Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
J Immunother Cancer ; 8(2)2020 08.
Article in English | MEDLINE | ID: mdl-32759235

ABSTRACT

BACKGROUND: The aggressive clinical behavior of poorly differentiated and anaplastic thyroid cancers (PDTC and ATC) has proven challenging to treat, and survival beyond a few months from diagnosis is rare. Although 30%-60% of these tumors contain mutations in the BRAF gene, inhibitors designed specifically to target oncogenic BRAF have shown limited and only short-lasting therapeutic benefits as single agents, thus highlighting the need for improved treatment strategies, including novel combinations. METHODS: Using a BRAFV600E-driven mouse model of ATC, we investigated the therapeutic efficacy of the combination of BRAF inhibition and oncolytic herpes simplex virus (oHSV). Analyses of samples from tumor-bearing mice were performed to immunologically characterize the effects of different treatments. These immune data were used to inform the incorporation of immune checkpoint inhibitors into triple combination therapies. RESULTS: We characterized the immune landscape in vivo following BRAF inhibitor treatment and detected only modest immune changes. We, therefore, hypothesized that the addition of oncolytic virotherapy to BRAF inhibition in thyroid cancer would create a more favorable tumor immune microenvironment, boost the inflammatory status of tumors and improve BRAF inhibitor therapy. First, we showed that thyroid cancer cells were susceptible to infection with oHSV and that this process was associated with activation of the immune tumor microenvironment in vivo. Next, we showed improved therapeutic responses when combining oHSV and BRAF inhibition in vivo, although no synergistic effects were seen in vitro, further confirming that the dominant effect of oHSV in this context was likely immune-mediated. Importantly, both gene and protein expression data revealed an increase in activation of T cells and natural killer (NK) cells in the tumor in combination-treated samples. The benefit of combination oHSV and BRAF inhibitor therapy was abrogated when T cells or NK cells were depleted in vivo. In addition, we showed upregulation of PD-L1 and CTLA-4 following combined treatment and demonstrated that blockade of the PD-1/PD-L1 axis or CTLA-4 further improved combination therapy. CONCLUSIONS: The combination of oHSV and BRAF inhibition significantly improved survival in a mouse model of ATC by enhancing immune-mediated antitumor effects, and triple combination therapies, including either PD-1 or CTLA-4 blockade, further improved therapy.


Subject(s)
Oncolytic Virotherapy/methods , Proto-Oncogene Proteins B-raf/metabolism , Thyroid Neoplasms/genetics , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Herpesvirus 1, Human/pathogenicity , Humans , Male , Mice , Thyroid Neoplasms/pathology
2.
J Immunother Cancer ; 7(1): 214, 2019 08 10.
Article in English | MEDLINE | ID: mdl-31399043

ABSTRACT

BACKGROUND: Oncolytic viruses preferentially replicate in tumors as compared to normal tissue and promote immunogenic cell death and induction of host systemic anti-tumor immunity. HSV-1 was chosen for further development as an oncolytic immunotherapy in this study as it is highly lytic, infects human tumor cells broadly, kills mainly by necrosis and is a potent activator of both innate and adaptive immunity. HSV-1 also has a large capacity for the insertion of additional, potentially therapeutic, exogenous genes. Finally, HSV-1 has a proven safety and efficacy profile in patients with cancer, talimogene laherparepvec (T-VEC), an oncolytic HSV-1 which expresses GM-CSF, being the only oncolytic immunotherapy approach that has received FDA approval. As the clinical efficacy of oncolytic immunotherapy has been shown to be further enhanced by combination with immune checkpoint inhibitors, developing improved oncolytic platforms which can synergize with other existing immunotherapies is a high priority. In this study we sought to further optimize HSV-1 based oncolytic immunotherapy through multiple approaches to maximize: (i) the extent of tumor cell killing, augmenting the release of tumor antigens and danger-associated molecular pattern (DAMP) factors; (ii) the immunogenicity of tumor cell death; and (iii) the resulting systemic anti-tumor immune response. METHODS: To sample the wide diversity amongst clinical strains of HSV-1, twenty nine new clinical strains isolated from cold sores from otherwise healthy volunteers were screened across a panel of human tumor cell lines to identify the strain with the most potent tumor cell killing ability, which was then used for further development. Following deletion of the genes encoding ICP34.5 and ICP47 to provide tumor selectivity, the extent of cell killing and the immunogenicity of cell death was enhanced through insertion of a gene encoding a truncated, constitutively highly fusogenic form of the envelope glycoprotein of gibbon ape leukemia virus (GALV-GP-R-). A number of further armed derivatives of this virus were then constructed intended to further enhance the anti-tumor immune response which was generated following fusion-enhanced, oncolytic virus replication-mediated cell death. These viruses expressed GMCSF, an anti-CTLA-4 antibody-like molecule, CD40L, OX40L and/or 4-1BB, each of which is expected to act predominantly at the site and time of immune response initiation. Expression of these proteins was confirmed by ELISA and/or western blotting. Immunogenic cell death was assessed by measuring the levels of HMGB1 and ATP from cell free supernatants from treated cells, and by measuring the surface expression of calreticulin. GALV-GP-R- mediated cell to cell fusion and killing was tested in a range of tumor cell lines in vitro. Finally, the in vivo therapeutic potential of these viruses was tested using human A549 (lung cancer) and MDA-MB-231(breast cancer) tumor nude mouse xenograft models and systemic anti-tumor effects tested using dual flank syngeneic 4434 (melanoma), A20 (lymphoma) mouse tumor models alone and in combination with a murine anti-PD1 antibody, and 9 L (gliosarcoma) tumors in rats. RESULTS: The twenty nine clinical strains of HSV-1 isolated and tested demonstrated a broad range of tumor cell killing abilities allowing the most potent strain to be identified which was then used for further development. Oncolytic ability was demonstrated to be further augmented by the expression of GALV-GP-R- in a range of tumor cell lines in vitro and in mouse xenograft models in nude mice. The expression of GALV-GP-R- was also demonstrated to lead to enhanced immunogenic cell death in vitro as confirmed by the increased release of HMGB1 and ATP and increased levels of calreticulin on the cell surface. Experiments using the rat 9 L syngeneic tumor model demonstrated that GALV-GP-R- expression increased abscopal uninjected (anenestic) tumor responses and data using mouse 4434 tumors demonstrated that virus treatment increased CD8+ T cell levels both in the injected and uninjected tumor, and also led to increased expression of PD-L1. A combination study using varying doses of a virus expressing GALV-GP-R- and mGM-CSF and an anti-murine PD1 antibody showed enhanced anti-tumor effects with the combination which was most evident at low virus doses, and also lead to immunological memory. Finally, treatment of mice with derivatives of this virus which additionally expressed anti-mCTLA-4, mCD40L, m4-1BBL, or mOX40L demonstrated enhanced activity, particularly in uninjected tumors. CONCLUSION: The new HSV-1 based platform described provides a potent and versatile approach to developing new oncolytic immunotherapies for clinical use. Each of the modifications employed was demonstrated to aid in optimizing the potential of the virus to both directly kill tumors and to lead to systemic therapeutic benefit. For clinical use, these viruses are expected to be most effective in combination with other anti-cancer agents, in particular PD1/L1-targeted immune checkpoint blockade. The first virus from this program (expressing GALV-GP-R- and hGM-CSF) has entered clinical development alone and in combination with anti-PD1 therapy in a number of tumor types (NCT03767348).


Subject(s)
Herpes Simplex/drug therapy , Herpesvirus 1, Human/pathogenicity , Immunotherapy/methods , Oncolytic Virotherapy/methods , Animals , Female , Humans , Male , Mice , Mice, Nude
3.
J Immunother Cancer ; 6(1): 78, 2018 08 06.
Article in English | MEDLINE | ID: mdl-30081947

ABSTRACT

In this White Paper, we discuss the current state of microbial cancer therapy. This paper resulted from a meeting ('Microbial Based Cancer Therapy') at the US National Cancer Institute in the summer of 2017. Here, we define 'Microbial Therapy' to include both oncolytic viral therapy and bacterial anticancer therapy. Both of these fields exploit tumor-specific infectious microbes to treat cancer, have similar mechanisms of action, and are facing similar challenges to commercialization. We designed this paper to nucleate this growing field of microbial therapeutics and increase interactions between researchers in it and related fields. The authors of this paper include many primary researchers in this field. In this paper, we discuss the potential, status and opportunities for microbial therapy as well as strategies attempted to date and important questions that need to be addressed. The main areas that we think will have the greatest impact are immune stimulation, control of efficacy, control of delivery, and safety. There is much excitement about the potential of this field to treat currently intractable cancer. Much of the potential exists because these therapies utilize unique mechanisms of action, difficult to achieve with other biological or small molecule drugs. By better understanding and controlling these mechanisms, we will create new therapies that will become integral components of cancer care.


Subject(s)
Bacteria , Biological Therapy/methods , Genetic Vectors , Neoplasms/prevention & control , Neoplasms/therapy , Viruses , Animals , Bacteria/genetics , Biological Therapy/standards , Biological Therapy/trends , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Clinical Studies as Topic , Combined Modality Therapy , Drug Evaluation, Preclinical , Genetic Engineering , Genetic Vectors/genetics , Humans , Neoplasms/etiology , Oncolytic Virotherapy , Treatment Outcome , Viruses/genetics
4.
Curr Opin Virol ; 13: 93-100, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26121656

ABSTRACT

Viruses have been suggested to be useful as anti-cancer agents since the early 20th century, although following the advent of chemotherapy and radiotherapy work largely stopped until the 1990s when a number of groups began to explore the use of engineered viruses. This overview summarizes the development of the field from the 1990s to the present day, an era when oncolytic viruses have now demonstrated clear clinical benefit to patients. The hurdles and challenges which needed to be overcome are discussed, and in particular the importance of the immune component in achieving a therapeutic effect is highlighted. Today, oncolytic therapy is generally thought of as an immunotherapy, the term 'oncolytic immunotherapy' having been widely adopted. With the advent of immuno-oncology drugs based on immune checkpoint blockade, a clear rationale for synergy between the two approaches, and initial pre-clinical and clinical data suggesting this to be the case, it might be expected that oncolytic immunotherapy combined with checkpoint blockade will provide a cornerstone of future cancer treatment.


Subject(s)
Immunotherapy , Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Animals , Humans , Neoplasms/immunology , Neoplasms/virology , Oncolytic Viruses/genetics
5.
J Clin Oncol ; 33(25): 2780-8, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26014293

ABSTRACT

PURPOSE: Talimogene laherparepvec (T-VEC) is a herpes simplex virus type 1-derived oncolytic immunotherapy designed to selectively replicate within tumors and produce granulocyte macrophage colony-stimulating factor (GM-CSF) to enhance systemic antitumor immune responses. T-VEC was compared with GM-CSF in patients with unresected stage IIIB to IV melanoma in a randomized open-label phase III trial. PATIENTS AND METHODS: Patients with injectable melanoma that was not surgically resectable were randomly assigned at a two-to-one ratio to intralesional T-VEC or subcutaneous GM-CSF. The primary end point was durable response rate (DRR; objective response lasting continuously ≥ 6 months) per independent assessment. Key secondary end points included overall survival (OS) and overall response rate. RESULTS: Among 436 patients randomly assigned, DRR was significantly higher with T-VEC (16.3%; 95% CI, 12.1% to 20.5%) than GM-CSF (2.1%; 95% CI, 0% to 4.5%]; odds ratio, 8.9; P < .001). Overall response rate was also higher in the T-VEC arm (26.4%; 95% CI, 21.4% to 31.5% v 5.7%; 95% CI, 1.9% to 9.5%). Median OS was 23.3 months (95% CI, 19.5 to 29.6 months) with T-VEC and 18.9 months (95% CI, 16.0 to 23.7 months) with GM-CSF (hazard ratio, 0.79; 95% CI, 0.62 to 1.00; P = .051). T-VEC efficacy was most pronounced in patients with stage IIIB, IIIC, or IVM1a disease and in patients with treatment-naive disease. The most common adverse events (AEs) with T-VEC were fatigue, chills, and pyrexia. The only grade 3 or 4 AE occurring in ≥ 2% of T-VEC-treated patients was cellulitis (2.1%). No fatal treatment-related AEs occurred. CONCLUSION: T-VEC is the first oncolytic immunotherapy to demonstrate therapeutic benefit against melanoma in a phase III clinical trial. T-VEC was well tolerated and resulted in a higher DRR (P < .001) and longer median OS (P = .051), particularly in untreated patients or those with stage IIIB, IIIC, or IVM1a disease. T-VEC represents a novel potential therapy for patients with metastatic melanoma.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antineoplastic Agents/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/drug effects , Herpesvirus 1, Human , Immunotherapy/methods , Melanoma/drug therapy , Melanoma/immunology , Oncolytic Virotherapy , Oncolytic Viruses , Skin Neoplasms/drug therapy , Skin Neoplasms/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Chills/chemically induced , Fatigue/chemically induced , Female , Fever/chemically induced , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Injections, Intralesional , Male , Melanoma/mortality , Melanoma/prevention & control , Melanoma/secondary , Middle Aged , Neoplasm Staging , Odds Ratio , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Skin Neoplasms/prevention & control , Survival Analysis , Time Factors , Treatment Outcome , United States/epidemiology
6.
Oncolytic Virother ; 3: 11-20, 2014.
Article in English | MEDLINE | ID: mdl-27512660

ABSTRACT

Oncolytic viruses that selectively lyse tumor cells with minimal damage to normal cells are a new area of therapeutic development in oncology. An attenuated herpesvirus encoding the granulocyte-macrophage colony stimulating factor (GM-CSF), known as talimogene laherparepvec (T-VEC), has been identified as an attractive oncolytic virus for cancer therapy based on preclinical tumor studies and results from early-phase clinical trials and a large randomized Phase III study in melanoma. In this review, we discuss the basic biology of T-VEC, describe the role of GM-CSF as an immune adjuvant, summarize the preclinical data, and report the outcomes of published clinical trials using T-VEC. The emerging data suggest that T-VEC is a safe and potentially effective antitumor therapy in malignant melanoma and represents the first oncolytic virus to demonstrate therapeutic activity against human cancer in a randomized, controlled Phase III study.

7.
BMC Cancer ; 10: 486, 2010 Sep 13.
Article in English | MEDLINE | ID: mdl-20836854

ABSTRACT

BACKGROUND: Delivery of small interfering RNA (siRNA) to tumours remains a major obstacle for the development of RNA interference (RNAi)-based therapeutics. Following the promising pre-clinical and clinical results with the oncolytic herpes simplex virus (HSV) OncoVEX GM-CSF, we aimed to express RNAi triggers from oncolytic HSV, which although has the potential to improve treatment by silencing tumour-related genes, was not considered possible due to the highly oncolytic properties of HSV. METHODS: To evaluate RNAi-mediated silencing from an oncolytic HSV backbone, we developed novel replicating HSV vectors expressing short-hairpin RNA (shRNA) or artificial microRNA (miRNA) against the reporter genes green fluorescent protein (eGFP) and ß-galactosidase (lacZ). These vectors were tested in non-tumour cell lines in vitro and tumour cells that are moderately susceptible to HSV infection both in vitro and in mice xenografts in vivo. Silencing was assessed at the protein level by fluorescent microscopy, x-gal staining, enzyme activity assay, and western blotting. RESULTS: Our results demonstrate that it is possible to express shRNA and artificial miRNA from an oncolytic HSV backbone, which had not been previously investigated. Furthermore, oncolytic HSV-mediated delivery of RNAi triggers resulted in effective and specific silencing of targeted genes in tumour cells in vitro and tumours in vivo, with the viruses expressing artificial miRNA being comprehensibly more effective. CONCLUSIONS: This preliminary data provide the first demonstration of oncolytic HSV-mediated expression of shRNA or artificial miRNA and silencing of targeted genes in tumour cells in vitro and in vivo. The vectors developed in this study are being adapted to silence tumour-related genes in an ongoing study that aims to improve the effectiveness of oncolytic HSV treatment in tumours that are moderately susceptible to HSV infection and thus, potentially improve response rates seen in human clinical trials.


Subject(s)
Gene Silencing , Gliosarcoma/therapy , MicroRNAs/physiology , Oncolytic Virotherapy , RNA Interference , RNA, Small Interfering/physiology , Simplexvirus/physiology , Animals , Blotting, Western , Cells, Cultured , Cricetinae , Genetic Therapy , Genetic Vectors/administration & dosage , Gliosarcoma/genetics , Gliosarcoma/virology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Herpes Simplex/genetics , Herpes Simplex/therapy , Herpes Simplex/virology , Humans , In Vitro Techniques , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Messenger/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
8.
Expert Opin Biol Ther ; 10(1): 89-103, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20420517

ABSTRACT

IMPORTANCE OF THE FIELD: Pain is a hugely important area of research attracting considerable academic and commercial interest. However, the application of RNA interference (RNAi) to the study of nociceptive processes and the development of new analgesics has been limited by the specific challenges associated with the delivery of RNAi triggers to the cell bodies of sensory neurons in the dorsal root ganglia (DRG). AREAS COVERED IN THIS REVIEW: In the past five years, delivery of small-interfering RNA (siRNA) to the DRG and spinal cord has achieved effective and specific silencing of targeted genes in various animal models of pain. However, delivery of short-hairpin RNA (shRNA) or artificial microRNA (miRNA) to sensory neurons in vivo has not been feasible using most delivery systems currently available. WHAT THE READER WILL GAIN: Replication-defective vectors based on herpes simplex virus (HSV), which are particularly efficient at targeting DRG neurons, have been recently engineered to express shRNA and artificial miRNA. Whilst silencing induced by siRNA is transient and requires relatively high doses of silencing triggers, HSV-mediated expression of shRNA/miRNA in sensory neurons allows silencing of targeted genes for at least one week following a single injection. TAKE HOME MESSAGE: The potential to use inducible or tissue-specific promoters and to simultaneously silence multiple gene targets, in addition to recent studies suggesting that artificial miRNAs may have improved safety profiles, hold clear advantages for the use of miRNA-based vectors for gene silencing in sensory neurons.


Subject(s)
Gene Silencing , Pain Management , RNA Interference , Sensory Receptor Cells/metabolism , Simplexvirus/genetics , Animals , Genetic Therapy , Humans , MicroRNAs/physiology , Pain/genetics , RNA, Small Interfering/pharmacology , Virus Replication
9.
Arch Otolaryngol Head Neck Surg ; 136(2): 151-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20157061

ABSTRACT

OBJECTIVE: To determine if prodrug conversion of fluorocytosine to fluorouracil by an engineered herpes virus, OncoVEX(GALV/CD), enhances oncolytic therapy of head and neck squamous cell carcinoma. DESIGN: We assessed the ability of OncoVEX(GALV/CD) and OncoVEX(GFP) to infect, replicate within, and lyse 4 head and neck squamous cell carcinoma lines in vitro. The effects of adding fluorocytosine with OncoVEX(GALV/CD) were evaluated. RESULTS: Head and neck squamous cell carcinoma was permissive to green fluorescent protein expression in100% of cells by OncoVEX(GFP) at a multiplicity of infection of 1 after 48 hours and supported logarithmic viral replication. Virus caused more than 60% cell death 6 days after exposure to virus at a multiplicity of infection of 0.1 in 3 of the 4 cell lines. Fluorocytosine did not enhance cytotoxicity induced by OncoVEX(GALV/CD) at a multiplicity of infection of 0.1. However, for the least-sensitive SCC25 cell line, virus at a multiplicity of infection of 0.01 was cytotoxic to only 4% of cells after 6 days but was cytotoxic to 35% of cells with fluorocytosine. CONCLUSIONS: OncoVEX(GALV/CD) efficiently infects, replicates within, and lyses head and neck squamous cell carcinoma at relatively low viral doses. Prodrug conversion by cytosine deaminase did not enhance therapy at viral doses that cause efficient cytotoxicity but may have beneficial effects in less-sensitive cell lines at low viral doses.


Subject(s)
Carcinoma, Squamous Cell/therapy , Flucytosine/therapeutic use , Head and Neck Neoplasms/therapy , Herpesvirus 1, Human/genetics , Oncolytic Virotherapy , Prodrugs/therapeutic use , Cell Line, Tumor , Cytosine Deaminase/biosynthesis , Gene Transfer Techniques , Genetic Engineering , Genetic Vectors , Humans , Leukemia Virus, Gibbon Ape/genetics
10.
Methods Mol Biol ; 542: 551-64, 2009.
Article in English | MEDLINE | ID: mdl-19565922

ABSTRACT

A large number of oncolytic viral vectors are currently under clinical development for cancer therapy. Herpes simplex virus type 1 (HSV-1) has demonstrated particular promise in this field, showing genetically engineered selective tumor replication and cytotoxicity in a wide variety of tumor types, without damaging healthy tissues. Enhanced activity has been observed when a range of therapeutic genes has been inserted into various oncolytic HSV genomes. Here, we discuss methods used to develop and characterize an oncolytic HSV virus that combines expression of a highly potent prodrug activating gene (yeast cytosine deaminase/uracil phosphoribosyltransferase fusion [Fcy::Fur]) and the fusogenic glycoprotein from gibbon ape leukemia virus (GALV) for enhanced local tumor control.


Subject(s)
Genetic Vectors/genetics , Glycoproteins/metabolism , Molecular Biology/methods , Neoplasms/therapy , Oncolytic Viruses/genetics , Prodrugs/pharmacology , Simplexvirus/genetics , Animals , Cell Death , Cell Line, Tumor , Cloning, Molecular , Humans , Plasmids , Recombinant Fusion Proteins , Saccharomyces cerevisiae
11.
Nucleic Acids Res ; 36(14): e86, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18583367

ABSTRACT

Considerable interest has been focused on inducing RNA interference (RNAi) in neurons to study gene function and identify new targets for disease intervention. Although small interfering RNAs (siRNAs) have been used to silence genes in neurons, in vivo delivery of RNAi remains a major challenge limiting its applications. We have developed a highly efficient method for in vivo gene silencing in dorsal root ganglia (DRG) using replication-defective herpes simplex viral (HSV-1) vectors. HSV-mediated delivery of short-hairpin RNA (shRNA) targeting reporter genes resulted in highly effective and specific silencing in neuronal and non-neuronal cells in culture and in the DRG of mice in vivo including in a transgenic mouse model. We further establish proof of concept by demonstrating in vivo silencing of the endogenous trpv1 gene. These data are the first to show silencing in DRG neurons in vivo by vector-mediated delivery of shRNA. Our results support the utility of HSV vectors for gene silencing in peripheral neurons and the potential application of this technology to the study of nociceptive processes and in pain gene target validation studies.


Subject(s)
Herpesvirus 1, Human/genetics , Neurons/metabolism , RNA Interference , RNA, Untranslated/metabolism , Animals , Cell Line , Cells, Cultured , Ganglia, Spinal/metabolism , Genetic Vectors , Mice , Mice, Inbred BALB C , RNA, Untranslated/genetics , Rats , TRPV Cation Channels/genetics
12.
Neurosurgery ; 61(3): 586-94; discussion 594-5, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17881973

ABSTRACT

OBJECTIVE: Several reports recently suggested that vascular endothelial growth factor (VEGF) may have a therapeutic benefit against experimental cerebral infarction animal models. In addition, bone marrow stromal cells (BMSCs) are known to have therapeutic potency in improving neurological deficits after occlusive cerebrovascular diseases. In the present study, we evaluated the hypothesis that intracerebral transplantation of VEGF gene-transferred BMSCs could provide a greater therapeutic effect than intracerebral transplantation of native (non-gene-transformed) BMSCs by using a transient middle cerebral artery occlusion (MCAO) rat model. METHODS: Adult Wistar rats (Japan SLC, Inc., Hamamatsu, Japan) were anesthetized. VEGF gene-transferred BMSCs engineered with a replication-deficient herpes simplex virus type 1 1764/4-/pR19-hVEGF165 vector, native BMSCs, or phosphate-buffered saline were administered intracerebrally 24 hours after transient MCAO. All animals underwent behavioral testing for 28 days, and the infarction volume was determined 14 days after MCAO. The brain water contents in the ipsilateral and contralateral hemispheres of the MCAO were measured 2 and 7 days after the MCAO. Fourteen days after MCAO, immunohistochemical staining for VEGF was performed. RESULTS: The group receiving VEGF-modified BMSCs demonstrated significant functional recovery compared with those receiving native BMSCs. Fourteen days after the MCAO, there was a significantly lower infarct volume without aggravating cerebral edema in the group treated with VEGF gene-modified BMSCs compared with the control groups. The transplanted VEGF gene-modified BMSCs strongly expressed VEGF protein for at least 14 days. CONCLUSION: Our data suggest that the intracerebral transplantation of VEGF gene-transferred BMSCs may provide a more potent autologous cell transplantation therapy for stroke than the transplantation of native BMSCs alone.


Subject(s)
Bone Marrow Transplantation/methods , Brain Ischemia/pathology , Gene Transfer Techniques , Genetic Vectors/genetics , Herpesvirus 1, Human/genetics , Vascular Endothelial Growth Factor A/genetics , Animals , Brain Ischemia/genetics , Brain Ischemia/surgery , Genetic Engineering/methods , Genetic Vectors/administration & dosage , Humans , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/surgery , Rats , Rats, Wistar , Stromal Cells/transplantation , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/therapeutic use
13.
J Neurosci Res ; 85(11): 2332-42, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17549731

ABSTRACT

Many therapeutic interventions using neurotrophic factors or pharmacological agents have focused on secondary degeneration after spinal cord injury (SCI) to reduce damaged areas and promote axonal regeneration and functional recovery. Hepatocyte growth factor (HGF), which was identified as a potent mitogen for mature hepatocytes and a mediator of inflammatory responses to tissue injury, has recently been highlighted as a potent neurotrophic and angiogenic factor in the central nervous system (CNS). In the present study, we revealed that the extent of endogenous HGF up-regulation was less than that of c-Met, an HGF receptor, during the acute phase of SCI and administered exogenous HGF into injured spinal cord using a replication-incompetent herpes simplex virous-1 (HSV-1) vector to determine whether HGF exerts beneficial effects and promotes functional recovery after SCI. This treatment resulted in the significant promotion of neuron and oligodendrocyte survival, angiogenesis, axonal regrowth, and functional recovery after SCI. These results suggest that HGF gene delivery to the injured spinal cord exerts multiple beneficial effects and enhances endogenous repair after SCI. This is the first study to demonstrate the efficacy of HGF for SCI.


Subject(s)
Hepatocyte Growth Factor/metabolism , Nerve Regeneration/physiology , Recovery of Function/physiology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/therapy , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression , Genetic Therapy , Genetic Vectors , Hepatocyte Growth Factor/administration & dosage , Immunohistochemistry , Injections, Spinal , Neovascularization, Physiologic , Nerve Growth Factors/administration & dosage , Nerve Growth Factors/metabolism , Neurons/metabolism , Oligodendroglia/metabolism , Proto-Oncogene Proteins c-met/metabolism , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
14.
J Virol ; 81(12): 6326-38, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17428858

ABSTRACT

Mature dendritic cells (DCs) are the most potent antigen-presenting cells within the human immune system. However, Herpes simplex virus type 1 (HSV-1) is able to interfere with DC biology and to establish latency in infected individuals. In this study, we provide new insights into the mechanism by which HSV-1 disarms DCs by the manipulation of CD83, a functionally important molecule for DC activation. Fluorescence-activated cell sorter (FACS) analyses revealed a rapid downmodulation of CD83 surface expression within 6 to 8 h after HSV-1 infection, in a manner strictly dependent on viral gene expression. Soluble CD83 enzyme-linked immunosorbent assays, together with Western blot analysis, demonstrated that CD83 rapidly disappears from the cell surface after contact with HSV-1 by a mechanism that involves protein degradation rather than shedding of CD83 from the cell surface into the medium. Infection experiments with an ICP0 deletion mutant demonstrated an important role for this viral immediate-early protein during CD83 degradation, since this particular mutant strain leads to strongly reduced CD83 degradation. This hypothesis was further strengthened by cotransfection of plasmids expressing CD83 and ICP0 into 293T cells, which led to significantly reduced accumulation of CD83. In strong contrast, transfection of plasmids expressing CD83 and a mutant ICP0 defective in its RING finger-mediated E3 ubiquitin ligase function did not reduce CD83 expression. Inhibition of the proteasome, the cellular protein degradation machinery, almost completely restored CD83 surface expression during HSV-1 infection, indicating that proteasome-mediated degradation and HSV-1 ICP0 play crucial roles in this novel viral immune escape mechanism.


Subject(s)
Antigens, CD/biosynthesis , Dendritic Cells/cytology , Herpesvirus 1, Human/metabolism , Immunoglobulins/biosynthesis , Membrane Glycoproteins/biosynthesis , Proteasome Endopeptidase Complex/metabolism , Cell Line , Cell Separation , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Herpes Simplex/immunology , Humans , Immediate-Early Proteins/metabolism , Kinetics , Leukocytes, Mononuclear/virology , Transfection , Ubiquitin-Protein Ligases/metabolism , CD83 Antigen
15.
Clin Cancer Res ; 12(22): 6737-47, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17121894

ABSTRACT

PURPOSE: To conduct a phase I clinical trial with a second-generation oncolytic herpes simplex virus (HSV) expressing granulocyte macrophage colony-stimulating factor (Onco VEXGM-CSF) to determine the safety profile of the virus, look for evidence of biological activity, and identify a dosing schedule for later studies. EXPERIMENTAL DESIGN: The virus was administered by intratumoral injection in patients with cutaneous or s.c. deposits of breast, head and neck and gastrointestinal cancers, and malignant melanoma who had failed prior therapy. Thirteen patients were in a single-dose group, where doses of 10(6), 10(7), and 10(8) plaque-forming units (pfu)/mL were tested, and 17 patients were in a multidose group testing a number of dose regimens. RESULTS: The virus was generally well tolerated with local inflammation, erythema, and febrile responses being the main side effects. The local reaction to injection was dose limiting in HSV-seronegative patients at 10(7) pfu/mL. The multidosing phase thus tested seroconverting HSV-seronegative patients with 10(6) pfu/mL followed by multiple higher doses (up to 10(8) pfu/mL), which was well tolerated by all patients. Biological activity (virus replication, local reactions, granulocyte macrophage colony-stimulating factor expression, and HSV antigen-associated tumor necrosis), was observed. The duration of local reactions and virus replication suggested that dosing every 2 to 3 weeks was appropriate. Nineteen of 26 patient posttreatment biopsies contained residual tumor of which 14 showed tumor necrosis, which in some cases was extensive, or apoptosis. In all cases, areas of necrosis also strongly stained for HSV. The overall responses to treatment were that three patients had stable disease, six patients had tumors flattened (injected and/or uninjected lesions), and four patients showed inflammation of uninjected as well as the injected tumor, which, in nearly all cases, became inflamed. CONCLUSIONS: Onco VEXGM-CSF is well tolerated and can be safely administered using the multidosing protocol described. Evidence of an antitumor effect was seen.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Neoplasms/therapy , Oncolytic Virotherapy/methods , Simplexvirus , Adult , Aged , Aged, 80 and over , Antibodies, Antinuclear/blood , Biopsy , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Carcinoma/pathology , Carcinoma/therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Cytokines/blood , Female , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/therapy , Humans , Male , Melanoma/pathology , Melanoma/therapy , Middle Aged , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses , Recombinant Proteins , Simplexvirus/immunology , Simplexvirus/metabolism , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Treatment Outcome
16.
Clin Cancer Res ; 12(22): 6853-62, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17121907

ABSTRACT

PURPOSE: Oncolytic herpes simplex virus type 1 (HSV-1) vectors show considerable promise as agents for cancer therapy. We have developed a novel recombinant HSV-1 virus (JS1/34.5-/47-) for purging of occult breast cancer cells from bone marrow of patients. Here, we evaluate the therapeutic efficacy of this oncolytic virus. EXPERIMENTAL DESIGN: Electron microscopy was used to determine whether human breast cancer and bone marrow cells are permissive for JS1/34.5-/47- infection. Subsequently, the biological effects of JS1/34.5-/47- infection on human breast cancer cells and bone marrow were established using cell proliferation and colony formation assays, and the efficiency of cell kill was evaluated. Finally, the efficiency of JS1/34.5-/47- purging of breast cancer cells was examined in cocultures of breast cancer cells with bone marrow as well as bone marrow samples from high-risk breast cancer patients. RESULTS: We show effective killing of human breast cancer cell lines with the JS1/34.5-/47- virus. Furthermore, we show that treatment with JS1/34.5-/47- can significantly inhibit the growth of breast cancer cell lines without affecting cocultured mononuclear hematopoietic cells. Finally, we have found that the virus is effective in destroying disseminated tumors cells in bone marrow taken from breast cancer patients, without affecting the hematopoietic contents in these samples. CONCLUSION: Collectively, our data show that the JS1/34.5-/47- virus can selectively target breast cancer cells while sparing hematopoietic cells, suggesting that JS1/34.5-/47- can be used to purge contaminating breast cancer cells from human bone marrow in the setting of autologous hematopoietic cell transplantation.


Subject(s)
Adenocarcinoma/pathology , Bone Marrow/pathology , Breast Neoplasms/pathology , Herpesvirus 1, Human , Oncolytic Viruses/physiology , Adenocarcinoma/virology , Biomarkers, Tumor/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/virology , Breast Neoplasms/therapy , Cell Death , Cell Line, Tumor , Cell Survival , Flow Cytometry , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/pathogenicity , Herpesvirus 1, Human/physiology , Humans , Keratin-19/metabolism , Mammary Glands, Human/pathology , Mammary Glands, Human/virology , Oncolytic Virotherapy/adverse effects
17.
Cancer Res ; 66(9): 4835-42, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16651439

ABSTRACT

We have previously developed an oncolytic herpes simplex virus-1 based on a clinical virus isolate, which was deleted for ICP34.5 to provide tumor selected replication and ICP47 to increase antigen presentation as well as tumor selective virus replication. A phase I/II clinical trial using a version of this virus expressing granulocyte macrophage colony-stimulating factor has shown promising results. The work reported here aimed to develop a version of this virus in which local tumor control was further increased through the combined expression of a highly potent prodrug activating gene [yeast cytosine deaminase/uracil phospho-ribosyltransferase fusion (Fcy::Fur)] and the fusogenic glycoprotein from gibbon ape leukemia virus (GALV), which it was hoped would aid the spread of the activated prodrug through the tumor. Viruses expressing the two genes individually or in combination were constructed and tested, showing (a) GALV and/or Fcy::Fur expression did not affect virus growth; (b) GALV expression causes cell fusion and increases the tumor cell killing at least 30-fold in vitro and tumor shrinkage 5- to 10-fold in vivo; (c) additional expression of Fcy::Fur combined with 5-fluorocytosine administration improves tumor shrinkage further. These results indicate, therefore, that the combined expression of the GALV protein and Fcy::Fur provides a highly potent oncolytic virus with improved capabilities for local tumor control. It is intended to enter the GALV/Fcy::Fur expressing virus into clinical development for the treatment of tumor types, such as pancreatic or lung cancer, where local control would be anticipated to be clinically advantageous.


Subject(s)
Fibrosarcoma/therapy , Flucytosine/pharmacokinetics , Leukemia Virus, Gibbon Ape/genetics , Membrane Glycoproteins/genetics , Oncolytic Virotherapy/methods , Simplexvirus/physiology , Animals , Biotransformation , Cell Fusion , Combined Modality Therapy , Cytosine Deaminase/biosynthesis , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Fibrosarcoma/virology , Flucytosine/pharmacology , Fluorouracil/pharmacokinetics , Fluorouracil/pharmacology , Humans , Leukemia Virus, Gibbon Ape/metabolism , Leukemia Virus, Gibbon Ape/physiology , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred BALB C , Pentosyltransferases/biosynthesis , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , Prodrugs/pharmacokinetics , Rats , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Simplexvirus/genetics , Virus Replication , Xenograft Model Antitumor Assays
18.
J Cereb Blood Flow Metab ; 26(9): 1176-88, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16421510

ABSTRACT

Occlusive cerebrovascular disease leads to brain ischemia that causes neurological deficits. Here we introduce a new strategy combining mesenchymal stromal cells (MSCs) and ex vivo hepatocyte growth factor (HGF) gene transferring with a multimutated herpes simplex virus type-1 vector in a rat transient middle cerebral artery occlusion (MCAO) model. Gene-transferred MSCs were intracerebrally transplanted into the rats' ischemic brains at 2 h (superacute) or 24 h (acute) after MCAO. Behavioral tests showed significant improvement of neurological deficits in the HGF-transferred MSCs (MSC-HGF)-treated group compared with the phosphate-buffered saline (PBS)-treated and MSCs-only-treated group. The significant difference of infarction areas on day 3 was detected only between the MSC-HGF group and the PBS group with the superacute treatment, but was detected among each group on day 14 with both transplantations. After the superacute transplantation, we detected abundant expression of HGF protein in the ischemic brain of the MSC-HGF group compared with others on day 1 after treatment, and it was maintained for at least 2 weeks. Furthermore, we determined that the increased expression of HGF was derived from the transferred HGF gene in gene-modified MSCs. The percentage of apoptosis-positive cells in the ischemic boundary zone (IBZ) was significantly decreased, while that of remaining neurons in the cortex of the IBZ was significantly increased in the MSC-HGF group compared with others. The present study shows that combined therapy is more therapeutically efficient than MSC cell therapy alone, and it may extend the therapeutic time window from superacute to acute phase.


Subject(s)
Bone Marrow Transplantation/methods , Genetic Therapy , Hepatocyte Growth Factor/genetics , Herpesvirus 1, Human/genetics , Stroke/therapy , Stromal Cells/transplantation , Animals , Apoptosis/physiology , Brain/pathology , Enzyme-Linked Immunosorbent Assay , Genetic Vectors , Hepatocyte Growth Factor/biosynthesis , Immunohistochemistry , In Situ Nick-End Labeling , Infarction, Middle Cerebral Artery/pathology , Middle Cerebral Artery/physiology , Rats , Rats, Wistar , Stroke/pathology , Treatment Outcome
19.
Stroke ; 36(12): 2725-30, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16282547

ABSTRACT

BACKGROUND AND PURPOSE: Fibroblast growth factor-2 (FGF-2) administration and bone marrow stromal cell (MSC) transplantation could improve neurological deficits after occlusive cerebrovascular disease. In the present study, we examined the effects of neurological improvement after transient middle cerebral artery occlusion (MCAO) in rats by a novel therapeutic strategy with FGF-2 gene-transferred MSCs by the herpes simplex virus type 1 (HSV-1) vector. METHODS: Adult Wistar rats were anesthetized. Nonmodified MSCs, FGF-2-modified MSCs with HSV-1 1764/-4/pR19/ssIL2-FGF-2, or PBS was administered intracerebrally 24 hours after transient right MCAO. All animals underwent behavioral tests for 21 days, and the infarction volume with 2-3-5-triphenylterazolium was detected 3 days and 14 days after the MCAO. Three days and 7 days after the MCAO, the FGF-2 production in the ipsilateral hemisphere of the MCAO was measured with ELISA. Seven and 14 days after the MCAO, immunohistochemical staining for FGF-2 was applied. RESULTS: The stroke animals receiving FGF-2-modified MSCs demonstrated significant functional recovery compared with the other groups. Fourteen days after the MCAO, there was a significant reduction in infarction volume only in FGF-2-modified MSC-treated group. FGF-2 production in the FGF-2-modified MSC-treated brain was significantly higher compared with the other groups at 3 and 7 days after MCAO. Administrated FGF-2-modified MSCs strongly expressed the FGF-2 protein, which was proven by ELISA. CONCLUSIONS: Our data suggest that the FGF-2 gene-modified MSCs with the HSV-1 vector can contribute to remarkable functional recovery after stroke compared with MSCs transplantation alone.


Subject(s)
Bone Marrow Transplantation , Fibroblast Growth Factor 2/metabolism , Genetic Vectors/administration & dosage , Herpesvirus 1, Human/immunology , Ischemic Attack, Transient/therapy , Stromal Cells/transplantation , Animals , Brain/metabolism , Cells, Cultured , Gene Transfer Techniques , Herpesvirus 1, Human/genetics , Immunohistochemistry , Ischemic Attack, Transient/metabolism , Rats , Rats, Wistar , Tissue Extracts/metabolism
20.
J Gen Virol ; 86(Pt 6): 1645-1657, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15914842

ABSTRACT

Herpes simplex virus type 1 (HSV-1) is able to establish latency in infected individuals. In order to characterize potential new immune-escape mechanisms, mature dendritic cells (DCs) were infected with HSV-1 and total cellular RNA was isolated from infected and mock-infected populations at different time points. RNA profiling on Affymetrix Human Genome U133A arrays demonstrated a dramatic downregulation of the migration-mediating surface molecules CCR7 and CXCR4, an observation that was further confirmed by RT-PCR and fluorescence-activated cell sorting analyses. Furthermore, migration assays revealed that, upon infection of mature DCs, CCR7- and CXCR4-mediated migration towards the corresponding CCL19 and CXCL12 chemokine gradients was strongly reduced. It is noteworthy that the infection of immature DCs with HSV-1 prior to maturation led to a failure of CCR7 and CXCR4 upregulation during DC maturation and, as a consequence, also induced a block in their migratory capacity. Additional migration assays with a Deltavhs mutant virus lacking the virion host shutoff (vhs) gene, which is known to degrade cellular mRNAs, suggested a vhs-independent mechanism. These results indicate that HSV-1-infected mature DCs are limited in their capacity to migrate to secondary lymphoid organs, the areas of antigen presentation and T-cell stimulation, thus inhibiting an antiviral immune response. This represents a novel, previously unrecognized mechanism for HSV-1 to escape the human immune system.


Subject(s)
Dendritic Cells/immunology , Herpesvirus 1, Human/immunology , Cell Differentiation , Cell Movement , Dendritic Cells/cytology , Down-Regulation , Herpes Simplex/immunology , Humans , Mutation , RNA, Messenger/analysis , Receptors, CCR7 , Receptors, CXCR4/analysis , Receptors, CXCR4/genetics , Receptors, Chemokine/analysis , Receptors, Chemokine/genetics , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleases , Time Factors , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...