151
|
High APLN Expression Predicts Poor Prognosis for Glioma Patients. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8393336. [PMID: 36193059 PMCID: PMC9526648 DOI: 10.1155/2022/8393336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/30/2022]
Abstract
Apelin (APLN) is an endogenous ligand of the G protein-coupled receptor APJ (APLNR). APLN/APLNR system was involved in a variety of pathological and physiological functions, such as tumorigenesis and development. However, its prognostic roles in patients with central nervous system (CNS) cancers remain unknown. The present study was designed to explore the expression profile, prognostic significance, and interaction network of APLN/APLNR by integrating data from Oncomine, GEPIA, LOGpc, STRING, GeneMANIA, and immunohistochemical staining. The results demonstrated that APLN and APLNR mRNA expression were significantly increased in CNS cancers, including both low-grade glioma (LGG) and glioblastoma (GBM), when compared with normal CNS tissues. The high APLN, but not APLNR, expression was significantly correlated with overall survival (OS), recurrence free survival (RFS), and progression free survival (PFS) of LGG patients. However, neither APLN nor APLNR expression was significantly related to prognostic value in terms of OS, disease free interval (DFI), disease specific survival (DSS), or progression free interval (PFI) for GBM patients. Additionally, immunohistochemistry staining confirmed the increased APLN expression in tissues of LGG patients with grade II than grade I. These results showed that an elevated APLN level could predict poor OS, RFS, and PFS for LGG patients, and it could be a promising prognostic biomarker for LGG.
Collapse
|
152
|
Hanna GK, Madany M, Tay ASMS, Edwards LA, Kim S, Michael JS, Nuno M, Thomas T, Li A, Berel D, Black KL, Fan X, Zhang W, Rudnick JD, Wang R, Yu JS. ZEB1 loss increases glioma stem cell tumorigenicity and resistance to chemoradiation. J Neurosurg 2022; 138:1313-1324. [PMID: 36115050 DOI: 10.3171/2022.7.jns22259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 07/15/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Glioblastoma has been known to be resistant to chemotherapy and radiation, whereas the underlying mechanisms of resistance have not been fully elucidated. The authors studied the role of the transcription factor ZEB1 (zinc finger E-box-binding homeobox 1 protein), which is associated with epithelial-mesenchymal transition (EMT) and is central to the stemness of glioblastoma, to determine its role in therapeutic resistance to radiation and chemotherapy. The authors previously demonstrated that ZEB1 is deleted in a majority of glioblastomas. METHODS The authors explored resistance to therapy in the context of ZEB1 loss and overexpression in glioma stem cells (GSCs) and in patient data. RESULTS Patients with ZEB1 loss had a shorter survival time than patients with wild-type ZEB1 in both the high- and low-MGMT groups. Consistent with the clinical data, mice implanted with ZEB1 knockdown GSCs showed shortened survival compared with mice inoculated with nonsilencing control (NS) short-hairpin RNA (shRNA) GSC glioblastoma. ZEB1-deleted GSCs demonstrated increased tumorigenicity with regard to proliferation and invasion. Importantly, GSCs that lose ZEB1 expression develop enhanced resistance to chemotherapy, radiotherapy, and combined chemoradiation. ZEB1 loss may lead to increased HER3 expression through the HER3/Akt pathway associated with this chemoresistance. Conversely, overexpression of ZEB1 in GSCs that are ZEB1 null leads to increased sensitivity to chemoradiation. CONCLUSIONS The study results indicate that ZEB1 loss in cancer stem cells confers resistance to chemoradiation and uncovers a potentially targetable cell surface receptor in these resistant cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Miriam Nuno
- Departments of1Neurosurgery and.,3Department of Biostatics, University of California, Davis, Sacramento, California
| | | | - Aiguo Li
- 4Neuro-Oncology Branch, National Institutes of Health/National Cancer Institute, Bethesda, Maryland; and
| | | | | | - Xuemo Fan
- 5Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles
| | - Wei Zhang
- 4Neuro-Oncology Branch, National Institutes of Health/National Cancer Institute, Bethesda, Maryland; and
| | - Jeremy D Rudnick
- Departments of1Neurosurgery and.,6Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles
| | - Rongfu Wang
- 7USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | | |
Collapse
|
153
|
Zhu Z, Kiang KMY, Li N, Liu J, Zhang P, Jin L, He X, Zhang S, Leung GKK. Folate enzyme MTHFD2 links one-carbon metabolism to unfolded protein response in glioblastoma. Cancer Lett 2022; 549:215903. [PMID: 36089117 DOI: 10.1016/j.canlet.2022.215903] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 11/28/2022]
Abstract
The mitochondrial folate enzyme methylenetetrahydrofolate dehydrogenase/cyclohydrolase (MTHFD2) has shown oncogenic roles in various cancers and may have non-metabolic functions. This study investigated the role of MTHFD2 in glioblastoma pathogenesis. We find that MTHFD2 expression is enriched in gliomas by analysing public databases and clinical specimens. RNA interference (RNAi) and inhibitor of MTHFD2 hamper the proliferation of glioblastoma and induce apoptosis in cell lines, glioma stem-like cells (GSCs) and patient-derived xenografts (PDX). Metabolomic analyses show that MTHFD2 depletion suppresses the central carbon metabolic pathways, including glycolysis, the pentose phosphate pathway (PPP), and the tricarboxylic acid (TCA) cycle. GSEA reveals a novel non-metabolic function of MTHFD2 in association with the unfolded protein response (UPR). MTHFD2 depletion activates the PERK/eIF2α axis which contributes to translation inhibition and apoptosis; these effects are attenuated by a PERK inhibitor. Mechanistically, MTHFD2 may be linked to UPR via the post-transcriptionally regulation of chaperone protein GRP78. In conclusion, MTHFD2 could be a promising therapeutic target for glioblastoma. Besides its canonical role, MTHFD2 may contribute to glioblastoma pathogenesis via UPR, highlighting a newly identified functional link between one-carbon metabolism and cell stress response.
Collapse
Affiliation(s)
- Zhiyuan Zhu
- Department of Functional Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Guangzhou, China; Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Karrie Mei-Yee Kiang
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Ning Li
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Jiaxin Liu
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Pingde Zhang
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Lei Jin
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Xiaozheng He
- Department of Functional Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Guangzhou, China
| | - Shizhong Zhang
- Department of Functional Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Guangzhou, China.
| | - Gilberto Ka-Kit Leung
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China.
| |
Collapse
|
154
|
Wei Y, Chen Q, Huang S, Liu Y, Li Y, Xing Y, Shi D, Xu W, Liu W, Ji Z, Wu B, Chen X, Jiang J. The Interaction between DNMT1 and High-Mannose CD133 Maintains the Slow-Cycling State and Tumorigenic Potential of Glioma Stem Cell. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202216. [PMID: 35798319 PMCID: PMC9475542 DOI: 10.1002/advs.202202216] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Indexed: 05/24/2023]
Abstract
The quiescent/slow-cycling state preserves the self-renewal capacity of cancer stem cells (CSCs) and leads to the therapy resistance of CSCs. The mechanisms maintaining CSCs quiescence remain largely unknown. Here, it is demonstrated that lower expression of MAN1A1 in glioma stem cell (GSC) resulted in the formation of high-mannose type N-glycan on CD133. Furthermore, the high-mannose type N-glycan of CD133 is necessary for its interaction with DNMT1. Activation of p21 and p27 by the CD133-DNMT1 interaction maintains the slow-cycling state of GSC, and promotes chemotherapy resistance and tumorigenesis of GSCs. Elimination of the CD133-DNMT1 interaction by a cell-penetrating peptide or MAN1A1 overexpression inhibits the tumorigenesis of GSCs and increases the sensitivity of GSCs to temozolomide. Analysis of glioma samples reveals that the levels of high-mannose type N-glycan are correlated with glioma recurrence. Collectively, the high mannose CD133-DNMT1 interaction maintains the slow-cycling state and tumorigenic potential of GSC, providing a potential strategy to eliminate quiescent GSCs.
Collapse
Affiliation(s)
- Yuanyan Wei
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Qihang Chen
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Sijing Huang
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Yingchao Liu
- Department of NeurosurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandong250021P. R. China
| | - Yinan Li
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Yang Xing
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Danfang Shi
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Wenlong Xu
- Division of NeurosurgeryZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Weitao Liu
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Zhi Ji
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Bingrui Wu
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Xiaoning Chen
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| | - Jianhai Jiang
- NHC Key Laboratory of Glycoconjuates ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200032P. R. China
| |
Collapse
|
155
|
CCAAT/Enhancer-Binding Protein Delta Regulates Glioblastoma Survival through Catalase-Mediated Hydrogen Peroxide Clearance. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4081380. [PMID: 36035213 PMCID: PMC9411925 DOI: 10.1155/2022/4081380] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 02/18/2022] [Accepted: 07/21/2022] [Indexed: 12/03/2022]
Abstract
It has long been documented that cancer cells show increased and persistent oxidative stress due to increased reactive oxygen species (ROS), which is necessary for their increased proliferative rate. Due to the high levels of ROS, cancer cells also stimulate the antioxidant system, which includes the enzymes superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPX), to eliminate ROS. However, overexpressed antioxidant enzymes often lead to drug resistance and therapeutic failure. Glioblastoma (GBM) is the most aggressive brain tumor and has the poorest prognosis. The transcription factor CCAAT/enhancer-binding protein delta (CEBPD) is highly expressed in GBM and correlates with drug resistance, prompting us to elucidate its role in GBM cell survival. In this study, we first demonstrated that loss of CEBPD significantly inhibited GBM cell viability and increased cell apoptosis. Furthermore, the expression of CAT was attenuated through promoter regulation following CEBPD knockdown, accelerating intracellular hydrogen peroxide (H2O2) accumulation. In addition, mitochondrial function was impaired in CEBPD knockdown cells. Together, we revealed the mechanism by which CEBPD-mediated CAT expression regulates H2O2 clearance for GBM cell survival.
Collapse
|
156
|
Acidosis induces RIPK1-dependent death of glioblastoma stem cells via acid-sensing ion channel 1a. Cell Death Dis 2022; 13:702. [PMID: 35961983 PMCID: PMC9374719 DOI: 10.1038/s41419-022-05139-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 01/21/2023]
Abstract
Eliciting regulated cell death, like necroptosis, is a potential cancer treatment. However, pathways eliciting necroptosis are poorly understood. It has been reported that prolonged activation of acid-sensing ion channel 1a (ASIC1a) induces necroptosis in mouse neurons. Glioblastoma stem cells (GSCs) also express functional ASIC1a, but whether prolonged activation of ASIC1a induces necroptosis in GSCs is unknown. Here we used a tumorsphere formation assay to show that slight acidosis (pH 6.6) induces necrotic cell death in a manner that was sensitive to the necroptosis inhibitor Nec-1 and to the ASIC1a antagonist PcTx1. In addition, genetic knockout of ASIC1a rendered GSCs resistant to acid-induced reduction in tumorsphere formation, while the ASIC1 agonist MitTx1 reduced tumorsphere formation also at neutral pH. Finally, a 20 amino acid fragment of the ASIC1 C-terminus, thought to interact with the necroptosis kinase RIPK1, was sufficient to reduce the formation of tumorspheres. Meanwhile, the genetic knockout of MLKL, the executive protein in the necroptosis cascade, did not prevent a reduction in tumor sphere formation, suggesting that ASIC1a induced an alternative cell death pathway. These findings demonstrate that ASIC1a is a death receptor on GSCs that induces cell death during prolonged acidosis. We propose that this pathway shapes the evolution of a tumor in its acidic microenvironment and that pharmacological activation of ASIC1a might be a potential new strategy in tumor therapy.
Collapse
|
157
|
Guo G, Gong K, Beckley N, Zhang Y, Yang X, Chkheidze R, Hatanpaa KJ, Garzon-Muvdi T, Koduru P, Nayab A, Jenks J, Sathe AA, Liu Y, Xing C, Wu SY, Chiang CM, Mukherjee B, Burma S, Wohlfeld B, Patel T, Mickey B, Abdullah K, Youssef M, Pan E, Gerber DE, Tian S, Sarkaria JN, McBrayer SK, Zhao D, Habib AA. EGFR ligand shifts the role of EGFR from oncogene to tumour suppressor in EGFR-amplified glioblastoma by suppressing invasion through BIN3 upregulation. Nat Cell Biol 2022; 24:1291-1305. [PMID: 35915159 PMCID: PMC9389625 DOI: 10.1038/s41556-022-00962-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 06/14/2022] [Indexed: 02/03/2023]
Abstract
The epidermal growth factor receptor (EGFR) is a prime oncogene that is frequently amplified in glioblastomas. Here we demonstrate a new tumour-suppressive function of EGFR in EGFR-amplified glioblastomas regulated by EGFR ligands. Constitutive EGFR signalling promotes invasion via activation of a TAB1-TAK1-NF-κB-EMP1 pathway, resulting in large tumours and decreased survival in orthotopic models. Ligand-activated EGFR promotes proliferation and surprisingly suppresses invasion by upregulating BIN3, which inhibits a DOCK7-regulated Rho GTPase pathway, resulting in small hyperproliferating non-invasive tumours and improved survival. Data from The Cancer Genome Atlas reveal that in EGFR-amplified glioblastomas, a low level of EGFR ligands confers a worse prognosis, whereas a high level of EGFR ligands confers an improved prognosis. Thus, increased EGFR ligand levels shift the role of EGFR from oncogene to tumour suppressor in EGFR-amplified glioblastomas by suppressing invasion. The tumour-suppressive function of EGFR can be activated therapeutically using tofacitinib, which suppresses invasion by increasing EGFR ligand levels and upregulating BIN3.
Collapse
Affiliation(s)
- Gao Guo
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ke Gong
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, School of Basic Medical Sciences, Taikang Medical School, Wuhan University, Wuhan, China
| | - Nicole Beckley
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yue Zhang
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaoyao Yang
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rati Chkheidze
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kimmo J Hatanpaa
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tomas Garzon-Muvdi
- Department of Neurosurgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Prasad Koduru
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Arifa Nayab
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jennifer Jenks
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Adwait Amod Sathe
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yan Liu
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chao Xing
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shwu-Yuan Wu
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pharamacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Cheng-Ming Chiang
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pharamacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bipasha Mukherjee
- Department of Neurosurgery, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Sandeep Burma
- Department of Neurosurgery, University of Texas Health San Antonio, San Antonio, TX, USA
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Bryan Wohlfeld
- Department of Neurosurgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Toral Patel
- Department of Neurosurgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bruce Mickey
- Department of Neurosurgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kalil Abdullah
- Department of Neurosurgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michael Youssef
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Edward Pan
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - David E Gerber
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Hematology-Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shulan Tian
- Department of Quantitative Heath Sciences, Mayo Clinic, Rochester, MN, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Samuel K McBrayer
- Department of Pediatrics and Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dawen Zhao
- Departments of Biomedical Engineering and Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Amyn A Habib
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- VA North Texas Health Care System, Dallas, TX, USA.
| |
Collapse
|
158
|
Ntafoulis I, Koolen SLW, Leenstra S, Lamfers MLM. Drug Repurposing, a Fast-Track Approach to Develop Effective Treatments for Glioblastoma. Cancers (Basel) 2022; 14:3705. [PMID: 35954371 PMCID: PMC9367381 DOI: 10.3390/cancers14153705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 12/10/2022] Open
Abstract
Glioblastoma (GBM) remains one of the most difficult tumors to treat. The mean overall survival rate of 15 months and the 5-year survival rate of 5% have not significantly changed for almost 2 decades. Despite progress in understanding the pathophysiology of the disease, no new effective treatments to combine with radiation therapy after surgical tumor debulking have become available since the introduction of temozolomide in 1999. One of the main reasons for this is the scarcity of compounds that cross the blood-brain barrier (BBB) and reach the brain tumor tissue in therapeutically effective concentrations. In this review, we focus on the role of the BBB and its importance in developing brain tumor treatments. Moreover, we discuss drug repurposing, a drug discovery approach to identify potential effective candidates with optimal pharmacokinetic profiles for central nervous system (CNS) penetration and that allows rapid implementation in clinical trials. Additionally, we provide an overview of repurposed candidate drug currently being investigated in GBM at the preclinical and clinical levels. Finally, we highlight the importance of phase 0 trials to confirm tumor drug exposure and we discuss emerging drug delivery technologies as an alternative route to maximize therapeutic efficacy of repurposed candidate drug.
Collapse
Affiliation(s)
- Ioannis Ntafoulis
- Brain Tumor Center, Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (I.N.); (S.L.)
| | - Stijn L. W. Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands;
- Department of Hospital Pharmacy, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Sieger Leenstra
- Brain Tumor Center, Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (I.N.); (S.L.)
| | - Martine L. M. Lamfers
- Brain Tumor Center, Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (I.N.); (S.L.)
| |
Collapse
|
159
|
Park S, Avera AD, Kim Y. BIOMANUFACTURING OF GLIOBLASTOMA ORGANOIDS EXHIBITING HIERARCHICAL AND SPATIALLY ORGANIZED TUMOR MICROENVIRONMENT VIA TRANSDIFFERENTIATION. Biotechnol Bioeng 2022; 119:3252-3274. [DOI: 10.1002/bit.28191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/14/2022] [Accepted: 07/20/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Seungjo Park
- Department of Chemical and Biological EngineeringThe University of AlabamaTuscaloosaAlabama
| | - Alexandra D. Avera
- Department of Chemical and Biological EngineeringThe University of AlabamaTuscaloosaAlabama
| | - Yonghyun Kim
- Department of Chemical and Biological EngineeringThe University of AlabamaTuscaloosaAlabama
| |
Collapse
|
160
|
Liang L, Cui R, Zhong S, Wang Z, He Z, Duan H, Guo X, Lu J, Hu H, Li C, Yu C, Yu Y, Guo C, Mou Y. Analysis of the potential role of photocurable hydrogel in patient-derived glioblastoma organoid culture through RNA sequencing. Biomater Sci 2022; 10:4902-4914. [PMID: 35861413 DOI: 10.1039/d2bm00589a] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Patient-derived glioblastoma organoid (GBO) growth in hydrogels recapitulates key features of parental tumors, making GBOs a useful tool for fundamental research on cancer biology and offer deeper insight into the development of innovative therapeutic strategies for cancer treatment. Matrigel as a natural hydrogel has been widely used for 3D culture in most tumor organoid studies, but the volatility in its biochemical and biophysical properties makes it difficult to be further applied in GBO cultures. Thus, several kinds of biomimetic hydrogels from synthetic or biological polymers have been developed for tumor organoid growth. Here, we innovatively utilize a photocurable hydrogel-based biomimetic instructive system containing gelatin methacryloyl (GelMA) mixed with a hyaluronic acid (HA) hydrogel as a scaffold for generating GBOs. Furthermore, we evaluated the GBO biological properties at the transcriptome level, which showed that GBOs cultured with this hydrogel retain the expression profile of key neurodevelopmental markers, driving mutations and alternative splicing of parental tumors. Notably, GBOs cultured with the photocurable hydrogel may provide a platform for precision cancer medicine, bridging the gap between basic research and clinical application. Although significant challenges remain, biomimetic hydrogels can provide an exceptional window for the construction of tumor organoids to ensure the accuracy of the research and clinical data.
Collapse
Affiliation(s)
- Lun Liang
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China.
| | - Run Cui
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China.
| | - Sheng Zhong
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China.
| | - Zhenning Wang
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China.
| | - Zhenqiang He
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China.
| | - Hao Duan
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China.
| | - Xiaoyu Guo
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China.
| | - Jie Lu
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China.
| | - Hongrong Hu
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China.
| | - Chang Li
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China.
| | - Chengwei Yu
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China.
| | - Yanjiao Yu
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China.
| | - Chengcheng Guo
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China.
| | - Yonggao Mou
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510000, China.
| |
Collapse
|
161
|
Frtús A, Smolková B, Uzhytchak M, Lunova M, Jirsa M, Henry SJW, Dejneka A, Stephanopoulos N, Lunov O. The interactions between DNA nanostructures and cells: A critical overview from a cell biology perspective. Acta Biomater 2022; 146:10-22. [PMID: 35523414 PMCID: PMC9590281 DOI: 10.1016/j.actbio.2022.04.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 11/18/2022]
Abstract
DNA nanotechnology has yielded remarkable advances in composite materials with diverse applications in biomedicine. The specificity and predictability of building 3D structures at the nanometer scale make DNA nanotechnology a promising tool for uses in biosensing, drug delivery, cell modulation, and bioimaging. However, for successful translation of DNA nanostructures to real-world applications, it is crucial to understand how they interact with living cells, and the consequences of such interactions. In this review, we summarize the current state of knowledge on the interactions of DNA nanostructures with cells. We identify key challenges, from a cell biology perspective, that influence progress towards the clinical translation of DNA nanostructures. We close by providing an outlook on what questions must be addressed to accelerate the clinical translation of DNA nanostructures. STATEMENT OF SIGNIFICANCE: Self-assembled DNA nanostructures (DNs) offers unique opportunities to overcome persistent challenges in the nanobiotechnology field. However, the interactions between engineered DNs and living cells are still not well defined. Critical systematization of current cellular models and biological responses triggered by DNs is a crucial foundation for the successful clinical translation of DNA nanostructures. Moreover, such an analysis will identify the pitfalls and challenges that are present in the field, and provide a basis for overcoming those challenges.
Collapse
Affiliation(s)
- Adam Frtús
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic
| | - Barbora Smolková
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic
| | - Mariia Uzhytchak
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic
| | - Mariia Lunova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic; Institute for Clinical & Experimental Medicine (IKEM), Prague, 14021, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), Prague, 14021, Czech Republic
| | - Skylar J W Henry
- School of Molecular Sciences, Arizona State University, Tempe, AZ, 85281, United States; Biodesign Center for Molecular Design and Biomimetics, Arizona State University, Tempe, AZ 85281, United States
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic
| | - Nicholas Stephanopoulos
- School of Molecular Sciences, Arizona State University, Tempe, AZ, 85281, United States; Biodesign Center for Molecular Design and Biomimetics, Arizona State University, Tempe, AZ 85281, United States.
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic.
| |
Collapse
|
162
|
Trastulla L, Noorbakhsh J, Vazquez F, McFarland J, Iorio F. Computational estimation of quality and clinical relevance of cancer cell lines. Mol Syst Biol 2022; 18:e11017. [PMID: 35822563 PMCID: PMC9277610 DOI: 10.15252/msb.202211017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 12/12/2022] Open
Abstract
Immortal cancer cell lines (CCLs) are the most widely used system for investigating cancer biology and for the preclinical development of oncology therapies. Pharmacogenomic and genome-wide editing screenings have facilitated the discovery of clinically relevant gene-drug interactions and novel therapeutic targets via large panels of extensively characterised CCLs. However, tailoring pharmacological strategies in a precision medicine context requires bridging the existing gaps between tumours and in vitro models. Indeed, intrinsic limitations of CCLs such as misidentification, the absence of tumour microenvironment and genetic drift have highlighted the need to identify the most faithful CCLs for each primary tumour while addressing their heterogeneity, with the development of new models where necessary. Here, we discuss the most significant limitations of CCLs in representing patient features, and we review computational methods aiming at systematically evaluating the suitability of CCLs as tumour proxies and identifying the best patient representative in vitro models. Additionally, we provide an overview of the applications of these methods to more complex models and discuss future machine-learning-based directions that could resolve some of the arising discrepancies.
Collapse
Affiliation(s)
| | - Javad Noorbakhsh
- Broad Institute of MIT and HarvardCambridgeMAUSA
- Present address:
Kojin TherapeuticsBostonMAUSA
| | - Francisca Vazquez
- Broad Institute of MIT and HarvardCambridgeMAUSA
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMAUSA
| | | | | |
Collapse
|
163
|
Nakod PS, Kondapaneni RV, Edney B, Kim Y, Rao SS. The impact of temozolomide and lonafarnib on the stemness marker expression of glioblastoma cells in multicellular spheroids. Biotechnol Prog 2022; 38:e3284. [PMID: 35768943 DOI: 10.1002/btpr.3284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 11/10/2022]
Abstract
Glioblastoma multiforme (GBM) is a highly malignant brain tumor with poor prognosis. The GBM microenvironment is highly heterogeneous and is composed of many cell types including astrocytes and endothelial cells (ECs) along with tumor cells, which are responsible for heightened resistance to standard chemotherapeutic drugs such as Temozolomide (TMZ). Here, we investigated how drug treatments impact stemness marker expression of GBM cells in multicellular tumor spheroid (MCTS) models. Co- and tri-culture MCTS constructed using U87-MG GBM cells, astrocytes and/or ECs were cultured for 7 days. At day 7, 5 μM lonafarnib (LNF), 100 μM TMZ, or combination of 5 μM LNF + 100 μM TMZ was added and the MCTS were cultured for an additional 48 h. We assessed the spheroid sizes and expression of stemness markers- NESTIN, SOX2, CD133, NANOG, and OCT4- through qRT-PCR and immunostaining. Following 48 h treatment with LNF, TMZ or their combination (LNF+TMZ), the spheroid sizes decreased compared to the untreated control. We also observed that the expression of most of the stemness markers significantly increased in the LNF+TMZ treated condition as compared to the untreated condition. These results indicate that drug treatment can influence the stemness marker expression of GBM cells in MCTS models and these aspects must be considered while evaluating therapies. In future, by incorporating other relevant cell types, we can further our understanding of their crosstalk, eventually leading to the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Pinaki S Nakod
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, USA
| | - Raghu Vamsi Kondapaneni
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, USA
| | - Brandon Edney
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Yonghyun Kim
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, USA
| | - Shreyas S Rao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, USA
| |
Collapse
|
164
|
Boccellato C, Rehm M. Glioblastoma, from disease understanding towards optimal cell-based in vitro models. Cell Oncol (Dordr) 2022; 45:527-541. [PMID: 35763242 PMCID: PMC9424171 DOI: 10.1007/s13402-022-00684-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2022] [Indexed: 11/24/2022] Open
Abstract
Abstract
Background
Glioblastoma (GBM) patients are notoriously difficult to treat and ultimately all succumb to disease. This unfortunate scenario motivates research into better characterizing and understanding this disease, and into developing novel research tools by which potential novel therapeutics and treatment options initially can be evaluated pre-clinically. Here, we provide a concise overview of glioblastoma epidemiology, disease classification, the challenges faced in the treatment of glioblastoma and current novel treatment strategies. From this, we lead into a description and assessment of advanced cell-based models that aim to narrow the gap between pre-clinical and clinical studies. Such invitro models are required to deliver reliable and meaningful data for the development and pre-validation of novel therapeutics and treatments.
Conclusions
The toolbox for GBM cell-based models has expanded substantially, with the possibility of 3D printing tumour tissues and thereby replicating invivo tissue architectures now looming on the horizon. A comparison of experimental cell-based model systems and techniques highlights advantages and drawbacks of the various tools available, based on which cell-based models and experimental approaches best suited to address a diversity of research questions in the glioblastoma research field can be selected.
Collapse
Affiliation(s)
- Chiara Boccellato
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany.
| | - Markus Rehm
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany.
- Stuttgart Research Center Systems Biology, University of Stuttgart, 70569, Stuttgart, Germany.
| |
Collapse
|
165
|
Cisplatin-Resistant CD44+ Lung Cancer Cells Are Sensitive to Auger Electrons. Int J Mol Sci 2022; 23:ijms23137131. [PMID: 35806135 PMCID: PMC9266901 DOI: 10.3390/ijms23137131] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer stem cells (CSCs) are resistant to conventional therapy and present a major clinical challenge since they are responsible for the relapse of many cancers, including non-small cell lung cancer (NSCLC). Hence, future successful therapy should also eradicate CSCs. Auger electrons have demonstrated promising therapeutic potential and can induce DNA damage while sparing surrounding cells. Here, we sort primary patient-derived NSCLC cells based on their expression of the CSC-marker CD44 and investigate the effects of cisplatin and a thymidine analog (deoxyuridine) labeled with an Auger electron emitter (125I). We show that the CD44+ populations are more resistant to cisplatin than the CD44− populations. Interestingly, incubation with the thymidine analog 5-[125I]iodo-2′-deoxyuridine ([125I]I-UdR) induces equal DNA damage, G2/M cell cycle arrest, and apoptosis in the CD44− and CD44+ populations. Our results suggest that Auger electron emitters can also eradicate resistant lung cancer CD44+ populations.
Collapse
|
166
|
Koh L, Novera W, Lim SW, Chong YK, Pang QY, Low D, Ang BT, Tang C. Integrative multi-omics approach to targeted therapy for glioblastoma. Pharmacol Res 2022; 182:106308. [PMID: 35714825 DOI: 10.1016/j.phrs.2022.106308] [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: 03/30/2022] [Revised: 05/19/2022] [Accepted: 06/10/2022] [Indexed: 11/30/2022]
Abstract
This review describes recent technological advances applied to glioblastoma (GBM), a brain tumor with dismal prognosis. International consortial efforts suggest the presence of molecular subtypes within histologically identical GBM tumors. This emphasizes that future treatment decisions should no longer be made based solely on morphological analyses, but must now take into consideration such molecular and cellular heterogeneity. The use of single-cell technologies has advanced our understanding and assignation of functional subtypes revealing therapeutic vulnerabilities. Our team has developed stratification approaches in the past few years, and we have been able to identify patient cohorts enriched for various signaling pathways. Importantly, our Glioportal brain tumor resource has been established under the National Neuroscience Institute Tissue Bank in 2021. This resource offers preclinical capability to validate working hypotheses established from patient clinical datasets. This review highlights recent developments with the ultimate goal of assigning functional meaning to molecular subtypes, revealing therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Lynnette Koh
- Department of Research, National Neuroscience Institute, Singapore.
| | - Wisna Novera
- Department of Research, National Neuroscience Institute, Singapore
| | - See Wee Lim
- Department of Research, National Neuroscience Institute, Singapore
| | - Yuk Kien Chong
- Department of Research, National Neuroscience Institute, Singapore
| | - Qing You Pang
- Department of Research, National Neuroscience Institute, Singapore
| | - David Low
- Department of Neurosurgery, National Neuroscience Institute, Singapore; Duke-National University of Singapore, Singapore
| | - Beng Ti Ang
- Department of Neurosurgery, National Neuroscience Institute, Singapore; Duke-National University of Singapore, Singapore
| | - Carol Tang
- Department of Research, National Neuroscience Institute, Singapore; Duke-National University of Singapore, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore.
| |
Collapse
|
167
|
Haddock S, Alban TJ, Turcan Ş, Husic H, Rosiek E, Ma X, Wang Y, Bale T, Desrichard A, Makarov V, Monette S, Wu W, Gardner R, Manova K, Boire A, Chan TA. Phenotypic and molecular states of IDH1 mutation-induced CD24-positive glioma stem-like cells. Neoplasia 2022; 28:100790. [PMID: 35398668 PMCID: PMC9014446 DOI: 10.1016/j.neo.2022.100790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/19/2022] [Accepted: 03/21/2022] [Indexed: 12/15/2022]
Abstract
Mutations in IDH1 and IDH2 drive the development of gliomas. These genetic alterations promote tumor cell renewal, disrupt differentiation states, and induce stem-like properties. Understanding how this phenotypic reprogramming occurs remains an area of high interest in glioma research. Previously, we showed that IDH mutation results in the development of a CD24-positive cell population in gliomas. Here, we demonstrate that this CD24-positive population possesses striking stem-like properties at the molecular and phenotypic levels. We found that CD24 expression is associated with stem-like features in IDH-mutant tumors, a patient-derived gliomasphere model, and a neural stem cell model of IDH1-mutant glioma. In orthotopic models, CD24-positive cells display enhanced tumor initiating potency compared to CD24-negative cells. Furthermore, CD24 knockdown results in changes in cell viability, proliferation rate, and gene expression that closely resemble a CD24-negative phenotype. Our data demonstrate that induction of a CD24-positive population is one mechanism by which IDH-mutant tumors acquire stem-like properties. These findings have significant implications for our understanding of the molecular underpinnings of IDH-mutant gliomas.
Collapse
|
168
|
Comprehensive Landscape of STEAP Family Members Expression in Human Cancers: Unraveling the Potential Usefulness in Clinical Practice Using Integrated Bioinformatics Analysis. DATA 2022. [DOI: 10.3390/data7050064] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The human Six-Transmembrane Epithelial Antigen of the Prostate (STEAP) family comprises STEAP1-4. Several studies have pointed out STEAP proteins as putative biomarkers, as well as therapeutic targets in several types of human cancers, particularly in prostate cancer. However, the relationships and significance of the expression pattern of STEAP1-4 in cancer cases are barely known. Herein, the Oncomine database and cBioPortal platform were selected to predict the differential expression levels of STEAP members and clinical prognosis. The most common expression pattern observed was the combination of the over- and underexpression of distinct STEAP genes, but cervical and gastric cancer and lymphoma showed overexpression of all STEAP genes. It was also found that STEAP genes’ expression levels were already deregulated in benign lesions. Regarding the prognostic value, it was found that STEAP1 (prostate), STEAP2 (brain and central nervous system), STEAP3 (kidney, leukemia and testicular) and STEAP4 (bladder, cervical, gastric) overexpression correlate with lower patient survival rate. However, in prostate cancer, overexpression of the STEAP4 gene was correlated with a higher survival rate. Overall, this study first showed that the expression levels of STEAP genes are highly variable in human cancers, which may be related to different patients’ outcomes.
Collapse
|
169
|
Da-Veiga MA, Rogister B, Lombard A, Neirinckx V, Piette C. Glioma Stem Cells in Pediatric High-Grade Gliomas: From Current Knowledge to Future Perspectives. Cancers (Basel) 2022; 14:cancers14092296. [PMID: 35565425 PMCID: PMC9099564 DOI: 10.3390/cancers14092296] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Pediatric high-grade glioma (pHGG) has a dismal prognosis in which the younger the patient, the more restricted the treatments are, in regard to the incurred risks. Current therapies destroy many tumor cells but fail to target the highly malignant glioma stem cells (GSCs) that adapt quickly to give rise to recurring, treatment-resistant cancers. Despite a lack of consensus around an efficient detection, GSCs are well described in adult brain tumors but remain poorly investigated in pediatric cases, mostly due to their rarity. An improved knowledge about GSC roles in pediatric tumors would provide a key leverage towards the elimination of this sub-population, based on targeted treatments. The aim of this review is to sum up the state of art about GSCs in pHGG. Abstract In children, high-grade gliomas (HGG) and diffuse midline gliomas (DMG) account for a high proportion of death due to cancer. Glioma stem cells (GSCs) are tumor cells in a specific state defined by a tumor-initiating capacity following serial transplantation, self-renewal, and an ability to recapitulate tumor heterogeneity. Their presence was demonstrated several decades ago in adult glioblastoma (GBM), and more recently in pediatric HGG and DMG. In adults, we and others have previously suggested that GSCs nest into the subventricular zone (SVZ), a neurogenic niche, where, among others, they find shelter from therapy. Both bench and bedside evidence strongly indicate a role for the GSCs and the SVZ in GBM progression, fostering the development of innovative targeting treatments. Such new therapeutic approaches are of particular interest in infants, in whom standard therapies are often limited due to the risk of late effects. The aim of this review is to describe current knowledge about GSCs in pediatric HGG and DMG, i.e., their characterization, the models that apply to their development and maintenance, the specific signaling pathways that may underlie their activity, and their specific interactions with neurogenic niches. Finally, we will discuss the clinical relevance of these observations and the therapeutic advantages of targeting the SVZ and/or the GSCs in infants.
Collapse
Affiliation(s)
- Marc-Antoine Da-Veiga
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000 Liège, Belgium; (M.-A.D.-V.); (B.R.); (A.L.); (V.N.)
| | - Bernard Rogister
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000 Liège, Belgium; (M.-A.D.-V.); (B.R.); (A.L.); (V.N.)
- Department of Neurology, CHU of Liège, 4000 Liège, Belgium
| | - Arnaud Lombard
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000 Liège, Belgium; (M.-A.D.-V.); (B.R.); (A.L.); (V.N.)
- Department of Neurosurgery, CHU of Liège, 4000 Liège, Belgium
| | - Virginie Neirinckx
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000 Liège, Belgium; (M.-A.D.-V.); (B.R.); (A.L.); (V.N.)
| | - Caroline Piette
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000 Liège, Belgium; (M.-A.D.-V.); (B.R.); (A.L.); (V.N.)
- Department of Pediatrics, Division of Hematology-Oncology, CHU Liège, 4000 Liège, Belgium
- Correspondence:
| |
Collapse
|
170
|
Almstedt E, Rosén E, Gloger M, Stockgard R, Hekmati N, Koltowska K, Krona C, Nelander S. Real-time evaluation of glioblastoma growth in patient-specific zebrafish xenografts. Neuro Oncol 2022; 24:726-738. [PMID: 34919147 PMCID: PMC9071311 DOI: 10.1093/neuonc/noab264] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Patient-derived xenograft (PDX) models of glioblastoma (GBM) are a central tool for neuro-oncology research and drug development, enabling the detection of patient-specific differences in growth, and in vivo drug response. However, existing PDX models are not well suited for large-scale or automated studies. Thus, here, we investigate if a fast zebrafish-based PDX model, supported by longitudinal, AI-driven image analysis, can recapitulate key aspects of glioblastoma growth and enable case-comparative drug testing. METHODS We engrafted 11 GFP-tagged patient-derived GBM IDH wild-type cell cultures (PDCs) into 1-day-old zebrafish embryos, and monitored fish with 96-well live microscopy and convolutional neural network analysis. Using light-sheet imaging of whole embryos, we analyzed further the invasive growth of tumor cells. RESULTS Our pipeline enables automatic and robust longitudinal observation of tumor growth and survival of individual fish. The 11 PDCs expressed growth, invasion and survival heterogeneity, and tumor initiation correlated strongly with matched mouse PDX counterparts (Spearman R = 0.89, p < 0.001). Three PDCs showed a high degree of association between grafted tumor cells and host blood vessels, suggesting a perivascular invasion phenotype. In vivo evaluation of the drug marizomib, currently in clinical trials for GBM, showed an effect on fish survival corresponding to PDC in vitro and in vivo marizomib sensitivity. CONCLUSIONS Zebrafish xenografts of GBM, monitored by AI methods in an automated process, present a scalable alternative to mouse xenograft models for the study of glioblastoma tumor initiation, growth, and invasion, applicable to patient-specific drug evaluation.
Collapse
Affiliation(s)
- Elin Almstedt
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Emil Rosén
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Marleen Gloger
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Rebecka Stockgard
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Neda Hekmati
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Katarzyna Koltowska
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Cecilia Krona
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Sven Nelander
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
171
|
Cho CF, Farquhar CE, Fadzen CM, Scott B, Zhuang P, von Spreckelsen N, Loas A, Hartrampf N, Pentelute BL, Lawler SE. A Tumor-Homing Peptide Platform Enhances Drug Solubility, Improves Blood-Brain Barrier Permeability and Targets Glioblastoma. Cancers (Basel) 2022; 14:cancers14092207. [PMID: 35565337 PMCID: PMC9103942 DOI: 10.3390/cancers14092207] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Glioblastoma (GBM) is a fatal and incurable brain cancer, and current treatment options have demonstrated limited success. Here, we describe the use of a dg-Bcan-Targeting-Peptide (BTP-7) that has BBB-penetrating properties and targets GBM. Conjugation of BTP-7 to an insoluble anti-cancer drug, camptothecin (CPT), improves drug solubility in aqueous solution, retains drug efficacy against patient-derived GBM stem cells (GSC), enhances BBB permeability, and enables therapeutic targeting to intracranial patient-derived GBM xenograft in mice, leading to higher toxicity in GBM cells compared to normal brain tissues and prolonged animal survival. This work demonstrates a proof-of-concept for BTP-7 as a tumor-targeting peptide for therapeutic delivery to GBM. Abstract Background: Glioblastoma (GBM) is the most common and deadliest malignant primary brain tumor, contributing significant morbidity and mortality among patients. As current standard-of-care demonstrates limited success, the development of new efficacious GBM therapeutics is urgently needed. Major challenges in advancing GBM chemotherapy include poor bioavailability, lack of tumor selectivity leading to undesired side effects, poor permeability across the blood–brain barrier (BBB), and extensive intratumoral heterogeneity. Methods: We have previously identified a small, soluble peptide (BTP-7) that is able to cross the BBB and target the human GBM extracellular matrix (ECM). Here, we covalently attached BTP-7 to an insoluble anti-cancer drug, camptothecin (CPT). Results: We demonstrate that conjugation of BTP-7 to CPT improves drug solubility in aqueous solution, retains drug efficacy against patient-derived GBM stem cells (GSC), enhances BBB permeability, and enables therapeutic targeting to intracranial GBM, leading to higher toxicity in GBM cells compared to normal brain tissues, and ultimately prolongs survival in mice bearing intracranial patient-derived GBM xenograft. Conclusion: BTP-7 is a new modality that opens the door to possibilities for GBM-targeted therapeutic approaches.
Collapse
Affiliation(s)
- Choi-Fong Cho
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (B.S.); (P.Z.); (N.v.S.); (S.E.L.)
- Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA;
- Correspondence:
| | - Charlotte E. Farquhar
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; (C.E.F.); (C.M.F.); (A.L.); (N.H.)
| | - Colin M. Fadzen
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; (C.E.F.); (C.M.F.); (A.L.); (N.H.)
| | - Benjamin Scott
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (B.S.); (P.Z.); (N.v.S.); (S.E.L.)
| | - Pei Zhuang
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (B.S.); (P.Z.); (N.v.S.); (S.E.L.)
| | - Niklas von Spreckelsen
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (B.S.); (P.Z.); (N.v.S.); (S.E.L.)
- Department of General Neurosurgery, Centre of Neurosurgery, Faculty of Medicine and University Hospital, University of Cologne, 50937 Cologne, Germany
| | - Andrei Loas
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; (C.E.F.); (C.M.F.); (A.L.); (N.H.)
| | - Nina Hartrampf
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; (C.E.F.); (C.M.F.); (A.L.); (N.H.)
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Bradley L. Pentelute
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA;
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; (C.E.F.); (C.M.F.); (A.L.); (N.H.)
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sean E. Lawler
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (B.S.); (P.Z.); (N.v.S.); (S.E.L.)
- Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02912, USA
| |
Collapse
|
172
|
Glioblastoma Embryonic-like Stem Cells Exhibit Immune-Evasive Phenotype. Cancers (Basel) 2022; 14:cancers14092070. [PMID: 35565200 PMCID: PMC9104850 DOI: 10.3390/cancers14092070] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/17/2022] [Accepted: 04/19/2022] [Indexed: 12/30/2022] Open
Abstract
Simple Summary Most glioblastoma (GBM) patients relapse after an initial response to treatment. These aggressive traits are often associated with the presence of glioma stem cells (GSCs) within the tumor bulk, which are thought to participate in GBM therapy resistance. Given GBM cellular heterogeneity, we hypothesized that GSCs might also display cellular hierarchies associated with different degrees of stemness. Based on single-cell RNAseq data from GBM patients, we identified a subpopulation of GSCs, named core-GSCs (c-GSCs), with a similar profile to embryonic stem cells and downregulation of immune-associated pathways. In addition, we developed an in vitro induced c-GSC (ic-GSC) model resembling their tumor counterpart. The characterization of immune-privileged c-GSCs provides a valuable resource to study immune evasion mechanisms in GBM and to identify potential unexplored targets to improve immunotherapy treatments. Abstract Background: Glioma stem cells (GSCs) have self-renewal and tumor-initiating capacities involved in drug resistance and immune evasion mechanisms in glioblastoma (GBM). Methods: Core-GSCs (c-GSCs) were identified by selecting cells co-expressing high levels of embryonic stem cell (ESC) markers from a single-cell RNA-seq patient-derived GBM dataset (n = 28). Induced c-GSCs (ic-GSCs) were generated by reprogramming GBM-derived cells (GBM-DCs) using induced pluripotent stem cell (iPSC) technology. The characterization of ic-GSCs and GBM-DCs was conducted by immunostaining, transcriptomic, and DNA methylation (DNAm) analysis. Results: We identified a GSC population (4.22% ± 0.59) exhibiting concurrent high expression of ESC markers and downregulation of immune-associated pathways, named c-GSCs. In vitro ic-GSCs presented high expression of ESC markers and downregulation of antigen presentation HLA proteins. Transcriptomic analysis revealed a strong agreement of enriched biological pathways between tumor c-GSCs and in vitro ic-GSCs (κ = 0.71). Integration of our epigenomic profiling with 833 functional ENCODE epigenetic maps identifies increased DNA methylation on HLA genes’ regulatory regions associated with polycomb repressive marks in a stem-like phenotype. Conclusions: This study unravels glioblastoma immune-evasive mechanisms involving a c-GSC population. In addition, it provides a cellular model with paired gene expression, and DNA methylation maps to explore potential therapeutic complements for GBM immunotherapy.
Collapse
|
173
|
Corgnac S, Damei I, Gros G, Caidi A, Terry S, Chouaib S, Deloger M, Mami-Chouaib F. Cancer stem-like cells evade CD8 +CD103 + tumor-resident memory T (T RM) lymphocytes by initiating an epithelial-to-mesenchymal transition program in a human lung tumor model. J Immunother Cancer 2022; 10:jitc-2022-004527. [PMID: 35418483 PMCID: PMC9014106 DOI: 10.1136/jitc-2022-004527] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2022] [Indexed: 02/06/2023] Open
Abstract
Background Cancer stem cells (CSC) define a population of rare malignant cells endowed with ‘stemness’ properties, such as self-renewing, multipotency and tumorigenicity. They are responsible for tumor initiation and progression, and could be associated with resistance to immunotherapies by negatively regulating antitumor immune response and acquiring molecular features enabling escape from CD8 T-cell immunity. However, the immunological hallmarks of human lung CSC and their potential interactions with resident memory T (TRM) cells within the tumor microenvironment have not been investigated. Methods We generated a non-small cell lung cancer model, including CSC line and clones, and autologous CD8+CD103+ TRM and CD8+CD103− non-TRM clones, to dissect out immune properties of CSC and their susceptibility to specific T-cell-mediated cytotoxic activity. Results Unlike their parental tumor cells, lung CSC are characterized by the initiation of an epithelial-to-mesenchymal transition program defined by upregulation of the SNAIL1 transcription factor and downregulation of phosphorylated-GSK-3β and cell surface E-cadherin. Acquisition of a CSC profile results in partial resistance to TRM-cell-mediated cytotoxicity, which correlates with decreased surface expression of the CD103 ligand E-cadherin and human leukocyte antigen-A2-neoepitope complexes. On the other hand, CSC gained expression of intercellular adhesion molecule (ICAM)-1 and thereby sensitivity to leukocyte function-associated antigen (LFA)-1-dependent non-TRM-cell-mediated killing. Cytotoxicity is inhibited by anti-ICAM-1 and anti-major histocompatibility complex class I neutralizing antibodies further emphasizing the role of LFA-1/ICAM-1 interaction in T-cell receptor-dependent lytic function. Conclusion Our data support the rational design of immunotherapeutic strategies targeting CSC to optimize their responsiveness to local CD8+CD103+ TRM cells for more efficient anticancer treatments.
Collapse
Affiliation(s)
- Stéphanie Corgnac
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Isabelle Damei
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Gwendoline Gros
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Aziza Caidi
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France.,Gustave Roussy, Plateforme de Bioinformatique, Université Paris-Saclay, INSERM US23, CNRS UMS 3655, Villejuif, France
| | - Stéphane Terry
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Salem Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France.,Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, UAE
| | - Marc Deloger
- Gustave Roussy, Plateforme de Bioinformatique, Université Paris-Saclay, INSERM US23, CNRS UMS 3655, Villejuif, France
| | - Fathia Mami-Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
174
|
Arunachalam E, Rogers W, Simpson GR, Möller-Levet C, Bolton G, Ismael M, Smith C, Keegen K, Bagwan I, Brend T, Short SC, Hong B, Otani Y, Kaur B, Annels N, Morgan R, Pandha H. HOX and PBX gene dysregulation as a therapeutic target in glioblastoma multiforme. BMC Cancer 2022; 22:400. [PMID: 35418059 PMCID: PMC9006463 DOI: 10.1186/s12885-022-09466-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 03/21/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most common high-grade malignant brain tumour in adults and arises from the glial cells in the brain. The prognosis of treated GBM remains very poor with 5-year survival rates of 5%, a figure which has not improved over the last few decades. Currently, there is a modest 14-month overall median survival in patients undergoing maximum safe resection plus adjuvant chemoradiotherapy. HOX gene dysregulation is now a widely recognised feature of many malignancies. METHODS In this study we have focused on HOX gene dysregulation in GBM as a potential therapeutic target in a disease with high unmet need. RESULTS We show significant dysregulation of these developmentally crucial genes and specifically that HOX genes A9, A10, C4 and D9 are strong candidates for biomarkers and treatment targets for GBM and GBM cancer stem cells. We evaluated a next generation therapeutic peptide, HTL-001, capable of targeting HOX gene over-expression in GBM by disrupting the interaction between HOX proteins and their co-factor, PBX. HTL-001 induced both caspase-dependent and -independent apoptosis in GBM cell lines. CONCLUSION In vivo biodistribution studies confirmed that the peptide was able to cross the blood brain barrier. Systemic delivery of HTL-001 resulted in improved control of subcutaneous murine and human xenograft tumours and improved survival in a murine orthotopic model.
Collapse
Affiliation(s)
- Einthavy Arunachalam
- Targeted Cancer Therapy, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7WG, UK
| | - William Rogers
- Targeted Cancer Therapy, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7WG, UK
| | - Guy R Simpson
- Targeted Cancer Therapy, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7WG, UK
| | - Carla Möller-Levet
- Targeted Cancer Therapy, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7WG, UK
| | - Gemma Bolton
- Targeted Cancer Therapy, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7WG, UK
- Surrey Technology Centre, HOX Therapeutics Ltd, Unit 2440 Occam Rd, Guildford, GU2 7YG, UK
| | - Mohammed Ismael
- Targeted Cancer Therapy, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7WG, UK
- Surrey Technology Centre, HOX Therapeutics Ltd, Unit 2440 Occam Rd, Guildford, GU2 7YG, UK
| | - Christopher Smith
- Targeted Cancer Therapy, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7WG, UK
| | - Karl Keegen
- Surrey Technology Centre, HOX Therapeutics Ltd, Unit 2440 Occam Rd, Guildford, GU2 7YG, UK
| | - Izhar Bagwan
- Department of Pathology, Royal Surrey County Hospital, Egerton Road, Guildford, GU2 7XX, Surrey, UK
| | - Tim Brend
- Faculty of Medicine and Health, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, LS9 7TF, UK
| | - Susan C Short
- Faculty of Medicine and Health, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, LS9 7TF, UK
| | - Bangxing Hong
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Centre at Houston, 7000 Fannin Street, Houston, TX, 77030, USA
| | - Yoshihiro Otani
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Centre at Houston, 7000 Fannin Street, Houston, TX, 77030, USA
| | - Balveen Kaur
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Centre at Houston, 7000 Fannin Street, Houston, TX, 77030, USA
| | - Nicola Annels
- Targeted Cancer Therapy, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7WG, UK
| | - Richard Morgan
- School of Biomedical Sciences, University of West London, St Mary's Road, Ealing, London, W5 5RF, UK
| | - Hardev Pandha
- Targeted Cancer Therapy, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7WG, UK.
| |
Collapse
|
175
|
Abstract
Tumors contain heterogeneous neoplastic cells and diverse stromal elements that collectively function as dynamic ecosystems, and this complicates predictive modeling ex vivo. LeBlanc et al. utilize single-cell analysis to demonstrate that patient-derived explants replicate tumor cell diversity and transient stromal cell types in patient surgical specimens. This suggests that patient-derived explants can be valuable as tumor models.
Collapse
Affiliation(s)
- Christopher G Hubert
- Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jeremy N Rich
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
176
|
dePadua M, Kulothungan P, Lath R, Prasad R, Madamchetty K, Atmakuri S, Ragamouni S, Gandhari M, Khandrika L, Jain J. Establishment and Characterization of Brain Cancer Primary Cell Cultures From Patients to Enable Phenotypic Screening for New Drugs. Front Pharmacol 2022; 13:778193. [PMID: 35370679 PMCID: PMC8970592 DOI: 10.3389/fphar.2022.778193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/07/2022] [Indexed: 11/24/2022] Open
Abstract
Aim: Desmoplastic infantile ganglioglioma (DIG), is a rare tumor arising mainly during the first 2 years of life. Molecular characterization of these benign yet rapidly proliferating tumors has been limited to evaluating a few mutations in few genes. Our aim was to establish a live cell culture to enable the understanding of the cellular processes driving the non-malignant growth of these tumors. Methods: Tumor tissue from a rare non-infantile 8-year-old female DIG patient was dissociated and digested using collagenase to establish live cultures. Both 2D monolayer and 3D neurospheres were successfully cultured and characterized for proliferative potential, intrinsic plasticity, presence of cancer stem cells and the expression of stem cell markers. Cells cultured as 3D were embedded as tissue blocks. Immunohistochemistry was performed in both tissue and 3D sections for markers including synaptophysin, vimentin, neurofilament and MIB-1. Mutation analysis by NGS was performed using a-100 gene panel. Results: Using immunohistochemistry, the 3D cultures were shown to express markers as in the original DIG tumor tissue indicating that the spheroid cultures were able to maintain the heterogeneity found in the original tumor. Cells continued proliferating past passage 10 indicative of immortalization. Enrichment of cancer stem cells was observed in neurospheres by FACS using CD133 antibody and RT-PCR. Mutation analysis indicated the presence of germline mutations in three genes and somatic mutations in two other genes. Conclusion: A spontaneous cell line-like cell culture with high percentage of stem cells has been established from a DIG tumor for the first time.
Collapse
Affiliation(s)
- Michelle dePadua
- Department of Pathology, Apollo Hospital, Apollo Health City, Hyderabad, India
| | | | - Rahul Lath
- Department of Neurosciences, Apollo Hospital, Apollo Health City, Hyderabad, India
| | - Ravikanti Prasad
- Department of Radiology, Apollo Hospital, Apollo Health City, Hyderabad, India
| | | | | | | | | | | | - Jugnu Jain
- Saarum Sciences Pvt Ltd., Hyderabad, India.,Sapien Biosciences Pvt Ltd., Hyderabad, India
| |
Collapse
|
177
|
Sarmiento BE, Callegari S, Ghotme KA, Akle V. Patient-Derived Xenotransplant of CNS Neoplasms in Zebrafish: A Systematic Review. Cells 2022; 11:cells11071204. [PMID: 35406768 PMCID: PMC8998145 DOI: 10.3390/cells11071204] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/11/2022] [Accepted: 03/28/2022] [Indexed: 02/01/2023] Open
Abstract
Glioblastoma and neuroblastoma are the most common central nervous system malignant tumors in adult and pediatric populations. Both are associated with poor survival. These tumors are highly heterogeneous, having complex interactions among different cells within the tumor and with the tumor microenvironment. One of the main challenges in the neuro-oncology field is achieving optimal conditions to evaluate a tumor’s molecular genotype and phenotype. In this respect, the zebrafish biological model is becoming an excellent alternative for studying carcinogenic processes and discovering new treatments. This review aimed to describe the results of xenotransplantation of patient-derived CNS tumors in zebrafish models. The reviewed studies show that it is possible to maintain glioblastoma and neuroblastoma primary cell cultures and transplant the cells into zebrafish embryos. The zebrafish is a suitable biological model for understanding tumor progression and the effects of different treatments. This model offers new perspectives in providing personalized care and improving outcomes for patients living with central nervous system tumors.
Collapse
Affiliation(s)
- Beatriz E. Sarmiento
- School of Medicine, Universidad de Los Andes, Bogotá 11711, Colombia; (B.E.S.); (S.C.)
| | - Santiago Callegari
- School of Medicine, Universidad de Los Andes, Bogotá 11711, Colombia; (B.E.S.); (S.C.)
| | - Kemel A. Ghotme
- Department of Neurosurgery, Fundación Santa Fe de Bogotá, Bogotá 111071, Colombia;
- Translational Neuroscience Research Lab, Faculty of Medicine, Universidad de La Sabana, Chía 250001, Colombia
| | - Veronica Akle
- School of Medicine, Universidad de Los Andes, Bogotá 11711, Colombia; (B.E.S.); (S.C.)
- Correspondence:
| |
Collapse
|
178
|
Hicks WH, Bird CE, Gattie LC, Shami ME, Traylor JI, Shi DD, McBrayer SK, Abdullah KG. Creation and Development of Patient-Derived Organoids for Therapeutic Screening in Solid Cancer. CURRENT STEM CELL REPORTS 2022. [DOI: 10.1007/s40778-022-00211-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
179
|
Ion Channel Drugs Suppress Cancer Phenotype in NG108-15 and U87 Cells: Toward Novel Electroceuticals for Glioblastoma. Cancers (Basel) 2022; 14:cancers14061499. [PMID: 35326650 PMCID: PMC8946312 DOI: 10.3390/cancers14061499] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 01/07/2023] Open
Abstract
Glioblastoma is a lethal brain cancer that commonly recurs after tumor resection and chemotherapy treatment. Depolarized resting membrane potentials and an acidic intertumoral extracellular pH have been associated with a proliferative state and drug resistance, suggesting that forced hyperpolarization and disruption of proton pumps in the plasma membrane could be a successful strategy for targeting glioblastoma overgrowth. We screened 47 compounds and compound combinations, most of which were ion-modulating, at different concentrations in the NG108-15 rodent neuroblastoma/glioma cell line. A subset of these were tested in the U87 human glioblastoma cell line. A FUCCI cell cycle reporter was stably integrated into both cell lines to monitor proliferation and cell cycle response. Immunocytochemistry, electrophysiology, and a panel of physiological dyes reporting voltage, calcium, and pH were used to characterize responses. The most effective treatments on proliferation in U87 cells were combinations of NS1643 and pantoprazole; retigabine and pantoprazole; and pantoprazole or NS1643 with temozolomide. Marker analysis and physiological dye signatures suggest that exposure to bioelectric drugs significantly reduces proliferation, makes the cells senescent, and promotes differentiation. These results, along with the observed low toxicity in human neurons, show the high efficacy of electroceuticals utilizing combinations of repurposed FDA approved drugs.
Collapse
|
180
|
Beiriger J, Habib A, Jovanovich N, Kodavali CV, Edwards L, Amankulor N, Zinn PO. The Subventricular Zone in Glioblastoma: Genesis, Maintenance, and Modeling. Front Oncol 2022; 12:790976. [PMID: 35359410 PMCID: PMC8960165 DOI: 10.3389/fonc.2022.790976] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/07/2022] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is a malignant tumor with a median survival rate of 15-16 months with standard care; however, cases of successful treatment offer hope that an enhanced understanding of the pathology will improve the prognosis. The cell of origin in GBM remains controversial. Recent evidence has implicated stem cells as cells of origin in many cancers. Neural stem/precursor cells (NSCs) are being evaluated as potential initiators of GBM tumorigenesis. The NSCs in the subventricular zone (SVZ) have demonstrated similar molecular profiles and share several distinctive characteristics to proliferative glioblastoma stem cells (GSCs) in GBM. Genomic and proteomic studies comparing the SVZ and GBM support the hypothesis that the tumor cells and SVZ cells are related. Animal models corroborate this connection, demonstrating migratory patterns from the SVZ to the tumor. Along with laboratory and animal research, clinical studies have demonstrated improved progression-free survival in patients with GBM after radiation to the ipsilateral SVZ. Additionally, key genetic mutations in GBM for the most part carry regulatory roles in the SVZ as well. An exciting avenue towards SVZ modeling and determining its role in gliomagenesis in the human context is human brain organoids. Here we comprehensively discuss and review the role of the SVZ in GBM genesis, maintenance, and modeling.
Collapse
Affiliation(s)
- Jamison Beiriger
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Ahmed Habib
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Nicolina Jovanovich
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Chowdari V. Kodavali
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Lincoln Edwards
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Nduka Amankulor
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Pascal O. Zinn
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| |
Collapse
|
181
|
Tan X, Mi T, Zhang Z, Jin L, Wang Z, Wu X, Wang J, Li M, Zhanghuang C, He D. Multiple transcriptome analysis of Piwil2-induced cancer stem cells, including piRNAs, mRNAs and miRNAs reveals the mechanism of tumorigenesis and development. Mol Biol Rep 2022; 49:6885-6898. [PMID: 35182325 DOI: 10.1007/s11033-022-07237-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/08/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Cancer stem cells play important roles in the process of tumorigenesis. Our research group obtained cancer stem cell-like cells named Piwil2-iCSCs by reprogramming human preputial fibroblasts (FBs) with the PIWIL2 gene, but the mechanism of Piwil2-iCSCs is still unclear. METHODS We sequenced the piRNAs, miRNAs and mRNAs of Piwil2-iCSCs and FBs, and analyzed the differences. Gene Ontology (GO) and, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses and gene set enrichment analysis (GSEA) were performed on the differentially expressed (DE) mRNAs. In addition, we analyzed the variable shear events and fusion genes in the Piwil2-iCSCs. Target gene prediction and functional enrichment analysis were performed for the DE miRNAs. RESULTS A total of 1119 DE mRNAs, 220 DE piRNAs, and 440 DE miRNAs were obtained between the Piwil2-iCSCs and FBs. Functional enrichment analysis showed that the genes with upregulated expression were mainly involved in DNA repair, mismatch repair, base excision repair, and nucleotide excision repair. Genes with downregulated expression were mainly involved in the TGF-β receptor signaling pathway, senescence and autophagy in cancer. More frequent shear events occurred in Piwil2-iCSCs and FBs, especially in intron retention (IR) events. We also identified three fusion genes MCM3AP-C21orf58, LRRFIP2-CAV3 and TMEM184B-DMC1. Enrichment analysis of DE miRNAs showed that they were associated with apoptosis, the TGF-β signaling pathway, and the stem cell regulatory signaling pathway. In particular, target gene prediction of the top three miRNAs with upregulated expression showed that they targeted SMAD, GREM1 and other genes to participate in the regulation of TGF-β and other pathways. CONCLUSION PIWIL2-induced cancer stem cells have significantly altered levels of miRNAs, piRNAs and mRNAs.TGF-β, autophagy, apoptosis and other pathways may play an important role in stem cell development. The occurrence of alternative splicing and fusion genes may be related to the occurrence of cancer stem cells.
Collapse
Affiliation(s)
- Xiaojun Tan
- Department of Urology, Children's Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400014, People's Republic of China.,Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, People's Republic of China.,China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing, Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, People's Republic of China
| | - Tao Mi
- Department of Urology, Children's Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400014, People's Republic of China.,Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, People's Republic of China.,China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing, Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, People's Republic of China
| | - Zhaoxia Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400014, People's Republic of China.,Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, People's Republic of China.,China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing, Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, People's Republic of China
| | - Liming Jin
- Department of Urology, Children's Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400014, People's Republic of China.,Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, People's Republic of China.,China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing, Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, People's Republic of China
| | - Zhang Wang
- Department of Urology, Children's Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400014, People's Republic of China.,Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, People's Republic of China.,China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing, Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, People's Republic of China
| | - Xin Wu
- Department of Urology, Children's Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400014, People's Republic of China.,Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, People's Republic of China.,China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing, Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, People's Republic of China
| | - Jinkui Wang
- Department of Urology, Children's Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400014, People's Republic of China.,Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, People's Republic of China.,China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing, Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, People's Republic of China
| | - Mujie Li
- Department of Urology, Children's Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400014, People's Republic of China.,Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, People's Republic of China.,China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing, Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, People's Republic of China
| | - Chenghao Zhanghuang
- Department of Urology, Children's Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400014, People's Republic of China.,Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, People's Republic of China.,China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing, Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, People's Republic of China
| | - Dawei He
- Department of Urology, Children's Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400014, People's Republic of China. .,Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, People's Republic of China. .,China International Science and Technology Cooperation Base of Child Development and Critical; National Clinical Research Center for Child Health and Disorders, Chongqing, Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, People's Republic of China.
| |
Collapse
|
182
|
Lu C, Chen X, Yan Y, Ren X, Wang X, Peng B, Cai Y, Liang Q, Xu Z, Peng J. Aberrant Expression of ADARB1 Facilitates Temozolomide Chemoresistance and Immune Infiltration in Glioblastoma. Front Pharmacol 2022; 13:768743. [PMID: 35177985 PMCID: PMC8844449 DOI: 10.3389/fphar.2022.768743] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 01/10/2022] [Indexed: 02/05/2023] Open
Abstract
Chemoresistance, especially temozolomide (TMZ) resistance, is a major clinical challenge in the treatment of glioblastoma (GBM). Exploring the mechanisms of TMZ resistance could help us identify effective therapies. Adenosine deaminases acting on RNA (ADARs) are very important in RNA modification through regulating the A-to-I RNA editing. Recent studies have shown that ADARs regulate multiple neurotransmitter receptors, which have been linked with the occurrence and progress of GBM. Here, data from several bioinformatics databases demonstrated that adenosine deaminase RNA specific B1 (ADARB1), also named ADAR2, was upregulated in both GBM tissues and cells, and had the prognostic value in GBM patients. Moreover, ADARB1 was found to be involved in AKT-mediated TMZ resistance in GBM cells. The KEGG analysis of ADARB1-associated co-expressed genes showed that ADARB1 was potentially involved in the mitochondrial respiratory chain complex. TISIDB and GEPIA databases were further used to analyze the role of ADARB1 in tumor-immune system interactions in GBM. These findings deepened our understanding of the function of ADARB1 in tumorigenesis and therapeutic response in GBM.
Collapse
Affiliation(s)
- Can Lu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Xi Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Xinxin Ren
- Key Laboratory of Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Xiang Wang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Bi Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Cai
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Qiuju Liang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China.,Department of Pathology, Xiangya Changde Hospital, Changde, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jinwu Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China.,Department of Pathology, Xiangya Changde Hospital, Changde, China
| |
Collapse
|
183
|
Jia Y, Wei Z, Zhang S, Yang B, Li Y. Instructive Hydrogels for Primary Tumor Cell Culture: Current Status and Outlook. Adv Healthc Mater 2022; 11:e2102479. [PMID: 35182456 DOI: 10.1002/adhm.202102479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 02/07/2022] [Indexed: 02/06/2023]
Abstract
Primary tumor organoids (PTOs) growth in hydrogels have emerged as an important in vitro model that recapitulates many characteristics of the native tumor tissue, and have important applications in fundamental cancer research and for the development of useful therapeutic treatment. This paper begins with reviewing the methods of isolation of primary tumor cells. Then, recent advances on the instructive hydrogels as biomimetic extracellular matrix for primary tumor cell culture and construction of PTO models are summarized. Emerging microtechnology for growth of PTOs in microscale hydrogels and the applications of PTOs are highlighted. This paper concludes with an outlook on the future directions in the investigation of instructive hydrogels for PTO growth.
Collapse
Affiliation(s)
- Yiyang Jia
- State Key Laboratory of Supramolecular Structure and Materials College of Chemistry Jilin University 2699 Qianjin Street Changchun 130012 China
| | - Zhentong Wei
- Department of Oncologic Gynecology The First Hospital of Jilin University Changchun 130021 China
| | - Songling Zhang
- Department of Oncologic Gynecology The First Hospital of Jilin University Changchun 130021 China
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials College of Chemistry Jilin University 2699 Qianjin Street Changchun 130012 China
- Joint Laboratory of Opto‐Functional Theranostics in Medicine and Chemistry The First Hospital of Jilin University Changchun 130021 China
| | - Yunfeng Li
- State Key Laboratory of Supramolecular Structure and Materials College of Chemistry Jilin University 2699 Qianjin Street Changchun 130012 China
- Joint Laboratory of Opto‐Functional Theranostics in Medicine and Chemistry The First Hospital of Jilin University Changchun 130021 China
| |
Collapse
|
184
|
Soroceanu L, Singer E, Dighe P, Sidorov M, Limbad C, Rodriquez-Brotons A, Rix P, Woo RWL, Dickinson L, Desprez PY, McAllister SD. Cannabidiol Inhibits RAD51 and Sensitizes Glioblastoma to Temozolomide in Multiple Orthotopic Tumor Models. Neurooncol Adv 2022; 4:vdac019. [PMID: 35356807 PMCID: PMC8962752 DOI: 10.1093/noajnl/vdac019] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background Cannabidiol (CBD), a nonpsychoactive cannabinoid with a low toxicity profile, has been shown to produce antitumor activity across cancers in part through selective production of reactive oxygen species (ROS) in tumor cells. The alkylating agent, temozolomide (TMZ), is standard of care for treatment of glioblastoma (GBM). It can trigger increased ROS to induce DNA damage. It has also been reported that downregulating the expression of RAD51, an important DNA damage repair protein, leads to sensitization of GBM to TMZ. Methods We determined the extent to which CBD enhanced the antitumor activity of TMZ in multiple orthotopic models of GBM. In addition, we investigated the potential for CBD to enhance the antitumor activity of TMZ through production of ROS and modulation of DNA repair pathways. Results CBD enhanced the activity of TMZ in U87 MG and U251 GBM cell lines and in patient-derived primary GBM163 cells leading to stimulation of ROS, activation of the ROS sensor AMP-activated protein kinase (AMPK), and upregulation of the autophagy marker LC3A. CBD produced a sensitization of U87 and GBM163-derived intracranial (i.c.) tumors to TMZ and significantly increased survival of tumor-bearing mice. However, these effects were not observed in orthotopic models derived from GBM with intact methylguanine methyltransferase (MGMT) expression. We further demonstrate that CBD inhibited RAD51 expression in MGMT-methylated models of GBM, providing a potential mechanism for tumor sensitization to TMZ by CBD. Conclusion These data support the potential therapeutic benefits of using CBD to enhance the antitumor activity of TMZ in GBM patients.
Collapse
Affiliation(s)
- Liliana Soroceanu
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Eric Singer
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Pratiksha Dighe
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Max Sidorov
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Chandani Limbad
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | | | - Peter Rix
- Launch Bioscience, San Diego, CA, USA
| | - Rinette W L Woo
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | | | - Pierre-Yves Desprez
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Sean D McAllister
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| |
Collapse
|
185
|
Uribe D, Niechi I, Rackov G, Erices JI, San Martín R, Quezada C. Adapt to Persist: Glioblastoma Microenvironment and Epigenetic Regulation on Cell Plasticity. BIOLOGY 2022; 11:313. [PMID: 35205179 PMCID: PMC8869716 DOI: 10.3390/biology11020313] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is the most frequent and aggressive brain tumor, characterized by great resistance to treatments, as well as inter- and intra-tumoral heterogeneity. GBM exhibits infiltration, vascularization and hypoxia-associated necrosis, characteristics that shape a unique microenvironment in which diverse cell types are integrated. A subpopulation of cells denominated GBM stem-like cells (GSCs) exhibits multipotency and self-renewal capacity. GSCs are considered the conductors of tumor progression due to their high tumorigenic capacity, enhanced proliferation, invasion and therapeutic resistance compared to non-GSCs cells. GSCs have been classified into two molecular subtypes: proneural and mesenchymal, the latter showing a more aggressive phenotype. Tumor microenvironment and therapy can induce a proneural-to-mesenchymal transition, as a mechanism of adaptation and resistance to treatments. In addition, GSCs can transition between quiescent and proliferative substates, allowing them to persist in different niches and adapt to different stages of tumor progression. Three niches have been described for GSCs: hypoxic/necrotic, invasive and perivascular, enhancing metabolic changes and cellular interactions shaping GSCs phenotype through metabolic changes and cellular interactions that favor their stemness. The phenotypic flexibility of GSCs to adapt to each niche is modulated by dynamic epigenetic modifications. Methylases, demethylases and histone deacetylase are deregulated in GSCs, allowing them to unlock transcriptional programs that are necessary for cell survival and plasticity. In this review, we described the effects of GSCs plasticity on GBM progression, discussing the role of GSCs niches on modulating their phenotype. Finally, we described epigenetic alterations in GSCs that are important for stemness, cell fate and therapeutic resistance.
Collapse
Affiliation(s)
- Daniel Uribe
- Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia 5090000, Chile; (D.U.); (I.N.); (J.I.E.); (R.S.M.)
| | - Ignacio Niechi
- Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia 5090000, Chile; (D.U.); (I.N.); (J.I.E.); (R.S.M.)
| | - Gorjana Rackov
- Department of Immunology and Oncology, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas (CNB-CSIC), 28049 Madrid, Spain;
| | - José I. Erices
- Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia 5090000, Chile; (D.U.); (I.N.); (J.I.E.); (R.S.M.)
| | - Rody San Martín
- Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia 5090000, Chile; (D.U.); (I.N.); (J.I.E.); (R.S.M.)
| | - Claudia Quezada
- Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia 5090000, Chile; (D.U.); (I.N.); (J.I.E.); (R.S.M.)
- Millennium Institute on Immunology and Immunotherapy, Universidad Austral de Chile, Valdivia 5090000, Chile
| |
Collapse
|
186
|
Barbieri F, Bosio AG, Pattarozzi A, Tonelli M, Bajetto A, Verduci I, Cianci F, Cannavale G, Palloni LMG, Francesconi V, Thellung S, Fiaschi P, Mazzetti S, Schenone S, Balboni B, Girotto S, Malatesta P, Daga A, Zona G, Mazzanti M, Florio T. Chloride intracellular channel 1 activity is not required for glioblastoma development but its inhibition dictates glioma stem cell responsivity to novel biguanide derivatives. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:53. [PMID: 35135603 PMCID: PMC8822754 DOI: 10.1186/s13046-021-02213-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 12/07/2021] [Indexed: 11/10/2022]
Abstract
Abstract
Background
Chloride intracellular channel-1 (CLIC1) activity controls glioblastoma proliferation. Metformin exerts antitumor effects in glioblastoma stem cells (GSCs) inhibiting CLIC1 activity, but its low potency hampers its translation in clinical settings.
Methods
We synthesized a small library of novel biguanide-based compounds that were tested as antiproliferative agents for GSCs derived from human glioblastomas, in vitro using 2D and 3D cultures and in vivo in the zebrafish model. Compounds were compared to metformin for both potency and efficacy in the inhibition of GSC proliferation in vitro (MTT, Trypan blue exclusion assays, and EdU labeling) and in vivo (zebrafish model), migration (Boyden chamber assay), invasiveness (Matrigel invasion assay), self-renewal (spherogenesis assay), and CLIC1 activity (electrophysiology recordings), as well as for the absence of off-target toxicity (effects on normal stem cells and toxicity for zebrafish and chick embryos).
Results
We identified Q48 and Q54 as two novel CLIC1 blockers, characterized by higher antiproliferative potency than metformin in vitro, in both GSC 2D cultures and 3D spheroids. Q48 and Q54 also impaired GSC self-renewal, migration and invasion, and displayed low systemic in vivo toxicity. Q54 reduced in vivo proliferation of GSCs xenotransplanted in zebrafish hindbrain. Target specificity was confirmed by recombinant CLIC1 binding experiments using microscale thermophoresis approach. Finally, we characterized GSCs from GBMs spontaneously expressing low CLIC1 protein, demonstrating their ability to grow in vivo and to retain stem-like phenotype and functional features in vitro. In these GSCs, Q48 and Q54 displayed reduced potency and efficacy as antiproliferative agents as compared to high CLIC1-expressing tumors. However, in 3D cultures, metformin and Q48 (but not Q54) inhibited proliferation, which was dependent on the inhibition dihydrofolate reductase activity.
Conclusions
These data highlight that, while CLIC1 is dispensable for the development of a subset of glioblastomas, it acts as a booster of proliferation in the majority of these tumors and its functional expression is required for biguanide antitumor class-effects. In particular, the biguanide-based derivatives Q48 and Q54, represent the leads to develop novel compounds endowed with better pharmacological profiles than metformin, to act as CLIC1-blockers for the treatment of CLIC1-expressing glioblastomas, in a precision medicine approach.
Collapse
|
187
|
Jiang L, Hao Y, Shao C, Wu Q, Prager BC, Gimple RC, Sulli G, Kim LJ, Zhang G, Qiu Z, Zhu Z, Fu XD, Rich JN. ADAR1-mediated RNA editing links ganglioside catabolism to glioblastoma stem cell maintenance. J Clin Invest 2022; 132:143397. [PMID: 35133980 PMCID: PMC8920333 DOI: 10.1172/jci143397] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 02/03/2022] [Indexed: 11/17/2022] Open
Abstract
Glioblastoma (GBM) is the most common and lethal primary malignant brain tumor, containing GBM stem cells (GSCs) that contribute to therapeutic resistance and relapse. Exposing potential GSC vulnerabilities may provide therapeutic strategies against GBM. Here, we interrogated the role of Adenosine-to-Inosine (A-to-I) RNA editing mediated by ADAR1 (adenosine deaminase acting on RNA 1) in GSCs and found that both ADAR1 and global RNA editomes were elevated in GSCs compared to normal neural stem cells (NSCs). ADAR1 inactivation or blocking the upstream JAK/STAT pathway through TYK2 inhibition impaired GSC self-renewal and stemness. Downstream of ADAR1, RNA editing of the 3'UTR of GM2A, a key ganglioside catabolism activator, proved to be critical, as interfering with ganglioside catabolism showed similar functional impact on GSCs as ADAR1 disruption. These findings reveal RNA editing links ganglioside catabolism to GSC self-renewal and stemness, exposing a potential vulnerability of GBM for therapeutic intervention.
Collapse
Affiliation(s)
- Li Jiang
- Department of Medicine, University of California, San Diego, San Diego, United States of America
| | - Yajing Hao
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, United States of America
| | - Changwei Shao
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, United States of America
| | - Qiulian Wu
- Hillman Cancer Center, Cancer Institute, University of Pittsburgh, Pittsburgh, United States of America
| | - Briana C Prager
- Stem Cell Biology, Cleveland Clinic, Cleveland, United States of America
| | - Ryan C Gimple
- Department of Medicine, University of California, San Diego, San Diego, United States of America
| | - Gabriele Sulli
- Department of Medicine, University of California, San Diego, San Diego, United States of America
| | - Leo Jk Kim
- Department of Medicine, University of California, San Diego, San Diego, United States of America
| | - Guoxin Zhang
- Department of Medicine, University of California, San Diego, San Diego, United States of America
| | - Zhixin Qiu
- Hillman Cancer Center, Cancer Institute, University of Pittsburgh, Pittsburgh, United States of America
| | - Zhe Zhu
- Department of Medicine, University of California, San Diego, San Diego, United States of America
| | - Xiang-Dong Fu
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, United States of America
| | - Jeremy N Rich
- Hillman Cancer Center, Cancer Institute, University of Pittsburgh, Pittsburgh, United States of America
| |
Collapse
|
188
|
Heffernan JM, McNamara JB, Vernon BL, Mehta S, Sirianni RW. PNJ scaffolds promote microenvironmental regulation of glioblastoma stem-like cell enrichment and radioresistance. Biomater Sci 2022; 10:819-833. [PMID: 34994746 PMCID: PMC8939461 DOI: 10.1039/d0bm01169j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Glioblastoma (GBM) brain tumors contain a subpopulation of self-renewing multipotent Glioblastoma stem-like cells (GSCs) that are believed to drive the near inevitable recurrence of GBM. We previously engineered temperature responsive scaffolds based on the polymer poly(N-isopropylacrylamide-co-Jeffamine M-1000 acrylamide) (PNJ) for the purpose of enriching GSCs in vitro from patient-derived samples. Here, we used PNJ scaffolds to study microenvironmental regulation of self-renewal and radiation response in patient-derived GSCs representing classical and proneural subtypes. GSC self-renewal was regulated by the composition of PNJ scaffolds and varied with cell type. PNJ scaffolds protected against radiation-induced cell death, particularly in conditions that also promoted GSC self-renewal. Additionally, cells cultured in PNJ scaffolds exhibited increased expression of the transcription factor HIF2α, which was not observed in neurosphere culture, providing a potential mechanistic basis for differences in radio-resistance. Differences in PNJ regulation of HIF2α in irradiated and untreated conditions also offered evidence of stem plasticity. These data show PNJ scaffolds provide a unique biomaterial for evaluating dynamic microenvironmental regulation of GSC self-renewal, radioresistance, and stem plasticity.
Collapse
Affiliation(s)
- John M. Heffernan
- Ivy Brain Tumor Center, Barrow Neurological Institute, 350 W Thomas Rd, Phoenix, AZ 85013, USA, School of Biological and Health Systems Engineering, Arizona State University, PO Box 879709, Tempe, AZ 85287, USA, Sonoran Biosciences, 1048 E Knight Ln, Tempe, AZ, USA
| | - James B. McNamara
- Ivy Brain Tumor Center, Barrow Neurological Institute, 350 W Thomas Rd, Phoenix, AZ 85013, USA, Department of Chemistry and Biochemistry, University of Arizona, 1306 E. University Blvd., Tucson, Arizona 85721, USA
| | - Brent L. Vernon
- School of Biological and Health Systems Engineering, Arizona State University, PO Box 879709, Tempe, AZ 85287, USA
| | - Shwetal Mehta
- Ivy Brain Tumor Center, Barrow Neurological Institute, 350 W Thomas Rd, Phoenix, AZ 85013, USA
| | - Rachael W. Sirianni
- Ivy Brain Tumor Center, Barrow Neurological Institute, 350 W Thomas Rd, Phoenix, AZ 85013, USA, School of Biological and Health Systems Engineering, Arizona State University, PO Box 879709, Tempe, AZ 85287, USA, Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA
| |
Collapse
|
189
|
Dekker LJM, Verheul C, Wensveen N, Leenders W, Lamfers MLM, Leenstra S, Luider TM. Effects of the IDH1 R132H Mutation on the Energy Metabolism: A Comparison between Tissue and Corresponding Primary Glioma Cell Cultures. ACS OMEGA 2022; 7:3568-3578. [PMID: 35128264 PMCID: PMC8811756 DOI: 10.1021/acsomega.1c06121] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/24/2021] [Indexed: 05/03/2023]
Abstract
The R132H mutation in the metabolic enzyme isocitrate dehydrogenase 1 (IDH1) is the most important prognostic factor for the survival of glioma patients. Subsequent studies led to the discovery of a panel of enzymes mainly involved in glutamate anaplerosis and aerobic glycolysis that change in abundance as a result of the IDH1 mutation. To further study these changes, appropriate glioma models are required that accurately mimic in vivo metabolism. To investigate how metabolism is affected by in vitro cell culture, we here compared surgically obtained snap-frozen glioma tissues with their corresponding primary glioma cell culture models with a previously developed targeted mass spectrometry proteomic assay. We determined the relative abundance of a panel of metabolic enzymes. Results confirmed increased glutamate use and decreased aerobic glycolysis in resected IDH1 R132H glioma tissue samples. However, these metabolic profiles were not reflected in the paired glioma primary cell cultures. We suggest that culture conditions and tumor microenvironment play a crucial role in maintaining the in vivo metabolic situation in cell culture models. For this reason, new models that more closely resemble the in vivo microenvironment, such as three-dimensional cell co-cultures or organotypic multicellular spheroid models, need to be developed and investigated.
Collapse
Affiliation(s)
- Lennard J M Dekker
- Laboratories of Neuro-Oncology/Clinical and Cancer Proteomics, Department of Neurology, Erasmus MC, Rotterdam, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Cassandra Verheul
- Department of Neurosurgery, Erasmus MC, Rotterdam, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Nicky Wensveen
- Laboratories of Neuro-Oncology/Clinical and Cancer Proteomics, Department of Neurology, Erasmus MC, Rotterdam, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - William Leenders
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Martine L M Lamfers
- Department of Neurosurgery, Erasmus MC, Rotterdam, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Sieger Leenstra
- Department of Neurosurgery, Erasmus MC, Rotterdam, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Theo M Luider
- Laboratories of Neuro-Oncology/Clinical and Cancer Proteomics, Department of Neurology, Erasmus MC, Rotterdam, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
190
|
Li J, Fan H, Zhou X, Xiang Y, Liu Y. Prognostic Significance and Gene Co-Expression Network of PLAU and PLAUR in Gliomas. Front Oncol 2022; 11:602321. [PMID: 35087738 PMCID: PMC8787124 DOI: 10.3389/fonc.2021.602321] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/16/2021] [Indexed: 02/05/2023] Open
Abstract
The urokinase-type plasminogen activator(PLAU) and its receptor PLAUR participate in a series of cell physiological activities on the extracellular surface. Abnormal expression of PLAU and PLAUR is associated with tumorigenesis. This study aims to evaluate the prognostic value of PLAU/PLAUR transcription expression in glioma and to explore how they affect the generation and progression of glioma. In this study, online databases are applied, such as Oncomine, GEPIA, CGGA, cBioPortal, and LinkedOmics. Overexpression of PLAU/PLAUR was found to be significantly associated with clinical variables including age, tumor type, WHO grade, histology, IDH-1 mutation, and 1p19q status. PLAU and PLAUR had a high correlation in transcriptional expression levels. High expression of PLAU and PLAUR predicted a poor prognosis in primary glioma and recurrent glioma patients, especially in lower grade gliomas. Cox regression analysis indicated that high expression of PLAU and PLAUR were independent prognostic factors for shorter overall survival in glioma patients. In gene co-expression network analysis PLAU and PLAUR and their co-expression genes were found to be involved in inflammatory activities and tumor-related signaling pathways. In conclusion, PLAU and PLAUR could be promising prognostic biomarkers and potential therapeutic targets of glioma patients.
Collapse
Affiliation(s)
- Junhong Li
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Huanhuan Fan
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu, China
| | - Xingwang Zhou
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yufan Xiang
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yanhui Liu
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
191
|
Liu F, Peng B, Li M, Ma J, Deng G, Zhang S, Sheu WC, Zou P, Wu H, Liu J, Chen AT, Mohammed FS, Zhou J. Targeted disruption of tumor vasculature via polyphenol nanoparticles to improve brain cancer treatment. CELL REPORTS. PHYSICAL SCIENCE 2022; 3:100691. [PMID: 35199059 PMCID: PMC8863382 DOI: 10.1016/j.xcrp.2021.100691] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Despite being effective for many other solid tumors, traditional anti-angiogenic therapy has been shown to be insufficient for the treatment of malignant glioma. Here, we report the development of polyphenol nanoparticles (NPs), which not only inhibit the formation of new vessels but also enable targeted disruption of the existing tumor vasculature. The NPs are synthesized through a combinatory iron-coordination and polymer-stabilization approach, which allows for high drug loading and intrinsic tumor vessel targeting. We study a lead NP consisting of quercetin and find that the NP after intravenous administration preferentially binds to VEGFR2, which is overexpressed in tumor vasculature. We demonstrate that the binding is mediated by quercetin, and the interaction of NPs with VEGFR2 leads to disruption of the existing tumor vasculature and inhibition of new vessel development. As a result, systemic treatment with the NPs effectively inhibits tumor growth and increases drug delivery to tumors.
Collapse
Affiliation(s)
- Fuyao Liu
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Bin Peng
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Miao Li
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Junning Ma
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Gang Deng
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Shenqi Zhang
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Wendy C. Sheu
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
| | - Pan Zou
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Haoan Wu
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Jun Liu
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Ann T. Chen
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
| | - Farrah S. Mohammed
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
| | - Jiangbing Zhou
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
- Lead contact
- Correspondence:
| |
Collapse
|
192
|
Nishida K, Sekida S, Anada T, Tanaka M. Modulation of Biological Responses of Tumor Cells Adhered to Poly(2-methoxyethyl acrylate) with Increasing Cell Viability under Serum-Free Conditions. ACS Biomater Sci Eng 2022; 8:672-681. [PMID: 35037460 DOI: 10.1021/acsbiomaterials.1c01469] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Circulating tumor cells in body fluids are important biomarkers in cancer diagnosis. The culture of tumor cells isolated from body fluids can provide intrinsic information about tumors and can be used to screen for the best anticancer drugs. However, the culture of primary tumor cells has been hindered by their low viability and difficulties in recapitulating the phenotype of primary tumors in in vitro culture. The culture of tumor cells under serum-free conditions is one of the methodologies to maintain the phenotype and genotype of primary tumors. Poly(2-methoxyethyl acrylate) (PMEA)-coated substrates have been investigated to prolong the proliferation of tumor cells under serum-free conditions. In this study, we investigated the detailed behavior and the mechanism of the increase in tumor cell viability after adherence to PMEA substrates. The blebbing formation of tumor cells on PMEA was attributed not to apoptosis but to the low adhesion strength of cells on PMEA. Moreover, blebbing tumor cells showed amoeboid movement and formed clusters with other cells via N-cadherin, leading to an increase in tumor cell viability. Furthermore, the behaviors of tumor cells adhered to PMEA under serum-free conditions were involved in the activation of the PI3K and Rho-associated protein kinase pathways. Thus, we propose that PMEA would be suitable for the development of devices to cultivate primary tumor cells under serum-free conditions for the label-free diagnosis of cancer.
Collapse
Affiliation(s)
- Kei Nishida
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Shogo Sekida
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Takahisa Anada
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.,Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Masaru Tanaka
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.,Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| |
Collapse
|
193
|
Pacini L, Cabal VN, Hermsen MA, Huang PH. EGFR Exon 20 Insertion Mutations in Sinonasal Squamous Cell Carcinoma. Cancers (Basel) 2022; 14:394. [PMID: 35053553 PMCID: PMC8774177 DOI: 10.3390/cancers14020394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 12/27/2022] Open
Abstract
Recurrent epidermal growth factor receptor (EGFR)-activating mutations have been identified in a rare form of head and neck cancer known as sinonasal squamous cell carcinoma (SNSCC), a malignant disease with a 5-year mortality rate of ~40%. Interestingly, the majority of EGFR mutations identified in patients with primary SNSCC are exon 20 insertions (Ex20ins), which is in contrast to non-small-cell lung cancer (NSCLC), where the EGFR exon 19 deletion and L858R mutations predominate. These studies demonstrate that EGFR Ex20ins mutations are not exclusive to lung cancer as previously believed, but are also involved in driving SNSCC pathogenesis. Here we review the landscape of EGFR mutations in SNSCC, with a particular focus on SNSCC associated with inverted sinonasal papilloma (ISP), a benign epithelial neoplasm. Taking lessons from NSCLC, we also discuss potential new treatment options for ISP-associated SNSCC harbouring EGFR Ex20ins in the context of targeted therapies, drug resistance and precision cancer medicine. Moving forward, further basic and translational work is needed to delineate the biology of EGFR Ex20ins in SNSCC in order to develop more effective treatments for patients with this rare disease.
Collapse
Affiliation(s)
- Laura Pacini
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM2 5NG, UK;
| | - Virginia N. Cabal
- Department Head and Neck Cancer, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Centro de Investigación Biomédica en Red (CIBER-ONC), 33011 Oviedo, Spain; (V.N.C.); (M.A.H.)
| | - Mario A. Hermsen
- Department Head and Neck Cancer, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Centro de Investigación Biomédica en Red (CIBER-ONC), 33011 Oviedo, Spain; (V.N.C.); (M.A.H.)
| | - Paul H. Huang
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM2 5NG, UK;
| |
Collapse
|
194
|
Malacrida A, Di Domizio A, Bentivegna A, Cislaghi G, Messuti E, Tabano SM, Giussani C, Zuliani V, Rivara M, Nicolini G. MV1035 Overcomes Temozolomide Resistance in Patient-Derived Glioblastoma Stem Cell Lines. BIOLOGY 2022; 11:70. [PMID: 35053068 PMCID: PMC8772739 DOI: 10.3390/biology11010070] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/15/2021] [Accepted: 12/29/2021] [Indexed: 11/26/2022]
Abstract
Glioblastoma (GBM, grade IV glioma) represents the most aggressive brain tumor and patients with GBM have a poor prognosis. Until now surgical resection followed by radiotherapy and temozolomide (TMZ) treatment represents the standard strategy for GBM. We showed that the imidazobenzoxazin-5-thione MV1035 is able to significantly reduce GBM U87-MG cells migration and invasiveness through inhibition of the RNA demethylase ALKBH5. In this work, we focus on the DNA repair protein ALKBH2, a further MV1035 target resulting from SPILLO-PBSS proteome-wide scale in silico analysis. Our data demonstrate that MV1035 inhibits the activity of ALKBH2, known to be involved in GBM TMZ resistance. MV1035 was used on both U87-MG and two patient-derived (PD) glioma stem cells (GSCs): in combination with TMZ, it has a significant synergistic effect in reducing cell viability and sphere formation. Moreover, MV1035 induces a reduction in MGMT expression in PD-GSCs cell lines most likely through a mechanism that acts on MGMT promoter methylation. Taken together our data show that MV1035 could act as an inhibitor potentially helpful to overcome TMZ resistance and able to reduce GBM migration and invasiveness.
Collapse
Affiliation(s)
- Alessio Malacrida
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy; (A.M.); (A.B.); (E.M.); (C.G.); (G.N.)
- Milan Center for Neuroscience, University of Milano-Bicocca, Piazza dell’Ateneo Nuovo 1, 20126 Milan, Italy
| | | | - Angela Bentivegna
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy; (A.M.); (A.B.); (E.M.); (C.G.); (G.N.)
- Milan Center for Neuroscience, University of Milano-Bicocca, Piazza dell’Ateneo Nuovo 1, 20126 Milan, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy
| | - Giacomo Cislaghi
- SPILLOproject, Via Stradivari 17, 20037 Paderno Dugnano, Italy; (A.D.D.); (G.C.)
| | - Eleonora Messuti
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy; (A.M.); (A.B.); (E.M.); (C.G.); (G.N.)
| | - Silvia Maria Tabano
- Laboratory of Medical Genetics, IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy;
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Carlo Giussani
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy; (A.M.); (A.B.); (E.M.); (C.G.); (G.N.)
- Neurosurgery Unit, Department of Neuroscience, S. Gerardo Hospital, 20900 Monza, Italy
| | - Valentina Zuliani
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy;
| | - Mirko Rivara
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy;
| | - Gabriella Nicolini
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy; (A.M.); (A.B.); (E.M.); (C.G.); (G.N.)
- Milan Center for Neuroscience, University of Milano-Bicocca, Piazza dell’Ateneo Nuovo 1, 20126 Milan, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy
| |
Collapse
|
195
|
Del Valle L, Reiss K, Parker Struckhoff A. Culture and Phenotyping of Glial Cell Cultures, Gliospheres, and Neurospheres. Methods Mol Biol 2022; 2422:217-232. [PMID: 34859409 DOI: 10.1007/978-1-0716-1948-3_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Cell cultures constitute an important tool for research as a way to reproduce pathological processes in a controlled system. However, the culture of brain-derived cells in monolayer presents significant challenges that obscure the fidelity of in vitro results. After a few number of passages, glial and neuronal cells begin to lose their morphological characteristics, and most importantly, their specific cellular markers and phenotype. In recent years, the discovery of neural progenitor cells, and the methodology to culture them in suspension maintaining their potentiality while still retaining the ability to differentiate into astrocytes, oligodendrocytes and neurons has been a significant contribution to the fields of neuroscience and neuropathology.In the brain, progenitor cells are located in the Germinal Matrix, the subventricular zone in what later would become the basal ganglia, and play an essential role in the homeostasis of the brain by providing the source to replace differentiated cells that have been lost or damaged by different pathological processes, such as senescence, injury, genetic conditions, or disease. The discovery of these neural stem cells in an organ traditionally thought to have limited or no regenerative capacity has opened the door to the development of novel treatments, which include cell replacement therapy. Here we describe the culture and differentiation of neural progenitor cells into neurospheres, and the phenotyping of the resulting cells using immunocytochemistry . The immunocytological methods outlined are not restricted to the analysis of neurosphere-derived cultures but are also applicable for cell typing of primary glial or cell line-derived samples.
Collapse
Affiliation(s)
- Luis Del Valle
- Department of Pathology and Medicine & Louisiana Cancer Research Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
| | - Krzysztof Reiss
- Departments of Medicine & Neurological Cancer Research, Louisiana Cancer Research Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Amanda Parker Struckhoff
- Neurological Cancer Research, Louisiana Cancer Research Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
196
|
Orthotopic brain tumor models derived from glioblastoma stem-like cells. Methods Cell Biol 2022; 170:1-19. [DOI: 10.1016/bs.mcb.2022.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
197
|
Belyashova AS, Galkin MV, Antipina NA, Pavlova GV, Golanov AV. Cell cultures in assessing radioresistance of glioblastomas. ZHURNAL VOPROSY NEIROKHIRURGII IMENI N. N. BURDENKO 2022; 86:126-132. [PMID: 36252203 DOI: 10.17116/neiro202286051126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
To date, no modern methods of treatment allow overcoming malignant potential of glial neoplasms and significant increase of survival. Analysis of glioblastoma radioresistance using cancer cell cultures is one of the perspective directions, as radiotherapy is standard and available treatment method for these neoplasms. This review summarizes current studies identifying many factors of radioresistance of glial tumors, such as hypoxia, microenvironment and metabolic features of tumor, stem cells, internal heterogeneity of tumor, microRNA, features of cell cycle, DNA damage and reparation. We obtained data on involvement of various molecular pathways in development of radioresistance such as MEK/ERK, c-MYC, PI3K/Akt, PTEN, Wnt, JAK/STAT, Notch, etc. Changes in activity of RAD51 APC, FZD1, LEF1, TCF4, WISP1, p53 and many others are determined in radioresistant cells. Further study of radioresistance pathways will allow development of specific target aptamers and inhibitors.
Collapse
Affiliation(s)
| | - M V Galkin
- Burdenko Neurosurgical Center, Moscow, Russia
| | | | - G V Pavlova
- Burdenko Neurosurgical Center, Moscow, Russia
| | - A V Golanov
- Burdenko Neurosurgical Center, Moscow, Russia
| |
Collapse
|
198
|
Zhou Z, Cong L, Cong X. Patient-Derived Organoids in Precision Medicine: Drug Screening, Organoid-on-a-Chip and Living Organoid Biobank. Front Oncol 2021; 11:762184. [PMID: 35036354 PMCID: PMC8755639 DOI: 10.3389/fonc.2021.762184] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Organoids are in vitro self-assembling, organ-like, three-dimensional cellular structures that stably retain key characteristics of the respective organs. Organoids can be generated from healthy or pathological tissues derived from patients. Cancer organoid culture platforms have several advantages, including conservation of the cellular composition that captures the heterogeneity and pharmacotypic signatures of the parental tumor. This platform has provided new opportunities to fill the gap between cancer research and clinical outcomes. Clinical trials have been performed using patient-derived organoids (PDO) as a tool for personalized medical decisions to predict patients' responses to therapeutic regimens and potentially improve treatment outcomes. Living organoid biobanks encompassing several cancer types have been established, providing a representative collection of well-characterized models that will facilitate drug development. In this review, we highlight recent developments in the generation of organoid cultures and PDO biobanks, in preclinical drug discovery, and methods to design a functional organoid-on-a-chip combined with microfluidic. In addition, we discuss the advantages as well as limitations of human organoids in patient-specific therapy and highlight possible future directions.
Collapse
Affiliation(s)
- Zilong Zhou
- Biobank, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Lele Cong
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xianling Cong
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
199
|
Fauß J, Sprang B, Leukel P, Sommer C, Nikolova T, Ringel F, Kim EL. ALDH1A3 Segregated Expression and Nucleus-Associated Proteasomal Degradation Are Common Traits of Glioblastoma Stem Cells. Biomedicines 2021; 10:biomedicines10010007. [PMID: 35052687 PMCID: PMC8772809 DOI: 10.3390/biomedicines10010007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 12/15/2022] Open
Abstract
Aldehyde dehydrogenase 1 isoforms A1 and A3 have been implicated as functional biomarkers associated with distinct molecular subtypes of glioblastoma and glioblastoma stem cells. However, the exact roles of these isoforms in different types of glioma cells remain unclear. The purpose of this study was to dissect the association of A1 or A3 isoforms with stem and non-stem glioblastoma cells. This study has undertaken a systematic characterization of A1 and A3 proteins in glioblastoma tissues and a panel of glioblastoma stem cells using immunocytochemical and immunofluorescence staining, Western blot and the subcellular fractionation methodology. Our main findings are (i) human GSCs express uniformly ALDH1A3 but not the ALDH1A1 isoform whereas non-stem glioma cells comparably express both isoforms; (ii) there is an abundance of ALDH1A3 peptides that prevail over the full-length form in glioblastoma stem cells but not in non-stem glioma cells; (iii) full-length ALDH1A3 and ALDH1A3 peptides are spatially segregated within the cell; and (vi) the abundance of full-length ALDH1A3 and ALDH1A3 peptides is sensitive to MG132-mediated proteasomal inhibition. Our study further supports the association of ALDH1A3 with glioblastoma stem cells and provide evidence for the regulation of ALDH1A3 activities at the level of protein turnover.
Collapse
Affiliation(s)
- Julian Fauß
- Laboratory of Experimental Neurooncology, Department of Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany; (J.F.); (B.S.)
| | - Bettina Sprang
- Laboratory of Experimental Neurooncology, Department of Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany; (J.F.); (B.S.)
| | - Petra Leukel
- Institute of Neuropathology, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany; (P.L.); (C.S.)
| | - Clemens Sommer
- Institute of Neuropathology, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany; (P.L.); (C.S.)
| | - Teodora Nikolova
- Institute of Toxicology, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany;
| | - Florian Ringel
- Department of Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany;
| | - Ella L. Kim
- Laboratory of Experimental Neurooncology, Department of Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany; (J.F.); (B.S.)
- Correspondence:
| |
Collapse
|
200
|
Głów D, Maire CL, Schwarze LI, Lamszus K, Fehse B. CRISPR-to-Kill (C2K)-Employing the Bacterial Immune System to Kill Cancer Cells. Cancers (Basel) 2021; 13:cancers13246306. [PMID: 34944926 PMCID: PMC8699370 DOI: 10.3390/cancers13246306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/01/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Reasoning that multiple DNA breaks will trigger programmed cell death, we generated lentiviral CRISPR-to-kill (C2K) vectors targeting highly repetitive SINE sequences for cancer gene therapy. In proof-of-concept experiments, C2K-Alu-vectors selectively killed human, but not murine cell lines, and efficiently inhibited the growth of patient-derived glioblastoma cell lines resistant to high-dose irradiation. In combination with tumor-targeting approaches, the C2K system might represent a promising tool for cancer gene therapy. Abstract CRISPR/Cas9 was described as a bacterial immune system that uses targeted introduction of DNA double-strand breaks (DSBs) to destroy invaders. We hypothesized that we can analogously employ CRISPR/Cas9 nucleases to kill cancer cells by inducing maximal numbers of DSBs in their genome and thus triggering programmed cell death. To do so, we generated CRISPR-to-kill (C2K) lentiviral particles targeting highly repetitive Short Interspersed Nuclear Element-Alu sequences. Our Alu-specific sgRNA has more than 15,000 perfectly matched target sites within the human genome. C2K-Alu-vectors selectively killed human, but not murine cell lines. More importantly, they efficiently inhibited the growth of cancer cells including patient-derived glioblastoma cell lines resistant to high-dose irradiation. Our data provide proof-of-concept for the potential of C2K as a novel treatment strategy overcoming common resistance mechanisms. In combination with tumor-targeting approaches, the C2K system might therefore represent a promising tool for cancer gene therapy.
Collapse
Affiliation(s)
- Dawid Głów
- Research Department, Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (D.G.); (L.I.S.)
| | - Cecile L. Maire
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (C.L.M.); (K.L.)
| | - Lea Isabell Schwarze
- Research Department, Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (D.G.); (L.I.S.)
| | - Katrin Lamszus
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (C.L.M.); (K.L.)
| | - Boris Fehse
- Research Department, Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (D.G.); (L.I.S.)
- Correspondence: ; Tel.: +49-40-7410-55518; Fax: +49-40-7410-55468
| |
Collapse
|