Keywords
diffuse midline glioma - H3.1 or H3.2 K27-mutant - H3-wildtype with EZHIP overexpression
-
EGFR-mutant - H3.3 K27-mutant
Introduction
H3 K27M-altered diffuse midline gliomas (DMG) are a rare primary central nervous system
(CNS) glioma subtype. This tumor was previously known, in children, as diffuse intrinsic
pontine glioma (DIPG).[1] However, genetic analysis showed that 80% of DIPG harbored an H3 K27 mutation associated
with poor prognosis. Analysis of a wider cohort of tumors for this mutation expanded
the classification in the World Health Organization (WHO) 2016 Update of Classification
of Tumours of the CNS.[2] In 2021, the WHO created a specific group of pediatric-type diffuse high-grade gliomas.
The group consists of four tumors (DMG, H3 K27-altered; diffuse hemispheric glioma,
H3 G34-mutant; diffuse pediatric-type high-grade glioma, H3-wildtype and IDH [isocitrate
dehydrogenase]-wildtype; infant-type hemispheric glioma) that differ molecularly and
prognostically (
[Table 1]). DMG H3 K27-altered may also occur in the adult population.[3]
Table 1
Subtype definitions for pediatric-type high-grade glioma
Pediatric-type high-grade glioma subtypes
|
DMG H3K27-altered
An infiltrative midline glioma with loss of H3 p.K28me3 (K27me3) and either an H3
c.83A > T p.K28M (K27M) substitution in one of the histone H3 isoforms, aberrant overexpression
of EZHIP, or an EGFR mutation (CNS WHO grade 4)
|
Diffuse hemispheric glioma H3G34-mutant
Malignant IDH-wildtype glioma with a G34R/V mutation in H3F3A, and located in the
cerebral hemispheres
|
Diffuse pediatric-type high-grade glioma, H3-wildtype and IDH wildtype
High-grade glioma predominantly located in the hemispheres, both IDH and H3-wildtype
|
Infant type hemispheric glioma
High-grade glioma predominantly found within the cerebral hemispheres of infants,
with mutations in ROS, ALK, MET, and NTRK receptor tyrosine kinases
|
Abbreviations: ALK, anaplastic lymphoma kinase; CNS, central nervous system; DMG,
diffuse midline glioma; EGFR, epidermal growth factor receptor gene; EZHIP, enhancer
of zest homolog inhibitory protein; ROS, proto-oncogene tyrosine protein kinase; MET,
mesenchymal epithelial transition; NTPK, nonreceptor proto-oncogene tyrosine protein
kinase; WHO, World Health Organization.
We review the features of DMG H3 K27-altered in the adult and pediatric populations
and compare their genetic, epidemiological, clinical, radiological, histopathological,
treatment, and prognostic characteristics.
Methodology
We conducted a PubMed search up to July 31, 2022 using the terms “diffuse midline
glioma” and “H3 K27M” that returned 110 articles. We excluded articles without or
with incomplete abstracts, books and documents, conference abstracts, letters, editorials,
comments, published errata, case reports, and retracted articles. Only articles in
English were considered. The search was supplemented by reviewing the article reference
lists ([Fig. 1]). Overall, 96 articles were included in the study (as one of 97 articles originally
identified was retracted). There has been an increase in the number of articles related
to this tumor type published per year over the previous decade ([Fig. 2]).[1]
[2]
[3]
[4]
[5]
[6]
[7]
[8]
[9]
[10]
[11]
[12]
[13]
[14]
[15]
[16]
[17]
[18]
[19]
[20]
[21]
[22]
[23]
[24]
[25]
[26]
[27]
[28]
[29]
[30]
[31]
[32]
[33]
[34]
[35]
[36]
[37]
[38]
[39]
[40]
[41]
[42]
[43]
[44]
[45]
[46]
[47]
[48]
[49]
[50]
[51]
[52]
[53]
[54]
[55]
[56]
[57]
[58]
[59]
[60]
[61]
[62]
[63]
[64]
[65]
[66]
[67]
[68]
[69]
[70]
[71]
[72]
[73]
[74]
[75]
[76]
[77]
[78]
[79]
[80]
[81]
[82]
[83]
[84]
[85]
[86]
[87]
[88]
[89]
[90]
[91]
[92]
[93]
[94]
[95]
[96] Although a systematic review was attempted, the data was not, ultimately, appropriate
for metanalysis and therefore a rigorous integrative qualitative/narrative review
was undertaken.
Fig. 1 Flow diagram presenting study selection in the review.
Fig. 2 Graphic presentation of the studies included in the review by year, showing the growth
in publications in recent years.
Definition and Classification
Definition and Classification
H3 K27M-Mutant Diffuse Midline Glioma
In 2016, the WHO created a new tumor subtype, DMG H3 K27M-mutant, a diagnostic entity
based on its epigenetic signature.[1] The WHO criteria for diagnosis of DMG H3 K27M-mutant are described in [Table 2].
Table 2
2016 WHO classification criteria for DMG H3 K27M-mutant
DMG H3 K27M-mutant DMG criteria
|
Infiltrative glioma
and
Located in the midline
and
Loss of H3 K27me3
and
K27M mutation (immunohistochemistry)
in either H3F3A
or
HIST1H3B/C
|
Results from molecular analysis are desirable
|
Abbreviations: DMG, diffuse midline glioma; WHO, World Health Organization.
H3 K27-Altered Diffuse Midline Glioma
Subsequently, in 2021, the WHO amended this classification to DMG H3 K27-altered to
encompass the varying mechanisms that can lead to the epigenetic alteration characteristic
of this tumor type [21]
[50] and based on these molecular changes distinguished four subtypes of DMG H3 K27-altered
([Table 3])[76]:
Table 3
20121 WHO classification criteria for DMG H3 K27-altered
DMG H3 K27-altered criteria
|
Infiltrative glioma
and
Loss of H3 p.K28me3 (K27me3) (immunohistochemistry)
and
Located in the midline
and one of below
H3 p.K28M (K27M) or p.K28I (K27I) mutation (for H3 K27–mutant subtypes)
or
Pathogenic mutation or amplification of
EGFR
(for the
EGFR
-mutant subtype)
or
Overexpression of EZHIP (for the H3-wildtype with EZHIP overexpression subtype)
or
Methylation profile of one of the subtypes of DMG
|
Results from the molecular analysis that enable discrimination of the H3.1 or H3.2
p.K28 (K27)-mutant subtype from the H3.3 p.K28 (K27)-mutant subtype are desirable.
|
Abbreviations: DMG, diffuse midline glioma; EGFR, epidermal growth factor receptor
gene; EZHIP, enhancer of zest homolog inhibitory protein; WHO, World Health Organization.
-
DMG, H3.3 K27-mutant,
-
DMG, H3.1 or H3.2 K27-mutant,
-
DMG, H3-wildtype with EZHIP overexpression,
-
DMG, EGFR-mutant.
Genetic Characteristics and Cell of Origin
Genetic Characteristics and Cell of Origin
The molecular profile of DMG H3 K27-altered is still under investigation. The timing
of H3 K27M mutation in children and the effect on embryonic, neonatal, and childhood
brain development have not been characterized.[42] In addition, the relationship between spinal cord DMG and supratentorial DMG regarding
genetic alterations is also not clear, but spinal cord high-grade glioma in children
and adults frequently harbors H3 K27M mutations.[66]
In a 2018 study by Castel et al, investigating the epigenetic profile of H3 K27M-mutant
tumors in a group of 215 children with pediatric high grade gliomas, it was found
that subtypes of H3 K27M-mutant tumors were more accurately differentiated based on
their methylation profile and gene expression, rather than their location, particularly
when comparing supra- and infratentorial tumors. This may suggest these subtypes arise
from a different precursor or epigenetic reorganization.[75] Up to now, there is no conclusive evidence regarding the cell of origin for these
tumors, but a few studies suggest an oligodendrocyte precursor cell, due to the strong
immunophenotypic resemblance of the tumor cells to oligodendrocytes and the temporal
and spatial relationship of tumor incidence to the expression of the precursor cells
in neural development.[95] This was later supported in a study by Filbin et al in 2018, who also found that,
compared to IDH-mutant gliomas, H3 K27M-mutant gliomas are largely composed of cells
resembling oligodendrocyte precursor cells.[71] They also reported that H3 K27M-mutant gliomas had a large component of highly undifferentiated
cells with high proliferative index, consistent with their aggressive behavior compared
to IDH-mutant glioma.[71]
DMG H3 K27M-mutants are characterized by a somatic gain of function mutation that
leads to a lysine 27 to methionine (p.lys27Met: K27M) substitution in histone 3 (H3)
variants.[92] The K27 residue is essential for all variants of the H3 histone, as methylation
of K27 has an inhibitory effect on gene transcription. Thus, H3 affects gene expression
with a wide influence on cellular differentiation, proliferation, and epigenetic regulation.[61] The H3 K27M mutation, causing reduced H3 K27 trimethylation, affects stability of
genetic transcription, inhibiting cellular differentiation while conversely promoting
proliferation, with overexpression of genes that promote gliomagenesis.[61]
This K27M substitution affects histone variants H3.1 and H3.3 and results from mutations
to the HIST1HH3B/C or H3F3A gene, with the H3F3A gene being most commonly affected.[61] H3.1K27M tumors are also associated with mutations in the ACVR1 (Activin A receptor
type 1), while H3.3K27M gliomas are associated with a gain of function mutation of
PDGFRA (platelet-derived growth factor receptor alpha) and loss of function p53 mutations.
Interestingly, up to one-third of DMG H3 K27M-mutant have ACVR1 mutations. Alteration
of function of this receptor induces hyperactive bone morphogenic protein (BMP) signaling,
which arrests oligodendrocyte differentiation and is potentially tumorigenic. In H3 K27M-mutant
mouse models, mice with additional ACVR1 mutations survive only 70 days compared to
those without, which have an average survival of 180 days.[62]
[87]
H3 K27 hypomethylation is not necessarily due to K27M mutation.[45] In 2020, Castel et al, described a subgroup of DMG with EZHIP (enhancer of zest
homolog inhibitory protein) overexpression and H3 K27 trimethylation loss, in the
absence of a H3 K27M-mutation, in a cohort of 10 patients with diffuse pontine glioma.[45] This has subsequently been confirmed by other groups.[46] EZHIP binds to PRC 2 (polycomb repressive methyltransferase complex 2) through the
EZH2 subunit. PRC2 is the complex responsible for modifying the lysine residue on
histone 3, resulting in methylation of the H3 protein to HK27me3. By binding to PRC2,
EZHIP leads to a reduction in HK27me3. H3 K27me3 hypomethylation leads to driver mutations
that contribute to oncogene activation through gene expression alteration.[6] These genetic alterations include, as mentioned above, TP53 loss, associated with
radiotherapy resistance, tumor immortality and self-renewal of neural stem cells,[6]
[82] and gain of function of PDGFR, associated with tumor cell proliferation, invasion,
and migration.[90] Finally, a subset of DMG H3 K27M-mutant has been associated with amplification or
pathogenic mutation of the EGFR (epidermal growth factor receptor) and overexpression
of EGFR is associated with aggressive behavior and migration.[91] Conversely, prognosis is improved in those DMG H3 K27M-mutant tumor subtypes with
RAS-MAPK pathway alterations, such as FGFR1 (fibroblast growth factor receptor).[47] Additional genetic alterations described in these tumors involve PPM1D (protein
phosphatase, Mg2 +/Mn2+ dependent 1D, MYC proto-oncogene transcription factor family,
NF1 (neurofibromin 1), ATRX (α-thalassemia mental retardation X-linked protein), CDK
(cyclin dependent kinase)4/CDK6, and CCND (cyclin) 1-3.[6]
[88]
[92]
[93]
The prognosis of DMG H3 K27M-altered varies between the pediatric and adult population
and some genomic differences are noted between the two populations. The H3.3 K27M
mutation rate in adults is significantly higher than in children.[8]
[65]
[73]
[82] However, H3.1 mutant DMG occur more often in the young.[15]
[38] Molecular profiling reveals higher frequencies of ATRX loss and H3.3 mutation in
adult than in pediatric H3 K27M-mutant DMG and loss of ATRX expression has been associated
with improved survival.[49]
[63] TERT (telomerase reverse transcriptase) promoter mutations and MGMT (O[6]-methylguanine DNA methyltransferase) promoter methylation is not detected in children,[8] but are present in a few adult patients. Loss of ATRX expression is observed mainly
in adult patients. TERT promoter mutations and MGMT promoter methylation are present
in up to 50% of IDH WT (wildtype)/H3-WT gliomas,[1] in opposition to H3 K27M-mutant DMG, where TERT promoter mutations occur in 0 to
10% and MGMT is usually not methylated.[8]
[77] Similar to children, adults with H3 K27M-mutant tumors harbor TP53 and FGFR1 mutations.
To a large extent, however, the molecular profile of DMG H3 K27M-altered tumors in
adults is similar to that in children and the key to novel treatments of these tumors
is likely to be the development of epigenetic modulators ([Table 4]).[48]
[77]
[86] Tumor diagnosis and monitoring using “liquid biopsy” to detect DMG H3K27-altered
mutations in cell-free DNA (cfDNA) in cerebrospinal fluid (CSF) or blood is a promising
alternative to biopsy of highly eloquent regions such as the brainstem. Ventricular
CSF is more reliable than lumbar CSF samples.[12]
Table 4
Comparison of DMG H3 K27-altered in children and adults
|
Children
|
Adults
|
Prevalence
|
0.54 cases per 1 million person-years
|
3–5% of adult primary brain tumors
|
Location
|
Primarily brainstem
|
Primarily thalamus, also brainstem, cerebellum, corpus callosum, hypothalamus, cerebral
hemispheres, spinal cord
|
Clinical presentation
|
Ataxia, cranial nerve palsies, developmental regression
|
Focal neurological deficits, hemiparesis, ataxia and cranial nerve palsies
|
Radiological features
|
Nonspecific
|
Nonspecific
|
Histopathology features
|
No significant differences
|
No significant differences
|
Mutation in H3F3A or HIST1HB gene
|
More frequently
|
Less frequently
|
Mainstay of treatment
|
Radiotherapy
|
Radiotherapy
|
Prognosis
|
Median survival 9 to 15 months
|
Median survival 8–27.6 months
|
Abbreviation: DMG, diffuse midline gliomas.
Epidemiology
Our epidemiological understanding of DMG H3 K27M-altered DMG is still incomplete as
the entity was only described in 2021.[7] This tumor is more common in children than in adults, but the mechanism for the
pediatric predilection is unclear ([Table 4]). Some studies have suggested that children are three times more likely than adults
to have mutations in their H3F3A or HIST1HB gene.[77]
Of midline pediatric primary brain tumors, 80% are H3 K27M-altered, and they comprise
75% of all pediatric brainstem tumors.[9]
[19]
[49] However, DMG constitute only 10 to 15% of pediatric brain tumors overall and the
prevalence of DMG is estimated at 0.54 cases per 1 million person-years.[69]
[83] DMG comprise only 3 to 5% of adult primary brain tumors.[25]
[55]
Anatomical Location
DMG H3 K27-altered are almost exclusively located in midline CNS structures. In adults,
the most common location is the thalamus, but the brainstem, cerebellum, corpus callosum,
hypothalamus, cerebral hemispheres, and spinal cord have also been reported.[54] In children, brainstem tumors predominate ([Table 4])[81] chiefly in the pons (51.9%), then in the thalamus/basal ganglia (36.5%) and spine
(9.6%).[15] This is in contrast to non-H3 K27-altered tumors that are more widely distributed
in children (31.3% thalamus/basal ganglia, 31.3% spinal cord, 12.5% pons, 12.5% midbrain/tectum,
12.5% other intracranial locations).[15] No difference in location has been found for H3.1 and H3.3 molecular subgroups.[15] For spinal cord DMG H3 K27-altered, H3 K27M-mutated tumors have a greater propensity
for the thoracic spine, but there is no difference in location between mutated and
wildtype tumors.[68]
Clinical Presentation
Clinical Features of Cranial DMG H3 K27-Altered
The clinical presentation of DMG H3 K27-altered correlates with location. Focal neurological
deficits, hemiparesis, ataxia and cranial nerve palsies, account for more than 50%
of presentations.[48] In patients who present with cranial nerve palsies, cranial nerves VI and VII are
most commonly affected, but cranial nerves III, IV, and X have also been reported.[57] Symptoms of raised intracranial pressure are not common, although impaired level
of consciousness is reported.[48] Interestingly, hydrocephalus at first presentation occurs in less than 10% of patients
with DMG H3 K27-altered. In children, ataxia and cranial nerve palsies are frequent
modes of presentation.[24] Symptomatic intratumoral hemorrhage is rare, occurring in up to 6% of patients,
and generally only symptomatic in the pediatric population.[16]
[48] In contrast to other brainstem gliomas, DMG H3 K27-altered typically present with
an acute course of months and in both adults and children, with an average duration
of symptoms of 2 to 3 months ([Table 4]).[24]
[40]
Clinical Features of Spinal Cord DMG H3 K27-Altered
In spinal cord DMG H3 K27-altered, the clinical presentation is generally nonspecific,
and varies with the spinal level. Local pain generally precedes the development of
neurological signs and symptoms, which most commonly comprise ataxia, sphincter dysfunction,
and limb weakness.[31]
Radiological Characteristics
Radiological Characteristics
Radiological Features of Cranial DMG H3 K27-Altered
DMG H3 K27-altered in both adults and children have similar radiological features,
but no pathognomonic or unique imaging characteristics have been described when compared
to other cranial gliomas apart from location ([Figs. 3] and [4], [Table 4]).[81] On T1-weighted magnetic resonance imaging (MRI), these tumors are hypointense and
enhanced with contrast with a diffuse or heterogenous pattern. On T2-weighted MRI,
they have heterogenous signal intensity.[54] Contrast enhancement may be patchy, nodular, cystic, homogenous or ring-like, and
rarely absent.[49]
[54]
[65] Extensive spread can occur craniocaudally to involve the cerebral hemispheres and
spinal cord, and there may be leptomeningeal spread. Cortical invasion and leptomeningeal
tumor spread are usually related to normal expression of ATRX.[26] Other MRI findings include peritumoral edema, diffusion restriction, and hemorrhage,
with peritumoral edema most common.[30] However, no studies have identified imaging features able to differentiate DMG H3 K27M-altered
and other high-grade gliomas.[81]
Fig. 3 Magnetic resonance imaging (MRI) of pediatric brainstem diffuse midline glioma (DMG)-H3 K27M-altered.
(A) T1-weighted MRI. The tumor is hypointense. (B) T1-weighted contrast-enhanced MRI. There is minimal and heterogeneous enhancement.
(C) T2-weighted MRI. The tumor is heterogeneously hyperintense.
Fig. 4 MRI of adult thalamic DMG-H3 K27M-altered. (A) T2-weighted MRI. The tumor is heterogeneously hyperintense. (B) Fluid attenuated inversion recovery MRI. The tumor is hyperintense. (C) T1-weighted contrast-enhanced MRI. There is significant heterogeneous enhancement.
(D) Apparent diffusion coefficient MRI. There is minimal diffusion restriction. (E) Cerebral blood volume MRI. There is patchy increase in perfusion. (F) MR spectroscopy. A glial tumor trace is identified.
On diffusion-weighted imaging (DWI), DMG usually do not, or only mildly, restrict
and this is correlated with the presence of the H3K27M-mutation. Differences between
H3 K27M-mutant and wildtype DMG have been noted. DWI and MRI perfusion-weighted imaging
may be useful in preoperative prediction of H3 K27M-mutation status. The normalized
tumoral and peritumoral relative apparent diffusion coefficient (rADC) values have
been reported to be lower and the relative cerebral blood volume (rCBV) and the normalized
maximum rCBV (nrCBV) values higher in DMG H3 K27M-mutant.[4] It has also been suggested that lower minimum rADC values in DMG H3 K27M-mutant
indicate more malignant histology, likely representing a more complex tissue microstructure.[13] Multiparametric MRI-based radiomics models may be useful to predict H3 K27M-mutant
status in DMG [14] using ADC histogram parameters. Additionally, myo-inositol/creatine plus phosphocreatine
(Ins/tCr) ratios were lower than in the wildtype DMG in both children and adults.[17]
[41] Moreover, when comparing DMG H3 K27M-mutant with H3 K27M-wildtype tumors, significant
differences were found in T2 signal intensity, with H3.1 and H3.3 mutant tumors demonstrating
higher signal intensity and wildtype tumors demonstrating homogeneous T2 signal, and
T1 signal homogeneity, with H3 K27M-mutants demonstrating more heterogeneous T1 signal
([Table 5]). No significant imaging differences have been found between H3.1 and H3.3 K27M
mutant tumors[15] ([Figs. 3] and [4]).
Table 5
Comparison of radiological features of DMG H3 K27M-mutant and wildtype
|
DMG H3 K27M-mutant
|
DMG wildtype
|
MRI
|
Hyperintense on T2, heterogeneous intensity on T1
|
Homogeneous intensity on T1 and T2
|
DWI
|
Low rADC values
|
High rADC values
|
PWI
|
High rCBV and rCBV values
|
Low rCBV and rCBV values
|
MR spectroscopy
|
Low Ins/tCr ratios
|
High Ins/tCr ratios
|
Abbreviations: Cr, creatine; DMG, diffuse midline gliomas; DWI, diffusion-weighted
imaging; Ins, inositol; MRI, magnetic resonance imaging; PWI, perfusion weighted imaging;
rADC, relative apparent diffusion coefficient; rCBV, relative cerebral blood volume.
Radiological Features of Spinal Cord DMG H3 K27-Altered
Studies evaluating the radiological features of spinal cord DMG have demonstrated
a predilection for the cervical spine.[78]
[85] They are isointense on T1-weighted images and hyperintense on T2-weighted. They
have either absent or heterogenous peripheral enhancement.[35] No significant difference has been described for MRI features of H3 K27M-mutant
and H3 K27 wildtype tumors[27] however, spinal DMG H3 K27M-mutant tumors are more likely to display lesional hemorrhage.[68]
Pathology
There are no significant histopathological differences between adult and pediatric
DMG H3 K27-altered tumors.[8] Macroscopically, DMG H3 K27-altered share similar appearances to other gliomas in
that they are infiltrative, enlarge, and distort invaded structures and have associated
necrosis and hemorrhage.[29] Microscopically, tumor cells are generally small and monomorphic, but polymorphism
similar to other gliomas may be seen.[82] Microvascular proliferation, necrosis, and frequent mitoses may be seen but are
not associated with prognosis.[84] Perineural or perivascular clustering does not occur.[1] According to the WHO 2021 classification, those tumors are grade IV, irrespective
of histopathological appearance.[1]
Immunophenotyping and molecular confirmation of characteristic mutations is essential
for diagnosis. Typically, DMG H3 K27-altered stain positive for OLIG2, MAP2, and S100.
GFAP (glial fibrillary acidic protein) immunoreactivity varies. The EGFR-mutant subtype
often shows positive straining for GFAP and less commonly OLIG2 and SOX10. Antibodies
against H3 p.K28M (K27M), H3 p.K28me3 (K27me3), and EZHIP (CXorf67) combined with
additional molecular analysis are key to confirm the diagnosis[45]
[96] ([Figs. 5] and [6]).
Fig. 5 Histopathology of diffuse midline glioma (DMG) H3 K27M-altered with high grade appearance.
(A–C) Sections show a hypercellular glial tumor. The tumor cells have hyperchromatic,
pleomorphic nuclei. Mitoses are numerous, being present in numbers up to 14 per 10
high-power field. There is no necrosis or microvascular proliferation. (D–F) isocitrate dehydrogenase (IDH) immunostain is negative, indicating the absence of
an IDH1 R132H mutation. α-thalassemia mental retardation X-linked protein (ATRX) is
retained. OLIG2 is positive, indicating the glial nature of the tumor. (G, H) H3K27M-positive (mutated), H3K27me3—lost. H3K27M immunostain is positive, establishing
the diagnosis of diffuse midline glioma, H3 K27-altered (central nervous system World
Health Organization grade 4).
Fig. 6 Histopathology of diffuse midline glioma (DMG) H3 K27M-altered with low grade appearance.
(A–C) Sections from a left thalamic lesion showed a moderately cellular glioma. The tumor
cell nuclei were moderately enlarged and angulated. No mitoses were identified, and
there was no necrosis or microvascular proliferation. (D–F) The isocitrate dehydrogenase (IDH) immunostain was negative, indicating a lack of
an IDH1 R132H mutation. Ki67 was elevated at approximately 10%. The H3K27M immunostain
was strongly positive, establishing the diagnosis of diffuse midline glioma, H3 K27-altered
(central nervous system World Health Organization grade 4). In addition, the lesion
underwent pyrosequencing for H3F3A which confirmed the presence of the K27M mutation.
Treatment
There is no effective treatment for DMG H3 K27M-altered DMG, including for chemotherapy
and targeted molecular agents. There is a paucity of literature on treatment of the
specific DMG H3 K27M-altered tumor subtypes; therefore, treatment decisions rely on
studies addressing DMG in general. Due to the predominantly eloquent location of these
tumors, surgical management is often limited to biopsy to avoid postoperative morbidity.
The current mainstay of treatment is radiotherapy, which has been shown to provide
a symptomatic and survival benefit with limited disease control.[58]
Radiotherapy in H3 K27M-Altered DMG
Fractionated external beam radiotherapy is the mainstay for DMG H3 K27M-altered, due
to the known effects in other gliomas.[43] The current treatment regimen is 54 Gy in 30 fractions.[58] In a systematic review by Gallito et al, 49 studies investigating the role of conventionally
fractionated, hyperfractionated, and hypofractionated radiotherapy in DIPG were evaluated.
Patients who received radiotherapy had a median survival of 11 months as opposed to
6 months without radiotherapy.[58] The median survival of patients who received hyperfractionated, hypofractionated
and conventionally fractionated radiotherapy was 7.9, 10.2, and 12 months, respectively.[58]
Chemotherapy and Targeted Agents in H3 K27M-Altered DMG
Despite no demonstrated impact on prognosis, chemotherapy is commonly used for DMG.
However, even in combination with radiotherapy, chemotherapy has shown no survival
benefit.[22] The most common agent is temozolomide due to its demonstrated effect in other gliomas.[70] Other chemotherapy and targeted agents, including panobinostat, gefitinib, thiotepa,
and busulfan, have been trialed, with no benefit.[22]
[39]
[51] In the study by Izzuddeen et al, children with DIPG were randomized to conventional
fractionated radiotherapy or hypofractionated radiotherapy with concurrent temozolomide.
The median survival was 11 and 12 months, respectively, with no significant difference,
but the patients who received radiotherapy and chemotherapy had a higher incidence
of hematological toxicity.[44] Cohen et al compared temozolomide and radiotherapy to combination chemotherapy with
carboplatin/cisplatin, etoposide, cyclophosphamide, and vincristine with radiotherapy,
with no improved event-free survival rate compared to combination chemotherapy in
DIPG.[94] Patients receiving temozolomide and radiotherapy had a 1-year overall survival of
40%, compared to 32% in those receiving combination chemotherapy and radiotherapy.
Interestingly, H3 K27M mutations cause DNA hypomethylation, including the MGMT promoter.[79] Therefore, expression of MGMT in DMG H3 K27M-altered tumors will lead to temozolomide
resistance in these patients.[34]
Identification of new therapeutic targets and corresponding targeted therapies against
them is an area of intense research and a detailed overview is beyond the scope of
this review. Abe et al suggested ALK2 receptor inhibitors may benefit patients with
DMG as the type I BMP receptor ALK2 is encoded by ACVR1 gene that is frequently mutated
in DMG HIST1H3B-mutant, but not H3.3 H3 K27M-mutant tumors.[34] This receptor mutation causes constitutive activation of the BMP signaling pathway
that can also be activated in ACVR1-wildtype DMG. Thus, the use of ALK2 inhibitors
may be warranted. The same group has suggested PARP (poly (ADP-ribose) polymerase)
inhibitors as PARP-mediated base excision facilitates repair of damaged DNA. DMG cells
express PARP, thus inhibiting this pathway may be a potential therapeutic .[34] Additionally, ONC206, a DRD2/3/4 (dopamine receptor D2/3/4) antagonist, is currently
in trial for newly diagnosed and recurrent DMG (ClinicalTrials.gov Identifier: NCT04732065.
The availability of new chemotherapy and targeted treatments would provide DMG patients
with the option of more efficacious drugs with improved tolerability in a more convenient
dosage format. Thus, intense ongoing research is required to give new hope to DMG
patients.
Surgery in H3 K27M-Altered DMG
Given the eloquent location of DMG H3 K27M-altered, surgical resection is almost always
contraindicated as no survival benefit has been demonstrated and there is a high risk
of neurological deterioration and death. No randomized study to date has investigated
the role of surgery in DMG. The primary aim of surgery is to obtain a biopsy for diagnosis,
prognosis, and research.[22]
[77] The single-center, retrospective study by Wang et al that investigated outcome for
those with DMG H3 K27M-mutant of the spinal cord found that surgical treatment, including
biopsy, subtotal resection, or aggressive resection, showed no significant survival
benefit.[31] Interestingly, another single-center, retrospective study by Dorfer et al reported
that surgical resection, as opposed to biopsy, was associated with a statistically
significant improvement in overall survival, in a pediatric population of both H3 K27M-mutant
(14 patients) and H3 K27M-wildtype (35 patients) thalamic gliomas.[33] However, this was associated with high postoperative surgical morbidity including
ataxia, visual field defects, and hemiparesis. A separate analysis of the DMG H3 K27-mutant
tumors was not done and due to the small number of patients, the inclusion of better
prognosis patients with pilocytic astrocytoma (almost 50% of the cohort) and the retrospective
data, this study should be interpreted with caution.
In the multicenter, retrospective study by Karremann et al, extent of resection was
not associated with improved survival in adults and children with DMG H3 K27M-mutant
and H3-wildtype tumors.[76] A trend to shorter survival in patients in the H3 K27M-mutant group who underwent
more than 90% resection compared to those with <90% resection was reported, with 10%
2-year survival for those with less than 90% resection and 0% for those with more
than 90% resection.[76] These findings were replicated by Park et al, who also reported surgical resection
was not associated with increased survival in adults with DMG (21.9 vs. 20.4 months),
including for gross total compared to subtotal resection (13.2 vs. 21.9 months).[20] Similarly, in the review of adult and pediatric patients with H3 K37M-mutant DMG
by Vuong et al, surgical resection was not associated with improved overall survival.[25]
Immunotherapy and Future Perspectives
The search for novel therapies has intensified recently, particularly in relation
to immunotherapy.[11] Phase 1 studies of chimeric antigen receptor (CAR) T-cells (CAR-T cells) in patients
with pontine DMG H3 K27M-mutant have demonstrated both radiological improvements,
with reduced T2/FLAIR signal extent on MRI, and clinical improvement. There was, however,
also CAR-T cell-mediated inflammation with brainstem edema and obstructive hydrocephalus
or transient worsening of clinical deficits.[5] Other forms of immunotherapy, including immune-modulatory and dendritic cell vaccines,
have shown initial promise.[74]
In children, crenolanib, a selective inhibitor of PDGFR-mediated phosphorylation,
has shown promising early phase results. It was well tolerated at doses slightly higher
than the established maximum tolerated dose in adults, with a similar toxicity spectrum.[18]
Combination treatments of personalized cytotoxic agents or targeted inhibitors, chosen
based on molecular analysis of biopsied tissue, are being examined in ongoing trials.[60]
[80] Additionally, novel methods of drug delivery, including CED (convection-enhancing
delivery), with small volume infusions via intraparenchymal catheters [59] or MRI-guided focused ultrasound that enhances focally drug delivery of targeted
chemotherapeutics to brain tumors are being investigated to bypass the blood–brain
barrier.[28] In a study by Gojo et al, mutations identified in patients with H3 K27M-mutant tumors,
including those in E545K, G118D, and ACVR1, were targeted in a personalized medicine
approach. However, the median survival of 16.5 months for the treatment group was
not different to 17.5 months for the control group.[60] Chi et al reported a promising response using a selective dopamine receptor D2/3
antagonist.[64] Clearly further research into new approaches is needed.
Prognosis
DMG H3 K27M-altered carry a poor prognosis for both adults and children, with a median
survival around 1 year and a poorer prognosis compared to most other gliomas ([Table 4]).[28]
[29]
[36]
[40]
[53]
[59]
[67]
[86]
[92] In adults, prognosis does not vary by anatomical site.[10] In children with DMG H3 K27M-mutant, the median survival is 9 to 15 months,[89] except in children with NF1 for whom all DMG have an extremely poor prognosis, independent
of the presence or absence of H3 K27M mutation.[52]
Adults fare slightly better with a median survival of 8 to 27.6 months.[8]
[49]
[65]
[72]
[77] The mechanisms of this survival advantage are unknown; however, as pediatric DMG
H3 K27M-mutant tumors occur more commonly in the brainstem, and adult tumors in the
thalamus, location may be important, with a greater morbidity in brainstem tumors.[10]
[49]
[73] Additionally, the molecular genetics of thalamic DMG H3 K27M-mutant tumors differ
compared to those in the brainstem, including in expression of the CDK6, TP53, K27M,
and IDH1 genes.[86] Additionally, TP53 mutation has been reported as a poor prognostic indicator, but
FGFR1 mutation and ATRX loss confers prolonged survival.[25] Thus, the molecular characteristics are likely more important than location to predict
prognosis and should be examined in all patients.
For spinal cord DMG, the H3 K27M mutation predicts a worse outcome than other gliomas,
although thoracic tumors have a significantly better prognosis than cervical.[37]
Conclusion
DMG H3 K27-altered are uniformly fatal primary CNS tumors for which the biology is
only beginning to be determined. Differences in location and prognosis between adults
and children are not fully understood. Current treatments are ineffective with research
efforts aimed at novel drug delivery mechanisms, targeted agents, and immunotherapy.
Further investigation is clearly required to improve outcomes.