1
|
Tang PC, Um S, Mayfield AB, Bracho OR, Castillo CD, Dinh CT, Dykxhoorn DM, Liu XZ. Interactions among Merlin, Arkadia, and SKOR2 mediate NF2-associated Schwann cell proliferation in human. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614711. [PMID: 39386608 PMCID: PMC11463491 DOI: 10.1101/2024.09.24.614711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
NF2-Related Schwannomatosis (previously referred to as Neurofibromatosis Type 2, or NF2) is a genetic-associated disease resulting from mutations in the gene, NF2. NF2 encodes the merlin protein, which acts as a tumor suppressor. Bilateral vestibular schwannoma (VS) is a hallmark of NF2. Although the exactly molecular mechanism mediating NF2-driven schwannomatosis remain unclear, it is known that defective Merlin protein functionality leads to abnormal cell proliferation. Herein, we utilized a human induced pluripotent stem cell (hiPSC)-based Schwann cell (SC) model to investigate the role of merlin in human SCs. SCs were derived from hiPSCs carrying a NF2 mutation (c.191 T > C; p. L64P), its isogenic wild-type control cell line, and a NF2 patient-derived hiPSC line. NF2 mutant SCs showed abnormal cellular morphology and proliferation. Proteomic analyses identified novel interaction partners for Merlin - Arkadia and SKOR2. Our results established a new model in which merlin interacts with Arkadia and SKOR2 and this interaction is required for the proper activation of the SMAD-dependent pathway in TGFβ signaling.
Collapse
Affiliation(s)
- Pei-Ciao Tang
- Equal contribution: Pei-Ciao Tang and Seyoung Um
- Lead contact: Pei-Ciao Tang
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, 33136, United States
| | - Seyoung Um
- Equal contribution: Pei-Ciao Tang and Seyoung Um
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, 33136, United States
| | | | - Olena R. Bracho
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, 33136, United States
| | - Christian Del Castillo
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, 33136, United States
| | - Christine T. Dinh
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, 33136, United States
- Sylvester Comprehensive Cancer Center, Miami, FL, 33136, United States
| | - Derek M. Dykxhoorn
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, United States
| | - Xue Zhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, 33136, United States
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, United States
| |
Collapse
|
2
|
Shelton WJ, Santos Horta E, Stephen Nix J, Gokden M, Rodriguez A. Functional precision medicine assay for recurrent meningioma: a proof of principle. Illustrative case. JOURNAL OF NEUROSURGERY. CASE LESSONS 2024; 8:CASE24242. [PMID: 39074389 DOI: 10.3171/case24242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/20/2024] [Indexed: 07/31/2024]
Abstract
BACKGROUND Meningiomas are the most prevalent primary central nervous system tumors. Although low-grade meningiomas are considered benign tumors, a subset of these can behave aggressively, showing progression and recurrence. In such cases, functional assays could influence treatment decisions and improve patient outcomes. OBSERVATIONS A 78-year-old female presented with a long-standing history of a supratentorial meningioma that was initially resected and treated with Gamma Knife radiosurgery. Surveillance revealed progression. She began systemic therapy with everolimus and octreotide but was lost to follow-up and did not continue the treatment. She returned because of a rapid decline in her neurological status. Biopsy with advanced molecular characterization by next-generation sequencing revealed NF2 and CREBBP mutations, and a commercial functional assay was done. This assay successfully isolated cancer stem cells (CSCs) from biopsy cores and identified potential drugs based on cellular sensitivity profiles. This is the first reported case in which a commercial functional drug screen was used for a meningioma. LESSONS In cases in which meningiomas exhibit specific genetic alterations and characteristics of aggressiveness, functional assays can be a useful tool for isolating CSCs. The authors report success in obtaining drug-screen profiling for a World Health Organization grade 1 meningioma. Multimodal approaches utilizing multi-omics analyses with functional assays can improve patient outcomes. https://thejns.org/doi/10.3171/CASE24242.
Collapse
Affiliation(s)
- William J Shelton
- Departments of Neurosurgery, University of Arkansas for Medical Sciences, College of Medicine, Little Rock, Arkansas
| | - Erika Santos Horta
- Departments of Medical Oncology, University of Arkansas for Medical Sciences, College of Medicine, Little Rock, Arkansas
| | - James Stephen Nix
- Departments of Pathology, University of Arkansas for Medical Sciences, College of Medicine, Little Rock, Arkansas
| | - Murat Gokden
- Departments of Pathology, University of Arkansas for Medical Sciences, College of Medicine, Little Rock, Arkansas
| | - Analiz Rodriguez
- Departments of Neurosurgery, University of Arkansas for Medical Sciences, College of Medicine, Little Rock, Arkansas
| |
Collapse
|
3
|
Vlashi R, Sun F, Zheng C, Zhang X, Liu J, Chen G. The molecular biology of NF2/Merlin on tumorigenesis and development. FASEB J 2024; 38:e23809. [PMID: 38967126 DOI: 10.1096/fj.202400019rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/06/2024]
Abstract
The neurofibromatosis type 2 (NF2) gene, known for encoding the tumor suppressor protein Merlin, is central to the study of tumorigenesis and associated cellular processes. This review comprehensively examines the multifaceted role of NF2/Merlin, detailing its structural characteristics, functional diversity, and involvement in various signaling pathways such as Wnt/β-catenin, Hippo, TGF-β, RTKs, mTOR, Notch, and Hedgehog. These pathways are crucial for cellular growth, proliferation, and differentiation. NF2 mutations are specifically linked to the development of schwannomas, meningiomas, and ependymomas, although the precise mechanisms of tumor formation in these specific cell types remain unclear. Additionally, the review explores Merlin's role in embryogenesis, highlighting the severe developmental defects and embryonic lethality caused by NF2 deficiency. The potential therapeutic strategies targeting these genetic aberrations are also discussed, emphasizing inhibitors of mTOR, HDAC, and VEGF as promising avenues for treatment. This synthesis of current knowledge underscores the necessity for ongoing research to elucidate the detailed mechanisms of NF2/Merlin and develop effective therapeutic strategies, ultimately aiming to improve the prognosis and quality of life for individuals with NF2 mutations.
Collapse
Affiliation(s)
- Rexhina Vlashi
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Fuju Sun
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Chenggong Zheng
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xingen Zhang
- Department of Orthopedics, Jiaxing Key Laboratory for Minimally Invasive Surgery in Orthopaedics & Skeletal Regenerative Medicine, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Jie Liu
- Department of Cancer Center, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, China
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
4
|
Nikanjam M, Wells K, Kato S, Adashek JJ, Block S, Kurzrock R. Reverse repurposing: Potential utility of cancer drugs in nonmalignant illnesses. MED 2024; 5:689-717. [PMID: 38749442 PMCID: PMC11246816 DOI: 10.1016/j.medj.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/03/2024] [Accepted: 04/18/2024] [Indexed: 06/02/2024]
Abstract
Growth and immune process dysregulation can result in both cancer and nonmalignant disease (hereditary or acquired, with and without predisposition to malignancy). Moreover, perhaps unexpectedly, many nonmalignant illnesses harbor genomic alterations indistinguishable from druggable oncogenic drivers. Therefore, targeted compounds used successfully to treat cancer may have therapeutic potential for nonmalignant conditions harboring the same target. MEK, PI3K/AKT/mTOR, fibroblast growth factor receptor (FGFR), and NRG1/ERBB pathway genes have all been implicated in both cancer and noncancerous conditions, and several cognate antagonists, as well as Bruton's tyrosine kinase inhibitors, JAK inhibitors, and CD20-directed antibodies, have established or theoretical therapeutic potential to bridge cancer and benign diseases. Intriguingly, pharmacologically tractable cancer drivers characterize a wide spectrum of disorders without malignant potential, including but not limited to Alzheimer's disease and a variety of other neurodegenerative conditions, rheumatoid arthritis, achondroplastic dwarfism, and endometriosis. Expanded repositioning of oncology agents in order to benefit benign but serious medical illnesses is warranted.
Collapse
Affiliation(s)
- Mina Nikanjam
- Division of Hematology-Oncology, University of California, San Diego, La Jolla, CA, USA.
| | - Kaitlyn Wells
- Department of Pharmacy, University of California, San Diego, La Jolla, CA, USA
| | - Shumei Kato
- Division of Hematology-Oncology, University of California, San Diego, La Jolla, CA, USA
| | - Jacob J Adashek
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - Shanna Block
- Department of Pharmacy, University of California, San Diego, La Jolla, CA, USA
| | - Razelle Kurzrock
- Division of Hematology-Oncology, Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA; WIN Consortium, Chevilly-Larue, France.
| |
Collapse
|
5
|
Martin P, Szkop KJ, Robert F, Bhattacharyya S, Beauchamp RL, Brenner J, Redmond NE, Huang S, Erdin S, Larsson O, Ramesh V. TSC2 loss in neural progenitor cells suppresses translation of ASD/NDD-associated transcripts in an mTORC1- and MNK1/2-reversible fashion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597393. [PMID: 38895292 PMCID: PMC11185676 DOI: 10.1101/2024.06.04.597393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Tuberous sclerosis complex (TSC) is an inherited neurodevelopmental disorder (NDD) with frequent manifestations of epilepsy and autism spectrum disorder (ASD). TSC is caused by inactivating mutations in TSC1 or TSC2 tumor suppressor genes, with encoded proteins hamartin (TSC1) and tuberin (TSC2) forming a functional complex inhibiting mechanistic target of rapamycin complex 1 (mTORC1) signaling. This has led to treatment with allosteric mTORC1 inhibitor rapamycin analogs ("rapalogs") for TSC tumors; however, rapalogs are ineffective for treating neurodevelopmental manifestations. mTORC1 signaling controls protein synthesis by regulating formation of the eIF4F complex, with further modulation by MNK1/2 kinases via phosphorylation of the eIF4F subunit eIF4E. While both these pathways modulate translation, comparing their impact on transcriptome-wide mRNA translation, as well as effects of inhibiting these pathways in TSC has not been explored. Here, employing CRISPR-modified, isogenic TSC2 patient-derived neural progenitor cells (NPCs), we have examined transcriptome-wide changes in mRNA translation upon TSC2 loss. Our results reveal dysregulated translation in TSC2 -Null NPCs, which significantly overlaps with the translatome from TSC1 -Null NPCs. Interestingly, numerous non-monogenic ASD-, NDD-and epilepsy-associated genes identified in patients harboring putative loss-of-function mutations, were translationally suppressed in TSC2 -Null NPCs. Importantly, translation of these ASD- and NDD-associated genes was reversed upon inhibition of either mTORC1 or MNK1/2 signaling using RMC-6272 or eFT-508, respectively. This study establishes the importance of mTORC1-eIF4F- and MNK-eIF4E-sensitive mRNA translation in TSC, ASD and other neurodevelopmental disorders laying the groundwork for evaluating drugs in clinical development that target these pathways as a treatment strategy for these disorders.
Collapse
|
6
|
Xu D, Yin S, Shu Y. NF2: An underestimated player in cancer metabolic reprogramming and tumor immunity. NPJ Precis Oncol 2024; 8:133. [PMID: 38879686 PMCID: PMC11180135 DOI: 10.1038/s41698-024-00627-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/02/2024] [Indexed: 06/19/2024] Open
Abstract
Neurofibromatosis type 2 (NF2) is a tumor suppressor gene implicated in various tumors, including mesothelioma, schwannomas, and meningioma. As a member of the ezrin, radixin, and moesin (ERM) family of proteins, merlin, which is encoded by NF2, regulates diverse cellular events and signalling pathways, such as the Hippo, mTOR, RAS, and cGAS-STING pathways. However, the biological role of NF2 in tumorigenesis has not been fully elucidated. Furthermore, cross-cancer mutations may exert distinct biological effects on tumorigenesis and treatment response. In addition to the functional inactivation of NF2, the codeficiency of other genes, such as cyclin-dependent kinase inhibitor 2A/B (CDKN2A/B), BRCA1-associated protein-1 (BAP1), and large tumor suppressor 2 (LATS2), results in unique tumor characteristics that should be considered in clinical treatment decisions. Notably, several recent studies have explored the metabolic and immunological features associated with NF2, offering potential insights into tumor biology and the development of innovative therapeutic strategies. In this review, we consolidate the current knowledge on NF2 and examine the potential connection between cancer metabolism and tumor immunity in merlin-deficient malignancies. This review may provide a deeper understanding of the biological roles of NF2 and guide possible therapeutic avenues.
Collapse
Affiliation(s)
- Duo Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shiyuan Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
7
|
Maruta H, He H. Rapamycin vs TORin-1 or Gleevec vs Nilotinib: Simple chemical evolution that converts PAK1-blockers to TOR-blockers or vice versa? Drug Discov Ther 2024; 18:134-139. [PMID: 38569833 DOI: 10.5582/ddt.2023.01097] [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] [Indexed: 04/05/2024]
Abstract
Both PAK1 (RAC/CDC42-activating kinase 1) and TOR (Target of Rapamycin) are among the major oncogenic/ageing kinases. However, they play the opposite role in our immune system, namely immune system is suppressed by PAK1, while it requires TOR. Thus, PAK1-blockers, would be more effective for therapy of cancers, than TOR-blockers. Since 2015 when we discovered genetically that PDGF-induced melanogenesis depends on "PAK1", we are able to screening a series of PAK1-blockers as melanogenesis-inhibitors which could eventually promote longevity. Interestingly, rapamycin, the first TOR-inhibitor, promotes melanogenesis, clearly indicating that TOR suppresses melanogenesis. However, a new TOR-inhibitor called TORin-1 no longer suppresses immune system, and blocks melanogenesis in cell culture. These observations strongly indicate that TORin-1 acts as PAK1-blockers, instead of TOR-blockers, in vivo. Thus, it is most likely that melanogenesis in cell culture could enable us to discriminate PAK1-blockers from TORblockers.
Collapse
Affiliation(s)
| | - Hong He
- Melbourne University Hospital (Austin Health), Melbourne, Australia
| |
Collapse
|
8
|
Yuan R, Wang B, Wang Y, Liu P. Gene Therapy for Neurofibromatosis Type 2-Related Schwannomatosis: Recent Progress, Challenges, and Future Directions. Oncol Ther 2024; 12:257-276. [PMID: 38760612 PMCID: PMC11187037 DOI: 10.1007/s40487-024-00279-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 04/30/2024] [Indexed: 05/19/2024] Open
Abstract
Neurofibromatosis type 2 (NF2)-related schwannomatosis is a rare autosomal dominant monogenic disorder caused by mutations in the NF2 gene. The hallmarks of NF2-related schwannomatosis are bilateral vestibular schwannomas (VS). The current treatment options for NF2-related schwannomatosis, such as observation with serial imaging, surgery, radiotherapy, and pharmacotherapies, have shown limited effectiveness and serious complications. Therefore, there is a critical demand for novel effective treatments. Gene therapy, which has made significant advancements in treating genetic diseases, holds promise for the treatment of this disease. This review covers the genetic pathogenesis of NF2-related schwannomatosis, the latest progress in gene therapy strategies, current challenges, and future directions of gene therapy for NF2-related schwannomatosis.
Collapse
Affiliation(s)
- Ruofei Yuan
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Bo Wang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Ying Wang
- Department of Neural Reconstruction, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Pinan Liu
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China.
- Department of Neural Reconstruction, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| |
Collapse
|
9
|
Chen Y, Steiner S, Hagedorn C, Kollar S, Pliego-Mendieta A, Haberecker M, Plock J, Britschgi C, Planas-Paz L, Pauli C. Acquired NF2 mutation confers resistance to TRK inhibition in an ex vivo LMNA::NTRK1-rearranged soft-tissue sarcoma cell model. J Pathol 2024; 263:257-269. [PMID: 38613194 DOI: 10.1002/path.6282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 03/05/2024] [Indexed: 04/14/2024]
Abstract
Genomic rearrangements of the neurotrophic receptor tyrosine kinase genes (NTRK1, NTRK2, and NTRK3) are the most common mechanism of oncogenic activation for this family of receptors, resulting in sustained cancer cell proliferation. Several targeted therapies have been approved for tumours harbouring NTRK fusions and a new generation of TRK inhibitors has already been developed due to acquired resistance. We established a patient-derived LMNA::NTRK1-rearranged soft-tissue sarcoma cell model ex vivo with an acquired resistance to targeted TRK inhibition. Molecular profiling of the resistant clones revealed an acquired NF2 loss of function mutation that was absent in the parental cell model. Parental cells showed continuous sensitivity to TRK-targeted treatment, whereas the resistant clones were insensitive. Furthermore, resistant clones showed upregulation of the MAPK and mTOR/AKT pathways in the gene expression based on RNA sequencing data and increased sensitivity to MEK and mTOR inhibitor therapy. Drug synergy was seen using trametinib and rapamycin in combination with entrectinib. Medium-throughput drug screening further identified small compounds as potential drug candidates to overcome resistance as monotherapy or in combination with entrectinib. In summary, we developed a comprehensive model of drug resistance in an LMNA::NTRK1-rearranged soft-tissue sarcoma and have broadened the understanding of acquired drug resistance to targeted TRK therapy. Furthermore, we identified drug combinations and small compounds to overcome acquired drug resistance and potentially guide patient care in a functional precision oncology setting. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Yanjiang Chen
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Sabrina Steiner
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Catherine Hagedorn
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Sarah Kollar
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Alicia Pliego-Mendieta
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Martina Haberecker
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Jan Plock
- Department of Plastic Surgery and Hand Surgery, Kantonsspital Aarau, Aarau, Switzerland
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Christian Britschgi
- Department of Hematology and Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Lara Planas-Paz
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Chantal Pauli
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
- Medical Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
10
|
Nghiemphu PL, Vitte J, Dombi E, Nguyen T, Wagle N, Ishiyama A, Sepahdari AR, Cachia D, Widemann BC, Brackmann DE, Doherty JK, Kalamarides M, Giovannini M. Imaging as an early biomarker to predict sensitivity to everolimus for progressive NF2-related vestibular schwannoma. J Neurooncol 2024; 167:339-348. [PMID: 38372904 PMCID: PMC11023969 DOI: 10.1007/s11060-024-04596-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 02/01/2024] [Indexed: 02/20/2024]
Abstract
PURPOSE NF2-related schwannomatosis (NF2) is characterized by bilateral vestibular schwannomas (VS) often causing hearing and neurologic deficits, with currently no FDA-approved drug treatment. Pre-clinical studies highlighted the potential of mTORC1 inhibition in delaying schwannoma progression. We conducted a prospective open-label, phase II study of everolimus for progressive VS in NF2 patients and investigated imaging as a potential biomarker predicting effects on growth trajectory. METHODS The trial enrolled 12 NF2 patients with progressive VS. Participants received oral everolimus daily for 52 weeks. Brain imaging was obtained quarterly. As primary endpoint, radiographic response (RR) was defined as ≥ 20% decrease in target VS volume. Secondary endpoints included other tumors RR, hearing outcomes, drug safety and quality of life (QOL). RESULTS Eight participants completed the trial and four discontinued the drug early due to significant volumetric VS progression. After 52 weeks of treatment, the median annual VS growth rate decreased from 77.2% at baseline to 29.4%. There was no VS RR and 3 of 8 (37.5%) participants had stable disease. Decreased or unchanged VS volume after 3 months of treatment was predictive of stabilization at 12 months. Seven of eight participants had stable hearing during treatment except one with a decline in word recognition score. Ten of twelve participants reported only minimal changes to their QOL scores. CONCLUSIONS Volumetric imaging at 3 months can serve as an early biomarker to predict long-term sensitivity to everolimus treatment. Everolimus may represent a safe treatment option to decrease the growth of NF2-related VS in patients who have stable hearing and neurological condition. TRN: NCT01345136 (April 29, 2011).
Collapse
Affiliation(s)
- Phioanh Leia Nghiemphu
- Department of Neurology, UCLA Neuro‑Oncology Program, David Geffen School of Medicine and Jonsson Comprehensive Cancer Center (JCCC), University of California, Los Angeles, Los Angeles, CA, USA
| | - Jeremie Vitte
- Department of Head and Neck Surgery, David Geffen School of Medicine and Jonsson Comprehensive Cancer Center (JCCC), University of California, Los Angeles, 675 Charles E Young Dr. S, MRL 2240, Los Angeles, CA, 90095-7286, USA
| | - Eva Dombi
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Thien Nguyen
- Department of Neurology, UCLA Neuro‑Oncology Program, David Geffen School of Medicine and Jonsson Comprehensive Cancer Center (JCCC), University of California, Los Angeles, Los Angeles, CA, USA
- Department of Pediatrics, Division of Pediatric Hematology, Oncology, Stem Cell Transplant and Regenerative Medicine, Stanford University, Palo Alto, CA, USA
| | - Naveed Wagle
- Department of Medicine, Division of Medical Oncology, Norris Cancer Center, University of Southern California, Los Angeles, CA, USA
- Department of Translational Neurosciences, Saint John's Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Akira Ishiyama
- Department of Head and Neck Surgery, David Geffen School of Medicine and Jonsson Comprehensive Cancer Center (JCCC), University of California, Los Angeles, 675 Charles E Young Dr. S, MRL 2240, Los Angeles, CA, 90095-7286, USA
| | - Ali R Sepahdari
- Department of Radiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Diagnostic Neuroradiology, Scripps Clinic Medical Group, La Jolla, CA, USA
| | - David Cachia
- Department of Neurosurgery, Division of Neuro-oncology, Medical University of South Carolina, Charleston, SC, USA
- Department of Medicine, Division of Hematology/Oncology, University of Massachusetts, Worcester, MA, USA
| | | | - Derald E Brackmann
- Department of Otolaryngology and Neurotology, House Clinic and Research Institute, Los Angeles, CA, USA
| | - Joni K Doherty
- Center for Neural Tumor Research, House Research Institute, Los Angeles, CA, USA
- Department of Otolaryngology - Head and Neck Surgery, University of Southern California, Los Angeles, CA, USA
| | - Michel Kalamarides
- Department of Neurosurgery, Hôpital Pitié-Salpêtrière, APHP, Sorbonne Université, Paris, France
| | - Marco Giovannini
- Department of Head and Neck Surgery, David Geffen School of Medicine and Jonsson Comprehensive Cancer Center (JCCC), University of California, Los Angeles, 675 Charles E Young Dr. S, MRL 2240, Los Angeles, CA, 90095-7286, USA.
| |
Collapse
|
11
|
Szulzewsky F, Thirimanne HN, Holland EC. Meningioma: current updates on genetics, classification, and mouse modeling. Ups J Med Sci 2024; 129:10579. [PMID: 38571886 PMCID: PMC10989216 DOI: 10.48101/ujms.v129.10579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 04/05/2024] Open
Abstract
Meningiomas, the most common primary brain tumors in adults, are often benign and curable by surgical resection. However, a subset is of higher grade, shows aggressive growth behavior as well as brain invasion, and often recurs even after several rounds of surgery. Increasing evidence suggests that tumor classification and grading primarily based on histopathology do not always accurately predict tumor aggressiveness and recurrence behavior. The underlying biology of aggressive treatment-resistant meningiomas and the impact of specific genetic aberrations present in these high-grade tumors is still only insufficiently understood. Therefore, an in-depth research into the biology of this tumor type is warranted. More recent studies based on large-scale molecular data such as whole exome/genome sequencing, DNA methylation sequencing, and RNA sequencing have provided new insights into the biology of meningiomas and have revealed new risk factors and prognostic subtypes. The most common genetic aberration in meningiomas is functional loss of NF2 and occurs in both low- and high-grade meningiomas, whereas NF2-wildtype meningiomas are enriched for recurrent mutations in TRAF7, KLF4, AKT1, PI3KCA, and SMO and are more frequently benign. Most meningioma mouse models are based on patient-derived xenografts and only recently have new genetically engineered mouse models of meningioma been developed that will aid in the systematic evaluation of specific mutations found in meningioma and their impact on tumor behavior. In this article, we review recent advances in the understanding of meningioma biology and classification and highlight the most common genetic mutations, as well as discuss new genetically engineered mouse models of meningioma.
Collapse
Affiliation(s)
- Frank Szulzewsky
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Eric C. Holland
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Seattle Translational Tumor Research Center, Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
12
|
Febres-Aldana CA, Fanaroff R, Offin M, Zauderer MG, Sauter JL, Yang SR, Ladanyi M. Diffuse Pleural Mesothelioma: Advances in Molecular Pathogenesis, Diagnosis, and Treatment. ANNUAL REVIEW OF PATHOLOGY 2024; 19:11-42. [PMID: 37722697 DOI: 10.1146/annurev-pathol-042420-092719] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/20/2023]
Abstract
Diffuse pleural mesothelioma (DPM) is a highly aggressive malignant neoplasm arising from the mesothelial cells lining the pleural surfaces. While DPM is a well-recognized disease linked to asbestos exposure, recent advances have expanded our understanding of molecular pathogenesis and transformed our clinical practice. This comprehensive review explores the current concepts and emerging trends in DPM, including risk factors, pathobiology, histologic subtyping, and therapeutic management, with an emphasis on a multidisciplinary approach to this complex disease.
Collapse
Affiliation(s)
- Christopher A Febres-Aldana
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA; ,
| | - Rachel Fanaroff
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA; ,
| | - Michael Offin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Marjorie G Zauderer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jennifer L Sauter
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA; ,
| | - Soo-Ryum Yang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA; ,
| | - Marc Ladanyi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA; ,
| |
Collapse
|
13
|
Matta SK, Kohio HP, Chandra P, Brown A, Doench JG, Philips JA, Ding S, Sibley LD. Genome-wide and targeted CRISPR screens identify RNF213 as a mediator of interferon gamma-dependent pathogen restriction in human cells. Proc Natl Acad Sci U S A 2024; 121:e2315865120. [PMID: 38147552 PMCID: PMC10769850 DOI: 10.1073/pnas.2315865120] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/15/2023] [Indexed: 12/28/2023] Open
Abstract
To define cellular immunity to the intracellular pathogen Toxoplasma gondii, we performed a genome-wide CRISPR loss-of-function screen to identify genes important for (interferon gamma) IFN-γ-dependent growth restriction. We revealed a role for the tumor suppressor NF2/Merlin for maximum induction of Interferon Stimulated Genes (ISG), which are positively regulated by the transcription factor IRF-1. We then performed an ISG-targeted CRISPR screen that identified the host E3 ubiquitin ligase RNF213 as necessary for IFN-γ-mediated control of T. gondii in multiple human cell types. RNF213 was also important for control of bacterial (Mycobacterium tuberculosis) and viral (Vesicular Stomatitis Virus) pathogens in human cells. RNF213-mediated ubiquitination of the parasitophorous vacuole membrane (PVM) led to growth restriction of T. gondii in response to IFN-γ. Moreover, overexpression of RNF213 in naive cells also impaired growth of T. gondii. Surprisingly, growth inhibition did not require the autophagy protein ATG5, indicating that RNF213 initiates restriction independent of a previously described noncanonical autophagy pathway. Mutational analysis revealed that the ATPase domain of RNF213 was required for its recruitment to the PVM, while loss of a critical histidine in the RZ finger domain resulted in partial reduction of recruitment to the PVM and complete loss of ubiquitination. Both RNF213 mutants lost the ability to restrict growth of T. gondii, indicating that both recruitment and ubiquitination are required. Collectively, our findings establish RNF213 as a critical component of cell-autonomous immunity that is both necessary and sufficient for control of intracellular pathogens in human cells.
Collapse
Affiliation(s)
- Sumit K. Matta
- Department of Molecular Microbiology, School of Medicine, Washington University in St. Louis, St Louis, MO63130
| | - Hinissan P. Kohio
- Department of Molecular Microbiology, School of Medicine, Washington University in St. Louis, St Louis, MO63130
| | - Pallavi Chandra
- Department of Medicine, Division of Infectious Diseases, School of Medicine, Washington University in St. Louis, St Louis, MO63130
| | - Adam Brown
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA02142
| | - John G. Doench
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA02142
| | - Jennifer A. Philips
- Department of Molecular Microbiology, School of Medicine, Washington University in St. Louis, St Louis, MO63130
- Department of Medicine, Division of Infectious Diseases, School of Medicine, Washington University in St. Louis, St Louis, MO63130
| | - Siyuan Ding
- Department of Molecular Microbiology, School of Medicine, Washington University in St. Louis, St Louis, MO63130
| | - L. David Sibley
- Department of Molecular Microbiology, School of Medicine, Washington University in St. Louis, St Louis, MO63130
| |
Collapse
|
14
|
Bhattacharyya S, Oblinger JL, Beauchamp RL, Kosa L, Robert F, Plotkin SR, Chang LS, Ramesh V. Preclinical evaluation of the third-generation, bi-steric mechanistic target of rapamycin complex 1-selective inhibitor RMC-6272 in NF2-deficient models. Neurooncol Adv 2024; 6:vdae024. [PMID: 38476930 PMCID: PMC10929445 DOI: 10.1093/noajnl/vdae024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024] Open
Abstract
Background NF2-associated meningiomas are progressive, highly morbid, and nonresponsive to chemotherapies, highlighting the need for improved treatments. We have established aberrant activation of the mechanistic target of rapamycin (mTOR) signaling in NF2-deficient tumors, leading to clinical trials with first- and second-generation mTOR inhibitors. However, results have been mixed, showing stabilized tumor growth without shrinkage offset by adverse side effects. To address these limitations, here we explored the potential of third-generation, bi-steric mTOR complex 1 (mTORC1) inhibitors using the preclinical tool compound RMC-6272. Methods Employing human NF2-deficient meningioma lines, we compared mTOR inhibitors rapamycin (first-generation), INK128 (second-generation), and RMC-6272 (third-generation) using in vitro dose-response testing, cell-cycle analysis, and immunoblotting. Furthermore, the efficacy of RMC-6272 was assessed in NF2-null 3D-spheroid meningioma models, and its in vivo potential was evaluated in 2 orthotopic meningioma mouse models. Results Treatment of meningioma cells revealed that, unlike rapamycin, RMC-6272 demonstrated superior growth inhibitory effects, cell-cycle arrest, and complete inhibition of phosphorylated 4E-BP1 (mTORC1 readout). Moreover, RMC-6272 had a longer retention time than INK128 and inhibited the expression of several eIF4E-sensitive targets on the protein level. RMC-6272 treatment of NF2 spheroids showed significant shrinkage in size as well as reduced proliferation. Furthermore, in vivo studies in mice revealed effective blockage of meningioma growth by RMC-6272, compared with vehicle controls. Conclusions Our study in preclinical models of NF2 supports possible future clinical evaluation of third-generation, investigational mTORC1 inhibitors, such as RMC-5552, as a potential treatment strategy for NF2.
Collapse
Affiliation(s)
- Srirupa Bhattacharyya
- Department of Neurology and Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Janet L Oblinger
- Center for Childhood Cancer, 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, Massachusetts, USA
| | - Lili Kosa
- Department of Neurology and Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Francis Robert
- Department of Biochemistry and Goodman Cancer Research Institute, McGill University, Montreal, Quebec, Canada
| | - Scott R Plotkin
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Long-Sheng Chang
- Center for Childhood Cancer, 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, Massachusetts, USA
| |
Collapse
|
15
|
Nozzoli F, Buccoliero AM, Massi D, Santoro R, Pecci R. External auditory canal ectopic atypical meningioma: A case report and brief literature review. Pathol Res Pract 2024; 253:154963. [PMID: 38029716 DOI: 10.1016/j.prp.2023.154963] [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: 10/09/2023] [Revised: 11/16/2023] [Accepted: 11/18/2023] [Indexed: 12/01/2023]
Abstract
Meningiomas are tumours typically derived from the meningothelial cells of the arachnoid mater. They most often arise in intracranial, intraspinal, or orbital locations. Ectopic meningiomas, described as primary meningiomas with no intracranial involvement, are definitely unconventional. In fact, most of the extracranial meningiomas described in the literature, particularly in the outer ear, are effectively spreads of disease with primary intracranial localization. We describe a case of a primary external auditory canal meningioma with demonstrated absence of intracranial involvement, and we provide a full radiological, histological, immunohistochemical and molecular characterization of the lesion.
Collapse
Affiliation(s)
- Filippo Nozzoli
- Section of Anatomic Pathology, Department of Health Sciences, University of Florence, Florence, Italy.
| | | | - Daniela Massi
- Section of Anatomic Pathology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Roberto Santoro
- Audiology and Robotic Oncologic Head and Neck Surgery, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Rudi Pecci
- Audiology and Robotic Oncologic Head and Neck Surgery, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
16
|
Jovanović A, Tošić N, Marjanović I, Komazec J, Zukić B, Nikitović M, Ilić R, Grujičić D, Janić D, Pavlović S. Germline Variants in Cancer Predisposition Genes in Pediatric Patients with Central Nervous System Tumors. Int J Mol Sci 2023; 24:17387. [PMID: 38139220 PMCID: PMC10744041 DOI: 10.3390/ijms242417387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/04/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Central nervous system (CNS) tumors comprise around 20% of childhood malignancies. Germline variants in cancer predisposition genes (CPGs) are found in approximately 10% of pediatric patients with CNS tumors. This study aimed to characterize variants in CPGs in pediatric patients with CNS tumors and correlate these findings with clinically relevant data. Genomic DNA was isolated from the peripheral blood of 51 pediatric patients and further analyzed by the next-generation sequencing approach. Bioinformatic analysis was done using an "in-house" gene list panel, which included 144 genes related to pediatric brain tumors, and the gene list panel Neoplasm (HP:0002664). Our study found that 27% of pediatric patients with CNS tumors have a germline variant in some of the known CPGs, like ALK, APC, CHEK2, ELP1, MLH1, MSH2, NF1, NF2 and TP53. This study represents the first comprehensive evaluation of germline variants in pediatric patients with CNS tumors in the Western Balkans region. Our results indicate the necessity of genomic research to reveal the genetic basis of pediatric CNS tumors, as well as to define targets for the application and development of innovative therapeutics that form the basis of the upcoming era of personalized medicine.
Collapse
Affiliation(s)
- Aleksa Jovanović
- Pediatric Oncology Department, National Cancer Research Center, 11000 Belgrade, Serbia; (A.J.); (D.J.)
| | - Nataša Tošić
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042 Belgrade, Serbia; (N.T.); (I.M.); (J.K.); (B.Z.)
| | - Irena Marjanović
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042 Belgrade, Serbia; (N.T.); (I.M.); (J.K.); (B.Z.)
| | - Jovana Komazec
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042 Belgrade, Serbia; (N.T.); (I.M.); (J.K.); (B.Z.)
| | - Branka Zukić
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042 Belgrade, Serbia; (N.T.); (I.M.); (J.K.); (B.Z.)
| | - Marina Nikitović
- Pediatric Radiation Oncology Department, National Cancer Research Center, 11000 Belgrade, Serbia;
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (R.I.); (D.G.)
| | - Rosanda Ilić
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (R.I.); (D.G.)
- Neurooncology Department, Neurosurgery Clinic, University Clinical Centre of Serbia, 11000 Belgrade, Serbia
| | - Danica Grujičić
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (R.I.); (D.G.)
- Neurooncology Department, Neurosurgery Clinic, University Clinical Centre of Serbia, 11000 Belgrade, Serbia
| | - Dragana Janić
- Pediatric Oncology Department, National Cancer Research Center, 11000 Belgrade, Serbia; (A.J.); (D.J.)
| | - Sonja Pavlović
- Laboratory for Molecular Biomedicine, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042 Belgrade, Serbia; (N.T.); (I.M.); (J.K.); (B.Z.)
| |
Collapse
|
17
|
Halabi R, Dakroub F, Haider MZ, Patel S, Amhaz NA, Reslan MA, Eid AH, Mechref Y, Darwiche N, Kobeissy F, Omeis I, Shaito AA. Unveiling a Biomarker Signature of Meningioma: The Need for a Panel of Genomic, Epigenetic, Proteomic, and RNA Biomarkers to Advance Diagnosis and Prognosis. Cancers (Basel) 2023; 15:5339. [PMID: 38001599 PMCID: PMC10670806 DOI: 10.3390/cancers15225339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Meningiomas are the most prevalent primary intracranial tumors. The majority are benign but can undergo dedifferentiation into advanced grades classified by World Health Organization (WHO) into Grades 1 to 3. Meningiomas' tremendous variability in tumor behavior and slow growth rates complicate their diagnosis and treatment. A deeper comprehension of the molecular pathways and cellular microenvironment factors implicated in meningioma survival and pathology is needed. This review summarizes the known genetic and epigenetic aberrations involved in meningiomas, with a focus on neurofibromatosis type 2 (NF2) and non-NF2 mutations. Novel potential biomarkers for meningioma diagnosis and prognosis are also discussed, including epigenetic-, RNA-, metabolomics-, and protein-based markers. Finally, the landscape of available meningioma-specific animal models is overviewed. Use of these animal models can enable planning of adjuvant treatment, potentially assisting in pre-operative and post-operative decision making. Discovery of novel biomarkers will allow, in combination with WHO grading, more precise meningioma grading, including meningioma identification, subtype determination, and prediction of metastasis, recurrence, and response to therapy. Moreover, these biomarkers may be exploited in the development of personalized targeted therapies that can distinguish between the 15 diverse meningioma subtypes.
Collapse
Affiliation(s)
- Reem Halabi
- Department of Biological and Chemical Sciences, Lebanese International University, Beirut 1105, Lebanon;
| | - Fatima Dakroub
- Department of Experimental Pathology, Microbiology and Immunology and Center for Infectious Diseases Research, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon;
| | - Mohammad Z. Haider
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (M.Z.H.); (A.H.E.)
| | - Stuti Patel
- Department of Biology, University of Florida, Gainesville, FL 32601, USA; (S.P.); (N.A.A.)
| | - Nayef A. Amhaz
- Department of Biology, University of Florida, Gainesville, FL 32601, USA; (S.P.); (N.A.A.)
| | - Mohammad A. Reslan
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut 1107, Lebanon; (M.A.R.); (N.D.); (F.K.)
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (M.Z.H.); (A.H.E.)
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409, USA;
| | - Nadine Darwiche
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut 1107, Lebanon; (M.A.R.); (N.D.); (F.K.)
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut 1107, Lebanon; (M.A.R.); (N.D.); (F.K.)
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Ibrahim Omeis
- Hammoud Hospital University Medical Center, Saida 652, Lebanon
- Division of Neurosurgery, Penn Medicine, Lancaster General Health, Lancaster, PA 17601, USA
| | - Abdullah A. Shaito
- Biomedical Research Center, College of Medicine, and Department of Biomedical Sciences at College of Health Sciences, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
18
|
Mohamed T, Melfi V, Colciago A, Magnaghi V. Hearing loss and vestibular schwannoma: new insights into Schwann cells implication. Cell Death Dis 2023; 14:629. [PMID: 37741837 PMCID: PMC10517973 DOI: 10.1038/s41419-023-06141-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/25/2023]
Abstract
Hearing loss (HL) is the most common and heterogeneous disorder of the sensory system, with a large morbidity in the worldwide population. Among cells of the acoustic nerve (VIII cranial nerve), in the cochlea are present the hair cells, the spiral ganglion neurons, the glia-like supporting cells, and the Schwann cells (SCs), which alterations have been considered cause of HL. Notably, a benign SC-derived tumor of the acoustic nerve, named vestibular schwannoma (VS), has been indicated as cause of HL. Importantly, SCs are the main glial cells ensheathing axons and forming myelin in the peripheral nerves. Following an injury, the SCs reprogram, expressing some stemness features. Despite the mechanisms and factors controlling their biological processes (i.e., proliferation, migration, differentiation, and myelination) have been largely unveiled, their role in VS and HL was poorly investigated. In this review, we enlighten some of the mechanisms at the base of SCs transformation, VS development, and progression, likely leading to HL, and we pose great attention on the environmental factors that, in principle, could contribute to HL onset or progression. Combining the biomolecular bench-side approach to the clinical bedside practice may be helpful for the diagnosis, prediction, and therapeutic approach in otology.
Collapse
Affiliation(s)
- Tasnim Mohamed
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Valentina Melfi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Via G. Balzaretti 9, 20133, Milan, Italy
- Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133, Milan, Italy
| | - Alessandra Colciago
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Valerio Magnaghi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Via G. Balzaretti 9, 20133, Milan, Italy.
| |
Collapse
|
19
|
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: 0] [Impact Index Per Article: 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
|
20
|
Le TPH, Nguyen NTT, Le DDT, Anwar MA, Lee SY. Lipid kinase PIP5Kα contributes to Hippo pathway activation via interaction with Merlin and by mediating plasma membrane targeting of LATS1. Cell Commun Signal 2023; 21:149. [PMID: 37337213 DOI: 10.1186/s12964-023-01161-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/07/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND The Hippo pathway plays a critical role in controlled cell proliferation. The tumor suppressor Merlin and large tumor suppressor kinase 1 (LATS1) mediate activation of Hippo pathway, consequently inhibiting the primary effectors, Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ). Phosphatidylinositol 4,5-bisphosphate (PIP2), a lipid present in the plasma membrane (PM), binds to and activates Merlin. Phosphatidylinositol 4-phosphate 5-kinase α (PIP5Kα) is an enzyme responsible for PIP2 production. However, the functional role of PIP5Kα in regulation of Merlin and LATS1 under Hippo signaling conditions remains unclear. METHODS PIP5Kα, Merlin, or LATS1 knockout or knockdown cells and transfected cells with them were used. LATS1, YAP, and TAZ activities were measured using biochemical methods and PIP2 levels were evaluated using cell imaging. Low/high cell density and serum starvation/stimulation conditions were tested. Colocalization of PIP5Kα and PIP2 with Merlin and LATS1, and their protein interactions were examined using transfection, confocal imaging, immunoprecipitation, western blotting, and/or pull-down experiments. Colony formation and adipocyte differentiation assays were performed. RESULTS We found that PIP5Kα induced LATS1 activation and YAP/TAZ inhibition in a kinase activity-dependent manner. Consistent with these findings, PIP5Kα suppressed cell proliferation and enhanced adipocyte differentiation of mesenchymal stem cells. Moreover, PIP5Kα protein stability and PIP2 levels were elevated at high cell density compared with those at low cell density, and both PIP2 and YAP phosphorylation levels initially declined, then recovered upon serum stimulation. Under these conditions, YAP/TAZ activity was aberrantly regulated by PIP5Kα deficiency. Mechanistically, either Merlin deficiency or LATS1 deficiency abrogated PIP5Kα-mediated YAP/TAZ inactivation. Additionally, the catalytic domain of PIP5Kα directly interacted with the band 4.1/ezrin/radixin/moesin domain of Merlin, and this interaction reinforced interaction of Merlin with LATS1. In accordance with these findings, PIP5Kα and PIP2 colocalized with Merlin and LATS1 in the PM. In PIP5Kα-deficient cells, Merlin colocalization with PIP2 was reduced, and LATS1 solubility increased. CONCLUSIONS Collectively, our results support that PIP5Kα serves as an activator of the Hippo pathway through interaction and colocalization with Merlin, which promotes PIP2-dependent Merlin activation and induces local recruitment of LATS1 to the PIP2-rich PM and its activation, thereby negatively regulating YAP/TAZ activity. Video Abstract.
Collapse
Affiliation(s)
- Truc Phan Hoang Le
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Gyeonggi, 16499, Republic of Korea
| | - Nga Thi Thanh Nguyen
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Gyeonggi, 16499, Republic of Korea
| | - Duong Duy Thai Le
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Gyeonggi, 16499, Republic of Korea
| | - Muhammad Ayaz Anwar
- Department of Applied Chemistry, Kyung Hee University International Campus, Yongin, Gyeonggi, 17104, Republic of Korea
| | - Sang Yoon Lee
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Gyeonggi, 16499, Republic of Korea.
- Institute of Medical Science, Ajou University School of Medicine, Suwon, Gyeonggi, 16499, Republic of Korea.
| |
Collapse
|
21
|
Pan F, Zhu M, Liang Y, Yuan C, Zhang Y, Wang Y, Fan H, Waldor MK, Ma Z. Membrane vesicle delivery of a streptococcal M protein disrupts the blood-brain barrier by inducing autophagic endothelial cell death. Proc Natl Acad Sci U S A 2023; 120:e2219435120. [PMID: 37276410 PMCID: PMC10268326 DOI: 10.1073/pnas.2219435120] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 05/08/2023] [Indexed: 06/07/2023] Open
Abstract
M family proteins are critical virulence determinants of Streptococci. Streptococcus equi subsp. zooepidemicus (SEZ) are Group C streptococci that cause meningitis in animals and humans. SzM, the M protein of SEZ, has been linked to SEZ brain invasion. Here, we demonstrate that SzM is important in SEZ disruption of the blood-brain barrier (BBB). SEZ release SzM-bound membrane vesicles (MVs), and endocytosis of these vesicles by human brain endothelial microvascular cells (hBMECs) results in SzM-dependent cytotoxicity. Furthermore, administration of SzM-bound MVs disrupted the murine BBB. A CRISPR screen revealed that SzM cytotoxicity in hBMECs depends on PTEN-related activation of autophagic cell death. Pharmacologic inhibition of PTEN activity prevented SEZ disruption of the murine BBB and delayed mortality. Our data show that MV delivery of SzM to host cells plays a key role in SEZ pathogenicity and suggests that MV delivery of streptococcal M family proteins is likely a common streptococcal virulence mechanism.
Collapse
Affiliation(s)
- Fei Pan
- Ministry of Agriculture Key Laboratory of Animal Bacteriology, the International Joint Laboratory of Animal Health and Food Safety, and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu210095, China
| | - Mingli Zhu
- Ministry of Agriculture Key Laboratory of Animal Bacteriology, the International Joint Laboratory of Animal Health and Food Safety, and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu210095, China
| | - Ying Liang
- Ministry of Agriculture Key Laboratory of Animal Bacteriology, the International Joint Laboratory of Animal Health and Food Safety, and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu210095, China
| | - Chen Yuan
- Ministry of Agriculture Key Laboratory of Animal Bacteriology, the International Joint Laboratory of Animal Health and Food Safety, and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu210095, China
| | - Yu Zhang
- Ministry of Agriculture Key Laboratory of Animal Bacteriology, the International Joint Laboratory of Animal Health and Food Safety, and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu210095, China
| | - Yuchang Wang
- Ministry of Agriculture Key Laboratory of Animal Bacteriology, the International Joint Laboratory of Animal Health and Food Safety, and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu210095, China
| | - Hongjie Fan
- Ministry of Agriculture Key Laboratory of Animal Bacteriology, the International Joint Laboratory of Animal Health and Food Safety, and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu210095, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou225009, China
| | - Matthew K. Waldor
- HHMI, Boston, MA02115
- Brigham and Women’s HospitalDivision of Infectious Diseases, Boston, MA02115
- Department of Microbiology, Harvard Medical School, Boston, MA02115
| | - Zhe Ma
- Ministry of Agriculture Key Laboratory of Animal Bacteriology, the International Joint Laboratory of Animal Health and Food Safety, and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu210095, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou225009, China
| |
Collapse
|
22
|
Nourbakhsh A, Dinh CT. Updates on Tumor Biology in Vestibular Schwannoma. Otolaryngol Clin North Am 2023; 56:421-434. [PMID: 37121611 DOI: 10.1016/j.otc.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Vestibular schwannomas (VSs) are benign tumors that develop after biallelic inactivation of the neurofibromatosis type 2 (NF2) gene that encodes the tumor suppressor merlin. Merlin inactivation leads to cell proliferation by dysregulation of receptor tyrosine kinase signaling and other intracellular pathways. In VS without NF2 mutations, dysregulation of non-NF2 genes can promote pathways favoring cell proliferation and tumorigenesis. The tumor microenvironment of VS consists of multiple cell types that influence VS tumor biology through complex intercellular networking and communications.
Collapse
Affiliation(s)
- Aida Nourbakhsh
- Department of Otolaryngology, University of Miami Miller School of Medicine, 1120 Northwest 14th Street, Suite 579, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, 1475 Northwest 12th Avenue, Miami, FL 33136, USA
| | - Christine T Dinh
- Department of Otolaryngology, University of Miami Miller School of Medicine, 1120 Northwest 14th Street, Suite 579, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, 1475 Northwest 12th Avenue, Miami, FL 33136, USA.
| |
Collapse
|
23
|
Ghalavand MA, Asghari A, Farhadi M, Taghizadeh-Hesary F, Garshasbi M, Falah M. The genetic landscape and possible therapeutics of neurofibromatosis type 2. Cancer Cell Int 2023; 23:99. [PMID: 37217995 DOI: 10.1186/s12935-023-02940-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 05/07/2023] [Indexed: 05/24/2023] Open
Abstract
Neurofibromatosis type 2 (NF2) is a genetic condition marked by the development of multiple benign tumors in the nervous system. The most common tumors associated with NF2 are bilateral vestibular schwannoma, meningioma, and ependymoma. The clinical manifestations of NF2 depend on the site of involvement. Vestibular schwannoma can present with hearing loss, dizziness, and tinnitus, while spinal tumor leads to debilitating pain, muscle weakness, or paresthesias. Clinical diagnosis of NF2 is based on the Manchester criteria, which have been updated in the last decade. NF2 is caused by loss-of-function mutations in the NF2 gene on chromosome 22, leading the merlin protein to malfunction. Over half of NF2 patients have de novo mutations, and half of this group are mosaic. NF2 can be managed by surgery, stereotactic radiosurgery, monoclonal antibody bevacizumab, and close observation. However, the nature of multiple tumors and the necessity of multiple surgeries over the lifetime, inoperable tumors like meningiomatosis with infiltration of the sinus or in the area of the lower cranial nerves, the complications caused by the operation, the malignancies induced by radiotherapy, and inefficiency of cytotoxic chemotherapy due to the benign nature of NF-related tumors have led a march toward exploring targeted therapies. Recent advances in genetics and molecular biology have allowed identifying and targeting of underlying pathways in the pathogenesis of NF2. In this review, we explain the clinicopathological characteristics of NF2, its genetic and molecular background, and the current knowledge and challenges of implementing genetics to develop efficient therapies.
Collapse
Affiliation(s)
- Mohammad Amin Ghalavand
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Alimohamad Asghari
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Skull Base Research Center, The Five Senses Health Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Farhadi
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Radiation Oncology Department, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Garshasbi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Masoumeh Falah
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Chiasson-MacKenzie C, Vitte J, Liu CH, Wright EA, Flynn EA, Stott SL, Giovannini M, McClatchey AI. Cellular mechanisms of heterogeneity in NF2-mutant schwannoma. Nat Commun 2023; 14:1559. [PMID: 36944680 PMCID: PMC10030849 DOI: 10.1038/s41467-023-37226-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 03/06/2023] [Indexed: 03/23/2023] Open
Abstract
Schwannomas are common sporadic tumors and hallmarks of familial neurofibromatosis type 2 (NF2) that develop predominantly on cranial and spinal nerves. Virtually all schwannomas result from inactivation of the NF2 tumor suppressor gene with few, if any, cooperating mutations. Despite their genetic uniformity schwannomas exhibit remarkable clinical and therapeutic heterogeneity, which has impeded successful treatment. How heterogeneity develops in NF2-mutant schwannomas is unknown. We have found that loss of the membrane:cytoskeleton-associated NF2 tumor suppressor, merlin, yields unstable intrinsic polarity and enables Nf2-/- Schwann cells to adopt distinct programs of ErbB ligand production and polarized signaling, suggesting a self-generated model of schwannoma heterogeneity. We validated the heterogeneous distribution of biomarkers of these programs in human schwannoma and exploited the synchronous development of lesions in a mouse model to establish a quantitative pipeline for studying how schwannoma heterogeneity evolves. Our studies highlight the importance of intrinsic mechanisms of heterogeneity across human cancers.
Collapse
Affiliation(s)
- Christine Chiasson-MacKenzie
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 149 13th Street, Charlestown, MA, 02129, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Jeremie Vitte
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA and Jonsson Comprehensive Cancer Center (JCCC), University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Ching-Hui Liu
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 149 13th Street, Charlestown, MA, 02129, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Emily A Wright
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 149 13th Street, Charlestown, MA, 02129, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Elizabeth A Flynn
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 149 13th Street, Charlestown, MA, 02129, USA
- Center for Engineering in Medicine and BioMEMS Resource Center, Surgical Services, Massachusetts General Hospital, Harvard Medical School, 114 16th Street, Charlestown, MA, 02129, USA
| | - Shannon L Stott
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 149 13th Street, Charlestown, MA, 02129, USA
- Center for Engineering in Medicine and BioMEMS Resource Center, Surgical Services, Massachusetts General Hospital, Harvard Medical School, 114 16th Street, Charlestown, MA, 02129, USA
| | - Marco Giovannini
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA and Jonsson Comprehensive Cancer Center (JCCC), University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Andrea I McClatchey
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 149 13th Street, Charlestown, MA, 02129, USA.
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
| |
Collapse
|
25
|
Graillon T, Tabouret E, Salgues B, Horowitz T, Padovani L, Appay R, Farah K, Dufour H, Régis J, Guedj E, Barlier A, Chinot O. Innovative treatments for meningiomas. Rev Neurol (Paris) 2023; 179:449-463. [PMID: 36959063 DOI: 10.1016/j.neurol.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/25/2023]
Abstract
Multi-recurrent high-grade meningiomas remain an unmet medical need in neuro-oncology when iterative surgeries and radiation therapy sessions fail to control tumor growth. Nevertheless, the last 10years have been marked by multiple advances in the comprehension of meningioma tumorigenesis via the discovery of new driver mutations, the identification of activated intracellular signaling pathways, and DNA methylation analyses, providing multiple potential therapeutic targets. Today, Anti-VEGF and mTOR inhibitors are the most used and probably the most active drugs in aggressive meningiomas. Peptide radioactive radiation therapy aims to target SSTR2A receptors, which are strongly expressed in meningiomas, but have an insufficient effect in most aggressive meningiomas, requiring the development of new techniques to increase the dose applied to the tumor. Based on the multiple potential intracellular targets, multiple targeted therapy clinical trials targeting Pi3K-Akt-mTOR and MAP kinase pathways as well as cell cycle and particularly, cyclin D4-6 are ongoing. Recently discovered driver mutations, SMO, Akt, and PI3KCA, offer new targets but are mostly observed in benign meningiomas, limiting their clinical relevance mainly to rare aggressive skull base meningiomas. Therefore, NF2 mutation remains the most frequent mutation and main challenging target in high-grade meningioma. Recently, inhibitors of focal adhesion kinase (FAK), which is involved in tumor cell adhesion, were tested in a phase 2 clinical trial with interesting but insufficient activity. The Hippo pathway was demonstrated to interact with NF2/Merlin and could be a promising target in NF2-mutated meningiomas with ongoing multiple preclinical studies and a phase 1 clinical trial. Recent advances in immune landscape comprehension led to the proposal of the use of immunotherapy in meningiomas. Except in rare cases of MSH2/6 mutation or high tumor mass burden, the activity of PD-1 inhibitors remains limited; however, its combination with various radiation therapy modalities is particularly promising. On the whole, therapeutic management of high-grade meningiomas is still challenging even with multiple promising therapeutic targets and innovations.
Collapse
Affiliation(s)
- T Graillon
- Aix-Marseille University, AP-HM, Inserm, MMG, Neurosurgery department, La Timone Hospital, Marseille, France.
| | - E Tabouret
- Aix-Marseille University, AP-HM, CNRS, INP, Inst Neurophysiopathol, CHU Timone, Service de Neurooncologie, Marseille, France
| | - B Salgues
- Nuclear Medicine Department, Groupe Hospitalier Pitié-Salpêtrière-Charles-Foix, Assistance publique-Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - T Horowitz
- AP-HM, CNRS, centrale Marseille, Institut Fresnel, Timone Hospital, CERIMED, Nuclear Medicine Department, Aix-Marseille University, Marseille, France
| | - L Padovani
- AP-HM, Timone Hospital, Radiotherapy Department, Marseille, France
| | - R Appay
- AP-HM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France; Aix-Marseille University, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - K Farah
- Aix-Marseille University, Institut de Neurosciences des Systèmes, UMR Inserm 1106, Functional Neurosurgery and Radiosurgery, Timone University Hospital, Marseille, France
| | - H Dufour
- Aix-Marseille University, AP-HM, Inserm, MMG, Neurosurgery department, La Timone Hospital, Marseille, France
| | - J Régis
- Aix-Marseille University, Institut de Neurosciences des Systèmes, UMR Inserm 1106, Functional Neurosurgery and Radiosurgery, Timone University Hospital, Marseille, France
| | - E Guedj
- AP-HM, CNRS, centrale Marseille, Institut Fresnel, Timone Hospital, CERIMED, Nuclear Medicine Department, Aix-Marseille University, Marseille, France
| | - A Barlier
- Aix-Marseille University, AP-HM, Inserm, MMG, Laboratory of Molecular Biology Hospital La Conception, Marseille, France
| | - O Chinot
- Aix-Marseille University, AP-HM, CNRS, INP, Inst Neurophysiopathol, CHU Timone, Service de Neurooncologie, Marseille, France
| |
Collapse
|
26
|
Li S, Wang C, Chen J, Lan Y, Zhang W, Kang Z, Zheng Y, Zhang R, Yu J, Li W. Signaling pathways in brain tumors and therapeutic interventions. Signal Transduct Target Ther 2023; 8:8. [PMID: 36596785 PMCID: PMC9810702 DOI: 10.1038/s41392-022-01260-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 01/05/2023] Open
Abstract
Brain tumors, although rare, contribute to distinct mortality and morbidity at all ages. Although there are few therapeutic options for brain tumors, enhanced biological understanding and unexampled innovations in targeted therapies and immunotherapies have considerably improved patients' prognoses. Nonetheless, the reduced response rates and unavoidable drug resistance of currently available treatment approaches have become a barrier to further improvement in brain tumor (glioma, meningioma, CNS germ cell tumors, and CNS lymphoma) treatment. Previous literature data revealed that several different signaling pathways are dysregulated in brain tumor. Importantly, a better understanding of targeting signaling pathways that influences malignant behavior of brain tumor cells might open the way for the development of novel targeted therapies. Thus, there is an urgent need for a more comprehensive understanding of the pathogenesis of these brain tumors, which might result in greater progress in therapeutic approaches. This paper began with a brief description of the epidemiology, incidence, risk factors, as well as survival of brain tumors. Next, the major signaling pathways underlying these brain tumors' pathogenesis and current progress in therapies, including clinical trials, targeted therapies, immunotherapies, and system therapies, have been systemically reviewed and discussed. Finally, future perspective and challenges of development of novel therapeutic strategies in brain tumor were emphasized.
Collapse
Affiliation(s)
- Shenglan Li
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Can Wang
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinyi Chen
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yanjie Lan
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Weichunbai Zhang
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhuang Kang
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yi Zheng
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Rong Zhang
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jianyu Yu
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenbin Li
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
27
|
Danish H, Brastianos P. Novel Medical Therapies in Meningiomas. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1416:213-223. [PMID: 37432630 DOI: 10.1007/978-3-031-29750-2_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
Meningiomas are the most common primary brain tumor in adults and have been historically managed with surgery and radiation therapy. However, in patients with inoperable, recurrent or high-grade tumors, medical therapy is often needed. Traditional chemotherapy and hormone therapy have been largely ineffective. However, with improved understanding of the molecular drivers in meningioma, there has been increasing interest in targeted molecular and immune therapies. In this chapter, we will discuss recent advances in meningioma genetics and biology and review current clinical trials with targeted molecular treatment and other novel therapies.
Collapse
Affiliation(s)
- Husain Danish
- Massachusetts General Hospital, Divisions of Neuro-Oncology and Hematology/Oncology, Departments of Neurology and Medicine, Harvard Medical School, Boston, MA, USA.
| | - Priscilla Brastianos
- Massachusetts General Hospital, Divisions of Neuro-Oncology and Hematology/Oncology, Departments of Neurology and Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
28
|
Jordan JT, Orr CC, Thalheimer RD, Cambillo JV, Beauchamp RL, Shaikh G, Muzikansky A, Stemmer-Rachamimov A, Giovannini M, Kalamarides M, Barker FG, Ramesh V, Plotkin SR. Prospective phase II trial of the dual mTORC1/2 inhibitor vistusertib for progressive or symptomatic meningiomas in persons with neurofibromatosis 2. Neurooncol Adv 2023; 5:vdad041. [PMID: 37215956 PMCID: PMC10195194 DOI: 10.1093/noajnl/vdad041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023] Open
Abstract
Background Meningiomas occur in 80% of persons with neurofibromatosis 2 (NF2) and cause significant mortality and morbidity, yet there are no effective medical treatments. NF2-deficient tumors have constitutive activation of mammalian/mechanistic target of rapamycin (mTOR), and treatment with mTORC1 inhibitors results in growth arrest in a minority of tumors, with paradoxical activation of the mTORC2/AKT pathway. We studied the effect of vistusertib, a dual mTORC1/mTORC2 inhibitor, in NF2 patients with progressive or symptomatic meningiomas. Methods Vistusertib was administered orally at 125 mg twice daily for 2 consecutive days each week. The primary endpoint was the imaging response in the target meningioma, defined as a volume decrease of 20% compared with the baseline. Secondary endpoints included toxicity, imaging response of nontarget tumors, quality of life, and genetic biomarkers. Results Eighteen participants (13 female), median age of 41 (range, 18-61) years, were enrolled. In target meningiomas, the best response was partial response (PR) in 1/18 tumors (6%) and stable disease (SD) in 17/18 tumors (94%). For all measured intracranial meningiomas and vestibular schwannomas, the best imaging response was PR in 6/59 tumors (10%) and SD in 53 (90%). Treatment-related grade 3/4 adverse events occurred in 14 (78%) participants, and 9 participants discontinued treatment due to side effects. Conclusions Although the study did not meet the primary endpoint, vistusertib treatment was associated with high rates of SD in progressive NF2-related tumors. However, this dosing regimen for vistusertib was poorly tolerated. Future studies of dual mTORC inhibitors for NF2 should focus on optimizing tolerability and evaluating the relevance of tumor stability in participants.
Collapse
Affiliation(s)
- Justin T Jordan
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Christina C Orr
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Raquel D Thalheimer
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Josephine V Cambillo
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Roberta L Beauchamp
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Ghalib Shaikh
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Alona Muzikansky
- Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anat Stemmer-Rachamimov
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marco Giovannini
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA and Jonsson Comprehensive Cancer Center (JCCC), University of California Los Angeles, Los Angeles, CA, USA
| | - Michel Kalamarides
- Department of Neurosurgery, Hopital Pitie-Salpetriere, Sorbonne Université, Paris, France
| | - Fred G Barker
- Neurosurgical Service, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vijaya Ramesh
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Scott R Plotkin
- Corresponding Author: Scott R. Plotkin, MD, PhD, Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, 55 Fruit Street, Yawkey 9E, Boston, MA 02114, USA ()
| |
Collapse
|
29
|
Bailo M, Gagliardi F, Boari N, Spina A, Piloni M, Castellano A, Mortini P. Meningioma and Other Meningeal Tumors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1405:73-97. [PMID: 37452935 DOI: 10.1007/978-3-031-23705-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Meningiomas develop from meningothelial cells and approximately account for more than 30 percent of central nervous system (CNS) tumors. They can occur anywhere in the dura, most often intracranially and at dural reflection sites. Half of the cases are usually at parasagittal/falcine and convexity locations; other common sites are sphenoid ridge, suprasellar, posterior fossa, and olfactory groove. The female-to-male ratio is approximately 2 or 3-1, and the median age at diagnosis is 65 years. Meningiomas are generally extremely slow-growing tumors; many are asymptomatic or paucisymptomatic at diagnosis and are discovered incidentally. Clinical manifestations, when present, are influenced by the tumor site and by the time course over which it develops. Meningiomas are divided into three grades. Grade I represents the vast majority of cases; they are considered typical or benign, although their CNS location can still lead to severe morbidity or mortality, resulting in a reported ten-year net survival of over 80%. Atypical (WHO grade II) meningiomas are considered "intermediate grade" malignancies and represent 5-7% of cases. They show a tendency for recurrence and malignant degeneration with a relevant increase in tumor cell migration and surrounding tissue infiltration; ten-year net survival is reported over 60%. The anaplastic subtype (WHO III) represents only 1-3% of cases, and it is characterized by a poor prognosis (ten-year net survival of 15%). The treatment of choice for these tumors stands on complete microsurgical resection in case the subsequent morbidities are assumed minimal. On the other hand, and in case the tumor is located in critical regions such as the skull base, or the patient may have accompanied comorbidities, or it is aimed to avoid intensive treatment, some other approaches, including stereotactic radiosurgery and radiotherapy, were recommended as safe and effective choices to be considered as a primary treatment option or complementary to surgery. Adjuvant radiosurgery/radiotherapy should be considered in the case of atypical and anaplastic histology, especially when a residual tumor is identifiable in postoperative imaging. A "watchful waiting" strategy appears reasonable for extremely old individuals and those with substantial comorbidities or low-performance status, while there is a reduced threshold for therapeutic intervention for relatively healthy younger individuals due to the expectation that tumor progression will inevitably necessitate proactive treatment. To treat and manage meningioma efficiently, the assessments of both neurosurgeons and radiation oncologists are essential. The possibility of other rarer tumors, including hemangiopericytomas, solitary fibrous tumors, lymphomas, metastases, melanocytic tumors, and fibrous histiocytoma, must be considered when a meningeal lesion is diagnosed, especially because the ideal diagnostic and therapeutic approaches might differ significantly in every tumor type.
Collapse
Affiliation(s)
- Michele Bailo
- Department of Neurosurgery and Gamma Knife Radiosurgery, I.R.C.C.S. Ospedale San Raffaele, Vita-Salute University, Via Olgettina 60, 20132, Milano, Italy.
| | - Filippo Gagliardi
- Department of Neurosurgery and Gamma Knife Radiosurgery, I.R.C.C.S. Ospedale San Raffaele, Vita-Salute University, Via Olgettina 60, 20132, Milano, Italy
| | - Nicola Boari
- Department of Neurosurgery and Gamma Knife Radiosurgery, I.R.C.C.S. Ospedale San Raffaele, Vita-Salute University, Via Olgettina 60, 20132, Milano, Italy
| | - Alfio Spina
- Department of Neurosurgery and Gamma Knife Radiosurgery, I.R.C.C.S. Ospedale San Raffaele, Vita-Salute University, Via Olgettina 60, 20132, Milano, Italy
| | - Martina Piloni
- Department of Neurosurgery and Gamma Knife Radiosurgery, I.R.C.C.S. Ospedale San Raffaele, Vita-Salute University, Via Olgettina 60, 20132, Milano, Italy
| | - Antonella Castellano
- Department of Neurosurgery and Gamma Knife Radiosurgery, I.R.C.C.S. Ospedale San Raffaele, Vita-Salute University, Via Olgettina 60, 20132, Milano, Italy
| | - Pietro Mortini
- Department of Neurosurgery and Gamma Knife Radiosurgery, I.R.C.C.S. Ospedale San Raffaele, Vita-Salute University, Via Olgettina 60, 20132, Milano, Italy
| |
Collapse
|
30
|
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
|
31
|
Shobeiri P, Seyedmirzaei H, Kalantari A, Mohammadi E, Rezaei N, Hanaei S. The Epidemiology of Brain and Spinal Cord Tumors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1394:19-39. [PMID: 36587379 DOI: 10.1007/978-3-031-14732-6_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
CNS tumors are a diverse group of neoplasms that emerge from a variety of different CNS cell types. These tumors may be benign, malignant, or borderline in nature. The majority of high grade glial tumors are fatal, with the exception of pilocytic astrocytoma. Primary malignant CNS tumors occur at a global annual rate of 2.1 to 5.8 per 100,000 persons. Males are more likely to develop malignant brain tumors than females, whereas benign meningiomas are more common in adult females. Additionally, gender inequalities in non-malignant tumors peak between the ages of 25 and 29 years. Only a small number of genetic variants have been associated with survival and prognosis. Notably, central nervous system (CNS) tumors exhibit significant age, gender, and race variation. Race is another factor that affects the incidence of brain and spinal cord tumors. Different races exhibit variation in terms of the prevalence of brain and CNS malignancies. This chapter discusses ongoing research on brain and spinal cord tumor epidemiology, as well as the associated risks and accompanied disorders.
Collapse
Affiliation(s)
- Parnian Shobeiri
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Homa Seyedmirzaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Amirali Kalantari
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Esmaeil Mohammadi
- Department of Pediatric Neurosurgery, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sara Hanaei
- Department of Neurosurgery, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
- Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
32
|
Hennigan RF, Thomson CS, Stachowski K, Nassar N, Ratner N. Merlin tumor suppressor function is regulated by PIP2-mediated dimerization. PLoS One 2023; 18:e0281876. [PMID: 36809290 PMCID: PMC9942953 DOI: 10.1371/journal.pone.0281876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/30/2023] [Indexed: 02/23/2023] Open
Abstract
Neurofibromatosis Type 2 is an inherited disease characterized by Schwann cell tumors of cranial and peripheral nerves. The NF2 gene encodes Merlin, a member of the ERM family consisting of an N-terminal FERM domain, a central α-helical region, and a C-terminal domain. Changes in the intermolecular FERM-CTD interaction allow Merlin to transition between an open, FERM accessible conformation and a closed, FERM-inaccessible conformation, modulating Merlin activity. Merlin has been shown to dimerize, but the regulation and function Merlin dimerization is not clear. We used a nanobody based binding assay to show that Merlin dimerizes via a FERM-FERM interaction, orientated with each C-terminus close to each other. Patient derived and structural mutants show that dimerization controls interactions with specific binding partners, including HIPPO pathway components, and correlates with tumor suppressor activity. Gel filtration experiments showed that dimerization occurs after a PIP2 mediated transition from closed to open conformation monomers. This process requires the first 18 amino acids of the FERM domain and is inhibited by phosphorylation at serine 518. The discovery that active, open conformation Merlin is a dimer represents a new paradigm for Merlin function with implications for the development of therapies designed to compensate for Merlin loss.
Collapse
Affiliation(s)
- Robert F. Hennigan
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States of America
- * E-mail:
| | - Craig S. Thomson
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States of America
| | - Kye Stachowski
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States of America
| | - Nicolas Nassar
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States of America
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States of America
| |
Collapse
|
33
|
Clark KL, George JW, Przygrodzka E, Plewes MR, Hua G, Wang C, Davis JS. Hippo Signaling in the Ovary: Emerging Roles in Development, Fertility, and Disease. Endocr Rev 2022; 43:1074-1096. [PMID: 35596657 PMCID: PMC9695108 DOI: 10.1210/endrev/bnac013] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Indexed: 01/09/2023]
Abstract
Emerging studies indicate that the Hippo pathway, a highly conserved pathway that regulates organ size control, plays an important role in governing ovarian physiology, fertility, and pathology. Specific to the ovary, the spatiotemporal expression of the major components of the Hippo signaling cascade are observed throughout the reproductive lifespan. Observations from multiple species begin to elucidate the functional diversity and molecular mechanisms of Hippo signaling in the ovary in addition to the identification of interactions with other signaling pathways and responses to various external stimuli. Hippo pathway components play important roles in follicle growth and activation, as well as steroidogenesis, by regulating several key biological processes through mechanisms of cell proliferation, migration, differentiation, and cell fate determination. Given the importance of these processes, dysregulation of the Hippo pathway contributes to loss of follicular homeostasis and reproductive disorders such as polycystic ovary syndrome (PCOS), premature ovarian insufficiency, and ovarian cancers. This review highlights what is currently known about the Hippo pathway core components in ovarian physiology, including ovarian development, follicle development, and oocyte maturation, while identifying areas for future research to better understand Hippo signaling as a multifunctional pathway in reproductive health and biology.
Collapse
Affiliation(s)
- Kendra L Clark
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA.,Veterans Affairs Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Jitu W George
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA.,Veterans Affairs Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Emilia Przygrodzka
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA.,Veterans Affairs Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Michele R Plewes
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA.,Veterans Affairs Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Guohua Hua
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science & Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Cheng Wang
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - John S Davis
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA.,Veterans Affairs Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA
| |
Collapse
|
34
|
Dahoud W, Handler J, Parimi V, Meyer CF, Wethington SL, Eshleman JR, Vang R, Ronnett BM, Xing D. Adult Granulosa Cell Tumor With Sarcomatous Transformation: A Case Study With Emphasis on Molecular Alterations. Int J Gynecol Pathol 2022; 41:600-607. [PMID: 34856571 PMCID: PMC9167042 DOI: 10.1097/pgp.0000000000000845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Adult granulosa cells tumors (AGCTs) are typically low-grade indolent tumors. On rare occasions, they undergo high-grade/sarcomatous transformation and behave aggressively. This transformation is postulated to occur as the result of acquired genetic alterations, some of which may be eligible for targeted therapy. Here we report a rare case of AGCT with sarcomatous transformation that harbored distinct molecular alterations from those typically seen with AGCTs supporting a molecularly driven approach to these malignancies. The patient is a 56-yr-old G3P3 woman with a history of multiple recurrences of ovarian AGCT for which the first diagnosis was made at the age of 25 when she was evaluated for infertility. The ovarian tumor displayed typical features of AGCT with low-grade, bland morphology. The first extraovarian spread of tumor involving the cul-de-sac was reported at the age of 39. After that, recurrences occurred every 2 to 3 yr with involvement of multiple anatomic sites and repeated surgical resections. At the age of 55 she developed a symptomatic recurrence in the pelvis and underwent resection of an isolated lesion (specimen 1) to no gross residual disease. Within 4 wk of resection she developed significant pelvic pain and imaging showed recurrence of the mass. Therefore, in 5 mo after the initial resection she underwent repeat excision of the lesion (specimen 2) and associated bowel. The sections from specimen 1 showed a biphasic morphology: a low-grade component with morphology and immunophenotype consistent with a typical AGCT and a high-grade spindle cell component with features consistent with a high-grade sarcoma. Specimen 2 featured a pure high-grade sarcoma characterized by coagulative tumor cell necrosis, readily recognizable mitoses, highly atypical cells with vesicular nuclei and prominent nucleoli. SF-1 positivity and the presence of FOXL2 C134W mutation in the sarcomatous component support the notion of transformation of typical AGCT. While detected TERT promoter C228T mutation may play a role in this process, we further identified genetic alterations affecting PI3K/AKT/mTOR pathway, including mutations in PIK3CA , PIK3R1 , AKT1 , and NF2 , which may also contribute to tumor progression/transformation. These findings provide rationale for molecular/pathway-based targeted therapy for patients with advanced AGCT.
Collapse
|
35
|
Co-Targeting MAP Kinase and Pi3K-Akt-mTOR Pathways in Meningioma: Preclinical Study of Alpelisib and Trametinib. Cancers (Basel) 2022; 14:cancers14184448. [PMID: 36139608 PMCID: PMC9496760 DOI: 10.3390/cancers14184448] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/01/2022] [Accepted: 09/09/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Multi recurrent or high-grade meningiomas remain an unmet medical need in neuro-oncology. Several studies have highlighted the potential therapeutic efficacy of mTor inhibitors to control tumoral growth of meningiomas. However, a positive feedback on AKT oncogenic pathway from these drugs may explain the modest success. Our aim was to target Pi3kinase upstream mTor, and MAP kinase pathway, overactivated in meningiomas, alone or in combined targeting in comparison to mTor targeting. Our in vitro results obtained on three meningioma cell lines and on a large series of fresh human meningiomas, including 35 WHO grade 1, 23 grade 2, and five grade 3, showed that co-targeting Pi3kinase and MAP kinase seemed promising, opening new therapeutic strategies in these tumors. Abstract Recurrent or high-grade meningiomas are an unmet medical need. Recently, we demonstrated that targeting mTOR by everolimus was relevant both in vitro and in humans. However, everolimus induces an AKT activation that may impact the anti-proliferative effect of the drug. Moreover, the MAP kinase pathway was shown to be involved in meningioma tumorigenesis. We therefore targeted both the Pi3k-AKT-mTOR and MAP kinase pathways by using combinations of the Pi3k inhibitor alpelisib and the MEK inhibitor trametinib. Our study was performed in vitro on the human meningioma cell lines and on a large series of primary cultures providing from 63 freshly operated meningiomas including 35 WHO grade 1, 23 grade 2, and five grade 3, half of which presented a NF2 genomic alteration. Alpelisib induced a higher inhibitory effect on cell viability and proliferation than everolimus in all cell lines and 32 randomly selected tumors no matter the genomic status, the histological subtype or grade. Trametinib also strongly inhibited cell proliferation and induced AKT activation. Combined treatment with alpelisib plus trametinib reversed the AKT activation induced by trametinib and induced an additive inhibitory effect irrespective of the cell lines or tumor features. Co-targeting pathways seems promising and may be considered particularly for aggressive meningioma.
Collapse
|
36
|
Prabhakar S, Beauchamp RL, Cheah PS, Yoshinaga A, Haidar EA, Lule S, Mani G, Maalouf K, Stemmer-Rachamimov A, Jung DH, Welling DB, Giovannini M, Plotkin SR, Maguire CA, Ramesh V, Breakefield XO. Gene replacement therapy in a schwannoma mouse model of neurofibromatosis type 2. Mol Ther Methods Clin Dev 2022; 26:169-180. [PMID: 35846573 PMCID: PMC9263409 DOI: 10.1016/j.omtm.2022.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 06/17/2022] [Indexed: 11/25/2022]
Abstract
Loss of function of the neurofibromatosis type 2 (NF2) tumor suppressor gene leads to the formation of schwannomas, meningiomas, and ependymomas, comprising ∼50% of all sporadic cases of primary nervous system tumors. NF2 syndrome is an autosomal dominant condition, with bi-allelic inactivation of germline and somatic alleles resulting in loss of function of the encoded protein merlin and activation of mammalian target of rapamycin (mTOR) pathway signaling in NF2-deficient cells. Here we describe a gene replacement approach through direct intratumoral injection of an adeno-associated virus vector expressing merlin in a novel human schwannoma model in nude mice. In culture, the introduction of an AAV1 vector encoding merlin into CRISPR-modified human NF2-null arachnoidal cells (ACs) or Schwann cells (SCs) was associated with decreased size and mTORC1 pathway activation consistent with restored merlin activity. In vivo, a single injection of AAV1-merlin directly into human NF2-null SC-derived tumors growing in the sciatic nerve of nude mice led to regression of tumors over a 10-week period, associated with a decrease in dividing cells and an increase in apoptosis, in comparison with vehicle. These studies establish that merlin re-expression via gene replacement in NF2-null schwannomas is sufficient to cause tumor regression, thereby potentially providing an effective treatment for NF2.
Collapse
Affiliation(s)
- Shilpa Prabhakar
- Department of Neurology and Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Roberta L. Beauchamp
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Pike See Cheah
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Center for Molecular Imaging Research, Massachusetts General Hospital, 25 Shattuck St, Boston, MA 02115, USA
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, JALAN UNIVERSITI 1 Serdang, 43400 Seri Kembangan, Selangor, Malaysia
| | - Akiko Yoshinaga
- Department of Neurology and Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Edwina Abou Haidar
- Department of Neurology and Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Sevda Lule
- Department of Neurology and Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Gayathri Mani
- Department of Neurology and Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Katia Maalouf
- Department of Neurology and Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Anat Stemmer-Rachamimov
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - David H. Jung
- Department of Otolaryngology, Massachusetts Eye and Ear and Harvard Medical School, Boston, MA 02114, USA
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, MA 02114, USA
| | - D. Bradley Welling
- Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Massachusetts Eye and Ear and Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, MA 02114, USA
| | - Marco Giovannini
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA and Jonsson Comprehensive Cancer Center (JCCC), University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Scott R. Plotkin
- Department of Neurology and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Casey A. Maguire
- Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| | - Vijaya Ramesh
- Department of Neurology and Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Xandra O. Breakefield
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
37
|
Deng J, Hua L, Bian L, Chen H, Chen L, Cheng H, Dou C, Geng D, Hong T, Ji H, Jiang Y, Lan Q, Li G, Liu Z, Qi S, Qu Y, Shi S, Sun X, Wang H, You Y, Yu H, Yue S, Zhang J, Zhang X, Wang S, Mao Y, Zhong P, Gong Y. Molecular diagnosis and treatment of meningiomas: an expert consensus (2022). Chin Med J (Engl) 2022; 135:1894-1912. [PMID: 36179152 PMCID: PMC9746788 DOI: 10.1097/cm9.0000000000002391] [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: 05/19/2022] [Indexed: 11/27/2022] Open
Abstract
ABSTRACT Meningiomas are the most common primary intracranial neoplasm with diverse pathological types and complicated clinical manifestations. The fifth edition of the WHO Classification of Tumors of the Central Nervous System (WHO CNS5), published in 2021, introduces major changes that advance the role of molecular diagnostics in meningiomas. To follow the revision of WHO CNS5, this expert consensus statement was formed jointly by the Group of Neuro-Oncology, Society of Neurosurgery, Chinese Medical Association together with neuropathologists and evidence-based experts. The consensus provides reference points to integrate key biomarkers into stratification and clinical decision making for meningioma patients. REGISTRATION Practice guideline REgistration for transPAREncy (PREPARE), IPGRP-2022CN234.
Collapse
Affiliation(s)
- Jiaojiao Deng
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Lingyang Hua
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai 200040, China
- Neurosurgical Institute of Fudan University, Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai 200040, China
| | - Liuguan Bian
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hong Chen
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ligang Chen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Hongwei Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Changwu Dou
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 750306, China
| | - Dangmurenjiapu Geng
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| | - Tao Hong
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Hongming Ji
- Department of Neurosurgery, Shanxi Medical University Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Yugang Jiang
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Qing Lan
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Soochow, Jiangsu 215004, China
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong 250063, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yan Qu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi 710038, China
| | - Songsheng Shi
- Department of Neurosurgery, Fujian Medical University Affiliated Union Hospital, Fuzhou, Fujian 350001, China
| | - Xiaochuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Haijun Wang
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Yongping You
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Hualin Yu
- Department of Neurosurgery, Kunming Medical University First Affiliated Hospital, Kunming, Yunnan 650032, China
| | - Shuyuan Yue
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jianming Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Xiaohua Zhang
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shuo Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ping Zhong
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai 200040, China
- Neurosurgical Institute of Fudan University, Shanghai 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai 200040, China
| | - Ye Gong
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
38
|
Okano A, Miyawaki S, Teranishi Y, Ohara K, Hongo H, Sakai Y, Ishigami D, Nakatomi H, Saito N. Advances in Molecular Biological and Translational Studies in World Health Organization Grades 2 and 3 Meningiomas: A Literature Review. Neurol Med Chir (Tokyo) 2022; 62:347-360. [PMID: 35871574 PMCID: PMC9464479 DOI: 10.2176/jns-nmc.2022-0114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/10/2022] [Indexed: 11/29/2022] Open
Abstract
The treatment of World Health Organization (WHO) grades 2 and 3 meningiomas remains difficult and controversial. The pathogenesis of high-grade meningiomas was expected to be elucidated to improve treatment strategies. The molecular biology of meningiomas has been clarified in recent years. High-grade meningiomas have been linked to NF2 mutations and 22q deletion. CDKN2A/B homozygous deletion and TERT promoter mutations are independent prognostic factors for WHO grade 3 meningiomas. In addition to 22q loss, 1p, 14p, and 9q loss have been linked to high-grade meningiomas. Meningiomas enriched in copy number alterations may be biologically invasive. Furthermore, several new comprehensive classifications of meningiomas have been proposed based on these molecular biological features, including DNA methylation status. The new classifications may have implications for treatment strategies for refractory aggressive meningiomas because they provide a more accurate prognosis compared to the conventional WHO classification. Although several systemic therapies, including molecular targeted therapies, may be effective in treating refractory aggressive meningiomas, these drugs are being tested. Systemic drug therapy for meningioma is expected to be developed in the future. Thus, this review aims to discuss the distinct genomic alterations observed in WHO grade 2 and 3 meningiomas, as well as their diagnostic and therapeutic implications and systemic drug therapies for high-grade meningiomas.
Collapse
Affiliation(s)
- Atsushi Okano
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo
| | - Satoru Miyawaki
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo
| | - Yu Teranishi
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo
| | - Kenta Ohara
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo
| | - Hiroki Hongo
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo
| | - Yu Sakai
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo
| | - Daiichiro Ishigami
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo
| | - Hirofumi Nakatomi
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo
- Department of Neurosurgery, Kyorin University
| | - Nobuhito Saito
- Department of Neurosurgery, Faculty of Medicine, The University of Tokyo
| |
Collapse
|
39
|
miR-127-5p Targets JAM3 to Regulate Ferroptosis, Proliferation, and Metastasis in Malignant Meningioma Cells. DISEASE MARKERS 2022; 2022:6423237. [PMID: 35818586 PMCID: PMC9271006 DOI: 10.1155/2022/6423237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/09/2022] [Accepted: 06/02/2022] [Indexed: 11/18/2022]
Abstract
Objective Meningiomas are one of the most common primary tumors of the central nervous system. Most of them are benign and can be cured by surgery, while a few meningiomas are malignant. Ferroptosis gene characteristics might be associated with drug therapy and survival in patients with clinically aggressive, unresectable meningiomas. This study explored the mechanism of differentially expressed miRNAs and ferroptosis in meningioma to provide a new reference to treat meningioma. Methods Bioinformatics analysis of differential miRNA profiles and functions in patients with meningioma was performed. The contents of lactate dehydrogenase (LDH), malondialdehyde (MDA), and Fe2+ were determined. Reactive oxygen species (ROS) values, as well as cell cycle changes, were analyzed by flow cytometry. The targets of miR-127-5p and JAM3 were detected by dual luciferase assays. Cell counting kit-8 (CCK8) and Transwell assays were used to analyze cell activity. Ki67 expression was analyzed by immunohistochemistry. Expression levels of miR-127-5p and JAM3 were analyzed by RT-qPCR. GPX4 expression was quantified by western blotting. Results miR-127-5p was expressed at low levels in IOMM-Lee cells, while JAM3 was highly expressed in IOMM-Lee cells. A dual luciferase assay demonstrated that miR-127-5p could target JAM3. Upregulation of miR-127-5p in IOMM-Lee cells resulted in cell cycle arrest and inhibition of cell activity. Upregulation of miR-127-5p increased LDH, MDA, and ROS levels and Fe2+ content and inhibited the expression of GPX4 protein. Upregulation of JAM3 reversed the results of miR-127-5p upregulation. Conclusion miR-127-5p regulated meningioma formation and ferroptosis through JAM3, providing insights for the development of new treatments for meningioma.
Collapse
|
40
|
Pellerino A, Bruno F, Palmiero R, Pronello E, Bertero L, Soffietti R, Rudà R. Clinical Significance of Molecular Alterations and Systemic Therapy for Meningiomas: Where Do We Stand? Cancers (Basel) 2022; 14:2256. [PMID: 35565385 PMCID: PMC9100910 DOI: 10.3390/cancers14092256] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 12/25/2022] Open
Abstract
Meningiomas are common intracranial tumors that can be treated successfully in most cases with surgical resection and/or adjuvant radiotherapy. However, approximately 20% of patients show an aggressive clinical course with tumor recurrence or progressive disease, resulting in significant morbidity and increased mortality. Despite several studies that have investigated different cytotoxic agents in aggressive meningiomas in the past several years, limited evidence of efficacy and clinical benefit has been reported thus far. Novel molecular alterations have been linked to a particular clinicopathological phenotype and have been correlated with grading, location, and prognosis of meningiomas. In this regard, SMO, AKT, and PIK3CA mutations are typical of anterior skull base meningiomas, whereas KLF4 mutations are specific for secretory histology, and BAP1 alterations are common in progressive rhabdoid meningiomas. Alterations in TERT, DMD, and BAP1 correlate with poor outcomes. Moreover, some actionable mutations, including SMO, AKT1, and PIK3CA, regulate meningioma growth and are under investigation in clinical trials. PD-L1 and/or M2 macrophage expression in the microenvironment provides evidence for the investigation of immunotherapy in progressive meningiomas.
Collapse
Affiliation(s)
- Alessia Pellerino
- Division of Neuro-Oncology, Department Neuroscience, University and City of Health and Science Hospital, 10126 Turin, Italy; (A.P.); (F.B.); (R.P.); (R.R.)
| | - Francesco Bruno
- Division of Neuro-Oncology, Department Neuroscience, University and City of Health and Science Hospital, 10126 Turin, Italy; (A.P.); (F.B.); (R.P.); (R.R.)
| | - Rosa Palmiero
- Division of Neuro-Oncology, Department Neuroscience, University and City of Health and Science Hospital, 10126 Turin, Italy; (A.P.); (F.B.); (R.P.); (R.R.)
| | - Edoardo Pronello
- Department of Neurology Unit, Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy;
| | - Luca Bertero
- Pathology Unit, Department of Medical Sciences, University and City of Health and Science Hospital, 10126 Turin, Italy;
| | - Riccardo Soffietti
- Division of Neuro-Oncology, Department Neuroscience, University and City of Health and Science Hospital, 10126 Turin, Italy; (A.P.); (F.B.); (R.P.); (R.R.)
| | - Roberta Rudà
- Division of Neuro-Oncology, Department Neuroscience, University and City of Health and Science Hospital, 10126 Turin, Italy; (A.P.); (F.B.); (R.P.); (R.R.)
- Department of Neurology, Castelfranco Veneto and Treviso Hospital, 31100 Treviso, Italy
| |
Collapse
|
41
|
Patel B, Desai R, Pugazenthi S, Butt OH, Huang J, Kim AH. Identification and Management of Aggressive Meningiomas. Front Oncol 2022; 12:851758. [PMID: 35402234 PMCID: PMC8984123 DOI: 10.3389/fonc.2022.851758] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/23/2022] [Indexed: 12/31/2022] Open
Abstract
Meningiomas are common primary central nervous system tumors derived from the meninges, with management most frequently entailing serial monitoring or a combination of surgery and/or radiation therapy. Although often considered benign lesions, meningiomas can not only be surgically inaccessible but also exhibit aggressive growth and recurrence. In such cases, adjuvant radiation and systemic therapy may be required for tumor control. In this review, we briefly describe the current WHO grading scale for meningioma and provide demonstrative cases of treatment-resistant meningiomas. We also summarize frequently observed molecular abnormalities and their correlation with intracranial location and recurrence rate. We then describe how genetic and epigenetic features might supplement or even replace histopathologic features for improved identification of aggressive lesions. Finally, we describe the role of surgery, radiotherapy, and ongoing systemic therapy as well as precision medicine clinical trials for the treatment of recurrent meningioma.
Collapse
Affiliation(s)
- Bhuvic Patel
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, United States
| | - Rupen Desai
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, United States
| | - Sangami Pugazenthi
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, United States
| | - Omar H. Butt
- Department of Medicine, Division of Medical Oncology, Washington University School of Medicine, St. Louis, MO, United States,The Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Jiayi Huang
- The Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, United States,Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, United States
| | - Albert H. Kim
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, United States,The Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, United States,*Correspondence: Albert H. Kim,
| |
Collapse
|
42
|
Varela L, Garcia-Rendueles MER. Oncogenic Pathways in Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms23063223. [PMID: 35328644 PMCID: PMC8952192 DOI: 10.3390/ijms23063223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 02/05/2023] Open
Abstract
Cancer and neurodegenerative diseases are two of the leading causes of premature death in modern societies. Their incidence continues to increase, and in the near future, it is believed that cancer will kill more than 20 million people per year, and neurodegenerative diseases, due to the aging of the world population, will double their prevalence. The onset and the progression of both diseases are defined by dysregulation of the same molecular signaling pathways. However, whereas in cancer, these alterations lead to cell survival and proliferation, neurodegenerative diseases trigger cell death and apoptosis. The study of the mechanisms underlying these opposite final responses to the same molecular trigger is key to providing a better understanding of the diseases and finding more accurate treatments. Here, we review the ten most common signaling pathways altered in cancer and analyze them in the context of different neurodegenerative diseases such as Alzheimer's (AD), Parkinson's (PD), and Huntington's (HD) diseases.
Collapse
Affiliation(s)
- Luis Varela
- Yale Center for Molecular and Systems Metabolism, Department of Comparative Medicine, School of Medicine, Yale University, 310 Cedar St. BML 330, New Haven, CT 06520, USA
- Correspondence: (L.V.); (M.E.R.G.-R.)
| | - Maria E. R. Garcia-Rendueles
- Precision Nutrition and Cancer Program, IMDEA Food Institute, Campus Excelencia Internacional UAM+CSIC, 28049 Madrid, Spain
- Correspondence: (L.V.); (M.E.R.G.-R.)
| |
Collapse
|
43
|
Pinker B, Barciszewska AM. mTOR Signaling and Potential Therapeutic Targeting in Meningioma. Int J Mol Sci 2022; 23:ijms23041978. [PMID: 35216092 PMCID: PMC8876623 DOI: 10.3390/ijms23041978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/27/2022] [Accepted: 01/30/2022] [Indexed: 12/30/2022] Open
Abstract
Meningiomas are the most frequent primary tumors arising in the central nervous system. They typically follow a benign course, with an excellent prognosis for grade I lesions through surgical intervention. Although radiotherapy is a good option for recurrent, progressive, or inoperable tumors, alternative treatments are very limited. mTOR is a protein complex with increasing therapeutical potential as a target in cancer. The current understanding of the mTOR pathway heavily involves it in the development of meningioma. Its activation is strongly dependent on PI3K/Akt signaling and the merlin protein. Both factors are commonly defective in meningioma cells, which indicates their likely function in tumor growth. Furthermore, regarding molecular tumorigenesis, the kinase activity of the mTORC1 complex inhibits many components of the autophagosome, such as the ULK1 or Beclin complexes. mTOR contributes to redox homeostasis, a vital component of neoplasia. Recent clinical trials have investigated novel chemotherapeutic agents for mTOR inhibition, showing promising results in resistant or recurrent meningiomas.
Collapse
Affiliation(s)
- Benjamin Pinker
- Medical Faculty, Karol Marcinkowski University of Medical Sciences, Fredry 10, 61-701 Poznan, Poland
- Correspondence:
| | - Anna-Maria Barciszewska
- Intraoperative Imaging Unit, Chair and Department of Neurosurgery and Neurotraumatology, Karol Marcinkowski University of Medical Sciences, Przybyszewskiego 49, 60-355 Poznan, Poland;
- Department of Neurosurgery and Neurotraumatology, Heliodor Swiecicki Clinical Hospital, Przybyszewskiego 49, 60-355 Poznan, Poland
| |
Collapse
|
44
|
Graillon T, Tabouret E, Chinot O. Chemotherapy and targeted therapies for meningiomas: what is the evidence? Curr Opin Neurol 2021; 34:857-867. [PMID: 34629433 DOI: 10.1097/wco.0000000000001002] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE OF REVIEW Although most meningiomas are slow growing tumors mainly controlled by surgery with or without radiotherapy, aggressive meningiomas that fail these conventional treatments constitute a rare situation, a therapeutic challenge and an unmet need in neuro-oncology. RECENT FINDING Mutational landscape in recurrent high-grade meningiomas includes mainly NF2 mutation or 22q chromosomal deletion, whereas telomerase reverse transcriptase promoter, BAP-1 and CDK2NA mutations were also found in aggressive meningiomas. Pi3K-Akt-mTOR pathway is currently the most relevant intracellular signaling pathway target in meningiomas with preliminary clinical activity observed. Assessment of drug activity with progression free survival rate at 6 months is challenging in regard to meningioma growth rate heterogeneity, so that 3-dimensional growth rate before and during treatment could be considered in the future to selected new active drugs. SUMMARY Despite a low evidence level, some systemic therapies may be considered for patients with recurrent meningioma not amenable to further surgery or radiotherapy. In recurrent high-grade meningioma, everolimus-octreotide combination, bevacizumab, sunitinib and peptide receptor radionuclide therapy exhibit a signal of activity that may justify their clinical use. Despite a lack of clear signal of activity to date, immunotherapy may offer new perspectives in the treatment of these refractory tumors.
Collapse
Affiliation(s)
- Thomas Graillon
- Aix Marseille Univ, APHM, INSERM, MMG, UMR1251, La Timone Hospital, neurosurgery department Marseille, France
| | - Emeline Tabouret
- Aix-Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, La Timone Hospital, Neurooncology Department, Marseille, France
| | - Olivier Chinot
- Aix-Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, La Timone Hospital, Neurooncology Department, Marseille, France
| |
Collapse
|
45
|
Sanchez LD, Bui A, Klesse LJ. Targeted Therapies for the Neurofibromatoses. Cancers (Basel) 2021; 13:cancers13236032. [PMID: 34885143 PMCID: PMC8657309 DOI: 10.3390/cancers13236032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/09/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022] Open
Abstract
Over the past several years, management of the tumors associated with the neurofibromatoses has been recognized to often require approaches that are distinct from their spontaneous counterparts. Focus has shifted to therapy aimed at minimizing symptoms given the risks of persistent, multiple tumors and new tumor growth. In this review, we will highlight the translation of preclinical data to therapeutic trials for patients with neurofibromatosis, particularly neurofibromatosis type 1 and neurofibromatosis type 2. Successful inhibition of MEK for patients with neurofibromatosis type 1 and progressive optic pathway gliomas or plexiform neurofibromas has been a significant advancement in patient care. Similar success for the malignant NF1 tumors, such as high-grade gliomas and malignant peripheral nerve sheath tumors, has not yet been achieved; nor has significant progress been made for patients with either neurofibromatosis type 2 or schwannomatosis, although efforts are ongoing.
Collapse
Affiliation(s)
- Lauren D. Sanchez
- Department of Pediatrics, Division of Neurology, UT Southwestern Medical Center, Dallas, TX 75235, USA;
| | - Ashley Bui
- Department of Pediatrics, Division of Hematology/Oncology, UT Southwestern Medical Center, Dallas, TX 75235, USA;
| | - Laura J. Klesse
- Department of Pediatrics, Division of Hematology/Oncology, UT Southwestern Medical Center, Dallas, TX 75235, USA;
- Correspondence:
| |
Collapse
|
46
|
John P, Waldt N, Liebich J, Kesseler C, Schnabel S, Angenstein F, Sandalcioglu IE, Scherlach C, Sahm F, Kirches E, Mawrin C. AKT1 E17K -mutated meningioma cell lines respond to treatment with the AKT inhibitor AZD5363. Neuropathol Appl Neurobiol 2021; 48:e12780. [PMID: 34837233 DOI: 10.1111/nan.12780] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/26/2021] [Accepted: 11/14/2021] [Indexed: 11/30/2022]
Abstract
AIMS Meningiomas are the most frequent primary brain tumours. Recently, knowledge about the molecular drivers underlying aggressive meningiomas has been expanded. A hotspot mutation in the AKT1 gene (AKT1E17K ), which is found in meningiomas at the convexity and especially at the skull base, has been associated with earlier tumour recurrence. METHODS Here, we analysed the effects of the AKT1E17K mutation and treatment response to the Akt inhibitor AZD5363 in transgenic meningioma cell clones and mouse xenografts modelling convexity or skull base meningiomas. RESULTS We show that the AKTE17K mutation significantly enhances meningioma cell proliferation and colony size in vitro, resulting in significantly shortened survival times of mice carrying convexity or skull base AKT1E17K xenografts. Treatment of mutant cells or xenografts (150 mg/kg/d) with AZD5363 revealed a significant decrease in cell proliferation and colony size and a prolongation of mouse survival. Western blots revealed activation of AKT1 kinase (phosphorylation at Ser273 and Thr308) by the E17K mutation in human meningioma samples and in our in vitro and in vivo models. CONCLUSIONS Our data suggest that AKT1E17K mutated meningiomas are a promising selective target for AZD5363.
Collapse
Affiliation(s)
- Peter John
- Department of Neuropathology, Otto von Guericke University, Magdeburg, Germany
| | - Natalie Waldt
- Department of Neuropathology, Otto von Guericke University, Magdeburg, Germany
| | - Josephine Liebich
- Department of Neuropathology, Otto von Guericke University, Magdeburg, Germany
| | - Christoph Kesseler
- Department of Neuropathology, Otto von Guericke University, Magdeburg, Germany
| | - Stefan Schnabel
- Department of Neurosurgery, Paracelsus Hospital Zwickau, Zwickau, Germany
| | | | - I Erol Sandalcioglu
- Department of Neurosurgery, Otto von Guericke University, Magdeburg, Germany
| | - Cordula Scherlach
- Department of Neuroradiology, University Hospital Leipzig, Heidelberg, Germany
| | - Felix Sahm
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Elmar Kirches
- Department of Neuropathology, Otto von Guericke University, Magdeburg, Germany
| | - Christian Mawrin
- Department of Neuropathology, Otto von Guericke University, Magdeburg, Germany
| |
Collapse
|
47
|
Discovery of putative tumor suppressors from CRISPR screens reveals rewired lipid metabolism in acute myeloid leukemia cells. Nat Commun 2021; 12:6506. [PMID: 34764293 PMCID: PMC8586352 DOI: 10.1038/s41467-021-26867-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 10/27/2021] [Indexed: 12/26/2022] Open
Abstract
CRISPR knockout fitness screens in cancer cell lines reveal many genes whose loss of function causes cell death or loss of fitness or, more rarely, the opposite phenotype of faster proliferation. Here we demonstrate a systematic approach to identify these proliferation suppressors, which are highly enriched for tumor suppressor genes, and define a network of 145 such genes in 22 modules. One module contains several elements of the glycerolipid biosynthesis pathway and operates exclusively in a subset of acute myeloid leukemia cell lines. The proliferation suppressor activity of genes involved in the synthesis of saturated fatty acids, coupled with a more severe loss of fitness phenotype for genes in the desaturation pathway, suggests that these cells operate at the limit of their carrying capacity for saturated fatty acids, which we confirm biochemically. Overexpression of this module is associated with a survival advantage in juvenile leukemias, suggesting a clinically relevant subtype. CRISPR-based knockout screens in cancer cells have suggested the existence of proliferation suppressor genes (PSG). Here, the authors develop an approach to systematically identify them, and reveal a PSG module involved in fatty acid synthesis and tumour suppression in acute myeloid leukemia cell lines.
Collapse
|
48
|
p21-Activated kinase 1 (PAK1) in aging and longevity: An overview. Ageing Res Rev 2021; 71:101443. [PMID: 34390849 DOI: 10.1016/j.arr.2021.101443] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/26/2021] [Accepted: 08/10/2021] [Indexed: 02/08/2023]
Abstract
The p21-activated kinases (PAKs) belong to serine/threonine kinases family, regulated by ∼21 kDa small signaling G proteins RAC1 and CDC42. The mammalian PAK family comprises six members (PAK1-6) that are classified into two groups (I and II) based on their domain architecture and regulatory mechanisms. PAKs are implicated in a wide range of cellular functions. PAK1 has recently attracted increasing attention owing to its involvement in oncogenesis, tumor progression, and metastasis as well as several life-limiting diseases and pathological conditions. In Caenorhabditis elegans, PAK1 functions limit the lifespan under basal conditions by inhibiting forkhead transcription factor DAF-16. Interestingly, PAK depletion extended longevity and attenuated the onset of age-related phenotypes in a premature-aging mouse model and delayed senescence in mammalian fibroblasts. These observations implicate PAKs as not only oncogenic but also aging kinases. Therefore, PAK-targeting genetic and/or pharmacological interventions, particularly PAK1-targeting, could be a viable strategy for developing cancer therapies with relatively no side effects and promoting healthy longevity. This review describes PAK family proteins, their biological functions, and their role in regulating aging and longevity using C. elegans. Moreover, we discuss the effect of small-molecule PAK1 inhibitors on the lifespan and healthspan of C. elegans.
Collapse
|
49
|
Maggio I, Franceschi E, Di Nunno V, Gatto L, Tosoni A, Angelini D, Bartolini S, Lodi R, Brandes AA. Discovering the Molecular Landscape of Meningioma: The Struggle to Find New Therapeutic Targets. Diagnostics (Basel) 2021; 11:1852. [PMID: 34679551 PMCID: PMC8534341 DOI: 10.3390/diagnostics11101852] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/27/2021] [Accepted: 10/04/2021] [Indexed: 02/06/2023] Open
Abstract
Meningiomas are the most common primary CNS tumors. They are usually benign but can present aggressive behavior in about 20% of cases. The genetic landscape of meningioma is characterized by the presence (in about 60% of cases) or absence of NF2 mutation. Low-grade meningiomas can also present other genetic alterations, particularly affecting SMO, TRAF7, KLF4 AKT1 and PI3KCA. In higher grade meningiomas, mutations of TERT promoter and deletion of CDKN2A/B seem to have a prognostic value. Furthermore, other genetic alterations have been identified, such as BAP1, DMD and PBRM1. Different subgroups of DNA methylation appear to be correlated with prognosis. In this review, we explored the genetic landscape of meningiomas and the possible therapeutic implications.
Collapse
Affiliation(s)
- Ilaria Maggio
- Medical Oncology Department, Azienda USL, Via Altura n. 3, 40139 Bologna, Italy; (I.M.); (V.D.N.); (L.G.)
| | - Enrico Franceschi
- Nervous System Medical Oncology Department, IRCSS Istituto di Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (A.T.); (D.A.); (S.B.); (A.A.B.)
| | - Vincenzo Di Nunno
- Medical Oncology Department, Azienda USL, Via Altura n. 3, 40139 Bologna, Italy; (I.M.); (V.D.N.); (L.G.)
- Nervous System Medical Oncology Department, IRCSS Istituto di Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (A.T.); (D.A.); (S.B.); (A.A.B.)
| | - Lidia Gatto
- Medical Oncology Department, Azienda USL, Via Altura n. 3, 40139 Bologna, Italy; (I.M.); (V.D.N.); (L.G.)
- Nervous System Medical Oncology Department, IRCSS Istituto di Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (A.T.); (D.A.); (S.B.); (A.A.B.)
| | - Alicia Tosoni
- Nervous System Medical Oncology Department, IRCSS Istituto di Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (A.T.); (D.A.); (S.B.); (A.A.B.)
| | - Daniele Angelini
- Nervous System Medical Oncology Department, IRCSS Istituto di Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (A.T.); (D.A.); (S.B.); (A.A.B.)
| | - Stefania Bartolini
- Nervous System Medical Oncology Department, IRCSS Istituto di Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (A.T.); (D.A.); (S.B.); (A.A.B.)
| | - Raffaele Lodi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; or
| | - Alba Ariela Brandes
- Nervous System Medical Oncology Department, IRCSS Istituto di Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (A.T.); (D.A.); (S.B.); (A.A.B.)
| |
Collapse
|
50
|
Amaravathi A, Oblinger JL, Welling DB, Kinghorn AD, Chang LS. Neurofibromatosis: Molecular Pathogenesis and Natural Compounds as Potential Treatments. Front Oncol 2021; 11:698192. [PMID: 34604034 PMCID: PMC8485038 DOI: 10.3389/fonc.2021.698192] [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: 04/20/2021] [Accepted: 09/01/2021] [Indexed: 12/22/2022] Open
Abstract
The neurofibromatosis syndromes, including NF1, NF2, and schwannomatosis, are tumor suppressor syndromes characterized by multiple nervous system tumors, particularly Schwann cell neoplasms. NF-related tumors are mainly treated by surgery, and some of them have been treated by but are refractory to conventional chemotherapy. Recent advances in molecular genetics and genomics alongside the development of multiple animal models have provided a better understanding of NF tumor biology and facilitated target identification and therapeutic evaluation. Many targeted therapies have been evaluated in preclinical models and patients with limited success. One major advance is the FDA approval of the MEK inhibitor selumetinib for the treatment of NF1-associated plexiform neurofibroma. Due to their anti-neoplastic, antioxidant, and anti-inflammatory properties, selected natural compounds could be useful as a primary therapy or as an adjuvant therapy prior to or following surgery and/or radiation for patients with tumor predisposition syndromes, as patients often take them as dietary supplements and for health enhancement purposes. Here we review the natural compounds that have been evaluated in NF models. Some have demonstrated potent anti-tumor effects and may become viable treatments in the future.
Collapse
Affiliation(s)
- Anusha Amaravathi
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Janet L Oblinger
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - D Bradley Welling
- Department of Otolaryngology Head & Neck Surgery, Harvard Medical School, Massachusetts Eye and Ear, and Massachusetts General Hospital, Boston, MA, United States
| | - A Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University College of Pharmacy, Columbus, OH, United States
| | - Long-Sheng Chang
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States.,Department of Otolaryngology-Head & Neck Surgery, The Ohio State University College of Medicine, Columbus, OH, United States
| |
Collapse
|