1
|
Kreatsoulas D, Damante M, Cua S, Lonser RR. Adjuvant convection-enhanced delivery for the treatment of brain tumors. J Neurooncol 2024; 166:243-255. [PMID: 38261143 PMCID: PMC10834622 DOI: 10.1007/s11060-023-04552-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024]
Abstract
BACKGROUND Malignant gliomas are a therapeutic challenge and remain nearly uniformly fatal. While new targeted chemotherapeutic agentsagainst malignant glioma have been developed in vitro, these putative therapeutics have not been translated into successful clinical treatments. The lack of clinical effectiveness can be the result of ineffective biologic strategies, heterogeneous tumor targets and/or the result of poortherapeutic distribution to malignant glioma cells using conventional nervous system delivery modalities (intravascular, cerebrospinal fluid and/orpolymer implantation), and/or ineffective biologic strategies. METHODS The authors performed a review of the literature for the terms "convection enhanced delivery", "glioblastoma", and "glioma". Selectclinical trials were summarized based on their various biological mechanisms and technological innovation, focusing on more recently publisheddata when possible. RESULTS We describe the properties, features and landmark clinical trials associated with convection-enhanced delivery for malignant gliomas.We also discuss future trends that will be vital to CED innovation and improvement. CONCLUSION Efficacy of CED for malignant glioma to date has been mixed, but improvements in technology and therapeutic agents arepromising.
Collapse
Affiliation(s)
- Daniel Kreatsoulas
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, The Ohio State University, N1019 Doan Hall, 410 W 10Th Avenue, Columbus, OH, 43210, USA.
| | - Mark Damante
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, The Ohio State University, N1019 Doan Hall, 410 W 10Th Avenue, Columbus, OH, 43210, USA
| | - Santino Cua
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, The Ohio State University, N1019 Doan Hall, 410 W 10Th Avenue, Columbus, OH, 43210, USA
| | - Russell R Lonser
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, The Ohio State University, N1019 Doan Hall, 410 W 10Th Avenue, Columbus, OH, 43210, USA
| |
Collapse
|
2
|
Liu X, Zhao Z, Dai W, Liao K, Sun Q, Chen D, Pan X, Feng L, Ding Y, Wei S. The Development of Immunotherapy for the Treatment of Recurrent Glioblastoma. Cancers (Basel) 2023; 15:4308. [PMID: 37686584 PMCID: PMC10486426 DOI: 10.3390/cancers15174308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 09/10/2023] Open
Abstract
Recurrent glioblastoma (rGBM) is a highly aggressive form of brain cancer that poses a significant challenge for treatment in neuro-oncology, and the survival status of patients after relapse usually means rapid deterioration, thus becoming the leading cause of death among patients. In recent years, immunotherapy has emerged as a promising strategy for the treatment of recurrent glioblastoma by stimulating the body's immune system to recognize and attack cancer cells, which could be used in combination with other treatments such as surgery, radiation, and chemotherapy to improve outcomes for patients with recurrent glioblastoma. This therapy combines several key methods such as the use of monoclonal antibodies, chimeric antigen receptor T cell (CAR-T) therapy, checkpoint inhibitors, oncolytic viral therapy cancer vaccines, and combination strategies. In this review, we mainly document the latest immunotherapies for the treatment of glioblastoma and especially focus on rGBM.
Collapse
Affiliation(s)
- Xudong Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; (X.L.); (Y.D.)
| | - Zihui Zhao
- School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China;
| | - Wufei Dai
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering Research, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China;
| | - Kuo Liao
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China;
| | - Qi Sun
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Q.S.); (L.F.)
| | - Dongjiang Chen
- Division of Neuro-Oncology, USC Keck Brain Tumor Center, University of Southern California Keck School of Medicine, Los Angeles, CA 90089, USA;
| | - Xingxin Pan
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA;
| | - Lishuang Feng
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Q.S.); (L.F.)
| | - Ying Ding
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; (X.L.); (Y.D.)
| | - Shiyou Wei
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
3
|
Oraee-Yazdani S, Akhlaghpasand M, Rostami F, Golmohammadi M, Tavanaei R, Shokri G, Hafizi M, Oraee-Yazdani M, Zali AR, Soleimani M. Case report: Stem cell-based suicide gene therapy mediated by the herpes simplex virus thymidine kinase gene reduces tumor progression in multifocal glioblastoma. Front Neurol 2023; 14:1060180. [PMID: 37034076 PMCID: PMC10075310 DOI: 10.3389/fneur.2023.1060180] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 02/28/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction The prognosis for glioblastoma multiforme (GBM), a malignant brain tumor, is poor despite recent advancements in treatments. Suicide gene therapy is a therapeutic strategy for cancer that requires a gene to encode a prodrug-activating enzyme which is then transduced into a vector, such as mesenchymal stem cells (MSCs). The vector is then injected into the tumor tissue and exerts its antitumor effects. Case presentation A 37-year-old man presented to our department with two evident foci of glioblastoma multiforme at the left frontal and left parietal lobes. The patient received an injection of bone marrow-derived MSCs delivering the herpes simplex virus thymidine kinase (HSV-tk) gene to the frontal focus of the tumor, followed by ganciclovir administration as a prodrug for 14 days. For follow-up, the patient was periodically assessed using magnetic resonance imaging (MRI). The growth and recurrence patterns of the foci were assessed. After the injection on 09 February 2019, the patient's follow-up appointment on 19 December 2019 MRI revealed a recurrence of parietal focus. However, the frontal focus had a slight and unremarkable enhancement. On the last follow-up (18 March 2020), the left frontal focus had no prominent recurrence; however, the size of the left parietal focus increased and extended to the contralateral hemisphere through the corpus callosum. Eventually, the patient passed away on 16 July 2020 (progression-free survival (PFS) = 293 days, overall survival (OS) = 513 days). Conclusion The gliomatous focus (frontal) treated with bone marrow-derived MSCs carrying the HSV-TK gene had a different pattern of growth and recurrence compared with the non-treated one (parietal). Trial registration IRCT20200502047277N2. Registered 10 May 2020-Retrospectively registered, https://eng.irct.ir/trial/48110.
Collapse
Affiliation(s)
- Saeed Oraee-Yazdani
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- *Correspondence: Saeed Oraee-Yazdani
| | - Mohammadhosein Akhlaghpasand
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Rostami
- Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Golmohammadi
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roozbeh Tavanaei
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Maryam Hafizi
- Department of Research and Development, Sodour Ahrar Shargh Company, Tehran, Iran
| | - Maryam Oraee-Yazdani
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali-Reza Zali
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
4
|
Wang EJ, Chen JS, Jain S, Morshed RA, Haddad AF, Gill S, Beniwal AS, Aghi MK. Immunotherapy Resistance in Glioblastoma. Front Genet 2021; 12:750675. [PMID: 34976006 PMCID: PMC8718605 DOI: 10.3389/fgene.2021.750675] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/27/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma is the most common malignant primary brain tumor in adults. Despite treatment consisting of surgical resection followed by radiotherapy and adjuvant chemotherapy, survival remains poor at a rate of 26.5% at 2 years. Recent successes in using immunotherapies to treat a number of solid and hematologic cancers have led to a growing interest in harnessing the immune system to target glioblastoma. Several studies have examined the efficacy of various immunotherapies, including checkpoint inhibitors, vaccines, adoptive transfer of lymphocytes, and oncolytic virotherapy in both pre-clinical and clinical settings. However, these therapies have yielded mixed results at best when applied to glioblastoma. While the initial failures of immunotherapy were thought to reflect the immunoprivileged environment of the brain, more recent studies have revealed immune escape mechanisms created by the tumor itself and adaptive resistance acquired in response to therapy. Several of these resistance mechanisms hijack key signaling pathways within the immune system to create a protumoral microenvironment. In this review, we discuss immunotherapies that have been trialed in glioblastoma, mechanisms of tumor resistance, and strategies to sensitize these tumors to immunotherapies. Insights gained from the studies summarized here may help pave the way for novel therapies to overcome barriers that have thus far limited the success of immunotherapy in glioblastoma.
Collapse
Affiliation(s)
- Elaina J. Wang
- Department of Neurological Surgery, The Warren Alpert School of Medicine, Brown University, Providence, RI, United States
| | - Jia-Shu Chen
- Department of Neurological Surgery, The Warren Alpert School of Medicine, Brown University, Providence, RI, United States
| | - Saket Jain
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Ramin A. Morshed
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Alexander F. Haddad
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Sabraj Gill
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Angad S. Beniwal
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Manish K. Aghi
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
5
|
Enhancement of Radio-Thermo-Sensitivity of 5-Iodo-2-Deoxyuridine-Loaded Polymeric-Coated Magnetic Nanoparticles Triggers Apoptosis in U87MG Human Glioblastoma Cancer Cell Line. Cell Mol Bioeng 2021; 14:365-377. [PMID: 34295445 DOI: 10.1007/s12195-021-00675-y] [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: 12/11/2019] [Accepted: 05/05/2021] [Indexed: 01/22/2023] Open
Abstract
Introduction With an emphasis on the radioresistant nature of glioblastoma cells, the aim of the present study was to evaluate the radio-thermo-sensitizing effects of PCL-PEG-coated Superparamagnetic iron oxide nanoparticles (SPIONs) as a carrier of 5-iodo-2-deoxyuridine (IUdR) in monolayer culture of U87MG human glioma cell line. Methods Following monolayer culture of U87MG cells, nanoparticle uptake was assessed using Prussian blue staining and ICP-OES method. The U87MG cells were treated with an appropriate concentration of free IUdR and PCL-PEG-coated SPIONs (MNPs) loaded with IUdR (IUdR/MNPs) for 24 h, subjected to hyperthermia (water bath and alternating magnetic field (AMF)) at 43 °C, and exposed to X-ray (2 Gy, 6 MV). The combined effects of hyperthermia with or without magnetic nanoparticles on radiosensitivity of the U87MG cells were evaluated using colony formation assay (CFA) and Flowcytometry. Results Prussian blue staining and ICP-OES showed that the nanoparticles were able to enter the cells. The results also indicated that IUdR/MNPs combined with X-ray radiation and hyperthermia significantly decreased the colony formation ability of monolayer cells (1.11, 1.41 fold) and increased the percentage of apoptotic (2.47, 4.1 fold) and necrotic cells (12.28, 29.34 fold), when compared to IUdR combined with X-ray and hyperthermia or IUdR/MNPs + X-ray. MTT results revealed that the presence of IUdR/MNPs significantly increased the toxicity of AMF hyperthermia compared to the water bath method. Conclusions Our study showed that SPIONs/PCL-PEG, as a carrier of IUdR, can enhance the cytotoxic effects of radiotherapy and hyperthermia and act as a radio-thermo-sensitizing agent. Graphic Abstract Supplementary Information The online version contains supplementary material available at 10.1007/s12195-021-00675-y.
Collapse
|
6
|
Intracerebral Administration of Autologous Mesenchymal Stem Cells as HSV-TK Gene Vehicle for Treatment of Glioblastoma Multiform: Safety and Feasibility Assessment. Mol Neurobiol 2021; 58:4425-4436. [PMID: 34021868 DOI: 10.1007/s12035-021-02393-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 04/13/2021] [Indexed: 10/21/2022]
Abstract
Widespread investigation has revealed the promising ability of suicidal genes in the treatment of glioma tumors; nevertheless, promoting their effects relies on the ability to apply suitable vehicles and techniques. In this study, the safety and feasibility of using bone marrow-derived mesenchymal stem cells (MSCs) in combination with prodrug for treatment of patients with primary and secondary glioblastoma multiform (GBM) was assessed. Five GBM patients were recruited. Following gross total resection of the tumor and adjuvant radiotherapy and chemotherapy, intracerebral injection of autologous MSCs transduced with lentivirus containing herpes simplex virus thymidine kinase (HSV-TK) was performed followed by intravenous administration of ganciclovir for 2 weeks. The treatment was well tolerated by all patients. Mild-to-moderate fever, headache, and cerebrospinal fluid leukocytosis were evident in three, two, and one patient, respectively. The progression-free survival (PFS) and overall survival (OS) of patients were 95.79 ± 51.40 and 128.85 ± 48.81 weeks, respectively. The 1-year PFS and OS were 60% and 100%, respectively, among our patients, and two patients had more than 3 years of OS and more than 2 years of PFS. It seems that intracerebral administration of bone marrow MSC containing the HSV-TK gene in combination with intravenous ganciclovir would be safe and feasible in the treatment of patients with GBM.
Collapse
|
7
|
Sheikh S, Ernst D, Keating A. Prodrugs and prodrug-activated systems in gene therapy. Mol Ther 2021; 29:1716-1728. [PMID: 33831557 PMCID: PMC8116605 DOI: 10.1016/j.ymthe.2021.04.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 03/06/2021] [Accepted: 04/02/2021] [Indexed: 12/11/2022] Open
Abstract
The inclusion of genes that control cell fate (so-called suicide, or kill-switch, genes) into gene therapy vectors is based on a compelling rationale for the safe and selective elimination of aberrant transfected cells. Prodrug-activated systems were developed in the 1980s and 1990s and rely on the enzymatic conversion of non-active prodrugs to active metabolites that lead to cell death. Although considerable effort and ingenuity has gone into vector design for gene therapy, less attention has been directed at the efficacy or associated adverse effects of the prodrug systems employed. In this review, we discuss prodrug systems employed in clinical trials and consider their role in the field of gene therapy. We highlight potential drawbacks associated with the use of specific prodrugs, such as systemic toxicity of the activated compound, the paucity of data on biodistribution of prodrugs, bystander effects, and destruction of genetically modified cells, and how these can inform future advances in cell therapies.
Collapse
Affiliation(s)
- Semira Sheikh
- Princess Margaret Cancer Centre, Toronto, ON, Canada; University of Toronto, Toronto, ON, Canada.
| | - Daniel Ernst
- Krembil Research Institute, Toronto, ON, Canada; University of Toronto, Toronto, ON, Canada
| | - Armand Keating
- Princess Margaret Cancer Centre, Toronto, ON, Canada; Krembil Research Institute, Toronto, ON, Canada; Schroeder Arthritis Institute, Toronto, ON, Canada; University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
8
|
Wang JL, Scheitler KM, Wenger NM, Elder JB. Viral therapies for glioblastoma and high-grade gliomas in adults: a systematic review. Neurosurg Focus 2021; 50:E2. [PMID: 33524943 DOI: 10.3171/2020.11.focus20854] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/09/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE High-grade gliomas (HGGs) inevitably recur and progress despite resection and standard chemotherapies and radiation. Viral therapies have emerged as a theoretically favorable adjuvant modality that might overcome intrinsic factors of HGGs that confer treatment resistance. METHODS The authors present the results of systematic searches of the MEDLINE and ClinicalTrials.gov databases that were performed for clinical trials published or registered up to July 15, 2020. RESULTS Fifty-one completed clinical trials were identified that made use of a virus-based therapeutic strategy to treat HGG. The two main types of viral therapies were oncolytic viruses and viral vectors for gene therapy. Among clinical trials that met inclusion criteria, 20 related to oncolytic viruses and 31 to gene therapy trials. No oncolytic viruses have progressed to phase III clinical trial testing, although there have been many promising early-phase results and no reported cases of encephalitis or death due to viral therapy. Three phase III trials in which viral gene therapy was used have been completed but have not resulted in any FDA-approved therapy. Recent efforts in this area have been focused on the delivery of suicide genes such as herpes simplex virus thymidine kinase and cytosine deaminase. CONCLUSIONS Decades of research efforts and an improving understanding of the immunomodulatory effects of viral therapies for gliomas are informing ongoing clinical efforts aimed at improving outcomes in patients with HGG. The available clinical data reveal varied efficacy among different virus-based treatment strategies.
Collapse
Affiliation(s)
- Joshua L Wang
- 1Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | | | - Nicole M Wenger
- 3Department of Neurosurgery, University of Maryland, Baltimore, Maryland
| | - J Bradley Elder
- 1Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
9
|
Molecular Imaging of Gene Therapy. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00064-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
10
|
Hossain JA, Latif MA, Ystaas LAR, Ninzima S, Riecken K, Muller A, Azuaje F, Joseph JV, Talasila KM, Ghimire J, Fehse B, Bjerkvig R, Miletic H. Long-term treatment with valganciclovir improves lentiviral suicide gene therapy of glioblastoma. Neuro Oncol 2020; 21:890-900. [PMID: 30958558 DOI: 10.1093/neuonc/noz060] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Suicide gene therapy for malignant gliomas has shown encouraging results in the latest clinical trials. However, prodrug application was most often restricted to short-term treatment (14 days), especially when replication-defective vectors were used. We previously showed that a substantial fraction of herpes simplex virus thymidine kinase (HSV-TK) transduced tumor cells survive ganciclovir (GCV) treatment in an orthotopic glioblastoma (GBM) xenograft model. Here we analyzed whether these TK+ tumor cells are still sensitive to prodrug treatment and whether prolonged prodrug treatment can enhance treatment efficacy. METHODS Glioma cells positive for TK and green fluorescent protein (GFP) were sorted from xenograft tumors recurring after suicide gene therapy, and their sensitivity to GCV was tested in vitro. GBM xenografts were treated with HSV-TK/GCV, HSV-TK/valganciclovir (valGCV), or HSV-TK/valGCV + erlotinib. Tumor growth was analyzed by MRI, and survival as well as morphological and molecular changes were assessed. RESULTS TK-GFP+ tumor cells from recurrent xenograft tumors retained sensitivity to GCV in vitro. Importantly, a prolonged period (3 mo) of prodrug administration with valganciclovir (valGCV) resulted in a significant survival advantage compared with short-term (3 wk) application of GCV. Recurrent tumors from the treatment groups were more invasive and less angiogenic compared with primary tumors and showed significant upregulation of epidermal growth factor receptor (EGFR) expression. However, double treatment with the EGFR inhibitor erlotinib did not increase therapeutic efficacy. CONCLUSION Long-term treatment with valGCV should be considered as a replacement for short-term treatment with GCV in clinical trials of HSV-TK mediated suicide gene therapy.
Collapse
Affiliation(s)
- Jubayer A Hossain
- Department of Biomedicine, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Md A Latif
- Department of Biomedicine, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Lars A R Ystaas
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Sandra Ninzima
- Department of Biomedicine, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center, Hamburg, Germany
| | - Arnaud Muller
- Bioinformatics Team, Center for Quantitative Biology, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Francisco Azuaje
- Bioinformatics Team, Center for Quantitative Biology, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Justin V Joseph
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Jiwan Ghimire
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center, Hamburg, Germany
| | - Rolf Bjerkvig
- Department of Biomedicine, University of Bergen, Bergen, Norway.,Norlux Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Hrvoje Miletic
- Department of Biomedicine, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
11
|
Araldi RP, Khalil C, Grignet PH, Teixeira MR, de Melo TC, Módolo DG, Fernandes LGV, Ruiz J, de Souza EB. Medical applications of clustered regularly interspaced short palindromic repeats (CRISPR/Cas) tool: A comprehensive overview. Gene 2020; 745:144636. [PMID: 32244056 DOI: 10.1016/j.gene.2020.144636] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/01/2020] [Accepted: 03/30/2020] [Indexed: 12/22/2022]
Abstract
Since the discovery of the double helix and the introduction of genetic engineering, the possibility to develop new strategies to manipulate the genome has fascinated scientists around the world. Currently scientists have the knowledge andabilitytoedit the genomes. Several methodologies of gene editing have been established, all of them working like "scissor", creating double strand breaks at specific spots. The introduction of a new technology, which was adapted from the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas bacterial immune system, has revolutionized the genetic therapy field, as it allows a much more precise editing of gene than the previously described tools and, therefore, to prevent and treat disease in humans. This review aims to revisit the genome editing history that led to the rediscovery of the CRISPR/Cas technology and to explore the technical aspects, applications and perspectives of this fascinating, powerful, precise, simpler and cheaper technology in different fields.
Collapse
Affiliation(s)
- Rodrigo Pinheiro Araldi
- Genetic Bases of Thyroid Tumors Laboratory, Department of Morphology and Genetics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil; Programa de Pós-graduação em Biociências, Universidade Federal da Integração Latino-Americana (UNILA), Foz do Iguaçu, PR, Brazil.
| | - Charbel Khalil
- Reviva Research and Application Center- Lebanese University, Middle East Institute of Health University Hospital, Beirut, Lebanon
| | - Pedro Henrique Grignet
- Instituto Latino-Americano de Ciências da Vida e da Natureza (ILACVN), Universidade Federal da Integração Latino-Americana (UNILA), Foz do Iguaçu, PR, Brazil
| | - Michelli Ramires Teixeira
- Instituto Latino-Americano de Ciências da Vida e da Natureza (ILACVN), Universidade Federal da Integração Latino-Americana (UNILA), Foz do Iguaçu, PR, Brazil
| | - Thatiana Correa de Melo
- Instituto Latino-Americano de Ciências da Vida e da Natureza (ILACVN), Universidade Federal da Integração Latino-Americana (UNILA), Foz do Iguaçu, PR, Brazil
| | | | | | - Jorge Ruiz
- Programa de Pós-graduação em Biociências, Universidade Federal da Integração Latino-Americana (UNILA), Foz do Iguaçu, PR, Brazil; Instituto Latino-Americano de Ciências da Vida e da Natureza (ILACVN), Universidade Federal da Integração Latino-Americana (UNILA), Foz do Iguaçu, PR, Brazil
| | - Edislane Barreiros de Souza
- Laboratory of Genetics, Molecular Biology and Mutagenesis, Faculdade de Ciências e Letras de Assis, Universidade Estadual Paulista "Júlio de Mesquita Filho" (UNESP), Assis, SP, Brazil
| |
Collapse
|
12
|
Tamura R, Miyoshi H, Yoshida K, Okano H, Toda M. Recent progress in the research of suicide gene therapy for malignant glioma. Neurosurg Rev 2019; 44:29-49. [PMID: 31781985 DOI: 10.1007/s10143-019-01203-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/14/2019] [Accepted: 10/28/2019] [Indexed: 12/15/2022]
Abstract
Malignant glioma, which is characterized by diffuse infiltration into the normal brain parenchyma, is the most aggressive primary brain tumor with dismal prognosis. Over the past 40 years, the median survival has only slightly improved. Therefore, new therapeutic modalities must be developed. In the 1990s, suicide gene therapy began attracting attention for the treatment of malignant glioma. Some clinical trials used a viral vector for suicide gene transduction; however, it was found that viral vectors cannot cover the large invaded area of glioma cells. Interest in this therapy was recently revived because some types of stem cells possess a tumor-tropic migratory capacity, which can be used as cellular delivery vehicles. Immortalized, clonal neural stem cell (NSC) line has been used for patients with recurrent high-grade glioma, which showed safety and efficacy. Embryonic and induced pluripotent stem cells may be considered as sources of NSC because NSC is difficult to harvest, and ethical issues have been raised. Mesenchymal stem cells are alternative candidates for cellular vehicle and are easily harvested from the bone marrow. In addition, a new type of nonlytic, amphotropic retroviral replicating vector encoding suicide gene has shown efficacy in patients with recurrent high-grade glioma in a clinical trial. This replicating viral capacity is another possible candidate as delivery vehicle to tackle gliomas. Herein, we review the concept of suicide gene therapy, as well as recent progress in preclinical and clinical studies in this field.
Collapse
Affiliation(s)
- Ryota Tamura
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiroyuki Miyoshi
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kazunari Yoshida
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Masahiro Toda
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
13
|
Luzzi S, Crovace AM, Del Maestro M, Giotta Lucifero A, Elbabaa SK, Cinque B, Palumbo P, Lombardi F, Cimini A, Cifone MG, Crovace A, Galzio R. The cell-based approach in neurosurgery: ongoing trends and future perspectives. Heliyon 2019; 5:e02818. [PMID: 31844735 PMCID: PMC6889232 DOI: 10.1016/j.heliyon.2019.e02818] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/11/2019] [Accepted: 11/06/2019] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Examination of the current trends and future perspectives of the cell-based therapies in neurosurgery. METHODS A PubMed/MEDLINE-based systematic review has been performed combining the main Medical Subject Headings (MeSH) regarding the cell- and tissue-based therapies with the "Brain", "Spinal Cord", "Spine" and "Skull" MeSH terms. Only articles in English published in the last 10 years and pertinent to neurosurgery have been selected. RESULTS A total of 1,173 relevant articles have been chosen. Somatic cells and gene-modification technologies have undergone the greatest development. Immunotherapies and gene therapies have been tested for the cure of glioblastoma, stem cells mainly for brain and spinal cord traumatic injuries. Stem cells have also found a rationale in the treatment of the cranial and spinal bony defects, and of the intervertebral disc degeneration, as well.Most of the completed or ongoing trials concerning the cell-based therapies in neurosurgery are on phase 2. Future perspectives involve the need to overcome issues related to immunogenicity, oncogenicity and routes for administration. Refinement and improvement of vector design and delivery are required within the gene therapies. CONCLUSION The last decade has been characterised by a progressive evolution of neurosurgery from a purely mechanical phase to a new biological one. This trend has followed the rapid and parallel development of translational medicine and nanotechnologies.The introduction of new technologies, the optimisation of the already existing ones, and the reduction of costs are among the main challenges of the foreseeable future.
Collapse
Affiliation(s)
- Sabino Luzzi
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Polo Didattico "Cesare Brusotti", Viale Brambilla, 74, Pavia, 27100, Italy
- Neurosurgery Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, Viale C. Golgi, 19, Pavia, 27100, Italy
| | - Alberto Maria Crovace
- Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro", Piazza G. Cesare, 11 – Policlinico di Bari, Bari, 70124, Italy
| | - Mattia Del Maestro
- Neurosurgery Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, Viale C. Golgi, 19, Pavia, 27100, Italy
- PhD School in Experimental Medicine, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Polo Didattico "Cesare Brusotti", Viale Brambilla, 74, Pavia, 27100, Italy
| | - Alice Giotta Lucifero
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Polo Didattico "Cesare Brusotti", Viale Brambilla, 74, Pavia, 27100, Italy
| | - Samer K. Elbabaa
- Pediatric Neurosurgery, Pediatric Neuroscience Center of Excellence, Arnold Palmer Hospital for Children, 1222 S. Orange Avenue, 2nd Floor, MP 154, Orlando, FL, 32806, USA
| | - Benedetta Cinque
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, via Coppito, L'Aquila, 67100, Italy
| | - Paola Palumbo
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, via Coppito, L'Aquila, 67100, Italy
| | - Francesca Lombardi
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, via Coppito, L'Aquila, 67100, Italy
| | - Annamaria Cimini
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, via Coppito, L'Aquila, 67100, Italy
| | - Maria Grazia Cifone
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, via Coppito, L'Aquila, 67100, Italy
| | - Antonio Crovace
- Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro", Piazza G. Cesare, 11 – Policlinico di Bari, Bari, 70124, Italy
| | - Renato Galzio
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Polo Didattico "Cesare Brusotti", Viale Brambilla, 74, Pavia, 27100, Italy
- Neurosurgery Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, Viale C. Golgi, 19, Pavia, 27100, Italy
| |
Collapse
|
14
|
Shi M, Sanche L. Convection-Enhanced Delivery in Malignant Gliomas: A Review of Toxicity and Efficacy. JOURNAL OF ONCOLOGY 2019; 2019:9342796. [PMID: 31428153 PMCID: PMC6679879 DOI: 10.1155/2019/9342796] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/06/2019] [Accepted: 06/25/2019] [Indexed: 12/15/2022]
Abstract
Malignant gliomas are undifferentiated or anaplastic gliomas. They remain incurable with a multitude of modalities, including surgery, radiation, chemotherapy, and alternating electric field therapy. Convection-enhanced delivery (CED) is a local treatment that can bypass the blood-brain barrier and increase the tumor uptake of therapeutic agents, while decreasing exposure to healthy tissues. Considering the multiple choices of drugs with different antitumor mechanisms, the supra-additive effect of concomitant radiation and chemotherapy, CED appears as a promising modality for the treatment of brain tumors. In this review, the CED-related toxicities are summarized and classified into immediate, early, and late side effects based on the time of onset, and local and systemic toxicities based on the location of toxicity. The efficacies of CED of various therapeutic agents including targeted antitumor agents, chemotherapeutic agents, radioisotopes, and immunomodulators are covered. The phase III trial PRECISE compares CED of IL13-PE38QQR, an interleukin-13 conjugated to Pseudomonas aeruginosa exotoxin A, to Gliadel® Wafer, a polymer loaded with carmustine. However, in this case, CED had no significant median survival improvement (11.3 months vs. 10 months) in patients with recurrent glioblastomas. In phase II studies, CED of recombinant poliovirus (PVSRIPO) had an overall survival of 21% vs. 14% for the control group at 24 months, and 21% vs. 4% at 36 months. CED of Tf-diphtheria toxin had a response rate of 35% in recurrent malignant gliomas patients. On the other hand, the TGF-β2 inhibitor Trabedersen, HSV-1-tk ganciclovir, and radioisotope 131I-chTNT-1/B mAb had a limited response rate. With this treatment, patients who received CED of the chemotherapeutic agent paclitaxel and immunomodulator, oligodeoxynucleotides containing CpG motifs (CpG-ODN), experienced intolerable toxicity. Toward the end of this article, an ideal CED treatment procedure is proposed and the methods for quality assurance of the CED procedure are discussed.
Collapse
Affiliation(s)
- Minghan Shi
- Department of Radiation Oncology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Léon Sanche
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
15
|
Kojima K, Miyoshi H, Nagoshi N, Kohyama J, Itakura G, Kawabata S, Ozaki M, Iida T, Sugai K, Ito S, Fukuzawa R, Yasutake K, Renault‐Mihara F, Shibata S, Matsumoto M, Nakamura M, Okano H. Selective Ablation of Tumorigenic Cells Following Human Induced Pluripotent Stem Cell-Derived Neural Stem/Progenitor Cell Transplantation in Spinal Cord Injury. Stem Cells Transl Med 2019; 8:260-270. [PMID: 30485733 PMCID: PMC6392358 DOI: 10.1002/sctm.18-0096] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 10/12/2018] [Indexed: 12/29/2022] Open
Abstract
Tumorigenesis is an important problem that needs to be addressed in the field of human stem/progenitor cell transplantation for the treatment of subacute spinal cord injury (SCI). When certain "tumorigenic" cell lines are transplanted into the spinal cord of SCI mice model, there is initial improvement of motor function, followed by abrupt deterioration secondary to the effect of tumor growth. A significant proportion of the transplanted cells remains undifferentiated after transplantation and is thought to increase the risk of tumorigenesis. In this study, using lentiviral vectors, we introduced the herpes simplex virus type 1 thymidine kinase (HSVtk) gene into a human induced pluripotent stem cell-derived neural stem/progenitor cell (hiPSC-NS/PC) line that is known to undergo tumorigenic transformation. Such approach enables selective ablation of the immature proliferating cells and thereby prevents subsequent tumor formation. In vitro, the HSVtk system successfully ablated the immature proliferative neural cells while preserving mature postmitotic neuronal cells. Similar results were observed in vivo following transplantation into the injured spinal cords of immune-deficient (nonobese diabetic-severe combined immune-deficient) mice. Ablation of the proliferating cells exerted a protective effect on the motor function which was regained after transplantation, simultaneously defending the spinal cord from the harmful tumor growth. These results suggest a potentially promising role of suicide genes in opposing tumorigenesis during stem cell therapy. This system allows both preventing and treating tumorigenesis following hiPSC-NS/PC transplantation without sacrificing the improved motor function. Stem Cells Translational Medicine 2019;8:260&270.
Collapse
Affiliation(s)
- Kota Kojima
- Department of PhysiologyKeio University School of MedicineTokyoJapan
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Hiroyuki Miyoshi
- Department of PhysiologyKeio University School of MedicineTokyoJapan
| | - Narihito Nagoshi
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Jun Kohyama
- Department of PhysiologyKeio University School of MedicineTokyoJapan
| | - Go Itakura
- Department of PhysiologyKeio University School of MedicineTokyoJapan
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Soya Kawabata
- Department of PhysiologyKeio University School of MedicineTokyoJapan
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Masahiro Ozaki
- Department of PhysiologyKeio University School of MedicineTokyoJapan
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Tsuyoshi Iida
- Department of PhysiologyKeio University School of MedicineTokyoJapan
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Keiko Sugai
- Department of PhysiologyKeio University School of MedicineTokyoJapan
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Shuhei Ito
- Department of PhysiologyKeio University School of MedicineTokyoJapan
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Ryuji Fukuzawa
- Department of PathologyInternational University of Health and WelfareChibaJapan
| | - Kaori Yasutake
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | | | - Shinsuke Shibata
- Department of PhysiologyKeio University School of MedicineTokyoJapan
| | - Morio Matsumoto
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Masaya Nakamura
- Department of Orthopaedic SurgeryKeio University School of MedicineTokyoJapan
| | - Hideyuki Okano
- Department of PhysiologyKeio University School of MedicineTokyoJapan
| |
Collapse
|
16
|
Hitti FL, Gonzalez-Alegre P, Lucas TH. Gene Therapy for Neurologic Disease: A Neurosurgical Review. World Neurosurg 2019; 121:261-273. [DOI: 10.1016/j.wneu.2018.09.097] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/10/2018] [Accepted: 09/12/2018] [Indexed: 01/01/2023]
|
17
|
Comparative effect of immunotherapy and standard therapy in patients with high grade glioma: a meta-analysis of published clinical trials. Sci Rep 2018; 8:11800. [PMID: 30087385 PMCID: PMC6081409 DOI: 10.1038/s41598-018-30296-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 07/20/2018] [Indexed: 01/26/2023] Open
Abstract
Immunotherapy holds great promise in the treatment of high grade glioma (HGG). We performed a comprehensive meta-analysis of clinical trials involving dendritic cell (DC) therapy and viral therapy (VT) for the treatment of HGG, in order to assess their clinical impact in comparison to standard treatments in terms of overall survival (OS) and progression-free survival (PFS). To our knowledge, this is the first meta-analysis to evaluate VT for the treatment of HGG, allowing comparison of different immunotherapeutic approaches. Thirteen eligible studies of 1043 cases were included in the meta-analysis. For DC vaccination, in terms of OS, both newly diagnosed patients (HR, 0.65) and patients who suffered from recurrent HGGs (HR = 0.63) presented markedly improved results compared to the control groups. PFS was also improved (HR = 0.49) but was not statistically significant (p = 0.1). A slight improvement was observed for newly diagnosed patients receiving VT in terms of OS (HR = 0.88) while PFS was inferior for patients in the experimental arm (HR = 1.16). Our results show that DC therapy greatly improves OS for patients with both newly diagnosed and recurrent HGGs. VT, however, did not provide any statistically significant improvements in terms of OS and PFS for patients with newly diagnosed HGGs.
Collapse
|
18
|
|
19
|
Hossain JA, Ystaas LR, Mrdalj J, Välk K, Riecken K, Fehse B, Bjerkvig R, Grønli J, Miletic H. Lentiviral HSV-Tk.007-mediated suicide gene therapy is not toxic for normal brain cells. J Gene Med 2018; 18:234-43. [PMID: 27490042 DOI: 10.1002/jgm.2895] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 07/28/2016] [Accepted: 07/28/2016] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Gene therapeutic strategies with suicide genes are currently investigated in clinical trials for brain tumors. Previously, we have shown that lentiviral vectors delivering the suicide gene HSV-Tk to experimental brain tumors promote a highly significant treatment effect and thus are promising vectors for clinical translation. METHODS In the present study, we tested lentiviral vectors delivering the suicide gene HSV-Tk.007, a highly active mutant of HSV-Tk, to rat brains as a preclinical toxicity study. We injected 10(6) vesicular stomatitis virus glycoprotein (VSV-G) pseudotyped functional lentiviral particles harboring the suicide gene HSV-Tk.007 into the brain of healthy, immunocompetent rats. During prodrug treatment with ganciclovir (GCV), we measured weight and assessed the behavior of the rats in an open field test. After 14 days of GCV treatment, we analyzed HSV-Tk.007 expression in different brain cell populations, as well as inflammatory responses and apoptosis. RESULTS During prodrug treatment with GCV, behavior experiments did not reveal differences between the treated rats and the control groups. Analysis of HSV-Tk expression in different brain cell populations showed that transduced normal brain cells survived GCV treatment. There were no statistically significant differences in the number of transduced cells between treatment and control groups. Furthermore, inflammatory responses and apoptosis of brain cells were not observed. CONCLUSIONS We show that HSV-Tk.007-mediated suicide gene therapy is not toxic to normal brain cells. This observation is of high relevance for the translation of lentivirus-mediated suicide gene therapies into the clinic for the treatment of brain tumor patients. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jubayer A Hossain
- Department of Biomedicine, University of Bergen, Bergen, Norway.,KG Jebsen Brain Tumour Research Center, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Lars Rømo Ystaas
- Department of Biomedicine, University of Bergen, Bergen, Norway.,KG Jebsen Brain Tumour Research Center, University of Bergen, Bergen, Norway
| | - Jelena Mrdalj
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway.,Norwegian Competence Center for Sleep Disorders, Haukeland University Hospital, Bergen, Norway
| | - Kristjan Välk
- Department of Biomedicine, University of Bergen, Bergen, Norway.,KG Jebsen Brain Tumour Research Center, University of Bergen, Bergen, Norway
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Rolf Bjerkvig
- Department of Biomedicine, University of Bergen, Bergen, Norway.,KG Jebsen Brain Tumour Research Center, University of Bergen, Bergen, Norway.,Norlux Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg
| | - Janne Grønli
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway.,Norwegian Competence Center for Sleep Disorders, Haukeland University Hospital, Bergen, Norway
| | - Hrvoje Miletic
- Department of Biomedicine, University of Bergen, Bergen, Norway. .,KG Jebsen Brain Tumour Research Center, University of Bergen, Bergen, Norway. .,Department of Pathology, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
20
|
Mastorakos P, Zhang C, Song E, Kim YE, Park HW, Berry S, Choi WK, Hanes J, Suk JS. Biodegradable brain-penetrating DNA nanocomplexes and their use to treat malignant brain tumors. J Control Release 2017; 262:37-46. [PMID: 28694032 DOI: 10.1016/j.jconrel.2017.07.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/21/2017] [Accepted: 07/07/2017] [Indexed: 11/30/2022]
Abstract
The discovery of powerful genetic targets has spurred clinical development of gene therapy approaches to treat patients with malignant brain tumors. However, lack of success in the clinic has been attributed to the inability of conventional gene vectors to achieve gene transfer throughout highly disseminated primary brain tumors. Here, we demonstrate ex vivo that small nanocomplexes composed of DNA condensed by a blend of biodegradable polymer, poly(β-amino ester) (PBAE), with PBAE conjugated with 5kDa polyethylene glycol (PEG) molecules (PBAE-PEG) rapidly penetrate healthy brain parenchyma and orthotopic brain tumor tissues in rats. Rapid diffusion of these DNA-loaded nanocomplexes observed in fresh tissues ex vivo demonstrated that they avoided adhesive trapping in the brain owing to their dense PEG coating, which was critical to achieving widespread transgene expression throughout orthotopic rat brain tumors in vivo following administration by convection enhanced delivery. Transgene expression with the PBAE/PBAE-PEG blended nanocomplexes (DNA-loaded brain-penetrating nanocomplexes, or DNA-BPN) was uniform throughout the tumor core compared to nanocomplexes composed of DNA with PBAE only (DNA-loaded conventional nanocomplexes, or DNA-CN), and transgene expression reached beyond the tumor edge, where infiltrative cancer cells are found, only for the DNA-BPN formulation. Finally, DNA-BPN loaded with anti-cancer plasmid DNA provided significantly enhanced survival compared to the same plasmid DNA loaded in DNA-CN in two aggressive orthotopic brain tumor models in rats. These findings underscore the importance of achieving widespread delivery of therapeutic nucleic acids within brain tumors and provide a promising new delivery platform for localized gene therapy in the brain.
Collapse
Affiliation(s)
- Panagiotis Mastorakos
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Clark Zhang
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eric Song
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Biotechnology Education, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Young Eun Kim
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Hee Won Park
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Sneha Berry
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Biotechnology Education, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Won Kyu Choi
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Justin Hanes
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Oncology, Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pharmacology & Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Jung Soo Suk
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
21
|
Gardeck AM, Sheehan J, Low WC. Immune and viral therapies for malignant primary brain tumors. Expert Opin Biol Ther 2017; 17:457-474. [DOI: 10.1080/14712598.2017.1296132] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Andrew M. Gardeck
- Departments of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
| | - Jordan Sheehan
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Walter C. Low
- Departments of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
22
|
Chandran JS, Scarrott JM, Shaw PJ, Azzouz M. Gene Therapy in the Nervous System: Failures and Successes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1007:241-257. [PMID: 28840561 DOI: 10.1007/978-3-319-60733-7_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Genetic disorders, caused by deleterious changes in the DNA sequence away from the normal genomic sequence, affect millions of people worldwide. Gene therapy as a treatment option for patients is an attractive proposition due to its conceptual simplicity. In principle, gene therapy involves correcting the genetic disorder by either restoring a normal functioning copy of a gene or reducing the toxicity arising from a mutated gene. In this way specific genetic function can be restored without altering the expression of other genes and the proteins they encode. The reality however is much more complex, and as a result the vector systems used to deliver gene therapies have by necessity continued to evolve and improve over time with respect to safety profile, efficiency, and long-term expression. In this chapter we examine the current approaches to gene therapy, assess the different gene delivery systems utilized, and highlight the failures and successes of relevant clinical trials. We do not intend for this chapter to be a comprehensive and exhaustive assessment of all clinical trials that have been conducted in the CNS, but instead will focus on specific diseases that have seen successes and failures with different gene therapy vehicles to gauge how preclinical models have informed the design of clinical trials.
Collapse
Affiliation(s)
- Jayanth S Chandran
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
| | - Joseph M Scarrott
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
| | - Mimoun Azzouz
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK.
| |
Collapse
|
23
|
Ultrasound-targeted microbubble destruction in gene therapy: A new tool to cure human diseases. Genes Dis 2016; 4:64-74. [PMID: 30258909 PMCID: PMC6136600 DOI: 10.1016/j.gendis.2016.08.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/01/2016] [Indexed: 01/11/2023] Open
Abstract
Human gene therapy has made significant advances in less than two decades. Within this short period of time, gene therapy has proceeded from the conceptual stage to technology development and laboratory research, and finally to clinical trials for the treatment of a variety of deadly diseases. Cardiovascular disease, cancer, and stroke are leading causes of death worldwide. Despite advances in medical, interventional, radiation and surgical treatments, the mortality rate remains high, and the need for novel therapies is great. Gene therapy provides an efficient approach to disease treatment. Notable advances in gene therapy have been made for genetic disorders, including severe combined immune deficiency, chronic granulomatus disorder, hemophilia and blindness, as well as for acquired diseases, including cancer and neurodegenerative and cardiovascular diseases. However, lack of an efficient delivery system to target cells as well as the difficulty of sustained expression of transgenes has hindered advancements in gene therapy. Ultrasound targeted microbubble destruction (UTMD) is a promising approach for target-specific gene delivery, and it has been successfully investigated for the treatment of many diseases in the past decade. In this paper, we review UTMD-mediated gene delivery for the treatment of cardiovascular diseases, cancer and stroke.
Collapse
|
24
|
Chimeric adeno-associated virus and bacteriophage: a potential targeted gene therapy vector for malignant glioma. Ther Deliv 2016; 5:975-90. [PMID: 25375341 DOI: 10.4155/tde.14.58] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The incipient development of gene therapy for cancer has fuelled its progression from bench to bedside in mere decades. Of all malignancies that exist, gliomas are the largest class of brain tumors, and are renowned for their aggressiveness and resistance to therapy. In order for gene therapy to achieve clinical success, a multitude of barriers ranging from glioma tumor physiology to vector biology must be overcome. Many viral gene delivery systems have been subjected to clinical investigation; however, with highly limited success. In this review, the current progress and challenges of gene therapy for malignant glioma are discussed. Moreover, we highlight the hybrid adeno-associated virus and bacteriophage vector as a potential candidate for targeted gene delivery to brain tumors.
Collapse
|
25
|
Lowenstein PR, Castro MG. The Long and Winding Road: From the High-Affinity Choline Uptake Site to Clinical Trials for Malignant Brain Tumors. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 76:147-73. [PMID: 27288077 DOI: 10.1016/bs.apha.2016.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Malignant brain tumors are one of the most lethal cancers. They originate from glial cells which infiltrate throughout the brain. Current standard of care involves surgical resection, radiotherapy, and chemotherapy; median survival is currently ~14-20 months postdiagnosis. Given that the brain immune system is deficient in priming systemic immune responses to glioma antigens, we proposed to reconstitute the brain immune system to achieve immunological priming from within the brain. Two adenoviral vectors are injected into the resection cavity or remaining tumor. One adenoviral vector expresses the HSV-1-derived thymidine kinase which converts ganciclovir into a compound only cytotoxic to dividing glioma cells. The second adenovirus expresses the cytokine fms-like tyrosine kinase 3 ligand (Flt3L). Flt3L differentiates precursors into dendritic cells and acts as a chemokine that attracts dendritic cells to the brain. HSV-1/ganciclovir killing of tumor cells releases tumor antigens that are taken up by dendritic cells within the brain tumor microenvironment. Tumor killing also releases HMGB1, an endogenous TLR2 agonist that activates dendritic cells. HMGB1-activated dendritic cells, loaded with glioma antigens, migrate to cervical lymph nodes to stimulate a systemic CD8+ T cells cytotoxic immune response against glioma. This immune response is specific to glioma tumors, induces immunological memory, and does neither cause brain toxicity nor autoimmune responses. An IND was granted by the FDA on 4/7/2011. A Phase I, first in person trial, to test whether reengineering the brain immune system is potentially therapeutic is ongoing.
Collapse
Affiliation(s)
- P R Lowenstein
- The Medical School, The University of Michigan, Ann Arbor, MI, United States.
| | - M G Castro
- The Medical School, The University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
26
|
Hulou MM, Cho CF, Chiocca EA, Bjerkvig R. Experimental therapies: gene therapies and oncolytic viruses. HANDBOOK OF CLINICAL NEUROLOGY 2016; 134:183-197. [PMID: 26948355 DOI: 10.1016/b978-0-12-802997-8.00011-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Glioblastoma is the most common and aggressive primary brain tumor in adults. Over the past three decades, the overall survival time has only improved by a few months, therefore novel alternative treatment modalities are needed to improve clinical management strategies. Such strategies should ultimately extend patient survival. At present, the extensive insight into the molecular biology of gliomas, as well as into genetic engineering techniques, has led to better decision processes when it comes to modifying the genome to accommodate suicide genes, cytokine genes, and tumor suppressor genes that may kill cancer cells, and boost the host defensive immune system against neoantigenic cytoplasmic and nuclear targets. Both nonreplicative viral vectors and replicating oncolytic viruses have been developed for brain cancer treatment. Stem cells, microRNAs, nanoparticles, and viruses have also been designed. These have been armed with transgenes or peptides, and have been used both in laboratory-based experiments as well as in clinical trials, with the aim of improving selective killing of malignant glioma cells while sparing normal brain tissue. This chapter reviews the current status of gene therapies for malignant gliomas and highlights the most promising viral and cell-based strategies under development.
Collapse
Affiliation(s)
- M Maher Hulou
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Choi-Fong Cho
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - E Antonio Chiocca
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Rolf Bjerkvig
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg; Department of Biomedicine, University of Bergen, Norway
| |
Collapse
|
27
|
Mangraviti A, Tzeng SY, Kozielski KL, Wang Y, Jin Y, Gullotti D, Pedone M, Buaron N, Liu A, Wilson DR, Hansen SK, Rodriguez FJ, Gao GD, DiMeco F, Brem H, Olivi A, Tyler B, Green JJ. Polymeric nanoparticles for nonviral gene therapy extend brain tumor survival in vivo. ACS NANO 2015; 9:1236-49. [PMID: 25643235 PMCID: PMC4342728 DOI: 10.1021/nn504905q] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Biodegradable polymeric nanoparticles have the potential to be safer alternatives to viruses for gene delivery; however, their use has been limited by poor efficacy in vivo. In this work, we synthesize and characterize polymeric gene delivery nanoparticles and evaluate their efficacy for DNA delivery of herpes simplex virus type I thymidine kinase (HSVtk) combined with the prodrug ganciclovir (GCV) in a malignant glioma model. We investigated polymer structure for gene delivery in two rat glioma cell lines, 9L and F98, to discover nanoparticle formulations more effective than the leading commercial reagent Lipofectamine 2000. The lead polymer structure, poly(1,4-butanediol diacrylate-co-4-amino-1-butanol) end-modified with 1-(3-aminopropyl)-4-methylpiperazine, is a poly(β-amino ester) (PBAE) and formed nanoparticles with HSVtk DNA that were 138 ± 4 nm in size and 13 ± 1 mV in zeta potential. These nanoparticles containing HSVtk DNA showed 100% cancer cell killing in vitro in the two glioma cell lines when combined with GCV exposure, while control nanoparticles encoding GFP maintained robust cell viability. For in vivo evaluation, tumor-bearing rats were treated with PBAE/HSVtk infusion via convection-enhanced delivery (CED) in combination with systemic administration of GCV. These treated animals showed a significant benefit in survival (p = 0.0012 vs control). Moreover, following a single CED infusion, labeled PBAE nanoparticles spread completely throughout the tumor. This study highlights a nanomedicine approach that is highly promising for the treatment of malignant glioma.
Collapse
Affiliation(s)
- Antonella Mangraviti
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Stephany Yi Tzeng
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- The Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Kristen Lynn Kozielski
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- The Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Yuan Wang
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an 710032, China
| | - Yike Jin
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - David Gullotti
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Mariangela Pedone
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Nitsa Buaron
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Chemical Engineering, Ben Gurion University of the Negev, Be’er Sheva 84105, Israel
| | - Ann Liu
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - David R. Wilson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- The Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Sarah K. Hansen
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- The Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Fausto J. Rodriguez
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Guo-Dong Gao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an 710032, China
| | - Francesco DiMeco
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico C. Besta, Milan 20133, Italy
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Alessandro Olivi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Betty Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Address correspondence to ,
| | - Jordan J. Green
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- The Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Material Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21231, United States
- Address correspondence to ,
| |
Collapse
|
28
|
Hirst TC, Vesterinen HM, Conlin S, Egan KJ, Antonic A, Lawson McLean A, Macleod MR, Grant R, Brennan PM, Sena ES, Whittle IR. A systematic review and meta-analysis of gene therapy in animal models of cerebral glioma: why did promise not translate to human therapy? ACTA ACUST UNITED AC 2015; 1:e00006. [PMID: 27668084 PMCID: PMC5020579 DOI: 10.1002/ebm2.6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 11/22/2014] [Accepted: 11/28/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND The development of therapeutics is often characterized by promising animal research that fails to translate into clinical efficacy; this holds for the development of gene therapy in glioma. We tested the hypothesis that this is because of limitations in the internal and external validity of studies reporting the use of gene therapy in experimental glioma. METHOD We systematically identified studies testing gene therapy in rodent glioma models by searching three online databases. The number of animals treated and median survival were extracted and studies graded using a quality checklist. We calculated median survival ratios and used random effects meta-analysis to estimate efficacy. We explored effects of study design and quality and searched for evidence of publication bias. RESULTS We identified 193 publications using gene therapy in experimental glioma, including 6,366 animals. Overall, gene therapy improved median survival by a factor of 1.60 (95% CI 1.53-1.67). Study quality was low and the type of gene therapy did not account for differences in outcome. Study design characteristics accounted for a significant proportion of between-study heterogeneity. We observed similar findings in a data subset limited to the most common gene therapy. CONCLUSION As the dysregulation of key molecular pathways is characteristic of gliomas, gene therapy remains a promising treatment for glioma. Nevertheless, we have identified areas for improvement in conduct and reporting of studies, and we provide a basis for sample size calculations. Further work should focus on genes of interest in paradigms recapitulating human disease. This might improve the translation of such therapies into the clinic.
Collapse
Affiliation(s)
- T C Hirst
- Centre for Clinical Brain Sciences Chancellors Building University of Edinburgh Edinburgh UK
| | - H M Vesterinen
- Centre for Clinical Brain Sciences Chancellors Building University of Edinburgh Edinburgh UK
| | - S Conlin
- Centre for Clinical Brain Sciences Chancellors Building University of Edinburgh Edinburgh UK
| | - K J Egan
- Centre for Clinical Brain Sciences Chancellors Building University of Edinburgh Edinburgh UK
| | - A Antonic
- Florey Neuroscience and Mental Health Institute University of Melbourne Victoria Australia
| | - A Lawson McLean
- Centre for Clinical Brain Sciences Chancellors Building University of Edinburgh Edinburgh UK
| | - M R Macleod
- Division of Clinical Neurosciences University of Edinburgh Western General Hospital Edinburgh UK
| | - R Grant
- Division of Clinical Neurosciences University of Edinburgh Western General Hospital Edinburgh UK
| | - P M Brennan
- Division of Clinical Neurosciences University of Edinburgh Western General Hospital Edinburgh UK
| | - E S Sena
- Centre for Clinical Brain Sciences Chancellors Building University of Edinburgh Edinburgh UK; Florey Neuroscience and Mental Health Institute University of Melbourne Victoria Australia
| | - I R Whittle
- Division of Clinical Neurosciences University of Edinburgh Western General Hospital Edinburgh UK
| |
Collapse
|
29
|
Zhou XL, Shi YL, Li X. Inhibitory effects of the ultrasound-targeted microbubble destruction-mediated herpes simplex virus-thymidine kinase/ganciclovir system on ovarian cancer in mice. Exp Ther Med 2014; 8:1159-1163. [PMID: 25187815 PMCID: PMC4151658 DOI: 10.3892/etm.2014.1877] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 06/25/2014] [Indexed: 12/23/2022] Open
Abstract
The aim of the present study was to investigate the effect of the ultrasound-targeted microbubble destruction mediated (UTMD) herpes simplex virus-thymidine kinase (HSV-TK) and ganciclovir (GCV) system on ovarian cancer (OC). This study was conducted between June and December 2012 in the Animal Biosafety Level III Laboratory of Wuhan University. Mice with OC were randomly divided into four groups: i) HSV-TK plus microbubbles (MBs) plus ultrasound (US) (n=15); ii) HSV-TK plus US (n=15); iii) HSV-TK (n=15); and iv) phosphate-buffered saline (n=15). The inhibitory effect and survival time in the experimental groups were compared with those in the control group. The TK protein expression was detected by western blot analysis. Tumor cell apoptosis was detected by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling and caspase-3 activity analysis. The data showed that the efficiency of HSV-TK gene transfection and the tumor inhibitory effects were significantly increased in the HSV-TK plus MBs plus US group compared with those in the control group (P<0.01). UTMD-mediated HSV-TK treatment has also improved the rat survival rate (P<0.01). In conclusion, UTMD can effectively transfect the HSV-TK gene into target tissues and exert a significant inhibitory effect on OC in mice.
Collapse
Affiliation(s)
- Xian-Long Zhou
- Emergency Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yu-Lu Shi
- Medical College of Wuhan University, Wuhan, Hubei 430072, P.R. China
| | - Xiong Li
- Department of Ultrasound, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
30
|
Burnett R, Ibañez CE, Pettersson PL, Chen CI, Parab S, Huang T, Robbins J, Bankiewicz K, Aghi M, Logg C, Kasahara N, Pertschuk D, Gruber HE, Jolly DJ. Maintaining therapeutic activity in the operating room: compatibility of a gamma-retroviral replicating vector with clinical materials and biofluids. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2014; 1:14024. [PMID: 26015967 PMCID: PMC4362351 DOI: 10.1038/mtm.2014.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Revised: 04/08/2014] [Accepted: 04/09/2014] [Indexed: 11/28/2022]
Abstract
Toca 511 is a novel retroviral replicating vector, encoding a modified yeast cytosine deaminase, administered to recurrent high grade glioma patients in Phase 1 trials by stereotactic, transcranial injection into the tumor or into the walls of the resection cavity. A key issue, with little published data, is vector biocompatibility with agents likely to be encountered in a neurosurgical setting. We tested biocompatibility of Toca 511 with: delivery devices; MRI contrast agents, including ProHance supporting coinjection for real time MRI-guided intratumoral delivery; hemostatic agents; biofluids (blood and cerebrospinal fluid); potential adjuvants; and a needleless vial adapter that reduces risk of accidental needle sticks. Toca 511 is stable upon thawing at ambient temperature for at least 6 hours, allowing sufficient time for administration, and its viability is not reduced in the presence of: stainless steel and silica-based delivery devices; the potential MRI contrast agent, Feraheme; ProHance at several concentrations; the hemostatic agent SURGIFOAM; blood; cerebrospinal fluid; and the needleless vial adapter. Toca 511 is not compatible with the hemostatic agent SURGICEL or with extended exposures to titanium-based biopsy needles.
Collapse
Affiliation(s)
- Ryan Burnett
- Tocagen Inc., Bunker Hill St. , San Diego, CA, USA
| | | | | | - Ching-I Chen
- Tocagen Inc., Bunker Hill St. , San Diego, CA, USA
| | | | | | - Joan Robbins
- Tocagen Inc., Bunker Hill St. , San Diego, CA, USA
| | - Krystof Bankiewicz
- Department of Neurological Surgery, University of California San Francisco (UCSF) , San Francisco, CA, USA
| | - Manish Aghi
- Department of Neurological Surgery, University of California San Francisco (UCSF) , San Francisco, CA, USA
| | - Christopher Logg
- Department of Medicine, University of California Los Angeles (UCLA) , Los Angeles, CA, USA
| | - Noriyuki Kasahara
- Department of Medicine, University of California Los Angeles (UCLA) , Los Angeles, CA, USA
| | | | | | | |
Collapse
|
31
|
Wilson TA, Karajannis MA, Harter DH. Glioblastoma multiforme: State of the art and future therapeutics. Surg Neurol Int 2014; 5:64. [PMID: 24991467 PMCID: PMC4078454 DOI: 10.4103/2152-7806.132138] [Citation(s) in RCA: 182] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 03/13/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most common and lethal primary malignancy of the central nervous system (CNS). Despite the proven benefit of surgical resection and aggressive treatment with chemo- and radiotherapy, the prognosis remains very poor. Recent advances of our understanding of the biology and pathophysiology of GBM have allowed the development of a wide array of novel therapeutic approaches, which have been developed. These novel approaches include molecularly targeted therapies, immunotherapies, and gene therapy. METHODS We offer a brief review of the current standard of care, and a survey of novel therapeutic approaches for treatment of GBM. RESULTS Despite promising results in preclinical trials, many of these therapies have demonstrated limited therapeutic efficacy in human clinical trials. Thus, although survival of patients with GBM continues to slowly improve, treatment of GBM remains extremely challenging. CONCLUSION Continued research and development of targeted therapies, based on a detailed understanding of molecular pathogenesis can reasonably be expected to yield improved outcomes for patients with GBM.
Collapse
Affiliation(s)
- Taylor A Wilson
- Department of Neurosurgery, Division of Oncology, New York University School of Medicine, NY, USA
| | - Matthias A Karajannis
- Department of Pediatrics, Division of Oncology, New York University School of Medicine, NY, USA
| | - David H Harter
- Department of Neurosurgery, Division of Oncology, New York University School of Medicine, NY, USA
| |
Collapse
|
32
|
Zhao F, Tian J, An L, Yang K. Prognostic utility of gene therapy with herpes simplex virus thymidine kinase for patients with high-grade malignant gliomas: a systematic review and meta analysis. J Neurooncol 2014; 118:239-246. [PMID: 24756350 DOI: 10.1007/s11060-014-1444-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 04/09/2014] [Indexed: 11/24/2022]
Abstract
The aim of this study was to assess the effectiveness of adding viral vector-mediated gene therapy with herpes simplex virus thymidine kinase (HSV-tk) to standard treatment, in comparison with standard treatment alone to treat patients with high-grade gliomas (HGGs). A literature search of the databases PubMed, Embase, the Cochrane Library, Web of Science, and Chinese biomedicine was performed to identify eligible studies. Three randomized controlled trials (involving a total of 532 patients) were included in this systematic review. A meta-analysis of included studies demonstrated a significant increase in median survival time (MST) in patients who were treated with HSV-tk gene therapy (mean deviation 0.59, 95% CI: 0.41-0.76, p < 0.0001). The results of pooled analysis for different patient groups show that overall survival (OS) for all HGG patients was improved by adding gene therapy [hazard ratio (HR) = 0.91, 95% CI: 0.74-1.13, p = 0.42], while a different result was seen for glioblastoma multiforme (GBM) patients (HR = 1.06, 95% CI: 0.80-1.41, p = 0.70). Furthermore, the combined results for tumor progression implied that standard therapy was superior to gene therapy [odds ratio (OR) = 1.31, p = 0.09]; yet differences in HR and OR between experimental groups and control groups had no statistical significance (p > 0.05). Based on the best available evidence, it appears that adding gene therapy with HSV-tk has some effect in treating HGG patients, especially with respect to MST. However, neither the pooled analysis of OS, nor the combined analysis of tumor progress indicates any significant advantage to adding gene therapy compared with standard treatment alone. More prospective studies are needed to draw solid conclusions about whether gene therapy has significant prognostic advantage.
Collapse
Affiliation(s)
- Fei Zhao
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Institution of Integrated Traditional Chinese with Western Medicine, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China.,School of Medicine, Northwest University of Nationality, Lanzhou, China
| | - Jinhui Tian
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| | - Lifeng An
- School of Medicine, Northwest University of Nationality, Lanzhou, China
| | - Kehu Yang
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China. .,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China.
| |
Collapse
|
33
|
Abstract
Despite extensive research, current glioma therapies are still unsatisfactory, and novel approaches are pressingly needed. In recent years, both nonreplicative viral vectors and replicating oncolytic viruses have been developed for brain cancer treatment, and the mechanistic background of their cytotoxicity has been unveiled. A growing number of clinical trials have convincingly established viral therapies to be safe in glioma patients, and maximum tolerated doses have generally not been reached. However, evidence for therapeutic benefit has been limited: new generations of therapeutic vectors need to be developed in order to target not only tumor cells but also the complex surrounding microenvironment. Such therapies could also direct long-lasting immune responses toward the tumor while reducing early antiviral reactions. Furthermore, viral delivery methods are to be improved and viral spread within the tumor will have to be enhanced. Here, we will review the outcome of completed glioma virus therapy trials as well as highlight the ongoing clinical activities. On this basis, we will give an overview of the numerous strategies to enhance therapeutic efficacy of new-generation viruses and novel treatment regimens. Finally, we will conclude with approaches that may be crucial to the development of successful glioma therapies in the future.
Collapse
Affiliation(s)
| | - E. Antonio Chiocca
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
34
|
Abstract
Glioblastoma multiforme represents the most common primary malignant tumor of the adult CNS. Unfortunately, the median survival after surgical intervention alone is less than 6 months and the addition of radiotherapy can extend this time to only 9 months. Consequently, efforts aimed at developing new therapies have focused on new treatment strategies that specifically target tumor cells and spare normal cells. One such modality, gene therapy, has shown promise in the spectrum of agents utilized against brain tumors. This review highlights the principles of gene therapy and discusses the results of recent clinical trials in which gene therapy has been employed against malignant brain tumors.
Collapse
Affiliation(s)
- Maciej S Lesniak
- Division of Neurological Surgery, The University of Chicago, Pritzker School of Medicine, 5841 S. Maryland Avenue, MC3026, Chicago, IL 60637, USA.
| |
Collapse
|
35
|
Hagiwara K, Kishimoto S, Ishihara M, Koyama Y, Mazda O, Sato T. In vivo gene transfer using pDNA/chitosan/chondroitin sulfate ternary complexes: influence of chondroitin sulfate on the stability of freeze-dried complexes and transgene expression in vivo. J Gene Med 2013; 15:83-92. [PMID: 23307647 DOI: 10.1002/jgm.2694] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 12/30/2012] [Accepted: 01/02/2013] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Chitosan has been investigated as a promising nonviral vector. However, several problems still remain, such as a relatively low transfection efficiency and instability under physiological conditions. We previously demonstrated that a chondroitin sulfate (CS) coating enhanced the transfection efficiency and physicochemical stability of plasmid DNA (pDNA)/chitosan complexes in vitro. In the present study, the effects of coating pDNA/chitosan complexes with CS on the stability in freeze-dry rehydration processes and gene expression in vivo were investigated. METHODS Freeze-drying storage at -20 °C, 4 °C, or room temperature, freezing storage at -20 °C, or liquid storage at 4 °C or room temperature, were examined for preservation conditions of pDNA/chitosan/CS ternary complexes by a gel retardation assay, measurements of sizes and zeta potentials, and a luciferase assay. Moreover, to determine the transfection efficiency of the ternary complexes in vivo, suicide gene therapy was carried out in Huh-7-implanted mice using herpes simplex virus thymidine kinase coding pDNA and ganciclovir. RESULTS The freeze-dried pDNA/chitosan/CS ternary complexes showed sufficient cell transfection ability in vitro and in vivo. In addition, ternary complexes were associated with a significant suppression of tumor growth and a histopathologically high anti-tumor effect by intratumoral injection to tumor-bearing mice. CONCLUSIONS The CS coating enhanced the preservation stability of the pDNA/chitosan complexes after freeze-drying-rehydration and their transgene expression in vivo.
Collapse
Affiliation(s)
- Kenji Hagiwara
- Department of Biosciences and Informatics, Keio University, Yokohama, Kanagawa, Japan
| | | | | | | | | | | |
Collapse
|
36
|
Wang K, Park JO, Zhang M. Treatment of glioblastoma multiforme using a combination of small interfering RNA targeting epidermal growth factor receptor and β-catenin. J Gene Med 2013; 15:42-50. [PMID: 23319157 DOI: 10.1002/jgm.2693] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 12/15/2012] [Accepted: 12/29/2012] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) and β-catenin are two key mediators of cell signal transduction implicated in the pathogenesis of a variety of tumors. There is emerging evidence indicating that they are overexpressed in glioblastoma multiforme (GBM) and both play significant roles in GBM carcinogenesis. Moreover, down-regulating EGFR individually only provides limited therapeutic efficacy. Therefore, we aimed to determine the feasibility and efficacy of gene therapy of GBM using combinatorial inhibition of EGFR and β-catenin in view of the cross-talk between these two signaling pathways. METHODS The down-regulatory effect of small interfering RNA (siRNA) targeting EGFR and β-catenin alone or in combination in human GBM cells U-87 MG was evaluated by Quantitative RT-PCR. Cell proliferation in the short- and long-term was investigated by alamar blue and clonogenic assays, respectively. An annexin-V assay was performed to detect apoptosis caused by siRNA treatment. The effect of downregulating EGFR and β-catenin on cell cycle progression, cell migration and invasive potential were also examined. RESULTS The siRNA treatment potently reduced gene expression of EGFR and β-catenin at the mRNA level. Simultaneous inhibition of EGFR and β-catenin greatly decreased GBM cell proliferation. Although no significant increase in apoptosis was demonstrated, combinatorial siRNA treatment delayed the progression of cell cycle with an increased proportion of cells arrested in the G0/1 phase. Furthermore, EGFR and β-catenin siRNA in combination significantly inhibited the migratory and invasive ability of GBM cells. CONCLUSIONS Simultaneous inhibition of EGFR and β-catenin expression could represent an effective therapy for human GBM, and warrants further study in vivo.
Collapse
Affiliation(s)
- Kui Wang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | | | | |
Collapse
|
37
|
Merienne N, Le Douce J, Faivre E, Déglon N, Bonvento G. Efficient gene delivery and selective transduction of astrocytes in the mammalian brain using viral vectors. Front Cell Neurosci 2013; 7:106. [PMID: 23847471 PMCID: PMC3701857 DOI: 10.3389/fncel.2013.00106] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 06/18/2013] [Indexed: 12/31/2022] Open
Abstract
Astrocytes are now considered as key players in brain information processing because of their newly discovered roles in synapse formation and plasticity, energy metabolism and blood flow regulation. However, our understanding of astrocyte function is still fragmented compared to other brain cell types. A better appreciation of the biology of astrocytes requires the development of tools to generate animal models in which astrocyte-specific proteins and pathways can be manipulated. In addition, it is becoming increasingly evident that astrocytes are also important players in many neurological disorders. Targeted modulation of protein expression in astrocytes would be critical for the development of new therapeutic strategies. Gene transfer is valuable to target a subpopulation of cells and explore their function in experimental models. In particular, viral-mediated gene transfer provides a rapid, highly flexible and cost-effective, in vivo paradigm to study the impact of genes of interest during central nervous system development or in adult animals. We will review the different strategies that led to the recent development of efficient viral vectors that can be successfully used to selectively transduce astrocytes in the mammalian brain.
Collapse
Affiliation(s)
- Nicolas Merienne
- Laboratory of Cellular and Molecular Neurotherapies, Department of Clinical Neurosciences, Lausanne University Hospital Lausanne, Switzerland
| | | | | | | | | |
Collapse
|
38
|
Current status of local therapy in malignant gliomas--a clinical review of three selected approaches. Pharmacol Ther 2013; 139:341-58. [PMID: 23694764 DOI: 10.1016/j.pharmthera.2013.05.003] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 05/12/2013] [Indexed: 12/21/2022]
Abstract
Malignant gliomas are the most frequently occurring, devastating primary brain tumors, and are coupled with a poor survival rate. Despite the fact that complete neurosurgical resection of these tumors is impossible in consideration of their infiltrating nature, surgical resection followed by adjuvant therapeutics, including radiation therapy and chemotherapy, is still the current standard therapy. Systemic chemotherapy is restricted by the blood-brain barrier, while methods of local delivery, such as with drug-impregnated wafers, convection-enhanced drug delivery, or direct perilesional injections, present attractive ways to circumvent these barriers. These methods are promising ways for direct delivery of either standard chemotherapeutic or new anti-cancer agents. Several clinical trials showed controversial results relating to the influence of a local delivery of chemotherapy on the survival of patients with both recurrent and newly diagnosed malignant gliomas. Our article will review the development of the drug-impregnated release, as well as convection-enhanced delivery and the direct injection into brain tissue, which has been used predominantly in gene-therapy trials. Further, it will focus on the use of convection-enhanced delivery in the treatment of patients with malignant gliomas, placing special emphasis on potential shortcomings in past clinical trials. Although there is a strong need for new or additional therapeutic strategies in the treatment of malignant gliomas, and although local delivery of chemotherapy in those tumors might be a powerful tool, local therapy is used only sporadically nowadays. Thus, we have to learn from our mistakes in the past and we strongly encourage future developments in this field.
Collapse
|
39
|
Simonato M, Bennett J, Boulis NM, Castro MG, Fink DJ, Goins WF, Gray SJ, Lowenstein PR, Vandenberghe LH, Wilson TJ, Wolfe JH, Glorioso JC. Progress in gene therapy for neurological disorders. Nat Rev Neurol 2013; 9:277-91. [PMID: 23609618 PMCID: PMC3908892 DOI: 10.1038/nrneurol.2013.56] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Diseases of the nervous system have devastating effects and are widely distributed among the population, being especially prevalent in the elderly. These diseases are often caused by inherited genetic mutations that result in abnormal nervous system development, neurodegeneration, or impaired neuronal function. Other causes of neurological diseases include genetic and epigenetic changes induced by environmental insults, injury, disease-related events or inflammatory processes. Standard medical and surgical practice has not proved effective in curing or treating these diseases, and appropriate pharmaceuticals do not exist or are insufficient to slow disease progression. Gene therapy is emerging as a powerful approach with potential to treat and even cure some of the most common diseases of the nervous system. Gene therapy for neurological diseases has been made possible through progress in understanding the underlying disease mechanisms, particularly those involving sensory neurons, and also by improvement of gene vector design, therapeutic gene selection, and methods of delivery. Progress in the field has renewed our optimism for gene therapy as a treatment modality that can be used by neurologists, ophthalmologists and neurosurgeons. In this Review, we describe the promising gene therapy strategies that have the potential to treat patients with neurological diseases and discuss prospects for future development of gene therapy.
Collapse
Affiliation(s)
- Michele Simonato
- Section of Pharmacology, Department of Medical Sciences, University of Ferrara, Fossato di Mortara 17-19, 44121 Ferrara, Italy. michele.simonato@ unife.it
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Potts MB, Silvestrini MT, Lim DA. Devices for cell transplantation into the central nervous system: Design considerations and emerging technologies. Surg Neurol Int 2013; 4:S22-30. [PMID: 23653887 PMCID: PMC3642746 DOI: 10.4103/2152-7806.109190] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 11/04/2012] [Indexed: 12/14/2022] Open
Abstract
Successful use of cell-based therapies for the treatment of neurological diseases is dependent upon effective delivery to the central nervous system (CNS). The CNS poses several challenges to the delivery of cell-based therapeutics, including the blood-brain barrier, anatomic complexity, and regional specificity. Targeted delivery methods are therefore required for the selective treatment of specific CNS regions. In addition, CNS tissues are mechanically and physiologically delicate and even minor injury to normal brain or spinal cord can cause devastating neurological deficits. Targeted delivery methods must therefore minimize tissue trauma. At present, direct injection into brain or spinal cord parenchyma promises to be the most versatile and accurate method of targeted CNS therapeutic delivery. While direct injection methods have already been employed in clinical trials of cell transplantation for a wide variety of neurological diseases, there are many shortcomings with the devices and surgical approaches currently used. Some of these technical limitations may hinder the clinical development of cell transplantation therapies despite validity of the underlying biological mechanisms. In this review, we discuss some of the important technical considerations of CNS injection devices such as targeting accuracy, distribution of infused therapeutic, and overall safety to the patient. We also introduce and discuss an emerging technology - radially branched deployment - that may improve our ability to safely distribute cell-based therapies and other therapeutic agents to the CNS. Finally, we speculate on future technological developments that may further enhance the efficacy of CNS therapeutic delivery.
Collapse
Affiliation(s)
- Matthew B Potts
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | | | | |
Collapse
|
41
|
Gene therapy with HSV1-sr39TK/GCV exhibits a stronger therapeutic efficacy than HSV1-TK/GCV in rat C6 glioma cells. ScientificWorldJournal 2013; 2013:951343. [PMID: 23533367 PMCID: PMC3603674 DOI: 10.1155/2013/951343] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2013] [Accepted: 01/29/2013] [Indexed: 11/18/2022] Open
Abstract
Although the combination of herpes simplex virus type 1 (HSV-1) thymidine kinase (TK) with ganciclovir (GCV) has been shown as a promising suicide gene treatment strategy for glioma, the almost immunodepressive dose of GCV required for its adequate in vivo efficacy has hampered its further clinical application. Therefore, In order to reduce the GCV dose required, we aim to compare the therapeutic efficacy of HSV1-sr39TK, an HSV1-TK mutant with increased GCV prodrug catalytic activity, with wildtype TK in C6 glioma cells. Accordingly, rat C6 glioma cells were first transfected with pCDNA-TK and pCDNA-sr39TK, respectively, and the gene transfection efficacy was verified by immunocytochemistry and western blot analysis. Then the in vivo sensitivity of these transfected C6-TK and C6-sr39TK cells to GCV was determined by 3-(4,5)-dimethylthiahiazo-(-z-y1)-3,5-di-phenytetrazoliumromide (MTT) colorimetric assay and Hoechst-propidium iodide (PI) staining. Finally, a subcutaneously C6 xenograft tumor model was established in the nude mice to test the in vitro efficacy of TK/GCV gene therapy. Our results showed that, as compared with wildtype TK, HSV1-sr39TK/GCV demonstrated a stronger therapeutic efficacy against C6 glioma both in vitro and in vivo, which, by reducing the required GCV dose, might warrant its future use in the treatment of glioma under clinical setting.
Collapse
|
42
|
Tobias A, Ahmed A, Moon KS, Lesniak MS. The art of gene therapy for glioma: a review of the challenging road to the bedside. J Neurol Neurosurg Psychiatry 2013; 84:213-22. [PMID: 22993449 PMCID: PMC3543505 DOI: 10.1136/jnnp-2012-302946] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Glioblastoma multiforme (GBM) is a highly invasive brain tumour that is unvaryingly fatal in humans despite even aggressive therapeutic approaches such as surgical resection followed by chemotherapy and radiotherapy. Unconventional treatment options such as gene therapy provide an intriguing option for curbing glioma related deaths. To date, gene therapy has yielded encouraging results in preclinical animal models as well as promising safety profiles in phase I clinical trials, but has failed to demonstrate significant therapeutic efficacy in phase III clinical trials. The most widely studied antiglioma gene therapy strategies are suicide gene therapy, genetic immunotherapy and oncolytic virotherapy, and we have attributed the challenging transition of these modalities into the clinic to four major roadblocks: (1) anatomical features of the central nervous system, (2) the host immune system, (3) heterogeneity and invasiveness of GBM and (4) limitations in current GBM animal models. In this review, we discuss possible ways to jump these hurdles and develop new gene therapies that may be used alone or in synergy with other modalities to provide a powerful treatment option for patients with GBM.
Collapse
Affiliation(s)
- Alex Tobias
- Brain Tumour Center, The University of Chicago, 5841 South Maryland Ave, MC 3026, Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
43
|
Abstract
Progress in improving the prognosis of patients with glioblastoma has been modest and has predominantly relied on informative imaging, optimization of medical and surgical treatment, and approval of new drugs with modest benefits on overall and/or progression-free survival. This has frustrated clinicians and demoralized patients but has underscored the importance of pursuing novel treatment strategies in hopes of mounting a decisive assault on this disease. Although initially not intuitive, the use of a pathogen to treat cancer has become a radical and sophisticated strategy to combat the aggressive phenotype of this disease. In fact, the engineering of viruses to fight cancer is a field that has now reached scientific maturity and has rapidly progressed from preclinical stages to clinical testing with considerable safety but disappointing efficacy. Here we review the milestones of this therapy focusing on landmark clinical trials, shed light on the limitations of this approach, and describe the recent and future strategies aimed at bringing promising efficacy to this mode of therapy.
Collapse
|
44
|
Larkin J, Goh XY, Vetter M, Pickering L, Swanton C. Epigenetic regulation in RCC: opportunities for therapeutic intervention? Nat Rev Urol 2012; 9:147-55. [DOI: 10.1038/nrurol.2011.236] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
45
|
Gomez-Manzano C, Jiang H, Alonso M, Yung WKA, Fueyo J. Gene therapy. HANDBOOK OF CLINICAL NEUROLOGY 2012; 104:331-8. [PMID: 22230451 DOI: 10.1016/b978-0-444-52138-5.00021-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Candelaria Gomez-Manzano
- Department of Neuro-oncology, The University of Texas, M. D Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | |
Collapse
|
46
|
Rueger MA, Ameli M, Li H, Winkeler A, Rueckriem B, Vollmar S, Galldiks N, Hesselmann V, Fraefel C, Wienhard K, Heiss WD, Jacobs AH. [18F]FLT PET for non-invasive monitoring of early response to gene therapy in experimental gliomas. Mol Imaging Biol 2011; 13:547-557. [PMID: 20563754 DOI: 10.1007/s11307-010-0361-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The purpose of this study was to investigate the potential of 3'-deoxy-3'-[¹⁸F]fluorothymidine ([¹⁸F]FLT) positron emission tomography (PET) to detect early treatment responses in gliomas. Human glioma cells were stably transduced with genes yielding therapeutic activity, sorted for different levels of exogenous gene expression, and implanted subcutaneously into nude mice. Multimodality imaging during prodrug therapy included (a) magnetic resonance imaging, (b) PET with 9-(4-[¹⁸F]fluoro-3-hydroxymethylbutyl)guanine assessing exogenous gene expression, and (c) repeat [¹⁸F]FLT PET assessing antiproliferative therapeutic response. All stably transduced gliomas responded to therapy with significant reduction in tumor volume and [¹⁸F]FLT accumulation within 3 days after initiation of therapy. The change in [¹⁸F]FLT uptake before and after treatment correlated to volumetrically calculated growth rates. Therapeutic efficacy as monitored by [¹⁸F]FLT PET correlated to levels of therapeutic gene expression measured in vivo. Thus, [¹⁸F]FLT PET assesses early antiproliferative effects, making it a promising radiotracer for the development of novel treatments for glioma.
Collapse
Affiliation(s)
- Maria A Rueger
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany.,Center for Molecular Medicine (CMMC), Cologne, Germany.,Departments of Neurology, University Hospital Cologne, Cologne, Germany
| | - Mitra Ameli
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany.,Departments of Neurology, University Hospital Cologne, Cologne, Germany
| | - Hongfeng Li
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany
| | - Alexandra Winkeler
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany.,Center for Molecular Medicine (CMMC), Cologne, Germany
| | | | - Stefan Vollmar
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany
| | - Norbert Galldiks
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany
| | - Volker Hesselmann
- Department of Radiology, University Hospital Cologne, Cologne, Germany
| | - Cornel Fraefel
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany
| | - Klaus Wienhard
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany
| | - Wolf-Dieter Heiss
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany
| | - Andreas H Jacobs
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany. .,Center for Molecular Medicine (CMMC), Cologne, Germany. .,European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany.
| |
Collapse
|
47
|
Castro MG, Candolfi M, Kroeger K, King GD, Curtin JF, Yagiz K, Mineharu Y, Assi H, Wibowo M, Ghulam Muhammad AKM, Foulad D, Puntel M, Lowenstein PR. Gene therapy and targeted toxins for glioma. Curr Gene Ther 2011; 11:155-80. [PMID: 21453286 DOI: 10.2174/156652311795684722] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 03/08/2011] [Indexed: 12/12/2022]
Abstract
The most common primary brain tumor in adults is glioblastoma. These tumors are highly invasive and aggressive with a mean survival time of 15-18 months from diagnosis to death. Current treatment modalities are unable to significantly prolong survival in patients diagnosed with glioblastoma. As such, glioma is an attractive target for developing novel therapeutic approaches utilizing gene therapy. This review will examine the available preclinical models for glioma including xenographs, syngeneic and genetic models. Several promising therapeutic targets are currently being pursued in pre-clinical investigations. These targets will be reviewed by mechanism of action, i.e., conditional cytotoxic, targeted toxins, oncolytic viruses, tumor suppressors/oncogenes, and immune stimulatory approaches. Preclinical gene therapy paradigms aim to determine which strategies will provide rapid tumor regression and long-term protection from recurrence. While a wide range of potential targets are being investigated preclinically, only the most efficacious are further transitioned into clinical trial paradigms. Clinical trials reported to date are summarized including results from conditionally cytotoxic, targeted toxins, oncolytic viruses and oncogene targeting approaches. Clinical trial results have not been as robust as preclinical models predicted; this could be due to the limitations of the GBM models employed. Once this is addressed, and we develop effective gene therapies in models that better replicate the clinical scenario, gene therapy will provide a powerful approach to treat and manage brain tumors.
Collapse
Affiliation(s)
- Maria G Castro
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Liu T, Ye L, He Y, Chen X, Peng J, Zhang X, Yi H, Peng F, Leng A. Combination gene therapy using VEGF-shRNA and fusion suicide gene yCDglyTK inhibits gastric carcinoma growth. Exp Mol Pathol 2011; 91:745-52. [PMID: 21840308 DOI: 10.1016/j.yexmp.2011.07.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2011] [Accepted: 07/29/2011] [Indexed: 01/15/2023]
Abstract
Clinical trials of suicide gene therapy have achieved limited success, which suggests a need for improvement. Angiogenesis plays a crucial role in the progression of cancers, which is greatly regulated by vascular endothelial growth factor (VEGF).The current study was designed to evaluate the anti-tumor effects of VEGF siRNA in combination with fusion suicide gene yCDglyTK. Introduction of a VEGF-targeted small hairpin RNA (shVEGF) to CDTK/5-FC system could induce cell apoptosis more effectively and decrease micro vessel density in xenograft tissue, thus resulted in a significant tumor growth delay in SGC7901 xenografts. These findings for the first time suggest the potential of combination gene therapy using suicide gene therapy and anti-angiogenesis gene therapy.
Collapse
Affiliation(s)
- Ting Liu
- Department of Gastroenterology, Xiangya Hospital of Central South University, Changsha, Hunan Province, 410008, China
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Soft tissue sarcomas comprise a heterogeneous group of aggressive tumors that have a relatively poor prognosis. Although conventional therapeutic regimens can effectively cytoreduce the overall tumor mass, they fail to consistently achieve a curative outcome. Alternative gene-based approaches that counteract the underlying neoplastic process by eliminating the clonal aberrations that potentiate malignant behavior have been proposed. As compared to the accumulation of gene alterations associated with epithelial carcinomas, sarcomas are frequently characterized by the unique presence of a single chromosomal translocation in each histological subtype. Similar to the Philadelphia chromosome associated with CML, these clonal abnormalities result in the fusion of two independent unrelated genes to generate a unique chimeric protein that displays aberrant activity believed to initiate cellular transformation. Secondary gene mutations may provide an additional growth advantage that further contributes to malignant progression. The recent clinical success of the tyrosine kinase inhibitor, STI571, suggests that therapeutic approaches specifically directed against essential survival factors in sarcoma cells may be effective. This review summarizes published approaches targeting a specific molecular mechanism associated with sarcomagenesis. The strategy and significance of published translational studies in six distinct areas are presented. These include: (1) the disruption of chimeric transcription factor activity; (2) inhibition of growth stimulatory post-translational modifications; (3) restoration of tumor suppressor function; (4) interference with angiogenesis; (5) induction of apoptotic pathways; and (6) introduction of toxic gene products. The potential for improving outcomes in sarcoma patients and the conceptual obstacles to be overcome are discussed.
Collapse
Affiliation(s)
- R J Olsen
- Department of Pathology and Microbiology University of Nebraska Medical Center Omaha NE 69198-6495 USA
| | | | | |
Collapse
|
50
|
Zheng X, Jiang F, Katakowski M, Lu Y, Chopp M. ADAM17 promotes glioma cell malignant phenotype. Mol Carcinog 2011; 51:150-64. [PMID: 21480393 DOI: 10.1002/mc.20772] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 02/04/2011] [Accepted: 03/01/2011] [Indexed: 01/06/2023]
Abstract
A disintegrin and metalloproteinase-17 (ADAM17) is involved in proteolytic ectodomain shedding of several membrane-bound growth factors and cytokines. The expression and activity of ADAM17 increase under some pathological conditions such as stroke and cancer. ADAM17 promotes neural progenitor cell migration and contributes to neurogenesis after stroke and breast cancer growth and invasion. In the present study, we sought to elucidate whether ADAM17 contributes to glioma progression. To this end, we examined the role of ADAM17 in the proliferation, invasion, and tube formation of U87 human glioma cells in vitro and tumor growth in vivo. Stable transfection of the U87 cell line with either a plasmid for over-expression of human ADAM17, or a siRNA to ADAM17 was employed in this study to establish high- or low-ADAM17 expression in glioma cells, respectively. For study of mechanism, the ADAM17 inhibitor TAPI-2 and the PI3K-AKT inhibitor LY294002 were used to counteract high-ADAM17 expression and the activated PI3K-AKT pathway, respectively. Proliferation of glioma cells were tested by thiazolyl blue tetrazolium bromide (MTT) assay, bromodeoxyuridine incorporation assay, growth curve, and sulforhodamine B assay. Matrigel invasion assays were used to assess the ability of U87 cells to penetrate the extracellular matrix (ECM). A Matrigel tube formation assay was performed to test capillary tube formation ability. EGFR-PI3K-Akt pathway activation in U87 cells under different ADAM17 expression levels were tested by Western blot. Our data show that ADAM17 promotes the U87 malignant phenotype by increased proliferation, invasion, angiogenesis, and in vivo tumor growth. Tumor growth in nude mice was significantly inhibited by ADAM17 inhibitor and A17-shRNA in vivo transfection. TGF-α, VEGF secretion, and VEGF expression was increased by ADAM17 and counteracted by ADAM17 siRNA, TAPI-2, and LY294002 in U87 cells. ADAM17 activated, whereas ADAM17 siRNA, TAPI-2, and LY294002 deactivated the EGFR-PI3K-AKT signal pathway, which correlated with U87 cell malignant phenotype changes. This study suggests ADAM17 contributes to glioma progression through activation of the EGFR-PI3K-AKT signal pathway.
Collapse
Affiliation(s)
- Xuguang Zheng
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan 48202, USA
| | | | | | | | | |
Collapse
|