1
|
Vitale G, Terrone G, Vitale S, Vitulli F, Aiello S, Bravaccio C, Pisano S, Bove I, Rizzo F, Seetahal-Maraj P, Wiese T. The Evolving Landscape of Therapeutics for Epilepsy in Tuberous Sclerosis Complex. Biomedicines 2023; 11:3241. [PMID: 38137462 PMCID: PMC10741146 DOI: 10.3390/biomedicines11123241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/30/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a rare multisystem genetic disorder characterized by benign tumor growth in multiple organs, including the brain, kidneys, heart, eyes, lungs, and skin. Pathogenesis stems from mutations in either the TSC1 or TSC2 gene, which encode the proteins hamartin and tuberin, respectively. These proteins form a complex that inhibits the mTOR pathway, a critical regulator of cell growth and proliferation. Disruption of the tuberin-hamartin complex leads to overactivation of mTOR signaling and uncontrolled cell growth, resulting in hamartoma formation. Neurological manifestations are common in TSC, with epilepsy developing in up to 90% of patients. Seizures tend to be refractory to medical treatment with anti-seizure medications. Infantile spasms and focal seizures are the predominant seizure types, often arising in early childhood. Drug-resistant epilepsy contributes significantly to morbidity and mortality. This review provides a comprehensive overview of the current state of knowledge regarding the pathogenesis, clinical manifestations, and treatment approaches for epilepsy and other neurological features of TSC. While narrative reviews on TSC exist, this review uniquely synthesizes key advancements across the areas of TSC neuropathology, conventional and emerging pharmacological therapies, and targeted treatments. The review is narrative in nature, without any date restrictions, and summarizes the most relevant literature on the neurological aspects and management of TSC. By consolidating the current understanding of TSC neurobiology and evidence-based treatment strategies, this review provides an invaluable reference that highlights progress made while also emphasizing areas requiring further research to optimize care and outcomes for TSC patients.
Collapse
Affiliation(s)
- Giovanni Vitale
- Neuroscience and Rare Diseases, Discovery and Translational Area, Roche Pharma Research and Early Development (pRED), F. Hoffmann–La Roche, 4070 Basel, Switzerland
| | - Gaetano Terrone
- Department of Translational Medical Sciences, Child and Adolescent Neuropsychiatry, University of Naples Federico II, 80138 Naples, Italy; (G.T.); (C.B.)
| | - Samuel Vitale
- School of Medicine and Surgery, University of Naples Federico II, 80138 Naples, Italy;
| | - Francesca Vitulli
- Department of Neurosciences and Reproductive and Dental Sciences, Division of Neurosurgery, University of Naples Federico II, 80138 Naples, Italy (I.B.)
| | - Salvatore Aiello
- Department of Translational Medical Sciences, Child and Adolescent Neuropsychiatry, University of Naples Federico II, 80138 Naples, Italy; (G.T.); (C.B.)
| | - Carmela Bravaccio
- Department of Translational Medical Sciences, Child and Adolescent Neuropsychiatry, University of Naples Federico II, 80138 Naples, Italy; (G.T.); (C.B.)
| | - Simone Pisano
- Department of Translational Medical Sciences, Child and Adolescent Neuropsychiatry, University of Naples Federico II, 80138 Naples, Italy; (G.T.); (C.B.)
| | - Ilaria Bove
- Department of Neurosciences and Reproductive and Dental Sciences, Division of Neurosurgery, University of Naples Federico II, 80138 Naples, Italy (I.B.)
| | - Francesca Rizzo
- Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, Italy;
| | | | - Thomas Wiese
- Neuroscience and Rare Diseases, Discovery and Translational Area, Roche Pharma Research and Early Development (pRED), F. Hoffmann–La Roche, 4070 Basel, Switzerland
| |
Collapse
|
2
|
Subependymal Giant Cell Astrocytomas in Tuberous Sclerosis Complex-Current Views on Their Pathogenesis and Management. J Clin Med 2023; 12:jcm12030956. [PMID: 36769603 PMCID: PMC9917805 DOI: 10.3390/jcm12030956] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/21/2023] [Accepted: 01/22/2023] [Indexed: 01/28/2023] Open
Abstract
Introduction, Tuberous sclerosis complex (TSC) is an autosomal-dominant disorder caused by mutations inactivating TSC1 or TSC2 genes and characterized by the presence of tumors involving many organs, including the brain, heart, kidneys, and skin. Subependymal giant cell astrocytoma (SEGA) is a slow-growing brain tumor almost exclusively associated with TSC. STATE OF THE ART Despite the fact that SEGAs are benign, they require well-considered decisions regarding the timing and modality of pharmacological or surgical treatment. In TSC children and adolescents, SEGA is the major cause of mortality and morbidity. CLINICAL IMPLICATIONS Until recently, surgical resection has been the standard therapy for SEGAs but the discovery of the role of the mTOR pathway and the introduction of mTOR inhibitors to clinical practice changed the therapeutic landscape of these tumors. In the current paper, we discuss the pros and cons of mTOR inhibitors and surgical approaches in SEGA treatment. FUTURE DIRECTIONS In 2021, the International Tuberous Sclerosis Complex Consensus Group proposed a new integrative strategy for SEGA management. In the following review, we discuss the proposed recommendations and report the results of the literature search for the latest treatment directions.
Collapse
|
3
|
Riley VA, Holmberg JC, Sokolov AM, Feliciano DM. Tsc2 shapes olfactory bulb granule cell molecular and morphological characteristics. Front Mol Neurosci 2022; 15:970357. [PMID: 36277492 PMCID: PMC9581303 DOI: 10.3389/fnmol.2022.970357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Tuberous Sclerosis Complex (TSC) is a neurodevelopmental disorder caused by mutations that inactivate TSC1 or TSC2. Hamartin and tuberin are encoded by TSC1 and TSC2 which form a GTPase activating protein heteromer that inhibits the Rheb GTPase from activating a growth promoting protein kinase called mammalian target of rapamycin (mTOR). Growths and lesions occur in the ventricular-subventricular zone (V-SVZ), cortex, olfactory tract, and olfactory bulbs (OB) in TSC. A leading hypothesis is that mutations in inhibitory neural progenitor cells cause brain growths in TSC. OB granule cells (GCs) are GABAergic inhibitory neurons that are generated through infancy by inhibitory progenitor cells along the V-SVZ. Removal of Tsc1 from mouse OB GCs creates cellular phenotypes seen in TSC lesions. However, the role of Tsc2 in OB GC maturation requires clarification. Here, it is demonstrated that conditional loss of Tsc2 alters GC development. A mosaic model of TSC was created by performing neonatal CRE recombinase electroporation into inhibitory V-SVZ progenitors yielded clusters of ectopic cytomegalic neurons with hyperactive mTOR complex 1 (mTORC1) in homozygous Tsc2 mutant but not heterozygous or wild type mice. Similarly, homozygous Tsc2 mutant GC morphology was altered at postnatal days 30 and 60. Tsc2 mutant GCs had hypertrophic dendritic arbors that were established by postnatal day 30. In contrast, loss of Tsc2 from mature GCs had negligible effects on mTORC1, soma size, and dendrite arborization. OB transcriptome profiling revealed a network of significantly differentially expressed genes following loss of Tsc2 during development that altered neural circuitry. These results demonstrate that Tsc2 has a critical role in regulating neural development and shapes inhibitory GC molecular and morphological characteristics.
Collapse
Affiliation(s)
| | | | | | - David M. Feliciano
- Department of Biological Sciences, Clemson University, Clemson, SC, United States
| |
Collapse
|
4
|
Subependymal giant-cell astrocytoma: a surgical review in the modern era of mTOR inhibitors. Neurochirurgie 2022; 68:627-636. [PMID: 35907444 DOI: 10.1016/j.neuchi.2022.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 04/05/2022] [Accepted: 07/15/2022] [Indexed: 11/21/2022]
Abstract
INTRODUCTION Surgical removal has been the historical treatment for subependymal giant-cell astrocytoma (SEGA) in tuberous sclerosis complex (TSC) patients. In the past decade, mTOR inhibitors have shown efficacy in the treatment of SEGA, significantly reducing tumor size. The aim of this study was to assess the safety and efficacy of surgical treatment at a time when mTOR inhibitors have changed standard treatment. MATERIAL AND METHODS We conducted a single-center retrospective study including all patients treated by surgery for SEGA from October 2003 to September 2019, with a review of all SEGA surgical case series, following PRISMA guidelines. Research focused on demographics, surgical indications, surgical approach, use of CSF shunt, morbidity and mortality, resection quality, recurrence rate and treatment of recurrence, follow-up and long-term clinical status. RESULTS Eleven patients were included, with a median age at surgery of 16.0 years. Gross total resection was achieved in 8 patients (72%), with no permanent morbidity. One patient needed further surgery for tumor recurrence. Eighteen studies were reviewed, totaling 263 TSC patients affected by SEGA and 286 surgical procedures. Gross total resection was achieved in 81.1% of cases, mortality was 4.9% and permanent morbidity 6.1%. Tumor recurrence occurred in 11.5% of cases, and was secondary to partial tumor resection at first surgery in the majority of cases. CONCLUSION Surgical treatment of SEGA is still a valid and effective option. Morbidity is low and complete disappearance of SEGA can be achieved in selected cases.
Collapse
|
5
|
Familial Neoplastic Syndromes. Neurol Clin 2022; 40:405-420. [DOI: 10.1016/j.ncl.2021.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
6
|
Barnett JR, Freedman JH, Zheng H, Thiele EA, Caruso P. Growth Curves of Subependymal Giant Cell Tumors in Tuberous Sclerosis Complex. AJNR. AMERICAN JOURNAL OF NEURORADIOLOGY 2021; 42:1891-1897. [PMID: 34615647 DOI: 10.3174/ajnr.a7231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/03/2021] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Growth of subependymal giant cell tumor and subependymal nodules has not been well-characterized. The purpose of this study was to determine whether growth curves can differentiate subependymal giant cell tumors from subependymal nodules. MATERIALS AND METHODS Brain MR imaging of patients with tuberous sclerosis complex were retrospectively reviewed from 2002 to 2018. All lesions in the region of the foramen of Monro were measured. Lesions were categorized on the basis of maximal diameter at the most recent scan: small lesions (<1 cm), indeterminate lesions (>1 cm), and resected lesions (>1 cm and surgically resected). Growth velocity and acceleration on serial imaging were analyzed, and growth rates were calculated between 0 and 20 years of age and compared among the 3 categories. RESULTS Forty-one patients were analyzed. The average age at the earliest scan was 5.9 (SD = 5.7) years. One hundred twenty-six small, 27 indeterminate, and 10 resected lesions were measured. Subependymal giant cell tumors grew faster than indeterminate lesions between 6 and 15 years of age. Indeterminate lesions grew faster than small lesions at 0-10 years of age. Resected lesions showed increased velocity and acceleration of growth compared with indeterminate lesions and small lesions on serial imaging. CONCLUSIONS Growth differentiates subependymal nodules and subependymal giant cell tumors within the first 20 years of life, and the use of velocity and acceleration of growth may refine the diagnostic criteria of subependymal giant cell tumors. Additionally, 6-15 years of age may be an important period to monitor subependymal giant cell tumors at the foramen of Monro because increased growth may help to identify subependymal giant cell tumors that will continue to grow and result in obstructive hydrocephalus.
Collapse
Affiliation(s)
- J R Barnett
- From the Carol and James Herscot Center for Tuberous Sclerosis Complex (J.R.B., J.H.F., E.A.T.), Massachusetts General Hospital, Boston, Massachusetts
| | - J H Freedman
- From the Carol and James Herscot Center for Tuberous Sclerosis Complex (J.R.B., J.H.F., E.A.T.), Massachusetts General Hospital, Boston, Massachusetts
| | - H Zheng
- Biostatistics Center (H.Z.), Massachusetts General Hospital, Boston, Massachusetts
| | - E A Thiele
- From the Carol and James Herscot Center for Tuberous Sclerosis Complex (J.R.B., J.H.F., E.A.T.), Massachusetts General Hospital, Boston, Massachusetts
| | - P Caruso
- Pediatric Neuroimaging (P.C.), Lenox Hill Radiology & Medical Imaging Associates, New York
| |
Collapse
|
7
|
Mo F, Pellerino A, Rudà R. Subependymal Giant Cell Astrocytomas (SEGAs): a Model of Targeting Tumor Growth and Epilepsy. Curr Treat Options Neurol 2021. [DOI: 10.1007/s11940-021-00673-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
8
|
Reis LB, Filippi-Chiela EC, Ashton-Prolla P, Visioli F, Rosset C. The paradox of autophagy in Tuberous Sclerosis Complex. Genet Mol Biol 2021; 44:e20200014. [PMID: 33821877 PMCID: PMC8022228 DOI: 10.1590/1678-4685-gmb-2020-0014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 01/17/2021] [Indexed: 12/21/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant genetic disorder caused by germline mutations in TSC1 or TSC2 genes, which leads to the hyperactivation of the mTORC1 pathway, an important negative regulator of autophagy. This leads to the development of hamartomas in multiple organs. The variability in symptoms presents a challenge for the development of completely effective treatments for TSC. One option is the treatment with mTORC1 inhibitors, which are targeted to block cell growth and restore autophagy. However, the therapeutic effect of rapamycin seems to be more efficient in the early stages of hamartoma development, an effect that seems to be associated with the paradoxical role of autophagy in tumor establishment. Under normal conditions, autophagy is directly inhibited by mTORC1. In situations of bioenergetics stress, mTORC1 releases the Ulk1 complex and initiates the autophagy process. In this way, autophagy promotes the survival of established tumors by supplying metabolic precursors during nutrient deprivation; paradoxically, excessive autophagy has been associated with cell death in some situations. In spite of its paradoxical role, autophagy is an alternative therapeutic strategy that could be explored in TSC. This review compiles the findings related to autophagy and the new therapeutic strategies targeting this pathway in TSC.
Collapse
Affiliation(s)
- Larissa Brussa Reis
- Hospital de Clínicas de Porto Alegre (HCPA), Serviço de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul (UFRGS), Programa de Pós-Graduação em Genética e Biologia Molecular, Porto Alegre, RS, Brazil
| | - Eduardo C Filippi-Chiela
- Hospital de Clínicas de Porto Alegre (HCPA), Serviço de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul (UFRGS), Instituto de Ciências Básicas da Saúde, Departamento de Ciências Morfológicas, Porto Alegre, RS, Brazil
| | - Patricia Ashton-Prolla
- Hospital de Clínicas de Porto Alegre (HCPA), Serviço de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul (UFRGS), Programa de Pós-Graduação em Genética e Biologia Molecular, Porto Alegre, RS, Brazil.,Hospital de Clínicas de Porto Alegre (HCPA), Serviço de Genética Médica, Porto Alegre, RS, Brazil
| | - Fernanda Visioli
- Universidade Federal do Rio Grande do Sul, Faculdade de Odontologia, Departamento de Patologia Oral, Porto Alegre, RS, Brazil
| | - Clévia Rosset
- Hospital de Clínicas de Porto Alegre (HCPA), Serviço de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul (UFRGS), Programa de Pós-Graduação em Genética e Biologia Molecular, Porto Alegre, RS, Brazil
| |
Collapse
|
9
|
Tomoto K, Fujimoto A, Inenaga C, Okanishi T, Imai S, Ogai M, Fukunaga A, Nakamura H, Sato K, Obana A, Masui T, Arai Y, Enoki H. Experience using mTOR inhibitors for subependymal giant cell astrocytoma in tuberous sclerosis complex at a single facility. BMC Neurol 2021; 21:139. [PMID: 33784976 PMCID: PMC8011204 DOI: 10.1186/s12883-021-02160-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 03/15/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Subependymal giant cell astrocytoma (SEGA) is occasionally seen in tuberous sclerosis complex (TSC). Two main options are currently available for treating SEGA: surgical resection or pharmacotherapy using mammalian target of rapamycin inhibitors (mTORi). We hypothesized that opportunities for surgical resection of SEGA would have reduced with the advent of mTORi. METHODS We retrospectively reviewed the charts of patients treated between August 1979 and July 2020, divided into a pre-mTORi era group (Pre-group) of patients treated before November 2012, and a post-mTORi era group (Post-group) comprising patients treated from November 2012, when mTORi became available in Japan for SEGA. We compared groups in terms of treatment with surgery or mTORi. We also reviewed SEGA size, rate of acute hydrocephalus, recurrence of SEGA, malignant transformation and adverse effects of mTORi. RESULTS In total, 120 patients with TSC visited our facility, including 24 patients with SEGA. Surgical resection was significantly more frequent in the Pre-group (6 of 7 patients, 86 %) than in the Post-group (2 of 17 patients, 12 %; p = 0.001). Acute hydrocephalus was seen in 1 patient (4 %), and no patients showed malignant transformation of SEGA. The group treated using mTORi showed significantly smaller SEGA compared with the group treated under a wait-and-see policy (p = 0.012). Adverse effects of pharmacotherapy were identified in seven (64 %; 6 oral ulcers, 1 irregular menstruation) of the 11 patients receiving mTORi. CONCLUSIONS The Post-group underwent surgery significantly less often than the Pre-group. Since the treatment option to use mTORi in the treatment of SEGA in TSC became available, opportunities for surgical resection have decreased in our facility.
Collapse
Affiliation(s)
- Kyoichi Tomoto
- Department of Neurosurgery, Seirei Hamamatsu General Hospital, 2-12-12 Sumiyoshi, Nakaku, Hamamatsu, 430-8558, Shizuoka, Japan
| | - Ayataka Fujimoto
- Department of Neurosurgery, Seirei Hamamatsu General Hospital, 2-12-12 Sumiyoshi, Nakaku, Hamamatsu, 430-8558, Shizuoka, Japan.
- Tuberous Sclerosis Complex Board, Seirei Hamamatsu General Hospital, 2-12-12 Sumiyoshi, Nakaku, Hamamatsu, 430-8558, Shizuoka, Japan.
| | - Chikanori Inenaga
- Department of Neurosurgery, Seirei Hamamatsu General Hospital, 2-12-12 Sumiyoshi, Nakaku, Hamamatsu, 430-8558, Shizuoka, Japan
| | - Tohru Okanishi
- Tuberous Sclerosis Complex Board, Seirei Hamamatsu General Hospital, 2-12-12 Sumiyoshi, Nakaku, Hamamatsu, 430-8558, Shizuoka, Japan
| | - Shin Imai
- Tuberous Sclerosis Complex Board, Seirei Hamamatsu General Hospital, 2-12-12 Sumiyoshi, Nakaku, Hamamatsu, 430-8558, Shizuoka, Japan
| | - Masaaki Ogai
- Tuberous Sclerosis Complex Board, Seirei Hamamatsu General Hospital, 2-12-12 Sumiyoshi, Nakaku, Hamamatsu, 430-8558, Shizuoka, Japan
| | - Akiko Fukunaga
- Tuberous Sclerosis Complex Board, Seirei Hamamatsu General Hospital, 2-12-12 Sumiyoshi, Nakaku, Hamamatsu, 430-8558, Shizuoka, Japan
| | - Hidenori Nakamura
- Tuberous Sclerosis Complex Board, Seirei Hamamatsu General Hospital, 2-12-12 Sumiyoshi, Nakaku, Hamamatsu, 430-8558, Shizuoka, Japan
| | - Keishiro Sato
- Tuberous Sclerosis Complex Board, Seirei Hamamatsu General Hospital, 2-12-12 Sumiyoshi, Nakaku, Hamamatsu, 430-8558, Shizuoka, Japan
| | - Akira Obana
- Tuberous Sclerosis Complex Board, Seirei Hamamatsu General Hospital, 2-12-12 Sumiyoshi, Nakaku, Hamamatsu, 430-8558, Shizuoka, Japan
| | - Takayuki Masui
- Tuberous Sclerosis Complex Board, Seirei Hamamatsu General Hospital, 2-12-12 Sumiyoshi, Nakaku, Hamamatsu, 430-8558, Shizuoka, Japan
| | - Yoshifumi Arai
- Tuberous Sclerosis Complex Board, Seirei Hamamatsu General Hospital, 2-12-12 Sumiyoshi, Nakaku, Hamamatsu, 430-8558, Shizuoka, Japan
| | - Hideo Enoki
- Tuberous Sclerosis Complex Board, Seirei Hamamatsu General Hospital, 2-12-12 Sumiyoshi, Nakaku, Hamamatsu, 430-8558, Shizuoka, Japan
| |
Collapse
|
10
|
Gaillard AL, Crombé A, Jecko V, Bessou P, Havez M, Pédespan JM, Van Gils J, Chateil JF. Magnetic resonance imaging diagnosis of subependymal giant cell astrocytomas in follow-up of children with tuberous sclerosis complex: should we always use contrast enhancement? Pediatr Radiol 2020; 50:1397-1408. [PMID: 32671416 DOI: 10.1007/s00247-020-04707-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/08/2020] [Accepted: 05/05/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Subependymal giant cell astrocytomas (SEGAs) arise in 10-26% of tuberous sclerosis complex (TSC) patients. SEGAs cause obstructive hydrocephalus and increase morbi-mortality. It is recommended that TSC patients be followed with contrast enhanced magnetic resonance imaging (CE-MRI), but repetitive use of gadolinium-based contrast-agents (GBCAs) may cause organ deposits. OBJECTIVE To compare the diagnostic performances of non-CE- and CE-MRI to differentiate SEGAs from subependymal nodules in TSC patients during follow-up. MATERIALS AND METHODS Thirty-five TSC patients (median age: 2.4 years) were enrolled in this retrospective single-center study from September 2007 to January 2019. Inclusion criteria were a certain diagnosis of TSC and at least three follow-up brain MRIs with GBCA injection. Two consecutive MRI scans per patient were selected and anonymized. Three radiologists performed a blinded review of non-enhanced and enhanced MRI sequences during different sessions. The diagnostic performances were compared (sensitivity, specificity, positive/negative predictive values, accuracy, inter/intra-observer agreements). RESULTS The accuracies for detecting SEGAs were good and similar between the non-enhanced and enhanced MRI sequences. The sensitivity and specificity of non-CE-MRI to diagnose SEGA ranged from 75% to 100% and from 94% to 100%, respectively. The differences in numbers of false-positive and false-negative patients between non-CE- and CE-MRI never exceeded one case. Nodules size >10 mm, location near the Monro foramen, hydrocephalus and modifications between two consecutive MRI scans were significantly associated with the diagnosis of SEGA for the three readers (all P-values <0.05). Inter- and intra-observer agreements were also excellent for non-enhanced and enhanced MRI sequences (kappa=0.85-1 and 0.81-0.93, respectively). CONCLUSION The performances of non-enhanced and enhanced MRI sequences are comparable for detecting SEGAs, questioning the need for systematic GBCA injections for TSC patients.
Collapse
Affiliation(s)
| | - Amandine Crombé
- Department of Diagnostic and Interventional Oncologic Imaging, Institut Bergonié, Bordeaux, France.,University of Bordeaux, Bordeaux, France
| | - Vincent Jecko
- Department of Neurosurgery, CHU Bordeaux, Bordeaux, France
| | - Pierre Bessou
- Unit of Pediatric Imaging, Pellegrin Hospital, Bordeaux, France
| | - Marion Havez
- Unit of Pediatric Imaging, Pellegrin Hospital, Bordeaux, France
| | | | | | - Jean-François Chateil
- Unit of Pediatric Imaging, Pellegrin Hospital, Bordeaux, France. .,University of Bordeaux/CNRS, CRMSB, UMR 5536, F-33076, Bordeaux, France.
| |
Collapse
|
11
|
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. [PMID: 32103336 DOI: 10.1007/s00381-020-04551-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 02/19/2020] [Indexed: 11/26/2022]
Abstract
PURPOSE Subependymal giant-cell astrocytomas (SEGAs) are low grade intraventricular tumors typically found in patients with tuberous sclerosis complex (TSC). The occurrence of SEGA in non TSC patients is very rare and from a genetic point of view these so-called solitary SEGA are thought to result either from somatic mutations in one of the TSC genes (TSC1 or TSC2) limited to the tumor, or be part of a "forme fruste" of TSC with somatic mosaicism. We report on three new cases of solitary SEGA with germline and somatic mutation analysis. METHODS We retrospectively analyzed TSC genes in three patients with a solitary SEGA using next-generation sequencing technique. RESULTS In the three patients, a somatic mutation of TSC1 or TSC2 was found only in the tumor cells: one patient had a TSC1 heterozygote mutation, involving the natural acceptor splicing site of intron 15 (c.1998-1G > A (p.?). Two patients had a TSC2 mutation located in the canonical splicing donor site of intron 5 (c.599 + 1G > A) in 70% of the alleles in one patient and in exon 9: c.949_955dup7 (p.V319DfxX21) in 25 of the alleles in the second patient. No other TSC mutations were found in patient's blood or tumor and those identified mutations were absent in blood DNA from parents and siblings. CONCLUSION We therefore conclude that solitary SEGA can occur with a TSC1 or TSC2 mutation limited to the tumor in patients without TSC.
Collapse
Affiliation(s)
- Martine Fohlen
- Service de neurochirurgie pediatrique, Foundation Rothschild Hospital, Paris, France.
| | - Ines Harzallah
- Service de Génétique Clinique, Chromosomique et Moléculaire, Centre de Compétence des Epilepsies rares (CReER), CHU-Hôpital Nord, Saint Etienne, France
| | - Marc Polivka
- Service d'anatomie et cytologie pathologiques, Hôpital Lariboisière, Paris, France
| | - Fabienne Giuliano
- Service de génétique, Centre Hospitalier Universitaire, Nice, France
| | - Linda Pons
- Service de Génétique, Centre de Référence des Épilepsies Rares (CReER), Hôpital Femme Mère Enfant, Bron, France
| | - Nathalie Streichenberger
- Centre de Pathologie et Neuropathologie Est, Hospices Civils de Lyon; Université Claude Bernard Lyon1, Lyon, France
- Institut NeuroMyogène, CNRS UMR 5310-INSERM U1217, Villeurbanne, France
| | - Georg Dorfmüller
- Service de neurochirurgie pediatrique, Foundation Rothschild Hospital, Paris, France
| | - Renaud Touraine
- Service de Génétique Clinique, Chromosomique et Moléculaire, Centre de Compétence des Epilepsies rares (CReER), CHU-Hôpital Nord, Saint Etienne, France
| |
Collapse
|
12
|
Giordano F, Moscheo C, Lenge M, Biagiotti R, Mari F, Sardi I, Buccoliero AM, Mongardi L, Aronica E, Guerrini R, Genitori L. Neurosurgical treatment of subependymal giant cell astrocytomas in tuberous sclerosis complex: a series of 44 surgical procedures in 31 patients. Childs Nerv Syst 2020; 36:951-960. [PMID: 31853898 DOI: 10.1007/s00381-019-04449-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 11/19/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Subependymal giant cell astrocytomas (SEGA) are benign tumors characteristic of tuberous sclerosis complex (TSC) that may cause hydrocephalus. Various treatments are nowadays available as mTOR inhibitors or surgery. Surgery is still a valid option especially for symptomatic and larger tumors. METHODS From January 1994 to December 2015, 31 TSC patients harboring SEGA underwent surgery at the Department of Neurosurgery of the Meyer Pediatric Hospital, Florence. Indications for surgery were tumor size and location, growth and cystization/hemorrhage, and hydrocephalus. Clinical data, preoperative and postoperative MRI, recurrence rate, further surgical procedures, and related complications were analyzed. RESULTS A total of 44 surgeries were performed in 31 TSC patients affected by SEGA, achieving gross total removal (GTR) and subtotal removal (STR), respectively, in 36 and 8 patients. Recurrences occurred in 11 patients; 9 of them underwent further surgical procedures and 2 were treated with mTOR pathway inhibitors. Surgical morbidity and mortality were, respectively, 22.7% and 2.3%. After a mean follow-up of 4.9 years, 90% of patients were tumor-free with good neurological status in 93.3%; twelve (40%) had a ventriculo-peritoneal shunt (VPS) for hydrocephalus. CONCLUSIONS The present series confirms that the surgical approach, combined with mTOR inhibitors, is still a valid option for the treatment of SEGAs.
Collapse
Affiliation(s)
- Flavio Giordano
- Department of Neurosurgery, Children's Hospital A. Meyer, University of Florence, viale Pieraccini 24, Florence, 50139, Italy.
| | - Carla Moscheo
- Neuro-oncology Unit, Children's Hospital A. Meyer, University of Florence, viale Pieraccini 24, Florence, 50139, Italy
| | - Matteo Lenge
- Department of Neurosurgery, Children's Hospital A. Meyer, University of Florence, viale Pieraccini 24, Florence, 50139, Italy.,3. Pediatric Neurology, Children's Hospital A. Meyer, University of Florence, viale Pieraccini 24, Florence, 50139, Italy
| | - Roberto Biagiotti
- Division of Prenatal Diagnosis, Children's Hospital A. Meyer, University of Florence, viale Pieraccini 24, Florence, 50139, Italy
| | - Francesco Mari
- 3. Pediatric Neurology, Children's Hospital A. Meyer, University of Florence, viale Pieraccini 24, Florence, 50139, Italy
| | - Iacopo Sardi
- Neuro-oncology Unit, Children's Hospital A. Meyer, University of Florence, viale Pieraccini 24, Florence, 50139, Italy
| | - Anna Maria Buccoliero
- Pathology Unit, Children's Hospital A. Meyer, University of Florence, viale Pieraccini 24, Florence, 50139, Italy
| | - Lorenzo Mongardi
- Neurosurgery, Sant'Anna Hospital, Via Aldo Moro, Ferrara, 44124, Italy
| | - Eleonora Aronica
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| | - Renzo Guerrini
- 3. Pediatric Neurology, Children's Hospital A. Meyer, University of Florence, viale Pieraccini 24, Florence, 50139, Italy
| | - Lorenzo Genitori
- Department of Neurosurgery, Children's Hospital A. Meyer, University of Florence, viale Pieraccini 24, Florence, 50139, Italy
| |
Collapse
|
13
|
Kútna V, Uttl L, Waltereit R, Krištofiková Z, Kaping D, Petrásek T, Hoschl C, Ovsepian SV. Tuberous Sclerosis (tsc2+/-) Model Eker Rats Reveals Extensive Neuronal Loss with Microglial Invasion and Vascular Remodeling Related to Brain Neoplasia. Neurotherapeutics 2020; 17:329-339. [PMID: 31820275 PMCID: PMC7007483 DOI: 10.1007/s13311-019-00812-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is a genetic disorder characterized by frequent noncancerous neoplasia in the brain, which can induce a range of severe neuropsychiatric symptoms in humans, resulting from out of control tissue growth. The causative spontaneous loss-of-function mutations have been also identified in rats. Herein, we studied histopathological and molecular changes in brain lesions of the Eker rat model carrying germline mutation of the tsc2 gene, predisposed to multiple neoplasias. Predominant subcortical tumors were analyzed, along with a rare form occurring within the pyriform lobe. The uniform composition of lesions supports the histochemical parity of malformations, with immunofluorescence data supporting their neuro-glial origin. Massive depletion of mature neurons and axonal loss were evident within lesions, with occasional necrotic foci implying advanced stage of pathology. Enrichment of mesenchymal-derived cell markers with hallmarks of neurogenesis and active microglia imply enhanced cell proliferation, with local immune response. The depletion of capillaries within the core was complemented by the formation of dense mesh of nascent vessels at the interface of neoplasia with healthy tissue, implying large-scale vascular remodeling. Taken as a whole, these findings present several novel features of brain tumors in Eker rat model, rendering it suitable for studies of the pathobiology and progression of primary brain tumors, with therapeutic interventions.
Collapse
Affiliation(s)
- Viera Kútna
- Department of Experimental Neurobiology, National Institute of Mental Health, Topolová 748, 250 67 Klecany, Czech Republic
| | - Libor Uttl
- Department of Experimental Neurobiology, National Institute of Mental Health, Topolová 748, 250 67 Klecany, Czech Republic
| | - Robert Waltereit
- University Hospital Carl Gustav Carus, Technical University, Department of Child and Adolescent Psychiatry, Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Zdenka Krištofiková
- Department of Experimental Neurobiology, National Institute of Mental Health, Topolová 748, 250 67 Klecany, Czech Republic
| | - Daniel Kaping
- Department of Experimental Neurobiology, National Institute of Mental Health, Topolová 748, 250 67 Klecany, Czech Republic
| | - Tomáš Petrásek
- Department of Experimental Neurobiology, National Institute of Mental Health, Topolová 748, 250 67 Klecany, Czech Republic
| | - Cyril Hoschl
- Department of Experimental Neurobiology, National Institute of Mental Health, Topolová 748, 250 67 Klecany, Czech Republic
- Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Ruská 87, 100 00 Prague 10, Czech Republic
| | - Saak V. Ovsepian
- Department of Experimental Neurobiology, National Institute of Mental Health, Topolová 748, 250 67 Klecany, Czech Republic
- Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Ruská 87, 100 00 Prague 10, Czech Republic
| |
Collapse
|
14
|
Bongaarts A, van Scheppingen J, Korotkov A, Mijnsbergen C, Anink JJ, Jansen FE, Spliet WGM, den Dunnen WFA, Gruber VE, Scholl T, Samueli S, Hainfellner JA, Feucht M, Kotulska K, Jozwiak S, Grajkowska W, Buccoliero AM, Caporalini C, Giordano F, Genitori L, Coras R, Blümcke I, Krsek P, Zamecnik J, Meijer L, Scicluna BP, Schouten-van Meeteren AYN, Mühlebner A, Mills JD, Aronica E. The coding and non-coding transcriptional landscape of subependymal giant cell astrocytomas. Brain 2020; 143:131-149. [PMID: 31834371 PMCID: PMC6935755 DOI: 10.1093/brain/awz370] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 09/13/2019] [Accepted: 10/01/2019] [Indexed: 12/12/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominantly inherited neurocutaneous disorder caused by inactivating mutations in TSC1 or TSC2, key regulators of the mechanistic target of rapamycin complex 1 (mTORC1) pathway. In the CNS, TSC is characterized by cortical tubers, subependymal nodules and subependymal giant cell astrocytomas (SEGAs). SEGAs may lead to impaired circulation of CSF resulting in hydrocephalus and raised intracranial pressure in patients with TSC. Currently, surgical resection and mTORC1 inhibitors are the recommended treatment options for patients with SEGA. In the present study, high-throughput RNA-sequencing (SEGAs n = 19, periventricular control n = 8) was used in combination with computational approaches to unravel the complexity of SEGA development. We identified 9400 mRNAs and 94 microRNAs differentially expressed in SEGAs compared to control tissue. The SEGA transcriptome profile was enriched for the mitogen-activated protein kinase (MAPK) pathway, a major regulator of cell proliferation and survival. Analysis at the protein level confirmed that extracellular signal-regulated kinase (ERK) is activated in SEGAs. Subsequently, the inhibition of ERK independently of mTORC1 blockade decreased efficiently the proliferation of primary patient-derived SEGA cultures. Furthermore, we found that LAMTOR1, LAMTOR2, LAMTOR3, LAMTOR4 and LAMTOR5 were overexpressed at both gene and protein levels in SEGA compared to control tissue. Taken together LAMTOR1-5 can form a complex, known as the 'Ragulator' complex, which is known to activate both mTORC1 and MAPK/ERK pathways. Overall, this study shows that the MAPK/ERK pathway could be used as a target for treatment independent of, or in combination with mTORC1 inhibitors for TSC patients. Moreover, our study provides initial evidence of a possible link between the constitutive activated mTORC1 pathway and a secondary driver pathway of tumour growth.
Collapse
Affiliation(s)
- Anika Bongaarts
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Anatoly Korotkov
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Caroline Mijnsbergen
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jasper J Anink
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Floor E Jansen
- Department of Pediatric Neurology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wim G M Spliet
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wilfred F A den Dunnen
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Victoria E Gruber
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Theresa Scholl
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Sharon Samueli
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | | | - Martha Feucht
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Katarzyna Kotulska
- Department of Neurology and Epileptology, Children's Memorial Health Institute, Warsaw, Poland
| | - Sergiusz Jozwiak
- Department of Neurology and Epileptology, Children's Memorial Health Institute, Warsaw, Poland
- Department of Child Neurology, Medical University of Warsaw, Warsaw, Poland
| | - Wieslawa Grajkowska
- Department of Pathology, Children's Memorial Health Institute, Warsaw, Poland
| | | | | | - Flavio Giordano
- Department of Neurosurgery, Anna Meyer Children's Hospital, Florence, Italy
| | - Lorenzo Genitori
- Department of Neurosurgery, Anna Meyer Children's Hospital, Florence, Italy
| | - Roland Coras
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Ingmar Blümcke
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Pavel Krsek
- Department of Paediatric Neurology, Charles University, 2nd Faculty of Medicine, Motol University Hospital, Prague, Czech Republic
| | - Josef Zamecnik
- Department of Pathology and Molecular Medicine, Charles University, 2nd Faculty of Medicine, Motol University Hospital, Prague, Czech Republic
| | - Lisethe Meijer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Brendon P Scicluna
- Center for Experimental and Molecular Medicine and Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Antoinette Y N Schouten-van Meeteren
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pediatric Oncology, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Angelika Mühlebner
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - James D Mills
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), The Netherlands
| |
Collapse
|
15
|
Jansen AC, Belousova E, Benedik MP, Carter T, Cottin V, Curatolo P, Dahlin M, D'Amato L, d'Augères GB, de Vries PJ, Ferreira JC, Feucht M, Fladrowski C, Hertzberg C, Jozwiak S, Lawson JA, Macaya A, Marques R, Nabbout R, O'Callaghan F, Qin J, Sander V, Sauter M, Shah S, Takahashi Y, Touraine R, Youroukos S, Zonnenberg B, Kingswood JC. Clinical Characteristics of Subependymal Giant Cell Astrocytoma in Tuberous Sclerosis Complex. Front Neurol 2019; 10:705. [PMID: 31333563 PMCID: PMC6616060 DOI: 10.3389/fneur.2019.00705] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/14/2019] [Indexed: 01/10/2023] Open
Abstract
Background: This study evaluated the characteristics of subependymal giant cell astrocytoma (SEGA) in patients with tuberous sclerosis complex (TSC) entered into the TuberOus SClerosis registry to increase disease Awareness (TOSCA). Methods: The study was conducted at 170 sites across 31 countries. Data from patients of any age with a documented clinical visit for TSC in the 12 months preceding enrollment or those newly diagnosed with TSC were entered. Results: SEGA were reported in 554 of 2,216 patients (25%). Median age at diagnosis of SEGA was 8 years (range, <1-51), with 18.1% diagnosed after age 18 years. SEGA growth occurred in 22.7% of patients aged ≤ 18 years and in 11.6% of patients aged > 18 years. SEGA were symptomatic in 42.1% of patients. Symptoms included increased seizure frequency (15.8%), behavioural disturbance (11.9%), and regression/loss of cognitive skills (9.9%), in addition to those typically associated with increased intracranial pressure. SEGA were significantly more frequent in patients with TSC2 compared to TSC1 variants (33.7 vs. 13.2 %, p < 0.0001). Main treatment modalities included surgery (59.6%) and mammalian target of rapamycin (mTOR) inhibitors (49%). Conclusions: Although SEGA diagnosis and growth typically occurs during childhood, SEGA can occur and grow in both infants and adults.
Collapse
Affiliation(s)
- Anna C. Jansen
- Pediatric Neurology Unit, Department of Pediatrics, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Elena Belousova
- Research and Clinical Institute of Pediatrics, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Mirjana P. Benedik
- Child Neurology Department, SPS Pediatrična Klinika, Ljubljana, Slovenia
| | - Tom Carter
- Tuberous Sclerosis Association, Nottingham, United Kingdom
| | - Vincent Cottin
- Hôpital Louis Pradel, Claude Bernard University Lyon 1, Lyon, France
| | - Paolo Curatolo
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University Hospital, Rome, Italy
| | - Maria Dahlin
- Neuropediatric Department, Astrid Lindgren Childrens Hospital, Stockholm, Sweden
| | | | | | - Petrus J. de Vries
- Division of Child and Adolescent Psychiatry, University of Cape Town, Cape Town, South Africa
| | - José C. Ferreira
- Neurology Department, Centro Hospitalar Lisboa Ocidental, Lisbon, Portugal
| | - Martha Feucht
- Medical University of Vienna, Universitätsklinik für Kinder-und Jugendheilkunde, Vienna, Austria
| | - Carla Fladrowski
- Associazione Sclerosi Tuberosa ONLUS, Milan, Italy
- European Tuberous Sclerosis Complex Association, In den Birken, Dattein, Germany
| | - Christoph Hertzberg
- Zentrum für Neuropädiatrie und Sozialpädiatrie, Vivantes-Klinikum Neukölln, Berlin, Germany
| | - Sergiusz Jozwiak
- Department of Child Neurology, Warsaw Medical University, Warsaw, Poland
- Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warsaw, Poland
| | - John A. Lawson
- The Tuberous Sclerosis Multidisciplinary Management Clinic, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Alfons Macaya
- Pediatric Neurology Section, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Ruben Marques
- Novartis Farma S.p.A., Origgio, Italy
- Institute of Biomedicine, University of Leon, León, Spain
| | - Rima Nabbout
- Department of Pediatric Neurology, Necker Enfants Malades Hospital, Paris Descartes University, Paris, France
| | - Finbar O'Callaghan
- Institute of Child Health, University College London, London, United Kingdom
| | - Jiong Qin
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Valentin Sander
- Neurology and Rehabilitation, Tallinn Children Hospital, Tallinn, Estonia
| | - Matthias Sauter
- Klinikum Kempten, Klinikverbund Kempten-Oberallgäu gGmbH, Kempten, Germany
| | - Seema Shah
- Novartis Healthcare Pvt. Ltd., Hyderabad, India
| | - Yukitoshi Takahashi
- National Epilepsy Center, Shizuoka Institute of Epilepsy and Neurological Disorders, NHO, Shizuoka, Japan
| | - Renaud Touraine
- Department of Genetics, CHU-Hôpital Nord, Saint Etienne, France
| | - Sotiris Youroukos
- First Department of Paediatrics, St. Sophia Children's Hospital, Athens University, Athens, Greece
| | - Bernard Zonnenberg
- Department of Internal Medicine, University Medical Center, Utrecht, Netherlands
| | - John C. Kingswood
- Cardiology Clinical Academic Group, Molecular and Clinical Sciences Research Centre, St. Georges University of London, London, United Kingdom
| |
Collapse
|
16
|
Mühlebner A, Bongaarts A, Sarnat HB, Scholl T, Aronica E. New insights into a spectrum of developmental malformations related to mTOR dysregulations: challenges and perspectives. J Anat 2019; 235:521-542. [PMID: 30901081 DOI: 10.1111/joa.12956] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2019] [Indexed: 12/20/2022] Open
Abstract
In recent years the role of the mammalian target of rapamycin (mTOR) pathway has emerged as crucial for normal cortical development. Therefore, it is not surprising that aberrant activation of mTOR is associated with developmental malformations and epileptogenesis. A broad spectrum of malformations of cortical development, such as focal cortical dysplasia (FCD) and tuberous sclerosis complex (TSC), have been linked to either germline or somatic mutations in mTOR pathway-related genes, commonly summarised under the umbrella term 'mTORopathies'. However, there are still a number of unanswered questions regarding the involvement of mTOR in the pathophysiology of these abnormalities. Therefore, a monogenetic disease, such as TSC, can be more easily applied as a model to study the mechanisms of epileptogenesis and identify potential new targets of therapy. Developmental neuropathology and genetics demonstrate that FCD IIb and hemimegalencephaly are the same diseases. Constitutive activation of mTOR signalling represents a shared pathogenic mechanism in a group of developmental malformations that have histopathological and clinical features in common, such as epilepsy, autism and other comorbidities. We seek to understand the effect of mTOR dysregulation in a developing cortex with the propensity to generate seizures as well as the aftermath of the surrounding environment, including the white matter.
Collapse
Affiliation(s)
- A Mühlebner
- Department of Neuropathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - A Bongaarts
- Department of Neuropathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - H B Sarnat
- Departments of Paediatrics, Pathology (Neuropathology) and Clinical Neurosciences, University of Calgary Cumming School of Medicine and Alberta Children's Hospital Research Institute (Owerko Centre), Calgary, AB, Canada
| | - T Scholl
- Department of Paediatric and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - E Aronica
- Department of Neuropathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Amsterdam, The Netherlands
| |
Collapse
|
17
|
Chan DL, Calder T, Lawson JA, Mowat D, Kennedy SE. The natural history of subependymal giant cell astrocytomas in tuberous sclerosis complex: a review. Rev Neurosci 2018; 29:295-301. [PMID: 29211682 DOI: 10.1515/revneuro-2017-0027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/11/2017] [Indexed: 11/15/2022]
Abstract
Tuberous sclerosis complex (TSC) is an auto-somal-dominant inherited condition with an incidence of approximately 1:6000 births, characterised by deregulated mTOR activity with multi-site hamartomas. Subependymal giant cell astrocytomas (SEGA) are one such hamartoma, affecting up to 24% of patients with TSC. Their intraventricular location may lead to life-threatening obstructive hydrocephalus. Current management is hampered by a lack of understanding regarding the natural history, behaviour and growth patterns of SEGA. We review the current literature to summarise what is known about SEGA in the following areas: (1) diagnostic criteria, (2) prevalence, (3) origin, (4) imaging characteristics, (5) growth rate, (6) genotype-phenotype correlation, (7) congenital SEGA and (8) SEGA as a marker of severity of other TSC manifestations.
Collapse
Affiliation(s)
- Denise L Chan
- Department of Neurology, Sydney Children's Hospital, Randwick, NSW 2031, Australia
| | - Tessa Calder
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW 2000, Australia
| | - John A Lawson
- Department of Neurology, Sydney Children's Hospital, Randwick, NSW 2031, Australia
| | - David Mowat
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW 2000, Australia
| | - Sean E Kennedy
- Department of Neurology, Sydney Children's Hospital, Randwick, NSW 2031, Australia
| |
Collapse
|
18
|
Fohlen M, Ferrand-Sorbets S, Delalande O, Dorfmüller G. Surgery for subependymal giant cell astrocytomas in children with tuberous sclerosis complex. Childs Nerv Syst 2018; 34:1511-1519. [PMID: 29766265 DOI: 10.1007/s00381-018-3826-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 05/06/2018] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Subependymal giant cell astrocytomas (SEGAs) are low-grade intraventricular glial tumors that develop in 10-15% of patients with tuberous sclerosis complex; they often cause hydrocephalus and are potentially accessible to a surgical treatment. Our aim is to evaluate morbidity and results after surgery in symptomatic and asymptomatic patients. METHOD We present a retrospective series of 18 pediatric patients operated on for SEGA between 2006 and 2016 at our institution. We reviewed surgical indications, preoperative clinical and radiologic data, surgical management, and clinical and radiological follow-up. RESULTS Mean age at surgery was 10.7 years. The surgical decision was based on clinical signs of raised intracranial pressure due to hydrocephalus in 8 and on radiological findings without any clinical signs in the other 10 patients (increased in SEGA volume with or without ventricular enlargement). Surgical treatment consisted in a frontal trans-ventricular microsurgical approach in 17 patients and an endoscopic approach in 1. External ventricular drainage was placed in all the patients but 1. Ventriculoperitoneal shunting (VPS) became necessary in 6 patients, all of them presenting with a preoperative active hydrocephalus. Morbidity appeared very low with meningitis occurring in 1 patient. Resection was complete in 15 children with no recurrence during a mean follow-up of 5.25 years and incomplete in 3 requiring a second surgery. CONCLUSION Surgery of SEGA represents a very effective treatment with low morbidity and no mortality in the present series. In patients operated before the onset of clinical signs of hydrocephalus, internal VPS could be avoided whereas in others, an additional shunt surgery became necessary. This gives arguments in favor of a regular MRI surveillance in tuberous sclerosis complex patients with SEGA in order to best propose resective surgery once a growth of tumor and/or ventricular size have been confirmed but before raised intracranial pressure occurs.
Collapse
Affiliation(s)
- Martine Fohlen
- Department of Pediatric Neurosurgery, Fondation Ophtalmologique A. de Rothschild, Paris, France.
| | - Sarah Ferrand-Sorbets
- Department of Pediatric Neurosurgery, Fondation Ophtalmologique A. de Rothschild, Paris, France
| | - Olivier Delalande
- Department of Pediatric Neurosurgery, Fondation Ophtalmologique A. de Rothschild, Paris, France
| | - Georg Dorfmüller
- Department of Pediatric Neurosurgery, Fondation Ophtalmologique A. de Rothschild, Paris, France
| |
Collapse
|
19
|
Mei GH, Liu XX, Zhou P, Shen M. Clinical and imaging features of subependymal giant cell astrocytoma: report of 20 cases. Chin Neurosurg J 2017. [DOI: 10.1186/s41016-017-0077-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
20
|
Abstract
Neoplasms in the central (CNS) and peripheral nervous system (PNS) in hereditary tumor syndromes play an important role in the neuropathological diagnostics. The benign and malignant PNS and CNS tumors that occur in the frequent neurofibromatosis type 1 (NF1) and type 2 (NF2) often represent essential factors for the course of the disease in those affected. Furthermore, certain clinical constellations (e.g. bilateral schwannomas of the auditory nerve, schwannomas at a young age and multiple meningiomas) can be important indications for a previously undiagnosed hereditary tumor disease. Other tumors occur practically regularly in association with certain germline defects, e.g. subependymal giant cell astrocytoma (SEGA) in tuberous sclerosis and dysplastic gangliocytoma of the cerebellum in Cowden's syndrome and can be indications in the diagnostics for an extended genetic counselling. This is not only important because many germline defects are based on new mutations, but also for the now established targeted therapy of certain tumors, e.g. inhibition of the mammalian target of rapamycin (mTOR) signaling pathway using temsirolimus for SEGA. Furthermore, knowledge about the possible constellations of genetic mosaics in hereditary tumor syndromes with the resulting (incomplete) syndrome manifestations is useful. This review article summarizes the most important hereditary tumor syndromes with involvement of the PNS and CNS.
Collapse
Affiliation(s)
- C Mawrin
- Institut für Neuropathologie, Otto-von-Guericke-Universität Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Deutschland.
| |
Collapse
|
21
|
Vijapura C, Saad Aldin E, Capizzano AA, Policeni B, Sato Y, Moritani T. Genetic Syndromes Associated with Central Nervous System Tumors. Radiographics 2017; 37:258-280. [DOI: 10.1148/rg.2017160057] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
22
|
Krishnan A, Kaza RK, Vummidi DR. Cross-sectional Imaging Review of Tuberous Sclerosis. Radiol Clin North Am 2016; 54:423-40. [PMID: 27153781 DOI: 10.1016/j.rcl.2015.12.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Tuberous sclerosis complex (TSC) is a multisystem, genetic disorder characterized by development of hamartomas in the brain, abdomen, and thorax. It results from a mutation in one of 2 tumor suppressor genes that activates the mammalian target of rapamycin pathway. This article discusses the origins of the disorder, the recently updated criteria for the diagnosis of TSC, and the cross-sectional imaging findings and recommendations for surveillance. Familiarity with the diverse radiological features facilitates diagnosis and helps in treatment planning and monitoring response to treatment of this multisystem disorder.
Collapse
Affiliation(s)
- Anant Krishnan
- Department of Diagnostic Radiology, The Oakland University William Beaumont School of Medicine and Beaumont Hospital, 3601 West 13 Mile Road, Royal Oak, MI 48073, USA.
| | - Ravi K Kaza
- Department of Radiology, University of Michigan Hospitals, 1500 East Medical Center Drive, UH B1 502 E, Ann Arbor, MI 48109, USA
| | - Dharshan R Vummidi
- Department of Radiology, University of Michigan Hospitals, 1500 East Medical Center Drive, CVC5581, Ann Arbor, MI 48109, USA
| |
Collapse
|
23
|
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. [PMID: 26861567 PMCID: PMC4748639 DOI: 10.1186/s13256-016-0818-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 01/24/2016] [Indexed: 11/10/2022] Open
Abstract
Background Subependymal giant cell astrocytoma is a rare tumor that occurs in the wall of the lateral ventricle and foramen of Monro and, rarely, in the third ventricle. It is one of the intracranial lesions found in tuberous sclerosis complex (which include subependymal nodules, cortical tubers, retinal astrocytoma and subependymal giant cell astrocytoma), but cases without such lesions have also been reported in the literature. It was described for the first time in 1908 by Vogt as part of the typical triad of tuberous sclerosis complex. At the 2012 Washington Consensus Conference, it was decided by the invited expert panel to document the definition of subependymal giant cell astrocytoma as a lesion at the caudothalamic groove with either a size of more than 1 cm in any direction or a subependymal lesion at any location that has shown serial growth on consecutive imaging regardless of size. Most subependymal giant cell astrocytomas will show avid enhancement after contrast administration; however, a growing subependymal lesion even in the absence of enhancement should be considered a subependymal giant cell astrocytoma. Case presentation We report a case of subependymal giant cell astrocytoma in a 10-year-old white girl, who had no clinical symptoms of tuberous sclerosis. A computed tomography scan revealed a voluminous mass in her perilateral ventricle. An extemporaneous examination was in favor of a benign ganglioglioma tumor. After fixation in 10 % neutral-buffered formalin, embedding in paraffin and staining with hematoxylin, eosin and safran, the definitive diagnosis was subependymal giant cell astrocytoma. Conclusions Subependymal giant cell astrocytoma is a rare tumor of the central nervous system whose diagnosis is based on clinical, radiological, histological and immunohistochemical arguments. For its rarity, we must consider this diagnosis when faced with a mass near the foramen of Monro in the pediatric population even if there are no other features of tuberous sclerosis complex.
Collapse
Affiliation(s)
- Layla Tahiri Elousrouti
- Department of Pathology, Hassan II University Hospital, Route Sidi Harazem, 30000, Fes, Morocco.
| | - Meryem Lamchahab
- Department of Pathology, Hassan II University Hospital, Route Sidi Harazem, 30000, Fes, Morocco.
| | - Nawal Bougtoub
- Department of Radiology, Hassan II University Hospital, Route de Sidi, Harazem, 30000, Fes, Morocco.
| | - Hinde Elfatemi
- Department of Pathology, Hassan II University Hospital, Route Sidi Harazem, 30000, Fes, Morocco.
| | - Laila Chbani
- Department of Pathology, Hassan II University Hospital, Route Sidi Harazem, 30000, Fes, Morocco.
| | - Taoufik Harmouch
- Department of Pathology, Hassan II University Hospital, Route Sidi Harazem, 30000, Fes, Morocco.
| | - Mustapha Maaroufi
- Department of Radiology, Hassan II University Hospital, Route de Sidi, Harazem, 30000, Fes, Morocco.
| | - Afaf Amarti Riffi
- Department of Pathology, Hassan II University Hospital, Route Sidi Harazem, 30000, Fes, Morocco.
| |
Collapse
|
24
|
Abstract
Neurocutaneous syndromes (or phakomatoses) are a diverse group of congenital disorders that encompass abnormalities of neuroectodermal and, sometimes, mesodermal development, hence commonly involving the skin, eye, and central nervous system. These are often inherited conditions and typically present in early childhood or adolescence. Some of the abnormalities and clinical symptoms may, however, be progressive, and there is an increased risk of neoplastic formation in many of the syndromes. As a group, neurocutaneous syndromes are characterized by distinctive cutaneous stigmata and neurologic symptomology, the latter often representing the most devastating and debilitating features of these diseases. Many of these syndromes are markedly heterogeneous in nature as they affect many organ systems. Given the incurable nature of these conditions and the broad spectrum of pathologies they comprise, treatments vary on a case-by-case basis and tend to be palliative rather than curative. With the advances in molecular genetics, however, greater understanding of biologic functions of the gene products and the correlative phenotypic expression is being attained, and this knowledge may guide future therapeutic developments. This chapter focuses on the cutaneous and neurologic pathology with emphasis on neuroimaging of selective neurocutaneous syndromes, including tuberous sclerosis, Sturge-Weber syndrome, Klippel-Trenaunay syndrome, ataxia-telangiectasia, and incontinentia pigmenti.
Collapse
Affiliation(s)
- Nitasha Klar
- Division of Neuroradiology, Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bernard Cohen
- Departments of Dermatology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Doris D M Lin
- Division of Neuroradiology, Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
25
|
Natural History and Current Treatment Options for Subependymal Giant Cell Astrocytoma in Tuberous Sclerosis Complex. Semin Pediatr Neurol 2015; 22:274-81. [PMID: 26706014 DOI: 10.1016/j.spen.2015.10.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Subependymal giant cell astrocytoma (SEGA) is a benign brain tumor associated with tuberous sclerosis complex (TSC). Since there were some discrepancies in SEGA classification, in 2012, a consensus statement defined SEGA as a lesion at the caudothalamic groove with either a size of more than 1cm in any direction or a subependymal lesion at any location that has shown serial growth on consecutive imaging regardless of size. This pointed that the documented tumor growing is an important diagnostic feature. Currently, there are 2 therapeutic options: surgical and medical with mammalian target of rapamycin (mTOR) inhibitors. Neurosurgery should consider a localization and size of the tumor, presence of hydrocephalus and surgeon's experience. Patients who are not qualified for surgery and require therapeutic intervention are subjected to mTOR inhibitors therapy. Everolimus, one of the mTOR inhibitors, has been recently approved in the US and Europe for treatment of patients with TSC and SEGA. Treatment with everolimus results in a rapid initial reduction in tumor volume, followed by a phase of slower reduction or stabilization of residual mass. mTOR inhibitor dose de-escalation strategy seems to be also logical in long-term SEGA treatment to reduce the risk of adverse effects while maintaining the therapeutic effect. However, duration of the treatment and the efficacy of dose reduction are still questionable. Thus, this review covers the various approaches that are currently being made toward improving the outcome of management of SEGA among patients with TSC.
Collapse
|
26
|
Curatolo P, Moavero R, Roberto D, Graziola F. Genotype/Phenotype Correlations in Tuberous Sclerosis Complex. Semin Pediatr Neurol 2015; 22:259-73. [PMID: 26706013 DOI: 10.1016/j.spen.2015.10.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant disorder characterized by the development of widespread hamartomatous lesions in various organs, including brain, skin, kidneys, heart, and eyes. Central nervous system is almost invariably involved, with up to 85% of patients presenting with epilepsy, and at least half of patients having intellectual disability or other neuropsychiatric disorders including autism spectrum disorder. TSC is caused by the mutation in one of the 2 genes TSC1, at 9q34, and TSC2, at 16p13.3. They respectively encode for hamartin and tuberin, which form an intracellular complex inhibiting the mammalian target of rapamycin. Mammalian target of rapamycin overactivation following the genetic defect determines the cell growth and proliferation responsible for TSC-related lesions, as well as the alterations in neuronal excitability and synaptogenesis leading to epilepsy and neuropsychiatric disorders. A causative mutation for the disorder is identified in about 85% of patients with a clinical diagnosis of TSC. Mosaicism and technology limits likely explain most of the no mutation identified cases. This review confirms that patients with TSC2 mutations considered as a group usually present a more severe phenotype, characterized by higher number of tubers, earlier age at seizure onset and higher prevalence of intellectual disability. However, the clinical phenotype of the disease presents a high variability, thus making the prediction of the phenotype on an individual basis still challenging. The increasing application of new molecular techniques to subjects with TSC has the potential to significantly reduce the rate of patients with no mutation demonstrated and to identify an increasing higher number of mutations. This would hopefully allow a better characterization of higher risk mutations, which might help clinicians to plan individualized surveillance plans. Furthermore, the increasing availability of disease registries to collect clinical and genetics data of patients help to define more valid and clinically oriented genotype or phenotype correlations.
Collapse
Affiliation(s)
- Paolo Curatolo
- (⁎)Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University Hospital of Rome, Rome, Italy.
| | - Romina Moavero
- (⁎)Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University Hospital of Rome, Rome, Italy; Child Neurology Unit, Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Denis Roberto
- (⁎)Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University Hospital of Rome, Rome, Italy
| | - Federica Graziola
- (⁎)Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University Hospital of Rome, Rome, Italy
| |
Collapse
|
27
|
Curatolo P, Moavero R, de Vries PJ. Neurological and neuropsychiatric aspects of tuberous sclerosis complex. Lancet Neurol 2015; 14:733-45. [PMID: 26067126 DOI: 10.1016/s1474-4422(15)00069-1] [Citation(s) in RCA: 377] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 04/21/2015] [Accepted: 04/27/2015] [Indexed: 01/22/2023]
Abstract
Tuberous sclerosis (also known as tuberous sclerosis complex [TSC]) is a multisystem genetic disorder that affects almost every organ in the body. Mutations in the TSC1 or TSC2 genes lead to disruption of the TSC1-TSC2 intracellular protein complex, causing overactivation of the mammalian target of rapamycin (mTOR) protein complex. The surveillance and management guidelines and clinical criteria for tuberous sclerosis were revised in 2012, and mTOR inhibitors are now recommended as treatment options for subependymal giant cell astrocytomas and renal angiomyolipomas-two common features of the disease. However, most morbidity and mortality caused by tuberous sclerosis is associated with neurological and neuropsychiatric manifestations. Treatment of epilepsy associated with tuberous sclerosis remains a major challenge, with more than 60% of patients having ongoing seizures. Tuberous-sclerosis-associated neuropsychiatric disorders (TAND) are multilevel and occur in most individuals with the disorder, but are rarely assessed and treated. Clinical trials of mTOR inhibitors to treat seizures and TAND are underway. Management of the neurological and neuropsychiatric manifestations of the disorder should be coordinated with treatment of other organ systems. In view of the age-related expression of manifestations from infancy to adulthood, continuity of clinical care and ongoing monitoring is paramount, and particular attention is needed to plan transition of patient care from childhood to adult services.
Collapse
Affiliation(s)
- Paolo Curatolo
- Neuroscience Department, Child Neurology and Psychiatry Division, University Hospital of Tor Vergata, Rome, Italy.
| | - Romina Moavero
- Neuroscience Department, Child Neurology and Psychiatry Division, University Hospital of Tor Vergata, Rome, Italy; Neuroscience Department, Child Neurology Division, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Petrus J de Vries
- Division of Child and Adolescent Psychiatry, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
28
|
Ng KH, Ng SM, Parker A. Annual review of children with tuberous sclerosis. Arch Dis Child Educ Pract Ed 2015; 100:114-21. [PMID: 25112285 DOI: 10.1136/archdischild-2013-304948] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 07/21/2014] [Indexed: 12/17/2022]
Abstract
Tuberous sclerosis complex is a complex childhood disorder characterised by the formation of hamartomas in multiple organs. Annual review of this disease is recommended to monitor the development of complications. We aim to provide a concise, evidence-based framework to assist clinicians during this annual review. We focus on the following areas: (A) what questions need to be asked during annual review, (B) which areas need emphasis on examination, (C) when is an investigation required and (D) referral to tertiary specialists and other members of the multidisciplinary team. It should also be noted that there are ongoing debates regarding screening intervals in certain areas. These include the frequency, modality of screening and degree of intervention for astrocytomas and renal angiomyolipomas. This review seeks to summarise the product of the ongoing debates, and provide evidence-based suggestions in light of the uncertainty.
Collapse
Affiliation(s)
- K H Ng
- The North West London Hospitals NHS Trust, Northwick Park Hospital, London, UK School of Clinical Medicine, University Of Cambridge, Cambridge, UK
| | - S M Ng
- School of Clinical Medicine, University Of Cambridge, Cambridge, UK
| | - A Parker
- School of Clinical Medicine, University Of Cambridge, Cambridge, UK Department of Paediatric Neurology, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
29
|
Franz DN, Belousova E, Sparagana S, Bebin EM, Frost M, Kuperman R, Witt O, Kohrman MH, Flamini JR, Wu JY, Curatolo P, de Vries PJ, Berkowitz N, Anak O, Niolat J, Jozwiak S. 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. [DOI: 10.1016/s1470-2045(14)70489-9] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
30
|
Cardamone M, Flanagan D, Mowat D, Kennedy SE, Chopra M, Lawson JA. Mammalian target of rapamycin inhibitors for intractable epilepsy and subependymal giant cell astrocytomas in tuberous sclerosis complex. J Pediatr 2014; 164:1195-200. [PMID: 24518170 DOI: 10.1016/j.jpeds.2013.12.053] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 11/21/2013] [Accepted: 12/27/2013] [Indexed: 11/28/2022]
Abstract
OBJECTIVES To evaluate the efficacy and side effects of oral mammalian target of rapamycin (mTOR) inhibitors in children and adolescents with tuberous sclerosis complex (TSC) and intractable epilepsy or subependymal giant cell astrocytoma (SEGA). STUDY DESIGN Single-center series of 13 children and adolescents with TSC who received sirolimus or everolimus (mTOR inhibitors). The anticonvulsant response was evaluated in 7 patients with TSC and refractory seizures. Six patients with SEGAs were treated with either sirolimus or everolimus for nonsurgical management. SEGA volumes were assessed longitudinally using 1.5-T magnetic resonance imaging. RESULTS Of the intractable seizure group (7 patients), 1 patient had >90% reduction, 4 had 50%-90% reduction, and 2 had <50% reduction. Three reported subjective improvements in learning. By 12 months of treatment, there were statistically significant reductions in the SEGA volumes in 4 patients who received mTOR inhibitors (P < .04). The mean SEGA volume after 6 months of treatment was 2.18 cm(3), which represents 33% reduction in the mean baseline volume of 3.26 cm(3). The mTOR inhibitors were well tolerated. Adverse effects include dyslipidaemia (3 of 13), gingivitis (1 of 13), anorexia (1 of 13), and mild gastrointestinal side effects (1 of 13). CONCLUSION This case series suggests that mTOR inhibitors can improve seizures in those with TSC and refractory epilepsy. They are also an effective treatment for reducing the volume of SEGAs in patients with TSC not amenable to surgery with an acceptable side effect profile.
Collapse
Affiliation(s)
- Michael Cardamone
- Department of Neurology, Sydney Children's Hospital, Randwick, New South Wales, Australia; The School of Women's and Children's Health, Medicine UNSW, University of New South Wales, Sydney, New South Wales, Australia.
| | - Danny Flanagan
- Department of Neurology, Sydney Children's Hospital, Randwick, New South Wales, Australia; The School of Women's and Children's Health, Medicine UNSW, University of New South Wales, Sydney, New South Wales, Australia
| | - David Mowat
- Department of Medical Genetics, Sydney Children's Hospital, Randwick, New South Wales, Australia; The School of Women's and Children's Health, Medicine UNSW, University of New South Wales, Sydney, New South Wales, Australia
| | - Sean E Kennedy
- Department of Nephrology, Sydney Children's Hospital, Randwick, New South Wales, Australia; The School of Women's and Children's Health, Medicine UNSW, University of New South Wales, Sydney, New South Wales, Australia
| | - Maya Chopra
- Department of Medical Genetics, Sydney Children's Hospital, Randwick, New South Wales, Australia
| | - John A Lawson
- Department of Neurology, Sydney Children's Hospital, Randwick, New South Wales, Australia; The School of Women's and Children's Health, Medicine UNSW, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
31
|
Rovira À, Ruiz-Falcó ML, García-Esparza E, López-Laso E, Macaya A, Málaga I, Vázquez É, Vicente J. Recommendations for the radiological diagnosis and follow-up of neuropathological abnormalities associated with tuberous sclerosis complex. J Neurooncol 2014; 118:205-223. [PMID: 24771286 DOI: 10.1007/s11060-014-1429-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 03/28/2014] [Indexed: 01/25/2023]
Abstract
Tuberous sclerosis complex (TSC) is a genetic condition with multisystem involvement, characterized by the development of tumors and other abnormalities in organs such as the brain, retina, skin, heart, kidneys, and lungs. Most patients have neuropathological abnormalities such as cortical tubers, white matter radial migration lines, subependymal nodules, and subependymal giant cell astrocytomas (SEGAs). These lesions are associated with different neurological manifestations that are frequently associated with TSC. These manifestations consist of epilepsy, intellectual disability, and neurobehavioral and psychiatric problems, including autism spectrum disorder. Hydrocephalus may also develop in patients with SEGAs due to ventricular obstruction, when this usually slow-growing tumor reaches sufficient size. Surgery has been the classical approach to treat SEGAs, although this treatment is associated with substantial morbidity and does not completely prevent tumor recurrence. Recently, the mammalian target of rapamycin (mTOR) inhibitor, everolimus, has been approved by the Food and Drug Administration and the European Medicines Agency for the treatment of patients with SEGAs associated with TSC. However, the treatment of SEGAs with these agents requires the development of guidelines that establish a differential diagnosis between SENs and SEGAs, in which neuroradiological examinations play an essential role. With the aim of improving the neuroradiological diagnosis and follow-up of the neuropathological abnormalities associated with TSC, a group of experts in this field has reviewed different aspects related to these issues and put together, a series of statements and recommendations intended to provide guidance to specialists involved in the management of TSC.
Collapse
Affiliation(s)
- Àlex Rovira
- Neuroradiology Unit (IDI), Hospital Universitari Vall d'Hebron, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - María Luz Ruiz-Falcó
- Neuropediatrics Department, Hospital Infantil Universitario Niño Jesús, Avenida de Menéndez Pelayo, 65, 28009, Madrid, Spain.
| | - Elena García-Esparza
- Neuroradiology Department, Hospital Infantil Universitario Niño Jesús, Avenida de Menéndez Pelayo, 65, 28009, Madrid, Spain
| | - Eduardo López-Laso
- Neuropediatrics Department, Hospital Universitario Reina Sofía, Avenida Menéndez Pidal, s/n, 14004, Córdoba, Spain
| | - Alfons Macaya
- Neuropediatrics Department, Hospital Universitari Vall D'Hebron, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Ignacio Málaga
- Child Neurology Unit, Pediatrics Department, Hospital Universitario Central de Asturias, Calle de Celestino Villamil, s/n, 33006, Oviedo, Spain
| | - Élida Vázquez
- Pediatric Radiology Department, Hospital Universitari Vall d'Hebron, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Josefina Vicente
- Neuroradiology Department, Hospital Universitario Reina Sofía, Avenida Menéndez Pidal, s/n, 14004, Córdoba, Spain
| |
Collapse
|
32
|
Abstract
Tuberous sclerosis complex (TSC) is a genetic multisystem disorder characterized by widespread hamartomas in several organs, including the brain, heart, skin, eyes, kidney, lung, and liver. The affected genes are TSC1 and TSC2, encoding hamartin and tuberin respectively. The hamartin-tuberin complex inhibits the mammalian-target-of-Rapamycin (mTOR) pathway, which controls cell growth and proliferation. Variations in the distribution, number, size, and location of lesions cause the clinical syndrome to vary even between relatives. About 85% of children and adolescents with TSC have CNS complications, including epilepsy, cognitive impairment, challenging behavioral problems, and autism-like symptoms. Epilepsy generally begins during the first year of life, with focal seizures and spasms. The discovery of the mTOR pathway upregulation in TSC-associated lesions presents new possibilities for treatment strategy. Increasing understanding of the molecular abnormalities caused by TSC may enable improved management of the disease.
Collapse
Affiliation(s)
- P Curatolo
- Department of Neuroscience, Pediatric Neurology Unit, Tor Vergata University of Rome, Rome, Italy.
| | | |
Collapse
|
33
|
Kotulska K, Borkowska J, Roszkowski M, Mandera M, Daszkiewicz P, Drabik K, Jurkiewicz E, Larysz-Brysz M, Nowak K, Grajkowska W, Domańska-Pakieła D, Jóźwiak S. Surgical treatment of subependymal giant cell astrocytoma in tuberous sclerosis complex patients. Pediatr Neurol 2014; 50:307-12. [PMID: 24507694 DOI: 10.1016/j.pediatrneurol.2013.12.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 11/28/2013] [Accepted: 12/03/2013] [Indexed: 02/06/2023]
Abstract
BACKGROUND Subependymal giant cell astrocytoma is a brain tumor associated with tuberous sclerosis complex. There are two treatment options for subependymal giant cell astrocytomas: surgery or mammalian target of rapamycin inhibitor. The analysis of outcome of subependymal giant cell astrocytoma surgery may help characterize the patients who may benefit from pharmacotherapy. METHODS Sixty-four subependymal giant cell astrocytoma surgeries in 57 tuberous sclerosis complex patients with at least a 12-month follow-up were included in the study. The tumor size, age of the patients, mutation in the TSC1 or TSC2 gene, indication for the surgery, and postsurgical complications were analyzed. RESULTS The mean age of patients at surgery was 9.7 years. Mean follow-up after surgery was 63.7 months. Thirty-seven (57.8%) tumors were symptomatic and 27 (42.2%) were asymptomatic. Patients with TSC2 mutations developed subependymal giant cell astrocytoma at a significantly younger age than individuals with TSC1 mutations. Four patients (6.2% of all surgeries) died after surgery. Surgery-related complications were reported in 0%, 46%, 83%, 81%, and 67% of patients with tumors <2 cm, between 2 and 3 cm, between 3 and 4 cm, >4 cm, and bilateral subependymal giant cell astrocytomas, respectively, and were most common in children younger than 3 years of age. The most common complications included hemiparesis, hydrocephalus, hematoma, and cognitive decline. CONCLUSIONS Our study indicates that subependymal giant cell astrocytoma surgery is associated with significant risk in individuals with bilateral subependymal giant cell astrocytomas, tumors bigger than 2 cm, and in children younger than 3 years of age. Therefore, tuberous sclerosis complex patients should be thoroughly screened for subependymal giant cell astrocytoma growth, and early treatment should be considered in selected patients.
Collapse
Affiliation(s)
- Katarzyna Kotulska
- Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warsaw, Poland; Department of Science, The Children's Memorial Health Institute, Warsaw, Poland.
| | - Julita Borkowska
- Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warsaw, Poland
| | - Marcin Roszkowski
- Department of Neurosurgery, The Children's Memorial Health Institute, Warsaw, Poland
| | - Marek Mandera
- Department of Pediatric Neurosurgery, Silesian Medical University, Katowice, Poland
| | - Paweł Daszkiewicz
- Department of Neurosurgery, The Children's Memorial Health Institute, Warsaw, Poland
| | - Krzysztof Drabik
- Department of Neurosurgery, The Children's Memorial Health Institute, Warsaw, Poland
| | - Elzbieta Jurkiewicz
- Department of Radiology, The Children's Memorial Health Institute, Warsaw, Poland
| | | | - Katarzyna Nowak
- Department of Radiology, The Children's Memorial Health Institute, Warsaw, Poland
| | - Wiesława Grajkowska
- Department of Pathology, The Children's Memorial Health Institute, Warsaw, Poland
| | - Dorota Domańska-Pakieła
- Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warsaw, Poland
| | - Sergiusz Jóźwiak
- Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warsaw, Poland
| |
Collapse
|
34
|
Ouyang T, Zhang N, Benjamin T, Wang L, Jiao J, Zhao Y, Chen J. Subependymal giant cell astrocytoma: current concepts, management, and future directions. Childs Nerv Syst 2014; 30:561-70. [PMID: 24549759 DOI: 10.1007/s00381-014-2383-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 02/06/2014] [Indexed: 12/16/2022]
Abstract
BACKGROUND Subependymal giant cell astrocytoma (SEGA) is the most common central nervous system tumor in patients with tuberous sclerosis complex (TSC). SEGAs are generally benign, non-infiltrative lesions, but they can lead to intracranial hypertension, obstructive hydrocephalus, focal neurologic deficits, and even sudden death. DISCUSSION Surgical resection has been the standard treatment for SEGAs, and it is generally curative with complete resection. However, not all SEGAs are amenable to safe and complete resection. Gamma Knife stereotactic radiosurgery provides another treatment option as a primary or adjuvant treatment for SEGAs, but it has highly variable response effects with sporadic cases demonstrating its efficacy. Recently, biologically targeted pharmacotherapy with mammalian target of rapamycin (mTOR) inhibitors such as sirolimus and everolimus has provided a safe and efficacious treatment option for patients with SEGAs. However, SEGAs can recur few months after drug discontinuation, indicating that mTOR inhibitors may need to be continued to avoid recurrence. Further studies are needed to evaluate the advantages and adverse effects of long-term treatment with mTOR inhibitors. This review presents an overview of the current knowledge and particularly highlights the surgical and medical options of SEGAs in patients with TSC.
Collapse
Affiliation(s)
- Taohui Ouyang
- Department of Neurosurgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | | | | | | | | | | | | |
Collapse
|
35
|
Canpolat M, Per H, Gumus H, Yikilmaz A, Unal E, Patiroglu T, Cinar L, Kurtsoy A, Kumandas S. Rapamycin has a beneficial effect on controlling epilepsy in children with tuberous sclerosis complex: results of 7 children from a cohort of 86. Childs Nerv Syst 2014; 30:227-40. [PMID: 23743820 DOI: 10.1007/s00381-013-2185-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Accepted: 05/23/2013] [Indexed: 01/03/2023]
Abstract
PURPOSE Tuberous sclerosis complex (TSC) is a genetic disorder characterized by the formation of hamartomas in various organ systems. We would like share our experience from 86 patients and the results of rapamycin treatment in seven children with TSC. METHODS Eighty-six children with TSC were enrolled into this retrospective study. The clinical features of seven children treated with oral rapamycin were presented in detail. RESULTS The most common complaint of administration was convulsion in 77 children (89.5%). Hypopigmented skin lesions, adenoma sebaceum, resistant epilepsy, intracardiac mass, renal angiomyolipomas, and West syndrome were detected (n = 83, 96.5%; n = 47, 54.7%; n = 36, 41.9%; n = 27, 31.4%; n = 18, 20.9%; and n = 13, 15.1%, respectively). Subependymal nodules were the most frequent finding in cranial imaging followed by cortical tubers and subependymal giant cell astrocytomas (n = 75, 87.2%; n = 71, 82.6%; and n = 8, 9.3%, respectively). Of the seven patients treated with rapamycin, the lesions of six children with facial adenoma sebaceum showed regression in various degrees. The frequency of convulsions decreased in five patients with resistant epilepsy within the first 6 months of the treatment, and complete control of convulsion for all patients was achieved in the second 6 months. CONCLUSION This is the first study that showed that rapamycin is an effective agent for controlling epilepsy without any significant side effect in children with TSC. Rapamycin seems to be effective after 6 months of therapy, and we recommend tapering the dosage after successful management of epilepsy.
Collapse
Affiliation(s)
- Mehmet Canpolat
- Department of Pediatrics, Division of Pediatric Neurology, Faculty of Medicine, Erciyes University, 38039, Kayseri, Turkey
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
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. Childs Nerv Syst 2014; 30:2037-42. [PMID: 25227171 PMCID: PMC4223570 DOI: 10.1007/s00381-014-2555-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 09/03/2014] [Indexed: 12/23/2022]
Abstract
PURPOSE Subependymal giant cell astrocytoma (SEGA) is a brain tumor associated with tuberous sclerosis complex (TSC). It usually grows in a second decade of life, but may develop in the first months of life. The aim of this work was to establish the incidence, clinical features, and outcome of congenital SEGA in TSC patients. METHODS Cohort of 452 TSC patients was reviewed to identify cases with growing or hydrocephalus producing SEGAs in the first 3 months of life. Clinical presentation, size of the tumor, growth rate, mutational analysis, treatment applied, and outcome were analyzed. RESULTS Ten (2.2 %) patients presented with SEGA in the first 3 months of life. All of them had documented SEGA growth and all developed hydrocephalus. In eight patients, mutational analysis was done, and in all of them, TSC2 gene mutations were identified. Mean maximum SEGA diameter at baseline was 21.8 mm. Mean SEGA growth rate observed postnatally was 2.78 mm per month and tended to be higher (5.43 mm per month) in patients with TSC2/PKD1 mutation than in other cases. Seven patients underwent SEGA surgery and surgery-related complications were observed in 57.1 % cases. One patient was successfully treated with everolimus as a primary treatment. CONCLUSIONS Congenital SEGA develops 2.2 % of TSC patients. Patients with TSC2 mutations, and especially with TSC2/PKD1 mutations, are more prone to develop SEGA earlier in childhood and should be screened for SEGA from birth. In young infants with SEGA, both surgery and mTOR inhibitor should be considered as a treatment option.
Collapse
Affiliation(s)
- Katarzyna Kotulska
- Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warsaw, Poland,
| | - Julita Borkowska
- Department of Neurology and Epileptology, The Children’s Memorial Health Institute, Warsaw, Poland
| | - Marek Mandera
- Department of Paediatric Neurosurgery, Silesian Medical University, Katowice, Poland
| | - Marcin Roszkowski
- Department of Neurosurgery, The Children’s Memorial Health Institute, Warsaw, Poland
| | - Elzbieta Jurkiewicz
- Department of Radiology, The Children’s Memorial Health Institute, Warsaw, Poland
| | - Wiesława Grajkowska
- Department of Pathology, The Children’s Memorial Health Institute, Warsaw, Poland
| | - Małgorzata Bilska
- Department of Neurology and Epileptology, The Children’s Memorial Health Institute, Warsaw, Poland
| | - Sergiusz Jóźwiak
- Department of Neurology and Epileptology, The Children’s Memorial Health Institute, Warsaw, Poland
| |
Collapse
|
37
|
Harter DH, Bassani L, Rodgers SD, Roth J, Devinsky O, Carlson C, Wisoff JH, Weiner HL. A management strategy for intraventricular subependymal giant cell astrocytomas in tuberous sclerosis complex. J Neurosurg Pediatr 2014; 13:21-8. [PMID: 24180681 DOI: 10.3171/2013.9.peds13193] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Subependymal giant cell astrocytomas (SEGAs) are benign tumors, most commonly associated with tuberous sclerosis complex (TSC). The vast majority of these tumors arise from the lateral ependymal surface adjacent to the foramen of Monro, therefore potentially encroaching on one or both foramina, and resulting in obstructive hydrocephalus that necessitates surgical decompression. The indications for surgery, intraoperative considerations, and evolution of the authors' management paradigm are presented. METHODS Patients with TSC who underwent craniotomy for SEGA resection at New York University Langone Medical Center between January 1997 and March 2011 were identified. Preoperative imaging, clinical characteristics, management decisions, operative procedures, and outcomes were reviewed. RESULTS Eighteen patients with TSC underwent 22 primary tumor resections for SEGAs. The indication for surgery was meaningful radiographic tumor progression in 16 of 21 cases. The average age at the time of operation was 10.3 years. Average follow-up duration was 52 months (range 12-124 months). The operative approach was intrahemispheric-transcallosal in 16 cases, transcortical-transventricular in 5, and neuroendoscopic in 1. Nine tumors were on the right, 9 on the left, and 3 were bilateral. Gross-total resection was documented in 16 of 22 cases in our series, with radical subtotal resection achieved in 4 cases, and subtotal resection (STR) in 2 cases. Two patients had undergone ventriculoperitoneal shunt placement preoperatively and 7 patients required shunt placement after surgery for moderate to severe ventriculomegaly. Two patients experienced tumor progression requiring reoperation; both of these patients had initially undergone STR. CONCLUSIONS The authors present their management strategy for TSC patients with SEGAs. Select patients underwent microsurgical resection of SEGAs with acceptable morbidity. Gross-total resection or radical STR was achieved in 90.9% of our series (20 of 22 primary tumor resections), with no recurrences in this group. Approximately half of our patient series required CSF diversionary procedures. There were no instances of permanent neurological morbidity associated with surgery.
Collapse
|
38
|
Peters JM, Taquet M, Prohl AK, Scherrer B, van Eeghen AM, Prabhu SP, Sahin M, Warfield SK. Diffusion tensor imaging and related techniques in tuberous sclerosis complex: review and future directions. FUTURE NEUROLOGY 2013; 8:583-597. [PMID: 24489482 DOI: 10.2217/fnl.13.37] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In this article, the authors aim to introduce the nonradiologist to diffusion tensor imaging (DTI) and its applications to both clinical and research aspects of tuberous sclerosis complex. Tuberous sclerosis complex is a genetic neurocutaneous syndrome with variable and unpredictable neurological comorbidity that includes refractory epilepsy, intellectual disability, behavioral abnormalities and autism spectrum disorder. DTI is a method for modeling water diffusion in tissue and can noninvasively characterize microstructural properties of the brain. In tuberous sclerosis complex, DTI measures reflect well-known pathological changes. Clinically, DTI can assist with detecting the epileptogenic tuber. For research, DTI has a putative role in identifying potential disease biomarkers, as DTI abnormalities of the white matter are associated with neurocognitive morbidity including autism. If indeed DTI changes parallel phenotypical changes related to the investigational treatment of epilepsy, cognition and behavior with mTOR inhibitors, it will facilitate future clinical trials.
Collapse
Affiliation(s)
- Jurriaan M Peters
- Department of Neurology & the Division of Epilepsy & Clinical Neurophysiology, Boston Children's Hospital, 300 Longwood Avenue, Fegan 9, Boston, MA 02115, USA ; Department of Radiology & the Computational Radiology Laboratory, Boston Children's Hospital, Boston, MA 02115, USA
| | - Maxime Taquet
- Department of Radiology & the Computational Radiology Laboratory, Boston Children's Hospital, Boston, MA 02115, USA ; ICTEAM Institute, Université catholique de Louvain, Place du Levant 2 bte L5.04.04, 1348 Louvain-La-Neuve, Belgium
| | - Anna K Prohl
- Department of Neurology & the Division of Epilepsy & Clinical Neurophysiology, Boston Children's Hospital, 300 Longwood Avenue, Fegan 9, Boston, MA 02115, USA ; Department of Radiology & the Computational Radiology Laboratory, Boston Children's Hospital, Boston, MA 02115, USA
| | - Benoit Scherrer
- Department of Radiology & the Computational Radiology Laboratory, Boston Children's Hospital, Boston, MA 02115, USA
| | - Agnies M van Eeghen
- Department of Neuroscience, ENCORE, Expertise Centre for Neurodevelopmental Disorders, Erasmus Medical Centre, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Sanjay P Prabhu
- Department of Radiology & the Computational Radiology Laboratory, Boston Children's Hospital, Boston, MA 02115, USA
| | - Mustafa Sahin
- Department of Neurology & the Division of Epilepsy & Clinical Neurophysiology, Boston Children's Hospital, 300 Longwood Avenue, Fegan 9, Boston, MA 02115, USA
| | - Simon K Warfield
- Department of Radiology & the Computational Radiology Laboratory, Boston Children's Hospital, Boston, MA 02115, USA
| |
Collapse
|
39
|
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; 17:348-52. [PMID: 23391693 DOI: 10.1016/j.ejpn.2012.12.008] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 12/13/2012] [Accepted: 12/17/2012] [Indexed: 10/27/2022]
Abstract
Subependymal giant cell astrocytoma (SEGA) is a type of brain tumour that develops in 10-15% of individuals with tuberous sclerosis complex (TSC). SEGAs can be unilateral or bilateral, developing from benign subependymal nodules (hamartomas) located near the foramen of Monro. These are usually slow-growing, glialneuronal tumours that develop within the first 2 decades of life. Traditionally, the management of SEGA involved monitoring using periodic neuroimaging, and surgical resection of tumours that exhibited growth and/or caused clinical signs of intracranial hypertension. Recent clinical research has demonstrated that mammalian target of rapamycin (mTOR) inhibitors can induce partial regression of SEGA associated with TSC and so might provide an acceptable alternative to neurosurgery for these tumours. This report summarizes the clinical recommendations for the management of SEGA made by a panel of European experts in March 2012. Current treatment options and outstanding questions are outlined.
Collapse
Affiliation(s)
- Sergiusz Jóźwiak
- Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warsaw, Poland.
| | | | | | | |
Collapse
|
40
|
Okura H, Kobayashi T, Koike M, Ohsawa M, Zhang D, Arai H, Uchiyama Y, Hino O. Tuberin activates and controls the distribution of Rac1 via association with p62 and ubiquitin through the mTORC1 signaling pathway. Int J Oncol 2013; 43:447-56. [PMID: 23759924 DOI: 10.3892/ijo.2013.1984] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 05/17/2013] [Indexed: 11/06/2022] Open
Abstract
Recent studies indicated that the tuberous sclerosis 2 (TSC2) gene product, tuberin, regulates Rac1 activity. However, the underlying mechanism by which tuberin regulates Rac1 activity has not been clearly elucidated to date. To better understand the molecular link between tuberin function and Rac1, we characterized the activity and distribution of Rac1 in mouse Tsc2-deficient renal tumor cells using restoration experiments with wild-type tuberin. Rac1 activity was significantly higher in tuberin-expressing cells compared with control Tsc2-deficient cells. Further, Rac1 activation was induced by rapamycin treatment or knockdown of raptor, but not rictor, in Tsc2-deficient cells, indicating that mTORC1 is an upstream negative regulator of Rac1. Intriguingly, Rac1 appeared to form cytoplasmic dots in Tsc2-deficient cells, but not in tuberin-expressing and since rapamycin treatment dispersed these dots, involvement of aberrant mTOR complex 1 (mTORC1) activation in the dot formation was suspected. Moreover, the dots were co-localized with p62/sequestosome-1 and ubiquitin. These findings imply that Rac1 distribution and/or its degradation may be regulated by tuberin through the mTORC1 signaling pathway.
Collapse
Affiliation(s)
- Hidehiro Okura
- Department of Pathology and Oncology, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
The authors report on 2 cases of subependymal giant cell astrocytoma (SEGA) with intratumoral hemorrhage causing acute hydrocephalus, necessitating emergent resection of the tumor. They review the literature and present their insights on the management of SEGA showing growth on serial imaging. Intratumoral hemorrhage causing acute hydrocephalus can occur not only in the pediatric ages but also in the early 20s in patients with SEGA. Awareness of this sequela is considered to be important in addressing surgical timing. The authors suggest early resection of the lesions when the evidence of growth has been confirmed, to prevent possible morbidity and mortality.
Collapse
Affiliation(s)
- Hideki Ogiwara
- Division of Neurosurgery, National Center for Child Health and Development, Tokyo, Japan.
| | | |
Collapse
|
42
|
Michelozzi C, Di Leo G, Galli F, Silva Barbosa F, Labriola F, Sardanelli F, Cornalba G. Subependymal nodules and giant cell tumours in tuberous sclerosis complex patients: prevalence on MRI in relation to gene mutation. Childs Nerv Syst 2013; 29:249-54. [PMID: 22936080 DOI: 10.1007/s00381-012-1892-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 08/07/2012] [Indexed: 11/26/2022]
Abstract
PURPOSE The purpose of this study was to estimate the association among the presence of subependymal nodules (SENs), subependymal giant cell tumours (SGCTs) and gene mutation in tuberous sclerosis complex (TSC) patients. METHODS Clinical records and images of 81 TSC patients were retrospectively reviewed by two neuroradiologists in consensus. All patients were assessed for gene mutations and were categorized as TSC1 or TSC2 mutation carriers, or no-mutations-identified (NMI) patients. They underwent a brain magnetic resonance imaging (MRI) using 0.1 mmol/kg of gadobutrol. Any enhancing SEN ≥ 1 cm and placed near the foramen of Monro was considered SGCT. Two MRI follow-up exams for each patient with SGCT were evaluated to assess tumour growth using Wilcoxon and chi-squared tests. RESULTS Of 81 patients, 44 (54%) were TSC2 mutation carriers, 20 (25%) TSC1 and 17 (21%) NMI. Nine (11%) had a unilateral and three (4%) a bilateral SGCT. Fifty of 81 patients (62%) showed at least one SEN. None of the 31 patients without SEN showed SGCTs, whilst 12 (24%) of the 50 patients with at least one SEN showed SGCTs (p = 0.003). The association between the presence of SGCT or SEN and gene mutation was not significant (p = 0.251 and p = 0.187, respectively). At follow-up, the median SGCT diameter increased from 14 to 15 mm (p = 0.017), whilst the median SGCT volume increased from 589 to 791 mm(3) (p = 0.006). CONCLUSIONS TSC patients with SENs are more likely to present with SGCT than those without SENs, in particular for TSC2 mutation carriers. The SGCT growth rate may be missed if based on the diameter instead of on the volume.
Collapse
Affiliation(s)
- Caterina Michelozzi
- Scuola di Specializzazione in Radiodiagnostica, Università degli Studi di Milano, Via Festa del Perdono 7, 20122 Milan, Italy.
| | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
OBJECTIVES The indications for surgery and outcomes of patients who underwent surgical removal of subependymal giant cell astrocytomas (SEGAs) in our institution between 2000 and 2011 were reviewed. METHODS We reviewed the clinical details of 16 patients with a diagnosis of Tuberous Sclerosis Complex (TSC) who underwent surgery for SEGA in Bristol since 2000. We collected information on age, sex, epilepsy history and cognitive status. We reviewed the indications for surgery, age at surgery, surgical approach, and the size and location of the lesions. We analysed mortality, completeness of tumour resection, intraoperative blood transfusion, shunt placements, and surgical complications. RESULTS 13 patients had surgery due to hydrocephalus. Increasing size of SEGA without hydrocephalus was an indication for surgery in two patients, and in one patient, the SEGA was removed because of its size and location at initial scan. 13 patients had complete tumour resection. One patient had tumour recurrence. Hydrocephalus failed to resolve or reoccurred in four patients post operatively necessitating shunt insertion. The surgical approach was transcortical in 14 patients and transcallosal in two. There was zero mortality in this series. There were no reports of cognitive decline or worsening epilepsy following surgery. CONCLUSION Surgery is a safe and effective treatment for SEGA. It is the authors' view that surgery remains the most appropriate treatment strategy for SEGAs that are amenable to surgery. More work needs to be undertaken to assess prospectively the neurocognitive impact of surgery, and the relative advantages of different surgical approaches.
Collapse
|
44
|
[Neuro-encephalic features of tuberous sclerosis complex]. Rev Med Interne 2012; 33:433-8. [PMID: 22658530 DOI: 10.1016/j.revmed.2012.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 01/04/2012] [Accepted: 04/01/2012] [Indexed: 11/22/2022]
Abstract
Tuberous sclerosis is a phacomatosis resulting from an autosomal dominant inheritance. It is characterized by the presence of multiple hamartomas in various organs, especially the brain, the skin, the kidneys and the heart. The diagnosis of tuberous sclerosis is based on imaging and clinical examination, where magnetic resonance imaging constitutes the key investigation showing characteristic brain lesions. Neuro-encephalic manifestations may be particularly severe, and may even be life threatening. The authors report personal cases series and review the literature highlighting epidemiology, clinical features and imaging of neuro-encephalic tuberous sclerosis.
Collapse
|
45
|
Katz JS, Milla SS, Wiggins GC, Devinsky O, Weiner HL, Roth J. Intraventricular lesions in tuberous sclerosis complex: a possible association with the caudate nucleus. J Neurosurg Pediatr 2012; 9:406-13. [PMID: 22462706 DOI: 10.3171/2011.12.peds11418] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Tuberous sclerosis complex (TSC) can manifest with 3 principal intracranial pathological entities: cortical tubers, subependymal nodules (SENs), and subependymal giant cell astrocytomas (SEGAs). The authors analyzed the location and growth of intraventricular lesions in a large cohort of patients with TSC. METHODS After institutional review board protocol approval, the authors retrospectively reviewed brain MRI scans of TSC patients for whom at least 1 electronically stored cranial MRI study was available. Collected data included location, size, and growth over time of all intraventricular lesions. RESULTS The authors reviewed 560 scans in 103 patients, who harbored 496 intraventricular lesions. Of the 496 lesions, 157 lesions were located along the caudate-thalamic groove (CTG) in 88 patients. Twenty SEGAs were operated on. The remaining 339 lesions were distributed along the lateral ventricle, always in contact with the course of the caudate nucleus, and were presumed to be SENs. Twenty-two patients with more than 4 years of follow-up had 34 lesions along the CTG, of which 23 were stable in size and 11 grew. All other intraventricular lesions were stable. Seven-Tesla MRI showed the intimate association of SENs and the caudate nucleus in 1 patient. CONCLUSIONS Intraventricular lesions in TSC patients are located throughout the lateral ventricular wall. Their location exclusively follows the course of the caudate nucleus. Only lesions along the CTG showed the potential to grow, and these were then identified as SEGAs. The remaining lesions were SENs. Understanding why these lesions develop in relation to the caudate nucleus may offer insights into therapy.
Collapse
Affiliation(s)
- Joel S Katz
- Department of Neurosurgery, Division of Pediatric Neurosurgery, New York, NY, USA
| | | | | | | | | | | |
Collapse
|
46
|
Kohrman MH. Emerging treatments in the management of tuberous sclerosis complex. Pediatr Neurol 2012; 46:267-75. [PMID: 22520346 DOI: 10.1016/j.pediatrneurol.2012.02.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 02/14/2012] [Indexed: 12/31/2022]
Abstract
Tuberous sclerosis complex is a genetic disorder characterized by the formation of nonmalignant hamartomas in the brain, heart, skin, kidney, lung, and other organs. It is associated with autism, epilepsy, and other neurocognitive and behavioral disabilities. Wide phenotypic variation occurs in disease severity and natural course: some patients demonstrate minimal effects, e.g., skin changes; others manifest profound seizures and mental retardation. Tuberous sclerosis complex is caused by mutations in either the tuberous sclerosis complex 1 or 2 gene (coding for hamartin and tuberin, respectively). The tuberous sclerosis complex 1/tuberous sclerosis complex 2 protein dimer complex is a crucial inhibitory element in the mammalian target of rapamycin pathway, regulating cell growth and proliferation. Until recently, few options existed, other than surgery, for treating symptoms of tuberous sclerosis complex related to the growth of hamartomas. Increased understanding of the genetic cause of the disease and underlying dysregulation of the mammalian target of rapamycin pathway has led to clinical trials of mammalian target of rapamycin inhibitors, including sirolimus and everolimus. This review gives an overview of tuberous sclerosis complex and its molecular causes, and summarizes results from recent clinical trials of mammalian target of rapamycin inhibitors in patients with the disease.
Collapse
Affiliation(s)
- Michael H Kohrman
- Department of Pediatrics, Comer Children's Hospital, University of Chicago, Chicago, Illinois 60637, USA.
| |
Collapse
|
47
|
Vinchon M, Baroncini M, Leblond P. [Tumors of the lateral ventricle in child: characteristics and specificities]. Neurochirurgie 2011; 57:230-6. [PMID: 22024608 DOI: 10.1016/j.neuchi.2011.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Accepted: 09/12/2011] [Indexed: 11/19/2022]
Abstract
Tumors of the lateral ventricle are not as rare in children as in adults and present complex problems which are to be dealt with often in a context of emergency, on account of their large size and the risk of life-threatening bleeding in a small child. Their diagnostic features are different in children, especially in infants and even more so in the fetus. The main surgical problems are the risk of bleeding and of hydraulic complications. From an oncological perspective, the main tumor types are choroid plexus tumors, subependymal giant-cell astrocytoma, and low-grade gliomas. The developmental outcome of these patients is often poor, on account of severe intracranial hypertension and young age at diagnosis.
Collapse
Affiliation(s)
- M Vinchon
- Neurochirurgie pédiatrique, hôpital Roger-Salengro, CHRU de Lille, rue Émile-Laine, 59037 Lille cedex, France.
| | | | | |
Collapse
|
48
|
Pascual-Castroviejo I. Neurosurgical treatment of tuberous sclerosis complex lesions. Childs Nerv Syst 2011; 27:1211-9. [PMID: 21607641 DOI: 10.1007/s00381-011-1488-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 05/09/2011] [Indexed: 11/27/2022]
Abstract
BACKGROUND Tuberous sclerosis complex (TSC) is an autosomal dominantly inherited syndrome. Renal disease is the main cause of death. Brain disorders are the origin of more frequent and severe problems, such as tumors, epilepsy, and mental retardation. Participation of neurosurgeons in the study and especially in the treatment of TSC patients is often required. MATERIALS AND METHODS Two types of pathological conditions mainly require neurosurgical interventions in TSC: subependymal giant cell astrocytomas (SGCA) and cortical tubers. SGCA are located in the cerebral region close to the foramina of Monroe, uni- or bilaterally, and originate in hamartomas that can grow slowly as well as rapidly, even suddenly, especially in cases with intratumoral cyst, causing increased intracranial pressure (ICP) with severe risk for visual loss and life. Neurosurgeons have to participate in the follow-up of the patients as soon as the risk of ICP exists to remove the tumor when the criteria of SGCA growth are present. The other intracranial lesions that require neurosurgical intervention by are the cortical tubers. CONCLUSION These dysplastic lesions are associated with TSC in almost the 100% of affected persons and are the cause of epilepsy in most patients. The seizures can be resistant to antiepileptic medication in many cases in which a tuber is identified as the origin of the focal seizures after functional studies, such as EEG, MR, PET, etc. In these cases, only surgical removal of the tuber and the perituberal epileptogenic foci can cure the epilepsy. Large tubers are more epileptogenic than smaller ones.
Collapse
|
49
|
Turner SG, Peters KB, Vredenburgh JJ, Desjardins A, Friedman HS, Reardon DA. Everolimus tablets for patients with subependymal giant cell astrocytoma. Expert Opin Pharmacother 2011; 12:2265-9. [PMID: 21806479 DOI: 10.1517/14656566.2011.601742] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Better understanding of aberrantly active molecular pathways in tumors offers potential to develop more specific and less toxic therapies. Abnormal mammalian target of rapamycin (mTOR) complex signaling and defects in TSC1 and TSC2 have been associated with the development of subependymal giant cell astrocytomas (SEGAs) in tuberous sclerosis complex (TSC) patients. Recently, mTOR inhibitors such as everolimus have shown encouraging benefit for patients with SEGAs. AREAS COVERED The authors discuss a molecular genetic pathway linked with TSC, specifically the role of two proteins whose functional absence is responsible for most SEGA tumors that arise in TSC patients. The authors also examine the rationale for targeted agents against this pathway therapeutically and describe the clinical evidence underlying the FDA approval of everolimus for patients with inoperable SEGAs. EXPERT OPINION Everolimus (Afinitor) selectively targets a molecular defect of SEGAs in TSC patients. Although surgery is effective, most SEGAs recur. An agent that inhibits an underlying molecular abnormality represents a particularly attractive therapeutic option for patients with inoperable or recurrent tumors. Studies are also underway to assess everolimus in treating other sequelae of TSC, and other gliomas. Finally, additional research aimed at better understanding aberrant cell signaling pathways may lead to the development of more effective therapeutics.
Collapse
Affiliation(s)
- Scott G Turner
- Duke University Medical Center, The Preston Robert Tisch Brain Tumor Center, Department of Surgery, Box 3624, Durham, NC 27710, USA
| | | | | | | | | | | |
Collapse
|
50
|
The management of subependymal giant cell tumors in tuberous sclerosis: a clinician's perspective. Childs Nerv Syst 2011; 27:1203-10. [PMID: 21305305 DOI: 10.1007/s00381-011-1406-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Accepted: 01/26/2011] [Indexed: 10/18/2022]
Abstract
BACKGROUND Tuberous sclerosis (TSC) is a genetic multisystem disorder associated with hamartomas in several organs including subependymal giant cell tumors (SGCT). SGCT have the potential to grow and therefore to become symptomatic and are one of the main causes of death in TSC individuals. Surgical resection is the procedure of choice for SGCT. However, the discovery of mTOR pathway upregulation in TSC-associated tumors and recent evidence that mTOR inhibitors may induce regression of SGCT open up new treatment strategies. Based on a review of the currently available literature and on personal experience, current options for the management of TSC patients and appropriate indications, taking into account benefits and risks of surgery and pharmacotherapy, are discussed. DISCUSSION An earlier diagnosis of SGCT in neurologically asymptomatic children may allow a precocious surgical removal of the tumor, thus minimizing surgery-related morbidity and mortality. Biologically targeted pharmacotherapy with mTOR inhibitors such as sirolimus and everolimus provides a safe and efficacious treatment option for patients with SGCT and has the potential to change the clinical management of these tumors. However, whether pharmacotherapy is sufficient to control growth or if it only delays the need for surgical removal of symptomatic SGCT remains unclear. Further studies are needed to determine the optimal levels of mTOR inhibitors that preserve maximal anti-tumor efficacy while minimizing side effects.
Collapse
|