• Open Access
    Review

    Update for astrocytomas: medical and surgical management considerations

    Matthew Willman 1
    Jonathan Willman 1
    John Figg 1
    Emma Dioso 2
    Sai Sriram 1
    Bankole Olowofela 1
    Kevin Chacko 1
    Jairo Hernandez 1
    Brandon Lucke-Wold 3*

    Explor Neurosci. 2023;2:1–26 DOI: https://doi.org/10.37349/en.2023.00009

    Received: September 06, 2022 Accepted: December 10, 2022 Published: February 23, 2023

    Academic Editor: Dirk M. Hermann, University of Duisburg-Essen, Germany

    This article belongs to the special issue Emerging Trends in Endovascular Management

    Abstract

    Astrocytomas include a wide range of tumors with unique mutations and varying grades of malignancy. These tumors all originate from the astrocyte, a star-shaped glial cell that plays a major role in supporting functions of the central nervous system (CNS), including blood-brain barrier (BBB) development and maintenance, water and ion regulation, influencing neuronal synaptogenesis, and stimulating the immunological response. In terms of epidemiology, glioblastoma (GB), the most common and malignant astrocytoma, generally occur with higher rates in Australia, Western Europe, and Canada, with the lowest rates in Southeast Asia. Additionally, significantly higher rates of GB are observed in males and non-Hispanic whites. It has been suggested that higher levels of testosterone observed in biological males may account for the increased rates of GB. Hereditary syndromes such as Cowden, Lynch, Turcot, Li-Fraumeni, and neurofibromatosis type 1 have been linked to increased rates of astrocytoma development. While there are a number of specific gene mutations that may influence malignancy or be targeted in astrocytoma treatment, O6-methylguanine-DNA methyltransferase (MGMT) gene function is an important predictor of astrocytoma response to chemotherapeutic agent temozolomide (TMZ). TMZ for primary and bevacizumab in the setting of recurrent tumor formation are two of the main chemotherapeutic agents currently approved in the treatment of astrocytomas. While stereotactic radiosurgery (SRS) has debatable implications for increased survival in comparison to whole-brain radiotherapy (WBRT), SRS demonstrates increased precision with reduced radiation toxicity. When considering surgical resection of astrocytoma, the extent of resection (EoR) is taken into consideration. Subtotal resection (STR) spares the margins of the T1 enhanced magnetic resonance imaging (MRI) region, gross total resection (GTR) includes the margins, and supramaximal resection (SMR) extends beyond the margin of the T1 and into the T2 region. Surgical resection, radiation, and chemotherapy are integral components of astrocytoma treatment.

    Hereditary risk factors, genetic mutations, and imaging modalities are discussed in reference to astrocytoma staging and mechanism of growth. In terms of the treatment of astrocytomas, chemotherapy, radiation therapy, and strategic surgical interventions are discussed

    Keywords

    Astrocytoma, surgical resection, chemotherapy, radiation, glioblastoma, diagnostic imaging

    Introduction

    As astrocytes are the most plentiful cell in the human central nervous system (CNS), it is not entirely surprising that astrocytomas are the most common form of primary malignant brain tumors in both adults and children [13]. Lacking the ability to produce electrical depolarization, astrocytes are often described as star-shaped glial cells that support the function of neurons [4]. Astrocytes have a plethora of roles in the CNS, including blood-brain barrier (BBB) development and maintenance, water and ion regulation, influencing neuronal synaptogenesis, and stimulating immunological response [4, 5]. The exact function and morphology of the astrocyte may be primarily determined by the spatial domain the cell occupies within the brain and the surrounding population of astrocytes [6, 7].

    The clinical presentation of astrocytoma symptoms is similar to most brain tumors with both focal and generalized neurologic signs. Focal symptoms include weakness, numbness, seizures, and aphasia, while generalized symptoms due to edema and increased intracranial pressure can cause headaches, vision deficits, and emesis [8, 9]. Diffuse headaches present at diagnosis in 50% of patients, with seizures observed in 20–40% of patients [10].

    Radiation, chemotherapy, and surgical resection play a role in the medical management of both low- and high-grade astrocytomas to alleviate symptoms, improve prognosis, and prevent malignant transformation [11, 12]. By summarizing the current understanding of astrocytomas, this paper strives to consolidate and clearly explain the accepted medical and surgical management of this condition to better educate the reader.

    Epidemiology

    Glioblastoma (GB) is both the most common astrocytoma and the most malignant, accounting for roughly 65% of astrocytomas [8, 13]. Based on 2003–2007 data gathered from region-specific populations over 40 years old, the age-adjusted incidence rate of astrocytic tumors per 100,000 is highest in Australia [9.58, 95% confidence interval (CI) = 9.30–9.86], Western Europe (8.45, 95% CI = 8.30–8.59), and Canada (8.26, 95% CI = 8.04–8.48), with lowest rates in Southeast Asia (1.91, 95% CI = 1.75–2.08) [14]. These trends in astrocytoma incidence based on the region are summarized in Figure 1.

    The age-adjusted incidence rates of astrocytomas per 100,000 in adult populations (> 40 years old) around the world. The highest rates are observed in Australia (9.58/100,000) and the lowest are observed in Southeast Asia (1.91/100,000) [14]

    When examining 244,808 patients with gliomas from 2000–2014, there was a higher incidence of astrocytomas in non-Hispanic whites compared to Hispanic whites (30% lower), blacks (52% lower), or Asians (52% lower) [15]. Overall, epidemiological studies have indicated an increased incidence of astrocytomas in males compared to female populations [16]. This sex preference is especially prominent in the development of GB, with an incidence 60% higher in both male children and adults [1517]. Elevated levels of testosterone and activation of androgen receptors on cancer cells are believed to play significant roles in the increased incidence of male GB, while the protective benefit of progesterone and estrogen has also been theorized [1820]. With the exception of low-grade pilocytic astrocytomas (PAs) that typically present in pediatric populations, the majority of astrocytomas are observed sporadically in adulthood after the age of 40 without a familial association [14, 21, 22]. While the mortality rate varies with different grades of astrocytomas and mutations present, GB has a median survival of 15 months with treatment and a 3-year survival rate of 3–5% [23]. There are numerous factors associated with improved overall survival rate, including female sex, younger age of diagnosis, lower comorbidity burden, presence of O6-methylguanine-DNA methyltransferase (MGMT) methylation in tumor genetics, reduced tumor size, single lesion, complete tumor resection, chemotherapy, and radiation treatment [24]. Although it could be explained by the patient population and socioeconomic elements, there appears to be a trend towards superior 30–90 day mortality and reduced readmission rates in academic research and high-volume facilities compared to community and integrated network cancer programs [24].

    Astrocytoma hereditary and environmental risk factors

    Therapeutic ionizing radiation exposure is one of the few environmental risk factors with a strong correlation with increased incidence of astrocytomas and other glioma formation [25]. Surprisingly, cigarette smoking and alcohol consumption have not been associated with higher rates of astrocytomas as observed in many other forms of cancer [26]. Although the risk factors of astrocytoma are poorly understood, germline genetic variants contribute significantly to the risk of developing gliomas like astrocytomas. For example, some genetic syndromes [Cowden, Lynch, Turcot, Li-Fraumeni, neurofibromatosis type 1 (NF1)] have been identified as potential risk factors. Conditional inactivation of NF1, phosphatase and tensin homolog (PTEN), and tumor protein p53 (TP53) tumor suppressor genes in murine models has induced the formation of malignant astrocytomas [27, 28]. Carlos-Escalante et al. [29] found an association between genetic variants in angiotensinogen (AGT, involved in angiogenesis), TP53 (tumor suppressor), and MGMT (DNA repair) and the risk of developing astrocytoma in a case-control study performed in 49 Mexican patients.

    Another genetic association involves B-Raf proto-oncogene (BRAF) abnormalities, in which tandem duplication-rearrangements and BRAF V600E mutations are observed in PA and pleomorphic xanthoastrocytoma, respectively [30]. The most common glial tumor in children is PA. While sporadic cases are associated with BRAF rearrangements, 15–20% of children develop PA in the context of NF1 [31]. The most common tumor in NF1 patients is the optic pathway tumor, also known as optic glioma. Although these are rare tumors (2–5% of childhood brain tumors), up to 70% are associated with NF1 [32]. Most of these tumors are grade I PAs. These tumors form within the optic pathway and brainstem to then expand into the optic nerve, producing a fusiform mass [27, 33, 34].

    Certain genetic markers have been associated with worse prognosis. Perdomo-Pantoja et al. [35] conducted a prospective study on 48 patients diagnosed with astrocytoma and proposed that the genetic variant AGT reference single nucleotide polymorphism 5050 (rs5050) GG-genotype can be detected safely and practically in blood/saliva samples and was identified as a risk factor for lower survival rates. Other genes [ST6 beta-galactoside alpha-2,6-sialyltransferase 1 (ST6GAL1), cytochrome P450 family 19 subfamily A member 1 (CYP19A1)] have also shown an association with worse prognosis [36].

    Astrocytoma genetics and mechanisms of growth

    As astrocytomas, like all cancers, are the unregulated overgrowth of cells brought about by changes in genetic expression, genetics play a significant role in astrocytoma advent, evolution, and treatment [37]. While some cancer states are strongly linked to specific genes, such as Li-Fraumeni with TP53 and breast and ovarian cancers with breast cancer 1 (BRCA1)/BRCA2, other cancers, such as astrocytomas rest at the intersection of a combination of genes [38, 39]. Significant research on the genetics of astrocytomas focuses on isocitrate dehydrogenase 1 (IDH1) and MGMT.

    IDH1 and IDH2 are genes that encode nicotinamide adenine dinucleotide phosphate (NADP+)-dependent IDHs which convert isocitrate to alpha-ketoglutarate [40, 41]. This activity plays a role in cellular processes affecting lipid synthesis, oxidative respiration, and signal transduction [42, 43]. The R132H mutation in IDH1 can lead to the transformation of alpha-ketoglutarate into 2-hydroxygluterate (2HG) [44]. Aberrations in IDH1/2 as well as the accumulation of 2HG have been linked to a multitude of cancers, including astrocytomas like GB [42, 44, 45]. While their presence is not ubiquitous in primary GB (5%), mutations in IDH1 are prevalent in secondary GB (80%) [46].

    Extensive exploration into the link between IDH1 and gliomas has led to a deeper understanding of the influence of IDH1 on current treatments. Investigation of this relationship has also opened the door for targeted therapeutic interventions. Mutation of IDH1 has proven to be a strong prognostic indicator in gliomas [45, 46]. Studies have shown better responses to treatment and improved outcomes in patients with IDH1 mutated GB and anaplastic astrocytoma (AA) [46, 47]. Therapies targeting IDH1-specific mutations including hypomethylating agents, IDH enzyme inhibition, and vaccination are being investigated with promising early findings [42].

    MGMT is another gene that’s been shown to influence patient response to treatment in astrocytoma. MGMT encodes a protein that repairs alkylated DNA via the transfer of a methyl group [48]. Temozolomide (TMZ) is an alkylating agent that has been a stalwart therapy in the treatment of astrocytoma [49]. As such, functional MGMT repairs damage caused by the alkylation of DNA by drugs like TMZ and can lead to drug resistance in tumors [50]. While the loss of function of MGMT via methylation is associated with different cancers, it is also associated with improved patient outcomes in the treatment of astrocytomas [48, 51, 52]. Studies have shown a significant association between methylated MGMT and longer survival as well as decreased tumor size over the course of treatment [51, 52]. Of note, there is no current consensus on the amount of methylation that distinguishes methylated from unmethylated MGMT. The mechanism of the improved outcome of TMZ treatment in MGMT-inhibited tumors is summarized in Figure 2 [53].

    In the setting of dysfunctional MGMT DNA repair, TMZ has an improved capacity to promote astrocytoma cellular injury

    Note. This figure contains (modified) images from Servier Medical Art (https://smart.servier.com). CC BY.

    While the BBB normally restricts immune cell access to the brain, malignancy in the brain often creates a privileged environment where leukocytes are granted access [54]. Despite that, often times immune response in brain tumors is limited by patient immunodeficiency or immunological escape of the cancer [54]. Immunological escape is a phenomenon whereby tumors elude immune action [55]. There are a few forms of immune escape exhibited by astrocytomas, including the restriction of antigen presentation via the downregulation of major histocompatibility complex (MHC) expression [56]. Other methods include the facilitation of cell death in leukocytes by increasing Fas-ligand expression, and secretion of immunosuppressive cytokines like transforming growth factor-β (TGF-β), and interleukin-10 (IL-10) [57, 58]. As immunological escape and immune suppression increase the difficulty of treating astrocytomas, genetically modified immune cells and augmentation of the immune response are of particular interest in the treatment of astrocytomas [59]. Of note, these methods may run the risk of autoimmunity.

    When examining GB mutation distribution based on sex, there appears to be a trend toward higher subclonal mutation burden in the majority of glioma subtypes of biological females over males [60]. Additionally, some mutations observed in astrocytomas appear to express some level of male or female preference. For example, mutations in the signaling pathway of mitogen-activated protein kinase (MAPK) tend to occur more often in the GB of biological female patients [60]. In the case of gliomas with mutations of the phosphoinositide 3-kinase (PI3K) pathway, these genetic changes occur more frequently in the helical domain for females and the kinase domain for males, demonstrating a sex preference for mutation location [61]. The increased mutation burden could be explained by increased mutations on the X chromosome, or potentially by different environmental exposures that may be more sex-specific.

    As genetics play a significant role in the development and progression of astrocytomas, further investigation into the influence of specific genes may illuminate a pathway to better understand and more effectively treat the disease.

    Grading astrocytomas

    The grading of astrocytomas is based on histological observations and cellular markers [62]. The following presents a discussion on some of the most common locations for each lesion type and prognosis based on tumor grading. The grading type and location are summarized in Table 1.

    While different types of astrocytomas can be found throughout the CNS, there are regions of the brain where specific astrocytomas tend to present more commonly

    GradeAstrocytoma typeCommon location
    1PACerebellum
    Subependymal giant cell astrocytoma (SEGA)Walls of the lateral ventricles or foramen of Monro
    2Diffuse astrocytomaFrontal, parietal, or temporal lobes
    Pleomorphic xanthoastrocytomaTemporal lobes
    3AAFrontal, parietal, or temporal lobes
    4GBFrontal, parietal, or temporal lobes
    Display full size

    Grade 1

    Grade 1 astrocytomas are slow-growing lesions, allowing these tumors to grow quite large without inducing symptoms as the brain gradually adapts to the tumor growth [63]. Grade 1 tumors include PA, which is the most common primary CNS tumor in children aged 5–14 [64]. Though commonly localized to the cerebellum, PAs can also be found in the brainstem, hypothalamus, spinal cord, and optic chiasm, with the latter having the least ideal outcome [6567]. With a 10-year survival over 90%, pediatric PAs are amenable to surgery and hold an excellent prognosis if completely excised [68]. PA in adults is not fully understood, though recurrence is still substantially less prevalent in those with complete resection than those without (4% vs. 38%) [69]. SEGAs are rarer, slow-growing pediatric astrocytomas that are typically localized in the walls of the lateral ventricle or foramen of Monro [70]. Similar to PAs, SEGAs also carry a favorable prognosis and are generally considered curative if complete resection is achieved [71]. Finally, though diffuse astrocytomas are typically considered grade 2, the recent fifth edition of World Health Organization (WHO) classifications has updated those pediatric diffuse astrocytomas with alterations in proto-oncogenes myeloblastosis (MYB) and MYB proto-oncogene like 1 (MYBL1) to grade 1, likely stemming from their improved prognosis (10-year overall survival of 95% with MYB/MYBL1 alterations compared to 38% otherwise) [7275].

    Grade 2

    Diffuse grade 2 astrocytomas present a much greater challenge for clinicians as tumor borders are no longer well-delineated, complicating resection and increasing the likelihood of recurrence [76]. Though prognostication can be complicated by the inability of histopathological features to accurately predict the disease course, clinical features such as the preoperative performance and extent of resection (EoR) may better inform physicians [77]. Nevertheless, some novel analyses focusing on oxidative enzymes and their products have established new methods of generating a prognosis based on histopathological evidence [78]. IDH mutations, for example, are thought to have a protective effect and display improved survival in glioma [79, 80]. Interestingly, in some instances mutant IDH grade 2 astrocytomas have displayed a 21% reduction in 5-year progression-free survival (PFS) but an improved survival following recurrence when compared to wild-type IDH tumors [81]. Nevertheless, evidence points to an overall limited prognostic value of IDH1 mutations in grade 2 astrocytomas [81, 82]. Overall, grade 2 astrocytomas display markedly poorer outcomes (65% 2-year survival) when compared to grade 1 tumors (98% 2-year survival) [83]. Diffuse astrocytomas are localized to the cerebrum in the frontal, parietal, or temporal lobes in 60% of cases [83]. Pleomorphic xanthoastrocytoma is another tumor with the potential to be grade 2 or 3 depending on the degree of anaplastic changes present. Presenting most frequently in the temporal lobes, grade 2 pleomorphic xanthoastrocytomas carry a favorable 3-year overall survival of 84% [84, 85].

    Grades 3 and 4

    Similar to grade 2 astrocytomas, grades 3 and 4 are largely localized to the supratentorial region, with 62% (grade 3) and 68% (grade 4) found in one of the frontal, parietal, or temporal lobes [83]. Despite this similarity, these tumors display significantly higher rates of metastasis and progression when compared to their low-grade counterparts. AA is a grade 3 astrocytoma and around half of the individuals with AA display progression at 3 years [86]. For grade 4 astrocytomas (GB), 2-year PFS is an abysmal 10% [87]. With this comes an especially poor 2-year survival prognosis of 50% for grade 3 and 19% for grade 4 tumors [83]. IDH mutations in high-grade astrocytomas are extremely relevant to prognosis, with wild-type IDH AAs typically displaying worse prognosis than IDH-mutated GB [47, 88]. Taken together, these findings communicate the diverse set of pathologies that grade 1 through 4 astrocytomas represent with their resulting prognosis.

    When evaluating depth within the brain, tumor location can impact prognosis. Especially in elderly populations, the closer the margin of a high-grade glioma is to the 3rd ventricle, generally the worse the prognosis [89, 90]. Specifically, periventricular and ventricular high-grade gliomas were associated with the worst outcome in elderly patients, when compared to more superficial tumors [89].

    It is important to note that the new 2021 WHO fifth edition classification of gliomas differs significantly from the previous 2016 fourth edition standards. Under the new system, diffuse gliomas are subcategorized into pediatric-type diffuse low-grade gliomas, pediatric-type diffuse high-grade gliomas, adult-type diffuse gliomas, and circumscribed astrocytic gliomas [91].

    Adult-type diffuse gliomas are further subcategorized into astrocytoma IDH-mutant (grade 2–4), oligodendroglioma IDH-mutant and 1p/19q-codeleted (grade 2–3), and GB IDH-wild-type (grade 4) [75, 91].

    Astrocytoma (IDH-mutant), which includes the now obsolete categories of GB (IDH-mutant), diffuse astrocytoma, and AA, can be classified as a grade 2, 3, or 4 tumor [75, 92]. Grading within this tumor type is based on both genetics and histology with higher proliferative tumors receiving a grade of 3 and more ominous findings such as microvascular expansion or necrosis receiving a grade of 4 [75, 92]. Additionally, cyclin dependent kinase inhibitor 2A (CDKN2A)/CDKN2B homozygous deletion tumors receive a default grade of 4 regardless of histological findings [75, 92]. Of note, astrocytoma (IDH-mutant) often includes a TP53, alpha-thalassemia/mental retardation, X-linked (ATRX), or CDKN2A/B mutation, but must not include a 1p/19q codeletion [75, 91].

    As the name implies, oligodendroglioma IDH-mutant and 1p/19q-codeleted must include both an IDH mutation and 1p/19q-codeletion [75]. It can be a grade 2 or 3 tumor based on histology and image findings [75, 92]. Common co-mutations include a telomerase reverse transcriptase (TERT) promoter mutation and a notch receptor 1 (NOTCH1) mutation [75, 92].

    GB IDH-wild-type is by default a grade 4 tumor [75, 92]. Defining features of this diffuse glioma tumor type are the presence of IDH-wild-type and the histological finding of necrosis or microvascular expansion or one of three key genetic features—epidermal growth factor receptor (EGFR) amplification, +7/−10 chromosome copy number changes, or a TERT promoter mutation [75, 91, 92].

    Under this new system, a number of tumor types are now obsolete including GB IDH-mutant, AA, diffuse astrocytoma, anaplastic pleomorphic xanthoastrocytoma, and anaplastic oligodendroglioma [75].

    Diagnostic imaging

    Magnetic resonance imaging (MRI) maintains a significant role in the standard of care among patients diagnosed with astrocytomas, especially as MRI has evolved to incorporate functional, hemodynamic, metabolic, cellular, and cytoarchitectural modalities [93]. Malignant gliomas typically demonstrate strong contrast enhancement, peritumoral edema [evident on T2-fluid-attenuated inversion recovery (FLAIR) sequences], mass effects, heterogeneity, central necrosis, and intratumoral hemorrhage on conventional MRI [9497]. In contrast, low-grade astrocytomas often present as well-defined homogeneous masses with little or no mass effect, vasogenic edema, or enhancement after contrast administration on conventional MRI [9496]. Yet, distinguishing low-grade astrocytomas from high-grade astrocytomas and metastases using conventional MRI findings alone is not always possible, as overlapping features may persist [98, 99]. The distinctions that exist between high- and low-grade astrocytomas when observed on different MRI modalities are summarized in Table 2.

    Differences in low- and high-grade astrocytomas among various diagnostic MRI sequences that are currently utilized in astrocytoma diagnoses

    Imaging modalityLow-grade astrocytomasHigh-grade astrocytomas
    MRIWell-defined homogeneous masses with little or no mass effect, vasogenic edema, or enhancement after contrast administrationStrong contrast enhancement, peritumoral edema (evident on T2-FLAIR sequences), mass effects, heterogeneity, central necrosis, and intratumoral hemorrhage on conventional MRI
    SWILow ITSS gradeHigh ITSS grade
    DWIHigh ADCLow ADC
    PWILow rCBVHigh rCBV
    DCEHigh KtransLow Ktrans
    MRSLow Lip-Lac/Cr, low Cho/CrHigh Lip-Lac/Cr, high Cho/Cr
    Display full size

    SWI: susceptibility-weighted imaging; ITSS: intratumoral susceptibility score; DWI: diffusion-weighted imaging; ADC: apparent diffusion coefficient; PWI: perfusion-weighted imaging; rCBV: relative cerebral blood volume; DCE: dynamic contrast enhancement; Ktrans: volume transfer coefficient; MRS: magnetic resonance spectroscopy; Lip-Lac/Cr: lipid-lactate to creatine; Cho/Cr: choline to creatine

    Within GB management, functional MRI (fMRI) has utility pre-operatively, particularly when resection may disrupt eloquent areas [97]. Notably, patients with tumors previously deemed unresectable have become candidates for a more aggressive surgical resection following the utilization of advanced MRI sequences and functional mapping. Diffusion tensor imaging (DTI) can assist in distinguishing between postoperative vascular damage and residual enhancing tumor [100, 101]. Serving a predictive role, DCE can pre-operatively measure contrast uptake, which may be associated with early disease progression [102]. Dynamic susceptibility contrast (DSC) can indicate tumor capillary permeability and neoangiogenesis, factors useful in characterizing glioma type, grade, and tumor recurrence after treatment [103].

    In 2019, Bulakbaşı and Paksoy [104] utilized advanced MRI techniques including SWI, DWI, PWI, MRS, and fMRI to differentiate low- and high-grade gliomas. ADC is determined using DWI, rCBV is approximated using PWI, Ktrans is determined using DCE, ITSS is calculated using SWI, and metabolic markers (i.e., Lip-Lac/ Cr and Cho/Cr) can be measured using MRS. Their study suggested low-grade gliomas have lower cellularity (high ADC), decreased angiogenesis (low rCBV), decreased capillary permeability (high Ktrans), and decreased mitotic activity (Cho/Cr), thus supporting the use of advanced MRI techniques in the differentiation of astrocytomas [104]. Challenges in utilizing these advanced MRI techniques include the diversity of imaging equipment and techniques available, as standardization is currently lacking across facilities [104].

    de Fatima Vasco Aragao et al. [99] evaluated the application of advanced MRI sequences using DWI, PWI, and MRS in differentiating PAs from high-grade astrocytomas. Their study suggested lower rCBV from PWI and higher ADC on DWI were suggestive of PAs, whereas higher Lip-Lac/Cr and Cho/Cr ratios on MRS favored high-grade astrocytomas [99, 104]. The accuracy of MRS in glioma grading is controversial, particularly as many studies have not included the broad range of low-grade gliomas (particularly PAs) [99]. Additionally, MRS patterns do not necessarily translate between adults and children, as is particularly notable in PAs [105].

    Per a 1991 study by Krouwer et al. [106], as low as a 5% gemistocytic component within a patient’s astrocytoma is sufficient to negatively impact prognosis. In 2016, Simkin et al. [107] described the use of MRI in diagnosing gemistocytic astrocytomas. Specifically, gemistocytic astrocytoma evaluation should be considered in patients with heterogeneous cystic lesions that display contrast enhancement and partial FLAIR suppression [107]. Recent literature has emerged on the use of MRI to predict tumor grade in IDH-mutant astrocytomas and oligodendrogliomas [108]. Joyner et al. [108] suggest a positive association between contrast enhancement and the grade of astrocytomas as well as oligodendrogliomas. Additionally, their study suggests a negative association with ADC and a positive association with rCBV in predicting astrocytoma grade, but not oligodendroglioma grade [108].

    Whole-brain radiotherapy vs. stereotactic radiosurgery

    Over the last 50 years, radiotherapy (RT) has been a key therapy in astrocytoma management, as an integral component of multimodal treatment alongside surgical and medical considerations [109]. Initial clinical trials in patients with malignant gliomas, including high-grade astrocytomas, utilized whole-brain RT (WBRT) in conjunction with surgical resection. The combination of WBRT using 5,000–6,000 rad in addition to surgical resection of malignant gliomas revealed significant survival benefit in comparison to surgical resection alone [110, 111]. Studies led by the Brain Tumor Study Group (BTSG) from the National Cancer Institute (NCI) examined the relationship between increasing WBRT dose titration and median survival [112]. Patients receiving doses of 5,000–6,000 rad demonstrated significantly longer median survival, compared to patients randomized to no radiation [112].

    With the advent of more targeted approaches in radiation oncology such as stereotactic radiosurgery (SRS), new techniques, protocols, and recommendations have emerged for studying and treating astrocytoma. SRS differs from WBRT in numerous ways, but the most important distinction is the high degree of precision SRS has directed against cancerous lesions while sparing healthy tissue [113]. SRS typically refers to multisource cobalt-60 gamma radiation or linear accelerator (LINAC)-based photon therapy [114]. Radiation Therapy Oncology Group (RTOG) 93-05 trial evaluated the use of standard RT with TMZ compared to SRS with TMZ and results demonstrated comparable overall survival between both arms. PFS, complete and partial response rates did not significantly differ, but significantly higher toxicity was observed in the standard RT with TMZ arm [113, 115]. Like other focal regimens, SRS has not produced a significantly different survival advantage relative to standard RT [116]. Other roles for SRS may exist, such as salvage therapy for recurrent astrocytoma and as an adjuvant to systemic immunotherapy, but further elucidation of this role is necessary through pre-clinical studies and randomized controlled trials (RCTs) [117, 118].

    Evidence-based guidelines for use of RT in treating high-grade astrocytoma have been established by the American Society for Radiation Oncology (ASTRO). Regarding ideal target volumes for RT against GB, ASTRO issued a “strong recommendation” for partial brain radiation therapy directed against brain areas with the highest recurrence risk, namely within 2 cm of the primary site [119]. This recommendation was supported by evidence indicating that partial brain RT did not decrease survival relative to WBRT [119]. Additionally, a “strong recommendation” was issued for two RT strategies for high-grade astrocytoma:

    • Two-phase strategy utilizing RT against the primary target volume encompassing edema and gross residual tumor/resection cavity plus a boost target volume encompassing gross residual tumor/ residual cavity [119].

    • One-phase single target volume including gross residual tumor/resection cavity with wide margins [119].

    Guidelines issued by the European Association for Neuro-Oncology (EANO) present similar recommendations as ASTRO for various subtypes of astrocytoma. Depending on various prognostic factors, the mainstay radiation recommendation is focused RT with a 1–2.5 cm margin, with SRS suggested as an option in the recurrent setting to improve the delivery of therapy against cancerous lesions and sparing healthy structures [120].

    Guidelines for RT for low-grade astrocytoma were examined and issued by the Spanish Society of Medical Oncology, regarding timing and dosage. Early RT demonstrated a significant increase in PFS, but no difference in overall survival. Based on level I evidence, high and low doses of radiation were demonstrated to be equivalent, with lower doses minimizing radio-necrosis and toxicity [121].

    Chemotherapies

    TMZ

    TMZ is part of a class of drugs known as alkylating agents and was first approved for use in the treatment of GB and AA in 1999 by the Food and Drug Administration (FDA) [122]. Landmark RCTs were conducted in 2005, demonstrating significant survival benefit when TMZ was administered as concomitant and adjuvant chemotherapy with RT. Compared to patients randomized to the RT-alone arm, patients in the TMZ with radiation arm at 28 months displayed a 37% death risk reduction and an overall 2.5-month survival benefit [123]. Follow-up assessing 2-year survival rate revealed that patients who had received the combination arm had a 26.5% survival rate compared to 10.4% for patients who had only received RT with significant survival benefit persisting through the fifth year of follow-up [123, 124]. The methylation status of MGMT further stratified patients with GB. Methylation of this gene in tumor tissue was associated with significant increases in median survival for patients receiving combinatorial therapy of 21.7 months compared to patients lacking methylation of MGMT which had a median survival of 12.7 months [50]. CeTeG/ NOA-09 trial investigated the use of TMZ with lomustine, another alkylating agent, in addition to RT in patients with newly diagnosed GB and who had methylation of the MGMT. The lomustine-TMZ combination treatment arm demonstrated a significant overall survival benefit of 16.7 months, compared to patients randomized to the TMZ-alone treatment arm [125]. The use of TMZ was also assessed in patients with low-grade glioma (astrocytoma, oligoastrocytoma, or oligodendroglioma) compared to RT-alone. Median overall survival has not been reached in this study and no significant differences in PFS were observed between TMZ-alone and RT-alone treatment groups [126]. American Society of Clinical Oncology (ASCO)-Society for Neuro-Oncology (SNO) guidelines issued recommendations for treating various molecular subtypes of astrocytoma graded II–IV using RT with adjuvant TMZ [127]. Additionally, TMZ offers greater tolerability relative to other chemotherapies such as the lone chemotherapeutic agent l,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) or the triple combination therapy procarbazine, lomustine [1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (CCNU)], and vincristine (PCV), further supporting TMZ’s role a first-line treatment for patients with astrocytoma [86].

    Bevacizumab

    Bevacizumab is a monoclonal antibody that is believed to neutralize vascular endothelial growth factor A (VEGF-A). Bevacizumab is FDA-approved for use in adult patients diagnosed with recurrent GB, following promising phase II clinical trials [128]. A double-blind placebo-controlled trial comparing the use of bevacizumab to placebo with standard RT-TMZ therapy in adult patients diagnosed with GB revealed no overall survival benefit [129]. While PFS was longer in patients receiving bevacizumab, there was a coinciding increase in adverse events such as thromboembolic events, neutropenia, worsening quality of life, and increased symptom burden [129]. Another phase III, randomized, double-blind placebo-controlled study elucidated similar findings as seen in previous literature, in which patients with newly diagnosed GB received bevacizumab compared to placebo following standard RT-TMZ [130]. PFS was found to be significantly increased in patients receiving bevacizumab compared to placebo, but no significant differences in overall survival emerged [130]. Several studies have examined the use of bevacizumab in combination with other agents [131136]. The addition of lomustine with bevacizumab was evaluated but did not confer a significant survival advantage compared to lomustine alone in patients with progressive GB [137].

    Other chemotherapies

    In early trials of adjuvant chemotherapy with RT for astrocytoma treatment, one of the first chemotherapies to demonstrate a survival advantage was carmustine, or BCNU [138, 139]. Meta-analysis of early clinical trials evaluated the use of BCNU in combination with RT, showing a 10% increase in survival at 1 year [139]. Additional trials were conducted by comparing BCNU with a cocktail of cytotoxic agents known as PCV [140, 141]. When compared to BCNU, patients treated with PCV demonstrated significant increases in time to progression and overall survival [140, 141]. These drugs remain second-line with respect to TMZ due to limited tolerability. Other classes of cytotoxic chemotherapies, such as platins, phosphoramides, and topoisomerase inhibitors, have been studied in the context of treating primary and recurrent high-grade astrocytomas. These classes have demonstrated minimal or limited activity and poor efficacy, with various clinical guidelines suggesting the use of these compounds only after trials of RT with TMZ or PCV have failed [109, 142].

    Radiation and chemotherapy treatment side effects

    Despite survival benefits with multimodal management of astrocytomas, iatrogenic toxicities and adverse events do exist and afflict patients both short- and long-term. Among the various clinical trials cited in this review, RT-related toxicities and adverse events include but are not limited to infection, worsening neurologic status and late cognitive decline, and significantly worsening Karnofsky performance scores [115, 123, 143]. Related to the use of chemotherapies such as TMZ, toxicities and adverse events include, but are not limited to hematologic toxicities and myelosuppressive effects such as neutropenia, anemia, thrombocytopenia, and lymphopenia [115]. Other adverse effects include moderate-to-severe fatigue, deep vein thrombosis, pulmonary embolism, and severe pneumonia [115]. The use of bevacizumab in patients with GBs exhibited numerous toxicities such as hypertension, leukopenia, thromboembolic events, non-CNS, and CNS hemorrhage [144]. The combination of lomustine and bevacizumab demonstrated similar toxicities to other chemotherapies used to treat astrocytomas [145]. These toxicities include leukopenia, intracranial hemorrhage, serious thromboembolic events, hypertension, liver toxicity, and limited gastrointestinal side effects [145]. In patients that are candidates for re-irradiation due to recurrence, some studies have found rates of neurologic toxicity related to radiation necrosis ranging from 5–20% among patients following SRS, while others have found high rates of therapy tolerance and few adverse events [143, 146]. Overall, each treatment comes with its own unique risks and potential reduction in quality of life.

    Surgical options

    Resection extent

    Resection of low-grade astrocytomas can be curative, but for higher-grade astrocytomas such as GBs, surgery serves a more palliative function [147149]. In addition, EoR plays a critical role in the outcome and prognosis after surgery. Gross total resection (GTR) is a term that has generally come to mean the resection of an entire lesion mapped with T1 enhancement on MRI [150]. In contrast, subtotal resection (STR) is the partial ablation of a lesion that spares the margins of the T1 enhancement, as seen in Figure 3.

    STR spares T1 MRI region margins, GTR includes the T1 margins, and SMR extends into the T2 MRI region

    Note. This figure contains (modified) images from Servier Medical Art (https://smart.servier.com). CC BY.

    A meta-analysis of IDH wild-type low-grade gliomas by Di Carlo et al. [151] demonstrated a significant inverse correlation between EoR and the hazard ratio of death. Similarly, a meta-analysis of low-grade gliomas by Brown et al. [152] showed that GTR compared with STR resulted in clinically and statistically significant reductions in relative risk of death and progression at the 2-, 5-, and 10-year mark. This suggests that GTR when compared to STR has the potential to improve both mortality and morbidity of patients with low-grade astrocytomas. Studies have shown that GTR provides similar benefits for spinal cord astrocytomas [153, 154]. However, given the delicate nature of the region, GTR is often not feasible for high-grade spinal cord astrocytomas [153, 154].

    Recent research has focused on expanding the zone of ablation even further than the bounds of GTR in what is now called SMR, as seen in Figure 3 [155]. This expanded zone of resection is typically mapped using MRI T2 FLAIR instead of T1 [150, 156]. SMR may be more indicative of the extent of GB infiltration given that studies have demonstrated tissue samples beyond the T1 weighted MRI region comprise as much as 89% cancerous tissue [157159]. A number of studies have suggested ideal average ranges of FLAIR resection ranging from 20–53% [160163]. A more recent study by Tripathi et al. [164] suggests that the ideal SMR volume for GBs depends on the physiology of the tumor, with a range of 10–29% for nodular, 10–59% for moderately diffuse, and 30–90% for highly diffuse. One retrospective analysis of 102 patients with GBs showed a statistically significant 6.06-month median overall survival improvement in SMR when compared with GTR [165]. An improvement in overall survival and PFS for SMR compared with GTR has been repeatedly supported by a plethora of studies [150, 156, 166168].

    Neuro navigation

    While the survival benefits of increased EoR are clear, advanced techniques and equipment are necessary to avoid collateral damage to functioning regions of the CNS when achieving GTR or SMR.

    One such commonly utilized system is intraoperative navigation technology. Integrating pre-operative images and tract mapping [magnetoencephalography (MEG), DTI, and fMRI] as a reference point, optical or electromagnetic sensors can locate surgical equipment in real-time in relation to the patient’s brain viewed as a three-dimensional (3D) field [169, 170]. While this allows for more precise ablation, the benefit is often constrained by intraoperative brain shift due to edema, tumor mass reduction, and loss of colony stimulating factor (CSF) [169, 170]. To overcome the limitations introduced by brain shift, intraoperative imaging has been introduced. While intraoperative MRI (iMRI) offers advantages in image resolution it can significantly increase operation time [171]. By comparison, interoperative ultrasound is less costly and may have similar levels of specificity and sensitivity to iMRI, but may require a higher level of training and experience [172]. Another method of monitoring eloquent regions of the brain intraoperatively is through motor evoked potentials (MEPs). In MEP, the brain and primary motor cortex are stimulated intraoperatively to produce corresponding action potential in muscles [173]. Using this method, the motor function of the patient can be monitored and eloquent regions of the brain can be more readily avoided [171, 173].

    Fluorescence

    The two most utilized fluorophores for astrocytoma visualization are 5-aminolevulinic acid (5-ALA) and sodium fluorescein (SF). A porphyrin precursor, 5-ALA accumulates in cancer cells and is metabolized to protoporphyrin IX (PpIX), a fluorophore that is best visualized under a violet, 370–440 nm wavelength light [174, 175]. A systemic review and meta-analysis by Gandhi et al. [176] demonstrated that the addition of 5-ALA to high-grade glioma resections was associated with a 26% increase in GTR rates. SF functions by binding proteins within the blood which collect in regions of tumor-induced BBB perturbation [175]. In this way, SF allows for tumor visualization under 465–490 nm wavelength light [175, 177]. Naik et al. [178] performed a meta-analysis comparing various resection guiding modalities and found that iMRI combined with 5-ALA produced the highest rates of GTR followed by iMRI, SF, and 5-ALA alone. However, another recent meta-analysis by Smith et al. [175] suggested that SF produced a 29.5% increase in GTR which was functionally equivalent to 5-ALA when compared to surgeries without fluorescent guidance. Looking forward, the combined use of both fluorophores may offer some cumulative advantages and may be the focus of future research [179].

    Awake surgery

    While numerous studies have demonstrated the safety of awake craniotomy (AC) compared to general anesthesia (GA), whether AC offers a clear advantage over GA remains a matter of debate [180182]. Some studies have suggested that AC increases the EoR or decreases hospital stay after surgery [180, 182188]. Furthermore, a recent multicenter cohort study by Gerritsen et al. [189] examined surgical outcomes of 1,047 patients with GBs in eloquent regions. They found that AC was associated with significant reductions in postoperative neurological deficits, an increase in overall survival, and an increase in PFS when compared with GA [189]. Currently, a number of prospective studies designed to answer the question of AC vs. GA efficacy (including NCT03861299 and NCT04708171) are underway [190, 191].

    Future treatment

    With unique intrinsic factors and a privileged microenvironment, astrocytomas continue to remain an elusive target for new therapeutic options. One of the reasons it has been challenging to develop new treatments for astrocytomas is the need to penetrate the BBB [192]. The BBB consists of a highly selective semipermeable border of endothelial cells, through which most conventional drugs cannot pass [193]. The selectivity of the BBB is one of the reasons that TMZ remains the standard chemotherapeutic agent in the treatment of astrocytomas. TMZ is able to cross the BBB due to its small size and lipophilic properties [194]. Two recent developments have shown promise in facilitating transport through the BBB [195, 196]. One of these is convection-enhanced delivery (CED), a technique in which a pressure gradient is generated at the tip of an infusion catheter allowing therapeutics to be delivered through the interstitial space into the CNS [195]. Since the development of CED in the 1990s, there have been many advancements over the years. Although CED in its current state has limited anti-tumor effects for astrocytomas, it may show promise in the near future with many current clinical trials seeking to improve the technique [197, 198]. Another method to cross the BBB employed by Gregory et al. [196] is nanoparticle technology. Researchers engineered a synthetic protein nanoparticle (SPNP) based on human serum albumin along with internalizing arginine-glycine-aspartic acid (iRGD) for cell penetration. An in vitro study using this SPNP against signal transducer and activator of transcription 3 inhibitor (STAT3i) in mice found that tumor regression and long-term survival were increased in mice with GB [196]. STAT3i downregulates signal transducer and activator of transcription 3 (STAT3), which impairs anti-tumor immunity and enhances tumor progression [196, 199]. As such, this technology shows promise for the treatment of high-grade astrocytomas.

    Another challenge in developing new therapies for astrocytomas is the tumor microenvironment. Astrocytomas invade the CNS, an immune-privileged site. This has been a significant obstacle in the development of effective immunotherapies. In recent years, immunotherapy treatments such as chimeric antigen receptor (CAR)-T cell therapy, monoclonal antibodies against programmed cell death protein 1 (PD-1), programmed cell death-ligand 1 (PD-L1), and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) have been approved by the FDA for the treatment of various cancers [200202]. However, currently, there are no immunotherapy drugs approved by the FDA for the treatment of astrocytomas [203]. With current clinical trials that seek to improve the shortcomings of CAR-T cell, anti-CTLA-4, anti-PD-1, and anti-PD-L1 therapies in regard to the treatment of astrocytomas, these immunotherapy drugs may show promise in the future. Additional immune checkpoints that are being explored as potential targets for treatment are cluster of differentiation 47 (CD47) and CD73 [203]. CD47 blockade has shown to be effective against GB in vitro and in animal models, however, clinical trials have not yet been conducted on this immunotherapy target [204]. CD73 pathway targeting agents are now reaching clinical trials and may prove successful as a target for immunotherapy [205]. Other immunotherapy options that are being developed for the treatment of astrocytomas are vaccine therapies including dendritic cell-based vaccines, heat shock protein vaccines, peptide vaccines, and viral-based vaccines [192].

    Astrocytomas often have high recurrence rates and may be resistant to chemotherapy and radiation therapy. One of the reasons behind this high recurrence rate may be glioma stem cells (GSCs) which allow for tumorigenicity and stem-cell renewal [203]. Additionally, different types of astrocytomas present with molecular heterogeneity, making it difficult to develop targeted therapeutics [206]. Despite this molecular heterogeneity, targeted therapies may show promise against specific astrocytomas. As discussed previously, some astrocytomas have been found to contain the BRAF V600E mutation, with the highest rate of the mutation found in pleomorphic xanthoastrocytomas [30]. Studies evaluating the use of BRAF V600E kinase inhibitors in various astrocytomas have had interesting results [207, 208]. A basket trial by Kaley et al. [207] looking at the BRAF V600E kinase inhibitor vemurafenib found that patients with pleomorphic xanthoastrocytomas responded best to the drug, while patients with other astrocytomas had variable response. Unfortunately, high-grade astrocytomas tend to have low rates of the BRAF V600E mutation, which may limit the application of vemurafenib and other BRAF V600E kinase inhibitors to a subgroup of astrocytomas [209]. An additional targeted therapeutic that shows potential against high-grade astrocytomas is Erdafitinib, a tyrosine kinase inhibitor of fibroblast growth factor receptor 1–4 (FGFR1–4) [208]. This drug is currently in phase II clinical trials (NCT04083976). Inhibitors of the mutated form of IDH may also prove beneficial against astrocytomas that display this mutation. Potential therapeutics against astrocytomas with mutant IDH1 include AG-5198 and ivosidenib. Additionally, enasidenib may prove to be a potential therapeutic against astrocytomas with mutant IDH2 [206].

    Conclusions

    Staging based on physical and genetic characteristics, imaging, RT, chemotherapy, surgical management, and targeted therapies all serve an integral function in the understanding and management of astrocytomas. Genetic mutations and a variety of imaging modalities are playing an increasingly more important role in the staging and consideration of treatment approaches for astrocytomas. With the ever-increasing interest in targeting the mechanism of each mutation that drives tumor formation, genetic characterization and targeted therapies for astrocytoma treatment will likely continue to develop in conjunction with surgical and radiation therapy. While not always improving the speed and efficiency of surgery, the newest surgical techniques may dramatically improve the field of resection and most importantly improve mortality and quality of life.

    Abbreviations

    5-ALA:

    5-aminolevulinic acid

    AA:

    anaplastic astrocytoma

    AC:

    awake craniotomy

    ADC:

    apparent diffusion coefficient

    ASTRO:

    American Society for Radiation Oncology

    BBB:

    blood-brain barrier

    BCNU:

    l,3-bis(2-chloroethyl)-1-nitrosourea

    BRAF:

    B-Raf proto-oncogene

    CD47:

    cluster of differentiation 47

    CDKN2A:

    cyclin dependent kinase inhibitor 2A

    CED:

    convection-enhanced delivery

    Cho/Cr:

    choline to creatine

    CI:

    confidence interval

    CNS:

    central nervous system

    DCE:

    dynamic contrast enhancement

    DWI:

    diffusion-weighted imaging

    EoR:

    extent of resection

    FDA:

    Food and Drug Administration

    FLAIR:

    fluid-attenuated inversion recovery

    fMRI:

    functional magnetic resonance imaging

    GA:

    general anesthesia

    GB:

    glioblastoma

    GTR:

    gross total resection

    IDH1:

    isocitrate dehydrogenase 1

    iMRI:

    intraoperative magnetic resonance imaging

    ITSS:

    intratumoral susceptibility score

    Ktrans:

    volume transfer coefficient

    Lip-Lac/Cr:

    lipid-lactate to creatine

    MGMT:

    O6-methylguanine-DNA methyltransferase

    MRI:

    magnetic resonance imaging

    MRS:

    magnetic resonance spectroscopy

    NF1:

    neurofibromatosis type 1

    PAs:

    pilocytic astrocytomas

    PCV:

    procarbazine, lomustine, and vincristine

    PFS:

    progression-free survival

    PWI:

    perfusion-weighted imaging

    rCBV:

    relative cerebral blood volume

    RT:

    radiotherapy

    SEGA:

    subependymal giant cell astrocytoma

    SF:

    sodium fluorescein

    SMR:

    supramaximal resection

    SRS:

    stereotactic radiosurgery

    STR:

    subtotal resection

    SWI:

    susceptibility-weighted imaging

    TMZ:

    temozolomide

    TP53:

    tumor protein p53

    WBRT:

    whole-brain radiotherapy

    Declarations

    Author contributions

    MW: Conceptualization, Writing—original draft, Writing—review & editing. JW: Writing—original draft, Writing—review & editing. JF: Writing—original draft. ED: Writing—original draft. SS: Writing—original draft. BO: Writing—original draft. KC: Writing—original draft. JH: Writing—original draft. BLW: Conceptualization, Writing—review & editing.

    Conflicts of interest

    The authors declare that they have no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2023.

    References

    Freeman MR. Specification and morphogenesis of astrocytes. Science. 2010;330:7748. [DOI] [PubMed] [PMC]
    Wang W. Increased incidence of second primary malignancy in patients with malignant astrocytoma: a population-based study. Biosci Rep. 2019;39:BSR20181968. [DOI] [PubMed] [PMC]
    Miller KD, Ostrom QT, Kruchko C, Patil N, Tihan T, Cioffi G, et al. Brain and other central nervous system tumor statistics, 2021. CA Cancer J Clin. 2021;71:381406. [DOI] [PubMed]
    Giovannoni F, Quintana FJ. The role of astrocytes in CNS inflammation. Trends Immunol. 2020;41:80519. [DOI] [PubMed] [PMC]
    Molofsky AV, Deneen B. Astrocyte development: a guide for the perplexed. Glia. 2015;63:13209. [DOI] [PubMed]
    Batiuk MY, Martirosyan A, Wahis J, de Vin F, Marneffe C, Kusserow C, et al. Identification of region-specific astrocyte subtypes at single cell resolution. Nat Commun. 2020;11:1220. [DOI] [PubMed] [PMC]
    Herrero-Navarro Á, Puche-Aroca L, Moreno-Juan V, Sempere-Ferràndez A, Espinosa A, Susín R, et al. Astrocytes and neurons share region-specific transcriptional signatures that confer regional identity to neuronal reprogramming. Sci Adv. 2021;7:eabe8978. [DOI] [PubMed] [PMC]
    Sathornsumetee S, Rich JN, Reardon DA. Diagnosis and treatment of high-grade astrocytoma. Neurol Clin. 2007;25:111139. [DOI] [PubMed]
    Hanif F, Muzaffar K, Perveen K, Malhi SM, Simjee ShU. Glioblastoma multiforme: a review of its epidemiology and pathogenesis through clinical presentation and treatment. Asian Pac J Cancer Prev. 2017;18:39. [DOI] [PubMed] [PMC]
    Omuro A, DeAngelis LM. Glioblastoma and other malignant gliomas: a clinical review. JAMA. 2013;310:184250. [DOI] [PubMed]
    Chaulagain D, Smolanka V, Smolanka A, Munakomi S, Havryliv T. The role of extent of resection on the prognosis of low-grade astrocytoma: a systematic review and meta-analysis. Egypt J Neurosurg. 2022;37:19. [DOI]
    Johnson DR, Galanis E. Medical management of high-grade astrocytoma: current and emerging therapies. Semin Oncol. 2014;41:51122. [DOI] [PubMed]
    Ohgaki H, Kleihues P. Epidemiology and etiology of gliomas. Acta Neuropathol. 2005;109:93108. [DOI] [PubMed]
    Leece R, Xu J, Ostrom QT, Chen Y, Kruchko C, Barnholtz-Sloan JS. Global incidence of malignant brain and other central nervous system tumors by histology, 2003–2007. Neuro Oncol. 2017;19:155364. [DOI] [PubMed] [PMC]
    Ostrom QT, Cote DJ, Ascha M, Kruchko C, Barnholtz-Sloan JS. Adult glioma incidence and survival by race or ethnicity in the United States from 2000 to 2014. JAMA Oncol. 2018;4:125462. [DOI] [PubMed] [PMC]
    Costa AR, Lança de Oliveira M, Cruz I, Gonçalves I, Cascalheira JF, Santos CRA. The sex bias of cancer. Trends Endocrinol Metab. 2020;31:78599. [DOI] [PubMed]
    Bello-Alvarez C, Camacho-Arroyo I. Impact of sex in the prevalence and progression of glioblastomas: the role of gonadal steroid hormones. Biol Sex Differ. 2021;12:28. [DOI] [PubMed] [PMC]
    Carteri RB, Kopczynski A, Rodolphi MS, Strogulski NR, Sartor M, Feldmann M, et al. Testosterone administration after traumatic brain injury reduces mitochondrial dysfunction and neurodegeneration. J Neurotrauma. 2019;36:224659. [DOI] [PubMed] [PMC]
    Rodríguez-Lozano DC, Velázquez-Vázquez DE, Del Moral-Morales Á, Camacho-Árroyo I. Dihydrotestosterone induces proliferation, migration, and invasion of human glioblastoma cell lines. Onco Targets Ther. 2020;13:881323. [DOI] [PubMed] [PMC]
    Carrano A, Juarez JJ, Incontri D, Ibarra A, Guerrero Cazares H. Sex-specific differences in glioblastoma. Cells. 2021;10:1783. [DOI] [PubMed] [PMC]
    Davis ME. Epidemiology and overview of gliomas. Semin Oncol Nurs. 2018;34:4209. [DOI] [PubMed]
    Tabash MA. Characteristics, survival and incidence rates and trends of pilocytic astrocytoma in children in the United States; SEER-based analysis. J Neurol Sci. 2019;400:14852. [DOI] [PubMed]
    Tykocki T, Eltayeb M. Ten-year survival in glioblastoma. A systematic review. J Clin Neurosci. 2018;54:713. [DOI] [PubMed]
    Hauser A, Dutta SW, Showalter TN, Sheehan JP, Grover S, Trifiletti DM. Impact of academic facility type and volume on post-surgical outcomes following diagnosis of glioblastoma. J Clin Neurosci. 2018;47:10310. [DOI] [PubMed]
    Karlsson P, Holmberg E, Lundell M, Mattsson A, Holm LE, Wallgren A. Intracranial tumors after exposure to ionizing radiation during infancy: a pooled analysis of two Swedish cohorts of 28,008 infants with skin hemangioma. Radiat Res. 1998;150:35764. [DOI] [PubMed]
    Bondy ML, Scheurer ME, Malmer B, Barnholtz-Sloan JS, Davis FG, Il’yasova D, et al.; Brain Tumor Epidemiology Consortium. Brain tumor epidemiology: consensus from the Brain Tumor Epidemiology Consortium. Cancer. 2008;113:195368. [DOI] [PubMed] [PMC]
    Chen YH, Gutmann DH. The molecular and cell biology of pediatric low-grade gliomas. Oncogene. 2014;33:201926. [DOI] [PubMed]
    Alcantara Llaguno S, Chen J, Kwon CH, Jackson EL, Li Y, Burns DK, et al. Malignant astrocytomas originate from neural stem/progenitor cells in a somatic tumor suppressor mouse model. Cancer Cell. 2009;15:4556. Erratum in: Cancer Cell. 2009;15:240. [DOI] [PubMed] [PMC]
    Carlos-Escalante JÁ, Gómez-Flores-Ramos L, Bian X, Perdomo-Pantoja Á, de Ándrade KC, Mejía-Pérez SI, et al. Landscape of germline genetic variants in AGT, MGMT, and TP53 in Mexican adult patients with astrocytoma. Cell Mol Neurobiol. 2021;41:128597. [DOI] [PubMed]
    Alentorn A, Labussière M, Sanson M, Delattre JY, Hoang-Xuan K, Idbaih Á. Genetics and brain gliomas. Presse Med. 2013;42:80613. French.
    Lynch TM, Gutmann DH. Neurofibromatosis 1. Neurol Clin. 2002;20:84165. [DOI] [PubMed]
    Listernick R, Louis DN, Packer RJ, Gutmann DH. Optic pathway gliomas in children with neurofibromatosis 1: consensus statement from the NF1 Optic Pathway Glioma Task Force. Ann Neurol. 1997;41:1439. [DOI] [PubMed]
    Stern J, Jakobiec FA, Housepian EM. The architecture of optic nerve gliomas with and without neurofibromatosis. Arch Ophthalmol. 1980;98:50511. [DOI] [PubMed]
    Kirkpatrick P. Neurosurgical management of aneurysmal subarachnoid haemorrhage. J Neurol Neurosurg Psychiatry. 2000;68:538E. [DOI] [PubMed] [PMC]
    Perdomo-Pantoja A, Mejía-Pérez SI, Reynoso-Noverón N, Gómez-Flores-Ramos L, Soto-Reyes E, Sánchez-Correa TE, et al. Ángiotensinogen rs5050 germline genetic variant as potential biomarker of poor prognosis in astrocytoma. PLoS One. 2018;13:e0206590. [DOI] [PubMed] [PMC]
    Wang T, Sun Y, Xiong Z, Wu J, Ding X, Guo X, et al. Association of ST6GAL1 and CYP19A1 polymorphisms in the 3’-UTR with astrocytoma risk and prognosis in a Chinese Han population. BMC Cancer. 2021;21:391. [DOI] [PubMed] [PMC]
    The genetics of cancer [Internet]. Credit the National Cancer Institute as the source; [cited 2022 Aug 10]. Available from: https://www.cancer.gov/about-cancer/causes-prevention/genetics
    Welcsh PL, King MC. BRCA1 and BRCA2 and the genetics of breast and ovarian cancer. Hum Mol Genet. 2001;10:70513. [DOI] [PubMed]
    Guha T, Malkin D. Inherited TP53 mutations and the Li-Fraumeni syndrome. Cold Spring Harb Perspect Med. 2017;7:a026187. [DOI] [PubMed] [PMC]
    IDH1 isocitrate dehydrogenase (NADP(+)) 1 [ Homo sapiens (human) ] [Internet]. Courtesy of the National Library of Medicine; [cited 2022 Aug 11]. Available from: https://www.ncbi.nlm.nih.gov/gene/3417
    IDH2 isocitrate dehydrogenase (NADP(+)) 2 [ Homo sapiens (human) ] [Internet]. Courtesy of the National Library of Medicine; [cited 2022 Aug 11]. Available from: https://www.ncbi.nlm.nih.gov/gene/3418
    Mondesir J, Willekens C, Touat M, de Botton S. IDH1 and IDH2 mutations as novel therapeutic targets: current perspectives. J Blood Med. 2016;7:17180. [DOI] [PubMed] [PMC]
    Reitman ZJ, Yan H. Isocitrate dehydrogenase 1 and 2 mutations in cancer: alterations at a crossroads of cellular metabolism. J Natl Cancer Inst. 2010;102:93241. [DOI] [PubMed] [PMC]
    Dang L, White DW, Gross S, Bennett BD, Bittinger MA, Driggers EM, et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature. 2009;462:73944. [DOI] [PubMed] [PMC]
    van den Bent MJ, Dubbink HJ, Marie Y, Brandes AA, Taphoorn MJ, Wesseling P, et al. IDH1 and IDH2 mutations are prognostic but not predictive for outcome in anaplastic oligodendroglial tumors: a report of the European Organization for Research and Treatment of Cancer Brain Tumor Group. Clin Cancer Res. 2010;16:1597604. [DOI] [PubMed]
    Alifieris C, Trafalis DT. Glioblastoma multiforme: pathogenesis and treatment. Pharmacol Ther. 2015;152:6382. [DOI] [PubMed]
    Hartmann C, Hentschel B, Wick W, Capper D, Felsberg J, Simon M, et al. Patients with IDH1 wild type anaplastic astrocytomas exhibit worse prognosis than IDH1-mutated glioblastomas, and IDH1 mutation status accounts for the unfavorable prognostic effect of higher age: implications for classification of gliomas. Acta Neuropathol. 2010;120:70718. [DOI] [PubMed]
    MGMT O-6-methylguanine-DNA methyltransferase [ Homo sapiens (human) ] [Internet]. Courtesy of the National Library of Medicine; [cited 2022 Aug 11]. Available from: https://www.ncbi.nlm.nih.gov/gene/4255
    Tomar MS, Kumar A, Srivastava C, Shrivastava A. Elucidating the mechanisms of temozolomide resistance in gliomas and the strategies to overcome the resistance. Biochim Biophys Acta Rev Cancer. 2021;1876:188616. [DOI] [PubMed]
    Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 2005;352:9971003. [DOI] [PubMed]
    Minniti G, Scaringi C, Arcella A, Lanzetta G, Di Stefano D, Scarpino S, et al. IDH1 mutation and MGMT methylation status predict survival in patients with anaplastic astrocytoma treated with temozolomide-based chemoradiotherapy. J Neurooncol. 2014;118:37783. [DOI] [PubMed]
    Hosoya T, Takahashi M, Honda-Kitahara M, Miyakita Y, Ohno M, Yanagisawa S, et al. MGMT gene promoter methylation by pyrosequencing method correlates volumetric response and neurological status in IDH wild-type glioblastomas. J Neurooncol. 2022;157:56171. [DOI] [PubMed] [PMC]
    Servier Medical Art [Internet]. [cited 2022 Jan 28]. Available from: https://smart.servier.com
    Muldoon LL, Alvarez JI, Begley DJ, Boado RJ, Del Zoppo GJ, Doolittle ND, et al. Immunologic privilege in the central nervous system and the blood-brain barrier. J Cereb Blood Flow Metab. 2013;33:1321. [DOI] [PubMed] [PMC]
    Peruzzi P, Prabhu V. Astrocytoma tumors [Internet]. Rolling Meadows: American Association of Neurological Surgeons; 2023 [cited 2022 Aug 10]. Available from: https://www.aans.org/Patients/Neurosurgical-Conditions-and-Treatments/Astrocytoma-Tumors
    Brown NF, Carter TJ, Ottaviani D, Mulholland P. Harnessing the immune system in glioblastoma. Br J Cancer. 2018;119:117181. [DOI] [PubMed] [PMC]
    Didenko VV, Ngo HN, Minchew C, Baskin DS. Apoptosis of T lymphocytes invading glioblastomas multiforme: a possible tumor defense mechanism. J Neurosurg. 2002;96:5804. [DOI] [PubMed] [PMC]
    Bodmer S, Strommer K, Frei K, Siepl C, de Tribolet N, Heid I, et al. Immunosuppression and transforming growth factor-beta in glioblastoma. Preferential production of transforming growth factor-beta 2. J Immunol. 1989;143:32229. [DOI] [PubMed]
    Pearson JRD, Cuzzubbo S, McArthur S, Durrant LG, Adhikaree J, Tinsley CJ, et al. Immune escape in glioblastoma multiforme and the adaptation of immunotherapies for treatment. Front Immunol. 2020;11:582106. [DOI] [PubMed] [PMC]
    Zhang H, Liao J, Zhang X, Zhao E, Liang X, Luo S, et al. Sex difference of mutation clonality in diffuse glioma evolution. Neuro Oncol. 2019;21:20113. [DOI] [PubMed] [PMC]
    Claus EB, Cannataro VL, Gaffney SG, Townsend JP. Environmental and sex-specific molecular signatures of glioma causation. Neuro Oncol. 2022;24:2936. Erratum in: Neuro Oncol. 2022;24:2012. [DOI] [PubMed] [PMC]
    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;131:80320. [DOI] [PubMed]
    Mamelak AN, Prados MD, Obana WG, Cogen PH, Edwards MS. Treatment options and prognosis for multicentric juvenile pilocytic astrocytoma. J Neurosurg. 1994;81:2430. [DOI] [PubMed]
    Johnson DR, Brown PD, Galanis E, Hammack JE. Pilocytic astrocytoma survival in adults: analysis of the Surveillance, Epidemiology, and End Results program of the National Cancer Institute. J Neurooncol. 2012;108:18793. [DOI] [PubMed]
    Dirven CM, Mooij JJ, Molenaar WM. Cerebellar pilocytic astrocytoma: a treatment protocol based upon analysis of 73 cases and a review of the literature. Childs Nerv Syst. 1997;13:1723. [DOI] [PubMed]
    Komotar RJ, Burger PC, Carson BS, Brem H, Olivi A, Goldthwaite PT, et al. Pilocytic and pilomyxoid hypothalamic/chiasmatic astrocytomas. Neurosurgery. 2004;54:7280. [DOI] [PubMed]
    Fernandez C, Figarella-Branger D, Girard N, Bouvier-Labit C, Gouvernet J, Paz Paredes A, et al. Pilocytic astrocytomas in children: prognostic factors—a retrospective study of 80 cases. Neurosurgery. 2003;53:54455. [DOI] [PubMed]
    Bornhorst M, Frappaz D, Packer RJ. Chapter 20 - Pilocytic astrocytomas. In: Berger MS, Weller M, editors. Handb Clin Neurol. Elsevier; 2016. pp. 329–44. [DOI] [PubMed]
    Mair MJ, Wöhrer A, Furtner J, Simonovska A, Kiesel B, Oberndorfer S, et al. Clinical characteristics and prognostic factors of adult patients with pilocytic astrocytoma. J Neurooncol. 2020;148:18798. [DOI] [PubMed] [PMC]
    Tahiri Elousrouti L, Lamchahab M, Bougtoub N, Elfatemi H, Chbani L, Harmouch T, et al. Subependymal giant cell astrocytoma (SEGA): a case report and review of the literature. J Med Case Rep. 2016;10:35. [DOI] [PubMed] [PMC]
    Campen CJ, Porter BE. Subependymal giant cell astrocytoma (SEGA) treatment update. Curr Treat Options Neurol. 2011;13:3805. [DOI] [PubMed] [PMC]
    Ryall S, Tabori U, Hawkins C. Pediatric low-grade glioma in the era of molecular diagnostics. Acta Neuropathol Commun. 2020;8:30. [DOI] [PubMed] [PMC]
    Liu S, Liu X, Zhuang W. Prognostic factors associated with survival in patients with diffuse astrocytoma. Front Surg. 2021;8:712350. [DOI] [PubMed] [PMC]
    Chiang J, Harreld JH, Tinkle CL, Moreira DC, Li X, Acharya S, et al. A single-center study of the clinicopathologic correlates of gliomas with a MYB or MYBL1 alteration. Acta Neuropathol. 2019;138:10912. [DOI] [PubMed] [PMC]
    Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro Oncol. 2021;23:123151. [DOI] [PubMed] [PMC]
    Wessels PH, Weber WE, Raven G, Ramaekers FC, Hopman AH, Twijnstra A. Supratentorial grade II astrocytoma: biological features and clinical course. Lancet Neurol. 2003;2:395403. [DOI] [PubMed]
    Lind-Landström T, Habberstad AH, Sundstrøm S, Torp SH. Prognostic value of histological features in diffuse astrocytomas WHO grade II. Int J Clin Exp Pathol. 2012;5:1528. [PubMed] [PMC]
    Zhang F, Liu Y, Zhang Z, Li J, Wan Y, Zhang L, et al. 5-hydroxymethylcytosine loss is associated with poor prognosis for patients with WHO grade II diffuse astrocytomas. Sci Rep. 2016;6:20882. [DOI] [PubMed] [PMC]
    Chen JR, Yao Y, Xu HZ, Qin ZY. Isocitrate dehydrogenase (IDH)1/2 mutations as prognostic markers in patients with glioblastomas. Medicine (Baltimore). 2016;95:e2583. [DOI] [PubMed] [PMC]
    Cancer Genome Atlas Research Network; Brat DJ, Verhaak RG, Aldape KD, Yung WK, Salama SR, Cooper LA, et al. Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. N Engl J Med. 2015;372:248198. [DOI] [PubMed] [PMC]
    Thon N, Eigenbrod S, Kreth S, Lutz J, Tonn JC, Kretzschmar H, et al. IDH1 mutations in grade II astrocytomas are associated with unfavorable progression-free survival and prolonged postrecurrence survival. Cancer. 2012;118:45260. [DOI] [PubMed]
    Ahmadi R, Stockhammer F, Becker N, Hohlen K, Misch M, Christians A, et al. No prognostic value of IDH1 mutations in a series of 100 WHO grade II astrocytomas. J Neurooncol. 2012;109:1522. [DOI] [PubMed]
    Dong X, Noorbakhsh A, Hirshman BR, Zhou T, Tang JA, Chang DC, et al. Survival trends of grade I, II, and III astrocytoma patients and associated clinical practice patterns between 1999 and 2010: a SEER-based analysis. Neurooncol Pract. 2016;3:2938. [DOI] [PubMed] [PMC]
    Shaikh N, Brahmbhatt N, Kruser TJ, Kam KL, Appin CL, Wadhwani N, et al. Pleomorphic xanthoastrocytoma: a brief review. CNS Oncol. 2019;8:CNS39. [DOI] [PubMed] [PMC]
    Lim S, Kim JH, Kim SA, Park ES, Ra YS, Kim CJ. Prognostic factors and therapeutic outcomes in 22 patients with pleomorphic xanthoastrocytoma. J Korean Neurosurg Soc. 2013;53:2817. [DOI] [PubMed] [PMC]
    Brandes AA, Nicolardi L, Tosoni A, Gardiman M, Iuzzolino P, Ghimenton C, et al. Survival following adjuvant PCV or temozolomide for anaplastic astrocytoma. Neuro Oncol. 2006;8:25360. [DOI] [PubMed] [PMC]
    Lakomy R, Kazda T, Selingerova I, Poprach A, Pospisil P, Belanova R, et al. Real-world evidence in glioblastoma: Stupp’s regimen after a decade. Front Oncol. 2020;10:840. [DOI] [PubMed] [PMC]
    Christians A, Adel-Horowski A, Banan R, Lehmann U, Bartels S, Behling F, et al. The prognostic role of IDH mutations in homogeneously treated patients with anaplastic astrocytomas and glioblastomas. Acta Neuropathol Commun. 2019;7:156. [DOI] [PubMed] [PMC]
    Yang W, Xu T, Garzon-Muvdi T, Jiang C, Huang J, Chaichana KL. Survival of ventricular and periventricular high-grade gliomas: a Surveillance, Epidemiology, and End Results program-based study. World Neurosurg. 2018;111:e32334. [DOI] [PubMed]
    Fyllingen EH, Bø LE, Reinertsen I, Jakola AS, Sagberg LM, Berntsen EM, et al. Survival of glioblastoma in relation to tumor location: a statistical tumor atlas of a population-based cohort. Acta Neurochir (Wien). 2021;163:1895905. [DOI] [PubMed] [PMC]
    Osborn AG, Louis DN, Poussaint TY, Linscott LL, Salzman KL. The 2021 World Health Organization classification of tumors of the central nervous system: what neuroradiologists need to know. AJNR Am J Neuroradiol. 2022;43:92837. [DOI] [PubMed]
    McNamara C, Mankad K, Thust S, Dixon L, Limback-Stanic C, D’Arco F, et al. 2021 WHO classification of tumours of the central nervous system: a review for the neuroradiologist. Neuroradiology. 2022;64:191950. [DOI] [PubMed]
    Mechtler L. Neuroimaging in neuro-oncology. Neurol Clin. 2009;27:171201. [DOI] [PubMed]
    Pierallini A, Bonamini M, Bozzao A, Pantano P, Stefano DD, Ferone E, et al. Supratentorial diffuse astrocytic tumours: proposal of an MRI classification. Eur Radiol. 1997;7:3959. [DOI] [PubMed]
    Tervonen O, Forbes G, Scheithauer BW, Dietz MJ. Diffuse “fibrillary” astrocytomas: correlation of MRI features with histopathologic parameters and tumor grade. Neuroradiology. 1992;34:1738. [DOI] [PubMed]
    Watanabe M, Tanaka R, Takeda N. Magnetic resonance imaging and histopathology of cerebral gliomas. Neuroradiology. 1992;34:4639. [DOI] [PubMed]
    Shukla G, Alexander GS, Bakas S, Nikam R, Talekar K, Palmer JD, et al. Advanced magnetic resonance imaging in glioblastoma: a review. Chin Clin Oncol. 2017;6:40. [DOI] [PubMed]
    Kono K, Inoue Y, Nakayama K, Shakudo M, Morino M, Ohata K, et al. The role of diffusion-weighted imaging in patients with brain tumors. AJNR Am J Neuroradiol. 2001;22:10818. [PubMed] [PMC]
    de Fatima Vasco Aragao M, Law M, Batista de Almeida D, Fatterpekar G, Delman B, Bader AS, et al. Comparison of perfusion, diffusion, and MR spectroscopy between low-grade enhancing pilocytic astrocytomas and high-grade astrocytomas. AJNR Am J Neuroradiol. 2014;35:1495502. [DOI] [PubMed] [PMC]
    Abdullah KG, Lubelski D, Nucifora PG, Brem S. Use of diffusion tensor imaging in glioma resection. Neurosurg Focus. 2013;34:E1. [DOI] [PubMed]
    Lee SK. Diffusion tensor and perfusion imaging of brain tumors in high-field MR imaging. Neuroimaging Clin N Am. 2012;22:12334. [DOI] [PubMed]
    Kim R, Choi SH, Yun TJ, Lee ST, Park CK, Kim TM, et al. Prognosis prediction of non-enhancing T2 high signal intensity lesions in glioblastoma patients after standard treatment: application of dynamic contrast-enhanced MR imaging. Eur Radiol. 2017;27:117685. [DOI] [PubMed]
    Chakravorty A, Steel T, Chaganti J. Accuracy of percentage of signal intensity recovery and relative cerebral blood volume derived from dynamic susceptibility-weighted, contrast-enhanced MRI in the preoperative diagnosis of cerebral tumours. Neuroradiol J. 2015;28:57483. [DOI] [PubMed] [PMC]
    Bulakbaşı N, Paksoy Y. Ádvanced imaging in adult diffusely infiltrating low-grade gliomas. Insights Imaging. 2019;10:122. Erratum in: Insights Imaging. 2020;11:57. [DOI] [PubMed] [PMC]
    Porto L, Kieslich M, Franz K, Lehrbecher T, Vlaho S, Pilatus U, et al. Spectroscopy of untreated pilocytic astrocytomas: do children and adults share some metabolic features in addition to their morphologic similarities? Childs Nerv Syst. 2010;26:8016. [DOI] [PubMed]
    Krouwer HG, Davis RL, Silver P, Prados M. Gemistocytic astrocytomas: a reappraisal. J Neurosurg. 1991;74:399406. [DOI] [PubMed]
    Simkin PM, Yang N, Tsui A, Kalnins RM, Fitt G, Gaillard F. Magnetic resonance imaging features of gemistocytic astrocytoma. J Med Imaging Radiat Oncol. 2016;60:73340. [DOI] [PubMed]
    Joyner DA, Garrett J, Batchala PP, Rama B, Ravicz JR, Patrie JT, et al. MRI features predict tumor grade in isocitrate dehydrogenase (IDH)-mutant astrocytoma and oligodendroglioma. Neuroradiology. 2023;65:1219. [DOI] [PubMed]
    Kim YZ, Kim CY, Lim DH. The overview of practical guidelines for gliomas by KSNO, NCCN, and EANO. Brain Tumor Res Treat. 2022;10:8393. [DOI] [PubMed] [PMC]
    Salazar OM, Rubin P, Feldstein ML, Pizzutiello R. High dose radiation therapy in the treatment of malignant gliomas: final report. Int J Radiat Oncol Biol Phys. 1979;5:173340. [DOI] [PubMed]
    Salazar OM, VanHoutte PJ, Bennett JM, Rubin P, Wheeler KT. High-dose radiation therapy with low-dose (pulsed) BCNU in malignant gliomas: an eastern cooperative oncology group (ECOG) report. Int J Radiat Oncol Biol Phys. 1982;8:9159. [DOI] [PubMed]
    Walker MD, Strike TA, Sheline GE. An analysis of dose-effect relationship in the radiotherapy of malignant gliomas. Int J Radiat Oncol Biol Phys. 1979;5:172531. [DOI] [PubMed]
    Chen JC, Girvigian MR. Stereotactic radiosurgery: instrumentation and theoretical aspects—part 1. Perm J. 2005;9:236. [DOI] [PubMed] [PMC]
    Yanagihara TK, Saadatmand HJ, Wang TJ. Reevaluating stereotactic radiosurgery for glioblastoma: new potential for targeted dose-escalation. J Neurooncol. 2016;130:397411. [DOI] [PubMed]
    Minniti G, Scaringi C, Lanzetta G, Terrenato I, Esposito V, Arcella A, et al. Standard (60 Gy) or short-course (40 Gy) irradiation plus concomitant and adjuvant temozolomide for elderly patients with glioblastoma: a propensity-matched analysis. Int J Radiat Oncol Biol Phys. 2015;91:10915. [DOI] [PubMed]
    Niyazi M, Brada M, Chalmers AJ, Combs SE, Erridge SC, Fiorentino A, et al. ESTRO-ACROP guideline “target delineation of glioblastomas”. Radiother Oncol. 2016;118:3542. [DOI] [PubMed]
    Kong DS, Lee JI, Park K, Kim JH, Lim DH, Nam DH. Efficacy of stereotactic radiosurgery as a salvage treatment for recurrent malignant gliomas. Cancer. 2008;112:204651. [DOI] [PubMed]
    Zeng J, See AP, Phallen J, Jackson CM, Belcaid Z, Ruzevick J, et al. Anti-PD-1 blockade and stereotactic radiation produce long-term survival in mice with intracranial gliomas. Int J Radiat Oncol Biol Phys. 2013;86:3439. [DOI] [PubMed] [PMC]
    Cabrera AR, Kirkpatrick JP, Fiveash JB, Shih HA, Koay EJ, Lutz S, et al. Radiation therapy for glioblastoma: executive summary of an American Society for Radiation Oncology evidence-based clinical practice guideline. Pract Radiat Oncol. 2016;6:21725. [DOI] [PubMed]
    Weller M, van den Bent M, Tonn JC, Stupp R, Preusser M, Cohen-Jonathan-Moyal E, et al.; European Association for Neuro-Oncology (EANO) Task Force on Gliomas. European Association for Neuro-Oncology (EANO) guideline on the diagnosis and treatment of adult astrocytic and oligodendroglial gliomas. Lancet Oncol. 2017;18:e31529. Erratum in: Lancet Oncol. 2017;18:e642. [DOI] [PubMed]
    Sepúlveda-Sánchez JM, Muñoz Langa J, Arráez MÁ, Fuster J, Hernández Laín Á, Reynés G, et al. Correction to: SEOM clinical guideline of diagnosis and management of low-grade glioma (2017). Clin Transl Oncol. 2018;20:1089. Erratum for: Clin Transl Oncol. 2018;20:3–15. [DOI]
    Tatar Z, Thivat E, Planchat E, Gimbergues P, Gadea E, Abrial C, et al. Temozolomide and unusual indications: review of literature. Cancer Treat Rev. 2013;39:12535. [DOI] [PubMed]
    Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al.; European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups, National Cancer Institute of Canada Clinical Trials Group. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:98796. [DOI] [PubMed]
    Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, et al.; European Organisation for Research and Treatment of Cancer Brain Tumour and Radiation Oncology Groups, National Cancer Institute of Canada Clinical Trials Group. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009;10:45966. [DOI] [PubMed]
    Herrlinger U, Tzaridis T, Mack F, Steinbach JP, Schlegel U, Sabel M, et al.; Neurooncology Working Group of the German Cancer Society. Lomustine-temozolomide combination therapy versus standard temozolomide therapy in patients with newly diagnosed glioblastoma with methylated MGMT promoter (CeTeG/NOA-09): a randomised, open-label, phase 3 trial. Lancet. 2019;393:67888. [DOI]
    Baumert BG, Hegi ME, van den Bent MJ, von Deimling A, Gorlia T, Hoang-Xuan K, et al. Temozolomide chemotherapy versus radiotherapy in high-risk low-grade glioma (EORTC 22033-26033): a randomised, open-label, phase 3 intergroup study. Lancet Oncol. 2016;17:152132. [DOI] [PubMed]
    Mohile NA, Messersmith H, Gatson NT, Hottinger AF, Lassman A, Morton J, et al. Therapy for diffuse astrocytic and oligodendroglial tumors in adults: ASCO-SNO guideline. J Clin Oncol. 2022;40:40326. [DOI] [PubMed]
    Kreisl TN, Kim L, Moore K, Duic P, Royce C, Stroud I, et al. Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol. 2009;27:7405. [DOI] [PubMed] [PMC]
    Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal DT, Vogelbaum MA, et al. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med. 2014;370:699708. [DOI] [PubMed] [PMC]
    Chinot OL, Wick W, Mason W, Henriksson R, Saran F, Nishikawa R, et al. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med. 2014;370:70922. [DOI] [PubMed]
    Gil-Gil MJ, Mesia C, Rey M, Bruna J. Bevacizumab for the treatment of glioblastoma. Clin Med Insights Oncol. 2013;7:CMO.S8503. [DOI] [PubMed] [PMC]
    Sathornsumetee S, Desjardins A, Vredenburgh JJ, McLendon RE, Marcello J, Herndon JE, et al. Phase II trial of bevacizumab and erlotinib in patients with recurrent malignant glioma. Neuro Oncol. 2010;12:130010. [DOI] [PubMed] [PMC]
    Reardon DA, Desjardins A, Vredenburgh JJ, Gururangan S, Sampson JH, Sathornsumetee S, et al. Metronomic chemotherapy with daily, oral etoposide plus bevacizumab for recurrent malignant glioma: a phase II study. Br J Cancer. 2009;101:198694. [DOI] [PubMed] [PMC]
    Verhoeff JJC, Lavini C, van Linde ME, Stalpers LJA, Majoie CBLM, Reijneveld JC, et al. Bevacizumab and dose-intense temozolomide in recurrent high-grade glioma. Ann Oncol. 2010;21:17237. [DOI] [PubMed]
    Soffietti R, Trevisan E, Bertero L, Cassoni P, Morra I, Fabrini MG, et al. Bevacizumab and fotemustine for recurrent glioblastoma: a phase II study of AINO (Italian Association of Neuro-Oncology). J Neurooncol. 2014;116:53341. [DOI] [PubMed] [PMC]
    Hasselbalch B, Lassen U, Hansen S, Holmberg M, Sørensen M, Kosteljanetz M, et al. Cetuximab, bevacizumab, and irinotecan for patients with primary glioblastoma and progression after radiation therapy and temozolomide: a phase II trial. Neuro Oncol. 2010;12:50816. [DOI] [PubMed] [PMC]
    Wick W, Gorlia T, Bendszus M, Taphoorn M, Sahm F, Harting I, et al. Lomustine and bevacizumab in progressive glioblastoma. N Engl J Med. 2017;377:195463. [DOI] [PubMed]
    Armstrong TS, Gilbert MR. Chemotherapy of astrocytomas: an overview. Semin Oncol Nurs. 1998;14:1825. [DOI] [PubMed]
    Fine HA, Dear KB, Loeffler JS, Black PM, Canellos GP. Meta-analysis of radiation therapy with and without adjuvant chemotherapy for malignant gliomas in adults. Cancer. 1993;71:258597. [DOI] [PubMed]
    Cairncross G, Macdonald D, Ludwin S, Lee D, Cascino T, Buckner J, et al. Chemotherapy for anaplastic oligodendroglioma. National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 1994;12:201321. [DOI] [PubMed]
    Levin VA, Silver P, Hannigan J, Wara WM, Gutin PH, Davis RL, et al. Superiority of post-radiotherapy adjuvant chemotherapy with CCNU, procarbazine, and vincristine (PCV) over BCNU for anaplastic gliomas: NCOG 6G61 final report. Int J Radiat Oncol Biol Phys. 1990;18:3214. [DOI] [PubMed]
    Grimm SA, Chamberlain MC. State of the art and perspectives in the treatment of glioblastoma. CNS Oncol. 2012;1:4970. [DOI] [PubMed] [PMC]
    Fokas E, Wacker U, Gross MW, Henzel M, Encheva E, Engenhart-Cabillic R. Hypofractionated stereotactic reirradiation of recurrent glioblastomas : a beneficial treatment option after high-dose radiotherapy? Strahlenther Onkol. 2009;185:23540. [DOI] [PubMed]
    Diaz RJ, Ali S, Qadir MG, De La Fuente MI, Ivan ME, Komotar RJ. The role of bevacizumab in the treatment of glioblastoma. J Neurooncol. 2017;133:45567. [DOI] [PubMed]
    Jakobsen JN, Urup T, Grunnet K, Toft A, Johansen MD, Poulsen SH, et al. Toxicity and efficacy of lomustine and bevacizumab in recurrent glioblastoma patients. J Neurooncol. 2018;137:43946. [DOI] [PubMed]
    Minniti G, Niyazi M, Alongi F, Navarria P, Belka C. Current status and recent advances in reirradiation of glioblastoma. Radiat Oncol. 2021;16:36. [DOI]
    De Leeuw BI, Van Baarsen KM, Snijders TJ, Robe PAJT. Interrelationships between molecular subtype, anatomical location, and extent of resection in diffuse glioma: a systematic review and meta-analysis. Neurooncol Adv. 2019;1:vdz032. [DOI] [PubMed] [PMC]
    Bonfield CM, Steinbok P. Pediatric cerebellar astrocytoma: a review. Childs Nerv Syst. 2015;31:167785. [DOI] [PubMed]
    Davis ME. Glioblastoma: overview of disease and treatment. Clin J Oncol Nurs. 2016;20:S28. [DOI] [PubMed] [PMC]
    Dimou J, Beland B, Kelly J. Supramaximal resection: a systematic review of its safety, efficacy and feasibility in glioblastoma. J Clin Neurosci. 2020;72:32834. [DOI] [PubMed]
    Di Carlo DT, Duffau H, Cagnazzo F, Benedetto N, Morganti R, Perrini P. IDH wild-type WHO grade II diffuse low-grade gliomas. A heterogeneous family with different outcomes. Systematic review and meta-analysis. Neurosurg Rev. 2020;43:38395. [DOI] [PubMed]
    Brown TJ, Bota DA, van Den Bent MJ, Brown PD, Maher E, Aregawi D, et al. Management of low-grade glioma: a systematic review and meta-analysis. Neurooncol Pract. 2019;6:24958. [DOI] [PubMed] [PMC]
    Nagoshi N, Tsuji O, Suzuki S, Nori S, Yagi M, Okada E, et al. Clinical outcomes and a therapeutic indication of intramedullary spinal cord astrocytoma. Spinal Cord. 2022;60:21622. [DOI] [PubMed]
    Ogunlade J, Wiginton JG 4th, Elia C, Odell T, Rao SC. Primary spinal astrocytomas: a literature review. Cureus. 2019;11:e5247. [DOI] [PubMed] [PMC]
    Khalafallah AM, Rakovec M, Bettegowda C, Jackson CM, Gallia GL, Weingart JD, et al. A crowdsourced consensus on supratotal resection versus gross total resection for anatomically distinct primary glioblastoma. Neurosurgery. 2021;89:7129. [DOI] [PubMed] [PMC]
    Jackson C, Choi J, Khalafallah AM, Price C, Bettegowda C, Lim M, et al. A systematic review and meta-analysis of supratotal versus gross total resection for glioblastoma. J Neurooncol. 2020;148:41931. [DOI] [PubMed]
    Barajas RF Jr, Phillips JJ, Parvataneni R, Molinaro A, Essock-Burns E, Bourne G, et al. Regional variation in histopathologic features of tumor specimens from treatment-naive glioblastoma correlates with anatomic and physiologic MR Imaging. Neuro Oncol. 2012;14:94254. [DOI] [PubMed] [PMC]
    Eidel O, Burth S, Neumann JO, Kieslich PJ, Sahm F, Jungk C, et al. Tumor infiltration in enhancing and non-enhancing parts of glioblastoma: a correlation with histopathology. PLoS One. 2017;12:e0169292. [DOI] [PubMed] [PMC]
    Kubben PL, Wesseling P, Lammens M, Schijns OE, Ter Laak-Poort MP, van Overbeeke JJ, et al. Correlation between contrast enhancement on intraoperative magnetic resonance imaging and histopathology in glioblastoma. Surg Neurol Int. 2012;3:158. [DOI] [PubMed] [PMC]
    Haddad AF, Young JS, Morshed RA, Berger MS. FLAIRectomy: resecting beyond the contrast margin for glioblastoma. Brain Sci. 2022;12:544. [DOI] [PubMed] [PMC]
    Li YM, Suki D, Hess K, Sawaya R. The influence of maximum safe resection of glioblastoma on survival in 1229 patients: can we do better than gross-total resection? J Neurosurg. 2016;124:97788. [DOI] [PubMed]
    Vivas-Buitrago T, Domingo RA, Tripathi S, De Biase G, Brown D, Akinduro OO, et al. Influence of supramarginal resection on survival outcomes after gross-total resection of IDH-wild-type glioblastoma. J Neurosurg. 2021;136:18. [DOI] [PubMed] [PMC]
    Pessina F, Navarria P, Cozzi L, Ascolese AM, Simonelli M, Santoro A, et al. Maximize surgical resection beyond contrast-enhancing boundaries in newly diagnosed glioblastoma multiforme: is it useful and safe? A single institution retrospective experience. J Neurooncol. 2017;135:12939. [DOI] [PubMed]
    Tripathi S, Vivas-Buitrago T, Domingo RA, Biase GD, Brown D, Akinduro OO, et al. IDH-wild-type glioblastoma cell density and infiltration distribution influence on supramarginal resection and its impact on overall survival: a mathematical model. J Neurosurg. 2022;136:156775. [DOI] [PubMed]
    Di L, Shah AH, Mahavadi A, Eichberg DG, Reddy R, Sanjurjo AD, et al. Radical supramaximal resection for newly diagnosed left-sided eloquent glioblastoma: safety and improved survival over gross-total resection. J Neurosurg. 2022;138:629. [DOI] [PubMed]
    Glenn CA, Baker CM, Conner AK, Burks JD, Bonney PA, Briggs RG, et al. An examination of the role of supramaximal resection of temporal lobe glioblastoma multiforme. World Neurosurg. 2018;114:e74755. [DOI] [PubMed]
    Lopez-Rivera V, Dono A, Lewis CT, Chandra A, Abdelkhaleq R, Sheth SA, et al. Extent of resection and survival outcomes of geriatric patients with glioblastoma: is there benefit from aggressive surgery? Clin Neurol Neurosurg. 2021;202:106474. [DOI] [PubMed]
    Shah AH, Mahavadi A, Di L, Sanjurjo A, Eichberg DG, Borowy V, et al. Survival benefit of lobectomy for glioblastoma: moving towards radical supramaximal resection. J Neurooncol. 2020;148:5018. [DOI] [PubMed] [PMC]
    Lukas RV, Wainwright DA, Ladomersky E, Sachdev S, Sonabend AM, Stupp R. Newly diagnosed glioblastoma: a review on clinical management. Oncology (Williston Park). 2019;33:91100. [PubMed] [PMC]
    Ma R, Taphoorn MJB, Plaha P. Advances in the management of glioblastoma. J Neurol Neurosurg Psychiatry. 2021;92:110311. [DOI] [PubMed]
    Matsumae M, Nishiyama J, Kuroda K. Intraoperative MR imaging during glioma resection. Magn Reson Med Sci. 2022;21:14867. [DOI] [PubMed] [PMC]
    Coburger J, Scheuerle A, Kapapa T, Engelke J, Thal DR, Wirtz CR, et al. Sensitivity and specificity of linear array intraoperative ultrasound in glioblastoma surgery: a comparative study with high field intraoperative MRI and conventional sector array ultrasound. Neurosurg Rev. 2015;38:499509. [DOI] [PubMed]
    Tanaka S, Tashiro T, Gomi A, Takanashi J, Ujiie H. Sensitivity and specificity in transcranial motor-evoked potential monitoring during neurosurgical operations. Surg Neurol Int. 2011;2:111. [DOI] [PubMed] [PMC]
    Traylor JI, Pernik MN, Sternisha AC, McBrayer SK, Abdullah KG. Molecular and metabolic mechanisms underlying selective 5-aminolevulinic acid-induced fluorescence in gliomas. Cancers (Basel). 2021;13:580. [DOI] [PubMed] [PMC]
    Smith EJ, Gohil K, Thompson CM, Naik A, Hassaneen W. Fluorescein-guided resection of high grade gliomas: a meta-analysis. World Neurosurg. 2021;155:1818.e7. [DOI] [PubMed]
    Gandhi S, Tayebi Meybodi A, Belykh E, Cavallo C, Zhao X, Syed MP, et al. Survival outcomes among patients with high-grade glioma treated with 5-aminolevulinic acid-guided surgery: a systematic review and meta-analysis. Front Oncol. 2019;9:620. [DOI] [PubMed] [PMC]
    Zhang N, Tian H, Huang D, Meng X, Guo W, Wang C, et al. Sodium fluorescein-guided resection under the YELLOW 560 nm surgical microscope filter in malignant gliomas: our first 38 cases experience. Biomed Res Int. 2017;2017:7865747. Erratum in: Biomed Res Int. 2018;2018:6348625. [DOI] [PubMed] [PMC]
    Naik A, Smith EJ, Barreau A, Nyaeme M, Cramer SW, Najafali D, et al. Comparison of fluorescein sodium, 5-ALA, and intraoperative MRI for resection of high-grade gliomas: a systematic review and network meta-analysis. J Clin Neurosci. 2022;98:2407. [DOI] [PubMed]
    Zeppa P, De Marco R, Monticelli M, Massara A, Bianconi A, Di Perna G, et al. Fluorescence-guided surgery in glioblastoma: 5-ALA, SF or both? Differences between fluorescent dyes in 99 consecutive cases. Brain Sci. 2022;12:555. [DOI] [PubMed] [PMC]
    Clavreul A, Aubin G, Delion M, Lemée JM, Ter Minassian A, Menei P. What effects does awake craniotomy have on functional and survival outcomes for glioblastoma patients? J Neurooncol. 2021;151:11321. [DOI] [PubMed]
    Sacko O, Lauwers-Cances V, Brauge D, Sesay M, Brenner A, Roux FE. Awake craniotomy vs surgery under general anesthesia for resection of supratentorial lesions. Neurosurgery. 2011;68:11929. [DOI] [PubMed]
    Gravesteijn BY, Keizer ME, Vincent AJPE, Schouten JW, Stolker RJ, Klimek M. Awake craniotomy versus craniotomy under general anesthesia for the surgical treatment of insular glioma: choices and outcomes. Neurol Res. 2018;40:8796. [DOI] [PubMed]
    Suarez-Meade P, Marenco-Hillembrand L, Prevatt C, Murguia-Fuentes R, Mohamed A, Alsaeed T, et al. Awake vs. asleep motor mapping for glioma resection: a systematic review and meta-analysis. Acta Neurochir (Wien). 2020;162:170920. [DOI] [PubMed]
    Zhang JJY, Lee KS, Voisin MR, Hervey-Jumper SL, Berger MS, Zadeh G. Awake craniotomy for resection of supratentorial glioblastoma: a systematic review and meta-analysis. Neurooncol Adv. 2020;2:vdaa111. [DOI] [PubMed] [PMC]
    Zigiotto L, Annicchiarico L, Corsini F, Vitali L, Falchi R, Dalpiaz C, et al. Effects of supra-total resection in neurocognitive and oncological outcome of high-grade gliomas comparing asleep and awake surgery. J Neurooncol. 2020;148:97108. [DOI] [PubMed]
    Bu LH, Zhang J, Lu JF, Wu JS. Glioma surgery with awake language mapping versus generalized anesthesia: a systematic review. Neurosurg Rev. 2021;44:19972011. [DOI] [PubMed]
    Moiraghi A, Roux A, Peeters S, Pelletier JB, Baroud M, Trancart B, et al. Feasibility, safety and impact on overall survival of awake resection for newly diagnosed supratentorial IDH-wildtype glioblastomas in adults. Cancers (Basel). 2021;13:2911. [DOI] [PubMed] [PMC]
    Lu VM, Phan K, Rovin RA. Comparison of operative outcomes of eloquent glioma resection performed under awake versus general anesthesia: a systematic review and meta-analysis. Clin Neurol Neurosurg. 2018;169:1217. [DOI] [PubMed]
    Gerritsen JKW, Zwarthoed RH, Kilgallon JL, Nawabi NL, Jessurun CAC, Versyck G, et al. Effect of awake craniotomy in glioblastoma in eloquent areas (GLIOMAP): a propensity score-matched analysis of an international, multicentre, cohort study. Lancet Oncol. 2022;23:80217. [DOI] [PubMed]
    Gerritsen JKW, Dirven CMF, De Vleeschouwer S, Schucht P, Jungk C, Krieg SM, et al. The PROGRAM study: awake mapping versus asleep mapping versus no mapping for high-grade glioma resections: study protocol for an international multicenter prospective three-arm cohort study. BMJ Open. 2021;11:e047306. Erratum in: BMJ Open. 2022;12:e047306corr1. [DOI] [PubMed] [PMC]
    Gerritsen JKW, Klimek M, Dirven CMF, Hoop EO, Wagemakers M, Rutten GJM, et al. The SAFE-trial: Safe surgery for glioblastoma multiforme: awake craniotomy versus surgery under general anesthesia. Study protocol for a multicenter prospective randomized controlled trial. Contemp Clin Trials. 2020;88:105876. [DOI] [PubMed]
    Wu A, Lim M. The challenges and future of immunotherapy for gliomas. Cancer J. 2021;27:3718. [DOI] [PubMed]
    Liang M, Gao C, Wang Y, Gong W, Fu S, Cui L, et al. Enhanced blood-brain barrier penetration and glioma therapy mediated by T7 peptide-modified low-density lipoprotein particles. Drug Deliv. 2018;25:165263. [DOI] [PubMed] [PMC]
    Agarwala SS, Kirkwood JM. Temozolomide, a novel alkylating agent with activity in the central nervous system, may improve the treatment of advanced metastatic melanoma. Oncologist. 2000;5:14451. [DOI] [PubMed]
    Mehta AM, Sonabend AM, Bruce JN. Convection-enhanced delivery. Neurotherapeutics. 2017;14:35871. [DOI] [PubMed] [PMC]
    Gregory JV, Kadiyala P, Doherty R, Cadena M, Habeel S, Ruoslahti E, et al. Systemic brain tumor delivery of synthetic protein nanoparticles for glioblastoma therapy. Nat Commun. 2020;11:5687. [DOI] [PubMed] [PMC]
    Shah AH, Heiss JD. Neurosurgical clinical trials for glioblastoma: current and future directions. Brain Sci. 2022;12:787. [DOI] [PubMed] [PMC]
    Vogelbaum MA, Brewer C, Barnett GH, Mohammadi AM, Peereboom DM, Ahluwalia MS, et al. First-in-human evaluation of the Cleveland Multiport Catheter for convection-enhanced delivery of topotecan in recurrent high-grade glioma: results of pilot trial 1. J Neurosurg. 2019;130:47685. [DOI] [PubMed]
    Piperi C, Papavassiliou KA, Papavassiliou AG. Pivotal role of STAT3 in shaping glioblastoma immune microenvironment. Cells. 2019;8:1398. [DOI] [PubMed] [PMC]
    McNutt M. Cancer immunotherapy. Science. 2013;342:1417. [DOI] [PubMed]
    Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of age. Nature. 2011;480:4809. [DOI] [PubMed] [PMC]
    Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015;27:45061. [DOI] [PubMed] [PMC]
    Rong L, Li N, Zhang Z. Emerging therapies for glioblastoma: current state and future directions. J Exp Clin Cancer Res. 2022;41:142. [DOI] [PubMed] [PMC]
    Gholamin S, Mitra SS, Feroze AH, Liu J, Kahn SA, Zhang M, et al. Disrupting the CD47-SIRPα anti-phagocytic axis by a humanized anti-CD47 antibody is an efficacious treatment for malignant pediatric brain tumors. Sci Transl Med. 2017;9:eaaf2968. [DOI] [PubMed]
    Vijayan D, Young A, Teng MWL, Smyth MJ. Targeting immunosuppressive adenosine in cancer. Nat Rev Cancer. 2017;17:765. Erratum for: Nat Rev Cancer. 2017;17:709–24. [DOI] [PubMed]
    Caccese M, Padovan M, D’Avella D, Chioffi F, Gardiman MP, Berti F, et al. Anaplastic astrocytoma: state of the art and future directions. Crit Rev Oncol Hematol. 2020;153:103062. [DOI] [PubMed]
    Kaley T, Touat M, Subbiah V, Hollebecque A, Rodon J, Lockhart AC, et al. BRAF inhibition in BRAFV600-mutant gliomas: results from the VE-BASKET study. J Clin Oncol. 2018;36:347784. [DOI] [PubMed] [PMC]
    Bahleda R, Italiano A, Hierro C, Mita A, Cervantes A, Chan N, et al. Multicenter phase I study of erdafitinib (JNJ-42756493), oral pan-fibroblast growth factor receptor inhibitor, in patients with advanced or refractory solid tumors. Clin Cancer Res. 2019;25:488897. [DOI] [PubMed]
    Schindler G, Capper D, Meyer J, Janzarik W, Omran H, Herold-Mende C, et al. Analysis of BRAF V600E mutation in 1,320 nervous system tumors reveals high mutation frequencies in pleomorphic xanthoastrocytoma, ganglioglioma and extra-cerebellar pilocytic astrocytoma. Acta Neuropathol. 2011;121:397405. [DOI] [PubMed]