1
|
Gan Y, Huang X, Guo W, Yan C, Zou G. Predicting synergistic anticancer drug combination based on low-rank global attention mechanism and bilinear predictor. Bioinformatics 2023; 39:btad607. [PMID: 37812255 PMCID: PMC10598574 DOI: 10.1093/bioinformatics/btad607] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/29/2023] [Accepted: 10/07/2023] [Indexed: 10/10/2023] Open
Abstract
MOTIVATION Drug combination therapy has exhibited remarkable therapeutic efficacy and has gradually become a promising clinical treatment strategy of complex diseases such as cancers. As the related databases keep expanding, computational methods based on deep learning model have become powerful tools to predict synergistic drug combinations. However, predicting effective synergistic drug combinations is still a challenge due to the high complexity of drug combinations, the lack of biological interpretability, and the large discrepancy in the response of drug combinations in vivo and in vitro biological systems. RESULTS Here, we propose DGSSynADR, a new deep learning method based on global structured features of drugs and targets for predicting synergistic anticancer drug combinations. DGSSynADR constructs a heterogeneous graph by integrating the drug-drug, drug-target, protein-protein interactions and multi-omics data, utilizes a low-rank global attention (LRGA) model to perform global weighted aggregation of graph nodes and learn the global structured features of drugs and targets, and then feeds the embedded features into a bilinear predictor to predict the synergy scores of drug combinations in different cancer cell lines. Specifically, LRGA network brings better model generalization ability, and effectively reduces the complexity of graph computation. The bilinear predictor facilitates the dimension transformation of the features and fuses the feature representation of the two drugs to improve the prediction performance. The loss function Smooth L1 effectively avoids gradient explosion, contributing to better model convergence. To validate the performance of DGSSynADR, we compare it with seven competitive methods. The comparison results demonstrate that DGSSynADR achieves better performance. Meanwhile, the prediction of DGSSynADR is validated by previous findings in case studies. Furthermore, detailed ablation studies indicate that the one-hot coding drug feature, LRGA model and bilinear predictor play a key role in improving the prediction performance. AVAILABILITY AND IMPLEMENTATION DGSSynADR is implemented in Python using the Pytorch machine-learning library, and it is freely available at https://github.com/DHUDBlab/DGSSynADR.
Collapse
Affiliation(s)
- Yanglan Gan
- School of Computer Science and Technology, Donghua University, Shanghai 201600, China
| | - Xingyu Huang
- School of Computer Science and Technology, Donghua University, Shanghai 201600, China
| | - Wenjing Guo
- School of Computer Science and Technology, Donghua University, Shanghai 201600, China
| | - Cairong Yan
- School of Computer Science and Technology, Donghua University, Shanghai 201600, China
| | - Guobing Zou
- School of Computer Engineering and Science, Shanghai University, Shanghai 200444, China
| |
Collapse
|
2
|
Kalita O, Kazda T, Reguli S, Jancalek R, Fadrus P, Slachta M, Pospisil P, Krska L, Vrbkova J, Hrabalek L, Smrcka M, Lipina R. Effects of Reoperation Timing on Survival among Recurrent Glioblastoma Patients: A Retrospective Multicentric Descriptive Study. Cancers (Basel) 2023; 15:cancers15092530. [PMID: 37173996 PMCID: PMC10177480 DOI: 10.3390/cancers15092530] [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: 02/14/2023] [Revised: 04/14/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Glioblastoma inevitably recurs, but no standard regimen has been established for treating this recurrent disease. Several reports claim that reoperative surgery can improve survival, but the effects of reoperation timing on survival have rarely been investigated. We, therefore, evaluated the relationship between reoperation timing and survival in recurrent GBM. A consecutive cohort of unselected patients (real-world data) from three neuro-oncology cancer centers was analyzed (a total of 109 patients). All patients underwent initial maximal safe resection followed by treatment according to the Stupp protocol. Those meeting the following criteria during progression were indicated for reoperation and were further analyzed in this study: (1) The tumor volume increased by >20-30% or a tumor was rediscovered after radiological disappearance; (2) The patient's clinical status was satisfactory (KS ≥ 70% and PS WHO ≤ gr. 2); (3) The tumor was localized without multifocality; (4) The minimum expected tumor volume reduction was above 80%. A univariate Cox regression analysis of postsurgical survival (PSS) revealed a statistically significant effect of reoperation on PSS from a threshold of 16 months after the first surgery. Cox regression models that stratified the Karnofsky score with age adjustment confirmed a statistically significant improvement in PSS for time-to-progression (TTP) thresholds of 22 and 24 months. The patient groups exhibiting the first recurrence at 22 and 24 months had better survival rates than those exhibiting earlier recurrences. For the 22-month group, the HR was 0.5 with a 95% CI of (0.27, 0.96) and a p-value of 0.036. For the 24-month group, the HR was 0.5 with a 95% CI of (0.25, 0.96) and a p-value of 0.039. Patients with the longest survival were also the best candidates for repeated surgery. Later recurrence of glioblastoma was associated with higher survival rates after reoperation.
Collapse
Affiliation(s)
- Ondrej Kalita
- Department of Neurosurgery, Faculty of Medicine and Dentistry, Palacky University in Olomouc, University Hospital Olomouc, Zdravotníků 248/7, 779 00 Olomouc, Czech Republic
- Department of Health Care Science, Faculty of Humanities, T. Bata University in Zlin, Stefanikova 5670, 760 01 Zlín, Czech Republic
| | - Tomas Kazda
- Department of Radiation Oncology, Faculty of Medicine, Masaryk University, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53 Brno, Czech Republic
| | - Stefan Reguli
- Department of Neurosurgery, Faculty of Medicine, University of Ostrava, University Hospital Ostrava, 17. Listopadu 1790/5, 708 52 Ostrava, Czech Republic
| | - Radim Jancalek
- Department of Neurosurgery, Faculty of Medicine, Masaryk University, St. Anne's University Hospital in Brno, Pekarska 664/53, 602 00 Brno, Czech Republic
| | - Pavel Fadrus
- Department of Neurosurgery, Faculty of Medicine, Masaryk University, University Hospital Brno, Jihlavská 20, 625 00 Brno, Czech Republic
| | - Marek Slachta
- Department of Neurosurgery, Faculty of Medicine and Dentistry, Palacky University in Olomouc, University Hospital Olomouc, Zdravotníků 248/7, 779 00 Olomouc, Czech Republic
| | - Petr Pospisil
- Department of Radiation Oncology, Faculty of Medicine, Masaryk University, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53 Brno, Czech Republic
| | - Lukas Krska
- Department of Neurosurgery, Faculty of Medicine, University of Ostrava, University Hospital Ostrava, 17. Listopadu 1790/5, 708 52 Ostrava, Czech Republic
| | - Jana Vrbkova
- Institute of Molecular and Translate Medicine, Faculty of Medicine and Dentistry, Palacky University in Olomouc, Hnevotinska 133/5, 779 00 Olomouc, Czech Republic
| | - Lumir Hrabalek
- Department of Neurosurgery, Faculty of Medicine and Dentistry, Palacky University in Olomouc, University Hospital Olomouc, Zdravotníků 248/7, 779 00 Olomouc, Czech Republic
| | - Martin Smrcka
- Department of Neurosurgery, Faculty of Medicine, Masaryk University, University Hospital Brno, Jihlavská 20, 625 00 Brno, Czech Republic
| | - Radim Lipina
- Department of Neurosurgery, Faculty of Medicine, University of Ostrava, University Hospital Ostrava, 17. Listopadu 1790/5, 708 52 Ostrava, Czech Republic
| |
Collapse
|
3
|
Bausart M, Rodella G, Dumont M, Ucakar B, Vanvarenberg K, Malfanti A, Préat V. Combination of local immunogenic cell death-inducing chemotherapy and DNA vaccine increases the survival of glioblastoma-bearing mice. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 50:102681. [PMID: 37105343 DOI: 10.1016/j.nano.2023.102681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/22/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023]
Abstract
Immunotherapy efficacy as monotherapy is negligible for glioblastoma (GBM). We hypothesized that combining therapeutic vaccination using a plasmid encoding an epitope derived from GBM-associated antigen (pTOP) with local delivery of immunogenic chemotherapy using mitoxantrone-loaded PEGylated PLGA-based nanoparticles (NP-MTX) would improve the survival of GBM-bearing mice by stimulating an antitumor immune response. We first proved that MTX retained its ability to induce cytotoxicity and immunogenic cell death of GBM cells after encapsulation. Intratumoral delivery of MTX or NP-MTX increased the frequency of IFN-γ-secreting CD8 T cells. NP-MTX mixed with free MTX in combination with pTOP DNA vaccine increased the median survival of GL261-bearing mice and increased M1-like macrophages in the brain. The addition of CpG to this combination abolished the survival benefit but led to increased M1 to M2 macrophage ratio and IFN-γ-secreting CD4 T cell frequency. These results highlight the benefits of combination strategies to potentiate immunotherapy and improve GBM outcome.
Collapse
Affiliation(s)
- Mathilde Bausart
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Giulia Rodella
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Mathilde Dumont
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Bernard Ucakar
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Kevin Vanvarenberg
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium.
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium.
| |
Collapse
|
4
|
Vedunova M, Turubanova V, Vershinina O, Savyuk M, Efimova I, Mishchenko T, Raedt R, Vral A, Vanhove C, Korsakova D, Bachert C, Coppieters F, Agostinis P, Garg AD, Ivanchenko M, Krysko O, Krysko DV. DC vaccines loaded with glioma cells killed by photodynamic therapy induce Th17 anti-tumor immunity and provide a four-gene signature for glioma prognosis. Cell Death Dis 2022; 13:1062. [PMID: 36539408 PMCID: PMC9767932 DOI: 10.1038/s41419-022-05514-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 12/12/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
Gliomas, the most frequent type of primary tumor of the central nervous system in adults, results in significant morbidity and mortality. Despite the development of novel, complex, multidisciplinary, and targeted therapies, glioma therapy has not progressed much over the last decades. Therefore, there is an urgent need to develop novel patient-adjusted immunotherapies that actively stimulate antitumor T cells, generate long-term memory, and result in significant clinical benefits. This work aimed to investigate the efficacy and molecular mechanism of dendritic cell (DC) vaccines loaded with glioma cells undergoing immunogenic cell death (ICD) induced by photosens-based photodynamic therapy (PS-PDT) and to identify reliable prognostic gene signatures for predicting the overall survival of patients. Analysis of the transcriptional program of the ICD-based DC vaccine led to the identification of robust induction of Th17 signature when used as a vaccine. These DCs demonstrate retinoic acid receptor-related orphan receptor-γt dependent efficacy in an orthotopic mouse model. Moreover, comparative analysis of the transcriptome program of the ICD-based DC vaccine with transcriptome data from the TCGA-LGG dataset identified a four-gene signature (CFH, GALNT3, SMC4, VAV3) associated with overall survival of glioma patients. This model was validated on overall survival of CGGA-LGG, TCGA-GBM, and CGGA-GBM datasets to determine whether it has a similar prognostic value. To that end, the sensitivity and specificity of the prognostic model for predicting overall survival were evaluated by calculating the area under the curve of the time-dependent receiver operating characteristic curve. The values of area under the curve for TCGA-LGG, CGGA-LGG, TCGA-GBM, and CGGA-GBM for predicting five-year survival rates were, respectively, 0.75, 0.73, 0.9, and 0.69. These data open attractive prospects for improving glioma therapy by employing ICD and PS-PDT-based DC vaccines to induce Th17 immunity and to use this prognostic model to predict the overall survival of glioma patients.
Collapse
Affiliation(s)
- Maria Vedunova
- grid.28171.3d0000 0001 0344 908XInstitute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Victoria Turubanova
- grid.28171.3d0000 0001 0344 908XInstitute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia ,grid.5342.00000 0001 2069 7798Cell Death Investigation and Therapy (CDIT) Laboratory, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Olga Vershinina
- grid.28171.3d0000 0001 0344 908XInstitute of Information Technology, Mathematics and Mechanics, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Maria Savyuk
- grid.28171.3d0000 0001 0344 908XInstitute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia ,grid.5342.00000 0001 2069 7798Cell Death Investigation and Therapy (CDIT) Laboratory, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Iuliia Efimova
- grid.5342.00000 0001 2069 7798Cell Death Investigation and Therapy (CDIT) Laboratory, Department of Human Structure and Repair, Ghent University, Ghent, Belgium ,grid.510942.bCancer Research Institute Ghent, Ghent, Belgium
| | - Tatiana Mishchenko
- grid.28171.3d0000 0001 0344 908XInstitute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Robrecht Raedt
- grid.5342.00000 0001 2069 77984Brain Team, Department of Head and Skin, Ghent University, Ghent, Belgium
| | - Anne Vral
- grid.5342.00000 0001 2069 7798Radiobiology Research Group, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Christian Vanhove
- grid.5342.00000 0001 2069 7798IBiTech-MEDISIP-Infinity Laboratory, Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Daria Korsakova
- grid.28171.3d0000 0001 0344 908XInstitute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Claus Bachert
- grid.5342.00000 0001 2069 7798Upper Airways Research Laboratory, Department of Head and Skin, Ghent University, Ghent, Belgium
| | - Frauke Coppieters
- grid.5342.00000 0001 2069 7798Center for Medical Genetics Ghent (CMGG), Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Patrizia Agostinis
- grid.5596.f0000 0001 0668 7884Laboratory of Cell Death Research & Therapy, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium ,grid.511459.dVIB Center for Cancer Biology Research, Leuven, Belgium
| | - Abhishek D. Garg
- grid.5596.f0000 0001 0668 7884Laboratory of Cell Stress & Immunity (CSI), Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Mikhail Ivanchenko
- grid.28171.3d0000 0001 0344 908XInstitute of Information Technology, Mathematics and Mechanics, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Olga Krysko
- grid.5342.00000 0001 2069 7798Cell Death Investigation and Therapy (CDIT) Laboratory, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Dmitri V. Krysko
- grid.28171.3d0000 0001 0344 908XInstitute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia ,grid.5342.00000 0001 2069 7798Cell Death Investigation and Therapy (CDIT) Laboratory, Department of Human Structure and Repair, Ghent University, Ghent, Belgium ,grid.510942.bCancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
5
|
Mouawad R, Neamati N. Inhibition of Protein Disulfide Isomerase (PDIA1) Leads to Proteasome-Mediated Degradation of Ubiquitin-like PHD and RING Finger Domain-Containing Protein 1 (UHRF1) and Increased Sensitivity of Glioblastoma Cells to Topoisomerase II Inhibitors. ACS Pharmacol Transl Sci 2022; 6:100-114. [PMID: 36654750 PMCID: PMC9841782 DOI: 10.1021/acsptsci.2c00186] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Indexed: 12/12/2022]
Abstract
Glioblastoma (GBM) is the most aggressive brain tumor, and the prognosis remains poor with current available treatments. PDIA1 is considered a promising therapeutic target in GBM. In this study, we demonstrate that targeting PDIA1 results in increased GBM cell death by topoisomerase II (Top-II) inhibitors resulting in proteasome-mediated degradation of the oncogenic protein UHRF1. Combination of the PDIA1 inhibitor, bepristat-2a, produces strong synergy with doxorubicin, etoposide, and mitoxantrone in GBM and other cancer cell lines. Our bioinformatics analysis of multiple datasets revealed downregulation of UHRF1, upon PDIA1 inhibition. In addition, PDIA1 inhibition results in proteasome-mediated degradation of UHRF1 protein. Interestingly, treatment of GBM cells with bepristat-2a results in increased apoptosis and resistance to ferroptosis. Our findings emphasize the importance of PDIA1 as a therapeutic target in GBM and present a promising new therapeutic approach using Top-II inhibitors for GBM treatment.
Collapse
|
6
|
Urbaniak A, Reed MR, Heflin B, Gaydos J, Piña-Oviedo S, Jędrzejczyk M, Klejborowska G, Stępczyńska N, Chambers TC, Tackett AJ, Rodriguez A, Huczyński A, Eoff RL, MacNicol AM. Anti-glioblastoma activity of monensin and its analogs in an organoid model of cancer. Biomed Pharmacother 2022; 153:113440. [PMID: 36076555 PMCID: PMC9472755 DOI: 10.1016/j.biopha.2022.113440] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 11/30/2022] Open
Abstract
Glioblastoma (GBM) remains the most frequently diagnosed primary malignant brain cancer in adults. Despite recent progress in understanding the biology of GBM, the clinical outcome for patients remains poor, with a median survival of approximately one year after diagnosis. One factor contributing to failure in clinical trials is the fact that traditional models used in GBM drug discovery poorly recapitulate patient tumors. Previous studies have shown that monensin (MON) analogs, namely esters and amides on C-26 were potent towards various types of cancer cell lines. In the present study we have investigated the activity of these molecules in GBM organoids, as well as in a host:tumor organoid model. Using a mini-ring cell viability assay we have identified seven analogs (IC50 = 91.5 ± 54.4–291.7 ± 68.8 nM) more potent than parent MON (IC50 = 612.6 ± 184.4 nM). Five of these compounds induced substantial DNA fragmentation in GBM organoids, suggestive of apoptotic cell death. The most active analog, compound 1, significantly reduced GBM cell migration, induced PARP degradation, diminished phosphorylation of STAT3, Akt and GSK3β, increased ɣH2AX signaling and upregulated expression of the autophagy associated marker LC3-II. To investigate the activity of MON and compound 1 in a tumor microenvironment, we developed human cerebral organoids (COs) from human induced pluripotent stem cells (iPSCs). The COs showed features of early developing brain such as multiple neural rosettes with a proliferative zone of neural stem cells (Nestin+), neurons (TUJ1 +), primitive ventricular system (SOX2 +/Ki67 +), intermediate zone (TBR2 +) and cortical plate (MAP2 +). In order to generate host:tumor organoids, we co-cultured RFP-labeled U87MG cells with fully formed COs. Compound 1 and MON reduced U87MG tumor size in the COs after four days of treatment and induced a significant reduction of PARP expression. These findings highlight the therapeutic potential of MON analogs towards GBM and support the application of organoid models in anti-cancer drug discovery.
Collapse
Affiliation(s)
- Alicja Urbaniak
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| | - Megan R Reed
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Billie Heflin
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - John Gaydos
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Sergio Piña-Oviedo
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Marta Jędrzejczyk
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland
| | - Greta Klejborowska
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland
| | - Natalia Stępczyńska
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland
| | - Timothy C Chambers
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Alan J Tackett
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Analiz Rodriguez
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Adam Huczyński
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland
| | - Robert L Eoff
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Angus M MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| |
Collapse
|
7
|
Han D, Teng L, Wang X, Zhen Y, Chen X, Yang M, Gao M, Yang G, Han M, Wang L, Xu J, Li Y, Shumadalova A, Zhao S. Phase I/II trial of local interstitial chemotherapy with arsenic trioxide in patients with newly diagnosed glioma. Front Neurol 2022; 13:1001829. [PMID: 36212657 PMCID: PMC9535358 DOI: 10.3389/fneur.2022.1001829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background Glioma is the most common primary brain tumor in adults with poor prognosis. The glioma patients benefit from STUPP strategy, including maximum and safe resection and adjuvant radiotherapy and chemotherapy. Arsenic trioxide could inhibit various tumors. However, it is a challenge to evaluate the efficiency and safety of srsenic trioxide in glioma patients. Objective The arsenic trioxide has the potent therapeutic effect on glioma. However, the safety and efficacy of local interstitial chemotherapy with arsenic trioxide in newly diagnosed glioma patients is unclear. Methods All patients received partial or complete tumor resection and intraoperative implantation of Ommaya reservoirs followed by standard radiotherapy. Arsenic trioxide with the starting dose 0.3 mg was administered via an Ommaya reservoir catheter inserted into the tumor cavity for 5 consecutive days every 3 months for a total of eight cycles unless tumor progression or excessive toxicity was observed. Results No hematological or grade 4 non-hematological toxicity was observed in any patient during arsenic trioxide treatment. The maximum tolerated dose of 1.5 mg of arsenic trioxide was safe and well tolerated. The median overall survival for WHO grade 3 glioma was 33.6 months, and for glioblastoma was 13.9 months. The median progression-free survival for WHO grade 2 glioma was 40.3 months, for grade 3 glioma was 21.5 months, and for glioblastoma was 9.5 months. Conclusion These results suggest that arsenic trioxide is safe and well tolerated with local delivery into the tumor cavity of the brain, and the dose recommended for a phase II trial is 1.5 mg.
Collapse
Affiliation(s)
- Dayong Han
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Institute of Brain Science, Harbin Medical University, Harbin, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Lei Teng
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Institute of Brain Science, Harbin Medical University, Harbin, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Xiaoxiong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Institute of Brain Science, Harbin Medical University, Harbin, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Yunbo Zhen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Institute of Brain Science, Harbin Medical University, Harbin, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Xiaofeng Chen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Institute of Brain Science, Harbin Medical University, Harbin, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Mingchun Yang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Institute of Brain Science, Harbin Medical University, Harbin, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Ming Gao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Institute of Brain Science, Harbin Medical University, Harbin, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Guang Yang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Institute of Brain Science, Harbin Medical University, Harbin, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Mingyang Han
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, China
| | - Ligang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Institute of Brain Science, Harbin Medical University, Harbin, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Jiajun Xu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Institute of Brain Science, Harbin Medical University, Harbin, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Yue Li
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Alina Shumadalova
- Department of General Chemistry, Bashkir State Medical University, Ufa, Russia
| | - Shiguang Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Institute of Brain Science, Harbin Medical University, Harbin, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, China
- *Correspondence: Shiguang Zhao
| |
Collapse
|
8
|
Feng Q, Xu X, Wei C, Li Y, Wang M, Lv C, Wu J, Dai Y, Han Y, Lesniak MS, Fan H, Zhang L, Cheng Y. The Dynamic Interactions between Nanoparticles and Macrophages Impact Their Fate in Brain Tumors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2103600. [PMID: 34643042 DOI: 10.1002/smll.202103600] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/10/2021] [Indexed: 06/13/2023]
Abstract
Functional nanomaterials such as iron oxide nanoparticles have been extensively explored for the diagnosis and treatment of central nervous system diseases. However, an insufficient understanding of the comprehensive nanomaterial-biological interactions in the brain hinders the nanomaterials from meeting the medical requirements for translational research. Here, FDA-approved ferumoxytol, an iron oxide nanoparticle, is chosen as the model nanomaterial for a systematic study of the dynamic interactions between ferumoxytol and immune cells, including microglia and macrophages, in the brain tumors. Strikingly, up to 90% of intratumorally injected ferumoxytol nanoparticles are recognized and phagocytized by tumor-associated microglia and macrophages. The dynamic trafficking progress of ferumoxytol in microglia and macrophages, including scavenger receptor-mediated endocytosis, lysosomal internalization, and extracellular vesicle-dominated excretion, is further studied. Importantly, the results demonstrate that extracellular vesicle-encapsulated nanoparticles could be gradually eliminated from the brain along with cerebrospinal fluid circulation over 21 days. Moreover, ferumoxytol exhibits no obvious long-term neurological toxicity after its injection. The study suggests that the dynamic biointeractions of nanoparticles with immune cells in the brain exert a key rate-limiting impact on the efficiency of targeting tumor cells and their in vivo fate and thus provide a deeper understanding of the nanomaterials in the brain for clinical applications.
Collapse
Affiliation(s)
- Qishuai Feng
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Collaborative Innovation Center for Brain Science, Tongji University, 1800 Yuntai Road, Shanghai, 200123, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 200092, China
| | - Xianyun Xu
- The First Rehabilitation Hospital of Shanghai, Tongji University School of Medicine, 349 Hangzhou Road, Shanghai, 200090, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Department of Pediatric Surgery, The First Affiliated Hospital of Gannan Medical University, Gannan Medical University, 1 Yixueyuan Road, Ganzhou, Jiangxi, 341000, China
| | - Chen Wei
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Collaborative Innovation Center for Brain Science, Tongji University, 1800 Yuntai Road, Shanghai, 200123, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 200092, China
| | - Yingze Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Collaborative Innovation Center for Brain Science, Tongji University, 1800 Yuntai Road, Shanghai, 200123, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 200092, China
| | - Min Wang
- Shanghai Key Lab of Tuberculofsis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, 507 Zhengmin Road, Shanghai, 200433, China
| | - Cheng Lv
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Collaborative Innovation Center for Brain Science, Tongji University, 1800 Yuntai Road, Shanghai, 200123, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 200092, China
| | - Jiaojiao Wu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Collaborative Innovation Center for Brain Science, Tongji University, 1800 Yuntai Road, Shanghai, 200123, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 200092, China
| | - Yalei Dai
- Shanghai Key Lab of Tuberculofsis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, 507 Zhengmin Road, Shanghai, 200433, China
| | - Yu Han
- Feinberg School of Medicine, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, IL, 60611, USA
| | - Maciej S Lesniak
- Feinberg School of Medicine, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, IL, 60611, USA
| | - Haiming Fan
- College of Chemistry and Materials Science, Northwest University, Xi'an, 710127, China
| | - Ling Zhang
- The First Rehabilitation Hospital of Shanghai, Tongji University School of Medicine, 349 Hangzhou Road, Shanghai, 200090, China
| | - Yu Cheng
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Collaborative Innovation Center for Brain Science, Tongji University, 1800 Yuntai Road, Shanghai, 200123, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 200092, China
| |
Collapse
|
9
|
McBain C, Lawrie TA, Rogozińska E, Kernohan A, Robinson T, Jefferies S. Treatment options for progression or recurrence of glioblastoma: a network meta-analysis. Cochrane Database Syst Rev 2021; 5:CD013579. [PMID: 34559423 PMCID: PMC8121043 DOI: 10.1002/14651858.cd013579.pub2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Glioblastoma (GBM) is a highly malignant brain tumour that almost inevitably progresses or recurs after first line standard of care. There is no consensus regarding the best treatment/s to offer people upon disease progression or recurrence. For the purposes of this review, progression and recurrence are considered as one entity. OBJECTIVES To evaluate the effectiveness of further treatment/s for first and subsequent progression or recurrence of glioblastoma (GBM) among people who have received the standard of care (Stupp protocol) for primary treatment of the disease; and to prepare a brief economic commentary on the available evidence. SEARCH METHODS We searched MEDLINE and Embase electronic databases from 2005 to December 2019 and the Cochrane Central Register of Controlled Trials (CENTRAL, in the Cochrane Library; Issue 12, 2019). Economic searches included the National Health Service Economic Evaluation Database (NHS EED) up to 2015 (database closure) and MEDLINE and Embase from 2015 to December 2019. SELECTION CRITERIA Randomised controlled trials (RCTs) and comparative non-randomised studies (NRSs) evaluating effectiveness of treatments for progressive/recurrent GBM. Eligible studies included people with progressive or recurrent GBM who had received first line radiotherapy with concomitant and adjuvant temozolomide (TMZ). DATA COLLECTION AND ANALYSIS Two review authors independently selected studies and extracted data to a pre-designed data extraction form. We conducted network meta-analyses (NMA) and ranked treatments according to effectiveness for each outcome using the random-effects model and Stata software (version 15). We rated the certainty of evidence using the GRADE approach. MAIN RESULTS We included 42 studies: these comprised 34 randomised controlled trials (RCTs) and 8 non-randomised studies (NRSs) involving 5236 participants. We judged most RCTs to be at a low risk of bias and NRSs at high risk of bias. Interventions included chemotherapy, re-operation, re-irradiation and novel therapies either used alone or in combination. For first recurrence, we included 11 interventions in the network meta-analysis (NMA) for overall survival (OS), and eight in the NMA for progression-free survival (PFS). Lomustine (LOM; also known as CCNU) was the most common comparator and was used as the reference treatment. No studies in the NMA evaluated surgery, re-irradiation, PCV (procarbazine, lomustine, vincristine), TMZ re-challenge or best supportive care. We could not perform NMA for second or later recurrence due to insufficient data. Quality-of-life data were sparse. First recurrence (NMA findings) Median OS across included studies in the NMA ranged from 5.5 to 12.6 months and median progression-free survival (PFS) ranged from 1.5 months to 4.2 months. We found no high-certainty evidence that any treatments tested were better than lomustine. These treatments included the following. Bevacizumab plus lomustine: Evidence suggested probably little or no difference in OS between bevacizumab (BEV) combined with lomustine (LOM) and LOM monotherapy (hazard ratio (HR) 0.91, 0.75 to 1.10; moderate-certainty evidence), although BEV + LOM may improve PFS (HR 0.57, 95% confidence interval (CI) 0.44 to 0.74; low-certainty evidence). Bevacizumab monotherapy: Low-certainty evidence suggested there may be little or no difference in OS (HR 1.22, 95% CI 0.84 to 1.76) and PFS (HR 0.90, 95% CI 0.58 to 1.38; low-certainty evidence) between BEV and LOM monotherapies; more evidence on BEV is needed. Regorafenib (REG): REG may improve OS compared with LOM (HR 0.50, 95% CI 0.33 to 0.76; low-certainty evidence). Evidence on PFS was very low certainty and more evidence on REG is needed. Temozolomide (TMZ) plus Depatux-M (ABT414): For OS, low-certainty evidence suggested that TMZ plus ABT414 may be more effective than LOM (HR 0.66, 95% CI 0.47 to 0.92) and may be more effective than BEV (HR 0.54, 95% CI 0.33 to 0.89; low-certainty evidence). This may be due to the TMZ component only and more evidence is needed. Fotemustine (FOM): FOM and LOM may have similar effects on OS (HR 0.89, 95% CI 0.51 to 1.57, low-certainty evidence). Bevacizumab and irinotecan (IRI): Evidence on BEV + irinotecan (IRI) versus LOM for both OS and PFS is very uncertain and there is probably little or no difference between BEV + IRI versus BEV monotherapy (OS: HR 0.95, 95% CI 0.70 to 1.30; moderate-certainty evidence). When treatments were ranked for OS, FOM ranked first, BEV + LOM second, LOM third, BEV + IRI fourth, and BEV fifth. Ranking does not take into account the certainty of the evidence, which also suggests there may be little or no difference between FOM and LOM. Other treatments Three studies evaluated re-operation versus no re-operation, with or without re-irradiation and chemotherapy, and these suggested possible survival advantages with re-operation within the context of being able to select suitable candidates for re-operation. A cannabinoid treatment in the early stages of evaluation, in combination with TMZ, merits further evaluation. Second or later recurrence Limited evidence from three heterogeneous studies suggested that radiotherapy with or without BEV may have a beneficial effect on survival but more evidence is needed. Evidence was insufficient to draw conclusions about the best radiotherapy dosage. Other evidence suggested that there may be little difference in survival with tumour-treating fields compared with physician's best choice of treatment. We found no reliable evidence on best supportive care. Severe adverse events (SAEs) The BEV+LOM combination was associated with significantly greater risk of SAEs than LOM monotherapy (RR 2.51, 95% CI 1.72 to 3.66, high-certainty evidence), and ranked joint worst with cediranib + LOM (RR 2.51, 95% CI 1.29 to 4.90; high-certainty evidence). LOM ranked best and REG ranked second best. Adding novel treatments to BEV was generally associated with a higher risk of severe adverse events compared with BEV alone. AUTHORS' CONCLUSIONS For treatment of first recurrence of GBM, among people previously treated with surgery and standard chemoradiotherapy, the combination treatments evaluated did not improve overall survival compared with LOM monotherapy and were often associated with a higher risk of severe adverse events. Limited evidence suggested that re-operation with or without re-irradiation and chemotherapy may be suitable for selected candidates. Evidence on second recurrence is sparse. Re-irradiation with or without bevacizumab may be of value in selected individuals, but more evidence is needed.
Collapse
Affiliation(s)
- Catherine McBain
- Clinical Oncology, The Christie NHS FT, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester, UK
| | | | | | - Ashleigh Kernohan
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Tomos Robinson
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Sarah Jefferies
- Department of Oncology, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
10
|
Mitoxantrone-Loaded PLGA Nanoparticles for Increased Sensitivity of Glioblastoma Cancer Cell to TRAIL-Induced Apoptosis. J Pharm Innov 2021. [DOI: 10.1007/s12247-021-09551-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
11
|
Anton S, Margold M, Kowalski T, Miller D, Schmieder K, Schlegel U, Seidel S. Complications of intracerebroventricular chemotherapy via subgaleal reservoir in primary central nervous system lymphoma: A single-institution experience on 1247 installations in 94 consecutive patients. Hematol Oncol 2021; 39:176-184. [PMID: 33316084 DOI: 10.1002/hon.2833] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 11/09/2020] [Accepted: 12/09/2020] [Indexed: 11/09/2022]
Abstract
The implantation of a subgaleal reservoir intracerebroventricular (ICV port) in order to apply ICV chemotherapy in patients with leptomeningeal cancer may be complicated by misplacement of the device, pericatheter leucencephalopathy, hemorrhage and iatrogenic ventriculitis/meningitis. Here we analyzed the occurrence of such complications in patients with primary central nervous system lymphoma (PCNSL) treated with systemic and ICV methotrexate- and cytarabine-based chemotherapy. We retrospectively reviewed the medical records of 94 consecutive patients (1247 installations), who had received an ICV port for intraventricular chemotherapy for treatment of histologically confirmed PCNSL at our institution between September 2005 and October 2018. Infectious and noninfectious complications were systematically recorded including clinical, laboratory, and imaging data. In 9/94 patients (9.6%), a misplacement of the ICV port seen on the postoperative computed tomography scan was corrected immediately and chemotherapy was then continued as planned. In 5/94 patients (5.3%), symptomatic noninfectious complications were observed (four patients with symptomatic pericatheter leucencephalopathy and one patient with surgical scar dehiscence with CSF leak). In 8/94 patients (8.5%), asymptomatic white matter lesions around the catheter were visible on cerebral magnetic resonance imaging after completion of therapy. The rate of infectious complications was 6/94 patients (6.4%). No complication was lethal or required intensive care monitoring. This retrospective study shows that complications of ICV treatment have to be expected in a fraction of patients, however, in this series these complications were manageable and did not result in long-term deficits.
Collapse
Affiliation(s)
- Seena Anton
- Department of Neurology, Knappschaftskrankenhaus University of Bochum, Bochum, Germany
| | - Michelle Margold
- Department of Neurology, Knappschaftskrankenhaus University of Bochum, Bochum, Germany
| | - Thomas Kowalski
- Department of Neurology, Knappschaftskrankenhaus University of Bochum, Bochum, Germany
| | - Dorothea Miller
- Department of Neurosurgery, Knappschaftskrankenhaus University of Bochum, Bochum, Germany
| | - Kirsten Schmieder
- Department of Neurosurgery, Knappschaftskrankenhaus University of Bochum, Bochum, Germany
| | - Uwe Schlegel
- Department of Neurology, Knappschaftskrankenhaus University of Bochum, Bochum, Germany
| | - Sabine Seidel
- Department of Neurology, Knappschaftskrankenhaus University of Bochum, Bochum, Germany
| |
Collapse
|
12
|
Zhang X, Zhang W, Mao XG, Cao WD, Zhen HN, Hu SJ. Malignant Intracranial High Grade Glioma and Current Treatment Strategy. Curr Cancer Drug Targets 2020; 19:101-108. [PMID: 29848277 DOI: 10.2174/1568009618666180530090922] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 12/06/2017] [Accepted: 12/19/2017] [Indexed: 12/17/2022]
Abstract
Malignant high-grade glioma (HGG) is the most common and extremely fatal type of primary intracranial tumor. These tumors recurred within 2 to 3 cm of the primary region of tumor resection in the majority of cases. Furthermore, the blood-brain barrier significantly limited the access of many systemically administered chemotherapeutics to the tumor, pointing towards a stringent need for new therapeutic patterns. Therefore, targeting therapy using local drug delivery for HGG becomes a priority for the development of novel therapeutic strategies. The main objectives to the effective use of chemotherapy for HGG include the drug delivery to the tumor region and the infusion of chemotherapeutic agents into the vascular supply of a tumor directly, which could improve the pharmacokinetic profile by enhancing drug delivery to the neoplasm tissue. Herein, we reviewed clinical and molecular features, different methods of chemotherapy application in HGGs, especially the existing and promising targeting therapies using local drug delivery for HGG which could effectively inhibit tumor invasion, proliferation and recurrence of HGG to combat the deadly disease. Undoubtedly, novel chemical medicines targeting these HGG may represent one of the most important directions in the Neuro-oncology.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Wei Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Xing-Gang Mao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Wei-Dong Cao
- Department of Neurosurgery, Navy General Hospital, PLA, Beijing, 100048, China
| | - Hai-Ning Zhen
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Shi-Jie Hu
- Department of Neuro-oncology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
13
|
Beccaria K, Canney M, Bouchoux G, Desseaux C, Grill J, Heimberger AB, Carpentier A. Ultrasound-induced blood-brain barrier disruption for the treatment of gliomas and other primary CNS tumors. Cancer Lett 2020; 479:13-22. [PMID: 32112904 DOI: 10.1016/j.canlet.2020.02.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/05/2020] [Accepted: 02/11/2020] [Indexed: 01/08/2023]
Abstract
The treatment of primary brain tumors, especially malignant gliomas, remains challenging. The failure of most treatments for this disease is partially explained by the blood-brain barrier (BBB), which prevents circulating molecules from entering the brain parenchyma. Ultrasound-induced BBB disruption (US-BBBD) has recently emerged as a promising strategy to improve the delivery of therapeutic agents to brain tumors. A large body of preclinical studies has demonstrated that the association of low-intensity pulsed ultrasound with intravenous microbubbles can transiently open the BBB in a localized manner. The safety of this technique has been assessed in numerous preclinical studies in both small and large animal models. A large panel of therapeutic agents have been delivered to the brain in preclinical models, demonstrating both tumor control and increased survival. This technique has recently entered clinical trials with encouraging preliminary data. In this review, we describe the mechanisms and histological effects of US-BBBD and summarize the preclinical studies published to date. We furthermore provide an overview of the current clinical development and future potential of this promising technology.
Collapse
Affiliation(s)
- Kévin Beccaria
- Department of Pediatric Neurosurgery, Necker Enfants Malades Hospital, APHP, Paris 5 University, Paris, France.
| | - Michael Canney
- CarThera, Institut Du Cerveau et de La Moelle épinière (ICM), Paris, F-75013, France
| | - Guillaume Bouchoux
- CarThera, Institut Du Cerveau et de La Moelle épinière (ICM), Paris, F-75013, France
| | - Carole Desseaux
- CarThera, Institut Du Cerveau et de La Moelle épinière (ICM), Paris, F-75013, France
| | - Jacques Grill
- Department of Pediatric Oncology, Gustave-Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France; UMR8203 "Vectorologie et Thérapeutiques Anticancéreuses," CNRS, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Alexandre Carpentier
- Department of Neurosurgery, Sorbonne Université, UPMC Univ Paris 06, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires La Pitié-Salpêtrière, Paris, France
| |
Collapse
|
14
|
El Demerdash N, Kedda J, Ram N, Brem H, Tyler B. Novel therapeutics for brain tumors: current practice and future prospects. Expert Opin Drug Deliv 2020; 17:9-21. [DOI: 10.1080/17425247.2019.1676227] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Nagat El Demerdash
- Department of Neurosurgery, Hunterian Neurosurgical Research Laboratory, Johns Hopkins University, Baltimore, MD, USA
| | - Jayanidhi Kedda
- Department of Neurosurgery, Hunterian Neurosurgical Research Laboratory, Johns Hopkins University, Baltimore, MD, USA
| | - Nivi Ram
- Department of Neurosurgery, Hunterian Neurosurgical Research Laboratory, Johns Hopkins University, Baltimore, MD, USA
| | - Henry Brem
- Department of Neurosurgery, Hunterian Neurosurgical Research Laboratory, Johns Hopkins University, Baltimore, MD, USA
- Departments of Biomedical Engineering, Oncology, and Ophthalmology, Johns Hopkins University, Baltimore, MD, USA
| | - Betty Tyler
- Department of Neurosurgery, Hunterian Neurosurgical Research Laboratory, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
15
|
Zhao YH, Wang ZF, Pan ZY, Péus D, Delgado-Fernandez J, Pallud J, Li ZQ. A Meta-Analysis of Survival Outcomes Following Reoperation in Recurrent Glioblastoma: Time to Consider the Timing of Reoperation. Front Neurol 2019; 10:286. [PMID: 30984099 PMCID: PMC6448034 DOI: 10.3389/fneur.2019.00286] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/05/2019] [Indexed: 12/16/2022] Open
Abstract
Background: Glioblastoma multiforme (GBM) inevitably recurs, but no standard regimen has been established for recurrent patients. Reoperation at recurrence alleviates mass effects, and the survival benefit has been reported in many studies. However, in most studies, the effect of reoperation timing on survival benefit was ignored. The aim of this meta-analysis was to investigate whether reoperation provided similar survival benefits in recurrent GBM patients when it was analyzed as a fixed or time-dependent covariate. Methods: A systematic literature search of PubMed, EMBASE, and Cochrane databases was performed to identify original articles that evaluated the associations between reoperation and prognosis in recurrent GBM patients. Results: Twenty-one articles involving 8,630 patients were included. When reoperation was considered as a fixed covariate, it was associated with better overall survival (OS) and post-progression survival (PPS) (OS: HR = 0.66, 95% CI 0.61-0.71, p < 0.001, I2 = 0%; PPS: HR = 0.70, 95% CI 0.57–0.88, p < 0.01, I2 = 70.2%). However, such a survival benefit was not observed when reoperation was considered as a time-dependent covariate (OS: HR = 2.19, 95% CI 1.47–3.27, p < 0.001; PPS: HR = 0.95, 95% CI 0.82–1.10, p = 0.51, I2 = 0%). The estimate bias caused by ignoring the time-dependent nature of reoperation was further demonstrated by the re-analysis of survival data in three included studies. Conclusions: The timing of reoperation may have an impact on the survival outcome in recurrent GBM patients, and survival benefits of reoperation in recurrent GBM may be overestimated when analyzed as fixed covariates. Proper analysis methodology should be used in future work to confirm the clinical benefits of reoperation.
Collapse
Affiliation(s)
- Yu-Hang Zhao
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Ze-Fen Wang
- Department of Physiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Zhi-Yong Pan
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Dominik Péus
- Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| | | | - Johan Pallud
- Department of Neurosurgery, Sainte-Anne Hospital, Paris, France.,Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - Zhi-Qiang Li
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
16
|
Multicenter, single arm, phase II trial on the efficacy of ortataxel in recurrent glioblastoma. J Neurooncol 2019; 142:455-462. [PMID: 30726533 DOI: 10.1007/s11060-019-03116-z] [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: 11/08/2018] [Accepted: 01/31/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND PURPOSE Glioblastoma (GBM) is the most aggressive and frequent subtype of all malignant gliomas. At the time of recurrence, therapeutic options are lacking. Ortataxel, a second-generation taxane was reported to be effective in pre-clinical and phase I clinical studies. The aim of this study was to evaluate a potential therapeutic activity of ortataxel in patients with GBM recurring after surgery and first line treatment. METHODS In this phase II study, according to a two stage design, adult patients with histologically confirmed GBM in recurrence after surgery or biopsy, standard radiotherapy and chemotherapy with temozolomide were considered eligible. Patients included were treated with ortataxel 75 mg/m2 i.v. every 3 weeks until disease progression. The primary objective of the study was to evaluate the activity of ortataxel in terms of progression free survival (PFS) at 6 months after the enrollment. PFS, overall survival at 9 months after the enrollment, objective response rate, compliance and safety were evaluated as secondary endpoints. RESULTS Between Nov 26, 2013 and Dec 12, 2015, 40 patients were recruited across six centres. The number of patients alive and free from progression at 6 months after the enrollment, observed in the first stage was four (11.4%), out of 35 patients included in the analysis, below the minimum number of events (7 out of 33) required to continue the study with the second stage The most important toxicities were neutropenia and hepatotoxicity that occurred in 13.2% of patients and leukopenia that occurred in 15.8% of patients. CONCLUSION Overall ortataxel treatment fail to demonstrate a significant activity in recurrent GBM patients. However in a limited number of patients the drug produced a benefit that lasted for a long time. TRIAL REGISTRATION This study is registered with ClinicalTrials.gov, number NCT01989884.
Collapse
|
17
|
Yu F, Asghar S, Zhang M, Zhang J, Ping Q, Xiao Y. Local strategies and delivery systems for the treatment of malignant gliomas. J Drug Target 2018; 27:367-378. [PMID: 30101621 DOI: 10.1080/1061186x.2018.1509982] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Glioma is one of the most common type of malignant tumours with high morbidity and mortality rates. Due to the particular features of the brain, such as blood-brain barrier or blood-tumour barrier, therapeutic agents are ineffective by systemic administration. The tumour inevitably recurs and devitalises patients. Herein, an overview of the localised gliomas treatment strategies is provided, including direct intratumoural/intracerebral injection, convection-enhanced delivery, and the implant of biodegradable polymer systems. The advantages and disadvantages of each therapy are discussed. Subsequently, we have reviewed the recent developments of therapeutic delivery systems aimed at transporting sufficient amounts of antineoplastic drugs into the brain tumour sites while minimising the potential side effects. To treat gliomas, localised and controlled delivery of drugs at their desired site of action is preferred as it reduces toxicity and increases treatment efficiency. Simultaneously, various drug delivery systems (DDS) have been used to enhance drug delivery to the brain. Use of non-conventional DDS for localised therapy has greatly expanded the spectrum of drugs available for the treatment of malignant tumours. Use smart DDS via localised delivery strategies, in combination with radiotherapy and multiple drug loading would serve as a promising approach to treat gliomas.
Collapse
Affiliation(s)
- Feng Yu
- a Department of Pharmaceutics, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Sajid Asghar
- b Faculty of Pharmaceutical Sciences , Government College University Faisalabad , Faisalabad , Pakistan
| | - Mei Zhang
- a Department of Pharmaceutics, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Jingwei Zhang
- a Department of Pharmaceutics, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Qineng Ping
- a Department of Pharmaceutics, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Yanyu Xiao
- a Department of Pharmaceutics, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , People's Republic of China
| |
Collapse
|
18
|
Abstract
The role of reoperation for glioblastoma multiforme (GBM) recurrence is currently unknown. However, multiple studies have indicated that survival and quality of life are improved with a repeat operation at the time of disease recurrence. Prognosis is likely interdependent on several factors, including age, functional status, initial resection status, disease location, and surgical efficacy. However, there are significant data indicating no survival benefit for reoperation. This comprehensive literature review considering the controversial question of whether to operate for progressive or recurrent GBM seeks to evaluate the current available evidence and report on its conclusions.
Collapse
|
19
|
Yu KKH, Taylor JT, Pathmanaban ON, Youshani AS, Beyit D, Dutko-Gwozdz J, Benson R, Griffiths G, Peers I, Cueppens P, Telfer BA, Williams KJ, McBain C, Kamaly-Asl ID, Bigger BW. High content screening of patient-derived cell lines highlights the potential of non-standard chemotherapeutic agents for the treatment of glioblastoma. PLoS One 2018; 13:e0193694. [PMID: 29499065 PMCID: PMC5834163 DOI: 10.1371/journal.pone.0193694] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 02/19/2018] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common primary brain malignancy in adults, yet survival outcomes remain poor. First line treatment is well established, however disease invariably recurs and improving prognosis is challenging. With the aim of personalizing therapy at recurrence, we have established a high content screening (HCS) platform to analyze the sensitivity profile of seven patient-derived cancer stem cell lines to 83 FDA-approved chemotherapy drugs, with and without irradiation. METHODS Seven cancer stem cell lines were derived from patients with GBM and, along with the established cell line U87-MG, each patient-derived line was cultured in tandem in serum-free conditions as adherent monolayers and three-dimensional neurospheres. Chemotherapeutics were screened at multiple concentrations and cells double-stained to observe their effect on both cell death and proliferation. Sensitivity was classified using high-throughput algorithmic image analysis. RESULTS Cell line specific drug responses were observed across the seven patient-derived cell lines. Few agents were seen to have radio-sensitizing effects, yet some drug classes showed a marked difference in efficacy between monolayers and neurospheres. In vivo validation of six drugs suggested that cell death readout in a three-dimensional culture scenario is a more physiologically relevant screening model and could be used effectively to assess the chemosensitivity of patient-derived GBM lines. CONCLUSION The study puts forward a number of non-standard chemotherapeutics that could be useful in the treatment of recurrent GBM, namely mitoxantrone, bortezomib and actinomycin D, whilst demonstrating the potential of HCS to be used for personalized treatment based on the chemosensitivity profile of patient tumor cells.
Collapse
Affiliation(s)
- Kenny Kwok-Hei Yu
- Brain Tumour Research Group, Stem Cell and Neurotherapies Laboratory, Division of Cell Matrix Biology & Regenerative Medicine, University of Manchester, Manchester, United Kingdom
| | - Jessica T. Taylor
- Brain Tumour Research Group, Stem Cell and Neurotherapies Laboratory, Division of Cell Matrix Biology & Regenerative Medicine, University of Manchester, Manchester, United Kingdom
| | - Omar N. Pathmanaban
- Manchester Centre for Clinical Neurosciences, Salford Royal Hospital, Manchester Academic Health Sciences Centre, Salford, United Kingdom
| | - Amir Saam Youshani
- Brain Tumour Research Group, Stem Cell and Neurotherapies Laboratory, Division of Cell Matrix Biology & Regenerative Medicine, University of Manchester, Manchester, United Kingdom
| | - Deniz Beyit
- Imagen Therapeutics, Manchester, United Kingdom
| | | | | | | | - Ian Peers
- Inferstats Consulting, Alderley Park, Biohub, Cheshire, United Kingdom
| | - Peter Cueppens
- Inferstats Consulting, Alderley Park, Biohub, Cheshire, United Kingdom
| | - Brian A. Telfer
- Division of Pharmacy & Optometry, School of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kaye J. Williams
- Division of Pharmacy & Optometry, School of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Catherine McBain
- Department of Clinical Oncology, The Christie NHS FT, Manchester, United Kingdom
| | - Ian D. Kamaly-Asl
- Children’s Brain Tumour Research Network (CBTRN), Royal Manchester Children’s Hospital, Manchester, United Kingdom
- Department of Neurosurgery, Royal Manchester Children’s Hospital, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Brian W. Bigger
- Brain Tumour Research Group, Stem Cell and Neurotherapies Laboratory, Division of Cell Matrix Biology & Regenerative Medicine, University of Manchester, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
20
|
Singh R, Bellat V, Wang M, Schweitzer ME, Wu YL, Tung CH, Souweidane MM, Law B. Volume of distribution and clearance of peptide-based nanofiber after convection-enhanced delivery. J Neurosurg 2017; 129:10-18. [PMID: 28885119 DOI: 10.3171/2017.2.jns162273] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Drug clearance may be a limiting factor in the clinical application of convection-enhanced delivery (CED). Peptide-based nanofibers (NFPs) have a high aspect ratio, and NFPs loaded with drugs could potentially maintain effective drug concentrations for an extended period sufficient for cancer therapy. The objective of this study was to assess the volume of distribution (Vd) and clearance of variable lengths of NFPs when administered using CED. METHODS NFPs composed of multiple methoxypolyethylene glycol (mPEG)-conjugated constructs (mPEG2000-KLDLKLDLKLDL-K( FITC)-CONH2, for which FITC is fluorescein isothiocyanate) were assembled in an aqueous buffer. The NFPs were approximately 5 nm in width and were formulated into different lengths: 100 nm (NFP-100), 400 nm (NFP-400), and 1000 nm (NFP-1000). The NFP surface was covalently conjugated with multiple Cy5.5 fluorophores as the optical reporters to track the post-CED distribution. Forty-two 6- to 8-week-old Ntv-a;p53fl/fl mice underwent CED to the striatum. Animals were killed immediately, 24 hours or 72 hours after CED. The brains were extracted and sectioned for assessing NFP Vd to volume of infusion (Vi) ratio, and clearance using fluorescence microscopy. RESULTS CED of NFPs was well tolerated by all the animals. The average Vd/Vi ratios for NFP-100, NFP-400, NFP-1000, and unconjugated positive control (free Cy5.5) were 1.87, 2.47, 1.07, and 3.0, respectively, which were statistically different (p = 0.003). The percentages remaining of the original infusion volume at 24 hours for NFP-100, -400, and -1000 were 40%, 90%, and 74%, respectively. The percentages remaining at 72 hours for NFP-100, -400, and -1000 were 15%, 30%, and 46%, respectively. Unconjugated Cy5.5 was not detected at 24 or 72 hours after CED. CONCLUSIONS CED of NFPs is feasible with Vd/Vi ratios and clearance rates comparable to other nanocarriers. Of the 3 NFPs, NFP-400 appears to provide the best distribution and slowest clearance after 24 hours. NFP provides a dynamic theranostic platform, with the potential to deliver clinically efficacious drug payload to brain tumor after CED.
Collapse
Affiliation(s)
| | - Vanessa Bellat
- 2Department of Radiology, Molecular Imaging Innovations Institute, and
| | | | | | | | - Ching-Hsuan Tung
- 2Department of Radiology, Molecular Imaging Innovations Institute, and
| | - Mark M Souweidane
- 1Department of Neurological Surgery.,3Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - Benedict Law
- 2Department of Radiology, Molecular Imaging Innovations Institute, and
| |
Collapse
|
21
|
Cohen-Pfeffer JL, Gururangan S, Lester T, Lim DA, Shaywitz AJ, Westphal M, Slavc I. Intracerebroventricular Delivery as a Safe, Long-Term Route of Drug Administration. Pediatr Neurol 2017; 67:23-35. [PMID: 28089765 DOI: 10.1016/j.pediatrneurol.2016.10.022] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 09/07/2016] [Accepted: 10/30/2016] [Indexed: 01/19/2023]
Abstract
Intrathecal delivery methods have been used for many decades to treat a broad range of central nervous system disorders. A literature review demonstrated that intracerebroventricular route is an established and well-tolerated method for prolonged central nervous system drug delivery in pediatric and adult populations. Intracerebroventricular devices were present in patients from one to 7156 days. The number of punctures per device ranged from 2 to 280. Noninfectious complication rates per patient (range, 1.0% to 33.0%) were similar to infectious complication rates (0.0% to 27.0%). Clinician experience and training and the use of strict aseptic techniques have been shown to reduce the frequency of complications.
Collapse
Affiliation(s)
| | | | | | - Daniel A Lim
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | | | - Manfred Westphal
- Department of Neurosurgery, University Clinic Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Irene Slavc
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
22
|
Masoumi S, Harisankar A, Gracias A, Bachinger F, Fufa T, Chandrasekar G, Gaunitz F, Walfridsson J, Kitambi SS. Understanding cytoskeleton regulators in glioblastoma multiforme for therapy design. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:2881-2897. [PMID: 27672311 PMCID: PMC5026218 DOI: 10.2147/dddt.s106196] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The cellular cytoskeleton forms the primary basis through which a cell governs the changes in size, shape, migration, proliferation, and forms the primary means through which the cells respond to their environment. Indeed, cell and tissue morphologies are used routinely not only to grade tumors but also in various high-content screening methods with an aim to identify new small molecules with therapeutic potential. This study examines the expression of various cytoskeleton regulators in glioblastoma multiforme (GBM). GBM is a very aggressive disease with a low life expectancy even after chemo- and radiotherapy. Cancer cells of GBM are notorious for their invasiveness, ability to develop resistance to chemo- and radiotherapy, and to form secondary site tumors. This study aims to gain insight into cytoskeleton regulators in GBM cells and to understand the effect of various oncology drugs, including temozolomide, on cytoskeleton regulators. We compare the expression of various cytoskeleton regulators in GBM-derived tumor and normal tissue, CD133-postive and -negative cells from GBM and neural cells, and GBM stem-like and differentiated cells. In addition, the correlation between the expression of cytoskeleton regulators with the clinical outcome was examined to identify genes associated with longer patient survival. This was followed by a small molecule screening with US Food and Drug Administration (FDA)-approved oncology drugs, and its effect on cellular cytoskeleton was compared to treatment with temozolomide. This study identifies various groups of cytoskeletal regulators that have an important effect on patient survival and tumor development. Importantly, this work highlights the advantage of using cytoskeleton regulators as biomarkers for assessing prognosis and treatment design for GBM.
Collapse
Affiliation(s)
| | - Aditya Harisankar
- Center for Hematology and Regenerative Medicine, Department of Medicine
| | - Aileen Gracias
- Department of Neuroscience, Karolinska Institutet, Solna, Sweden
| | | | - Temesgen Fufa
- Department of Microbiology Tumor and Cell Biology; Department of Neurosurgery, University Hospital, Leipzig, Germany
| | | | - Frank Gaunitz
- Department of Neurosurgery, University Hospital, Leipzig, Germany
| | | | | |
Collapse
|
23
|
Novel chemical library screen identifies naturally occurring plant products that specifically disrupt glioblastoma-endothelial cell interactions. Oncotarget 2016; 6:18282-92. [PMID: 26286961 PMCID: PMC4621891 DOI: 10.18632/oncotarget.4957] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 07/09/2015] [Indexed: 12/23/2022] Open
Abstract
Tumor growth is not solely a consequence of autonomous tumor cell properties. Rather, tumor cells act upon and are acted upon by their microenvironment. It is tumor tissue biology that ultimately determines tumor growth. Thus, we developed a compound library screen for agents that could block essential tumor-promoting effects of the glioblastoma (GBM) perivascular stem cell niche (PVN). We modeled the PVN with three-dimensional primary cultures of human brain microvascular endothelial cells in Matrigel. We previously demonstrated stimulated growth of GBM cells in this PVN model and used this to assay PVN function. We screened the Microsource Spectrum Collection library for drugs that specifically blocked PVN function, without any direct effect on GBM cells themselves. Three candidate PVN-disrupting agents, Iridin, Tigogenin and Triacetylresveratrol (TAR), were identified and evaluated in secondary in vitro screens against a panel of primary GBM isolates as well as in two different in vivo intracranial models. Iridin and TAR significantly inhibited intracranial tumor growth and prolonged survival in these mouse models. Together these data identify Iridin and TAR as drugs with novel GBM tissue disrupting effects and validate the importance of preclinical screens designed to address tumor tissue function rather than the mechanisms of autonomous tumor cell growth.
Collapse
|
24
|
Second surgery for recurrent glioblastoma: A concise overview of the current literature. Clin Neurol Neurosurg 2016; 142:60-64. [PMID: 26811867 DOI: 10.1016/j.clineuro.2016.01.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 12/29/2015] [Accepted: 01/05/2016] [Indexed: 12/17/2022]
Abstract
Optimal treatment for recurrent glioblastoma continues to evolve. Currently, however, there is no consensus in the literature on the role of reoperation in the management of these patients, as several studies provide evidence for a longer overall survival in selected patients with recurrent glioblastoma who underwent second surgery and other studies report a limited impact of second surgery in the clinical course. In this paper, a review of the current literature was performed to analyze the role of reoperation in patients with recurrent glioblastoma and to report the overall survival from diagnosis, progression-free survival and quality of life. Using PubMed and Ovid Medline databases, we performed a review of the literature of the last seven years, finding a total of 28 studies and 2279 patients who underwent second surgery, that were included in the final analysis. The median overall survival from diagnosis and the median survival from second surgery were 18.5 months and 9.7 months, respectively. Extent of resection at reoperation improves overall survival, even in patients with subtotal resection at initial operation. Preoperative performance status and age are important predictors of a longer survival, reason why younger patients with a good preoperative performance status could benefit from reoperation.
Collapse
|
25
|
Delivery of local therapeutics to the brain: working toward advancing treatment for malignant gliomas. Ther Deliv 2015; 6:353-69. [PMID: 25853310 DOI: 10.4155/tde.14.114] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Malignant gliomas, including glioblastoma and anaplastic astrocytomas, are characterized by their propensity to invade surrounding brain parenchyma, making curative resection difficult. These tumors typically recur within two centimeters of the resection cavity even after gross total removal. As a result, there has been an emphasis on developing therapeutics aimed at achieving local disease control. In this review, we will summarize the current developments in the delivery of local therapeutics, namely direct injection, convection-enhanced delivery and implantation of drug-loaded polymers, as well as the application of these therapeutics in future methods including microchip drug delivery and local gene therapy.
Collapse
|
26
|
Rhun EL, Taillibert S, Chamberlain MC. The future of high-grade glioma: Where we are and where are we going. Surg Neurol Int 2015; 6:S9-S44. [PMID: 25722939 PMCID: PMC4338495 DOI: 10.4103/2152-7806.151331] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 10/15/2014] [Indexed: 01/12/2023] Open
Abstract
High-grade glioma (HGG) are optimally treated with maximum safe surgery, followed by radiotherapy (RT) and/or systemic chemotherapy (CT). Recently, the treatment of newly diagnosed anaplastic glioma (AG) has changed, particularly in patients with 1p19q codeleted tumors. Results of trials currenlty ongoing are likely to determine the best standard of care for patients with noncodeleted AG tumors. Trials in AG illustrate the importance of molecular characterization, which are germane to both prognosis and treatment. In contrast, efforts to improve the current standard of care of newly diagnosed glioblastoma (GB) with, for example, the addition of bevacizumab (BEV), have been largely disappointing and furthermore molecular characterization has not changed therapy except in elderly patients. Novel approaches, such as vaccine-based immunotherapy, for newly diagnosed GB are currently being pursued in multiple clinical trials. Recurrent disease, an event inevitable in nearly all patients with HGG, continues to be a challenge. Both recurrent GB and AG are managed in similar manner and when feasible re-resection is often suggested notwithstanding limited data to suggest benefit from repeat surgery. Occassional patients may be candidates for re-irradiation but again there is a paucity of data to commend this therapy and only a minority of selected patients are eligible for this approach. Consequently systemic therapy continues to be the most often utilized treatment in recurrent HGG. Choice of therapy, however, varies and revolves around re-challenge with temozolomide (TMZ), use of a nitrosourea (most often lomustine; CCNU) or BEV, the most frequently used angiogenic inhibitor. Nevertheless, no clear standard recommendation regarding the prefered agent or combination of agents is avaliable. Prognosis after progression of a HGG remains poor, with an unmet need to improve therapy.
Collapse
Affiliation(s)
- Emilie Le Rhun
- Department of Neuro-oncology, Roger Salengro Hospital, University Hospital, Lille, and Neurology, Department of Medical Oncology, Oscar Lambret Center, Lille, France, Inserm U-1192, Laboratoire de Protéomique, Réponse Inflammatoire, Spectrométrie de Masse (PRISM), Lille 1 University, Villeneuve D’Ascq, France
| | - Sophie Taillibert
- Neurology, Mazarin and Radiation Oncology, Pitié Salpétrière Hospital, University Pierre et Marie Curie, Paris VI, Paris, France
| | - Marc C. Chamberlain
- Department of Neurology and Neurological Surgery, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
27
|
Toxicity evaluation of prolonged convection-enhanced delivery of small-molecule kinase inhibitors in naïve rat brainstem. Childs Nerv Syst 2015; 31:221-6. [PMID: 25269544 DOI: 10.1007/s00381-014-2568-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 09/23/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE Convection-enhanced delivery (CED), a local drug delivery technique, is typically performed as a single session and drug concentrations therefore decline quickly post CED. Prolonged CED (pCED) overcomes this problem by performing a long-term infusion to maintain effective drug concentrations for an extended period. The purpose of the current study was to assess the toxicity of using pCED to deliver single and multi-drug therapy in naïve rat brainstem. METHODS Sixteen rats underwent pCED of three small-molecule kinase inhibitors in the pons. Single and multi-drug combinations were delivered continuously for 7 days using ALZET mini-osmotic pumps (model 2001, rate of 1 μl/h). Rats were monitored daily for neurological signs of toxicity. Rats were sacrificed 10 days post completion of infusion, and appropriate tissue sections were analyzed for histological signs of toxicity. RESULTS Two rats exhibited signs of neurological deficits, which corresponded with diffuse inflammation, necrosis, and parenchymal damage on histological analysis. The remaining rats showed no neurological or histological signs of toxicity. CONCLUSION The neurological deficits in the two rats were likely due to injury from physical force, such as cannula movement post insertion and subsequent encephalitis. The remaining rats showed no toxicity and therefore brainstem targeting using pCED to infuse single and multi-drug therapy was well tolerated in these rats.
Collapse
|
28
|
|
29
|
Weller M, Cloughesy T, Perry JR, Wick W. Standards of care for treatment of recurrent glioblastoma--are we there yet? Neuro Oncol 2013; 15:4-27. [PMID: 23136223 PMCID: PMC3534423 DOI: 10.1093/neuonc/nos273] [Citation(s) in RCA: 530] [Impact Index Per Article: 48.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 09/17/2012] [Indexed: 12/21/2022] Open
Abstract
Newly diagnosed glioblastoma is now commonly treated with surgery, if feasible, or biopsy, followed by radiation plus concomitant and adjuvant temozolomide. The treatment of recurrent glioblastoma continues to be a moving target as new therapeutic principles enrich the standards of care for newly diagnosed disease. We reviewed PubMed and American Society of Clinical Oncology abstracts from January 2006 to January 2012 to identify clinical trials investigating the treatment of recurrent or progressive glioblastoma with nitrosoureas, temozolomide, bevacizumab, and/or combinations of these agents. At recurrence, a minority of patients are eligible for second surgery or reirradiation, based on appropriate patient selection. In temozolomide-pretreated patients, progression-free survival rates at 6 months of 20%-30% may be achieved either with nitrosoureas, temozolomide in various dosing regimens, or bevacizumab. Combination regimens among these agents or with other drugs have not produced evidence for superior activity but commonly produce more toxicity. More research is needed to better define patient profiles that predict benefit from the limited therapeutic options available after the current standard of care has failed.
Collapse
Affiliation(s)
- Michael Weller
- Department of Neurology, University Hospital Zurich, Frauenklinikstrasse 26, CH-8091 Zurich, Switzerland.
| | | | | | | |
Collapse
|
30
|
Buonerba C, Di Lorenzo G, Marinelli A, Federico P, Palmieri G, Imbimbo M, Conti P, Peluso G, De Placido S, Sampson JH. A comprehensive outlook on intracerebral therapy of malignant gliomas. Crit Rev Oncol Hematol 2011; 80:54-68. [DOI: 10.1016/j.critrevonc.2010.09.001] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Revised: 08/12/2010] [Accepted: 09/01/2010] [Indexed: 11/15/2022] Open
|
31
|
Xu JF, Fang J, Shen Y, Zhang JM, Liu WG, Shen H. Should we reoperate for recurrent high-grade astrocytoma? J Neurooncol 2011; 105:291-9. [PMID: 21590314 DOI: 10.1007/s11060-011-0585-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Accepted: 04/08/2011] [Indexed: 12/18/2022]
Abstract
Despite optimal treatment of post-operative radiotherapy and chemotherapy for newly diagnosed high-grade astrocytoma, nearly all patients eventually recur. However, the efficacy of reoperation for recurrent astrocytoma is still debatable as to different surgical indications. To investigate the therapeutic effect of reoperation on patients with recurrent high-grade astrocytoma more objectively, a retrospective case-matched study was carried out. The clinical data of 63 cases of recurrent high-grade astrocytoma treated between January 2006 and December 2008 were studied. A total of 21 cases received reoperation immediately after tumor recurrence, while 42 cases without reoperation were matched by gender, age, Karnofsky Performance Scale (KPS) score, histopathology, recurrent interval after the first operation, extent of initial surgery, adjuvant treatment and characteristics of recurrent tumor. The study showed that the median survival time was 7 months in the reoperation group, while in non-reoperation group, it was 4 months. There was significant difference on univariate analysis (P < 0.001). Moreover, the median duration time of progression-free survival (PFS) after tumor recurrence was significantly (P < 0.001) longer in the reoperation group (5 months) than that in the non-reoperation group (2.5 months). The prognostic factors of recurrent high-grade astrocytoma included reoperation, KPS score and tumor location. It was indicated that reoperation could prolong the survival time and improve the quality of survival in patients of recurrent high-grade astrocytoma.
Collapse
Affiliation(s)
- Jin-fang Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou City, 310009, People's Republic of China
| | | | | | | | | | | |
Collapse
|
32
|
Efficacy and safety of intratumoral thermotherapy using magnetic iron-oxide nanoparticles combined with external beam radiotherapy on patients with recurrent glioblastoma multiforme. J Neurooncol 2010; 103:317-24. [PMID: 20845061 PMCID: PMC3097345 DOI: 10.1007/s11060-010-0389-0] [Citation(s) in RCA: 760] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Accepted: 08/26/2010] [Indexed: 01/10/2023]
Abstract
Therapy options at the time of recurrence of glioblastoma multiforme are often limited. We investigated whether treatment with a new intratumoral thermotherapy procedure using magnetic nanoparticles improves survival outcome. In a single-arm study in two centers, 66 patients (59 with recurrent glioblastoma) received neuronavigationally controlled intratumoral instillation of an aqueous dispersion of iron-oxide (magnetite) nanoparticles and subsequent heating of the particles in an alternating magnetic field. Treatment was combined with fractionated stereotactic radiotherapy. A median dose of 30 Gy using a fractionation of 5 × 2 Gy/week was applied. The primary study endpoint was overall survival following diagnosis of first tumor recurrence (OS-2), while the secondary endpoint was overall survival after primary tumor diagnosis (OS-1). Survival times were calculated using the Kaplan–Meier method. Analyses were by intention to treat. The median overall survival from diagnosis of the first tumor recurrence among the 59 patients with recurrent glioblastoma was 13.4 months (95% CI: 10.6–16.2 months). Median OS-1 was 23.2 months while the median time interval between primary diagnosis and first tumor recurrence was 8.0 months. Only tumor volume at study entry was significantly correlated with ensuing survival (P < 0.01). No other variables predicting longer survival could be determined. The side effects of the new therapeutic approach were moderate, and no serious complications were observed. Thermotherapy using magnetic nanoparticles in conjunction with a reduced radiation dose is safe and effective and leads to longer OS-2 compared to conventional therapies in the treatment of recurrent glioblastoma.
Collapse
|
33
|
Radiation Therapy for Prevention of Heterotopic Ossification about the Elbow. Strahlenther Onkol 2009; 185:506-11. [DOI: 10.1007/s00066-009-1968-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Accepted: 01/09/2009] [Indexed: 11/27/2022]
|
34
|
Salmaggi A, Erbetta A, Silvani A, Maderna E, Pollo B. Intracerebral haemorrhage in primary and metastatic brain tumours. Neurol Sci 2008; 29 Suppl 2:S264-5. [DOI: 10.1007/s10072-008-0958-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|