Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 202
Filter
1.
Oncoimmunology ; 11(1): 2066050, 2022.
Article in English | MEDLINE | ID: mdl-35558159

ABSTRACT

Systemic relapse after radiotherapy and surgery is the major cause of disease-related mortality in sarcoma patients. Combining radiotherapy and immunotherapy is under investigation as a means to improve response rates. However, the immune contexture of sarcoma is understudied. Here, we use a retrospective cohort of sarcoma patients, treated with neoadjuvant radiotherapy, and TCGA data. We explore therapeutic targets of relevance to sarcoma, using genomics and multispectral immunohistochemistry to provide insights into the tumor immune microenvironment across sarcoma subtypes. Differential gene expression between radioresponsive myxoid liposarcoma (MLPS) and more radioresistant undifferentiated pleomorphic sarcoma (UPS) indicated UPS contained higher transcript levels of a number of immunotherapy targets (CD73/NT5E, CD39/ENTPD1, CD25/IL2RA, and 4-1BB/TNFRSF9). We focused on 4-1BB/TNFRSF9 and other costimulatory molecules. In TCGA data, 4-1BB correlated to an inflamed and exhausted phenotype. OX40/TNFRSF4 and 4-1BB/TNFRSF9 were highly expressed in sarcoma subtypes versus other cancers. Despite OX40 and 4-1BB being described as Treg markers, we identified that they delineate distinct tumor immune profiles. This was true for sarcoma and other cancers. While only a limited number of samples could be analyzed, spatial analysis of OX40 expression identified two diverse phenotypes of OX40+ Tregs, one associated with and one independent of tertiary lymphoid structures (TLSs). Patient stratification is of intense interest for immunotherapies. We provide data supporting the viewpoint that a cohort of sarcoma patients, appropriately selected, are promising candidates for immunotherapies. Spatial profiling of OX40+ Tregs, in relation to TLSs, could be an additional metric to improve future patient stratification.


Subject(s)
Sarcoma , Soft Tissue Neoplasms , Adult , Humans , Neoplasm Recurrence, Local , Retrospective Studies , Sarcoma/genetics , Sarcoma/therapy , T-Lymphocytes, Regulatory , Tumor Microenvironment
3.
J Environ Manage ; 307: 114503, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35078064

ABSTRACT

Conserving aquatic resources in the West African Sahel requires water management tools to assess the ecological status of surface water bodies threatened by mounting pressures from agricultural intensification and urbanization. Macroinvertebrate communities of Sahelian rivers were examined to test if a multi-metric index approach could be developed to assess the ecological quality of rivers. A total of 40 sample sites falling within a continuum ranging from "unimpaired reference sites" to "impaired sites" were assessed during this study. Macroinvertebrates were sampled with a hand net following a multi-habitat sampling approach. Key environmental parameters, both physico-chemical and hydro-morphologic, were recorded. More than 20 candidate metrics were evaluated in four categories: composition, functional feeding, diversity, and tolerance. We used detailed analysis procedures to exclude unsuitable metrics from the data set. After excluding redundant metrics, six-core metrics were selected to compose the Sahel River Multimetric Index (SRMI): Total-taxa, Shannon & Weiner index, EPT-taxa, ASPT-NEPBIOS and ASPT-SASS and Collector-filterers. The final index derived from these metrics was divided into five ecological quality classes (high, good, moderate, poor, and bad). The results showed that the SRMI responded to a set of environmental parameters associated with a gradient of human pressures affecting the ecological integrity of water bodies (R2≥|0.50|; p < 0.05; p < 0.001). This work produced a data base and analysis that confirms the usefulness of an unprecedented and promising tool for biological monitoring and decision-making in Sahelian regions' water management.


Subject(s)
Environmental Monitoring , Rivers , Animals , Burkina Faso , Ecosystem , Humans , Invertebrates
5.
Clin Exp Immunol ; 200(1): 33-44, 2020 04.
Article in English | MEDLINE | ID: mdl-31784984

ABSTRACT

Glioblastoma (GBM) is an aggressive cancer with a very poor prognosis. Generally viewed as weakly immunogenic, GBM responds poorly to current immunotherapies. To understand this problem more clearly we used a combination of natural killer (NK) cell functional assays together with gene and protein expression profiling to define the NK cell response to GBM and explore immunosuppression in the GBM microenvironment. In addition, we used transcriptome data from patient cohorts to classify GBM according to immunological profiles. We show that glioma stem-like cells, a source of post-treatment tumour recurrence, express multiple immunomodulatory cell surface molecules and are targeted in preference to normal neural progenitor cells by natural killer (NK) cells ex vivo. In contrast, GBM-infiltrating NK cells express reduced levels of activation receptors within the tumour microenvironment, with hallmarks of transforming growth factor (TGF)-ß-mediated inhibition. This NK cell inhibition is accompanied by expression of multiple immune checkpoint molecules on T cells. Single-cell transcriptomics demonstrated that both tumour and haematopoietic-derived cells in GBM express multiple, diverse mediators of immune evasion. Despite this, immunome analysis across a patient cohort identifies a spectrum of immunological activity in GBM, with active immunity marked by co-expression of immune effector molecules and feedback inhibitory mechanisms. Our data show that GBM is recognized by the immune system but that anti-tumour immunity is restrained by multiple immunosuppressive pathways, some of which operate in the healthy brain. The presence of immune activity in a subset of patients suggests that these patients will more probably benefit from combination immunotherapies directed against multiple immunosuppressive pathways.


Subject(s)
Brain Neoplasms/immunology , Gene Expression Profiling/methods , Glioblastoma/immunology , Immune Tolerance/immunology , Killer Cells, Natural/immunology , Neoplastic Stem Cells/immunology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Cohort Studies , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , Gene Expression Regulation, Neoplastic/immunology , Gene Regulatory Networks/immunology , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Immune Tolerance/genetics , Killer Cells, Natural/metabolism , Neoplastic Stem Cells/metabolism , Phenotype , Prognosis , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
8.
Environ Monit Assess ; 190(1): 2, 2017 Dec 02.
Article in English | MEDLINE | ID: mdl-29209819

ABSTRACT

Awareness of sustainable management of water and its biological resources is rising in West Africa, but application of effective tools for biomonitoring and detecting habitats at risk in aquatic ecosystems is limited. In this study, we provide key environmental descriptors to characterize reference sites by applying the following "a priori criteria" (physical and chemical, hydro-morphological, and land use parameters) by exploring their potential to determine suitable reference sites. Using data collected from 44 sites, we identified 37 criteria that reliably identify reference conditions in semi-arid rivers by reflecting the impacts of multiple pressures ranging from low to very high intensity of human uses and impairments. We integrated all these impacts in an overall pressures index, which showed that protected areas can reasonably be considered as credible reference sites as far as they show low overall impact levels from cumulative pressures. We recommend that development of bio-indicator standards should be based on the collection and integration of all the available information, especially quantitative, spatially-explicit data, from benthic macroinvertebrates and fish. Rigorous standardization of bio-indicator protocols will make them more easily applicable for management and conservation of aquatic ecosystem resources in semi-arid zones of Africa.


Subject(s)
Ecosystem , Environmental Monitoring/methods , Rivers/chemistry , Africa, Western , Animals , Ecology , Fishes , Humans
9.
Gene Ther ; 24(1): 21-30, 2017 01.
Article in English | MEDLINE | ID: mdl-27779616

ABSTRACT

The anti-tumour effects associated with oncolytic virus therapy are mediated significantly through immune-mediated mechanisms, which depend both on the type of virus and the route of delivery. Here, we show that intra-tumoral oncolysis by Reovirus induced the priming of a CD8+, Th1-type anti-tumour response. By contrast, systemically delivered Vesicular Stomatitis Virus expressing a cDNA library of melanoma antigens (VSV-ASMEL) promoted a potent anti-tumour CD4+ Th17 response. Therefore, we hypothesised that combining the Reovirus-induced CD8+ T cell response, with the VSV-ASMEL CD4+ Th17 helper response, would produce enhanced anti-tumour activity. Consistent with this, priming with intra-tumoral Reovirus, followed by an intra-venous VSV-ASMEL Th17 boost, significantly improved survival of mice bearing established subcutaneous B16 melanoma tumours. We also show that combination of either therapy alone with anti-PD-1 immune checkpoint blockade augmented both the Th1 response induced by systemically delivered Reovirus in combination with GM-CSF, and also the Th17 response induced by VSV-ASMEL. Significantly, anti-PD-1 also uncovered an anti-tumour Th1 response following VSV-ASMEL treatment that was not seen in the absence of checkpoint blockade. Finally, the combination of all three treatments (priming with systemically delivered Reovirus, followed by double boosting with systemic VSV-ASMEL and anti-PD-1) significantly enhanced survival, with long-term cures, compared to any individual, or double, combination therapies, associated with strong Th1 and Th17 responses to tumour antigens. Our data show that it is possible to generate fully systemic, highly effective anti-tumour immunovirotherapy by combining oncolytic viruses, along with immune checkpoint blockade, to induce complementary mechanisms of anti-tumour immune responses.


Subject(s)
Cell Cycle Checkpoints , Immunotherapy/methods , Melanoma/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cell Line, Tumor , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Melanoma-Specific Antigens/genetics , Melanoma-Specific Antigens/immunology , Mice , Oncolytic Viruses/genetics , Reoviridae/genetics , Reoviridae/immunology , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/virology , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/virology , Vesiculovirus/genetics , Vesiculovirus/immunology
10.
Gene Ther ; 23(4): 357-68, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26814609

ABSTRACT

Oncolytic strains of vaccinia virus are currently in clinical development with clear evidence of safety and promising signs of efficacy. Addition of therapeutic genes to the viral genome may increase the therapeutic efficacy of vaccinia. We evaluated the therapeutic potential of vaccinia virus expressing the sodium iodide symporter (NIS) in prostate cancer models, combining oncolysis, external beam radiotherapy and NIS-mediated radioiodide therapy. The NIS-expressing vaccinia virus (VV-NIS), GLV-1h153, was tested in in vitro analyzes of viral cell killing, combination with radiotherapy, NIS expression, cellular radioiodide uptake and apoptotic cell death in PC3, DU145, LNCaP and WPMY-1 human prostate cell lines. In vivo experiments were carried out in PC3 xenografts in CD1 nude mice to assess NIS expression and tumor radioiodide uptake. In addition, the therapeutic benefit of radioiodide treatment in combination with viral oncolysis and external beam radiotherapy was measured. In vitro viral cell killing of prostate cancers was dose- and time-dependent and was through apoptotic mechanisms. Importantly, combined virus therapy and iodizing radiation did not adversely affect oncolysis. NIS gene expression in infected cells was functional and mediated uptake of radioiodide both in vitro and in vivo. Therapy experiments with both xenograft and immunocompetent Transgenic Adenocarcinoma of the Mouse Prostate (TRAMP) mouse models showed that the addition of radioiodide to VV-NIS-infected tumors was more effective than each single-agent therapy, restricting tumor growth and increasing survival. In conclusion, VV-NIS is effective in prostate cancer models. This treatment modality would be an attractive complement to existing clinical radiotherapy practice.


Subject(s)
Genetic Therapy/methods , Oncolytic Virotherapy/methods , Prostatic Neoplasms/therapy , Symporters/genetics , Animals , Cell Line, Tumor , Humans , Male , Mice , Mice, Nude , Oncolytic Viruses/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/virology , Random Allocation , Symporters/metabolism , Transfection , Vaccinia virus/genetics , Xenograft Model Antitumor Assays
11.
Leukemia ; 29(9): 1799-810, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25814029

ABSTRACT

The naturally occurring oncolytic virus (OV), reovirus, replicates in cancer cells causing direct cytotoxicity, and can activate innate and adaptive immune responses to facilitate tumour clearance. Reovirus is safe, well tolerated and currently in clinical testing for the treatment of multiple myeloma, in combination with dexamethasone/carfilzomib. Activation of natural killer (NK) cells has been observed after systemic delivery of reovirus to cancer patients; however, the ability of OV to potentiate NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC) is unexplored. This study elucidates the potential of oncolytic reovirus for the treatment of chronic lymphocytic leukaemia (CLL), both as a direct cytotoxic agent and as an immunomodulator. We demonstrate that reovirus: (i) is directly cytotoxic against CLL, which requires replication-competent virus; (ii) phenotypically and functionally activates patient NK cells via a monocyte-derived interferon-α (IFNα)-dependent mechanism; and (iii) enhances ADCC-mediated killing of CLL in combination with anti-CD20 antibodies. Our data provide strong preclinical evidence to support the use of reovirus in combination with anti-CD20 immunotherapy for the treatment of CLL.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Mammalian orthoreovirus 3/immunology , Oncolytic Viruses/immunology , Rituximab/immunology , Rituximab/therapeutic use , Aged , Aged, 80 and over , Antineoplastic Agents/immunology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cytopathogenic Effect, Viral , Female , Humans , Immunity, Innate , Immunologic Factors/immunology , Immunologic Factors/therapeutic use , Immunophenotyping , Immunotherapy , Killer Cells, Natural/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Lymphocyte Activation/immunology , Male , Middle Aged , Neoplasm Staging , Virus Replication
12.
Br J Cancer ; 112(4): 693-703, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25628092

ABSTRACT

BACKGROUND: Paediatric high grade glioma (pHGG) and diffuse intrinsic pontine glioma (DIPG) are highly aggressive brain tumours. Their invasive phenotype contributes to their limited therapeutic response, and novel treatments that block brain tumour invasion are needed. METHODS: Here, we examine the migratory characteristics and treatment effect of small molecule glycogen synthase kinase-3 inhibitors, lithium chloride (LiCl) and the indirubin derivative 6-bromoindirubin-oxime (BIO), previously shown to inhibit the migration of adult glioma cells, on two pHGG cell lines (SF188 and KNS42) and one patient-derived DIPG line (HSJD-DIPG-007) using 2D (transwell membrane, immunofluorescence, live cell imaging) and 3D (migration on nanofibre plates and spheroid invasion in collagen) assays. RESULTS: All lines were migratory, but there were differences in morphology and migration rates. Both LiCl and BIO reduced migration and instigated cytoskeletal rearrangement of stress fibres and focal adhesions when viewed by immunofluorescence. In the presence of drugs, loss of polarity and differences in cellular movement were observed by live cell imaging. CONCLUSIONS: Ours is the first study to demonstrate that it is possible to pharmacologically target migration of paediatric glioma in vitro using LiCl and BIO, and we conclude that these agents and their derivatives warrant further preclinical investigation as potential anti-migratory therapeutics for these devastating tumours.


Subject(s)
Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Movement , Glioma/pathology , Glioma/therapy , Molecular Targeted Therapy , Cell Line, Tumor , Cell Movement/drug effects , Child , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Glycogen Synthase Kinase 3/antagonists & inhibitors , Humans , Indoles/pharmacology , Lithium Chloride/pharmacology , Neoplasm Invasiveness , Oximes/pharmacology , Protein Kinase Inhibitors/pharmacology , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Spheroids, Cellular/physiology
13.
Clin Exp Immunol ; 180(1): 98-107, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25469725

ABSTRACT

Human natural killer (NK) cells play an important role in anti-viral immunity. However, studying their activation kinetics during infection is highly problematic. A clinical trial of a therapeutic virus provided an opportunity to study human NK cell activation in vivo in a controlled manner. Ten colorectal cancer patients with liver metastases received between one and five doses of oncolytic reovirus prior to surgical resection of their tumour. NK cell surface expression of the interferon-inducible molecules CD69 and tetherin peaked 24-48 h post-infection, coincident with a peak of interferon-induced gene expression. The interferon response and NK cell activation were transient, declining by 96 h post-infection. Furthermore, neither NK cell activation nor the interferon response were sustained in patients undergoing multiple rounds of virus treatment. These results show that reovirus modulates human NK cell activity in vivo and suggest that this may contribute to any therapeutic effect of this oncolytic virus. Detection of a single, transient peak of activation, despite multiple treatment rounds, has implications for the design of reovirus-based therapy. Furthermore, our results suggest the existence of a post-infection refractory period when the interferon response and NK cell activation are blunted. This refractory period has been observed previously in animal models and may underlie the enhanced susceptibility to secondary infections that is seen following viral infection.


Subject(s)
Immunity, Cellular , Killer Cells, Natural/immunology , Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses/immunology , Reoviridae/immunology , Aged , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Female , Humans , Interferons/immunology , Killer Cells, Natural/pathology , Lectins, C-Type/immunology , Male , Middle Aged , Neoplasms/immunology , Neoplasms/therapy
14.
Surgeon ; 12(4): 210-20, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24502935

ABSTRACT

BACKGROUND: Despite mankind's many achievements, we are yet to find a cure for cancer. We are now approaching a new era which recognises the promise of harnessing the immune system for anti-cancer therapy. Pathogens have been implicated for decades as potential anti-cancer agents, but implementation into clinical therapy has been plagued with significant drawbacks. Newer 'designer' agents have addressed some of these concerns, in particular, a new breed of oncolytic virus: JX-594, a genetically engineered pox virus, is showing promise. OBJECTIVE: To review the current literature on the use of oncolytic viruses in the treatment of cancer; both by direct oncolysis and stimulation of the immune system. The review will provide a background and historical progression for the surgeon on tumour immunology, and the interplay between oncolytic viruses, immune cells, inflammation on tumourigenesis. METHODS: A literature review was performed using the Medline database. CONCLUSIONS: Viral therapeutics hold promise as a novel treatment modality for the treatment of disseminated malignancy. It provides a multi-pronged attack against tumour burden; direct tumour cell lysis, exposure of tumour-associated antigens (TAA), induction of immune danger signals, and recognition by immune effector cells.


Subject(s)
Cancer Vaccines/therapeutic use , Immunity, Cellular , Neoplasms/therapy , Oncolytic Viruses/immunology , Vaccination/methods , Humans , Neoplasms/immunology
15.
Oncogene ; 33(13): 1700-12, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-23624923

ABSTRACT

Melanoma is an aggressive skin cancer that carries an extremely poor prognosis when local invasion, nodal spread or systemic metastasis has occurred. Recent advances in melanoma biology have revealed that RAS-RAF-MEK-ERK signaling has a pivotal role in governing disease progression and treatment resistance. Proof-of-concept clinical studies have shown that direct BRAF inhibition yields impressive responses in advanced disease but these are short-lived as treatment resistance rapidly emerges. Therefore, there is a pressing need to develop new targeted strategies for BRAF mutant melanoma. As such, oncolytic viruses represent a promising cancer-specific approach with significant activity in melanoma. This study investigated interactions between genetically-modified vaccinia virus (GLV-1h68) and radiotherapy in melanoma cell lines with BRAF mutant, Ras mutant or wild-type genotype. Preclinical studies revealed that GLV-1h68 combined with radiotherapy significantly increased cytotoxicity and apoptosis relative to either single agent in (V600D)BRAF/(V600E)BRAF mutant melanoma in vitro and in vivo. The mechanism of enhanced cytotoxicity with GLV-1h68/radiation (RT) was independent of viral replication and due to attenuation of JNK, p38 and ERK MAPK phosphorylation specifically in BRAF mutant cells. Further studies showed that JNK pathway inhibition sensitized BRAF mutant cells to GLV-1h68-mediated cell death, mimicking the effect of RT. GLV-1h68 infection activated MAPK signaling in (V600D)BRAF/(V600E)BRAF mutant cell lines and this was associated with TNF-α secretion which, in turn, provided a prosurvival signal. Combination GLV-1h68/RT (or GLV-1h68/JNK inhibition) caused abrogation of TNF-α secretion. These data provide a strong rationale for combining GLV-1h68 with irradiation in (V600D/E)BRAF mutant tumors.


Subject(s)
JNK Mitogen-Activated Protein Kinases/genetics , Melanoma/therapy , Oncolytic Virotherapy/methods , Proto-Oncogene Proteins B-raf/genetics , Tumor Necrosis Factor-alpha/metabolism , Vaccinia virus/physiology , Animals , Cell Death , Cell Line, Tumor , Female , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Melanoma/genetics , Melanoma/metabolism , Melanoma/virology , Mice , Mice, Inbred BALB C , Mice, Nude , Random Allocation , Tumor Necrosis Factor-alpha/genetics , Xenograft Model Antitumor Assays
16.
Int J Cancer ; 134(5): 1091-101, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-23982804

ABSTRACT

Reovirus is an oncolytic virus (OV), which acts by both direct tumor cell killing and priming of antitumor immunity. A major obstacle for effective oncolytic virotherapy is effective delivery of OV to tumor cells. Ovarian cancer is often confined to the peritoneal cavity and therefore i.p. delivery of reovirus may provide the ideal locoregional delivery, avoiding systemic dissemination. However, ovarian cancer is associated with an accumulation of ascitic fluid, which may interfere with oncolytic viral therapy. Here, we investigated the effect of ascites on reovirus-induced oncolysis against primary ovarian cancer cells and ovarian cancer cell lines. In the absence of ascites, reovirus was cytotoxic against ovarian cancer cells; however, cytotoxicity was abrogated in the presence of ascitic fluid. Neutralizing antibodies (NAb) were identified as the cause of this inhibition. Loading OV onto cell carriers may facilitate virus delivery in the presence of NAb and immune cells which have their own antitumor effector activity are particularly appealing. Immature dendritic cells (iDC), Lymphokine-activated killer (LAK) cells and LAKDC cocultures were tested as potential carriers for reovirus for tumor cell killing and immune cell priming. Reovirus-loaded LAKDC, and to a lesser degree iDC, were able to: (i) protect from NAb and hand-off reovirus for tumor cell killing; (ii) induce a proinflammatory cytokine milieu (IFNÉ£, IL-12, IFNα and TNFα) and (iii) generate an innate and specific antitumor adaptive immune response. Hence, LAKDC pulsed with reovirus represent a novel, clinically practical treatment for ovarian cancer to maximise both direct and innate/adaptive immune-mediated tumor cell killing.


Subject(s)
Antibodies, Neutralizing/immunology , Ascites/immunology , Dendritic Cells/immunology , Killer Cells, Lymphokine-Activated/immunology , Oncolytic Virotherapy , Ovarian Neoplasms/therapy , Reoviridae/immunology , Apoptosis , Cytokines/biosynthesis , Female , Humans , Ovarian Neoplasms/immunology , Tumor Cells, Cultured
17.
Oral Oncol ; 49(2): 108-18, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22925693

ABSTRACT

OBJECTIVE: Oncolytic forms of attenuated Vaccinia virus are now in clinical development, assessing the compatibility of this novel treatment with radiotherapy may reveal exploitable synergistic relationships. MATERIALS AND METHODS: In vitro analyses of cell killing, cell cycle effects and caspase activation were carried out on HN3, HN5, CAL27, Detroit, SIHN5B, and PJ41 cells. In vivo studies of the virus and X-radiation were performed on H&N xenografts in CD1 nude mice. RESULTS: Cell killing in vitro was demonstrated to be dose- and time-dependent. Infection causes an increase in S-phase and sub-G1 cells. A dose dependent increase in active caspase-3 indicated induction of apoptosis. Xenografts injected with Vaccinia stabilised and frequently completely regressed. Combination with radiation generated additional cell death, induction of caspase activity and in vivo further improved long term regression rates. CONCLUSIONS: These data support continued exploration of this therapy combination and indicates potential for clinical trials in head and neck cancer.


Subject(s)
Head and Neck Neoplasms/therapy , Oncolytic Virotherapy , Vaccinia virus , Caspase 3/metabolism , Caspase 7/metabolism , Cell Cycle , Cell Line, Tumor , Combined Modality Therapy , Enzyme Activation , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/radiotherapy , Humans
18.
Gene Ther ; 20(1): 7-15, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22170342

ABSTRACT

Oncolytic viruses (OV) are promising treatments for cancer, with several currently undergoing testing in randomised clinical trials. Measles virus (MV) has not yet been tested in models of human melanoma. This study demonstrates the efficacy of MV against human melanoma. It is increasingly recognised that an essential component of therapy with OV is the recruitment of host antitumour immune responses, both innate and adaptive. MV-mediated melanoma cell death is an inflammatory process, causing the release of inflammatory cytokines including type-1 interferons and the potent danger signal HMGB1. Here, using human in vitro models, we demonstrate that MV enhances innate antitumour activity, and that MV-mediated melanoma cell death is capable of stimulating a melanoma-specific adaptive immune response.


Subject(s)
Measles virus/immunology , Melanoma/immunology , Oncolytic Viruses/immunology , Cell Death/immunology , Cell Line, Tumor , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Measles virus/pathogenicity , Melanoma/pathology , Melanoma/virology , Oncolytic Viruses/pathogenicity , Up-Regulation
19.
Gene Ther ; 20(5): 521-8, 2013 May.
Article in English | MEDLINE | ID: mdl-22895509

ABSTRACT

Oncolytic reovirus is currently under active investigation in a range of tumour types. Early phase studies have shown that this agent has modest monotherapy efficacy and its future development is likely to focus on combination regimens with cytotoxic chemotherapy. Indeed, phase I/II clinical trials have confirmed that reovirus can be safely combined with cytotoxic drugs, including a platin-taxane doublet regimen, which is currently being tested in a phase III clinical trial in patients with relapsed/metastatic head and neck cancer. Therefore, we have tested this triple (reovirus, cisplatin, paclitaxel) combination therapy in a panel of four head and neck cancer cell lines. Using the combination index (CI) method, the triple therapy demonstrated synergistic cytotoxicity in vitro in both malignant and non-malignant cell lines. In head and neck cancer cell lines, this was associated with enhanced caspase 3 and 7 cleavage, but no increase in viral replication. In vitro analyses confirmed colocalisation of markers of reovirus infection and caspase 3. Triple therapy was significantly more effective than reovirus or cisplatin-paclitaxel in athymic nude mice. These data suggest that the combination of reovirus plus platin-taxane doublet chemotherapy has significant activity in head and neck cancer and underpin the current phase III study in this indication.


Subject(s)
Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Animals , Antineoplastic Agents , Cell Line, Tumor , Cisplatin/administration & dosage , Clinical Trials, Phase I as Topic , Combined Modality Therapy , Head and Neck Neoplasms/virology , Humans , Mice , Orthoreovirus/genetics , Paclitaxel/administration & dosage
20.
Br J Cancer ; 106(3): 496-507, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22240799

ABSTRACT

BACKGROUND: There are still no effective treatments for superficial bladder cancer (SBC)/non-muscle invasive bladder cancer. Following treatment, 20% of patients still develop metastatic disease. Superficial bladder cancer is often multifocal, has high recurrences after surgical resection and recurs after intravesical live Bacillus Calmette-Guérin. Oncovex(GALV/CD), an oncolytic herpes simplex virus-1, has shown enhanced local tumour control by combining oncolysis with the expression of a highly potent pro-drug activating gene and the fusogenic glycoprotein. METHODS: In vitro fusion/prodrug/apoptotic cell-based assays. In vivo orthotopic bladder tumour model, visualised by computed microtomography. RESULTS: Treatment of seven human bladder carcinoma cell lines with the virus resulted in tumour cell killing through oncolysis, pro-drug activation and glycoprotein fusion. Oncovex(GALV/CD) and mitomycin C showed a synergistic effect, whereas the co-administration with cisplatin or gemcitabine showed an antagonistic effect in vitro. Transitional cell cancer (TCC) cells follow an apoptotic cell death pathway after infection with Oncovex(GALV/CD) with or without 5-FC. In vivo results showed that intravesical treatment with Oncovex(GALV/CD) + prodrug (5-FC) reduced the average tumour volume by over 95% compared with controls. DISCUSSION: Our in vitro and in vivo results indicate that Oncovex(GALV/CD) can improve local tumour control within the bladder, and potentially alter its natural history.


Subject(s)
Carcinoma, Transitional Cell/therapy , Glycoproteins/therapeutic use , Neoplasm Recurrence, Local/therapy , Oncolytic Virotherapy , Prodrugs/therapeutic use , Urinary Bladder Neoplasms/therapy , Administration, Intravesical , Animals , Apoptosis/drug effects , Carcinoma, Transitional Cell/pathology , Cell Line, Tumor/drug effects , Cricetinae , Disease Models, Animal , Female , Fluorouracil/pharmacology , Glycoproteins/pharmacology , Herpesvirus 1, Human/genetics , Humans , Leukemia Virus, Gibbon Ape/genetics , Neoplasm Recurrence, Local/pathology , Prodrugs/administration & dosage , Prodrugs/pharmacology , Rats , Rats, Inbred F344 , Urinary Bladder Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...