Translating Molecular Biomarkers of Gliomas to Clinical Practice


Acknowledgments

The authors are grateful to their NYU Langone Medical Center colleagues Matija Snuderl, MD, for contributing illustrated cases of cytogenetics analysis and methylation array; Cyrus Hedvat, MD, PhD, for providing an illustrated case of loss of heterozygosity analysis; and Elad Mashiach for assisting with the preparation of images and diagrams, and the editing of the chapter.

Glioma classification and grading have traditionally been based on the histomorphology of the tumors. Recent advances have identified new molecular markers with diagnostic, prognostic, and/or predictive (ie, therapeutic) significance ( Box 4.1 , Table 4.1 ). Since the publication of the World Health Organization (WHO) guidelines in 2007, there has been a rapid expansion of molecular data on central nervous system (CNS) tumors that has improved clinicians’ diagnostic, prognostic, and therapeutic abilities. Although most of this information has not yet been translated into tangible clinical advances, many changes have been implemented in the revised WHO guidelines for the classification of tumors of the CNS (2016) for gliomas. This chapter reviews recently identified genetic markers that have had a significant impact on the molecular classification of gliomas. Many have been shown to be essential in better diagnosing CNS tumors, reliably determining the prognosis, and allowing better clinical management. Based on the revised WHO classification, each tumor type is discussed separately, accompanied by the relevant molecular profiles.

Box 4.1
Definition and types of biomarkers

The National Institutes of Health Biomarkers Definitions Working Group defined a biomarker as “a characteristic that is objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention.” Biomarkers in gliomas have been investigated particularly for their use in identifying patients with a disease or a disease subtype (diagnostic biomarkers), stratifying the patients’ prognoses and natural history of the disease (prognostic biomarkers), and identifying patients who may achieve a particular outcome based on a particular treatment and attempting to personalize clinical treatment (predictive biomarkers). Several of the biomarkers discussed in this chapter have distinct roles as diagnostic, prognostic and/or predictive biomarkers, and these are discussed in the text and summarized in Table 4.1 and Figs. 4.1 and 4.5 .

Table 4.1
Overview of common chromosomal, genetic, epigenetic and phenotypic alterations in gliomas and their use as biomarkers
Gene/Phenotype a Gene Family/Alternative Name Chromosomal Location Driver Gene b Typical Mutation c Copy Number Alteration Translocation Partner Detection Method Adult Glioma Tumor Type Pediatric Glioma Tumor Type Biomarker Clinical Utility References
Chromosomal
1p/19q Codeletion FISH, LOH, MLPA, 450K-MA OD, AOD Diag, Prog, Pred (chemo + radiotherapy)
CIC Transcription repressor 19q13.2 TSG R215Q/W Del IHC (loss of staining), others OD, AOD Diag, Prog
FUBP1 DNA-binding protein 1p31.1 TSG Many Del IHC (loss of staining), others OD, AOD Diag, Prog
7 or 7q Single copy gain FISH, others DA, AA, GBM
10 or 10q Single copy loss FISH, others GBM
Genetic
ACVR1 RSTK 2q23-q24 TSG Few qRT-PCR, Seq Midline HGG, DIPG
BRAF RAF kinase 7q34 ONC V600E MS-IHC, qRT-PCR, Seq PXA, EGBM PA, PXA, cortical HGG Diag
BRAF Amp KIAA1549, others FISH, others; qRT-PCR, Seq PA PA, PMA Diag
CDKN2A Kinase inhibitor/p14, p16 9p21 TSG Del FISH, MLPA, 450K-MA OD, AOD, DA, AA, GBM PXA
CDKN2B Kinase inhibitor 9p21 Del FISH, MLPA, 450K-MA OD, AOD, DA, AA, GBM PXA
EGFR RTK 7p12 ONC Amp SEPT14 FISH, others; qRT-PCR, Seq Classic GBM Cortical HGG Diag
EGFR EGFRvIII, A289D/T/V MS-IHC, qRT-PCR, Seq Classic GBM Diag
FGFR1 RTK/CD331 8p11.23-p11.22 K656E TACC1 qRT-PCR, Seq; FISH, others PA, midline HGG/DIPG
FGFR3 RTK/CD333 4p16.3 ONC TACC3 FISH, qRT-PCR, Seq GBM
IDH1 Dehydrogenase 2q34 ONC R132H, others MS-IHC, qRT-PCR, Seq OD, AOD, DA, AA, GBM Cortical HGG Diag, Prog
IDH2 Dehydrogenase 15q26.1 ONC R172K, others qRT-PCR, Seq OD, AOD, DA, AA, GBM Diag, Prog
MDM2 Ubiquitin protein ligase 12q13-q14 ONC Few Amp FISH, MLPA, 450K-MA GBM
MDM4 p53 regulator 1q32 ONC Few Amp FISH, MLPA, 450K-MA GBM
MET RTK 7q31 ONC Amp FISH, MLPA, 450K-MA GBM
MYC Transcription factor 8q24 ONC Amp FISH, MLPA, 450K-MA Astrocytoma, GBM
NF1 RAS negative regulator 17q11.2 TSG Many Del qRT-PCR, Seq; FISH, others Mesenchymal GBM PA, midline HGG
NOTCH1 receptor 9q34.3 TSG F357del Amp qRT-PCR, Seq; FISH, others OD
NTRK2 RTK 9q22.1 Amp QKI FISH, others; qRT-PCR, Seq PA, non-brainstem HGG
PDGFRA RTK/CD140a 4q12 ONC Many Amp KDR qRT-PCR, Seq; FISH, others Proneural GBM Midline HGG, DIPG Prog
PIK3CA PI3 kinase 3q26.3 ONC H1047L/R/Y Amp qRT-PCR, Seq; FISH, others OD, AOD, GBM Midline HGG, DIPG
PIK3R1 Regulatory subunit of PI3 kinase 5q13.1 TSG G376R Del qRT-PCR, Seq; FISH, others OD, AOD, GBM Midline HGG, DIPG
PTEN Phosphatase 10q23 TSG R130 d /Q Del qRT-PCR, Seq; FISH, others Astrocytoma, classical GBM Prog
PTPN11 Phosphatase 12q24.1 ONC Many qRT-PCR, Seq PA
RB1 Ligand 13q14.2 TSG R445 d , X445_splice Del qRT-PCR, Seq; FISH, others Mesenchymal GBM
TERT Telomerase 5p15.33 Promoter qRT-PCR, Seq OD, AOD, astrocytoma, GBM Diag, Prog
TP53 Transcription factor 17p13.1 TSG R273C/H/L, R248Q/W (IHC), qRT-PCR, Seq Astrocytoma, GBM Midline/cortical HGG
Epigenetic
ATRX Chromatin remodeler Xq21.1 TSG F2113fs IHC (loss of staining), others DA, AA, GBM Cortical HGG Diag, Prog
DAXX Chromatin remodeler 6p21.3 TSG Amp FISH, MLPA, 450K-MA Cortical HGG
HIST1H3B Histone 6p22.2 ONC H3.1 K27M qRT-PCR, Seq Midline HGG, DIPG Diag, Prog
H3F3A Histone 1q42.12 ONC H3.3 K27M MS-IHC, qRT-PCR, Seq Midline HGG, DIPG Midline HGG, DIPG Diag, Prog
H3F3A Histone 1q42.12 ONC H3.3 G34R/V MS-IHC, qRT-PCR, Seq Cortical HGG Diag, Prog
MGMT DNA cysteine MT 10q26 Promoter methylation MS-PCR, 450K-MA GBM Prog, Pred (temozolomide)
SETD2 Histone lysine MT 3p21.31 TSG Many Del qRT-PCR, Seq; FISH, others Cortical HGG
TET2 Demethylase 4q24 TSG Few qRT-PCR, Seq GBM
Phenotypic
2-HG 2-hydroxyglutarate MRS, mass spectrometry OD, AOD, DA, AA, GBM
G-CIMP Glioma–CpG island methylator phenotype 450K-MA OD, AOD, DA, AA, GBM
Abbreviations: AA, anaplastic astrocytoma; AOD, anaplastic oligodendroglioma; CD, cluster of differentiation; DA, diffuse astrocytoma; Diag, diagnostic biomarker; DIPG, diffuse intrinsic pontine glioma; EGBM, epithelioid glioblastoma; EGFR; epidermal growth factor receptor; EGFRvIII, deleted exons 2 to 7 EGFR; FISH, fluorescence in situ hybridization; fs, frame shift; GBM, glioblastoma; HGG, high-grade (III–IV) glioma; IHC, immunohistochemistry; LGG, low-grade (I-II) glioma; LOH, loss of heterozygosity; MLPA, multiplex ligation-dependent probe amplification; MRS, magnetic resonance spectroscopy; MS-IHC, mutation-specific immunohistochemistry; MS-PCR, methylation-specific polymerase chain reaction; MT, methyltransferase; OD, oligodendroglioma; ONC, oncogene; PA, pilocytic astrocytoma; PMA, pilomyxoid astrocytoma; Pred, predictive biomarker; Prog, prognostic biomarker; PXA, pleomorphic xanthoastrocytoma; qRT-PCR, quantitative reverse transcriptase polymerase chain reaction; RSTK, receptor serine/threonine kinase; RTK, receptor tyrosine kinase; Seq, targeted nucleotide sequencing; TSG, tumor suppressor gene; 450K-MA, 450K CpG methylation array.

a Gene symbols, gene families, and chromosomal locations according to the Human Genome Organisation Gene Nomenclature Committee ( www.genenames.org ) and Catalogue of Somatic Mutations in Cancer ( cancer.sanger.ac.uk ).

b Driver genes that contain driver gene mutations as defined by Vogelstein and colleagues.

c Gene mutations and copy number alterations based on the Merged Cohort of LGG and GBM (The Cancer Genome Atlas [TCGA], 2016) database (1102 samples) generated by TCGA Research Network ( http://www.cbioportal.org/index.do ). In addition, the translocation partners for BRAF, EGFR, FGFR1, NTRK2, and PDGFRA are listed.

d Change to a termination codon (nonsense mutation).

Adult diffuse gliomas

Adult diffuse gliomas are infiltrating glial neoplasms that include astrocytomas and oligodendrogliomas. In the 2007 edition of the WHO Classification of Tumours of the Central Nervous System , these entities were diagnosed and classified as grade II (diffuse) or grade III (anaplastic) based on histologic features. In cases in which a morphologic distinction between these two entities was not clear, a diagnosis of oligoastrocytoma was appropriate. Following major advances in our understanding of molecular gliomagenesis, the revised WHO Classification of Tumours of the Central Nervous System (2016) has refined the diagnostic criteria for astrocytomas and oligodendrogliomas by incorporating clinically relevant molecular information about the mutation status of isocitrate dehydrogenase 1/2 ( IDH1/2 ), and alpha thalassemia/mental retardation syndrome X-linked ( ATRX ) genes and codeletion of chromosome arms 1p and 19q. After an initial IDH mutation, oligodendrogliomas are thought to develop via subsequent telomerase reverse transcriptase ( TERT ) promoter mutations and codeletion of 1p/19q, whereas IDH -mutant astrocytomas develop with subsequent alterations of TP53 and/or ATRX. The diagnosis of oligoastrocytoma is now strongly discouraged ( Fig. 4.1 ).

Fig. 4.1, Molecular classification model of adult diffuse gliomas based on the combined findings 102 103 of characteristic genomic alterations in astrocytic and oligodendroglial tumors. 3 24 104 105 The analysis of common mutations in IDH1/2 , ATRX and the TERT promoter and codeletion of 1p/19q allows the classification of these tumors into 5 molecular subgroups that define the biological and clinical behavior of gliomas more accurately than the classification based solely on the histopathologic tumor types. Additional gene mutations and copy number alterations are associated with these subgroups.

Point mutations in cytosolic IDH1 and mitochondrial IDH2 most commonly by substitution of arginine to histidine (R132H) or to lysine (R172K), respectively, alter their catalytic activity such that they produce high levels of the oncometabolite 2-hydroxyglutarate (2-HG), instead of α-ketoglutarate. The presence of 2-HG results in disruption of tet methylcytosine dioxygenase 2 (TET2) activity, leading to aberrant histone regulation and development of the glioma–CpG island methylator phenotype (G-CIMP).

G-CIMP is an epigenetic molecular profile that was noted and named after the observation of a subset of gliomas within The Cancer Genome Atlas (TCGA) database that showed concerted hypermethylation at a large number of loci. In general, CIMP gliomas are lower-grade, often IDH -mutated, tumors. Mutation of IDH is the molecular basis for the G-CIMP phenotype. Overall, IDH -mutant, G-CIMP high infiltrating gliomas are associated with a favorable prognosis compared with IDH wild-type tumors. IDH status is an even stronger predictor of patient outcome than histologic grade in infiltrating gliomas.

IDH mutations can be detected by immunohistochemical analysis of formalin-fixed, paraffin-embedded (FFPE) tissue using the IDH1 R132H mutant-specific antibody ( Fig. 4.2 A, B ). Direct Sanger sequencing, although requiring more tissue specimens, has the advantage over immunohistochemistry (IHC) of not only detecting IDH1 R132H but also detecting other noncanonical IDH mutations. This technique is highly sensitive, but is limited because the specimens must contain at least 50% neoplastic cells to ensure reliability. Another method, pyrosequencing, has a higher sensitivity than Sanger sequencing because it can detect as little as 10% mutant alleles. Moreover, clinical efforts have been undertaken to determine whether IDH mutations can be detected indirectly, and magnetic resonance spectroscopy has been proposed as a reliable technique to achieve this goal by detecting the levels of 2-HG.

Fig. 4.2, Immunohistochemical assessment of IDH1, ATRX, and TP53 expression in gliomas. ( A , B ) Oligodendroglioma, IDH1 mutated, ATRX preserved (WHO grade II). Tumor cells show diffuse cytoplasmic immunoreactivity to IDH R132H. ( C , D ) Anaplastic astrocytoma, ATRX mutated, IDH1 mutated, 1p/19q intact (WHO grade III). The nuclear expression of ATRX is not detected in tumor cells, but is retained in endothelial cells. A biphenotypic pattern of microgemistocytes and small round cells with scanty cytoplasm is seen on hematoxylin and eosin (H&E) staining. ( E , F ) Giant cell glioblastoma, TP53 mutated, IDH1 mutated (WHO grade IV). Tumor cells show strong, diffuse immunoreactivity to TP53. Note the presence of bizarre-looking giant cells, including some multinucleated cells, and vascular proliferation on H&E staining (original magnification ×20 for all panels).

Diffuse Astrocytomas

Diffuse astrocytomas are now defined based on their IDH status as (1) diffuse astrocytoma, IDH mutant (most common); (2) diffuse astrocytoma, IDH wild-type or (3) diffuse astrocytoma, not otherwise specified (NOS; given when IDH testing is unavailable or inconclusive). A characteristic feature of IDH -mutated astrocytomas is the presence of a mutation in ATRX and frequent mutation of TP53 (see Fig. 4.1 ; Fig. 4.2 ).

ATRX is a nuclear chromatin remodeling protein that is encoded by the ATRX gene on chromosome Xq21.1. Loss-of-function mutations in ATRX are associated with alterations in replication and activation of the alternative lengthening of telomeres (ALT) pathway. In diffuse gliomas, ATRX mutation is a useful marker for astrocytic differentiation, and is frequently seen in combination with mutations of IDH and TP53 . In contrast, IDH -mutant, 1p/19q codeleted oligodendrogliomas only rarely harbor concurrent mutations in ATRX to the point that they are considered to be mutually exclusive. For these reasons, IHC or sequencing for ATRX has a role both in confirming a diagnosis of astrocytoma and in ruling out a diagnosis of oligodendroglioma ( Fig. 4.2 C, D). IHC is sensitive and able to detect 82% to 89% of mutants that are detectable by sequencing. Importantly, although they often exist in tandem, the presence of an ATRX mutation does not confirm the presence of a concurrent IDH mutation. The prognostic implications of ATRX mutation are limited, and are mostly related to increased accuracy in the diagnosis of diffuse gliomas as astrocytic versus oligodendroglial. In non–1p/19q-codeleted gliomas, ATRX mutation has been associated with better treatment outcomes.

Oligodendrogliomas

Changes to the diagnostic criteria of oligodendroglioma and anaplastic oligodendroglioma include the requirement of both an IDH mutation and a 1p/19q codeletion (see Fig. 4.1 ). There are 4 distinct types of oligodendroglioma tumors: (1) IDH mutant and 1p/19q codeleted, (2) NOS, (3) anaplastic IDH mutant and 1p/19q codeleted, and (4) anaplastic NOS (when only classic histology is available). The diagnosis of oligodendroglioma NOS should only be given after further analysis excludes potential diagnoses such as dysembryoplastic neuroepithelial tumor, clear cell ependymoma, neurocytoma, pilocytic astrocytoma, and several other tumor types that are histologically similar to oligodendroglioma.

Loss of 1p/19q has more than just a diagnostic significance. It has been associated with a favorable prognosis and an increased sensitivity to chemotherapy. Moreover, 1p/19q-codeleted anaplastic oligodendrogliomas that show polysomy for chromosomes 1p and 19q have intermediate survival between 1p/19q-retained tumors and 1p/19q-codeleted oligodendrogliomas in a euploid background. Codeletion of 1p/19q can be detected by fluorescence in situ hybridization (FISH), loss of heterozygosity (LOH) capillary gel electrophoresis, or as a change in copy number in the 450K CpG methylation array (450K-MA) results ( Figs. 4.3 and 4.4 A ). However, only FISH allows the enumeration of absolute numbers of chromosomes and determination of ploidy status.

Fig. 4.3, FISH and LOH analyses of loss of chromosome arms 1p/19q in oligodendrogliomas. ( A – C ) FISH using probes for 1p or 19q ( red signal ) and control probes for 1q or 19p ( green ) with 4′,6-diamidino-2-phenylindole (DAPI) nuclear counterstain ( blue ) shows maintenance of 1p with 2 red and 2 green signals ( A ) and classic absolute loss of 1 copy of 1p, whereas 2 1q signals remain intact ( B ). In anaplastic oligodendroglioma, concurrent polysomy indicated by multiple 1q signals with ≈50% deletion of 1p/19q ( C ) is a marker of early recurrence. 16 Similar results were obtained for chromosome 19 (not shown). ( D – G ) Alternatively, LOH can be evaluated by comparing normal (blood) ( D , F ) and tumor DNA samples ( E , G ) for the presence of allelic loss of 1p and/or 19q using fluorescence-labeled polymorphic chromosomal markers for 1p (7 loci) ( D , E ) and 19q (4 loci) ( F , G ) followed by capillary gel electrophoresis. The blood sample shows 2 alleles for each heterozygous marker, but the decrease in peak height (bottom numbers in the boxes below the electropherograms) of 1 of the 2 alleles indicates that the tumor has undergone LOH for both 1p and 19q.

Fig. 4.4, Genomic copy number alterations in gliomas determined by methylation array. In addition to genome-wide DNA methylation patterns, the copy number dosages of investigated markers along chromosomes 1 to 22 are determined. Copy numbers greater than average are indicated in green, and copy numbers less than average in red. Typical copy number profiles of ( A ) oligodendroglioma with 1p/19q codeletion, ( B ) pleomorphic xanthoastrocytoma with deletion of CDKN2A/B on chromosome arm 9p21, and ( C ) glioblastoma with gain of chromosome 7 ( EGFR ), and loss of chromosome 9 ( CDKN2A/B ) and 10 ( PTEN ) are shown.

Several other recurrent molecular alterations can be found in oligodendrogliomas. CIC (Capicua transcriptional repressor), a gene that resides in chromosome 19q, plays a pivotal role in regulating RAS (rat sarcoma oncogene)/MAPK (mitogen-activated protein kinase) signaling. Somatic mutations in CIC are present in as many as 69% of oligodendrogliomas. FUBP1 (far-upstream element binding protein) is a gene located on chromosome 1p. It has been suggested that inactivating somatic mutations in this gene (along with CIC ) are also associated with the development of oligodendroglioma. Tumors with such alterations seem to cluster with 1p/19q-codeleted oligodendrogliomas. Recent work has identified that TERT is overexpressed in oligodendrogliomas (see Fig. 4.1 ). Oligodendrogliomas are therefore now recognized to contain at least 4 different recurrent molecular genetic correlates: IDH mutation, 1p/19q loss, CIC mutation, and TERT promoter mutation. Mutations in the NOTCH1 gene (discussed later) were also observed in 31% of oligodendrogliomas.

Adult glioblastomas

Glioblastomas are high-grade, infiltrative astrocytomas with atypical nuclei, mitotic activity, extensive vascular proliferation, and/or necrosis. According to the revised WHO classification of CNS tumors, glioblastomas can be molecularly classified as IDH wild-type, IDH -mutant glioblastomas or NOS (see Fig. 4.1 ). Histologic IDH wild-type variants include giant cell ( Fig. 4.2 E, F), gliosarcoma, and the newly described epithelioid glioblastoma. About 90% of glioblastomas (usually primary) present as IDH wild-type; glioblastomas are found to be IDH mutated in about 10% of cases, and these are most often secondary glioblastomas that have progressed from lower-grade IDH -mutant gliomas.

Recent efforts to molecularly subclassify glioblastomas based on genetic and epigenetic expression profiling have consistently identified the classical, mesenchymal and proneural subtypes characterized by epidermal growth factor receptor ( EGFR ) amplification, neurofibromin 1 ( NF1 ) loss, and platelet-derived growth factor receptor A ( PDGFRA ) amplification, respectively. Notably, the proneural subtype was also associated with IDH1/2 mutations, G-CIMP phenotype as well as TP53 mutations. Additional mutations in histone H3F3A-K27 and H3F3A-G34 have defined epigenetic subgroups of glioblastoma in young adult (and pediatric) patients, which are addressed later.

IDH -mutated glioblastomas typically arise in younger patients following secondary transformation of a lower-grade astrocytoma. As such, the molecular features in IDH -mutant glioblastomas are often similar to those seen in IDH -mutant diffuse or anaplastic astrocytomas. IDH -mutant glioblastomas often harbor concurrent ATRX and TP53 mutations. Despite their high-grade histology, IDH -mutant glioblastomas show clinical behavior similar to that seen in lower-grade IDH -mutant astrocytomas. In contrast, IDH wild-type glioblastomas most commonly present as de novo tumors arising in patients older than 55 years. These tumors not only show vast histomorphologic heterogeneity but also harbor a wide variety of genetic alterations (see Fig. 4.1 ; Fig. 4.4 C). Data from TCGA show frequent alterations in 3 core signaling pathways: the CDK4/6-p16 INK4a -RB1-E2F, p14 ARF -MDM2-MDM4-p53, and RTK-RAS-PI3K pathways. These pathways and their common associated alterations are discussed below.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here