1
|
Chang M, Sherief M, Ioannou M, Chinnasamy V, Chen L, Frost M, Mattson-Hoss M, Sarnoff H, Kamson DO, Holdhoff M, Mukherjee D, Bettegowda C, Rincon-Torroella J, Croog V, Huang P, Rodriguez FJ, Lucas CHG, Schreck KC. NF1 expression profiling in IDH-wildtype glioblastoma: genomic associations and survival outcomes. Acta Neuropathol Commun 2024; 12:172. [PMID: 39472976 PMCID: PMC11520828 DOI: 10.1186/s40478-024-01875-z] [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] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 10/12/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND NF1 inactivation is associated with sensitivity to MEK inhibitor targeted therapy in low-grade and some high-grade gliomas. NF1 loss may also be a harbinger of exploitable vulnerabilities in IDH-wildtype glioblastoma (GBM). Accurate and consistent detection of NF1 loss, however, is fraught given the large gene size, challenges with complete coverage and variant calling upon sequencing, and mechanisms of mRNA and protein regulation that result in early degradation in the absence of genomic alterations. Here, we seek to perform a composite analysis for NF1 loss accounting for genomic alterations and protein expression via immunohistochemistry. We also characterize the landscape of NF1 alterations in GBM. METHODS We assembled a single-institution, retrospective cohort of 542 IDH-wildtype GBM with somatic next generation sequencing to investigate the frequency and nature of detected NF1 alterations. We selected 69 GBMs from which to build a tissue microarray (TMA) of 44 NF1-wildtype and 25 NF1-mutant cases. We performed NF1 immunohistochemistry using two different NF1 antibodies (NFC, Sigma-Aldrich; and iNF-07E, iNFixion Bioscience) and correlated results with clinical, genomic, and other immunohistochemical features. RESULTS In our retrospective cohort, we identified 88 IDH-wildtype GBM with NF1 alterations (16%). NF1 alterations were mutually exclusive with EGFR and MDM2 alterations (p-adj < 0.001, 0.05, respectively), but co-occurred with PIK3R1 alterations (Log2(OR) = - 1.6, p-adj = 0.03). Of the 63 scorable sporadic GBMs in the TMA, 14 harbored NF1 inactivating alterations and of those, 12 (86%) demonstrated minimal NF1 immunoreactivity by NFC antibody, compared to 8 (57%) by iNF-07E antibody. Among the 42 scorable NF1-wildtype GBM in the TMA, NF1 immunostaining was minimal in 18 (43%) by NFC antibody compared to 4 (10%) by iNF-07E antibody, potentially reflecting false positives or differential protein regulation. Minimal immunoreactivity by NFC antibody was associated with decreased median overall survival (8.5 vs. 16.4 months, p = 0.011). Cox proportional hazards model correcting for prognostic variables in this subset revealed HR 3.23 (95% CI 1.29-8.06, p = 0.01) associated with decreased NF1 expression by IHC. CONCLUSION NF1 immunostaining may serve as a sensitive surrogate marker of NF1 genomic inactivation and a valuable extension to next-generation sequencing for defining NF1 status. Minimal NF1 immunoreactivity is a poor prognostic marker, even in IDH-wildtype glioblastoma without apparent NF1 genomic alterations, but the underlying molecular mechanism requires further investigation.
Collapse
Affiliation(s)
- Michael Chang
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mohamed Sherief
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Maria Ioannou
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Viveka Chinnasamy
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lucy Chen
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | | - David O Kamson
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthias Holdhoff
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Debraj Mukherjee
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chetan Bettegowda
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Victoria Croog
- Department of Radiation Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peng Huang
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Fausto J Rodriguez
- Division of Neuropathology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Calixto-Hope G Lucas
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Karisa C Schreck
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Bouley SJ, Grassetti AV, Allaway RJ, Wood MD, Hou HW, Burdon Dasbach IR, Seibel W, Wu J, Gerber SA, Dragnev KH, Walker JA, Sanchez Y. Chemical genetic screens reveal defective lysosomal trafficking as synthetic lethal with NF1 loss. J Cell Sci 2024; 137:jcs262343. [PMID: 39016685 PMCID: PMC11361638 DOI: 10.1242/jcs.262343] [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: 06/11/2024] [Accepted: 06/28/2024] [Indexed: 07/18/2024] Open
Abstract
Neurofibromatosis type 1, a genetic disorder caused by pathogenic germline variations in NF1, predisposes individuals to the development of tumors, including cutaneous and plexiform neurofibromas (CNs and PNs), optic gliomas, astrocytomas, juvenile myelomonocytic leukemia, high-grade gliomas and malignant peripheral nerve sheath tumors (MPNSTs), which are chemotherapy- and radiation-resistant sarcomas with poor survival. Loss of NF1 also occurs in sporadic tumors, such as glioblastoma (GBM), melanoma, breast, ovarian and lung cancers. We performed a high-throughput screen for compounds that were synthetic lethal with NF1 loss, which identified several leads, including the small molecule Y102. Treatment of cells with Y102 perturbed autophagy, mitophagy and lysosome positioning in NF1-deficient cells. A dual proteomics approach identified BLOC-one-related complex (BORC), which is required for lysosome positioning and trafficking, as a potential target of Y102. Knockdown of a BORC subunit using siRNA recapitulated the phenotypes observed with Y102 treatment. Our findings demonstrate that BORC might be a promising therapeutic target for NF1-deficient tumors.
Collapse
Affiliation(s)
- Stephanie J. Bouley
- Department of Molecular and Systems Biology, Geisel School of Medicine, Hanover, NH 03755, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Andrew V. Grassetti
- Department of Molecular and Systems Biology, Geisel School of Medicine, Hanover, NH 03755, USA
- Department of Biochemistry and Cellular Biology, Geisel School of Medicine, Hanover, NH 03755, USA
| | - Robert J. Allaway
- Department of Molecular and Systems Biology, Geisel School of Medicine, Hanover, NH 03755, USA
| | - Matthew D. Wood
- Department of Pharmacology and Toxicology, Geisel School of Medicine, Hanover, NH 03755, USA
| | - Helen W. Hou
- Department of Pharmacology and Toxicology, Geisel School of Medicine, Hanover, NH 03755, USA
| | - India R. Burdon Dasbach
- Department of Molecular and Systems Biology, Geisel School of Medicine, Hanover, NH 03755, USA
| | - William Seibel
- Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Jimmy Wu
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA
| | - Scott A. Gerber
- Department of Molecular and Systems Biology, Geisel School of Medicine, Hanover, NH 03755, USA
- Department of Biochemistry and Cellular Biology, Geisel School of Medicine, Hanover, NH 03755, USA
| | - Konstantin H. Dragnev
- Department of Medicine, Geisel School of Medicine, Hanover, NH 03755, USA
- Section of Medical Oncology, Geisel School of Medicine, Hanover, NH 03755, USA
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03766, USA
| | - James A. Walker
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Yolanda Sanchez
- Department of Molecular and Systems Biology, Geisel School of Medicine, Hanover, NH 03755, USA
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03766, USA
| |
Collapse
|
3
|
Stauffer S, Roth JS, Hernandez ER, Kowalczyk JT, Sealover NE, Hebron KE, James A, Isanogle KA, Riffle LA, Ileva L, Luo X, Chen JQ, Kedei N, Kortum RL, Lei H, Shern JF, Kalen JD, Edmondson EF, Hall MD, Difilippantonio S, Thiele CJ, Yohe ME. Preclinical Therapeutic Efficacy of RAF/MEK/ERK and IGF1R/AKT/mTOR Inhibition in Neuroblastoma. Cancers (Basel) 2024; 16:2320. [PMID: 39001383 PMCID: PMC11240493 DOI: 10.3390/cancers16132320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/12/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Activating mutations in the RAS/MAPK pathway are observed in relapsed neuroblastoma. Preclinical studies indicate that these tumors have an increased sensitivity to inhibitors of the RAS/MAPK pathway, such as MEK inhibitors. MEK inhibitors do not induce durable responses as single agents, indicating a need to identify synergistic combinations of targeted agents to provide therapeutic benefit. We previously showed preclinical therapeutic synergy between a MEK inhibitor, trametinib, and a monoclonal antibody specific for IGF1R, ganitumab in RAS-mutated rhabdomyosarcoma. Neuroblastoma cells, like rhabdomyosarcoma cells, are sensitive to the inhibition of the RAS/MAPK and IGF1R/AKT/mTOR pathways. We hypothesized that the combination of trametinib and ganitumab would be effective in RAS-mutated neuroblastoma. In this study, trametinib and ganitumab synergistically suppressed neuroblastoma cell proliferation and induced apoptosis in cell culture. We also observed a delay in tumor initiation and prolongation of survival in heterotopic and orthotopic xenograft models treated with trametinib and ganitumab. However, the growth of both primary and metastatic tumors was observed in animals receiving the combination of trametinib and ganitumab. Therefore, more preclinical work is necessary before testing this combination in patients with relapsed or refractory RAS-mutated neuroblastoma.
Collapse
Affiliation(s)
- Stacey Stauffer
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute, NIH, 8560 Progress Drive, Frederick, MD 21701, USA
| | - Jacob S Roth
- Early Translation Branch, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Edjay R Hernandez
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Joshua T Kowalczyk
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Nancy E Sealover
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Services, Bethesda, MD 20814, USA
| | - Katie E Hebron
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute, NIH, 8560 Progress Drive, Frederick, MD 21701, USA
| | - Amy James
- Animal Research Technical Support, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Kristine A Isanogle
- Animal Research Technical Support, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Lisa A Riffle
- Small Animal Imaging Program, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Lilia Ileva
- Small Animal Imaging Program, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Xiaoling Luo
- Collaborative Protein Technology Resource, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Jin-Qiu Chen
- Collaborative Protein Technology Resource, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Noemi Kedei
- Collaborative Protein Technology Resource, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Robert L Kortum
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Services, Bethesda, MD 20814, USA
| | - Haiyan Lei
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Jack F Shern
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Joseph D Kalen
- Small Animal Imaging Program, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Elijah F Edmondson
- Molecular Histopathology Laboratory, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Matthew D Hall
- Early Translation Branch, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Simone Difilippantonio
- Animal Research Technical Support, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Carol J Thiele
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Marielle E Yohe
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute, NIH, 8560 Progress Drive, Frederick, MD 21701, USA
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bethesda, MD 20892, USA
| |
Collapse
|
4
|
Zhao J, Cui X, Zhan Q, Zhang K, Su D, Yang S, Hong B, Wang Q, Ju J, Cheng C, Li C, Wan C, Wang Y, Zhou J, Kang C. CRISPR-Cas9 library screening combined with an exosome-targeted delivery system addresses tumorigenesis/TMZ resistance in the mesenchymal subtype of glioblastoma. Theranostics 2024; 14:2835-2855. [PMID: 38773970 PMCID: PMC11103500 DOI: 10.7150/thno.92703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/20/2024] [Indexed: 05/24/2024] Open
Abstract
Rationale: The large-scale genomic analysis classifies glioblastoma (GBM) into three major subtypes, including classical (CL), proneural (PN), and mesenchymal (MES) subtypes. Each of these subtypes exhibits a varying degree of sensitivity to the temozolomide (TMZ) treatment, while the prognosis corresponds to the molecular and genetic characteristics of the tumor cell type. Tumors with MES features are predominantly characterized by the NF1 deletion/alteration, leading to sustained activation of the RAS and PI3K-AKT signaling pathways in GBM and tend to acquire drug resistance, resulting in the worst prognosis compared to other subtypes (PN and CL). Here, we used the CRISPR/Cas9 library screening technique to detect TMZ-related gene targets that might play roles in acquiring drug resistance, using overexpressed KRAS-G12C mutant GBM cell lines. The study identified a key therapeutic strategy to address the chemoresistance against the MES subtype of GBM. Methods: The CRISPR-Cas9 library screening was used to discover genes associated with TMZ resistance in the U87-KRAS (U87-MG which is overexpressed KRAS-G12C mutant) cells. The patient-derived GBM primary cell line TBD0220 was used for experimental validations in vivo and in vitro. Chromatin isolation by RNA purification (ChIRP) and chromatin immunoprecipitation (ChIP) assays were used to elucidate the silencing mechanism of tumor suppressor genes in the MES-GBM subtype. The small-molecule inhibitor EPIC-0412 was obtained through high-throughput screening. Transmission electron microscopy (TEM) was used to characterize the exosomes (Exos) secreted by GBM cells after TMZ treatment. Blood-derived Exos-based targeted delivery of siRNA, TMZ, and EPIC-0412 was optimized to tailor personalized therapy in vivo. Results: Using the genome-wide CRISPR-Cas9 library screening, we found that the ERBIN gene could be epigenetically regulated in the U87-KRAS cells. ERBIN overexpression inhibited the RAS signaling and downstream proliferation and invasion effects of GBM tumor cells. EPIC-0412 treatment inhibited tumor proliferation and EMT progression by upregulating the ERBIN expression both in vitro and in vivo. Genome-wide CRISPR-Cas9 screening also identified RASGRP1(Ras guanine nucleotide-releasing protein 1) and VPS28(Vacuolar protein sorting-associated protein 28) genes as synthetically lethal in response to TMZ treatment in the U87-KRAS cells. We found that RASGRP1 activated the RAS-mediated DDR pathway by promoting the RAS-GTP transformation. VPS28 promoted the Exos secretion and decreased intracellular TMZ concentration in GBM cells. The targeted Exos delivery system encapsulating drugs and siRNAs together showed a powerful therapeutic effect against GBM in vivo. Conclusions: We demonstrate a new mechanism by which ERBIN is epigenetically silenced by the RAS signaling in the MES subtype of GBM. Restoration of the ERBIN expression with EPIC-0412 significantly inhibits the RAS signaling downstream. RASGRP1 and VPS28 genes are associated with the promotion of TMZ resistance through RAS-GDP to RAS-GTP transformation and TMZ efflux, as well. A quadruple combination therapy based on a targeted Exos delivery system demonstrated significantly reduced tumor burden in vivo. Therefore, our study provides new insights and therapeutic approaches for regulating tumor progression and TMZ resistance in the MES-GBM subtype.
Collapse
Affiliation(s)
- Jixing Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Xiaoteng Cui
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Qi Zhan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Kailiang Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, China
| | - Dongyuan Su
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Shixue Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Biao Hong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Qixue Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Jiasheng Ju
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Chunchao Cheng
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Chen Li
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunxiao Wan
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
| | - Yunfei Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Junhu Zhou
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Chunsheng Kang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| |
Collapse
|
5
|
Liang S, Xu S, Zhou S, Chang C, Shao Z, Wang Y, Chen S, Huang Y, Guo Y. IMAGGS: a radiogenomic framework for identifying multi-way associations in breast cancer subtypes. J Genet Genomics 2024; 51:443-453. [PMID: 37783335 DOI: 10.1016/j.jgg.2023.09.010] [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: 09/04/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/04/2023]
Abstract
Investigating correlations between radiomic and genomic profiling in breast cancer (BC) molecular subtypes is crucial for understanding disease mechanisms and providing personalized treatment. We present a well-designed radiogenomic framework image-gene-gene set (IMAGGS), which detects multi-way associations in BC subtypes by integrating radiomic and genomic features. Our dataset consists of 721 patients, each of whom has 12 ultrasound (US) images captured from different angles and gene mutation data. To better characterize tumor traits, 12 multi-angle US images are fused using two distinct strategies. Then, we analyze complex many-to-many associations between phenotypic and genotypic features using a machine learning algorithm, deviating from the prevalent one-to-one relationship pattern observed in previous studies. Key radiomic and genomic features are screened using these associations. In addition, gene set enrichment analysis is performed to investigate the joint effects of gene sets and delve deeper into the biological functions of BC subtypes. We further validate the feasibility of IMAGGS in a glioblastoma multiforme dataset to demonstrate the scalability of IMAGGS across different modalities and diseases. Taken together, IMAGGS provides a comprehensive characterization for diseases by associating imaging, genes, and gene sets, paving the way for biological interpretation of radiomics and development of targeted therapy.
Collapse
Affiliation(s)
- Shuyu Liang
- Department of Electronic Engineering, School of Information Science and Technology, Fudan University, Shanghai 200433, China; The Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention (MICCAI) of Shanghai, Shanghai 200032, China
| | - Sicheng Xu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200433, China
| | - Shichong Zhou
- Department of Ultrasound, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Cai Chang
- Department of Ultrasound, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhiming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yuanyuan Wang
- Department of Electronic Engineering, School of Information Science and Technology, Fudan University, Shanghai 200433, China; The Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention (MICCAI) of Shanghai, Shanghai 200032, China
| | - Sheng Chen
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Yunxia Huang
- Department of Ultrasound, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Yi Guo
- Department of Electronic Engineering, School of Information Science and Technology, Fudan University, Shanghai 200433, China; The Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention (MICCAI) of Shanghai, Shanghai 200032, China.
| |
Collapse
|
6
|
Begagić E, Pugonja R, Bečulić H, Čeliković A, Tandir Lihić L, Kadić Vukas S, Čejvan L, Skomorac R, Selimović E, Jaganjac B, Juković-Bihorac F, Jusić A, Pojskić M. Molecular Targeted Therapies in Glioblastoma Multiforme: A Systematic Overview of Global Trends and Findings. Brain Sci 2023; 13:1602. [PMID: 38002561 PMCID: PMC10669565 DOI: 10.3390/brainsci13111602] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
This systematic review assesses current molecular targeted therapies for glioblastoma multiforme (GBM), a challenging condition with limited treatment options. Using PRISMA methodology, 166 eligible studies, involving 2526 patients (61.49% male, 38.51% female, with a male-to-female ratio of 1.59/1), were analyzed. In laboratory studies, 52.52% primarily used human glioblastoma cell cultures (HCC), and 43.17% employed animal samples (mainly mice). Clinical participants ranged from 18 to 100 years, with 60.2% using combined therapies and 39.8% monotherapies. Mechanistic categories included Protein Kinase Phosphorylation (41.6%), Cell Cycle-Related Mechanisms (18.1%), Microenvironmental Targets (19.9%), Immunological Targets (4.2%), and Other Mechanisms (16.3%). Key molecular targets included Epidermal Growth Factor Receptor (EGFR) (10.8%), Mammalian Target of Rapamycin (mTOR) (7.2%), Vascular Endothelial Growth Factor (VEGF) (6.6%), and Mitogen-Activated Protein Kinase (MEK) (5.4%). This review provides a comprehensive assessment of molecular therapies for GBM, highlighting their varied efficacy in clinical and laboratory settings, ultimately impacting overall and progression-free survival in GBM management.
Collapse
Affiliation(s)
- Emir Begagić
- Department of General Medicine, School of Medicine, Unversity of Zenica, Travnička 1, 72000 Zenica, Bosnia and Herzegovina; (E.B.)
| | - Ragib Pugonja
- Department of Anatomy, School of Medicine, University of Zenica, Travnička 1, 72000 Zenica, Bosnia and Herzegovina;
- Department of General Medicine, Primary Health Care Center, Nikole Šubića Zrinjskog bb., 72260 Busovača, Bosnia and Herzegovina
| | - Hakija Bečulić
- Department of General Medicine, Primary Health Care Center, Nikole Šubića Zrinjskog bb., 72260 Busovača, Bosnia and Herzegovina
- Department of Neurosurgery, Cantonal Hospital Zenica, Crkvice 76, 72000 Zenica, Bosnia and Herzegovina
| | - Amila Čeliković
- Department of General Medicine, School of Medicine, Unversity of Zenica, Travnička 1, 72000 Zenica, Bosnia and Herzegovina; (E.B.)
| | - Lejla Tandir Lihić
- Department of Neurology, Cantonal Hospital Zenica, Crkvice 76, 72000 Zenica, Bosnia and Herzegovina
| | - Samra Kadić Vukas
- Department of Neurology, Cantonal Hospital Zenica, Crkvice 76, 72000 Zenica, Bosnia and Herzegovina
| | - Lejla Čejvan
- Department of General Medicine, School of Medicine, Unversity of Zenica, Travnička 1, 72000 Zenica, Bosnia and Herzegovina; (E.B.)
| | - Rasim Skomorac
- Department of Neurosurgery, Cantonal Hospital Zenica, Crkvice 76, 72000 Zenica, Bosnia and Herzegovina
- Department of Surgery, School of Medicine, University of Zenica, Travnička 1, 72000 Zenica, Bosnia and Herzegovina;
| | - Edin Selimović
- Department of Surgery, School of Medicine, University of Zenica, Travnička 1, 72000 Zenica, Bosnia and Herzegovina;
| | - Belma Jaganjac
- Department of Histology, School of Medicine, University of Zenica, Travnička 1, 72000 Zenica, Bosnia and Herzegovina; (B.J.)
| | - Fatima Juković-Bihorac
- Department of Histology, School of Medicine, University of Zenica, Travnička 1, 72000 Zenica, Bosnia and Herzegovina; (B.J.)
- Department of Pathology, School of Medicine, University of Zenica, Travnička 1, 72000 Zenica, Bosnia and Herzegovina
- Department of Pathology, Cantonal Hospital Zenica, Crkvice 76, 72000 Zenica, Bosnia and Herzegovina
| | - Aldin Jusić
- Department of Neurosurgery, Cantonal Hospital Zenica, Crkvice 76, 72000 Zenica, Bosnia and Herzegovina
| | - Mirza Pojskić
- Department of Neurosurgery, University Hospital Marburg, Baldingerstr., 35033 Marburg, Germany
| |
Collapse
|
7
|
Romo CG, Piotrowski AF, Campian JL, Diarte J, Rodriguez FJ, Bale TA, Dahiya S, Gutmann DH, Lucas CHG, Prichett L, Mellinghoff I, Blakeley JO. Clinical, histological, and molecular features of gliomas in adults with neurofibromatosis type 1. Neuro Oncol 2023; 25:1474-1486. [PMID: 36840626 PMCID: PMC10398805 DOI: 10.1093/neuonc/noad033] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND People with NF1 have an increased prevalence of central nervous system malignancy. However, little is known about the clinical course or pathologic features of NF1-associated gliomas in adults, limiting clinical care and research. METHODS Adults (≥18 years) with NF1 and histologically confirmed non-optic pathway gliomas (non-OPGs) at Johns Hopkins Hospital, Memorial Sloan Kettering Cancer Center, and Washington University presenting between 1990 and 2020 were identified. Retrospective data were collated, and pathology was reviewed centrally. RESULTS Forty-five patients, comprising 23 females (51%), met eligibility criteria, with a median of age 37 (18-68 years) and performance status of 80% (30%-100%). Tissue was available for 35 patients. Diagnoses included infiltrating (low-grade) astrocytoma (9), glioblastoma (7), high-grade astrocytoma with piloid features (4), pilocytic astrocytoma (4), high-grade astrocytoma (3), WHO diagnosis not reached (4) and one each of gliosarcoma, ganglioglioma, embryonal tumor, and diffuse midline glioma. Seventy-one percent of tumors were midline and underwent biopsy only. All 27 tumors evaluated were IDH1-wild-type, independent of histology. In the 10 cases with molecular testing, the most common genetic variants were NF1, EGFR, ATRX, CDKN2A/B, TP53, TERT, and MSH2/3 mutation. While the treatments provided varied, the median overall survival was 24 months [2-267 months] across all ages, and 38.5 [18-109] months in individuals with grade 1-2 gliomas. CONCLUSIONS Non-OPGs in adults with NF1, including low-grade tumors, often have an aggressive clinical course, indicating a need to better understand the pathobiology of these NF1-associated gliomas.
Collapse
Affiliation(s)
- Carlos G Romo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anna F Piotrowski
- Departments of Neurology and Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jian L Campian
- Departments of Neurology and Pathology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jose Diarte
- Departments of Neurology and Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Fausto J Rodriguez
- Department of Pathology, University of California Los Angeles, Los Angeles, California, USA
| | - Tejus A Bale
- Departments of Neurology and Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sonika Dahiya
- Departments of Neurology and Pathology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - David H Gutmann
- Departments of Neurology and Pathology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Calixto-Hope G Lucas
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Laura Prichett
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ingo Mellinghoff
- Departments of Neurology and Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jaishri O Blakeley
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
8
|
Muzyka L, Goff NK, Choudhary N, Koltz MT. Systematic Review of Molecular Targeted Therapies for Adult-Type Diffuse Glioma: An Analysis of Clinical and Laboratory Studies. Int J Mol Sci 2023; 24:10456. [PMID: 37445633 DOI: 10.3390/ijms241310456] [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: 04/17/2023] [Revised: 06/05/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
Gliomas are the most common brain tumor in adults, and molecularly targeted therapies to treat gliomas are becoming a frequent topic of investigation. The current state of molecular targeted therapy research for adult-type diffuse gliomas has yet to be characterized, particularly following the 2021 WHO guideline changes for classifying gliomas using molecular subtypes. This systematic review sought to characterize the current state of molecular target therapy research for adult-type diffuse glioma to better inform scientific progress and guide next steps in this field of study. A systematic review was conducted in accordance with PRISMA guidelines. Studies meeting inclusion criteria were queried for study design, subject (patients, human cell lines, mice, etc.), type of tumor studied, molecular target, respective molecular pathway, and details pertaining to the molecular targeted therapy-namely the modality, dose, and duration of treatment. A total of 350 studies met the inclusion criteria. A total of 52 of these were clinical studies, 190 were laboratory studies investigating existing molecular therapies, and 108 were laboratory studies investigating new molecular targets. Further, a total of 119 ongoing clinical trials are also underway, per a detailed query on clinicaltrials.gov. GBM was the predominant tumor studied in both ongoing and published clinical studies as well as in laboratory analyses. A few studies mentioned IDH-mutant astrocytomas or oligodendrogliomas. The most common molecular targets in published clinical studies and clinical trials were protein kinase pathways, followed by microenvironmental targets, immunotherapy, and cell cycle/apoptosis pathways. The most common molecular targets in laboratory studies were also protein kinase pathways; however, cell cycle/apoptosis pathways were the next most frequent target, followed by microenvironmental targets, then immunotherapy pathways, with the wnt/β-catenin pathway arising in the cohort of novel targets. In this systematic review, we examined the current evidence on molecular targeted therapy for adult-type diffuse glioma and discussed its implications for clinical practice and future research. Ultimately, published research falls broadly into three categories-clinical studies, laboratory testing of existing therapies, and laboratory identification of novel targets-and heavily centers on GBM rather than IDH-mutant astrocytoma or oligodendroglioma. Ongoing clinical trials are numerous in this area of research as well and follow a similar pattern in tumor type and targeted pathways as published clinical studies. The most common molecular targets in all study types were protein kinase pathways. Microenvironmental targets were more numerous in clinical studies, whereas cell cycle/apoptosis were more numerous in laboratory studies. Immunotherapy pathways are on the rise in all study types, and the wnt/β-catenin pathway is increasingly identified as a novel target.
Collapse
Affiliation(s)
- Logan Muzyka
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, 1501 Red River Street, Austin, TX 78712, USA
| | - Nicolas K Goff
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, 1501 Red River Street, Austin, TX 78712, USA
| | - Nikita Choudhary
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, 1501 Red River Street, Austin, TX 78712, USA
| | - Michael T Koltz
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, 1501 Red River Street, Austin, TX 78712, USA
| |
Collapse
|
9
|
Wisinski KB, Flamand Y, Wilson MA, Luke JJ, Tawbi HA, Hong F, Mitchell EP, Zwiebel JA, Chen H, Gray RJ, Li S, McShane LM, Rubinstein LV, Patton D, Williams PM, Hamilton SR, Behrens RJ, Pennington KP, Conley BA, Arteaga CL, Harris LN, O'Dwyer PJ, Chen AP, Flaherty KT. Trametinib in Patients With NF1-, GNAQ-, or GNA11-Mutant Tumors: Results From the NCI-MATCH ECOG-ACRIN Trial (EAY131) Subprotocols S1 and S2. JCO Precis Oncol 2023; 7:e2200421. [PMID: 37053535 PMCID: PMC10309549 DOI: 10.1200/po.22.00421] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 02/13/2023] [Indexed: 04/15/2023] Open
Abstract
PURPOSE NCI-MATCH is a precision medicine trial using genomic testing to allocate patients with advanced malignancies to targeted treatment subprotocols. This report combines two subprotocols evaluating trametinib, a MEK1/2 inhibitor, in patients with Neurofibromatosis 1 (NF1[S1] or GNA11/Q [S2]) altered tumors. METHODS Eligible patients had tumors with deleterious inactivating NF1 or GNA11/Q mutations by the customized Oncomine AmpliSeq panel. Prior MEK inhibitor treatment was excluded. Glioblastomas (GBMs) were permitted, including malignancies associated with germline NF1 mutations (S1 only). Trametinib was administered at 2 mg once daily over 28-day cycles until toxicity or disease progression. Primary end point was objective response rate (ORR). Secondary end points included progression-free survival (PFS) at 6 months, PFS, and overall survival. Exploratory analyses included co-occurring genomic alterations and PTEN loss. RESULTS Fifty patients were eligible and started therapy: 46 with NF1 mutations (S1) and four with GNA11 mutations (S2). In the NF1 cohort, nonsense single-nucleotide variants were identified in 29 and frameshift deletions in 17 tumors. All in S2 had nonuveal melanoma and GNA11 Q209L variant. Two partial responses (PR) were noted in S1, one patient each with advanced lung cancer and GBM for an ORR of 4.3% (90% CI, 0.8 to 13.1). One patient with melanoma in S2 had a PR (ORR, 25%; 90% CI, 1.3 to 75.1). Prolonged stable disease (SD) was also noted in five patients (four in S1 and one in S2) with additional rare histologies. Adverse events were as previously described with trametinib. Comutations in TP53 and PIK3CA were common. CONCLUSION Although these subprotocols did not meet the primary end point for ORR, significant responses or prolonged SD noted in some disease subtypes warrants further investigation.
Collapse
Affiliation(s)
- Kari B. Wisinski
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI
| | - Yael Flamand
- Dana Farber Cancer Institute—ECOG-ACRIN Biostatistics Center, Boston, MA
| | - Melissa A. Wilson
- Department of Oncology, Division of Hematology/Medical Oncology, St Luke's University Health Network, Easton, PA
| | - Jason J. Luke
- Division of Hematology/Oncology, University of Pittsburgh Medical Center, Hillman Cancer Center, Pittsburgh, PA
| | | | - Fangxin Hong
- Dana Farber Cancer Institute—ECOG-ACRIN Biostatistics Center, Boston, MA
| | | | - James A. Zwiebel
- Investigational Drug Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Helen Chen
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Robert J. Gray
- Dana Farber Cancer Institute—ECOG-ACRIN Biostatistics Center, Boston, MA
| | - Shuli Li
- Dana Farber Cancer Institute—ECOG-ACRIN Biostatistics Center, Boston, MA
| | - Lisa M. McShane
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Lawrence V. Rubinstein
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - David Patton
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, Bethesda, MD
| | | | | | | | | | - Barbara A. Conley
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | | - Lyndsay N. Harris
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | | - Alice P. Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | |
Collapse
|
10
|
Development of a prognostic model for children with neuroblastoma based on necroptosis-related genes. Front Genet 2022; 13:947000. [PMID: 35991559 PMCID: PMC9389598 DOI: 10.3389/fgene.2022.947000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Neuroblastoma (NBL) is a rare malignant tumor of the peripheral sympathetic nervous system in children with a low overall survival rate. Recent studies have revealed the important role of necroptosis in the occurrence and development of many kinds of tumors. In this study, a prognostic model based on necroptosis-related genes was constructed for NBL. Methods: Expression profiles and clinical information for patients with NBL were downloaded from TARGET. Data for necroptosis-related genes were extracted for Cox regression and lasso regression analyses to evaluate factors associated with prognosis and to construct a prognostic model. Data from the GEO datasets GSE62564 and GSE85047 were used for external verification. Associations between risk scores were calculated, and immune infiltration, drug sensitivity, and mutation analyses were conducted. Functional enrichment analyses of genes in the prognostic model were performed. Results: Six necroptosis-related genes (i.e., CYLD, JAK1, APC, ERH, CNBP, and BAX) were selected to construct a prognostic risk model. The risk score was highly correlated with levels of infiltration of multiple immune cells and sensitivity to common antineoplastic drugs. In addition, the risk score was identified as an independent prognostic factor for patients with NBL. Conclusion: We constructed and validated a prognostic model based on necroptosis-related genes, providing insights into the development and progression of NBL and a basis for improved management. In addition to providing a tool for clinical decision-making, these findings support the importance of necroptosis in NBL and may guide the development of therapeutic strategies targeting this process.
Collapse
|
11
|
Del Bello B, Gamberucci A, Marcolongo P, Maellaro E. The autophagy inducer trehalose stimulates macropinocytosis in NF1-deficient glioblastoma cells. Cancer Cell Int 2022; 22:232. [PMID: 35864494 PMCID: PMC9306097 DOI: 10.1186/s12935-022-02652-5] [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: 04/13/2022] [Accepted: 06/29/2022] [Indexed: 12/03/2022] Open
Abstract
Background Glioblastoma is a highly aggressive brain tumor. A big effort is required to find novel molecules which can cross the blood–brain barrier and efficiently kill these tumor cells. In this perspective, trehalose (α-glucopyranosyl‐[1→1]‐α‐d‐glucopyranoside), found in various dietary sources and used as a safe nutrient supplement, attracted our attention for its pleiotropic effects against tumor cells. Methods Human glioblastoma cell lines U373-MG and T98G were exposed to trehalose and analyzed at different time points. Cell proliferation was evaluated at medium term, and clonogenic capacity and cell morphology were evaluated at long term. Western blot was used to evaluate biochemical markers of autophagy (also measured in cells co-treated with EIPA or chloroquine), and mTOR, AMPK and ERK 1/2 signalling. Macropinocytosis was evaluated morphologically by bright-field microscopy; in cells loaded with the fluorescein-conjugated fluid-phase tracer dextran, macropinocytic vacuoles were also visualized by fluorescence microscopy, and the extent of macropinocytosis was quantified by flow cytometry. Results The long-term effect of trehalose on U373-MG and T98G cell lines was impressive, as indicated by a dramatic reduction in clonogenic efficiency. Mechanistically, trehalose proved to be an efficient autophagy inducer in macropinocytosis-deficient T98G cells and an efficient inducer of macropinocytosis and eventual cell death by methuosis in U373-MG glioblastoma cells, proved to be poorly responsive to induction of autophagy. These two processes appeared to act in a mutually exclusive manner; indeed, co-treatment of U373-MG cells with the macropinocytosis inhibitor, EIPA, significantly increased the autophagic response. mTOR activation and AMPK inhibition occurred in a similar way in the two trehalose-treated cell lines. Interestingly, ERK 1/2 was activated only in macropinocytosis-proficient U373-MG cells harbouring loss-of-function mutations in the negative RAS regulator, NF1, suggesting a key role of RAS signalling. Conclusions Our results indicate that trehalose is worthy of further study as a candidate molecule for glioblastoma therapy, due to its capacity to induce a sustained autophagic response, ultimately leading to loss of clonogenic potential, and more interestingly, to force macropinocytosis, eventually leading to cell death by methuosis, particularly in tumor cells with RAS hyperactivity. As a further anticancer strategy, stimulation of macropinocytosis may be exploited to increase intracellular delivery of anticancer drugs.
Collapse
Affiliation(s)
- Barbara Del Bello
- Department of Molecular and Developmental Medicine, University of Siena, Via A. Moro, 53100, Siena, Italy
| | - Alessandra Gamberucci
- Department of Molecular and Developmental Medicine, University of Siena, Via A. Moro, 53100, Siena, Italy
| | - Paola Marcolongo
- Department of Molecular and Developmental Medicine, University of Siena, Via A. Moro, 53100, Siena, Italy
| | - Emilia Maellaro
- Department of Molecular and Developmental Medicine, University of Siena, Via A. Moro, 53100, Siena, Italy.
| |
Collapse
|
12
|
Izquierdo E, Carvalho DM, Mackay A, Temelso S, Boult JK, Pericoli G, Fernandez E, Das M, Molinari V, Grabovska Y, Rogers RF, Ajmone-Cat MA, Proszek PZ, Stubbs M, Depani S, O'Hare P, Yu L, Roumelioti G, Choudhary JS, Clarke M, Fairchild AR, Jacques TS, Grundy RG, Howell L, Picton S, Adamski J, Wilson S, Gray JC, Zebian B, Marshall LV, Carceller F, Grill J, Vinci M, Robinson SP, Hubank M, Hargrave D, Jones C. DIPG Harbors Alterations Targetable by MEK Inhibitors, with Acquired Resistance Mechanisms Overcome by Combinatorial Inhibition. Cancer Discov 2022; 12:712-729. [PMID: 34737188 PMCID: PMC7612484 DOI: 10.1158/2159-8290.cd-20-0930] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/04/2021] [Accepted: 10/19/2021] [Indexed: 11/16/2022]
Abstract
The survival of children with diffuse intrinsic pontine glioma (DIPG) remains dismal, with new treatments desperately needed. In a prospective biopsy-stratified clinical trial, we combined detailed molecular profiling and drug screening in newly established patient-derived models in vitro and in vivo. We identified in vitro sensitivity to MEK inhibitors in DIPGs harboring MAPK pathway alterations, but treatment of patient-derived xenograft models and a patient at relapse failed to elicit a significant response. We generated trametinib-resistant clones in a BRAFG469V model through continuous drug exposure and identified acquired mutations in MEK1/2 with sustained pathway upregulation. These cells showed hallmarks of mesenchymal transition and expression signatures overlapping with inherently trametinib-insensitive patient-derived cells, predicting sensitivity to dasatinib. Combined trametinib and dasatinib showed highly synergistic effects in vitro and on ex vivo brain slices. We highlight the MAPK pathway as a therapeutic target in DIPG and show the importance of parallel resistance modeling and combinatorial treatments for meaningful clinical translation. SIGNIFICANCE We report alterations in the MAPK pathway in DIPGs to confer initial sensitivity to targeted MEK inhibition. We further identify for the first time the mechanism of resistance to single-agent targeted therapy in these tumors and suggest a novel combinatorial treatment strategy to overcome it in the clinic. This article is highlighted in the In This Issue feature, p. 587.
Collapse
Affiliation(s)
- Elisa Izquierdo
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Diana M. Carvalho
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Alan Mackay
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Sara Temelso
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Jessica K.R. Boult
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Giulia Pericoli
- Department of Haematology/Oncology, Gene and Cell Therapy, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| | - Elisabet Fernandez
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Molina Das
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Valeria Molinari
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Yura Grabovska
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Rebecca F. Rogers
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | | | - Paula Z. Proszek
- Molecular Diagnostics, Royal Marsden Hospital NHS Trust, Sutton, United Kingdom
| | - Mark Stubbs
- Division of Cancer Therapeutics, Institute of Cancer Research, London, United Kingdom
| | - Sarita Depani
- Department of Haematology and Oncology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Patricia O'Hare
- Department of Haematology and Oncology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Lu Yu
- Division of Cancer Biology, Institute of Cancer Research, London, United Kingdom
| | - Georgia Roumelioti
- Division of Cancer Biology, Institute of Cancer Research, London, United Kingdom
| | - Jyoti S. Choudhary
- Division of Cancer Biology, Institute of Cancer Research, London, United Kingdom
| | - Matthew Clarke
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Amy R. Fairchild
- UCL Great Ormond Street Institute for Child Health, London, United Kingdom
| | - Thomas S. Jacques
- UCL Great Ormond Street Institute for Child Health, London, United Kingdom
| | - Richard G. Grundy
- Children's Brain Tumour Research Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Lisa Howell
- Alder Hey Children's NHS Foundation Trust, Liverpool, United Kingdom
| | - Susan Picton
- Leeds Children's Hospital, Leeds, United Kingdom
| | - Jenny Adamski
- Birmingham Women's and Children's Hospital, Birmingham, United Kingdom
| | - Shaun Wilson
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Juliet C. Gray
- Centre for Cancer Immunology, University of Southampton, Southampton, United Kingdom
| | - Bassel Zebian
- Department of Neurosurgery, Kings College Hospital NHS Trust, London, United Kingdom
| | - Lynley V. Marshall
- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
- Children & Young People's Unit, Royal Marsden Hospital NHS Trust, Sutton, United Kingdom
| | - Fernando Carceller
- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
- Children & Young People's Unit, Royal Marsden Hospital NHS Trust, Sutton, United Kingdom
| | - Jacques Grill
- Department of Pediatric and Adolescent Oncology and INSERM Unit U891, Team “Genomics and Oncogenesis of Pediatric Brain Tumors,” Gustave Roussy and University Paris-Saclay, Villejuif, France
| | - Maria Vinci
- Department of Haematology/Oncology, Gene and Cell Therapy, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| | - Simon P. Robinson
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Michael Hubank
- Molecular Diagnostics, Royal Marsden Hospital NHS Trust, Sutton, United Kingdom
| | - Darren Hargrave
- Department of Haematology and Oncology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
- UCL Great Ormond Street Institute for Child Health, London, United Kingdom
| | - Chris Jones
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
13
|
Clinicopathological and genomic characterization of BCORL1-driven high-grade endometrial stromal sarcomas. Mod Pathol 2021; 34:2200-2210. [PMID: 34302054 DOI: 10.1038/s41379-021-00873-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 12/28/2022]
Abstract
BCORL1 is a transcriptional corepressor homologous to BCOR. We describe 12 BCORL1-altered uterine sarcomas with striking resemblance to BCOR-altered endometrial stromal sarcoma (BCOR-ESS), including 5 with BCORL1 rearrangements (JAZF1-BCORL1, EP300-BCORL1, or internal BCORL1 rearrangement), 5 with inactivating BCORL1 mutations (T513fs*22, P600fs*1, R945*, R1196*, or R1265fs*4) and 2 with homozygous BCORL1 deletion. The median patient age was 57.5 years (range 33-79). An association with aggressive clinical behavior was identified. Diagnoses assigned prior to genomic testing varied: 7 tumors were previously diagnosed as ESS, 2 as high-grade uterine sarcomas, 2 as myxoid uterine leiomyosarcomas, and 1 as a uterine spindle cell neoplasm consistent with leiomyosarcoma. Tumors harbored frequent gelatinous, mucomyxoid-like appearance by gross examination and unique histology with morphological overlap with BCOR-ESS. Key microscopic features included (1) a spindle cell appearance, most often with at least focal myxoid stroma, (2) variable amounts of hypocellular fibromyxoid spindle areas with lower grade atypia and/or (3) variable amounts of epithelioid areas with higher grade atypia. Specifically, spindle and epithelioid components were present in 100 and 75% of sarcomas, respectively; myxoid stroma was identified in 83%, collagen plaques or fibrosis in 50%, and high-grade nuclear atypia was present in 42%. Like BCOR-ESS, 50% of BCORL1-altered sarcomas exhibited CDK4 amplification or CDKN2A loss. In contrast, 33% harbored NF1 alterations, while 25% had other alterations in the NF2-mTOR pathway, expanding potential therapeutic targets. In conclusion, inactivating BCORL1 genomic alterations may define a distinct subset of high-grade endometrial stromal sarcomas with biological overlap with BCOR-ESS, both of which may mimic myxoid leiomyosarcomas.
Collapse
|
14
|
Luo M, Xia Y, Wang F, Zhang H, Su D, Su C, Yang C, Wu S, An S, Lin S, Fu L. PD0325901, an ERK inhibitor, enhances the efficacy of PD-1 inhibitor in non-small cell lung carcinoma. Acta Pharm Sin B 2021; 11:3120-3133. [PMID: 34729305 PMCID: PMC8546891 DOI: 10.1016/j.apsb.2021.03.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 02/03/2021] [Accepted: 02/10/2021] [Indexed: 12/29/2022] Open
Abstract
ERK pathway regulated the programmed death ligand-1 (PD-L1) expression which was linked to the response of programmed death-1 (PD-1)/PD-L1 blockade therapy. So it is deducible that ERK inhibitor could enhance the efficacy of PD-1 inhibitor in cancer immunotherapy. In this study, PD0325901, an oral potent ERK inhibitor, strongly enhanced the efficacy of PD-1 antibody in vitro and in vivo models in non-small cell lung carcinoma (NSCLC) cells. Mechanistically, PD0325901 or shRNA-ERK1/2 significantly downregulated the PD-L1 expression in NSCLC cells and increased the CD3+ T cells infiltration and functions in tumor tissue. There was a positive correlation between the p-ERK1/2 expression and PD-L1 expression in patients with NSCLC. And the patients with low p-ERK1/2 expression were observed a high response rate of PD-1/PD-L1 blockage therapy. Our results demonstrate that PD0325901, an ERK inhibitor, can enhance the efficacy of PD-1 blockage against NSCLC in vitro and in vivo models. And the combination of ERK inhibitor such as PD0325901 and PD-1/PD-L1 blockage is a promising regimen and encouraged to be further confirmed in the treatment of patients with NSCLC.
Collapse
Affiliation(s)
- Min Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yuhui Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Fang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Hong Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Danting Su
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Chaoyue Su
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Pharmacy College, Guangzhou Medical University, Guangzhou 510182, China
| | - Chuan Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Shaocong Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Sainan An
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Suxia Lin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Corresponding authors. Tel.: +86 20 873431-63, fax: +86 20 87343170.
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Corresponding authors. Tel.: +86 20 873431-63, fax: +86 20 87343170.
| |
Collapse
|
15
|
Zaki MM, Mashouf LA, Woodward E, Langat P, Gupta S, Dunn IF, Wen PY, Nahed BV, Bi WL. Genomic landscape of gliosarcoma: distinguishing features and targetable alterations. Sci Rep 2021; 11:18009. [PMID: 34504233 PMCID: PMC8429571 DOI: 10.1038/s41598-021-97454-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 07/19/2021] [Indexed: 12/20/2022] Open
Abstract
Gliosarcoma is an aggressive brain tumor with histologic features of glioblastoma (GBM) and soft tissue sarcoma. Despite its poor prognosis, its rarity has precluded analysis of its underlying biology. We used a multi-center database to characterize the genomic landscape of gliosarcoma. Sequencing data was obtained from 35 gliosarcoma patients from Genomics Evidence Neoplasia Information Exchange (GENIE) 5.0, a database curated by the American Association of Cancer Research (AACR). We analyzed genomic alterations in gliosarcomas and compared them to GBM (n = 1,449) and soft tissue sarcoma (n = 1,042). 30 samples were included (37% female, median age 59 [IQR: 49–64]). Nineteen common genes were identified in gliosarcoma, defined as those altered in > 5% of samples, including TERT Promoter (92%), PTEN (66%), and TP53 (60%). Of the 19 common genes in gliosarcoma, 6 were also common in both GBM and soft tissue sarcoma, 4 in GBM alone, 0 in soft tissue sarcoma alone, and 9 were more distinct to gliosarcoma. Of these, BRAF harbored an OncoKB level 1 designation, indicating its status as a predictive biomarker of response to an FDA-approved drug in certain cancers. EGFR, CDKN2A, NF1, and PTEN harbored level 4 designations in solid tumors, indicating biological evidence of these biomarkers predicting a drug-response. Gliosarcoma contains molecular features that overlap GBM and soft tissue sarcoma, as well as its own distinct genomic signatures. This may play a role in disease classification and inclusion criteria for clinical trials. Gliosarcoma mutations with potential therapeutic indications include BRAF, EGFR, CDKN2A, NF1, and PTEN.
Collapse
Affiliation(s)
- Mark M Zaki
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA.,Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Leila A Mashouf
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Eleanor Woodward
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Pinky Langat
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Saksham Gupta
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Ian F Dunn
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Patrick Y Wen
- Center for NeuroOncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Brian V Nahed
- Center for NeuroOncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Wenya Linda Bi
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA.
| |
Collapse
|
16
|
Ronsley R, Hounjet CD, Cheng S, Rassekh SR, Duncan WJ, Dunham C, Gardiner J, Ghag A, Ludemann JP, Wensley D, Rehmus W, Sargent MA, Hukin J. Trametinib therapy for children with neurofibromatosis type 1 and life-threatening plexiform neurofibroma or treatment-refractory low-grade glioma. Cancer Med 2021; 10:3556-3564. [PMID: 33939292 PMCID: PMC8178485 DOI: 10.1002/cam4.3910] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose To describe a series of children with extensive PNF or treatment refractory PLGG treated on a compassionate basis with trametinib. Methods We report on six patients with NF‐1 treated with trametinib on a compassionate basis at British Columbia Children's Hospital since 2017. Data were collected retrospectively from the patient record. RAPNO and volumetric criteria were used to evaluate the response of intracranial and extracranial lesions, respectively. Results Subjects were 21 months to 14 years old at the time of initiation of trametinib therapy and 3/6 subjects are male. Duration of therapy was 4–28 months at the time of this report. All patients had partial response or were stable on analysis. Two patients with life‐threatening PNF had a partial radiographic response in tandem with significant clinical improvement and developmental catch up. One subject discontinued therapy after 6 months due to paronychia and inadequate response. The most common adverse effect (AE) was grade 1–2 paronychia or dermatitis in 5/6 patients. There were no grade 3 or 4 AEs. At the time of this report, five patients remain on therapy. Conclusion Trametinib is an effective therapy for advanced PNF and refractory PLGG in patients with NF‐1 and is well tolerated in children. Further data and clinical trials are required to assess tolerance, efficacy and durability of response, and length of treatment required in such patients.
Collapse
Affiliation(s)
- Rebecca Ronsley
- Division of Hematology, Oncology & BMT, Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Celine D Hounjet
- Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Sylvia Cheng
- Division of Hematology, Oncology & BMT, Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Shahrad Rod Rassekh
- Division of Hematology, Oncology & BMT, Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Walter J Duncan
- Division of Pediatric Cardiology, Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Christopher Dunham
- Division of Anatomic Pathology, Department of Pathology, University of British Columbia, Vancouver, Canada
| | - Jane Gardiner
- Division of Pediatric Ophthalmology, Department of Surgery, University of British Columbia, Vancouver, Canada
| | - Arvindera Ghag
- Division of Pediatric Orthopedic Surgery, Department of Surgery, University of British Columbia, Vancouver, Canada
| | - Jeffrey P Ludemann
- Division of Pediatric Otolaryngology, Department of Surgery, University of British Columbia, Vancouver, Canada
| | - David Wensley
- Division of Pediatric Respiratory Medicine, Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Wingfield Rehmus
- Division of Dermatology, Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Michael A Sargent
- Division of Pediatric Neuro-Radiology, Department of Radiology, University of British Columbia, Vancouver, Canada
| | - Juliette Hukin
- Division of Hematology, Oncology & BMT, Department of Pediatrics, University of British Columbia, Vancouver, Canada.,Division of Neurology, Department of Pediatrics, University of British Columbia, Vancouver, Canada
| |
Collapse
|
17
|
Harder A. MEK inhibitors - novel targeted therapies of neurofibromatosis associated benign and malignant lesions. Biomark Res 2021; 9:26. [PMID: 33863389 PMCID: PMC8052700 DOI: 10.1186/s40364-021-00281-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 03/30/2021] [Indexed: 12/18/2022] Open
Abstract
MAP/ERK kinase 1 and 2 (MEK 1/2) inhibitors (MEKi) are investigated in several trials to treat lesions that arise from pathogenic variants of the Neurofibromatosis type 1 and type 2 genes (NF1, NF2). These trials showed that MEKi are capable to shrink volume of low grade gliomas and plexiform neurofibromas in NF1. Targeting other lesions being associated with a high morbidity in NF1 seems to be promising. Due to involvement of multiple pathways in NF2 associated lesions as well as in malignant tumors, MEKi are also used in combination therapies. This review outlines the current state of MEKi application in neurofibromatosis and associated benign and malignant lesions.
Collapse
Affiliation(s)
- Anja Harder
- Institute of Pathology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 14, 06120, Halle (Saale), Germany. .,Institute of Neuropathology, University Hospital Münster, Münster, Germany. .,Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, the Brandenburg Medical School Theodor Fontane and the University of Potsdam, Potsdam, Germany.
| |
Collapse
|
18
|
Attina G, Maurizi P, Triarico S, Capozza MA, Romano A, Mastrangelo S, Ruggiero A. Management of Children with Optic Gliomas and Neurofibromatosis Type 1. BIOMEDICAL AND PHARMACOLOGY JOURNAL 2020; 13:1601-1606. [DOI: 10.13005/bpj/2035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Optic pathway gliomas (OPG) are a common cancer in children with neurofibromatosis type 1. OPGs can cause clinical symptoms such as reduction of visual acuity, alterations of the visual field, pallor of the optical papilla, strabismus, endocrinological alterations up to diencephalic syndrome.The current guidelines provide for wait and see as the main approach if the tumor is not causing visual deterioration and adopting treatment only in the event of significant impairment of the visual function. Therefore, it is essential to early detect the visual deterioration changes as well as the identification of children eligible for treatment.
Collapse
Affiliation(s)
- Giorgio Attina
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Palma Maurizi
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Silvia Triarico
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Michele Antonio Capozza
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Alberto Romano
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Stefano Mastrangelo
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Antonio Ruggiero
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| |
Collapse
|
19
|
Park SY, Mittal S, Dong J, Jeong K, Martinez-Ledesma E, Piao Y, Khan S, Henry V, Verhaak RGW, Majd N, Balasubramaniyan V, de Groot JF. Depletion of CLK2 sensitizes glioma stem-like cells to PI3K/mTOR and FGFR inhibitors. Am J Cancer Res 2020; 10:3765-3783. [PMID: 33294266 PMCID: PMC7716149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 09/10/2020] [Indexed: 06/12/2023] Open
Abstract
The Cdc2-like kinases (CLKs) regulate RNA splicing and have been shown to suppress cell growth. Knockdown of CLK2 was found to block glioma stem-like cell (GSC) growth in vivo through the AKT/FOXO3a/p27 pathway without activating mTOR and MAPK signaling, suggesting that these pathways mediate resistance to CLK2 inhibition. We identified CLK2 binding partners using immunoprecipitation assays and confirmed their interactions in vitro in GSCs. We then tested the cellular viability of several signaling inhibitors in parental and CLK2 knockdown GSCs. Our results demonstrate that CLK2 binds to 14-3-3τ isoform and prevents its ubiquitination in GSCs. Stable CLK2 knockdown increased PP2A activity and activated PI3K signaling. Treatment with a PI3K/mTOR inhibitor in CLK2 knockdown cells led to a modest reduction in cell viability compared to drug treatment alone at a lower dose. However, FGFR inhibitor in CLK2 knockdown cells led to a decrease in cell viability and increased apoptosis. Reduced expression of CLK2 in glioblastoma, in combination with FGFR inhibitors, led to synergistic apoptosis induction and cell cycle arrest compared to blockade or either kinase alone.
Collapse
Affiliation(s)
- Soon Young Park
- Department of Cell Developmental and Cancer Biology, Oregon Health and Science UniversityPortland, Oregon, USA
| | - Sandeep Mittal
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Jianwen Dong
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Kangjin Jeong
- Department of Cell Developmental and Cancer Biology, Oregon Health and Science UniversityPortland, Oregon, USA
| | | | - Yuji Piao
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Sabbir Khan
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Verlene Henry
- Department of Neuro-Surgery, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Roel GW Verhaak
- The Jackson Laboratory for Genomic MedicineFarmington, Connecticut, USA
| | - Nazanin Majd
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | | | - John F de Groot
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| |
Collapse
|
20
|
Schreck KC, Allen AN, Wang J, Pratilas CA. Combination MEK and mTOR inhibitor therapy is active in models of glioblastoma. Neurooncol Adv 2020; 2:vdaa138. [PMID: 33235998 PMCID: PMC7668446 DOI: 10.1093/noajnl/vdaa138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background RAS effector signaling pathways such as PI3K/mTOR and ERK are frequently dysregulated in glioblastoma. While small molecule targeted therapies against these pathways have appeared promising in preclinical studies, they have been disappointing in clinical trials due to toxicity and de novo and adaptive resistance. To identify predictors of glioblastoma sensitivity to dual pathway inhibition with mTORC1/2 and MEK inhibitors, we tested these agents, alone and in combination, in a cohort of genomically characterized glioblastoma cell lines. Methods Seven genomically characterized, patient-derived glioblastoma neurosphere cell lines were evaluated for their sensitivity to the dual mTORC1/2 kinase inhibitor sapanisertib (MLN0128, TAK-228) alone or in combination with the MEK1/2 inhibitor trametinib (GSK1120212), using assessment of proliferation and evaluation of the downstream signaling consequences of these inhibitors. Results Sapanisertib inhibited cell growth in neurosphere lines, but induced apoptosis only in a subset of lines, and did not completely inhibit downstream mTOR signaling via ribosomal protein S6 (RPS6). Growth sensitivity to MEK inhibitor monotherapy was observed in a subset of lines defined by loss of NF1, was predicted by an ERK-dependent expression signature, and was associated with effective phospho-RPS6 inhibition. In these lines, combined MEK/mTOR treatment further inhibited growth and induced apoptosis. Combined MEK and mTOR inhibition also led to modest antiproliferative effects in lines with intact NF1 and insensitivity to MEK inhibitor monotherapy. Conclusions These data demonstrate that combined MEK/mTOR inhibition is synergistic in glioblastoma cell lines and may be more potent in NF1-deficient glioblastoma.
Collapse
Affiliation(s)
- Karisa C Schreck
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amy N Allen
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, USA.,Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jiawan Wang
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, USA.,Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Christine A Pratilas
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
21
|
Whitehouse JP, Howlett M, Hii H, Mayoh C, Wong M, Barahona P, Ajuyah P, White CL, Buntine MK, Dyke JM, Lee S, Valvi S, Stanley J, Andradas C, Carline B, Kuchibhotla M, Ekert PG, Cowley MJ, Gottardo NG, Endersby R. A Novel Orthotopic Patient-Derived Xenograft Model of Radiation-Induced Glioma Following Medulloblastoma. Cancers (Basel) 2020; 12:cancers12102937. [PMID: 33053751 PMCID: PMC7600047 DOI: 10.3390/cancers12102937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022] Open
Abstract
Radiation-induced glioma (RIG) is a highly aggressive brain cancer arising as a consequence of radiation therapy. We report a case of RIG that arose in the brain stem following treatment for paediatric medulloblastoma, and the development and characterisation of a matched orthotopic patient-derived xenograft (PDX) model (TK-RIG915). Patient and PDX tumours were analysed using DNA methylation profiling, whole genome sequencing (WGS) and RNA sequencing. While initially thought to be a diffuse intrinsic pontine glioma (DIPG) based on disease location, results from methylation profiling and WGS were not consistent with this diagnosis. Furthermore, clustering analyses based on RNA expression suggested the tumours were distinct from primary DIPG. Additional gene expression analysis demonstrated concordance with a published RIG expression profile. Multiple genetic alterations that enhance PI3K/AKT and Ras/Raf/MEK/ERK signalling were discovered in TK-RIG915 including an activating mutation in PIK3CA, upregulation of PDGFRA and AKT2, inactivating mutations in NF1, and a gain-of-function mutation in PTPN11. Additionally, deletion of CDKN2A/B, increased IDH1 expression, and decreased ARID1A expression were observed. Detection of phosphorylated S6, 4EBP1 and ERK via immunohistochemistry confirmed PI3K pathway and ERK activation. Here, we report one of the first PDX models for RIG, which recapitulates the patient disease and is molecularly distinct from primary brain stem glioma. Genetic interrogation of this model has enabled the identification of potential therapeutic vulnerabilities in this currently incurable disease.
Collapse
Affiliation(s)
- Jacqueline P. Whitehouse
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Centre for Child Health Research, University of Western Australia, Nedlands 6009, Australia
| | - Meegan Howlett
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Centre for Child Health Research, University of Western Australia, Nedlands 6009, Australia
| | - Hilary Hii
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
| | - Chelsea Mayoh
- Children’s Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington 2033, Australia; (C.M.); (M.W.); (P.B.); (P.A.); (P.G.E.); (M.J.C.)
- School of Women’s and Children’s Health, UNSW Sydney, Kensington 2033, Australia
| | - Marie Wong
- Children’s Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington 2033, Australia; (C.M.); (M.W.); (P.B.); (P.A.); (P.G.E.); (M.J.C.)
- School of Women’s and Children’s Health, UNSW Sydney, Kensington 2033, Australia
| | - Paulette Barahona
- Children’s Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington 2033, Australia; (C.M.); (M.W.); (P.B.); (P.A.); (P.G.E.); (M.J.C.)
| | - Pamela Ajuyah
- Children’s Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington 2033, Australia; (C.M.); (M.W.); (P.B.); (P.A.); (P.G.E.); (M.J.C.)
| | - Christine L. White
- Genetics and Molecular Pathology Laboratory, Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia; (C.L.W.); (M.K.B.)
- Department of Molecular and Translational Science, Monash University, Melbourne 3168, Victoria, Australia
| | - Molly K. Buntine
- Genetics and Molecular Pathology Laboratory, Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia; (C.L.W.); (M.K.B.)
- Department of Molecular and Translational Science, Monash University, Melbourne 3168, Victoria, Australia
| | - Jason M. Dyke
- Department of Neuropathology, PathWest Laboratory Medicine, Royal Perth Hospital, Perth 6000, Australia;
- Pathology and Laboratory Medicine, University of Western Australia, Nedlands 6009, Australia
| | - Sharon Lee
- Department of Neurosurgery, Perth Children’s Hospital, Nedlands 6009, Australia;
| | - Santosh Valvi
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Department of Paediatric and Adolescent Oncology/Haematology, Perth Children’s Hospital, Nedlands 6009, Australia
- Division of Paediatrics, University of Western Australia Medical School, Nedlands 6009, Australia
| | - Jason Stanley
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Centre for Child Health Research, University of Western Australia, Nedlands 6009, Australia
| | - Clara Andradas
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Centre for Child Health Research, University of Western Australia, Nedlands 6009, Australia
| | - Brooke Carline
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
| | - Mani Kuchibhotla
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
| | - Paul G. Ekert
- Children’s Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington 2033, Australia; (C.M.); (M.W.); (P.B.); (P.A.); (P.G.E.); (M.J.C.)
- School of Women’s and Children’s Health, UNSW Sydney, Kensington 2033, Australia
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville 3052, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne 3000, Victoria, Australia
| | - Mark J. Cowley
- Children’s Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington 2033, Australia; (C.M.); (M.W.); (P.B.); (P.A.); (P.G.E.); (M.J.C.)
- School of Women’s and Children’s Health, UNSW Sydney, Kensington 2033, Australia
| | - Nicholas G. Gottardo
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Centre for Child Health Research, University of Western Australia, Nedlands 6009, Australia
- Department of Paediatric and Adolescent Oncology/Haematology, Perth Children’s Hospital, Nedlands 6009, Australia
| | - Raelene Endersby
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Centre for Child Health Research, University of Western Australia, Nedlands 6009, Australia
- Correspondence:
| |
Collapse
|
22
|
Nix JS, Yuan M, Imada EL, Ames H, Marchionni L, Gutmann DH, Rodriguez FJ. Global microRNA profiling identified miR-10b-5p as a regulator of neurofibromatosis 1 (NF1)-glioma migration. Neuropathol Appl Neurobiol 2020; 47:96-107. [PMID: 32603552 DOI: 10.1111/nan.12641] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 06/14/2020] [Accepted: 06/30/2020] [Indexed: 01/12/2023]
Abstract
AIMS Neurofibromatosis 1 (NF1) is an autosomal-dominant cancer predisposition syndrome caused by loss of function alterations involving the NF1 locus on chromosome 17. The most common brain tumours encountered in affected patients are low-grade gliomas (pilocytic astrocytomas), although high-grade gliomas are also observed at increased frequency. While bi-allelic NF1 loss characterizes these tumours, previous studies have suggested noncoding RNA molecules (microRNA, miR) may have important roles in dictating glioma biology. METHODS To explore the contributions of miRs in NF1-associated gliomas, we analysed five high-grade gliomas (NF1-HGG) and five PAs (NF1-PA) using global microRNA profiling with NanoString-based microarrays followed by functional experiments with glioma cell lines. RESULTS miR-10b-5p, miR-135b-5p, miR-196a-5p, miR-196b-5p, miR-1247-5p and miR-320a (adjusted P < 0.05) were increased> 3-fold in NF1-HGG relative to NF1-PA tumours. In addition, miR-378b and miR-1305 were decreased 6.8- and 6-fold, respectively, whereas miR-451a was increased 2.7-fold (adjusted P < 0.05) in NF1-PAs compared to non-neoplastic NF1 patient brain specimens (n = 2). As miR-10b-5p was the microRNA overexpressed the most in NF1-high-grade glioma compared to NF1-low-grade glioma (5.76 fold), we examined its levels in glioma cell lines. miR-10b-5p levels were highest in adult glioma cell lines and lowest in paediatric low-grade glioma lines (P = 0.02). miR-10b-5p knockdown resulted in decreased invasion in NF1-deficient LN229 high-grade glioma line, whereas its overexpression in the NF1-PA derived line (JHH-NF1-PA1) led to increased invasion. There was no change in cell growth (viability and proliferation). CONCLUSIONS These proof-of-concept experiments support a role for microRNA regulation in NF1-glioma biology.
Collapse
Affiliation(s)
- J S Nix
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - M Yuan
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - E L Imada
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - H Ames
- Department of Pathology, University of Maryland, Baltimore, MD, USA
| | - L Marchionni
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - D H Gutmann
- Department of Neurology, Washington University, St. Louis, Missouri, USA
| | - F J Rodriguez
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Departments of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
23
|
Cho HJ, Zhao J, Jung SW, Ladewig E, Kong DS, Suh YL, Lee Y, Kim D, Ahn SH, Bordyuh M, Kang HJ, Sa JK, Seo YJ, Kim ST, Lim DH, Dho YS, Lee JI, Seol HJ, Choi JW, Park WY, Park CK, Rabadan R, Nam DH. Distinct genomic profile and specific targeted drug responses in adult cerebellar glioblastoma. Neuro Oncol 2020; 21:47-58. [PMID: 30085274 DOI: 10.1093/neuonc/noy123] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background Despite extensive efforts on the genomic characterization of gliomas, very few studies have reported the genetic alterations of cerebellar glioblastoma (C-GBM), a rare and lethal disease. Here, we provide a systematic study of C-GBM to better understand its specific genomic features. Methods We collected a cohort of C-GBM patients and compared patient demographics and tumor pathologies with supratentorial glioblastoma (S-GBM). To uncover the molecular characteristics, we performed DNA and mRNA sequencing and DNA methylation arrays on 19, 6, and 4 C-GBM cases, respectively. Moreover, chemical drug screening was conducted to identify potential therapeutic options for C-GBMs. Results Despite differing anatomical origins of C-GBM and S-GBM, neither histological, cytological, nor patient demographics appeared significantly different between the 2 types. However, we observed striking differences in mutational patterns, including frequent alterations of ATRX, PDGFRA, NF1, and RAS and absence of EGFR alterations in C-GBM. These results show a distinct evolutionary path in C-GBM, suggesting specific therapeutic targeted options. Targeted-drug screening revealed that C-GBMs were more responsive to mitogen-activated protein kinase kinase (MEK) inhibitor and resistant to epidermal growth factor receptor inhibitors than S-GBMs. Also, differential expression analysis indicated that C-GBMs may have originated from oligodendrocyte progenitor cells, suggesting that different types of cells can undergo malignant transformation according to their location in brain. Master regulator analysis with differentially expressed genes between C-GBM and proneural S-GBM revealed NR4A1 as a potential therapeutic target. Conclusions Our results imply that unique gliomagenesis mechanisms occur in adult cerebellum and new treatment strategies are needed to provide greater therapeutic benefits for C-GBM patients. Key Points 1. Distinct genomic profiles of 19 adult cerebellar GBMs were characterized. 2. MEK inhibitor was highly sensitive to cerebellar GBM compared with supratentorial GBM. 3. Master regulator analysis revealed NR4A1 as a potential therapeutic target in cerebellar GBM.
Collapse
Affiliation(s)
- Hee Jin Cho
- Institute for Refractory Cancer Research, Seoul, Korea.,Research Institute for Future Medicine, Seoul, Korea
| | - Junfei Zhao
- Department of Systems Biology, Columbia University, New York, New York, USA.,Department of Biomedical Informatics, Columbia University, New York, New York, USA
| | - Sang Won Jung
- Institute for Refractory Cancer Research, Seoul, Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
| | - Erik Ladewig
- Department of Systems Biology, Columbia University, New York, New York, USA.,Department of Biomedical Informatics, Columbia University, New York, New York, USA
| | - Doo-Sik Kong
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yeon-Lim Suh
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yeri Lee
- Institute for Refractory Cancer Research, Seoul, Korea.,Research Institute for Future Medicine, Seoul, Korea
| | - Donggeon Kim
- Institute for Refractory Cancer Research, Seoul, Korea.,Research Institute for Future Medicine, Seoul, Korea
| | - Sun Hee Ahn
- Institute for Refractory Cancer Research, Seoul, Korea
| | - Mykola Bordyuh
- Department of Systems Biology, Columbia University, New York, New York, USA.,Department of Biomedical Informatics, Columbia University, New York, New York, USA
| | - Hyun Ju Kang
- Institute for Refractory Cancer Research, Seoul, Korea.,Research Institute for Future Medicine, Seoul, Korea
| | - Jason K Sa
- Institute for Refractory Cancer Research, Seoul, Korea.,Research Institute for Future Medicine, Seoul, Korea
| | - Yun Jee Seo
- Institute for Refractory Cancer Research, Seoul, Korea
| | - Sung Tae Kim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Do Hoon Lim
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yun-Sik Dho
- Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jung-Il Lee
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ho Jun Seol
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jung Won Choi
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
| | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Raul Rabadan
- Department of Systems Biology, Columbia University, New York, New York, USA.,Department of Biomedical Informatics, Columbia University, New York, New York, USA
| | - Do-Hyun Nam
- Institute for Refractory Cancer Research, Seoul, Korea.,Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
24
|
Nix JS, Blakeley J, Rodriguez FJ. An update on the central nervous system manifestations of neurofibromatosis type 1. Acta Neuropathol 2020; 139:625-641. [PMID: 30963251 DOI: 10.1007/s00401-019-02002-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 03/30/2019] [Accepted: 04/01/2019] [Indexed: 02/06/2023]
Abstract
Neurofibromatosis 1 (NF1) is an autosomal dominant genetic disorder that presents with variable phenotypes as a result of mutations in the neurofibromatosis type 1 (NF1) gene and subsequently, abnormal function of the protein product, neurofibromin. Patients with NF1 are at increased risk for central nervous system (CNS) manifestations including structural, functional, and neoplastic disease. The mechanisms underlying the varied manifestations of NF1 are incompletely understood, but the loss of functional neurofibromin, resulting in sustained activation of the oncoprotein RAS, is responsible for tumorigenesis throughout the body, including the CNS. Much of our understanding of NF1-related CNS manifestations is from a combination of data from animal models and natural history studies of people with NF1 and CNS disease. Data from animal models suggest the importance of both Nf1 mutations and somatic genetic alterations, such as Tp53 loss, for development of neoplasms, as well as the role of the timing of the acquisition of such alterations on the variability of CNS manifestations. A variety of non-neoplastic structural (macrocephaly, hydrocephalus, aqueductal stenosis, and vasculopathy) and functional (epilepsy, impaired cognition, attention deficits, and autism spectrum disorder) abnormalities occur with variable frequency in individuals with NF1. In addition, there is increasing evidence that similar appearing CNS neoplasms in people with and without the NF1 syndrome are due to distinct oncogenic pathways. Gliomas in people with NF1 show alterations in the RAS/MAPK pathway, generally in the absence of BRAF alterations (common to sporadic pilocytic astrocytomas) or IDH or histone H3 mutations (common to diffuse gliomas subsets). A subset of low-grade astrocytomas in these patients remain difficult to classify using standard criteria, and occasionally demonstrate morphologic features resembling subependymal giant cell astrocytomas that afflict patients with tuberous sclerosis complex ("SEGA-like astrocytomas"). There is also emerging evidence that NF1-associated high-grade astrocytomas have frequent co-existing alterations such as ATRX mutations and an alternative lengthening of telomeres (ALT) phenotype responsible for unique biologic properties. Ongoing efforts are seeking to improve diagnostic accuracy for CNS neoplasms in the setting of NF1 versus sporadic tumors. In addition, MEK inhibitors, which act on the RAS/MAPK pathway, continue to be studied as rational targets for the treatment of NF1-associated tumors, including CNS tumors.
Collapse
|
25
|
Lin DI, Hemmerich A, Edgerly C, Duncan D, Severson EA, Huang RSP, Ramkissoon SH, Connor YD, Shea M, Hecht JL, Ali SM, Vergilio JA, Ross JS, Elvin JA. Genomic profiling of BCOR-rearranged uterine sarcomas reveals novel gene fusion partners, frequent CDK4 amplification and CDKN2A loss. Gynecol Oncol 2020; 157:357-366. [PMID: 32156473 DOI: 10.1016/j.ygyno.2020.02.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/11/2020] [Accepted: 02/14/2020] [Indexed: 02/09/2023]
Abstract
OBJECTIVE Genomic alterations of BCOR via ZC3H7B-BCOR fusion or BCOR internal tandem duplication (ITD) define a subset of endometrial stromal sarcoma (ESS). The goals of this study were to: 1) determine the molecular landscape of BCOR-rearranged ESS, 2) to identify novel BCOR fusion gene partners in ESS and their associated clinicopathological characteristics, and 3) to potentially unravel targetable genomic alterations in BCOR-mutated ESS. METHODS A retrospective database search of a CLIA-certified molecular laboratory was performed for uterine sarcomas that contained BCOR rearrangements or BCOR ITD. The cases were previously assayed by comprehensive genomic profiling via both DNA- and RNA-based targeted next generation sequencing during the course of clinical care. Clinicopathological and genomic data was centrally re-reviewed. RESULTS We identify largest cohort of BCOR-rearranged ESS to date (n = 40), which included 31 cases with canonical ZC3H7B-BCOR fusion as well as 8 cases with novel BCOR gene rearrangement partners, such as BCOR-L3MBTL2, EP300-BCOR, BCOR-NUTM2G, BCOR-RALGPS1, BCOR-MAP7D2, RGAG1-BCOR, ING3-BCOR, BCOR-NUGGC, KMT2D-BCOR, CREBBP-BCOR and 1 case with BCOR internal rearrangement. Re-review of cases with novel rearrangements demonstrated sarcomas with spindle, epithelioid or small round cell components and frequent myxoid stromal change. Comprehensive genomic profiling revealed high frequency of CDK4 and MDM2 amplification in 38% and 45% of BCOR-rearranged cases, respectively, and homozygous deletion of CDKN2A, which encodes an inhibitor of CDK4 in 28% of cases. Notably, CDK4 and MDM2 amplification was absent in all cases from 15 different ESS cases harboring BCOR ITD. CONCLUSIONS Alterations of CDK4 pathway members, for which targeted therapy is clinically available (i.e. palbociclib), via CDK4 amplification or CDKN2A loss, contributes to the pathogenesis of BCOR-rearranged uterine sarcomas, which may have therapeutic implications.
Collapse
Affiliation(s)
- Douglas I Lin
- Foundation Medicine Inc., Cambridge, MA, United States of America.
| | - Amanda Hemmerich
- Foundation Medicine Inc., Morrisville, NC, United States of America
| | - Claire Edgerly
- Foundation Medicine Inc., Morrisville, NC, United States of America
| | - Daniel Duncan
- Foundation Medicine Inc., Morrisville, NC, United States of America
| | - Eric A Severson
- Foundation Medicine Inc., Morrisville, NC, United States of America
| | | | - Shakti H Ramkissoon
- Foundation Medicine Inc., Morrisville, NC, United States of America; Wake Forest Comprehensive Cancer Center, Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - Yamicia D Connor
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Meghan Shea
- Department of Internal Medicine, Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Jonathan L Hecht
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Siraj M Ali
- Foundation Medicine Inc., Cambridge, MA, United States of America
| | - Jo-Anne Vergilio
- Foundation Medicine Inc., Cambridge, MA, United States of America
| | - Jeffrey S Ross
- Foundation Medicine Inc., Cambridge, MA, United States of America; Upstate Medical University, Syracuse, NY, United States of America
| | - Julia A Elvin
- Foundation Medicine Inc., Cambridge, MA, United States of America
| |
Collapse
|
26
|
Kessler T, Berberich A, Casalini B, Drüschler K, Ostermann H, Dormann A, Walter S, Hai L, Schlesner M, Herold-Mende C, Jungk C, Unterberg A, Bendszus M, Sahm K, von Deimling A, Winkler F, Platten M, Wick W, Sahm F, Wick A. Molecular profiling-based decision for targeted therapies in IDH wild-type glioblastoma. Neurooncol Adv 2020; 2:vdz060. [PMID: 32642725 PMCID: PMC7212885 DOI: 10.1093/noajnl/vdz060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Molecular profiling allows tumor classification as well as assessment of diagnostic, prognostic, and treatment-related molecular changes. Translation into clinical practice and relevance for patients has not been demonstrated yet. Methods We analyzed clinical and molecular data of isocitrate dehydrogenase wild-type glioblastoma patients with sufficient clinical follow-up from the Heidelberg Neuro-Oncology Center and with molecular analysis of tumor tissue that consisted of DNA methylation array data, genome-scale copy number variations, gene panel sequencing, and partly mTOR immunohistochemistry between October 2014 and April 2018. Results Of 536 patients screened, molecular assessment was performed in 253 patients (47%) in a prospective routine clinical setting with further clinical appointments. Therapy decision was directly based on the molecular assessment in 97 (38%) patients. Of these, genetic information from MGMT (n = 68), EGFR (n = 7), CDKN2A/B (n = 8), alterations of the PI3K–AKT–mTOR pathway (n = 5), and BRAF (n = 3) have been the most frequently used for decision making with a positive overall survival signal for patients with glioblastoma harboring an unmethylated MGMT promoter treated according to the molecular assignment. Based on detected molecular alterations and possible targeted therapies, we generated an automated web-based prioritization algorithm. Conclusion Molecular decision making in clinical practice was mainly driven by MGMT promoter status in elderly patients and study inclusion criteria. A reasonable number of patients have been treated based on other molecular aberrations. This study prepares for complex molecular decisions in a routine clinical decision making.
Collapse
Affiliation(s)
- Tobias Kessler
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology and Neuro-oncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Anne Berberich
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Belen Casalini
- Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katharina Drüschler
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology and Neuro-oncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Hannah Ostermann
- Department of Neurology and Neuro-oncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Andrea Dormann
- Department of Neurology and Neuro-oncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Sandy Walter
- Department of Neurology and Neuro-oncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Ling Hai
- Junior Research Group Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias Schlesner
- Junior Research Group Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Christine Jungk
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas Unterberg
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Katharina Sahm
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andreas von Deimling
- Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Winkler
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology and Neuro-oncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Platten
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Wolfgang Wick
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neurology and Neuro-oncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Felix Sahm
- Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Antje Wick
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
27
|
Optic Pathway Glioma in Type 1 Neurofibromatosis: Review of Its Pathogenesis, Diagnostic Assessment, and Treatment Recommendations. Cancers (Basel) 2019; 11:cancers11111790. [PMID: 31739524 PMCID: PMC6896195 DOI: 10.3390/cancers11111790] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/12/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022] Open
Abstract
Type 1 neurofibromatosis (NF1) is a dominantly inherited condition predisposing to tumor development. Optic pathway glioma (OPG) is the most frequent central nervous system tumor in children with NF1, affecting approximately 15-20% of patients. The lack of well-established prognostic markers and the wide clinical variability with respect to tumor progression and visual outcome make the clinical management of these tumors challenging, with significant differences among distinct centers. We reviewed published articles on OPG diagnostic protocol, follow-up and treatment in NF1. Cohorts of NF1 children with OPG reported in the literature and patients prospectively collected in our center were analyzed with regard to clinical data, tumor anatomical site, diagnostic workflow, treatment and outcome. In addition, we discussed the recent findings on the pathophysiology of OPG development in NF1. This review provides a comprehensive overview about the clinical management of NF1-associated OPG, focusing on the most recent advances from preclinical studies with genetically engineered models and the ongoing clinical trials.
Collapse
|
28
|
Eckerdt F, Bell JB, Beauchamp EM, Clymer J, Blyth GT, Kosciuczuk EM, Ma Q, Chen DZ, Horbinski C, Goldman S, Munshi HG, Hashizume R, Platanias LC. Potent Antineoplastic Effects of Combined PI3Kα-MNK Inhibition in Medulloblastoma. Mol Cancer Res 2019; 17:1305-1315. [PMID: 30842251 PMCID: PMC6548590 DOI: 10.1158/1541-7786.mcr-18-1193] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 01/15/2019] [Accepted: 03/01/2019] [Indexed: 12/28/2022]
Abstract
Medulloblastoma is a highly malignant pediatric brain tumor associated with poor outcome. Developing treatments that target the cancer stem cell (CSC) population in medulloblastoma are important to prevent tumor relapse and induce long-lasting clinical responses. We utilized medulloblastoma neurospheres that display CSC characteristics and found activation of the PI3K/AKT pathway in sphere-forming cells. Of all class IA PI3Ks, only the PI3Kα isoform was required for sphere formation by medulloblastoma cells. Knockdown of p110α, but not p110β or p110δ, significantly disrupted cancer stem cell frequencies as determined by extreme limiting dilution analysis (ELDA), indicating an essential role for the PI3Kα catalytic isoform in medulloblastoma CSCs. Importantly, pharmacologic inhibition of the MAPK-interacting kinase (MNK) enhanced the antineoplastic effects of targeted PI3Kα inhibition in medulloblastoma. This indicates that MNK signaling promotes survival in medulloblastoma, suggesting dual PI3Kα and MNK inhibition may provide a novel approach to target and eliminate medulloblastoma CSCs. We also observed a significant reduction in tumor formation in subcutaneous and intracranial mouse xenograft models, which further suggests that this combinatorial approach may represent an efficient therapeutic strategy for medulloblastoma. IMPLICATIONS: These findings raise the possibility of a unique therapeutic approach for medulloblastoma, involving MNK targeting to sensitize medulloblastoma CSCs to PI3Kα inhibition.
Collapse
Affiliation(s)
- Frank Eckerdt
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jonathan B Bell
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Elspeth M Beauchamp
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Medicine Service, Jesse Brown VA Medical Center, Chicago, Illinois
| | - Jessica Clymer
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
- Division of Hematology/Oncology/Stem Cell Transplantation, Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Gavin T Blyth
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Ewa M Kosciuczuk
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Medicine Service, Jesse Brown VA Medical Center, Chicago, Illinois
| | - Quanhong Ma
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - David Z Chen
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Craig Horbinski
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Stewart Goldman
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
- Division of Hematology/Oncology/Stem Cell Transplantation, Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Hidayatullah G Munshi
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Medicine Service, Jesse Brown VA Medical Center, Chicago, Illinois
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Rintaro Hashizume
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Leonidas C Platanias
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois.
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Medicine Service, Jesse Brown VA Medical Center, Chicago, Illinois
| |
Collapse
|
29
|
Molecular genetics and therapeutic targets of pediatric low-grade gliomas. Brain Tumor Pathol 2019; 36:74-83. [PMID: 30929113 DOI: 10.1007/s10014-019-00340-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/19/2019] [Indexed: 12/27/2022]
Abstract
Pediatric low-grade gliomas (PLGGs) have relatively favorable prognosis and some resectable PLGGs, such as cerebellar pilocytic astrocytoma, can be cured by surgery alone. However, many PLGG cases are unresectable and some of them undergo tumor progression. Therefore, a multidisciplinary approach is necessary to treat PLGG patients. Recent genomic analysis revealed a broad genomic landscape underlying PLGG. Notably, the majority of PLGGs present MAPK pathway-associated genomic alterations and MAPK signaling-dependent tumor progression. Following preclinical evidence, many clinical trials based on molecular target therapy have been conducted on PLGG patients, some of whom exhibited durable response to target therapy. Here, we provide an overview of PLGG genetics and the evidence supporting the application of molecular target therapy in these patients.
Collapse
|
30
|
Khani P, Nasri F, Khani Chamani F, Saeidi F, Sadri Nahand J, Tabibkhooei A, Mirzaei H. Genetic and epigenetic contribution to astrocytic gliomas pathogenesis. J Neurochem 2018; 148:188-203. [PMID: 30347482 DOI: 10.1111/jnc.14616] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 09/16/2018] [Accepted: 10/17/2018] [Indexed: 12/30/2022]
Abstract
Astrocytic gliomas are the most common and lethal form of intracranial tumors. These tumors are characterized by a significant heterogeneity in terms of cytopathological, transcriptional, and (epi)genomic features. This heterogeneity has made these cancers one of the most challenging types of cancers to study and treat. To uncover these complexities and to have better understanding of the disease initiation and progression, identification, and characterization of underlying cellular and molecular pathways related to (epi)genetics of astrocytic gliomas is crucial. Here, we discuss and summarize molecular and (epi)genetic mechanisms that provide clues as to the pathogenesis of astrocytic gliomas.
Collapse
Affiliation(s)
- Pouria Khani
- Department of Medical Genetics and Molecular Biology, Faculty of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Nasri
- Department of Medical Immunology, Faculty of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Fateme Khani Chamani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzane Saeidi
- Department of Medical Genetics, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Javid Sadri Nahand
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Tabibkhooei
- Department of Neurosurgery, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
31
|
Leung EY, Askarian-Amiri ME, Singleton DC, Ferraro-Peyret C, Joseph WR, Finlay GJ, Broom RJ, Kakadia PM, Bohlander SK, Marshall E, Baguley BC. Derivation of Breast Cancer Cell Lines Under Physiological (5%) Oxygen Concentrations. Front Oncol 2018; 8:425. [PMID: 30370249 PMCID: PMC6194255 DOI: 10.3389/fonc.2018.00425] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 09/11/2018] [Indexed: 11/13/2022] Open
Abstract
Background: Most human breast cancer cell lines currently in use were developed and are cultured under ambient (21%) oxygen conditions. While this is convenient in practical terms, higher ambient oxygen could increase oxygen radical production, potentially modulating signaling pathways. We have derived and grown a series of four human breast cancer cell lines under 5% oxygen, and have compared their properties to those of established breast cancer lines growing under ambient oxygen. Methods: Cell lines were characterized in terms of appearance, cellular DNA content, mutation spectrum, hormone receptor status, pathway utilization and drug sensitivity. Results: Three of the four lines (NZBR1, NZBR2, and NZBR4) were triple negative (ER-, PR-, HER2-), with NZBR1 also over-expressing EGFR. NZBR3 was HER2+ and ER+ and also over-expressed EGFR. Cell lines grown in 5% oxygen showed increased expression of the hypoxia-inducible factor 1 (HIF-1) target gene carbonic anhydrase 9 (CA9) and decreased levels of ROS. As determined by protein phosphorylation, NZBR1 showed low AKT pathway utilization while NZBR2 and NZBR4 showed low p70S6K and rpS6 pathway utilization. The lines were characterized for sensitivity to 7-hydroxytamoxifen, doxorubicin, paclitaxel, the PI3K inhibitor BEZ235 and the HER inhibitors lapatinib, afatinib, dacomitinib, and ARRY-380. In some cases they were compared to established breast cancer lines. Of particular note was the high sensitivity of NZBR3 to HER inhibitors. The spectrum of mutations in the NZBR lines was generally similar to that found in commonly used breast cancer cell lines but TP53 mutations were absent and mutations in EVI2B, LRP1B, and PMS2, which have not been reported in other breast cancer lines, were detected. The results suggest that the properties of cell lines developed under low oxygen conditions (5% O2) are similar to those of commonly used breast cancer cell lines. Although reduced ROS production and increased HIF-1 activity under 5% oxygen can potentially influence experimental outcomes, no difference in sensitivity to estrogen or doxorubicin was observed between cell lines cultured in 5 vs. 21% oxygen.
Collapse
Affiliation(s)
- Euphemia Y Leung
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand.,Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Marjan E Askarian-Amiri
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand.,Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Dean C Singleton
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Carole Ferraro-Peyret
- Univ Lyon, Claude Bernard University, Cancer Research Center of Lyon, INSERM 1052, CNRS5286, Faculty of Pharmacy, Lyon, France.,Hospices Civils de Lyon, Molecular Biology of Tumors, GHE Hospital, Bron, France
| | - Wayne R Joseph
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Graeme J Finlay
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand.,Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Reuben J Broom
- Auckland City Hospital-Oncology, Grafton, Auckland, New Zealand
| | - Purvi M Kakadia
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Stefan K Bohlander
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Elaine Marshall
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Bruce C Baguley
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| |
Collapse
|
32
|
Wilson KM, Mathews-Griner LA, Williamson T, Guha R, Chen L, Shinn P, McKnight C, Michael S, Klumpp-Thomas C, Binder ZA, Ferrer M, Gallia GL, Thomas CJ, Riggins GJ. Mutation Profiles in Glioblastoma 3D Oncospheres Modulate Drug Efficacy. SLAS Technol 2018; 24:28-40. [PMID: 30289729 DOI: 10.1177/2472630318803749] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Glioblastoma (GBM) is a lethal brain cancer with a median survival time of approximately 15 months following treatment. Common in vitro GBM models for drug screening are adherent and do not recapitulate the features of human GBM in vivo. Here we report the genomic characterization of nine patient-derived, spheroid GBM cell lines that recapitulate human GBM characteristics in orthotopic xenograft models. Genomic sequencing revealed that the spheroid lines contain alterations in GBM driver genes such as PTEN, CDKN2A, and NF1. Two spheroid cell lines, JHH-136 and JHH-520, were utilized in a high-throughput drug screen for cell viability using a 1912-member compound library. Drug mechanisms that were cytotoxic in both cell lines were Hsp90 and proteasome inhibitors. JHH-136 was uniquely sensitive to topoisomerase 1 inhibitors, while JHH-520 was uniquely sensitive to Mek inhibitors. Drug combination screening revealed that PI3 kinase inhibitors combined with Mek or proteasome inhibitors were synergistic. However, animal studies to test these drug combinations in vivo revealed that Mek inhibition alone was superior to the combination treatments. These data show that these GBM spheroid lines are amenable to high-throughput drug screening and that this dataset may deliver promising therapeutic leads for future GBM preclinical studies.
Collapse
Affiliation(s)
- Kelli M Wilson
- 1 Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore MD, USA
| | - Lesley A Mathews-Griner
- 2 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, USA
| | - Tara Williamson
- 1 Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore MD, USA
| | - Rajarshi Guha
- 2 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, USA
| | - Lu Chen
- 2 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, USA
| | - Paul Shinn
- 2 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, USA
| | - Crystal McKnight
- 2 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, USA
| | - Sam Michael
- 2 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, USA
| | - Carleen Klumpp-Thomas
- 2 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, USA
| | - Zev A Binder
- 3 Department of Neurosurgery, Perelmen School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marc Ferrer
- 2 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, USA
| | - Gary L Gallia
- 1 Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore MD, USA
| | - Craig J Thomas
- 2 Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, USA
| | - Gregory J Riggins
- 1 Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore MD, USA
| |
Collapse
|
33
|
Unruh D, Mirkov S, Wray B, Drumm M, Lamano J, Li YD, Haider QF, Javier R, McCortney K, Saratsis A, Scholtens DM, Sarkaria JN, James CD, Horbinski C. Methylation-dependent Tissue Factor Suppression Contributes to the Reduced Malignancy of IDH1-mutant Gliomas. Clin Cancer Res 2018; 25:747-759. [PMID: 30266764 DOI: 10.1158/1078-0432.ccr-18-1222] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/15/2018] [Accepted: 09/24/2018] [Indexed: 01/08/2023]
Abstract
PURPOSE Gliomas with isocitrate dehydrogenase 1 mutations (IDH1mut) are less aggressive than IDH1 wild-type (IDH1wt) gliomas and have global genomic hypermethylation. Yet it is unclear how specific hypermethylation events contribute to the IDH1mut phenotype. Previously, we showed that the gene encoding the procoagulant tissue factor (TF), F3, is among the most hypermethylated and downregulated genes in IDH1mut gliomas, correlating with greatly reduced thrombosis in patients with IDH1mut glioma. Because TF also increases the aggressiveness of many cancers, the current study explored the contribution of TF suppression to the reduced malignancy of IDH1mut gliomas.Experimental Design: TF expression was manipulated in patient-derived IDH1mut and IDH1wt glioma cells, followed by evaluation of in vitro and in vivo behavior and analyses of cell signaling pathways. RESULTS A demethylating agent, decitabine, increased F3 transcription and TF-dependent coagulative activity in IDH1mut cells, but not in IDH1wt cells. TF induction enhanced the proliferation, invasion, and colony formation of IDH1mut cells, and increased the intracranial engraftment of IDH1mut GBM164 from 0% to 100% (P = 0.0001). Conversely, TF knockdown doubled the median survival of mice engrafted with IDH1wt/EGFRvIIIamp GBM6, and caused complete regression of IDH1wt/EGFRamp GBM12 (P = 0.001). In vitro and in vivo effects were linked to activation of receptor tyrosine kinases (RTK) by TF through a Src-dependent intracellular pathway, even when extracellular RTK stimulation was blocked. TF stimulated invasion predominately through upregulation of β-catenin. CONCLUSIONS These data show that TF suppression is a component of IDH1mut glioma behavior, and that it may therefore be an attractive target against IDH1wt gliomas.
Collapse
Affiliation(s)
- Dusten Unruh
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Snezana Mirkov
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Brian Wray
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
| | - Michael Drumm
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Jonathan Lamano
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Yuping D Li
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Qazi F Haider
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Rodrigo Javier
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Kathleen McCortney
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Amanda Saratsis
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Denise M Scholtens
- Department of Preventive Medicine, Northwestern University, Chicago, Illinois
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - C David James
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois
| | - Craig Horbinski
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois. .,Department of Pathology, Northwestern University, Chicago, Illinois
| |
Collapse
|
34
|
Young JS, Prados MD, Butowski N. Using genomics to guide treatment for glioblastoma. Pharmacogenomics 2018; 19:1217-1229. [PMID: 30203716 DOI: 10.2217/pgs-2018-0078] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma has been shown to have many different genetic mutations found both within and between tumor samples. Molecular testing and genomic sequencing has helped to classify diagnoses and clarify difficult to interpret histopathological specimens. Genomic information also plays a critical role in prognostication for patients, with IDH mutations and MGMT methylation having significant impact of the response to chemotherapy and overall survival of patients. Unfortunately, personalized medicine and targeted therapy against specific mutations have not been shown to improve patient outcomes. As technology continues to improve, exome and RNA sequencing will play a role in the design of clinical trials, classification of patient subgroups and identification of rare mutations that can be targeted by small-molecule inhibitors and biologic agents.
Collapse
Affiliation(s)
- Jacob S Young
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | - Michael D Prados
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | - Nicholas Butowski
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
35
|
McNeill RS, Stroobant EE, Smithberger E, Canoutas DA, Butler MK, Shelton AK, Patel SD, Limas JC, Skinner KR, Bash RE, Schmid RS, Miller CR. PIK3CA missense mutations promote glioblastoma pathogenesis, but do not enhance targeted PI3K inhibition. PLoS One 2018; 13:e0200014. [PMID: 29975751 PMCID: PMC6033446 DOI: 10.1371/journal.pone.0200014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/18/2018] [Indexed: 12/12/2022] Open
Abstract
Background Glioblastoma (GBM) is the most common adult primary brain tumor. Multimodal treatment is empiric and prognosis remains poor. Recurrent PIK3CA missense mutations (PIK3CAmut) in GBM are restricted to three functional domains: adaptor binding (ABD), helical, and kinase. Defining how these mutations influence gliomagenesis and response to kinase inhibitors may aid in the clinical development of novel targeted therapies in biomarker-stratified patients. Methods We used normal human astrocytes immortalized via expression of hTERT, E6, and E7 (NHA). We selected two PIK3CAmut from each of 3 mutated domains and induced their expression in NHA with (NHARAS) and without mutant RAS using lentiviral vectors. We then examined the role of PIK3CAmut in gliomagenesis in vitro and in mice, as well as response to targeted PI3K (PI3Ki) and MEK (MEKi) inhibitors in vitro. Results PIK3CAmut, particularly helical and kinase domain mutations, potentiated proximal PI3K signaling and migration of NHA and NHARASin vitro. Only kinase domain mutations promoted NHA colony formation, but both helical and kinase domain mutations promoted NHARAS tumorigenesis in vivo. PIK3CAmut status had minimal effects on PI3Ki and MEKi efficacy. However, PI3Ki/MEKi synergism was pronounced in NHA and NHARAS harboring ABD or helical mutations. Conclusion PIK3CAmut promoted differential gliomagenesis based on the mutated domain. While PIK3CAmut did not influence sensitivity to single agent PI3Ki, they did alter PI3Ki/MEKi synergism. Taken together, our results demonstrate that a subset of PIK3CAmut promote tumorigenesis and suggest that patients with helical domain mutations may be most sensitive to dual PI3Ki/MEKi treatment.
Collapse
Affiliation(s)
- Robert S McNeill
- Pathobiology and Translational Science Graduate Program, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Emily E Stroobant
- Department of Chemistry, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Erin Smithberger
- Pathobiology and Translational Science Graduate Program, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Demitra A Canoutas
- Department of Biology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Madison K Butler
- Department of Biology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Abigail K Shelton
- Pathobiology and Translational Science Graduate Program, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Shrey D Patel
- Department of Chemistry, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Juanita C Limas
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Kasey R Skinner
- Neurosciences Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Ryan E Bash
- Departments of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Ralf S Schmid
- Neurosciences Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America.,Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - C Ryan Miller
- Pathobiology and Translational Science Graduate Program, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America.,Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America.,Neurosciences Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America.,Departments of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America.,Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America.,Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| |
Collapse
|
36
|
Baro M, Lopez Sambrooks C, Quijano A, Saltzman WM, Contessa J. Oligosaccharyltransferase Inhibition Reduces Receptor Tyrosine Kinase Activation and Enhances Glioma Radiosensitivity. Clin Cancer Res 2018; 25:784-795. [PMID: 29967251 DOI: 10.1158/1078-0432.ccr-18-0792] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/21/2018] [Accepted: 06/27/2018] [Indexed: 12/31/2022]
Abstract
PURPOSE Parallel signaling reduces the effects of receptor tyrosine kinase (RTK)-targeted therapies in glioma. We hypothesized that inhibition of protein N-linked glycosylation, an endoplasmic reticulum co- and posttranslational modification crucial for RTK maturation and activation, could provide a new therapeutic approach for glioma radiosensitization.Experimental Design: We investigated the effects of a small-molecule inhibitor of the oligosaccharyltransferase (NGI-1) on EGFR family receptors, MET, PDGFR, and FGFR1. The influence of glycosylation state on tumor cell radiosensitivity, chemotherapy-induced cell toxicity, DNA damage, and cell-cycle arrest were determined and correlated with glioma cell receptor expression profiles. The effects of NGI-1 on xenograft tumor growth were tested using a nanoparticle formulation validated by in vivo molecular imaging. A mechanistic role for RTK signaling was evaluated through the expression of a glycosylation-independent CD8-EGFR chimera. RESULTS NGI-1 reduced glycosylation, protein levels, and activation of most RTKs. NGI-1 also enhanced the radiosensitivity and cytotoxic effects of chemotherapy in those glioma cells with elevated ErbB family activation, but not in cells without high levels of RTK activation. NGI-1 radiosensitization was associated with increases in both DNA damage and G1 cell-cycle arrest. Combined treatment of glioma xenografts with fractionated radiotherapy and NGI-1 significantly reduced tumor growth compared with controls. Expression of the CD8-EGFR eliminated the effects of NGI-1 on G1 arrest, DNA damage, and cellular radiosensitivity, identifying RTK inhibition as the principal mechanism for the NGI-1 effect. CONCLUSIONS This study suggests that oligosaccharyltransferase inhibition with NGI-1 is a novel approach to radiosensitize malignant gliomas with enhanced RTK signaling.See related commentary by Wahl and Lawrence, p. 455.
Collapse
Affiliation(s)
- Marta Baro
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut
| | | | - Amanda Quijano
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Joseph Contessa
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut. .,Department of Pharmacology, Yale University, New Haven, Connecticut
| |
Collapse
|
37
|
McNeill RS, Canoutas DA, Stuhlmiller TJ, Dhruv HD, Irvin DM, Bash RE, Angus SP, Herring LE, Simon JM, Skinner KR, Limas JC, Chen X, Schmid RS, Siegel MB, Van Swearingen AED, Hadler MJ, Sulman EP, Sarkaria JN, Anders CK, Graves LM, Berens ME, Johnson GL, Miller CR. Combination therapy with potent PI3K and MAPK inhibitors overcomes adaptive kinome resistance to single agents in preclinical models of glioblastoma. Neuro Oncol 2018; 19:1469-1480. [PMID: 28379424 DOI: 10.1093/neuonc/nox044] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Glioblastoma (GBM) is the most common and aggressive primary brain tumor. Prognosis remains poor despite multimodal therapy. Developing alternative treatments is essential. Drugs targeting kinases within the phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) effectors of receptor tyrosine kinase (RTK) signaling represent promising candidates. Methods We previously developed a non-germline genetically engineered mouse model of GBM in which PI3K and MAPK are activated via Pten deletion and KrasG12D in immortalized astrocytes. Using this model, we examined the influence of drug potency on target inhibition, alternate pathway activation, efficacy, and synergism of single agent and combination therapy with inhibitors of these 2 pathways. Efficacy was then examined in GBM patient-derived xenografts (PDX) in vitro and in vivo. Results PI3K and mitogen-activated protein kinase kinase (MEK) inhibitor potency was directly associated with target inhibition, alternate RTK effector activation, and efficacy in mutant murine astrocytes in vitro. The kinomes of GBM PDX and tumor samples were heterogeneous, with a subset of the latter harboring MAPK hyperactivation. Dual PI3K/MEK inhibitor treatment overcame alternate effector activation, was synergistic in vitro, and was more effective than single agent therapy in subcutaneous murine allografts. However, efficacy in orthotopic allografts was minimal. This was likely due to dose-limiting toxicity and incomplete target inhibition. Conclusion Drug potency influences PI3K/MEK inhibitor-induced target inhibition, adaptive kinome reprogramming, efficacy, and synergy. Our findings suggest that combination therapies with highly potent, brain-penetrant kinase inhibitors will be required to improve patient outcomes.
Collapse
Affiliation(s)
- Robert S McNeill
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Demitra A Canoutas
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Timothy J Stuhlmiller
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Harshil D Dhruv
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - David M Irvin
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Ryan E Bash
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Steven P Angus
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Laura E Herring
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Jeremy M Simon
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Kasey R Skinner
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Juanita C Limas
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Xin Chen
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Ralf S Schmid
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Marni B Siegel
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Amanda E D Van Swearingen
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Michael J Hadler
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Erik P Sulman
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Jann N Sarkaria
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Carey K Anders
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Lee M Graves
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Michael E Berens
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Gary L Johnson
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - C Ryan Miller
- Pathobiology and Translational Science Graduate Program, Departments of Pathology and Laboratory Medicine, Biology, Pharmacology, Genetics, Medicine, and Neurology, Divisions of Neuropathology and Hematology/Oncology, Curriculum in Genetics and Molecular Biology, Lineberger Comprehensive Cancer Center, Proteomics Core Facility, Neurosciences Center, Carolina Institute for Developmental Disabilities, and Biological and Biomedical Sciences Program, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina; Cancer & Cell Biology Division, TGen, Phoenix, Arizona; Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota; Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| |
Collapse
|
38
|
Versano Z, Shany E, Freedman S, Tuval-Kochen L, Leitner M, Paglin S, Toren A, Yalon M. MutT homolog 1 counteracts the effect of anti-neoplastic treatments in adult and pediatric glioblastoma cells. Oncotarget 2018; 9:27547-27563. [PMID: 29938005 PMCID: PMC6007941 DOI: 10.18632/oncotarget.25547] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 05/19/2018] [Indexed: 11/29/2022] Open
Abstract
Glioblastoma, a fatal disease in both adult and pediatric patients, currently has limited treatment options that offer no more than temporary relief. Our experiments with adult and pediatric glioblastoma cell lines showed that radiation induces a dose-dependent increase in the level of MutT homolog 1 (MTH1) - an enzyme that hydrolyzes oxidized purine nucleoside triphosphates. Similarly, the combination of vorinostat, which is a histone deacetylase inhibitor, and ABT-888, which is a PARP-1 inhibitor, enhanced clonogenic death and increased the MTH1 level, relative to each treatment alone. This result suggests that the MTH1 level is directly related to the damage that is inflicted upon the cells, and its activity protects them against anti-neoplastic therapy. Indeed, the MTH1 inhibitor TH588 and MTH1 siRNA increased glioblastoma's response to both radiation and the combination of vorinostat and ABT-888. TH588 also inhibited glioblastoma's capacity for migration and invasion. In normal fibroblasts, low radiation doses and the combination of vorinostat and ABT-888 decreased the level of the enzyme. TH588 did not alter the fibroblasts’ response to radiation and only mildly affected their response to the combination of vorinostat and ABT-888. In summary, the inhibition of MTH1 is required to better realize the therapeutic potential of anti-neoplastic treatments in glioblastoma.
Collapse
Affiliation(s)
- Ziv Versano
- Pediatric Hemato-Oncology, Edmond and Lilly Safra Children's Hospital and Cancer Research Center, Sheba Medical Center, Ramat Gan 52621, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Eitan Shany
- Pediatric Hemato-Oncology, Edmond and Lilly Safra Children's Hospital and Cancer Research Center, Sheba Medical Center, Ramat Gan 52621, Israel
| | - Shany Freedman
- Pediatric Hemato-Oncology, Edmond and Lilly Safra Children's Hospital and Cancer Research Center, Sheba Medical Center, Ramat Gan 52621, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Liron Tuval-Kochen
- Pediatric Hemato-Oncology, Edmond and Lilly Safra Children's Hospital and Cancer Research Center, Sheba Medical Center, Ramat Gan 52621, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Moshe Leitner
- Pediatric Hemato-Oncology, Edmond and Lilly Safra Children's Hospital and Cancer Research Center, Sheba Medical Center, Ramat Gan 52621, Israel
| | - Shoshana Paglin
- Pediatric Hemato-Oncology, Edmond and Lilly Safra Children's Hospital and Cancer Research Center, Sheba Medical Center, Ramat Gan 52621, Israel
| | - Amos Toren
- Pediatric Hemato-Oncology, Edmond and Lilly Safra Children's Hospital and Cancer Research Center, Sheba Medical Center, Ramat Gan 52621, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Michal Yalon
- Pediatric Hemato-Oncology, Edmond and Lilly Safra Children's Hospital and Cancer Research Center, Sheba Medical Center, Ramat Gan 52621, Israel.,The Talpiot Medical Leadership Program, Chaim Sheba Medical Center, Ramat Gan 52621, Israel
| |
Collapse
|
39
|
Mukherjee J, Johannessen TC, Ohba S, Chow TT, Jones L, Pandita A, Pieper RO. Mutant IDH1 Cooperates with ATRX Loss to Drive the Alternative Lengthening of Telomere Phenotype in Glioma. Cancer Res 2018; 78:2966-2977. [PMID: 29545335 PMCID: PMC10578296 DOI: 10.1158/0008-5472.can-17-2269] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 12/04/2017] [Accepted: 03/12/2018] [Indexed: 11/16/2022]
Abstract
A subset of tumors use a recombination-based alternative lengthening of telomere (ALT) pathway to resolve telomeric dysfunction in the absence of TERT. Loss-of-function mutations in the chromatin remodeling factor ATRX are associated with ALT but are insufficient to drive the process. Because many ALT tumors express the mutant isocitrate dehydrogenase IDH1 R132H, including all lower grade astrocytomas and secondary glioblastoma, we examined a hypothesized role for IDH1 R132H in driving the ALT phenotype during gliomagenesis. In p53/pRb-deficient human astrocytes, combined deletion of ATRX and expression of mutant IDH1 were sufficient to create tumorigenic cells with ALT characteristics. The telomere capping complex component RAP1 and the nonhomologous DNA end joining repair factor XRCC1 were each downregulated consistently in these tumorigenic cells, where their coordinate reexpression was sufficient to suppress the ALT phenotype. RAP1 or XRCC1 downregulation cooperated with ATRX loss in driving the ALT phenotype. RAP1 silencing caused telomere dysfunction in ATRX-deficient cells, whereas XRCC1 silencing suppressed lethal fusion of dysfunctional telomeres by allowing IDH1-mutant ATRX-deficient cells to use homologous recombination and ALT to resolve telomeric dysfunction and escape cell death. Overall, our studies show how expression of mutant IDH1 initiates telomeric dysfunction and alters DNA repair pathway preferences at telomeres, cooperating with ATRX loss to defeat a key barrier to gliomagenesis.Significance: Studies show how expression of mutant IDH1 initiates telomeric dysfunction and alters DNA repair pathway preferences at telomeres, cooperating with ATRX loss to defeat a key barrier to gliomagenesis and suggesting new therapeutic options to treat low-grade gliomas. Cancer Res; 78(11); 2966-77. ©2018 AACR.
Collapse
Affiliation(s)
- Joydeep Mukherjee
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Tor-Christian Johannessen
- The Kristian Gerhard Jebsen Brain Tumor Research Centre, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Shigeo Ohba
- Department of Neurosurgery, Fujita Health University, Toyoake, Aichi, Japan
| | - Tracy T Chow
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, California
| | - Lindsey Jones
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | | | - Russell O Pieper
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California.
| |
Collapse
|
40
|
Combinatorial Drug Testing in 3D Microtumors Derived from GBM Patient-Derived Xenografts Reveals Cytotoxic Synergy in Pharmacokinomics-informed Pathway Interactions. Sci Rep 2018; 8:8412. [PMID: 29849102 PMCID: PMC5976646 DOI: 10.1038/s41598-018-26840-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 05/15/2018] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma multiforme (GBM), the most common form of primary malignant brain cancer in adults, is a devastating disease for which effective treatment has remained elusive for over 75 years. One reason for the minimal progress during this time is the lack of accurate preclinical models to represent the patient's tumor's in vivo environment, causing a disconnect in drug therapy effectiveness between the laboratory and clinic. While patient-derived xenografts (PDX's or xenolines) are excellent human tumor representations, they are not amenable to high throughput testing. Therefore, we developed a miniaturized xenoline system (microtumors) for drug testing. Nineteen GBM xenolines were profiled for global kinase (kinomic) activity revealing actionable kinase targets associated with intracranial tumor growth rate. Kinase inhibitors for these targets (WP1066, selumetinib, crizotinib, and cediranib) were selected for single and combination therapy using a fully human-derived three-dimensional (3D) microtumor model of GBM xenoline cells embedded in HuBiogel for subsequent molecular and phenotype assays. GBM microtumors closely resembled orthotopically-implanted tumors based on immunohistochemical analysis and displayed kinomic and morphological diversity. Drug response testing could be reproducibly performed in a 96-well format identifying several synergistic combinations. Our findings indicate that 3D microtumors can provide a suitable high-throughput model for combination drug testing.
Collapse
|
41
|
Haas B, Klinger V, Keksel C, Bonigut V, Kiefer D, Caspers J, Walther J, Wos-Maganga M, Weickhardt S, Röhn G, Timmer M, Frötschl R, Eckstein N. Inhibition of the PI3K but not the MEK/ERK pathway sensitizes human glioma cells to alkylating drugs. Cancer Cell Int 2018; 18:69. [PMID: 29755294 PMCID: PMC5935937 DOI: 10.1186/s12935-018-0565-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 03/14/2018] [Indexed: 12/19/2022] Open
Abstract
Background Intrinsic chemoresistance of glioblastoma (GBM) is frequently owed to activation of the PI3K and MEK/ERK pathways. These signaling cascades are tightly interconnected however the quantitative contribution of both to intrinsic resistance is still not clear. Here, we aimed at determining the activation status of these pathways in human GBM biopsies and cells and investigating the quantitative impact of both pathways to chemoresistance. Methods Receptor tyrosine kinase (RTK) pathways in temozolomide (TMZ) treatment naive or TMZ resistant human GBM biopsies and GBM cells were investigated by proteome profiling and immunoblotting of a subset of proteins. Resistance to drugs and RTK pathway inhibitors was assessed by MTT assays. Apoptotic rates were determined by Annexin V staining and DNA damage with comet assays and immunoblotting. Results We analyzed activation of RTK pathways by proteome profiling of tumor samples of patients which were diagnosed a secondary GBM and underwent surgery and patients which underwent a second surgery after TMZ treatment due to recurrence of the tumor. We observed substantial activation of the PI3K and MEK/ERK pathways in both groups. However, AKT and CREB phosphorylation was reduced in biopsies of resistant tumors while ERK phosphorylation remained unchanged. Subsequent proteome profiling revealed that multiple RTKs and downstream targets are also activated in three GBM cell lines. We then systematically describe a mechanism of resistance of GBM cell lines and human primary GBM cells to the alkylating drugs TMZ and cisplatin. No specific inhibitor of the upstream RTKs sensitized cells to drug treatment. In contrast, we were able to restore sensitivity to TMZ and cisplatin by inhibiting PI3K in all cell lines and in human primary GBM cells. Interestingly, an opposite effect was observed when we inhibited the MEK/ERK signaling cascade with two different inhibitors. Conclusions Temozolomide treatment naive and TMZ resistant GBM biopsies show a distinct activation pattern of the MEK/ERK and PI3K signaling cascades indicating a role of these pathways in resistance development. Both pathways are also activated in GBM cell lines, however, only the PI3K pathway seems to play a crucial role in resistance to alkylating agents and might serve as drug target for chemosensitization.
Collapse
Affiliation(s)
- Bodo Haas
- 1Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Veronika Klinger
- 1Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany.,2Institute of Pharmacy, University of Bonn, 53121 Bonn, Germany
| | - Christina Keksel
- 1Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany.,3Applied Pharmacy, University of Applied Sciences Kaiserslautern, Campus Pirmasens, Carl-Schurz-Str. 10-16, 66953 Pirmasens, Germany
| | - Verena Bonigut
- 1Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany.,3Applied Pharmacy, University of Applied Sciences Kaiserslautern, Campus Pirmasens, Carl-Schurz-Str. 10-16, 66953 Pirmasens, Germany
| | - Daniela Kiefer
- 1Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany.,3Applied Pharmacy, University of Applied Sciences Kaiserslautern, Campus Pirmasens, Carl-Schurz-Str. 10-16, 66953 Pirmasens, Germany
| | - Julia Caspers
- 1Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany.,4Faculty of Applied Natural Sciences, Cologne University of Applied Sciences, Kaiser-Wilhelm-Allee, 51368 Leverkusen, Germany
| | - Julia Walther
- 1Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany.,2Institute of Pharmacy, University of Bonn, 53121 Bonn, Germany
| | - Maria Wos-Maganga
- 1Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Sandra Weickhardt
- 1Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Gabriele Röhn
- 5Department of General Neurosurgery, Center for Neurosurgery, University Hospital Cologne, 50937 Cologne, Germany
| | - Marco Timmer
- 5Department of General Neurosurgery, Center for Neurosurgery, University Hospital Cologne, 50937 Cologne, Germany
| | - Roland Frötschl
- 1Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Niels Eckstein
- 3Applied Pharmacy, University of Applied Sciences Kaiserslautern, Campus Pirmasens, Carl-Schurz-Str. 10-16, 66953 Pirmasens, Germany
| |
Collapse
|
42
|
Freret ME, Gutmann DH. Insights into optic pathway glioma vision loss from mouse models of neurofibromatosis type 1. J Neurosci Res 2018; 97:45-56. [PMID: 29704429 DOI: 10.1002/jnr.24250] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 04/09/2018] [Indexed: 12/12/2022]
Abstract
Neurofibromatosis type 1 (NF1) is a common cancer predisposition syndrome caused by mutations in the NF1 gene. The NF1-encoded protein (neurofibromin) is an inhibitor of the oncoprotein RAS and controls cell growth and survival. Individuals with NF1 are prone to developing low-grade tumors of the optic nerves, chiasm, tracts, and radiations, termed optic pathway gliomas (OPGs), which can cause vision loss. A paucity of surgical tumor specimens and of patient-derived xenografts for investigative studies has limited our understanding of human NF1-associated OPG (NF1-OPG). However, mice genetically engineered to harbor Nf1 gene mutations develop optic gliomas that share many features of their human counterparts. These genetically engineered mouse (GEM) strains have provided important insights into the cellular and molecular determinants that underlie mouse Nf1 optic glioma development, maintenance, and associated vision loss, with relevance by extension to human NF1-OPG disease. Herein, we review our current understanding of NF1-OPG pathobiology and describe the mechanisms responsible for tumor initiation, growth, and associated vision loss in Nf1 GEM models. We also discuss how Nf1 GEM and other preclinical models can be deployed to identify and evaluate molecularly targeted therapies for OPG, particularly as they pertain to future strategies aimed at preventing or improving tumor-associated vision loss in children with NF1.
Collapse
Affiliation(s)
- Morgan E Freret
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
43
|
Wood MD, Mukherjee J, Pieper RO. Neurofibromin knockdown in glioma cell lines is associated with changes in cytokine and chemokine secretion in vitro. Sci Rep 2018; 8:5805. [PMID: 29643433 PMCID: PMC5895785 DOI: 10.1038/s41598-018-24046-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 03/22/2018] [Indexed: 01/21/2023] Open
Abstract
The neurofibromin-1 tumor suppressor gene (NF1) is altered in approximately 20% of sporadic glioblastoma (GBM) cases. NF1 deficient GBM frequently shows a mesenchymal gene expression signature, suggesting a relationship between NF1 status and the tumor microenvironment. To identify changes in the production of secreted cytokines/chemokines in NF1 deficient glioma, we applied cytokine arrays to conditioned media from a panel of three GBM cell lines after siRNA-mediated NF1 knockdown. We identified increased secretion of platelet-derived growth factor AA (PDGF-AA), chitinase-3-like protein 1 (CHI3L1), interleukin-8 (IL-8), and endoglin (ENG) in different subsets of these cell lines. Secretion was associated with induction of the corresponding messenger RNA, suggesting a mechanism involving transcriptional upregulation. By contrast, in non-transformed immortalized normal human astrocytes, PDGF-AA secretion was increased upon NF1 knockdown, while secreted CHI3L1, ENG, and IL-8 were reduced or unchanged. Analysis of The Cancer Genome Atlas confirmed a relationship between glioma NF1 status and ENG and CHI3L1 in tumor samples. Overall, this study identifies candidate changes in secreted proteins from NF1 deficient glioma cells that could influence the tumor microenvironment, and suggests a direct link between NF1 loss and increased tumor cell production of CHI3L1 and endoglin, two factors implicated in mesenchymal identity in glioblastoma.
Collapse
Affiliation(s)
- Matthew D Wood
- Department of Pathology, Division of Neuropathology, University of California San Francisco, San Francisco, CA, USA
- Department of Pathology, Oregon Health & Science University, Portland, OR, USA
| | - Joydeep Mukherjee
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Russell O Pieper
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
44
|
Allaway RJ, Wood MD, Downey SL, Bouley SJ, Traphagen NA, Wells JD, Batra J, Melancon SN, Ringelberg C, Seibel W, Ratner N, Sanchez Y. Exploiting mitochondrial and metabolic homeostasis as a vulnerability in NF1 deficient cells. Oncotarget 2018; 9:15860-15875. [PMID: 29662612 PMCID: PMC5882303 DOI: 10.18632/oncotarget.19335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 07/09/2017] [Indexed: 11/26/2022] Open
Abstract
Neurofibromatosis type 1 is a disease caused by mutation of neurofibromin 1 (NF1), loss of which results in hyperactive Ras signaling and a concomitant increase in cell proliferation and survival. Patients with neurofibromatosis type 1 frequently develop tumors such as plexiform neurofibromas and malignant peripheral nerve sheath tumors. Mutation of NF1 or loss of the NF1 protein is also observed in glioblastoma, lung adenocarcinoma, and ovarian cancer among other sporadic cancers. A therapy that selectively targets NF1 deficient tumors would substantially advance our ability to treat these malignancies. To address the need for these therapeutics, we developed and conducted a synthetic lethality screen to discover molecules that target yeast lacking the homolog of NF1, IRA2. One of the lead candidates that was observed to be synthetic lethal with ira2Δ yeast is Y100. Here, we describe the mechanisms by which Y100 targets ira2Δ yeast and NF1-deficient tumor cells. Y100 treatment disrupted proteostasis, metabolic homeostasis, and induced the formation of mitochondrial superoxide in NF1-deficient cancer cells. Previous studies also indicate that NF1/Ras-dysregulated tumors may be sensitive to modulators of oxidative and ER stress. We hypothesize that the use of Y100 and molecules with related mechanisms of action represent a feasible therapeutic strategy for targeting NF1 deficient cells.
Collapse
Affiliation(s)
- Robert J. Allaway
- Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
| | - Matthew D. Wood
- Department of Pharmacology and Toxicology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
- Current address: Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Sondra L. Downey
- Department of Pharmacology and Toxicology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
| | - Stephanie J. Bouley
- Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
| | - Nicole A. Traphagen
- Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
| | - Jason D. Wells
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
| | - Jaya Batra
- Department of Pharmacology and Toxicology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
- Current address: Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sir Norman Melancon
- Department of Pharmacology and Toxicology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
- Current address: Vanderbilt School of Medicine, Nashville, TN 37232, USA
| | - Carol Ringelberg
- Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
- Bioinformatics Shared Resource, Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756, USA
| | - William Seibel
- Division of Oncology, Cincinnati Children’s Hospital Medical Center, Cancer and Blood Diseases Institute, Cincinnati, OH 45229, USA
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cancer and Blood Diseases Institute, Cincinnati, OH 45229, USA
| | - Yolanda Sanchez
- Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
- Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756, USA
| |
Collapse
|
45
|
Khatua S, Gutmann DH, Packer RJ. Neurofibromatosis type 1 and optic pathway glioma: Molecular interplay and therapeutic insights. Pediatr Blood Cancer 2018; 65. [PMID: 29049847 DOI: 10.1002/pbc.26838] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 08/21/2017] [Accepted: 09/02/2017] [Indexed: 12/17/2022]
Abstract
Children with neurofibromatosis type 1 (NF1) are predisposed to develop central nervous system neoplasms, the most common of which are low-grade gliomas (LGGs). The absence of human NF1 associated LGG-derived cell lines, coupled with an inability to generate patient-derived xenograft models, represents barriers to profile molecularly targeted therapies for these tumors. Thus, genetically engineered mouse models have been identified to evaluate the interplay between Nf1-deficient tumor cells and nonneoplastic stromal cells to evaluate potential therapies for these neoplasms. Future treatments might also consider targeting the nonneoplastic cells in NF1-LGGs to reduce tumor growth and neurologic morbidity in affected children.
Collapse
Affiliation(s)
- Soumen Khatua
- Department of Pediatrics, MD Anderson Cancer Center, Houston, Texas
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Roger J Packer
- Center for Neuroscience and Behavioral Medicine, Children's National Medical Center, Washington, District of Columbia
| |
Collapse
|
46
|
Abstract
Neurofibromatosis type 1 (NF1) is one of the most common brain tumor predisposition syndromes, in which affected children are prone to the development of low-grade gliomas. While NF1-associated gliomas can be found in several brain regions, the majority arise in the optic nerves, chiasm, tracts, and radiations (optic pathway gliomas; OPGs). Owing to their location, 35-50% of affected children present with reduced visual acuity. Unfortunately, despite tumor stabilization following chemotherapy, vision does not improve in most children. For this reasons, more effective therapies are being sought that reflect a deeper understanding of the NF1 gene and the use of authenticated Nf1 genetically-engineered mouse strains. The implementation of these models for drug discovery and validation has galvanized molecularly-targeted clinical trials in children with NF1-OPG. Future research focused on defining the cellular and molecular factors that underlie optic glioma development and progression also has the potential to provide personalized risk assessment strategies for this pediatric population.
Collapse
Affiliation(s)
| | - David H. Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis MO
| |
Collapse
|
47
|
Masgras I, Ciscato F, Brunati AM, Tibaldi E, Indraccolo S, Curtarello M, Chiara F, Cannino G, Papaleo E, Lambrughi M, Guzzo G, Gambalunga A, Pizzi M, Guzzardo V, Rugge M, Vuljan SE, Calabrese F, Bernardi P, Rasola A. Absence of Neurofibromin Induces an Oncogenic Metabolic Switch via Mitochondrial ERK-Mediated Phosphorylation of the Chaperone TRAP1. Cell Rep 2017; 18:659-672. [PMID: 28099845 DOI: 10.1016/j.celrep.2016.12.056] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 11/04/2016] [Accepted: 12/18/2016] [Indexed: 01/21/2023] Open
Abstract
Mutations in neurofibromin, a Ras GTPase-activating protein, lead to the tumor predisposition syndrome neurofibromatosis type 1. Here, we report that cells lacking neurofibromin exhibit enhanced glycolysis and decreased respiration in a Ras/ERK-dependent way. In the mitochondrial matrix of neurofibromin-deficient cells, a fraction of active ERK1/2 associates with succinate dehydrogenase (SDH) and TRAP1, a chaperone that promotes the accumulation of the oncometabolite succinate by inhibiting SDH. ERK1/2 enhances both formation of this multimeric complex and SDH inhibition. ERK1/2 kinase activity is favored by the interaction with TRAP1, and TRAP1 is, in turn, phosphorylated in an ERK1/2-dependent way. TRAP1 silencing or mutagenesis at the serine residues targeted by ERK1/2 abrogates tumorigenicity, a phenotype that is reverted by addition of a cell-permeable succinate analog. Our findings reveal that Ras/ERK signaling controls the metabolic changes orchestrated by TRAP1 that have a key role in tumor growth and are a promising target for anti-neoplastic strategies.
Collapse
Affiliation(s)
- Ionica Masgras
- CNR Institute of Neuroscience and Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Francesco Ciscato
- CNR Institute of Neuroscience and Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Anna Maria Brunati
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Elena Tibaldi
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | | | | | - Federica Chiara
- Department of Cardiac, Thoracic, and Vascular Sciences, University of Padova, 35128 Padova, Italy
| | - Giuseppe Cannino
- CNR Institute of Neuroscience and Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Elena Papaleo
- Computational Biology Laboratory, Unit of Statistics, Bioinformatics and Registry, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
| | - Matteo Lambrughi
- Computational Biology Laboratory, Unit of Statistics, Bioinformatics and Registry, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
| | - Giulia Guzzo
- CNR Institute of Neuroscience and Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Alberto Gambalunga
- Department of Cardiac, Thoracic, and Vascular Sciences, University of Padova, 35128 Padova, Italy
| | - Marco Pizzi
- Department of Medicine, University of Padova, 35128 Padova, Italy
| | | | - Massimo Rugge
- Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Stefania Edith Vuljan
- Department of Cardiac, Thoracic, and Vascular Sciences, University of Padova, 35128 Padova, Italy
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic, and Vascular Sciences, University of Padova, 35128 Padova, Italy
| | - Paolo Bernardi
- CNR Institute of Neuroscience and Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Andrea Rasola
- CNR Institute of Neuroscience and Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy.
| |
Collapse
|
48
|
Byron SA, Tran NL, Halperin RF, Phillips JJ, Kuhn JG, de Groot JF, Colman H, Ligon KL, Wen PY, Cloughesy TF, Mellinghoff IK, Butowski NA, Taylor JW, Clarke JL, Chang SM, Berger MS, Molinaro AM, Maggiora GM, Peng S, Nasser S, Liang WS, Trent JM, Berens ME, Carpten JD, Craig DW, Prados MD. Prospective Feasibility Trial for Genomics-Informed Treatment in Recurrent and Progressive Glioblastoma. Clin Cancer Res 2017; 24:295-305. [PMID: 29074604 DOI: 10.1158/1078-0432.ccr-17-0963] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 08/15/2017] [Accepted: 10/03/2017] [Indexed: 01/16/2023]
Abstract
Purpose: Glioblastoma is an aggressive and molecularly heterogeneous cancer with few effective treatment options. We hypothesized that next-generation sequencing can be used to guide treatment recommendations within a clinically acceptable time frame following surgery for patients with recurrent glioblastoma.Experimental Design: We conducted a prospective genomics-informed feasibility trial in adults with recurrent and progressive glioblastoma. Following surgical resection, genome-wide tumor/normal exome sequencing and tumor RNA sequencing were performed to identify molecular targets for potential matched therapy. A multidisciplinary molecular tumor board issued treatment recommendations based on the genomic results, blood-brain barrier penetration of the indicated therapies, drug-drug interactions, and drug safety profiles. Feasibility of generating genomics-informed treatment recommendations within 35 days of surgery was assessed.Results: Of the 20 patients enrolled in the study, 16 patients had sufficient tumor tissue for analysis. Exome sequencing was completed for all patients, and RNA sequencing was completed for 14 patients. Treatment recommendations were provided within the study's feasibility time frame for 15 of 16 (94%) patients. Seven patients received treatment based on the tumor board recommendations. Two patients reached 12-month progression-free survival, both adhering to treatments based on the molecular profiling results. One patient remained on treatment and progression free 21 months after surgery, 3 times longer than the patient's previous time to progression. Analysis of matched nonenhancing tissue from 12 patients revealed overlapping as well as novel putatively actionable genomic alterations.Conclusions: Use of genome-wide molecular profiling is feasible and can be informative for guiding real-time, central nervous system-penetrant, genomics-informed treatment recommendations for patients with recurrent glioblastoma. Clin Cancer Res; 24(2); 295-305. ©2017 AACRSee related commentary by Wick and Kessler, p. 256.
Collapse
Affiliation(s)
- Sara A Byron
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Nhan L Tran
- Departments of Cancer Biology and Neurosurgery, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Rebecca F Halperin
- Quantitative Medicine & Systems Biology Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Joanna J Phillips
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California.,Department of Neuropathology, University of California, San Francisco, San Francisco, California
| | - John G Kuhn
- College of Pharmacy, University of Texas Health Science Center, San Antonio, Texas
| | - John F de Groot
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Howard Colman
- Department of Neurosurgery, University of Utah Huntsman Cancer Institute, Salt Lake City, Utah
| | - Keith L Ligon
- Center for Neuro-Oncology, Dana-Farber Cancer Center, Boston, Massachusetts.,Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Pathology, Harvard Medical School, Boston, Massachusetts
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts
| | - Timothy F Cloughesy
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.,Neuro-Oncology Program, The Ronald Reagan UCLA Medical Center, University of California, Los Angeles, Los Angeles, California
| | - Ingo K Mellinghoff
- Department of Neurology and Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nicholas A Butowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Jennie W Taylor
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Jennifer L Clarke
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Susan M Chang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Mitchel S Berger
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California.,Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Gerald M Maggiora
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Sen Peng
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Sara Nasser
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Winnie S Liang
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, Arizona.,Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Jeffrey M Trent
- Genetic Basis of Human Disease Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Michael E Berens
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - John D Carpten
- Department of Translational Genomics, University of Southern California, Los Angeles, California
| | - David W Craig
- Department of Translational Genomics, University of Southern California, Los Angeles, California
| | - Michael D Prados
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California.
| |
Collapse
|
49
|
Narayan RS, Gasol A, Slangen PLG, Cornelissen FMG, Lagerweij T, Veldman HYYE, Dik R, van den Berg J, Slotman BJ, Würdinger T, Haas-Kogan DA, Stalpers LJA, Baumert BG, Westerman BA, Theys J, Sminia P. Identification of MEK162 as a Radiosensitizer for the Treatment of Glioblastoma. Mol Cancer Ther 2017; 17:347-354. [PMID: 28958992 DOI: 10.1158/1535-7163.mct-17-0480] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/11/2017] [Accepted: 09/08/2017] [Indexed: 11/16/2022]
Abstract
Glioblastoma (GBM) is a highly aggressive and lethal brain cancer type. PI3K and MAPK inhibitors have been studied preclinically in GBM as monotherapy, but not in combination with radiotherapy, which is a key component of the current standard treatment of GBM. In our study, GBM cell lines and patient representative primary cultures were grown as multicellular spheroids. Spheroids were treated with a panel of small-molecule drugs including MK2206, RAD001, BEZ235, MLN0128, and MEK162, alone and in combination with irradiation. Following treatment, spheroid growth parameters (growth rate, volume reduction, and time to regrow), cell-cycle distribution and expression of key target proteins were evaluated. In vivo, the effect of irradiation (3 × 2 Gy) without or with MEK162 (50 mg/kg) was studied in orthotopic GBM8 brain tumor xenografts with endpoints tumor growth and animal survival. The MAPK-targeting agent MEK162 was found to enhance the effect of irradiation as demonstrated by growth inhibition of spheroids. MEK162 downregulated and dephosphorylated the cell-cycle checkpoint proteins CDK1/CDK2/WEE1 and DNA damage response proteins p-ATM/p-CHK2. When combined with radiation, this led to a prolonged DNA damage signal. In vivo data on tumor-bearing animals demonstrated a significantly reduced growth rate, increased growth delay, and prolonged survival time. In addition, RNA expression of responsive cell cultures correlated to mesenchymal stratification of patient expression data. In conclusion, the MAPK inhibitor MEK162 was identified as a radiosensitizer in GBM spheroids in vitro and in orthotopic GBM xenografts in vivo The data are supportive for implementation of this targeted agent in an early-phase clinical study in GBM patients. Mol Cancer Ther; 17(2); 347-54. ©2017 AACRSee all articles in this MCT Focus section, "Developmental Therapeutics in Radiation Oncology."
Collapse
Affiliation(s)
- Ravi S Narayan
- Department of Radiation Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Ana Gasol
- Department of Radiation Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Paul L G Slangen
- Department of Radiation Oncology, VU University Medical Center, Amsterdam, the Netherlands.,Department of Neurosurgery, Neuro-oncology Research Group, VU University Medical Center, Amsterdam, the Netherlands
| | - Fleur M G Cornelissen
- Department of Radiation Oncology, VU University Medical Center, Amsterdam, the Netherlands.,Department of Neurosurgery, Neuro-oncology Research Group, VU University Medical Center, Amsterdam, the Netherlands
| | - Tonny Lagerweij
- Department of Neurosurgery, Neuro-oncology Research Group, VU University Medical Center, Amsterdam, the Netherlands
| | - Hou Y Y E Veldman
- Department of Neurosurgery, Neuro-oncology Research Group, VU University Medical Center, Amsterdam, the Netherlands
| | - Rogier Dik
- Department of Radiation Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Jaap van den Berg
- Department of Radiation Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Ben J Slotman
- Department of Radiation Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Tom Würdinger
- Department of Neurosurgery, Neuro-oncology Research Group, VU University Medical Center, Amsterdam, the Netherlands
| | - Daphne A Haas-Kogan
- Department or Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Lukas J A Stalpers
- Department of Radiation Oncology, Academic Medical Center, Amsterdam, the Netherlands
| | - Brigitta G Baumert
- Department of Radiation Oncology (MAASTRO), Maastricht University Medical Center (MUMC) and GROW (School for Oncology), Maastricht, the Netherlands
| | - Bart A Westerman
- Department of Neurosurgery, Neuro-oncology Research Group, VU University Medical Center, Amsterdam, the Netherlands
| | - Jan Theys
- Department of Radiation Oncology (MAASTRO), Maastricht University Medical Center (MUMC) and GROW (School for Oncology), Maastricht, the Netherlands
| | - Peter Sminia
- Department of Radiation Oncology, VU University Medical Center, Amsterdam, the Netherlands.
| |
Collapse
|
50
|
Allen CE, Laetsch TW, Mody R, Irwin MS, Lim MS, Adamson PC, Seibel NL, Parsons DW, Cho YJ, Janeway K. Target and Agent Prioritization for the Children's Oncology Group-National Cancer Institute Pediatric MATCH Trial. J Natl Cancer Inst 2017; 109:2972640. [PMID: 28376230 DOI: 10.1093/jnci/djw274] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 10/13/2016] [Indexed: 12/15/2022] Open
Abstract
Over the past decades, outcomes for children with cancer have improved dramatically through serial clinical trials based in large measure on dose intensification of cytotoxic chemotherapy for children with high-risk malignancies. Progress made through such dose intensification, in general, is no longer yielding further improvements in outcome. With the revolution in sequencing technologies and rapid development of drugs that block specific proteins and pathways, there is now an opportunity to improve outcomes for pediatric cancer patients through mutation-based targeted therapeutic strategies. The Children's Oncology Group (COG), in partnership with the National Cancer Institute (NCI), is planning a trial entitled the COG-NCI Pediatric Molecular Analysis for Therapeutic Choice (Pediatric MATCH) protocol utilizing an umbrella design. This protocol will have centralized infrastructure and will consist of a biomarker profiling protocol and multiple single-arm phase II trials of targeted therapies. Pediatric patients with recurrent or refractory solid tumors, lymphomas, or histiocytoses with measurable disease will be eligible. The Pediatric MATCH Target and Agent Prioritization (TAP) committee includes membership representing COG disease committees, the Food and Drug Administration, and the NCI. The TAP Committee systematically reviewed target and agent pairs for inclusion in the Pediatric MATCH trial. Fifteen drug-target pairs were reviewed by the TAP Committee, with seven recommended for further development as initial arms of the Pediatric MATCH trial. The current evidence for availability, efficacy, and safety of targeted agents in children for each class of mutation considered for inclusion in the Pediatric MATCH trial is discussed in this review.
Collapse
Affiliation(s)
- Carl E Allen
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX, USA.,Division of Pediatric Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Theodore W Laetsch
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Pauline Allen Gill Center for Cancer and Blood Disorders, Children's Health, Dallas, TX, USA
| | - Rajen Mody
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Meredith S Irwin
- Department of Pediatrics, Division of Hematology/Oncology, Hospital for Sick Children, Toronto, ON, Canada
| | - Megan S Lim
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Hospital of the University of Pennsylvania and Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Peter C Adamson
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nita L Seibel
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, USA
| | - D Williams Parsons
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX, USA.,Division of Pediatric Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Y Jae Cho
- Division of Pediatric Neurology, Doernbecher Children's Hospital, Portland, OR, USA.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Katherine Janeway
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer Center and Blood Disorder Center, Boston, MA, USA
| | | |
Collapse
|