1
|
Su D, Han L, Shi C, Li Y, Qian S, Feng Z, Yu L. An updated review of HSV-1 infection-associated diseases and treatment, vaccine development, and vector therapy application. Virulence 2024; 15:2425744. [PMID: 39508503 PMCID: PMC11562918 DOI: 10.1080/21505594.2024.2425744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/24/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is a globally widespread virus that causes and associates with a wide range of diseases, including herpes simplex encephalitis, herpes simplex keratitis, and herpes labialis. The interaction between HSV-1 and the host involves complex immune response mechanisms, including recognition of viral invasion, maintenance of latent infection, and triggering of reactivation. Antiviral therapy is the core treatment for HSV-1 infections. Meanwhile, vaccine development employs different strategies and methods, and several promising vaccine types have emerged, such as live attenuated, protein subunit, and nucleic acid vaccines, offering new possibilities for the prevention of HSV-1 infection. Moreover, HSV-1 can be modified into a therapeutic vector for gene therapy and tumour immunotherapy. This review provides an in-depth summary of HSV-1 infection-associated innate and adaptive immune responses, disease pathogenesis, current therapeutic approaches, recent advances in vaccine development, and vector therapy applications for cancer treatment. Through a systematic review of multiple aspects of HSV-1, this study aims to provide a comprehensive and detailed reference for the public on the prevention, control, and treatment of HSV-1.
Collapse
Affiliation(s)
- Dan Su
- Department of Endocrine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang, Henan, China
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan, P.R.China
| | - Liping Han
- Department of Endocrine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang, Henan, China
| | - Chengyu Shi
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan, P.R.China
| | - Yaoxin Li
- Department of Endocrine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang, Henan, China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan, P.R.China
| | - Shaoju Qian
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan, P.R.China
| | - Zhiwei Feng
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan, P.R.China
| | - Lili Yu
- Department of Endocrine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang, Henan, China
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan, P.R.China
| |
Collapse
|
2
|
Wang T, Zhu X, Yi H, Gu J, Liu S, Izenwasser S, Lemmon VP, Roy S, Hao S. Viral vector-mediated gene therapy for opioid use disorders. Exp Neurol 2021; 341:113710. [PMID: 33781732 DOI: 10.1016/j.expneurol.2021.113710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/26/2021] [Accepted: 03/24/2021] [Indexed: 12/13/2022]
Abstract
Chronic exposure to opioids typically results in adverse consequences. Opioid use disorder (OUD) is a disease of the CNS with behavioral, psychological, neurobiological, and medical manifestations. OUD induces a variety of changes of neurotransmitters/neuropeptides in the nervous system. Existing pharmacotherapy, such as opioid maintenance therapy (OMT) is the mainstay for the treatment of OUD, however, current opioid replacement therapy is far from effective for the majority of patients. Pharmacological therapy for OUD has been challenging for many reasons including debilitating side-effects. Therefore, developing an effective, non-pharmacological approach would be a critical advancement in improving and expanding treatment for OUD. Viral vector mediated gene therapy provides a potential new approach for treating opioid abused patients. Gene therapy can supply targeting gene products directly linked to the mechanisms of OUD to restore neurotransmitter and/or neuropeptides imbalance, and avoid the off-target effects of systemic administration of drugs. The most commonly used viral vectors in rodent studies of treatment of opioid-used disorder are based on recombinant adenovirus (AV), adeno-associated virus (AAV), lentiviral (LV) vectors, and herpes simplex virus (HSV) vectors. In this review, we will focus on the recent progress of viral vector mediated gene therapy in OUD, especially morphine tolerance and withdrawal.
Collapse
Affiliation(s)
- Tao Wang
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Xun Zhu
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Hyun Yi
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Jun Gu
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Shue Liu
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Sari Izenwasser
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Vance P Lemmon
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Sabita Roy
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Shuanglin Hao
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America.
| |
Collapse
|
3
|
Soukupová M, Zucchini S, Trempat P, Ingusci S, Perrier-Biollay C, Barbieri M, Cattaneo S, Bettegazzi B, Falzoni S, Berthommé H, Simonato M. Improvement of HSV-1 based amplicon vectors for a safe and long-lasting gene therapy in non-replicating cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:399-412. [PMID: 33869657 PMCID: PMC8044385 DOI: 10.1016/j.omtm.2021.03.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 03/25/2021] [Indexed: 11/22/2022]
Abstract
A key factor for developing gene therapy strategies for neurological disorders is the availability of suitable vectors. Currently, the most advanced are adeno-associated vectors that, while being safe and ensuring long-lasting transgene expression, have a very limited cargo capacity. In contrast, herpes simplex virus-based amplicon vectors can host huge amounts of foreign DNA, but concerns exist about their safety and ability to express transgenes long-term. We aimed at modulating and prolonging amplicon-induced transgene expression kinetics in vivo using different promoters and preventing transgene silencing. To pursue the latter, we deleted bacterial DNA sequences derived from vector construction and shielded the transgene cassette using AT-rich and insulator-like sequences (SAm technology). We employed luciferase and GFP as reporter genes. To determine transgene expression kinetics, we injected vectors in the hippocampus of mice that were longitudinally scanned for bioluminescence for 6 months. To evaluate safety, we analyzed multiple markers of damage and performed patch clamp electrophysiology experiments. All vectors proved safe, and we managed to modulate the duration of transgene expression, up to obtaining a stable, long-lasting expression using the SAm technology. Therefore, these amplicon vectors represent a flexible, efficient, and safe tool for gene delivery in the brain.
Collapse
Affiliation(s)
- Marie Soukupová
- Department of Neuroscience and Rehabilitation, Section of Pharmacology, University of Ferrara, 44121 Ferrara, Italy
| | - Silvia Zucchini
- Department of Neuroscience and Rehabilitation, Section of Pharmacology, University of Ferrara, 44121 Ferrara, Italy.,Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy
| | - Pascal Trempat
- Bioviron, Université Claude Bernard Lyon 1, 69100 Villeurbanne, France
| | - Selene Ingusci
- Department of Neuroscience and Rehabilitation, Section of Pharmacology, University of Ferrara, 44121 Ferrara, Italy
| | | | - Mario Barbieri
- Department of Neuroscience and Rehabilitation, Section of Pharmacology, University of Ferrara, 44121 Ferrara, Italy
| | - Stefano Cattaneo
- School of Medicine, University Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Barbara Bettegazzi
- School of Medicine, University Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Simonetta Falzoni
- Department of Medical Sciences, Section of Pathology, Oncology and Experimental Biology, University of Ferrara, 44121 Ferrara, Italy
| | - Hervé Berthommé
- Bioviron, Université Claude Bernard Lyon 1, 69100 Villeurbanne, France
| | - Michele Simonato
- Department of Neuroscience and Rehabilitation, Section of Pharmacology, University of Ferrara, 44121 Ferrara, Italy.,Division of Neuroscience, IRCCS San Raffaele Hospital, 20132 Milan, Italy
| |
Collapse
|
4
|
Wedekind MF, Cripe TP. Oncolytic Viruses and Their Potential as a Therapeutic Opportunity in Osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1258:77-89. [PMID: 32767235 DOI: 10.1007/978-3-030-43085-6_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Osteosarcoma remains an unmet medical need. Oncolytic viruses are gaining traction as novel cancer therapeutics. These viruses are either naturally nonpathogenic or engineered to be safe by specific genetic deletions yet retain the ability to infect and kill human cancer cells and elicit anticancer immunity. Some versions are being specifically designed and tested in patients with osteosarcoma, though due to their generalized mechanism of action most are being tested in patients across a broad range of cancer types. The activity of these viruses is impacted not only by the susceptibility of tumor cells to infection but also by the tumor microenvironment (TME) and by tumor immunogenicity. Here we review the field of oncolytic viruses with a particular emphasis on highlighting any available data in preclinical osteosarcoma models or in patients with osteosarcoma. While in general the viruses have been shown safe to administer to patients by a variety of routes, their therapeutic efficacy to date has been limited. Given the low rate of adverse events and the likely absence of long-term side effects, the utility of oncolytic viruses will most likely be realized when used in combination with other agents.
Collapse
Affiliation(s)
| | - Timothy P Cripe
- Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
5
|
Venkatesan JK, Rey-Rico A, Cucchiarini M. Current Trends in Viral Gene Therapy for Human Orthopaedic Regenerative Medicine. Tissue Eng Regen Med 2019; 16:345-355. [PMID: 31413939 PMCID: PMC6675832 DOI: 10.1007/s13770-019-00179-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/09/2019] [Accepted: 01/12/2019] [Indexed: 12/29/2022] Open
Abstract
Background Viral vector-based therapeutic gene therapy is a potent strategy to enhance the intrinsic reparative abilities of human orthopaedic tissues. However, clinical application of viral gene transfer remains hindered by detrimental responses in the host against such vectors (immunogenic responses, vector dissemination to nontarget locations). Combining viral gene therapy techniques with tissue engineering procedures may offer strong tools to improve the current systems for applications in vivo. Methods The goal of this work is to provide an overview of the most recent systems exploiting biomaterial technologies and therapeutic viral gene transfer in human orthopaedic regenerative medicine. Results Integration of tissue engineering platforms with viral gene vectors is an active area of research in orthopaedics as a means to overcome the obstacles precluding effective viral gene therapy. Conclusions In light of promising preclinical data that may rapidly expand in a close future, biomaterial-guided viral gene therapy has a strong potential for translation in the field of human orthopaedic regenerative medicine.
Collapse
Affiliation(s)
- Jagadeesh Kumar Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr, Bldg 37, 66421 Homburg/Saar, Germany
| | - Ana Rey-Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr, Bldg 37, 66421 Homburg/Saar, Germany
- Cell Therapy and Regenerative Medicine Unit, Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña, Campus de A Coruña, 15071 A Coruña, Spain
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr, Bldg 37, 66421 Homburg/Saar, Germany
| |
Collapse
|
6
|
Ventosa M, Wu Z, Lim F. Sustained FXN expression in dorsal root ganglia from a nonreplicative genomic HSV-1 vector. J Gene Med 2017; 19:376-386. [PMID: 29044877 DOI: 10.1002/jgm.2993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/06/2017] [Accepted: 10/07/2017] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Friedreich's ataxia (FA) is an autosomal recessive neurodegenerative disease caused by mutations in the frataxin gene (FXN), which lead to reduced levels of the essential mitochondrial protein frataxin. Currently, there is no effective cure. METHODS With the aim of developing a gene therapy for FA neuropathology, we describe the construction and preliminary characterization of a high-capacity nonreplicative genomic herpes simplex virus type 1 vector (H24B-FXNlac vector) carrying a reduced version of the human FXN genomic locus, comprising the 5-kb promoter and the FXN cDNA with the inclusion of intron 1. RESULTS We show that the transgene cassette contains the elements necessary to preserve physiological neuronal regulation of human FXN expression. Transduction of cultured fetal rat dorsal root ganglia neurons with the H24B-FXNlac vector results in sustained expression of human FXN transcripts and frataxin protein. Rat footpad inoculation with the H24B-FXNlac vector results in human FXN transgene delivery to the dorsal root ganglia, with expression persisting for at least 1 month. CONCLUSIONS The results of the present study support the feasibility of using this vector for sustained neuronal expression of human frataxin for FA gene therapy.
Collapse
Affiliation(s)
- Maria Ventosa
- Department of Neurosurgery, University of Michigan and VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Zetang Wu
- Department of Neurology, University of Michigan and VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Filip Lim
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
7
|
Collins DE, Reuter JD, Rush HG, Villano JS. Viral Vector Biosafety in Laboratory Animal Research. Comp Med 2017; 67:215-221. [PMID: 28662750 PMCID: PMC5482513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/16/2016] [Accepted: 12/28/2016] [Indexed: 06/07/2023]
Abstract
Viral vector research presents unique occupational health and safety challenges to institutions due to the rapid development of both in vivo and in vitro gene-editing technologies. Risks to human and animal health make it incumbent on institutions to appropriately evaluate viral vector usage in research on the basis of available information and governmental regulations and guidelines. Here we review the factors related to risk assessment regarding viral vector usage in animals and the relevant regulatory documents associated with this research, and we highlight the most commonly used viral vectors in research today. This review is particularly focused on the background, use in research and associated health and environmental risks related to adenoviral, adeno-associated viral, lentiviral, and herpesviral vectors.
Collapse
|
8
|
Adler B, Sattler C, Adler H. Herpesviruses and Their Host Cells: A Successful Liaison. Trends Microbiol 2017; 25:229-241. [DOI: 10.1016/j.tim.2016.11.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 11/08/2016] [Accepted: 11/15/2016] [Indexed: 12/11/2022]
|
9
|
Condreay JP, Kost TA, Mickelson CA. Emerging Considerations in Virus-Based Gene Transfer Systems. BIOLOGICAL SAFETY 2016:221-246. [DOI: 10.1128/9781555819637.ch10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
10
|
Abstract
For decades, effective cancer gene therapy has been a tantalising prospect; for a therapeutic modality potentially able to elicit highly effective and selective responses, definitive efficacy outcomes have often seemed out of reach. However, steady progress in vector development and accumulated experience from previous clinical studies has finally led the field to its first licensed therapy. Following a pivotal phase III trial, Imlygic (talimogene laherparepvec/T-Vec) received US approval as a treatment for cutaneous and subcutaneous melanoma in October 2015, followed several weeks later by its European authorisation. These represent the first approvals for an oncolytic virotherapy. Imlygic is an advanced-generation herpesvirus-based vector optimised for oncolytic and immunomodulatory activities. Many other oncolytic agents currently remain in development, providing hope that current success will be followed by other diverse vectors that may ultimately come to constitute a new class of clinical anti-cancer agents. In this review, we discuss some of the key oncolytic viral agents developed in the adenovirus and herpesvirus classes, and the prospects for further enhancing their efficacy by combining them with novel immunotherapeutic approaches.
Collapse
Affiliation(s)
- Alan E. Bilsland
- Institute of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre, Glasgow, G61 1QH, UK
| | | | - T. R. Jeffry Evans
- Institute of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre, Glasgow, G61 1QH, UK
| |
Collapse
|
11
|
Waters AM, Friedman GK, Ring EK, Beierle EA. Oncolytic virotherapy for pediatric malignancies: future prospects. Oncolytic Virother 2016; 5:73-80. [PMID: 27579298 PMCID: PMC4996251 DOI: 10.2147/ov.s96932] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Pediatric solid tumors remain a major health concern, with nearly 16,000 children diagnosed each year. Of those, ~2,000 succumb to their disease, and survivors often suffer from lifelong disability secondary to toxic effects of current treatments. Countless multimodality treatment regimens are being explored to make advances against this deadly disease. One targeted treatment approach is oncolytic virotherapy. Conditionally replicating viruses can infect tumor cells while leaving normal cells unharmed. Four viruses have been advanced to pediatric clinical trials, including herpes simplex virus-1, Seneca Valley virus, reovirus, and vaccinia virus. In this review, we discuss the mechanism of action of each virus, pediatric preclinical studies conducted to date, past and ongoing pediatric clinical trials, and potential future direction for these novel viral therapeutics.
Collapse
Affiliation(s)
- Alicia M Waters
- Department of Surgery, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gregory K Friedman
- Department of Pediatrics, Division of Hematology-Oncology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eric K Ring
- Department of Pediatrics, Division of Hematology-Oncology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Elizabeth A Beierle
- Department of Surgery, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
12
|
Katsu-Jiménez Y, Loría F, Corona JC, Díaz-Nido J. Gene Transfer of Brain-derived Neurotrophic Factor (BDNF) Prevents Neurodegeneration Triggered by FXN Deficiency. Mol Ther 2016; 24:877-89. [PMID: 26849417 PMCID: PMC4881769 DOI: 10.1038/mt.2016.32] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 01/21/2016] [Indexed: 02/07/2023] Open
Abstract
Friedreich's ataxia is a predominantly neurodegenerative disease caused by recessive mutations that produce a deficiency of frataxin (FXN). Here, we have used a herpesviral amplicon vector carrying a gene encoding for brain-derived neurotrophic factor (BDNF) to drive its overexpression in neuronal cells and test for its effect on FXN-deficient neurons both in culture and in the mouse cerebellum in vivo. Gene transfer of BDNF to primary cultures of mouse neurons prevents the apoptosis which is triggered by the knockdown of FXN gene expression. This neuroprotective effect of BDNF is also observed in vivo in a viral vector-based knockdown mouse cerebellar model. The injection of a lentiviral vector carrying a minigene encoding for a FXN-specific short hairpin ribonucleic acid (shRNA) into the mouse cerebellar cortex triggers a FXN deficit which is accompanied by significant apoptosis of granule neurons as well as loss of calbindin in Purkinje cells. These pathological changes are accompanied by a loss of motor coordination of mice as assayed by the rota-rod test. Coinjection of a herpesviral vector encoding for BDNF efficiently prevents both the development of cerebellar neuropathology and the ataxic phenotype. These data demonstrate the potential therapeutic usefulness of neurotrophins like BDNF to protect FXN-deficient neurons from degeneration.
Collapse
Affiliation(s)
- Yurika Katsu-Jiménez
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC) and Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Instituto de Investigaciones Sanitarias Hospital Puerta de Hierro-Majadahonda (IDIPHIM), Madrid, Spain
| | - Frida Loría
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC) and Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Instituto de Investigaciones Sanitarias Hospital Puerta de Hierro-Majadahonda (IDIPHIM), Madrid, Spain
| | - Juan Carlos Corona
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC) and Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Instituto de Investigaciones Sanitarias Hospital Puerta de Hierro-Majadahonda (IDIPHIM), Madrid, Spain
- Current address: Hospital Infantil de México “Federico Gómez”, México, D.F., México
| | - Javier Díaz-Nido
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC) and Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Instituto de Investigaciones Sanitarias Hospital Puerta de Hierro-Majadahonda (IDIPHIM), Madrid, Spain
| |
Collapse
|
13
|
Ma F, Wang C, Yoder WE, Westlund KN, Carlson CR, Miller CS, Danaher RJ. Efficacy of Herpes Simplex Virus Vector Encoding the Human Preproenkephalin Gene for Treatment of Facial Pain in Mice. J Oral Facial Pain Headache 2016; 30:42-50. [PMID: 26817032 DOI: 10.11607/ofph.1512] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
AIMS To determine whether herpes simplex virus-based vectors can efficiently transduce mouse trigeminal ganglion (TG) neurons and attenuate preexisting nerve injury-induced whisker pad mechanical hypersensitivity in a trigeminal inflammatory compression (TIC) neuropathic pain model. METHODS Tissue transduction efficiencies of replication-conditional and replication-defective vectors to mouse whisker pads after topical administration and subcutaneous injection were assessed using quantitative real-time PCR (qPCR). Tissue tropism and transgene expression were assessed using qPCR and reverse-transcriptase qPCR following topical application of the vectors. Whisker pad mechanical sensitivities of TIC-injured mice were determined using graduated von Frey fibers before and after application of human preproenkephalin expressing replication-conditional vector (KHPE). Data were analyzed using one-way analysis of variance (ANOVA) and post hoc tests. RESULTS Transduction of target TGs was 8- to 50-fold greater after topical application than subcutaneous injection and ≥ 100-fold greater for replication-conditional than replication-defective vectors. Mean KHPE loads remained constant in TGs (4.5-9.8 × 10(4) copies/TG) over 3 weeks but were below quantifiable levels (10 copies/tissue) within 2 weeks of application in other nontarget cephalic tissues examined. Transgene expression in TGs was maximal during 2 weeks after topical application (100-200 cDNA copies/mL) and was below quantifiable levels (1 cDNA copy/mL) in all nontarget tissues. Topical KHPE administration reduced TIC-related mechanical hypersensitivity on whisker pads 4-fold (P < .05) for at least 1 week. CONCLUSION Topically administered KHPE produced a significant antinociceptive effect in the TIC mouse model of chronic facial neuropathic pain. This is the first report in which a gene therapeutic approach reduced trigeminal pain-related behaviors in an established pain state in mice.
Collapse
|
14
|
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
|
15
|
Cole G, McCaffrey J, Ali AA, McCarthy HO. DNA vaccination for prostate cancer: key concepts and considerations. Cancer Nanotechnol 2015; 6:2. [PMID: 26161151 PMCID: PMC4488504 DOI: 10.1186/s12645-015-0010-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 05/16/2015] [Indexed: 11/10/2022] Open
Abstract
While locally confined prostate cancer is associated with a low five year mortality rate, advanced or metastatic disease remains a major challenge for healthcare professionals to treat and is usually terminal. As such, there is a need for the development of new, efficacious therapies for prostate cancer. Immunotherapy represents a promising approach where the host's immune system is harnessed to mount an anti-tumour effect, and the licensing of the first prostate cancer specific immunotherapy in 2010 has opened the door for other immunotherapies to gain regulatory approval. Among these strategies DNA vaccines are an attractive option in terms of their ability to elicit a highly specific, potent and wide-sweeping immune response. Several DNA vaccines have been tested for prostate cancer and while they have demonstrated a good safety profile they have faced problems with low efficacy and immunogenicity compared to other immunotherapeutic approaches. This review focuses on the positive aspects of DNA vaccines for prostate cancer that have been assessed in preclinical and clinical trials thus far and examines the key considerations that must be employed to improve the efficacy and immunogenicity of these vaccines.
Collapse
Affiliation(s)
- Grace Cole
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL Northern Ireland UK
| | - Joanne McCaffrey
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL Northern Ireland UK
| | - Ahlam A Ali
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL Northern Ireland UK
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL Northern Ireland UK
| |
Collapse
|