Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
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.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
Curr Pharm Biotechnol ; 13(9): 1834-41, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21740364

ABSTRACT

There has been interest in using viruses to treat cancer for over a century. Recent clinical efforts, driven on by significant preclinical advances, have focussed on the safety of using replication-competent viruses. Recently published clinical trials of six oncolytic viruses (adenovirus, reovirus, measles, herpes simplex, Newcastle disease virus and vaccinia) have added to the accumulating data that endorse oncolytic viruses as a safe and well tolerated treatment approach. Conclusive evidence of efficacy remains to be demonstrated, but randomised clinical trials are now underway.


Subject(s)
Neoplasms/therapy , Neoplasms/virology , Oncolytic Virotherapy/methods , Oncolytic Viruses/physiology , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans , Virus Replication
15.
Br J Cancer ; 106(1): 92-8, 2012 Jan 03.
Article in English | MEDLINE | ID: mdl-22134504

ABSTRACT

BACKGROUND: Tumour cell lysates are an excellent source of many defined and undefined tumour antigens and have been used clinically in immunotherapeutic regimes but with limited success. METHODS: We conjugated Mel888 melanoma lysates to rabbit haemorrhagic disease virus virus-like particles (VLP), which can act as vehicles to deliver multiple tumour epitopes to dendritic cells (DC) to effectively activate antitumour responses. RESULTS: Virus-like particles did not stimulate the phenotypic maturation of DC although, the conjugation of lysates to VLP (VLP-lysate) did overcome lysate-induced suppression of DC activation. Lysate-conjugated VLP enhanced delivery of antigenic proteins to DC, while the co-delivery of VLP-lysates with OK432 resulted in cross-priming of naïve T cells, with expansion of a MART1(+) population of CD8(+) T cells and generation of a specific cytotoxic response against Mel888 tumour cell targets. The responses generated with VLP-lysate and OK432 were superior to those stimulated by unconjugated lysate with OK432. CONCLUSION: Collectively, these results show that the combination of VLP-lysate with OK432 delivered to DC overcomes the suppressive effects of lysates, and enables priming of naïve T cells with superior ability to specifically kill their target tumour cells.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Virion/immunology , Blotting, Western , CD8-Positive T-Lymphocytes/immunology , Electrophoresis, Polyacrylamide Gel , Humans , Subcellular Fractions
16.
Br J Cancer ; 105(6): 787-95, 2011 Sep 06.
Article in English | MEDLINE | ID: mdl-21847125

ABSTRACT

BACKGROUND: Optimal cellular immunotherapy for cancer should ideally harness both the innate and adaptive arms of the immune response. Lymphokine-activated killer cells (LAKs) can trigger early innate killing of tumour targets, whereas long-term adaptive-specific tumour control requires priming of CD8+ cytotoxic lymphocytes (CTLs) following acquisition of tumour-associated antigens (TAAs) by antigen-presenting cells such as dendritic cells (DCs). As DCs stimulate both innate and adaptive effectors, combination cell therapy using LAKs and DCs has the potential to maximise anti-tumour immune priming. METHODS: Reciprocal activation between human clinical grade LAKs and DCs on co-culture, and its immune consequences, was monitored by cell phenotype, cytokine release and priming of both innate and adaptive cytotoxicity against melanoma targets. RESULTS: Co-culture of DCs and LAKs led to phenotypic activation of natural killer (NK) cells within the LAK population, which was associated with increased production of inflammatory cytokines and enhanced innate cytotoxicity against tumour cell targets. The LAKs reciprocally matured DCs, and the combination of LAKs and DCs, on addition of melanoma cells, supported priming of specific anti-tumour CTLs better than DCs alone. CONCLUSION: Clinical-grade LAKs/DCs represents a practical, effective combination cell immunotherapy for stimulation of both innate and adaptive anti-tumour immunity in cancer patients.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Dendritic Cells/immunology , Immunotherapy/methods , Killer Cells, Lymphokine-Activated/immunology , Melanoma/therapy , Adaptive Immunity , Cell Line, Tumor , Coculture Techniques , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Humans , Immunity, Innate
17.
Expert Opin Biol Ther ; 10(3): 369-79, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20132058

ABSTRACT

IMPORTANCE OF THE FIELD: Dendritic cells (DC) are a clear choice for use in cancer immunotherapy, and much research has focused on generating DC for clinical use. Although DC therapy has been successful in inducing specific anti-tumour immune responses, these have rarely translated into clinical efficacy. AREAS COVERED IN THIS REVIEW: We examine some of the components of generating DC for therapy, including their culture, antigen loading and delivery, and discuss why DC therapy has not yet delivered substantial clinical benefit. We also examine more novel approaches, such as the potential for combination DC-based immunomodulatory strategies. WHAT THE READER WILL GAIN: Given the highly immunosuppressive tumour environment, many of the approaches to DC vaccination are unlikely to result in effective therapy, as even successfully primed T cells may fail to infiltrate tumours or be anergized after entry. Broader approaches against multiple tumour-associated antigens in the context of overcoming tumour immune suppression are likely to prove more successful. The combination of oncolytic viral therapy with DC vaccines may promote an inflammatory tumour environment, inducing optimal DC activation, T cell priming and effective therapy. TAKE HOME MESSAGE: Evolving DC-based therapeutic strategies addressing multiple components of tumour-immune system interactions may yield substantial benefits for patients.


Subject(s)
Cell Transplantation , Dendritic Cells/cytology , Neoplasms/therapy , Dendritic Cells/immunology , Humans , Neoplasms/immunology
18.
Gene Ther ; 16(5): 689-99, 2009 May.
Article in English | MEDLINE | ID: mdl-19282847

ABSTRACT

Reovirus is a naturally occurring oncolytic virus currently in early clinical trials. However, the rapid induction of neutralizing antibodies represents a major obstacle to successful systemic delivery. This study addresses, for the first time, the ability of cellular carriers in the form of T cells and dendritic cells (DC) to protect reovirus from systemic neutralization. In addition, the ability of these cellular carriers to manipulate the subsequent balance of anti-viral versus anti-tumour immune response is explored. Reovirus, either neat or loaded onto DC or T cells, was delivered intravenously into reovirus-naive or reovirus-immune C57Bl/6 mice bearing lymph node B16tk melanoma metastases. Three and 10 days after treatment, reovirus delivery, carrier cell trafficking, metastatic clearance and priming of anti-tumour/anti-viral immunity were assessed. In naive mice, reovirus delivered either neat or through cell carriage was detectable in the tumour-draining lymph nodes 3 days after treatment, though complete clearance of metastases was only obtained when the virus was delivered on T cells or mature DC (mDC); neat reovirus or loaded immature DC (iDC) gave only partial early tumour clearance. Furthermore, only T cells carrying reovirus generated anti-tumour immune responses and long-term tumour clearance; reovirus-loaded DC, in contrast, generated only an anti-viral immune response. In reovirus-immune mice, however, the results were different. Neat reovirus was completely ineffective as a therapy, whereas mDC--though not iDC--as well as T cells, effectively delivered reovirus to melanoma in vivo for therapy and anti-tumour immune priming. Moreover, mDC were more effective than T cells over a range of viral loads. These data show that systemically administered neat reovirus is not optimal for therapy, and that DC may be an appropriate vehicle for carriage of significant levels of reovirus to tumours. The pre-existing immune status against the virus is critical in determining the balance between anti-viral and anti-tumour immunity elicited when reovirus is delivered by cell carriage, and the viral dose and mode of delivery, as well as the immune status of patients, may profoundly affect the success of any clinical anti-tumour viral therapy. These findings are therefore of direct translational relevance for the future design of clinical trials.


Subject(s)
Dendritic Cells/transplantation , Melanoma, Experimental/secondary , Melanoma, Experimental/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , T-Lymphocytes/transplantation , Adaptive Immunity , Animals , Cell Death , Cytotoxicity, Immunologic , Lymph Nodes/virology , Lymphatic Metastasis , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Reoviridae/immunology , Reoviridae/isolation & purification , Treatment Outcome , Tumor Cells, Cultured , Viral Load
19.
Gene Ther ; 16(1): 78-92, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18784747

ABSTRACT

Transfer of healthy autologous tissue as a microvascular free flap facilitates reconstruction during ablative cancer surgery. In addition to filling surgical defects, free flaps might concentrate viral vectors at the tumour bed and mediate local therapeutic effects. We evaluated the magnitude, topography and duration of luciferase gene expression after plasmid and adenoviral delivery in rat superficial inferior epigastric (SIE) flaps. For plasmid delivery, luciferase expression was significantly increased by all transduction routes (topical, intraflap injection, intravascular) (P<0.01) at day 1, but not at day 7. The spread of luciferase expression was significantly different between the 4 groups at 1 day (P=0.026) and was greatest for flaps transduced by intravascular injection. For adenoviral transduction, total radiance was significantly different between the transduced groups at 1, 14 and 28 days (P<0.05 for all comparisons). The highest levels of radiance were seen in the intravascular group. There was a statistically significant difference in the spread of light emission between the 3 groups at 1 (P=0.009) and 14 (P=0.013) days, but this was no longer evident at 28 days. Intravascular adenoviral delivery yields high-level, diffuse and durable gene expression in rat SIE flaps and is suitable for examination in therapeutic models.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Plasmids/pharmacology , Surgical Flaps , Animals , Gene Expression , Genetic Vectors/genetics , Genetic Vectors/metabolism , Injections , Lac Operon , Luciferases/analysis , Luciferases/genetics , Male , Models, Animal , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution , Transduction, Genetic/methods
20.
Gene Ther ; 15(18): 1257-70, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18401435

ABSTRACT

Reovirus is a promising unmodified double-stranded RNA (dsRNA) anti-cancer oncolytic virus, which is thought to specifically target cells with activated Ras. Although reovirus has been tested in a wide range of preclinical models and has entered early clinical trials, it has not previously been tested for the treatment of human melanoma. Here, we show that reovirus effectively kills and replicates in both human melanoma cell lines and freshly resected tumour; intratumoural injection also causes regression of melanoma in a xenograft in vivo model. Reovirus-induced melanoma death is blocked by caspase inhibition and is dependent on constituents of the Ras/RalGEF/p38 pathway. Reovirus melanoma killing is more potent than, and distinct from, chemotherapy or radiotherapy-induced cell death; a range of inflammatory cytokines and chemokines are released by infected tumour cells, while IL-10 secretion is abrogated. Furthermore, the inflammatory response generated by reovirus-infected tumour cells causes bystander toxicity against reovirus-resistant tumour cells and activates human myeloid dendritic cells (DC) in vitro. Hence, reovirus is suitable for clinical testing in melanoma, and may provide a useful danger signal to reverse the immunologically suppressive environment characteristic of this tumour.


Subject(s)
Melanoma/therapy , Oncolytic Virotherapy/methods , Reoviridae/physiology , Skin Neoplasms/therapy , Animals , Apoptosis , Cell Line, Tumor , Cell Survival , Chromones/pharmacology , Cytokines/immunology , Cytotoxicity Tests, Immunologic , Dendritic Cells/immunology , Flow Cytometry , Humans , Imidazoles/pharmacology , Melanoma/immunology , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Pyridines/pharmacology , Signal Transduction/physiology , Skin Neoplasms/immunology , Tumor Cells, Cultured , Virus Replication , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...