U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

De Vleeschouwer S, editor. Glioblastoma [Internet]. Brisbane (AU): Codon Publications; 2017 Sep 27. doi: 10.15586/codon.glioblastoma.2017.ch15

Cover of Glioblastoma

Glioblastoma [Internet].

Show details

Chapter 15Pediatric Glioblastoma

and .

Author Information and Affiliations

Abstract

Glioblastoma in children, when compared with adults, is relatively rare. Despite this rarity, it is apparent from the limited number of publications that pediatric glioblastoma is quite distinct from their adult counterparts. The differences pertain to the molecular genetics, effectiveness of the adjuvant therapies, and possibly the prognosis after treatment. With a plethora of path-breaking translational research coming through in recent times, a host of new information is now available on pediatric glioblastomas that holds great promise as far as the future treatment options are concerned. This chapter is an attempt to highlight the key clinical aspects of pediatric glioblastoma in the light of the emerging clinical and laboratory evidence.

Key words:

High-grade glioma, Neuroimaging, Pediatric glioblastoma, Radiotherapy, Supratentorial

Introduction

Glioblastoma (GBM) is the commonest and the most lethal primary brain tumor in adults (1). In contrast, GBM accounts for no more than 3–15% of primary central nervous system (CNS) tumors in children (27). This is despite the fact that CNS tumors are the most common solid tumors of childhood, and 40–50% of these tumors are constituted by the astrocytomas (8). Naturally, this relative rarity has been a great hurdle in properly deciphering the enigma of pediatric glioblastomas (p-GBM). Nevertheless, GBM remains an equally devastating disease in children with substantial morbidity and mortality. The reported median survival in p-GBM ranges from 13 to 73 months with a 5-year survival of less than 20% (2, 4, 6, 913). A few reports, however, reveal a relatively better prognosis and long-term survival figures in p-GBM as compared with adults (4, 911).

Maximal safe tumor resection followed by concurrent and adjuvant chemoradiation using oral temozolomide (TMZ) is the current standard of care in adult GBM (12). In fact, no such standard exists in children although a similar management policy is employed by most neurosurgeons across the globe. While there is sufficient evidence for the prognostic impact of maximal surgical excision of the visible tumor mass (4, 9, 10, 14), the concerns of irradiation on the developing brain and contradictory results of various chemotherapy regimens in p-GBM make the treatment decisions rather complicated and difficult in children.

A number of key clinical and laboratory investigations have led to a far better understanding of tumor biology of p-GBM today. Unlike in the past, these tumors are now considered to be distinctly different biological diseases compared with the adults. Numerous novel targeted drug therapies are emerging for the postoperative management of these tumors and hold great promise in times to come. However, it has to be agreed that the translation of the laboratory research into clinical patient management and patient outcomes have been relatively disappointing.

Epidemiology

Tumors of the CNS are the second most common childhood tumors after leukemia and are the commonest solid tumors in childhood (15). The overall incidence of primary CNS tumors in childhood is estimated to be approximately 30 per million (16). While astrocytic tumors account for 40–50% of the CNS tumors in children, high-grade gliomas are relatively rare. Estimation of the true incidence of p-GBM is often hampered by the fact that most studies tend to analyze GBMs (WHO grade IV) and anaplastic astrocytomas (WHO grade III) together, probably to derive a larger sample size for analysis. There is also a lack of consensus regarding the definition of pediatric age group. While the majority of studies consider 18 years as the cutoff, some studies consider 16 years or even 21 years for the same. On the contrary, many researchers also include adolescents in their analysis, thereby adding a lot of heterogeneity in the literature that hampers their holistic analysis.

As per the Central Brain Tumor Registry of the United States (CBTRUS) 2012 data, the incidence of pediatric high-grade glioma is approximately 0.85 per 100,000 (17). Most studies estimate the incidence of pediatric high-grade gliomas to be between 8 and 12% (18, 19). When only the GBM is considered, its incidence in the pediatric age group varies from 3 to 15% (27).

p-GBMs are most commonly reported in the second decade of life although their occurrence have been reported even in utero (2, 4, 5, 9, 20, 21). The highest incidence of p-GBM is seen between ages 15 and 19. This probably reflects the cumulative effects of different genetic insults in the eventual tumorigenesis. As far as gender predilection is concerned, most studies point towards a male predilection, the reasons for which are rather unknown. While it is unclear whether the patients’ gender has any effect on the disease outcome, GBM in very young patients (<5 years) may have a slightly better prognosis compared to the older children.

As far as location is concerned, p-GBM is most commonly seen in the supratentorial brain, when the brainstem is excluded (12, 15, 20). Primary spinal cord high-grade gliomas constitute only 3% of the pediatric high-grade gliomas (22). In the supratentorial compartment, cerebral hemispheres are affected in nearly 50% of the cases. The incidence of deeper midline structure involvement, for example, thalamus, corpus callosum, hypothalamus, etc., is fortunately low (4, 23). In the infratentorial compartment, cerebellum is an extremely uncommon site with 1–2% of the GBMs in children affecting this site (24). Brainstem high-grade gliomas constitute nearly 20% of the intrinsic tumors in this area (25). Interestingly, when the nonbrainstem high-grade gliomas are analyzed, younger children are particularly susceptible compared with the older children and adolescents (26).

p-GBM remains a multifactorial disease similar to the malignancies at other age groups and systems. Prior history of ionizing radiations, particularly for hematologic malignancies like leukemia, is a proven factor in tumorigenesis (27, 28). Certain syndromes like Neurofibromatosis-1, Li-Fraumeni syndrome (characterized by inactivation of the tumor suppressor gene p53), and Turcot syndrome are also known to be associated with high-grade gliomas in children (23). We studied the association of matrix metalloproteinase (MMP)-1 gene of 110 patients with adult GBM and found a very high prevalence of 2G allele in these patients. A very strong association between 2G/2G genotype and GBM was detected in our study which indicated likely susceptibility for GBM in patients harboring this particular variant of MMP-1 (29). We also noted, in a separate study, that MMP-2 gene was not responsible for an increased susceptibility to high-grade gliomas in our population, unlike MMP-1 (30). These factors are discussed in detail below. However, it has to be agreed that the genetic syndromes constitute only a minuscule part of the entire spectrum of pediatric high-grade gliomas, the majority of which are sporadic without any clearly known predisposition. A number of genetic factors are now known to be associated with GBM in general and p-GBM in particular. These include p53 mutation, PDGFR mutation, H3K27M, etc., to name a few.

Pathology and Molecular Biology

GROSS AND MICROSCOPIC FEATURES

GBMs originate from the astrocytes, the chief glial constituent of the CNS. The gross and microscopic features of p-GBM are no different from the adults (5). They are diffusely infiltrative despite their often apparent well-demarcated nature on imaging or even at surgery. These are usually dusky red or yellowish pink, friable, and vascularized tumors. Presence of thrombosed vessels inside the tumor mass is very much characteristic. There may be foci of hemorrhage or necrosis inside the lesion. Calcific components are rare but can be seen particularly in secondary GBMs. The usual sites of affections are supratentorial cerebral lobes like the frontal/temporal lobe. As mentioned before, deeper midline location, infratentorial compartment, and spinal cord locations are relatively uncommon (12, 15, 20). Although extracranial distant metastasis is rare, secondary dissemination inside the brain or the leptomeninges does occur in nearly 17% of patients (31).

Microscopically, these tumors are typically characterized by four histopathological hallmarks, namely, hypercellularity, nuclear atypia, pseudopalisading necrosis, and vascular endothelial cell proliferation. Multinucleated cells, bizarre nuclei, and neovascularization with glomeruloid formation are often detected. There are abundant mitotic figures with a high MIB labeling index, indicating a highly aggressive growth potential of the tumor. Satellite lesions are frequently seen. In one of the studies from our center, nearly 11% of GBMs were found to be multiple, the majority of the patients in that study being adults. Although these figures may not necessarily apply in pediatric population, our study showed that a high mitotic index (>40%), satellitosis, and a higher proportion of small cells correlate with tumor multiplicity in GBM (32).

PATHOLOGICAL VARIANTS/PATTERNS

WHO recognizes three variants of GBMs: giant-cell GBM, gliosarcoma, and, most recently, the epithelioid GBM. While the former two variants are relatively rare in children, epithelioid GBM, characterized by large eosinophilic cells, prominent melanoma-like nuclei, and often rhabdoid cells, is more common in children. These tumors tend to occur in the midline and are typically characterized by positive immunoreactivity for BRAF V600E, indicating their origin from a pre-existing low-grade precursor (33). GBM with primitive neuroectodermal components, small-cell GBM, and granular cell GBM are not true variants but specific patterns recognized by WHO. While the former is associated with a high risk of CSF spread, the latter two patterns portend poor outcomes despite the lack of necrosis inside the tumor (33).

MOLECULAR BIOLOGY

Incorporating the latest evidence emanating from laboratory research all over the world, WHO has updated its classification of brain tumors in 2016 (33). GBM is now classified as per the Isocitrate Dehydrogenase (IDH) gene mutation status. As a result, GBM can be either IDH mutation positive or IDH wild type. While the former group represents the secondary GBMs, the latter represents de novo lesions occurring mostly in elderly patients. But studies on pediatric high-grade gliomas, including GBMs, have noted a very low incidence of IDH mutation, particularly in younger children (34). Thus, for all practical purposes, p-GBM is almost always IDH wild type, although as Pollack et al. showed, the incidence of secondary GBM (IDH mutated) may be higher in adolescents and younger adults (35). Lack of IDH mutation negatively impacts the outcome to therapy.

As far as the underlying genetic alterations are concerned, it is now wellknown that p-GBMs have a higher incidence of p53 mutation/overexpression than mutation of epithelial growth factor receptor (EGFR) or deletion of phosphatase and tensin homologue (PTEN), the signatures of adult GBM. P53 mutation is particularly common in young children (<3 years). Interestingly, there may be overexpression of p53 in p-GBM even in the face of absent TP53 mutation (5, 36). Although, traditionally it was believed that PTEN mutation (with inactivation of its tumor suppressive effect on the downstream AKT pathway) played little role in p-GBM, certain recent studies have questioned the traditional belief and have noted activation of the AKT pathway, a feature that may negatively affect the patient outcomes (37). ATRX mutations have been reported in a fraction of p-GBMs, usually in the presence of p-53 mutation. Such tumors affect older children and are usually associated with better prognosis (38).

Recent studies have identified histone mutations (H3.3) in the DNA of pediatric high-grade glioma patients (39, 40). In fact, the H3K27M variant, wherein lysine is replaced by methionine at 27 positions, has been identified as an exclusive finding in pediatric high-grade gliomas. In addition, in slightly older children, replacement of glycine by valine or arginine at amino acid 34 of the H3.3 nucleic acid (G34V/R) is also frequently identified. While H3K27M variant is associated with poor prognosis, the outcome in patients with G34V/R is thought to be relatively better. Hemizygous deletions of ODZ3 have been described in the epithelioid variant of p-GBM. This particular variant, as already stated, shows BRAF V600E mutation, indicating its origin from low-grade pilomyxoid astrocytomas.

Vascular endothelial growth factor (VEGF) is commonly expressed by adult GBM cells. It is responsible for increased vascularity, tumor progression, and infiltration capacity of GBMs. As a result, anti-VEGF (bevacizumab) therapy is frequently employed in adult GBMs. The expression of VEGF is, however, relatively low in p-GBM, and it may be responsible for comparative ineffectiveness of anti-VEGF therapy in children (41). Somatic mutations of PDGFRA have also been recently reported in pediatric HGGs. This is in contrast to EGFRA mutations seen in adults. This has prompted anti-PDGFRA therapy in the form of receptor tyrosine kinase inhibitor imatinib (42, 43).

Apart from these clearly defined molecular aberrations, the pediatric HGGs seem to possess a significantly higher incidence of gains at 1q and losses at 16q and 4q. The 1p19q co-deletions are characteristic of oligodendroglial lineage. Some GBMs, particularly the secondary GBM, may have partial deletion of either 1p or more commonly 19q arms, and this may potentially confer a better prognosis (44). These GBMs are rare and were previously classified as GBM with oligodendroglial component (2007, WHO). As far as the O6-Methylguanine-DNA Methyltransferase (MGMT) promoter methylation status is concerned, it assumes an important prognostic significance in GBMs. Inactivation of MGMT generally correlates with chemoresponsiveness of these tumors. Studies on the expression of MGMT in p-GBM have revealed little difference in the promoter methylation status in children, with some studies noting even an overexpression of MGMT in tumors in children (45). It may be one of the reasons for reduced efficacy of TMZ in children compared with the adults. Whenever present, the prognostic significance of the inactivating hypermethylation of MGMT confers a survival benefit to the affected children. Thus, a number of key molecular signatures of GBMs are now available for diagnosis and prognostication of high-grade gliomas in general.

Clinical Features

Symptoms of GBM are protean and are largely nonspecific. The duration of symptoms is usually short, often spanning a few months (2, 4, 20). The most common presentations are headache, vomiting, diplopia, and altered sensorium, indicating raised intracranial tension. It is reported as an initial symptom in 80–100% of patients (4, 20, 46). Children with GBM may present with acute neurological deterioration, usually from intratumoral hemorrhage, although an episode of seizure may also bring about such a dramatic presentation. The incidence of seizures as a clinical feature is estimated to be around 30%, being more common when frontotemporal lobes are affected or in the setting of secondary GBMs (4, 20, 46). Some authors have noted a relatively higher incidence of seizures in p-GBM, unlike in adults. Focal symptoms like neurological deficits, cranial nerve dysfunction, cerebellar symptoms, etc., depend on the location of the lesion. Neurological deficits, when present preoperatively, are known to affect postoperative prognosis negatively (47). Compared to the older children, infants and young children often present with nonspecific complaints, such as failure to thrive, lethargy, nausea/emesis, and macrocephaly, which, at times, can be difficult to diagnose. Assessment of the functional status is critically important in a disease like GBM. A number of scales are available to determine the functional status of children with brain tumors that are used preoperatively as well as during the posttreatment period. Karnofsky performance scale (KPS) is one such commonly utilized scale (4). A cutoff score of 80 generally differentiates good performance status from the poorly performing patients. In addition, neurological function scale (NFS) is another similar assessment tool in children (10). In our own study, the prognostic significance of preoperative performance status of these patients on the postoperative outcome was clearly demonstrated (4).

Neuroimaging

Neuroimaging plays an important part in the diagnosis, management, and prognostication of GBMs. Computed tomography (CT) and magnetic resonance imaging (MRI) form the backbones as far as the radiological assessment of these tumors is concerned. On CT and conventional sequences of MRI, these tumors appear as irregular, heterogeneously contrast-enhancing masses with significant perilesional edema. Although, some of the anaplastic astrocytomas may not enhance, GBMs almost always enhance. Necrosis, hemorrhage, and a garland pattern of enhancement are often characteristic of GBMs. The common differential diagnosis includes metastasis, lymphoma, brain abscess, etc. Although contrast-enhanced computed tomogram is usually characteristic, MRI provides finer details needed for surgical as well as radiotherapy (RT) planning. Magnetic resonance spectroscopy typically displays choline peak with reduced N-acetyl aspartate in the region of the tumor although no such peaks may be seen in areas of necrosis. Diffusion-weighted images may show restricted diffusion with low apparent diffusion coefficient in the cellular parts of the tumor. While contrast images delineate the portions of the tumor with blood–brain barrier disruption, T2 fluid-attenuated inversion recovery (FLAIR) images clearly demonstrate the nonenhancing and edematous portions. Figure 1 shows the different radiological characteristics of GBM. Apart from the above diagnostic information, recent MRI techniques like functional MRI, tractography, etc., help in planning tumor resections, especially in eloquent locations. Perfusion-weighted MRI, although not routinely used, shows increased vascularity inside the tumor, a characteristic feature in high-grade gliomas. It can be performed by utilizing one of the three techniques, namely, magnetic resonance perfusion imaging, dynamic susceptibility contrast-MRI, and dynamic contrast-enhanced MRI (48).

Figure 1. (A).

Figure 1

(A). Post-contrast computed tomography of the head shows a left frontal irregularly enhancing intraaxial mass of size approximately 4.5 × 4.5 cm with perilesional edema. The mass is causing effacement of the adjacent lateral ventricle. The mass (more...)

An important role of neuroimaging is to assess response to therapy. Volumetric MRI is able to provide the extent of tumor excision, an important prognostic variable in GBMs. Moreover, the response assessment criteria like McDonald’s criteria and RANO criteria rely on the posttreatment neuroimaging. In this regard, neuroimaging plays an important role in deciphering the true nature of two interesting radiological phenomena in high-grade gliomas, namely, “pseudo-progression” and “pseudoresponse” (48, 49). The former typically occurs after 3 months of chemoradiation when an erroneous observation of tumor progression is made when, in actuality, there is none. Different neuroimaging modalities like MR spectroscopy, diffusion-weighted MRI, positron emission tomography (PET), and perfusion-weighted MRI, etc., usually reveal the true nature of this spurious radiological finding. In true tumor progression, the typical metabolite profile of elevated choline, choline: creatinine >2, and reduced N-acetyl aspartate peaks would be seen in MRS while there will be diffusion restriction on DWI. PET and perfusion-weighted MRI usually shows hypermetabolism and increased blood flow inside the enhancing area if there is true tumor progression, while the findings will be contradictory in pseudoprogression. Pseudo response, on the contrary, occurs as early as 24 h of anti-VEGF (Bevacizumab) therapy and is characterized by the lack of enhancement on contrast images even though the tumor is still there. Such pseudo responses are detected using T2 flair and diffusion-weighted MRI. While T2 flair shows the nonenhancing tumor as a hyperintense area, persistent diffusion restriction in the suspected area is usually diagnostic of residual tumor tissue. Neuroimaging has prognostic significance as well. In a recent study, Wangaryattawanich et al. (49) from MD Anderson Cancer Hospital showed that preoperative eloquent tumor location (P = 0.007), deep white matter invasion (P = 0.006), tumor volume (measured on contrast T1) >35,000 cm3 (P = 0.08), and volume of nonenhancing tumor/brain edema volume (measured in T2 flair) >85,000 mm3 (P = 0.003) were associated with poor survival in GBM.

Management

SURGERY

Maximal surgical resection followed by chemoradiotherapy remains the best current treatment in adult GBMs. Numerous studies have reiterated the utility of maximal tumor removal on both progression free as well as overall survival (OS) in p-GBM (4, 9, 10, 13, 14, 51). The Children’s Cancer Group (CCG) study-945 showed that children with HGG who underwent a surgical resection of 90% or greater had a progression-free survival (PFS) of 35 ± 7% as compared with a 5-year PFS of 17 ± 4% in patients who did not (50, 51). Reporting on probably the largest single-center experience of p-GBM, we have also shown that the extent of tumor excision was a strong predictor of long progression-free survival as well as OS (4). The utility of maximal surgical excision has been proven in a recent multiple propensity analysis, the scientific value of which is as good as a randomized clinical trial (14). The extent of resection is, however, dependent on the location as well as extensions of the tumor (4). Brainstem location, midline supratentorial tumors, tumors affecting eloquent area, etc., are often difficult to excise completely without incurring significant neurological deficits. Apart from providing tissue for diagnosis, surgical debulking relieves tumor-related mass effect and potentiates the effect of the adjuvant therapy. Different intraoperative imaging techniques may allow larger extents of tumor excision which in turn translates into better survival outcomes. These advanced techniques include intraoperative neuronavigation, intraoperative ultrasound, intraoperative MRI, intraoperative cortical mapping, etc. Recent technological advances utilizing microfluidic chips allow for rapid analysis of the operative specimen for molecular signatures like IDH mutation within no time (51, 52). Therefore, it is possible now to make a molecular diagnosis even intraoperatively. Such advances have the potential of facilitating intraoperative decision-making regarding the radicalism of the surgical excision in the future.

RADIATION THERAPY

Radiotherapy is an integral part of the comprehensive management basket of p-GBM. This is more so as the role of chemotherapy is not yet clear in these patients unlike their adult counterparts. Usually, the radiotherapy dose ranges from 50 to 60 Gy fractionated over 5–6 weeks (52, 53). Trials on hypo/hyper fractionation of the total dose have not shown any better results (54). It is routinely used in children aged more than 3 years. The primary reason why it should not be used before 3 years of age is that RT may lead to adverse neurocognitive complications due to its damaging effects on the developing brain. Moreover, it is believed that the tumors in the early years of life are rather indolent, responding less completely to irradiation (55, 56). The various long-term sequels of childhood cranial irradiation include endocrine dysfunctions, neurocognitive impairments, psychosocial and behavioral abnormalities, ototoxicity, growth abnormalities, and heightened chances of secondary malignancies (57). There has been a change in the way RT is administered in these patients. Previously, RT protocols encompassed the whole brain RT with additional boost at the site of the tumor with a 2-cm margin. However, with improvements in technology and accurate delineation of tumor margins, made possible by newer generation MRI scanners, currently RT is delivered using 3-dimensional conformal techniques. Thus, many of the earlier concerns with radiation treatment are no longer there. The conformal radiation treatment technologies include intensity-modulated RT, stereotactic RT, and proton beam RT (57). The latter techniques employ head fixation using rigid frames to enable precisely localized radiation. Therefore, recent advances have made RT in pediatric high-grade gliomas rather safe and effective.

CHEMOTHERAPY

Chemotherapy forms an important and integral part of the comprehensive treatment regime in adult GBMs. The same, however, cannot be said about the pediatric patients. Sposto et al. (CCG 943 trial) were the first to prove the effectiveness of chemotherapy (concomitant vincristine and adjuvant eight cycles of PCV regimen comprising procarbazine, CCNU, and vincristine) in high-grade gliomas (57). This regimen demonstrated a statistically significant improvement in the outcome of patients with GBM treated with chemotherapy compared with RT-alone group (5-year PFS: 42% vs. 18%). However, the regimen never reproduced similar results thereafter and hence failed to become a standard regime. It was later found that many of the patients included in that trial actually had low-grade gliomas. A subsequent trial looked at eight-drug chemotherapeutic regimen consisting of vincristine, carmustine, procarbazine, hydroxyurea, cisplatin, cytosine arabinoside, prednisone, and dimethyl-triazenoimidazole-carboxamide (DTIC) in children younger than 2 years (50, 58). This trial did not show any benefit in p-GBM. Moreover, the regimen failed to show any effectiveness in older children as well. Intensive chemotherapy after surgical excision has shown promising results in children, provided the tumor excision was complete. The treatment using HIT-GBM-C protocol showed 5-year OS rate for these patients, with total resections of 63% versus 17% for historical controls. The OS was 19% at 60 months from diagnosis (50, 59). Some trials have also looked into the high-dose chemotherapy with bone marrow rescue in p-GBM, but these trials are mainly applicable for recurrent cases and their results have not been consistently proven. Unacceptable drug toxicity is usually the major handicap in these trials (60).

The landmark trial by Stupp et al. showed that the addition of concomitant and adjuvant TMZ improved progression-free survival and OS in adult GBMs (12). Five years’ OS in this study for the TMZ arm was 9.8% versus 1.9% for the radiation alone arm. This trial has established the current therapeutic standard of concurrent and adjuvant TMZ in adult GBM. The Stupp trial, however, did not include p-GBMs. Thus, despite its landmark findings, the Stupp trial has not really helped matters as far as the p-GBM are concerned. Most studies indicate that TMZ chemotherapy does not affect survival figures in children, although a recent study has shown otherwise. A trial similar to the Stupp trial involving the pediatric patients did not show any benefit of TMZ in children (61). MGMT promoter methylation, whenever present, potentiates the activity of TMZ even in children. Thus, the ambiguity of the results of these studies has put a question mark on the routine use of chemotherapy in p-GBM vis-à-vis adult GBM, at least as of now. The PCV regimen is still used at many centers, often as a salvage therapy after disease recurrence.

ANTIANGIOGENESIS INHIBITORS

p-GBMs express VEGF similar to the adults. The clinical trials with anti-VEGF therapy (bevacizumab), however, have been rather disappointing in children (62). In a study of 10 patients with supratentorial HGGs and two studies with diffuse intrinsic pontine glioma (DIPG), clinical responses to bevacizumab were inferior to those in adult patients (62). Trials of combining chemotherapeutic agent (irinotecan) with bevacizumab has not improved the outcome either (63). Thalidomide, another antiangiogenic agent, has also failed to prove its efficacy in clinical trials when combined with RT. In particular, this combination led to rather high and unacceptable toxicities (64).

MOLECULAR TARGETED THERAPY

Recent insights into the molecular biology of gliomas in general and pediatric high-grade glioma in particular have led to the development of a number of agents directed specifically against these molecular targets. These include monoclonal antibodies like imatinib (anti-PDGFR) (65); erlotinib; gefitinib (anti-EGFR) (66, 67); and tipifarnib, a farnesyltransferase inhibitor (68). Most of these agents are in Stage I/II trials and have not really lived up to the expectations. One of the primary reasons for lack of expected success could be the fact that a number of tumorigenic pathways act simultaneously in these tumors, thereby negating the effect of blockade of a particular pathway. Similar to the above agents, lobradimil, a bradykinin agonist, was tried with chemotherapy in a Phase II trial involving pediatric high-grade glioma, with a view to enhance the drug permeability of the chemotherapeutic agents. The fate was unfortunately no different (68).

EMERGING NEWER DRUGS AND THERAPEUTIC MODALITIES

A number of other drugs and therapeutic modalities are being tested currently in the laboratories that hold great promise in the days to come. These include integrin inhibitors (cilengitide), EGFR inhibitors (cetuximab, nimotuzumab), novel antiangiogenic agents (enzastaurin, cediranib), histone deacetylase inhibitors (vorinostat, valproic acid), and dendritic cell vaccines. These agents have been tested in children with HGG and have shown good response in the clinical trials. Some agents are in the pre-trial recruitment phase and are likely to enter clinical trials in days to come. These include boron neutron capture therapy, cytomegalovirus-specific cytotoxic T cells, IL-13-PE38QR (an enzymatically active portion of pseudomonas exotoxin A conjugated with human interleukin-13), smoothened inhibitor LDE225, telomerase inhibitors, gamma secretase inhibitors, poly(ADP-ribose) polymerase inhibitor, etc., to name a few. Details of these advances are beyond the scope of this chapter and can be found in other articles (69).

Factors Affecting Outcome

The OS in p-GBM varies from 10 to 73 months (2, 4, 6, 912). Majority of the studies emphasize on improved survival figures in p-GBM compared with the adults (4, 911). There are studies on the long-term outcomes in p-GBM, defined as survival beyond 3 years of diagnosis. Although the factors determining the outcomes are still being studied, a number of factors are already known to predict survival in p-GBM in particular (Table 1). Of all the reported factors, the extent of surgical tumor excision probably remains the strongest predictor of outcome in p-GBM as of today.

TABLE 1. Factors Predicting a Longer Survival in p-GBM.

TABLE 1

Factors Predicting a Longer Survival in p-GBM.

Conclusion

p-GBM is a rare but distinctly different biological disease compared with the adults. Specific sets of genetic aberrations characterize p-GBMs. In the absence of concrete evidence for adjuvant chemotherapy, maximal surgical excision followed by adjuvant RT (in children >3 years of age) remains the current best treatment strategy for these tumors. Prognosis in the majority of children is better than the adults which in turn may be explained by a different biological make up of these tumors. With rapid scientific advances being made in this field, newer targeted molecular and other treatment strategies are likely to emerge and change the future course as well as the prognosis of p-GBM.

Conflict of interest: The authors declare no potential conflicts of interest with respect to research, authorship, and/or publication of this manuscript.

Copyright and permission statement: To the best of our knowledge, the materials included in this chapter do not violate copyright laws. All original sources have been appropriately acknowledged and/or referenced. Where relevant, appropriate permissions have been obtained from the original copyright holder(s).

References

1.
Adamson C, Kanu OO, Mehta AI, Di C, Lin N, Mattox AK, et al. Glioblastoma multiforme: A review of where we have been and where we are going. Expert Opin Investig Drugs. 2009 Aug;18(8):1061–83. [PubMed: 19555299] [CrossRef]
2.
Perkins SM, Rubin JB, Leonard JR, Smyth MD, El Naqa I, Michalski JM, et al. Glioblastoma in children: A single-institution experience. Int J Radiat Oncol Biol Phys. 2011 July;80(4):1117–21. [PubMed: 21220190] [CrossRef]
3.
Sánchez-Herrera F, Castro-Sierra E, Gordillo-Domínguez LF, Vaca-Ruiz MA, Santana-Montero B, Perezpeña-Diazconti M, et al. Glioblastoma multiforme in children: Experience at Hospital Infantil de Mexico Federico Gomez. Childs Nerv Syst. 2010 May;25(5):551–7. [PubMed: 19148652] [CrossRef]
4.
Das KK, Mehrotra A, Nair AP, Kumar S, Srivastava AK, Sahu RN, et al. Pediatric glioblastoma: Clinico-radiological features and factors affecting the outcome. Childs Nerv Syst. 2012 Dec;28(12):2055–62. [PubMed: 22903238] [CrossRef]
5.
Suri V, Das P, Pathak P, Jain A, Sharma MC, Borkar SA, et al. Pediatric glioblastomas: A histopathological and molecular genetic study. Neuro Oncol. 2009 Jun;11(3):274–80. [PMC free article: PMC2718971] [PubMed: 18981259] [CrossRef]
6.
Faury D, Nantel A, Dunn SE, Guiot MC, Haque T, Hauser P, et al. Molecular profiling identifies prognostic subgroups of pediatric glioblastoma and shows increased YB-1 expression in tumors. J Clin Oncol. 2007;25:1196–208. [PubMed: 17401009] [CrossRef]
7.
Dohrmann GJ, Farwell JR, Flannery JT. Astrocytomas in childhood: A population-based study. Surg Neurol. 1985 Jan;23(1):64–8. [PubMed: 3964979] [CrossRef]
8.
Song KS, Phi JH, Cho BK, Wang KC, Lee JY, Kim DG, et al. Long-term outcomes in children with glioblastoma. J Neurosurg Pediatr. 2010 Aug;6(2):145–9. [PubMed: 20672935] [CrossRef]
9.
Nikitović M, Stanić D, Pekmezović T, Gazibara MS, Bokun J, Paripović L. Pediatric glioblastoma: A single institution experience. Childs Nerv Syst. 2016 Jan;32(1):97–103. [PubMed: 26537911] [CrossRef]
10.
Mahvash M, Hugo H-H, Maslehaty H, Mehdorn HM, Stark AM. Glioblastoma multiforme in children: Report of 13 cases and review of the literature. Pediatr Neurol. 2011 Sep;45(3):178–80. [PubMed: 21824566] [CrossRef]
11.
Broniscer A, Gajjar A. Supratentorial high-grade astrocytoma and diffuse brainstem glioma: Two challenges for the pediatric oncologist. Oncologist. 2004;9(2):197–206. [PubMed: 15047924] [CrossRef]
12.
Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005 Mar;352(10):987–96. [PubMed: 15758009] [CrossRef]
13.
Adams H, Adams HH, Jackson C, Rincon-Torroella J, Jallo GI, Quiñones-Hinojosa A. Evaluating extent of resection in pediatric glioblastoma: A multiple propensity score-adjusted population-basedanalysis. Childs Nerv Syst. 2016 Mar;32(3):493–503. [PubMed: 26767842] [CrossRef]
14.
Packer J. Diagnosis and treatment of pediatric brain tumors. Curr Opin Neurol. 1994 Dec;7(6):484–91. [PubMed: 7866579] [CrossRef]
15.
Staneczek W, Jänisch W. Epidemiology of primary tumors of the central nervous system in children and adolescents. A population-based study. Pathologe. 1994 Aug;15(4):207–15. [PubMed: 7971795] [CrossRef]
16.
CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2004–2008. Hinsdale, IL: CBTRUS; 2012. http://www​.cbtrus.org​/2012-NPCR-SEER/CBTRUS​_Report_2004-2008_3-23-2012.
17.
Broniscer A. Past, present, and future strategies in the treatment of high-grade glioma in children. Cancer Invest. 2006 Feb;24(1):77–81. [PubMed: 16466996] [CrossRef]
18.
Finlay JL, Zacharoulis S. The treatment of high grade gliomas and diffuse intrinsic pontine tumors of childhood and adolescence: A historical—And futuristic—Perspective. J. Neurooncol. 2005 Dec;75(3):253–66. [PubMed: 16195805] [CrossRef]
19.
Artico M, Cervoni L, Celli P, Salvati M, Palma L. Supra-tentorial glioblastoma in children: A series of 27 surgically treated cases. Childs Nerv Syst. 1993 Feb;9(1):7–9. [PubMed: 8481945] [CrossRef]
20.
Milano GM, Cerri C, Ferruzzi V, Capolsini I, Mastrodicasa E, Genitori LL, et al. Congenital glioblastoma. Pediatr. Blood Cancer. 2009 Jul;53(1):124–6. [PubMed: 19309717] [CrossRef]
21.
Wolff B, Ng A, Roth D, Parthey K, Warmuth-Metz M, Eyrich M, et al. Pediatric high grade glioma of the spinal cord: Results of the HIT- GBM database. J. Neurooncol. 2012 Mar;107(1):139–46. [PubMed: 21964697] [CrossRef]
22.
Fangusaro J. Pediatric high grade glioma: A review and update on tumor clinical characteristics and biology. Front Oncol. 2012 Aug;2:105. [PMC free article: PMC3426754] [PubMed: 22937526] [CrossRef]
23.
Sgouros S, Fineron PW, Hockley AD. Cerebellar astrocytoma of childhood: Long-term follow-up. Childs Nerv Syst. 1995 Feb;11(2):89–96. [PubMed: 7758018] [CrossRef]
24.
Epstein F, Wisoff J. Intra-axial tumors of the cervicomedullary junction. J Neurosurg. 1987 Oct;67(4):483–87. [PubMed: 3309202] [CrossRef]
25.
Fangusaro J. Pediatric high- grade gliomas and diffuse intrinsic pontine gliomas. J Child Neurol. 2009 Nov;24(11):1409–17. [PubMed: 19638636] [CrossRef]
26.
Neglia JP, Robison LL, Stovall M, Liu Y, Packer RJ, Hammond S, et al. New primary neoplasms of the central nervous system in survivors of childhood cancer: A report from the Childhood Cancer Survivor Study. J Natl Cancer Inst. 2006 Nov;98(21):1528–37. [PubMed: 17077355] [CrossRef]
27.
Walter AW, Hancock ML, Pui CH, Hudson MM, Ochs JS, Rivera GK, et al. Secondary brain tumors in children treated for acute lymphoblastic leukemia at St Jude Children’s Research Hospital. J Clin Oncol. 1998 Dec;16(12):3761–7. [PubMed: 9850019] [CrossRef]
28.
Malik N, Kumar R, Prasad KN, Kawal P, Srivastava A, Mahapatra AK. Association of matrix metalloproteinase-1 gene polymorphism with glioblastoma multiforme in a northern Indian population. J Neurooncol. 2011 May;102(3):347–52. [PubMed: 20706772] [CrossRef]
29.
Kumar R, Malik N, Tungaria A, Kawal P. Matrix metalloproteinase-2 gene polymorphism is not associated with increased glioblastoma multiforme susceptibility: An Indian institutional experience. Neurol India. 2011 Mar–Apr;59(2):236–40. [PubMed: 21483125] [CrossRef]
30.
Wagner S, Benesch M, Berthold F, Gnekow AK, Rutkowski S, Sträter R, et al. Secondary dissemination in children with high-grade malignant gliomas and diffuse intrinsic pontine gliomas. Br J Cancer. 2006 Oct;95(8):991–7. [PMC free article: PMC2360717] [PubMed: 17047647] [CrossRef]
31.
Singh G, Mehrotra A, Sardhara J, Das KK, Jamdar J, Pal L, et al. Multiple glioblastomas: Are they different from their solitary counterparts? Asian J Neurosurg. 2015 Oct–Dec;10(4):266–71. [PMC free article: PMC4558800] [PubMed: 26425153] [CrossRef]
32.
Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. The 2016 World Health Organization classification of tumors of the central nervous system: A summary. Acta Neuropathol. 2016 Jun;131(6):803–20. [PubMed: 27157931] [CrossRef]
33.
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 Feb;360(8):765–73. [PMC free article: PMC2820383] [PubMed: 19228619] [CrossRef]
34.
Pollack IF, Hamilton RL, Sobol RW, Nikiforova MN, Lyons-Weiler MA, LaFramboise WA, et al. IDH1 mutations are common in malignant gliomas arising in adolescents: A report from the Children’s Oncology Group. Childs Nerv Syst. 2011 Jan;27(1):87–94. [PMC free article: PMC3014378] [PubMed: 20725730] [CrossRef]
35.
Pollack IF, Finkelstein SD, Woods J, Burnham J, Holmes EJ, Hamilton RL, et al. Expression of p53 and prognosis in children with malignant gliomas. N Engl J Med. 2002 Feb;346(6):420–7. [PubMed: 11832530] [CrossRef]
36.
Pollack IF, Hamilton RL, Burger PC, Brat DJ, Rosenblum MK, Murdoch GH, et al. Akt activation is a common event in pediatric malignant gliomas and a potential adverse prognostic marker: A report from the Children’s Oncology Group. J Neurooncol. 2010 Sep;99(2):155–63. [PMC free article: PMC2954435] [PubMed: 20607350] [CrossRef]
37.
Heaphy CM, de Wilde RF, Jiao Y, Klein AP, Edil BH, Shi C, et al. Altered telomeres in tumors with ATRX and DAXX mutations. Science. 2011 Jul;333(6041):425. [PMC free article: PMC3174141] [PubMed: 21719641] [CrossRef]
38.
Simon JA, Kingston RE. Mechanisms of polycomb gene silencing: Knowns and unknowns. Nat Rev Mol Cell Biol. 2009 Oct;10(10):697–708. [PubMed: 19738629] [CrossRef]
39.
Wu G, Broniscer A, McEachron TA, Lu C, Paugh BS, Becksfort J, et al. Somatic histone H3 alterations in pediatric diffuse intrinsic pontine gliomas and non-brainstem glioblastomas. Nat Genet. 2012 Jan;44(3):251–3. [PMC free article: PMC3288377] [PubMed: 22286216] [CrossRef]
40.
Lai A, Filka E, McGibbon B, Nghiemphu PL, Graham C, Yong WH, et al. Phase II pilot study of bevacizumab in combination with temozolomide and regional radiation therapy for up-front treatment of patients with newly diagnosed glioblastoma multiforme: Interim analysis of safety and tolerability. Int J Radiat Oncol Biol Phys. 2008 Aug;71(5):1372–80. [PubMed: 18355978] [CrossRef]
41.
Paugh BS, Zhu X, Qu C, Endersby R, Diaz AK, Zhang J, et al. Novel oncogenic PDGFRA mutations in pediatric high-grade gliomas. Cancer Res. 2013 Oct;73(20):6219–29. [PMC free article: PMC3800209] [PubMed: 23970477] [CrossRef]
42.
Raymond E, Brandes AA, Dittrich C, Fumoleau P, Coudert B, Clement PM, et al. Phase II study of imatinib in patients with recurrent gliomas of various histologies: A European Organisation for Research and Treatment of Cancer Brain Tumor Group study. J Clin Oncol. 2008 Oct;26(28):4659–65. [PMC free article: PMC2653126] [PubMed: 18824712] [CrossRef]
43.
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 Jun;59(6):539–43. [PubMed: 10850866] [CrossRef]
44.
Lee JY, Park CK, Park SH, Wang KC, Cho BK, Kim SK. MGMT promoter gene methylation in pediatric glioblastoma: Analysis using MS-MLPA. Childs Nerv Syst. 2011 Nov;27(11):1877–83. [PubMed: 21789683] [CrossRef]
45.
Cervoni L, Celli P, Salvati M, Palma L. Supratentorial glioblastoma in children: A series of 27 surgically treated cases. Childs Nerv Syst. 1993 Feb;9(1):7–9. [PubMed: 8481945] [CrossRef]
46.
Chaichana K, Parker S, Olivi A, Quiñones-Hinojosa A. A proposed classification system that projects outcomes based on preoperative variables for adult patients with glioblastoma multiforme. J Neurosurg. 2010 May;112(5):997–1004. [PubMed: 19817542] [CrossRef]
47.
Huang RY, Neagu MR, Reardon DA, Wen PY. Pitfalls in the neuroimaging of glioblastoma in the era of antiangiogenic and immuno/targeted therapy—Detecting illusive disease, defining response. Front Neurol. 2015 Feb;6:33. [PMC free article: PMC4337341] [PubMed: 25755649] [CrossRef]
48.
Wen PY, Macdonald DR, Reardon DA, Cloughesy TF, Sorensen AG, Galanis E, et al. Updated response assessment criteria for high-grade gliomas: Response assessment in neuro-oncology working group. J Clin Oncol. 2010 Apr;28(11):1963–72. [PubMed: 20231676] [CrossRef]
49.
Wangaryattawanich P, Hatami M, Wang J, Thomas G, Flanders A, Kirby J, et al. Multicenter imaging outcomes study of The Cancer Genome Atlas glioblastoma patient cohort: Imaging predictors of overall and progression-free survival. Neuro Oncol. 2015 Nov;17(11):1525–37. [PMC free article: PMC4648306] [PubMed: 26203066] [CrossRef]
50.
Finlay JL, Boyett JM, Yates AJ, Wisoff JH, Milstein JM, Geyer JR, et al. Randomized phase III trial in childhood high-grade astrocytoma comparing vincristine, lomustine, and prednisone with the eight-drugs-in-1-day regimen. Childrens Cancer Group. J Clin Oncol. 1995 Jan;13(1):112–23. [PubMed: 7799011] [CrossRef]
51.
Aibaidula A, Zhao W, Wu JS, Chen H, Shi ZF, Zheng LL, et al. Microfluidics for rapid detection of isocitrate dehydrogenase 1 mutation for intraoperative application. J Neurosurg. 2016 Jun;124(6):1611–18. [PubMed: 26544771] [CrossRef]
52.
Mallick S, Gandhi AK, Joshi NP, Kumar A, Puri T, Sharma DN, et al. Outcomes of pediatric glioblastoma treated with adjuvant chemoradiation with temozolomide and correlation with prognostic factors. Indian J Med Paediatr Oncol. 2015 Apr–Jun;36(2):99–104. [PMC free article: PMC4477385] [PubMed: 26157286] [CrossRef]
53.
Fallai C, Olmi P. Hyper-fractionated and accelerated radiation therapy in central nervous system tumors (malignant gliomas, pediatric tumors, and brain metas-tases). Radiother. Oncol. 1997 Jun;43(3):235–46. [PubMed: 9215782] [CrossRef]
54.
Geyer JR, Finlay JL, Boyett JM, Wisoff J, Yates A, Mao L, et al. Survival of infants with malignant astrocytomas. A Report from the Childrens Cancer Group. Cancer. 1995 Feb;75(4):1045–50. [PubMed: 7842407] [CrossRef]
55.
Sanders RP, Kocak M, Burger PC, Merchant TE, Gajjar A, Broniscer A. High-grade astrocytoma in very young children. Pediatr Blood Cancer. 2007 Dec;49(7):888–93. [PubMed: 17554787]
56.
Knab B, Connell PP. Radiotherapy for pediatric brain tumors: When and how. Expert Rev Anticancer Ther. 2007 Dec;7 12 Suppl:69–77. [PubMed: 18076312] [CrossRef]
57.
Sposto R, Ertel IJ, Jenkin RD, Boesel CP, Venes JL, Ortega JA, et al. The effectiveness of chemotherapy for treatment of high grade astrocytoma in children: Results of a randomized trial. A report from the Children’s Cancer Study Group. J Neurooncol. 1989 Jul;7(2):165–77. [PubMed: 2550594] [CrossRef]
58.
Wolff JE, Driever PH, Erdlenbruch B, Kortmann RD, Rutkowski S, Pietsch T, et al. Intensive chemotherapy improves survival in pediatric high-grade glioma after gross total resection: Results of the HIT-GBM-C protocol. Cancer. 2010 Feb;116(3):705–12. [PubMed: 19957326] [CrossRef]
59.
Finlay JL, Dhall G, Boyett JM, Dunkel IJ, Gardner SL, Goldman S, et al. Myeloablative chemotherapy with autologous bone marrow rescue in children and adolescents with recurrent malignant astrocytoma: Outcome compared with conventional chemotherapy: A report from the Children’s Oncology Group. Pediatr Blood Cancer. 2008 Dec;51(6):806–11. [PMC free article: PMC2844080] [PubMed: 18802947] [CrossRef]
60.
Cohen KJ, Pollack IF, Zhou T, Buxton A, Holmes EJ, Burger PC, et al. Temozolomide in the treatment of high-grade gliomas in children: A report from the Children’s Oncology Group. Neuro Oncol. 2011 Mar;13(3):317–23. [PMC free article: PMC3064602] [PubMed: 21339192] [CrossRef]
61.
Narayana A, Kunnakkat S, Chacko-Mathew J, Gardner S, Karajannis M, Raza S, et al. Bevacizumab in recurrent high-grade pediatric gliomas. Neuro Oncol. 2010 Sep;12(9):985–90. [PMC free article: PMC2940690] [PubMed: 20363768] [CrossRef]
62.
Gururangan S, Chi SN, Young Poussaint T, Onar-Thomas A, Gilbertson RJ, Vajapeyam S, et al. Lack of efficacy of bevacizumab plus irinotecan in children with recurrent malignant glioma and diffuse brainstem glioma: A Pediatric Brain Tumor Consortium study. J Clin Oncol. 2010 Jun;28(18):3069–75. [PMC free article: PMC2903337] [PubMed: 20479404] [CrossRef]
63.
Turner CD, Chi S, Marcus KJ, MacDonald T, Packer RJ, Poussaint TY, et al. Phase II study of thalidomide and radiation in children with newly diagnosed brain stem gliomas and glioblastoma multiforme. J Neurooncol. 2007 Mar;82(1):95–101. [PubMed: 17031553] [CrossRef]
64.
Pollack IF, Jakacki RI, Blaney SM, Hancock ML, Kieran MW, Phillips P, et al. Phase I trial of imatinib in children with newly diagnosed brainstem and recurrent malignant gliomas: A Pediatric Brain Tumor Consortium report. Neuro Oncol. 2007;9:145–60. [PMC free article: PMC1871662] [PubMed: 17293590] [CrossRef]
65.
Broniscer A, Baker SJ, Stewart CF, Merchant TE, Laningham FH, Schaiquevich P, et al. Phase I and pharmacokinetic studies of erlotinib administered concurrently with radiotherapy for children, adolescents, and young adults with high-grade glioma. Clin Cancer Res. 2009 Jan;15(2):701–7. [PMC free article: PMC2629527] [PubMed: 19147777] [CrossRef]
66.
Geyer JR, Stewart CF, Kocak M, Broniscer A, Phillips P, Douglas JG, et al. A phase I and biology study of gefitinib and radiation in children with newly diagnosed brain stem gliomas or supratentorial malignant gliomas. Eur J Cancer. 2010 Dec;46(18):3287–93. [PMC free article: PMC2988095] [PubMed: 20708924] [CrossRef]
67.
Fouladi M, Nicholson HS, Zhou T, Laningham F, Helton KJ, Holmes E, et al. A phase II study of the farnesyl transferase inhibitor, tipifarnib, in children with recurrent or progressive high-grade glioma, medulloblastoma/primitive neuroectodermal tumor, or brainstem glioma: A Children’s Oncology Group study. Cancer. 2007 Dec;110(11):2535–41. [PubMed: 17932894] [CrossRef]
68.
Warren K, Jakacki R, Widemann B, Aikin A, Libucha M, Packer R, et al. Phase II trial of intravenous lobradimil and carboplatin in childhood brain tumors: A report from the Children’s Oncology Group. Cancer Chemother Pharmacol. 2006 Sep;58(3):343–47. [PubMed: 16408203] [CrossRef]
69.
MacDonald TJ, Aguilera D, Kramm CM. Treatment of high-grade glioma in children and adolescents. Neuro Oncol. 2011 Oct;13(10):1049–58. [PMC free article: PMC3177659] [PubMed: 21784756] [CrossRef]
Copyright: The Authors.

Licence: This open access article is licenced under Creative Commons Attribution 4.0 International (CC BY 4.0). https://creativecommons.org/licenses/by-nc/4.0/

Bookshelf ID: NBK469983PMID: 29251872DOI: 10.15586/codon.glioblastoma.2017.ch15

Views

Related Items in Bookshelf

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...