1
|
Kameda-Smith MM, Zhu H, Luo EC, Suk Y, Xella A, Yee B, Chokshi C, Xing S, Tan F, Fox RG, Adile AA, Bakhshinyan D, Brown K, Gwynne WD, Subapanditha M, Miletic P, Picard D, Burns I, Moffat J, Paruch K, Fleming A, Hope K, Provias JP, Remke M, Lu Y, Reya T, Venugopal C, Reimand J, Wechsler-Reya RJ, Yeo GW, Singh SK. Characterization of an RNA binding protein interactome reveals a context-specific post-transcriptional landscape of MYC-amplified medulloblastoma. Nat Commun 2022; 13:7506. [PMID: 36473869 PMCID: PMC9726987 DOI: 10.1038/s41467-022-35118-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 11/18/2022] [Indexed: 12/12/2022] Open
Abstract
Pediatric medulloblastoma (MB) is the most common solid malignant brain neoplasm, with Group 3 (G3) MB representing the most aggressive subgroup. MYC amplification is an independent poor prognostic factor in G3 MB, however, therapeutic targeting of the MYC pathway remains limited and alternative therapies for G3 MB are urgently needed. Here we show that the RNA-binding protein, Musashi-1 (MSI1) is an essential mediator of G3 MB in both MYC-overexpressing mouse models and patient-derived xenografts. MSI1 inhibition abrogates tumor initiation and significantly prolongs survival in both models. We identify binding targets of MSI1 in normal neural and G3 MB stem cells and then cross referenced these data with unbiased large-scale screens at the transcriptomic, translatomic and proteomic levels to systematically dissect its functional role. Comparative integrative multi-omic analyses of these large datasets reveal cancer-selective MSI1-bound targets sharing multiple MYC associated pathways, providing a valuable resource for context-specific therapeutic targeting of G3 MB.
Collapse
Affiliation(s)
- Michelle M. Kameda-Smith
- grid.25073.330000 0004 1936 8227Centre for Discovery in Cancer Research (CDCR), McMaster University, Hamilton, ON Canada ,grid.25073.330000 0004 1936 8227Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON Canada ,grid.25073.330000 0004 1936 8227Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON Canada
| | - Helen Zhu
- grid.419890.d0000 0004 0626 690XComputational Biology Program, Ontario Institute for Cancer Research, Toronto, Canada ,grid.17063.330000 0001 2157 2938Department of Medical Biophysics, University of Toronto, Toronto, Canada ,grid.231844.80000 0004 0474 0428University Health Network, Toronto, ON Canada ,grid.494618.6Vector Institute Toronto, Toronto, ON Canada
| | - En-Ching Luo
- grid.266100.30000 0001 2107 4242Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA USA ,grid.266100.30000 0001 2107 4242Stem Cell Program, University of California San Diego, La Jolla, CA USA ,grid.468218.10000 0004 5913 3393Sanford Consortium for Regenerative Medicine, La Jolla, CA USA
| | - Yujin Suk
- grid.25073.330000 0004 1936 8227Centre for Discovery in Cancer Research (CDCR), McMaster University, Hamilton, ON Canada ,grid.25073.330000 0004 1936 8227Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON Canada ,grid.25073.330000 0004 1936 8227Michael G DeGroote School of Medicine, McMaster University, Hamilton, Canada
| | - Agata Xella
- grid.479509.60000 0001 0163 8573Tumor Initiation and Maintenance Program, National Cancer Institute-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA USA
| | - Brian Yee
- grid.266100.30000 0001 2107 4242Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA USA ,grid.266100.30000 0001 2107 4242Stem Cell Program, University of California San Diego, La Jolla, CA USA ,grid.468218.10000 0004 5913 3393Sanford Consortium for Regenerative Medicine, La Jolla, CA USA
| | - Chirayu Chokshi
- grid.25073.330000 0004 1936 8227Centre for Discovery in Cancer Research (CDCR), McMaster University, Hamilton, ON Canada ,grid.25073.330000 0004 1936 8227Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON Canada
| | - Sansi Xing
- grid.25073.330000 0004 1936 8227Centre for Discovery in Cancer Research (CDCR), McMaster University, Hamilton, ON Canada ,grid.25073.330000 0004 1936 8227Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON Canada
| | - Frederick Tan
- grid.266100.30000 0001 2107 4242Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA USA ,grid.266100.30000 0001 2107 4242Stem Cell Program, University of California San Diego, La Jolla, CA USA ,grid.468218.10000 0004 5913 3393Sanford Consortium for Regenerative Medicine, La Jolla, CA USA
| | - Raymond G. Fox
- grid.266100.30000 0001 2107 4242Departments of Pharmacology and Medicine, University of California San Diego School of Medicine, Sanford Consortium for Regenerative Medicine, La Jolla, CA USA
| | - Ashley A. Adile
- grid.25073.330000 0004 1936 8227Centre for Discovery in Cancer Research (CDCR), McMaster University, Hamilton, ON Canada ,grid.25073.330000 0004 1936 8227Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON Canada
| | - David Bakhshinyan
- grid.25073.330000 0004 1936 8227Centre for Discovery in Cancer Research (CDCR), McMaster University, Hamilton, ON Canada ,grid.25073.330000 0004 1936 8227Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON Canada
| | - Kevin Brown
- grid.17063.330000 0001 2157 2938Donnelly Centre, Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - William D. Gwynne
- grid.25073.330000 0004 1936 8227Centre for Discovery in Cancer Research (CDCR), McMaster University, Hamilton, ON Canada ,grid.25073.330000 0004 1936 8227Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON Canada
| | - Minomi Subapanditha
- grid.25073.330000 0004 1936 8227Centre for Discovery in Cancer Research (CDCR), McMaster University, Hamilton, ON Canada
| | - Petar Miletic
- grid.25073.330000 0004 1936 8227Centre for Discovery in Cancer Research (CDCR), McMaster University, Hamilton, ON Canada ,grid.25073.330000 0004 1936 8227Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON Canada
| | - Daniel Picard
- grid.14778.3d0000 0000 8922 7789Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Ian Burns
- grid.25073.330000 0004 1936 8227Michael G DeGroote School of Medicine, McMaster University, Hamilton, Canada
| | - Jason Moffat
- grid.17063.330000 0001 2157 2938Donnelly Centre, Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Kamil Paruch
- grid.10267.320000 0001 2194 0956Department of Chemistry, CZ Openscreen, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic ,grid.483343.bInternational Clinical Research Center, St. Anne’s University Hospital in Brno, 602 00 Brno, Czech Republic
| | - Adam Fleming
- grid.25073.330000 0004 1936 8227McMaster University, Departments of Pediatrics, Hematology and Oncology Division, Hamilton, Canada
| | - Kristin Hope
- grid.25073.330000 0004 1936 8227Centre for Discovery in Cancer Research (CDCR), McMaster University, Hamilton, ON Canada ,grid.25073.330000 0004 1936 8227Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON Canada
| | - John P. Provias
- grid.25073.330000 0004 1936 8227McMaster University, Departments of Neuropathology, Hamilton, Canada
| | - Marc Remke
- grid.14778.3d0000 0000 8922 7789Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Yu Lu
- grid.25073.330000 0004 1936 8227Centre for Discovery in Cancer Research (CDCR), McMaster University, Hamilton, ON Canada ,grid.25073.330000 0004 1936 8227Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON Canada
| | - Tannishtha Reya
- grid.266100.30000 0001 2107 4242Departments of Pharmacology and Medicine, University of California San Diego School of Medicine, Sanford Consortium for Regenerative Medicine, La Jolla, CA USA ,grid.239585.00000 0001 2285 2675Present Address: Herbert Irving Comprehensive Cancer Center, Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY USA
| | - Chitra Venugopal
- grid.25073.330000 0004 1936 8227Centre for Discovery in Cancer Research (CDCR), McMaster University, Hamilton, ON Canada ,grid.25073.330000 0004 1936 8227Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON Canada
| | - Jüri Reimand
- grid.419890.d0000 0004 0626 690XComputational Biology Program, Ontario Institute for Cancer Research, Toronto, Canada ,grid.17063.330000 0001 2157 2938Department of Medical Biophysics, University of Toronto, Toronto, Canada ,grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Robert J. Wechsler-Reya
- grid.479509.60000 0001 0163 8573Tumor Initiation and Maintenance Program, National Cancer Institute-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA USA ,grid.239585.00000 0001 2285 2675Present Address: Herbert Irving Comprehensive Cancer Center, Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY USA
| | - Gene W. Yeo
- grid.266100.30000 0001 2107 4242Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA USA ,grid.266100.30000 0001 2107 4242Stem Cell Program, University of California San Diego, La Jolla, CA USA ,grid.468218.10000 0004 5913 3393Sanford Consortium for Regenerative Medicine, La Jolla, CA USA
| | - Sheila K. Singh
- grid.25073.330000 0004 1936 8227Centre for Discovery in Cancer Research (CDCR), McMaster University, Hamilton, ON Canada ,grid.25073.330000 0004 1936 8227Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON Canada ,grid.25073.330000 0004 1936 8227Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON Canada ,grid.25073.330000 0004 1936 8227McMaster University, Department of Pediatrics, Hamilton, Canada
| |
Collapse
|
2
|
Totsch SK, Schlappi C, Kang KD, Ishizuka AS, Lynn GM, Fox B, Beierle EA, Whitley RJ, Markert JM, Gillespie GY, Bernstock JD, Friedman GK. Oncolytic herpes simplex virus immunotherapy for brain tumors: current pitfalls and emerging strategies to overcome therapeutic resistance. Oncogene 2019; 38:6159-6171. [PMID: 31289361 PMCID: PMC6771414 DOI: 10.1038/s41388-019-0870-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/21/2019] [Accepted: 06/22/2019] [Indexed: 12/25/2022]
Abstract
Malignant tumors of the central nervous system (CNS) continue to be a leading cause of cancer-related mortality in both
children and adults. Traditional therapies for malignant brain tumors consist of surgical resection and adjuvant chemoradiation;
such approaches are often associated with extreme morbidity. Accordingly, novel, targeted therapeutics for neoplasms of the CNS,
such as immunotherapy with oncolytic engineered herpes simplex virus (HSV) therapy, are urgently warranted. Herein, we discuss
treatment challenges related to HSV virotherapy delivery, entry, replication, and spread, and in so doing focus on host antiviral
immune responses and the immune microenvironment. Strategies to overcome such challenges including viral re-engineering,
modulation of the immunoregulatory microenvironment and combinatorial therapies with virotherapy, such as checkpoint inhibitors,
radiation, and vaccination are also examined in detail.
Collapse
Affiliation(s)
- Stacie K Totsch
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Charles Schlappi
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kyung-Don Kang
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - Brandon Fox
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Elizabeth A Beierle
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Richard J Whitley
- Division of Pediatric Infectious Disease, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - James M Markert
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - G Yancey Gillespie
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Joshua D Bernstock
- Avidea Technologies, Inc, Baltimore, MD, USA. .,Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, USA. .,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Gregory K Friedman
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA. .,Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA. .,Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
3
|
Yokoda R, Nagalo BM, Vernon B, Oklu R, Albadawi H, DeLeon TT, Zhou Y, Egan JB, Duda DG, Borad MJ. Oncolytic virus delivery: from nano-pharmacodynamics to enhanced oncolytic effect. Oncolytic Virother 2017; 6:39-49. [PMID: 29184854 PMCID: PMC5687448 DOI: 10.2147/ov.s145262] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
With the advancement of a growing number of oncolytic viruses (OVs) to clinical development, drug delivery is becoming an important barrier to overcome for optimal therapeutic benefits. Host immunity, tumor microenvironment and abnormal vascularity contribute to inefficient vector delivery. A number of novel approaches for enhanced OV delivery are under evaluation, including use of nanoparticles, immunomodulatory agents and complex viral–particle ligands along with manipulations of the tumor microenvironment. This field of OV delivery has quickly evolved to bioengineering of complex nanoparticles that could be deposited within the tumor using minimal invasive image-guided delivery. Some of the strategies include ultrasound (US)-mediated cavitation-enhanced extravasation, magnetic viral complexes delivery, image-guided infusions with focused US and targeting photodynamic virotherapy. In addition, strategies that modulate tumor microenvironment to decrease extracellular matrix deposition and increase viral propagation are being used to improve tumor penetration by OVs. Some involve modification of the viral genome to enhance their tumoral penetration potential. Here, we highlight the barriers to oncolytic viral delivery, and discuss the challenges to improving it and the perspectives of establishing new modes of active delivery to achieve enhanced oncolytic effects.
Collapse
Affiliation(s)
- Raquel Yokoda
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Bolni M Nagalo
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Brent Vernon
- Department of Biomedical Engineering, Arizona State University, Tempe
| | - Rahmi Oklu
- Division of Vascular and Interventional Radiology, Department of Radiology, Mayo Clinic, Scottsdale, AZ
| | - Hassan Albadawi
- Division of Vascular and Interventional Radiology, Department of Radiology, Mayo Clinic, Scottsdale, AZ
| | - Thomas T DeLeon
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Yumei Zhou
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Jan B Egan
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| | - Dan G Duda
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Mitesh J Borad
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale
| |
Collapse
|
4
|
Campadelli-Fiume G, Petrovic B, Leoni V, Gianni T, Avitabile E, Casiraghi C, Gatta V. Retargeting Strategies for Oncolytic Herpes Simplex Viruses. Viruses 2016; 8:63. [PMID: 26927159 PMCID: PMC4810253 DOI: 10.3390/v8030063] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 12/22/2015] [Accepted: 12/30/2015] [Indexed: 02/08/2023] Open
Abstract
Most of the oncolytic herpes simplex viruses (HSVs) exhibit a high safety profile achieved through attenuation. They carry defects in virulence proteins that antagonize host cell response to the virus, including innate response, apoptosis, authophagy, and depend on tumor cell proliferation. They grow robustly in cancer cells, provided that these are deficient in host cell responses, which is often the case. To overcome the attenuation limits, a strategy is to render the virus highly cancer-specific, e.g., by retargeting their tropism to cancer-specific receptors, and detargeting from natural receptors. The target we selected is HER-2, overexpressed in breast, ovarian and other cancers. Entry of wt-HSV requires the essential glycoproteins gD, gH/gL and gB. Here, we reviewed that oncolytic HSV retargeting was achieved through modifications in gD: the addition of a single-chain antibody (scFv) to HER-2 coupled with appropriate deletions to remove part of the natural receptors' binding sites. Recently, we showed that also gH/gL can be a retargeting tool. The insertion of an scFv to HER-2 at the gH N-terminus, coupled with deletions in gD, led to a recombinant capable to use HER-2 as the sole receptor. The retargeted oncolytic HSVs can be administered systemically by means of carrier cells-forcedly-infected mesenchymal stem cells. Altogether, the retargeted oncolytic HSVs are highly cancer-specific and their replication is not dependent on intrinsic defects of the tumor cells. They might be further modified to express immunomodulatory molecules.
Collapse
Affiliation(s)
- Gabriella Campadelli-Fiume
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy.
| | - Biljana Petrovic
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy.
| | - Valerio Leoni
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy.
| | - Tatiana Gianni
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy.
| | - Elisa Avitabile
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy.
| | - Costanza Casiraghi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy.
| | - Valentina Gatta
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy.
| |
Collapse
|
5
|
Cambuli FM, Correa BR, Rezza A, Burns SC, Qiao M, Uren PJ, Kress E, Boussouar A, Galante PAF, Penalva LOF, Plateroti M. A Mouse Model of Targeted Musashi1 Expression in Whole Intestinal Epithelium Suggests Regulatory Roles in Cell Cycle and Stemness. Stem Cells 2015; 33:3621-34. [PMID: 26303183 DOI: 10.1002/stem.2202] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 06/30/2015] [Accepted: 07/28/2015] [Indexed: 12/21/2022]
Abstract
The intestinal epithelium is very peculiar for its continuous cell renewal, fuelled by multipotent stem cells localized within the crypts of Lieberkühn. Several lines of evidence have established the evolutionary conserved RNA-binding protein Musashi1 as a marker of adult stem cells, including those of the intestinal epithelium, and revealed its roles in stem cell self-renewal and cell fate determination. Previous studies from our laboratories have shown that Musashi1 controls stem cell-like features in medulloblastoma, glioblastoma, and breast cancer cells, and has pro-proliferative and pro-tumorigenic properties in intestinal epithelial progenitor cells in vitro. To undertake a detailed study of Musashi1's function in the intestinal epithelium in vivo, we have generated a mouse model, referred to as v-Msi, overexpressing Musashi1 specifically in the entire intestinal epithelium. Compared with wild type litters, v-Msi1 mice exhibited increased intestinal crypt size accompanied by enhanced proliferation. Comparative transcriptomics by RNA-seq revealed Musashi1's association with gut stem cell signature, cell cycle, DNA replication, and drug metabolism. Finally, we identified and validated three novel mRNA targets that are stabilized by Musashi1, Ccnd1 (Cyclin D1), Cdk6, and Sox4. In conclusion, the targeted expression of Musashi1 in the intestinal epithelium in vivo increases the cell proliferation rate and strongly suggests its action on stem cells activity. This is due to the modulation of a complex network of gene functions and pathways including drug metabolism, cell cycle, and DNA synthesis and repair.
Collapse
Affiliation(s)
- F M Cambuli
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Lyon, France
| | - B R Correa
- Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, Texas, USA.,Centro de Oncologia Molecular, Hospital Sírio-Libanês, São Paulo, Brazil
| | - A Rezza
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Lyon, France
| | - S C Burns
- Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, Texas, USA
| | - M Qiao
- Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, Texas, USA
| | - P J Uren
- Molecular and Computational Biology Section, Division of Biological Sciences, University of Southern California, Los Angeles, California, USA
| | - E Kress
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Lyon, France
| | - A Boussouar
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Lyon, France
| | - P A F Galante
- Centro de Oncologia Molecular, Hospital Sírio-Libanês, São Paulo, Brazil
| | - L O F Penalva
- Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, Texas, USA.,Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, Texas, USA
| | - M Plateroti
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Lyon, France
| |
Collapse
|
6
|
Pediatric cancer gone viral. Part I: strategies for utilizing oncolytic herpes simplex virus-1 in children. MOLECULAR THERAPY-ONCOLYTICS 2015; 2:S2372-7705(16)30017-1. [PMID: 26436135 PMCID: PMC4589755 DOI: 10.1038/mto.2015.15] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Progress for improving outcomes in pediatric patients with solid tumors remains slow. In addition, currently available therapies are fraught with numerous side effects, often causing significant life-long morbidity for long-term survivors. The use of viruses to kill tumor cells based on their increased vulnerability to infection is gaining traction, with several viruses moving through early and advanced phase clinical testing. The prospect of increased efficacy and decreased toxicity with these agents is thus attractive for pediatric cancer. In part I of this two-part review, we focus on strategies for utilizing oncolytic engineered herpes simplex virus (HSV) to target pediatric malignancies. We discuss mechanisms of action, routes of delivery, and the role of preexisting immunity on antitumor efficacy. Challenges to maximizing oncolytic HSV in children are examined, and we highlight how these may be overcome through various arming strategies. We review the preclinical and clinical evidence demonstrating safety of a variety of oncolytic HSVs. In Part II, we focus on the antitumor efficacy of oncolytic HSV in pediatric tumor types, pediatric clinical advances made to date, and future prospects for utilizing HSV in pediatric patients with solid tumors.
Collapse
|
7
|
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
|
8
|
Ady JW, Heffner J, Klein E, Fong Y. Oncolytic viral therapy for pancreatic cancer: current research and future directions. Oncolytic Virother 2014; 3:35-46. [PMID: 27512661 PMCID: PMC4918362 DOI: 10.2147/ov.s53858] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The development of targeted agents and chemotherapies for pancreatic cancer has only modestly affected clinical outcome and not changed 5-year survival. Fortunately the genetic and molecular mechanisms underlying pancreatic cancer are being rapidly uncovered and are providing opportunities for novel targeted therapies. Oncolytic viral therapy is one of the most promising targeted agents for pancreatic cancer. This review will look at the current state of the development of these self-replicating nanoparticles in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Justin W Ady
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Jacqueline Heffner
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Elizabeth Klein
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Yuman Fong
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
9
|
Xu C, Li H, Su C, Li Z. Viral therapy for pancreatic cancer: tackle the bad guys with poison. Cancer Lett 2013; 333:1-8. [PMID: 23354590 DOI: 10.1016/j.canlet.2013.01.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 01/15/2013] [Accepted: 01/18/2013] [Indexed: 12/15/2022]
Abstract
Pancreatic cancer is one of the most devastating diseases with very poor prognosis. Only a small proportion is curable by surgical resection, whilst standard chemotherapy for patients with advanced disease has only modest effect with substantial toxicity. Therefore, there is an urgent need for the development of novel therapeutic approaches to improve the patient outcome. Recently the viral therapy is emerging as a novel effective therapeutic approach for cancer with the potential to selectively treat both primary tumor and metastatic lesions. This review provides an overview of the current status of viral treatment for pancreatic cancer, both in the laboratories and in clinical settings.
Collapse
Affiliation(s)
- Can Xu
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | | | | | | |
Collapse
|
10
|
Wirth T. A short perspective on gene therapy: Clinical experience on gene therapy of gliomablastoma multiforme. World J Exp Med 2011; 1:10-6. [PMID: 24520527 PMCID: PMC3905579 DOI: 10.5493/wjem.v1.i1.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 12/12/2011] [Accepted: 12/16/2011] [Indexed: 02/06/2023] Open
Abstract
More than two decades have passed since the first gene therapy clinical trial was conducted. During this time, we have gained much knowledge regarding gene therapy in general, but also learned to understand the fear that persists in society. We have experienced drawbacks and successes. More than 1700 clinical trials have been conducted where gene therapy is used as a means for therapy. In the very first trial, patients with advanced melanoma were treated with tumor infiltrating lymphocytes genetically modified ex-vivo to express tumor necrosis factor. Around the same time the first gene therapy trial was conducted, the ethical aspects of performing gene therapy on humans was intensively discussed. What are the risks involved with gene therapy? Can we control the technology? What is ethically acceptable and what are the indications gene therapy can be used for? Initially, gene therapy was thought to be implemented mainly for the treatment of monogenetic diseases, such as adenosine deaminase deficiency. However, other therapeutic areas have become of interest and currently cancer is the most studied therapeutic area for gene therapy based medicines. In this review I will be giving a short introduction into gene therapy and will direct the discussion to where we should go from here. Furthermore, I will focus on the use of the Herpes simplex virus-thymidine kinase for gene therapy of malignant gliomas and highlight the efficacy of gene therapy for the treatment of malignant gliomas, but other strategies will also be mentioned.
Collapse
Affiliation(s)
- Thomas Wirth
- Thomas Wirth, AI Virtanen Institute, Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Neulaniementie 2, FIN-70211 Kuopio, Finland
| |
Collapse
|
11
|
Rezza A, Skah S, Roche C, Nadjar J, Samarut J, Plateroti M. The overexpression of the putative gut stem cell marker Musashi-1 induces tumorigenesis through Wnt and Notch activation. J Cell Sci 2010; 123:3256-65. [PMID: 20826465 DOI: 10.1242/jcs.065284] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The RNA-binding protein Musashi-1 (Msi1) has been proposed as a marker of intestinal epithelial stem cells. These cells are responsible for the continuous renewal of the intestinal epithelium. Although the function of Msi1 has been studied in several organs from different species and in mammalian cell lines, its function and molecular regulation in mouse intestinal epithelium progenitor cells are still undefined. We describe here that, in these cells, the expression of Msi1 is regulated by the canonical Wnt pathway, through a mechanism involving a functional Tcf/Lef binding site on its promoter. An in vitro study in intestinal epithelium primary cultures showed that Msi1 overexpression promotes progenitor proliferation and activates Wnt and Notch pathways. Moreover, Msi1-overexpressing cells exhibit tumorigenic properties in xenograft experiments. These data point to a positive feedback loop between Msi1 and Wnt in intestinal epithelial progenitors. They also suggest that Msi1 has oncogenic properties in these cells, probably through induction of both the Wnt and Notch pathways.
Collapse
Affiliation(s)
- Amelie Rezza
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, INRA, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie, 69364 Lyon Cedex 07, France
| | | | | | | | | | | |
Collapse
|
12
|
Hukkanen V, Paavilainen H, Mattila RK. Host responses to herpes simplex virus and herpes simplex virus vectors. Future Virol 2010. [DOI: 10.2217/fvl.10.35] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Herpes simplex virus (HSV) is a well-known, ubiquitous pathogen of humans. Engineered mutants of HSV can also be exploited as vectors in gene therapy or for virotherapy of tumors. HSV has multiple abilities to evade and modulate the innate and adaptive responses of the host. The increasing knowledge on the mutual interactions of the invading HSV with the host defenses will contribute to our deeper understanding of the relationship between HSV and the host, and thereby lead to future development of more effective and specific HSV vectors for treatment of human diseases. The future advances of HSV vaccines and vaccine vectors are based on the knowlegde of the complex interplay between HSV and the host defenses.
Collapse
Affiliation(s)
| | - Henrik Paavilainen
- Department of Virology, University of Turku, Kiinamyllynkatu 13, FIN-20520 Turku, Finland
| | - Riikka K Mattila
- Institute of Diagnostics, University of Oulu, Aapistie 5A, FIN-90014, Finland
| |
Collapse
|
13
|
Shah AC, Parker JN, Shimamura M, Cassady KA. Spontaneous and Engineered Compensatory HSV Mutants that Counteract the Host Antiviral PKR Response. Viruses 2009; 1:510-22. [PMID: 21994558 PMCID: PMC3185541 DOI: 10.3390/v1030510] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2009] [Revised: 10/22/2009] [Accepted: 10/22/2009] [Indexed: 01/25/2023] Open
Abstract
A virulent recombinant HSV lacking the diploid γ134.5 gene (Δγ134.5) have been investigated over the last two decades both for anti-tumor therapy and as vaccine vectors. The first generation vectors, while safe, are incapable of sustained replication in the majority of treated patients. An interferon inducible host antiviral kinase, protein kinase R (PKR), limits late viral protein synthesis and replication of Δγ134.5 viruses. This review describes the development of new Δγ134.5 vectors, through serial passage selection and direct viral genome engineering, which demonstrate selective PKR evasion in targeted cells and improved viral replication without restoring neurovirulence.
Collapse
Affiliation(s)
- Amish C. Shah
- University of Pennsylvania, Department of Pediatrics, Children’s Hospital of Philadelphia, 34 Street and Civic Center Boulevard, Philadelphia, PA 19104-4399, USA; E-Mail: (A.C.S.)
| | - Jacqueline N. Parker
- University of Alabama at Birmingham, Department of Pediatrics, Division of Infectious Disease, 1600 6th Avenue South, CHB 118C, Birmingham, AL 35222, USA; E-Mails: (J.N.P.); (M.S.)
| | - Masako Shimamura
- University of Alabama at Birmingham, Department of Pediatrics, Division of Infectious Disease, 1600 6th Avenue South, CHB 118C, Birmingham, AL 35222, USA; E-Mails: (J.N.P.); (M.S.)
| | - Kevin A. Cassady
- University of Alabama at Birmingham, Department of Pediatrics, Division of Infectious Disease, 1600 6th Avenue South, CHB 118C, Birmingham, AL 35222, USA; E-Mails: (J.N.P.); (M.S.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-205-996-7910; Fax: +1-205-934-0460
| |
Collapse
|
14
|
Dorer DE, Nettelbeck DM. Targeting cancer by transcriptional control in cancer gene therapy and viral oncolysis. Adv Drug Deliv Rev 2009; 61:554-71. [PMID: 19394376 DOI: 10.1016/j.addr.2009.03.013] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Accepted: 03/05/2009] [Indexed: 01/02/2023]
Abstract
Cancer-specificity is the key requirement for a drug or treatment regimen to be effective against malignant disease--and has rarely been achieved adequately to date. Therefore, targeting strategies need to be implemented for future therapies to ensure efficient activity at the site of patients' tumors or metastases without causing intolerable side-effects. Gene therapy and viral oncolysis represent treatment modalities that offer unique opportunities for tumor targeting. This is because both the transfer of genes with anti-cancer activity and viral replication-induced cell killing, respectively, facilitate the incorporation of multiple mechanisms restricting their activity to cancer. To this end, cellular mechanisms of gene regulation have been successfully exploited to direct therapeutic gene expression and viral cell lysis to cancer cells. Here, transcriptional targeting has been the role model and most widely investigated. This approach exploits cellular gene regulatory elements that mediate cell type-specific transcription to restrict the expression of therapeutic genes or essential viral genes, ideally to cancer cells. In this review, we first discuss the rationale for such promoter targeting and its limitations. We then give an overview how tissue-/tumor-specific promoters are being identified and characterized. Strategies to apply and optimize such promoters for the engineering of targeted viral gene transfer vectors and oncolytic viruses-with respect to promoter size, selectivity and activity in the context of viral genomes-are described. Finally, we discuss in more detail individual examples for transcriptionally targeted virus drugs. First highlighting oncolytic viruses targeted by prostate-specific promoters and by the telomerase promoter as representatives of tissue-targeted and pan-cancer-specific virus drugs respectively, and secondly recent developments of the last two years.
Collapse
Affiliation(s)
- Dominik E Dorer
- Helmholtz-University Group Oncolytic Adenoviruses, German Cancer Research Center (DKFZ) and Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | | |
Collapse
|
15
|
Tyler MA, Sonabend AM, Ulasov IV, Lesniak MS. Vector therapies for malignant glioma: shifting the clinical paradigm. Expert Opin Drug Deliv 2008; 5:445-58. [PMID: 18426385 DOI: 10.1517/17425247.5.4.445] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Malignant glioma represents one of the most aggressive and devastating forms of human cancer. At present, there exists no successful treatment for this disease. Gene therapy, or vector therapy, has emerged as a viable experimental treatment method for intracranial malignancies. OBJECTIVE Vector therapy paradigms that have entered the clinical arena have shown adequate safety; however, the majority of the studies failed to observe significant clinical benefits. As such, researchers have refocused their efforts on developing novel vectors as well as new delivery methods to enhance the therapeutic effect of a particular vector. In this review, we discuss common vector therapy approaches used in clinical trials, their drawbacks and potential ways of overcoming these challenges. METHODS We focus on the experimental evaluation of cell-based vector therapies and adenoviral and herpes simplex virus type 1 vectors in the treatment of malignant glioma. CONCLUSION Vector therapy remains a promising treatment strategy for malignant glioma. Although significant questions remain to be answered, early clinical data suggest safety of this approach and future studies will likely address the efficacy of the proposed therapy.
Collapse
Affiliation(s)
- Matthew A Tyler
- University of Chicago, The Brain Tumor Center, 5841 S. Maryland Avenue, MC 3026, Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
16
|
Cattaneo R, Miest T, Shashkova EV, Barry MA. Reprogrammed viruses as cancer therapeutics: targeted, armed and shielded. Nat Rev Microbiol 2008; 6:529-40. [PMID: 18552863 PMCID: PMC3947522 DOI: 10.1038/nrmicro1927] [Citation(s) in RCA: 299] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Virotherapy is currently undergoing a renaissance, based on our improved understanding of virus biology and genetics and our better knowledge of many different types of cancer. Viruses can be reprogrammed into oncolytic vectors by combining three types of modification: targeting, arming and shielding. Targeting introduces multiple layers of cancer specificity and improves safety and efficacy; arming occurs through the expression of prodrug convertases and cytokines; and coating with polymers and the sequential usage of different envelopes or capsids provides shielding from the host immune response. Virus-based therapeutics are beginning to find their place in cancer clinical practice, in combination with chemotherapy and radiation.
Collapse
Affiliation(s)
- Roberto Cattaneo
- Department of Molecular Medicine, Rochester, MayoClinic, Minnesota 55905, USA.
| | | | | | | |
Collapse
|
17
|
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
|
18
|
Kanai R, Tomita H, Hirose Y, Ohba S, Goldman S, Okano H, Kawase T, Yazaki T. Augmented therapeutic efficacy of an oncolytic herpes simplex virus type 1 mutant expressing ICP34.5 under the transcriptional control of musashi1 promoter in the treatment of malignant glioma. Hum Gene Ther 2007; 18:63-73. [PMID: 17238803 DOI: 10.1089/hum.2006.107] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Although second-generation replication-conditional herpes simplex virus type 1 (HSV-1) vectors defective for both ribonucleotide reductase (RR) and the virulence factor gamma(1)34.5 have been proven safe through a number of animal experiments and clinical trials, their therapeutic efficacy was also markedly reduced. To overcome this situation, we concentrated on the use of a tumor-specific promoter in this study, to express ICP34.5 selectively in malignant glioma cells. As a molecular marker for malignant glioma, we focused on the neural RNA-binding protein, Musashi1. On the basis of the results of defective vector dvM345, as reported previously, we created, via homologous recombination, a novel HSV-1 vector termed KeM34.5, which expresses ICP34.5 under the transcriptional control of the musashi1 gene promoter (P/musashi1). Cytotoxicity mediated by KeM34.5 was significantly enhanced in human glioma cell lines (U87MG, U87MG-E6, U251, and T98G), resulting in an approximately 2-log increase in viral yield, compared with its parental vector G207. This virus also showed much higher therapeutic efficacy in the in vivo glioma model, while maintaining the desirable neuroattenuated phenotype. These results suggest that oncolytic HSV-1 expressing ICP34.5 under the transcriptional control of the musashi1 gene promoter could be a promising therapeutic agent for the treatment of malignant glioma.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Female
- Gene Expression Regulation, Viral/genetics
- Genetic Therapy
- Genetic Vectors
- Glioma/genetics
- Glioma/metabolism
- Glioma/therapy
- Glioma/virology
- Herpesvirus 1, Human/genetics
- Herpesvirus 1, Human/metabolism
- Humans
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Mutation
- Neoplasm Transplantation
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/therapy
- Neoplasms, Experimental/virology
- Nerve Tissue Proteins/genetics
- Oncolytic Virotherapy
- Promoter Regions, Genetic
- RNA-Binding Proteins/genetics
- Transduction, Genetic
- Viral Proteins/biosynthesis
- Viral Proteins/genetics
Collapse
Affiliation(s)
- Ryuichi Kanai
- Molecular Neurosurgery Laboratory, Department of Neurosurgery, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Kanai R, Eguchi K, Takahashi M, Goldman S, Okano H, Kawase T, Yazaki T. Enhanced therapeutic efficacy of oncolytic herpes vector G207 against human non-small cell lung cancer--expression of an RNA-binding protein, Musashi1, as a marker for the tailored gene therapy. J Gene Med 2007; 8:1329-40. [PMID: 16955534 DOI: 10.1002/jgm.965] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Oncolytic herpes vectors like G207 have shown considerable promise in the treatment of solid tumors, but their potency must be enhanced for the full achievement of therapeutic efficacy. Deletion of the innate gamma34.5 gene made these vectors extremely safe, but their efficacy was also severely attenuated. Use of tumor-specific promoters is one method to direct toxicity and enhance efficacy against tumors. Recently, Musashi1 has been shown expressed in some tumor tissues. METHODS Eleven human cancer cell lines including five non-small cell lung cancers (NSCLCs) were investigated. Musashi1 mRNA expression was examined by RT-PCR analysis. Western blotting was also performed. Transcriptional activity of P/musashi1 in NSCLCs was assayed by GFP reporter plasmids. Then we constructed a defective amplicon vector containing musashi1 promoter/ICP34.5 with G207 as helper virus (dvM345). In vitro cytotoxicity against NSCLCs and growth characteristics of helper virus were examined. A Lu-99 subcutaneous tumor model was used in an animal study. The tumor volume treated with G207 alone or dvM345 was measured. RESULTS Musashi1 mRNA was detected in four cell lines. Two in five NSCLCs were positive, and P/musashi1 was proved functional within them. Against these cell lines, dvM345 showed enhanced cytotoxicity, and helper viral growth was augmented. A subcutaneous tumor study confirmed the enhanced therapeutic efficacy of G207 by dvM345 without compromising safety. CONCLUSIONS These results suggest that Musashi1 might be involved in the development of several carcinomas including NSCLC. In the context of oncolytic herpes vector strategy, the P/musashi1-ICP34.5 method could be used for the treatment of cancers expressing Musashi1.
Collapse
MESH Headings
- Animals
- Base Sequence
- Biomarkers, Tumor/genetics
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/therapy
- Cell Line, Tumor
- Female
- Genetic Therapy/methods
- Genetic Vectors
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/therapy
- Mice
- Mice, Nude
- Neoplasm Transplantation
- Nerve Tissue Proteins/genetics
- Oncolytic Virotherapy/methods
- Oncolytic Viruses/genetics
- Promoter Regions, Genetic
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- RNA-Binding Proteins/genetics
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Ryuichi Kanai
- Molecular Neurosurgery Laboratory, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | | | |
Collapse
|
20
|
Kanai R, Tomita H, Hirose Y, Ohba S, Goldman S, Okano H, Kawase T, Yazaki T. Augmented Therapeutic Efficacy of an Oncolytic Herpes Simplex Virus Type 1 Mutant Expressing ICP34.5 Under the Transcriptional Control ofmusashi1Promoter in the Treatment of Malignant Glioma. Hum Gene Ther 2006. [DOI: 10.1089/hum.2007.18.ft-280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
21
|
Campbell SA, Mulvey M, Mohr I, Gromeier M. Attenuation of herpes simplex virus neurovirulence with picornavirus cis-acting genetic elements. J Virol 2006; 81:791-9. [PMID: 17079296 PMCID: PMC1797477 DOI: 10.1128/jvi.00714-06] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Viral pathogenesis depends on a suitable milieu in target host cells permitting viral gene expression, propagation, and spread. In many instances, viral genomes can be manipulated to select for propagation in certain tissues or cell types. This has been achieved for the neurotropic poliovirus (PV) by exchange of the internal ribosomal entry site (IRES), which is responsible for translation of the uncapped plus-strand RNA genome. The IRES of human rhinovirus type 2 (HRV2) confers neuron-specific replication deficits to PV but has no effect on viral propagation in malignant glioma cells. We report here that placing the critical gamma(1)34.5 virulence genes of herpes simplex virus type 1 (HSV) under translation control of the HRV2 IRES results in neuroattenuation in mice. In contrast, IRES insertion permits HSV propagation in malignant glioma cell lines that do not support replication of HSV recombinants carrying gamma(1)34.5 deletions. Our observations indicate that the conditions for alternative translation initiation at the HRV2 IRES in malignant glioma cells differ from those in normal central nervous system (CNS) cells. Picornavirus regulatory sequences mediating cell type-specific gene expression in the CNS can be utilized to target cancerous cells at the level of translation regulation outside their natural context.
Collapse
Affiliation(s)
- Stephanie A Campbell
- Division of Neurological Surgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | |
Collapse
|
22
|
Cutter JL, Kurozumi K, Chiocca EA, Kaur B. Gene therapeutics: the future of brain tumor therapy? Expert Rev Anticancer Ther 2006; 6:1053-64. [PMID: 16831077 DOI: 10.1586/14737140.6.7.1053] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Primary glioblastoma multiforme is an aggressive brain tumor that has no cure. Current treatments include gross resection of the tumor, radiation and chemotherapy. Despite valiant efforts, prognosis remains dismal. A promising new technique involves the use of oncolytic viruses that can specifically replicate and lyse in cancers, without spreading to normal tissues. Currently, these are being tested in relevant preclinical models and clinical trials as a therapeutic modality for many types of cancer. Results from recent clinical trials with oncolytic viruses have revealed the safety of this approach, although evidence for efficacy remains elusive. Oncolytic viral strategies are summarized in this review, with a focus on therapies used in brain tumors.
Collapse
Affiliation(s)
- Jennifer L Cutter
- Dardinger Laboratory for Neuro-Oncology and Neurosciences, Department of Neurological Surgery and Comprehensive Cancer Center, The Ohio State University Medical Center, 410 West 12th Avenue, Columbus, OH 43210, USA.
| | | | | | | |
Collapse
|