1
|
SAITO R, CHAMBERS JK, UCHIDA K. The expression of platelet-derived growth factor and its receptor in canine and feline meningiomas. J Vet Med Sci 2023; 85:1057-1062. [PMID: 37558425 PMCID: PMC10600539 DOI: 10.1292/jvms.23-0300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/31/2023] [Indexed: 08/11/2023] Open
Abstract
Feline meningiomas usually have benign biological behavior, while canine and human meningiomas are often classified as grade 2 or 3. Activation of the platelet-derived growth factor (PDGF) and its receptor signal pathway through PDGFβ/Rβ autocrine and paracrine is considered to play an important role in the tumor proliferation and malignant transformation of human meningiomas. However, there have been few studies about the expression of these molecules in canine meningiomas and no studies about their expression in feline meningiomas. We analyzed the PDGFα/Rα and PDGFβ/Rβ expression in canine and feline meningiomas by immunohistochemistry and western blotting. Immunohistochemically, most canine meningiomas showed the expression of PDGFα (42/44; 95.5%), PDGFRα (44/44; 100%) and PDGFRβ (35/44; 79.5%), and a few showed the expression of PDGFβ (8/44; 18.2%). In contrast, feline meningiomas were immunopositive for PDGFRα and PDGFRβ in all cases (14/14; 100%), while no or a few cases expressed PDGFα (0/14; 0%) and PDGFβ (2/14; 14.3%). Western blotting revealed specific bands for PDGFα, PDGFRα and PDGFRβ, but not for PDGFβ in a canine meningioma. In a feline meningioma, specific bands for PDGFRα and PDGFRβ were detected, but not for PDGFα and PDGFβ. These results suggested that canine meningiomas commonly express PDGFα/Rα, and thus autocrine or paracrine PDGFα/Rα signaling may be involved in their initiation and progression. Moreover, PDGF negativity may be related to benign biological behavior and a low histopathological grade in feline meningioma.
Collapse
Affiliation(s)
- Ryo SAITO
- Laboratory of Veterinary Pathology, Graduate School of
Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - James K CHAMBERS
- Laboratory of Veterinary Pathology, Graduate School of
Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuyuki UCHIDA
- Laboratory of Veterinary Pathology, Graduate School of
Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
2
|
Go KO, Kim YZ. Brain Invasion and Trends in Molecular Research on Meningioma. Brain Tumor Res Treat 2023; 11:47-58. [PMID: 36762808 PMCID: PMC9911709 DOI: 10.14791/btrt.2022.0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 02/05/2023] Open
Abstract
Meningiomas are the most common primary brain tumors in adults. The treatment of non-benign meningiomas remains a challenging task, and after the publication of the 2021 World Health Organization classification, the importance of molecular biological classification is emerging. In this article, we introduce the mechanisms of brain invasion in atypical meningioma and review the genetic factors involved along with epigenetic regulation. First, it is important to understand the three major steps for brain invasion of meningeal cells: 1) degradation of extracellular matrix by proteases, 2) promotion of tumor cell migration to resident cells by adhesion molecules, and 3) neovascularization and supporting cells by growth factors. Second, the genomic landscape of meningiomas should be analyzed by major categories, such as germline mutations in NF2 and somatic mutations in non-NF2 genes (TRAF7, KLF4, AKT1, SMO, and POLR2A). Finally, epigenetic alterations in meningiomas are being studied, with a focus on DNA methylation, histone modification, and RNA interference. Increasing knowledge of the molecular landscape of meningiomas has allowed the identification of prognostic and predictive markers that can guide therapeutic decision-making processes and the timing of follow-up.
Collapse
Affiliation(s)
- Kyeong-O Go
- Department of Neurosurgery, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, Korea
| | - Young Zoon Kim
- Division of Neuro Oncology and Department of Neurosurgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea.
| |
Collapse
|
3
|
Receptor-Tyrosine Kinase Inhibitor Ponatinib Inhibits Meningioma Growth In Vitro and In Vivo. Cancers (Basel) 2021; 13:cancers13235898. [PMID: 34885009 PMCID: PMC8657092 DOI: 10.3390/cancers13235898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/04/2021] [Accepted: 11/17/2021] [Indexed: 11/17/2022] Open
Abstract
To date, there is no standard-of-care systemic therapy for the treatment of aggressive meningiomas. Receptor tyrosine kinases (RTK) are frequently expressed in aggressive meningiomas and are associated with poor survival. Ponatinib is a FDA- and EMA-approved RTK inhibitor and its efficacy in meningioma has not been studied so far. Therefore, we investigated ponatinib as a potential drug candidate against meningioma. Cell viability and cell proliferation of ponatinib-treated meningioma cells were assessed using crystal violet assay, manual counting and BrdU assay. Treated meningioma cell lines were subjected to flow cytometry to evaluate the effects on cell cycle and apoptosis. Meningioma-bearing mice were treated with ponatinib to examine antitumor effects in vivo. qPCR was performed to assess the mRNA levels of tyrosine kinase receptors after ponatinib treatment. Full-length cDNA sequencing was carried out to assess differential gene expression. IC50 values of ponatinib were between 171.2 and 341.9 nM in three meningioma cell lines. Ponatinib induced G0/G1 cell cycle arrest and subsequently led to an accumulation of cells in the subG1-phase. A significant induction of apoptosis was observed in vitro. In vivo, ponatinib inhibited meningioma growth by 72.6%. Mechanistically, this was associated with downregulation of PDGFRA/B and FLT3 mRNA levels, and mitochondrial dysfunction. Taken together, ponatinib is a promising candidate for targeted therapy in the treatment of aggressive meningioma.
Collapse
|
4
|
Sharma P, Katiyar V, Sharma R, Gurjar HK, Krishnan S. Letter: Role of Tyrosine Kinase Inhibitors in Recurrent Meningiomas: Controversies and Promises. Neurosurgery 2018; 82:E181-E183. [DOI: 10.1093/neuros/nyy055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
5
|
Peyre M, Salaud C, Clermont-Taranchon E, Niwa-Kawakita M, Goutagny S, Mawrin C, Giovannini M, Kalamarides M. PDGF activation in PGDS-positive arachnoid cells induces meningioma formation in mice promoting tumor progression in combination with Nf2 and Cdkn2ab loss. Oncotarget 2016; 6:32713-22. [PMID: 26418719 PMCID: PMC4741724 DOI: 10.18632/oncotarget.5296] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/14/2015] [Indexed: 11/25/2022] Open
Abstract
The role of PDGF-B and its receptor in meningeal tumorigenesis is not clear. We investigated the role of PDGF-B in mouse meningioma development by generating autocrine stimulation of the arachnoid through the platelet-derived growth factor receptor (PDGFR) using the RCAStv-a system. To specifically target arachnoid cells, the cells of origin of meningioma, we generated the PGDStv-a mouse (Prostaglandin D synthase). Forced expression of PDGF-B in arachnoid cells in vivo induced the formation of Grade I meningiomas in 27% of mice by 8 months of age. In vitro, PDGF-B overexpression in PGDS-positive arachnoid cells lead to increased proliferation.We found a correlation of PDGFR-B expression and NF2 inactivation in a cohort of human meningiomas, and we showed that, in mice, Nf2 loss and PDGF over-expression in arachnoid cells induced meningioma malignant transformation, with 40% of Grade II meningiomas. In these mice, additional loss of Cdkn2ab resulted in a higher incidence of malignant meningiomas with 60% of Grade II and 30% of Grade III meningiomas. These data suggest that chronic autocrine PDGF signaling can promote proliferation of arachnoid cells and is potentially sufficient to induce meningiomagenesis. Loss of Nf2 and Cdkn2ab have synergistic effects with PDGF-B overexpression promoting meningioma malignant transformation.
Collapse
Affiliation(s)
- Matthieu Peyre
- Department of Neurosurgery, AP-HP, Hôpital Pitié-Salpêtrière, Paris, France.,Université Paris 6 - Pierre et Marie Curie, Paris, France.,CRICM INSERM U1127 CNRS UMR 7225, Institut du Cerveau et de la Moelle Epinière, Paris, France
| | - Céline Salaud
- Université Paris 6 - Pierre et Marie Curie, Paris, France.,CRICM INSERM U1127 CNRS UMR 7225, Institut du Cerveau et de la Moelle Epinière, Paris, France
| | - Estelle Clermont-Taranchon
- Université Paris 6 - Pierre et Marie Curie, Paris, France.,CRICM INSERM U1127 CNRS UMR 7225, Institut du Cerveau et de la Moelle Epinière, Paris, France
| | - Michiko Niwa-Kawakita
- Inserm U944, CNRS U7212, Université Paris VII, Institut Universitaire d'Hématologie, Paris, France
| | | | - Christian Mawrin
- Department of Neuropathology, Otto-von-Guericke Universität, Magdeburg, Germany
| | - Marco Giovannini
- Department of Head and Neck Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Michel Kalamarides
- Department of Neurosurgery, AP-HP, Hôpital Pitié-Salpêtrière, Paris, France.,Université Paris 6 - Pierre et Marie Curie, Paris, France.,CRICM INSERM U1127 CNRS UMR 7225, Institut du Cerveau et de la Moelle Epinière, Paris, France
| |
Collapse
|
6
|
Abstract
The efficacy of surgery and radiation has been well validated in the treatment of meningiomas, with efficacy depending on tumor pathology, size, symptomatology and rate of progression. The role of medical therapy has the least amount of data but is being increasingly investigated for tumors that are inoperable or those tumors that recur and/or progress despite standard therapy. In this review, current data on the use of chemotherapeutic agents in the management of meningiomas will be reviewed, including cytotoxic, biologic, targeted molecular and hormonal agents.
Collapse
Affiliation(s)
- Wendy J Sherman
- Northwestern University Department of Neurology, 710 North Lake Shore Drive, Abbott Hall, Room 1123, Chicago, IL 60611, USA
| | | |
Collapse
|
7
|
Peyre M, Kalamarides M. Molecular genetics of meningiomas: Building the roadmap towards personalized therapy. Neurochirurgie 2014; 64:22-28. [PMID: 25245924 DOI: 10.1016/j.neuchi.2014.06.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 06/25/2014] [Accepted: 06/29/2014] [Indexed: 11/17/2022]
Abstract
New advances have recently been made in the field of molecular genetics and mouse modeling of meningiomas, opening new perspectives for future treatments. Recent genome-wide genotyping and exome sequencing studies have confirmed the pivotal role of NF2 in meningioma tumorigenesis, concerning roughly half of the tumors, and unraveled new mutations in non-NF2 meningiomas concerning AKT1, SMO, KLF4 and TRAF7. The molecular mechanisms underlying tumorigenesis of high histological grades have been progressively deciphered with the recent discovery of TERT promoter mutations in progressing tumors. A better understanding of the genetics and clinical behavior of high-grade meningiomas is mandatory in order to better design future clinical trials. New genetically engineered mouse models of benign and histologically aggressive meningioma represent a substantial resource for the establishment of relevant pre-clinical trials. By studying the mechanisms underlying these new tumorigenesis pathways and the corresponding mouse models, we should be able to offer personalized chemotherapy to patients with surgery- and radiation-refractory meningiomas in the near future.
Collapse
Affiliation(s)
- M Peyre
- Service de Neurochirurgie, AP-HP, Hôpital Pitié Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France; Inserm, UMR S975, Institut du Cerveau et de la Moelle Épinière, 75013 Paris, France; Université Paris 6 - Pierre-et-Marie-Curie, 75013 Paris, France
| | - M Kalamarides
- Service de Neurochirurgie, AP-HP, Hôpital Pitié Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France; Inserm, UMR S975, Institut du Cerveau et de la Moelle Épinière, 75013 Paris, France; Université Paris 6 - Pierre-et-Marie-Curie, 75013 Paris, France.
| |
Collapse
|
8
|
Kaley TJ, Wen P, Schiff D, Ligon K, Haidar S, Karimi S, Lassman AB, Nolan CP, DeAngelis LM, Gavrilovic I, Norden A, Drappatz J, Lee EQ, Purow B, Plotkin SR, Batchelor T, Abrey LE, Omuro A. Phase II trial of sunitinib for recurrent and progressive atypical and anaplastic meningioma. Neuro Oncol 2014; 17:116-21. [PMID: 25100872 DOI: 10.1093/neuonc/nou148] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND No proven effective medical therapy for surgery and radiation-refractory meningiomas exists. Sunitinib malate (SU011248) is a small-molecule tyrosine kinase inhibitor that targets vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor, abundant in meningiomas. METHODS This was a prospective, multicenter, investigator-initiated single-arm phase II trial. The primary cohort enrolled patients with surgery and radiation-refractory recurrent World Health Organization (WHO) grades II-III meningioma. An exploratory cohort enrolled patients with WHO grade I meningioma, hemangiopericytoma, or hemangioblastoma. Sunitinib was administered at 50 mg/d for days 1-28 of every 42-day cycle. The primary endpoint was the rate of 6-month progression-free survival (PFS6), with secondary endpoints of radiographic response rate, safety, PFS, and overall survival. Exploratory objectives include analysis of tumoral molecular markers and MR perfusion imaging. RESULTS Thirty-six patients with high-grade meningioma (30 atypical and 6 anaplastic) were enrolled. Patients were heavily pretreated (median number of 5 recurrences, range 2-10). PFS6 rate was 42%, meeting the primary endpoint. Median PFS was 5.2 months (95% CI: 2.8-8.3 mo), and median overall survival was 24.6 months (95% CI: 16.5-38.4 mo). Thirteen patients enrolled in the exploratory cohort. Overall toxicity included 1 grade 5 intratumoral hemorrhage, 2 grade 3 and 1 grade 4 CNS/intratumoral hemorrhages, 1 grade 3 and 1 grade 4 thrombotic microangiopathy, and 1 grade 3 gastrointestinal perforation. Expression of VEGFR2 predicted PFS of a median of 1.4 months in VEGFR2-negative patients versus 6.4 months in VEGFR2-positive patients (P = .005). CONCLUSION Sunitinib is active in recurrent atypical/malignant meningioma patients. A randomized trial should be performed.
Collapse
Affiliation(s)
- Thomas J Kaley
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Patrick Wen
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - David Schiff
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Keith Ligon
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Sam Haidar
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Sasan Karimi
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Andrew B Lassman
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Craig P Nolan
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Lisa M DeAngelis
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Igor Gavrilovic
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Andrew Norden
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Jan Drappatz
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Eudocia Quant Lee
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Benjamin Purow
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Scott R Plotkin
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Tracy Batchelor
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Lauren E Abrey
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Antonio Omuro
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| |
Collapse
|
9
|
Abstract
Although advances in surgery, radiation therapy and stereotactic radiosurgery have significantly improved the treatment of meningiomas, there remains an important subset of patients who remain refractory to conventional therapy. Treatment with chemotherapeutic agents such as hydroxyurea and alpha-interferon has provided minimal benefit. In this review, the role of newly emerging novel therapies for meningiomas, with a focus on targeted molecular agents, will be discussed.
Collapse
Affiliation(s)
- Patrick Y Wen
- Center for Neuro-Oncology Dana-Farber/Brigham and Women's Cancer Center, and Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | | |
Collapse
|
10
|
Yew A, Trang A, Nagasawa DT, Spasic M, Choy W, Garcia HM, Yang I. Chromosomal alterations, prognostic factors, and targeted molecular therapies for malignant meningiomas. J Clin Neurosci 2013; 20:17-22. [DOI: 10.1016/j.jocn.2012.02.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 02/13/2012] [Indexed: 12/29/2022]
|
11
|
Pfister C, Pfrommer H, Tatagiba MS, Roser F. Vascular endothelial growth factor signals through platelet-derived growth factor receptor β in meningiomas in vitro. Br J Cancer 2012; 107:1702-13. [PMID: 23047550 PMCID: PMC3493872 DOI: 10.1038/bjc.2012.459] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background: Vascular endothelial growth factor (VEGF)-mediated angiogenesis mediates tumour growth and metastasis. Meningiomas are primarily benign, slow-growing, highly vascularised tumours. Aside from VEGF, there is little data on the function of major angiogenic proteins in meningiomas. Methods: The VEGFA, platelet-derived growth factor B (PDGFB), and their respective receptors – VEGF receptor 2 (KDR) and PDGF receptor β (PDGFRβ) – were quantified using real-time PCR and a TaqMan Protein Assay in meningiomas in vivo and in vitro. The effect of VEGFA and PDGFB on cell proliferation and the tyrosine phosphorylation of PDGFRβ were examined. Results: Most meningiomas displayed no KDR protein expression but elevated PDGFRβ levels. Exogenous VEGFA stimulation significantly increased cell proliferation. The PDGFRβ inhibition before stimulation with VEGFA abolished the proliferative stimuli. The VEGFA induced concentration-dependent PDGFRβ tyrosine phosphorylation comparable to PDGFB-induced PDGFRβ tyrosine phosphorylation. The PDGFRβ inhibitors gambogic acid, sunitinib, and tandutinib equally impaired the migration of meningioma cells. In addition, gambogic acid suppressed the VEGFA-induced PDGFRβ tyrosine phosphorylation. Conclusion: Collectively, our data suggest that VEGFA primarily regulates VEGF-mediated migration through PDGFRβ in meningiomas. The inhibitory effect of gambogic acid and tandutinib against meningioma growth in vitro suggests that selective PDGFRβ inhibitors, in combination with VEGF inhibitors, should be evaluated further as potential therapies for recurrent and malignant meningiomas.
Collapse
Affiliation(s)
- C Pfister
- Department of Neurosurgery, University of Tuebingen, Hoppe-Seyler-Strasse 3, Tuebingen 72076, Germany.
| | | | | | | |
Collapse
|
12
|
Horak P, Wöhrer A, Hassler M, Hainfellner J, Preusser M, Marosi C. Imatinib mesylate treatment of recurrent meningiomas in preselected patients: a retrospective analysis. J Neurooncol 2012; 109:323-30. [PMID: 22610940 DOI: 10.1007/s11060-012-0896-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 04/30/2012] [Indexed: 12/20/2022]
Abstract
Some unresectable and symptomatic meningiomas recur after conventional radiation therapy or stereotactic radiosurgery and are a therapeutic challenge. Evidence-based data from medical therapy for patients with recurrent meningioma can be deemed insufficient. Because of the prevalent expression of PDGF receptors in meningiomas, the tyrosine kinase imatinib mesylate has attracted interest as a treatment option for this patient group. In this retrospective study we analyzed 18 patients with recurrent meningiomas who were treated at our institution between 1996 and 2008. Nine patients with positive immunohistochemical staining of at least one of the PDGF receptors were given a daily oral dose of 400 mg imatinib mesylate as first, second, or third-line systemic therapy. Immunohistochemical staining was performed on formalin-fixed and paraffin-embedded tumor tissue with antibodies against PDGFR-α and β, c-Kit, Arg, and c-Abl. Imatinib mesylate at a dose of 400-800 mg/day was well tolerated. Of nine patients treated with imatinib, seven had stable disease and two had progressed at the first scan after three months. We observed no complete or partial responses, although prolonged disease stabilization with progression-free survival of 66.7 % at six months was observed. Overall median progression-free survival was 16 months. We conclude that single-agent imatinib mesylate might be a well-tolerated therapeutic option with high achievement of disease stabilization for preselected patients with recurrent meningiomas. Because of the small cohort, non-randomized design, and highly diverse patient population, we propose future prospective studies to validate our results.
Collapse
Affiliation(s)
- Peter Horak
- Department of Medicine I, Clinical Division of Oncology, and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | |
Collapse
|
13
|
Aberrant signaling pathways in meningiomas. J Neurooncol 2010; 99:315-24. [DOI: 10.1007/s11060-010-0381-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 08/20/2010] [Indexed: 12/24/2022]
|
14
|
Wen PY, Quant E, Drappatz J, Beroukhim R, Norden AD. Medical therapies for meningiomas. J Neurooncol 2010; 99:365-78. [PMID: 20820875 DOI: 10.1007/s11060-010-0349-8] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Accepted: 08/06/2010] [Indexed: 12/31/2022]
Abstract
Meningiomas are the most common primary brain tumor in adults. Although the majority of these tumors can be effectively treated with surgery and radiation therapy, an important subset of patients have inoperable tumors, or develop recurrent disease after surgery and radiotherapy, and require some form of medical therapy. There are increasing numbers of studies evaluating various medical therapies but the results remain disappointing. Chemotherapies and hormonal therapies have been generally ineffective, although somatostatin analogues may have therapeutic potential. There is also increasing interest in targeted molecular therapies. Agents inhibiting platelet derived growth factor receptors and epidermal growth factor receptors have shown little efficacy, but molecular agents inhibiting vascular endothelial growth factor receptors appear to have some promise. As with other tumors, advances in the medical therapies for meningiomas will require improved understanding of the molecular pathogenesis of these tumors, more predictive preclinical models, and efficient mechanisms for conducting clinical trials, given the small population of eligible patients.
Collapse
Affiliation(s)
- Patrick Y Wen
- Center for Neuro-Oncology, Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, MA 02115, USA.
| | | | | | | | | |
Collapse
|
15
|
Johnson MD, O'Connell MJ, Pilcher W, Reeder JE. Fibroblast growth factor receptor-3 expression in meningiomas with stimulation of proliferation by the phosphoinositide 3 kinase-Akt pathway. J Neurosurg 2010; 112:934-9. [PMID: 19698046 DOI: 10.3171/2009.7.jns09726] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECT Fibroblast growth factor receptors (FGFRs)-1, -2, and -3 are expressed in the developing brain and may participate in CNS neoplasia. Fibroblast growth receptor-3 has not been demonstrated in the human CNS or its tumors. Nonetheless, it has been implicated in the pathogenesis of several other forms of neoplasia. METHODS Twenty-four human meningiomas were evaluated using Western blot analysis for expression of FGFR3, its ligand acidic FGF, and concomitant phosphorylation/activation of p44/42 mitogen-activated protein kinase (MAPK), Akt, and STAT3. Mutations in exons 7 and 10 of the FGFR3 gene were analyzed by polymerase chain reaction in 10 meningiomas. Primary meningioma cells cultured from 10 human meningiomas were also treated with acidic FGF and evaluated for cell proliferation or activation/phosphorylation of p44/42 MAPK, Akt, and STAT3. RESULTS Immunoblotting demonstrated the presence of FGFR3 in 12 (71%) of 17 primarily fibroblastic and transitional WHO Grade I meningiomas. The FGFR3 was detected in 4 (80%) of 5 WHO Grade II, and 2 of 2 Grade III tumors. Acidic FGF was detected in 3 (18%) of 17 Grade I, 1 (20%) of 5 Grade II, and 1 (50%) of 2 Grade III meningiomas. In WHO Grade I meningiomas, 3 of 6 tumors with no detectable FGFR3 had no detectable p-STAT3. In WHO Grades II and III meningiomas, FGFR3 expression was associated with p-STAT3, p-Akt, and p-p44/42 MAPK expression. No mutations were demonstrated in exons 7 or 10 by polymerase chain reaction in any meningioma. Treatment with acidic FGF, a ligand for FGFR3, stimulated meningioma cell proliferation and activation of Akt and STAT3 in primary meningioma cell cultures. CONCLUSIONS These findings suggest that FGFR3 and acidic FGF are expressed in adult human leptomeninges as well as WHO Grades I and II meningiomas. Fibroblast growth factor receptor-3 activation stimulates meningioma cell proliferation by activation of the phosphoinositide 3 kinase-Akt-PRAS40-mTOR and STAT3 pathways.
Collapse
Affiliation(s)
- Mahlon D Johnson
- Department of Pathology and Laboratory Medicine, Division of Neuropathology, University of Rochester Medical Center, Rochester, New York 14623, USA.
| | | | | | | |
Collapse
|
16
|
Wen PY, Yung WKA, Lamborn KR, Norden AD, Cloughesy TF, Abrey LE, Fine HA, Chang SM, Robins HI, Fink K, Deangelis LM, Mehta M, Di Tomaso E, Drappatz J, Kesari S, Ligon KL, Aldape K, Jain RK, Stiles CD, Egorin MJ, Prados MD. Phase II study of imatinib mesylate for recurrent meningiomas (North American Brain Tumor Consortium study 01-08). Neuro Oncol 2010; 11:853-60. [PMID: 19293394 DOI: 10.1215/15228517-2009-010] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Platelet-derived growth factor (PDGF) and its receptors (PDGFR) are frequently coexpressed in meningiomas, potentially contributing to their pathogenesis. The North American Brain Tumor Consortium conducted a phase II study to evaluate the therapeutic potential of imatinib mesylate (Gleevec), a PDGFR inhibitor, in patients with recurrent meningiomas. Patients were stratified into benign (WHO grade I) meningiomas or atypical (WHO grade II) and malignant (WHO grade III) meningiomas. The primary end point was 6-month progression-free survival (6M-PFS). Patients requiring enzyme-inducing antiepileptic drugs were ineligible. Patients received imatinib at a dose of 600 mg/day for the first 4-week cycle and then gradually increased to 800 mg/day for subsequent cycles, if there were no unacceptable toxicities. Plasma concentrations of imatinib and its active metabolite, CGP74588, were assessed. Twenty-three heavily pretreated patients were enrolled into the study (13 benign, 5 atypical, and 5 malignant meningiomas), of whom 22 were eligible. The study was closed prematurely due to slow accrual. Tissue was available only from a minority of patients, but in these specimens there was uniform distribution of PDGFR, the drug target. Imatinib was generally well tolerated. Of 19 patients evaluable for response, 10 progressed at the first scan, and 9 were stable. There were no complete or partial responses. Overall median PFS was 2 months (range, 0.7-34 months); 6M-PFS was 29.4%. For benign meningiomas, median PFS was 3 months (range, 1.1-34 months); 6M-PFS was 45%. For atypical and malignant meningiomas, median PFS was 2 months (range, 0.7-3.7 months); 6M-PFS was 0%. Cycle 1 trough concentrations of imatinib and CGP74588 were 2,129 +/- 1,600 ng/ml and 517 +/- 326 ng/ml, respectively. Single-agent imatinib was well tolerated but had no significant activity in recurrent meningiomas. Trough plasma concentrations of imatinib exceeded those associated with imatinib activity in chronic myelogenous leukemia.
Collapse
Affiliation(s)
- Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, SW430D, 44 Binney St., Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Meningiomas are mostly benign tumours originating from the arachnoid cap cells, represent 13-26% of all intracranial tumours. They are more common in older age and in females. Deletion in NF2 gene and exposure to ionizing radiation are established risk factors, while the role of sex hormones is yet not clarified. Five-year survival for typical meningiomas exceeds 80%, but is poorer (5-year survival <60%) in malignant and atypical meningiomas. Papillary and haemangiopericytic morphology, large tumour size, high mitotic index, absence of progesterone receptors, deletions and loss of heterozygosity are poor prognostic factors. Complete surgical excision is the standard treatment. Radiotherapy is currently used in the clinical practice in atypical, malignant or recurrent meningioma at a total dose of 45-60Gy. However, the role of adjuvant irradiation is still controversial and has to be compared in a randomised prospective setting with a policy of watchful waiting. Radiosurgery has gained more and more importance in the management of meningiomas, especially in meningiomas that cannot be completely resected as for many skull base meningiomas. Medical therapy for patients with recurrent, progressive and symptomatic disease after repeated surgery, radiosurgery and radiotherapy is investigational. Hormonal therapy with progesterone antagonists has shown modest results, while chemotherapy with hydroxyurea appears moderately active.
Collapse
|
18
|
Abstract
✓ Although advances in surgery, radiation therapy, and stereotactic radiosurgery have significantly improved the treatment of meningiomas, there remains an important subset of patients whose tumors are refractory to conventional therapy. Treatment with traditional chemotherapeutic agents has provided minimal benefit. In this review, the role of targeted molecular therapies for recurrent or progressive meningiomas is discussed.
Collapse
|
19
|
Johnson MD, Sade B, Milano MT, Lee JH, Toms SA. New prospects for management and treatment of inoperable and recurrent skull base meningiomas. J Neurooncol 2007; 86:109-22. [PMID: 17624496 DOI: 10.1007/s11060-007-9434-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2007] [Accepted: 06/07/2007] [Indexed: 12/25/2022]
Abstract
Skull base, including optic nerve, cavernous sinus, clival and foramen magnum tumors represent a major challenge for neurosurgeons and neuro-oncologists. Growth regulatory signaling pathways for these tumors are of increasing interest as potential targets for new chemotherapy. Those differentially activated in various grades of meningiomas are currently being identified as well. This article reviews some recent findings pathways that appear to regulate meningioma growth. Potential targets for novel therapies are also discussed.
Collapse
Affiliation(s)
- Mahlon D Johnson
- Department of Pathology and Laboratory Medicine, Division of Neuropathology, University of Rochester Medical Center, 601 Elmwood Ave. Box 626, Rochester, NY 14623, USA.
| | | | | | | | | |
Collapse
|
20
|
Smith JS, Lal A, Harmon-Smith M, Bollen AW, McDermott MW. Association between absence of epidermal growth factor receptor immunoreactivity and poor prognosis in patients with atypical meningioma. J Neurosurg 2007; 106:1034-40. [PMID: 17564176 DOI: 10.3171/jns.2007.106.6.1034] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT The clinical behavior of meningiomas is variable. Because multiple growth factor receptors have been identified in these tumors, the authors sought to assess the capacity of the expression patterns of a subset of these receptors to stratify meningioma cases. METHODS Eighty-four meningiomas were analyzed, including 36 benign, 29 atypical, and 19 malignant lesions. Immunohistochemical staining was performed for epidermal growth factor receptor (EGFR), platelet-derived growth factor receptor (PDGFR)-beta, basic fibroblast growth factor receptor (BFGFR), and MIB-1. Survival analyses were performed using follow-up data obtained in patients with newly diagnosed tumors. Immunoreactivity for EGFR was observed in 47% of benign, 48% of atypical, and 42% of malignant tumors. Staining for BFGFR was identified in 89% of benign, 97% of atypical, and 95% of malignant lesions. Immunostaining for PDGFR-beta was evident in all the lesions assessed. Mean MIB-I indices for benign, atypical, and malignant cases were 3.6 (range 0.5-15.3), 8.2 (range 1.5-23.1) and 18.3 (range 1.0-55.8), respectively. Overall mean follow-up duration was 9.0 years (range 5.1-18.8 years). Lack of EGFR immunoreactivity was identified as a strong predictor of shorter overall survival in patients with atypical meningioma (p = 0.003, log-rank test). This association was not evident in cases of benign or malignant meningiomas. CONCLUSIONS There is a significant association between EGFR immunoreactivity and prolonged survival in patients with atypical meningioma. Given the variable behavior of atypical meningiomas, EGFR assessment could improve existing strategies for patient stratification and treatment.
Collapse
Affiliation(s)
- Justin S Smith
- Department of Neurological Surgery, Brain Tumor Research Center, University of California, San Francisco, California 94143-0112, USA.
| | | | | | | | | |
Collapse
|
21
|
Andrae J, Afink G, Zhang XQ, Wurst W, Nistér M. Forced expression of platelet-derived growth factor B in the mouse cerebellar primordium changes cell migration during midline fusion and causes cerebellar ectopia. Mol Cell Neurosci 2004; 26:308-21. [PMID: 15207855 DOI: 10.1016/j.mcn.2004.02.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2003] [Revised: 02/02/2004] [Accepted: 02/11/2004] [Indexed: 11/22/2022] Open
Abstract
The platelet-derived growth factor (PDGF) and receptors are expressed in the developing central nervous system and in brain tumors. To investigate the role of PDGF during normal cerebellar development, we created transgenic mice where PDGF-B was introduced into the endogenous Engrailed1 locus (En1). These mice expressed PDGF-B in all types of cells that constitute the developing cerebellum, with localized high expression in the ventral midline of the cerebellar anlage. This affected cell migration in the midline during fusion of the cerebellar anlage and caused misplacement of midline structures. PDGFR-alpha- and laminin alpha1-positive meningeal cells migrated inwards, attracted by the ectopic transgene expression in the ventral neuroepithelium. Other cells followed the meningeal cells and in the adult mouse, cells from all cortical cell layers were found misplaced in the midline. Moreover, the transgene caused an enhancement of capillary vessels. The findings indicate that normal PDGF signaling is important for proper neural tube fusion. It also illustrates that meningeal structures can influence the process.
Collapse
Affiliation(s)
- Johanna Andrae
- Department of Genetics and Pathology, Uppsala University, Rudbeck Laboratory, SE-751 85 Uppsala, Sweden
| | | | | | | | | |
Collapse
|
22
|
Abstract
BACKGROUND Meningiomas are biologically complex and clinically and surgically challenging. These features, combined with the rewarding potential for cure, make them of great interest to neurologists, neurosurgeons, and neuroscientists alike. REVIEW SUMMARY Initially, we review the clinical context of meningiomas, particularly recent changes in histopathological classification, diagnosis, and neuroimaging. Secondly, the underlying basic science as it has evolved over the last decades is summarized. The status of areas recently of intense interest, such as steroid hormone receptors and oncogenic viruses is described. Additionally, emerging areas of great promise, such as cytogenetics and molecular biology are presented. Lastly, we describe recent advances in management. In particular, skull-base surgery, image-guided surgery, and advances in radiotherapy are emphasized. The possible impact of basic research on management and outcome is also outlined. CONCLUSIONS Although usually benign and amenable to cure, meningiomas still present significant diagnostic and treatment challenges. Advances in basic science, surgery, and adjuvant therapy are widening the potential for safe, effective, evidence-based management leading to even better outcomes
Collapse
Affiliation(s)
- Katharine J Drummond
- Department of Neurosurgery, The Brigham and Women's Hospital, Boston, Massachusetts, USA.
| | | | | |
Collapse
|
23
|
Johnson MD, Okedli E, Woodard A, Toms SA, Allen GS. Evidence for phosphatidylinositol 3-kinase—Akt—p70S6K pathway activation and transduction of mitogenic signals by platelet-derived growth factor in human meningioma cells. J Neurosurg 2002; 97:668-75. [PMID: 12296652 DOI: 10.3171/jns.2002.97.3.0668] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Object. The intracellular events transducing mitogenic signals from platelet-derived growth factor—β (PDGFβ) receptor tyrosine kinases are not precisely known. In this study the authors evaluated whether the phosphatidylinositol 3-kinase (PI3-K)—Akt—p70S6K pathway is expressed in meningiomas, regulates their growth, and transduces mitogenic signals of PDGF-BB.
Methods. Nine meningioma tumors obtained in humans were evaluated using Western blot analysis for phosphorylated (activated) Akt and phosphorylated p70S6K. Cells cultured from seven of these meningiomas were also screened using Western blot analysis for Akt and for phosphorylated Akt and p70S6K. The authors also evaluated whether PDGF-BB stimulation of meningioma cells was associated with the phosphorylation of Akt and p70S6K known to activate these kinases. In addition, the effects of wortmannin, an inhibitor of PI3-K, on proliferation and activation of Akt and p70S6K in meningioma cells stimulated with PDGF-BB were evaluated.
Western blots of lysates from meningiomas demonstrated phosphorylated Akt and p70S6K. Treatment with PDGF-BB stimulated phosphorylation of Akt and p70S6K in each meningioma cell culture. Wortmannin (500 and 1000 nM) significantly decreased PDGF-BB stimulation of meningioma cells (p < 0.001) while it reduced Akt and p70S6K phosphorylation but not mitogen-activated protein kinase/extracellular signal—regulated kinase (MAPK/ERK) phosphorylation.
Conclusions. These findings indicate that Akt and p70S6K are constitutively expressed and activated in meningioma cells and that the PI3-K—Akt—p70S6K pathway may participate in transduction of mitogenic signals in meningiomas independent of the Raf-1—MEK-1—MAPK/ERK cascade.
Collapse
Affiliation(s)
- Mahlon D Johnson
- Department of Pathology, Vanderbilt Medical School, Nashville, Tennessee 37232, USA.
| | | | | | | | | |
Collapse
|
24
|
Johnson MD, Woodard A, Okediji EJ, Toms SA, Allen GS. Lovastatin is a potent inhibitor of meningioma cell proliferation: evidence for inhibition of a mitogen associated protein kinase. J Neurooncol 2002; 56:133-42. [PMID: 11995814 DOI: 10.1023/a:1014588214966] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lovastatin inhibits 3-hydroxy 3-methylglutaryl coenzyme A (HMG-CoA) reductase the rate limiting enzyme for synthesis of mevalonic acid, a precursor for cholesterol, farnesyl and geranylgeranyl pyrophosphate isoprenoids. Recent studies suggest it also has growth inhibitory properties. Posttranslational farnesyl or geranylgeranylation of low molecular weight GTP-binding proteins such as RAS and RHO are thought to be an essential step in activation of phosphorylation cascades such as the RAS-RAF-1-MEK-1-MAPK/ERK pathway which stimulate cell proliferation. In this study, we evaluated lovastatin effects on meningioma cell proliferation and activation of the MEK-1-MAPK/ERK pathway. The effect of lovastatin on cell proliferation was assessed in eight human meningioma cell cultures stimulated by platelet derived growth factor (PDGF)-BB, cerebrospinal fluid (CSF), and fetal bovine serum (FBS). Concomitant lovastatin effects on phosphorylation/activation of mitogen-activated protein kinase/extracellular signal regulated kinase (MAPK/ERK) kinase (MEK-1) and MAPK/ERK were assessed by Western blot. Whether lovastatin acts via a mevalonate-dependent mechanism was also evaluated. Coadministration of lovastatin completely blocked PDGF-BB, CSF, and FBS stimulation of [3H]-thymidine incorporation and cell proliferation. Lovastatin inhibited PDGF-BB's stimulatory effect in a dose dependent manner. Concomitant with its growth inhibitory effects, lovastatin reduced phosphorylation/activation of MEK-1/2 in five meningiomas and MAPK/ERK in seven. Coadministration of mevalonate with lovastatin partially restored PDGF's mitogenic effect. Lovastatin is a potent inhibitor of meningioma cell proliferation which may act in part by reducing activation of MEK-1-MAPK/ERK pathway. Additional studies are warranted to assess whether lovastatin and similar HMG-CoA reductase inhibitors represent a new adjunctive chemotherapy for recurrent meningiomas.
Collapse
Affiliation(s)
- Mahlon D Johnson
- Department of Pathology, Vanderbilt Medical School, Nashville Veterans Administration Medical Center, TN 37212, USA.
| | | | | | | | | |
Collapse
|
25
|
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 2001; 8 Suppl 1:49-53. [PMID: 11386826 DOI: 10.1054/jocn.2001.0877] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In this study, we detected the expression of platelet-derived growth factor (PDGF) and its receptor in 61 human meningiomas by immunohistochemistry and in situ hybridisation. The results showed that almost all the 61 meningiomas expressed PDGFBB and PDGF beta receptor and the positive rate of PDGFAA was 49%. Only two meningiomas expressed PDGF alpha receptor. The positive rate and the immunostaining intensity of PDGFBB and PDGF beta receptor were higher in atypical meningiomas than in benign types. There was no significant difference between the different types of benign meningiomas. The expression of PDGFB chain mRNA was coincident with that of PDGFB chain protein. There was no correlation between the expression of PDGFAA and the types or grades of meningiomas. The correlation between overexpression of PDGFBB/R beta and tumour grade provides a useful parameter in evaluating the prognosis of patients with meningiomas. The proliferative activity of meningiomas was evaluated by the proliferating cell nuclear antigen labelling index (PCNA LI). In the 61 meningiomas, the average PCNA LI (%) was 1.8+/-1.3, 1.9+/-1.3, 1.7+/-0.8 and 11.6+/-5.3 (in fibrous, meningothelial, transitional and atypical meningiomas, respectively). Statistic analysis shows that the PCNA LI is higher in atypical meningiomas than that in benign types, and there was no significant difference between the different types of benign meningiomas. The expression of PDGFBB and PDGF beta receptor was significant enhanced in ascending order from low PCNA LI meningiomas to high ones. This result suggested that PDGFBB/R beta autocrine loop may stimulate the growth of meningiomas. In this study, we also detected the cell apoptosis of meningiomas by terminal deoxynucleotidyl transferase mediated dUTP nick end labelling (TUNEL) method. The average apoptosis labelling index (%) was 0.11+/-0.05, 0.08+/-0.04, 0.09+/-0.03 and 0.35+/-0.15 in fibrous, meningothelial, transitional and atypical meningiomas respectively. The apoptosis labelling index was higher in atypical meningiomas than that in benign types. There was a positive correlation between the apoptosis labelling index and PC NA LI of meningioma. When the positive rate of PDGFBB and/or PDGF beta receptor was higher in meningioma, the apoptotic cells was also increased. In conclusion, the overexpression of PDGFBB and its relevant receptor PDGFR beta in meningiomas was correlated with grade of meningiomas and the proliferative activity of meningiomas; PDGFBB/R beta autocrine loop may play an critical role in the pathogenesis of meningiomas.
Collapse
Affiliation(s)
- S Y Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
| | | |
Collapse
|
26
|
Johnson MD, Woodard A, Kim P, Frexes-Steed M. Evidence for mitogen-associated protein kinase activation and transduction of mitogenic signals by platelet-derived growth factor in human meningioma cells. J Neurosurg 2001; 94:293-300. [PMID: 11213968 DOI: 10.3171/jns.2001.94.2.0293] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Coexpression of platelet-derived growth factor (PDGF)-BB and activated PDGF-beta receptor in meningioma cells indicates that this cytokine may act as an autocrine or paracrine stimulant of meningioma growth. The intracellular events transducing signals from PDGF-beta receptor tyrosine kinases are unknown. In this study the authors evaluated whether or not mitogen-activated protein kinases (MAPKs) are expressed in meningiomas, regulate their growth, and transduce mitogenic signals of PDGF-BB. METHODS Ten human meningioma tumors as well as cells cultured from two normal leptomeninges and 10 additional human meningiomas were evaluated using Western blot analysis to determine the presence of MAPK and phosphorylated (activated) MAPK. The effects of PD098059, a selective inhibitor of MAPK phosphorylation/activation, on proliferation of meningioma cells stimulated with 10% fetal bovine serum was also evaluated. Last, the authors evaluated whether PDGF-BB stimulation of meningioma cells was associated with activation of MAPK. Western blots of lysates from meningiomas and from cultured leptomeningeal and meningioma cells demonstrated MAPK and phosphorylated MAPK. Treatment with PD098059 produced a 52 to 84% (x = 69.8) loss in [3H]thymidine incorporation, which was associated with a partial or complete loss of phosphorylated MAPK after 3 days of treatment. The PDGF-BB produced a significant increase in [3H]thymidine incorporation and phosphorylation of MAPK at 1 and 3 days. Coadministration of PD098059 completely blocked PDGF-BB's stimulation of [3H]thymidine incorporation and cell proliferation concomitant with reduced MAPK phosphorylation. CONCLUSIONS The findings indicate that MAPK is constitutively expressed in leptomeningeal and meningioma cells and transduces mitogenic signals of PDGF, contributing to the growth of human meningiomas.
Collapse
Affiliation(s)
- M D Johnson
- Department of Pathology, Veterans Administration Medical Center and Vanderbilt Medical School, Nashville, Tennessee 37212, USA.
| | | | | | | |
Collapse
|
27
|
Pallini R, Casalbore P, Mercanti D, Maggiano N, Larocca LM. Phenotypic change of human cultured meningioma cells. J Neurooncol 2000; 49:9-17. [PMID: 11131990 DOI: 10.1023/a:1006436903976] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
One objection to using cell cultures for studying the proliferation of tumors is the potential for phenotypic changes that may occur in vitro. Here, we compared the antigen pattern expression of cultured meningioma cells with that of the primary tumor. Cell cultures established from 9 intracranial meningiomas and deparaffinized sections of the resected tumors were analyzed for immunophenotyping with the following antibodies: vimentin, cytokeratin, epithelial membrane antigen, S-100, neuron-specific enolase, synaptophisin, factor VIII-related antigen, CD4, CD31, CD34, CD45RB, CD68-PGM1, CD68-KP, and myeloid/histiocyte antigen (MAC387). Overall, the cultured meningioma cells retained the main feature of the primary tumor, being positive both for mesenchymal antigens and for epithelial antigens. Interestingly, the cultured meningioma cells abundantly expressed the CD68 antigens at early passage. The CD68 antigens, which are normally found on hematopoietic cells like macrophages and monocytes, were not detectable on meningioma cells in situ. Our results show that phenotypic changes on human meningioma cells may occur in vitro. This phenomenon suggests caution when transposing the in vitro results to the in vivo condition.
Collapse
Affiliation(s)
- R Pallini
- Institute of Neurosurgery, CNR, Rome, Italy.
| | | | | | | | | |
Collapse
|
28
|
Jensen RL, Petr M, Wurster RD. Calcium channel antagonist effect on in vitro meningioma signal transduction pathways after growth factor stimulation. Neurosurgery 2000; 46:692-702; discussion 702-3. [PMID: 10719866 DOI: 10.1097/00006123-200003000-00032] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE We have previously demonstrated that calcium channel antagonists inhibit the growth of human meningiomas in culture after stimulation with growth factors. This study examined the effects of these drugs on signaling transduction pathways in an attempt to elucidate potential mechanisms by which this growth inhibition is mediated. METHODS Primary cell cultures from patients with intracranial meningiomas were established. Cell growth studies were performed with inhibitors and stimulators of tyrosine kinase signal transduction. Intracellular calcium changes and inositol phosphate production were measured after growth factor exposure, with or without pretreatment by calcium channel antagonists. RESULTS The growth of meningiomas in culture can be inhibited by tyrosine kinase receptor inhibitors. Inhibitors and stimulators of phospholipase C can stimulate or inhibit the growth of in vitro meningiomas, respectively. Calcium channel antagonists inhibit intracellular calcium changes induced by serum and epidermal growth factor. Inositol phosphate production is increased after growth factor stimulation, and calcium channel antagonists potentiate this effect. CONCLUSION Calcium channel antagonists interfere with intracellular signaling pathways of cultured meningioma cells. This inhibition is unrelated to voltage-sensitive calcium channels. The findings of this project may aid in the understanding of the signal transduction mechanisms involved in growth factor-mediated meningioma proliferation and may lead to clinically relevant strategies for growth inhibition.
Collapse
Affiliation(s)
- R L Jensen
- Department of Neurosurgery, University of Utah, Salt Lake City 84112, USA
| | | | | |
Collapse
|
29
|
Schrell UM, Koch HU, Marschalek R, Schrauzer T, Anders M, Adams E, Fahlbusch R. Formation of autocrine loops in human cerebral meningioma tissue by leukemia inhibitor factor, interleukin-6, and oncostatin M: inhibition of meningioma cell growth in vitro by recombinant oncostatin M. J Neurosurg 1998; 88:541-8. [PMID: 9488310 DOI: 10.3171/jns.1998.88.3.0541] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECT It has been demonstrated that growth of cerebral meningiomas found in humans is controlled by a variety of factors, including growth factors, aminergic agents, neuropeptides, and steroids. To further our knowledge of this process, the authors investigated the presence and function of the cytokines leukemia inhibitory factor (LIF), interleukin-6 (IL-6), and oncostatin M (OSM) on meningioma cell proliferation. METHODS Active transcription of LIF, IL-6, and OSM, their related receptors (LIF-R, IL-6-R, and gp130), and the consecutive signal-transducing molecules (STAT 1, STAT 3, and STAT 5a) were analyzed in reverse transcriptase-polymerase chain reaction experiments. The presence of endogenous LIF, IL-6, and OSM proteins was demonstrated in the supernatant of cultured meningioma cells using the enzyme-linked immunosorbent assay and Western blot experiments, thus indicating an autocrine signaling pathway for all three cytokines. The biological function of all three cytokines was evaluated by studying their effects on meningioma cell growth. Recombinant LIF and IL-6 showed no significant growth modulating effects; however, recombinant OSM decreased meningioma cell growth by 66%. The antiproliferative potency of OSM was demonstrated by cell count experiments, the [3H]thymidine incorporation assay, and cell cycle analysis. CONCLUSIONS These in vitro data support the concept that growth of meningioma cells may be modulated by cytokines, and they also indicate that recombinant OSM may be one future candidate for use in the adjuvant treatment of inoperable and recurrent meningiomas.
Collapse
Affiliation(s)
- U M Schrell
- Department of Neurosurgery, University of Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | | | | | |
Collapse
|
30
|
Maxwell M, Shih SD, Galanopoulos T, Hedley-Whyte ET, Cosgrove GR. Familial meningioma: analysis of expression of neurofibromatosis 2 protein Merlin. Report of two cases. J Neurosurg 1998; 88:562-9. [PMID: 9488313 DOI: 10.3171/jns.1998.88.3.0562] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Meningiomas are primarily benign brain tumors thought to arise through multistep tumorigenesis, involving both the activation of oncogenes and the loss of tumor suppressor genes. The recently isolated neurofibromatosis 2 (NF2) tumor suppressor gene has been found to be mutated in a large proportion of meningiomas. Almost all cases of familial meningioma occur in association with NF2. Familial meningioma in isolation from NF2 (sporadic) is exceedingly rare, with only 14 reports since 1959. The authors report the existence of a family lacking any stigmata of NF2, in which two members had spinal meningiomas. Tumor specimens were subjected to immunocytochemical analysis for the NF2 protein product Merlin, which has been implicated in the tumorigenesis of meningioma. Merlin immunoreactivity was present in both tumor specimens, implying that the NF2 tumor suppressor gene was not deleted in these tumors. This supports the hypothesis that a second tumor suppressor gene locus, other than NF2, acts in the formation of familial sporadic meningioma. The results are discussed in the context of putative oncogenic mechanisms of familial meningiomas.
Collapse
Affiliation(s)
- M Maxwell
- Department of Neuropathology, Massachusetts General Hospital, Harvard Medical School, Boston 02114-2696, USA.
| | | | | | | | | |
Collapse
|
31
|
Nakayama Y, Sueishi K, Oka K, Kono S, Tomonaga M. Stromal angiogenesis in human glioma: a role of platelet-derived endothelial cell growth factor. SURGICAL NEUROLOGY 1998; 49:181-7; discussion 187-8. [PMID: 9457269 DOI: 10.1016/s0090-3019(97)00038-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Although several tumor angiogenic factors have been identified previously and characterized, it is not yet fully clear how tumor angiogenic factors induce endothelial cell transformation and proliferation. Platelet-derived endothelial cell growth factor (PD-ECGF) has been recently discovered to be an endothelial cell growth factor initially purified from human platelets. However, there has been no previous report describing the significance of PD-ECGF in the growth of brain tumors by angiogenic stimulation. We report the immunohistochemical localization of PD-ECGF in human gliomas and meningiomas, and discuss whether PD-ECGF could play a role in the modulation of stromal angiogenesis in human glioblastoma multiforme. METHODS Twenty-eight cases of glioma (11 glioblastomas and 17 astrocytomas) derived from the neuroectoderm in embryogenesis and 12 meningiomas from the mesoderm were investigated by both immunohistochemical localization of the PD-ECGF and a semiquantitative assay to determine the degree of stromal angiogenesis. RESULTS Numerous PD-ECGF positive cells were observed within and around the blood vessels of glioblastoma multiforme, especially on the borders of tumor tissue. The PD-ECGF positive cells were negative for anti-von Willebrand factor (vWF) and antiglial fibrillary acidic protein (GFAP) antibodies and were positive for antimacrophage (HAM-56). The expression of PD-ECGF by macrophages closely correlated with the degree of stromal vascularity in glioblastoma multiforme; no such correlation was found in either astrocytoma or meningioma. Proliferating cell nuclear antigen (PCNA) was found to be positive in some endothelial cells of stromal vessels in glioblastoma multiforme. These findings suggest that PD-ECGF expressed by macrophages plays an important role in the growth of glioblastoma multiforme with stromal angiogenesis.
Collapse
Affiliation(s)
- Y Nakayama
- Department of Neurosurgery, School of Medicine, Fukuoka University, Japan
| | | | | | | | | |
Collapse
|
32
|
Abstract
This paper initially reviews ligand and receptor systems for the PDGF family and the signalling systems they use as well as their role in neural developments. It then describes the putative role of this family in astrocytoma, meningioma, and pituitary adenoma pathogenesis. Potential therapies with receptor antagonists or dominant negative mutants are discussed in the final sections.
Collapse
Affiliation(s)
- M Kirsch
- Neurosurgical Laboratories, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | | |
Collapse
|
33
|
Schrell UM, Rittig MG, Anders M, Kiesewetter F, Marschalek R, Koch UH, Fahlbusch R. Hydroxyurea for treatment of unresectable and recurrent meningiomas. I. Inhibition of primary human meningioma cells in culture and in meningioma transplants by induction of the apoptotic pathway. J Neurosurg 1997; 86:845-52. [PMID: 9126901 DOI: 10.3171/jns.1997.86.5.0845] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Meningiomas, which invade intracranial bone structures and the adjacent connective tissue, are frequently unresectable because of their aggressive and recalcitrant growth behavior. They have a high recurrence rate, and in approximately 10% of these tumors there is an increased risk of malignancy. Significant morbidity and mortality rates associated with recurrent meningiomas demand nonsurgical approaches. To date, adjuvant hormonal treatment has not proven beneficial. The anticancer drug hydroxyurea was therefore tested for its potential use in the treatment of meningiomas. Early-passaged cell cultures were established from 20 different meningiomas. The addition of 5 x 10(-4) and 10(-3) M hydroxyurea over a period of 5 to 9 days resulted in a remarkable decrease in cell proliferation and even blocked tumor cell growth when compared with untreated cells. A significant arrest of meningioma cell growth in the S phase of the cell cycle was revealed on DNA flow cytometry. Electron micrographs of hydroxyurea-treated tumor cells showed ultrastructural features consistent with apoptosis, and light microscopy demonstrated DNA fragmentation by in situ DNA strand break labeling. Short-term treatment of meningioma cell cultures with hydroxyurea for 24 to 48 hours resulted in discrete oligonucleosomal fragments (DNA ladder), another characteristic sign of apoptosis. In addition to the in vitro studies, tissue from five different meningiomas was transplanted into nude mice followed by treatment with 0.5 mg/g body weight hydroxyurea over 15 days. In situ DNA strand break labeling demonstrated DNA fragmentation in distinct regions with different tumor cell densities in all hydroxyurea-treated meningioma transplants. These data provide evidence that hydroxyurea is a powerful inhibitor of meningioma cell growth, most likely by causing apoptosis in the tumor cells. Thus, hydroxyurea may be a suitable chemotherapeutic agent for the long-term treatment of unresectable or semi- to malignant meningiomas, or for preventing recurrent growth of meningiomas after resection.
Collapse
Affiliation(s)
- U M Schrell
- Department of Neurosurgery, University of Erlangen-Nürnberg, Germany
| | | | | | | | | | | | | |
Collapse
|
34
|
Schrell UMH, Koch U, Marschalek R, Schrauzer T, Anders M, Adams E, Fahlbusch R. Formation of autocrine loops in human cerebral meningioma tissue by leukemia inhibitor factor, interleukin-6, and oncostatin M: inhibition of meningioma cell growth in vitro by recombinant oncostatin M. Neurosurg Focus 1997. [DOI: 10.3171/foc.1997.2.4.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
It has been demonstrated that growth of cerebral meningiomas found in humans is controlled by a variety of factors, including growth factors, aminergic agents, neuropeptides, and steroids. The authors investigated the presence and function of the cytokines leukemia inhibitory factor (LIF), interleukin-6 (IL-6), and oncostatin M (OSM) on meningioma cell proliferation.
Active transcription of LIF, IL-6, OSM, their related receptors (LIF-R, IL-6-R, gp130), and the consecutive signal-transducing molecules (STAT 1, STAT 3, and STAT 5a) were analyzed in reverse transcriptase-polymerase chain reaction experiments.
The presence of endogenous LIF, IL-6, and OSM proteins was demonstrated in the supernatant of cultured meningioma cells using enzyme-linked immunosorbent assay and Western blot experiments, thus indicating an autocrine signaling pathway for all three cytokines.
The biological function of all three cytokines was evaluated by studying their effects on meningioma cell growth. Recombinant LIF and IL-6 showed no significant growth modulating effects; however, recombinant OSM decreased meningioma cell growth by 66%. The antiproliferative potency of OSM was demonstrated by cell count experiments, [3H]thymidine incorporation assay, and cell cycle analysis. These in vitro data support the concept that growth of meningioma cells may be modulated by cytokines and also indicates that recombinant OSM may be one of the future candidates for use in the adjuvant treatment of inoperable and recurrent meningiomas.
Collapse
|
35
|
Schrell UMH, Rittig MG, Anders M, Kiesewetter F, Marschalek R, Koch UH, Fahlbusch R. Hydroxyurea for treatment of unresectable and recurrent meningiomas. I. Inhibition of primary human meningioma cells in culture and in meningioma transplants by induction of the apoptotic pathway. Neurosurg Focus 1997. [DOI: 10.3171/foc.1997.2.4.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Meningiomas, which invade intracranial bone structures and the adjacent connective tissue, are frequently unresectable because of their aggressive and recalcitrant growth behavior. They have a high recurrence rate, and in approximately 10% of these tumors there is an increased risk of malignancy. Significant morbidity and mortality rates associated with recurrent meningiomas demand nonsurgical approaches. To date, adjuvant hormonal treatment has not proven beneficial. The anticancer drug hydroxyurea was therefore tested for its potential use in the treatment of meningiomas.
Early-passaged cell cultures were established from 20 different meningiomas. The addition of 5 X 10−4 and 10−3 M hydroxyurea over a period of 5 to 9 days resulted in a remarkable decrease in cell proliferation and even blocked tumor cell growth when compared with untreated cells. A significant arrest of meningioma cell growth in the S phase of the cell cycle was revealed on DNA flow cytometry.
Electron micrographs of hydroxyurea-treated tumor cells showed ultrastructural features consistent with apoptosis, and light microscopy demonstrated DNA fragmentation by in situ DNA strand break labeling. Short-term treatment of meningioma cell cultures with hydroxyurea for 24 to 48 hours resulted in discrete oligonucleosomal fragments (DNA ladder), another characteristic sign of apoptosis. In addition to the in vitro studies, tissue from five different meningiomas was transplanted into nude mice followed by treatment with 0.5 mg/g body weight hydroxyurea over 15 days. In situ DNA strand break labeling demonstrated DNA fragmentation in distinct regions with different tumor cell densities in all hydroxyurea-treated meningioma transplants.
These data provide evidence that hydroxyurea is a powerful inhibitor of meningioma cell growth, most likely by causing apoptosis in the tumor cells. Thus, hydroxyurea may be a suitable chemotherapeutic agent for the long-term treatment of unresectable or semi- to malignant meningiomas, or for preventing recurrent growth of meningiomas after resection.
Collapse
|
36
|
Todo T, Adams EF, Fahlbusch R, Dingermann T, Werner H. Autocrine growth stimulation of human meningioma cells by platelet-derived growth factor. J Neurosurg 1996; 84:852-8; discussion 858-9. [PMID: 8622161 DOI: 10.3171/jns.1996.84.5.0852] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The authors have previously shown that meningioma-derived conditioned medium profoundly stimulates the in vitro proliferation of meningioma cells. In this paper, self mitogenic agents found in the conditioned medium-autocrine growth-stimulatory factors actually secreted by human meningioma cells-are characterized as proteins related to the B chain of platelet-derived growth factor (PDGF) and possibly to the A chain of PDGF as well. The addition to conditioned medium of a neutralizing antibody against PDGF-BB caused a significant inhibition of the conditioned medium-stimulated DNA synthesis in all three meningioma cultures studied. A similar neutralizing effect was observed with an anti-PDGF-AA antibody in one meningioma culture studied. Gel filtration chromatography of concentrated conditioned medium from two different meningiomas using a Sephadex G-100 column revealed similar profiles from both conditioned media with a major peak of mitogenic activity against meningioma cells at a molecular weight (M(r)) of approximately 32 to 36 kD, accompanied by a minor peak at approximately 22 kD. The major peak mitogenic activity was significantly reduced by addition of an anti-PDGF-BB antibody. Western blot analysis of protein extracts from five meningioma specimens was performed using a monoclonal antibody against the B chain of PGDF, and a major band of PDGF-B immunoreactivity was detected at an M(r) of approximately 19 kD in all five meningiomas under both reducing and nonreducing conditions. Exogenous human and porcine PDGFs both exhibited a significant dose-dependent stimulation of DNA synthesis in two of three and three of five meningioma cultures examined, respectively. Although not all meningiomas investigated proved to share the biological activity associated with PDGF and these results may be preliminary, it seems that the autocrine growth-stimulatory loop established by PDGF-B-related molecules plays an important functional role in meningioma cell proliferation.
Collapse
Affiliation(s)
- T Todo
- Department of Neurosurgery, University of Tokyo, Japan
| | | | | | | | | |
Collapse
|
37
|
Jensen RL, Lee YS, Guijrati M, Origitano TC, Wurster RD, Reichman OH. Inhibition of in vitro meningioma proliferation after growth factor stimulation by calcium channel antagonists: Part II--Additional growth factors, growth factor receptor immunohistochemistry, and intracellular calcium measurements. Neurosurgery 1995; 37:937-46; discussion 946-7. [PMID: 8559343 DOI: 10.1227/00006123-199511000-00013] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
We have previously reported that calcium channel antagonists can block both the growth of meningiomas in culture and the potent growth stimulation of meningioma cells by epidermal growth factor (EGF) and platelet-derived growth factor (PDGF). This study further defines the nature of this growth inhibition. Primary meningioma cultures were established, and cells were characterized. Fibroblast growth factor or insulin-like growth factor-I growth stimulation in the presence of calcium channel antagonists was examined. In addition, the effects of ethylene glycol-bis-(aminoethylether) N,N,N',N"-tetraacetic acid and Bay K 8644, a calcium channel agonist, on the growth factors were analyzed. Growth factor receptor immunohistochemistry was performed on the original tumors and the in vitro meningioma cells. Twelve of 17 (71%) meningiomas in this study were positive for the EGF receptor, and 14 of 17 (82%) were positive for the PDGF receptor. Five of six (83%) of the culture cells were positive for the EGF receptor, and four of five (80%) were positive for the PDGF receptor. Intracellular calcium changes were quantified using the intracellular calcium-chelating, fluorescent dye, Fura-2. The growth stimulation of fibroblast growth factor and insulin-like growth factor-I on meningioma cells in culture was decreased in a dose-dependent manner by calcium channel antagonists. The growth stimulation of fibroblast growth factor and insulin-like growth factor-I was not affected by a reduction of extracellular calcium, whereas the growth stimulation of EGF and PDGF was. Interestingly, intracellular calcium was not increased after exposure to growth factors but was increased after serum stimulation. This increase could be blocked by preincubation with verapamil. Calcium channel antagonists can inhibit proliferation of meningioma cells in culture after stimulation with a number of growth factors. These drugs might disrupt intracellular calcium homeostasis or interfere with key elements of the growth factor signal transduction pathways. These mechanisms as well as the potential clinical relevance of these findings are discussed.
Collapse
Affiliation(s)
- R L Jensen
- Department of Neurological Surgery, Loyola University, Maywood, Illinois, USA
| | | | | | | | | | | |
Collapse
|
38
|
Inhibition of In Vitro Meningioma Proliferation after Growth Factor Stimulation by Calcium Channel Antagonists. Neurosurgery 1995. [DOI: 10.1097/00006123-199511000-00013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
39
|
Zhao JF, Sharma S. Expression of the ROS1 oncogene for tyrosine receptor kinase in adult human meningiomas. CANCER GENETICS AND CYTOGENETICS 1995; 83:148-54. [PMID: 7553586 DOI: 10.1016/0165-4608(95)00043-o] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Oncogenes have been implicated in the promotion and progression of cancer in humans. Expression of the ROS1 oncogene, a member of the receptor tyrosine kinase superfamily, was examined in human meningiomas by coupled reverse transcription and polymerase chain reaction (RT-PCR) assays. Two sets of region-specific oligonucleotides, specific for different regions of the ROS1 messenger ribonucleic acid (mRNA), were used in RT-PCR assays to independently examine ROS1 transcripts from primary human meningiomas. ROS1 was expressed at high levels in approximately 55% (17 of 31) of the meningiomas examined, but not expressed in non-neoplastic brain samples. The commonplace expression of the ROS1 oncogene in meningiomas suggests a role for this oncogene in the etiology of these tumors.
Collapse
Affiliation(s)
- J F Zhao
- Department of Microbiology and Immunology, University of Tennessee, Memphis 38163, USA
| | | |
Collapse
|
40
|
Wang JL, Zhang ZJ, Hartman M, Smits A, Westermark B, Muhr C, Nistér M. 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 Cancer 1995; 64:223-8. [PMID: 7657383 DOI: 10.1002/ijc.2910640402] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Mutations in the TP53 tumor suppressor gene have been studied in different types of brain tumors. Little is known about this genetic event in human meningioma, a mostly benign tumor. To investigate the frequency of TP53 gene mutations in human tumors derived from meningeal tissues, paraffin-embedded tissues from 30 cases (including 2 malignant and 4 atypical meningiomas, as well as 2 hemangioblastomas and 3 hemangiopericytomas) were screened by immunohistochemistry. Polymerase chain reaction/single strand conformational polymorphism (PCR/SSCP) and direct DNA sequencing were thereafter performed in selected cases. Nuclear p53 staining was not seen in any of the 19 benign meningiomas tested, while atypical meningiomas, hemangioblastomas, and hemangiopericytomas displayed nuclear staining in a subpopulation of tumor cells in 4 out of 5, 2 out of 2, and 3 out of 3 cases, respectively. One malignant meningioma showed an intense nuclear staining and a band shift in SSCP. In this case, we identified a mutation in the TP53 gene at codon 161 changing GCC to ACC and resulting in an alteration of alanine to threonine in this position. Our results indicate that TP53 gene mutation may be considered as a marker for malignant transformation in meningioma. p53 immunoreactivity, even in the absence of detectable gene mutation, is also associated with atypia and does not appear in regular benign meningiomas.
Collapse
Affiliation(s)
- J L Wang
- Department of Pathology, University Hospital, Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
41
|
Mauro A, Di Sapio A, Mocellini C, Schiffer D. Control of meningioma cell growth by platelet-derived growth factor (PDGF). J Neurol Sci 1995; 131:135-43. [PMID: 7595638 DOI: 10.1016/0022-510x(95)00106-c] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
We have examined the possible involvement of PDGF and PDGF receptors in the growth control of five meningiomas, analyzing the biopsy specimens and the primary cultures derived from the same tumors. Light and electron microscopy demonstrated that MAbs against PDGF beta-receptors immunodecorate meningioma cells in vivo and in vitro, while those against alpha-receptors gave negative results. The effects of PDGF isoforms AA, AB, BB and of PDGF neutralizing antibodies on meningioma cultures were examined using [3H]thymidine incorporation analysis. Only with PDGF-AB and -BB a mitogenic effect was observed, while PDGF-neutralizing antibodies produced a reduction of [3H]thymidine incorporation. The production of PDGF-like growth factors by meningioma cells was tested analyzing the effects of meningioma culture-conditioned media on the growth of Swiss 3T3 cells. In all cases meningioma conditioned media stimulated the in vitro growth of 3T3 fibroblasts and this stimulatory effect was strongly reduced by PDGF-neutralizing antibodies. Furthermore, Northern blot analysis demonstrated expression of c-sis/PDGF-B and PDGF beta-receptors mRNA in all meningioma biopsies and in all the derived cultures. Our results provide strong evidence that PDGF-B chain and PDGF beta-receptors are involved in growth control mechanisms of human meningiomas through autocrine and/or paracrine mechanisms.
Collapse
MESH Headings
- Antibodies, Monoclonal
- Blotting, Northern
- Cell Division/drug effects
- Cell Division/physiology
- Culture Media, Conditioned
- Gene Expression/physiology
- Humans
- Meningioma/pathology
- Microscopy, Immunoelectron
- Neutralization Tests
- Platelet-Derived Growth Factor/immunology
- Platelet-Derived Growth Factor/pharmacology
- Receptor, Platelet-Derived Growth Factor beta
- Receptors, Platelet-Derived Growth Factor/genetics
- Receptors, Platelet-Derived Growth Factor/immunology
- Receptors, Platelet-Derived Growth Factor/physiology
- Thymidine
- Tritium
- Tumor Cells, Cultured/cytology
- Tumor Cells, Cultured/ultrastructure
Collapse
Affiliation(s)
- A Mauro
- 2nd Department of Neurology, University of Turin, Italy
| | | | | | | |
Collapse
|
42
|
In Vitro Growth Inhibition of Growth Factor-stimulated Meningioma Cells by Calcium Channel Antagonists. Neurosurgery 1995. [DOI: 10.1097/00006123-199502000-00017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
43
|
Jensen RL, Origitano TC, Lee YS, Weber M, Wurster RD. In vitro growth inhibition of growth factor-stimulated meningioma cells by calcium channel antagonists. Neurosurgery 1995; 36:365-73; discussion 373-4. [PMID: 7731518 DOI: 10.1227/00006123-199502000-00017] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Studies have shown that a majority of meningiomas contain receptors for platelet-derived growth factor and epidermal growth factor and that these growth factors promote the proliferation of meningioma cells in culture. Although the mechanism of action has not been elucidated, intracellular calcium appears to be part of the signal transduction mechanism. Because alterations in intracellular calcium could interrupt this pathway and decrease cellular proliferation, we investigated the effects of calcium channel-blocking agents on the growth of meningioma cells in vitro. Primary meningioma cell cultures were established, and the cells were characterized by light and electron microscopy and by immunohistochemical studies. Then, the cultures were given growth factors and/or various calcium channel antagonists, and growth rates were measured. A dose-response decrease in cell growth was seen when verapamil, nifedipine, or diltiazem (voltage-dependent calcium channel-blocking agents) was added to serum-containing media. Also, these drugs blocked the growth stimulation of epidermal growth factor and platelet-derived growth factor in a similar fashion. Dantrolene, which inhibits the release of sequestered intracellular calcium, was also an effective blocker of the mitogenic stimulation of these growth factors.
Collapse
Affiliation(s)
- R L Jensen
- Department of Neurological Surgery, Loyola University, Maywood, Illinois, USA
| | | | | | | | | |
Collapse
|
44
|
Kuratsu JI, Seto H, Kochi M, Ushio Y. Expression of PDGF, PDGF-receptor, EGF-receptor and sex hormone receptors on meningioma. Acta Neurochir (Wien) 1994; 131:289-93. [PMID: 7754837 DOI: 10.1007/bf01808629] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The expression of platelet derived growth factors (PDGF), the PDGF-Receptor (R) (alpha and beta types), epidermal growth factor (EGF)-Receptor (R) and sex hormone (oestrogen and progesterone) receptors was studied in 22 meningiomas. All tumours were PDGF-R beta type positive and 21 (95%) were PDGF positive. Only 2 (9%) were PDGF-R alpha type positive, 13 (59%) were EGF-R positive. The expression of these proteins was not related to the histological type or the malignancy of the meningiomas although the expression of PDGF and PDGF-R beta tended to be stronger in malignant meningiomas. Oestrogen and progesterone receptor protein were examined in 19 patients (10 females and 9 males). None of the meningioma cells revealed oestrogen receptor protein while 17 (89%) of the 19 meningiomas were positive for progesterone receptor protein. The expression of progesterone receptor was not related to histological type or malignancy. Our studies suggest that the autocrine system, through PDGF and PDGF-R type beta, may play an important role in the tumourigenicity of meningiomas. EGF-R was present in almost half and progesterone receptor in most of the meningiomas. There was no correlation between the expression of either PDGF, PDGF-R or EGF-R and the expression of progesterone receptor protein.
Collapse
Affiliation(s)
- J I Kuratsu
- Department of Neurosurgery, Kumamoto University Medical School, Japan
| | | | | | | |
Collapse
|
45
|
Black PM, Carroll R, Glowacka D, Riley K, Dashner K. Platelet-derived growth factor expression and stimulation in human meningiomas. J Neurosurg 1994; 81:388-93. [PMID: 8057146 DOI: 10.3171/jns.1994.81.3.0388] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The platelet-derived growth factor (PDGF) family consists of subunits A and B and receptors alpha and beta. This paper evaluates the potential role of the homodimer PDGF-BB as a growth factor in meningiomas. It analyzes the expression of messenger RNA in members of the PDGF family in these tumors, measures the growth response of meningiomas to exogenous PDGF-BB in culture, and examines the induction of the c-fos proto-oncogene by PDGF-BB. Northern blot analysis was carried out on tissue from 20 meningiomas to measure the expression of PDGF-A, PDGF-B, PDGF-alpha receptor (PDGF-alpha-R) and PDGF-beta receptor (PDGF-beta-R). All tumors expressed PDGF-A and PDGF-B subunits. Nineteen of the 20 tumors expressed PDGF-beta-R and none expressed PDGF-alpha-R as measured by this technique. Because the beta receptor is selectively sensitive to stimulation by the PDGF-B subunit, these data suggest that meningiomas might be susceptible to stimulation by PDGF-BB. To test this hypothesis, the effect of exogenous PDGF-BB on meningioma growth was evaluated by incubating cells from 10 human meningiomas. Tritiated thymidine incorporation was used to evaluate stimulation of growth over a 48-hour period using PDGF-BB concentrations of 1, 3, or 6 ng/ml. Linear regression analysis and multiple-factor analysis of variance were used to measure PDGF-BB effects. Three of the 10 tumor specimens responded significantly to PDGF-BB, with a three- to sixfold increase in thymidine incorporation over 72 hours of exposure, and there was a significant overall growth-stimulating effect of PDGF-BB in the 10 tumor specimens tested. In the last set of experiments, the functionality of the PDGF-beta-R was determined by examining the induction of the proto-oncogene c-fos by PDGF-BB in meningioma cell cultures. A significant increase in c-fos protein was observed 3 hours after PDGF-BB addition. These findings demonstrate that PDGF-A, PDGF-B, and PDGF-beta-R are expressed in meningiomas and suggest that the beta receptor is functional: when it is activated, c-fos levels are increased, and an increase in meningioma cell division is observed after the addition of PDGF-BB. These studies support the hypothesis that PDGF acts as a growth factor in meningiomas.
Collapse
Affiliation(s)
- P M Black
- Brain Tumor Center, Brigham and Women's Hospital, Boston, Massachusetts
| | | | | | | | | |
Collapse
|
46
|
Johnson MD, Horiba M, Winnier AR, Arteaga CL. The epidermal growth factor receptor is associated with phospholipase C-gamma 1 in meningiomas. Hum Pathol 1994; 25:146-53. [PMID: 8119714 DOI: 10.1016/0046-8177(94)90270-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In this study we evaluated phospholipase C-gamma 1 (PLC-gamma 1) expression, activity, and association with the epidermal growth factor (EGF) receptor in a series of human meningiomas as well as cultured meningioma cells. Phospholipase C-gamma 1 was detectable by immunoblot and immunohistochemistry in 13 of 13 meningioma specimens. Epidermal growth factor receptors were detected by immunoblot in six of nine meningiomas (67%) and by immunohistochemistry in 13 of 19 meningiomas (68%) but not in normal leptomeningeal cells. In two of three meningiomas EGF receptors and/or a 170-kd phosphotyrosine band precipitated with a PLC-gamma 1 antiserum. Both PLC-gamma 1 and EGF receptors also exhibited the same pattern of immunostaining on meningioma tissue sections. Phospholipase C-gamma 1 catalytic activity, measured in a PIP2 hydrolysis assay, was higher in nine EGF receptor-positive meningiomas than in six EGF receptor-negative meningiomas (P = .05; t test). Finally, treatment of cultured meningioma cells with transforming growth factor-alpha induced a 78% increase in PLC-gamma 1 catalytic activity. Thus, these data are consistent with the possibility that the EGF receptor tyrosine kinase regulates PLC-gamma 1 activity in native meningioma tissue.
Collapse
Affiliation(s)
- M D Johnson
- Nashville Veterans Administration Medical Center, TN
| | | | | | | |
Collapse
|
47
|
Abstract
Treatment objectives for meningiomas of the cranial base include relief of neurologic disability and prevention of clinical progression or recurrence with the least morbidity. Recent advances in skull base surgical techniques, through an appreciation of skull base anatomy and institutional specialization, have contributed major improvements to the outlook for patients with these tumors, and previously inoperable cases may now often be removed completely with acceptable risk. Since significant morbidity may be incurred during surgical resection of these difficult lesions, especially in terms of cranial nerve dysfunction, the value of aggressive surgical resection must be weighted against the often indolent natural history of these lesions, and must be individualized in each patient. Completeness of resection is the major prognostic factor determining the outcome of patients with typical benign meningiomas in terms of length of survival, risk of recurrence, and neurological disability. Various means of prognosticating the growth potential of a given tumor are being investigated, though none have yet been confirmed for their predictive value in typical, histologically benign meningiomas. The role of external beam radiotherapy has not been subjected to adequately controlled, prospective studies, and there is currently insufficient followup to assess the risks and benefits of stereotactic radiosurgery. Advances in the clinical management of tumors of the skull base has had perhaps the greatest impact for patients with meningiomas who constitute a large portion of tumors seen in these locations. Although the majority have benign histological features, skull base meningiomas can present a formidable challenge due to their proximity to vital structures, surgical inaccessibility, and occasional aggressive features. The combination in recent years of advances in skull base surgical techniques, adjuvant therapy, and rehabilitation methods have dramatically improved the outcome for these tumors.
Collapse
Affiliation(s)
- R Desai
- Department of Neurosurgery, College of Physicians and Surgeons of Columbia University, New York, New York
| | | |
Collapse
|
48
|
|
49
|
Brandis A, Mirzai S, Tatagiba M, Walter GF, Samii M, Ostertag H. Immunohistochemical detection of female sex hormone receptors in meningiomas: correlation with clinical and histological features. Neurosurgery 1993; 33:212-7; discussion 217-8. [PMID: 8367042 DOI: 10.1227/00006123-199308000-00005] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Sixty-one meningiomas from 60 patients were screened for estrogen receptors and progesterone receptors (PgR) with monoclonal antibodies in an immunohistochemical assay. In addition, 43 of the cases were evaluated for tumor size and peritumoral edema, as seen on computed tomographic scans and magnetic resonance images. Sixty-one percent of the tumors contained significant amounts of PgR, whereas no estrogen receptor-positive tumor was observed. Thirteen percent of all tumors were classified as nonbenign variants (atypical and anaplastic meningiomas) and were more frequently found in male patients (P < 0.05). Nonbenign tumors more frequently showed an absence of PgR (P < 0.05), and there was a tendency for PgR-negative tumors to be larger than PgR-positive ones. No correlation was found between PgR status and edema. It is concluded that PgR status in meningiomas is related to tumor differentiation and may be of prognostic value with regard to biological behavior and clinical outcome.
Collapse
Affiliation(s)
- A Brandis
- Institute of Neuropathology, Hannover Medical School, Germany
| | | | | | | | | | | |
Collapse
|
50
|
Seymour L, Dajee D, Bezwoda WR. Tissue platelet derived-growth factor (PDGF) predicts for shortened survival and treatment failure in advanced breast cancer. Breast Cancer Res Treat 1993; 26:247-52. [PMID: 8251649 DOI: 10.1007/bf00665802] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
In a study of plasma and tissue platelet derived growth factor (PDGF) concentration in patients with breast cancer, elevated levels of plasma PDGF were found in a significant proportion, 11/37 (30%), of patients. Sixteen patients (43%) had tumors which expressed PDGF-AA and 6 patients had tumors which in addition expressed the BB isoform of PDGF. All patients with elevated plasma levels of platelet derived growth factor had tumors which expressed the growth factor on immunohistochemical staining of tumor cells. Furthermore there was a significant correlation between plasma levels of platelet derived growth factor and the intensity of tissue staining. Patients with stage four breast cancer with tumors which were positive for platelet derived growth factor had a significantly lower response rate to chemotherapy as well as significantly shorter duration of survival. In addition, patients with stage four breast cancer who had elevated plasma PDGF levels had a significantly shorter survival. These results indicate that elevated plasma levels of platelet derived growth factor in patients with breast cancer are derived from the tumor cells and suggest that platelet derived growth factor may play a significant role in control tumor cell growth.
Collapse
Affiliation(s)
- L Seymour
- Department of Medicine, University of the Witwatersrand, South Africa
| | | | | |
Collapse
|