Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nat Commun ; 12(1): 1930, 2021 03 26.
Article in English | MEDLINE | ID: mdl-33772027

ABSTRACT

In our clinical trials of oncolytic vesicular stomatitis virus expressing interferon beta (VSV-IFNß), several patients achieved initial responses followed by aggressive relapse. We show here that VSV-IFNß-escape tumors predictably express a point-mutated CSDE1P5S form of the RNA-binding Cold Shock Domain-containing E1 protein, which promotes escape as an inhibitor of VSV replication by disrupting viral transcription. Given time, VSV-IFNß evolves a compensatory mutation in the P/M Inter-Genic Region which rescues replication in CSDE1P5S cells. These data show that CSDE1 is a major cellular co-factor for VSV replication. However, CSDE1P5S also generates a neo-epitope recognized by non-tolerized T cells. We exploit this predictable neo-antigenesis to drive, and trap, tumors into an escape phenotype, which can be ambushed by vaccination against CSDE1P5S, preventing tumor escape. Combining frontline therapy with escape-targeting immunotherapy will be applicable across multiple therapies which drive tumor mutation/evolution and simultaneously generate novel, targetable immunopeptidomes associated with acquired treatment resistance.


Subject(s)
DNA-Binding Proteins/immunology , Interferon-beta/immunology , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , RNA-Binding Proteins/immunology , Vesicular stomatitis Indiana virus/immunology , Virus Replication/immunology , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Humans , Immunotherapy/methods , Interferon-beta/metabolism , Mice, Inbred C57BL , Mutation , Oncolytic Viruses/metabolism , Oncolytic Viruses/physiology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vesicular stomatitis Indiana virus/metabolism , Vesicular stomatitis Indiana virus/physiology
2.
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
3.
Cancers (Basel) ; 12(11)2020 Oct 31.
Article in English | MEDLINE | ID: mdl-33142841

ABSTRACT

Oncolytic virotherapy (OVT) has received significant attention in recent years, especially since the approval of talimogene Laherparepvec (T-VEC) in 2015 by the Food and Drug administration (FDA). Mechanistic studies of oncolytic viruses (OVs) have revealed that most, if not all, OVs induce direct oncolysis and stimulate innate and adaptive anti-tumour immunity. With the advancement of tumour modelling, allowing characterisation of the effects of tumour microenvironment (TME) components and identification of the cellular mechanisms required for cell death (both direct oncolysis and anti-tumour immune responses), it is clear that a "one size fits all" approach is not applicable to all OVs, or indeed the same OV across different tumour types and disease locations. This article will provide an unbiased review of oncolytic reovirus (clinically formulated as pelareorep), including the molecular and cellular requirements for reovirus oncolysis and anti-tumour immunity, reports of pre-clinical efficacy and its overall clinical trajectory. Moreover, as it is now abundantly clear that the true potential of all OVs, including reovirus, will only be reached upon the development of synergistic combination strategies, reovirus combination therapeutics will be discussed, including the limitations and challenges that remain to harness the full potential of this promising therapeutic agent.

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.
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
6.
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
7.
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
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.
Cancer Immunol Res ; 5(11): 1029-1045, 2017 11.
Article in English | MEDLINE | ID: mdl-29038298

ABSTRACT

Understanding how incompletely cleared primary tumors transition from minimal residual disease (MRD) into treatment-resistant, immune-invisible recurrences has major clinical significance. We show here that this transition is mediated through the subversion of two key elements of innate immunosurveillance. In the first, the role of TNFα changes from an antitumor effector against primary tumors into a growth promoter for MRD. Second, whereas primary tumors induced a natural killer (NK)-mediated cytokine response characterized by low IL6 and elevated IFNγ, PD-L1hi MRD cells promoted the secretion of IL6 but minimal IFNγ, inhibiting both NK-cell and T-cell surveillance. Tumor recurrence was promoted by trauma- or infection-like stimuli inducing VEGF and TNFα, which stimulated the growth of MRD tumors. Finally, therapies that blocked PD-1, TNFα, or NK cells delayed or prevented recurrence. These data show how innate immunosurveillance mechanisms, which control infection and growth of primary tumors, are exploited by recurrent, competent tumors and identify therapeutic targets in patients with MRD known to be at high risk of relapse. Cancer Immunol Res; 5(11); 1029-45. ©2017 AACR.


Subject(s)
Killer Cells, Natural/immunology , Melanoma, Experimental/immunology , Neoplasm Recurrence, Local/immunology , Skin Neoplasms/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Antibodies/therapeutic use , Cell Line, Tumor , Cytokines/immunology , Female , Ganciclovir/therapeutic use , Immunologic Surveillance , Melanoma, Experimental/therapy , Mice, Inbred C57BL , Mice, Transgenic , Oncolytic Virotherapy , Paclitaxel/therapeutic use , Reoviridae , Skin Neoplasms/therapy , T-Lymphocytes/transplantation
10.
R Soc Open Sci ; 4(5): 160855, 2017 May.
Article in English | MEDLINE | ID: mdl-28572987

ABSTRACT

High trait anxiety has been associated with detriments in emotional face processing. By contrast, relatively little is known about the effects of state anxiety on emotional face processing. We investigated the effects of state anxiety on recognition of emotional expressions (anger, sadness, surprise, disgust, fear and happiness) experimentally, using the 7.5% carbon dioxide (CO2) model to induce state anxiety, and in a large observational study. The experimental studies indicated reduced global (rather than emotion-specific) emotion recognition accuracy and increased interpretation bias (a tendency to perceive anger over happiness) when state anxiety was heightened. The observational study confirmed that higher state anxiety is associated with poorer emotion recognition, and indicated that negative effects of trait anxiety are negated when controlling for state anxiety, suggesting a mediating effect of state anxiety. These findings may have implications for anxiety disorders, which are characterized by increased frequency, intensity or duration of state anxious episodes.

11.
Mol Ther Oncolytics ; 5: 75-86, 2017 Jun 16.
Article in English | MEDLINE | ID: mdl-28547002

ABSTRACT

Pediatric high-grade glioma (pHGG) and diffuse intrinsic pontine glioma (DIPG) are invasive tumors with poor survival. Oncolytic virotherapy, initially devised as a direct cytotoxic treatment, is now also known to act via immune-mediated mechanisms. Here we investigate a previously unreported mechanism of action: the inhibition of migration and invasion in pediatric brain tumors. We evaluated the effect of oncolytic herpes simplex virus 1716 (HSV1716) on the migration and invasion of pHGG and DIPG both in vitro using 2D (scratch assay, live cell imaging) and 3D (spheroid invasion in collagen) assays and in vivo using an orthotopic xenograft model of DIPG invasion. HSV1716 inhibited migration and invasion in pHGG and DIPG cell lines. pHGG cells demonstrated reduced velocity and changed morphology in the presence of virus. HSV1716 altered pHGG cytoskeletal dynamics by stabilizing microtubules, inhibiting glycogen synthase kinase-3, and preventing localized clustering of adenomatous polyposis coli (APC) to the leading edge of cells. HSV1716 treatment also reduced tumor infiltration in a mouse orthotopic xenograft DIPG model. Our results demonstrate that HSV1716 targets the migration and invasion of pHGG and DIPG and indicates the potential of an oncolytic virus (OV) to be used as a novel anti-invasive treatment strategy for pediatric brain tumors.

12.
Mol Ther Oncolytics ; 3: 16030, 2016.
Article in English | MEDLINE | ID: mdl-27933315

ABSTRACT

We showed previously that therapy with Vesicular Stomatitis Virus (VSV) expressing tumor-associated proteins eradicates established tumors. We show here that when cellular cDNA were cloned into VSV which retained their own poly-A signal, viral species emerged in culture which had deleted the cellular poly-A signal and also contained a truncated form of the protein coding sequence. Typically, the truncation occurred such that a Tyrosine-encoding codon was converted into a STOP codon. We believe that the truncation of tumor-associated proteins expressed from VSV in this way occurred to preserve the ability of the virus to replicate efficiently. Truncated cDNA expressed from VSV were significantly more effective than full length cDNA in treating established tumors. Moreover, tumor therapy with truncated cDNA was completely abolished by depletion of CD4+ T cells, whereas therapy with full length cDNA was CD8+ T cell dependent. These data show that the type/potency of antitumor immune responses against self-tumor-associated proteins can be manipulated in vivo through the nature of the self protein (full length or truncated). Therefore, in addition to generation of neoantigens through sequence mutation, immunological tolerance against self-tumor-associated proteins can be broken through manipulation of protein integrity, allowing for rational design of better self-immunogens for cancer immunotherapy.

13.
Neuro Oncol ; 18(4): 518-27, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26409567

ABSTRACT

BACKGROUND: Systemic delivery of a complementary cDNA library expressed from the vesicular stomatitis virus (VSV) treats tumors by vaccinating against a wide range of tumor associated antigens (TAAs). For subcutaneous B16 melanomas, therapy was achieved using a specific combination of self-TAAs (neuroblastoma-Ras, cytochrome c, and tyrosinase-related protein 1) expressed from VSV. However, for intracranial B16 tumors, a different combination was therapeutic (consisting of VSV-expressed hypoxia-inducible factor [HIF]-2α, Sox-10, c-Myc, and tyrosinase-related protein 1). Therefore, we tested the hypothesis that tumors of different histological types growing in the brain share a common immunogenic signature which can be exploited for immunotherapy. METHODS: Syngeneic tumors, including GL261 gliomas, in the brains of immune competent mice were analyzed for their antigenic profiles or were treated with systemic viroimmunotherapy. RESULTS: Several different histological types of tumors growing intracranially, as well as freshly resected human brain tumor explants, expressed a HIF-2α(Hi) phenotype imposed by brain-derived CD11b+ cells. This location-specific antigen expression was exploited therapeutically against intracranial GL261 gliomas using systemically delivered VSV expressing HIF-2α, Sox-10, and c-Myc. Viroimmunotherapy was enhanced by immune checkpoint inhibitors, associated with the de-repression of antitumor T-helper cell type 1 (Th1) interferon-γ and Th17 T cell responses. CONCLUSIONS: Since different tumor types growing in the same location in the brain share a location-specific phenotype, we suggest that antigen-specific immunotherapies should be based upon expression of both histological type-specific tumor antigens and location-specific antigens. Our findings support clinical application of VSV-TAA therapy with checkpoint inhibition for aggressive brain tumors and highlight the importance of the intracranial microenvironment in sculpting a location-specific profile of tumor antigen expression.


Subject(s)
Antigens, Neoplasm/immunology , Brain Neoplasms/therapy , Cell Cycle Checkpoints/immunology , Glioma/therapy , Immunotherapy , Melanoma, Experimental/therapy , Oncolytic Virotherapy , Vesiculovirus/physiology , Animals , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Cycle , Combined Modality Therapy , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Glioma/immunology , Glioma/metabolism , Glioma/pathology , Humans , Interferon-gamma , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Mol Ther ; 23(5): 845-856, 2015 May.
Article in English | MEDLINE | ID: mdl-25544599

ABSTRACT

We used a VSV-cDNA library to treat recurrent melanoma, identifying immunogenic antigens, allowing us to target recurrences with immunotherapy or chemotherapy. Primary B16 melanoma tumors were induced to regress by frontline therapy. Mice with recurrent tumors were treated with VSV-cDNA immunotherapy. A Th17 recall response was used to screen the VSV-cDNA library for individual viruses encoding rejection antigens, subsequently targeted using immunotherapy or chemotherapy. Recurrent tumors were effectively treated with a VSV-cDNA library using cDNA from recurrent B16 tumors. Recurrence-associated rejection antigens identified included Topoisomerase-IIα, YB-1, cdc7 kinase, and BRAF. Fourteen out of 16 recurrent tumors carried BRAF mutations (595-605 region) following frontline therapy, even though the parental B16 tumors were BRAF wild type. The emergence of mutated BRAF-containing recurrences served as an excellent target for BRAF-specific immune-(VSV-BRAF), or chemo-(PLX-4720) therapies. Successful PLX-4720 therapy of recurrent tumors was associated with the development of a broad spectrum of T-cell responses. VSV-cDNA technology can be used to identify recurrence specific antigens. Emergence of mutated BRAF may be a major effector of melanoma recurrence which could serve as a target for chemo or immune therapy. This study suggests a rationale for offering patients with initially wild-type BRAF melanomas an additional biopsy to screen for mutant BRAF upon recurrence.


Subject(s)
Melanoma/genetics , Melanoma/pathology , Mutation , Proto-Oncogene Proteins B-raf/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Antigens, Neoplasm/immunology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Base Sequence , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm , Ganciclovir/pharmacology , Gene Expression , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/pharmacology , Male , Melanoma/drug therapy , Melanoma/immunology , Melanoma, Experimental , Mice , Molecular Sequence Data , Neoplasm Recurrence, Local , Proto-Oncogene Proteins B-raf/chemistry , Thymidine Kinase/genetics
15.
Mol Ther ; 22(11): 1936-48, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25059678

ABSTRACT

Previously, we showed that vesicular stomatitis virus (VSV) engineered to express a cDNA library from human melanoma cells (ASMEL, Altered Self Melanoma Epitope Library) was an effective systemic therapy to treat subcutaneous (s.c.) murine B16 melanomas. Here, we show that intravenous treatment with the same ASMEL VSV-cDNA library was an effective treatment for established intra-cranial (i.c.) melanoma brain tumors. The optimal combination of antigens identified from the ASMEL which treated s.c. B16 tumors (VSV-N-RAS+VSV-CYTC-C+VSV-TYRP-1) was ineffective against i.c. B16 brain tumors. In contrast, combination of VSV-expressed antigens-VSV-HIF-2α+VSV-SOX-10+VSV-C-MYC+VSV-TYRP1-from ASMEL which was highly effective against i.c. B16 brain tumors, had no efficacy against the same tumors growing subcutaneously. Correspondingly, i.c. B16 tumors expressed a HIF-2α(Hi), SOX-10(Hi), c-myc(Hi), TYRP1, N-RAS(lo)Cytc(lo) antigen profile, which differed significantly from the HIF-2α(lo), SOX-10(lo), c-myc(lo), TYRP1, N-RAS(Hi)Cytc(Hi) phenotype of s.c. B16 tumors, and was imposed upon the tumor cells by CD11b(+) cells within the local brain tumor microenvironment. Combining T-cell costimulation with systemic VSV-cDNA treatment, long-term cures of mice with established i.c. tumors were achieved in about 75% of mice. Our data show that the anatomical location of a tumor profoundly affects the profile of antigens that it expresses.


Subject(s)
Antigens, Viral, Tumor/metabolism , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Vesiculovirus/genetics , Administration, Intravenous , Animals , Cell Line, Tumor , Gene Library , Genetic Vectors/administration & dosage , Humans , Immunotherapy/methods , Mice , Mice, Inbred C57BL , Organ Specificity , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Microenvironment , Vesiculovirus/metabolism
16.
Mol Ther ; 22(10): 1851-63, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24957982

ABSTRACT

Optimum clinical protocols require systemic delivery of oncolytic viruses in the presence of an intact immune system. We show that preconditioning with immune modulators, or loading virus onto carrier cells ex vivo, enhances virus-mediated antitumor activity. Our early trials of systemic reovirus delivery showed that after infusion reovirus could be recovered from blood cells--but not from plasma--suggesting that rapid association with blood cells may protect virus from neutralizing antibody. We therefore postulated that stimulation of potential carrier cells directly in vivo before intravenous viral delivery would enhance delivery of cell-associated virus to tumor. We show that mobilization of the CD11b(+) cell compartment by granulocyte macrophage-colony stimulating factor immediately before intravenous reovirus, eliminated detectable tumor in mice with small B16 melanomas, and achieved highly significant therapy in mice bearing well-established tumors. Unexpectedly, cytokine conditioning therapy was most effective in the presence of preexisting neutralizing antibody. Consistent with this, reovirus bound by neutralizing antibody effectively accessed monocytes/macrophages and was handed off to tumor cells. Thus, preconditioning with cytokine stimulated recipient cells in vivo for enhanced viral delivery to tumors. Moreover, preexisting neutralizing antibody to an oncolytic virus may, therefore, even be exploited for systemic delivery to tumors in the clinic.


Subject(s)
Cytokines/metabolism , Gene Transfer Techniques , Genetic Vectors/genetics , Oncolytic Viruses/genetics , Transduction, Genetic , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/metabolism , Antibodies, Viral , CD11b Antigen/metabolism , Cytokines/pharmacology , Cytotoxicity, Immunologic/drug effects , Female , Gene Expression Regulation/drug effects , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Immunity/drug effects , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mammalian orthoreovirus 3/genetics , Mammalian orthoreovirus 3/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/mortality , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Oncolytic Virotherapy , Oncolytic Viruses/immunology , Receptors, Fc/genetics , Receptors, Fc/metabolism , Tumor Burden
17.
Mol Ther ; 21(2): 348-57, 2013 02.
Article in English | MEDLINE | ID: mdl-23011032

ABSTRACT

Injection of oncolytic vesicular stomatitis virus (VSV) into established B16ova melanomas results in tumor regression, in large part by inducing innate immune reactivity against the viral infection, mediated by MyD88- and type III interferon (IFN)-, but not TLR-4-, signaling. We show here that intratumoral (IT) treatment with lipopolysaccharide (LPS), a TLR-4 agonist, significantly enhanced the local therapy induced by VSV by combining activation of different innate immune pathways. Therapy was further enhanced by co-recruiting a potent antitumor, adaptive T-cell response by using a VSV engineered to express the ovalbumin tumor-associated antigen ova, in combination with LPS. However, the combination of IT LPS with systemically delivered VSV resulted in rapid morbidity and mortality in the majority of mice. Decreasing the intravenous (IV) dose of VSV to levels at which toxicity was ameliorated did not enhance therapy compared with IT LPS alone. Toxicity of the systemic VSV + IT LPS regimen was associated with rapidly elevated levels of serum tumor necrosis factor-α (TNF-α) and interleukin (IL)-6, which neither systemic VSV, nor IT LPS, alone induced. These data show that therapy associated with direct IT injections of oncolytic viruses can be significantly enhanced by combination with agonists of innate immune activation pathways, which are not themselves activated by the virus alone. Importantly, they also highlight possible, unforeseen dangers of combination therapies in which an immunotherapy, even delivered locally at the tumor site, may systemically sensitize the patient to a cytokine shock-like response triggered by IV delivery of oncolytic virus.


Subject(s)
Oncolytic Virotherapy/adverse effects , Oncolytic Virotherapy/methods , Toll-Like Receptor 4/agonists , Vesiculovirus/genetics , Animals , Antigens, Neoplasm/immunology , Cell Line, Tumor , Combined Modality Therapy/adverse effects , Combined Modality Therapy/methods , Genetic Therapy , Genetic Vectors , Immunity, Innate , Immunotherapy , Interferon-alpha/metabolism , Interleukin-6/blood , Interleukin-6/immunology , Lipopolysaccharides/metabolism , Mice , Mice, Inbred C57BL , Oncolytic Viruses/genetics , Signal Transduction , T-Lymphocytes/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology , Vesiculovirus/immunology
18.
Nat Biotechnol ; 30(4): 337-43, 2012 Mar 18.
Article in English | MEDLINE | ID: mdl-22426030

ABSTRACT

Multiple intravenous injections of a cDNA library, derived from human melanoma cell lines and expressed using the highly immunogenic vector vesicular stomatitis virus (VSV), cured mice with established melanoma tumors. Successful tumor eradication was associated with the ability of mouse lymphoid cells to mount a tumor-specific CD4(+) interleukin (IL)-17 recall response in vitro. We used this characteristic IL-17 response to screen the VSV-cDNA library and identified three different VSV-cDNA virus clones that, when used in combination but not alone, achieved the same efficacy against tumors as the complete parental virus library. VSV-expressed cDNA libraries can therefore be used to identify tumor rejection antigens that can cooperate to induce anti-tumor responses. This technology should be applicable to antigen discovery for other cancers, as well as for other diseases in which immune reactivity against more than one target antigen contributes to disease pathology.


Subject(s)
Antigens, Neoplasm/immunology , Antigens, Neoplasm/isolation & purification , Interleukin-17/immunology , Melanoma/immunology , Melanoma/therapy , Neoplasms, Experimental/therapy , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Epitopes, T-Lymphocyte/immunology , Gene Library , Genetic Vectors , HSP70 Heat-Shock Proteins/metabolism , Humans , Lymphocytes/immunology , Melanoma/genetics , Melanoma, Experimental , Mice , Neoplasms, Experimental/immunology , Vesiculovirus
19.
Mol Ther ; 19(10): 1802-12, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21792179

ABSTRACT

We have previously reported that a burst of vascular endothelial growth factor (VEGF) signaling to tumor-associated endothelium induces a proviral state, during which systemically delivered oncolytic reovirus can replicate in endothelium, thereby inducing immune-mediated vascular collapse and significant antitumor therapy. Using chimeric receptors, we show here that induction of the proviral state proceeds through VEGFR2, but not VEGFR1, signaling in endothelial cells. In contrast, innate immune activation by reovirus-exposed endothelial cells was predominantly through VEGFR1. By screening conventional chemotherapies for their ability to induce similar effects in combination with reovirus both in vitro and in vivo, we observed that the proviral state could also be induced in endothelial cells exposed to VEGF during rebound from paclitaxel-mediated inhibition of VEGF signaling. We translated these in vitro findings in vivo by careful scheduling of paclitaxel chemotherapy with systemic virotherapy, neither of which alone had therapeutic effects against B16 tumors. Systemic availability of reovirus during endothelial cell recovery from paclitaxel treatment allowed for endothelial replication of the virus, immune-mediated therapy, and tumor cures. Therefore, careful scheduling of combination viro- and chemotherapies, which preclinical testing suggests are individually ineffective against tumor cells, can lead to rational new clinical protocols for systemic treatments with oncolytic viruses.


Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms/therapy , Oncolytic Virotherapy , Vascular Endothelial Growth Factor A/biosynthesis , Combined Modality Therapy , Drug Administration Schedule , Humans , Killer Cells, Natural/immunology , Lymphocyte Activation , Neoplasms/drug therapy , Neoplasms/immunology , Paclitaxel/administration & dosage , Reoviridae/physiology , Virus Replication
20.
Nat Med ; 17(7): 854-9, 2011 Jun 19.
Article in English | MEDLINE | ID: mdl-21685898

ABSTRACT

Effective cancer immunotherapy requires the release of a broad spectrum of tumor antigens in the context of potent immune activation. We show here that a cDNA library of normal tissue, expressed from a highly immunogenic viral platform, cures established tumors of the same histological type from which the cDNA library was derived. Immune escape occurred with suboptimal vaccination, but tumor cells that escaped the immune pressure were readily treated by second-line virus-based immunotherapy. This approach has several major advantages. Use of the cDNA library leads to presentation of a broad repertoire of (undefined) tumor-associated antigens, which reduces emergence of treatment-resistant variants and also permits rational, combined-modality approaches in the clinic. Finally, the viral vectors can be delivered systemically, without the need for tumor targeting, and are amenable to clinical-grade production. Therefore, virus-expressed cDNA libraries represent a novel paradigm for cancer treatment addressing many of the key issues that have undermined the efficacy of immuno- and virotherapy to date.


Subject(s)
Antigens, Neoplasm/genetics , Cancer Vaccines/genetics , Gene Library , Neoplasms/therapy , Animals , Cancer Vaccines/immunology , DNA, Complementary/genetics , Genetic Vectors/genetics , Male , Mice , Mice, Inbred C57BL , Neoplasms/genetics , Neoplasms/immunology , Polymerase Chain Reaction , Prostatic Neoplasms/therapy , Vesiculovirus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...