1
|
Bhatt DK, Wekema L, Carvalho Barros LR, Chammas R, Daemen T. A systematic analysis on the clinical safety and efficacy of onco-virotherapy. MOLECULAR THERAPY-ONCOLYTICS 2021; 23:239-253. [PMID: 34761104 PMCID: PMC8551473 DOI: 10.1016/j.omto.2021.09.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/13/2021] [Accepted: 09/29/2021] [Indexed: 12/12/2022]
Abstract
Several onco-virotherapy candidates have been developed and clinically evaluated for the treatment of cancer, and several are approved for clinical use. In this systematic review we explored the clinical impact of onco-virotherapy compared to other cancer therapies by analyzing factors such as trial design, patient background, therapy design, delivery strategies, and study outcomes. For this purpose, we retrieved clinical studies from three platforms: ClinicalTrials.gov, PubMed, and EMBASE. We found that most studies were performed in patients with advanced and metastatic tumors, using a broad range of genetically engineered vectors and mainly administered intratumorally. Therapeutic safety was the most frequently assessed outcome, while relatively few studies focused on immunological antitumor responses. Moreover, only 59 out of 896 clinical studies were randomized controlled trials reporting comparative data. This systemic review thus reveals the need of more, and better controlled, clinical studies to increase our understanding on the application of onco-virotherapy either as a single treatment or in combination with other cancer immunotherapies.
Collapse
Affiliation(s)
- Darshak K Bhatt
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands.,Center for Translational Research in Oncology, Instituto do Câncer do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, CEP 01246-000, Brazil
| | - Lieske Wekema
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Luciana Rodrigues Carvalho Barros
- Center for Translational Research in Oncology, Instituto do Câncer do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, CEP 01246-000, Brazil
| | - Roger Chammas
- Center for Translational Research in Oncology, Instituto do Câncer do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, CEP 01246-000, Brazil
| | - Toos Daemen
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| |
Collapse
|
2
|
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: 4.7] [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
|
3
|
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: 0.2] [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
|
4
|
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: 1.3] [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
|
5
|
Kolberg M, Høland M, Lind GE, Ågesen TH, Skotheim RI, Hall KS, Mandahl N, Smeland S, Mertens F, Davidson B, Lothe RA. Protein expression of BIRC5, TK1, and TOP2A in malignant peripheral nerve sheath tumours--A prognostic test after surgical resection. Mol Oncol 2015; 9:1129-39. [PMID: 25769404 PMCID: PMC5528761 DOI: 10.1016/j.molonc.2015.02.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 01/22/2015] [Accepted: 02/10/2015] [Indexed: 11/26/2022] Open
Abstract
No consensus treatment regime exists beyond surgery for malignant peripheral nerve sheath tumours (MPNST), and the purpose of the present study was to find new approaches to stratify patients with good and poor prognosis and to better guide therapeutic intervention for this aggressive soft tissue cancer. From a total of 67 MPNSTs from Scandinavian patients with and without neurofibromatosis type 1, 30 MPNSTs were investigated by genome‐wide RNA expression profiling and 63 MPNSTs by immunohistochemical (IHC) analysis, and selected genes were submitted to analyses of disease‐specific survival. The potential drug target genes survivin (BIRC5), thymidine kinase 1 (TK1), and topoisomerase 2‐alpha (TOP2A), all encoded on chromosome arm 17q, were up‐regulated in MPNST as compared to benign neurofibromas. Each of them was found to be independent prognostic markers on the gene expression level, as well as on the protein level. A prognostic profile was identified by combining the nuclear expression scores of the three proteins. For patients with completely resected tumours only 15% in the high risk group were alive after two years, as compared to 78% in the low risk group. In conclusion, we found a novel protein expression profile which identifies MPNST patients with inferior prognosis even after assumed curative surgery. The tested proteins are drug targets; therefore the expression profile may provide predictive information guiding the design of future clinical trials. Importantly, as the effect is seen on the protein level using IHC, the biomarker panel can be readily implemented in routine clinical testing.
Collapse
Affiliation(s)
- Matthias Kolberg
- Department of Molecular Oncology, Institute for Cancer Research, Division of Cancer Medicine Surgery and Transplantation, Oslo University Hospital, Oslo, Norway; Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Maren Høland
- Department of Molecular Oncology, Institute for Cancer Research, Division of Cancer Medicine Surgery and Transplantation, Oslo University Hospital, Oslo, Norway; Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Guro E Lind
- Department of Molecular Oncology, Institute for Cancer Research, Division of Cancer Medicine Surgery and Transplantation, Oslo University Hospital, Oslo, Norway; Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Trude H Ågesen
- Department of Molecular Oncology, Institute for Cancer Research, Division of Cancer Medicine Surgery and Transplantation, Oslo University Hospital, Oslo, Norway; Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Rolf I Skotheim
- Department of Molecular Oncology, Institute for Cancer Research, Division of Cancer Medicine Surgery and Transplantation, Oslo University Hospital, Oslo, Norway; Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kirsten Sundby Hall
- Department of Oncology, Division of Cancer Medicine Surgery and Transplantation, Oslo University Hospital, Oslo, Norway
| | - Nils Mandahl
- Department of Clinical Genetics, Skåne University Hospital, Lund, Sweden
| | - Sigbjørn Smeland
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Oncology, Division of Cancer Medicine Surgery and Transplantation, Oslo University Hospital, Oslo, Norway
| | - Fredrik Mertens
- Department of Clinical Genetics, Skåne University Hospital, Lund, Sweden
| | - Ben Davidson
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Pathology, Division of Diagnostics and Intervention, Oslo University Hospital, Oslo, Norway
| | - Ragnhild A Lothe
- Department of Molecular Oncology, Institute for Cancer Research, Division of Cancer Medicine Surgery and Transplantation, Oslo University Hospital, Oslo, Norway; Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
6
|
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
|
7
|
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: 3.2] [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
|
8
|
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
|
9
|
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: 64] [Impact Index Per Article: 4.6] [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
|
10
|
van Putten EH, Dirven CM, van den Bent MJ, Lamfers ML. Sitimagene ceradenovec: a gene-based drug for the treatment of operable high-grade glioma. Future Oncol 2011; 6:1691-710. [PMID: 21142657 DOI: 10.2217/fon.10.134] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The field of gene therapy for malignant glioma has made important advances since the first gene transfer studies were performed 20 years ago. Multiple Phase I/II trials and two Phase III trials have been performed and have demonstrated the feasibility and safety of intratumoral vector delivery in the brain. Sitimagene ceradenovec is an adenoviral vector encoding the herpes simplex thymidine kinase gene, developed by Ark Therapeutics Group plc (UK and Finland) for the treatment of patients with operable high-grade glioma. In preclinical and Phase I/II clinical studies, sitimagene ceradenovec exhibited a significant increase in survival. Although the preliminary results of a Phase III clinical study demonstrated a significant positive effect of sitimagene ceradenovec treatment on time to reintervention or death when compared with standard care treatment (hazard ratio: 1.43; 95% CI: 1.06-1.93; p < 0.05), the European Committee for Medicinal Products for Human Use did not consider the data to provide sufficient evidence of clinical benefit. Further clinical evaluation, powered to demonstrate a benefit on a robust end point, is required. This article focuses on sitimagene ceradenovec and provides an overview of the developments in the field of gene therapy for malignant glioma.
Collapse
Affiliation(s)
- Erik Hp van Putten
- Department of Neurosurgery, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands.
| | | | | | | |
Collapse
|
11
|
|
12
|
Asadi-Moghaddam K, Chiocca EA. Gene- and viral-based therapies for brain tumors. Neurotherapeutics 2009; 6:547-57. [PMID: 19560744 PMCID: PMC3052738 DOI: 10.1016/j.nurt.2009.04.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2009] [Revised: 04/15/2009] [Accepted: 04/17/2009] [Indexed: 01/21/2023] Open
Abstract
Advances in understanding and controlling genes and their expression have set the stage to alter genetic material to fight or prevent disease with brain tumors being among one of the first human malignancies to be targeted by gene therapy. All proteins are coded for by DNA and most neoplastic diseases ultimately result from the expression or lack thereof with one or more proteins (e.g., coded by oncogenes or tumor suppressor genes, respectively). In theory, therefore, diseases could be treated by expression of the appropriate protein in the affected cells. Gene therapy is an experimental treatment that involves introducing genetic material (DNA or RNA) into cells, and it has made important advances in the past decade. Within this short time span, it has moved from the conceptual laboratory research stage to clinical translational trials for brain tumors. The most efficient approaches for gene delivery are based on viral vectors, which have been proven relatively safe in the CNS, despite occasional cases of morbidity and death in non-neurosurgical trials. However, the human response to various viral vectors can not be predicted in a reliable manner from animal experimentation, nor can size, consistency, and extent of experimental brain tumors in mouse models reflect the large, necrotic, infiltrative nature of malignant gliomas. Furthermore, the problem of delivering genetic vectors into solid brain tumors and the efficiency in situ gene transfer remains one of the most significant hurdles in gene therapy.
Collapse
Affiliation(s)
- Kaveh Asadi-Moghaddam
- Department of Neurological Surgery, Dardinger Center for Neuro-oncology and Neurosciences, James Cancer Hospital/Solove Research Institute, The Ohio State University Medical Center, N-1017 Doan Hall, 410 W. 10th Avenue, 43210-1240 Columbus, OH
| | - E. Antonio Chiocca
- Department of Neurological Surgery, Dardinger Center for Neuro-oncology and Neurosciences, James Cancer Hospital/Solove Research Institute, The Ohio State University Medical Center, N-1017 Doan Hall, 410 W. 10th Avenue, 43210-1240 Columbus, OH
| |
Collapse
|
13
|
Yasuhara T, Date I. Gene therapy for Parkinson's disease. JOURNAL OF NEURAL TRANSMISSION. SUPPLEMENTUM 2009:301-309. [PMID: 20411788 DOI: 10.1007/978-3-211-92660-4_25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Parkinson's disease is characterized by the degeneration of the nigrostriatal dopaminergic neurons with the manifestation of tremor, rigidity, akinesia, and disturbances of postural reflexes. Medication using L-DOPA and surgeries including deep brain stimulation are the established therapies for Parkinson's disease. Cell therapies are also effective and have rapidly developed with the recent advancement in molecular biological technology including gene transfer. In this review, ex vivo gene therapy using genetically engineered cell transplantation for Parkinson's disease model of animals is described, including catecholamine/neurotrophic factor-secreting cell transplantation with or without encapsulation, as well as in vivo gene therapy using direct injection of viral vector to increase dopamine-production, ameliorate the survival of dopaminergic neurons, correct the deteriorated microenvironment, or normalize genetic abnormality. Furthermore, the future directions for clinical application are described together with recent clinical trials of gene therapy.
Collapse
Affiliation(s)
- Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, 2-5-1, Shikata-cho, Okayama, 700-8558, Japan.
| | | |
Collapse
|
14
|
Schenk-Braat EAM, van Mierlo MMKB, Wagemaker G, Bangma CH, Kaptein LCM. An inventory of shedding data from clinical gene therapy trials. J Gene Med 2007; 9:910-21. [PMID: 17880045 DOI: 10.1002/jgm.1096] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Viruses are the most commonly used vectors for clinical gene therapy. The risk of dissemination of a viral vector into the environment via excreta from the treated patient, a phenomenon called shedding, is a major safety concern for the environment. Despite the significant number of clinical gene therapy trials that have been conducted worldwide, there is currently no overview of actual shedding data available. In this article, an inventory of shedding data obtained from a total of 100 publications on clinical gene therapy trials using retroviral, adenoviral, adeno-associated viral and pox viral vectors is presented. In addition, the experimental set-up for shedding analysis including the assays used and biological materials tested is summarized. The collected data based on the analysis of 1619 patients in total demonstrate that shedding of these vectors occurs in practice, mainly determined by the type of vector and the route of vector administration. Due to the use of non-quantitative assays, the lack of information on assay sensitivity in most publications, and the fact that assay sensitivity is expressed in various ways, general conclusions cannot be made as to the level of vector shedding. The evaluation of the potential impact and consequences of the observations is complicated by the high degree of variety in the experimental design of shedding analysis between trials. This inventory can be supportive to clinical gene therapy investigators for the establishment of an evidence-based risk assessment to be included in a clinical protocol application, as well as to national regulatory authorities for the ongoing development of regulatory guidelines regarding gene therapy.
Collapse
|
15
|
Abstract
The advent of gene therapy in the early 1990's raised expectations for brain tumor therapies; however, whereas clinical trials in patients with malignant gliomas provided evidence of safety, therapeutic benefit was not convincing. These early forays resembled the historical introductions of other therapies that seemed promising, only to fail in human trials. Nevertheless, re-study in the laboratory and retesting in iterative laboratory-clinic processes enabled therapies with strong biological rationales to ultimately show evidence of success in humans and become accepted. Examples, such as organ transplantation, monoclonal antibody therapy and antiangiogenic therapy, provide solace that a strategy's initial lack of success in humans provides an opportunity for its further refinement in the laboratory and development of solutions that will translate into patient success stories. The authors herein summarize results from clinical trials of gene therapy for malignant gliomas, and discuss the influence of these results on present thought in preclinical research.
Collapse
Affiliation(s)
- Giulia Fulci
- Brain Tumor Research Center, Simches Research Building CRPZN-3800, Neurosurgery Service, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA.
| | | |
Collapse
|
16
|
Barzon L, Zanusso M, Colombo F, Palù G. Clinical trials of gene therapy, virotherapy, and immunotherapy for malignant gliomas. Cancer Gene Ther 2006; 13:539-54. [PMID: 16410822 DOI: 10.1038/sj.cgt.7700930] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Despite advances in surgical and adjuvant therapy, the prognosis for malignant gliomas remains dismal. This gloomy scenario has been recently brightened by the increasing understanding of the genetic and biological mechanisms at the basis of brain tumor development. These findings are being translated into innovative therapeutic approaches, including gene therapy, virotherapy, and vaccination, some of which have already been experimented in clinical trials. The advantages and disadvantages of all these different therapeutic modalities for malignant gliomas will be critically discussed, providing perspective for future investigations.
Collapse
Affiliation(s)
- L Barzon
- Department of Histology, Microbiology and Medical Biotechnologies, University of Padova, Padova, and Division of Neurosurgery, San Bortolo Hospital, Vicenza, Italy.
| | | | | | | |
Collapse
|
17
|
Pulkkanen KJ, Yla-Herttuala S. Gene therapy for malignant glioma: current clinical status. Mol Ther 2006; 12:585-98. [PMID: 16095972 DOI: 10.1016/j.ymthe.2005.07.357] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2005] [Revised: 07/06/2005] [Accepted: 07/06/2005] [Indexed: 12/24/2022] Open
Abstract
Glioblastoma is an aggressive brain tumor with a dismal prognosis. Gene therapy may offer a new option for the treatment of these patients. Several gene therapy approaches have shown anti-tumor efficiency in experimental studies, and the first clinical trials for the treatment of malignant glioma were conducted in the 1990s. HSV-tk gene therapy has been the pioneering and most commonly used approach, but oncolytic conditionally replicating adenoviruses and herpes simplex virus mutant vectors, p53, interleukins, interferons, and antisense oligonucleotides have also been used. During the past few years, adenoviruses have become the most popular gene transfer vectors, and some recent randomized, controlled trials have shown significant anti-tumor efficacy in clinical use. However, efficient gene delivery into the brain still presents a major problem, and there is a lack of definitive phase III trials, which would avoid potential problems associated with a small number of patients, inadvertent patient selection, and overinterpretation of results based on a few long-time survivors. For clinical efficacy, median survival is one of the most rigorous endpoints. It is used here to evaluate the usefulness of various treatment approaches and current clinical status of gene therapy for malignant glioma.
Collapse
Affiliation(s)
- Kalevi J Pulkkanen
- Department of Molecular Medicine, AI Virtanen Institute, University of Kuopio, Finland
| | | |
Collapse
|
18
|
King GD, Curtin JF, Candolfi M, Kroeger K, Lowenstein PR, Castro MG. Gene therapy and targeted toxins for glioma. Curr Gene Ther 2006; 5:535-57. [PMID: 16457645 PMCID: PMC1629033 DOI: 10.2174/156652305774964631] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The most common primary brain tumor in adults is glioblastoma. These tumors are highly invasive and aggressive with a mean survival time of nine to twelve 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)
- Gwendalyn D King
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | | | | | | | | | | |
Collapse
|
19
|
Abstract
The long-term survival of children with brain tumor has improved considerably in the last three decades, owing to advances in neuroimaging, neurosurgical, and radiation therapy modalities, coupled with the application of conventional chemotherapy. MRI, MR spectroscopy and diffusion-weighted MRI have contributed to more accurate diagnosis, prognostication and better treatment planning. Neurosurgical treatment has been advanced by the use of functional MRI, and intraoperative image-guided stereotactic techniques and electrophysiologic monitoring. The use of 3-D conformal and intensity-modulated radiation therapy, stereotactic radiosurgery, and radiosensitizing agents has made radiation therapy safer and more effective. Conventional chemotherapy, administered either alone or combined with radiation therapy has improved survival and quality of life of children with brain tumors. These improved outcomes have also occurred, due, in part, to their treatment on collaborative national and international studies. Recent promising diagnostic and therapeutic strategies have resulted from advances in understanding molecular brain tumor biology. Important new approaches include the refinement of drug-delivery strategies, the evaluation of biologic markers to stratify patients for optimal treatment and to exploit these molecular differences using "targeted" therapeutic strategies. These approaches include blocking tumor cell drug resistance mechanisms, immunotherapy, inhibition of molecular signal transduction pathways important in tumorigenesis, anti-angiogenic therapy, and gene therapy. The thrust of such approaches for children with brain tumors is especially directed at reducing the toxicity of therapy and improving quality-of-life, as well as increasing disease-free survival.
Collapse
Affiliation(s)
- Patricia L Robertson
- Department of Pediatrics and Neurology, Division of Pediatric Neurology, University of Michigan Health System, 1500 E. Medical Center Dr., L3215 Women's Hospital, Ann Arbor, 48109-0203, USA.
| |
Collapse
|
20
|
Abstract
For many types of childhood brain tumors, including malignant gliomas, disease progression at the primary site is the predominant mode of treatment failure. Accordingly, interest has been directed during the last decade on exploring strategies to enhance the delivery of therapeutically active agents into the tumor microenvironment. Two approaches that have been the focus of considerable attention in the treatment of adult malignant brain tumors include interstitial administration of chemotherapeutic agents using time-release polymers and convection-enhanced delivery of immunotoxin conjugates targeted to receptors overexpressed in brain tumors relative to normal brain cells. Although it remains to be determined whether these approaches will lead to meaningful improvements in disease control and long-term prognosis in children with brain tumors, the encouraging results from studies in adults support the rationale for further exploring these strategies in the pediatric setting.
Collapse
Affiliation(s)
- Ian F Pollack
- Department of Neurosurgery, Children's Hospital of Pittsburgh, University of Pittsburgh Brain Tumor Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | | |
Collapse
|
21
|
Tasciotti E, Giacca M. Fusion of the Human Immunodeficiency Virus Type 1 Tat Protein Transduction Domain to Thymidine Kinase Increases Bystander Effect and Induces Enhanced Tumor KillingIn Vivo. Hum Gene Ther 2005; 16:1389-403. [PMID: 16390270 DOI: 10.1089/hum.2005.16.1389] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The clinical success of suicide gene therapy using herpes simplex virus type 1 thymidine kinase (TK) is largely dependent on the capacity of this enzyme to effectively induce the death of bystander cells. We have shown that fusion of TK to an 11-amino acid peptide from the basic domain of the human immunodeficiency virus type 1 Tat protein (Tat11) imparts cell membrane-translocating ability to the enzyme and significantly increases its cytotoxic activity. Here we report on the efficacy of this strategy in two different mouse models of adoptive tumorigenesis, based on the implantation of human Kaposi sarcoma (KS-IMM) cells in nude mice or of B16F10 melanoma cells in syngeneic C57BL/6J mice. Experiments were performed by the subcutaneous injection of mixtures of unmodified tumor cells containing different fractions of TK or Tat11-TK producing cells followed by animal treatment with ganciclovir (GCV). In both systems we consistently found that mice bearing tumors containing Tat11-TK cells displayed significantly retarded tumor growth and prolonged survival as compared with mice inoculated with cells expressing unmodified TK. Collectively, these results demonstrate that fusion of Tat11 to TK imparts remarkable intercellular trafficking capability to the enzyme. This modification of TK might constitute an important step in the optimization of TK suicide gene strategy for gene therapy of cellular proliferation.
Collapse
MESH Headings
- Animals
- Bystander Effect/physiology
- Disease Models, Animal
- Gene Products, tat/genetics
- Gene Transfer Techniques
- Genes, Transgenic, Suicide/genetics
- Genetic Therapy
- HIV-1
- Humans
- Melanoma/genetics
- Melanoma/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Nude
- Neoplasm Transplantation
- Sarcoma, Kaposi/genetics
- Sarcoma, Kaposi/therapy
- Thymidine Kinase/genetics
- Thymidine Kinase/therapeutic use
- Transfection
- Tumor Cells, Cultured
- tat Gene Products, Human Immunodeficiency Virus
Collapse
Affiliation(s)
- Ennio Tasciotti
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology, 34012 Trieste, Italy
| | | |
Collapse
|
22
|
Tasciotti E, Giacca M. Fusion of the Human Immunodeficiency Virus Type 1 Tat Protein Transduction Domain to Thymidine Kinase Increases Bystander Effect and Induces Enhanced Tumor Killing In Vivo. Hum Gene Ther 2005. [DOI: 10.1089/hum.2005.16.ft-143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
23
|
Abstract
Pediatric brain tumors are the commonest cause of cancer-related death in children. The last four decades have seen only a 35% increase in 5-year survival rate of children with these tumors. The therapeutic successes achieved are due to advances in neuroimaging, surgical techniques, radiotherapy, and induction of newer chemotherapeutic agents along with molecular targeted therapy. Neuroimaging advances include the use of MRA, MRS, DSA, and PET scans. With the use of stereotactic surgery, intraoperative mapping, and imaging, surgical resection has improved with significant decrease in morbidity. A major development has been the use of precision guided radiotherapy utilizing technologies like 3D-CRT, SRS, and IMRT, thereby decreasing radiation to normal tissues. Induction of newer drugs and high-dose chemotherapy with peripheral stem cell support has improved survival and delayed radiation in younger children and infants with brain tumors. Intense ongoing research is profiling novel molecular targets for therapeutic intervention. Newer therapeutic strategies like blood brain barrier disruption, immunotherapy, and gene therapy are in clinical trials. This review article intends to give the reader an overview of current therapeutic strategies and research involved in the treatment of children with brain tumors.
Collapse
Affiliation(s)
- Soumen Khatua
- Division of Pediatric Oncology, Tata Memorial Center, Mumbai, India.
| | | |
Collapse
|
24
|
Hwang KS, Cho WK, Yoo J, Yun HJ, Kim S, Im DS. Adenovirus-mediated interleukin-12 gene transfer combined with cytosine deaminase followed by 5-fluorocytosine treatment exerts potent antitumor activity in Renca tumor-bearing mice. BMC Cancer 2005; 5:51. [PMID: 15910693 PMCID: PMC1168891 DOI: 10.1186/1471-2407-5-51] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2004] [Accepted: 05/24/2005] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Therapeutic gene transfer affords a clinically feasible and safe approach to cancer treatment but a more effective modality is needed to improve clinical outcomes. Combined transfer of therapeutic genes with different modes of actions may be a means to this end. Interleukin-12 (IL-12), a heterodimeric immunoregulatory cytokine composed of covalently linked p35 and p40 subunits, has antitumor activity in animal models. The enzyme/prodrug strategy using cytosine deaminase (CD) and 5-fluorocytosine (5-FC) has been used for cancer gene therapy. We have evaluated the antitumor effect of combining IL-12 with CD gene transfer in mice bearing renal cell carcinoma (Renca) tumors. METHODS Adenoviral vectors were constructed encoding one or both subunits of murine IL-12 (Ad.p35, Ad.p40 and Ad.IL-12) or cytosine deaminase (Ad.CD). The functionality of the IL-12 or CD gene products expressed from these vectors was validated by splenic interferon (IFN)-gamma production or viability assays in cultured cells. Ad.p35 plus Ad.p40, or Ad.IL-12, with or without Ad.CD, were administered (single-dose) intratumorally to Renca tumor-bearing mice. The animals injected with Ad.CD also received 5-FC intraperitoneally. The antitumor effects were then evaluated by measuring tumor regression, mean animal survival time, splenic natural killer (NK) cell activity and IFN-gamma production. RESULTS The inhibition of tumor growth in mice treated with Ad.p35 plus Ad.p40 and Ad.CD, followed by injection of 5-FC, was significantly greater than that in mice treated with Ad.CD/5-FC, a mixture of Ad.p35 plus Ad.p40, or Ad.GFP (control). The combined gene transfer increased splenic NK cell activity and IFN-gamma production by splenocytes. Ad.CD/5-FC treatment significantly increased the antitumor effect of Ad.IL-12 in terms of tumor growth inhibition and mean animal survival time. CONCLUSION The results suggest that adenovirus-mediated IL-12 gene transfer combined with Ad.CD followed by 5-FC treatment may be useful for treating cancers.
Collapse
Affiliation(s)
- Kyung-Sun Hwang
- Laboratory of Gene Therapy and Virology, Korea Research Institute of Bioscience and Biotechnology, Yusong, Daejeon, Republic of Korea
| | - Won-Kyung Cho
- Laboratory of Gene Therapy and Virology, Korea Research Institute of Bioscience and Biotechnology, Yusong, Daejeon, Republic of Korea
| | - Jinsang Yoo
- Laboratory of Gene Therapy and Virology, Korea Research Institute of Bioscience and Biotechnology, Yusong, Daejeon, Republic of Korea
| | - Hwan-Jung Yun
- Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Samyong Kim
- Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Dong-Soo Im
- Laboratory of Gene Therapy and Virology, Korea Research Institute of Bioscience and Biotechnology, Yusong, Daejeon, Republic of Korea
| |
Collapse
|
25
|
Chiocca EA, Broaddus WC, Gillies GT, Visted T, Lamfers MLM. Neurosurgical delivery of chemotherapeutics, targeted toxins, genetic and viral therapies in neuro-oncology. J Neurooncol 2004; 69:101-17. [PMID: 15527083 DOI: 10.1023/b:neon.0000041874.02554.b3] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Local delivery of biologic agents, such as gene and viruses, has been tested preclinically with encouraging success, and in some instances clinical trials have also been performed. In addition, the positive pressure infusion of various therapeutic agents is undergoing human testing and approval has already been granted for routine clinical use of biodegradable implants that diffuse a chemotherapeutic agent into peritumoral regions. Safety in glioma patients has been shown, but anticancer efficacy needs additional refinements in the technologies employed. In this review, we will describe these modalities and provide a perspective on needed improvements that should render them more successful.
Collapse
Affiliation(s)
- E Antonio Chiocca
- Molecular Neuro-Oncology Laboratories, Neurosurgery Service, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | | | | | | | | |
Collapse
|
26
|
Nestler U, Wakimoto H, Siller-Lopez F, Aguilar LK, Chakravarti A, Muzikansky A, Stemmer-Rachamimov A, Chiocca EA, Aguilar-Cordova E, Hochberg FH. The combination of adenoviral HSV TK gene therapy and radiation is effective in athymic mouse glioblastoma xenografts without increasing toxic side effects. J Neurooncol 2004; 67:177-88. [PMID: 15072465 DOI: 10.1023/b:neon.0000021897.53969.ca] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECT In mouse models of prostate and breast cancer therapeutic effects are enhanced when adenoviral HSV TK gene therapy is combined with ionizing radiation. In the present study, we adopted this approach for the treatment of human glioblastoma xenografts in an athymic mouse model and assessed treatment results as well as toxic side effects. METHODS About 72 nude mice received intracerebral inoculations of 2 x 10(5) U87deltaEGFR cells. On day 7 after tumor implantation the study population was randomized into six treatment arms: (1) intratumoral buffer inoculation on day 7, (2) intratumoral adenoviral vector injection (2 x 10(9) vp) on day 7, (3) single dose radiation (2.1 Gy) on day 9, (4) adenoviral injection + radiation, (5) adenoviral injection + ganciclovir (GCV) (20 microg/g twice daily from day 8 to 17), (6) adenoviral injection + GCV + radiation. On day 21 half of the animals were sacrificed for histological evaluation of the brain tumors, the other half was assessed for survival. RESULTS This study showed significantly prolonged median survival time of 5 days for the GCV treated groups. The addition of radiation decreased the frequency of neurological symptoms and delayed the onset of deficits without altering the expression of thymidine kinase in the tumor cells. CONCLUSIONS We conclude that adenoviral HSV TK gene therapy in combination with adjuvant radiotherapy does not generate increased toxic side effects in glioblastoma treatment. The prolonged survival time of animals receiving gene therapy and the reduced occurrence of neurological symptoms in irradiated mice constitute promising features of the combined treatment.
Collapse
Affiliation(s)
- Ulf Nestler
- Molecular Neuro-Oncology Laboratory, Department of Radiation Oncology, Massachusetts General Hospital East, Charlestown, MA, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
The field of cancer gene therapy is in continuous expansion, and technology is quickly moving ahead as far as gene targeting and regulation of gene expression are concerned. This review focuses on the endocrine aspects of gene therapy, including the possibility to exploit hormone and hormone receptor functions for regulating therapeutic gene expression, the use of endocrine-specific genes as new therapeutic tools, the effects of viral vector delivery and transgene expression on the endocrine system, and the endocrine response to viral vector delivery. Present ethical concerns of gene therapy and the risk of germ cell transduction are also discussed, along with potential lines of innovation to improve cell and gene targeting.
Collapse
Affiliation(s)
- Luisa Barzon
- Department of Histology, Microbiology, and Medical Biotechnologies, University of Padova, I-35121 Padua, Italy
| | | | | |
Collapse
|
28
|
Abstract
Retrovirus (RV) has been one of the earliest recombinant vectors to be investigated in the context of cancer gene therapy. Experiments in cell culture and in animal brain tumor models have demonstrated the feasibility of RV mediated gene transduction and killing of glioma cells by toxicity generating transgenes. Phase I and II clinical studies in patients with recurrent malignant glioma have shown a favorable safety profile and some efficacy of RV mediated gene therapy. On the other hand, a prospective randomized phase III clinical study of RV gene therapy in primary malignant glioma failed to demonstrate significant extension of the progression-free or overall survival times in RV treated patients. The failure of this RV gene therapy study may be due to the low tumor cell transduction rate observed in vivo. The biological effects of the treatment may also heavily depend on the choice of transgene/prodrug system and on the vector delivery methods. Retrovirus clinical trials in malignant glioma have nevertheless produced a substantial amount of data and have contributed toward the identification of serious shortcomings of the non-replicating virus vector gene therapy strategy. Novel types of therapeutic virus vector systems are currently being designed and new clinical protocols are being created based on the lessons learned from the RV gene therapy trials in patients with malignant brain tumors.
Collapse
Affiliation(s)
- Nikolai G Rainov
- Department of Neurological Science, University of Liverpool, Liverpool, UK.
| | | |
Collapse
|
29
|
Rainov NG, Kramm CM. Recombinant retrovirus vectors for treatment of malignant brain tumors. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2003; 55:185-203. [PMID: 12968537 DOI: 10.1016/s0074-7742(03)01008-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Affiliation(s)
- Nikolai G Rainov
- Department of Neurological Science, University of Liverpool, Walton Centre for Neurology and Neurosurgery NHS Trust, Liverpool L9 7LJ, United Kingdom
| | | |
Collapse
|
30
|
Niranjan A, Wolfe D, Tamura M, Soares MK, Krisky DM, Lunsford LD, Li S, Fellows-Mayle W, DeLuca NA, Cohen JB, Glorioso JC. Treatment of rat gliosarcoma brain tumors by HSV-based multigene therapy combined with radiosurgery. Mol Ther 2003; 8:530-42. [PMID: 14529825 DOI: 10.1016/s1525-0016(03)00232-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Our laboratory has employed replication-defective herpes simplex virus type 1 gene transfer vectors for treatment of animal models of human malignant glioblastoma. The base vectors were defective for the immediate early (IE) genes ICP4, ICP27, and ICP22 but expressed the IE gene ICP0, which can arrest tumor cell division, and an IE thymidine kinase (alpha-tk) gene construct that mediates suicide gene therapy (SGT) in the presence of ganciclovir (GCV). Previously, we reported that SGT using ICP0/alpha-tk vectors in nude mouse models of glioblastoma was improved by coexpression of the gap-junction-forming protein connexin43 (Cx43) or human tumor necrosis factor alpha (TNF alpha). We also showed that further gains in therapeutic outcome could be achieved by combining TNF alpha-enhanced SGT with gamma-knife radiosurgery (GKR). To expand these observations, we have first repeated these studies in immunocompetent rats with brain tumors derived from implanted 9L gliosarcoma cells and second compared the most efficient vector from this study with a new recombinant vector, NUREL-C2, which expressed both TNF alpha and Cx43 along with ICP0 and alpha-tk. Results from the first part indicated that our ICP0/alpha-tk/TNF alpha vector in combination with GKR provides an effective therapy although this treatment was not statistically better than GKR combined with the ICP0/alpha-tk/Cx43 vector. Our observations in the second part suggested that NUREL-C2 may be more effective than the ICP0/alpha-tk/TNF alpha vector in combination treatments with GCV (P = 0.08) or GCV plus GKR (P = 0.10). GKR significantly enhanced the efficacy of NUREL-C2/GCV treatment (P = 0.02) as well as other virus/GCV treatments (P < or = 0.05). Conversely, the efficacy of GKR was significantly improved by both the ICP0/alpha-tk/TNF alpha vector and NUREL-C2 in combination with GCV (P = 0.02 and P < 0.01, respectively). Together these results indicate that NUREL-C2 may be an attractive candidate for Phase I gene-therapy safety studies in patients with recurrent malignant glioblastoma.
Collapse
Affiliation(s)
- Ajay Niranjan
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
MacDonald TJ, Rood BR, Santi MR, Vezina G, Bingaman K, Cogen PH, Packer RJ. Advances in the diagnosis, molecular genetics, and treatment of pediatric embryonal CNS tumors. Oncologist 2003; 8:174-86. [PMID: 12697942 DOI: 10.1634/theoncologist.8-2-174] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Embryonal central nervous system (CNS) tumors are the most common group of malignant brain tumors in children. The diagnosis and classification of tumors belonging to this family have been controversial; however, utilization of molecular genetics is helping to refine traditional histopathologic and clinical classification schemes. Currently, this group of tumors includes medulloblastomas, supratentorial primitive neuroectodermal tumors, atypical teratoid/rhabdoid tumors, ependymoblastomas, and medulloepitheliomas. While the survival of older children with nonmetastatic medulloblastomas has improved considerably within the past two decades, the outcomes for infants and for those with metastatic medulloblastomas or other high-risk embryonal CNS tumors remain poor. It is anticipated that the emerging field of molecular biology will greatly aid in the future stratification and therapy for pediatric patients with malignant embryonal tumors. In this review, recent advances in the diagnosis, molecular genetics, and treatment of the most common pediatric embryonal CNS tumors are discussed.
Collapse
Affiliation(s)
- Tobey J MacDonald
- Departments of Hematology/Oncology, Children's Hospital National Medical Center, Washington, DC 20010, USA.
| | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Gene therapy holds great promise. Somatic gene therapy has the potential to treat a wide range of disorders, including inherited conditions, cancers, and infectious diseases. Early progress has already been made in the treatment of a range of disorders. Ethical issues surrounding somatic gene therapy are primarily those concerned with safety. Germline gene therapy is theoretically possible but raises serious ethical concerns concerning future generations.
Collapse
Affiliation(s)
- Kevin R Smith
- School of Contemporary Sciences, University of Abertay Dundee, Dundee, Scotland, UK.
| |
Collapse
|
33
|
Treatment of relapsed malignant glioma with an adenoviral vector containing the herpes simplex thymidine kinase gene followed by ganciclovir. Mol Ther 2003; 7:851-8. [PMID: 12788659 DOI: 10.1016/s1525-0016(03)00100-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Between November 1998 and December 2001, we treated 14 patients with advanced recurrent high-grade gliomas with a total dose of 4.6 x 10(8), 4.6 x 10(9), 4.6 x 10(10), or 4.6 x 10(11) viral particles (VP) of a replication-incompetent adenoviral vector harboring the herpes simplex virus thymidine kinase gene driven by the adenoviral major late promoter (IG.Ad.MLPI.TK), followed by ganciclovir (GCV) treatment. The VP-to-infectious-unit ratio was 40. The vector was administered by 50 intraoperative wound-bed injections of 0.2 ml each (total volume 10 ml). The study's primary objective was to determine the safety of this treatment and establish the maximum tolerated dose (MTD). Injection of all doses of IG.Ad.MLPI.TK followed by GCV was safely tolerated and MTD was not reached. All patients had recurrence or progression of the tumor 1-24 months (median 3.5 months) after gene therapy. The overall median survival was 4 months. Four patients survived longer than 1 year following gene therapy. One patient is still alive, with histologically confirmed progression of the tumor, 29 months after treatment. Ten patients died within 8 months of treatment, all from progression of the tumor. In 5 patients residual and measurable tumor was visible on the direct (<48 h) postoperative MRI. No objective radiological response was documented on subsequent MRI. None of the patients came to autopsy. In conclusion, the administration of 4.6 x 10(11) VP of IG.Ad.MLPI.TK by 50 injections into the wound bed following resection of recurrent malignant glioma, followed by GCV treatment, was well tolerated.
Collapse
|
34
|
Abstract
Malignant gliomas remain amongst the most difficult cancer to treat. Viral-based gene therapies have been employed for the last decade in preclinical and clinical modes as a novel treatment modality. In this review, such therapies are summarized. The overwhelming majority of clinical studies point one to conclude that methodologies that will increase tumor infection/transduction will lead to enhanced therapeutic results.
Collapse
Affiliation(s)
- E Antonio Chiocca
- Molecular Neuro-oncology Laboratory, Neurosurgery Service, Massachusetts General Hospital-East, Charlestown, Massachusetts 02129, USA.
| | | | | |
Collapse
|
35
|
Abstract
PURPOSE Brain tumors were the first human malignancy to be targeted by therapeutic transfer of nucleic acids into somatic cells, a process also known as gene therapy. Malignant brain tumor cells in the adult brain have some unique biologic features, such as high mitotic activity on an essentially postmitotic background and virtually no tumor spread outside of the central nervous system. Brain tumors seem therefore to offer major advantages in the design of tumor-selective gene therapy strategies, and the role of gene therapy in malignant glioma has been investigated since the late 1980s, initially in numerous laboratory studies and later on in clinical trials. DESIGN Retrovirus has been one of the earliest recombinant virus vectors used in brain tumors. Experiments in cell culture and in animal models have demonstrated the feasibility of retrovirus-mediated transduction and subsequent killing of glioma cells by toxic transgenes. Phase I and II clinical studies in patients with recurrent malignant glioma have shown a favorable safety profile and some efficacy of retrovirus-mediated gene therapy. However, the only prospective, randomized, phase III clinical study of retrovirus gene therapy in primary malignant glioma failed to demonstrate significant extension of progression-free or overall survival. Adenovirus- and herpes simplex virus type 1-based vectors have been actively investigated along with retrovirus, but their clinical use is still limited, mostly because of safety concerns. To increase efficacy, novel generations of therapeutic adenovirus and herpes simplex virus type 1 rely more on genetically engineered and tumor-selective lytic properties and less on the actual transfer of therapeutic genes. CONCLUSIONS The failure of most clinical gene therapy protocols to produce a significant and unequivocal benefitto brain tumor patients seems to be mainly due to the low tumor cell transduction rates observed in vivo, but it may also depend on the respective physical delivery strategy of the vector. Standard radiologic criteria for assessing the efficacy of clinical treatments may also not be fully applicable to the specific metabolic changes and blood-brain barrier permeability phenomena caused in brain tumors by virus-mediated gene therapy. Clinical trials in malignant glioma have nevertheless produced a substantial amount of data and have contributed to the continuous improvement of vector systems, delivery methods, and clinical protocols.
Collapse
Affiliation(s)
- Nikolai G Rainov
- Department of Neurological Science, University of Liverpool, Liverpool L9 7LJ, UK.
| | | |
Collapse
|
36
|
Castro MG, Cowen R, Williamson IK, David A, Jimenez-Dalmaroni MJ, Yuan X, Bigliari A, Williams JC, Hu J, Lowenstein PR. Current and future strategies for the treatment of malignant brain tumors. Pharmacol Ther 2003; 98:71-108. [PMID: 12667889 DOI: 10.1016/s0163-7258(03)00014-7] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Glioblastoma (GB) is the most common subtype of primary brain tumor in adults. These tumors are highly invasive, very aggressive, and often infiltrate critical neurological areas within the brain. The mean survival time after diagnosis of GB has remained unchanged during the last few decades, in spite of advances in surgical techniques, radiotherapy, and also chemotherapy; patients' survival ranges from 9 to 12 months after initial diagnosis. In the same time frame, with our increasing understanding and knowledge of the physiopathology of several cancers, meaningful advances have been made in the treatment and control of several cancers, such as breast, prostate, and hematopoietic malignancies. Although a number of the genetic lesions present in GB have been elucidated and our understanding of the progressions of this cancer has increased dramatically over the last few years, it has not yet been possible to harness this information towards developing effective cures. In this review, we will focus on the classical ways in which GB is currently being treated, and will introduce a novel therapeutic modality, i.e., gene therapy, which we believe will be used in combination with classical treatment strategies to prolong the life-span of patients and to ultimately be able to control and/or cure these brain tumors. We will discuss the use of several vector systems that are needed to introduce the therapeutic genes within either the tumor mass, if these are not resectable, or the tumor bed, after successful tumor resection. We also discuss different therapeutic modalities that could be exploited using gene therapy, i.e., conditional cytotoxic approach, direct cytotoxicity, immunotherapy, inhibition of angiogenesis, and the use of pro-apoptotic genes. The advantages and disadvantages of each of the current vector systems available to transfer genes into the CNS are also discussed. With the advances in molecular techniques, both towards the elucidation of the physiopathology of GB and the development of novel, more efficient and less toxic vectors to deliver putative therapeutic genes into the CNS, it should be possible to develop new rationale and effective therapeutic approaches to treat this devastating cancer.
Collapse
Affiliation(s)
- M G Castro
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Research Pavilion, 8700 Beverly Boulevard, Suite 5090, Los Angeles, CA 90048, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Biagi E, Bollard C, Rousseau R, Brenner M. Gene Therapy for Pediatric Cancer: State of the Art and Future Perspectives. J Biomed Biotechnol 2003; 2003:13-24. [PMID: 12686719 PMCID: PMC179759 DOI: 10.1155/s1110724303209025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
While modern treatments have led to a dramatic improvement in survival for pediatric malignancy, toxicities are high and a significant proportion of patients remain resistant. Gene transfer offers the prospect of highly specific therapies for childhood cancer. "Corrective" genes may be transferred to overcome the genetic abnormalities present in the precancerous cell. Alternatively, genes can be introduced to render the malignant cell sensitive to therapeutic drugs. The tumor can also be attacked by decreasing its blood supply with genes that inhibit vascular growth. Another possible approach is to modify normal tissues with genes that make them more resistant to conventional drugs and/or radiation, thereby increasing the therapeutic index. Finally, it may be possible to attack the tumor indirectly by using genes that modify the behavior of the immune system, either by making the tumor more immunogenic, or by rendering host effector cells more efficient. Several gene therapy applications have already been reported for pediatric cancer patients in preliminary Phase 1 studies. Although no major clinical success has yet been achieved, improvements in gene delivery technologies and a better understanding of mechanisms of tumor progression and immune escape have opened new perspectives for the cure of pediatric cancer by combining gene therapy with standard therapeutic available treatments.
Collapse
Affiliation(s)
- Ettore Biagi
- Center for Cell and Gene Therapy, Department of Pediatrics-Hematology/Oncology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Catherine Bollard
- Center for Cell and Gene Therapy, Department of Pediatrics-Hematology/Oncology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Raphael Rousseau
- Center for Cell and Gene Therapy, Department of Pediatrics-Hematology/Oncology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Malcolm Brenner
- Center for Cell and Gene Therapy, Department of Pediatrics-Hematology/Oncology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
38
|
Abstract
The grave outlook for malignant glioma patients in spite of improvements to current modalities has ushered in new approaches to therapy. Viruses have emerged on the scene and gained attention for their ability to play essentially two roles: first, as vectors for therapeutic gene delivery and second, as engineered infectious agents capable of selectively lysing tumor cells. To date, clinical brain tumor trials using viruses for gene delivery have employed retroviral or adenoviral vectors to introduce ganciclovir susceptibility to tumors in the form of the HSV1-TK gene. Clinical oncolytic studies, on the other hand, have evaluated a conditionally replicating HSV as an antineoplastic agent. Despite some promise afforded by these trials, further studies are warranted; the investigation of additional viruses to play these roles is inevitable and is now precedented.
Collapse
Affiliation(s)
- Peter E Fecci
- Departments of Neurosurgery and Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | |
Collapse
|
39
|
Abstract
Malignant gliomas cause 2% of cancer deaths in western countries, and even the most intensive combinations of radiotherapy and chemotherapy cannot be curative. New chemotherapeutic drugs and alternative therapeutic modalities are strongly needed. Huge efforts are directed towards the development of innovative strategies for targeting and mending the specific molecular alterations in tumor cells (translational research). This review aims to summarize the most promising lines of investigational research in the field of neuro-oncology, such as non-cytotoxic drugs, immunotoxins, inhibitors of angiogenesis and gene therapy approaches, which will probably offer new therapy options for brain tumor patients.
Collapse
Affiliation(s)
- Umberto Basso
- Department of Medical Oncology, Azienda Ospedale-Università of Padova, Italy
| | | | | | | |
Collapse
|
40
|
Harrington KJ, Melcher AA, Bateman AR, Ahmed A, Vile RG. Cancer gene therapy: Part 2. Candidate transgenes and their clinical development. Clin Oncol (R Coll Radiol) 2002; 14:148-69. [PMID: 12069125 DOI: 10.1053/clon.2001.0004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Kevin J Harrington
- CRC Centre for Cell and Molecular Biology, Institute for Cancer Research, London, UK.
| | | | | | | | | |
Collapse
|
41
|
Hsich G, Sena-Esteves M, Breakefield XO. Critical issues in gene therapy for neurologic disease. Hum Gene Ther 2002; 13:579-604. [PMID: 11916483 DOI: 10.1089/10430340252837198] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Gene therapy for the nervous system is a newly emerging field with special issues related to modes of delivery, potential toxicity, and realistic expectations for treatment of this vital and highly complex tissue. This review focuses on the potential for gene delivery to the brain, as well as possible risks and benefits of these procedures. This includes discussion of appropriate vectors, such as adeno-associated virus, lentivirus, gutless adenovirus, and herpes simplex virus hybrid amplicons, and cell vehicles, such as neuroprogenitor cells. Routes of delivery for focal and global diseases are enumerated, including use of migratory cells, facilitation of vascular delivery across the blood-brain barrier, cerebrospinal fluid delivery, and convection injection. Attention is given to examples of diseases falling into different etiologic types: metabolic deficiency states, including Canavan disease and lysosomal storage disorders; and degenerative conditions, including Parkinson's disease and other neurodegenerative conditions.
Collapse
Affiliation(s)
- Gary Hsich
- Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital, and Neuroscience Program, Harvard Medical School, Boston, MA 02114, USA
| | | | | |
Collapse
|
42
|
Xu CT, Huang LT, Pan BR. Current gene therapy for stomach carcinoma. World J Gastroenterol 2001; 7:752-9. [PMID: 11819868 PMCID: PMC4695588 DOI: 10.3748/wjg.v7.i6.752] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2001] [Revised: 05/29/2001] [Accepted: 06/06/2001] [Indexed: 02/06/2023] Open
Affiliation(s)
- C T Xu
- Editorial Department, the Journal of Fourth Military Medical University, Xi'an, Shaanxi Province, China.
| | | | | |
Collapse
|
43
|
Abstract
Childhood brain tumors are collectively the most common solid neoplasm and the leading cause of cancer-related death in children. They are a diverse group of diseases and outcome is extremely variable. Current treatment is dependent on histology, location, and in some instances, patient age. Advances in treatment have led to improved survival for some patients, but for many the outcome remains dismal despite aggressive treatment. A growing body of work is aimed at improving the outcome for children with brain tumors not only through clinical trials, but also by focusing on the biologic underpinning of these diseases that have been poorly understood.
Collapse
Affiliation(s)
- A T Reddy
- Departments of Pediatrics and Neurology, University of Alabama at Birmingham, Children's Hospital of Alabama, 1600 7th Ave. South, Suite 516 ACC, Birmingham, AL 35233, USA.
| |
Collapse
|
44
|
Garin MI, Garrett E, Tiberghien P, Apperley JF, Chalmers D, Melo JV, Ferrand C. Molecular mechanism for ganciclovir resistance in human T lymphocytes transduced with retroviral vectors carrying the herpes simplex virus thymidine kinase gene. Blood 2001; 97:122-9. [PMID: 11133751 DOI: 10.1182/blood.v97.1.122] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The herpes simplex virus thymidine kinase gene type 1 (HSV-Tk) ganciclovir (GCV) system is a novel therapeutic strategy for the modulation of graft-versus-host disease (GVHD), a major complication of allogeneic stem cell transplantation (allo-SCT). Retroviral-mediated gene transfer of the HSV-Tk gene into donor T lymphocytes before allo-SCT may allow their in vivo selective depletion after treatment with GCV. The expression of the HSV-Tk gene was analyzed in vitro in CEM cells, a human lymphoblastoid cell line, transduced with 2 different vectors, each containing the HSV-Tk gene and a selectable marker gene. GCV-resistant clones were identified within the clones expressing the marker gene. Characterization of the molecular events leading to this resistance revealed a 227-bp deletion in the HSV-Tk gene due to the presence of cryptic splice donor and acceptor sites within the HSV-Tk gene sequence. Furthermore, it was confirmed that this deletion was present in human primary T cells transduced with either vector and in 12 patients who received transduced donor T cells, together with a T-cell-depleted allo-SCT. In vivo circulating transduced T cells containing the truncated HSV-Tk gene were identified in all patients immediately after infusion and up to 800 days after transplantation. In patients who received GCV as treatment for GVHD, a progressive increase in the proportion of transduced donor T cells carrying the deleted HSV-Tk gene was observed. These results suggest that the limitations within the HSV-Tk/GCV system can be improved by developing optimized retroviral vectors to ensure maximal killing of HSV-Tk-transduced cells.
Collapse
Affiliation(s)
- M I Garin
- Department of Haematology, Imperial College School of Medicine, Hammersmith Hospital, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
A number of exciting advances have been reported over the past few years in the understanding and treatment of children with brain tumors. The present review highlights many of the publications from this period, focusing on their relevance within the major diagnostic and treatment domains of pediatric oncology (surgery, radiation therapy, chemotherapy, neuropathology, and neuroradiology). Although many of the publications cited provide confirmation of previously reported work, when taken together they form a good framework of the state of the field from the past few years.
Collapse
Affiliation(s)
- M W Kieran
- Pediatric Medical Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA.
| |
Collapse
|
46
|
Rainov NG. A phase III clinical evaluation of herpes simplex virus type 1 thymidine kinase and ganciclovir gene therapy as an adjuvant to surgical resection and radiation in adults with previously untreated glioblastoma multiforme. Hum Gene Ther 2000; 11:2389-401. [PMID: 11096443 DOI: 10.1089/104303400750038499] [Citation(s) in RCA: 478] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Previous uncontrolled clinical trials have shown the in vivo retrovirus (RV)-mediated transduction of glioblastoma cells with the herpes simplex virus thymidine kinase (HSV-tk) gene and subsequent systemic treatment with ganciclovir to be feasible and well tolerated. However, because of continued tumor progression in most patients, the antitumor effect could not be determined using historical controls. Here, we describe a phase III, multicenter, randomized, open-label, parallel-group, controlled trial of the technique in the treatment of 248 patients with newly diagnosed, previously untreated glioblastoma multiforme (GBM). Patients received, in equal numbers, either standard therapy (surgical resection and radiotherapy) or standard therapy plus adjuvant gene therapy during surgery. Progression-free median survival in the gene therapy group was 180 days compared with 183 days in control subjects. Median survival was 365 versus 354 days, and 12-month survival rates were 50 versus 55% in the gene therapy and control groups, respectively. These differences were not significant. Therefore, the adjuvant treatment improved neither time to tumor progression nor overall survival time, although the feasibility and good biosafety profile of this gene therapy strategy were further supported. The failure of this specific protocol may be due mainly to the presumably poor rate of delivery of the HSV-tk gene to tumor cells. In addition, the current mode of manual injection of vector-producing cells with a nonmigratory fibroblast phenotype limits the distribution of these cells and the released replication-deficient RV vectors to the immediate vicinity of the needle track. Further evaluation of the RV-mediated gene therapy strategy must incorporate refinements such as improved delivery of vectors and transgenes to the tumor cells, noninvasive in vivo assessment of transduction rates, and improved delivery of the prodrug across the blood-brain and blood-tumor barrier to the transduced tumor cells.
Collapse
Affiliation(s)
- N G Rainov
- Department of Neurosurgery, Martin-Luther Universität, Halle-Wittenberg, D-06097 Halle, Germany.
| |
Collapse
|
47
|
Alvarez RD, Gomez-Navarro J, Wang M, Barnes MN, Strong TV, Arani RB, Arafat W, Hughes JV, Siegal GP, Curiel DT. Adenoviral-mediated suicide gene therapy for ovarian cancer. Mol Ther 2000; 2:524-30. [PMID: 11082326 DOI: 10.1006/mthe.2000.0194] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The purpose of this phase I study was to determine the potential efficacy of adenoviral-mediated suicide gene therapy in women with recurrent ovarian cancer. Fourteen patients were treated intraperitoneally with herpes simplex virus-thymidine kinase (HSV-TK)-encoding adenovirus (AdHSV-TK) in dosages ranging between 1x10(9) and 1x10(11) pfu. Beginning 2 days later, ganciclovir (GCV) was administered intravenously at a dose of 5 mg/kg bid for 14 days. Transient vector-associated fever was experienced by 4 of 14 (29%) treated patients. Other possible vector-associated constitutional symptoms, abdominal pain, and gastrointestinal symptoms were experienced by 6 of 14 (43%) treated patients. No other dose-limiting vector-specific side effects were noted. Of the 13 patients evaluable for response, 5 (38%) had stable disease and 8 (62%) had evidence of progressive disease. Molecular analysis of evaluable ascites samples demonstrated the presence of transgene DNA and RNA in most patients 2 days following Ad HSV-TK administration. Ten of 11 evaluable patients had an increase in anti-adenovirus antibody titer. These results suggest that treatment with AdHSV-TK in combination with GCV is feasible in the context of human ovarian cancer and tolerated at the dosages studied.
Collapse
Affiliation(s)
- R D Alvarez
- Department of Obstetrics and Gynecology, National Gene Vector Laboratory, Birmingham, Alabama 35233, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
|
49
|
Abstract
"Gene therapy" can be defined as the transfer of genetic material into a patient's cells for therapeutic purposes. To date, a diverse and creative assortment of treatment strategies utilizing gene therapy have been devised, including gene transfer for modulating the immune system, enzyme prodrug ("suicide gene") therapy, oncolytic therapy, replacement/therapeutic gene transfer, and antisense therapy. For malignant glioma, gene-directed prodrug therapy using the herpes simplex virus thymidine kinase gene was the first gene therapy attempted clinically. A variety of different strategies have now been pursued experimentally and in clinical trials. Although, to date, gene therapy for brain tumors has been found to be reasonably safe, concerns still exist regarding issues related to viral delivery, transduction efficiency, potential pathologic response of the brain, and treatment efficacy. Improved viral vectors are being sought, and potential use of gene therapy in combination with other treatments is being investigated.
Collapse
Affiliation(s)
- K Bansal
- Departments of Neurosurgery and Molecular Genetics, College of Medicine, University of Illinois at Chicago, 912 South Wood Street, Chicago, IL 60612, USA
| | | |
Collapse
|
50
|
Niranjan A, Moriuchi S, Lunsford LD, Kondziolka D, Flickinger JC, Fellows W, Rajendiran S, Tamura M, Cohen JB, Glorioso JC. Effective treatment of experimental glioblastoma by HSV vector-mediated TNF alpha and HSV-tk gene transfer in combination with radiosurgery and ganciclovir administration. Mol Ther 2000; 2:114-20. [PMID: 10947938 DOI: 10.1006/mthe.2000.0101] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Experiments were carried out in a nude mouse model of human glioblastoma to determine whether gamma-knife radiosurgery combined with herpes simplex virus thymidine kinase (tk) suicide gene therapy and tumor necrosis factor alpha (TNFalpha) gene transfer provided an improved multimodality treatment of this disease. Animals were inoculated intracerebrally with 2 x 10(5) U-87MG human glioblastoma cells to establish brain tumors. At 3 days postinoculation, the tumor region was injected with 2 x 10(6) infectious particles of highly defective herpes simplex viral vectors expressing the viral tk gene with the kinetics of a viral immediate early gene either alone (T.1) or together with TNF alpha (TH:TNF). Subgroups of animals were given daily intraperitoneal injections of ganciclovir (GCV) for 10 days and/or subjected to gamma-knife radiosurgery on the fifth day post tumor-cell implantation. Comparisons of animal survival showed that the TH:TNF vector in combination with radiosurgery and GCV administration provided the most effective therapy; eight of nine animals survived for 75 days compared to four of eight using the next best protocol. These findings suggest that gene therapy in combination with more conventional therapeutic methods may provide an improved strategy for extending the life expectancy of patients afflicted with this ultimately fatal disease.
Collapse
Affiliation(s)
- A Niranjan
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|