Next Article in Journal
A Clinico-Genetic Score Incorporating Disease-Free Intervals and Chromosome 8q Copy Numbers: A Novel Prognostic Marker for Recurrence and Survival Following Liver Resection in Patients with Liver Metastases of Uveal Melanoma
Previous Article in Journal
A Novel Chimeric Oncolytic Virus Mediates a Multifaceted Cellular Immune Response in a Syngeneic B16 Melanoma Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Subependymal Giant Cell Astrocytoma: The Molecular Landscape and Treatment Advances

by
Emanuela Pucko
1,
Dorota Sulejczak
2,* and
Robert P. Ostrowski
1,*
1
Department of Neurooncology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5 St., 02-106 Warsaw, Poland
2
Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5 St., 02-106 Warsaw, Poland
*
Authors to whom correspondence should be addressed.
Cancers 2024, 16(19), 3406; https://doi.org/10.3390/cancers16193406
Submission received: 26 August 2024 / Revised: 27 September 2024 / Accepted: 4 October 2024 / Published: 7 October 2024
(This article belongs to the Section Molecular Cancer Biology)

Abstract

:

Simple Summary

Subependymal giant cell astrocytoma is a slow-growing brain tumor affecting children and young adults. Despite the characteristic molecular and clinical features, the severity of the disease varies from patient to patient, and, in addition, tumors in their early stages can be asymptomatic for a long time. Morbidity and mortality are due to the location of the tumor, which can obstruct the flow of cerebrospinal fluid, leading to progressive hydrocephalus and even death. The etiology and the response to treatment are still poorly understood due to the diversity of the background and the course of the disease. Therefore, this review aims to present and discuss the latest research on SEGA, its diagnosis, and treatment.

Abstract

Subependymal giant cell astrocytoma (SEGA) is most often found in patients with TSC (Tuberous Sclerosis Complex). Although it has been classified as a benign tumor, it may create a serious medical problem leading to grave consequences, including young patient demise. Surgery and chemotherapy belong to the gold standard of treatment. A broader pharmacological approach involves the ever-growing number of rapalogs and ATP-competitive inhibitors, as well as compounds targeting other kinases, such as dual PI3K/mTOR inhibitors and CK2 kinase inhibitors. Novel approaches may utilize noncoding RNA-based therapeutics and are extensively investigated to this end. The purpose of our review was to characterize SEGA and discuss the latest trends in the diagnosis and therapy of this disease.

1. Introduction to Subependymal Giant Cell Astrocytoma Etiology, Clinical Presentation, and Diagnostics

Subependymal giant cell astrocytoma (SEGA) is a rare benign tumor of astrocytic origin characterized by poor growth [1]. This tumor is included among first-degree brain tumors, according to the World Health Organization (WHO) [2] and accounts for about 2% of all pediatric tumors [3]. SEGA is often diagnosed in the first or second decade of life, although it can be detected in newborns and fetuses [4]. SEGAs originate from subependymal nodules (SENs), which most often do not cause symptoms in the patient [5]. SEGAs often grow on the side of the wall of the lateral ventricle, in the area of Monro’s septum, and they can be found in the third ventricle as well [6]. Although they are benign tumors, their location and growth potential can pose a risk. Despite their small size and slow growth, when they enlarge, they may block the flow of cerebrospinal fluid, which leads to a number of different clinical symptoms, including increased intracranial pressure associated with headaches, photophobia, double vision, and ataxia [7]. Blockage of CSF passage may also lead to hydrocephalus, an increase in intracranial pressure, and possibly fatal outcomes [6]. Additionally, SEGA may also produce seizures, blurred vision, and behavioral dysfunction. Despite being grade 1 of histological malignancy, clinically aggressive cases have also been reported [6,8,9].
SEGA diagnosis involves clinical assessment and imaging techniques. Magnetic resonance imaging shows tumors as areas of contrast enhancement in the characteristic locations described above. MRI allows detection and localiza tion of the tumor(s), assessment of their size and rate of growth, and assessment of their potential to cause hydrocephalus. A variety of methods of SEGA tumor size assessment in MRI studies have been so far applied, for example, ITK-Snap (pixel clustering, geodesic active contours, region competition methods), 3D Slicer (level-set thresholding), and NIRFast (k-means clustering, Markov random fields) [10]. SEGA is a well-defined/circumscribed tumor mass on a radiology scan, be it an MRI or CT. Neuroimaging after surgery may also visualize residual tumors in cases of incomplete resection. Gadolinium enhancement and tumor growth on consecutive neuroimaging scans allow us to distinguish SEGA from SEN. MRI surveillance currently allows the diagnosis of the majority of SEGA cases even before hydrocephalus develops. It is very important that patients with SEGA diagnosed in childhood undergo scrupulous brain neuroimaging assessment on a regular basis [11,12,13].
As mentioned, SEGAs can initially be asymptomatic. However, as the tumor(s) grow, neurological symptoms may appear. Therefore, neurological examinations are very important in diagnosing SEGA and, subsequently, in patient care. Symptoms such as nausea, headaches, or visual disturbances may indicate tumor growth. Genetic testing is not routinely performed for the diagnosis of SEGA, but the detection of mutations characteristic of TSC can help in diagnosing the disease [5].
In this review, we summarize SEGA etiology and classification alongside the clinical presentation and diagnostic information in the introductory chapter. Then, we cover the neuropathological characteristics of SEGA and SEN and move to the condition principally associated with SEGA, i.e., Tuberous Sclerosis Complex (TSC). We describe TSC etiology and genetic background as well as the genetic and molecular mechanisms of SEGA formation upon TSC. The therapeutic approach chapter deals with both conservative and candidate therapies, the latter emerging from experimental and clinical studies. The pathogenetic background of SEGA and tumor biology, as well as conservative and novel therapeutic approaches, are supported by Tables and Figures summarizing the relevant information. We conclude on the insidious nature of SEGA tumors that, despite being non-malignant tumors, require novel therapies, especially for pediatric cases with unsatisfactory outcomes of standard therapy.

2. Neuropathological Characteristics of SEGA

SEGA is characterized by neuronal–glial structure and a low proliferation index. SEGA-forming cells are large and polygonal or epithelioid cellular in shape with abundant cytoplasm and vascular–parenchymal stroma alongside the presence of vascular calcifications [1]. As SEGA cells are capable of differentiating bidirectionally, the expression of glial and neuronal markers may coexist within the same tumor cells. Bidirectional cell differentiation may be the result of a defect in progenitor cell differentiation during brain development [14]. Thus, in descriptive terms, SEGAs are composed of ganglion-like astrocytes. These are large cells of the gemistocytic type, with abundant eosinophilic cytoplasm and a peripheral location of the nucleus and distinct nucleolus [15,16].
Part of the SEGA tumor-forming cells shows astroglial differentiation, reflected by immunoreactivity of glial fibrillary acidic protein (GFAP) and S100, whereas beta-tubulin class III protein and features of neuronal differentiation in the form of expression of neurofilament proteins and neuron-specific enolase are present in a distinct subpopulation of tumor cells [1]. Interestingly, some of the cells do not express GFAP, and some cells only weakly express neurofilaments [17]. SEGA also showed the expression of markers present in progenitor cells emerging from the periventricular zone around the lateral ventricle. Bidirectional differentiation of cancer cells may, therefore, result from a defect in the differentiation of progenitor cells during brain development [14,18]. The prototype SEGA cells are radial glial cells that play a role during embryonic development, especially upon neuronal migration towards gray matter structures in the course of maturation of the central nervous system (CNS) [19].
SEGA tumor shows variable immunoreactivity. Therefore, specific antibodies are needed to immunolabel both glial and neuronal markers of SEGA cells. TTF-1, a 38 kDa DNA-binding protein, is encoded by the homeobox gene NKX2-1 located on chromosome 14q13 and is temporarily expressed during embryonic development of the ventral forebrain [20]. Detailed animal studies have demonstrated the involvement of thyroid transcription factor (TTF-1) in cells of the midbrain and diencephalic origin of the developing brain [21,22]. TTF-1 has also been shown to be transiently expressed in some brain areas in the postnatal period [23]. It seems important that TTF-1 protein expression can be examined when diagnosing primary brain tumors of uncertain nature [24]. TTF-1 expression has been detected in brain tumors, including the pituitary gland and spindle cell oncocytoma [25]. Interestingly, TTF-1 as a marker can influence SEGA histiogenesis. Studies have revealed a sizable mRNA content of TTF-1 in ependymal and subependymal cells of the third ventricle, pointing towards the diagnostic utility of this marker in distinguishing SEGAs from its mimics, especially considering that TTF-1 immunopositivity was seen in all cases of SEGA [20].
SEGA is difficult to clearly define histopathologically because SENs and SEGA are oftentimes barely distinguishable. SENs are hyperdense, often calcified nodular masses present in the majority of TSC patients in whom they arise embryonically but also during the neonatal period. SENs are composed of spindle GFAP-positive cells intermingled with groups of balloon cells, which are often PAS and vimentin-positive. SENs originate from the subependymal zone of the periventricular region and can also be found near the caudate nucleus or the foramen of Monro. SENs are classified as a major feature for the diagnosis of TSC [26]. In addition to that, both SEN and SEGA show increased levels of cytoplasmic proteins phospho-S6K, phospho-S6, and phospho-Stat3 downstream of mTORC1 [27].
However, the size of SEN is less than 5 mm (while SEGA > 5 mm), and the growth is lacking in most instances. They are also characterized by the lack of contrast enhancement on CT and MRI, in contrast to SEGA [28,29]. SEGA, but not SEN, may manifest itself with hydrocephalus, focal neurological deficit, or symptoms of increased intracranial pressure. However, approximately 5–15% of TSC SENs transform into SEGAs [30]. Interestingly, in the analysis of the expression of Akt, Erk, and mTOR (mammalian target of rapamycin) pathways in SEN and SEGA by the team of Siedlecka and colleagues, it was shown that there is an upregulation of p-Erk, p-Mek, or p-RSK1 in SEGA but not in the SEN sample, while p-Akt, p-GSK3β, and p-PDK1 were upregulated in both SEN and SEGA from the same tuberous sclerosis disease patient [31]. Those authors proposed a research hypothesis that activation of PI3K/Akt leads to upregulation of mTOR. According to their hypothesis, it is the activation of Erk that triggers the transformation of SEN into SEGA [31]. Many authors also report that dysregulation of the mTOR pathway leads to uncontrolled cell division and tumor development [5,32,33].

3. Tuberous Sclerosis Complex

SEGA tumors are most often diagnosed in patients with Tuberous Sclerosis Complex, which is also known as Bourneville-Pringle disease [3]. It is a genetic disorder that eventually leads to benign tumors in various organs of the body, including the heart, kidneys, lungs, skin, brain, and heart [5]. Data show that SEGAs occur in approximately 5–20% of patients with TSC [34,35,36,37]. However, there are also cases of SEGA in patients without clinical symptoms of TSC [38,39,40], and even though it is a rare phenomenon, it is estimated that there are approximately 48 cases of patients described in the literature, counting from the time of the first patient described by Hyalmagi et al. in 1979 [41,42]. The majority of TSC cases are associated with a sporadic mutation (de novo mutation) [43,44], while a smaller number of patients inherit the mutated gene from one of their parents in an autosomal dominant manner [45]. Sporadic TSC cases more often result from TSC2 than TSC1 mutations. Those cases harbor large deletions, missense mutations, and splice-junction mutations of TSC2 (mostly exons 16, 33, and 40) and mostly small deletions and nonsense mutations for TSC1 cases (predominately exons 15 and 17) [46,47]. Familial TSC cases with biallelic inactivation of TSC1 or TSC2 are caused by their mutations (including nonsense mutations, deletions, and splice-site mutations), with more frequent than in sporadic cases involvement of TSC1 [48,49,50,51].
TSC disease is extensive, multi-organ, and very heterogeneous in terms of phenotype, manifesting itself in a wide spectrum of symptoms, including skin lesions, hyperplastic lesions within the CNS, and changes in internal organs, including the kidneys, liver, heart, and lungs [52,53]. The diagnosis of TSC is based on the presence of a pathogenic mutation or the presence of two major or one major and two minor symptoms, according to the diagnostic criteria developed by the 2021 International Tuberous Sclerosis Complex Consensus Group [54]. It is important to stress that genetic testing is recommended for all patients who cannot be diagnosed on the basis of clinical symptoms alone [55].
TSC may manifest several principal intracranial pathological entities, including cortical tubers and SEGAs [56]. SENs are small lesions located in the subependymal space of the lateral ventricle; their characteristics, as determined by histopathological analysis, resemble that of SEGA, while differences comprise smaller size, absence of growth, and absence of contrast enhancement of SEN on MRI with gadolinium compared to SEGA. Patients’ cortical tubers comprise abnormal neurons and glial cells located in the cerebral cortex resulting from abnormal cellular differentiation and disturbed neuronal migration. Cortical tubers have been linked with autism and epilepsy associated with TCS [32]. However, non-growing cortical tubers are most often clinically benign, unlike a large and growing SEGA tumor that can lead to life-threatening symptoms [57].
The International TSC Clinical Consensus Group has recommended that independent genetic identification of pathogenic variants of the TSC1 or TSC2 gene should be sufficient to diagnose tuberous sclerosis disease regardless of clinical data. Furthermore, based on two major or one major and two minor clinical criteria, a clinical diagnosis of the disease can be established [54]. Literature data show that increasingly often, patients are diagnosed prenatally using magnetic resonance imaging (MRI) or ultrasound examination, and already at this stage, the changes in fetal heart and brain related to tuberous sclerosis disease can be observed [58].

4. Genetic Background of TSC

Tuberous sclerosis TSC is most commonly associated with a mutation in one of two tumor suppressor genes: the TSC1 gene (which codes for hamartin), located on chromosome 9 (9q34), or the TSC2 gene (which codes for tuberin), located on chromosome 16 (16p13.3) [11,59] (Table 1). Mutations in the TSC1 or TSC2 genes are not observed in only 20% of TSC patients. Additionally, the disease in such patients without mutations has a less severe course [60]. Interestingly, isolated cases of SEGA without mutations in the TSC1/TSC2 genes caused by epigenetic changes in tuberin or hamartin have been described [61]. TSC2 gene mutations occur four times more often than TSC1 gene mutations and occur in approximately one-third of all TSC patients, but unfortunately, they also determine a more severe clinical course. Animal studies show that the TSC2 gene mutation is a more common cause of severe phenotypic features, including neurological symptoms, including epilepsy, and patients with PKD (polycystic kidney disease) and TSC2 mutations are more often exposed to early symptoms of polycystic kidney disease [6].
As already mentioned, the TSC1 gene encodes a protein called hamartin with a molecular weight of 130 kDa, and the TSC2 gene encodes tuberin with a molecular weight of 180 kDa. Both proteins form a protein complex called TSC1/TSC2 heterodimer and limit the activity of the mTORC1 (mammalian target of rapamycin complex 1-mTORC1) signaling pathway, which controls various cell functions, such as growth, survival, and proliferation, by inhibiting the small Ras GTPase homolog enriched in the brain (Rheb). When hamartin–tuberin is inactive, increased levels of active Rheb continuously activate the mTOR pathway, specifically mTORC1, and subsequently the phosphorylation of S6K1 and 4E-BP1 proteins [62].
The mTOR kinase belongs to a class of evolutionarily conserved threonine and serine kinases. mTOR is composed of two separate multi-subunit complexes called mTOR complex 1/2 (mTORC1/2). mTOR kinase catalyzes the phosphorylation of insulin growth factor receptor (IGF-1R), AKT kinase, 4E-binding protein 1 (4E-BP1), ribosomal protein S6 kinase (S6K), and transcription factor EB (TFEB). The signaling pathways that include the mTOR kinase are responsible for cell growth, survival, proliferation, cell migration, protein translation, nucleotide synthesis, lysosome biogenesis, and many other processes [63]. Unfortunately, mutations, amplifications, or deletions in genes related to mTOR or its complexes (mTORC1 and mTORC2) change the functioning of the mTOR signaling pathway and lead to uncontrolled cell proliferation, avoidance of apoptosis, and the development of neoplasms, including brain tumors [64].
Additionally, oxygen deprivation stress and hypoxia itself induce mTORC1 inhibition through AMPK activation and REDD1 induction, which results in TSC activation. In contrast, induction of DNA damage response signaling inhibits mTORC1 through the induction of p53 target genes such as REDD1, LKB AMPKβ, PTEN, and TSC2 [65].
Another multiprotein complex of mTOR, i.e., mTORC2, consists of mTOR, GβL/mLST8, Rictor (rapamycin-insensitive companion of mTOR), Protor/PRR5 (proline-rich protein 5), DEPTOR, and mSIN1 (mammalian stress-activated protein kinase-interacting protein 1) involved in the regulation of cell proliferation, survival, and migration. mTORC2 is activated by growth factors, which in turn activate several signaling pathways associated with tuberous sclerosis receptor tyrosine kinases (RTKs), IGF-1, Wnt, TNFα, inflammatory cytokines, and the Ras signaling cascade. RTK-mediated Ras signaling was found to activate the mTORC1 pathway via MAPK/ERK and its effector p90RSK [66]. The activation of the pathway may also occur through activating mutations in mTOR and mTORC1 and overexpression/amplification of mTORC1 and mTORC2 components.
Table 1. The genetic alterations upon TSC and SEGA.
Table 1. The genetic alterations upon TSC and SEGA.
ConditionMutations Reported Reference
TSC, familialFrameshift > splicing, nonsense TSC1 mutations[67]
TSC, familialLower proportion of TSC2 mutations in familial cases of TSC than in de novo cases; Dominant in frequency were frameshifts followed by nonsense mutations [51]
TSC, familialIdentification of TSC1 mutations appears to be twice as likely in familial cases as in sporadic cases. Mutations in TSC2 are associated with more severe diseases, including seizures and cognitive dysfunction.[50]
TSC, familialGermline
mutations in TSC1 (9q34.3) encoding hamartin
and TSC2 (16p113.3) encoding tuberin
[68]
TSC, sporadicSmall TSC2 mutations > small TSC1 mutations > large TSC2 mutations[46]
TSC, sporadicNonsense, missense > indels among TSC2 mutations[48]
TSC, sporadicSporadic TSC cases more often result from TSC2 than TSC1 mutations[49]
TSC, sporadicPathogenic variants in TSC2 > TSC1: 23% nonsense, 22% missense, 19% splice, 18% deletions, 8% large deletions, 2% in-frame deletions[69]
SEGA; TSC-relatedTSC2 gene deletions affecting the adjacent PKD1 have the highest risk of early SEGA development.[70]
SEGA; TSC-relatedTSC1 nonsense > deletions > insertions; missense or TSC2 nonsence > deletions, splice sites > missense; insertions. Also found were somatic mutations in genes involved in transcriptional and translational regulation, cell cycle regulation, signal transduction, cell adhesion, resistance to anti-cancer drugs, energy metabolism, ubiquitin–proteasome system functioning, immune homeostasis, and cytoskeleton stabilization.[71]
SEGA-like; solitaryNF1 splice
mutations
[72]
SEGA; solitaryTSC1 or TSC2 mutation
limited to the tumor
[73]
SEGA; solitaryTSC2 somatic mosaic mutation, including extra-tumor tissues[74]
SEGA; solitaryEGFR amplification, CDKN2A/B homozygous deletion, chromosomal +7/−10 alterations, and TERT promoter mutation, typical molecular abnormalities usually found in GBM, were observed.[9]

5. The Mechanisms of Sega Formation in Tuberous Sclerosis

It is worth mentioning that theories regarding the origin of SEGAs are controversial partly due to a limited amount of research, including a limited set of data on the expression of the mTOR pathway in SEGA cells. Even though it cannot be concluded that a specific pathway is responsible for the formation of tumors, it can be assumed that the mTOR pathway plays a large role in the formation of SEGA [75,76]. Despite reports that showed a mutation and loss of heterozygosity in TSC2 in SEGA, one belief implying that the formation of SEGA is caused only by a mutation of the TSC1 or TSC2 gene has been challenged as being incorrect, mainly because cases of patients with SEGA without clinical symptoms associated with tuberous sclerosis and without TSC1/TSC2 mutations have been described [39,61,77]. The analysis of TSC1/TSC2 mutations in SEGA DNA from 13 patients showed that nine patients had a TSC2 mutation, two had a TSC1 mutation, and two had no mutation [78].
In the literature, there are several other hypotheses regarding the mechanisms of SEGA tumor development. One of those is that SEGA is associated with mosaicism, which quite frequently occurs in TSC (up to 15% of cases), although it may lead to SEGA without TSC as well [79,80].
On the other hand, SEGA patients may have two independent inactivating somatic mutations in TSC1 or TSC2, and thereby, both copies of the TSC1 or TSC2 gene are lost in the cells that form the tumor, but the mutations in other patients’ cells are absent [6]. Nevertheless, patients with SEGA should be carefully examined for features of TSC by physicians with expertise in TSC and subsequently monitored for an extended period of time, especially in pediatric cases [78].
Interestingly, the research revealed many genes potentially involved in the development of SEGA, including the ANXA1, GPNMB, LTF, RND3 and NPTX1 S100A11 genes, sFRP4, which may act as the effector genes in SEGA, and their expression can be modulated by rapamycin [45,81,82].
In addition, Akt phosphorylation was increased in SEGA, and a high level of proteins indicating mTOR activation was also found, including phospho-S6K, phospho-S6, and phospho-STAT3 [83]. Moreover, SEGA expressed the immunoreactive phosphorylated isoforms of MEK1/2 and ERK1/2, which in turn might indicate incorrect MAP kinase signal transduction [82].
In SEGA and in cortical tumors of patients with TSC1, there is an increased expression of growth factors and their receptors, including VEGF and HIF1α (hypoxia-inducible factor-1 α), a transcription factor that regulates the level of VEGF, which contributes to excessive cell growth and proliferation. VEGF modulates the activity of the mTOR pathway cascade, which is activated in TSCs [84].

6. Therapeutic Approach for SEGA Tumors

In recent years, the therapeutic approach to the treatment of SEGA has changed. Until recently, surgery used to be the standard procedure, especially in cases of acute clinical cases, but unfortunately, complete removal of the lesion was not possible in all cases [85,86] (Table 2).
Surgeries carried a risk of postoperative complications, which include headaches, memory deficits, paresis, acute hydrocephalus, intratumoral hemorrhages, cerebral infarction, secondary sepsis, meningitis, epilepsy, cardiac arrest, and lung infection leading to respiratory failure [32,90,91]. Additionally, surgical treatment of brain tumors, especially in children, was associated with postoperative neurological deficits and higher mortality compared to other age groups [92]. Another incentive to search for methods other than tumor resection was tumor recurrence, which requires reoperation and may result in further clinical complications and even the death of the patient [32,93].
SEGA tumors respond slowly and progressively to fractionated radiotherapy, similar to other low-grade intracranial tumors. One SEGA tumor’s response to intercurrent everolimus administration suggests an additive effect of radiation and that the drug could be clinically exploitable. Everolimus leads to reduced tumor volume, which facilitates more focused radiation that reduces the radiation-induced side effects as compared with pancranial irradiation. In summary, patients without hydrocephalus may be a population in which induction treatment with everolimus followed by fractionated stereotactic radiotherapy could be an alternative to surgery [12].
Recently, however, the discovery of molecules responsible for the pathogenesis of cancer has led to the introduction of targeted pharmacotherapies to SEGA treatment aimed at inhibiting various kinases and signaling pathways, including mTOR kinase [3,36,81,83,94,95,96,97,98]. For some time, recommendations on SEGA treatment assessed the effectiveness of mTOR inhibitors, but their place in the treatment strategy was variable. In 2021, recommendations for everolimus were updated, which is now recommended for people with asymptomatic SEGA growth, patients with mild and moderate symptoms, and those who do not qualify for surgery or prefer medical treatment to surgery [54].
The discovery of mTOR kinase inhibitors dates back to the 1970s, when a natural substance with antifungal properties was isolated from the bacteria Streptomyces hygroscopicus, found in the soil of the island of Rapa Nui (Easter Island, Chile), and therefore the isolated substance was named rapamycin in honor of the island of Rapa Nui. Interestingly, rapamycin, in addition to its antifungal properties, also showed anticancer activity, which was described in 1984 by Eng and colleagues [99]. The identification of rapamycin targets sparked interest in the therapeutic potential of rapamycin in anticancer therapy [99,100]. Scientists have shown that the expression of FKBP12 and FKBP51 is the rate-limiting factor that determines the response to the drug rapamycin in cell lines and tissues. This discovery triggered the synthesis of synthetic/semi-synthetic derivatives of rapamycin that were active against mTOR [101]. Sirolimus is a biochemical, functional form of rapamycin that disrupts the molecular interaction between mTOR and Raptor by targeting mTORC1 and is approved for the treatment of TSC manifestations, including SEGA. Sirolimus was also approved as an immunosuppressant used in organ transplantation and in the treatment of lymphangioleiomyomatosis. In addition to that, sirolimus is now being tested clinically as a preventive treatment for TSC [64,89]. Nab-sirolimus (sirolimus based on albumin-bound nanoparticles) showed an antitumor effect in perivascular epithelial cell tumors [102]. Studies using Nab-sirolimus are underway in patients with a variety of tumors with genetic mutations in the mTOR pathway and TSC1 and TSC2 [103].
Everolimus was designed and synthesized as an immunosuppressive drug by replacing the H of the hydroxyl group (C-40) with a hydroxyl group [104]. Everolimus was approved for the treatment of advanced, non-functional neuroendocrine tumors of the lung or gastrointestinal tract [105]. This drug is administered orally, is metabolized by the CYP3A enzyme, and can penetrate into the brain. Everolimus treatment has been shown to reduce the size of SEGAs and the frequency of seizures [88]. Rapamycin and its derivatives, temsirolimus and everolimus, are indicated in the treatment of SEGA patients who are not eligible for surgical treatment [106]. The action of the rapamycin compound is to form a rapamycin-acceptor protein FKBP12 (FRB-FK506 binding protein 12) complex; mTOR binds to FKBP12 and rapamycin via the FKBP-rapamycin binding (FRB) domain [107,108] (see Figure 1). Thus, FKBP12 inhibits the activity of mTOR and the activity of mTOR effector proteins (p70S6K and 4E-BP1). Consequently, this causes the accumulation of cells in the G1 phase of the cell cycle and the induction of apoptosis [109]. In order to oppose the abnormal PI3K-Akt-mTOR signaling, the use of dual PI3K/mTOR inhibitors is advocated, as delineated below, among others, due to interrupting the rapalog-induced negative feedback loop that augments Akt activation [110]. Moreover, other inhibitors of mTOR kinase, i.e., ATP competitive inhibitors, are being developed to overcome the drawbacks of rapalogs, including immune suppression and feedback activation [111]. Other therapeutic attempts utilized targeting different groups of kinase, including those of the MAPK family for SEGA treatment, appear less effective. For example, inhibiting ERK signaling using the U0126 inhibitor significantly reduces SEGA cell proliferation and migration but does not affect the size of cancer cells [81,82].
Everolimus is indicated as an alternative therapy for large SEGA tumors in patients with TSC [87,112,113,114,115]. This treatment allows for an initial rapid reduction in the tumor mass and subsequent stabilization of its growth [116,117]. However, it turned out that after discontinuing the drug, the cancer process quickly recurs, prompting us to examine the safety endpoints of long-term pharmacotherapy with mTOR inhibitors [118]. Thus, continuous and long-term therapy with a risk of tumor recurrence after discontinuation of treatment can be seen as a disadvantage of the pharmacological approach to SEGA. As with any therapy, patients may experience side effects, e.g., aphthous ulcers, acne rash, inflammation of the oral mucosa, hematological complications, fever, nasopharyngitis, fatigue, otitis media, and upper respiratory tract infections [114,119]. There is also a risk of reduced insulin secretion and insulin resistance, pneumonia, sepsis, and amenorrhea, while the long-term side effects of mTOR are largely unknown [106,120,121].
Amongst studies on potential treatments for SEGA that may become standard of care treatments over time, several approaches can be named, including repositioned drugs, novel anticancer drugs, gene therapy, and novel surgical techniques (Table 3).
It is also worth mentioning that dual PI3K/mTOR inhibitors exert anticancer effects and can be considered for the treatment of SEGA brain tumors. It is known for the fact that SEGA, although assigned WHO grade 1 of histological malignancy, may resemble malignant gliomas due to its atypical histological features present at times. SEGA and malignant gliomas have a substantial portion of oncogenic signaling pathways and vulnerabilities in common, and several therapeutic agents developed for malignant gliomas originally might as well work for SEGA. This notion is supported by the results from clinical research and quite a few experimental works in cell lines derived from both high-grade and low-grade astrocytic tumors [127,133,134,135].
Paxalisib as a PI3K/mTOR inhibitor is in use for patients with newly diagnosed GBM with unmethylated MGMT promoter status following surgical resection and initial chemoradiation with temozolomide [124]. Paxalisib has entered a clinical trial for diffuse midline glioma as a monotherapy or in combination with ONC201, a TRAIL inducer [122,136]. Paxalisib can cross the blood–brain barrier and inhibit PI3K, a master regulator of neoplasm cell growth and cell division activated in SEGA; hence, it has the potential for the treatment of this astrocytoma [123].
Apitolisib (GDC-0980), a novel dual PI3K/mTOR inhibitor of mTORC1/2 and PI3K class I tested in solid tumors, including those of the brain, inhibits growth and induces apoptosis in human glioblastoma cells. Apitolisib (GDC-0980) induces time- and dose-dependent cytotoxicity and apoptosis in the tested A-172 and U-118-MG GBM glioma cell lines; the strongest apoptosis induction was demonstrated in the A-172 line after 48 h of incubation with 20 μM GDC-0980, where 46.47% of cells were apoptotic. Researchers found that dual PI3K/mTOR blockade by GDC-0980 significantly suppressed human GBM cell survival and induced apoptosis [137].
Voxtalisib has demonstrated synergistic effects with low-intensity pulsed ultrasound to inhibit tumorigenesis in glioblastoma’s CSCs while inhibiting PI3K/AKT/mTOR signaling [138]. Others have demonstrated synergistic effects of voxtalisib in combination with temozolomide and radiotherapy for glioma [139].
A promising drug that has been shown to inhibit the mTOR protein kinase is metformin, which is used in the treatment of type 2 diabetes. Interestingly, metformin has also shown anticancer properties [140]. One research group conducted a study where they determined the effect of metformin in patients with tuberous sclerosis. It was observed that patients taking metformin had a reduction in SEGA tumor volume compared to placebo and a reduction in the frequency of epileptic seizures. It is likely that the beneficial effect is mainly due to the inhibitory effect of metformin on the mTOR pathway via activation of adenosine monophosphate-activated protein kinase (AMPK) [130]. Importantly, metformin penetrates the blood–brain barrier and is distributed in many areas of the brain after oral administration [141,142]. Moreover, metformin does not interact with the cytochrome p450 system; therefore, it is unlikely to interfere with the metabolism of other mTOR inhibitors, including everolimus and rapamycin. It has been shown that the metabolism of metformin is not disturbed by antiepileptic drugs such as cannabidiol or carbamazepine [130,143,144].
CK2 kinase inhibitors are also interesting candidate compounds for SEGA treatment, which have shown a reduction in cell viability and proliferation of the T98G malignant glioma cell line and the SEGA cell line, derived from a pediatric case of tuberous sclerosis complex (TSC). Cell cultures were incubated with selected CK2 inhibitors, including 4,5,6,7-tetrabromo-1H-benzimidazole (TBI), 2-dimethylamino-4,5,6,7-tetrabromo-1H-benzimidazole (DMAT), and 4,5,6,7-tetrabromo-1H-benzotriazole (TBB). Studies have shown that the tested CK2 inhibitors reduce cell growth and viability, but the strongest cytotoxic effect on SEGA and T98G cells was caused by the TBI inhibitor [127]. Promising compounds appear to be pentabromobenzyl isothiourea bromide derivatives, which cause a reduction in cell viability and proliferation of the T98G malignant glioma cell line and the SEGA cell line, derived from a pediatric case of TSC. Studies have shown that among those compounds, the best anti-proliferative effect was demonstrated by the compound ZKK13 [145]. The mainstay of current treatment options for SEGA is depicted in Figure 2.

7. Non-Pharmacological Treatment Modalities and Biopharmaceuticals

It is worth considering other treatment methods for SEGA, e.g., endoscopic removal of tumors. The advantage of this method is the possibility of adding a septostomy to the tumor resection, but the disadvantage is that this method is applicable to tumors less than 3 cm in size and the wide attachment of the tumor to the basal ganglia [146].
Recently, laser methods have been substantially developed, including laser interstitial thermal therapy (LITT). Amongst the limitations of its use are tumor size < 2 cm and wide attachment of the tumor to the base. Moreover, active hydrocephalus is a contraindication to LITT in SEGA due to a risk of acute hydrocephalus and swelling of the basal ganglia associated with LITT [128,129,146,147]. Thus, although LITT is a promising method, there are no long-term treatment results so far [128,148].
Another form of therapy is the use of radiosurgery using the Gamma Knife™ method, which may have the effect of reducing the size of the SEGA tumor, but unfortunately, this therapy carries an additional burden, which is the risk of developing secondary radiation-induced tumors. Moreover, such therapy is not suitable for large tumors accompanied by hydrocephalus [94,132,148].
In recent years, research advances in SEGA molecular biology have also provided the background for emerging novel therapies. Gene therapy on lentivirus restoring human TSC1 reduced growth and proliferation of TSC1-deficient neural cells. TSC1 replenishment, along with Rapamycin, further decreased proliferation and growth in TSC1-deficient tumors in mice [126].
Moreover, alterations of non-coding RNAs that may result in the dysregulation of matrix metalloproteinases (MMPs) and their endogenous tissue inhibitors (TIMPs) have been found in SEGA cells [131]. MMP/TIMP system abnormalities have been implicated in tumor recurrence and local neuroinflammation of SEGA tumors, among others [131]. Since miR-320d was found to be lowly expressed in SEGA cells, this miRNA or its mimetics could be utilized in prospective therapies. Although the disturbed MMP/TIMP system is not specific for SEGA, it can be controlled by different sets of non-coding RNAs across different tumor types, hence the importance of revealing the exact mechanism [149,150,151,152]. In addition, small non-coding RNAs have been implicated in the control of DNA replication, cell cycle, protein degradation, immune and hypoxic responses, as well as p53, PI3K/Akt, and MAPK signaling of SEGA cells, although further studies are needed to devise therapeutic systems based on such knowledge [125]. To this end, nanotechnologies may come in handy in overcoming the limitations in the clinical translation of ncRNA and other therapies as well [153,154,155,156].
In summary of the established and prospective treatments for SEGA, gross total resection of SEGA can be curative. Moreover, novel minimally invasive surgery techniques offer an option to reach a tumor such as SEGA with fewer side effects and complications and speedy recovery as compared to older techniques. However, with respect to surgery, there is a risk of a tumor growing back if not suppressed with chemotherapy with rapalogs.
Several authors, however, postulated a decreased role of surgery reserved for refractory cases because of favorable responses to mTOR inhibitors in terms of tumor suppression along with the betterment of clinical symptoms. Others imply that fractionated radiotherapy should be firmly included in the treatment of SEGA because the tumor slowly but progressively responds to fractionated radiotherapy. On the other hand, rapalog treatment cessation carries a risk of regrowth, as does the aftermath of radiotherapy. Thus, the combined therapies based on the above approaches and tailored to a particular patient appear to be optimal treatments until the beneficial effects of novel therapies are substantiated.

8. Conclusions

It should be emphasized that the growth of a histologically benign SEGA tumor is often associated with severe complications, including epilepsy, increased intracranial pressure, and hydrocephalus, which may lead to serious neurological deterioration and even death of the young patient. Hence, the indications for surgical interventions are solid. Preoperative administration of mTOR inhibitors to large SEGAs and tumors occurring in deep, hard-to-reach brain structures helps reduce tumor size and enable complete resection. Patients with acute obstructive hydrocephalus who have clinical symptoms of increased intracranial pressure are often treated with the combination therapy of surgery and mTOR inhibitors. They benefit from such neoadjuvant therapy based on the use of mTOR inhibitors because it reduces the size of the tumor, thus facilitating surgery and reducing the number of postoperative complications [157]. In the future, the design and synthesis of new and specific inhibitors of the mTOR pathway or mTOR-related pathways may eliminate cancer cells alone or in combination with chemotherapy drugs and immunotherapeutic agents.
It is worth emphasizing again that Nab-sirolimus (albumin-bound sirolimus nanoparticle) has been approved by the FDA for clinical use against human cancers [158]. High hopes are invested in the nanoformulation of mTOR signaling pathway inhibitors for improved effectiveness towards SEGAs and limited toxicity to healthy tissues. The new discoveries and inventions, including genes involved, therapeutic compounds, and treatment procedures, will provide grounds for significant improvement in SEGA treatment, as once the discovery of the TSC1 and TSC2 genes, rapamycin, and acting components of the mTOR pathway led to the creation of new treatment opportunities for patients with tuberous sclerosis.

Author Contributions

Conceptualization and original draft preparation, E.P.; writing—review and editing, R.P.O. and D.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Foundation for Development of Diagnostics and Therapy in Warsaw.

Acknowledgments

The authors thank Anna Eliza Lisiecka for her assistance with graphical artwork.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Buccoliero, A.M.; Franchi, A.; Castiglione, F.; Gheri, C.F.; Mussa, F.; Giordano, F.; Genitori, L.; Taddei, G.L. Subependymal giant cell astrocytoma (SEGA): Is it an astrocytoma? Morphological, immunohistochemical and ultrastructural study. Neuropathology 2009, 29, 25–30. [Google Scholar] [CrossRef] [PubMed]
  2. Buccoliero, A.M.; Caporalini, C.; Giordano, F.; Mussa, F.; Scagnet, M.; Moscardi, S.; Baroni, G.; Genitori, L.; Taddei, G.L. Subependymal giant cell astrocytoma: A lesion with activated mTOR pathway and constant expression of glutamine synthetase. Clin. Neuropathol. 2016, 35, 295–301. [Google Scholar] [CrossRef] [PubMed]
  3. Jóźwiak, S.; Mandera, M.; Młynarski, W. Natural History and Current Treatment Options for Subependymal Giant Cell Astrocytoma in Tuberous Sclerosis Complex. Semin. Pediatr. Neurol. 2015, 22, 274–281. [Google Scholar] [CrossRef] [PubMed]
  4. Grajkowska, W.; Kotulska, K.; Jurkiewicz, E.; Matyja, E. Brain lesions in tuberous sclerosis complex. Review. Folia Neuropathol. 2010, 48, 139–149. [Google Scholar] [PubMed]
  5. Navarro-Ballester, A.; Álvaro-Ballester, R.; Lara-Martínez, M. Beyond Benign: A Case of Subependymal Giant Cell Astrocytomas Provoking Hydrocephalus in Tuberous Sclerosis Complex. Acta Med. Litu. 2024, 31, 61–67. [Google Scholar] [CrossRef]
  6. Gao, C.; Zabielska, B.; Jiao, F.; Mei, D.; Wang, X.; Kotulska, K.; Jozwiak, S. Subependymal Giant Cell Astrocytomas in Tuberous Sclerosis Complex-Current Views on Their Pathogenesis and Management. J. Clin. Med. 2023, 12, 956. [Google Scholar] [CrossRef]
  7. Jansen, A.C.; Belousova, E.; Benedik, M.P.; Carter, T.; Cottin, V.; Curatolo, P.; Dahlin, M.; D’Amato, L.; d’Augères, G.B.; de Vries, P.J.; et al. Clinical Characteristics of Subependymal Giant Cell Astrocytoma in Tuberous Sclerosis Complex. Front. Neurol. 2019, 10, 705. [Google Scholar] [CrossRef]
  8. Almubarak, A.O.; Abdullah, J.; Al Hindi, H.; AlShail, E. Infantile atypical subependymal giant cell astrocytoma. Neurosciences 2020, 25, 61–64. [Google Scholar] [CrossRef]
  9. Yamada, S.; Tanikawa, M.; Matsushita, Y.; Fujinami, R.; Yamada, H.; Sakomi, K.; Sakata, T.; Inagaki, H.; Yokoo, H.; Ichimura, K.; et al. SEGA-like circumscribed astrocytoma in a non-NF1 patient, harboring molecular profile of GBM. A case report. Neuropathology 2024, 44, 190–199. [Google Scholar] [CrossRef]
  10. Stawiski, K.; Trelińska, J.; Baranska, D.; Dachowska, I.; Kotulska, K.; Jóźwiak, S.; Fendler, W.; Młynarski, W. What are the true volumes of SEGA tumors? Reliability of planimetric and popular semi-automated image segmentation methods. Magma 2017, 30, 397–405. [Google Scholar] [CrossRef]
  11. Calderón-Garcidueñas, A.L.; Piña-Ballantyne, S.A.; Espinosa-Aguilar, E.J. Subependymal Giant Cell Astrocytoma Non-Associated with Tuberous Sclerosis Complex and Expression of OCT-4 and INI-1: A Case Report. Cureus 2023, 15, e39187. [Google Scholar] [CrossRef] [PubMed]
  12. Kamel, R.; Van den Berge, D. Radiotherapy for subependymal giant cell astrocytoma: Time to challenge a historical ban? A case report and review of the literature. J. Med. Case Rep. 2024, 18, 330. [Google Scholar] [CrossRef] [PubMed]
  13. Mo, F.; Pellerino, A.; Rudà, R. Subependymal Giant Cell Astrocytomas (SEGAs): A Model of Targeting Tumor Growth and Epilepsy. Curr. Treat. Options Neurol. 2021, 23, 18. [Google Scholar] [CrossRef]
  14. Ess, K.C.; Kamp, C.A.; Tu, B.P.; Gutmann, D.H. Developmental origin of subependymal giant cell astrocytoma in tuberous sclerosis complex. Neurology 2005, 64, 1446–1449. [Google Scholar] [CrossRef] [PubMed]
  15. Altermatt, H.J.; Scheithauer, B.W. Cytomorphology of subependymal giant cell astrocytoma. Acta Cytol. 1992, 36, 171–175. [Google Scholar]
  16. Mizuguchi, M.; Takashima, S. Neuropathology of tuberous sclerosis. Brain Dev. 2001, 23, 508–515. [Google Scholar] [CrossRef]
  17. Bonnin, J.M.; Rubinstein, L.J.; Papasozomenos, S.C.; Marangos, P.J. Subependymal giant cell astrocytoma. Significance and possible cytogenetic implications of an immunohistochemical study. Acta Neuropathol. 1984, 62, 185–193. [Google Scholar] [CrossRef]
  18. Scheithauer, B.W. The neuropathology of tuberous sclerosis. J. Dermatol. 1992, 19, 897–903. [Google Scholar] [CrossRef]
  19. Phi, J.H.; Park, S.H.; Chae, J.H.; Hong, K.H.; Park, S.S.; Kang, J.H.; Jun, J.K.; Cho, B.K.; Wang, K.C.; Kim, S.K. Congenital subependymal giant cell astrocytoma: Clinical considerations and expression of radial glial cell markers in giant cells. Childs Nerv. Syst. 2008, 24, 1499–1503. [Google Scholar] [CrossRef]
  20. Dutta, R.; Sharma, M.C.; Suri, V.; Sarkar, C.; Garg, A.; Suri, A.; Kale, S.S. TTF-1: A Well-Favored Addition to the Immunohistochemistry Armamentarium as a Diagnostic Marker of SEGA. World Neurosurg. 2022, 159, e62–e69. [Google Scholar] [CrossRef]
  21. Bingle, C.D. Thyroid transcription factor-1. Int. J. Biochem. Cell Biol. 1997, 29, 1471–1473. [Google Scholar] [CrossRef] [PubMed]
  22. Lazzaro, D.; Price, M.; de Felice, M.; Di Lauro, R. The transcription factor TTF-1 is expressed at the onset of thyroid and lung morphogenesis and in restricted regions of the foetal brain. Development 1991, 113, 1093–1104. [Google Scholar] [CrossRef] [PubMed]
  23. Lee, B.J.; Cho, G.; Norgren, R.; Junier, M.-P.; Tapia, V.; Costa, M.; Ojeda, S. TTF-1, a Homeodomain Gene Required for Diencephalic Morphogenesis, Is Postnatally Expressed in the Neuroendocrine Brain in a Developmentally Regulated and Cell-Specific Fashion. Mol. Cell. Neurosci. 2001, 17, 107–126. [Google Scholar] [CrossRef] [PubMed]
  24. Hang, J.F.; Hsu, C.Y.; Lin, S.C.; Wu, C.C.; Lee, H.J.; Ho, D.M. Thyroid transcription factor-1 distinguishes subependymal giant cell astrocytoma from its mimics and supports its cell origin from the progenitor cells in the medial ganglionic eminence. Mod. Pathol. 2017, 30, 318–328. [Google Scholar] [CrossRef]
  25. Schmid, S.; Solomon, D.A.; Perez, E.; Thieme, A.; Kleinschmidt-DeMasters, B.K.; Giannini, C.; Reinhardt, A.; Asa, S.L.; Mete, O.; Stichel, D.; et al. Genetic and epigenetic characterization of posterior pituitary tumors. Acta Neuropathol. 2021, 142, 1025–1043. [Google Scholar] [CrossRef]
  26. Feliciano, D.M. The Neurodevelopmental Pathogenesis of Tuberous Sclerosis Complex (TSC). Front. Neuroanat. 2020, 14, 39. [Google Scholar] [CrossRef]
  27. Bongaarts, A.; Giannikou, K.; Reinten, R.J.; Anink, J.J.; Mills, J.D.; Jansen, F.E.; Spliet, G.M.W.; den Dunnen, W.F.A.; Coras, R.; Blümcke, I.; et al. Subependymal giant cell astrocytomas in Tuberous Sclerosis Complex have consistent TSC1/TSC2 biallelic inactivation, and no BRAF mutations. Oncotarget 2017, 8, 95516–95529. [Google Scholar] [CrossRef]
  28. WHO Classification of Tumours Editorial Board. Central Nervous System Tumours: Who Classification of Tumours, 5th ed.; WHO Classification of Tumours Editorial Board, IARC Publications: Lyon, France, 2022; Volume 6, p. 584. [Google Scholar]
  29. Cuccia, V.; Zuccaro, G.; Sosa, F.; Monges, J.; Lubienieky, F.; Taratuto, A.L. Subependymal giant cell astrocytoma in children with tuberous sclerosis. Childs Nerv. Syst. 2003, 19, 232–243. [Google Scholar] [CrossRef]
  30. Mei, G.-H.; Liu, X.-X.; Zhou, P.; Shen, M. Clinical and imaging features of subependymal giant cell astrocytoma: Report of 20 cases. Chin. Neurosurg. J. 2017, 3, 14. [Google Scholar] [CrossRef]
  31. Siedlecka, M.; Szlufik, S.; Grajkowska, W.; Roszkowski, M.; Jóźwiak, J. Erk activation as a possible mechanism of transformation of subependymal nodule into subependymal giant cell astrocytoma. Folia Neuropathol. 2015, 53, 8–14. [Google Scholar] [CrossRef]
  32. Danassegarane, G.; Tinois, J.; Sahler, Y.; Aouaissia, S.; Riffaud, L. Subependymal giant-cell astrocytoma: A surgical review in the modern era of mTOR inhibitors. Neurochirurgie 2022, 68, 627–636. [Google Scholar] [CrossRef] [PubMed]
  33. Magri, L.; Galli, R. mTOR signaling in neural stem cells: From basic biology to disease. Cell Mol. Life Sci. 2013, 70, 2887–2898. [Google Scholar] [CrossRef] [PubMed]
  34. Fogarasi, A.; De Waele, L.; Bartalini, G.; Jozwiak, S.; Laforgia, N.; Verhelst, H.; Petrak, B.; Pedespan, J.M.; Witt, O.; Castellana, R.; et al. EFFECTS: An expanded access program of everolimus for patients with subependymal giant cell astrocytoma associated with tuberous sclerosis complex. BMC Neurol. 2016, 16, 126. [Google Scholar] [CrossRef]
  35. Northrup, H.; Krueger, D.A. Tuberous sclerosis complex diagnostic criteria update: Recommendations of the 2012 Iinternational Tuberous Sclerosis Complex Consensus Conference. Pediatr. Neurol. 2013, 49, 243–254. [Google Scholar] [CrossRef] [PubMed]
  36. Roth, J.; Roach, E.S.; Bartels, U.; Jóźwiak, S.; Koenig, M.K.; Weiner, H.L.; Franz, D.N.; Wang, H.Z. Subependymal giant cell astrocytoma: Diagnosis, screening, and treatment. Recommendations from the International Tuberous Sclerosis Complex Consensus Conference 2012. Pediatr. Neurol. 2013, 49, 439–444. [Google Scholar] [CrossRef] [PubMed]
  37. Weidman, D.R.; Pole, J.D.; Bouffet, E.; Taylor, M.D.; Bartels, U. Dose-level response rates of mTor inhibition in tuberous sclerosis complex (TSC) related subependymal giant cell astrocytoma (SEGA). Pediatr. Blood Cancer 2015, 62, 1754–1760. [Google Scholar] [CrossRef] [PubMed]
  38. Appalla, D.; Depalma, A.; Calderwood, S. Mammalian Target of Rapamycin Inhibitor Induced Complete Remission of a Recurrent Subependymal Giant Cell Astrocytoma in a Patient without Features of Tuberous Sclerosis Complex. Pediatr. Blood Cancer 2016, 63, 1276–1278. [Google Scholar] [CrossRef]
  39. O’Rawe, M.; Chandran, A.S.; Joshi, S.; Simonin, A.; Dyke, J.M.; Lee, S. A case of subependymal giant cell astrocytoma without tuberous sclerosis complex and review of the literature. Childs Nerv. Syst. 2021, 37, 1381–1385. [Google Scholar] [CrossRef]
  40. Tahiri Elousrouti, L.; Lamchahab, M.; Bougtoub, N.; Elfatemi, H.; Chbani, L.; Harmouch, T.; Maaroufi, M.; Amarti Riffi, A. Subependymal giant cell astrocytoma (SEGA): A case report and review of the literature. J. Med. Case Rep. 2016, 10, 35. [Google Scholar] [CrossRef]
  41. Cobourn, K.D.; Chesney, K.M.; Mueller, K.; Fayed, I.; Tsering, D.; Keating, R.F. Isolated subependymal giant cell astrocytoma (SEGA) in the absence of clinical tuberous sclerosis: Two case reports and literature review. Childs Nerv. Syst. 2024, 40, 73–78. [Google Scholar] [CrossRef]
  42. Halmagyi, G.M.; Bignold, L.P.; Allsop, J.L. Recurrent subependymal giant-cell astrocytoma in the absence of tuberous sclerosis. Case report. J. Neurosurg. 1979, 50, 106–109. [Google Scholar] [CrossRef] [PubMed]
  43. Corlette, L.; Reid, A.; Roberts-Thomson, S.; Christie, M.; Gaillard, F. Solitary subependymal giant cell astrocytoma: Case report and review of the literature. J. Clin. Neurosci. 2020, 82, 26–28. [Google Scholar] [CrossRef] [PubMed]
  44. Suzuki, M.; Kondo, A.; Ogino, I.; Akiyama, O.; Fujita, N.; Shimizu, Y.; Arai, H. A case of solitary subependymal giant cell astrocytoma with histopathological anaplasia and TSC2 gene alteration. Childs Nerv. Syst. 2021, 37, 1357–1362. [Google Scholar] [CrossRef] [PubMed]
  45. De Waele, L.; Lagae, L.; Mekahli, D. Tuberous sclerosis complex: The past and the future. Pediatr. Nephrol. 2015, 30, 1771–1780. [Google Scholar] [CrossRef] [PubMed]
  46. Dabora, S.L.; Jozwiak, S.; Franz, D.N.; Roberts, P.S.; Nieto, A.; Chung, J.; Choy, Y.S.; Reeve, M.P.; Thiele, E.; Egelhoff, J.C.; et al. Mutational analysis in a cohort of 224 tuberous sclerosis patients indicates increased severity of TSC2, compared with TSC1, disease in multiple organs. Am. J. Hum. Genet. 2001, 68, 64–80. [Google Scholar] [CrossRef]
  47. Man, A.; Di Scipio, M.; Grewal, S.; Suk, Y.; Trinari, E.; Ejaz, R.; Whitney, R. The Genetics of Tuberous Sclerosis Complex and Related mTORopathies: Current Understanding and Future Directions. Genes 2024, 15, 332. [Google Scholar] [CrossRef]
  48. Avgeris, S.; Fostira, F.; Vagena, A.; Ninios, Y.; Delimitsou, A.; Vodicka, R.; Vrtel, R.; Youroukos, S.; Stravopodis, D.J.; Vlassi, M.; et al. Mutational analysis of TSC1 and TSC2 genes in Tuberous Sclerosis Complex patients from Greece. Sci. Rep. 2017, 7, 16697. [Google Scholar] [CrossRef]
  49. Caban, C.; Khan, N.; Hasbani, D.M.; Crino, P.B. Genetics of tuberous sclerosis complex: Implications for clinical practice. Appl. Clin. Genet. 2017, 10, 1–8. [Google Scholar] [CrossRef]
  50. Rosset, C.; Netto, C.B.O.; Ashton-Prolla, P. TSC1 and TSC2 gene mutations and their implications for treatment in Tuberous Sclerosis Complex: A review. Genet. Mol. Biol. 2017, 40, 69–79. [Google Scholar] [CrossRef]
  51. Sancak, O.; Nellist, M.; Goedbloed, M.; Elfferich, P.; Wouters, C.; Maat-Kievit, A.; Zonnenberg, B.; Verhoef, S.; Halley, D.; van den Ouweland, A. Mutational analysis of the TSC1 and TSC2 genes in a diagnostic setting: Genotype—Phenotype correlations and comparison of diagnostic DNA techniques in Tuberous Sclerosis Complex. Eur. J. Hum. Genet. 2005, 13, 731–741. [Google Scholar] [CrossRef]
  52. Barron, R.P.; Kainulainen, V.T.; Forrest, C.R.; Krafchik, B.; Mock, D.; Sàndor, G.K. Tuberous sclerosis: Clinicopathologic features and review of the literature. J. Craniomaxillofac. Surg. 2002, 30, 361–366. [Google Scholar] [CrossRef] [PubMed]
  53. Crino, P.B.; Nathanson, K.L.; Henske, E.P. The tuberous sclerosis complex. N. Engl. J. Med. 2006, 355, 1345–1356. [Google Scholar] [CrossRef] [PubMed]
  54. Northrup, H.; Aronow, M.E.; Bebin, E.M.; Bissler, J.; Darling, T.N.; de Vries, P.J.; Frost, M.D.; Fuchs, Z.; Gosnell, E.S.; Gupta, N.; et al. Updated International Tuberous Sclerosis Complex Diagnostic Criteria and Surveillance and Management Recommendations. Pediatr. Neurol. 2021, 123, 50–66. [Google Scholar] [CrossRef] [PubMed]
  55. Patniyot, I.; Qubty, W. Managing Headache Disorders Associated with Tuberous Sclerosis and Neurofibromatosis. Curr. Pain Headache Rep. 2022, 26, 281–288. [Google Scholar] [CrossRef] [PubMed]
  56. Crainic, N.; Furtner, J.; Pallud, J.; Bielle, F.; Lombardi, G.; Rudà, R.; Idbaih, A. Rare Neuronal, Glial and Glioneuronal Tumours in Adults. Cancers 2023, 15, 1120. [Google Scholar] [CrossRef]
  57. Gelot, A.B.; Represa, A. Progression of Fetal Brain Lesions in Tuberous Sclerosis Complex. Front. Neurosci. 2020, 14, 899. [Google Scholar] [CrossRef]
  58. Moavero, R.; Coniglio, A.; Garaci, F.; Curatolo, P. Is mTOR inhibition a systemic treatment for tuberous sclerosis? Ital. J. Pediatr. 2013, 39, 57. [Google Scholar] [CrossRef]
  59. Leung, A.K.; Robson, W.L. Tuberous sclerosis complex: A review. J. Pediatr. Health Care 2007, 21, 108–114. [Google Scholar] [CrossRef]
  60. Napolioni, V.; Curatolo, P. Genetics and molecular biology of tuberous sclerosis complex. Curr. Genom. 2008, 9, 475–487. [Google Scholar] [CrossRef]
  61. Beaumont, T.L.; Godzik, J.; Dahiya, S.; Smyth, M.D. Subependymal giant cell astrocytoma in the absence of tuberous sclerosis complex: Case report. J. Neurosurg. Pediatr. 2015, 16, 134–137. [Google Scholar] [CrossRef]
  62. Yalon, M.; Ben-Sira, L.; Constantini, S.; Toren, A. Regression of subependymal giant cell astrocytomas with RAD001 (Everolimus) in tuberous sclerosis complex. Child’s Nerv. Syst. 2011, 27, 179–181. [Google Scholar] [CrossRef] [PubMed]
  63. Nakamura, J.L.; Garcia, E.; Pieper, R.O. S6K1 plays a key role in glial transformation. Cancer Res. 2008, 68, 6516–6523. [Google Scholar] [CrossRef] [PubMed]
  64. Panwar, V.; Singh, A.; Bhatt, M.; Tonk, R.K.; Azizov, S.; Raza, A.S.; Sengupta, S.; Kumar, D.; Garg, M. Multifaceted role of mTOR (mammalian target of rapamycin) signaling pathway in human health and disease. Signal Transduct. Target. Ther. 2023, 8, 375. [Google Scholar] [CrossRef]
  65. Cui, D.; Qu, R.; Liu, D.; Xiong, X.; Liang, T.; Zhao, Y. The Cross Talk between p53 and mTOR Pathways in Response to Physiological and Genotoxic Stresses. Front. Cell Dev. Biol. 2021, 9, 775507. [Google Scholar] [CrossRef] [PubMed]
  66. Carriere, A.; Romeo, Y.; Jaquez, A.; Moreau, J.; Bonneil, E.; Thibault, P.; Fingar, D.; Roux, P. ERK1/2 phosphorylate Raptor to promote Ras-dependent activation of mTOR complex 1 (mTORC1). J. Biol. Chem. 2010, 286, 567–577. [Google Scholar] [CrossRef] [PubMed]
  67. Jones, A.C.; Daniells, C.E.; Snell, R.G.; Tachataki, M.; Idziaszczyk, S.A.; Krawczak, M.; Sampson, J.R.; Cheadle, J.P. Molecular genetic and phenotypic analysis reveals differences between TSC1 and TSC2 associated familial and sporadic tuberous sclerosis. Hum. Mol. Genet. 1997, 6, 2155–2161. [Google Scholar] [CrossRef] [PubMed]
  68. Piña-Ballantyne, S.A.; Espinosa-Aguilar, E.J.; Calderón-Garcidueñas, A.L. The Clinicopathological Features of the Solitary Subependymal Giant Cell Astrocytoma: A Systematic Review. Neurol. India 2024, 72, 708–717. [Google Scholar] [CrossRef]
  69. Ogórek, B.; Hamieh, L.; Hulshof, H.M.; Lasseter, K.; Klonowska, K.; Kuijf, H.; Moavero, R.; Hertzberg, C.; Weschke, B.; Riney, K.; et al. TSC2 pathogenic variants are predictive of severe clinical manifestations in TSC infants: Results of the EPISTOP study. Genet. Med. 2020, 22, 1489–1497. [Google Scholar] [CrossRef]
  70. Kotulska, K.; Borkowska, J.; Mandera, M.; Roszkowski, M.; Jurkiewicz, E.; Grajkowska, W.; Bilska, M.; Jóźwiak, S. Congenital subependymal giant cell astrocytomas in patients with tuberous sclerosis complex. Child’s Nerv. Syst. 2014, 30, 2037–2042. [Google Scholar] [CrossRef]
  71. Giannikou, K.; Zhu, Z.; Kim, J.; Winden, K.D.; Tyburczy, M.E.; Marron, D.; Parker, J.S.; Hebert, Z.; Bongaarts, A.; Taing, L.; et al. Subependymal giant cell astrocytomas are characterized by mTORC1 hyperactivation, a very low somatic mutation rate, and a unique gene expression profile. Mod. Pathol. 2021, 34, 264–279. [Google Scholar] [CrossRef]
  72. Palsgrove, D.N.; Brosnan-Cashman, J.A.; Giannini, C.; Raghunathan, A.; Jentoft, M.; Bettegowda, C.; Gokden, M.; Lin, D.; Yuan, M.; Lin, M.T.; et al. Subependymal giant cell astrocytoma-like astrocytoma: A neoplasm with a distinct phenotype and frequent neurofibromatosis type-1-association. Mod. Pathol. 2018, 31, 1787–1800. [Google Scholar] [CrossRef] [PubMed]
  73. Fohlen, M.; Harzallah, I.; Polivka, M.; Giuliano, F.; Pons, L.; Streichenberger, N.; Dorfmüller, G.; Touraine, R. Identification of TSC1 or TSC2 mutation limited to the tumor in three cases of solitary subependymal giant cell astrocytoma using next-generation sequencing technology. Childs Nerv. Syst. 2020, 36, 961–965. [Google Scholar] [CrossRef] [PubMed]
  74. Sasaki, T.; Uda, T.; Kuki, I.; Kunihiro, N.; Okazaki, S.; Niida, Y.; Goto, T. TSC2 somatic mosaic mutation, including extra-tumor tissue, may be the developmental cause of solitary subependymal giant cell astrocytoma. Childs Nerv. Syst. 2022, 38, 77–83. [Google Scholar] [CrossRef] [PubMed]
  75. Kumari, K.; Sharma, M.C.; Kakkar, A.; Malgulwar, P.B.; Pathak, P.; Suri, V.; Sarkar, C.; Chandra, S.P.; Faruq, M.; Gupta, R.K.; et al. Role of mTOR signaling pathway in the pathogenesis of subependymal giant cell astrocytoma—A study of 28 cases. Neurol. India 2016, 64, 988–994. [Google Scholar] [PubMed]
  76. Popova, N.V.; Jücker, M. The Role of mTOR Signaling as a Therapeutic Target in Cancer. Int. J. Mol. Sci. 2021, 22, 1743. [Google Scholar] [CrossRef] [PubMed]
  77. Yindeedej, V.; Rojnueangnit, K.; Chotsakulthong, P.; Thamwongskul, C. Bleeding solitary SEGA in non-tuberous sclerosis complex adolescent: A case illustration and review of literature. Childs Nerv. Syst. 2024, 40, 2199–2207. [Google Scholar] [CrossRef]
  78. Zabielska, B.; Rzewuska, N.; Jóźwiak, S. Subependymal Giant Cell Astrocytoma Tumors in Patients without Clinical Manifestation of Tuberous Sclerosis Complex: A Diagnostic Puzzle. Pediatr. Neurol. 2024, 150, 40–42. [Google Scholar] [CrossRef]
  79. Hall, A.; Westlake, G.; Short, B.P.; Pearson, M.; Ess, K.C. TSC1 Mosaicism Leading to Subependymal Giant Cell Astrocytoma but Not Tuberous Sclerosis Complex. Pediatr. Neurol. 2021, 123, 38–39. [Google Scholar] [CrossRef]
  80. Klonowska, K.; Giannikou, K.; Grevelink, J.M.; Boeszoermenyi, B.; Thorner, A.R.; Herbert, Z.T.; Afrin, A.; Treichel, A.M.; Hamieh, L.; Kotulska, K.; et al. Comprehensive genetic and phenotype analysis of 95 individuals with mosaic tuberous sclerosis complex. Am. J. Hum. Genet. 2023, 110, 979–988. [Google Scholar] [CrossRef]
  81. Ebrahimi-Fakhari, D.; Franz, D.N. Pharmacological treatment strategies for subependymal giant cell astrocytoma (SEGA). Expert Opin. Pharmacother. 2020, 21, 1329–1336. [Google Scholar] [CrossRef]
  82. Tyburczy, M.E.; Kotulska, K.; Pokarowski, P.; Mieczkowski, J.; Kucharska, J.; Grajkowska, W.; Roszkowski, M.; Jozwiak, S.; Kaminska, B. Novel proteins regulated by mTOR in subependymal giant cell astrocytomas of patients with tuberous sclerosis complex and new therapeutic implications. Am. J. Pathol. 2010, 176, 1878–1890. [Google Scholar] [CrossRef] [PubMed]
  83. Jóźwiak, S.; Nabbout, R.; Curatolo, P. Management of subependymal giant cell astrocytoma (SEGA) associated with tuberous sclerosis complex (TSC): Clinical recommendations. Eur. J. Paediatr. Neurol. 2013, 17, 348–352. [Google Scholar] [CrossRef] [PubMed]
  84. Parker, W.E.; Orlova, K.A.; Heuer, G.G.; Baybis, M.; Aronica, E.; Frost, M.; Wong, M.; Crino, P.B. Enhanced epidermal growth factor, hepatocyte growth factor, and vascular endothelial growth factor expression in tuberous sclerosis complex. Am. J. Pathol. 2011, 178, 296–305. [Google Scholar] [CrossRef] [PubMed]
  85. Amin, S.; Carter, M.; Edwards, R.J.; Pople, I.; Aquilina, K.; Merrifield, J.; Osborne, J.P.; O’Callaghan, F.J. The outcome of surgical management of subependymal giant cell astrocytoma in tuberous sclerosis complex. Eur. J. Paediatr. Neurol. 2013, 17, 36–44. [Google Scholar] [CrossRef]
  86. Dias, S.F.; Richards, O.; Elliot, M.; Chumas, P. Pediatric-Like Brain Tumors in Adults. Adv. Tech. Stand. Neurosurg. 2024, 50, 147–183. [Google Scholar] [CrossRef]
  87. Karita, H.; Tsuda, K.; Kono, M.; Yamamoto, T.; Ihara, S. Neoadjuvant Therapy with Everolimus for Subependymal Giant Cell Astrocytoma: A Case Report. NMC Case Rep. J. 2023, 10, 291–297. [Google Scholar] [CrossRef]
  88. Krueger, D.A.; Care, M.M.; Holland, K.; Agricola, K.; Tudor, C.; Mangeshkar, P.; Wilson, K.A.; Byars, A.; Sahmoud, T.; Franz, D.N. Everolimus for subependymal giant-cell astrocytomas in tuberous sclerosis. N. Engl. J. Med. 2010, 363, 1801–1811. [Google Scholar] [CrossRef]
  89. Capal, J.; Ritter, D.; Franz, D.; Griffith, M.; Currans, K.; Kent, B.; Bebin, M.; Northrup, H.; Koenig, M.; Mizuno, T.; et al. Preventative treatment of tuberous sclerosis complex with sirolimus: Phase I safety and efficacy results. Ann. Child Neurol. Soc. 2024, 2, 106–119. [Google Scholar] [CrossRef]
  90. Sun, P.; Kohrman, M.; Liu, J.; Guo, A.; Rogerio, J.; Krueger, D. Outcomes of resecting subependymal giant cell astrocytoma (SEGA) among patients with SEGA-related tuberous sclerosis complex: A national claims database analysis. Curr. Med. Res. Opin. 2012, 28, 657–663. [Google Scholar] [CrossRef]
  91. Sun, P.; Krueger, D.; Liu, J.; Guo, A.; Rogerio, J.; Kohrman, M. Surgical resection of subependymal giant cell astrocytomas (SEGAs) and changes in SEGA-related conditions: A US national claims database study. Curr. Med. Res. Opin. 2012, 28, 651–656. [Google Scholar] [CrossRef]
  92. Kotulska, K.; Borkowska, J.; Roszkowski, M.; Mandera, M.; Daszkiewicz, P.; Drabik, K.; Jurkiewicz, E.; Larysz-Brysz, M.; Nowak, K.; Grajkowska, W.; et al. Surgical treatment of subependymal giant cell astrocytoma in tuberous sclerosis complex patients. Pediatr. Neurol. 2014, 50, 307–312. [Google Scholar] [CrossRef] [PubMed]
  93. Yamamoto, K.; Yamada, K.; Nakahara, T.; Ishihara, A.; Takaki, S.; Kochi, M.; Ushio, Y. Rapid regrowth of solitary subependymal giant cell astrocytoma—Case report. Neurol. Med.-Chir. 2002, 42, 224–227. [Google Scholar] [CrossRef] [PubMed]
  94. Campen, C.J.; Porter, B.E. Subependymal Giant Cell Astrocytoma (SEGA) Treatment Update. Curr. Treat. Options Neurol. 2011, 13, 380–385. [Google Scholar] [CrossRef] [PubMed]
  95. Sasongko, T.H.; Ismail, N.F.; Nik Abdul Malik, N.M.; Zabidi-Hussin, Z.A. Rapamycin and its analogues (rapalogs) for Tuberous Sclerosis Complex-associated tumors: A systematic review on non-randomized studies using meta-analysis. Orphanet J. Rare Dis. 2015, 10, 95. [Google Scholar] [CrossRef] [PubMed]
  96. Śmiałek, D.; Kotulska, K.; Duda, A.; Jóźwiak, S. Effect of mTOR Inhibitors in Epilepsy Treatment in Children with Tuberous Sclerosis Complex Under 2 Years of Age. Neurol. Ther. 2023, 12, 931–946. [Google Scholar] [CrossRef]
  97. Tomoto, K.; Fujimoto, A.; Inenaga, C.; Okanishi, T.; Imai, S.; Ogai, M.; Fukunaga, A.; Nakamura, H.; Sato, K.; Obana, A.; et al. Experience using mTOR inhibitors for subependymal giant cell astrocytoma in tuberous sclerosis complex at a single facility. BMC Neurol. 2021, 21, 139. [Google Scholar] [CrossRef]
  98. Yang, G.; Yang, L.; Yang, X.; Shi, X.; Wang, J.; Liu, Y.; Ju, J.; Zou, L. Efficacy and safety of a mammalian target of rapamycin inhibitor in pediatric patients with tuberous sclerosis complex: A systematic review and meta-analysis. Exp. Ther. Med. 2015, 9, 626–630. [Google Scholar] [CrossRef]
  99. Eng, C.P.; Sehgal, S.N.; Vézina, C. Activity of rapamycin (AY-22,989) against transplanted tumors. J. Antibiot. 1984, 37, 1231–1237. [Google Scholar] [CrossRef]
  100. Georgakis, G.V.; Younes, A. From Rapa Nui to rapamycin: Targeting PI3K/Akt/mTOR for cancer therapy. Expert Rev. Anticancer. Ther. 2006, 6, 131–140. [Google Scholar] [CrossRef]
  101. Ballou, L.M.; Lin, R.Z. Rapamycin and mTOR kinase inhibitors. J. Chem. Biol. 2008, 1, 27–36. [Google Scholar] [CrossRef]
  102. Wagner, A.J.; Ravi, V.; Riedel, R.F.; Ganjoo, K.; Van Tine, B.A.; Chugh, R.; Cranmer, L.; Gordon, E.M.; Hornick, J.L.; Du, H.; et al. nab-Sirolimus for Patients with Malignant Perivascular Epithelioid Cell Tumors. J. Clin. Oncol. 2021, 39, 3660–3670. [Google Scholar] [CrossRef] [PubMed]
  103. Gordon, E.M.; Angel, N.L.; Omelchenko, N.; Chua-Alcala, V.S.; Moradkhani, A.; Quon, D.; Wong, S. A Phase I/II Investigation of Safety and Efficacy of Nivolumab and nab-Sirolimus in Patients with a Variety of Tumors with Genetic Mutations in the mTOR Pathway. Anticancer. Res. 2023, 43, 1993–2002. [Google Scholar] [CrossRef] [PubMed]
  104. Schuler, W.; Sedrani, R.; Cottens, S.; Häberlin, B.; Schulz, M.; Schuurman, H.J.; Zenke, G.; Zerwes, H.G.; Schreier, M.H. SDZ RAD, a new rapamycin derivative: Pharmacological properties in vitro and in vivo. Transplantation 1997, 64, 36–42. [Google Scholar] [CrossRef] [PubMed]
  105. Yao, J.C.; Fazio, N.; Singh, S.; Buzzoni, R.; Carnaghi, C.; Wolin, E.; Tomasek, J.; Raderer, M.; Lahner, H.; Voi, M.; et al. Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): A randomised, placebo-controlled, phase 3 study. Lancet 2016, 387, 968–977. [Google Scholar] [CrossRef] [PubMed]
  106. Sadowski, K.; Kotulska, K.; Jóźwiak, S. Management of side effects of mTOR inhibitors in tuberous sclerosis patients. Pharmacol. Rep. 2016, 68, 536–542. [Google Scholar] [CrossRef] [PubMed]
  107. Banaszynski, L.A.; Liu, C.W.; Wandless, T.J. Characterization of the FKBP.rapamycin.FRB ternary complex. J. Am. Chem. Soc. 2005, 127, 4715–4721. [Google Scholar] [CrossRef]
  108. Edwards, S.R.; Wandless, T.J. The Rapamycin-binding Domain of the Protein Kinase Mammalian Target of Rapamycin Is a Destabilizing Domain. J. Biol. Chem. 2007, 282, 13395–13401. [Google Scholar] [CrossRef]
  109. Ali, E.S.; Mitra, K.; Akter, S.; Ramproshad, S.; Mondal, B.; Khan, I.N.; Islam, M.T.; Sharifi-Rad, J.; Calina, D.; Cho, W.C. Recent advances and limitations of mTOR inhibitors in the treatment of cancer. Cancer Cell Int. 2022, 22, 284. [Google Scholar] [CrossRef]
  110. Zhao, H.-f.; Wang, J.; Shao, W.; Wu, C.-p.; Chen, Z.-p.; To, S.-s.T.; Li, W.-p. Recent advances in the use of PI3K inhibitors for glioblastoma multiforme: Current preclinical and clinical development. Mol. Cancer 2017, 16, 100. [Google Scholar] [CrossRef]
  111. Hillmann, P.; Fabbro, D. PI3K/mTOR Pathway Inhibition: Opportunities in Oncology and Rare Genetic Diseases. Int. J. Mol. Sci. 2019, 20, 5792. [Google Scholar] [CrossRef]
  112. Curran, M.P. Everolimus: In patients with subependymal giant cell astrocytoma associated with tuberous sclerosis complex. Paediatr. Drugs 2012, 14, 51–60. [Google Scholar] [CrossRef] [PubMed]
  113. Franz, D.N.; Agricola, K.; Mays, M.; Tudor, C.; Care, M.M.; Holland-Bouley, K.; Berkowitz, N.; Miao, S.; Peyrard, S.; Krueger, D.A. Everolimus for subependymal giant cell astrocytoma: 5-year final analysis. Ann. Neurol. 2015, 78, 929–938. [Google Scholar] [CrossRef] [PubMed]
  114. Franz, D.N.; Belousova, E.; Sparagana, S.; Bebin, E.M.; Frost, M.; Kuperman, R.; Witt, O.; Kohrman, M.H.; Flamini, J.R.; Wu, J.Y.; et al. Efficacy and safety of everolimus for subependymal giant cell astrocytomas associated with tuberous sclerosis complex (EXIST-1): A multicentre, randomised, placebo-controlled phase 3 trial. Lancet 2013, 381, 125–132. [Google Scholar] [CrossRef] [PubMed]
  115. Fukumura, S.; Watanabe, T.; Takayama, R.; Minagawa, K.; Tsutsumi, H. Everolimus Treatment for an Early Infantile Subependymal Giant Cell Astrocytoma with Tuberous Sclerosis Complex. J. Child Neurol. 2015, 30, 1192–1195. [Google Scholar] [CrossRef] [PubMed]
  116. Franz, D.N.; Belousova, E.; Sparagana, S.; Bebin, E.M.; Frost, M.; Kuperman, R.; Witt, O.; Kohrman, M.H.; Flamini, J.R.; Wu, J.Y.; et al. Everolimus for subependymal giant cell astrocytoma in patients with tuberous sclerosis complex: 2-year open-label extension of the randomised EXIST-1 study. Lancet Oncol. 2014, 15, 1513–1520. [Google Scholar] [CrossRef]
  117. Krueger, D.A.; Care, M.M.; Agricola, K.; Tudor, C.; Mays, M.; Franz, D.N. Everolimus long-term safety and efficacy in subependymal giant cell astrocytoma. Neurology 2013, 80, 574–580. [Google Scholar] [CrossRef]
  118. Tran, L.H.; Zupanc, M.L. Long-Term Everolimus Treatment in Individuals with Tuberous Sclerosis Complex: A Review of the Current Literature. Pediatr. Neurol. 2015, 53, 23–30. [Google Scholar] [CrossRef]
  119. French, J.A.; Lawson, J.A.; Yapici, Z.; Ikeda, H.; Polster, T.; Nabbout, R.; Curatolo, P.; de Vries, P.J.; Dlugos, D.J.; Berkowitz, N.; et al. Adjunctive everolimus therapy for treatment-resistant focal-onset seizures associated with tuberous sclerosis (EXIST-3): A phase 3, randomised, double-blind, placebo-controlled study. Lancet 2016, 388, 2153–2163. [Google Scholar] [CrossRef]
  120. Trelinska, J.; Dachowska, I.; Kotulska, K.; Baranska, D.; Fendler, W.; Jozwiak, S.; Mlynarski, W. Factors affecting response to everolimus therapy for subependymal giant cell astrocytomas associated with tuberous sclerosis. Pediatr. Blood Cancer 2015, 62, 616–621. [Google Scholar] [CrossRef]
  121. Vergès, B.; Cariou, B. mTOR inhibitors and diabetes. Diabetes Res. Clin. Pract. 2015, 110, 101–108. [Google Scholar] [CrossRef]
  122. Jackson, E.R.; Duchatel, R.J.; Staudt, D.E.; Persson, M.L.; Mannan, A.; Yadavilli, S.; Parackal, S.; Game, S.; Chong, W.C.; Jayasekara, W.S.N.; et al. ONC201 in combination with paxalisib for the treatment of H3K27-altered diffuse midline glioma. Cancer Res. 2023, 83, 2421–2437. [Google Scholar] [CrossRef] [PubMed]
  123. Rogers, H.A.; Estranero, J.; Gudka, K.; Grundy, R.G. The therapeutic potential of targeting the PI3K pathway in pediatric brain tumors. Oncotarget 2017, 8, 2083–2095. [Google Scholar] [CrossRef] [PubMed]
  124. Szklener, K.; Mazurek, M.; Wieteska, M.; Wacławska, M.; Bilski, M.; Mańdziuk, S. New Directions in the Therapy of Glioblastoma. Cancers 2022, 14, 5377. [Google Scholar] [CrossRef] [PubMed]
  125. Bongaarts, A.; van Scheppingen, J.; Korotkov, A.; Mijnsbergen, C.; Anink, J.J.; Jansen, F.E.; Spliet, W.G.M.; den Dunnen, W.F.A.; Gruber, V.E.; Scholl, T.; et al. The coding and non-coding transcriptional landscape of subependymal giant cell astrocytomas. Brain 2020, 143, 131–149. [Google Scholar] [CrossRef] [PubMed]
  126. Tang, X.; Angst, G.; Haas, M.; Yang, F.; Wang, C. The Characterization of a Subependymal Giant Astrocytoma-Like Cell Line from Murine Astrocyte with mTORC1 Hyperactivation. Int. J. Mol. Sci. 2021, 22, 4116. [Google Scholar] [CrossRef] [PubMed]
  127. Pucko, E.; Ostrowski, R.; Matyja, E. Novel small molecule protein kinase CK2 inhibitors exert potent antitumor effects on T98G and SEGA cells in vitro. Folia Neuropathol. 2019, 57, 239–248. [Google Scholar] [CrossRef]
  128. Aum, D.J.; Reynolds, R.A.; McEvoy, S.D.; Wong, M.; Roland, J.L.; Smyth, M.D. Laser interstitial thermal therapy compared with open resection for treating subependymal giant cell astrocytoma. J. Neurosurg. Pediatr. 2024, 33, 95–104. [Google Scholar] [CrossRef]
  129. Dadey, D.Y.; Kamath, A.A.; Leuthardt, E.C.; Smyth, M.D. Laser interstitial thermal therapy for subependymal giant cell astrocytoma: Technical case report. Neurosurg. Focus 2016, 41, E9. [Google Scholar] [CrossRef]
  130. Amin, S.; Mallick, A.A.; Edwards, H.; Cortina-Borja, M.; Laugharne, M.; Likeman, M.; O’Callaghan, F.J.K. The metformin in tuberous sclerosis (MiTS) study: A randomised double-blind placebo-controlled trial. eClinicalMedicine 2021, 32, 100715. [Google Scholar] [CrossRef]
  131. Bongaarts, A.; de Jong, J.M.; Broekaart, D.W.M.; van Scheppingen, J.; Anink, J.J.; Mijnsbergen, C.; Jansen, F.E.; Spliet, W.G.M.; den Dunnen, W.F.A.; Gruber, V.E.; et al. Dysregulation of the MMP/TIMP Proteolytic System in Subependymal Giant Cell Astrocytomas in Patients with Tuberous Sclerosis Complex: Modulation of MMP by MicroRNA-320d In Vitro. J. Neuropathol. Exp. Neurol. 2020, 79, 777–790. [Google Scholar] [CrossRef]
  132. Park, K.J.; Kano, H.; Kondziolka, D.; Niranjan, A.; Flickinger, J.C.; Lunsford, L.D. Gamma Knife surgery for subependymal giant cell astrocytomas. Clinical article. J. Neurosurg. 2011, 114, 808–813. [Google Scholar] [CrossRef] [PubMed]
  133. De Roxas, R.C.; Suratos, C.T.R.; Fernandez, M.L.L. Temozolomide as Treatment in Lowgrade Glioma: A Systematic Review. J. Neuro-Oncol. Neurosci. 2016, 1, 7. [Google Scholar] [CrossRef]
  134. Grajkowska, W.; Kotulska, K.; Jurkiewicz, E.; Roszkowski, M.; Daszkiewicz, P.; Jóźwiak, S.; Matyja, E. Subependymal giant cell astrocytomas with atypical histological features mimicking malignant gliomas. Folia Neuropathol. 2011, 49, 39–46. [Google Scholar] [PubMed]
  135. Hafazalla, K.; Sahgal, A.; Jaja, B.; Perry, J.R.; Das, S. Procarbazine, CCNU and vincristine (PCV) versus temozolomide chemotherapy for patients with low-grade glioma: A systematic review. Oncotarget 2018, 9, 33623–33633. [Google Scholar] [CrossRef] [PubMed]
  136. Duchatel, R.J.; Jackson, E.R.; Parackal, S.G.; Kiltschewskij, D.; Findlay, I.J.; Mannan, A.; Staudt, D.E.; Thomas, B.C.; Germon, Z.P.; Laternser, S.; et al. PI3K/mTOR is a therapeutically targetable genetic dependency in diffuse intrinsic pontine glioma. J. Clin. Investig. 2024, 134, e170329. [Google Scholar] [CrossRef]
  137. Omeljaniuk, W.J.; Krętowski, R.; Ratajczak-Wrona, W.; Jabłońska, E.; Cechowska-Pasko, M. Novel Dual PI3K/mTOR Inhibitor, Apitolisib (GDC-0980), Inhibits Growth and Induces Apoptosis in Human Glioblastoma Cells. Int. J. Mol. Sci. 2021, 22, 11511. [Google Scholar] [CrossRef]
  138. Tutak, I.; Ozdil, B.; Uysal, A. Voxtalisib and low intensity pulsed ultrasound combinatorial effect on glioblastoma multiforme cancer stem cells via PI3K/AKT/mTOR. Pathol. Res. Pract. 2022, 239, 154145. [Google Scholar] [CrossRef]
  139. Wen, P.Y.; Omuro, A.; Ahluwalia, M.S.; Fathallah-Shaykh, H.M.; Mohile, N.; Lager, J.J.; Laird, A.D.; Tang, J.; Jiang, J.; Egile, C.; et al. Phase I dose-escalation study of the PI3K/mTOR inhibitor voxtalisib (SAR245409, XL765) plus temozolomide with or without radiotherapy in patients with high-grade glioma. Neuro Oncol. 2015, 17, 1275–1283. [Google Scholar] [CrossRef]
  140. Srinivasan, A.; Naga Shashi Kiran, B.; Koduvath, A.; Pal, A.; Suresh, A.; Kannan, A.; Ravichandran, M. Metformin for the management of tuberous sclerosis: What does the evidence tell us? EXCLI J. 2021, 20, 1474–1475. [Google Scholar] [CrossRef]
  141. Łabuzek, K.; Suchy, D.; Gabryel, B.; Bielecka, A.; Liber, S.; Okopień, B. Quantification of metformin by the HPLC method in brain regions, cerebrospinal fluid and plasma of rats treated with lipopolysaccharide. Pharmacol. Rep. 2010, 62, 956–965. [Google Scholar] [CrossRef]
  142. Seliger, C.; Genbrugge, E.; Gorlia, T.; Chinot, O.; Stupp, R.; Nabors, B.; Weller, M.; Hau, P. Use of metformin and outcome of patients with newly diagnosed glioblastoma: Pooled analysis. Int. J. Cancer 2020, 146, 803–809. [Google Scholar] [CrossRef] [PubMed]
  143. Nadeem, M.D.; Memon, S.; Qureshi, K.; Farooq, U.; Memon, U.A.; Aparna, F.; Kachhadia, M.P.; Shahzeen, F.; Ali, S.; Varrassi, G.; et al. Seizing the Connection: Exploring the Interplay between Epilepsy and Glycemic Control in Diabetes Management. Cureus 2023, 15, e45606. [Google Scholar] [CrossRef] [PubMed]
  144. Tornio, A.; Niemi, M.; Neuvonen, P.J.; Backman, J.T. Drug interactions with oral antidiabetic agents: Pharmacokinetic mechanisms and clinical implications. Trends Pharmacol. Sci. 2012, 33, 312–322. [Google Scholar] [CrossRef] [PubMed]
  145. Pucko, E.; Matyja, E.; Koronkiewicz, M.; Ostrowski, R.P.; Kazimierczuk, Z. Potent Antitumour Effects of Novel Pentabromobenzylisothioureas Studied on Human Glial-derived Tumour Cell Lines. Anticancer Res. 2018, 38, 2691–2705. [Google Scholar] [CrossRef] [PubMed]
  146. Frassanito, P.; Noya, C.; Tamburrini, G. Current trends in the management of subependymal giant cell astrocytomas in tuberous sclerosis. Child’s Nerv. Syst. 2020, 36, 2527–2536. [Google Scholar] [CrossRef]
  147. Reese, J.C.; Fadel, H.A.; Pawloski, J.A.; Samir, M.; Haider, S.; Komatar, R.J.; Luther, E.; Morell, A.A.; Ivan, M.E.; Robin, A.M.; et al. Laser interstitial thermal therapy for deep-seated perivascular brain tumors is not associated with distal ischemia. J. Neurooncol. 2024, 166, 265–272. [Google Scholar] [CrossRef]
  148. Boop, S.; Bonda, D.; Randle, S.; Leary, S.; Vitanza, N.; Crotty, E.; Novotny, E.; Friedman, S.; Ellenbogen, R.G.; Durfy, S.; et al. A Comparison of Clinical Outcomes for Subependymal Giant Cell Astrocytomas Treated with Laser Interstitial Thermal Therapy, Open Surgical Resection, and mTOR Inhibitors. Pediatr. Neurosurg. 2023, 58, 150–159. [Google Scholar] [CrossRef]
  149. Abba, M.; Patil, N.; Allgayer, H. MicroRNAs in the Regulation of MMPs and Metastasis. Cancers 2014, 6, 625–645. [Google Scholar] [CrossRef]
  150. Beylerli, O.; Gareev, I.; Sufianov, A.; Ilyasova, T.; Zhang, F. The role of microRNA in the pathogenesis of glial brain tumors. Non-Coding RNA Res. 2022, 7, 71–76. [Google Scholar] [CrossRef]
  151. Dibdiakova, K.; Majercikova, Z.; Galanda, T.; Richterova, R.; Kolarovszki, B.; Racay, P.; Hatok, J. Relationship between the Expression of Matrix Metalloproteinases and Their Tissue Inhibitors in Patients with Brain Tumors. Int. J. Mol. Sci. 2024, 25, 2858. [Google Scholar] [CrossRef]
  152. Zhou, W.; Yu, X.; Sun, S.; Zhang, X.; Yang, W.; Zhang, J.; Zhang, X.; Jiang, Z. Increased expression of MMP-2 and MMP-9 indicates poor prognosis in glioma recurrence. Biomed. Pharmacother. 2019, 118, 109369. [Google Scholar] [CrossRef] [PubMed]
  153. Adylova, A.; Kapanova, G.; Datkhayeva, Z.; Raganina, K.; Tanbayeva, G.; Baigonova, K. Nanotechnology-based cancer chemoprevention in glioblastoma. Folia Neuropathol. 2023, 61, 235–241. [Google Scholar] [CrossRef] [PubMed]
  154. Singh, R.R.; Mondal, I.; Janjua, T.; Popat, A.; Kulshreshtha, R. Engineered smart materials for RNA based molecular therapy to treat Glioblastoma. Bioact. Mater. 2024, 33, 396–423. [Google Scholar] [CrossRef] [PubMed]
  155. Xiao, X.; Zhang, Y.; Zhang, Y.; Wang, L.; Luo, H. Study on the Treatment of Tuberous Sclerosis with Rapamycin Nanomicelles Based on Abdominal Ultrasound. J. Nanosci. Nanotechnol. 2021, 21, 962–970. [Google Scholar] [CrossRef] [PubMed]
  156. Yoon, M.S. Nanotechnology-Based Targeting of mTOR Signaling in Cancer. Int. J. Nanomed. 2020, 15, 5767–5781. [Google Scholar] [CrossRef]
  157. Jiang, T.; Du, J.; Raynald; Wang, J.; Li, C. Presurgical Administration of mTOR Inhibitors in Patients with Large Subependymal Giant Cell Astrocytoma Associated with Tuberous Sclerosis Complex. World Neurosurg. 2017, 107, 1053.e1–1053.e6. [Google Scholar] [CrossRef]
  158. Wagner, A.J.; Ravi, V.; Riedel, R.F.; Ganjoo, K.; Van Tine, B.A.; Chugh, R.; Cranmer, L.; Gordon, E.M.; Hornick, J.L.; Du, H.; et al. Phase II Trial of nab-Sirolimus in Patients with Advanced Malignant Perivascular Epithelioid Cell Tumors (AMPECT): Long-Term Efficacy and Safety Update. J. Clin. Oncol. 2024, 42, 1472–1476. [Google Scholar] [CrossRef]
Figure 1. Schema depicting ternary complex formed by FRB of mTOR, FKBP12, and rapamycin leading to allosteric inhibition of mTOR (and hence its downstream targets), thereby also opposing the overstimulation of mTOR by PI3K/AKT signaling.
Figure 1. Schema depicting ternary complex formed by FRB of mTOR, FKBP12, and rapamycin leading to allosteric inhibition of mTOR (and hence its downstream targets), thereby also opposing the overstimulation of mTOR by PI3K/AKT signaling.
Cancers 16 03406 g001
Figure 2. A summary of established (left and top boxes) and investigational (right and bottom boxes) therapeutics for SEGA. The latter is currently growing in number due to novel emerging therapies, including non-coding RNA-based therapeutics and nanocarrier technologies.
Figure 2. A summary of established (left and top boxes) and investigational (right and bottom boxes) therapeutics for SEGA. The latter is currently growing in number due to novel emerging therapies, including non-coding RNA-based therapeutics and nanocarrier technologies.
Cancers 16 03406 g002
Table 2. Results of surgery and chemotherapies of SEGA.
Table 2. Results of surgery and chemotherapies of SEGA.
Therapeutic Approach
for SEGA Tumors
Effect of TherapyReferences
SurgeryAfter surgical excision, the tumor may grow back.[32]
RadiotherapySEGA responds slowly and progressively to fractionated radiotherapy.[12]
Chemotherapy;
Everolimus (mTOR kinase
inhibitor)
35% of patients had at least 50% reduction in SEGA volume after 6–9 months of treatment with everolimus.[54,87,88]
Chemotherapy;
Sirolimus (mTOR kinase
inhibitor)
Reductions in SEGA volume of treatment with sirolimus[89]
Table 3. Investigational therapies for the treatment of SEGA.
Table 3. Investigational therapies for the treatment of SEGA.
Drug/Therapeutic ModalityExperimental/Clinical StudyMajor OutcomesReferences
Dual PI3K/mTOR inhibitorsclinical studies May provide survival benefit over standard care for gliomas; to be determined for SEGANCT05009992, NCT03970447 [122,123,124]
ERK inhibitorprimary human derived SEGA cultureDecreased proliferation in a similar manner to treatment with rapamycin[125]
Gene therapySEGA-like cell lineRecombinant lentivirus encoding human TSC1 restored the TSC1 level[126]
Inhibitors of kinase CK2: 4,5,6,7-tetrabromo-1H-benzimidazole (TBI); 2-dimethylamino-4,5,6,7-tetrabromo-1H-
benzimidazole (DMAT); 4,5, 6,7-tetrabromo-1H-benzotriazole (TBB)
cell lines established from human SEGA tumorReduced SEGA cell growth and viability[127]
Laser-induced interstitial thermotherapyclinical studiesTumor shrinkage; less invasive surgical alternative to open resection of SEGAs[128,129]
Metforminclinical studyThe effect of metformin on reducing SEGA volume was observed in patients[130]
MicroRNA-320d mimiccell cultureAmeliorated MMP/TIMP proteolytic system, dysregulated in SEGA[131]
Radiosurgeryclinical studyReducing the size of the SEGA tumor[132]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pucko, E.; Sulejczak, D.; Ostrowski, R.P. Subependymal Giant Cell Astrocytoma: The Molecular Landscape and Treatment Advances. Cancers 2024, 16, 3406. https://doi.org/10.3390/cancers16193406

AMA Style

Pucko E, Sulejczak D, Ostrowski RP. Subependymal Giant Cell Astrocytoma: The Molecular Landscape and Treatment Advances. Cancers. 2024; 16(19):3406. https://doi.org/10.3390/cancers16193406

Chicago/Turabian Style

Pucko, Emanuela, Dorota Sulejczak, and Robert P. Ostrowski. 2024. "Subependymal Giant Cell Astrocytoma: The Molecular Landscape and Treatment Advances" Cancers 16, no. 19: 3406. https://doi.org/10.3390/cancers16193406

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop