• Resumen
  • Sinónimos
  • Signos y Síntomas
  • Causas y Herencia
  • Frecuencia
  • Diagnóstico
  • Tratamiento
  • Investigaciones
  • Referencias
  • Programas & Recursos
  • Informe completo

Glioma

Imprimir

Última actualización: 8/16/2023
Años publicados: 2019, 2023


Reconocimiento

NORD gratefully acknowledges Etienne Leveille, MD, Yale School of Medicine and Jennie Taylor, MD, MPH, Associate Professor, Neurology and Neurological Surgery, University of California, San Francisco, for assistance in the preparation of this report.


Resumen

Summary

A glioma is a tumor of the central nervous system that arises from glial stem or progenitor cells. Glial cells are a type of cell widely present in the nervous system. Gliomas mostly occur in the brain and, rarely, in the spinal cord. They occur at various ages depending on the subtype. Gliomas can compress areas of the brain where they occur and cause various symptoms including headaches, nausea, vomiting, cognitive impairment, seizures, gait imbalance, language impairment (aphasia), numbness or weakness of one side of the body (hemiparesis), visual changes and personality changes. The treatment of gliomas often requires a combination of neurosurgical interventions, radiotherapy and chemotherapy.

Introduction


Classification of gliomas is based on the microscopic appearance of the tumor and the molecular characteristics such as gene changes (mutations) in the tumor. Tumors are often graded 1-4 based on severity, with 4 being the most aggressive. Information about specific tumor types is available from the National Brain Tumor Society.

  • Sección siguiente >
  • < Sección anterior
  • Sección siguiente >

Sinónimos

  • glial tumor
  • glial cell tumor
  • glial neoplasm
  • < Sección anterior
  • Sección siguiente >
  • < Sección anterior
  • Sección siguiente >

Signos y Síntomas

The symptoms associated with gliomas are similar among all types but can vary depending on the individual and the location of the tumor. Seizures (focal or generalized), language impairment (aphasia), weakness of part of the body (hemiparesis), sensory changes on part of the body and headaches are common. Other possible symptoms include gait disturbances, fatigue, dizziness, visual changes, vomiting and changes in urination. Psychological symptoms such as cognitive impairment, personality changes, depression, anxiety and memory impairment can also occur. Most of the symptoms are a consequence of the compressive effect of the tumor and fluid that surrounds it on the brain. Malignant gliomas (grade 3 and 4) are also associated with the development of blood clots in the deep veins, notably of the legs, (deep vein thrombosis) that can dislodge and migrate to occlude the arteries of the lungs (pulmonary embolism).

Gliomas can develop at any age. The average age at which they occur greatly varies depending on the subtype of glioma. Similarly, the survival rate greatly depends on the subtype of glioma.

In addition to being used for diagnosis and classification, the gene mutations present in tumor cells are also used to predict disease course and survival (prognosis).

Over time, gliomas can increase in grade and, therefore, become more malignant (malignant progression). The rate of malignant progression depends on the subtype of glioma and on the genetic characteristics of affected cells. Higher grade tumors are typically associated with lower survival rates.

Cells in gliomas have an altered glucose metabolism and can develop their own blood vessel network which allows them to sustain the high energy requirements for cell division and growth. Inflammation and accumulation of fluid around the tumor are also features of gliomas. Over time, certain types of gliomas can grow and invade healthy brain tissue extensively.

  • < Sección anterior
  • Sección siguiente >
  • < Sección anterior
  • Sección siguiente >

Causas y Herencia

Gliomas are caused by the accumulation of genetic mutations in glial stem or progenitor cells, leading to their uncontrolled growth. Mutated genes are typically involved in functions such as tumor suppression, DNA repair and regulation of cell growth. Examples of mutated genes in certain types of gliomas include TP53, PTEN (tumor suppressor genes), ATRX (involved in the remodeling of chromatin, a DNA-RNA-protein complex), TERT (encoding a subunit of telomerase, an enzyme that can lead to an infinite division potential in cells) BRAF (involved in cell growth) and IDH (involved in cellular metabolism).

The exact underlying cause of glioma development in most individuals is unknown. The only established environmental risk factor associated with gliomas is exposure to ionizing radiation, such as in atomic bomb survivors. Malignant gliomas can arise on their own (de novo) or can result from further accumulation of genetic mutations in low-grade gliomas (malignant progression). Cells from malignant gliomas have typically lost their specialized structure and function (de-differentiation or anaplasia). Initially, all cells in a glioma contain the same genetic code and are identical. Over time, different mutations accumulate in different cells of the tumor, thus leading to different subclones and a genetically heterogeneous tumor. Changes not affecting the genetic code directly, but rather how it is read and expressed (epigenetic modifications) are also involved in the growth and development of gliomas.

  • < Sección anterior
  • Sección siguiente >
  • < Sección anterior
  • Sección siguiente >

Frecuencia

Excluding metastases from other cancers that reach the central nervous system, gliomas make up 26% of all brain tumors, (primary brain tumors) and 81% of all malignant brain tumors. They develop in approximately 6.6 per 100,000 individuals each year and 2.94 per 100,000 individuals under age 14. The median age (meaning that half of affected individuals are younger than this age and the other half are older) for the development of glioma is between 12 and 65 years, depending on the subtype. Pilocytic astrocytoma is the most common glioma in individuals under age 14 (34.4% of all gliomas) whereas glioblastoma is the most common glioma in adults (56.6% of all gliomas) (for more information on these types of gliomas, choose the specific glioma name as your search term in the Rare Disease Database).

Gliomas are slightly more common in males. They tend to affect older individuals and are more common in countries with a higher level of development, as these countries generally have a larger proportion of older individuals. There are also several syndromes associated with a higher risk of glioma.

  • < Sección anterior
  • Sección siguiente >
  • < Sección anterior
  • Sección siguiente >

Diagnóstico

A diagnosis of glioma requires an extensive patient history as well as a complete physical and neurological examination. Signs that further investigation might be required include a new onset of seizures, abrupt onset of cognitive decline and presence of other neurological symptoms. Headaches that develop or worsen abruptly, that begin to occur after age 50, that awaken the affected person from sleep even when they are mild and that are associated with cognitive impairment are warning signs that could indicate a brain mass.

The presence of a brain tumor can be suspected with medical imaging. Magnetic resonance imaging (MRI) is the imaging modality of choice for initial evaluation of glioma. The final diagnosis, and therefore plan for treatment, can only be determined after a piece of tumor tissue is analyzed microscopically. Further characterization can be done by testing the DNA of the affected cells to determine if mutations in genes associated with certain subtypes of glioma are present.

  • < Sección anterior
  • Sección siguiente >
  • < Sección anterior
  • Sección siguiente >

Tratamiento

Treatment
A large multi-disciplinary team of medical specialists and health professionals is required for the therapeutic management of patients with gliomas. Patients will usually come to the emergency room or be referred by their primary care physician for MRI. The MRI of the patient’s brain will then be interpreted by a radiologist or neuroradiologist and identify a mass. After an initial diagnosis is made, the patient will be considered for neurosurgery to safely remove as much of the tumor as possible (surgical resection. After surgery, a neuropathologist will examine and characterize the tumor under the microscope.

The age of the patient, clinical symptoms, imaging findings and pathology analysis help to determine the best treatment options and the prognosis for the patient. Therapeutic management depends on the type of glioma, its size and location and the specific characteristics of the patient. Especially in patients where the tumor cannot be entirely removed because it is invading the brain in crucial areas or is not accessible, chemotherapy and radiotherapy will follow surgery. The collaboration of radiation oncologists and medical oncologists or neuro oncologists will therefore be required. Examples of chemotherapy for glioma include temozolomide and lomustine. These two medications are part of a drug class known as alkylating agents. Their therapeutic effect is to damage DNA of tumor cells that leads to tumor cell death. Well circumscribed gliomas might be managed by surgical resection only. MRI will usually be performed at certain intervals to assess the progression of the tumor and the effects of treatment.

In addition to chemotherapy, medication prescribed to individuals with gliomas might include anti-epileptic medication (if the patient has seizures), anti-coagulation medication (if blood clots develop) and corticosteroids (to alleviate neurological symptoms caused by the accumulation of fluid around the tumor). Neurologists and possibly other medical specialists might be required for prescriptions and follow-up of affected individuals.

Patients might have to undergo rehabilitation after surgery to recover function affected by the tumor and surgery. Rehabilitation teams comprise many health professionals, including physiotherapists, occupational therapists and nurses. Some patients may benefit from palliative care where they will receive optimal treatment to minimize their symptoms and pain, including analgesic medication, anti-epileptic medication and medication to prevent vomiting (antiemetics).

  • < Sección anterior
  • Sección siguiente >
  • < Sección anterior
  • Sección siguiente >

Investigaciones

The development of potential new treatments for gliomas is a very active field of medical research. Investigational therapies for gliomas include new ways to deliver anticancer drugs, innovations in chemotherapy and radiotherapy, utilization of factors to slow (inhibit) tumor growth or to destroy the tumor, stimulation of the patient’s own immune system to fight cancer cells (immunotherapy) and gene therapy.

Information on current clinical trials is posted on the Internet at www.clinicaltrials.gov. All studies receiving U.S. government funding, and some supported by private industry, are posted on this government web site.

For information about clinical trials being conducted at the NIH Clinical Center in Bethesda, MD, contact the NIH Patient Recruitment Office:

Toll-free: (800) 411-1222
TTY: (866) 411-1010
Email: prpl@cc.nih.gov

Some current clinical trials also are posted on the following page on the NORD website:
https://rarediseases.org/for-patients-and-families/information-resources/news-patient-recruitment/

For information about clinical trials sponsored by private sources, in the main, contact:
www.centerwatch.com

For more information about clinical trials conducted in Europe, contact:
https://www.clinicaltrialsregister.eu/

  • < Sección anterior
  • Sección siguiente >
  • < Sección anterior
  • Sección siguiente >

Referencias

JOURNAL ARTICLES
Louis DN, Perry A, Wesseling P, et al. The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro Oncol. 2021;23(8):1231-1251. doi:10.1093/neuonc/noab106

Bready D, Placantonakis DG. Molecular pathogenesis of low-grade glioma. Neurosurg Clin N Am 2019;30:17-25.

Smits A, Jakola AS. Clinical presentation, natural history, and prognosis of diffuse low-grade gliomas. Neurosurg Clin N Am 2019;30:35-42.

Jain KK. A Critical Overview of targeted therapies for glioblastoma. Front Oncol 2018;8:419.

Lapointe S, Perry A, Butowski NA. Primary brain tumours in adults. Lancet 2018;392:432-46.

Ostrom QT, Gittleman H, Truitt G, Boscia A, Kruchko C, Barnholtz-Sloan JS. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2011-2015. Neuro Oncol 2018;20:iv1-iv86.

Zang L, Kondengaden SM, Che F, Wang L, Heng X. Potential epigenetic-based therapeutic targets for glioma. Front Mol Neurosci 2018;11:408.

Alfonso JCL, Talkenberger K, Seifert M, et al. The biology and mathematical modelling of glioma invasion: a review. J R Soc Interface 2017;14.

Nandakumar P, Mansouri A, Das S. The role of ATRX in glioma biology. Front Oncol 2017;7:236.

Ashby LS, Smith KA, Stea B. Gliadel wafer implantation combined with standard radiotherapy and concurrent followed by adjuvant temozolomide for treatment of newly diagnosed high-grade glioma: a systematic literature review. World J Surg Oncol 2016;14:225.

Bell RJ, Rube HT, Xavier-Magalhaes A, et al. Understanding TERT promoter mutations: a common path to immortality. Mol Cancer Res 2016;14:315-23.

Jaber M, Wolfer J, Ewelt C, et al. The value of 5-aminolevulinic acid in low-grade gliomas and high-grade gliomas lacking glioblastoma imaging features: an analysis based on fluorescence, magnetic resonance imaging, 18F-fluoroethyl tyrosine positron emission tomography, and tumor molecular factors. Neurosurgery 2016;78:401-11; discussion 11.

Kresak JL, Walsh M. Neurofibromatosis: A review of NF1, NF2, and schwannomatosis. J Pediatr Genet 2016;5:98-104.

Perry A, Wesseling P. Histologic classification of gliomas. Handb Clin Neurol 2016;134:71-95.

Masui K, Mischel PS, Reifenberger G. Molecular classification of gliomas. Handb Clin Neurol 2016;134:97-120.

Solomon DA, Wood MD, Tihan T, et al. Diffuse midline gliomas with histone H3-K27M mutation: a series of 47 cases assessing the spectrum of morphologic variation and associated genetic alterations. Brain Pathol 2016;26:569-80.

Penman CL, Faulkner C, Lowis SP, Kurian KM. Current understanding of BRAF alterations in diagnosis, prognosis, and therapeutic targeting in pediatric low-grade gliomas. Front Oncol 2015;5:54.

Posti JP, Bori M, Kauko T, et al. Presenting symptoms of glioma in adults. Acta Neurol Scand 2015;131:88-93.

Weller M, Wick W, Aldape K, et al. Glioma. Nat Rev Dis Primers 2015;1:15017.

Ahmed R, Oborski MJ, Hwang M, Lieberman FS, Mountz JM. Malignant gliomas: current perspectives in diagnosis, treatment, and early response assessment using advanced quantitative imaging methods. Cancer Manag Res 2014;6:149-70.

Ostrom QT, Bauchet L, Davis FG, et al. The epidemiology of glioma in adults: a “state of the science” review. Neuro Oncol 2014;16:896-913.

Parker M, Mohankumar KM, Punchihewa C, et al. C11orf95-RELA fusions drive oncogenic NF-kappaB signalling in ependymoma. Nature 2014;506:451-5.

Stupp R, Brada M, van den Bent MJ, Tonn JC, Pentheroudakis G, Group EGW. High-grade glioma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2014;25 Suppl 3:iii93-101.

Weller M, van den Bent M, Hopkins K, et al. EANO guideline for the diagnosis and treatment of anaplastic gliomas and glioblastoma. Lancet Oncol 2014;15:e395-403.

Boots-Sprenger SH, Sijben A, Rijntjes J, et al. Significance of complete 1p/19q co-deletion, IDH1 mutation and MGMT promoter methylation in gliomas: use with caution. Mod Pathol. 2013;26:922-9.

Lin ZX. Glioma-related edema: new insight into molecular mechanisms and their clinical implications. Chin J Cancer 2013;32:49-52.

Omuro A, DeAngelis LM. Glioblastoma and other malignant gliomas: a clinical review. JAMA 2013;310:1842-50.

Zhang K, Wang XQ, Zhou B, Zhang L. The prognostic value of MGMT promoter methylation in Glioblastoma multiforme: a meta-analysis. Fam Cancer 2013;12:449-58.

Jakola AS, Myrmel KS, Kloster R, et al. Comparison of a strategy favoring early surgical resection vs a strategy favoring watchful waiting in low-grade gliomas. JAMA 2012;308:1881-8.

Wright BL, Lai JT, Sinclair AJ. Cerebrospinal fluid and lumbar puncture: a practical review. J Neurol 2012;259:1530-45.

Onishi M, Ichikawa T, Kurozumi K, Date I. Angiogenesis and invasion in glioma. Brain Tumor Pathol 2011;28:13-24.

Schneider T, Mawrin C, Scherlach C, Skalej M, Firsching R. Gliomas in adults. Dtsch Arztebl Int 2010;107:799-807; quiz 8.

Yan H, Parsons DW, Jin G, et al. IDH1 and IDH2 mutations in gliomas. N Engl J Med 2009;360:765-73.

Wen PY, Kesari S. Malignant gliomas in adults. N Engl J Med 2008;359:492-507.

Parsons DW, Jones S, Zhang X, et al. An integrated genomic analysis of human glioblastoma multiforme. Science 2008;321:1807-12.

Friedman HS, Kerby T, Calvert H. Temozolomide and treatment of malignant glioma. Clin Cancer Res 2000;6:2585-97.

Plate KH, Breier G, Weich HA, Mennel HD, Risau W. Vascular endothelial growth factor and glioma angiogenesis: coordinate induction of VEGF receptors, distribution of VEGF protein and possible in vivo regulatory mechanisms. Int J Cancer 1994;59:520-9.

INTERNET
Chheda MG, Wen PY. Uncommon brain tumors, UpToDate. Last updated: Apr 24, 2023. https://www.uptodate.com/contents/uncommon-brain-tumors Accessed August 16, 2023.

Brook I. Brain Abscess Differential Diagnoses, Medscape. Last updated: March 2, 2021.
https://reference.medscape.com/article/212946-differential Accessed August 16, 2023.

Glioma. Genetic and Rare Diseases Information Center. Last updated: July 15, 2015.
https://rarediseases.info.nih.gov/diseases/6513/glioma Accessed August 16, 2023.

Glioma. American Brain Tumor Association. May 2018.
https://www.abta.org/tumor_types/glioma/ Accessed August 16, 2023.

Louis DN, Schiff D, Batchelor T Classification and pathologic diagnosis of gliomas, UpToDate. Last updated: Aug 19, 2022.
https://www.uptodate.com/contents/classification-and-pathologic-diagnosis-of-gliomas Accessed August 16, 2023.

Batchelor T, Louis DN, Molecular pathogenesis of diffuse gliomas. UpToDate. Last updated: Mar 16, 2023. https://www.uptodate.com/contents/molecular-pathogenesis-of-diffuse-gliomas Accessed August 16, 2023.

Tumor Types: Understanding Brain Tumors. National Brain Tumor Society. https://braintumor.org/brain-tumors/about-brain-tumors/brain-tumor-types/ Accessed August 16, 2023.

  • < Sección anterior
  • Sección siguiente >

Programas & Recursos

Programas de asistencia RareCare®

Programas de Asistencia Adicional

Programa de Asistencia MedicAlert

NORD y la Fundación MedicAlert se han asociado en un nuevo programa para brindar protección a pacientes con enfermedades raras en situaciones de emergencia.

Aprende más https://rarediseases.org/patient-assistance-programs/medicalert-assistance-program/

Programa de Apoyo Educativo de Enfermedades Raras

Asegurarse de que los pacientes y los cuidadores estén equipados con las herramientas que necesitan para vivir su mejor vida mientras manejan su condición rara es una parte vital de la misión de NORD.

Aprende más https://rarediseases.org/patient-assistance-programs/rare-disease-educational-support/

Programa de descanso para cuidadores raros

Este programa de asistencia, primero en su tipo, está diseñado para los cuidadores de un niño o adulto diagnosticado con un trastorno raro.

Aprende más https://rarediseases.org/patient-assistance-programs/caregiver-respite/

Organizaciones de pacientes


National Organization for Rare Disorders