1
|
Le Hars M, Joussain C, Jégu T, Epstein AL. Non-replicative herpes simplex virus genomic and amplicon vectors for gene therapy - an update. Gene Ther 2025; 32:173-183. [PMID: 39533042 DOI: 10.1038/s41434-024-00500-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/22/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Two major types of defective vectors have been derived from herpes simplex virus type 1 (HSV-1), non-replicative genomic vectors (nrHSV-1), and amplicon vectors. This review recapitulates the main features of both vector types and summarizes recent improvements in our understanding of virus/vector biology, particularly with regard to the critical role played by the overpowering of antiviral cellular defenses and the epigenetic control of viral gene expression. Over the past years, significant breakthroughs in vector design, genetic engineering, and HSV-1 biology have accelerated the development of nrHSV-1 vectors. The low immunogenicity and enhanced safety profiles allowed the successful translation of these vectors into several clinical trials, with some being approved by the FDA. Regarding amplicons, despite their advantage in carrying very large or multiple transgenes, and their potential to avoid genome dilution in dividing cells, the absence of production procedures capable of generating large amounts of helper-free amplicons at reasonable cost with GMP compliance, still limits the translation of these outstanding vectors to clinical trials.
Collapse
Affiliation(s)
- Matthieu Le Hars
- UMR U1179 INSERM - University of Versailles Saint Quentin en Yvelines (UVSQ)-Paris Saclay, Montigny-le-Bretonneux, France
| | - Charles Joussain
- UMR U1179 INSERM - University of Versailles Saint Quentin en Yvelines (UVSQ)-Paris Saclay, Montigny-le-Bretonneux, France
| | | | | |
Collapse
|
2
|
Stergiopoulos GM, Iankov I, Galanis E. Personalizing Oncolytic Immunovirotherapy Approaches. Mol Diagn Ther 2024; 28:153-168. [PMID: 38150172 DOI: 10.1007/s40291-023-00689-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2023] [Indexed: 12/28/2023]
Abstract
Development of successful cancer therapeutics requires exploration of the differences in genetics, metabolism, and interactions with the immune system among malignant and normal cells. The clinical observation of spontaneous tumor regression following natural infection with microorganism has created the premise of their use as cancer therapeutics. Oncolytic viruses (OVs) originate from viruses with attenuated virulence in humans, well-characterized vaccine strains of known human pathogens, or engineered replication-deficient viral vectors. Their selectivity is based on receptor expression level and post entry restriction factors that favor replication in the tumor, while keeping the normal cells unharmed. Clinical trials have demonstrated a wide range of patient responses to virotherapy, with subgroups of patients significantly benefiting from OV administration. Tumor-specific gene signatures, including antiviral interferon-stimulated gene (ISG) expression profile, have demonstrated a strong correlation with tumor permissiveness to infection. Furthermore, the combination of OVs with immunotherapeutics, including anticancer vaccines and immune checkpoint inhibitors [ICIs, such as anti-PD-1/PD-L1 or anti-CTLA-4 and chimeric antigen receptor (CAR)-T or CAR-NK cells], could synergistically improve the therapeutic outcome. Creating response prediction algorithms represents an important step for the transition to individualized immunovirotherapy approaches in the clinic. Integrative predictors could include tumor mutational burden (TMB), inflammatory gene signature, phenotype of tumor-infiltrating lymphocytes, tumor microenvironment (TME), and immune checkpoint receptor expression on both immune and target cells. Additionally, the gut microbiota has recently been recognized as a systemic immunomodulatory factor and could further be used in the optimization of individualized immunovirotherapy algorithms.
Collapse
Affiliation(s)
| | - Ianko Iankov
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Evanthia Galanis
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Oncology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
3
|
Xiong F, Yang H, Song YG, Qin HB, Zhang QY, Huang X, Jing W, Deng M, Liu Y, Liu Z, Shen Y, Han Y, Lu Y, Xu X, Holmes TC, Luo M, Zhao F, Luo MH, Zeng WB. An HSV-1-H129 amplicon tracer system for rapid and efficient monosynaptic anterograde neural circuit tracing. Nat Commun 2022; 13:7645. [PMID: 36496505 PMCID: PMC9741617 DOI: 10.1038/s41467-022-35355-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
Monosynaptic viral tracers are essential tools for dissecting neuronal connectomes and for targeted delivery of molecular sensors and effectors. Viral toxicity and complex multi-injection protocols are major limiting application barriers. To overcome these barriers, we developed an anterograde monosynaptic H129Amp tracer system based on HSV-1 strain H129. The H129Amp tracer system consists of two components: an H129-dTK-T2-pacFlox helper which assists H129Amp tracer's propagation and transneuronal monosynaptic transmission. The shared viral features of tracer/helper allow for simultaneous single-injection and subsequent high expression efficiency from multiple-copy of expression cassettes in H129Amp tracer. These improvements of H129Amp tracer system shorten experiment duration from 28-day to 5-day for fast-bright monosynaptic tracing. The lack of toxic viral genes in the H129Amp tracer minimizes toxicity in postsynaptic neurons, thus offering the potential for functional anterograde mapping and long-term tracer delivery of genetic payloads. The H129Amp tracer system is a powerful tracing tool for revealing neuronal connectomes.
Collapse
Affiliation(s)
- Feng Xiong
- grid.9227.e0000000119573309State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China ,grid.9227.e0000000119573309Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Hong Yang
- grid.9227.e0000000119573309State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Yi-Ge Song
- grid.33199.310000 0004 0368 7223Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hai-Bin Qin
- grid.9227.e0000000119573309State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Qing-Yang Zhang
- grid.9227.e0000000119573309State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Xian Huang
- grid.33199.310000 0004 0368 7223Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Jing
- grid.33199.310000 0004 0368 7223Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Manfei Deng
- grid.33199.310000 0004 0368 7223Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Liu
- grid.410717.40000 0004 0644 5086National Institute of Biological Sciences, Beijing, China
| | - Zhixiang Liu
- grid.410717.40000 0004 0644 5086National Institute of Biological Sciences, Beijing, China
| | - Yin Shen
- grid.49470.3e0000 0001 2331 6153Eye Center, Renmin Hospital, Wuhan University, Wuhan, China
| | - Yunyun Han
- grid.49470.3e0000 0001 2331 6153Eye Center, Renmin Hospital, Wuhan University, Wuhan, China
| | - Youming Lu
- grid.33199.310000 0004 0368 7223Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangmin Xu
- grid.266093.80000 0001 0668 7243Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA USA ,grid.266093.80000 0001 0668 7243Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, CA USA
| | - Todd C. Holmes
- grid.266093.80000 0001 0668 7243Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, CA USA ,grid.266093.80000 0001 0668 7243Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA USA
| | - Minmin Luo
- grid.410717.40000 0004 0644 5086National Institute of Biological Sciences, Beijing, China ,grid.510934.a0000 0005 0398 4153Chinese Institute for Brain Research, Beijing, China
| | - Fei Zhao
- grid.510934.a0000 0005 0398 4153Chinese Institute for Brain Research, Beijing, China ,grid.24696.3f0000 0004 0369 153XSchool of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Min-Hua Luo
- grid.9227.e0000000119573309State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China ,grid.9227.e0000000119573309Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China ,grid.266093.80000 0001 0668 7243Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, CA USA
| | - Wen-Bo Zeng
- grid.9227.e0000000119573309State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
4
|
Wang CH, Naik NG, Liao LL, Wei SC, Chao YC. Global Screening of Antiviral Genes that Suppress Baculovirus Transgene Expression in Mammalian Cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 6:194-206. [PMID: 28831401 PMCID: PMC5554962 DOI: 10.1016/j.omtm.2017.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 07/13/2017] [Indexed: 12/31/2022]
Abstract
Although baculovirus has been used as a safe and convenient gene delivery vector in mammalian cells, baculovirus-mediated transgene expression is less effective in various mammalian cell lines. Identification of the negative regulators in host cells is necessary to improve baculovirus-based expression systems. Here, we performed high-throughput shRNA library screening, targeting 176 antiviral innate immune genes, and identified 43 host restriction factor genes in a human A549 lung carcinoma cell line. Among them, suppression of receptor interaction protein kinase 1 (RIP1, also known as RIPK1) significantly increased baculoviral transgene expression without resulting in significant cell death. Silencing of RIP1 did not affect viral entry or cell viability, but it did inhibit nuclear translocation of the IRF3 and NF-κB transcription factors. Also, activation of downstream signaling mediators (such as TBK1 and IRF7) was affected, and subsequent interferon and cytokine gene expression levels were abolished. Further, Necrostatin-1 (Nec-1)—an inhibitor of RIP1 kinase activity—dramatically increased baculoviral transgene expression in RIP1-silenced cells. Using baculovirus as a model system, this study presents an initial investigation of large numbers of human cell antiviral innate immune response factors against a “nonadaptive virus.” In addition, our study has made baculovirus a more efficient gene transfer vector for some of the most frequently used mammalian cell systems.
Collapse
Affiliation(s)
- Chia-Hung Wang
- Department of Life Science and Institute of Genome Science, National Yang Ming University, No. 115, Section 2, Linong St., Beitou District, Taipei 112, Taiwan.,Institute of Molecular Biology, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei 115, Taiwan
| | - Nenavath Gopal Naik
- Institute of Molecular Biology, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei 115, Taiwan
| | - Lin-Li Liao
- Institute of Molecular Biology, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei 115, Taiwan
| | - Sung-Chan Wei
- Institute of Molecular Biology, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei 115, Taiwan
| | - Yu-Chan Chao
- Institute of Molecular Biology, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei 115, Taiwan
| |
Collapse
|
5
|
Ebrahimi S, Ghorbani E, Khazaei M, Avan A, Ryzhikov M, Azadmanesh K, Hassanian SM. Interferon-Mediated Tumor Resistance to Oncolytic Virotherapy. J Cell Biochem 2017; 118:1994-1999. [DOI: 10.1002/jcb.25917] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 01/26/2017] [Indexed: 12/30/2022]
Affiliation(s)
- Safieh Ebrahimi
- Department of Medical Biochemistry; School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| | - Elnaz Ghorbani
- Department of Microbiology; Al-Zahra University; Tehran Iran
| | - Majid Khazaei
- Department of Medical Physiology; School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| | - Amir Avan
- Metabolic Syndrome Research Center; School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
- Molecular Medicine Group; Department of Modern Sciences and Technologies; School of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Mikhail Ryzhikov
- Department of Molecular Microbiology and Immunology; St. Louis University School of Medicine; Saint Louis Missouri
| | | | - Seyed Mahdi Hassanian
- Department of Medical Biochemistry; School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
- Metabolic Syndrome Research Center; School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
- Microanatomy Research Center; Mashhad University of Medical Sciences; Mashhad Iran
| |
Collapse
|
6
|
Ribosomal protein s15 phosphorylation mediates LRRK2 neurodegeneration in Parkinson's disease. Cell 2014; 157:472-485. [PMID: 24725412 DOI: 10.1016/j.cell.2014.01.064] [Citation(s) in RCA: 206] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 12/08/2013] [Accepted: 01/23/2014] [Indexed: 12/13/2022]
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are a common cause of familial and sporadic Parkinson's disease (PD). Elevated LRRK2 kinase activity and neurodegeneration are linked, but the phosphosubstrate that connects LRRK2 kinase activity to neurodegeneration is not known. Here, we show that ribosomal protein s15 is a key pathogenic LRRK2 substrate in Drosophila and human neuron PD models. Phosphodeficient s15 carrying a threonine 136 to alanine substitution rescues dopamine neuron degeneration and age-related locomotor deficits in G2019S LRRK2 transgenic Drosophila and substantially reduces G2019S LRRK2-mediated neurite loss and cell death in human dopamine and cortical neurons. Remarkably, pathogenic LRRK2 stimulates both cap-dependent and cap-independent mRNA translation and induces a bulk increase in protein synthesis in Drosophila, which can be prevented by phosphodeficient T136A s15. These results reveal a novel mechanism of PD pathogenesis linked to elevated LRRK2 kinase activity and aberrant protein synthesis in vivo.
Collapse
|
7
|
Tumor Restrictions to Oncolytic Virus. Biomedicines 2014; 2:163-194. [PMID: 28548066 PMCID: PMC5423468 DOI: 10.3390/biomedicines2020163] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 03/17/2014] [Accepted: 03/28/2014] [Indexed: 02/06/2023] Open
Abstract
Oncolytic virotherapy has advanced since the days of its conception but therapeutic efficacy in the clinics does not seem to reach the same level as in animal models. One reason is premature oncolytic virus clearance in humans, which is a reasonable assumption considering the immune-stimulating nature of the oncolytic agents. However, several studies are beginning to reveal layers of restriction to oncolytic virotherapy that are present before an adaptive neutralizing immune response. Some of these barriers are present constitutively halting infection before it even begins, whereas others are raised by minute cues triggered by virus infection. Indeed, we and others have noticed that delivering viruses to tumors may not be the biggest obstacle to successful therapy, but instead the physical make-up of the tumor and its capacity to mount antiviral defenses seem to be the most important efficacy determinants. In this review, we summarize the constitutive and innate barriers to oncolytic virotherapy and discuss strategies to overcome them.
Collapse
|
8
|
Farzad LM, Suzuki M. Feasibility of Applying Helper-Dependent Adenoviral Vectors for Cancer Immunotherapy. Biomedicines 2014; 2:110-131. [PMID: 28548063 PMCID: PMC5423480 DOI: 10.3390/biomedicines2010110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/25/2014] [Accepted: 02/26/2014] [Indexed: 01/08/2023] Open
Abstract
Adenoviruses (Ads) infect a broad range of tissue types, and derived vectors have been extensively used for gene therapy. Helper-dependent Ad vectors (HDAds), devoid of viral coding sequences, allow for insertion of large or multiple transgenes in a single vector and have been preclinically used for the study of genetic disorders. However, the clinical application of Ad vectors including HDAds for genetic disorders has been hampered by an acute toxic response. This characteristic, while disadvantageous for gene replacement therapy, could be strategically advantageous for the activation of an immune response if HDAds were used as an adjunct treatment in cancer. Cancer treatments including immunotherapy are frequently limited by the inhibitory environment produced by both tumors and their stroma, each of which express numerous inhibitory molecules. Hence, multiple inhibitory mechanisms must be overcome for development of anti-tumor immunity. The large coding capacity of HDAds can accommodate multiple immune modulating transgenes that could produce a combined effect to overcome tumor-derived inhibition and ensure intratumoral effector T-cell proliferation and function. In this review, we discuss the potential advantages of HDAds to cancer immunotherapy based on potent host immune responses to Ads.
Collapse
Affiliation(s)
- Lisa M Farzad
- Department of Medicine, Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Masataka Suzuki
- Department of Medicine, Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
9
|
Differential type I interferon-dependent transgene silencing of helper-dependent adenoviral vs. adeno-associated viral vectors in vivo. Mol Ther 2013; 21:796-805. [PMID: 23319058 DOI: 10.1038/mt.2012.277] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
We previously dissected the components of the innate immune response to Helper-dependent adenoviral vectors (HDAds) using genetic models, and demonstrated that multiple pattern recognition receptor signaling pathways contribute to this host response to HDAds in vivo. Based on analysis of cytokine expression profiles, type I interferon (IFN) mRNA is induced in host mouse livers at 1 hour post-injection. This type I IFN signaling amplifies cytokine expression in liver independent of the nature of vector DNA sequences after 3 hours post-injection. This type I IFN signaling in response to HDAds administration contributes to transcriptional silencing of both HDAd prokaryotic and eukaryotic DNA in liver. This silencing occurs early and is mediated by epigenetic modification as shown by in vivo chromatin immunoprecipitation (ChIP) with anti-histone deacetylase (HDAC) and promyelocytic leukemia protein (PML). In contrast, self-complementary adeno-associated viral vectors (scAAVs) showed significantly lower induction of type I IFN mRNA in liver compared to HDAds at both early and late time points. These results show that the type I IFN signaling dependent transgene silencing differs between AAV and HDAd vectors after liver-directed gene transfer.
Collapse
|
10
|
Sutlu T, Nyström S, Gilljam M, Stellan B, Applequist SE, Alici E. Inhibition of intracellular antiviral defense mechanisms augments lentiviral transduction of human natural killer cells: implications for gene therapy. Hum Gene Ther 2012; 23:1090-100. [PMID: 22779406 DOI: 10.1089/hum.2012.080] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Adoptive immunotherapy with genetically modified natural killer (NK) cells is a promising approach for cancer treatment. Yet, optimization of highly efficient and clinically applicable gene transfer protocols for NK cells still presents a challenge. In this study, we aimed at identifying conditions under which optimum lentiviral gene transfer to NK cells can be achieved. Our results demonstrate that stimulation of NK cells with interleukin (IL)-2 and IL-21 supports efficient transduction using a VSV-G pseudotyped lentiviral vector. Moreover, we have identified that inhibition of innate immune receptor signaling greatly enhances transduction efficiency. We were able to boost the efficiency of lentiviral genetic modification on average 3.8-fold using BX795, an inhibitor of the TBK1/IKKɛ complex acting downstream of RIG-I, MDA-5, and TLR3. We have also observed that the use of BX795 enhances lentiviral transduction efficiency in a number of human and mouse cell lines, indicating a broadly applicable, practical, and safe approach that has the potential of being applicable to various gene therapy protocols.
Collapse
Affiliation(s)
- Tolga Sutlu
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, SE-14186 Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
11
|
Jerusalinsky D, Baez MV, Epstein AL. Herpes simplex virus type 1-based amplicon vectors for fundamental research in neurosciences and gene therapy of neurological diseases. ACTA ACUST UNITED AC 2011; 106:2-11. [PMID: 22108428 DOI: 10.1016/j.jphysparis.2011.11.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 10/11/2011] [Accepted: 11/04/2011] [Indexed: 12/24/2022]
Abstract
Somatic manipulation of the nervous system without the involvement of the germinal line appears as a powerful counterpart of the transgenic strategy. The use of viral vectors to produce specific, transient and localized knockout, knockdown, ectopic expression or overexpression of a gene, leads to the possibility of analyzing both in vitro and in vivo molecular basis of neural function. In this approach, viral particles engineered to carry transgenic sequences are delivered into discrete brain regions, to transduce cells that will express the transgenic products. Amplicons are replication-incompetent helper-dependent vectors derived from herpes simplex virus type 1 (HSV-1), with several advantages that potentiate their use in neurosciences: (1) minimal toxicity: amplicons do not encode any virus proteins, are neither toxic for the infected cells nor pathogenic for the inoculated animals and elicit low levels of adaptive immune responses; (2) extensive transgene capacity to carry up to 150-kb of foreign DNA; i.e., entire genes with regulatory sequences could be delivered; (3) widespread cellular tropism: amplicons can experimentally infect several cell types including glial cells, though naturally the virus infects mainly neurons and epithelial cells; (4) since the viral genome does not integrate into cellular chromosomes there is low probability to induce insertional mutagenesis. Recent investigations on gene transfer into the brain using these vectors, have focused on gene therapy of inherited genetic diseases affecting the nervous system, such as ataxias, or on neurodegenerative disorders using experimental models of Parkinson's or Alzheimer's disease. Another group of studies used amplicons to investigate complex neural functions such as neuroplasticity, anxiety, learning and memory. In this short review, we summarize recent data supporting the potential of HSV-1 based amplicon vector model for gene delivery and modulation of gene expression in primary cultures of neuronal cells and into the brain of living animals.
Collapse
Affiliation(s)
- Diana Jerusalinsky
- Instituto de Biología Celular y Neurociencia (IBCN), CONICET-UBA. Buenos Aires, Argentina.
| | | | | |
Collapse
|
12
|
Suzuki M, Cela R, Bertin TK, Sule G, Cerullo V, Rodgers JR, Lee B. NOD2 signaling contributes to the innate immune response against helper-dependent adenovirus vectors independently of MyD88 in vivo. Hum Gene Ther 2011; 22:1071-82. [PMID: 21561248 DOI: 10.1089/hum.2011.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
We previously demonstrated that Toll-like receptor/myeloid differentiation primary response gene 88 (MyD88) signaling is required for maximal innate and acquired [T helper cell type 1 (Th1)] immune responses following systemic administration of helper-dependent adenoviral vectors (HDAds). However, MyD88-deficient mice injected with HDAdLacZ exhibited only partial reduction of innate immune cytokine expression compared with wild-type mice, suggesting MyD88-independent pathways also respond to HDAds. We now show that NOD2, a nucleotide-binding and oligomerization domain (NOD)-like receptor known to detect muramyl dipeptides in bacterial peptidoglycans, also contributes to innate responses to HDAds, but not to humoral or Th1 immune responses. We established NOD2/MyD88 double-deficient mice that, when challenged with HDAds, showed a significant reduction of the innate response compared with mice deficient for either gene singly, suggesting that NOD2 signaling contributes to the innate response independently of MyD88 signaling following systemic administration of HDAds. In addition, NOD2-deficient mice exhibited significantly higher transgene expression than did wild-type mice at an early time point (before development of an acquired response), but not at a later time point (after development of an acquired response). These results indicate that the intracellular sensor NOD2 is required for innate responses to HDAds and can limit transgene expression during early phases of infection.
Collapse
Affiliation(s)
- Masataka Suzuki
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
de Silva S, Lotta LT, Burris CA, Bowers WJ. Virion-associated cofactor high-mobility group DNA-binding protein-1 facilitates transposition from the herpes simplex virus/Sleeping Beauty amplicon vector platform. Hum Gene Ther 2010; 21:1615-22. [PMID: 20568967 DOI: 10.1089/hum.2010.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The development of the integration-competent, herpes simplex virus/Sleeping Beauty (HSV/SB) amplicon vector platform has created a means to efficiently and stably deliver therapeutic transcription units (termed "transgenons") to neurons within the mammalian brain. Furthermore, an investigation into the transposition capacity of the HSV/SB vector system revealed that the amplicon genome provides an optimal substrate for the transposition of transgenons at least 12 kb in length [de Silva, S., Mastrangelo, M.A., Lotta, L.T., Jr., Burris, C.A., Federoff, H.J., and Bowers, W.J. ( 2010 ). Gene Ther. 17, 424-431]. These results prompted an investigation into the factors that may contribute toward efficient transposition from the HSV/SB amplicon. One of the cellular cofactors known to play a key role during SB-mediated transposition is the high-mobility group DNA-binding protein-1 (HMGB1). Our present investigation into the role of HMGB1 during amplicon-based transposition revealed that transposition is not strictly dependent on the presence of cellular HMGB1, contrary to what had been previously demonstrated with plasmid-based SB transposition. We have shown for the first time that during amplicon preparation, biologically active HMGB1 derived from the packaging cell line is copackaged into amplicon vector particles. As a result, HSV/SB amplicon virions arrive prearmed with HMGB1 protein at levels sufficient for facilitating SB-mediated transposition in the transduced mammalian cell.
Collapse
Affiliation(s)
- Suresh de Silva
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | | | | | | |
Collapse
|
14
|
Tsitoura E, Epstein AL. Constitutive and Inducible Innate Responses in Cells Infected by HSV-1-Derived Amplicon Vectors. Open Virol J 2010; 4:96-102. [PMID: 20811588 DOI: 10.2174/1874357901004030096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 01/11/2010] [Accepted: 01/12/2010] [Indexed: 12/26/2022] Open
Abstract
Amplicons are helper-dependent herpes simplex virus type 1 (HSV-1)-based vectors that can deliver very large foreign DNA sequences and, as such, are good candidates both for gene delivery and vaccine development. However, many studies have shown that innate constitutive or induced cellular responses, elicited or activated by the entry of HSV-1 particles, can play a significant role in the control of transgenic expression and in the induction of inflammatory responses. Moreover, transgene expression from helper-free amplicon stocks is often weak and transient, depending on the particular type of infected cells, suggesting that cellular responses could be also responsible for the silencing of amplicon-mediated transgene expression. This review summarizes the current experimental evidence underlying these latter concepts, focusing on the impact on transgene expression of very-early interactions between amplicon particles and the infected cells, and speculates on possible ways to counteract the cellular protective mechanisms, thus allowing stable transgene expression without enhancement of vector toxicity.
Collapse
Affiliation(s)
- Eliza Tsitoura
- Université de Lyon, Lyon, F-69003, France; CNRS, UMR5534, Centre de Génétique Moléculaire et Cellulaire, Villeurbanne, F-69622, France
| | | |
Collapse
|
15
|
Recognition of virus infection and innate host responses to viral gene therapy vectors. Mol Ther 2010; 18:1422-9. [PMID: 20551916 DOI: 10.1038/mt.2010.124] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The innate immune and inflammatory response represents one of the key stumbling blocks limiting the efficacy of viral-based therapies. Numerous human diseases could be corrected or ameliorated if viruses were harnessed to safely and effectively deliver therapeutic genes to diseased cells and tissues in vivo. Recent studies have shown that host cells recognize viruses using an elaborate network of sensor proteins localized at the plasma membrane, in endosomes, or in the cytosol. Three classes of sensors have been implicated in sensing viruses in mammalian cells-Toll-like receptors (TLRs), retinoid acid-inducible gene (RIG)-I-like receptors (RLRs), and nucleotide oligomerization domain (NOD)-like receptors (NLRs). The interaction of virus-associated nucleic acids with these sensor molecules triggers a signaling cascade that activates the principal host defense program aimed to limit or eliminate virus infection and restore tissue homeostasis. In addition, recent data strongly suggest that host cells can mount innate immune responses to viruses without prior recognition of their nucleic acids. To deliver therapeutic genes into the nuclei of diseased cells, viral gene therapy vectors must be efficient at penetrating either the plasma or endosomal membrane. The therapeutic use of high numbers of virus particles disturbs cellular homeostasis, triggering cell damage and stress pathways, or "sensing of modified self". Accumulating data indicate that the sensing of modified self might represent a powerful framework explaining the innate immune response activation by viral gene therapy vectors.
Collapse
|
16
|
Suzuki M, Cerullo V, Bertin TK, Cela R, Clarke C, Guenther M, Brunetti-Pierri N, Lee B. MyD88-dependent silencing of transgene expression during the innate and adaptive immune response to helper-dependent adenovirus. Hum Gene Ther 2010; 21:325-36. [PMID: 19824822 DOI: 10.1089/hum.2009.155] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Activation of the host innate immune response after systemic administration of adenoviral vectors constitutes a principal impediment to successful clinical gene replacement therapies. Although helper-dependent adenoviruses (HDAds) lack all viral functional genes, systemic administration of a high dose of HDAd still elicits a potent innate immune response in host animals. Toll-like receptors (TLRs) are innate receptors that sense microbial products and trigger the maturation of antigen-presenting cells and cytokine production via MyD88-dependent signaling (except TLR3). Here we show that mice lacking MyD88 exhibit a dramatic reduction in proinflammatory cytokines after intravenous injection of a high dose of HDAd, and show significantly reduced induction of the adaptive immune response when compared with wild-type and TLR2-deficient mice. Importantly, MyD88(-/-) mice also show significantly higher and longer sustained transgene expression than do wild-type mice. Chromatin immunoprecipitation studies using wild-type and MyD88-deficient primary mouse embryonic fibroblasts showed significant MyD88-dependent transcriptional silencing of the HDAd-encoded transgenes. Our results demonstrate that MyD88 signaling, activated by systemic delivery of HDAd, initiates an innate immune response that suppresses transgene expression at the transcriptional level before initiation of the adaptive immune response.
Collapse
Affiliation(s)
- Masataka Suzuki
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Since its emergence onto the gene therapy scene nearly 25 years ago, the replication-defective Herpes Simplex Virus Type-1 (HSV-1) amplicon has gained significance as a versatile gene transfer platform due to its extensive transgene capacity, widespread cellular tropism, minimal immunogenicity, and its amenability to genetic manipulation. Herein, we detail the recent advances made with respect to the design of the HSV amplicon, its numerous in vitro and in vivo applications, and the current impediments this virus-based gene transfer platform faces as it navigates a challenging path towards future clinical testing.
Collapse
|
18
|
Epstein AL. HSV-1-derived amplicon vectors: recent technological improvements and remaining difficulties--a review. Mem Inst Oswaldo Cruz 2009; 104:399-410. [PMID: 19547864 DOI: 10.1590/s0074-02762009000300002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Accepted: 05/15/2009] [Indexed: 01/04/2023] Open
Abstract
Amplicons are defective and non-integrative vectors derived from herpes simplex virus type 1. As the vector genome carries no virus genes, amplicons are both non-toxic for the infected cells and non-pathogenic for the inoculated organisms. In addition, the large transgenic capacity of amplicons, which allow delivery of up to 150 Kbp of foreign DNA, makes these vectors one of the most powerful, interesting and versatile gene delivery platforms. We present here recent technological developments that have significantly improved and extended the use of amplicons, both in cultured cells and in living organisms. In addition, this review also discusses the many difficulties still pending to be solved, in order to achieve stable and physiologically regulated transgene expression.
Collapse
Affiliation(s)
- Alberto Luis Epstein
- Centre de Génétique Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, Université de Lyon, Villeurbanne, France.
| |
Collapse
|
19
|
Tsitoura E, Thomas J, Cuchet D, Thoinet K, Mavromara P, Epstein AL. Infection with herpes simplex type 1-based amplicon vectors results in an IRF3/7-dependent, TLR-independent activation of the innate antiviral response in primary human fibroblasts. J Gen Virol 2009; 90:2209-20. [PMID: 19515829 DOI: 10.1099/vir.0.012203-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amplicons are helper-dependent herpes simplex virus type 1 (HSV-1)-based vectors that can deliver very large, foreign DNA sequences and, as such, are good candidates for both gene delivery and vaccine development. However, many studies have shown that innate immune responses induced by virus vectors can play a significant role in the control of transgenic expression and in the induction of inflammatory responses. Furthermore, amplicons are very interesting tools to study innate cellular responses elicited by entry of HSV-1 particles in the absence of any virus gene expression. For these reasons, in this study we characterized the innate antiviral response established in human fibroblasts of limited passage (HFFF-2) infected by amplicons. Our results indicate that infection with amplicons triggered an interferon (IFN)-regulatory factors 3 and 7 (IRF3/7)-dependent antiviral response, rendered the cells resistant to vesicular stomatitis virus infection and induced significant changes in the pattern of cellular gene expression, including the upregulation of Toll-like receptor 3 (TLR3), IRF7 and IFN-stimulated genes (ISGs). In contrast, we observed only a mild and contained type I IFN response in infected cells. Amplicon infection induced nuclear translocation and subsequent degradation of IRF3, without hyperphosphorylation of the protein. Inhibition of endosome-resident TLR signalling by blocking lysosome maturation or the knockdown of TLR3 and 4 did not abolish the cellular response to amplicons, whereas knockdown of IRF3 and 7 inhibited ISG and IFN-beta expression severely. Therefore, our results confirm the existence of TLR-independent, IRF3/7-dependent activation pathways triggered by HSV-1 particles in human fibroblasts.
Collapse
|
20
|
Progress and prospects: biological properties and technological advances of herpes simplex virus type 1-based amplicon vectors. Gene Ther 2009; 16:709-15. [PMID: 19369969 DOI: 10.1038/gt.2009.42] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The last two years have seen significant advances in our understanding of the cellular innate responses elicited or activated by the entry of amplicon particles, which may, in part, explain the transient nature of transgene expression often observed in cells infected with helper-free amplicon stocks. At the technological level, the most consistent progress has been in strategies to enhance the stability of transgene cassettes, either through integration into host chromosomes or through the conversion of the amplicon genome into a replication-competent extrachromosomal element.
Collapse
|
21
|
Suzuki M, Kasai K, Ohtsuki A, Godlewski J, Nowicki MO, Chiocca EA, Saeki Y. ICP0 inhibits the decrease of HSV amplicon-mediated transgene expression. Mol Ther 2009; 17:707-15. [PMID: 19223864 DOI: 10.1038/mt.2008.306] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The herpes simplex virus (HSV) amplicon vector produces an initial host response that limits transgene expression. In this study, we hypothesized that restoration of the HSV gene infected cell protein (ICP0) into the amplicon could circumvent this host response and thus overcome silencing of encoded transgenes. To test this, we constructed an amplicon vector that encodes the ICP0 under control of its native promoter (ICP0+ amplicon). Expression of ICP0 was transient and, at a multiplicity of infection (MOI) of 1, did not significantly alter interferon (IFN)-based responses against the vector or cell kinetics/apoptosis of infected cells. Chromatin immunoprecipitation (ChIP) PCR analysis revealed that conventional amplicon DNA became associated with histone deacetylase 1 (HDAC1) immediately after infection, whereas ICP0+ amplicon DNA remained relatively unbound by HDAC1 for at least 72 hours after infection. Mice administered systemic ICP0+ amplicon exhibited significantly greater and more sustained transgene expression in their livers than did those receiving conventional amplicon, likely due to increased transcriptional or post-transcriptional activity rather than increased copy numbers of vector DNA. These findings indicate that restoration of ICP0 expression may be employed within HSV amplicon constructs to decrease transgene silencing in vitro and in vivo.
Collapse
Affiliation(s)
- Masataka Suzuki
- Dardinger Laboratory for Neuro-oncology and Neurosciences, Department of Neurological Surgery, James Comprehensive Cancer Center and The Ohio State University Medical Center, Columbus, Ohio 43210, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Otsuki A, Patel A, Kasai K, Suzuki M, Kurozumi K, Chiocca EA, Saeki Y. Histone deacetylase inhibitors augment antitumor efficacy of herpes-based oncolytic viruses. Mol Ther 2008; 16:1546-1555. [PMID: 18648350 DOI: 10.1038/mt.2008.155] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2008] [Accepted: 06/25/2008] [Indexed: 12/15/2022] Open
Abstract
Replication-conditional (oncolytic) mutants of herpes simplex virus (HSV), are considered promising therapeutic alternatives for human malignancies, and chemotherapeutic adjuvants are increasingly sought to augment their efficacy. Histone deacetylase (HDAC) inhibitors are a new class of antineoplastic agents because of their potent activity in growth arrest, differentiation, and apoptotic death of cancer cells. The ability of the HDAC inhibitors to upregulate exogenous transgene expression and inhibit interferon (IFN) responses prompted our exploration of their use in improving the antitumor efficacy of oncolytic HSV. We discovered that the yield of viral progeny increased significantly when cultured glioma cells were treated with HDAC inhibitors before viral infection. Valproic acid (VPA), a commonly used antiepileptic agent with HDAC inhibitory activity, proved most effective when used to treat glioma cells before viral infection, but not concomitantly with viral infection. Pretreatment with VPA inhibited the induction of several IFN-responsive antiviral genes, augmented the transcriptional level of viral genes, and improved viral propagation, even in the presence of type I IFNs. Moreover, VPA pretreatment improved the propagation and therapeutic efficacy of oncolytic HSV in a human glioma xenograft model in vivo. These findings indicate that HDAC inhibitors can improve the efficacy of tumor virotherapies.
Collapse
Affiliation(s)
- Akihiro Otsuki
- Dardinger Laboratory for Neuro-oncology and Neurosciences, Department of Neurological Surgery, The Ohio State University Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, 43210 USA
| | | | | | | | | | | | | |
Collapse
|