Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Proc Natl Acad Sci U S A ; 115(7): E1540-E1549, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29386395

ABSTRACT

Inhibition of immune checkpoints programmed death 1 (PD-1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4) on T cells results in durable antitumor activity in melanoma patients. Despite high frequency of melanoma brain metastases (BrM) and associated poor prognosis, the activity and mechanisms of immune checkpoint inhibitors (ICI) in metastatic tumors that develop within the "immune specialized" brain microenvironment, remain elusive. We established a melanoma tumor transplantation model with intracranial plus extracranial (subcutaneous) tumor, mimicking the clinically observed coexistence of metastases inside and outside the brain. Strikingly, intracranial ICI efficacy was observed only when extracranial tumor was present. Extracranial tumor was also required for ICI-induced increase in CD8+ T cells, macrophages, and microglia in brain tumors, and for up-regulation of immune-regulatory genes. Combined PD-1/CTLA-4 blockade had a superior intracranial efficacy over the two monotherapies. Cell depletion studies revealed that NK cells and CD8+ T cells were required for intracranial anti-PD-1/anti-CTLA-4 efficacy. Rather than enhancing CD8+ T cell activation and expansion within intracranial tumors, PD-1/CTLA-4 blockade dramatically (∼14-fold) increased the trafficking of CD8+ T cells to the brain. This was mainly through the peripheral expansion of homing-competent effector CD8+ T cells and potentially further enhanced through up-regulation of T cell entry receptors intercellular adhesion molecule 1 and vascular adhesion molecule 1 on tumor vasculature. Our study indicates that extracranial activation/release of CD8+ T cells from PD-1/CTLA-4 inhibition and potentiation of their recruitment to the brain are paramount to the intracranial anti-PD-1/anti-CTLA-4 activity, suggesting augmentation of these processes as an immune therapy-enhancing strategy in metastatic brain cancer.


Subject(s)
Antibodies, Monoclonal/pharmacology , Brain Neoplasms/therapy , CD8-Positive T-Lymphocytes/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/therapy , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Brain Neoplasms/immunology , Brain Neoplasms/secondary , Female , Granzymes/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/immunology , Skin Neoplasms/immunology , Skin Neoplasms/secondary , Skin Neoplasms/therapy , Tumor Burden , Tumor Cells, Cultured
2.
J Immunol ; 183(7): 4312-21, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19734207

ABSTRACT

Oncolytic virotherapy may mediate antitumor effects via direct oncolysis or immune-mediated tumor regression. Although the ability of oncolytic viruses to generate adaptive antitumor immunity has been characterized, their interactions with the innate immune system are relatively unclear. Using a human in vitro system, this study investigates the innate immunological consequences of reovirus therapy and its potential to activate NK cell-mediated antitumor activity. Dendritic cells (DC) loaded with reovirus-infected human melanoma Mel888 cells (DC-MelReo), but not reovirus-infected tumor cells alone, induced IFN-gamma production within the NK cell population upon coculture with PBMC, in a cell-to-cell contact-dependent manner. DC-MelReo secreted the chemokines CCL2, 3, 4, 5, 7, 8, 11, and CXCL10; these culture supernatants induced NK cell chemotaxis. Coculture of DC-MelReo with purified NK cells induced reciprocal contact-dependent phenotypic DC maturation, while DC-MelReo elicited up-regulation of the activation marker CD69 on NK cells, in a partially contact and partially IL-12 dependent manner. Significantly, DC-MelReo induced NK cell cytotoxicity toward tumor cells by a type I IFN dependent mechanism. These data demonstrate that tumor infection by reovirus can act via DC to induce NK cell recruitment, activation, and cytotoxicity, along with reciprocal DC maturation. These findings suggest that reciprocal DC-NK cell interactions, following reovirus therapy, may play an important role in altering the immune milieu of the tumor microenvironment and mediating tumor regression.


Subject(s)
Cell Communication/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Melanoma, Experimental/immunology , Melanoma, Experimental/prevention & control , Reoviridae/immunology , Cell Line, Tumor , Cells, Cultured , Chemotaxis, Leukocyte/immunology , Coculture Techniques , Cytotoxicity, Immunologic , Dendritic Cells/pathology , Humans , Killer Cells, Natural/pathology , Melanoma, Experimental/virology , Oncolytic Virotherapy/methods , Reoviridae Infections/immunology , Reoviridae Infections/prevention & control , Reoviridae Infections/virology
3.
J Immunother Cancer ; 9(3)2021 03.
Article in English | MEDLINE | ID: mdl-33741729

ABSTRACT

BACKGROUND: Multiple myeloma (MM) remains an incurable disease and oncolytic viruses offer a well-tolerated addition to the therapeutic arsenal. Oncolytic reovirus has progressed to phase I clinical trials and its direct lytic potential has been extensively studied. However, to date, the role for reovirus-induced immunotherapy against MM, and the impact of the bone marrow (BM) niche, have not been reported. METHODS: This study used human peripheral blood mononuclear cells from healthy donors and in vitro co-culture of MM cells and BM stromal cells to recapitulate the resistant BM niche. Additionally, the 5TGM1-Kalw/RijHSD immunocompetent in vivo model was used to examine reovirus efficacy and characterize reovirus-induced immune responses in the BM and spleen following intravenous administration. Collectively, these in vitro and in vivo models were used to characterize the development of innate and adaptive antimyeloma immunity following reovirus treatment. RESULTS: Using the 5TGM1-Kalw/RijHSD immunocompetent in vivo model we have demonstrated that reovirus reduces both MM tumor burden and myeloma-induced bone disease. Furthermore, detailed immune characterization revealed that reovirus: (i) increased natural killer (NK) cell and CD8+ T cell numbers; (ii) activated NK cells and CD8+ T cells and (iii) upregulated effector-memory CD8+ T cells. Moreover, increased effector-memory CD8+ T cells correlated with decreased tumor burden. Next, we explored the potential for reovirus-induced immunotherapy using human co-culture models to mimic the myeloma-supportive BM niche. MM cells co-cultured with BM stromal cells displayed resistance to reovirus-induced oncolysis and bystander cytokine-killing but remained susceptible to killing by reovirus-activated NK cells and MM-specific cytotoxic T lymphocytes. CONCLUSION: These data highlight the importance of reovirus-induced immunotherapy for targeting MM cells within the BM niche and suggest that combination with agents which boost antitumor immune responses should be a priority.


Subject(s)
Bone Marrow/immunology , CD8-Positive T-Lymphocytes/immunology , Killer Cells, Natural/immunology , Multiple Myeloma/therapy , Oncolytic Virotherapy , Oncolytic Viruses/immunology , Reoviridae/immunology , Spleen/immunology , Tumor Microenvironment/immunology , Animals , Bone Marrow/virology , CD8-Positive T-Lymphocytes/virology , Cell Line, Tumor , Coculture Techniques , Cytokines/immunology , Cytotoxicity, Immunologic , Female , Humans , Killer Cells, Natural/virology , Male , Mice, Inbred C57BL , Multiple Myeloma/immunology , Multiple Myeloma/virology , Oncolytic Viruses/pathogenicity , Reoviridae/pathogenicity , Spleen/virology , Tumor Escape
4.
Methods Mol Biol ; 2058: 229-236, 2020.
Article in English | MEDLINE | ID: mdl-31486041

ABSTRACT

Oncolytic virus therapy is a rapidly expanding branch of cancer immunotherapy and represents a genuine opportunity to improve currently available treatment options. However, as single agents oncolytic viruses have shown only moderate clinical benefit and many challenges remain before their full potential is realized. Central to this is the efficient delivery of the virus to the tumor site and potentiation of the antitumor immune response. This chapter describes the loading of oncolytic reovirus onto monocytes which act as carriers for delivery of the virus to the tumor site and, as antigen presenting cells, may also thereby potentiate the development of an adaptive antitumor immune response.


Subject(s)
Gene Transfer Techniques , Genetic Vectors/genetics , Oncolytic Viruses/genetics , Reoviridae/genetics , Animals , Antibodies, Viral/immunology , Cell- and Tissue-Based Therapy , Genetic Therapy , Humans , Immunity , Immunoconjugates/administration & dosage , Immunoconjugates/immunology , Immunomodulation , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Mice , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/therapy , Oncolytic Virotherapy , Reoviridae/immunology , Transduction, Genetic
5.
Clin Cancer Res ; 14(22): 7358-66, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-19010851

ABSTRACT

PURPOSE: Early clinical trials are under way exploring the direct oncolytic potential of reovirus. This study addresses whether tumor infection by reovirus is also able to generate bystander, adaptive antitumor immunity. EXPERIMENTAL DESIGN: Reovirus was delivered intravenously to C57BL/6 mice bearing lymph node metastases from the murine melanoma, B16-tk, with assessment of nodal metastatic clearance, priming of antitumor immunity against the tumor-associated antigen tyrosinase-related protein-2, and cytokine responses. In an in vitro human system, the effect of reovirus infection on the ability of Mel888 melanoma cells to activate and load dendritic cells for cytotoxic lymphocyte (CTL) priming was investigated. RESULTS: In the murine model, a single intravenous dose of reovirus reduced metastatic lymph node burden and induced antitumor immunity (splenocyte response to tyrosinase-related protein-2 and interleukin-12 production in disaggregated lymph nodes). In vitro human assays revealed that uninfected Mel888 cells failed to induce dendritic cell maturation or support priming of an anti-Mel888 CTL response. In contrast, reovirus-infected Mel888 cells (reo-Mel) matured dendritic cells in a reovirus dose-dependent manner. When cultured with autologous peripheral blood lymphocytes, dendritic cells loaded with reo-Mel induced lymphocyte expansion, IFN-gamma production, specific anti-Mel888 cell cytotoxicity, and cross-primed CD8+ T cells specific against the human tumor-associated antigen MART-1. CONCLUSION: Reovirus infection of tumor cells reduces metastatic disease burden and primes antitumor immunity. Future clinical trials should be designed to explore both direct cytotoxic and immunotherapeutic effects of reovirus.


Subject(s)
Antineoplastic Agents/therapeutic use , Cytotoxicity, Immunologic/immunology , Neoplasms, Experimental/therapy , Oncolytic Virotherapy/methods , Reoviridae Infections/immunology , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Interferon-gamma/biosynthesis , Intramolecular Oxidoreductases/immunology , Lymphatic Metastasis/pathology , Lymphocyte Activation/immunology , MART-1 Antigen , Mammalian orthoreovirus 3/immunology , Mice , Mice, Inbred C57BL , Neoplasm Proteins/immunology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/virology , Polymerase Chain Reaction , T-Lymphocytes, Cytotoxic/immunology
6.
Cancer Immunol Res ; 7(5): 828-840, 2019 05.
Article in English | MEDLINE | ID: mdl-30940643

ABSTRACT

Antitumor T-cell responses raised by first-line therapies such as chemotherapy, radiation, tumor cell vaccines, and viroimmunotherapy tend to be weak, both quantitatively (low frequency) and qualitatively (low affinity). We show here that T cells that recognize tumor-associated antigens can directly kill tumor cells if used at high effector-to-target ratios. However, when these tumor-reactive T cells were present at suboptimal ratios, direct T-cell-mediated tumor cell killing was reduced and the ability of tumor cells to evolve away from a coapplied therapy (oncolytic or suicide gene therapy) was promoted. This T-cell-mediated increase in therapeutic resistance was associated with C to T transition mutations that are characteristic of APOBEC3 cytosine deaminase activity and was induced through a TNFα and protein kinase C-dependent pathway. Short hairpin RNA inhibition of endogenous APOBEC3 reduced rates of tumor escape from oncolytic virus or suicide gene therapy to those seen in the absence of antitumor T-cell coculture. Conversely, overexpression of human APOBEC3B in tumor cells enhanced escape from suicide gene therapy and oncolytic virus therapy both in vitro and in vivo Our data suggest that weak affinity or low frequency T-cell responses against tumor antigens may contribute to the ability of tumor cells to evolve away from first-line therapies. We conclude that immunotherapies need to be optimized as early as possible so that, if they do not kill the tumor completely, they do not promote treatment resistance.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy , Melanoma, Experimental/therapy , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Line, Tumor , Cytidine Deaminase/genetics , Cytidine Deaminase/immunology , Female , Ganciclovir/therapeutic use , Mammalian orthoreovirus 3 , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Mice, Inbred C57BL , Mice, Transgenic , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/immunology , Mutation , Oncolytic Virotherapy , Tumor Escape
7.
Cancer Immunol Res ; 6(10): 1161-1173, 2018 10.
Article in English | MEDLINE | ID: mdl-30209061

ABSTRACT

Immunotherapy is showing promise for otherwise incurable cancers. Oncolytic viruses (OVs), developed as direct cytotoxic agents, mediate their antitumor effects via activation of the immune system. However, OVs also stimulate antiviral immune responses, including the induction of OV-neutralizing antibodies. Current dogma suggests that the presence of preexisting antiviral neutralizing antibodies in patients, or their development during viral therapy, is a barrier to systemic OV delivery, rendering repeat systemic treatments ineffective. However, we have found that human monocytes loaded with preformed reovirus-antibody complexes, in which the reovirus is fully neutralized, deliver functional replicative reovirus to tumor cells, resulting in tumor cell infection and lysis. This delivery mechanism is mediated, at least in part, by antibody receptors (in particular FcγRIII) that mediate uptake and internalization of the reovirus/antibody complexes by the monocytes. This finding has implications for oncolytic virotherapy and for the design of clinical OV treatment strategies. Cancer Immunol Res; 6(10); 1161-73. ©2018 AACR.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Melanoma, Experimental/therapy , Monocytes/immunology , Oncolytic Virotherapy , Oncolytic Viruses , Reoviridae , Animals , Cell Line , Chlorocebus aethiops , Female , Humans , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Mice, Inbred C57BL , Receptors, IgG/immunology
8.
Sci Transl Med ; 10(422)2018 01 03.
Article in English | MEDLINE | ID: mdl-29298869

ABSTRACT

Immune checkpoint inhibitors, including those targeting programmed cell death protein 1 (PD-1), are reshaping cancer therapeutic strategies. Evidence suggests, however, that tumor response and patient survival are determined by tumor programmed death ligand 1 (PD-L1) expression. We hypothesized that preconditioning of the tumor immune microenvironment using targeted, virus-mediated interferon (IFN) stimulation would up-regulate tumor PD-L1 protein expression and increase cytotoxic T cell infiltration, improving the efficacy of subsequent checkpoint blockade. Oncolytic viruses (OVs) represent a promising form of cancer immunotherapy. For brain tumors, almost all studies to date have used direct intralesional injection of OV, because of the largely untested belief that intravenous administration will not deliver virus to this site. We show, in a window-of-opportunity clinical study, that intravenous infusion of oncolytic human Orthoreovirus (referred to herein as reovirus) leads to infection of tumor cells subsequently resected as part of standard clinical care, both in high-grade glioma and in brain metastases, and increases cytotoxic T cell tumor infiltration relative to patients not treated with virus. We further show that reovirus up-regulates IFN-regulated gene expression, as well as the PD-1/PD-L1 axis in tumors, via an IFN-mediated mechanism. Finally, we show that addition of PD-1 blockade to reovirus enhances systemic therapy in a preclinical glioma model. These results support the development of combined systemic immunovirotherapy strategies for the treatment of both primary and secondary tumors in the brain.


Subject(s)
Brain Neoplasms/therapy , Oncolytic Viruses/pathogenicity , Animals , Glioma/therapy , Humans , Immunotherapy/methods , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/metabolism
9.
Clin Cancer Res ; 17(9): 2767-76, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21389099

ABSTRACT

PURPOSE: Dendritic cells (DC) may be the most effective way of delivering oncolytic viruses to patients. Reovirus, a naturally occurring oncolytic virus, is currently undergoing early clinical trials; however, intravenous delivery of the virus is hampered by pre-existing antiviral immunity. Systemic delivery via cell carriage is a novel approach currently under investigation and initial studies have indicated its feasibility by using a variety of cell types and viruses. This study addressed the efficacy of human DC to transport virus in the presence of human neutralizing serum. EXPERIMENTAL DESIGN: Following reovirus-loading, DC or T cells were cocultured with melanoma cells with or without neutralizing serum; the melanoma cells were then analyzed for cell death. Following reovirus loading, cells were examined by electron microscopy to identify mechanisms of delivery. The phagocytic function of reovirus-loaded DC was investigated by using labeled tumor cells and the ability of reovirus-loaded DC to prime T cells was also investigated. RESULTS: In the presence of human neutralizing serum DC, but not T cells, were able to deliver reovirus for melanoma cell killing in vitro. Electron microscopy suggested that DC protected the virus by internalization, whereas with T cells it remained bound to the surface and hence accessible to neutralizing antibodies. Furthermore, DC loaded with reovirus were fully functional with regard to phagocytosis and priming of specific antitumor immune responses. CONCLUSIONS: The delivery of reovirus via DC could be a promising new approach offering the possibility of combining systemic viral therapy for metastatic disease with induction of an antitumor immune response.


Subject(s)
Antibodies, Neutralizing/adverse effects , Dendritic Cells/virology , Oncolytic Virotherapy , Oncolytic Viruses/metabolism , Reoviridae/physiology , Virus Internalization , Cell Line, Tumor , Cytotoxicity, Immunologic/physiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/physiology , Drug Carriers , Endocytosis/physiology , Humans , Melanoma/pathology , Melanoma/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/physiology , Reoviridae/metabolism , Skin Neoplasms/pathology , Skin Neoplasms/therapy , T-Lymphocytes/immunology , T-Lymphocytes/virology , Treatment Outcome , Viral Load/physiology
10.
Curr Opin Investig Drugs ; 10(12): 1372-82, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19943208

ABSTRACT

JX-594 is a replication-competent Wyeth strain vaccinia virus that was genetically modified to inactive the endogenous thymidine kinase gene and to express human GM-CSF and LacZ genes. In development by Jennerex Inc and licensee Green Cross Corp, the modified virus is a novel therapy for treatment-refractive metastatic malignancies from various sites of origin. Targeted oncolytic virotherapy has demonstrated promise in preclinical studies, and more than ten viral species have subsequently entered clinical trials. JX-594 has been modified to augment the intrinsic targeting and oncolytic potential of the vaccinia virus and to enhance antitumor immunity by the expression of the GM-CSF transgene in situ. In vitro and in vivo animal studies have demonstrated the replication specificity of JX-594 for cancer cell lines and tumors, and the restriction of serum human GM-CSF expression to tumor-bearing animals, resulting in significantly reduced tumor burden and an increase in median survival. In phase I trials, JX-594 was well tolerated, with mild systemic toxicity reported. In a phase I trial in seven patients with melanoma, one partial response and one complete response after surgery were observed. In another phase I trial in patients with hepatic carcinoma, three out of ten evaluable patients had a partial response and six had stable disease; the MTD was also established. A phase II trial in patients (expected n = 30) with unresectable primary hepatocellular carcinoma was recruiting at the time of publication, with completion expected in March 2010, and a phase III trial in patients with hepatocellular carcinoma was planned for the second half of 2010. Further clinical investigations are needed to explore the potential of this agent as a single therapy and as part of multimodal treatment regimens.


Subject(s)
Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses , Animals , Clinical Trials as Topic , Humans , Neoplasm Metastasis , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses/genetics , Thymidine Kinase/genetics , Treatment Outcome , Vaccinia virus/genetics , Virus Replication
11.
Clin Cancer Res ; 15(13): 4374-4381, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19509134

ABSTRACT

PURPOSE: Reovirus is a naturally occurring oncolytic virus in clinical trials. Although tumor infection by reovirus can generate adaptive antitumor immunity, its therapeutic importance versus direct viral oncolysis is undefined. This study addresses the requirement for viral oncolysis and replication, and the relative importance of antitumor immunity and direct oncolysis in therapy. EXPERIMENTAL DESIGN: Nonantigen specific T cells loaded with reovirus were delivered i.v. to C57BL/6 and severe combined immunodeficient mice bearing lymph node and splenic metastases from the murine melanoma, B16ova, with assessment of viral replication, metastatic clearance by tumor colony outgrowth, and immune priming. Human cytotoxic lymphocyte priming assays were done with dendritic cells loaded with Mel888 cells before the addition of reovirus. RESULTS: B16ova was resistant to direct oncolysis in vitro, and failed to support reovirus replication in vitro or in vivo. Nevertheless, reovirus purged lymph node and splenic metastases in C57BL/6 mice and generated antitumor immunity. In contrast, reovirus failed to reduce tumor burden in severe combined immunodeficient mice bearing either B16ova or reovirus-sensitive B16tk metastases. In the human system, reovirus acted solely as an adjuvant when added to dendritic cells already loaded with Mel888, supporting priming of specific antitumor cytotoxic lymphocyte, in the absence of significant direct tumor oncolysis; UV-treated nonreplicating reovirus was similarly immunogenic. CONCLUSION: The immune response is critical in mediating the efficacy of reovirus, and does not depend upon direct viral oncolysis or replication. The findings are of direct relevance to fulfilling the potential of this novel anticancer agent.


Subject(s)
Cytotoxicity, Immunologic/physiology , Mammalian orthoreovirus 3/physiology , Melanoma, Experimental/therapy , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Virus Replication/physiology , Animals , Cell Line, Tumor , Cells, Cultured , Humans , Immunotherapy, Adoptive/methods , Mammalian orthoreovirus 3/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/virology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Oncolytic Viruses/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/transplantation , T-Lymphocytes, Cytotoxic/virology , Treatment Outcome , Virus Replication/immunology
SELECTION OF CITATIONS
SEARCH DETAIL