1
|
McDowall S, Aung-Htut M, Wilton S, Li D. Antisense oligonucleotides and their applications in rare neurological diseases. Front Neurosci 2024; 18:1414658. [PMID: 39376536 PMCID: PMC11456401 DOI: 10.3389/fnins.2024.1414658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/20/2024] [Indexed: 10/09/2024] Open
Abstract
Rare diseases affect almost 500 million people globally, predominantly impacting children and often leading to significantly impaired quality of life and high treatment costs. While significant contributions have been made to develop effective treatments for those with rare diseases, more rapid drug discovery strategies are needed. Therapeutic antisense oligonucleotides can modulate target gene expression with high specificity through various mechanisms determined by base sequences and chemical modifications; and have shown efficacy in clinical trials for a few rare neurological conditions. Therefore, this review will focus on the applications of antisense oligonucleotides, in particular splice-switching antisense oligomers as promising therapeutics for rare neurological diseases, with key examples of Duchenne muscular dystrophy and spinal muscular atrophy. Challenges and future perspectives in developing antisense therapeutics for rare conditions including target discovery, antisense chemical modifications, animal models for therapeutic validations, and clinical trial designs will also be briefly discussed.
Collapse
Affiliation(s)
- Simon McDowall
- School of Human Sciences, The University of Western Australia, Crawley, WA, Australia
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Nedlands, WA, Australia
| | - May Aung-Htut
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Nedlands, WA, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia
| | - Steve Wilton
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Nedlands, WA, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia
| | - Dunhui Li
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Nedlands, WA, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia
| |
Collapse
|
2
|
Hu X, Li J, Fu M, Zhao X, Wang W. The JAK/STAT signaling pathway: from bench to clinic. Signal Transduct Target Ther 2021; 6:402. [PMID: 34824210 PMCID: PMC8617206 DOI: 10.1038/s41392-021-00791-1] [Citation(s) in RCA: 1168] [Impact Index Per Article: 292.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/09/2021] [Accepted: 09/21/2021] [Indexed: 02/08/2023] Open
Abstract
The Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway was discovered more than a quarter-century ago. As a fulcrum of many vital cellular processes, the JAK/STAT pathway constitutes a rapid membrane-to-nucleus signaling module and induces the expression of various critical mediators of cancer and inflammation. Growing evidence suggests that dysregulation of the JAK/STAT pathway is associated with various cancers and autoimmune diseases. In this review, we discuss the current knowledge about the composition, activation, and regulation of the JAK/STAT pathway. Moreover, we highlight the role of the JAK/STAT pathway and its inhibitors in various diseases.
Collapse
Affiliation(s)
- Xiaoyi Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu, 610041, Sichuan, P. R. China
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, 610041, Chengdu, P. R. China
| | - Jing Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu, 610041, Sichuan, P. R. China
| | - Maorong Fu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu, 610041, Sichuan, P. R. China
| | - Xia Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu, 610041, Sichuan, P. R. China.
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, 610041, Chengdu, P. R. China.
| | - Wei Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu, 610041, Sichuan, P. R. China.
| |
Collapse
|
3
|
Xie T, Fan Z, Zhang R, Tian X, Han G, Liu Z, Zhang Z. In situ imaging of intracellular human telomerase RNA with molecular beacon-functionalized gold nanoparticles. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2020; 12:2385-2390. [PMID: 32930264 DOI: 10.1039/d0ay00461h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Since the expression level of human telomerase RNA (hTR) in tumor cells is much higher than that in normal cells, the determination of hTR is of prime importance in biological research of tumors. In this work, we report molecular beacon-functionalized gold nanoparticles for hTR imaging in live cells. The molecular beacon has a loop-and-stem structure with hTR recognition sequences and a red fluorophore Cy5. In the presence of hTR, the hTR sequence could be hybridized with the loop of molecular beacon to form a duplex DNA chain and thus the fluorescence state switched from "off" to "on". After co-incubation with cells, the probe could readily permeate into cells, leading to the in situ imaging of intracellular hTR. The proposed approach could be used to differentiate tumor cells from normal ones and assess hTR expression levels in different tumor cells. Furthermore, the proposed approach allowed us to dynamically monitor the expression level of hTR in live cells and holds great potential for application in tumor diagnosis and hTR-related drug delivery.
Collapse
Affiliation(s)
- Tao Xie
- Institute of Physical Science and Information Technology, School of Chemical and Chemical Engineering, School of Life Science, Anhui University, Hefei, Anhui 230601, China.
| | - Ziyan Fan
- Institute of Physical Science and Information Technology, School of Chemical and Chemical Engineering, School of Life Science, Anhui University, Hefei, Anhui 230601, China.
| | - Ruilong Zhang
- Institute of Physical Science and Information Technology, School of Chemical and Chemical Engineering, School of Life Science, Anhui University, Hefei, Anhui 230601, China.
| | - Xiaohe Tian
- Institute of Physical Science and Information Technology, School of Chemical and Chemical Engineering, School of Life Science, Anhui University, Hefei, Anhui 230601, China.
| | - Guangmei Han
- Institute of Physical Science and Information Technology, School of Chemical and Chemical Engineering, School of Life Science, Anhui University, Hefei, Anhui 230601, China.
| | - Zhengjie Liu
- Institute of Physical Science and Information Technology, School of Chemical and Chemical Engineering, School of Life Science, Anhui University, Hefei, Anhui 230601, China.
| | - Zhongping Zhang
- Institute of Physical Science and Information Technology, School of Chemical and Chemical Engineering, School of Life Science, Anhui University, Hefei, Anhui 230601, China.
| |
Collapse
|
4
|
Targeting STAT3 in Cancer with Nucleotide Therapeutics. Cancers (Basel) 2019; 11:cancers11111681. [PMID: 31671769 PMCID: PMC6896109 DOI: 10.3390/cancers11111681] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 10/22/2019] [Accepted: 10/24/2019] [Indexed: 12/18/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) plays a critical role in promoting the proliferation and survival of tumor cells. As a ubiquitously-expressed transcription factor, STAT3 has commonly been considered an "undruggable" target for therapy; thus, much research has focused on targeting upstream pathways to reduce the expression or phosphorylation/activation of STAT3 in tumor cells. Recently, however, novel approaches have been developed to directly inhibit STAT3 in human cancers, in the hope of reducing the survival and proliferation of tumor cells. Several of these agents are nucleic acid-based, including the antisense molecule AZD9150, CpG-coupled STAT3 siRNA, G-quartet oligodeoxynucleotides (GQ-ODNs), and STAT3 decoys. While the AZD9150 and CpG-STAT3 siRNA interfere with STAT3 expression, STAT3 decoys and GQ-ODNs target constitutively activated STAT3 and modulate its ability to bind to target genes. Both STAT3 decoy and AZD9150 have advanced to clinical testing in humans. Here we will review the current understanding of the structures, mechanisms, and potential clinical utilities of the nucleic acid-based STAT3 inhibitors.
Collapse
|
5
|
Walters BJ, Azam AB, Gillon CJ, Josselyn SA, Zovkic IB. Advanced In vivo Use of CRISPR/Cas9 and Anti-sense DNA Inhibition for Gene Manipulation in the Brain. Front Genet 2016; 6:362. [PMID: 26793235 PMCID: PMC4709581 DOI: 10.3389/fgene.2015.00362] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 12/19/2015] [Indexed: 12/28/2022] Open
Abstract
Gene editing tools are essential for uncovering how genes mediate normal brain-behavior relationships and contribute to neurodegenerative and neuropsychiatric disorders. Recent progress in gene editing technology now allows neuroscientists unprecedented access to edit the genome efficiently. Although many important tools have been developed, here we focus on approaches that allow for rapid gene editing in the adult nervous system, particularly CRISPR/Cas9 and anti-sense nucleotide-based techniques. CRISPR/Cas9 is a flexible gene editing tool, allowing the genome to be manipulated in diverse ways. For instance, CRISPR/Cas9 has been successfully used to knockout genes, knock-in mutations, overexpress or inhibit gene activity, and provide scaffolding for recruiting specific epigenetic regulators to individual genes and gene regions. Moreover, the CRISPR/Cas9 system may be modified to target multiple genes at one time, affording simultaneous inhibition and overexpression of distinct genetic targets. Although many of the more advanced applications of CRISPR/Cas9 have not been applied to the nervous system, the toolbox is widely accessible, such that it is poised to help advance neuroscience. Anti-sense nucleotide-based technologies can be used to rapidly knockdown genes in the brain. The main advantage of anti-sense based tools is their simplicity, allowing for rapid gene delivery with minimal technical expertise. Here, we describe the main applications and functions of each of these systems with an emphasis on their many potential applications in neuroscience laboratories.
Collapse
Affiliation(s)
- Brandon J Walters
- Department of Neuroscience and Mental Health, The Hospital for Sick Children Toronto, ON, Canada
| | - Amber B Azam
- Department of Psychology, University of Toronto Mississauga Mississauga, ON, Canada
| | - Colleen J Gillon
- Department of Neuroscience and Mental Health, The Hospital for Sick ChildrenToronto, ON, Canada; Department of Physiology, University of TorontoToronto, ON, Canada
| | - Sheena A Josselyn
- Department of Neuroscience and Mental Health, The Hospital for Sick ChildrenToronto, ON, Canada; Department of Physiology, University of TorontoToronto, ON, Canada
| | - Iva B Zovkic
- Department of Psychology, University of Toronto Mississauga Mississauga, ON, Canada
| |
Collapse
|
6
|
Abstract
Silencing of abnormally activated genes can be accomplished in a highly specific manner using nucleic acid based approaches. The focus of this review includes the different nucleic acid based inhibition strategies such as antisense oligodeoxynucleotides, small interfering RNA (siRNA), dominant-negative constructs, G-quartet oligonucleotides and decoy oligonucleotides, their mechanism of action and the effectiveness of these approaches to targeting the STAT (signal transducer and activator of transcription) proteins in cancer. Among the STAT proteins, especially STAT3, followed by STAT5, are the most frequently activated oncogenic STATs, which have emerged as plausible therapeutic cancer targets. Both STAT3 and STAT5 have been shown to regulate numerous oncogenic signaling pathways including proliferation, survival, angiogenesis and migration/invasion.
Collapse
Affiliation(s)
- Malabika Sen
- Department of Otolaryngology; University of Pittsburgh School of Medicine; Pittsburgh, PA USA
| | | |
Collapse
|
7
|
Zhou Y, Ness SA. Myb proteins: angels and demons in normal and transformed cells. Front Biosci (Landmark Ed) 2011; 16:1109-31. [PMID: 21196221 DOI: 10.2741/3738] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A key regulator of proliferation, differentiation and cell fate, the c-Myb transcription factor regulates the expression of hundreds of genes and is in turn regulated by numerous pathways and protein interactions. However, the most unique feature of c-Myb is that it can be converted into an oncogenic transforming protein through a few mutations that completely change its activity and specificity. The c-Myb protein is a myriad of interactions and activities rolled up in a protein that controls proliferation and differentiation in many different cell types. Here we discuss the background and recent progress that have led to a better understanding of this complex protein, and outline the questions that have yet to be answered.
Collapse
Affiliation(s)
- Ye Zhou
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131-0001, USA
| | | |
Collapse
|
8
|
Assessment of a balloon-tipped catheter modified for intracerebral convection-enhanced delivery. J Neurooncol 2008; 89:159-68. [DOI: 10.1007/s11060-008-9612-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2007] [Accepted: 04/22/2008] [Indexed: 10/22/2022]
|
9
|
Abstract
Gliomas are the most common major subgroup of primary CNS tumours. Approximately 17,000 new cases are reported each year and, of these, 11,500 patients die. Glioblastoma multiforme (GBM) is highly proliferative and typically invades distal portions of the brain, thereby making complete surgical resection of these tumours nearly impossible. Moreover, GBMs are often resistant to current chemotherapy and radiation regimens. Therefore, there is a need for better therapeutic interventions. One class of proteins that is involved in the formation of malignant brain tumours is protein kinase C (PKC) and these kinases have not been thoroughly explored for their chemotherapeutic value in GBMs. The PKC isozyme, PKCeta (PKC-eta) increases cell proliferation and resistance to radiation of GBM cell lines. These properties make PKCeta an attractive target for chemotherapeutic intervention in the management of GBMs.
Collapse
Affiliation(s)
- Patrick M Martin
- Department of Pathology, University of Virginia, Charlottesville, VA, USA.
| | | |
Collapse
|
10
|
Phuphanich S, Brat DJ, Olson JJ. Delivery systems and molecular targets of mechanism-based therapies for GBM. Expert Rev Neurother 2006; 4:649-63. [PMID: 15853584 DOI: 10.1586/14737175.4.4.649] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common malignant brain tumor of adults and is in great need of novel diagnostic and therapeutic approaches. Diagnosis is beginning to consider a tumor's genetic status and in the future may incorporate gene expression or proteomic profiles. Genetic alterations in gliomas that are being used in classification include TP53 and retinoblastoma pathway disruption, PTEN mutations, epidermal growth factor receptor amplification and 1p/19q losses. Molecular mechanisms are being exploited to treat glioblastoma multiforme. Tyrosine kinase inhibitors directed at epidermal growth factor receptor (ZD1839, OSI-774) are being explored. Farnesyltransferase inhibitors (R115777) block activation of the ras pathway and may be effective. Antagonists of the endothelin receptor (e.g., atrasentan) expressed on blood vessels may block the high degree of angiogenesis in gliomas. Tumors lacking methylthioadenosine phosphorylase are sensitive to inhibitors of de novo adenosine synthesis (SDX-102) since they lack a salvage pathway. Future goals are to tailor therapies to a tumor's molecular, proteomic or genomic status ,and manage glioblastoma multiformes as in chronic diseases in a multidisciplinary clinical setting.
Collapse
Affiliation(s)
- Surasak Phuphanich
- Department of Hematology-Oncology, Winship Cancer Institute, Suite C-5002, 1365 C Clifton Road NE, Atlanta, GA 30322, USA.
| | | | | |
Collapse
|
11
|
Zhang Z, Weinschenk T, Schluesener HJ. Uptake, cellular distribution and novel cellular binding proteins of immunostimulatory CpG oligodeoxynucleotides in glioblastoma cells. Mol Cell Biochem 2005; 272:35-46. [PMID: 16010970 DOI: 10.1007/s11010-005-6605-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Glioblastomas are the most malignant and most frequent brain tumors and exciting targets of gene and immunotherapy. Despite rapid development of experimental therapy little is known about the cellular behaviour of therapeutic oligodeoxynucleotides (ODNs). Here we designed uptake, cellular distribution and cellular binding proteins of immunostimulatory CpG-ODNs in glioblastoma cells by flow cytometry, fluorescence microscopy and mass spectrometry. Our data show that the phosphorothioate (PS) CpG-ODNs uptake in T98G and C6 cells is dose-, time-, temperature-dependent and independent of the CpG dinucleotides. Uptake can be inhibited by sodium azide, polyanions but not by chloroquine. After internalisation FITC labelled CpG-ODNs showed a spotted distribution in cytoplasm. Dozens of cellular binding proteins were identified using mass spectrometry. The binding of ODNs to proteins is dependent on modification and sequence but independent on CpG motif. ODNs bind to cellular proteins that are important for RNA processing and transport. Furthermore, three novel membrane proteins were identified, which might contribute to uptake of ODNs. ODNs binding to these proteins might interfere with the physiological function and thus might cause unwanted effects. Such binding also might influence the uptake efficiency or cellular distribution of therapeutic ODNs.
Collapse
Affiliation(s)
- Zhiren Zhang
- Institute of Brain Research, University of Tuebingen, Tuebingen, Germany.
| | | | | |
Collapse
|
12
|
Agius LM. Justification of glioma biology beyond a cellular basis of interpretation. Med Hypotheses 2003; 61:486-94. [PMID: 13679018 DOI: 10.1016/s0306-9877(03)00202-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Gliomas as neoplasms primarily arising from and constituted by glial cells would appear to implicate cell types that inherently reflect variation of aspects of a putative reparative process. The prominence of an astrocytic type cell of origin would further perhaps constitute a system of malignant transformation based on aberrant progression in cell proliferation and of cell pathology related to aspects on one hand of a gliosis and on the other of an autonomous process of progressiveness. In such terms, perhaps, one might consider the molecular aspects of gliomatous pathogenesis as simply a process of integral aberration of various aspects of astrocytic or glial cell responsiveness outside the normal confines of the normal reparative process and inherently beyond a strict cellular basis of interpretation in pathobiologic terms of such processes as anti-apoptosis and amplification of growth factor receptivity.
Collapse
Affiliation(s)
- L M Agius
- St. Luke's Hospital, Gwardamangia, University of Malta, Malta.
| |
Collapse
|
13
|
Shi W, Siemann DW. Inhibition of renal cell carcinoma angiogenesis and growth by antisense oligonucleotides targeting vascular endothelial growth factor. Br J Cancer 2002; 87:119-26. [PMID: 12085267 PMCID: PMC2364273 DOI: 10.1038/sj.bjc.6600416] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2002] [Revised: 04/22/2002] [Accepted: 04/23/2002] [Indexed: 01/12/2023] Open
Abstract
Angiogenesis is critical for growth and metastatic spread of solid tumours. It is tightly controlled by specific regulatory factors. Vascular endothelial growth factor has been implicated as the key factor in tumour angiogenesis. In the present studies we evaluated the effects of blocking vascular endothelial growth factor production by antisense phosphorothioate oligodeoxynucleotides on the growth and angiogenic activity of a pre-clinical model of renal cell carcinoma (Caki-1). In vitro studies showed that treating Caki-1 cells with antisense phosphorothioate oligodeoxynucleotides directed against vascular endothelial growth factor mRNA led to a reduction in expressed vascular endothelial growth factor levels sufficient to impair the proliferation and migration of co-cultured endothelial cells. The observed effects were antisense sequence specific, dose dependent, and could be achieved at a low, non-toxic concentration of phosphorothioate oligodeoxynucleotides. When vascular endothelial growth factor antisense treated Caki-1 cells were injected into nude mice and evaluated for their angiogenic potential, the number of vessels initiated were approximately half that induced by untreated Caki-1 cells. To test the anti-tumour efficacy of vascular endothelial growth factor antisense, phosphorothioate oligodeoxynucleotides were administrated to nude mice bearing macroscopic Caki-1 xenografts. The results showed that the systemic administration of two doses of vascular endothelial growth factor antisense phosphorothioate oligodeoxynucleotides given 1 and 4 days after the tumours reached a size of approximately 200 mm(3) significantly increased the time for tumours to grow to 1000 mm(3).
Collapse
Affiliation(s)
- W Shi
- Department of Pharmacology and Experimental Therapeutics, University of Florida, Box 100267, 1600 SW Archer Road, Gainesville, FL 32610, USA
| | | |
Collapse
|
14
|
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
|
15
|
Elepfandt P, Rupprecht S, Schöning-Burkhardt B, Volk HD, Woiciechowsky C. Oligodeoxynucleotides induce brain inflammation in rats when infused intracerebroventricularly. Neurosci Lett 2002; 322:107-10. [PMID: 11958855 DOI: 10.1016/s0304-3940(02)00093-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Antisense oligodeoxynucleotides (ODNs) can inhibit gene expression in a specific manner. However, several studies described problems with cerebral ODN application. Here, we investigated the immune effects (interleukin-6 (IL-6) release, cell invasion into cerebrospinal fluid (CSF) and brain parenchyma) of 'non-sense' randomized ODNs with different counterions (NH(4)(+), Na(+)) and modifications (with or without thioat-backbone) which were administered intracerebroventricularly for 48 h using osmotic mini-pumps in a rat model. All animals receiving ODNs showed increased IL-6 levels in the CSF as well as cell invasion into the CSF and brain parenchyma (P<0.05). However, the use of thioat-backbone and ammonium as the counterion induced the highest IL-6 levels (7210+/-1696 pg/ml, P<0.05) and the highest cell numbers in the CSF (31.6+/-15.5x10(5)/ml, P<0.05) as well as brain parenchyma (268.1+/-143.2 HIS-48+ cells/mm(2), P<0.01; and 31.3+/-10.7 OX-6+cells/mm(2), P<0.05) compared with the other groups.
Collapse
Affiliation(s)
- Peter Elepfandt
- Department of Neurosurgery, Charité Medical School, Humboldt University of Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | | | | | | | | |
Collapse
|
16
|
Brandes AA, Basso U, Pasetto LM. Changing boundaries in the treatment of malignant gliomas. Expert Rev Anticancer Ther 2001; 1:357-70. [PMID: 12113103 DOI: 10.1586/14737140.1.3.357] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Malignant gliomas are still among the most lethal and difficult tumors to treat; even the most intensive combinations of radio- and chemotherapy are not curative and yield only a modest impact on survival for most of these patients, as long-term survivors are less than 10%. There is a major need for new chemotherapeutic drugs and alternative therapeutic modalities. This review aims to define the best standard treatment in the common clinical practice and also summarizes the most promising lines of investigational research in the field of neuro-oncology, which will probably offer new and long-awaited valid therapy options for brain tumor patients.
Collapse
Affiliation(s)
- A A Brandes
- Divisione di Oncologia-Direzione Azienda Ospedale-Universita, Via Giustiniani 2, 35100 Padova, Italy.
| | | | | |
Collapse
|
17
|
Stoessl AJ. Antisense strategies for the treatment of neurological disease. Expert Opin Ther Pat 2001. [DOI: 10.1517/13543776.11.4.547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
18
|
Engelhard HH, Duncan HA, Kim S, Criswell PS, Van Eldik L. Therapeutic effects of sodium butyrate on glioma cells in vitro and in the rat C6 glioma model. Neurosurgery 2001; 48:616-24; discussion 624-5. [PMID: 11270553 DOI: 10.1097/00006123-200103000-00035] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVE Preliminary in vitro studies have indicated that sodium butyrate inhibits the proliferation of cultured glioma cells and induces cellular differentiation, making it potentially useful as a therapeutic agent for patients with glioblastoma multiforme. The purpose of this study was to expand on the preliminary research by investigating the effects of sodium butyrate on multiple cell lines, explanted cells from glioblastoma tumor specimens, and in vivo in the rat C6 glioma brain tumor model. METHODS Four malignant glioma cell lines (A-172, T98G, U118MG, and C6) and two primary cell cultures derived from human glioblastoma tumor specimens were treated with 2 mmol/L sodium butyrate for up to 72 hours. Sodium butyrate-induced effects on cell morphology, proliferation, cell cycle distribution, migration, glial fibrillary acidic protein staining, and S100beta protein content were determined. For in vivo studies, a total of 64 male Wistar-Furth rats underwent operations to implant C6 glioma cells stereotactically or were used as controls. The rats were treated with escalating doses of sodium butyrate by microinfusion with Alzet minipumps (Durect Corp., Cupertino, CA). RESULTS Sodium butyrate treatment in vitro produced changes in morphology and glial fibrillary acidic protein expression indicative of cellular differentiation. In cell lines and explanted cells, sodium butyrate consistently inhibited glioblastoma cell proliferation (to 51 +/- 6% that of controls) and migration (to 46 +/- 17%). Intratumoral infusion of 40 mmol/L sodium butyrate prolonged the survival of Wistar-Furth rats with intracerebral C6 tumors (P = 0.013) without detectable toxicity. CONCLUSION These data support further consideration of direct interstitial infusion of sodium butyrate in a Phase I clinical study for patients with recurrent glioblastoma multiforme.
Collapse
Affiliation(s)
- H H Engelhard
- Department of Neurosurgery, University of Illinois at Chicago, 60612, USA.
| | | | | | | | | |
Collapse
|
19
|
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
|
20
|
Abstract
BACKGROUND Over the past two decades, significant advances have been made in the fields of virology and molecular biology, and in understanding the genetic alterations present in brain tumors. The knowledge gained has been exploited for use in gene therapy. OBJECTIVE The purpose of this article is to present an introduction to the field of brain tumor gene therapy for the practicing clinician. RESULTS A variety of gene therapy strategies have now been used in the laboratory and in clinical trials for brain tumors. They can be divided into five categories: 1) gene-directed enzyme prodrug ("suicide gene") therapy (GDEPT); 2) gene therapy designed to boost the activity of the immune system against cancer cells; 3) oncolytic virus therapy; 4) transfer of potentially therapeutic genes--such as tumor suppressor genes--into cancer cells; and 5) antisense therapy. GDEPT is the strategy that has been most extensively studied. CONCLUSIONS To date, gene therapy has been found to be reasonably safe and concerns related to adverse events such as insertional mutagenesis have not been realized. Although patients have not been cured, the development of this therapy could still be considered to be at an early stage. Current research is addressing factors that could be limiting the successful clinical application of gene therapy, which remains an intriguing experimental option for patients with malignant brain tumors.
Collapse
Affiliation(s)
- H H Engelhard
- Departments of Neurosurgery and Molecular Genetics, The University of Illinois at Chicago, Chicago, Illinois 60612, USA
| |
Collapse
|