1
|
Géczi D, Klekner Á, Balogh I, Penyige A, Szilágyi M, Virga J, Bakó A, Nagy B, Torner B, Birkó Z. Identification of Deregulated miRNAs and mRNAs Involved in Tumorigenesis and Detection of Glioblastoma Patients Applying Next-Generation RNA Sequencing. Pharmaceuticals (Basel) 2025; 18:431. [PMID: 40143207 PMCID: PMC11944724 DOI: 10.3390/ph18030431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/10/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
(1) Background: Glioblastoma (GBM) is one of the most aggressive brain tumors with a poor prognosis. Therefore, new insights into GBM diagnosis and treatment are required. In addition to differentially expressed mRNAs, miRNAs may have the potential to be applied as diagnostic biomarkers. (2) Methods: In this study, profiling of human miRNAs in combination with mRNAs was performed on total RNA isolated from tissue samples of five control and five GBM patients, using a high-throughput RNA sequencing (RNA-Seq) approach. (3) Results: A total of 35 miRNAs and 365 mRNAs were upregulated, while 82 miRNAs and 1225 mRNAs showed significant downregulation between tissue samples of GBM patients compared to the control samples using the iDEP tool to analyze RNA-Seq data. To validate our results, the expression of five miRNAs (hsa-miR-196a-5p, hsa-miR-21-3p, hsa-miR-10b-3p, hsa-miR-383-5p, and hsa-miR-490-3p) and fourteen mRNAs (E2F2, HOXD13, VEGFA, CDC45, AURKB, HOXC10, MYBL2, FABP6, PRLHR, NEUROD6, CBLN1, HRH3, HCN1, and RELN) was determined by RT-qPCR assay. The miRNet tool was used to build miRNA-target interaction. Furthermore, a protein-protein interaction (PPI) network was created from the miRNA targets by applying the NetworkAnalyst 3.0 tool. Based on the PPI network, a functional enrichment analysis of the target proteins was also carried out. (4) Conclusions: We identified an miRNA panel and several deregulated mRNAs that could play an important role in tumor development and distinguish GBM patients from healthy controls with high sensitivity and specificity using total RNA isolated from tissue samples.
Collapse
Affiliation(s)
- Dóra Géczi
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (D.G.); (I.B.); (A.P.); (M.S.); (B.N.); (B.T.)
| | - Álmos Klekner
- Department of Neurosurgery, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary;
| | - István Balogh
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (D.G.); (I.B.); (A.P.); (M.S.); (B.N.); (B.T.)
- Division of Clinical Genetics, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - András Penyige
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (D.G.); (I.B.); (A.P.); (M.S.); (B.N.); (B.T.)
| | - Melinda Szilágyi
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (D.G.); (I.B.); (A.P.); (M.S.); (B.N.); (B.T.)
| | - József Virga
- Department of Oncology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (J.V.); (A.B.)
| | - Andrea Bakó
- Department of Oncology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (J.V.); (A.B.)
| | - Bálint Nagy
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (D.G.); (I.B.); (A.P.); (M.S.); (B.N.); (B.T.)
| | - Bernadett Torner
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (D.G.); (I.B.); (A.P.); (M.S.); (B.N.); (B.T.)
| | - Zsuzsanna Birkó
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (D.G.); (I.B.); (A.P.); (M.S.); (B.N.); (B.T.)
| |
Collapse
|
2
|
Sadeghinasab A, Fatahiasl J, Tahmasbi M, Razmjoo S, Yousefipour M. Assessing Glioblastoma Treatment Response Using Machine Learning Approach Based on Magnetic Resonance Images Radiomics: An Exploratory Study. Health Sci Rep 2025; 8:e70323. [PMID: 39741746 PMCID: PMC11683675 DOI: 10.1002/hsr2.70323] [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: 10/01/2024] [Revised: 12/07/2024] [Accepted: 12/18/2024] [Indexed: 01/03/2025] Open
Abstract
Background and Objectives Assessing treatment response in glioblastoma multiforme (GBM) tumors necessitates developing more objective and quantitative approaches. A machine learning-based approach is presented in this exploratory study for GBM patients' treatment response assessment based on radiomics extracted from magnetic resonance (MR) images. Methods MR images from 77 GBM patients were acquired at two post-surgery stages and preprocessed. From these images, 107 radiomics were extracted from the segmented tumoral cavities. The most informative features for training machine learning (ML) classifiers were identified using the Spearman correlation analysis of features retained by the forward sequential and LASSO algorithms. Applied machine learning models included support vector machine (SVM), random forest (RF), K-nearest neighbors (KNN), AdaBoost, categorical boosting (CatBoost), light gradient boosting machine (LightGBM), extreme gradient boosting (XGBoost), Naïve Bayes (NB) and logistic regression (LR). Ten-fold cross-validation was used to validate the models. Statistical analysis was conducted using SPSS version 27; p-value < 0.05 was considered significant. Results The Naïve Bayes classifier demonstrated the highest performance among the trained models, achieving an AUC (area under the receiver operating characteristic curve) of 0.86 ± 0.13 when trained on the seven features selected by the forward sequential algorithm and an AUC of 0.84 ± 0.14 when trained using the five features chosen by the LASSO algorithm. The second-best performance was observed with the KNN classifier, which achieved an AUC of 0.80 ± 0.17 when trained on the features selected by the forward sequential algorithm. Conclusion Findings demonstrated that MRI-based radiomics could be used as distinctive features to train ML models for GBM patients' treatment response assessment. Trained ML classifiers based on these features serve as aiding tools to expedite the quantitative assessment of GBM patients' treatment response besides qualitative evaluations.
Collapse
Affiliation(s)
- Amirreza Sadeghinasab
- Department of Radiologic Technology, School of Allied Medical Sciences, AhvazJundishapur University of Medical SciencesAhvazIran
| | - Jafar Fatahiasl
- Department of Radiologic Technology, School of Allied Medical Sciences, AhvazJundishapur University of Medical SciencesAhvazIran
| | - Marziyeh Tahmasbi
- Department of Radiologic Technology, School of Allied Medical Sciences, AhvazJundishapur University of Medical SciencesAhvazIran
| | - Sasan Razmjoo
- Department of Clinical Oncology and Clinical Research Development Center, Golestan HospitalAhvaz Jundishapur University of Medical SciencesAhvazIran
| | - Mohammad Yousefipour
- Department of Computer Engineering, Faculty of EngineeringShahid Chamran University of AhvazAhvazIran
| |
Collapse
|
3
|
Shetty K, Yadav KS. Temozolomide nano-in-nanofiber delivery system with sustained release and enhanced cellular uptake by U87MG cells. Drug Dev Ind Pharm 2024; 50:420-431. [PMID: 38502031 DOI: 10.1080/03639045.2024.2332906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 03/15/2024] [Indexed: 03/20/2024]
Abstract
OBJECTIVE The study was aimed at formulating temozolomide (TMZ) loaded gelatin nanoparticles (GNPs) encapsulated into polyvinyl alcohol (PVA) nanofibers (TMZ-GNPs-PVA NFs) as the nano-in-nanofiber delivery system. The secondary objective was to explore the sustained releasing ability of this system and to assess its enhanced cellular uptake against U87MG glioma cells in vitro. SIGNIFICANCE Nano-in-nanofibers are the emerging drug delivery systems for treating a wide range of diseases including cancers as they overcome the challenges experienced by nanoparticles and nanofibers alone. METHODS The drug-loaded GNPs were formulated by one-step desolvation method. The Design of Experiments (DoE) was used to optimize nanoparticle size and entrapment efficiency. The optimized drug-loaded nanoparticles were then encapsulated within nanofibers using blend electrospinning technique. The U87MG glioma cells were used to investigate the uptake of the formulation. RESULTS A 32 factorial design was used to optimize the mean particle size (145.7 nm) and entrapment efficiency (87.6%) of the TMZ-loaded GNPs which were subsequently ingrained into PVA nanofibers by electrospinning technique. The delivery system achieved a sustained drug release for up to seven days (in vitro). The SEM results ensured that the expected nano-in-nanofiber delivery system was achieved. The uptake of TMZ-GNPs-PVA NFs by cells was increased by a factor of 1.964 compared to that of the pure drug. CONCLUSION The nano-in-nanofiber drug delivery system is a potentially useful therapeutic strategy for the management of glioblastoma multiforme.
Collapse
Affiliation(s)
- Karishma Shetty
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS (Deemed to be University), Mumbai, India
| | - Khushwant S Yadav
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS (Deemed to be University), Mumbai, India
| |
Collapse
|
4
|
Hei B, Liu RE, Li M. Ursolic acid inhibits glioblastoma through suppressing TGFβ-mediated epithelial-mesenchymal transition (EMT) and angiogenesis. Heliyon 2024; 10:e27722. [PMID: 38501006 PMCID: PMC10945258 DOI: 10.1016/j.heliyon.2024.e27722] [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: 06/06/2023] [Revised: 03/02/2024] [Accepted: 03/06/2024] [Indexed: 03/20/2024] Open
Abstract
Found in many fruits and plants, Ursolic acid (UA), a pentacyclic triterpene that occurs naturally, is recognized for its anti-cancer effects, especially in combating glioblastoma. However, the intricate molecular mechanisms underpinning its anti-tumor actions are still not fully understood, despite the recognition of these effects. By examining the functions of epithelial-mesenchymal transition (EMT) and angiogenesis, crucial for glioblastoma progression, and their regulation through Transforming Growth Factor Beta (TGFβ) - a key marker for glioblastoma, our research aims to fill this knowledge gap. This study explores how ursolic acid can block the progression of glioblastoma by precisely targeting TGFβ-triggered EMT and angiogenesis. The findings show that UA successfully blocks the spread, movement, and invasion of glioblastoma cells. Accompanying this, there is a significant reduction in the expression of TGFβ and crucial EMT indicators like snail and vimentin. Furthermore, UA shows a reduction in angiogenesis that depends on the dosage, highlighted by decreased vascular endothelial growth factor (VEGF) in human umbilical vein endothelial cells (HUVECs). Interestingly, increased TGFβ expression in U87 and U251 glioblastoma cell lines was found to weaken UA's anti-tumor properties, shedding more light on TGFβ's critical function in glioblastoma's pathology. Supporting these laboratory results, UA also showed considerable inhibition of tumor growth in a glioblastoma xenograft mouse model. Overall, our research emphasizes Ursolic acid's promise as a new treatment for glioblastoma and clarifies its action mechanism, mainly by inhibiting TGFβ signaling and thereby EMT and angiogenesis.
Collapse
Affiliation(s)
- Bo Hei
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Jiangxi, China
- Department of Neurosurgery, Peking University People's Hospital, Peking University, Beijing, China
- Department of Neurosurgery, Army General Hospital, Beijing, China
| | - Ru-en Liu
- Department of Neurosurgery, Peking University People's Hospital, Peking University, Beijing, China
| | - Meihua Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Jiangxi, China
| |
Collapse
|
5
|
Kiel K, Król SK, Bronisz A, Godlewski J. MiR-128-3p - a gray eminence of the human central nervous system. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102141. [PMID: 38419943 PMCID: PMC10899074 DOI: 10.1016/j.omtn.2024.102141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
MicroRNA-128-3p (miR-128-3p) is a versatile molecule with multiple functions in the physiopathology of the human central nervous system. Perturbations of miR-128-3p, which is enriched in the brain, contribute to a plethora of neurodegenerative disorders, brain injuries, and malignancies, as this miRNA is a crucial regulator of gene expression in the brain, playing an essential role in the maintenance and function of cells stemming from neuronal lineage. However, the differential expression of miR-128-3p in pathologies underscores the importance of the balance between its high and low levels. Significantly, numerous reports pointed to miR-128-3p as one of the most depleted in glioblastoma, implying it is a critical player in the disease's pathogenesis and thus may serve as a therapeutic agent for this most aggressive form of brain tumor. In this review, we summarize the current knowledge of the diverse roles of miR-128-3p. We focus on its involvement in the neurogenesis and pathophysiology of malignant and neurodegenerative diseases. We also highlight the promising potential of miR-128-3p as an antitumor agent for the future therapy of human cancers, including glioblastoma, and as the linchpin of brain development and function, potentially leading to the development of new therapies for neurological conditions.
Collapse
Affiliation(s)
- Klaudia Kiel
- Tumor Microenvironment Laboratory, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego Street, Warsaw, Poland
| | - Sylwia Katarzyna Król
- Department of Neurooncology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego Street, Warsaw, Poland
| | - Agnieszka Bronisz
- Tumor Microenvironment Laboratory, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego Street, Warsaw, Poland
| | - Jakub Godlewski
- Department of Neurooncology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego Street, Warsaw, Poland
| |
Collapse
|
6
|
Zhang Q, Dai Z, Chen Y, Li Q, Guo Y, Zhu Z, Tu M, Cai L, Lu X. Endosome associated trafficking regulator 1 promotes tumor growth and invasion of glioblastoma multiforme via inhibiting TNF signaling pathway. J Neurooncol 2024; 166:113-127. [PMID: 38191954 DOI: 10.1007/s11060-023-04527-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 11/30/2023] [Indexed: 01/10/2024]
Abstract
PURPOSE Endosome associated trafficking regulator 1 (ENTR1) is a novel endosomal protein, which can affect multiple cellular biological behavior by remodeling plasma membrane structures. However, little is known regarding its function and underlying mechanisms in glioblastoma multiforme. METHODS Expression profile and clinical signature were obtained from The Public Database of human tumor. Immunohistochemical staining and western blotting assays were used to measure ENTR1 expression level. Human primary GBM tumor cells and human GBM cell lines A172, U87 and U251 were used to clarify the precise role of ENTR1. CCK-8 assays, wound healing and transwell invasion assays were designed to investigate cell viability, invasion and migration of GBM cells, respectively. Underlying molecular mechanisms of ENTR1 were determined via RNA-seq analysis. Tumor formation assay was used to validate the influence of ENTR1 in vivo. RESULTS Compared with normal brain tissues, ENTR1 was highly expressed in gliomas and correlated with malignant grades of gliomas and poor overall survival time. The proliferation and invasion of GBM cells could be weaken and the sensitivity to temozolomide (TMZ) chemotherapy increased after knocking down ENTR1. Overexpression of ENTR1 could reverse this effect. RNA-seq analysis showed that tumor necrosis factor (TNF) signaling pathway might be a putative regulatory target of ENTR1. Tumor formation assay validated that ENTR1 was a significant factor in tumor growth. CONCLUSION Our results indicated that ENTR1 played an important role in cell proliferation, invasion and chemotherapeutic sensitivity of GBM, suggesting that ENTR1 might be a novel prognostic marker and significant therapeutic target for GBM.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Zhang'an Dai
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yingyu Chen
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Qun Li
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yuhang Guo
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zhangzhang Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Ming Tu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Lin Cai
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Xianghe Lu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
7
|
Whitehead CA, Morokoff AP, Kaye AH, Drummond KJ, Mantamadiotis T, Stylli SS. Invadopodia associated Thrombospondin-1 contributes to a post-therapy pro-invasive response in glioblastoma cells. Exp Cell Res 2023; 431:113743. [PMID: 37591452 DOI: 10.1016/j.yexcr.2023.113743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/19/2023]
Abstract
A critical challenge in the treatment of glioblastoma (GBM) is its highly invasive nature which promotes cell migration throughout the brain and hinders surgical resection and effective drug delivery. GBM cells demonstrate augmented invasive capabilities following exposure to the current gold standard treatment of radiotherapy (RT) and concomitant and adjuvant temozolomide (TMZ), resulting in rapid disease recurrence. Elucidating the mechanisms employed by post-treatment invasive GBM cells is critical to the development of more effective therapies. In this study, we utilized a Nanostring® Cancer Progression gene expression panel to identify candidate genes that may be involved in enhanced GBM cell invasion after treatment with clinically relevant doses of RT/TMZ. Our findings identified thrombospondin-1 (THBS1) as a pro-invasive gene that is upregulated in these cells. Immunofluorescence staining revealed that THBS1 localised within functional matrix-degrading invadopodia that formed on the surface of GBM cells. Furthermore, overexpression of THBS1 resulted in enhanced GBM cell migration and secretion of MMP-2, which was reduced with silencing of THBS1. The preliminary data demonstrates that THBS1 is associated with invadopodia in GBM cells and is likely involved in the invadopodia-mediated invasive process in GBM cells exposed to RT/TMZ treatment. Therapeutic inhibition of THBS1-mediated invadopodia activity, which facilitates GBM cell invasion, should be further investigated as a treatment for GBM.
Collapse
Affiliation(s)
- Clarissa A Whitehead
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Andrew P Morokoff
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia; Department of Neurosurgery, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Andrew H Kaye
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia; Department of Neurosurgery, Hadassah Hebrew University Medical Centre, Jerusalem, Israel
| | - Katharine J Drummond
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia; Department of Neurosurgery, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Theo Mantamadiotis
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia; Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC, Australia; The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Stanley S Stylli
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia; Department of Neurosurgery, Royal Melbourne Hospital, Parkville, VIC, Australia.
| |
Collapse
|
8
|
Dreyer CA, VanderVorst K, Natwick D, Bell G, Sood P, Hernandez M, Angelastro JM, Collins SR, Carraway KL. A complex of Wnt/planar cell polarity signaling components Vangl1 and Fzd7 drives glioblastoma multiforme malignant properties. Cancer Lett 2023; 567:216280. [PMID: 37336284 PMCID: PMC10582999 DOI: 10.1016/j.canlet.2023.216280] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/21/2023]
Abstract
Targeting common oncogenic drivers of glioblastoma multiforme (GBM) in patients has remained largely ineffective, raising the possibility that alternative pathways may contribute to tumor aggressiveness. Here we demonstrate that Vangl1 and Fzd7, components of the non-canonical Wnt planar cell polarity (Wnt/PCP) signaling pathway, promote GBM malignancy by driving cellular proliferation, migration, and invasiveness, and engage Rho GTPases to promote cytoskeletal rearrangements and actin dynamics in migrating GBM cells. Mechanistically, we uncover the existence of a novel Vangl1/Fzd7 complex at the leading edge of migrating GBM cells and propose that this complex is critical for the recruitment of downstream effectors to promote tumor progression. Moreover, we observe that depletion of FZD7 results in a striking suppression of tumor growth and latency and extends overall survival in an intracranial mouse xenograft model. Our observations support a novel mechanism by which Wnt/PCP components Vangl1 and Fzd7 form a complex at the leading edge of migratory GBM cells to engage downstream effectors that promote actin cytoskeletal rearrangements dynamics. Our findings suggest that interference with Wnt/PCP pathway function may offer a novel therapeutic strategy for patients diagnosed with GBM.
Collapse
Affiliation(s)
- Courtney A Dreyer
- Department of Biochemistry and Molecular Medicine and University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Kacey VanderVorst
- Department of Biochemistry and Molecular Medicine and University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Dean Natwick
- Department of Microbiology and Molecular Genetics, University of California Davis, Davis, CA, USA
| | - George Bell
- Department of Microbiology and Molecular Genetics, University of California Davis, Davis, CA, USA
| | - Prachi Sood
- Department of Biochemistry and Molecular Medicine and University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Maria Hernandez
- Department of Biochemistry and Molecular Medicine and University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - James M Angelastro
- Department of Molecular Biosciences, University of California Davis School of Veterinary Medicine, Davis, CA, USA
| | - Sean R Collins
- Department of Microbiology and Molecular Genetics, University of California Davis, Davis, CA, USA
| | - Kermit L Carraway
- Department of Biochemistry and Molecular Medicine and University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
9
|
Kringel R, Lamszus K, Mohme M. Chimeric Antigen Receptor T Cells in Glioblastoma-Current Concepts and Promising Future. Cells 2023; 12:1770. [PMID: 37443804 PMCID: PMC10340625 DOI: 10.3390/cells12131770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/20/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Glioblastoma (GBM) is a highly aggressive primary brain tumor that is largely refractory to treatment and, therefore, invariably relapses. GBM patients have a median overall survival of 15 months and, given this devastating prognosis, there is a high need for therapy improvement. One of the therapeutic approaches currently tested in GBM is chimeric antigen receptor (CAR)-T cell therapy. CAR-T cells are genetically altered T cells that are redirected to eliminate tumor cells in a highly specific manner. There are several challenges to CAR-T cell therapy in solid tumors such as GBM, including restricted trafficking and penetration of tumor tissue, a highly immunosuppressive tumor microenvironment (TME), as well as heterogeneous antigen expression and antigen loss. In addition, CAR-T cells have limitations concerning safety, toxicity, and the manufacturing process. To date, CAR-T cells directed against several target antigens in GBM including interleukin-13 receptor alpha 2 (IL-13Rα2), epidermal growth factor receptor variant III (EGFRvIII), human epidermal growth factor receptor 2 (HER2), and ephrin type-A receptor 2 (EphA2) have been tested in preclinical and clinical studies. These studies demonstrated that CAR-T cell therapy is a feasible option in GBM with at least transient responses and acceptable adverse effects. Further improvements in CAR-T cells regarding their efficacy, flexibility, and safety could render them a promising therapy option in GBM.
Collapse
Affiliation(s)
| | | | - Malte Mohme
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (R.K.); (K.L.)
| |
Collapse
|
10
|
Tritz ZP, Ayasoufi K, Wolf DM, Owens CA, Malo CS, Himes BT, Fain CE, Goddery EN, Yokanovich LT, Jin F, Hansen MJ, Parney IF, Wang C, Moynihan KD, Irvine DJ, Wittrup KD, Marcano RMD, Vile RG, Johnson AJ. Anti-PD-1 and Extended Half-life IL2 Synergize for Treatment of Murine Glioblastoma Independent of Host MHC Class I Expression. Cancer Immunol Res 2023; 11:763-776. [PMID: 36921098 PMCID: PMC10239322 DOI: 10.1158/2326-6066.cir-22-0570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 01/20/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023]
Abstract
Glioblastoma (GBM) is the most common malignant brain tumor in adults, responsible for approximately 225,000 deaths per year. Despite preclinical successes, most interventions have failed to extend patient survival by more than a few months. Treatment with anti-programmed cell death protein 1 (anti-PD-1) immune checkpoint blockade (ICB) monotherapy has been beneficial for malignant tumors such as melanoma and lung cancers but has yet to be effectively employed in GBM. This study aimed to determine whether supplementing anti-PD-1 ICB with engineered extended half-life IL2, a potent lymphoproliferative cytokine, could improve outcomes. This combination therapy, subsequently referred to as enhanced checkpoint blockade (ECB), delivered intraperitoneally, reliably cures approximately 50% of C57BL/6 mice bearing orthotopic GL261 gliomas and extends median survival of the treated cohort. In the CT2A model, characterized as being resistant to CBI, ECB caused a decrease in CT2A tumor volume in half of measured animals similar to what was observed in GL261-bearing mice, promoting a trending survival increase. ECB generates robust immunologic responses, features of which include secondary lymphoid organ enlargement and increased activation status of both CD4 and CD8 T cells. This immunity is durable, with long-term ECB survivors able to resist GL261 rechallenge. Through employment of depletion strategies, ECB's efficacy was shown to be independent of host MHC class I-restricted antigen presentation but reliant on CD4 T cells. These results demonstrate ECB is efficacious against the GL261 glioma model through an MHC class I-independent mechanism and supporting further investigation into IL2-supplemented ICB therapies for tumors of the central nervous system.
Collapse
Affiliation(s)
| | | | | | | | - Courtney S. Malo
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN
| | - Benjamin T. Himes
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN
- Mayo Clinic Department of Neurologic Surgery, Rochester, MN
| | - Cori E. Fain
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN
| | - Emma N. Goddery
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN
| | | | - Fang Jin
- Mayo Clinic Department of Immunology, Rochester, MN
| | | | - Ian F. Parney
- Mayo Clinic Department of Immunology, Rochester, MN
- Mayo Clinic Department of Neurologic Surgery, Rochester, MN
| | - Chensu Wang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
| | - Kelly D. Moynihan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Darrell J. Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA
- Howard Hughes Medical Institute, Chevy Chase, MD
| | - K. Dane Wittrup
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | | | - Richard G. Vile
- Mayo Clinic Department of Immunology, Rochester, MN
- Mayo Clinic Department of Molecular Medicine, Rochester, MN
| | - Aaron J. Johnson
- Mayo Clinic Department of Immunology, Rochester, MN
- Mayo Clinic Department of Molecular Medicine, Rochester, MN
- Mayo Clinic Department of Neurology, Rochester, MN
| |
Collapse
|
11
|
Panovska D, Nazari P, Cole B, Creemers PJ, Derweduwe M, Solie L, Van Gassen S, Claeys A, Verbeke T, Cohen EF, Tolstorukov MY, Saeys Y, Van der Planken D, Bosisio FM, Put E, Bamps S, Clement PM, Verfaillie M, Sciot R, Ligon KL, De Vleeschouwer S, Antoranz A, De Smet F. Single-cell molecular profiling using ex vivo functional readouts fuels precision oncology in glioblastoma. Cell Mol Life Sci 2023; 80:147. [PMID: 37171617 PMCID: PMC11071868 DOI: 10.1007/s00018-023-04772-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/06/2023] [Accepted: 03/29/2023] [Indexed: 05/13/2023]
Abstract
BACKGROUND Functional profiling of freshly isolated glioblastoma (GBM) cells is being evaluated as a next-generation method for precision oncology. While promising, its success largely depends on the method to evaluate treatment activity which requires sufficient resolution and specificity. METHODS Here, we describe the 'precision oncology by single-cell profiling using ex vivo readouts of functionality' (PROSPERO) assay to evaluate the intrinsic susceptibility of high-grade brain tumor cells to respond to therapy. Different from other assays, PROSPERO extends beyond life/death screening by rapidly evaluating acute molecular drug responses at single-cell resolution. RESULTS The PROSPERO assay was developed by correlating short-term single-cell molecular signatures using mass cytometry by time-of-flight (CyTOF) to long-term cytotoxicity readouts in representative patient-derived glioblastoma cell cultures (n = 14) that were exposed to radiotherapy and the small-molecule p53/MDM2 inhibitor AMG232. The predictive model was subsequently projected to evaluate drug activity in freshly resected GBM samples from patients (n = 34). Here, PROSPERO revealed an overall limited capacity of tumor cells to respond to therapy, as reflected by the inability to induce key molecular markers upon ex vivo treatment exposure, while retaining proliferative capacity, insights that were validated in patient-derived xenograft (PDX) models. This approach also allowed the investigation of cellular plasticity, which in PDCLs highlighted therapy-induced proneural-to-mesenchymal (PMT) transitions, while in patients' samples this was more heterogeneous. CONCLUSION PROSPERO provides a precise way to evaluate therapy efficacy by measuring molecular drug responses using specific biomarker changes in freshly resected brain tumor samples, in addition to providing key functional insights in cellular behavior, which may ultimately complement standard, clinical biomarker evaluations.
Collapse
Affiliation(s)
- Dena Panovska
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Pouya Nazari
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Basiel Cole
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Pieter-Jan Creemers
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Marleen Derweduwe
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Lien Solie
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
- Department of Neurosurgery, University Hospitals (UZ) Leuven, Leuven, Belgium
- Laboratory of Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Sofie Van Gassen
- Data Mining and Modeling for Biomedicine Group, VIB Inflammation Research Center, Ghent University, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Annelies Claeys
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Tatjana Verbeke
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Elizabeth F Cohen
- Department of Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael Y Tolstorukov
- Department of Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Yvan Saeys
- Data Mining and Modeling for Biomedicine Group, VIB Inflammation Research Center, Ghent University, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | | | - Francesca M Bosisio
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Eric Put
- Neurosurgery Department, Faculty of Medicine and Life Sciences UHasselt, Hasselt, Belgium
| | - Sven Bamps
- Neurosurgery Department, Faculty of Medicine and Life Sciences UHasselt, Hasselt, Belgium
| | - Paul M Clement
- Department of Oncology, KU Leuven/UZ Leuven, Leuven, Belgium
| | - Michiel Verfaillie
- Europaziekenhuizen, Cliniques de l'Europe, Sint-Elisabeth, Brussels, Belgium
| | - Raf Sciot
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Keith L Ligon
- Department of Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Steven De Vleeschouwer
- Department of Neurosurgery, University Hospitals (UZ) Leuven, Leuven, Belgium
- Laboratory of Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Asier Antoranz
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium
| | - Frederik De Smet
- Department of Imaging and Pathology, KU Leuven, Herestraat 49, Box 1032, Leuven, Belgium.
- Leuven Institute for single-cell omics (LISCO), Leuven, Belgium.
| |
Collapse
|
12
|
Bhaduri S, Kelly CL, Lesbats C, Sharkey J, Ressel L, Mukherjee S, Platt MD, Delikatny EJ, Poptani H. Metabolic changes in glioblastomas in response to choline kinase inhibition: In vivo MRS in rodent models. NMR IN BIOMEDICINE 2023; 36:e4855. [PMID: 36269130 PMCID: PMC10078495 DOI: 10.1002/nbm.4855] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 06/16/2023]
Abstract
Changes in glioblastoma (GBM) metabolism was investigated in response to JAS239, a choline kinase inhibitor, using MRS. In addition to the inhibition of phosphocholine synthesis, we investigated changes in other key metabolic pathways associated with GBM progression and treatment response. Three syngeneic rodent models of GBM were used: F98 (N = 12) and 9L (N = 8) models in rats and GL261 (N = 10) in mice. Rodents were intracranially injected with GBM cells in the right cortex and tumor growth was monitored using T2 -weighted images. Animals were treated once daily with intraperitoneal injections of 4 mg/kg JAS239 (F98 rats, n = 6; 9L rats, n = 6; GL261 mice, n = 5) or saline (control group, F98 rats, n = 6; 9L rats, n = 2; GL261 mice, n = 5) for five consecutive days. Single voxel spectra were acquired on Days 0 (T0, baseline) and 6 (T6, end of treatment) from the tumor as well as the contralateral normal brain using a PRESS sequence. Changes in metabolite ratios (tCho/tCr, tCho/NAA, mI/tCr, Glx/tCr and (Lip + Lac)/Cr) were used to assess metabolic pathway alterations in response to JAS239. Tumor growth arrest was noted in all models in response to JAS239 treatment compared with saline-treated animals, with a significant reduction (p < 0.05) in the F98 model. A reduction in tCho/tCr was observed with JAS239 treatment in all GBM models, indicating reduced phospholipid metabolism, with the highest reduction in 9L followed by GL261 and F98 tumors. A significant reduction (p < 0.05) in the tCho/NAA ratio was observed in the 9L model. A significant reduction in mI/tCr (p < 0.05) was found in JAS239-treated F98 tumors compared with the saline-treated animals. A non-significant trend of reduction in Glx/tCr was observed only in F98 and 9L tumors. JAS239-treated F98 tumors also showed a significant increase in Lip + Lac (p < 0.05), indicating increased cell death. This study demonstrated the utility of MRS in assessing metabolic changes in GBM in response to choline kinase inhibition.
Collapse
Affiliation(s)
- Sourav Bhaduri
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| | - Claire Louise Kelly
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| | - Clémentine Lesbats
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
- Division of Radiotherapy and ImagingThe Institute of Cancer ResearchLondonUK
| | - Jack Sharkey
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| | - Lorenzo Ressel
- Department of Veterinary Anatomy Physiology and PathologyUniversity of LiverpoolChesterUK
| | - Soham Mukherjee
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| | - Mark David Platt
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| | - Edward J. Delikatny
- Department of Radiology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Harish Poptani
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| |
Collapse
|
13
|
Riva M, Wouters R, Nittner D, Ceusters J, Sterpin E, Giovannoni R, Himmelreich U, Gsell W, VAN Ranst M, Coosemans A. Radiation dose-escalation and dose-fractionation modulate the immune microenvironment, cancer stem cells and vasculature in experimental high-grade gliomas. J Neurosurg Sci 2023; 67:55-65. [PMID: 33056947 DOI: 10.23736/s0390-5616.20.05060-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND In the context of high-grade gliomas (HGGs), very little evidence is available concerning the optimal radiotherapy (RT) schedule to be used in radioimmunotherapy combinations. This studied was aimed at shedding new light in this field by analyzing the effects of RT dose escalation and dose fractionation on the tumor microenvironment of experimental HGGs. METHODS Neurospheres (NS) CT-2A HGG-bearing C57BL/6 mice were treated with stereotactic RT. For dose-escalation experiments, mice received 2, 4 or 8 Gy as single administrations. For dose-fractionation experiments, mice received 4 Gy as a single fraction or multiple (1.33x3 Gy) fractions. The impact of the RT schedule on murine survival and tumor immunity was evaluated. Modifications of glioma stem cells (GSCs), tumor vasculature and tumor cell replication were also assessed. RESULTS RT dose-escalation was associated with an improved immune profile, with higher CD8+ T cells and CD8+ T cells/regulatory T cells (Tregs) ratio (P=0.0003 and P=0.0022, respectively) and lower total tumor associated microglia/macrophages (TAMs), M2 TAMs and monocytic myeloid derived suppressor cells (mMDSCs) (P=0.0011, P=0.0024 and P<0.0001, respectively). The progressive increase of RT dosages prolonged survival (P<0.0001) and reduced tumor vasculature (P=0.069), tumor cell proliferation (P<0.0001) and the amount of GSCs (P=0.0132 or lower). Compared to the unfractionated regimen, RT dose-fractionation negatively affected tumor immunity by inducing higher total TAMs, M2 TAMs and mMDSCs (P=0.0051, P=0.0036 and P=0.0436, respectively). Fractionation also induced a shorter survival (P=0.0078), a higher amount of GSCs (P=0.0015 or lower) and a higher degree of tumor cell proliferation (P=0.0003). CONCLUSIONS This study demonstrates that RT dosage and fractionation significantly influence survival, tumor immunity and GSCs in experimental HGGs. These findings should be taken into account when aiming at designing more synergistic and effective radio-immunotherapy combinations.
Collapse
Affiliation(s)
- Matteo Riva
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Catholic University of Leuven, Leuven, Belgium - .,Department of Neurosurgery, UcL Namur, Mont-Godinne University Hospital, Yvoir, Belgium -
| | - Roxanne Wouters
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Catholic University of Leuven, Leuven, Belgium
| | - David Nittner
- Center for the Biology of Disease, Catholic University of Leuven Center for Human Genetics - InfraMouse, VIB, Catholic University of University of Leuven, Leuven, Belgium
| | - Jolien Ceusters
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Catholic University of Leuven, Leuven, Belgium
| | - Edmond Sterpin
- Laboratory of Experimental Radiotherapy, Department of Oncology, Catholic University of Leuven, Leuven, Belgium
| | | | - Uwe Himmelreich
- Biomedical MRI, Department of Imaging and Pathology and Molecular Small Animal Imaging Center (MoSAIC), Catholic University of Leuven, Leuven, Belgium
| | - Willy Gsell
- Biomedical MRI, Department of Imaging and Pathology and Molecular Small Animal Imaging Center (MoSAIC), Catholic University of Leuven, Leuven, Belgium
| | - Marc VAN Ranst
- Laboratory for Clinical and Epidemiological Virology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - An Coosemans
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Catholic University of Leuven, Leuven, Belgium.,Department of Gynecology and Obstetrics, Leuven Cancer Institute, UZ Leuven, Leuven, Belgium
| |
Collapse
|
14
|
Wang M, Malfanti A, Bastiancich C, Préat V. Synergistic effect of doxorubicin lauroyl hydrazone derivative delivered by α-tocopherol succinate micelles for the treatment of glioblastoma. Int J Pharm X 2022; 5:100147. [PMID: 36620521 PMCID: PMC9813532 DOI: 10.1016/j.ijpx.2022.100147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
We hypothesized that tocopherol succinate (TOS) and D-α-tocopherol polyethylene2000 succinate (TPGS2000) micelles could work as a drug delivery system while enhancing the anti-cancer efficacy of doxorubicin lauryl hydrazone derivative (DOXC12) for the treatment of glioblastoma. The DOXC12-TOS-TPGS2000 micelles were formulated with synthesized DOXC12 and TPGS2000. They showed a high drug loading of hydrophobic DOXC12 (29%), a size of <100 nm and a pH sensitive drug release behaviour. In vitro, fast uptake of DOXC12-TOS-TPGS2000 micelles by GL261 cells was observed. For cytotoxicity, DOXC12-TOS-TPGS2000 micelles were evaluated on two glioblastoma cell lines and showed synergism between DOXC12 and TOS-TPGS2000. The higher cytotoxicity of DOXC12-TOS-TPGS2000 micelles was mainly caused by necrosis. The DOXC12-TOS-TPGS2000 micelles seem to be a promising delivery system for enhancing the anticancer efficacy of doxorubicin in glioblastoma (GBM).
Collapse
Affiliation(s)
- Mingchao Wang
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Alessio Malfanti
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Chiara Bastiancich
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium,Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France,Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Véronique Préat
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium,Corresponding author.
| |
Collapse
|
15
|
Wang W, Zhang M, Zhang Q, Mohammadniaei M, Shen J, Sun Y. Brain-targeted antigen-generating nanoparticles improve glioblastoma prognosis. J Control Release 2022; 352:399-410. [PMID: 36309097 DOI: 10.1016/j.jconrel.2022.10.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 10/10/2022] [Accepted: 10/20/2022] [Indexed: 11/07/2022]
Abstract
The exploration of multifunctional nanomedicine has prompted interest in improving glioblastoma (GBM) prognosis. In this study, we constructed tumor microenvironment (TME)-responsive magnetic therapeutic nanoparticles (BK@MTNPs) as a multifunctional drug delivery platform. It contains the following components. [Des-arg(Sheets et al., 2020 [9])]bradykinin (BK), which contributes to the transient opening of the blood-brain barrier (BBB) and targeting of GBM cells; nanoparticles (NPs) encapsulated in MTNPs, which act as an in vivo magnetic resonance (MR) imaging agent; crizotinib, which is an inhibitor of protein kinase c-Met; and the immune drug anti-PDL1 antibody. These components were loaded into BK@MTNPs for complete tumoricidal effects. Abundant glutathione in the TME can promote BK@MTNP degradation by interrupting the disulfide bonds between cysteine residues. Such BK@MTNPs support a synergistic tumoricidal effect by inducing DNA damage, activating the transcription of the tumor suppressor gene PTEN, inhibiting glioblastoma stem cell function, activating cytotoxic T lymphocytes, and reprogramming tumor-associated macrophages. BK@MTNPs showed a significant increase in antitumor activity compared with free drugs in vitro. Furthermore, in mice bearing orthotopic GBM, treatment with BK@MTNPs resulted in marked tumor inhibition and greatly extended survival time with minimal side effects. This study demonstrates the advantages of chemo-immunotherapeutic NPs accumulated in the GBM area and their effective inhibition of GBM growth, thus establishing a delivery platform to promote antitumor immunity against GBM.
Collapse
Affiliation(s)
- Wentao Wang
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Ming Zhang
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark; School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China.
| | - Qicheng Zhang
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China
| | - Mohsen Mohammadniaei
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Jian Shen
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China
| | - Yi Sun
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark.
| |
Collapse
|
16
|
Bhere D, Choi SH, van de Donk P, Hope D, Gortzak K, Kunnummal A, Khalsa J, Revai Lechtich E, Reinshagen C, Leon V, Nissar N, Bi WL, Feng C, Li H, Zhang YS, Liang SH, Vasdev N, Essayed WI, Quevedo PV, Golby A, Banouni N, Palagina A, Abdi R, Fury B, Smirnakis S, Lowe A, Reeve B, Hiller A, Chiocca EA, Prestwich G, Wakimoto H, Bauer G, Shah K. Target receptor identification and subsequent treatment of resected brain tumors with encapsulated and engineered allogeneic stem cells. Nat Commun 2022; 13:2810. [PMID: 35589724 PMCID: PMC9120173 DOI: 10.1038/s41467-022-30558-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/03/2022] [Indexed: 11/09/2022] Open
Abstract
Cellular therapies offer a promising therapeutic strategy for the highly malignant brain tumor, glioblastoma (GBM). However, their clinical translation is limited by the lack of effective target identification and stringent testing in pre-clinical models that replicate standard treatment in GBM patients. In this study, we show the detection of cell surface death receptor (DR) target on CD146-enriched circulating tumor cells (CTC) captured from the blood of mice bearing GBM and patients diagnosed with GBM. Next, we developed allogeneic "off-the-shelf" clinical-grade bifunctional mesenchymal stem cells (MSCBif) expressing DR-targeted ligand and a safety kill switch. We show that biodegradable hydrogel encapsulated MSCBif (EnMSCBif) has a profound therapeutic efficacy in mice bearing patient-derived invasive, primary and recurrent GBM tumors following surgical resection. Activation of the kill switch enhances the efficacy of MSCBif and results in their elimination post-tumor treatment which can be tracked by positron emission tomography (PET) imaging. This study establishes a foundation towards a clinical trial of EnMSCBif in primary and recurrent GBM patients.
Collapse
Affiliation(s)
- Deepak Bhere
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29201, USA
| | - Sung Hugh Choi
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Pim van de Donk
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - David Hope
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Kiki Gortzak
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Amina Kunnummal
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jasneet Khalsa
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Esther Revai Lechtich
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Clemens Reinshagen
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Victoria Leon
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Nabil Nissar
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Wenya Linda Bi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Cheng Feng
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Hongbin Li
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yu Shrike Zhang
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Steven H Liang
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Neil Vasdev
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Walid Ibn Essayed
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Pablo Valdes Quevedo
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Alexandra Golby
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Naima Banouni
- Department of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Anna Palagina
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Reza Abdi
- Department of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Brian Fury
- UC Davis Institute for Regenerative Cures, Davis, CA, 95817, USA
| | - Stelios Smirnakis
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Alarice Lowe
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Brock Reeve
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Arthur Hiller
- Amasa Therapeutics Inc., 1 Harmony Lane, Andover, MA, 01810, USA
| | - E Antonio Chiocca
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Glenn Prestwich
- Department of Medicinal Chemistry, College of Pharmacy University of Utah, Salt Lake City, UT, 84112, USA
- Washington State University Health Sciences, Spokane, WA, 99202, USA
| | - Hiroaki Wakimoto
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Gerhard Bauer
- UC Davis Institute for Regenerative Cures, Davis, CA, 95817, USA
| | - Khalid Shah
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA.
| |
Collapse
|
17
|
Zhang H, Zhao H, Wang H, Yin Z, Huang K, Yu M. High PLA2 level is correlated with glioblastoma progression via regulating DNA replication. J Cell Mol Med 2022; 26:1466-1472. [PMID: 35166019 PMCID: PMC8899163 DOI: 10.1111/jcmm.17140] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 11/21/2021] [Accepted: 12/01/2021] [Indexed: 12/05/2022] Open
Abstract
Phospholipases A2 (PLA2) are a superfamily of enzymes, playing a critical role in the development of various human cancers. However, the mechanism of PLA2 as an oncogene in glioblastoma remains largely unknown. In this study, we explored the effects of PLA2 on glioblastoma and investigated the underlying mechanism. The results showed that PLA2 was highly expressed in glioblastoma. Patients with a high PLA2 level have low overall survival than those with low PLA2 expression. PLA2 overexpression promoted glioblastoma cell proliferation and viability and inhibited cell apoptosis by inducing cell cycle transition from G1 to S stage. Knockdown of PLA2 inhibited tumor growth in the xenograft mice model. In addition, PLA2 knockdown decreased the protein level of MCM2 and MCM5. These findings identify PLA2 as an oncogene in glioblastoma progression and provide a promising strategy to treat glioblastoma in the future.
Collapse
Affiliation(s)
- Haiyun Zhang
- Department of Laboratory Medicine, The Sixth People's Hospital of Nantong, Jiangsu, China
| | - Hanwei Zhao
- Department of Critical Care Medicine, 902 Hospital of PLA, Bengbu, China
| | - Hongliang Wang
- Department of Laboratory Medicine, The Sixth People's Hospital of Nantong, Jiangsu, China
| | - Zhongbo Yin
- Department of Laboratory Medicine, The Sixth People's Hospital of Nantong, Jiangsu, China
| | - Kai Huang
- Department of Orthopaedics, Changshu No. 2 People's Hospital (The 5th Clinical Medical College of Yangzhou University), Changshu, China
| | - Minhong Yu
- Department of Laboratory Medicine, The Sixth People's Hospital of Nantong, Jiangsu, China.,Medical Laboratory Department, Daqing people's Hospital of Heilongjiang Province, Daqing, China
| |
Collapse
|
18
|
Berg AL, Rowson-Hodel A, Hu M, Keeling M, Wu H, VanderVorst K, Chen JJ, Hatakeyama J, Jilek J, Dreyer CA, Wheeler MR, Yu AM, Li Y, Carraway KL. The Cationic Amphiphilic Drug Hexamethylene Amiloride Eradicates Bulk Breast Cancer Cells and Therapy-Resistant Subpopulations with Similar Efficiencies. Cancers (Basel) 2022; 14:cancers14040949. [PMID: 35205696 PMCID: PMC8869814 DOI: 10.3390/cancers14040949] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/02/2022] [Accepted: 02/02/2022] [Indexed: 12/07/2022] Open
Abstract
The resistance of cancer cell subpopulations, including cancer stem cell (CSC) populations, to apoptosis-inducing chemotherapeutic agents is a key barrier to improved outcomes for cancer patients. The cationic amphiphilic drug hexamethylene amiloride (HMA) has been previously demonstrated to efficiently kill bulk breast cancer cells independent of tumor subtype or species but acts poorly toward non-transformed cells derived from multiple tissues. Here, we demonstrate that HMA is similarly cytotoxic toward breast CSC-related subpopulations that are resistant to conventional chemotherapeutic agents, but poorly cytotoxic toward normal mammary stem cells. HMA inhibits the sphere-forming capacity of FACS-sorted human and mouse mammary CSC-related cells in vitro, specifically kills tumor but not normal mammary organoids ex vivo, and inhibits metastatic outgrowth in vivo, consistent with CSC suppression. Moreover, HMA inhibits viability and sphere formation by lung, colon, pancreatic, brain, liver, prostate, and bladder tumor cell lines, suggesting that its effects may be applicable to multiple malignancies. Our observations expose a key vulnerability intrinsic to cancer stem cells and point to novel strategies for the exploitation of cationic amphiphilic drugs in cancer treatment.
Collapse
Affiliation(s)
- Anastasia L. Berg
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Ashley Rowson-Hodel
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Michelle Hu
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Michael Keeling
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Hao Wu
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Kacey VanderVorst
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Jenny J. Chen
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Jason Hatakeyama
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Joseph Jilek
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Courtney A. Dreyer
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Madelyn R. Wheeler
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Kermit L. Carraway
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Correspondence:
| |
Collapse
|
19
|
Kasapidou PM, de Montullé EL, Dembélé KP, Mutel A, Desrues L, Gubala V, Castel H. Hyaluronic acid-based hydrogels loaded with chemoattractant and anticancer drug - new formulation for attracting and tackling glioma cells. SOFT MATTER 2021; 17:10846-10861. [PMID: 34806746 DOI: 10.1039/d1sm01003d] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Over the last few years, significant interest has emerged in the development of localised therapeutic strategies for the treatment of glioblastoma (GBM). The concept of attracting and trapping residual tumour cells within a confined area to facilitate their eradication has developed progressively. Herein, we propose a new design of hyaluronic acid-based hydrogel which can be utilized as a matrix containing a soluble chemoattractant to attract residual glioma cells and chemotherapeutic agents to eradicate them in a less invasive and more efficient way compared to the currently available methods. Hydrogels were prepared at different crosslinking densities, e.g. low and high density, by crosslinking hyaluronic acid with various concentrations of adipic acid dihydrazide and U87MG GBM cell morphology, survival and CD44 expression were evaluated. As a proof-of-concept, hydrogels were loaded with a small peptide chemokine, human urotensin II (hUII), and the migration and survival of U87MG GBM cells were studied. Chemoattractant-containing hydrogels were also loaded with chemotherapeutic drugs to promote cell death in culture. The results showed that U87MG cells were able to invade the hydrogel network and to migrate in response to the chemoattractant hUII. In addition, in static condition, hydrogels loaded with doxorubicin demonstrated significant cytotoxicity leading to less than 80% U87MG cell viability after 48 hours when compared to the control sample. In addition, in in vitro invasive assays, it was originally shown that the chemoattractant effect of hUII can be effective before the cytotoxic action of doxorubicin on the U87MG cells trapped in the hydrogel. Our results provide new insights into a promising approach which can be readily translated in vivo for the treatment of one of the most devastating brain tumours.
Collapse
Affiliation(s)
- Paraskevi M Kasapidou
- Medway School of Pharmacy, University of Kent, Central Avenue, Chatham, ME4 4TB, UK
- Normandie Univ, UNIROUEN, INSERM U1239, DC2N, 76000 Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
| | - Emmanuel Laillet de Montullé
- Normandie Univ, UNIROUEN, INSERM U1239, DC2N, 76000 Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
| | - Kleouforo-Paul Dembélé
- Normandie Univ, UNIROUEN, INSERM U1239, DC2N, 76000 Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
| | - Alexandre Mutel
- Normandie Univ, UNIROUEN, INSERM U1239, DC2N, 76000 Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
| | - Laurence Desrues
- Normandie Univ, UNIROUEN, INSERM U1239, DC2N, 76000 Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
| | - Vladimir Gubala
- Medway School of Pharmacy, University of Kent, Central Avenue, Chatham, ME4 4TB, UK
| | - Hélène Castel
- Normandie Univ, UNIROUEN, INSERM U1239, DC2N, 76000 Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
| |
Collapse
|
20
|
Bouché M, Dong YC, Sheikh S, Taing K, Saxena D, Hsu JC, Chen MH, Salinas RD, Song H, Burdick JA, Dorsey J, Cormode DP. Novel Treatment for Glioblastoma Delivered by a Radiation Responsive and Radiopaque Hydrogel. ACS Biomater Sci Eng 2021; 7:3209-3220. [PMID: 34160196 PMCID: PMC8411482 DOI: 10.1021/acsbiomaterials.1c00385] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Successful treatment of glioblastoma (GBM) is hampered by primary tumor recurrence after surgical resection and poor prognosis, despite adjuvant radiotherapy and chemotherapy. In search of improved outcomes for this disease, quisinostat appeared as a lead compound in drug screening. A delivery system was devised for this drug and to exploit current clinical methodology: an injectable hydrogel, loaded with both the quisinostat drug and radiopaque gold nanoparticles (AuNP) as contrast agent, that can release these payloads as a response to radiation. This hydrogel grants high local drug concentrations, overcoming issues with current standards of care. Significant hydrogel degradation and quisinostat release were observed due to the radiation trigger, providing high in vitro anticancer activity. In vivo, the combination of radiotherapy and the radiation-induced delivery of quisinostat from the hydrogel, successfully inhibited tumor growth in a mice model bearing xenografted human GBM tumors with a total response rate of 67%. Long-term tolerability was observed after intratumoral injection of the quisinostat loaded hydrogel. The AuNP payload enabled precise image-guided radiation delivery and the monitoring of hydrogel degradation using computed tomography (CT). These exciting results highlight this hydrogel as a versatile imageable drug delivery platform that can be activated simultaneously to radiation therapy and potentially offers improved treatment for GBM.
Collapse
Affiliation(s)
- Mathilde Bouché
- Department of Radiology, University of Pennsylvania, 3400 Spruce Street, 1 Silverstein Philadelphia, Pennsylvania 19104, United States
| | - Yuxi C Dong
- Department of Radiology, University of Pennsylvania, 3400 Spruce Street, 1 Silverstein Philadelphia, Pennsylvania 19104, United States
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Saad Sheikh
- Department of Radiation Oncology, University of Pennsylvania, 3400 Civic Center Boulevard Atrium, Philadelphia, Pennsylvania 19104, United States
| | - Kimberly Taing
- Department of Radiology, University of Pennsylvania, 3400 Spruce Street, 1 Silverstein Philadelphia, Pennsylvania 19104, United States
| | - Deeksha Saxena
- Department of Radiation Oncology, University of Pennsylvania, 3400 Civic Center Boulevard Atrium, Philadelphia, Pennsylvania 19104, United States
| | - Jessica C Hsu
- Department of Radiology, University of Pennsylvania, 3400 Spruce Street, 1 Silverstein Philadelphia, Pennsylvania 19104, United States
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Minna H Chen
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Ryan D Salinas
- Department of Neurosurgery, University of Pennsylvania, 3400 Spruce Street, Philadelphia, Pennsylvania 19104, United States
| | - Hongjun Song
- Department of Neuroscience, University of Pennsylvania, 3400 Spruce Street, Philadelphia, Pennsylvania 19104, United States
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Jay Dorsey
- Department of Radiation Oncology, University of Pennsylvania, 3400 Civic Center Boulevard Atrium, Philadelphia, Pennsylvania 19104, United States
| | - David P Cormode
- Department of Radiology, University of Pennsylvania, 3400 Spruce Street, 1 Silverstein Philadelphia, Pennsylvania 19104, United States
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
21
|
Bhattacharya S. Glioblastoma Multiforme and its Cell Interruption. CURRENT CANCER THERAPY REVIEWS 2021. [DOI: 10.2174/1573394716999201007125709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glioblastoma (GBM) is a fatal type of brain cancer or primary glial neoplasm, mostly
targeting aged populations. The average survival is only 15 months from the date of occurrence. It
is often observed that, the invasive nature of the tumour is the main reason for poor prognosis and
strong recurrence of GBM in patients even after prescribed treatments. Despite all types of therapies,
it is necessary to understand the various molecular mechanisms and signalling pathways to
identify targets for GBM treatment. This compilation is specifically designed to discuss Wnt signalling
and Hedgehog-GLI1 pathways, which have positive and negative regulation against GBM.
The recent research finding associated with different signalling pathways for GBM has also been
discussed within this article.
Collapse
Affiliation(s)
- Sankha Bhattacharya
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road (NH-95), Ghal Kalan, Moga, Punjab 142001, India
| |
Collapse
|
22
|
Reimunde P, Pensado-López A, Carreira Crende M, Lombao Iglesias V, Sánchez L, Torrecilla-Parra M, Ramírez CM, Anfray C, Torres Andón F. Cellular and Molecular Mechanisms Underlying Glioblastoma and Zebrafish Models for the Discovery of New Treatments. Cancers (Basel) 2021; 13:1087. [PMID: 33802571 PMCID: PMC7961726 DOI: 10.3390/cancers13051087] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) is the most common of all brain malignant tumors; it displays a median survival of 14.6 months with current complete standard treatment. High heterogeneity, aggressive and invasive behavior, the impossibility of completing tumor resection, limitations for drug administration and therapeutic resistance to current treatments are the main problems presented by this pathology. In recent years, our knowledge of GBM physiopathology has advanced significantly, generating relevant information on the cellular heterogeneity of GBM tumors, including cancer and immune cells such as macrophages/microglia, genetic, epigenetic and metabolic alterations, comprising changes in miRNA expression. In this scenario, the zebrafish has arisen as a promising animal model to progress further due to its unique characteristics, such as transparency, ease of genetic manipulation, ethical and economic advantages and also conservation of the major brain regions and blood-brain-barrier (BBB) which are similar to a human structure. A few papers described in this review, using genetic and xenotransplantation zebrafish models have been used to study GBM as well as to test the anti-tumoral efficacy of new drugs, their ability to interact with target cells, modulate the tumor microenvironment, cross the BBB and/or their toxicity. Prospective studies following these lines of research may lead to a better diagnosis, prognosis and treatment of patients with GBM.
Collapse
Affiliation(s)
- Pedro Reimunde
- Department of Medicine, Campus de Oza, Universidade da Coruña, 15006 A Coruña, Spain
- Department of Neurosurgery, Hospital Universitario Lucus Augusti, 27003 Lugo, Spain
| | - Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Martín Carreira Crende
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
| | - Vanesa Lombao Iglesias
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
| | - Marta Torrecilla-Parra
- IMDEA Research Institute of Food and Health Sciences, 28049 Madrid, Spain; (M.T.-P.); (C.M.R.)
| | - Cristina M. Ramírez
- IMDEA Research Institute of Food and Health Sciences, 28049 Madrid, Spain; (M.T.-P.); (C.M.R.)
| | - Clément Anfray
- IRCCS Istituto Clinico Humanitas, Via A. Manzoni 56, 20089 Rozzano, Milan, Italy;
| | - Fernando Torres Andón
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
- IRCCS Istituto Clinico Humanitas, Via A. Manzoni 56, 20089 Rozzano, Milan, Italy;
| |
Collapse
|
23
|
Zhou M, Li H, Chen K, Ding W, Yang C, Wang X. CircSKA3 Downregulates miR-1 Through Methylation in Glioblastoma to Promote Cancer Cell Proliferation. Cancer Manag Res 2021; 13:509-514. [PMID: 33500664 PMCID: PMC7826074 DOI: 10.2147/cmar.s279097] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/07/2020] [Indexed: 11/29/2022] Open
Abstract
Background Circular RNA circSKA3 plays an oncogenic role in breast cancer, while its role in glioblastoma (GBM) is unknown. This study aimed to explore the role of circSKA3 in GBM. Methods Differential expression of circSKA3 and miR-1 in GBM and adjacent non-cancer tissue samples were analyzed by RT-qPCR. GBM cells were transfected with circSKA3 expression vector or miR-1 mimic, followed by RT-qPCR to explore the potential crosstalk between them. Methylation-specific PCR (MSP) was carried out to assess the role of circSKA3 in regulating the methylation of miR-1 gene. The role of circSKA3 and miR-1 in regulating GBM cell proliferation was analyzed by CCK-8 assay. Results We found that circSKA3 was upregulated in GBM and inversely correlated with miR-1 across GBM tissues. High expression levels of circSKA3 and low expression levels of miR-1 were significantly correlated with the poor survival of GBM patients. In GBM cells, overexpression of circSKA3 increased the methylation of miR-1 gene and decreased the expression of miR-1. CCK-8 assay showed that overexpression of circSKA3 reduced the inhibitory effects of miR-1 on cell proliferation. Conclusion Therefore, circSKA3 may downregulate miR-1 through methylation in GBM to promote cancer cell proliferation.
Collapse
Affiliation(s)
- Meng Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou City, Guangdong Province 510630, People's Republic of China
| | - Huan Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, People's Republic of China
| | - Ke'en Chen
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou City, Guangdong Province 510630, People's Republic of China
| | - Weilong Ding
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou City, Guangdong Province 510630, People's Republic of China
| | - Chengyou Yang
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou City, Guangdong Province 510630, People's Republic of China
| | - Xiangyu Wang
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou City, Guangdong Province 510630, People's Republic of China
| |
Collapse
|
24
|
Bastiancich C, Da Silva A, Estève MA. Photothermal Therapy for the Treatment of Glioblastoma: Potential and Preclinical Challenges. Front Oncol 2021; 10:610356. [PMID: 33520720 PMCID: PMC7845694 DOI: 10.3389/fonc.2020.610356] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/01/2020] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma (GBM) is a very aggressive primary malignant brain tumor and finding effective therapies is a pharmaceutical challenge and an unmet medical need. Photothermal therapy may be a promising strategy for the treatment of GBM, as it allows the destruction of the tumor using heat as a non-chemical treatment for disease bypassing the GBM heterogeneity limitations, conventional drug resistance mechanisms and side effects on peripheral healthy tissues. However, its development is hampered by the distinctive features of this tumor. Photoabsorbing agents such as nanoparticles need to reach the tumor site at therapeutic concentrations, crossing the blood-brain barrier upon systemic administration. Subsequently, a near infrared light irradiating the head must cross multiple barriers to reach the tumor site without causing any local damage. Its power intensity needs to be within the safety limit and its penetration depth should be sufficient to induce deep and localized hyperthermia and achieve tumor destruction. To properly monitor the therapy, imaging techniques that can accurately measure the increase in temperature within the brain must be used. In this review, we report and discuss recent advances in nanoparticle-mediated plasmonic photothermal therapy for GBM treatment and discuss the preclinical challenges commonly faced by researchers to develop and test such systems.
Collapse
Affiliation(s)
- Chiara Bastiancich
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Anabela Da Silva
- Aix Marseille Univ, CNRS, Centrale Marseille, Institut Fresnel, Marseille, France
| | - Marie-Anne Estève
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France.,APHM, Hôpital de la Timone, Service Pharmacie, Marseille, France
| |
Collapse
|
25
|
Wouters R, Bevers S, Riva M, De Smet F, Coosemans A. Immunocompetent Mouse Models in the Search for Effective Immunotherapy in Glioblastoma. Cancers (Basel) 2020; 13:E19. [PMID: 33374542 PMCID: PMC7793150 DOI: 10.3390/cancers13010019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/19/2020] [Accepted: 12/20/2020] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive intrinsic brain tumor in adults. Despite maximal therapy consisting of surgery and radio/chemotherapy, GBM remains largely incurable with a median survival of less than 15 months. GBM has a strong immunosuppressive nature with a multitude of tumor and microenvironment (TME) derived factors that prohibit an effective immune response. To date, all clinical trials failed to provide lasting clinical efficacy, despite the relatively high success rates of preclinical studies to show effectivity of immunotherapy. Various factors may explain this discrepancy, including the inability of a single mouse model to fully recapitulate the complexity and heterogeneity of GBM. It is therefore critical to understand the features and limitations of each model, which should probably be combined to grab the full spectrum of the disease. In this review, we summarize the available knowledge concerning immune composition, stem cell characteristics and response to standard-of-care and immunotherapeutics for the most commonly available immunocompetent mouse models of GBM.
Collapse
Affiliation(s)
- Roxanne Wouters
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000 Leuven, Belgium; (R.W.); (S.B.); (M.R.)
- Oncoinvent, A.S., 0484 Oslo, Norway
| | - Sien Bevers
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000 Leuven, Belgium; (R.W.); (S.B.); (M.R.)
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium;
| | - Matteo Riva
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000 Leuven, Belgium; (R.W.); (S.B.); (M.R.)
- Department of Neurosurgery, Mont-Godinne Hospital, UCL Namur, 5530 Yvoir, Belgium
| | - Frederik De Smet
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium;
| | - An Coosemans
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000 Leuven, Belgium; (R.W.); (S.B.); (M.R.)
| |
Collapse
|
26
|
Bozzato E, Bastiancich C, Préat V. Nanomedicine: A Useful Tool against Glioma Stem Cells. Cancers (Basel) 2020; 13:cancers13010009. [PMID: 33375034 PMCID: PMC7792799 DOI: 10.3390/cancers13010009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 02/06/2023] Open
Abstract
The standard of care therapy of glioblastoma (GBM) includes invasive surgical resection, followed by radiotherapy and concomitant chemotherapy. However, this therapy has limited success, and the prognosis for GBM patients is very poor. Although many factors may contribute to the failure of current treatments, one of the main causes of GBM recurrences are glioma stem cells (GSCs). This review focuses on nanomedicine strategies that have been developed to eliminate GSCs and the benefits that they have brought to the fight against cancer. The first section describes the characteristics of GSCs and the chemotherapeutic strategies that have been used to selectively kill them. The second section outlines the nano-based delivery systems that have been developed to act against GSCs by dividing them into nontargeted and targeted nanocarriers. We also highlight the advantages of nanomedicine compared to conventional chemotherapy and examine the different targeting strategies that have been employed. The results achieved thus far are encouraging for the pursuit of effective strategies for the eradication of GSCs.
Collapse
Affiliation(s)
- Elia Bozzato
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Chiara Bastiancich
- Institute Neurophysiopathol, INP, CNRS, Aix-Marseille University, 13005 Marseille, France;
| | - Véronique Préat
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université Catholique de Louvain, 1200 Brussels, Belgium;
- Correspondence:
| |
Collapse
|
27
|
Alghamdi M, Chierchini F, Eigel D, Taplan C, Miles T, Pette D, Welzel PB, Werner C, Wang W, Neto C, Gumbleton M, Newland B. Poly(ethylene glycol) based nanotubes for tuneable drug delivery to glioblastoma multiforme. NANOSCALE ADVANCES 2020; 2:4498-4509. [PMID: 36132909 PMCID: PMC9418774 DOI: 10.1039/d0na00471e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/20/2020] [Indexed: 06/16/2023]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of malignant brain tumour, which is associated with a poor two-year survival rate and a high rate of fatal recurrence near the original tumour. Focal/local drug delivery devices hold promise for improving therapeutic outcomes for GBM by increasing drug concentrations locally at the tumour site, or by facilitating the use of potent anti-cancer drugs that are poorly permeable across the blood brain barrier (BBB). For inoperable tumours, stereotactic delivery to the tumour necessitates the development of nanoscale/microscale injectable drug delivery devices. Herein we assess the ability of a novel class of polymer nanotube (based on poly(ethylene glycol) (PEG)) to load doxorubicin (a mainstay breast cancer therapeutic with poor BBB permeability) and release it slowly. The drug loading properties of the PEG nanotubes could be tuned by varying the degree of carboxylic acid functionalisation and hence the capacity of the nanotubes to electrostatically bind and load doxorubicin. 70% of the drug was released over the first seven days followed by sustained drug release for the remaining two weeks tested. Unloaded PEG nanotubes showed no toxicity to any of the cell types analysed, whereas doxorubicin loaded nanotubes decreased GBM cell viability (C6, U-87 and U-251) in a dose dependent manner in 2D in vitro culture. Finally, doxorubicin loaded PEG nanotubes significantly reduced the viability of in vitro 3D GBM models whilst unloaded nanotubes showed no cytotoxicity. Taken together, these findings show that polymer nanotubes could be used to deliver alternative anti-cancer drugs for local therapeutic strategies against brain cancers.
Collapse
Affiliation(s)
- Majed Alghamdi
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University King Edward VII Avenue Cardiff CF10 3NB UK
- School of Pharmacy, King Abdulaziz University Jeddah 21589 Saudi Arabia
| | - Filippo Chierchini
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University King Edward VII Avenue Cardiff CF10 3NB UK
| | - Dimitri Eigel
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden Hohe Straße 6 D-01069 Dresden Germany
| | - Christian Taplan
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden Hohe Straße 6 D-01069 Dresden Germany
| | - Thomas Miles
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University King Edward VII Avenue Cardiff CF10 3NB UK
| | - Dagmar Pette
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden Hohe Straße 6 D-01069 Dresden Germany
| | - Petra B Welzel
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden Hohe Straße 6 D-01069 Dresden Germany
| | - Carsten Werner
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden Hohe Straße 6 D-01069 Dresden Germany
| | - Wenxin Wang
- Charles Institute of Dermatology, School of Medicine, University College Dublin Ireland
| | - Catia Neto
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University King Edward VII Avenue Cardiff CF10 3NB UK
| | - Mark Gumbleton
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University King Edward VII Avenue Cardiff CF10 3NB UK
| | - Ben Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University King Edward VII Avenue Cardiff CF10 3NB UK
- Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden Hohe Straße 6 D-01069 Dresden Germany
| |
Collapse
|
28
|
Mena-Hernández J, Jung-Cook H, Llaguno-Munive M, García-López P, Ganem-Rondero A, López-Ramírez S, Barragán-Aroche F, Rivera-Huerta M, Mayet-Cruz L. Preparation and Evaluation of Mebendazole Microemulsion for Intranasal Delivery: an Alternative Approach for Glioblastoma Treatment. AAPS PharmSciTech 2020; 21:264. [PMID: 32980937 DOI: 10.1208/s12249-020-01805-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
Although mebendazole (MBZ) has demonstrated antitumor activity in glioblastoma models, the drug has low aqueous solubility and therefore is poorly absorbed. Considering that other strategies are needed to improve its bioavailability, the current study was aimed to develop and evaluate novel microemulsions of MBZ (MBZ-NaH ME) for intranasal administration. MBZ raw materials were characterized by FTIR, DSC, and XDP. Subsequently, the raw material that contained mainly polymorph C was selected to prepare microemulsions. Two different oleic acid (OA) systems were selected. Formulation A was composed of OA and docosahexaenoic acid (3:1% w/w), while formulation B was composed of OA and Labrafil M2125 (1:1% w/w). Sodium hyaluronate (NaH) at 0.1% was selected as a mucoadhesive agent. MBZ MEs showed a particle size of 209 nm and 145 nm, respectively, and the pH was suitable for nasal formulations (4.5-6.5). Formulation B, which showed the best solubility and rheological behavior, was selected for intranasal evaluation. The nasal toxicity study revealed no damage in the epithelium. Furthermore, formulation B improved significantly the median survival time in the orthotopic C6 rat model compared to the control group. Moreover, NIRF signal intensity revealed a decrease in tumor growth in the treated group with MBZ-MaH ME, which was confirmed by histologic examinations. Results suggest that the intranasal administration of mebendazole-loaded microemulsion might be appropriated for glioblastoma treatment. Graphical abstract.
Collapse
|
29
|
Wan Y, Liang F, Wei M, Liu Y. Long non-coding RNA LINC00525 regulates the proliferation and epithelial to mesenchymal transition of human glioma cells by sponging miR-338-3p. AMB Express 2020; 10:156. [PMID: 32857271 PMCID: PMC7455684 DOI: 10.1186/s13568-020-01094-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 08/19/2020] [Indexed: 12/19/2022] Open
Abstract
Long non-coding RNA (LncRNA) LINC00525 has been shown to be upregulated in several human cancers and deduced to possess caner regulatory role. The regulation of molecular mechanics of human glioma by lncRNA-LINC00525 through microRNA sponging in glioma is elusive. The lncRNA-LINC00525 showed significant (P < 0.05) upregulation in glioma cancer cells. The upregulation of lncRNA-LINC00525 was upto 6.6-fold in glioma cells relative to the normal cells. Knockdown of lncRNA-LINC00525 significantly declined the proliferation of the glioma cancer cells. Additionally, the colony formation was inhibited by around 60% in glioma cells. The wound healing and transwell assays revealed significant (P < 0.05) inhibition of migration and invasion potential under lncRNA-LINC00525 knockdown. The western blotting study of biomarkers of epithelial to mesenchymal transition (EMT) revealed that lncRNA-LINC00525 gene silencing reduced the expression of mesenchymal molecular markers but increased the protein levels of epithelial markers. miR-338-3p was predicted to be interacting with lncRNA-LINC00525 in glioma and was shown to mediated the regulatory role of lncRNA-LINC00525. Taken together, the results of present study are supportive of the prognostic applicability of lncRNA-LINC00525 against human glioma together with its therapeutic potential against the said malignancy.
Collapse
|
30
|
Garanti T, Alhnan MA, Wan KW. RGD-decorated solid lipid nanoparticles enhance tumor targeting, penetration and anticancer effect of asiatic acid. Nanomedicine (Lond) 2020; 15:1567-1583. [DOI: 10.2217/nnm-2020-0035] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: Asiatic acid (AA) is a promising anticancer agent, however, its delivery to glioblastoma is a major challenge. This work investigates the beneficial therapeutic efficacy of RGD-conjugated solid lipid nanoparticles (RGD-SLNs) for the selective targeting of AA to gliblastoma. Materials & methods: AA-containing RGD-SLNs were prepared using two different PEG-linker size. Targetability and efficacy were tested using monolayer cells and spheroid tumor models. Results: RGD-SLNs significantly improved cytotoxicity of AA against U87-MG monolayer cells and enhanced cellular uptake compared with non-RGD-containing SLNs. In spheroid models, AA-containing RGD-SLNs showed superior control in tumor growth, improved cytotoxicity and enhanced spheroid penetration when compared with AA alone or non-RGD-containing SLNs. Conclusion: This study illustrates the potential of AA-loaded RGD-SLNs as efficacious target-specific treatment for glioblastoma.
Collapse
Affiliation(s)
- Tanem Garanti
- Faculty of Pharmacy, Cyprus International University, Haspolat, Nicosia, 99258, Cyprus via Mersin 10, Turkey
| | - Mohamed A Alhnan
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King’s College London, London, UK
| | - Ka-Wai Wan
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston, Lancashire, PR1 2HE, UK
| |
Collapse
|
31
|
Le Joncour V, Filppu P, Hyvönen M, Holopainen M, Turunen SP, Sihto H, Burghardt I, Joensuu H, Tynninen O, Jääskeläinen J, Weller M, Lehti K, Käkelä R, Laakkonen P. Vulnerability of invasive glioblastoma cells to lysosomal membrane destabilization. EMBO Mol Med 2020; 11:emmm.201809034. [PMID: 31068339 PMCID: PMC6554674 DOI: 10.15252/emmm.201809034] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The current clinical care of glioblastomas leaves behind invasive, radio‐ and chemo‐resistant cells. We recently identified mammary‐derived growth inhibitor (MDGI/FABP3) as a biomarker for invasive gliomas. Here, we demonstrate a novel function for MDGI in the maintenance of lysosomal membrane integrity, thus rendering invasive glioma cells unexpectedly vulnerable to lysosomal membrane destabilization. MDGI silencing impaired trafficking of polyunsaturated fatty acids into cells resulting in significant alterations in the lipid composition of lysosomal membranes, and subsequent death of the patient‐derived glioma cells via lysosomal membrane permeabilization (LMP). In a preclinical model, treatment of glioma‐bearing mice with an antihistaminergic LMP‐inducing drug efficiently eradicated invasive glioma cells and secondary tumours within the brain. This unexpected fragility of the aggressive infiltrating cells to LMP provides new opportunities for clinical interventions, such as re‐positioning of an established antihistamine drug, to eradicate the inoperable, invasive, and chemo‐resistant glioma cells from sustaining disease progression and recurrence.
Collapse
Affiliation(s)
- Vadim Le Joncour
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Pauliina Filppu
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Maija Hyvönen
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Minna Holopainen
- Helsinki University Lipidomics Unit, Helsinki Institute of Life Science (HiLIFE) and Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
| | - S Pauliina Turunen
- Research Programs Unit, Genome-Scale Biology, University of Helsinki, Helsinki, Finland.,Department of Microbiology, Tumour and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Harri Sihto
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Isabel Burghardt
- Department of Neurology and Brain Tumour Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Heikki Joensuu
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Oncology, Helsinki University Hospital, Helsinki, Finland
| | - Olli Tynninen
- Department of Pathology, Haartman Institute, University of Helsinki and HUSLAB, Helsinki, Finland
| | | | - Michael Weller
- Department of Neurology and Brain Tumour Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Kaisa Lehti
- Research Programs Unit, Genome-Scale Biology, University of Helsinki, Helsinki, Finland.,Department of Microbiology, Tumour and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Reijo Käkelä
- Helsinki University Lipidomics Unit, Helsinki Institute of Life Science (HiLIFE) and Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
| | - Pirjo Laakkonen
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland .,Laboratory Animal Centre, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| |
Collapse
|
32
|
Whitehead CA, Kaye AH, Drummond KJ, Widodo SS, Mantamadiotis T, Vella LJ, Stylli SS. Extracellular vesicles and their role in glioblastoma. Crit Rev Clin Lab Sci 2019:1-26. [PMID: 31865806 DOI: 10.1080/10408363.2019.1700208] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Research on the role of extracellular vesicles (EVs) in disease pathogenesis has been rapidly growing over the last two decades. As EVs can mediate intercellular communication, they can ultimately facilitate both normal and pathological processes through the delivery of their bioactive cargo, which may include nucleic acids, proteins and lipids. EVs have emerged as important regulators of brain tumors, capable of transferring oncogenic proteins, receptors, and small RNAs that may support brain tumor progression, including in the most common type of brain cancer, glioma. Investigating the role of EVs in glioma is crucial, as the most malignant glioma, glioblastoma (GBM), is incurable with a dismal median survival of 12-15 months. EV research in GBM has primarily focused on circulating brain tumor-derived vesicles in biofluids, such as blood and cerebrospinal fluid (CSF), investigating their potential as diagnostic and prognostic biomarkers. Gaining a greater understanding of the role of EVs and their cargo in brain tumor progression may contribute to the discovery of novel diagnostics and therapeutics. In this review, we summarize the known and emerging functions of EVs in glioma biology and pathogenesis, as well as their emerging biomarker potential.
Collapse
Affiliation(s)
- Clarissa A Whitehead
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia
| | - Andrew H Kaye
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,Department of Neurosurgery, Hadassah Hebrew University Medical Centre, Jerusalem, Israel
| | - Katharine J Drummond
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, Australia
| | - Samuel S Widodo
- Department of Microbiology & Immunology, School of Biomedical Sciences, The University of Melbourne, Parkville, Australia
| | - Theo Mantamadiotis
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,Department of Microbiology & Immunology, School of Biomedical Sciences, The University of Melbourne, Parkville, Australia
| | - Laura J Vella
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Stanley S Stylli
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,Department of Neurosurgery, Hadassah Hebrew University Medical Centre, Jerusalem, Israel
| |
Collapse
|
33
|
Venkatesh VS, Lou E. Tunneling nanotubes: A bridge for heterogeneity in glioblastoma and a new therapeutic target? Cancer Rep (Hoboken) 2019; 2:e1185. [PMID: 32729189 PMCID: PMC7941610 DOI: 10.1002/cnr2.1185] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/10/2019] [Accepted: 04/10/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The concept of tumour heterogeneity is not novel but is fast becoming a paradigm by which to explain part of the highly recalcitrant nature of aggressive malignant tumours. Glioblastoma is a prime example of such difficult-to-treat, invasive, and incurable malignancies. With the advent of the post-genomic age and increased access to next-generation sequencing technologies, numerous publications have described the presence and extent of intratumoural and intertumoural heterogeneity present in glioblastoma. Moreover, there have been numerous reports more directly correlating the heterogeneity of glioblastoma to its refractory, reoccurring, and inevitably terminal nature. It is therefore prudent to consider the different forms of heterogeneity seen in glioblastoma and how to harness this understanding to better strategize novel therapeutic approaches. One of the most central questions of tumour heterogeneity is how these numerous different cell types (both tumour and non-tumour) in the tumour mass communicate. RECENT FINDINGS This chapter provides a brief review on the variable heterogeneity of glioblastoma, with a focus on cellular heterogeneity and on modalities of communication that can induce further molecular diversity within the complex and ever-evolving tumour microenvironment. We provide particular emphasis on the emerging role of actin-based cellular conduits called tunnelling nanotubes (TNTs) and tumour microtubes (TMs) and outline the perceived current problems in the field that need to be resolved before pharmacological targeting of TNTs can become a reality. CONCLUSIONS We conclude that TNTs and TMs provide a new and exciting avenue for the therapeutic targeting of glioblastoma and that numerous inroads have already made into TNT and TM biology. However, to target TMs and TNTs, several advances must be made before this aim can become a reality.
Collapse
Affiliation(s)
| | - Emil Lou
- Division of Hematology, Oncology and TransplantationUniversity of MinnesotaMinneapolisMinnesota
| |
Collapse
|
34
|
Duwa R, Emami F, Lee S, Jeong JH, Yook S. Polymeric and lipid-based drug delivery systems for treatment of glioblastoma multiforme. J IND ENG CHEM 2019. [DOI: 10.1016/j.jiec.2019.06.050] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
35
|
Ramírez-Expósito MJ, Martínez-Martos JM. Differential Effects of Doxazosin on Renin-Angiotensin-System- Regulating Aminopeptidase Activities in Neuroblastoma and Glioma Tumoral Cells. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2019; 18:29-36. [DOI: 10.2174/1871527317666181029111739] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 07/13/2018] [Accepted: 10/10/2018] [Indexed: 11/22/2022]
Abstract
Background:
It has been described that doxazosin, an antihypertensive drug, also promotes
glioblastoma cells death by inhibiting cell proliferation, arresting cell cycle and inducing apoptosis.
Doxazosin has also demonstrated several modulator effects on renin-angiotensin system (RAS)-
regulating aminopeptidase activities, which are highly involved in tumor growth in experimental
glioma. Therefore, it remains to elucidate if the anti-tumoral effects of doxazosin could also be mediated
by the proteolytic regulatory components of the RAS.
Objective:
To analyze the effects of doxazosin on cell growth and on RAS-regulating proteolytic regulatory
aspartyl aminopeptidase (ASAP), aminopeptidase A (APA), aminopeptidase N (APN), aminopeptidase
B (APB) and insulin-regulated aminopeptidase (IRAP) specific activities in the human neuroblastoma
NB69 and astroglioma U373-MG tumoral cell lines.
Methods:
Human neuroblastoma NB69 and astroglioma U373-MG cell lines were treated with doxazosin
50-500 μM for 24h or 48h. The effects on cell growth and on RAS-regulating aminopeptidase
specific activities were analyzed.
Results:
Doxazosin treatments promote a concentration-dependent inhibition on cell growth in both
NB69 and U373-MG cells, being NB69 cells more sensitive to the drug than U373-MG cells. However,
its effects on RAS-regulating aminopeptidase specific activities depend on the concentration
used, the duration of the treatment and the cell type. These data confirm the existence of a different
dynamic progression of RAS cascade in each tumoral cell line as a consequence of the treatment with
doxazosin and time of action, which also implies a very dynamic metabolism of the peptides which
participate in each step of RAS cascade.
Conclusion:
Our results indicate that doxazosin modifies the proteolytic regulatory enzymes of RAS
cascade, modulating the bioactive efficacy of the different angiotensin peptides, and therefore, of their
functional roles as initiators/promoters of cell proliferation as autocrine/paracrine mediators.
Collapse
Affiliation(s)
- María Jesús Ramírez-Expósito
- Experimental and Clinical Physiopathology Research Group CTS1039, Department of Health Sciences, Faculty of Health Sciences, University of Jaen, Jaen, Spain
| | - José Manuel Martínez-Martos
- Experimental and Clinical Physiopathology Research Group CTS1039, Department of Health Sciences, Faculty of Health Sciences, University of Jaen, Jaen, Spain
| |
Collapse
|
36
|
Bastiancich C, Bozzato E, Luyten U, Danhier F, Bastiat G, Préat V. Drug combination using an injectable nanomedicine hydrogel for glioblastoma treatment. Int J Pharm 2019; 559:220-227. [PMID: 30703501 DOI: 10.1016/j.ijpharm.2019.01.042] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/14/2019] [Accepted: 01/17/2019] [Indexed: 01/25/2023]
Abstract
Lauroyl-gemcitabine lipid nanocapsules (GemC12-LNC) hydrogel, administered intratumorally or perisurgically in the tumor resection cavity, increases animal survival in several orthotopic GBM models. We hypothesized that GemC12-LNC can be used as nanodelivery platform for other drugs, to obtain a combined local therapeutic approach for GBM. Paclitaxel (PTX) was selected as a model molecule and PTX-GemC12-LNC formulation was evaluated in terms of physicochemical and mechanical properties. The PTX-GemC12-LNC hydrogel stability and drug release were evaluated over time showing no significant differences compared to GemC12-LNC. The drug combination was evaluated on several GBM cell lines showing increased cytotoxic activity compared to the original formulation and synergy between PTX and GemC12. Our results suggest that GemC12-LNC hydrogel can be used as nanodelivery platform for dual drug delivery to encapsulate active agents with different mechanisms of action to achieve a better antitumor efficacy against GBM or other solid tumors.
Collapse
Affiliation(s)
- Chiara Bastiancich
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Elia Bozzato
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Urszula Luyten
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Fabienne Danhier
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Guillaume Bastiat
- Micro & Nanomedecines Translationnelles - MINT, UNIV Angers, INSERM U1066, CNRS UMR 6021, UBL Université Bretagne Loire, Angers, France
| | - Véronique Préat
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium.
| |
Collapse
|
37
|
Repurposing of idebenone as a potential anti-cancer agent. Biochem J 2019; 476:245-259. [DOI: 10.1042/bcj20180384] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 01/01/2019] [Accepted: 01/01/2019] [Indexed: 12/22/2022]
Abstract
AbstractGlioblastoma (GB) represents the most common and aggressive form of malignant primary brain tumour associated with high rates of morbidity and mortality. In the present study, we considered the potential use of idebenone (IDE), a Coenzyme Q10 analogue, as a novel chemotherapeutic agent for GB. On two GB cell lines, U373MG and U87MG, IDE decreased the viable cell number and enhanced the cytotoxic effects of two known anti-proliferative agents: temozolomide and oxaliplatin. IDE also affected the clonogenic and migratory capacity of both GB cell lines, at 25 and 50 µM, a concentration equivalent to that transiently reached in plasma after oral intake that is deemed safe for humans. p21 protein expression was decreased in both cell lines, indicating that IDE likely exerts its effects through cell cycle dysregulation, and this was confirmed in U373MG cells only by flow cytometric cell cycle analysis which showed S-phase arrest. Caspase-3 protein expression was also significantly decreased in U373MG cells indicating IDE-induced apoptosis that was confirmed by flow cytometric Annexin V/propidium iodide staining. No major decrease in caspase-3 expression was observed in U87MG cells nor apoptosis as observed by flow cytometry analysis. Overall, the present study demonstrates that IDE has potential as an anti-proliferative agent for GB by interfering with several features of glioma pathogenesis such as proliferation and migration, and hence might be a drug that could be repurposed for aiding cancer treatments. Furthermore, the synergistic combinations of IDE with other agents aimed at different pathways involved in this type of cancer are promising.
Collapse
|
38
|
Was H, Krol SK, Rotili D, Mai A, Wojtas B, Kaminska B, Maleszewska M. Histone deacetylase inhibitors exert anti-tumor effects on human adherent and stem-like glioma cells. Clin Epigenetics 2019; 11:11. [PMID: 30654849 PMCID: PMC6337817 DOI: 10.1186/s13148-018-0598-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 12/17/2018] [Indexed: 12/29/2022] Open
Abstract
Background The diagnosis of glioblastoma (GBM), a most aggressive primary brain tumor with a median survival of 14.6 months, carries a dismal prognosis. GBMs are characterized by numerous genetic and epigenetic alterations, affecting patient survival and treatment response. Epigenetic mechanisms are deregulated in GBM as a result of aberrant expression/activity of epigenetic enzymes, including histone deacetylases (HDAC) which remove acetyl groups from histones regulating chromatin accessibility. Nevertheless, the impact of class/isoform-selective HDAC inhibitors (HDACi) on glioma cells, including glioma stem cells, had not been systematically determined. Results Comprehensive analysis of the public TCGA dataset revealed the increased expression of HDAC 1, 2, 3, and 7 in malignant gliomas. Knockdown of HDAC 1 and 2 in human GBM cells significantly decreased cell proliferation. We tested the activity of 2 new and 3 previously described HDACi with different class/isoform selectivity on human GBM cells. All tested compounds exerted antiproliferative properties on glioma cells. However, the HDACi 1 and 4 blocked proliferation of glioblastoma cells leading to G2/M growth arrest without affecting astrocyte survival. Moreover, 1 and 4 at low micromolar concentrations displayed cytotoxic and antiproliferative effects on sphere cultures enriched in glioma stem cells. Conclusions We identified two selective HDAC inhibitors that blocked proliferation of glioblastoma cells, but did not affect astrocyte survival. These new and highly effective inhibitors should be considered as promising candidates for further investigation in preclinical GBM models. Electronic supplementary material The online version of this article (10.1186/s13148-018-0598-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Halina Was
- Laboratory of Molecular Neurobiology, Neurobiology Center, The Nencki Institute of Experimental Biology, 3 Pasteur Str, 02-093, Warsaw, Poland.,Laboratory of Molecular Oncology, Military Institute of Medicine, 128 Szaserow Str, 04-141, Warsaw, Poland
| | - Sylwia K Krol
- Laboratory of Molecular Neurobiology, Neurobiology Center, The Nencki Institute of Experimental Biology, 3 Pasteur Str, 02-093, Warsaw, Poland
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Roma, P.le A. Moro 5, 00185, Rome, Italy
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Roma, P.le A. Moro 5, 00185, Rome, Italy.,Pasteur Institute, Cenci-Bolognetti Foundation, Sapienza University of Rome, 00185, Rome, Italy
| | - Bartosz Wojtas
- Laboratory of Molecular Neurobiology, Neurobiology Center, The Nencki Institute of Experimental Biology, 3 Pasteur Str, 02-093, Warsaw, Poland
| | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Neurobiology Center, The Nencki Institute of Experimental Biology, 3 Pasteur Str, 02-093, Warsaw, Poland
| | - Marta Maleszewska
- Laboratory of Molecular Neurobiology, Neurobiology Center, The Nencki Institute of Experimental Biology, 3 Pasteur Str, 02-093, Warsaw, Poland.
| |
Collapse
|
39
|
González-Morales A, Zabaleta A, García-Moure M, Alonso MM, Fernández-Irigoyen J, Santamaría E. Oncolytic adenovirus Delta-24-RGD induces a widespread glioma proteotype remodeling during autophagy. J Proteomics 2018; 194:168-178. [PMID: 30503830 DOI: 10.1016/j.jprot.2018.11.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 11/16/2018] [Accepted: 11/28/2018] [Indexed: 02/06/2023]
Abstract
Adenovirus Delta-24-RGD has shown a remarkable efficacy in a phase I clinical trial for glioblastoma. Delta-24-RGD induces autophagy in glioma cells, however, the molecular derangements associated with Delta-24-RGD infection remains poorly understood. Here, proteomics was applied to characterize the glioma metabolic disturbances soon after Delta-24-RGD internalization and late in infection. Minutes post-infection, a rapid survival reprogramming of glioma cells was evidenced by an early c-Jun activation and a time-dependent dephosphorylation of multiple survival kinases. At 48 h post-infection (hpi), a severe intracellular proteostasis impairment was characterized, detecting differentially expressed proteins related to mRNA splicing, cytoskeletal organization, oxidative response, and inflammation. Specific kinase-regulated protein interactomes for Delta-24-RGD-modulated proteome revealed interferences with the activation dynamics of protein kinases C and A (PKC, PKA), tyrosine-protein kinase Src (c-Src), glycogen synthase kinase-3 (GSK-3) as well as serine/threonine-protein phosphatases 1 and 2A (PP1, PP2A) at 48hpi, in parallel with adenoviral protein overproduction. Moreover, the late activation of the nuclear factor kappa B (NF-κB) correlates with the extracellular increment of specific cytokines involved in migration, and activation of different inflammatory cells. Taken together, our integrative analysis provides further insights into the effects triggered by Delta-24-RGD in the modulation of tumor suppression and immune response against glioma. SIGNIFICANCE: The current study provides new insights regarding the molecular mechanisms governing the glioma metabolism during Delta-24-RGD oncolytic adenoviral therapy. The compilation and analysis of intracellular and extracellular proteomics have led us to characterize: i) the cell survival reprogramming during Delta-24-RGD internalization, ii) the proteostatic disarrangement induced by Delta-24-RGD during the autophagic stage, iii) the protein interactomes for Delta-24-RGD-modulated proteome, iv) the regulatory effects on kinase dynamics induced by Delta-24-RGD late in infection, and v) the overproduction of multitasking cytokines upon Delta-24-RGD treatment. We consider that the quantitative molecular maps generated in this study may establish the foundations for the development of complementary adenoviral based-vectors to increase the potency against glioma.
Collapse
Affiliation(s)
- Andrea González-Morales
- Clinical Neuroproteomics Group, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea 3, 31008 Pamplona, Spain; IDISNA, Navarra Institute for Health Research, Pamplona, Spain; Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea 3, 31008 Pamplona, Spain
| | - Aintzane Zabaleta
- IDISNA, Navarra Institute for Health Research, Pamplona, Spain; Oncohematology Area, University Hospital of Navarra, Center for Applied Medical Research, CIBERONC, Pamplona, Spain
| | - Marc García-Moure
- IDISNA, Navarra Institute for Health Research, Pamplona, Spain; Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain; Department of Pediatrics, University Hospital of Navarra, Pamplona, Spain
| | - Marta M Alonso
- IDISNA, Navarra Institute for Health Research, Pamplona, Spain; Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain; Department of Pediatrics, University Hospital of Navarra, Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Group, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea 3, 31008 Pamplona, Spain; IDISNA, Navarra Institute for Health Research, Pamplona, Spain; Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea 3, 31008 Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Group, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea 3, 31008 Pamplona, Spain; IDISNA, Navarra Institute for Health Research, Pamplona, Spain; Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea 3, 31008 Pamplona, Spain.
| |
Collapse
|
40
|
Bastiancich C, Lemaire L, Bianco J, Franconi F, Danhier F, Préat V, Bastiat G, Lagarce F. Evaluation of lauroyl-gemcitabine-loaded hydrogel efficacy in glioblastoma rat models. Nanomedicine (Lond) 2018; 13:1999-2013. [DOI: 10.2217/nnm-2018-0057] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Chiara Bastiancich
- Louvain Drug Research Institute, Advanced Drug Delivery & Biomaterials, Université Catholique de Louvain, Avenue Mounier, 1200 Brussels, Belgium
- Micro & Nanomédecines Translationnelles-MINT, UNIV Angers, INSERM U1066, CNRS UMR 6021, UBL Université Bretagne Loire, Angers, France
| | - Laurent Lemaire
- Micro & Nanomédecines Translationnelles-MINT, UNIV Angers, INSERM U1066, CNRS UMR 6021, UBL Université Bretagne Loire, Angers, France
- PRISM Plate-forme de recherche en imagerie et spectroscopie multi-modales, PRISM-Icat Angers, UBL, France
| | - John Bianco
- Louvain Drug Research Institute, Advanced Drug Delivery & Biomaterials, Université Catholique de Louvain, Avenue Mounier, 1200 Brussels, Belgium
| | - Florence Franconi
- Micro & Nanomédecines Translationnelles-MINT, UNIV Angers, INSERM U1066, CNRS UMR 6021, UBL Université Bretagne Loire, Angers, France
- PRISM Plate-forme de recherche en imagerie et spectroscopie multi-modales, PRISM-Icat Angers, UBL, France
| | - Fabienne Danhier
- Louvain Drug Research Institute, Advanced Drug Delivery & Biomaterials, Université Catholique de Louvain, Avenue Mounier, 1200 Brussels, Belgium
| | - Veronique Préat
- Louvain Drug Research Institute, Advanced Drug Delivery & Biomaterials, Université Catholique de Louvain, Avenue Mounier, 1200 Brussels, Belgium
| | - Guillaume Bastiat
- Micro & Nanomédecines Translationnelles-MINT, UNIV Angers, INSERM U1066, CNRS UMR 6021, UBL Université Bretagne Loire, Angers, France
| | - Frederic Lagarce
- Micro & Nanomédecines Translationnelles-MINT, UNIV Angers, INSERM U1066, CNRS UMR 6021, UBL Université Bretagne Loire, Angers, France
- Pharmacy Department, CHU Angers, Angers University Hospital, Angers, France
| |
Collapse
|
41
|
González-Morales A, Zabaleta A, Guruceaga E, Alonso MM, García-Moure M, Fernández-Irigoyen J, Santamaría E. Spatial and temporal proteome dynamics of glioma cells during oncolytic adenovirus Delta-24-RGD infection. Oncotarget 2018; 9:31045-31065. [PMID: 30123426 PMCID: PMC6089549 DOI: 10.18632/oncotarget.25774] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/22/2018] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive type of malignant glioma. Oncolytic adenoviruses are being modified to exploit the aberrant expression of proteins in tumor cells to increase the antiglioma efficacy. E1A mutant adenovirus Delta-24-RGD (DNX-2401) has shown a favorable toxicity profile and remarkable efficacy in a first-in-human phase I clinical trial. However, the comprehensive modulation of glioma metabolism in response to Delta-24-RGD infection is poorly understood. Integrating mass spectrometry based-quantitative proteomics, physical and functional interaction data, and biochemical approaches, we conducted a cell-wide study of cytosolic, nuclear, and secreted glioma proteomes throughout the early time course of Delta-24-RGD infection. In addition to the severe proteostasis impairment detected during the first hours post-infection (hpi), Delta-24-RGD induces a transient inhibition of signal transducer and activator of transcription 3 (STAT3), and transcription factor AP-1 (c-JUN) between 3 and 10hpi, increasing the nuclear factor kappa B (NF-κB) activity at 6hpi. Furthermore, Delta-24-RGD specifically modulates the activation dynamics of protein kinase C (PKC), extracellular signal-regulated kinase 1/2 (ERK1/2), and p38 mitogen-activated protein kinase (p38 MAPK) pathways early in infection. At extracellular level, Delta-24-RGD triggers a time -dependent dynamic production of multitasking cytokines, and chemotactic factors, suggesting potential pleiotropic effects on the immune system reactivation. Taken together, these data help us to understand the mechanisms used by Delta-24-RGD to exploit glioma proteome organization. Further mining of this proteomic resource may enable design and engineering complementary adenoviral based-vectors to increase the specificity and potency against glioma.
Collapse
Affiliation(s)
- Andrea González-Morales
- Clinical Neuroproteomics Group, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea, Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea, Pamplona, Spain
| | - Aintzane Zabaleta
- IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Oncohematology Area, University Hospital of Navarra, Center for Applied Medical Research, CIBERONC, Pamplona, Spain
| | - Elizabeth Guruceaga
- IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Bioinformatics Unit, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Marta M Alonso
- IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, University Hospital of Navarra, Pamplona, Spain
| | - Marc García-Moure
- IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, University Hospital of Navarra, Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Group, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea, Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea, Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Group, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea, Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Irunlarrea, Pamplona, Spain
| |
Collapse
|
42
|
Brawanski K, Brockhoff G, Hau P, Vollmann-Zwerenz A, Freyschlag C, Lohmeier A, Riemenschneider MJ, Thomé C, Brawanski A, Proescholdt MA. Efficacy of D,L-methadone in the treatment of glioblastoma in vitro. CNS Oncol 2018; 7:CNS18. [PMID: 29916277 PMCID: PMC6200059 DOI: 10.2217/cns-2018-0006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 05/30/2018] [Indexed: 12/13/2022] Open
Abstract
AIM Recently, D,L-methadone has been put forward as adjuvant treatment in glioblastoma (GBM). METHODS We analyzed the μ-opioid receptor expression in a set of GBM cell lines and investigated the efficacy of D,L-methadone alone and in combination with temozolomide (TMZ). Results & conclusion: Expression of the μ-opioid receptor was similar in the tested cell lines. High concentrations of D,L-methadone induced apoptosis in all cell lines and showed treatment interaction with TMZ. However, in lower dosages, reflecting clinically attainable concentrations, D,L-methadone alone showed no efficacy, and induced even higher proliferation in one specific cell line. Also, no interaction with TMZ was observed. These results suggest caution to the premature use of D,L-methadone in the treatment of GBM patients.
Collapse
Affiliation(s)
| | - Gero Brockhoff
- Department of Gynecology & Obstetrics, University Hospital Regensburg, Regensburg, Germany
| | - Peter Hau
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
- Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, Regensburg, Germany
| | - Arabel Vollmann-Zwerenz
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
- Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, Regensburg, Germany
| | | | - Annette Lohmeier
- Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, Regensburg, Germany
- Department of Neurosurgery, University Hospital Regensburg, Regensburg, Germany
| | - Markus J Riemenschneider
- Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, Regensburg, Germany
- Department of Neuropathology, University Hospital Regensburg, Regensburg, Germany
| | - Claudius Thomé
- Department of Neurosurgery, University Hospital Innsbruck, Innsbruck, Austria
| | - Alexander Brawanski
- Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, Regensburg, Germany
- Department of Neurosurgery, University Hospital Regensburg, Regensburg, Germany
| | - Martin A Proescholdt
- Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, Regensburg, Germany
- Department of Neurosurgery, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
43
|
Repression of Septin9 and Septin2 suppresses tumor growth of human glioblastoma cells. Cell Death Dis 2018; 9:514. [PMID: 29724999 PMCID: PMC5938713 DOI: 10.1038/s41419-018-0547-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 03/27/2018] [Accepted: 03/27/2018] [Indexed: 01/18/2023]
Abstract
Glioblastoma (GBM) is the most common primary malignancy of the central nervous system (CNS) with <10% 5-year survival rate. The growth and invasion of GBM cells into normal brain make the resection and treatment difficult. A better understanding of the biology of GBM cells is crucial to the targeted therapies for the disease. In this study, we identified Septin9 (SEPT9) and Septin2 (SEPT2) as GBM-related genes through integrated multi-omics analysis across independent transcriptomic and proteomic studies. Further studies revealed that expression of SEPT9 and SEPT2 was elevated in glioma tissues and cell lines (A172, U87-MG). Knockdown of SEPT9 and SEPT2 in A172/U87-MG was able to inhibit GBM cell proliferation and arrest cell cycle progression in the S phase in a synergistic mechanism. Moreover, suppression of SEPT9 and SEPT2 decreased the GBM cell invasive capability and significantly impaired the growth of glioma xenografts in nude mice. Furthermore, the decrease in GBM cell growth caused by SEPT9 and SEPT2 RNAi appears to involve two parallel signaling pathway including the p53/p21 axis and MEK/ERK activation. Together, our integration of multi-omics analysis has revealed previously unrecognized synergistic role of SEPT9 and SEPT2 in GBM, and provided novel insights into the targeted therapy of GBM.
Collapse
|
44
|
Jannetti SA, Carlucci G, Carney B, Kossatz S, Shenker L, Carter LM, Salinas B, Brand C, Sadique A, Donabedian PL, Cunanan KM, Gönen M, Ponomarev V, Zeglis BM, Souweidane MM, Lewis JS, Weber WA, Humm JL, Reiner T. PARP-1-Targeted Radiotherapy in Mouse Models of Glioblastoma. J Nucl Med 2018; 59:1225-1233. [PMID: 29572254 DOI: 10.2967/jnumed.117.205054] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 03/05/2018] [Indexed: 12/18/2022] Open
Abstract
The DNA repair enzyme poly(ADP-ribose) polymerase 1 (PARP-1) is overexpressed in glioblastoma, with overall low expression in healthy brain tissue. Paired with the availability of specific small molecule inhibitors, PARP-1 is a near-ideal target to develop novel radiotherapeutics to induce DNA damage and apoptosis in cancer cells, while sparing healthy brain tissue. Methods: We synthesized an 131I-labeled PARP-1 therapeutic and investigated its pharmacology in vitro and in vivo. A subcutaneous tumor model was used to quantify retention times and therapeutic efficacy. A potential clinical scenario, intratumoral convection-enhanced delivery, was mimicked using an orthotopic glioblastoma model combined with an implanted osmotic pump system to study local administration of 131I-PARPi (PARPi is PARP inhibitor). Results:131I-PARPi is a 1(2H)-phthalazinone, similar in structure to the Food and Drug Administration-approved PARP inhibitor AZD-2281. In vitro studies have shown that 131I-PARPi and AZD-2281 share similar pharmacologic profiles. 131I-PARPi delivered 134.1 cGy/MBq intratumoral injected activity. Doses to nontarget tissues, including liver and kidney, were significantly lower. Radiation damage and cell death in treated tumors were shown by p53 activation in U87-MG cells transfected with a p53-bioluminescent reporter. Treated mice showed significantly longer survival than mice receiving vehicle (29 vs. 22 d, P < 0.005) in a subcutaneous model. Convection-enhanced delivery demonstrated efficient retention of 131I-PARPi in orthotopic brain tumors, while quickly clearing from healthy brain tissue. Conclusion: Our results demonstrate 131I-PARPi's high potential as a therapeutic and highlight PARP's relevance as a target for radionuclide therapy. Radiation plays an integral role in brain tumor therapy, and radiolabeled PARP therapeutics could ultimately lead to improvements in the standard of care.
Collapse
Affiliation(s)
- Stephen A Jannetti
- Department of Biochemistry, Hunter College-The City University of New York, New York, New York.,Department of Biochemistry, The Graduate Center, The City University of New York, New York, New York.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Giuseppe Carlucci
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Center for Advanced Imaging Innovation and Research, New York University Langone Medical Center, New York, New York
| | - Brandon Carney
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Chemistry, The Graduate Center, The City University of New York, New York, New York.,Department of Chemistry, Hunter College-The City University of New York, New York, New York
| | - Susanne Kossatz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Larissa Shenker
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lukas M Carter
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Beatriz Salinas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christian Brand
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ahmad Sadique
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Patrick L Donabedian
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kristen M Cunanan
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vladimir Ponomarev
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brian M Zeglis
- Department of Biochemistry, Hunter College-The City University of New York, New York, New York.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Pharmacology, Weill-Cornell Medical College, New York, New York.,Department of Radiology, Weill-Cornell Medical College, New York, New York
| | - Mark M Souweidane
- Department of Neurological Surgery, Weill-Cornell Medical College, New York, New York.,Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Pharmacology, Weill-Cornell Medical College, New York, New York.,Department of Radiology, Weill-Cornell Medical College, New York, New York
| | - Wolfgang A Weber
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill-Cornell Medical College, New York, New York
| | - John L Humm
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York .,Department of Radiology, Weill-Cornell Medical College, New York, New York
| |
Collapse
|
45
|
Zuccarini M, Giuliani P, Ziberi S, Carluccio M, Iorio PD, Caciagli F, Ciccarelli R. The Role of Wnt Signal in Glioblastoma Development and Progression: A Possible New Pharmacological Target for the Therapy of This Tumor. Genes (Basel) 2018; 9:genes9020105. [PMID: 29462960 PMCID: PMC5852601 DOI: 10.3390/genes9020105] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/12/2018] [Accepted: 02/13/2018] [Indexed: 12/26/2022] Open
Abstract
Wnt is a complex signaling pathway involved in the regulation of crucial biological functions such as development, proliferation, differentiation and migration of cells, mainly stem cells, which are virtually present in all embryonic and adult tissues. Conversely, dysregulation of Wnt signal is implicated in development/progression/invasiveness of different kinds of tumors, wherein a certain number of multipotent cells, namely “cancer stem cells”, are characterized by high self-renewal and aggressiveness. Hence, the pharmacological modulation of Wnt pathway could be of particular interest, especially in tumors for which the current standard therapy results to be unsuccessful. This might be the case of glioblastoma multiforme (GBM), one of the most lethal, aggressive and recurrent brain cancers, probably due to the presence of highly malignant GBM stem cells (GSCs) as well as to a dysregulation of Wnt system. By examining the most recent literature, here we point out several factors in the Wnt pathway that are altered in human GBM and derived GSCs, as well as new molecular strategies or experimental drugs able to modulate/inhibit aberrant Wnt signal. Altogether, these aspects serve to emphasize the existence of alternative pharmacological targets that may be useful to develop novel therapies for GBM.
Collapse
Affiliation(s)
- Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, via dei Vestini 29, 66100 Chieti, Italy.
- Aging Research Center and Translational Medicine (CeSI-MeT), via L. Polacchi 11, 66100 Chieti, Italy.
| | - Patricia Giuliani
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, via dei Vestini 29, 66100 Chieti, Italy.
- Aging Research Center and Translational Medicine (CeSI-MeT), via L. Polacchi 11, 66100 Chieti, Italy.
| | - Sihana Ziberi
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, via dei Vestini 29, 66100 Chieti, Italy.
- Aging Research Center and Translational Medicine (CeSI-MeT), via L. Polacchi 11, 66100 Chieti, Italy.
- StemTeCh Group, via L. Polacchi 11, 66100 Chieti, Italy.
| | - Marzia Carluccio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, via dei Vestini 29, 66100 Chieti, Italy.
- Aging Research Center and Translational Medicine (CeSI-MeT), via L. Polacchi 11, 66100 Chieti, Italy.
- StemTeCh Group, via L. Polacchi 11, 66100 Chieti, Italy.
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, via dei Vestini 29, 66100 Chieti, Italy.
- Aging Research Center and Translational Medicine (CeSI-MeT), via L. Polacchi 11, 66100 Chieti, Italy.
| | - Francesco Caciagli
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, via dei Vestini 29, 66100 Chieti, Italy.
- Aging Research Center and Translational Medicine (CeSI-MeT), via L. Polacchi 11, 66100 Chieti, Italy.
| | - Renata Ciccarelli
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, via dei Vestini 29, 66100 Chieti, Italy.
- Aging Research Center and Translational Medicine (CeSI-MeT), via L. Polacchi 11, 66100 Chieti, Italy.
- StemTeCh Group, via L. Polacchi 11, 66100 Chieti, Italy.
| |
Collapse
|
46
|
STAT3 Gene Silencing by Aptamer-siRNA Chimera as Selective Therapeutic for Glioblastoma. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 10:398-411. [PMID: 29499951 PMCID: PMC5862137 DOI: 10.1016/j.omtn.2017.12.021] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 12/28/2017] [Accepted: 12/28/2017] [Indexed: 12/21/2022]
Abstract
Glioblastoma (GBM) is the most frequent and aggressive primary brain tumor in adults, and despite advances in neuro-oncology, the prognosis for patients remains dismal. The signal transducer and activator of transcription-3 (STAT3) has been reported as a key regulator of the highly aggressive mesenchymal GBM subtype, and its direct silencing (by RNAi oligonucleotides) has revealed a great potential as an anti-cancer therapy. However, clinical use of oligonucleotide-based therapies is dependent on safer ways for tissue-specific targeting and increased membrane penetration. The objective of this study is to explore the use of nucleic acid aptamers as carriers to specifically drive a STAT3 siRNA to GBM cells in a receptor-dependent manner. Using an aptamer that binds to and antagonizes the oncogenic receptor tyrosine kinase PDGFRβ (Gint4.T), here we describe the design of a novel aptamer-siRNA chimera (Gint4.T-STAT3) to target STAT3. We demonstrate the efficient delivery and silencing of STAT3 in PDGFRβ+ GBM cells. Importantly, the conjugate reduces cell viability and migration in vitro and inhibits tumor growth and angiogenesis in vivo in a subcutaneous xenograft mouse model. Our data reveals Gint4.T-STAT3 conjugate as a novel molecule with great translational potential for GBM therapy.
Collapse
|
47
|
Haj A, Doenitz C, Schebesch KM, Ehrensberger D, Hau P, Putnik K, Riemenschneider MJ, Wendl C, Gerken M, Pukrop T, Brawanski A, Proescholdt MA. Extent of Resection in Newly Diagnosed Glioblastoma: Impact of a Specialized Neuro-Oncology Care Center. Brain Sci 2017; 8:brainsci8010005. [PMID: 29295569 PMCID: PMC5789336 DOI: 10.3390/brainsci8010005] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/07/2017] [Accepted: 12/19/2017] [Indexed: 02/05/2023] Open
Abstract
Treatment of glioblastoma (GBM) consists of microsurgical resection followed by concomitant radiochemotherapy and adjuvant chemotherapy. The best outcome regarding progression free (PFS) and overall survival (OS) is achieved by maximal resection. The foundation of a specialized neuro-oncology care center (NOC) has enabled the implementation of a large technical portfolio including functional imaging, awake craniotomy, PET scanning, fluorescence-guided resection, and integrated postsurgical therapy. This study analyzed whether the technically improved neurosurgical treatment structure yields a higher rate of complete resection, thus ultimately improving patient outcome. Patients and methods: The study included 149 patients treated surgically for newly diagnosed GBM. The neurological performance score (NPS) and the Karnofsky performance score (KPS) were measured before and after resection. The extent of resection (EOR) was volumetrically quantified. Patients were stratified into two subcohorts: treated before (A) and after (B) the foundation of the Regensburg NOC. The EOR and the PFS and OS were evaluated. Results: Prognostic factors for PFS and OS were age, preoperative KPS, O6-methylguanine-DNA-methyltransferase (MGMT) promoter methylation status, isocitrate dehydrogenase 1 (IDH1) mutation status and EOR. Patients with volumetrically defined complete resection had significantly better PFS (9.4 vs. 7.8 months; p = 0.042) and OS (18.4 vs. 14.5 months; p = 0.005) than patients with incomplete resection. The frequency of transient or permanent postoperative neurological deficits was not higher after complete resection in both subcohorts. The frequency of complete resection was significantly higher in subcohort B than in subcohort A (68.2% vs. 34.8%; p = 0.007). Accordingly, subcohort B showed significantly longer PFS (8.6 vs. 7.5 months; p = 0.010) and OS (18.7 vs. 12.4 months; p = 0.001). Multivariate Cox regression analysis showed complete resection, age, preoperative KPS, and MGMT promoter status as independent prognostic factors for PFS and OS. Our data show a higher frequency of complete resection in patients with GBM after the establishment of a series of technical developments that resulted in significantly better PFS and OS without increasing surgery-related morbidity.
Collapse
Affiliation(s)
- Amer Haj
- Wilhelm Sander Neuro-Oncology Unit, University Medical Center Regensburg, 93053 Regensburg, Germany.
- Department of Neurosurgery, University Medical Center Regensburg, 93053 Regensburg, Germany.
| | - Christian Doenitz
- Wilhelm Sander Neuro-Oncology Unit, University Medical Center Regensburg, 93053 Regensburg, Germany.
- Department of Neurosurgery, University Medical Center Regensburg, 93053 Regensburg, Germany.
| | - Karl-Michael Schebesch
- Wilhelm Sander Neuro-Oncology Unit, University Medical Center Regensburg, 93053 Regensburg, Germany.
- Department of Neurosurgery, University Medical Center Regensburg, 93053 Regensburg, Germany.
| | - Denise Ehrensberger
- Wilhelm Sander Neuro-Oncology Unit, University Medical Center Regensburg, 93053 Regensburg, Germany.
- Department of Neurosurgery, University Medical Center Regensburg, 93053 Regensburg, Germany.
| | - Peter Hau
- Wilhelm Sander Neuro-Oncology Unit, University Medical Center Regensburg, 93053 Regensburg, Germany.
- Department of Neurology, University Medical Center Regensburg, 93053 Regensburg, Germany.
| | - Kurt Putnik
- Wilhelm Sander Neuro-Oncology Unit, University Medical Center Regensburg, 93053 Regensburg, Germany.
- Department of Radiation Oncology, University Medical Center Regensburg, 93053 Regensburg, Germany.
| | - Markus J Riemenschneider
- Wilhelm Sander Neuro-Oncology Unit, University Medical Center Regensburg, 93053 Regensburg, Germany.
- Department of Neuropathology, University Medical Center Regensburg, 93053 Regensburg, Germany.
| | - Christina Wendl
- Wilhelm Sander Neuro-Oncology Unit, University Medical Center Regensburg, 93053 Regensburg, Germany.
- Department of Neuroradiology, University Medical Center Regensburg, 93053 Regensburg, Germany.
| | - Michael Gerken
- Wilhelm Sander Neuro-Oncology Unit, University Medical Center Regensburg, 93053 Regensburg, Germany.
- Tumor Center Regensburg, Institute of Quality Assurance and Health Services Research, University of Regensburg, 93053 Regensburg, Germany.
| | - Tobias Pukrop
- Wilhelm Sander Neuro-Oncology Unit, University Medical Center Regensburg, 93053 Regensburg, Germany.
- Department of Hematology and Oncology, University Medical Center Regensburg, 93053 Regensburg, Germany.
| | - Alexander Brawanski
- Wilhelm Sander Neuro-Oncology Unit, University Medical Center Regensburg, 93053 Regensburg, Germany.
- Department of Neurosurgery, University Medical Center Regensburg, 93053 Regensburg, Germany.
| | - Martin A Proescholdt
- Wilhelm Sander Neuro-Oncology Unit, University Medical Center Regensburg, 93053 Regensburg, Germany.
- Department of Neurosurgery, University Medical Center Regensburg, 93053 Regensburg, Germany.
| |
Collapse
|
48
|
Wang X, Zhang Q, Lv L, Fu J, Jiang Y, Xin H, Yao Q. Glioma and microenvironment dual targeted nanocarrier for improved antiglioblastoma efficacy. Drug Deliv 2017; 24:1401-1409. [PMID: 28933201 PMCID: PMC8241031 DOI: 10.1080/10717544.2017.1378940] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Drug delivery systems based on nanoparticles (nano-DDS) have aroused attentions for the treatment of glioblastoma (GBM), the most malignant brain cancer with a dismal prognosis. However, there are still numerous unmet challenges for traditional nano-DDS, such as the poor nanoparticle penetration, short retention in the GBM parenchyma and low glioma targeting ability. Herein, we used Pep-1 and CREKA peptides to construct a novel multifunctional GBM targeting nano-DDS (PC-NP). Pep-1 was used to overcome the blood-brain tumor barrier (BBTB) and home to glioma cells via interleukin-13 receptor-α2-mediated endocytosis, and CREKA was used to bind to fibrin-fibronectin complexes abundantly expressed in tumor microenvironment for enhanced retention in the GBM. Biological studies showed that the cellular uptake of PC-NP by U87MG cells was significantly enhanced compared with the non-targeting NP. Furthermore, CREKA modification increased the binding capacity of PC-NP to fibrin-fibronectin complexes as confirmed by the competition experiment. In accordance with the increased cellular uptake, PC-NP remarkably increased the cytotoxicity of its payload paclitaxel (PTX) against U87MG cells with an IC50 of 0.176 μg/mL. In vivo fluorescence imaging and antiglioma efficacy evaluation further confirmed that PC-NP accumulated effectively and penetrated deeply into GBM tissue. PC-NP-PTX exhibited a median survival time as long as 61 days in intracranial GBM-bearing mice. In conclusion, our findings indicated PC-NP as a promising nano-DDS for GBM targeting delivery of anticancer drugs.
Collapse
Affiliation(s)
- Xiuzhen Wang
- a Department of Medicinal Chemistry, School of Pharmacy , China Pharmaceutical University , Nanjing , China.,b School of Pharmacy , Nanjing Medical University , Nanjing , China
| | - Qing Zhang
- b School of Pharmacy , Nanjing Medical University , Nanjing , China
| | - Lingyan Lv
- b School of Pharmacy , Nanjing Medical University , Nanjing , China
| | - Junjie Fu
- b School of Pharmacy , Nanjing Medical University , Nanjing , China
| | - Yan Jiang
- b School of Pharmacy , Nanjing Medical University , Nanjing , China
| | - Hongliang Xin
- b School of Pharmacy , Nanjing Medical University , Nanjing , China
| | - Qizheng Yao
- a Department of Medicinal Chemistry, School of Pharmacy , China Pharmaceutical University , Nanjing , China
| |
Collapse
|
49
|
Bastiancich C, Bastiat G, Lagarce F. Gemcitabine and glioblastoma: challenges and current perspectives. Drug Discov Today 2017; 23:416-423. [PMID: 29074439 DOI: 10.1016/j.drudis.2017.10.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 09/22/2017] [Accepted: 10/12/2017] [Indexed: 12/15/2022]
Abstract
Gemcitabine is a nucleoside analog currently used for the treatment of various solid tumors as a single agent or in combination with other chemotherapeutic drugs. Its use against highly aggressive brain tumors (glioblastoma) has been evaluated in preclinical and clinical trials leading to controversial results. Gemcitabine can inhibit DNA chain elongation, is a potent radiosensitizer and it can enhance antitumor immune activity, but it also presents some drawbacks (e.g., short half-life, side effects, chemoresistance). The aim of this review is to discuss the challenges related to the use of gemcitabine for glioblastoma and to report recent studies that suggest overcoming these obstacles opening new perspectives for its use in the field (e.g., gemcitabine derivatives and/or nanomedicines).
Collapse
Affiliation(s)
- Chiara Bastiancich
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, Angers, France; Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Guillaume Bastiat
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, Angers, France
| | - Frederic Lagarce
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, Angers, France; Pharmacy Department, CHU Angers, Angers University Hospital, France.
| |
Collapse
|
50
|
Bastiancich C, Bianco J, Vanvarenberg K, Ucakar B, Joudiou N, Gallez B, Bastiat G, Lagarce F, Préat V, Danhier F. Injectable nanomedicine hydrogel for local chemotherapy of glioblastoma after surgical resection. J Control Release 2017; 264:45-54. [DOI: 10.1016/j.jconrel.2017.08.019] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 08/18/2017] [Indexed: 12/28/2022]
|