1
|
Ruiz-García C, Lassaletta L, López-Larrubia P, Varela-Nieto I, Murillo-Cuesta S. Tumors of the nervous system and hearing loss: Beyond vestibular schwannomas. Hear Res 2024; 447:109012. [PMID: 38703433 DOI: 10.1016/j.heares.2024.109012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/08/2024] [Accepted: 04/18/2024] [Indexed: 05/06/2024]
Abstract
Hearing loss is a common side effect of many tumor treatments. However, hearing loss can also occur as a direct result of certain tumors of the nervous system, the most common of which are the vestibular schwannomas (VS). These tumors arise from Schwann cells of the vestibulocochlear nerve and their main cause is the loss of function of NF2, with 95 % of cases being sporadic and 5 % being part of the rare neurofibromatosis type 2 (NF2)-related Schwannomatosis. Genetic variations in NF2 do not fully explain the clinical heterogeneity of VS, and interactions between Schwann cells and their microenvironment appear to be critical for tumor development. Preclinical in vitro and in vivo models of VS are needed to develop prognostic biomarkers and targeted therapies. In addition to VS, other tumors can affect hearing. Meningiomas and other masses in the cerebellopontine angle can compress the vestibulocochlear nerve due to their anatomic proximity. Gliomas can disrupt several neurological functions, including hearing; in fact, glioblastoma multiforme, the most aggressive subtype, may exhibit early symptoms of auditory alterations. Besides, treatments for high-grade tumors, including chemotherapy or radiotherapy, as well as incomplete resections, can induce long-term auditory dysfunction. Because hearing loss can have an irreversible and dramatic impact on quality of life, it should be considered in the clinical management plan of patients with tumors, and monitored throughout the course of the disease.
Collapse
Affiliation(s)
- Carmen Ruiz-García
- Department of Otorhinolaryngology, La Paz University Hospital. Paseo La Castellana 261, Madrid 28046, Spain; Research in Otoneurosurgery. Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital - Universidad Autónoma de Madrid), Paseo La Castellana 261, Madrid 28046, Spain; Neuropathology of Hearing and Myelinopathies, Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM. Arturo Duperier 4, Madrid 28029, Spain; PhD Program in Medicine and Surgery, Autonomous University of Madrid, Madrid, Spain
| | - Luis Lassaletta
- Department of Otorhinolaryngology, La Paz University Hospital. Paseo La Castellana 261, Madrid 28046, Spain; Research in Otoneurosurgery. Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital - Universidad Autónoma de Madrid), Paseo La Castellana 261, Madrid 28046, Spain; Biomedical Research Networking Centre On Rare Diseases (CIBERER), Institute of Health Carlos III, Monforte de Lemos 9-11, Madrid 28029, Spain
| | - Pilar López-Larrubia
- Biomedical Magnetic Resonance, Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM. Arturo Duperier 4, Madrid 28029, Spain
| | - Isabel Varela-Nieto
- Research in Otoneurosurgery. Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital - Universidad Autónoma de Madrid), Paseo La Castellana 261, Madrid 28046, Spain; Neuropathology of Hearing and Myelinopathies, Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM. Arturo Duperier 4, Madrid 28029, Spain; Biomedical Research Networking Centre On Rare Diseases (CIBERER), Institute of Health Carlos III, Monforte de Lemos 9-11, Madrid 28029, Spain.
| | - Silvia Murillo-Cuesta
- Research in Otoneurosurgery. Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital - Universidad Autónoma de Madrid), Paseo La Castellana 261, Madrid 28046, Spain; Neuropathology of Hearing and Myelinopathies, Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM. Arturo Duperier 4, Madrid 28029, Spain; Biomedical Research Networking Centre On Rare Diseases (CIBERER), Institute of Health Carlos III, Monforte de Lemos 9-11, Madrid 28029, Spain.
| |
Collapse
|
2
|
Bhattacharyya S, Oblinger JL, Beauchamp RL, Yin Z, Erdin S, Koundinya P, Ware AD, Ferrer M, Jordan JT, Plotkin SR, Xu L, Chang LS, Ramesh V. Proteasomal pathway inhibition as a potential therapy for NF2-associated meningioma and schwannoma. Neuro Oncol 2023; 25:1617-1630. [PMID: 36806881 PMCID: PMC10479743 DOI: 10.1093/neuonc/noad037] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Neurofibromatosis 2 (NF2) is an inherited disorder caused by bi-allelic inactivation of the NF2 tumor suppressor gene. NF2-associated tumors, including schwannoma and meningioma, are resistant to chemotherapy, often recurring despite surgery and/or radiation, and have generally shown cytostatic response to signal transduction pathway inhibitors, highlighting the need for improved cytotoxic therapies. METHODS Leveraging data from our previous high-throughput drug screening in NF2 preclinical models, we identified a class of compounds targeting the ubiquitin-proteasome pathway (UPP), and undertook studies using candidate UPP inhibitors, ixazomib/MLN9708, pevonedistat/MLN4924, and TAK-243/MLN7243. Employing human primary and immortalized meningioma (MN) cell lines, CRISPR-modified Schwann cells (SCs), and mouse Nf2-/- SCs, we performed dose response testing, flow cytometry-based Annexin V and cell cycle analyses, and RNA-sequencing to identify potential underlying mechanisms of apoptosis. In vivo efficacy was also assessed in orthotopic NF2-deficient meningioma and schwannoma tumor models. RESULTS Testing of three UPP inhibitors demonstrated potent reduction in cell viability and induction of apoptosis for ixazomib or TAK-243, but not pevonedistat. In vitro analyses revealed that ixazomib or TAK-243 downregulates expression of c-KIT and PDGFRα, as well as the E3 ubiquitin ligase SKP2 while upregulating genes associated with endoplasmic reticulum stress-mediated activation of the unfolded protein response (UPR). In vivo treatment of mouse models revealed delayed tumor growth, suggesting a therapeutic potential. CONCLUSIONS This study demonstrates the efficacy of proteasomal pathway inhibitors in meningioma and schwannoma preclinical models and lays the groundwork for use of these drugs as a promising novel treatment strategy for NF2 patients.
Collapse
Affiliation(s)
- Srirupa Bhattacharyya
- Department of Neurology and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Janet L Oblinger
- Center for Childhood Cancer & Blood Diseases, Nationwide Children's Hospital and Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Roberta L Beauchamp
- Department of Neurology and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Zhenzhen Yin
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Serkan Erdin
- Department of Neurology and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Priya Koundinya
- Department of Neurology and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Anna D Ware
- Department of Neurology and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Marc Ferrer
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Justin T Jordan
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Scott R Plotkin
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Lei Xu
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Long-Sheng Chang
- Center for Childhood Cancer & Blood Diseases, Nationwide Children's Hospital and Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Vijaya Ramesh
- Department of Neurology and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
3
|
Galldiks N, Angenstein F, Werner JM, Bauer EK, Gutsche R, Fink GR, Langen KJ, Lohmann P. Use of advanced neuroimaging and artificial intelligence in meningiomas. Brain Pathol 2022; 32:e13015. [PMID: 35213083 PMCID: PMC8877736 DOI: 10.1111/bpa.13015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/09/2021] [Accepted: 08/02/2021] [Indexed: 01/04/2023] Open
Abstract
Anatomical cross‐sectional imaging methods such as contrast‐enhanced MRI and CT are the standard for the delineation, treatment planning, and follow‐up of patients with meningioma. Besides, advanced neuroimaging is increasingly used to non‐invasively provide detailed insights into the molecular and metabolic features of meningiomas. These techniques are usually based on MRI, e.g., perfusion‐weighted imaging, diffusion‐weighted imaging, MR spectroscopy, and positron emission tomography. Furthermore, artificial intelligence methods such as radiomics offer the potential to extract quantitative imaging features from routinely acquired anatomical MRI and CT scans and advanced imaging techniques. This allows the linking of imaging phenotypes to meningioma characteristics, e.g., the molecular‐genetic profile. Here, we review several diagnostic applications and future directions of these advanced neuroimaging techniques, including radiomics in preclinical models and patients with meningioma.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany.,Center for Integrated Oncology (CIO), Universities of Aachen, Cologne, Germany
| | - Frank Angenstein
- Functional Neuroimaging Group, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Magdeburg, Germany.,Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany.,Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| | - Jan-Michael Werner
- Department of Neurology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Elena K Bauer
- Department of Neurology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Robin Gutsche
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
| | - Gereon R Fink
- Department of Neurology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany.,Center for Integrated Oncology (CIO), Universities of Aachen, Cologne, Germany.,Department of Nuclear Medicine, University Hospital Aachen, Aachen, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany.,Department of Stereotaxy and Functional Neurosurgery, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
4
|
Welling DB, Collier KA, Burns SS, Oblinger JL, Shu E, Miles‐Markley BA, Hofmeister CC, Makary MS, Slone HW, Blakeley JO, Mansouri SA, Neff BA, Jackler RK, Mortazavi A, Chang L. Early phase clinical studies of AR-42, a histone deacetylase inhibitor, for neurofibromatosis type 2-associated vestibular schwannomas and meningiomas. Laryngoscope Investig Otolaryngol 2021; 6:1008-1019. [PMID: 34667843 PMCID: PMC8513424 DOI: 10.1002/lio2.643] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/16/2021] [Accepted: 08/10/2021] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVES Two pilot studies of AR-42, a pan-histone deacetylase inhibitor, in human neurofibromatosis type 2 (NF2), vestibular schwannomas (VS), and meningiomas are presented. Primary endpoints included safety, and intra-tumoral pharmacokinetics (PK) and pharmacodynamics (PD). METHODS Pilot 1 is a subset analysis of a phase 1 study of AR-42 in solid tumors, which included NF2 or sporadic meningiomas. Tumor volumes and treatment-related adverse events (TRAEs) are reported (NCT01129193).Pilot 2 is a phase 0 surgical study of AR-42 assessing intra-tumoral PK and PD. AR-42 was administered for 3 weeks pre-operatively. Plasma and tumor drug concentrations and p-AKT expression were measured (NCT02282917). RESULTS Pilot 1: Five patients with NF2 and two with sporadic meningiomas experienced a similar incidence of TRAEs to the overall phase I trial. The six evaluable patients had 15 tumors (8 VS, 7 meningiomas). On AR-42, tumor volume increased in six, remained stable in eight, and decreased in one tumor. The annual percent growth rate decreased in eight, remained stable in three, and increased in four tumors. Pilot 2: Four patients with sporadic VS and one patient with meningioma experienced no grade 3/4 toxicities. Expression of p-AKT decreased in three of four VS. All tumors had higher AR-42 concentrations than plasma. CONCLUSIONS AR-42 is safe. Tumor volumes showed a mixed response, but most slowed growth. On a 40-mg regimen, drug concentrated in tumors and growth pathways were suppressed in most tumors, suggesting this may be a well-tolerated and effective dose. A phase 2 study of AR-42 for NF2-associated tumors appears warranted. LEVEL OF EVIDENCE 1b, 4.
Collapse
Affiliation(s)
- D. Bradley Welling
- Department of Otolaryngology Head and Neck SurgeryHarvard Medical School, Massachusetts Eye and Ear Infirmary, Massachusetts General HospitalBostonMassachusettsUSA
| | - Katharine A. Collier
- Division of Medical Oncology, Department of Internal MedicineThe Ohio State University College of Medicine and the Comprehensive Cancer CenterColumbusOhioUSA
| | - Sarah S. Burns
- Center for Childhood Cancer and Blood diseasesAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOhioUSA
| | - Janet L. Oblinger
- Center for Childhood Cancer and Blood diseasesAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOhioUSA
| | - Edina Shu
- Department of Otolaryngology Head and Neck SurgeryHarvard Medical School, Massachusetts Eye and Ear Infirmary, Massachusetts General HospitalBostonMassachusettsUSA
| | - Beth A. Miles‐Markley
- Department of Otolaryngology‐Head and Neck SurgeryThe Ohio State University College of MedicineColumbusOhioUSA
| | - Craig C. Hofmeister
- Department of Hematology & OncologyWinship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Mina S. Makary
- Department of RadiologyThe Ohio State University College of MedicineColumbusOhioUSA
| | - H. Wayne Slone
- Department of RadiologyThe Ohio State University College of MedicineColumbusOhioUSA
| | - Jaishri O. Blakeley
- Departments of Neurology, Neurosurgery, & OncologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - S. Alireza Mansouri
- Departments of Neurology, Neurosurgery, & OncologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Brian A. Neff
- Department of Otolaryngology Head and Neck SurgeryMayo ClinicRochesterMinnesotaUSA
| | - Robert K. Jackler
- Department of Otolaryngology Head and Neck SurgeryStanford UniversityPalo AltoCaliforniaUSA
| | - Amir Mortazavi
- Division of Medical Oncology, Department of Internal MedicineThe Ohio State University College of Medicine and the Comprehensive Cancer CenterColumbusOhioUSA
| | - Long‐Sheng Chang
- Center for Childhood Cancer and Blood diseasesAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOhioUSA
- Department of Otolaryngology‐Head and Neck SurgeryThe Ohio State University College of MedicineColumbusOhioUSA
| |
Collapse
|
5
|
Current Understanding of Neurofibromatosis Type 1, 2, and Schwannomatosis. Int J Mol Sci 2021; 22:ijms22115850. [PMID: 34072574 PMCID: PMC8198724 DOI: 10.3390/ijms22115850] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 12/16/2022] Open
Abstract
Neurofibromatosis (NF) is a neurocutaneous syndrome characterized by the development of tumors of the central or peripheral nervous system including the brain, spinal cord, organs, skin, and bones. There are three types of NF: NF1 accounting for 96% of all cases, NF2 in 3%, and schwannomatosis (SWN) in <1%. The NF1 gene is located on chromosome 17q11.2, which encodes for a tumor suppressor protein, neurofibromin, that functions as a negative regulator of Ras/MAPK and PI3K/mTOR signaling pathways. The NF2 gene is identified on chromosome 22q12, which encodes for merlin, a tumor suppressor protein related to ezrin-radixin-moesin that modulates the activity of PI3K/AKT, Raf/MEK/ERK, and mTOR signaling pathways. In contrast, molecular insights on the different forms of SWN remain unclear. Inactivating mutations in the tumor suppressor genes SMARCB1 and LZTR1 are considered responsible for a majority of cases. Recently, treatment strategies to target specific genetic or molecular events involved in their tumorigenesis are developed. This study discusses molecular pathways and related targeted therapies for NF1, NF2, and SWN and reviews recent clinical trials which involve NF patients.
Collapse
|
6
|
Chang LS, Oblinger JL, Burns SS, Huang J, Anderson LW, Hollingshead MG, Shen R, Pan L, Agarwal G, Ren Y, Roberts RD, O'Keefe BR, Kinghorn AD, Collins JM. Targeting Protein Translation by Rocaglamide and Didesmethylrocaglamide to Treat MPNST and Other Sarcomas. Mol Cancer Ther 2020; 19:731-741. [PMID: 31848295 PMCID: PMC7056570 DOI: 10.1158/1535-7163.mct-19-0809] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/02/2019] [Accepted: 12/13/2019] [Indexed: 01/30/2023]
Abstract
Malignant peripheral nerve sheath tumors (MPNST) frequently overexpress eukaryotic initiation factor 4F components, and the eIF4A inhibitor silvestrol potently suppresses MPNST growth. However, silvestrol has suboptimal drug-like properties, including a bulky structure, poor oral bioavailability (<2%), sensitivity to MDR1 efflux, and pulmonary toxicity in dogs. We compared ten silvestrol-related rocaglates lacking the dioxanyl ring and found that didesmethylrocaglamide (DDR) and rocaglamide (Roc) had growth-inhibitory activity comparable with silvestrol. Structure-activity relationship analysis revealed that the dioxanyl ring present in silvestrol was dispensable for, but may enhance, cytotoxicity. Both DDR and Roc arrested MPNST cells at G2-M, increased the sub-G1 population, induced cleavage of caspases and PARP, and elevated the levels of the DNA-damage response marker γH2A.X, while decreasing the expression of AKT and ERK1/2, consistent with translation inhibition. Unlike silvestrol, DDR and Roc were not sensitive to MDR1 inhibition. Pharmacokinetic analysis confirmed that Roc had 50% oral bioavailability. Importantly, Roc, when administered intraperitoneally or orally, showed potent antitumor effects in an orthotopic MPNST mouse model and did not induce pulmonary toxicity in dogs as found with silvestrol. Treated tumors displayed degenerative changes and had more cleaved caspase-3-positive cells, indicative of increased apoptosis. Furthermore, Roc effectively suppressed the growth of osteosarcoma, Ewing sarcoma, and rhabdomyosarcoma cells and patient-derived xenografts. Both Roc- and DDR-treated sarcoma cells showed decreased levels of multiple oncogenic kinases, including insulin-like growth factor-1 receptor. The more favorable drug-like properties of DDR and Roc and the potent antitumor activity of Roc suggest that these rocaglamides could become viable treatments for MPNST and other sarcomas.
Collapse
Affiliation(s)
- Long-Sheng Chang
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio.
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University College of Medicine, Columbus, Ohio
- Department of Pathology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Janet L Oblinger
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| | - Sarah S Burns
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| | - Jie Huang
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| | - Larry W Anderson
- Division of Cancer Treatment and Diagnosis, Center for Cancer Research, National Cancer Institute, NIH, Frederick, Maryland
| | - Melinda G Hollingshead
- Division of Cancer Treatment and Diagnosis, Center for Cancer Research, National Cancer Institute, NIH, Frederick, Maryland
| | - Rulong Shen
- Department of Pathology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Li Pan
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University College of Pharmacy, Columbus, Ohio
| | - Garima Agarwal
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University College of Pharmacy, Columbus, Ohio
| | - Yulin Ren
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University College of Pharmacy, Columbus, Ohio
| | - Ryan D Roberts
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| | - Barry R O'Keefe
- Division of Cancer Treatment and Diagnosis, Center for Cancer Research, National Cancer Institute, NIH, Frederick, Maryland
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, NIH, Frederick, Maryland
| | - A Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University College of Pharmacy, Columbus, Ohio
| | - Jerry M Collins
- Division of Cancer Treatment and Diagnosis, Center for Cancer Research, National Cancer Institute, NIH, Frederick, Maryland
| |
Collapse
|
7
|
AlSahlawi A, Aljelaify R, Magrashi A, AlSaeed M, Almutairi A, Alqubaishi F, Alturkistani A, AlObaid A, Abouelhoda M, AlMubarak L, AlTassan N, Abedalthagafi M. New insights into the genomic landscape of meningiomas identified FGFR3 in a subset of patients with favorable prognoses. Oncotarget 2019; 10:5549-5559. [PMID: 31565188 PMCID: PMC6756861 DOI: 10.18632/oncotarget.27178] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/12/2019] [Indexed: 12/26/2022] Open
Abstract
Background: With a prevalence of 170 000 adults in the US alone, meningiomas are the most common primary intracranial tumors. The management of skull base meningiomas is challenging due to their complexity and proximity to crucial nearby structures. The identification of oncogenic mutations has provided further insights into the tumorigenesis of meningioma and the possibility of targeted therapy.
This study aimed to further investigate the association of mutational profiles with anatomical distribution, histological subtype, WHO grade, and recurrence in patients with meningioma. Methods: Tissue samples were collected from 71 patients diagnosed with meningioma from 2008 to 2016. A total of 51 cases were skull based. Samples were subjected to targeted sequencing using a next generation customized cancer gene panel (n = 66 genes analyzed).
Results: We detected genomic alterations (GAs) in 68 tumors, averaging 1.56 ± 1.07 genomic alterations (GAs) per sample. NF2 was the most frequently altered gene (36/71 cases). Interestingly, we identified a number of mutations in non-NF2 genes, including a hotspot TERTp c.−124: G > A mutation that may be related to poor prognosis and FGFR3 mutations that may represent biomarkers of a favorable prognosis as reported in other cancers.
Conclusions: We demonstrate that comprehensive genomic profiling in our population can reveal a potential new prognostic biomarkers of skull base meningioma. These mutations can enhance diagnostic accuracy and clinical decision-making. Among our findings were the identification of a TERTp mutation and the first report of FGFR3 mutations that may represent biomarkers for the identification of skull base meningioma patients with a favorable prognosis.
Collapse
Affiliation(s)
- Aysha AlSahlawi
- Genomics Research Department, Saudi Human Genome Project, King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia.,Montreal Neurological Institute, Montreal, Canada.,Neurosurgery Department, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Rasha Aljelaify
- Genomics Research Department, Saudi Human Genome Project, King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia.,Saudi Human Genome Program, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Amna Magrashi
- Genetics Department, King Faisal Specialists Hospital and Research Center, Riyadh, Saudi Arabia
| | - Mariam AlSaeed
- Genomics Research Department, Saudi Human Genome Project, King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia.,Saudi Human Genome Program, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Amal Almutairi
- Genomics Research Department, Saudi Human Genome Project, King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia.,Saudi Human Genome Program, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Fatimah Alqubaishi
- Genomics Research Department, Saudi Human Genome Project, King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia.,Saudi Human Genome Program, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | | | - Abdullah AlObaid
- Neurosurgery Department, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Mohamed Abouelhoda
- Saudi Human Genome Program, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia.,Genetics Department, King Faisal Specialists Hospital and Research Center, Riyadh, Saudi Arabia
| | - Latifa AlMubarak
- Genomics Research Department, Saudi Human Genome Project, King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia.,Saudi Human Genome Program, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Nada AlTassan
- Saudi Human Genome Program, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia.,Genetics Department, King Faisal Specialists Hospital and Research Center, Riyadh, Saudi Arabia
| | - Malak Abedalthagafi
- Genomics Research Department, Saudi Human Genome Project, King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia.,Genetics Department, King Faisal Specialists Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
8
|
Allaway R, Angus SP, Beauchamp RL, Blakeley JO, Bott M, Burns SS, Carlstedt A, Chang LS, Chen X, Clapp DW, Desouza PA, Erdin S, Fernandez-Valle C, Guinney J, Gusella JF, Haggarty SJ, Johnson GL, La Rosa S, Morrison H, Petrilli AM, Plotkin SR, Pratap A, Ramesh V, Sciaky N, Stemmer-Rachamimov A, Stuhlmiller TJ, Talkowski ME, Welling DB, Yates CW, Zawistowski JS, Zhao WN. Traditional and systems biology based drug discovery for the rare tumor syndrome neurofibromatosis type 2. PLoS One 2018; 13:e0197350. [PMID: 29897904 PMCID: PMC5999111 DOI: 10.1371/journal.pone.0197350] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 05/01/2018] [Indexed: 11/18/2022] Open
Abstract
Neurofibromatosis 2 (NF2) is a rare tumor suppressor syndrome that manifests with multiple schwannomas and meningiomas. There are no effective drug therapies for these benign tumors and conventional therapies have limited efficacy. Various model systems have been created and several drug targets have been implicated in NF2-driven tumorigenesis based on known effects of the absence of merlin, the product of the NF2 gene. We tested priority compounds based on known biology with traditional dose-concentration studies in meningioma and schwann cell systems. Concurrently, we studied functional kinome and gene expression in these cells pre- and post-treatment to determine merlin deficient molecular phenotypes. Cell viability results showed that three agents (GSK2126458, Panobinostat, CUDC-907) had the greatest activity across schwannoma and meningioma cell systems, but merlin status did not significantly influence response. In vivo, drug effect was tumor specific with meningioma, but not schwannoma, showing response to GSK2126458 and Panobinostat. In culture, changes in both the transcriptome and kinome in response to treatment clustered predominantly based on tumor type. However, there were differences in both gene expression and functional kinome at baseline between meningioma and schwannoma cell systems that may form the basis for future selective therapies. This work has created an openly accessible resource (www.synapse.org/SynodosNF2) of fully characterized isogenic schwannoma and meningioma cell systems as well as a rich data source of kinome and transcriptome data from these assay systems before and after treatment that enables single and combination drug discovery based on molecular phenotype.
Collapse
Affiliation(s)
| | | | - Steve P. Angus
- University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Roberta L. Beauchamp
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Jaishri O. Blakeley
- Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Marga Bott
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Lake Nona-Orlando, FL, United States of America
| | - Sarah S. Burns
- Center for Childhood Cancer and Blood Diseases, The Research Institute at Nationwide Children's Hospital and Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States of America
| | | | - Long-Sheng Chang
- Center for Childhood Cancer and Blood Diseases, The Research Institute at Nationwide Children's Hospital and Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Xin Chen
- University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - D. Wade Clapp
- Indiana University, School of Medicine, Indianapolis, IN, United States of America
| | - Patrick A. Desouza
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Serkan Erdin
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Cristina Fernandez-Valle
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Lake Nona-Orlando, FL, United States of America
| | | | - James F. Gusella
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Stephen J. Haggarty
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Gary L. Johnson
- University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | | | - Helen Morrison
- Leibniz-Institute on Aging–Fritz-Lipmann Institute (FLI), Jena, Germany
| | - Alejandra M. Petrilli
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Lake Nona-Orlando, FL, United States of America
| | - Scott R. Plotkin
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Abhishek Pratap
- Sage Bionetworks, Seattle, WA, United States of America
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA, United States of America
| | - Vijaya Ramesh
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Noah Sciaky
- University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Anat Stemmer-Rachamimov
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Tim J. Stuhlmiller
- University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Michael E. Talkowski
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
| | - D. Bradley Welling
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Massachusetts General Hospital and Harvard University, Boston, MA, United States of America
| | - Charles W. Yates
- Indiana University, School of Medicine, Indianapolis, IN, United States of America
| | - Jon S. Zawistowski
- University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Wen-Ning Zhao
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
| |
Collapse
|
9
|
Abstract
Neurofibromatosis (NF) encompasses a group of distinct genetic disorders in which affected children and adults are prone to the development of benign and malignant tumors of the nervous system. The purpose of this review is to discuss the spectrum of CNS tumors arising in individuals with NF type 1 (NF1) and NF type 2 (NF2), their pathogenic etiologies, and the rational treatment options for people with these neoplasms. This article is a review of preclinical and clinical data focused on the treatment of the most common CNS tumors encountered in children and adults with NF1 and NF2. Although children with NF1 are at risk for developing low-grade gliomas of the optic pathway and brainstem, individuals with NF2 typically manifest low-grade tumors affecting the cranial nerves (vestibular schwannomas), meninges (meningiomas), and spinal cord (ependymomas). With the identification of the NF1 and NF2 genes, molecularly targeted therapies are beginning to emerge, as a result of a deeper understanding of the mechanisms underlying NF1 and NF2 protein function. As we enter into an era of precision oncology, a more comprehensive awareness of the factors that increase the risk of developing CNS cancers in affected individuals, coupled with a greater appreciation of the cellular and molecular determinants that maintain tumor growth, will undoubtedly yield more effective therapies for these cancer predisposition syndromes.
Collapse
Affiliation(s)
- Jian Campian
- All authors: Washington University School of Medicine, St. Louis, MO
| | - David H Gutmann
- All authors: Washington University School of Medicine, St. Louis, MO
| |
Collapse
|