51
Views
14
CrossRef citations to date
0
Altmetric
Review

Diagnostic and molecular pathology of meningiomas

, &
Pages 1671-1683 | Published online: 09 Jan 2014

References

  • Kleihues P, Cavenee WK. Pathology and Genetics Of Tumours Of The Nervous System (2nd Edition). IARC Press, Lyon, France, 314 (2000).
  • Graham DI, Lantos PL. Greenfield's Neuropathology (7th Edition). Arnold, London, UK (2002).
  • DeMonte F, Marmor E, Al-Mefty O. Meningiomas, in Brain Tumors.An Encyclopedic Approach. Kaye AH, Laws ER (Eds). Churchill Livingstone, London, UK, 719–750 (2001).
  • Rausing A, Ybo W, Stenflo J. Intracranial meningioma – a population study of ten years. Acta Neurol. Scand.46(1), 102–110 (1970).
  • Ironside JW, Moss TH, Louis DN, Lowe JS, Well RO. Diagnostic Pathology of Nervous System Tumours. Churchill Livingstone, London, UK (2002).
  • Grunberg SM, Rankin C, Townsend J et al. Phase III double-blind randomized placebo controlled study of mifepristone (RU486) for the treatment of unresectable meningioma. ACSO Proc.20, 56A (2001).
  • McCutcheon IE, Flyvbjerg A, Hill H et al. Antitumor activity of the growth hormone receptor antagonist pegvisomant against human meningiomas in nude mice. J. Neurosurg.94, 487–492 (2001).
  • Perry A, Gutmann DH, Reifenberger G. Molecular pathogenesis of meningiomas. J. Neurooncol.70(2), 183–202 (2004).
  • Sanders WP, Chundi VV. Extra-axial Tumors Including Pituitary and Parasellar in Neuroimaging. Orrison WW (Ed.). WB Saunders, PA, USA, 612–718 (2000).
  • Goldstein RA, Jorden MA, Harsh GR IV. Meningiomas: natural history, diagnosis, and imaging. In: Cancer of the Nervous System. Black PM, Loeffler JS (Eds). Lippincott Williams & Wilkins, PA, USA, 279–314 (2005).
  • Bendszus M, Rao G, Burger R et al. Is there a benefit of preoperative meningioma embolization? Neurosurgery47(6), 1306–1311 (2000).
  • Nakasu S, Hirano A, Shimura T, Llena JF. Incidental meningiomas in autopsy study. Surg. Neurol.27(4), 319–322 (1987).
  • Kleihues P, Burger PC, Scheithauer BW. Histological Typing of Tumours of the Central Nervous System (2nd Edition). Springer Verlag, Berlin, Germany (1993).
  • Roggendorf W, Schuster T, Peiffer J. Proliferative potential of meningiomas determined with the monoclonal antibody Ki-67. Acta Neuropathol. (Berl.)73(4), 361–364 (1987).
  • Prayson RA. Malignant meningioma: a clinicopathologic study of 23 patients including MIB1 and p53 immunohistochemistry. Am. J. Clin. Pathol.105(6), 719–726 (1996).
  • Maier H, Wanschitz J, Sedivy R et al. Proliferation and DNA fragmentation in meningioma subtypes. Neuropathol. Appl. Neurobiol.23(6), 496–506 (1997).
  • Johnston MC. A radioautographic study of the migration and fate of cranial neural crest cells in the chick embryo. Anat. Rec.156(2), 143–155 (1966).
  • Couly GF, Coltey PM, Le Douarin NM. The developmental fate of the cephalic mesoderm in quail-chick chimeras. Development114(1), 1–15 (1992).
  • Bagnall KM, Higgins SJ, Sanders EJ. The contribution made by a single somite to the vertebral column: experimental evidence in support of resegmentation using the chick-quail chimaera model. Development103(1), 69–85 (1988).
  • Halata Z, Grim M, Christ B. Origin of spinal cord meninges, sheaths of peripheral nerves, and cutaneous receptors including Merkel cells. An experimental and ultrastructural study with avian chimeras. Anat. Embryol. (Berl.)182(6), 529–537 (1990).
  • Catala M. Embryonic and fetal development of structures associated with the cerebro-spinal fluid in man and other species. Part I: the ventricular system, meninges and choroid plexuses. Arch. Anat. Cytol. Pathol.46(3), 153–169 (1998).
  • Kros J, de Greve K, van Tilborg A et al. NF2 status of meningiomas is associated with tumour localization and histology. J. Pathol.194(3), 367–372 (2001).
  • Hartmann C, Sieberns J, Gehlhaar C et al. NF2 Mutations in secretory and other rare variants of meningiomas. Brain Pathol.16(1), 15–19 (2006).
  • Wellenreuther R, Kraus JA, Lenartz D et al. Analysis of the neurofibromatosis 2 gene reveals molecular variants of meningioma. Am. J. Pathol.146(4), 827–832 (1995).
  • Evans JJ, Jeun SS, Lee JH et al. Molecular alterations in the neurofibromatosis type 2 gene and its protein rarely occurring in meningothelial meningiomas. J. Neurosurg.94(1), 111–117 (2001).
  • Ruttledge MH, Xie YG, Han F et al. Deletions on chromosome 22 in sporadic meningioma. Genes Chromosomes Cancer10(2), 122–130 (1994).
  • Zankl H, Zang KD. Cytological and cytogenetical studies on brain tumors. 4. Identification of the missing G chromosome in human meningiomas as no. 22 by fluorescence technique. Humangenetik14(2), 167–169 (1972).
  • Mark J, Levan G, Mitelman F. Identification by fluorescence of the G chromosome lost in human meningomas. Hereditas71(1), 163–168 (1972).
  • Dumanski JP, Carlbom E, Collins VP, Nordenskjold M. Deletion mapping of a locus on human chromosome 22 involved in the oncogenesis of meningioma. Proc. Natl Acad. Sci. USA84(24), 9275–9279 (1987).
  • Lindblom A, Ruttledge M, Collins VP, Nordenskjold M, Dumanski JP. Chromosomal deletions in anaplastic meningiomas suggest multiple regions outside chromosome 22 as important in tumor progression. Int. J. Cancer56(3), 354–357 (1994).
  • Meese E, Blin N, Zang KD. Loss of heterozygosity and the origin of meningioma. Hum. Genet.77(4), 349–351 (1987).
  • Dumanski JP, Rouleau GA, Nordenskjold M, Collins VP. Molecular genetic analysis of chromosome 22 in 81 cases of meningioma. Cancer Res.50(18), 5863–5867 (1990).
  • Trofatter JA, MacCollin MM, Rutter JL et al. A novel moesin-, ezrin-, radixin-like gene is a candidate for the neurofibromatosis 2 tumor suppressor. Cell75(4), 826 (1993).
  • Rouleau GA, Merel P, Lutchman M et al. Alteration in a new gene encoding a putative membrane-organizing protein causes neuro-fibromatosis type 2. Nature363(6429), 515–521 (1993).
  • Lallemand D, Curto M, Saotome I, Giovannini M, McClatchey AI. NF2 deficiency promotes tumorigenesis and metastasis by destabilizing adherens junctions. Genes Dev.17(9), 1090–1100 (2003).
  • Kissil JL, Johnson KC, Eckman MS, Jacks T. Merlin phosphorylation by p21-activated kinase 2 and effects of phosphorylation on merlin localization. J. Biol. Chem.277(12), 10394–10399 (2002).
  • Shaw RJ, Paez JG, Curto M et al. The Nf2 tumor suppressor, merlin, functions in Rac-dependent signaling. Dev. Cell1(1), 63–72 (2001).
  • Ikeda K, Saeki Y, Gonzalez-Agosti C, Ramesh V, Chiocca EA. Inhibition of NF2-negative and NF2-positive primary human meningioma cell proliferation by overexpression of merlin due to vector-mediated gene transfer. J. Neurosurg.91(1), 85–92 (1999).
  • Kalamarides M, Niwa-Kawakita M, Leblois H et al. Nf2 gene inactivation in arachnoidal cells is rate-limiting for meningioma development in the mouse. Genes Dev.16(9), 1060–1065 (2002).
  • Louis DN, Ramesh V, Gusella JF. Neuropathology and molecular genetics of neurofibromatosis 2 and related tumors. Brain Pathol.5(2), 163–172 (1995).
  • Kimura Y, Koga H, Araki N et al. The involvement of calpain-dependent proteolysis of the tumor suppressor NF2 (merlin) in schwannomas and meningiomas. Nat. Med.4(8), 915–922 (1998).
  • Lomas J, Bello MJ, Arjona D et al. Genetic and epigenetic alteration of the NF2 gene in sporadic meningiomas. Genes Chromosomes Cancer42(3), 314–319 (2005).
  • De Vitis LR, Tedde A, Vitelli F et al. Screening for mutations in the neurofibromatosis type 2 (NF2) gene in sporadic meningiomas. Hum. Genet.97(5), 632–637 (1996).
  • Harada T, Irving RM, Xuereb JH et al. Molecular genetic investigation of the neurofibromatosis type 2 tumor suppressor gene in sporadic meningioma. J. Neurosurg.84(5), 847–851 (1996).
  • Lekanne Deprez RH, Bianchi AB, Groen NA et al. Frequent NF2 gene transcript mutations in sporadic meningiomas and vestibular schwannomas. Am. J. Hum. Genet.54(6), 1022–1029 (1994).
  • Merel P, Hoang-Xuan K, Sanson M et al. Predominant occurrence of somatic mutations of the NF2 gene in meningiomas and schwannomas. Genes Chromosomes Cancer13(3), 211–216 (1995).
  • Papi L, De Vitis LR, Vitelli F et al. Somatic mutations in the neurofibromatosis type 2 gene in sporadic meningiomas. Hum. Genet.95(3), 347–351 (1995).
  • Lee JH, Sundaram V, Stein DJ et al. Reduced expression of schwannomin/merlin in human sporadic meningiomas. Neurosurgery40(3), 578–587 (1997).
  • Wellenreuther R, Waha A, Vogel Y et al. Quantitative analysis of neurofibromatosis type 2 gene transcripts in meningiomas supports the concept of distinct molecular variants. Lab. Invest.77(6), 601–606 (1997).
  • Lomas J, Bello MJ, Alonso ME et al. Loss of chromosome 22 and absence of NF2 gene mutation in a case of multiple meningiomas. Hum. Pathol.33(3), 375–378 (2002).
  • Ruttledge MH, Sarrazin J, Rangaratnam S et al. Evidence for the complete inactivation of the NF2 gene in the majority of sporadic meningiomas. Nat. Genet.6(2), 180–184 (1994).
  • Sevenet N, Lellouch-Tubiana A, Schofield D et al. Spectrum of hSNF5/INI1 somatic mutations in human cancer and genotype-phenotype correlations. Hum. Mol. Genet.8(13), 2359–2368 (1999).
  • Biegel JA, Zhou JY, Rorke LB et al. Germ-line and acquired mutations of INI1 in atypical teratoid and rhabdoid tumors. Cancer Res.59(1), 74–79 (1999).
  • Schmitz U, Mueller W, Weber M et al. INI1 mutations in meningiomas at a potential hotspot in exon 9. Br. J. Cancer84(2), 199–201 (2001).
  • Lekanne Deprez RH, Riegman PH, Groen NA et al. Cloning and characterization of MN1, a gene from chromosome 22q11, which is disrupted by a balanced translocation in a meningioma. Oncogene10(8), 1521–1528 (1995).
  • Meester-Smoor MA, Vermeij M, van Helmond MJ et al. Targeted disruption of the Mn1 oncogene results in severe defects in development of membranous bones of the cranial skeleton. Mol. Cell. Biol.25(10), 4229–4236 (2005).
  • Peyrard M, Seroussi E, Sandberg-Nordqvist AC et al. The human LARGE gene from 22q12.3-q13.1 is a new, distinct member of the glycosyltransferase gene family. Proc. Natl Acad. Sci. USA96(2), 598–603 (1999).
  • Kuns R, Kissil JL, Newsham IF et al. Protein 4.1B expression is induced in mammary epithelial cells during pregnancy and regulates their proliferation. Oncogene24(43), 6502–6515 (2005).
  • Gutmann DH, Donahoe J, Perry A et al. Loss of DAL-1, a protein 4.1-related tumor suppressor, is an important early event in the pathogenesis of meningiomas. Hum. Mol. Genet.9(10), 1495–1500 (2000).
  • Nunes F, Shen Y, Niida Y et al. Inactivation patterns of NF2 and DAL-1/4.1B (EPB41L3) in sporadic meningioma. Cancer Genet. Cytogenet.162(2), 135–139 (2005).
  • Perry A, Cai DX, Scheithauer BW et al. Merlin, DAL-1, and progesterone receptor expression in clinicopathologic subsets of meningioma: a correlative immunohistochemical study of 175 cases. J. Neuropathol. Exp. Neurol.59(10), 872–879 (2000).
  • Gutmann DH, Hirbe AC, Huang ZY, Haipek CA. The protein 4.1 tumor suppressor, DAL-1, impairs cell motility, but regulates proliferation in a cell-type-specific fashion. Neurobiol. Dis.8(2), 266–278 (2001).
  • Martinez-Glez V, Bello MJ, Franco-Hernandez C et al. Mutational analysis of the DAL-1/4.1B tumour-suppressor gene locus in meningiomas. Int. J. Mol. Med.16(4), 771–774 (2005).
  • Heinrich B, Hartmann C, Stemmer-Rachamimov AO, Louis DN, MacCollin M. Multiple meningiomas: investigating the molecular basis of sporadic and familial forms. Int. J. Cancer103(4), 483–488 (2003).
  • Yi C, McCarty JH, Troutman SA et al. Loss of the putative tumor suppressor band 4.1B/Dal1 gene is dispensable for normal development and does not predispose to cancer. Mol. Cell. Biol.25(22), 10052–10059 (2005).
  • Robb VA, Li W, Gascard P et al. Identification of a third Protein 4.1 tumor suppressor, Protein 4.1R, in meningioma pathogenesis. Neurobiol. Dis.13(3), 191–202 (2003).
  • Piaskowski S, Rieske P, Szybka M et al. GADD45A and EPB41 as tumor suppressor genes in meningioma pathogenesis. Cancer Genet. Cytogenet.162(1), 63–67 (2005).
  • Xie J, Johnson RL, Zhang X et al. Mutations of the PATCHED gene in several types of sporadic extracutaneous tumors. Cancer Res.57(12), 2369–2372 (1997).
  • Kimonis VE, Mehta SG, Digiovanna JJ, Bale SJ, Pastakia B. Radiological features in 82 patients with nevoid basal cell carcinoma (NBCC or Gorlin) syndrome. Genet. Med.6(6), 495–502 (2004).
  • Cai DX, Banerjee R, Scheithauer BW et al. Chromosome 1p and 14q FISH analysis in clinicopathologic subsets of meningioma: diagnostic and prognostic implications. J. Neuropathol. Exp. Neurol.60(6), 628–636 (2001).
  • Cai DX, James CD, Scheithauer BW, Couch FJ, Perry A. PS6K amplification characterizes a small subset of anaplastic meningiomas. Am. J. Clin. Pathol.115(2), 213–218 (2001).
  • Buschges R, Ichimura K, Weber RG, Reifenberger G, Collins VP. Allelic gain and amplification on the long arm of chromosome 17 in anaplastic meningiomas. Brain Pathol.12(2), 145–153 (2002).
  • Lamszus K, Kluwe L, Matschke J et al. Allelic losses at 1p, 9q, 10q, 14q, and 22q in the progression of aggressive meningiomas and undifferentiated meningeal sarcomas. Cancer Genet. Cytogenet.110(2), 103–110 (1999).
  • Ozaki S, Nishizaki T, Ito H, Sasaki K. Comparative genomic hybridization analysis of genetic alterations associated with malignant progression of meningioma. J. Neurooncol.41(2), 167–174 (1999).
  • Weber RG, Bostrom J, Wolter M et al. Analysis of genomic alterations in benign, atypical, and anaplastic meningiomas: toward a genetic model of meningioma progression. Proc. Natl Acad. Sci. USA94(26), 14719–14724 (1997).
  • von Deimling A, Fimmers R, Schmidt MC et al. Comprehensive allelotype and genetic anaysis of 466 human nervous system tumors. J. Neuropathol. Exp. Neurol.59(6), 544–558 (2000).
  • Buschges R, Bostrom J, Wolter M et al. Analysis of human meningiomas for aberrations of the MADH2, MADH4, APM-1 and DCC tumor suppressor genes on the long arm of chromosome 18. Int. J. Cancer92(4), 551–554 (2001).
  • Simon M, von Deimling A, Larson JJ et al. Allelic losses on chromosomes 14, 10, and 1 in atypical and malignant meningiomas: a genetic model of meningioma progression. Cancer Res.55(20), 4696–4701 (1995).
  • Simon M, Kokkino AJ, Warnick RE et al. Role of genomic instability in meningioma progression. Genes Chromosomes Cancer16(4), 265–269 (1996).
  • Bostrom J, Muhlbauer A, Reifenberger G. Deletion mapping of the short arm of chromosome 1 identifies a common region of deletion distal to D1S496 in human meningiomas. Acta Neuropathol. (Berl.)94(5), 479–485 (1997).
  • Lamszus K. Meningioma pathology, genetics, and biology. J. Neuropathol. Exp. Neurol.63(4), 275–286 (2004).
  • Bostrom J, Meyer-Puttlitz B, Wolter M et al. Alterations of the tumor suppressor genes CDKN2A (p16(INK4a)), p14(ARF), CDKN2B (p15(INK4b)), and CDKN2C (p18(INK4c)) in atypical and anaplastic meningiomas. Am. J. Pathol.159(2), 661–669 (2001).
  • Lomas J, Bello MJ, Arjona D et al. Analysis of p73 gene in meningiomas with deletion at 1p. Cancer Genet. Cytogenet.129(1), 88–91 (2001).
  • Lusis EA, Watson MA, Chicoine MR et al. Integrative genomic analysis identifies NDRG2 as a candidate tumor suppressor gene frequently inactivated in clinically aggressive meningioma. Cancer Res.65(16), 7121–7126 (2005).
  • Joachim T, Ram Z, Rappaport ZH et al. Comparative analysis of the NF2, TP53, PTEN, KRAS, NRAS and HRAS genes in sporadic and radiation-induced human meningiomas. Int. J. Cancer94(2), 218–221 (2001).
  • Peters N, Wellenreuther R, Rollbrocker B et al. Analysis of the PTEN gene in human meningiomas. Neuropathol. Appl. Neurobiol.24(1), 3–8 (1998).
  • Bostrom J, Cobbers JM, Wolter M et al. Mutation of the PTEN (MMAC1) tumor suppressor gene in a subset of glioblastomas but not in meningiomas with loss of chromosome arm 10q. Cancer Res.58(1), 29–33 (1998).
  • Simon M, Park TW, Koster G et al. Alterations of INK4a(p16-p14ARF)/INK4b(p15) expression and telomerase activation in meningioma progression. J. Neurooncol.55(3), 149–158 (2001).
  • Perry A, Banerjee R, Lohse CM, Kleinschmidt-DeMasters BK, Scheithauer BW. A role for chromosome 9p21 deletions in the malignant progression of meningiomas and the prognosis of anaplastic meningiomas. Brain Pathol.12(2), 183–190 (2002).
  • Wang JL, Zhang ZJ, Hartman M et al. Detection of TP53 gene mutation in human meningiomas: a study using immunohistochemistry, polymerase chain reaction/single-strand conformation polymorphism and DNA sequencing techniques on paraffin-embedded samples. Int. J. Cancer64(4), 223–228 (1995).
  • Bickerstaff ER, Small JM, Guest IA. The relapsing course of certain meningiomas in relation to pregnancy and menstruation. J. Neurol. Neurosurg. Psychiatry21(2), 89–91 (1958).
  • Roelvink NC, Kamphorst W, van Alphen HA, Rao BR. Pregnancy-related primary brain and spinal tumors. Arch. Neurol.44(2), 209–215 (1987).
  • Blaauw G, Blankenstein MA, Lamberts SW. Sex steroid receptors in human meningiomas. Acta Neurochir. (Wien.)79(1), 42–47 (1986).
  • Carroll RS, Zhang J, Black PM. Expression of estrogen receptors α and β human meningiomas. J. Neurooncol.42(2), 109–116 (1999).
  • Perry A, Lusis EA, Gutmann DH. Meningothelial hyperplasia: a detailed clinicopathologic, immunohistochemical and genetic study of 11 cases. Brain Pathol.15(2), 109–115 (2005).
  • Matsuda Y, Kawamoto K, Kiya K et al. Antitumor effects of antiprogesterones on human meningioma cells in vitro and in vivo. J. Neurosurg.80(3), 527–534 (1994).
  • Konstantinidou AE, Korkolopoulou P, Mahera H et al. Hormone receptors in non-malignant meningiomas correlate with apoptosis, cell proliferation and recurrence-free survival. Histopathology43(3), 280–290 (2003).
  • Rubinstein AB, Loven D, Geier A, Reichenthal E, Gadoth N. Hormone receptors in initially excised versus recurrent intracranial meningiomas. J. Neurosurg.81(2), 184–187 (1994).
  • Jacobs HM, van Spriel AB, Koehorst SG et al. The truncated estrogen receptor α variant lacking exon 5 is not involved in progesterone receptor expression in meningiomas. J. Steroid Biochem. Mol. Biol.71(5–6), 167–172 (1999).
  • Bouillot P, Pellissier JF, Devictor B et al. Quantitative imaging of estrogen and progesterone receptors, estrogen-regulated protein, and growth fraction: immunocytochemical assays in 52 meningiomas. Correlation with clinical and morphological data. J. Neurosurg.81(5), 765–773 (1994).
  • Probst-Cousin S, Villagran-Lillo R, Lahl R et al. Secretory meningioma: clinical, histologic, and immunohistochemical findings in 31 cases. Cancer79(10), 2003–2015 (1997).
  • Yang SY, Xu GM. Expression of PDGF and its receptor as well as their relationship to proliferating activity and apoptosis of meningiomas in human meningiomas. J. Clin. Neurosci.8(Suppl. 1), 49–53 (2001).
  • Black PM, Carroll R, Glowacka D, Riley K, Dashner K. Platelet-derived growth factor expression and stimulation in human meningiomas. J. Neurosurg.81(3), 388–393 (1994).
  • Carroll RS, Black PM, Zhang J et al. Expression and activation of epidermal growth factor receptors in meningiomas. J. Neurosurg.87(2), 315–323 (1997).
  • Torp SH, Helseth E, Dalen A, Unsgaard G. Expression of epidermal growth factor receptor in human meningiomas and meningeal tissue. APMIS100(9), 797–802 (1992).
  • Nordqvist AC, Peyrard M, Pettersson H et al. A high ratio of insulin-like growth factor II/insulin-like growth factor binding protein 2 messenger RNA as a marker for anaplasia in meningiomas. Cancer Res.57(13), 2611–2614 (1997).
  • Goldman CK, Bharara S, Palmer CA et al. Brain edema in meningiomas is associated with increased vascular endothelial growth factor expression. Neurosurgery40(6), 1269–1277 (1997).
  • Pagotto U, Arzberger T, Hopfner U et al. Expression and localization of endothelin-1 and endothelin receptors in human meningiomas. Evidence for a role in tumoral growth. J. Clin. Invest.96(4), 2017–2025 (1995).
  • Langford LA, Piatyszek MA, Xu R et al. Telomerase activity in ordinary meningiomas predicts poor outcome. Hum. Pathol.28(4), 416–420 (1997).
  • Simon M, Park TW, Leuenroth S et al. Telomerase activity and expression of the telomerase catalytic subunit, hTERT, in meningioma progression. J. Neurosurg.92(5), 832–840 (2000).
  • Falchetti ML, Larocca LM, Pallini R. Telomerase in brain tumors. Childs Nerv. Syst.18(3–4), 112–117 (2002).
  • Stafford SL, Perry A, Suman VJ et al. Primarily resected meningiomas: outcome and prognostic factors in 581 Mayo Clinic patients, 1978 through 1988. Mayo Clin. Proc.73(10), 936–942 (1998).
  • Stangl AP, Wellenreuther R, Lenartz D et al. Clonality of multiple meningiomas. J. Neurosurg.86(5), 853–858 (1997).
  • von Deimling A, Kraus JA, Stangl AP et al. Evidence for subarachnoid spread in the development of multiple meningiomas. Brain Pathol.5(1), 11–14 (1995).
  • Zhu J, Frosch MP, Busque L et al. Analysis of meningiomas by methylation- and transcription-based clonality assays. Cancer Res.55(17), 3865–3872 (1995).
  • Wu JK, MacGillavry M, Kessaris C et al. Clonal analysis of meningiomas. Neurosurgery38(6), 1196–1200 (1996).
  • Borovich B, Doron Y. Recurrence of intracranial meningiomas: the role played by regional multicentricity. J. Neurosurg.64(1), 58–63 (1986).
  • McCarthy BJ, Davis FG, Freels S et al. Factors associated with survival in patients with meningioma. J. Neurosurg.88(5), 831–839 (1998).
  • Jaaskelainen J. Seemingly complete removal of histologically benign intracranial meningioma: late recurrence rate and factors predicting recurrence in 657 patients. A multivariate analysis. Surg. Neurol.26(5), 461–469 (1986).
  • Kinjo T, al-Mefty O, Ciric I. Grade zero removal of supratentorial convexity meningiomas. Neurosurgery33(3), 394–399 (1995).
  • Simon M, Bostrom J, Koch P, Schramm J. Inter-institutional variance of postoperative radiotherapy and follow up for meningiomas in Germany. Impact of changes of the WHO classification. J. Neurol. Neurosurg. Psychiatry77(6), 767–73 (2005).
  • Willis J, Smith C, Ironside JW et al. The accuracy of meningioma grading: a 10-year retrospective audit. Neuropathol. Appl. Neurobiol.31(2), 141–149 (2005).
  • Perry A, Stafford SL, Scheithauer BW, Suman VJ, Lohse CM. Meningioma grading: an analysis of histologic parameters. Am. J. Surg. Pathol.21(12), 1455–1465 (1997).
  • Maillo A, Orfao A, Sayagues JM et al. New classification scheme for the prognostic stratification of meningioma on the basis of chromosome 14 abnormalities, patient age, and tumor histopathology. J. Clin. Oncol.21(17), 3285–3295 (2003).
  • Mihaila D, Gutierrez JA, Rosenblum ML et al. Meningiomas: analysis of loss of heterozygosity on chromosome 10 in tumor progression and the delineation of four regions of chromosomal deletion in common with other cancers. Clin. Cancer Res.9(12), 4435–4442 (2003).
  • Yamasaki F, Yoshioka H, Hama S et al. Recurrence of meningiomas. Cancer89(5), 1102–1110 (2000).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.