire
J
,
Fang
Y
, et al
Intratumoral heterogeneity of receptor tyrosine kinases EGFR and PDGFRA amplification in glioblastoma defines subpopulations with distinct growth factor response
.
Proc Natl Acad Sci U S A
2012
;
109
:
3041
6
.
82.
Little
SE
,
Popov
S
,
Jury
A
,
Bax
DA
,
Doey
L
,
Al-Sarraj
S
, et al
Receptor tyrosine kinase genes amplified in glioblastoma exhibit a mutual exclusivity in variable proportions reflective of individual tumor heterogeneity
.
Cancer Res
2012
;
72
:
1614
20
.
83.
Schulte
A
,
Gunther
HS
,
Martens
T
,
Zapf
S
,
Riethdorf
S
,
Wulfing
C
, et al
Glioblastoma stem-like cell lines with either maintenance or loss of high-level EGFR amplification, generated via modulation of ligand concentration
.
Clin Cancer Res
2012
;
18
:
1901
13
.
84.
Fujisawa
H
,
Kurrer
M
,
Reis
RM
,
Yonekawa
Y
,
Kleihues
P
,
Ohgaki
H
. 
Acquisition of the glioblastoma phenotype during astrocytoma progression is associated with LOH on chromosome 10q25-qter
.
Am J Pathol
1999
;
155
:
387
94
.
85.
Perry
A
,
Miller
CR
,
Gujrati
M
,
Scheithauer
BW
,
Zambrano
SC
,
Jost
SC
, et al
Malignant gliomas with primitive neuroectodermal tumor-like components: a clinicopathologic and genetic study of 53 cases
.
Brain Pathol
2009
;
19
:
81
90
.
86.
Beier
D
,
Hau
P
,
Proescholdt
M
,
Lohmeier
A
,
Wischhusen
J
,
Oefner
PJ
, et al
CD133(+) and CD133(−) glioblastoma-derived cancer stem cells show differential growth characteristics and molecular profiles
.
Cancer Res
2007
;
67
:
4010
5
.
87.
Ichimura
K
,
Schmidt
EE
,
Miyakawa
A
,
Goike
HM
,
Collins
VP
. 
Distinct patterns of deletion on 10p and 10q suggest involvement of multiple tumor suppressor genes in the development of astrocytic gliomas of different malignancy grades
.
Genes Chromosomes Cancer
1998
;
22
:
9
15
.
88.
Karlbom
AE
,
James
CD
,
Boethius
J
,
Cavenee
WK
,
Collins
VP
,
Nordenskjöld
M
, et al
Loss of heterozygosity in malignant gliomas involves at least three distinct regions on chromosome 10
.
Hum Genet
1993
;
92
:
169
74
.
89.
Rasheed
BK
,
McLendon
RE
,
Friedman
HS
,
Friedman
AH
,
Fuchs
HE
,
Bigner
DD
, et al
Chromosome 10 deletion mapping in human gliomas: a common deletion region in 10q25
.
Oncogene
1995
;
10
:
2243
6
.
90.
Tohma
Y
,
Gratas
C
,
Biernat
W
,
Peraud
A
,
Fukuda
M
,
Yonekawa
Y
, et al
PTEN (MMAC1) mutations are frequent in primary glioblastomas (de novo) but not in secondary glioblastomas
.
J Neuropathol Exp Neurol
1998
;
57
:
684
9
.
91.
Cancer Genome Atlas Research Network
. 
Comprehensive genomic characterization defines human glioblastoma genes and core pathways
.
Nature
2008
;
455
:
1061
8
.
92.
Motomura
K
,
Mittelbronn
M
,
Paulus
W
,
Brokinkel
B
,
Keyvani
K
,
Sure
U
, et al
DMBT1 homozygous deletion in diffuse astrocytomas is associated with unfavorable clinical outcome
.
J Neuropathol Exp Neurol
2012
;
71
:
702
7
.
93.
Pierscianek
D
,
Kim
YH
,
Motomura
K
,
Mittelbronn
M
,
Paulus
W
,
Brokinkel
B
, et al
MET gain in diffuse astrocytomas is associated with poorer outcome
.
Brain Pathol
2013
;
23
:
13
8
.