1
|
Kong D, Zha L, Yao Y, Zhang Z, Gao J, Zhang R, Chen L, Wang Z. Effects of HMGA2 on the biological characteristics and stemness acquisition of gastric cancer cells. Arab J Gastroenterol 2024; 25:135-142. [PMID: 38378354 DOI: 10.1016/j.ajg.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 09/01/2023] [Accepted: 01/02/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND AND STUDY AIMS The high mobility group A2 (HMGA2), a nonhistone nuclear binding protein, modulates transcription by altering the chromatin architecture of the target gene DNA in its specific AT-hooks region. HMGA2 overexpression has been observed in embryonic tissue and many malignant neoplasms. This study sought to verify whether HMGA2 plays a role in the biological functions of gastric cancer cells, such as cell proliferation, invasiveness, migration, and stem cell acquisition, and to provide some ideas for further research on the metastatic mechanism of gastric cancer. PATIENTS AND METHODS HMGA2's effects on the proliferation, invasiveness, and migration capabilities of gastric cancer cells were individually detected by BrdU, Transwell, and wound healing assays. Western blotting and immunofluorescence were used to evaluate whether HMGA2 could promote the acquisition of gastric cancer cells. Biostatistical analyses were performed using SPSS 17.0 for Windows. RESULTS HMGA2 expression levels in gastric cancer cell lines were significantly higher than those in human immortalized gastric epithelial cell lines (p < 0.01). Gastric cancer cell proliferation was inhibited when HMGA2 was overexpressed (p < 0.05). The invasiveness and migration capabilities of gastric cancer cells with HMGA2 overexpression were enhanced more than those of the corresponding control groups (p < 0.05). HMGA2 overexpression promotes the stemness acquisition of stem cells from gastric cancer cells. CONCLUSIONS This study verified that the HMGA2 structural transcription factor promotes invasiveness, migration, and acquisition of gastric cancer cells. Furthermore, our findings provide significant insight for further research on the metastatic mechanism of gastric cancer.
Collapse
Affiliation(s)
- Dequan Kong
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China.
| | - Lang Zha
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yaben Yao
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Zhenyu Zhang
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Jun Gao
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Rui Zhang
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Lei Chen
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Ziwei Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
2
|
Hartman TW, Radichev E, Ali HM, Alaba MO, Hoffman M, Kassa G, Sani R, Gadhamshetty V, Ragi S, Messerli SM, de la Puente P, Sandhurst ES, Do T, Lushbough C, Gnimpieba EZ. BASIN: A Semi-automatic Workflow, with Machine Learning Segmentation, for Objective Statistical Analysis of Biomedical and Biofilm Image Datasets. J Mol Biol 2023; 435:167895. [PMID: 36463932 PMCID: PMC10280363 DOI: 10.1016/j.jmb.2022.167895] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/01/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022]
Abstract
Micrograph comparison remains useful in bioscience. This technology provides researchers with a quick snapshot of experimental conditions. But sometimes a two- condition comparison relies on researchers' eyes to draw conclusions. Our Bioimage Analysis, Statistic, and Comparison (BASIN) software provides an objective and reproducible comparison leveraging inferential statistics to bridge image data with other modalities. Users have access to machine learning-based object segmentation. BASIN provides several data points such as images' object counts, intensities, and areas. Hypothesis testing may also be performed. To improve BASIN's accessibility, we implemented it using R Shiny and provided both an online and offline version. We used BASIN to process 498 image pairs involving five bioscience topics. Our framework supported either direct claims or extrapolations 57% of the time. Analysis results were manually curated to determine BASIN's accuracy which was shown to be 78%. Additionally, each BASIN version's initial release shows an average 82% FAIR compliance score.
Collapse
Affiliation(s)
- Timothy W Hartman
- Biomedical Engineering Department, University of South Dakota Sioux Falls, 4800 N Career Avenue, Sioux Falls, SD 57107, United States
| | - Evgeni Radichev
- Biomedical Engineering Department, University of South Dakota Sioux Falls, 4800 N Career Avenue, Sioux Falls, SD 57107, United States
| | - Hafiz Munsub Ali
- Biomedical Engineering Department, University of South Dakota Sioux Falls, 4800 N Career Avenue, Sioux Falls, SD 57107, United States
| | - Mathew Olakunle Alaba
- Biomedical Engineering Department, University of South Dakota Sioux Falls, 4800 N Career Avenue, Sioux Falls, SD 57107, United States
| | - Mariah Hoffman
- Biomedical Engineering Department, University of South Dakota Sioux Falls, 4800 N Career Avenue, Sioux Falls, SD 57107, United States
| | - Gideon Kassa
- Biomedical Engineering Department, University of South Dakota Sioux Falls, 4800 N Career Avenue, Sioux Falls, SD 57107, United States
| | - Rajesh Sani
- Chemical and Biological Engineering Department, South Dakota School of Mines and Technology, 501 E St. Joseph Street, Rapid City, SD 57701, United States
| | - Venkata Gadhamshetty
- Civil and Environmental Engineering Department, South Dakota School of Mines and Technology, 501 E St. Joseph Street, Rapid City, SD 57701, United States
| | - Shankarachary Ragi
- Electrical Engineering Department, South Dakota School of Mines and Technology, 501 E St. Joseph Street, Rapid City, SD 57701, United States
| | - Shanta M Messerli
- Biomedical Engineering Department, University of South Dakota Sioux Falls, 4800 N Career Avenue, Sioux Falls, SD 57107, United States; Cancer Biology and Immunotherapies Group, Sanford Research, 2301 E 60(th) Street North, Sioux Falls, SD 57104, United States; Department of Biology and Microbiology, South Dakota State University, Brookings, SD 57006, United States
| | - Pilar de la Puente
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 E 60(th) Street North, Sioux Falls, SD 57104, United States
| | - Eric S Sandhurst
- Biomedical Engineering Department, University of South Dakota Sioux Falls, 4800 N Career Avenue, Sioux Falls, SD 57107, United States
| | - Tuyen Do
- Biomedical Engineering Department, University of South Dakota Sioux Falls, 4800 N Career Avenue, Sioux Falls, SD 57107, United States
| | - Carol Lushbough
- Biomedical Engineering Department, University of South Dakota Sioux Falls, 4800 N Career Avenue, Sioux Falls, SD 57107, United States
| | - Etienne Z Gnimpieba
- Biomedical Engineering Department, University of South Dakota Sioux Falls, 4800 N Career Avenue, Sioux Falls, SD 57107, United States.
| |
Collapse
|
3
|
Parkhurst A, Wang SZ, Findlay TR, Malebranche KJ, Odabas A, Alt J, Maxwell MJ, Kaur H, Peer CJ, Figg WD, Warren KE, Slusher BS, Eberhart CG, Raabe EH, Rubens JA. Dual mTORC1/2 inhibition compromises cell defenses against exogenous stress potentiating Obatoclax-induced cytotoxicity in atypical teratoid/rhabdoid tumors. Cell Death Dis 2022; 13:410. [PMID: 35484114 PMCID: PMC9050713 DOI: 10.1038/s41419-022-04868-9] [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: 11/18/2021] [Revised: 04/08/2022] [Accepted: 04/19/2022] [Indexed: 11/09/2022]
Abstract
AbstractAtypical teratoid/rhabdoid tumors (AT/RT) are the most common malignant brain tumors of infancy and have a dismal 4-year event-free survival (EFS) of 37%. We have previously shown that mTOR activation contributes to AT/RT’s aggressive growth and poor survival. Targeting the mTOR pathway with the dual mTORC1/2 inhibitor TAK-228 slows tumor growth and extends survival in mice bearing orthotopic xenografts. However, responses are primarily cytostatic with limited durability. The aim of this study is to understand the impact of mTOR inhibitors on AT/RT signaling pathways and design a rational combination therapy to drive a more durable response to this promising therapy. We performed RNASeq, gene expression studies, and protein analyses to identify pathways disrupted by TAK-228. We find that TAK-228 decreases the expression of the transcription factor NRF2 and compromises AT/RT cellular defenses against oxidative stress and apoptosis. The BH3 mimetic, Obatoclax, is a potent inducer of oxidative stress and apoptosis in AT/RT. These complementary mechanisms of action drive extensive synergies between TAK-228 and Obatoclax slowing AT/RT cell growth and inducing apoptosis and cell death. Combination therapy activates the integrative stress response as determined by increased expression of phosphorylated EIF2α, ATF4, and CHOP, and disrupts the protective NOXA.MCL-1.BIM axis, forcing stressed cells to undergo apoptosis. Combination therapy is well tolerated in mice bearing orthotopic xenografts of AT/RT, slows tumor growth, and extends median overall survival. This novel combination therapy could be added to standard upfront therapies or used as a salvage therapy for relapsed disease to improve outcomes in AT/RT.
Collapse
|
4
|
Non-adhesive and highly stable biodegradable nanoparticles that provide widespread and safe transgene expression in orthotopic brain tumors. Drug Deliv Transl Res 2021; 10:572-581. [PMID: 32323162 DOI: 10.1007/s13346-020-00759-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Several generations of poly(β-amino ester) (PBAE) polymers have been developed for efficient cellular transfection. However, PBAE-based gene vectors, similar to other cationic materials, cannot readily provide widespread gene transfer in the brain due to adhesive interactions with the extracellular matrix (ECM). We thus engineered eight vector candidates using previously identified lead PBAE polymer variants but endowed them with non-adhesive surface coatings to facilitate their spread through brain ECM. Specifically, we screened for the ability to provide widespread gene transfer in tumor spheroids and healthy mouse brains. We then confirmed that a lead formulation provided widespread transgene expression in orthotopically established brain tumor models with an excellent in vivo safety profile. Lastly, we developed a method to store it long-term while fully retaining its brain-penetrating property. This new platform provides a broad utility in evaluating novel genetic targets for gene therapy of brain tumors and neurological disorders in preclinical and clinical settings. Graphical abstract We engineered biodegradable DNA-loaded brain-penetrating nanoparticles (DNA-BPN) possessing small particle diameters (< 70 nm) and non-adhesive surface coatings to facilitate their spread through brain tumor extracellular matrix (ECM). These DNA-BPN provide widespread gene transfer in models recapitulating the ECM barrier, including three-dimensional multicellular tumor spheroids and mice with orthotopically established brain tumor.
Collapse
|
5
|
Arnold A, Yuan M, Price A, Harris L, Eberhart CG, Raabe EH. Synergistic activity of mTORC1/2 kinase and MEK inhibitors suppresses pediatric low-grade glioma tumorigenicity and vascularity. Neuro Oncol 2021; 22:563-574. [PMID: 31841591 DOI: 10.1093/neuonc/noz230] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Pediatric low-grade glioma (pLGG) is the most common childhood brain tumor. Many patients with unresectable or recurrent/refractory tumors have significant lifelong disability. The majority of pLGG have mutations increasing the activity of the Ras/mitogen-activated protein kinase (MAPK) pathway. Activation of mammalian target of rapamycin (mTOR) is also a hallmark of pLGG. We therefore hypothesized that the dual target of rapamycin complexes 1 and 2 (TORC1/2) kinase inhibitor TAK228 would synergize with the mitogen-activated extracellular signal-regulated kinase (MEK) inhibitor trametinib in pLGG. METHODS We tested TAK228 and trametinib in patient-derived pLGG cell lines harboring drivers of pLGG including BRAFV600E and neurofibromatosis type 1 loss. We measured cell proliferation, pathway inhibition, cell death, and senescence. Synergy was analyzed via MTS assay using the Chou-Talalay method. In vivo, we tested for overall survival and pathway inhibition and performed immunohistochemistry for proliferation and vascularization. We performed a scratch assay and measured angiogenesis protein activation in human umbilical vein endothelial cells (HUVECs). RESULTS TAK228 synergized with trametinib in pLGG at clinically relevant doses in all tested cell lines, suppressing proliferation, inducing apoptosis, and causing senescence in a cell line-dependent manner. Combination treatment increased median survival by 70% and reduced tumor volume compared with monotreatment and control cohorts. Vascularization of tumors decreased as measured by CD31 and CD34. Combination treatment blocked activation of focal adhesion kinase (FAK) and sarcoma proto-oncogene non-receptor tyrosine kinase (SRC) in HUVEC cells and reduced HUVEC migration compared with each drug alone. CONCLUSIONS The combination of TAK228 and trametinib synergized to suppress the growth of pLGG. These agents synergized to reduce tumor vascularity and endothelial cell growth and migration by blocking activation of FAK and SRC.
Collapse
Affiliation(s)
- Antje Arnold
- Johns Hopkins School of Medicine, Department of Pathology, Division of Neuropathology, Baltimore, Maryland
| | - Ming Yuan
- Johns Hopkins School of Medicine, Department of Pathology, Division of Neuropathology, Baltimore, Maryland
| | - Antionette Price
- Johns Hopkins School of Medicine, Department of Pathology, Division of Neuropathology, Baltimore, Maryland
| | - Lauren Harris
- Johns Hopkins University Krieger School of Arts and Sciences, Department of Molecular and Cell Biology, Baltimore, Maryland
| | - Charles G Eberhart
- Johns Hopkins School of Medicine, Department of Pathology, Division of Neuropathology, Baltimore, Maryland
| | - Eric H Raabe
- Johns Hopkins School of Medicine, Department of Pathology, Division of Neuropathology, Baltimore, Maryland.,Johns Hopkins School of Medicine, Sidney Kimmel Comprehensive Cancer Center, Division of Pediatric Oncology, Baltimore, Maryland
| |
Collapse
|
6
|
Ho B, Johann PD, Grabovska Y, De Dieu Andrianteranagna MJ, Yao F, Frühwald M, Hasselblatt M, Bourdeaut F, Williamson D, Huang A, Kool M. Molecular subgrouping of atypical teratoid/rhabdoid tumors-a reinvestigation and current consensus. Neuro Oncol 2021; 22:613-624. [PMID: 31889194 PMCID: PMC7229260 DOI: 10.1093/neuonc/noz235] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Atypical teratoid/rhabdoid tumors (ATRTs) are known to exhibit molecular and clinical heterogeneity even though SMARCB1 inactivation is the sole recurrent genetic event present in nearly all cases. Indeed, recent studies demonstrated 3 molecular subgroups of ATRTs that are genetically, epigenetically, and clinically distinct. As these studies included different numbers of tumors, various subgrouping techniques, and naming, an international working group sought to align previous findings and to reach a consensus on nomenclature and clinicopathological significance of ATRT subgroups. Methods We integrated various methods to perform a meta-analysis on published and unpublished DNA methylation and gene expression datasets of ATRTs and associated clinicopathological data. Results In concordance with previous studies, the analyses identified 3 main molecular subgroups of ATRTs, for which a consensus was reached to name them ATRT-TYR, ATRT-SHH, and ATRT-MYC. The ATRT-SHH subgroup exhibited further heterogeneity, segregating further into 2 subtypes associated with a predominant supratentorial (ATRT-SHH-1) or infratentorial (ATRT-SHH-2) location. For each ATRT subgroup we provide an overview of its main molecular and clinical characteristics, including SMARCB1 alterations and pathway activation. Conclusions The introduction of a common classification, characterization, and nomenclature of ATRT subgroups will facilitate future research and serve as a common ground for subgrouping patient samples and ATRT models, which will aid in refining subgroup-based therapies for ATRT patients.
Collapse
Affiliation(s)
- Ben Ho
- Division of Hematology and Oncology, Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Pascal D Johann
- Hopp Children's Cancer Center, Heidelberg, Germany.,Division of Pediatric Neuro-oncology, German Cancer Research Center and German Cancer Research Consortium, Heidelberg, Germany.,Department of Pediatric Hematology and Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Yura Grabovska
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Mamy Jean De Dieu Andrianteranagna
- Departments of Genetics and of Oncopediatry and Young Adults, Curie Institute, Paris, France.,INSERM U830, Laboratory of Translational Research in Pediatric Oncology, SIREDO Pediatric Oncology Center, Curie Institute, Paris, France
| | - Fupan Yao
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Michael Frühwald
- University Children's Hospital Augsburg, Swabian Children's Cancer Center, Augsburg, Germany
| | - Martin Hasselblatt
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Franck Bourdeaut
- Departments of Genetics and of Oncopediatry and Young Adults, Curie Institute, Paris, France.,INSERM U830, Laboratory of Translational Research in Pediatric Oncology, SIREDO Pediatric Oncology Center, Curie Institute, Paris, France
| | - Daniel Williamson
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Annie Huang
- Division of Hematology and Oncology, Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Marcel Kool
- Hopp Children's Cancer Center, Heidelberg, Germany.,Division of Pediatric Neuro-oncology, German Cancer Research Center and German Cancer Research Consortium, Heidelberg, Germany
| |
Collapse
|
7
|
Wang Q, Wang G, Xu X, Chen Z. miR-760 mediated the proliferation and metastasis of hepatocellular carcinoma cells by regulating HMGA2. Pathol Res Pract 2021; 222:153420. [PMID: 33887625 DOI: 10.1016/j.prp.2021.153420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND The purpose of our study was to investigate the roles of miR-760 and its potential mechanisms in HCC. METHODS The functions of miR-760 were identified and measured by MTT, colony formation, transwell, and flow cytometry assays. Luciferase assay was applied to verify the direct binding of miR-760 on HMGA2 3'untranslated region (3'UTR). Then, in vitro experiment was used to investigate the biological effects of miR-760 and HMGA2. Luciferase and ChIP assays were used to detect the validity of SP1 binding sites on the miR-760 promoter. RESULTS We demonstrated that miR-760 overexpression suppressed cell proliferation, migration, and invasion in HCC. Besides, HMGA2 was demonstrated as a direct target gene of miR-760. Furthermore, we found that methylation may result in the downregulation of miR-760, and SP1 could inhibit the transcription of miR-760. CONCLUSIONS Our study demonstrated that SP1/miR-760/HMGA2 may serve as a molecular regulatory axis for HCC treatment.
Collapse
Affiliation(s)
- Quhui Wang
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Gang Wang
- Department of Anesthesiology, Union Hospital Affiliated With Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Xiaodong Xu
- Department of General Surgery, The 4th Affiliated Hospital of Nantong University, Yancheng, 224000, China
| | - Zhong Chen
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China; Department of General Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China.
| |
Collapse
|
8
|
HMGA2 as a Critical Regulator in Cancer Development. Genes (Basel) 2021; 12:genes12020269. [PMID: 33668453 PMCID: PMC7917704 DOI: 10.3390/genes12020269] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/01/2021] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
The high mobility group protein 2 (HMGA2) regulates gene expression by binding to AT-rich regions of DNA. Akin to other DNA architectural proteins, HMGA2 is highly expressed in embryonic stem cells during embryogenesis, while its expression is more limited at later stages of development and in adulthood. Importantly, HMGA2 is re-expressed in nearly all human malignancies, where it promotes tumorigenesis by multiple mechanisms. HMGA2 increases cancer cell proliferation by promoting cell cycle entry and inhibition of apoptosis. In addition, HMGA2 influences different DNA repair mechanisms and promotes epithelial-to-mesenchymal transition by activating signaling via the MAPK/ERK, TGFβ/Smad, PI3K/AKT/mTOR, NFkB, and STAT3 pathways. Moreover, HMGA2 supports a cancer stem cell phenotype and renders cancer cells resistant to chemotherapeutic agents. In this review, we discuss these oncogenic roles of HMGA2 in different types of cancers and propose that HMGA2 may be used for cancer diagnostic, prognostic, and therapeutic purposes.
Collapse
|
9
|
Shahab S, Rubens J, Kaur H, Sweeney H, Eberhart CG, Raabe EH. MEK Inhibition Suppresses Growth of Atypical Teratoid/Rhabdoid Tumors. J Neuropathol Exp Neurol 2020; 79:746-753. [PMID: 32472116 DOI: 10.1093/jnen/nlaa042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/05/2020] [Accepted: 04/25/2020] [Indexed: 11/12/2022] Open
Abstract
Atypical teratoid/rhabdoid (AT/RT) tumors are the most common malignant brain tumor of infancy and have a poor prognosis. We have previously identified very high expression of LIN28A and/or LIN28B in AT/RT tumors and showed that AT/RT have corresponding increased expression of the mitogen-activated protein (MAP) kinase pathway. Binimetinib is a novel inhibitor of mitogen-activated protein kinase (MAP2K1 or MEK), and is currently in pediatric phase II clinical trials for low-grade glioma. We hypothesized that binimetinib would inhibit growth of AT/RT cells by suppressing the MAP kinase pathway. Binimetinib inhibited AT/RT growth at nanomolar concentrations. Binimetinib decreased cell proliferation and induced apoptosis in AT/RT cells and significantly reduced AT/RT tumor growth in flank xenografts. Our data suggest that MAP kinase pathway inhibition could offer a potential avenue for treating these highly aggressive tumors.
Collapse
Affiliation(s)
- Shubin Shahab
- Division of Pediatric Oncology, Department of Oncology.,Sidney Kimmel Comprehensive Cancer Center
| | - Jeffrey Rubens
- Division of Pediatric Oncology, Department of Oncology.,Sidney Kimmel Comprehensive Cancer Center
| | - Harpreet Kaur
- Division of Pediatric Oncology, Department of Oncology.,Sidney Kimmel Comprehensive Cancer Center
| | | | - Charles G Eberhart
- Sidney Kimmel Comprehensive Cancer Center.,Division of Neuropathology, Department of Pathology (CGE, EHR), Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Eric H Raabe
- Division of Pediatric Oncology, Department of Oncology.,Sidney Kimmel Comprehensive Cancer Center.,Division of Neuropathology, Department of Pathology (CGE, EHR), Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
10
|
Ouchi K, Miyachi M, Yagyu S, Kikuchi K, Kuwahara Y, Tsuchiya K, Iehara T, Hosoi H. Oncogenic role of HMGA2 in fusion-negative rhabdomyosarcoma cells. Cancer Cell Int 2020; 20:192. [PMID: 32489328 PMCID: PMC7247181 DOI: 10.1186/s12935-020-01282-z] [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] [Received: 08/22/2019] [Accepted: 05/19/2020] [Indexed: 11/21/2022] Open
Abstract
Background Rhabdomyosarcoma (RMS) is the most common pediatric soft tissue sarcoma. There are two subtypes, fusion gene-positive RMS (FP-RMS) and fusion gene-negative RMS (FN-RMS), depending on the presence of a fusion gene, either PAX3-FOXO1 or PAX7-FOXO1. These fusion genes are thought to be oncogenic drivers of FP-RMS. By contrast, the underlying mechanism of FN-RMS has not been thoroughly investigated. It has recently been shown that HMGA2 is specifically positive in pathological tissue from FN-RMS, but the role of HMGA2 in FN-RMS remains to be clarified. Methods In this study, we used FN-RMS cell lines to investigate the function of HMGA2. Gene expression, cell growth, cell cycle, myogenic differentiation, tumor formation in vivo, and cell viability under drug treatment were assessed. Results We found that HMGA2 was highly expressed in FN-RMS cells compared with FP-RMS cells and that knockdown of HMGA2 in FN-RMS cells inhibited cell growth and induced G1 phase accumulation in the cell cycle and myogenic differentiation. Additionally, we showed using both gain-of-function and loss-of-function assays that HMGA2 was required for tumor formation in vivo. Consistent with these findings, the HMGA2 inhibitor netropsin inhibited the cell growth of FN-RMS. Conclusions Our results suggest that HMGA2 has important role in the oncogenicity of FP-RMS and may be a potential therapeutic target in patients with FN-RMS.
Collapse
Affiliation(s)
- Kazutaka Ouchi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji, Kamigyo-ku, Kyoto, 602-8566 Japan
| | - Mitsuru Miyachi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji, Kamigyo-ku, Kyoto, 602-8566 Japan
| | - Shigeki Yagyu
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji, Kamigyo-ku, Kyoto, 602-8566 Japan
| | - Ken Kikuchi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji, Kamigyo-ku, Kyoto, 602-8566 Japan
| | - Yasumichi Kuwahara
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji, Kamigyo-ku, Kyoto, 602-8566 Japan.,Department of Molecular Biochemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji, Kamigyo-ku, Kyoto, 602-8566 Japan
| | - Kunihiko Tsuchiya
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji, Kamigyo-ku, Kyoto, 602-8566 Japan
| | - Tomoko Iehara
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji, Kamigyo-ku, Kyoto, 602-8566 Japan
| | - Hajime Hosoi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji, Kamigyo-ku, Kyoto, 602-8566 Japan
| |
Collapse
|
11
|
Poore B, Yuan M, Arnold A, Price A, Alt J, Rubens JA, Slusher BS, Eberhart CG, Raabe EH. Inhibition of mTORC1 in pediatric low-grade glioma depletes glutathione and therapeutically synergizes with carboplatin. Neuro Oncol 2020; 21:252-263. [PMID: 30239952 DOI: 10.1093/neuonc/noy150] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Pediatric low-grade glioma (pLGG) often initially responds to front-line therapies such as carboplatin, but more than 50% of treated tumors eventually progress and require additional therapy. With the discovery that pLGG often contains mammalian target of rapamycin (mTOR) activation, new treatment modalities and combinations are now possible for patients. The purpose of this study was to determine if carboplatin is synergistic with the mTOR complex 1 inhibitor everolimus in pLGG. METHODS We treated 4 pLGG cell lines and 1 patient-derived xenograft line representing various pLGG genotypes, including neurofibromatosis type 1 loss, proto-oncogene B-Raf (BRAF)-KIAA1549 fusion, and BRAFV600E mutation, with carboplatin and/or everolimus and performed assays for growth, cell proliferation, and cell death. Immunohistochemistry as well as in vivo and in vitro metabolomics studies were also performed. RESULTS Carboplatin synergized with everolimus in all of our 4 pLGG cell lines (combination index <1 at Fa 0.5). Combination therapy was superior at inhibiting tumor growth in vivo. Combination treatment increased levels of apoptosis as well as gamma-H2AX phosphorylation compared with either agent alone. Everolimus treatment suppressed the conversion of glutamine and glutamate into glutathione both in vitro and in vivo. Exogenous glutathione reversed the effects of carboplatin and everolimus. CONCLUSIONS The combination of carboplatin and everolimus was effective at inducing cell death and slowing tumor growth in pLGG models. Everolimus decreased the amount of available glutathione inside the cell, preventing the detoxification of carboplatin and inducing increased DNA damage and apoptosis.
Collapse
Affiliation(s)
- Brad Poore
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ming Yuan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Antje Arnold
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Antoinette Price
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jesse Alt
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jeffrey A Rubens
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Barbara S Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Charles G Eberhart
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Eric H Raabe
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
12
|
Zhang S, Mo Q, Wang X. Oncological role of HMGA2 (Review). Int J Oncol 2019; 55:775-788. [PMID: 31432151 DOI: 10.3892/ijo.2019.4856] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/17/2019] [Indexed: 11/06/2022] Open
Abstract
The high mobility group A2 (HMGA2) protein is a non‑histone architectural transcription factor that modulates the transcription of several genes by binding to AT‑rich sequences in the minor groove of B‑form DNA and alters the chromatin structure. As a result, HMGA2 influences a variety of biological processes, including the cell cycle process, DNA damage repair process, apoptosis, senescence, epithelial‑mesenchymal transition and telomere restoration. In addition, the overexpression of HMGA2 is a feature of malignancy, and its elevated expression in human cancer predicts the efficacy of certain chemotherapeutic agents. Accumulating evidence has suggested that the detection of HMGA2 can be used as a routine procedure in clinical tumour analysis.
Collapse
Affiliation(s)
- Shizhen Zhang
- Department of Breast Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Qiuping Mo
- Department of Surgical Oncology and Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Xiaochen Wang
- Department of Breast Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
13
|
Wang SZ, Poore B, Alt J, Price A, Allen SJ, Hanaford AR, Kaur H, Orr BA, Slusher BS, Eberhart CG, Raabe EH, Rubens JA. Unbiased Metabolic Profiling Predicts Sensitivity of High MYC-Expressing Atypical Teratoid/Rhabdoid Tumors to Glutamine Inhibition with 6-Diazo-5-Oxo-L-Norleucine. Clin Cancer Res 2019; 25:5925-5936. [PMID: 31300448 DOI: 10.1158/1078-0432.ccr-19-0189] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 05/13/2019] [Accepted: 07/02/2019] [Indexed: 12/21/2022]
Abstract
PURPOSE Atypical teratoid/rhabdoid tumors (AT/RT) are aggressive infantile brain tumors with poor survival. Recent advancements have highlighted significant molecular heterogeneity in AT/RT with an aggressive subgroup featuring overexpression of the MYC proto-oncogene. We perform the first comprehensive metabolic profiling of patient-derived AT/RT cell lines to identify therapeutic susceptibilities in high MYC-expressing AT/RT. EXPERIMENTAL DESIGN Metabolites were extracted from AT/RT cell lines and separated in ultra-high performance liquid chromatography mass spectrometry. Glutamine metabolic inhibition with 6-diazo-5-oxo-L-norleucine (DON) was tested with growth and cell death assays and survival studies in orthotopic mouse models of AT/RT. Metabolic flux analysis was completed to identify combination therapies to act synergistically to improve survival in high MYC AT/RT. RESULTS Unbiased metabolic profiling of AT/RT cell models identified a unique dependence of high MYC AT/RT on glutamine for survival. The glutamine analogue, DON, selectively targeted high MYC cell lines, slowing cell growth, inducing apoptosis, and extending survival in orthotopic mouse models of AT/RT. Metabolic flux experiments with isotopically labeled glutamine revealed DON inhibition of glutathione (GSH) synthesis. DON combined with carboplatin further slowed cell growth, induced apoptosis, and extended survival in orthotopic mouse models of high MYC AT/RT. CONCLUSIONS Unbiased metabolic profiling of AT/RT identified susceptibility of high MYC AT/RT to glutamine metabolic inhibition with DON therapy. DON inhibited glutamine-dependent synthesis of GSH and synergized with carboplatin to extend survival in high MYC AT/RT. These findings can rapidly translate into new clinical trials to improve survival in high MYC AT/RT.
Collapse
Affiliation(s)
- Sabrina Z Wang
- Division of Pediatric Oncology, Johns Hopkins University, School of Medicine, Baltimore, Maryland.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Brad Poore
- Division of Pediatric Oncology, Johns Hopkins University, School of Medicine, Baltimore, Maryland.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Jesse Alt
- Johns Hopkins Drug Discovery, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Antoinette Price
- Division of Neuropathology, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Sariah J Allen
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Allison R Hanaford
- Division of Neuropathology, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Harpreet Kaur
- Division of Pediatric Oncology, Johns Hopkins University, School of Medicine, Baltimore, Maryland.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Brent A Orr
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Barbara S Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins University, School of Medicine, Baltimore, Maryland.,Department of Neurology, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Charles G Eberhart
- Division of Neuropathology, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Eric H Raabe
- Division of Pediatric Oncology, Johns Hopkins University, School of Medicine, Baltimore, Maryland. .,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, Maryland.,Division of Neuropathology, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Jeffrey A Rubens
- Division of Pediatric Oncology, Johns Hopkins University, School of Medicine, Baltimore, Maryland. .,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| |
Collapse
|
14
|
Mansoori B, Mohammadi A, Asadzadeh Z, Shirjang S, Minouei M, Abedi Gaballu F, Shajari N, Kazemi T, Gjerstorff MF, Duijf PHG, Baradaran B. HMGA2 and Bach-1 cooperate to promote breast cancer cell malignancy. J Cell Physiol 2019; 234:17714-17726. [PMID: 30825204 DOI: 10.1002/jcp.28397] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 12/12/2022]
Abstract
During breast cancer progression, tumor cells acquire multiple malignant features. The transcription factors and cell cycle regulators high mobility group A2 (HMGA2) and BTB and CNC homology 1 (Bach-1) are overexpressed in several cancers, but the mechanistic understanding of how HMGA2 and Bach-1 promote cancer development has been limited. We found that HMGA2 and Bach-1 are overexpressed in breast cancer tissues and their expression correlates positively in tumors but not in normal tissues. Individual HMGA2 or Bach-1 knockdown downregulates expression of both proteins, suggesting a mutual stabilizing effect between the two proteins. Importantly, combined HMGA2 and Bach-1 knockdown additively decrease cell proliferation, migration, epithelial-to-mesenchymal transition, and colony formation, while promoting apoptotic cell death via upregulation of caspase-3 and caspase-9. First the first time, we show that HMGA2 and Bach-1 overexpression in tumors correlate positively and that the proteins cooperatively suppress a broad range of malignant cellular properties, such as proliferation, migration, clonogenicity, and evasion of apoptotic cell death. Thus, our observations suggest that combined targeting of HMGA2 and Bach1 may be an effective therapeutic strategy to treat breast cancer.
Collapse
Affiliation(s)
- Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Shirjang
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahsa Minouei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Neda Shajari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morten F Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Pascal H G Duijf
- University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
15
|
Nesvick CL, Nageswara Rao AA, Raghunathan A, Biegel JA, Daniels DJ. Case-based review: atypical teratoid/rhabdoid tumor. Neurooncol Pract 2018; 6:163-178. [PMID: 31386032 DOI: 10.1093/nop/npy037] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Atypical teratoid/rhabdoid tumor (AT/RT) is a rare CNS cancer that typically occurs in children younger than 3 years of age. Histologically, AT/RTs are embryonal tumors that contain a rhabdoid component as well as areas with primitive neuroectodermal, mesenchymal, and epithelial features. Compared to other CNS tumors of childhood, AT/RTs are characterized by their rapid growth, short symptomatic prodrome, and large size upon presentation, often leading to brain compression and intracranial hypertension requiring urgent intervention. For decades, the mainstay of care has been a combination of maximal safe surgical resection followed by adjuvant chemotherapy and radiotherapy. Despite advances in each of these modalities, the relative paucity of data on these tumors, their inherently aggressive course, and a lack of molecular data have limited advances in treatment over the past 3 decades. Recent large-scale, multicenter interdisciplinary studies, however, have significantly advanced our understanding of the molecular pathogenesis of these tumors. Multiple clinical trials testing molecularly targeted therapies are underway, offering hope for patients with AT/RT and their families.
Collapse
Affiliation(s)
- Cody L Nesvick
- Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Amulya A Nageswara Rao
- Department of Pediatric and Adolescent Medicine, Division of Pediatric Hematology/Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Aditya Raghunathan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jaclyn A Biegel
- Department of Pathology and Laboratory Medicine, Children's Hospital of Los Angeles, Keck School of Medicine of University of Southern California, USA
| | - David J Daniels
- Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
16
|
Rubens JA, Wang SZ, Price A, Weingart MF, Allen SJ, Orr BA, Eberhart CG, Raabe EH. The TORC1/2 inhibitor TAK228 sensitizes atypical teratoid rhabdoid tumors to cisplatin-induced cytotoxicity. Neuro Oncol 2018; 19:1361-1371. [PMID: 28582547 DOI: 10.1093/neuonc/nox067] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background Atypical teratoid/rhabdoid tumors (AT/RTs) are deadly pediatric brain tumors driven by LIN28. Mammalian target of rapamycin (mTOR) is activated in many deadly, drug-resistant cancers and governs important cellular functions such as metabolism and survival. LIN28 regulates mTOR in normal cells. We therefore hypothesized that mTOR is activated downstream of LIN28 in AT/RT, and the brain-penetrating mTOR complex 1 and 2 (mTORC1/2) kinase inhibitor TAK228 would reduce AT/RT tumorigenicity. Methods Activation of mTOR in AT/RT was determined by measuring pS6 and pAKT (Ser473) by immunohistochemistry on tissue microarray of 18 primary AT/RT tumors. In vitro growth assays (BrdU and MTS), death assays (CC3, c-PARP by western blot), and survival curves of AT/RT orthotopic xenograft models were used to measure the efficacy of TAK228 alone and in combination with cisplatin. Results Lentiviral short hairpin RNA-mediated knockdown of LIN28A led to decreased mTOR activation. Primary human AT/RT had high levels of pS6 and pAKT (Ser473) in 21% and 87% of tumors by immunohistochemistry. TAK228 slowed cell growth, induced apoptosis in vitro, and nearly doubled median survival of orthotopic xenograft models of AT/RT. TAK228 combined with cisplatin synergistically slowed cell growth and enhanced cisplatin-induced apoptosis. Suppression of AKT sensitized cells to cisplatin-induced apoptosis and forced activation of AKT protected cells. Combined treatment with TAK228 and cisplatin significantly extended survival of orthotopic xenograft models of AT/RT compared with each drug alone. Conclusions TAK228 has efficacy in AT/RT as a single agent and synergizes with conventional chemotherapies by sensitizing tumors to cisplatin-induced apoptosis. These results suggest TAK228 may be an effective new treatment for AT/RT.
Collapse
Affiliation(s)
- Jeffrey A Rubens
- Division of Neuropathology and Sidney Kimmel Comprehensive Cancer Center and Division of Pediatric Oncology and Bloomberg Children's Hospital, Johns Hopkins Hospital, Baltimore, Maryland; St Jude Children's Research Hospital, Memphis, Tennessee
| | - Sabrina Z Wang
- Division of Neuropathology and Sidney Kimmel Comprehensive Cancer Center and Division of Pediatric Oncology and Bloomberg Children's Hospital, Johns Hopkins Hospital, Baltimore, Maryland; St Jude Children's Research Hospital, Memphis, Tennessee
| | - Antoinette Price
- Division of Neuropathology and Sidney Kimmel Comprehensive Cancer Center and Division of Pediatric Oncology and Bloomberg Children's Hospital, Johns Hopkins Hospital, Baltimore, Maryland; St Jude Children's Research Hospital, Memphis, Tennessee
| | - Melanie F Weingart
- Division of Neuropathology and Sidney Kimmel Comprehensive Cancer Center and Division of Pediatric Oncology and Bloomberg Children's Hospital, Johns Hopkins Hospital, Baltimore, Maryland; St Jude Children's Research Hospital, Memphis, Tennessee
| | - Sariah J Allen
- Division of Neuropathology and Sidney Kimmel Comprehensive Cancer Center and Division of Pediatric Oncology and Bloomberg Children's Hospital, Johns Hopkins Hospital, Baltimore, Maryland; St Jude Children's Research Hospital, Memphis, Tennessee
| | - Brent A Orr
- Division of Neuropathology and Sidney Kimmel Comprehensive Cancer Center and Division of Pediatric Oncology and Bloomberg Children's Hospital, Johns Hopkins Hospital, Baltimore, Maryland; St Jude Children's Research Hospital, Memphis, Tennessee
| | - Charles G Eberhart
- Division of Neuropathology and Sidney Kimmel Comprehensive Cancer Center and Division of Pediatric Oncology and Bloomberg Children's Hospital, Johns Hopkins Hospital, Baltimore, Maryland; St Jude Children's Research Hospital, Memphis, Tennessee
| | - Eric H Raabe
- Division of Neuropathology and Sidney Kimmel Comprehensive Cancer Center and Division of Pediatric Oncology and Bloomberg Children's Hospital, Johns Hopkins Hospital, Baltimore, Maryland; St Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
17
|
Casaos J, Huq S, Lott T, Felder R, Choi J, Gorelick N, Peters M, Xia Y, Maxwell R, Zhao T, Ji C, Simon T, Sesen J, Scotland SJ, Kast RE, Rubens J, Raabe E, Eberhart CG, Jackson EM, Brem H, Tyler B, Skuli N. Ribavirin as a potential therapeutic for atypical teratoid/rhabdoid tumors. Oncotarget 2018; 9:8054-8067. [PMID: 29487714 PMCID: PMC5814281 DOI: 10.18632/oncotarget.23883] [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: 03/09/2017] [Accepted: 11/11/2017] [Indexed: 11/25/2022] Open
Abstract
Atypical teratoid/rhabdoid tumors (AT/RT) are highly aggressive, malignant tumors and are the most common malignant brain tumor in children under 6 months of age. Currently, there is no standard treatment for AT/RT. Recent studies have reported potential anti-tumoral properties of ribavirin, a guanosine analog and anti-viral molecule approved by the Food and Drug Administration for treatment of hepatitis C. We previously demonstrated that ribavirin inhibited glioma cell growth in vitro and in vivo. Based on these results and the fact that no pre-clinical model of ribavirin in AT/RT exists, we decided to investigate the effect of ribavirin on several human AT/RT cell lines (BT12, BT16, and BT37) both in vitro and in vivo. We provide evidence that ribavirin has a significant impact on AT/RT cell growth and increases cell cycle arrest and cell death, potentially through modulation of the eIF4E and/or EZH2 pathways. Interestingly, using scratch wound and transwell Boyden chamber assays, we observed that ribavirin also impairs AT/RT cell migration, invasion, and adhesion. Finally, we demonstrate that ribavirin significantly improves the survival of mice orthotopically implanted with BT12 cells. Our work establishes that ribavirin is effective against AT/RT by decreasing tumoral cell growth and dissemination and could represent a new therapeutic option for children with this deadly disease.
Collapse
Affiliation(s)
- Joshua Casaos
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Sakibul Huq
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Tarik Lott
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Raphael Felder
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - John Choi
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Noah Gorelick
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Michael Peters
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Yuanxuan Xia
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Russell Maxwell
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Tianna Zhao
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Chenchen Ji
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Thomas Simon
- Center for Vascular and Inflammatory Diseases, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Julie Sesen
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA.,INSERM U1037, Centre de Recherche en Cancérologie de Toulouse, CRCT, 31100 Toulouse, France
| | - Sarah J Scotland
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | | | - Jeffrey Rubens
- Pathology Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Eric Raabe
- Pathology Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Charles G Eberhart
- Pathology Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Eric M Jackson
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Henry Brem
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Betty Tyler
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Nicolas Skuli
- Hunterian Neurosurgical Research Laboratory, Neurosurgery Department, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA.,INSERM U1037, Centre de Recherche en Cancérologie de Toulouse, CRCT, 31100 Toulouse, France
| |
Collapse
|
18
|
Clinicopathological and prognostic significance of HMGA2 overexpression in gastric cancer: a meta-analysis. Oncotarget 2017; 8:100478-100489. [PMID: 29245994 PMCID: PMC5725036 DOI: 10.18632/oncotarget.19001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 06/18/2017] [Indexed: 12/29/2022] Open
Abstract
Background High mobility group protein A2 (HMGA2) overexpression has been reported to be closely related to tumor progression [1-4] and indicate significantly worse overall survival in gastric cancer [5-8]. However, a final consensus regarding this issue has not yet been reached. Thus, we conducted a meta-analysis to evaluate the association between HMGA2 expression and prognosis of gastric cancer patients. Methods The Cochrane Library, Embase, PubMed, Web of Science and China Biology Medicine databases were searched to identify eligible literature published prior to September 2016. In the included studies, the level of HMGA2 amplification was evaluated by immunohistochemistry. We performed a meta-analysis, and pooled relative risk (RRs), hazard ratio (HRs), and 95% confidence intervals (CIs) were analyzed using Review Manager 5.3. Results Six studies [5-7, 9-11] involving 712 gastric cancer patients were included and stratified by HMGA2 amplification magnitude. The results of the analysis indicated that higher HMGA2 levels were associated with several clinicopathological parameters and predicted poor prognosis in terms of overall survival (OS). Conclusions The results of the present study indicate that higher HMGA2 levels were significantly associated with TNM stage, lymph node status, vascular invasion, and poor OS in patients with gastric cancer. In conclusion, HMGA2 may serve as a promising prognostic biomarker in gastric cancer.
Collapse
|
19
|
Miyahara H, Yadavilli S, Natsumeda M, Rubens JA, Rodgers L, Kambhampati M, Taylor IC, Kaur H, Asnaghi L, Eberhart CG, Warren KE, Nazarian J, Raabe EH. The dual mTOR kinase inhibitor TAK228 inhibits tumorigenicity and enhances radiosensitization in diffuse intrinsic pontine glioma. Cancer Lett 2017; 400:110-116. [PMID: 28450157 DOI: 10.1016/j.canlet.2017.04.019] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 03/13/2017] [Accepted: 04/19/2017] [Indexed: 11/18/2022]
Abstract
Diffuse intrinsic pontine glioma (DIPG) is an invasive and treatment-refractory pediatric brain tumor. Primary DIPG tumors harbor a number of mutations including alterations in PTEN, AKT, and PI3K and exhibit activation of mammalian Target of Rapamycin Complex 1 and 2 (mTORC1/2). mTORC1/2 regulate protein translation, cell growth, survival, invasion, and metabolism. Pharmacological inhibition of mTORC1 is minimally effective in DIPG. However, the activity of dual TORC kinase inhibitors has not been examined in this tumor type. Nanomolar levels of the mTORC1/2 inhibitor TAK228 reduced expression of p-AKTS473 and p-S6S240/244 and suppressed the growth of DIPG lines JHH-DIPG1, SF7761, and SU-DIPG-XIII. TAK228 induced apoptosis in DIPG cells and cooperated with radiation to further block proliferation and enhance apoptosis. TAK228 monotherapy inhibited the tumorigenicity of a murine orthotopic model of DIPG, more than doubling median survival (p = 0.0017) versus vehicle. We conclude that dual mTOR inhibition is a promising potential candidate for DIPG treatment.
Collapse
Affiliation(s)
- Hiroaki Miyahara
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sridevi Yadavilli
- Research Center for Genetic Medicine, Children's National Health System, Washington, District of Columbia 20010, USA
| | - Manabu Natsumeda
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeffrey A Rubens
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Louis Rodgers
- National Cancer Institute, National Institute of Health, Bethesda, MD 20892, USA
| | - Madhuri Kambhampati
- Research Center for Genetic Medicine, Children's National Health System, Washington, District of Columbia 20010, USA
| | - Isabella C Taylor
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Harpreet Kaur
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laura Asnaghi
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles G Eberhart
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Katherine E Warren
- National Cancer Institute, National Institute of Health, Bethesda, MD 20892, USA
| | - Javad Nazarian
- Research Center for Genetic Medicine, Children's National Health System, Washington, District of Columbia 20010, USA; Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia 20052, USA
| | - Eric H Raabe
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
20
|
Wang X, Lee RS, Alver BH, Haswell JR, Wang S, Mieczkowski J, Drier Y, Gillespie SM, Archer TC, Wu JN, Tzvetkov EP, Troisi EC, Pomeroy SL, Biegel JA, Tolstorukov MY, Bernstein BE, Park PJ, Roberts CWM. SMARCB1-mediated SWI/SNF complex function is essential for enhancer regulation. Nat Genet 2016; 49:289-295. [PMID: 27941797 PMCID: PMC5285474 DOI: 10.1038/ng.3746] [Citation(s) in RCA: 233] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 11/18/2016] [Indexed: 02/07/2023]
Abstract
SMARCB1 (also known as SNF5, INI1, and BAF47), a core subunit of the SWI/SNF (BAF) chromatin-remodeling complex, is inactivated in nearly all pediatric rhabdoid tumors. These aggressive cancers are among the most genomically stable, suggesting an epigenetic mechanism by which SMARCB1 loss drives transformation. Here we show that, despite having indistinguishable mutational landscapes, human rhabdoid tumors exhibit distinct enhancer H3K27ac signatures, which identify remnants of differentiation programs. We show that SMARCB1 is required for the integrity of SWI/SNF complexes and that its loss alters enhancer targeting-markedly impairing SWI/SNF binding to typical enhancers, particularly those required for differentiation, while maintaining SWI/SNF binding at super-enhancers. We show that these retained super-enhancers are essential for rhabdoid tumor survival, including some that are shared by all subtypes, such as SPRY1, and other lineage-specific super-enhancers, such as SOX2 in brain-derived rhabdoid tumors. Taken together, our findings identify a new chromatin-based epigenetic mechanism underlying the tumor-suppressive activity of SMARCB1.
Collapse
Affiliation(s)
- Xiaofeng Wang
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Ryan S Lee
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Burak H Alver
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeffrey R Haswell
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Su Wang
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
| | - Jakub Mieczkowski
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Yotam Drier
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Shawn M Gillespie
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Tenley C Archer
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer N Wu
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Evgeni P Tzvetkov
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Emma C Troisi
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Scott L Pomeroy
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jaclyn A Biegel
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles and Keck School of Medicine at the University of Southern California, Los Angeles, California, USA
| | - Michael Y Tolstorukov
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Bradley E Bernstein
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Peter J Park
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA.,Division of Genetics, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Charles W M Roberts
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA.,Comprehensive Cancer Center and Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
21
|
Atypical teratoid/rhabdoid tumours: clinicopathological characteristics, prognostic factors and outcomes of 22 children from 2010 to 2015 in China. Pathology 2016; 48:555-63. [PMID: 27558890 DOI: 10.1016/j.pathol.2016.05.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 05/08/2016] [Accepted: 05/20/2016] [Indexed: 12/15/2022]
Abstract
Atypical teratoid/rhabdoid tumours (AT/RTs) are rare, highly malignant tumours of the central nervous system (CNS) with poor prognosis that usually affect young children. The aim of this study was to assess the clinicopathological features and prognostic factors of AT/RTs. Here, we describe the clinicopathological and immunohistochemical characteristics, along with the treatments and outcomes, of 22 patients with AT/RTs treated in our hospital from 2010 to 2015. Morphologically, cytoplasmic vacuoles, the most common characteristic in our cases, were observed in 68% of the cases. Similarly, vesicular nuclei were detected in 68% of the cases. However, rhabdoid cells were found in only 59.1% of the cases and were not observed in 40.9% of the cases. Immunohistochemical analysis revealed loss of nuclear INI1 expression in all 22 cases. Age, surgical resection and adjuvant therapy, but not tumour location, were associated with AT/RTs patient prognosis. Our results showed that cells with cytoplasmic vacuoles or with vesicular nuclei are more common than rhabdoid cells in patients with AT/RTs and that a lack of INI1 protein expression is the most useful marker for the differential diagnosis of AT/RTs. Young age is a negative prognostic factor, whereas gross total surgical resection and adjuvant therapy are positive prognostic factors for AT/RT patients.
Collapse
|
22
|
Hashizume R, Zhang A, Mueller S, Prados MD, Lulla RR, Goldman S, Saratsis AM, Mazar AP, Stegh AH, Cheng SY, Horbinski C, Haas-Kogan DA, Sarkaria JN, Waldman T, James CD. Inhibition of DNA damage repair by the CDK4/6 inhibitor palbociclib delays irradiated intracranial atypical teratoid rhabdoid tumor and glioblastoma xenograft regrowth. Neuro Oncol 2016; 18:1519-1528. [PMID: 27370397 DOI: 10.1093/neuonc/now106] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 04/20/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Radiation therapy is the most commonly used postsurgical treatment for primary malignant brain tumors. Consequently, investigating the efficacy of chemotherapeutics combined with radiation for treating malignant brain tumors is of high clinical relevance. In this study, we examined the cyclin-dependent kinase 4/6 inhibitor palbociclib, when used in combination with radiation for treating human atypical teratoid rhabdoid tumor (ATRT) as well as glioblastoma (GBM). METHODS Evaluation of treatment antitumor activity in vitro was based upon results from cell proliferation assays, clonogenicity assays, flow cytometry, and immunocytochemistry for DNA double-strand break repair. Interpretation of treatment antitumor activity in vivo was based upon bioluminescence imaging, animal subject survival analysis, and staining of tumor sections for markers of proliferation and apoptosis. RESULTS For each of the retinoblastoma protein (RB)-proficient tumor models examined (2 ATRTs and 2 GBMs), one or more of the combination therapy regimens significantly (P < .05) outperformed both monotherapies with respect to animal subject survival benefit. Among the combination therapy regimens, concurrent palbociclib and radiation treatment and palbociclib treatment following radiation consistently outperformed the sequence in which radiation followed palbociclib treatment. In vitro investigation revealed that the concurrent use of palbociclib with radiation, as well as palbociclib following radiation, inhibited DNA double-strand break repair and promoted increased tumor cell apoptosis. CONCLUSIONS Our results support further investigation and possible clinical translation of palbociclib as an adjuvant to radiation therapy for patients with malignant brain tumors that retain RB expression.
Collapse
Affiliation(s)
- Rintaro Hashizume
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., A.Z., C.D.J., A.M.S., C.H.); Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., C.D.J.); Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (A.H.S., S.-Y.C.), Northwestern Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.M.S., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois (A.P.M., C.D.J.); Department of Pediatrics, Division of Hematology/Oncology, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois (R.R.L., S.G.); Department of Neurological Surgery, University of California San Francisco, San Francisco, California (S.M., M.D.P.); Department of Pediatrics, University of California San Francisco, San Francisco, California (S.M.); Department of Radiation Oncology, Brigham and Women's Hospital, Boston, Massachusetts (D.A.H.-K.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Lombardi Cancer Center, Georgetown University, Washington, DC (T.W.)
| | - Ali Zhang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., A.Z., C.D.J., A.M.S., C.H.); Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., C.D.J.); Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (A.H.S., S.-Y.C.), Northwestern Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.M.S., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois (A.P.M., C.D.J.); Department of Pediatrics, Division of Hematology/Oncology, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois (R.R.L., S.G.); Department of Neurological Surgery, University of California San Francisco, San Francisco, California (S.M., M.D.P.); Department of Pediatrics, University of California San Francisco, San Francisco, California (S.M.); Department of Radiation Oncology, Brigham and Women's Hospital, Boston, Massachusetts (D.A.H.-K.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Lombardi Cancer Center, Georgetown University, Washington, DC (T.W.)
| | - Sabine Mueller
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., A.Z., C.D.J., A.M.S., C.H.); Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., C.D.J.); Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (A.H.S., S.-Y.C.), Northwestern Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.M.S., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois (A.P.M., C.D.J.); Department of Pediatrics, Division of Hematology/Oncology, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois (R.R.L., S.G.); Department of Neurological Surgery, University of California San Francisco, San Francisco, California (S.M., M.D.P.); Department of Pediatrics, University of California San Francisco, San Francisco, California (S.M.); Department of Radiation Oncology, Brigham and Women's Hospital, Boston, Massachusetts (D.A.H.-K.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Lombardi Cancer Center, Georgetown University, Washington, DC (T.W.)
| | - Michael D Prados
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., A.Z., C.D.J., A.M.S., C.H.); Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., C.D.J.); Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (A.H.S., S.-Y.C.), Northwestern Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.M.S., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois (A.P.M., C.D.J.); Department of Pediatrics, Division of Hematology/Oncology, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois (R.R.L., S.G.); Department of Neurological Surgery, University of California San Francisco, San Francisco, California (S.M., M.D.P.); Department of Pediatrics, University of California San Francisco, San Francisco, California (S.M.); Department of Radiation Oncology, Brigham and Women's Hospital, Boston, Massachusetts (D.A.H.-K.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Lombardi Cancer Center, Georgetown University, Washington, DC (T.W.)
| | - Rishi R Lulla
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., A.Z., C.D.J., A.M.S., C.H.); Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., C.D.J.); Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (A.H.S., S.-Y.C.), Northwestern Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.M.S., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois (A.P.M., C.D.J.); Department of Pediatrics, Division of Hematology/Oncology, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois (R.R.L., S.G.); Department of Neurological Surgery, University of California San Francisco, San Francisco, California (S.M., M.D.P.); Department of Pediatrics, University of California San Francisco, San Francisco, California (S.M.); Department of Radiation Oncology, Brigham and Women's Hospital, Boston, Massachusetts (D.A.H.-K.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Lombardi Cancer Center, Georgetown University, Washington, DC (T.W.)
| | - Stewart Goldman
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., A.Z., C.D.J., A.M.S., C.H.); Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., C.D.J.); Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (A.H.S., S.-Y.C.), Northwestern Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.M.S., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois (A.P.M., C.D.J.); Department of Pediatrics, Division of Hematology/Oncology, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois (R.R.L., S.G.); Department of Neurological Surgery, University of California San Francisco, San Francisco, California (S.M., M.D.P.); Department of Pediatrics, University of California San Francisco, San Francisco, California (S.M.); Department of Radiation Oncology, Brigham and Women's Hospital, Boston, Massachusetts (D.A.H.-K.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Lombardi Cancer Center, Georgetown University, Washington, DC (T.W.)
| | - Amanda M Saratsis
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., A.Z., C.D.J., A.M.S., C.H.); Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., C.D.J.); Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (A.H.S., S.-Y.C.), Northwestern Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.M.S., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois (A.P.M., C.D.J.); Department of Pediatrics, Division of Hematology/Oncology, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois (R.R.L., S.G.); Department of Neurological Surgery, University of California San Francisco, San Francisco, California (S.M., M.D.P.); Department of Pediatrics, University of California San Francisco, San Francisco, California (S.M.); Department of Radiation Oncology, Brigham and Women's Hospital, Boston, Massachusetts (D.A.H.-K.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Lombardi Cancer Center, Georgetown University, Washington, DC (T.W.)
| | - Andrew P Mazar
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., A.Z., C.D.J., A.M.S., C.H.); Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., C.D.J.); Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (A.H.S., S.-Y.C.), Northwestern Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.M.S., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois (A.P.M., C.D.J.); Department of Pediatrics, Division of Hematology/Oncology, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois (R.R.L., S.G.); Department of Neurological Surgery, University of California San Francisco, San Francisco, California (S.M., M.D.P.); Department of Pediatrics, University of California San Francisco, San Francisco, California (S.M.); Department of Radiation Oncology, Brigham and Women's Hospital, Boston, Massachusetts (D.A.H.-K.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Lombardi Cancer Center, Georgetown University, Washington, DC (T.W.)
| | - Alexander H Stegh
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., A.Z., C.D.J., A.M.S., C.H.); Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., C.D.J.); Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (A.H.S., S.-Y.C.), Northwestern Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.M.S., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois (A.P.M., C.D.J.); Department of Pediatrics, Division of Hematology/Oncology, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois (R.R.L., S.G.); Department of Neurological Surgery, University of California San Francisco, San Francisco, California (S.M., M.D.P.); Department of Pediatrics, University of California San Francisco, San Francisco, California (S.M.); Department of Radiation Oncology, Brigham and Women's Hospital, Boston, Massachusetts (D.A.H.-K.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Lombardi Cancer Center, Georgetown University, Washington, DC (T.W.)
| | - Shi-Yuan Cheng
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., A.Z., C.D.J., A.M.S., C.H.); Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., C.D.J.); Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (A.H.S., S.-Y.C.), Northwestern Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.M.S., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois (A.P.M., C.D.J.); Department of Pediatrics, Division of Hematology/Oncology, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois (R.R.L., S.G.); Department of Neurological Surgery, University of California San Francisco, San Francisco, California (S.M., M.D.P.); Department of Pediatrics, University of California San Francisco, San Francisco, California (S.M.); Department of Radiation Oncology, Brigham and Women's Hospital, Boston, Massachusetts (D.A.H.-K.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Lombardi Cancer Center, Georgetown University, Washington, DC (T.W.)
| | - Craig Horbinski
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., A.Z., C.D.J., A.M.S., C.H.); Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., C.D.J.); Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (A.H.S., S.-Y.C.), Northwestern Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.M.S., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois (A.P.M., C.D.J.); Department of Pediatrics, Division of Hematology/Oncology, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois (R.R.L., S.G.); Department of Neurological Surgery, University of California San Francisco, San Francisco, California (S.M., M.D.P.); Department of Pediatrics, University of California San Francisco, San Francisco, California (S.M.); Department of Radiation Oncology, Brigham and Women's Hospital, Boston, Massachusetts (D.A.H.-K.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Lombardi Cancer Center, Georgetown University, Washington, DC (T.W.)
| | - Daphne A Haas-Kogan
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., A.Z., C.D.J., A.M.S., C.H.); Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., C.D.J.); Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (A.H.S., S.-Y.C.), Northwestern Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.M.S., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois (A.P.M., C.D.J.); Department of Pediatrics, Division of Hematology/Oncology, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois (R.R.L., S.G.); Department of Neurological Surgery, University of California San Francisco, San Francisco, California (S.M., M.D.P.); Department of Pediatrics, University of California San Francisco, San Francisco, California (S.M.); Department of Radiation Oncology, Brigham and Women's Hospital, Boston, Massachusetts (D.A.H.-K.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Lombardi Cancer Center, Georgetown University, Washington, DC (T.W.)
| | - Jann N Sarkaria
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., A.Z., C.D.J., A.M.S., C.H.); Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., C.D.J.); Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (A.H.S., S.-Y.C.), Northwestern Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.M.S., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois (A.P.M., C.D.J.); Department of Pediatrics, Division of Hematology/Oncology, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois (R.R.L., S.G.); Department of Neurological Surgery, University of California San Francisco, San Francisco, California (S.M., M.D.P.); Department of Pediatrics, University of California San Francisco, San Francisco, California (S.M.); Department of Radiation Oncology, Brigham and Women's Hospital, Boston, Massachusetts (D.A.H.-K.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Lombardi Cancer Center, Georgetown University, Washington, DC (T.W.)
| | - Todd Waldman
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., A.Z., C.D.J., A.M.S., C.H.); Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., C.D.J.); Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (A.H.S., S.-Y.C.), Northwestern Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.M.S., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois (A.P.M., C.D.J.); Department of Pediatrics, Division of Hematology/Oncology, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois (R.R.L., S.G.); Department of Neurological Surgery, University of California San Francisco, San Francisco, California (S.M., M.D.P.); Department of Pediatrics, University of California San Francisco, San Francisco, California (S.M.); Department of Radiation Oncology, Brigham and Women's Hospital, Boston, Massachusetts (D.A.H.-K.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Lombardi Cancer Center, Georgetown University, Washington, DC (T.W.)
| | - C David James
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., A.Z., C.D.J., A.M.S., C.H.); Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., C.D.J.); Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (A.H.S., S.-Y.C.), Northwestern Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.M.S., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois (R.H., R.R.L., S.G., A.P.M., A.H.S., S.-Y.C., C.H., C.D.J.); Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois (A.P.M., C.D.J.); Department of Pediatrics, Division of Hematology/Oncology, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois (R.R.L., S.G.); Department of Neurological Surgery, University of California San Francisco, San Francisco, California (S.M., M.D.P.); Department of Pediatrics, University of California San Francisco, San Francisco, California (S.M.); Department of Radiation Oncology, Brigham and Women's Hospital, Boston, Massachusetts (D.A.H.-K.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Lombardi Cancer Center, Georgetown University, Washington, DC (T.W.)
| |
Collapse
|
23
|
Kaur H, Ali SZ, Huey L, Hütt-Cabezas M, Taylor I, Mao XG, Weingart M, Chu Q, Rodriguez FJ, Eberhart CG, Raabe EH. The transcriptional modulator HMGA2 promotes stemness and tumorigenicity in glioblastoma. Cancer Lett 2016; 377:55-64. [PMID: 27102002 DOI: 10.1016/j.canlet.2016.04.020] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/08/2016] [Accepted: 04/11/2016] [Indexed: 01/17/2023]
Abstract
Glioblastoma (GBM) contains a population of stem-like cells that promote tumor invasion and resistance to therapy. Identifying and targeting stem cell factors in GBM may lead to the development of more effective therapies. High Mobility Group AT-hook 2 (HMGA2) is a transcriptional modulator that mediates motility and self-renewal in normal and cancer stem cells. We identified increased expression of HMGA2 in the majority of primary human GBM tumors and cell lines compared to normal brain. Additionally, HMGA2 expression was increased in CD133+ GBM neurosphere cells compared to CD133- cells. Targeting HMGA2 with lentiviral short hairpin RNA (shRNA) led to decreased GBM stemness, invasion, and tumorigenicity. Ectopic expression of HMGA2 in GBM cell lines promoted stemness, invasion, and tumorigenicity. Our data suggests that targeting HMGA2 in GBM may be therapeutically beneficial.
Collapse
Affiliation(s)
- Harpreet Kaur
- Division of Neuropathology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA; Division of Pediatric Oncology, Johns Hopkins University, Bloomberg Children's Hospital, Room 11379, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Sabeen Zulfiqar Ali
- Division of Neuropathology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Lauren Huey
- Division of Neuropathology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Marianne Hütt-Cabezas
- Division of Neuropathology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA; Division of Pediatric Oncology, Johns Hopkins University, Bloomberg Children's Hospital, Room 11379, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Isabella Taylor
- Division of Pediatric Oncology, Johns Hopkins University, Bloomberg Children's Hospital, Room 11379, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Xing-Gang Mao
- Division of Neuropathology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Melanie Weingart
- Division of Neuropathology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Qian Chu
- Division of Neuropathology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Fausto J Rodriguez
- Division of Neuropathology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Charles G Eberhart
- Division of Neuropathology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Eric H Raabe
- Division of Neuropathology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA; Division of Pediatric Oncology, Johns Hopkins University, Bloomberg Children's Hospital, Room 11379, 1800 Orleans St, Baltimore, MD 21287, USA.
| |
Collapse
|
24
|
Disrupting NOTCH Slows Diffuse Intrinsic Pontine Glioma Growth, Enhances Radiation Sensitivity, and Shows Combinatorial Efficacy With Bromodomain Inhibition. J Neuropathol Exp Neurol 2015; 74:778-90. [PMID: 26115193 DOI: 10.1097/nen.0000000000000216] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
NOTCH regulates stem cells during normal development and stemlike cells in cancer, but the roles of NOTCH in the lethal pediatric brain tumor diffuse intrinsic pontine glioma (DIPG) remain unknown. Because DIPGs express stem cell factors such as SOX2 and MYCN, we hypothesized that NOTCH activity would be critical for DIPG growth. We determined that primary DIPGs expressed high levels of NOTCH receptors, ligands, and downstream effectors. Treatment of the DIPG cell lines JHH-DIPG1 and SF7761 with the γ-secretase inhibitor MRK003 suppressed the level of the NOTCH effectors HES1, HES4, and HES5; inhibited DIPG growth by 75%; and caused a 3-fold induction of apoptosis. Short hairpin RNAs targeting the canonical NOTCH pathway caused similar effects. Pretreatment of DIPG cells with MRK003 suppressed clonogenic growth by more than 90% and enhanced the efficacy of radiation therapy. The high level of MYCN in DIPG led us to test sequential therapy with the bromodomain inhibitor JQ1 and MRK003, and we found that JQ1 and MRK003 inhibited DIPG growth and induced apoptosis. Together, these results suggest that dual targeting of NOTCH and MYCN in DIPG may be an effective therapeutic strategy in DIPG and that adding a γ-secretase inhibitor during radiation therapy may be efficacious initially or during reirradiation.
Collapse
|
25
|
Cai J, Shen G, Liu S, Meng Q. Downregulation of HMGA2 inhibits cellular proliferation and invasion, improves cellular apoptosis in prostate cancer. Tumour Biol 2015; 37:699-707. [DOI: 10.1007/s13277-015-3853-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 07/28/2015] [Indexed: 10/23/2022] Open
|