Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
J Clin Invest ; 134(1)2024 Jan 02.
Article in English | MEDLINE | ID: mdl-37988172

ABSTRACT

BACKGROUNDSystemic administration of adeno-associated virus (AAV) can trigger life-threatening inflammatory responses, including thrombotic microangiopathy (TMA), acute kidney injury due to atypical hemolytic uremic syndrome-like complement activation, immune-mediated myocardial inflammation, and hepatic toxicity.METHODSWe describe the kinetics of immune activation following systemic AAV serotype 9 (AAV9) administration in 38 individuals following 2 distinct prophylactic immunomodulation regimens. Group 1 received corticosteroids and Group 2 received rituximab plus sirolimus in addition to steroids to prevent anti-AAV antibody formation.RESULTSGroup 1 participants had a rapid increase in immunoglobulin M (IgM) and IgG. Increase in D-dimer, decline in platelet count, and complement activation are indicative of TMA. All Group 1 participants demonstrated activation of both classical and alternative complement pathways, as indicated by depleted C4 and elevated soluble C5b-9, Ba, and Bb antigens. Group 2 patients did not have a significant change in IgM or IgG and had minimal complement activation.CONCLUSIONSThis study demonstrates that TMA in the setting of AAV gene therapy is antibody dependent (classical pathway) and amplified by the alternative complement pathway. Critical time points and interventions are identified to allow for management of immune-mediated events that impact the safety and efficacy of systemic gene therapy.


Subject(s)
Dependovirus , Thrombotic Microangiopathies , Humans , Dependovirus/genetics , Thrombotic Microangiopathies/therapy , Immunoglobulin M , Immunoglobulin G
2.
Mol Cancer Ther ; 20(8): 1481-1494, 2021 08.
Article in English | MEDLINE | ID: mdl-34045231

ABSTRACT

Oncolytic vaccinia viruses have promising efficacy and safety profiles in cancer therapy. Although antitumor activity can be increased by manipulating viral genes, the relative efficacy of individual modifications has been difficult to assess without side-by-side comparisons. This study sought to compare the initial antitumor activity after intravenous administration of five vaccinia virus variants of the same Western Reserve backbone and thymidine kinase gene deletion in RIP-Tag2 transgenic mice with spontaneous pancreatic neuroendocrine tumors. Tumors had focal regions of infection at 5 days after all viruses. Natural killer (NK) cells were restricted to these sites of infection, but CD8+ T cells and tumor cell apoptosis were widespread and varied among the viruses. Antitumor activity of virus VV-A34, bearing amino acid substitution A34K151E to increase viral spreading, and virus VV-IL2v, expressing a mouse IL2 variant (mIL2v) with attenuated IL2 receptor alpha subunit binding, was similar to control virus VV-GFP. However, antitumor activity was significantly greater after virus VV-A34/IL2v, which expressed mIL2v together with A34K151E mutation and viral B18R gene deletion, and virus VV-GMCSF that expressed mouse GM-CSF. Both viruses greatly increased expression of CD8 antigens Cd8a/Cd8b1 and cytotoxicity genes granzyme A, granzyme B, Fas ligand, and perforin-1 in tumors. VV-A34/IL2v led to higher serum IL2 and greater tumor expression of death receptor ligand TRAIL, but VV-GMCSF led to higher serum GM-CSF, greater expression of leukocyte chemokines and adhesion molecules, and more neutrophil recruitment. Together, the results show that antitumor activity is similarly increased by viral expression of GM-CSF or IL2v combined with additional genetic modifications.


Subject(s)
Apoptosis , Cytokines/metabolism , Immunity , Neuroendocrine Tumors/therapy , Oncolytic Virotherapy/methods , Pancreatic Neoplasms/therapy , Vaccinia virus/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Female , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Interleukin-2/genetics , Interleukin-2/immunology , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/virology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/virology , Tumor Cells, Cultured
3.
Cancer Res ; 78(4): 922-937, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29259007

ABSTRACT

Oncolytic viruses pose many questions in their use in cancer therapy. In this study, we assessed the potential of mpJX-594 (mouse-prototype JX-594), a replication-competent vaccinia virus administered by intravenous injection, to target the tumor vasculature, produce immune activation and tumor cell killing more widespread than the infection, and suppress invasion and metastasis. These actions were examined in RIP-Tag2 transgenic mice with pancreatic neuroendocrine tumors that developed spontaneously and progressed as in humans. mpJX-594 initially infected tumor vascular endothelial cells, leading to vascular pruning and prolonged leakage in tumors but not in normal organs; parallel effects were observed in U87 gliomas. Viral infection spread to tumor cells, where tumor cell killing was much more widespread than the infection. Widespread tumor cell killing at 5 days was prevented by depletion of CD8+ T lymphocytes and did not require GM-CSF, as mpJX-594 variants that expressed human, mouse, or no GM-CSF produced equivalent amounts of killing. The antivascular, antitumor, and antimetastatic effects of mpJX-594 were amplified by concurrent or sequential administration of sunitinib, a multitargeted receptor tyrosine kinase inhibitor. These effects were not mimicked by selective inhibition of VEGFR2 despite equivalent vascular pruning, but were accompanied by suppression of regulatory T cells and greater influx of activated CD8+ T cells. Together, our results showed that mpJX-594 targets tumor blood vessels, spreads secondarily to tumor cells, and produces widespread CD8+ T-cell-dependent tumor cell killing in primary tumors and metastases, and that these effects can be amplified by coadministration of sunitinib.Significance: These findings reveal multiple unrecognized features of the antitumor properties of oncolytic vaccinia viruses, all of which can be amplified by the multitargeted kinase inhibitor sunitinib. Cancer Res; 78(4); 922-37. ©2017 AACR.


Subject(s)
Antineoplastic Agents/therapeutic use , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , Sunitinib/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Humans , Mice , Mice, Transgenic , Sunitinib/pharmacology , Vaccinia virus/immunology
4.
Cancer Cell ; 28(2): 210-24, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-26212250

ABSTRACT

Oncolytic viruses designed to attack malignant cells can in addition infect and destroy tumor vascular endothelial cells. We show here that this expanded tropism of oncolytic vaccinia virus to the endothelial compartment is a consequence of VEGF-mediated suppression of the intrinsic antiviral response. VEGF/VEGFR2 signaling through Erk1/2 and Stat3 leads to upregulation, nuclear localization, and activation of the transcription repressor PRD1-BF1/Blimp1. PRD1-BF1 does not contribute to the mitogenic effects of VEGF, but directly represses genes involved in type I interferon (IFN)-mediated antiviral signaling. In vivo suppression of VEGF signaling diminishes PRD1-BF1/Blimp1 expression in tumor vasculature and inhibits intravenously administered oncolytic vaccinia delivery to and consequent spread within the tumor.


Subject(s)
Neoplasms/virology , Oncolytic Viruses/physiology , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Animals , Cell Line , Cell Line, Tumor , Cells, Cultured , Gene Expression/drug effects , Gene Expression Profiling , Host-Pathogen Interactions , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/virology , Humans , Mice, Inbred C57BL , Microscopy, Fluorescence , Neoplasms/blood supply , Neoplasms/therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/virology , Positive Regulatory Domain I-Binding Factor 1 , RNA Interference , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcriptional Activation/drug effects , Vaccinia virus/physiology
5.
Methods Mol Biol ; 1317: 343-57, 2015.
Article in English | MEDLINE | ID: mdl-26072416

ABSTRACT

Primary liver cancer (hepatocellular carcinoma; HCC) in patients not eligible for surgery or transplant is currently treated by locoregional therapeutic approaches, including trans-arterial chemoembolization and radiofrequency ablation. Sorafenib (Nexavar; Bayer/Onyx) is currently the only approved systemic therapy for patients having failed locoregional interventions. Oncolytic viruses are designed to selectively replicate within, and subsequently lyse, cancer cells by a unique mechanisms-of-action that is not cross-resistant with approved therapies (Kirn et al., Nat Med 7:781-787, 2001; Parato et al., Nat Rev Cancer 5:965-976, 2005; Chiocca, Nat Rev Cancer 2:938-950, 2002; Heise and Kern, J Clin Invest 105:847-851, 2000). Given that these therapeutics are self-amplifying in tumors, the impact of dose on patient outcome is unclear. Pexa-Vec (JX-594) is an oncolytic and immunotherapeutic vaccinia virus which was shown to be well tolerated by intratumoral injection and intravenous infusions in Phase 1 trials (Park et al., Lancet Oncol 9:533-542, 2008; Breitbach et al., Nature 477:99-102, 2011). We present the design of a randomized dose-finding trial of Pexa-Vec in patients with advanced HCC in which Pexa-Vec was delivered by intratumoral injection three times every 2 weeks at one of two dose levels (1 × 10(8) plaque forming units (pfu) versus 1 × 10(9) pfu).


Subject(s)
Carcinoma, Hepatocellular/therapy , Injections, Intralesional , Liver Neoplasms/therapy , Oncolytic Virotherapy/methods , Vaccinia virus/physiology , Carcinoma, Hepatocellular/surgery , Follow-Up Studies , Humans , Image Processing, Computer-Assisted , Kaplan-Meier Estimate , Liver Neoplasms/surgery , Oncolytic Virotherapy/adverse effects , Treatment Outcome
6.
Curr Opin Virol ; 13: 49-54, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25900822

ABSTRACT

Oncolytic immunotherapies (OI) selectively infect, amplify within and destroy cancer cells, thereby representing a novel class of anti-cancer therapy. In addition to this primary mechanism-of-action (MOA), OI based on vaccinia have been shown to selectively target tumor-associated vasculature, triggering an acute reduction in tumor perfusion. This review focuses on a third complementary MOA for this product class: the induction of active immunotherapy. While the active immunotherapy approach has been validated by recent product approvals, the field is still faced with significant challenges. Tumors have evolved diverse mechanisms to hide from immune-mediated destruction. Here we hypothesize that oncolytic immunotherapy replication within tumors may tip the immune balance to allow for the effective induction and execution of adaptive anti-tumor immunity, resulting in long-term tumor control following OI clearance. This immune activation against the cancer can be augmented through OI 'arming' for the expression of immunostimulatory transgene products from the virus genome. With the first vaccinia OI (Pexa-Vec, thymidine kinase-inactivated vaccinia expressing Granulocyte-colony stimulating factor [GM-CSF]) now in advanced-stage clinical trials, it has become more important than ever to understand the complimentary MOA that contributes to tumor destruction and control in patients.


Subject(s)
Immunotherapy , Neoplasms/therapy , Oncolytic Viruses/genetics , Vaccinia virus/genetics , Animals , Humans , Neoplasms/immunology , Neoplasms/virology , Oncolytic Viruses/immunology , Vaccinia virus/immunology
7.
Oncolytic Virother ; 4: 25-31, 2015.
Article in English | MEDLINE | ID: mdl-27512667

ABSTRACT

Oncolytic immunotherapeutics (OIs) are viruses designed to preferentially replicate in and lyse cancer cells, thereby triggering antitumor immunity. Numerous oncolytic platforms are currently in clinical development. Here we review preclinical and clinical experience with Pexa-Vec (pexastimogene devacirepvec, JX-594). Pexa-Vec is derived from a vaccinia vaccine strain that has been engineered to target cancer cells and express the therapeutic transgene granulocyte macrophage colony-stimulating factor (GM-CSF) in order to stimulate antitumor immunity. Key to its ability to target metastatic disease is the evolution of unique vaccinia virus characteristics that allow for effective systemic dissemination. Multiple mechanisms of action (MOA) for Pexa-Vec have been demonstrated in preclinical models and patients: 1) tumor cell infection and lysis, 2) antitumor immune response induction, and 3) tumor vascular disruption. This review will summarize data on the Pexa-Vec MOA as well as provide an overview of the Pexa-Vec clinical development program from multiple Phase I studies, Phase II studies in renal cell cancer and colorectal cancer, through Phase IIb clinical testing in patients with advanced hepatocellular carcinoma (primary liver cancer).

8.
Mol Ther ; 23(3): 602-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25531693

ABSTRACT

Pexa-Vec (pexastimogene devacirepvec, JX-594) is an oncolytic and immunotherapeutic vaccinia virus designed to destroy cancer cells through viral lysis and induction of granulocyte-macrophage colony-stimulating factor (GM-CSF)-driven tumor-specific immunity. Pexa-Vec has undergone phase 1 and 2 testing alone and in combination with other therapies in adult patients, via both intratumoral and intravenous administration routes. We sought to determine the safety of intratumoral administration in pediatric patients. In a dose-escalation study using either 10(6) or 10(7) plaque-forming units per kilogram, we performed one-time injections in up to three tumor sites in five pediatric patients and two injections in one patient. Ages at study entry ranged from 4 to 21 years, and their cancer diagnoses included neuroblastoma, hepatocellular carcinoma, and Ewing sarcoma. All toxicities were ≤ grade 3. The most common side effects were sinus fever and sinus tachycardia. All three patients at the higher dose developed asymptomatic grade 1 treatment-related skin pustules that resolved within 3-4 weeks. One patient showed imaging evidence suggestive of antitumor biological activity. The two patients tested for cellular immunoreactivity to vaccinia antigens showed strong responses. Overall, our study suggests Pexa-Vec is safe to administer to pediatric patients by intratumoral administration and could be studied further in this patient population.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Cancer Vaccines/immunology , Gamma Rays , Immunotherapy/methods , Oncolytic Virotherapy/methods , Vaccinia virus/immunology , Adolescent , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cancer Vaccines/administration & dosage , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Child , Child, Preschool , Female , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Humans , Injections, Intralesional , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Male , Neoplasm Staging , Neuroblastoma/immunology , Neuroblastoma/pathology , Neuroblastoma/therapy , Sarcoma, Ewing/immunology , Sarcoma, Ewing/pathology , Sarcoma, Ewing/therapy , Vaccination , Vaccinia virus/genetics , Young Adult
9.
Mol Ther ; 23(1): 202-14, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25292189

ABSTRACT

Oncolytic viral therapy utilizes a tumor-selective replicating virus which preferentially infects and destroys cancer cells and triggers antitumor immunity. The Western Reserve strain of vaccinia virus (VV) is the most virulent strain of VV in animal models and has been engineered for tumor selectivity through two targeted gene deletions (vvDD). We performed the first-in-human phase 1, intratumoral dose escalation clinical trial of vvDD in 16 patients with advanced solid tumors. In addition to safety, we evaluated signs of vvDD replication and spread to distant tumors, pharmacokinetics and pharmacodynamics, clinical and immune responses to vvDD. Dose escalation proceeded without dose-limiting toxicities to a maximum feasible dose of 3 × 10(9) pfu. vvDD replication in tumors was reproducible. vvDD genomes and/or infectious particles were recovered from injected (n = 5 patients) and noninjected (n = 2 patients) tumors. At the two highest doses, vvDD genomes were detected acutely in blood in all patients while delayed re-emergence of vvDD genomes in blood was detected in two patients. Fifteen of 16 patients exhibited late symptoms, consistent with ongoing vvDD replication. In summary, intratumoral injection of the oncolytic vaccinia vvDD was well-tolerated in patients and resulted in selective infection of injected and noninjected tumors and antitumor activity.


Subject(s)
Breast Neoplasms/therapy , Colonic Neoplasms/therapy , Melanoma/therapy , Pancreatic Neoplasms/therapy , Skin Neoplasms/therapy , Vaccinia virus/immunology , Virus Replication/genetics , Aged , Animals , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Dose-Response Relationship, Immunologic , Female , Gene Deletion , Humans , Injections, Intralesional , Male , Melanoma/immunology , Melanoma/pathology , Middle Aged , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Oncolytic Viruses/growth & development , Oncolytic Viruses/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Vaccinia virus/genetics , Vaccinia virus/growth & development
10.
Sci Transl Med ; 5(185): 185ra63, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23677592

ABSTRACT

Oncolytic viruses cause direct cytolysis and cancer-specific immunity in preclinical models. The goal of this study was to demonstrate induction of functional anticancer immunity that can lyse target cancer cells in humans. Pexa-Vec (pexastimogene devacirepvec; JX-594) is a targeted oncolytic and immunotherapeutic vaccinia virus engineered to express human granulocyte-macrophage colony-stimulating factor (GM-CSF). Pexa-Vec demonstrated replication, GM-CSF expression, and tumor responses in previous phase 1 trials. We now evaluated whether Pexa-Vec induced functional anticancer immunity both in the rabbit VX2 tumor model and in patients with diverse solid tumor types in phase 1. Antibody-mediated complement-dependent cancer cell cytotoxicity (CDC) was induced by intravenous Pexa-Vec in rabbits; transfer of serum from Pexa-Vec-treated animals to tumor-bearing animals resulted in tumor necrosis and improved survival. In patients with diverse tumor types treated on a phase 1 trial, CDC developed within 4 to 8 weeks in most patients; normal cells were resistant to the cytotoxic effects. T lymphocyte activation in patients was evidenced by antibody class switching. We determined that patients with the longest survival duration had the highest CDC activity, and identified candidate target tumor cell antigens. Thus, we demonstrated that Pexa-Vec induced polyclonal antibody-mediated CDC against multiple tumor antigens both in rabbits and in patients with diverse solid tumor types.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/immunology , Complement System Proteins/immunology , Immunotherapy , Neoplasms/immunology , Oncolytic Virotherapy , Oncolytic Viruses/immunology , Vaccinia virus/immunology , Animals , Antigens, Neoplasm/immunology , Antineoplastic Agents/metabolism , Cell Survival , Disease Models, Animal , Humans , Necrosis , Neoplasms/pathology , Neoplasms/therapy , Rabbits , Serum/metabolism , Survival Analysis
11.
Cancer Res ; 73(4): 1265-75, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23393196

ABSTRACT

Efforts to selectively target and disrupt established tumor vasculature have largely failed to date. We hypothesized that a vaccinia virus engineered to target cells with activation of the ras/MAPK signaling pathway (JX-594) could specifically infect and express transgenes (hGM-CSF, ß-galactosidase) in tumor-associated vascular endothelial cells in humans. Efficient replication and transgene expression in normal human endothelial cells in vitro required either VEGF or FGF-2 stimulation. Intravenous infusion in mice resulted in virus replication in tumor-associated endothelial cells, disruption of tumor blood flow, and hypoxia within 48 hours; massive tumor necrosis ensued within 5 days. Normal vessels were not affected. In patients treated with intravenous JX-594 in a phase I clinical trial, we showed dose-dependent endothelial cell infection and transgene expression in tumor biopsies of diverse histologies. Finally, patients with advanced hepatocellular carcinoma, a hypervascular and VEGF-rich tumor type, were treated with JX-594 on phase II clinical trials. JX-594 treatment caused disruption of tumor perfusion as early as 5 days in both VEGF receptor inhibitor-naïve and -refractory patients. Toxicities to normal blood vessels or to wound healing were not evident clinically or on MRI scans. This platform technology opens up the possibility of multifunctional engineered vaccinia products that selectively target and infect tumor-associated endothelial cells, as well as cancer cells, resulting in transgene expression, vasculature disruption, and tumor destruction in humans systemically.


Subject(s)
Carcinoma, Hepatocellular/therapy , Liver Neoplasms/therapy , Neovascularization, Pathologic/prevention & control , Oncolytic Viruses/physiology , Vaccinia virus/physiology , Animals , Blotting, Western , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Cells, Cultured , Clinical Trials, Phase I as Topic , Endothelial Cells/drug effects , Endothelial Cells/physiology , Endothelial Cells/virology , Female , Fibroblast Growth Factor 2/pharmacology , Humans , Liver Neoplasms/blood supply , Liver Neoplasms/virology , Magnetic Resonance Imaging , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/therapy , Neoplasms, Experimental/virology , Neovascularization, Pathologic/virology , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Rabbits , Receptors, Vascular Endothelial Growth Factor/metabolism , Time Factors , Treatment Outcome , Vaccinia virus/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Virus Replication
12.
Nat Med ; 19(3): 329-36, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23396206

ABSTRACT

Oncolytic viruses and active immunotherapeutics have complementary mechanisms of action (MOA) that are both self amplifying in tumors, yet the impact of dose on subject outcome is unclear. JX-594 (Pexa-Vec) is an oncolytic and immunotherapeutic vaccinia virus. To determine the optimal JX-594 dose in subjects with advanced hepatocellular carcinoma (HCC), we conducted a randomized phase 2 dose-finding trial (n=30). Radiologists infused low- or high-dose JX-594 into liver tumors (days 1, 15 and 29); infusions resulted in acute detectable intravascular JX-594 genomes. Objective intrahepatic Modified Response Evaluation Criteria in Solid Tumors (mRECIST) (15%) and Choi (62%) response rates and intrahepatic disease control (50%) were equivalent in injected and distant noninjected tumors at both doses. JX-594 replication and granulocyte-macrophage colony-stimulating factor (GM-CSF) expression preceded the induction of anticancer immunity. In contrast to tumor response rate and immune endpoints, subject survival duration was significantly related to dose (median survival of 14.1 months compared to 6.7 months on the high and low dose, respectively; hazard ratio 0.39; P=0.020). JX-594 demonstrated oncolytic and immunotherapy MOA, tumor responses and dose-related survival in individuals with HCC.


Subject(s)
Carcinoma, Hepatocellular/therapy , Liver Neoplasms/therapy , Oncolytic Virotherapy , Vaccinia virus/genetics , Aged , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/mortality , Dose-Response Relationship, Immunologic , Female , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Immunotherapy , Liver Neoplasms/immunology , Liver Neoplasms/mortality , Male , Middle Aged , Oncolytic Viruses/metabolism , Survival Rate , Vaccinia virus/physiology , Virus Replication
13.
Cancer Res ; 73(1): 97-107, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23090117

ABSTRACT

Natural killer (NK) cell clearance of tumor cell emboli following surgery is thought to be vital in preventing postoperative metastases. Using a mouse model of surgical stress, we transferred surgically stressed NK cells into NK-deficient mice and observed enhanced lung metastases in tumor-bearing mice as compared with mice that received untreated NK cells. These results establish that NK cells play a crucial role in mediating tumor clearance following surgery. Surgery markedly reduced NK cell total numbers in the spleen and affected NK cell migration. Ex vivo and in vivo tumor cell killing by NK cells were significantly reduced in surgically stressed mice. Furthermore, secreted tissue signals and myeloid-derived suppressor cell populations were altered in surgically stressed mice. Significantly, perioperative administration of oncolytic parapoxvirus ovis (ORFV) and vaccinia virus can reverse NK cell suppression, which correlates with a reduction in the postoperative formation of metastases. In human studies, postoperative cancer surgery patients had reduced NK cell cytotoxicity, and we show for the first time that oncolytic vaccinia virus markedly increases NK cell activity in patients with cancer. These data provide direct in vivo evidence that surgical stress impairs global NK cell function. Perioperative therapies aimed at enhancing NK cell function will reduce metastatic recurrence and improve survival in surgical cancer patients.


Subject(s)
Killer Cells, Natural/immunology , Neoplasm Metastasis/prevention & control , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neoplastic Cells, Circulating/immunology , Oncolytic Virotherapy/methods , Surgical Procedures, Operative/adverse effects , Animals , Flow Cytometry , Humans , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Metastasis/immunology , Neoplasms, Experimental/surgery , Neoplastic Cells, Circulating/pathology , Oncolytic Viruses , Stress, Physiological/immunology
14.
Mol Ther ; 20(4): 749-58, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22186794

ABSTRACT

Oncolytic viruses are generally designed to be cancer selective on the basis of a single genetic mutation. JX-594 is a thymidine kinase (TK) gene-inactivated oncolytic vaccinia virus expressing granulocyte-macrophage colony-stimulating factor (GM-CSF) and lac-Z transgenes that is designed to destroy cancer cells through replication-dependent cell lysis and stimulation of antitumoral immunity. JX-594 has demonstrated a favorable safety profile and reproducible tumor necrosis in a variety of solid cancer types in clinical trials. However, the mechanism(s) responsible for its cancer-selectivity have not yet been well described. We analyzed the replication of JX-594 in three model systems: primary normal and cancer cells, surgical explants, and murine tumor models. JX-594 replication, transgene expression, and cytopathic effects were highly cancer-selective, and broad spectrum activity was demonstrated. JX-594 cancer-selectivity was multi-mechanistic; replication was activated by epidermal growth factor receptor (EGFR)/Ras pathway signaling, cellular TK levels, and cancer cell resistance to type-I interferons (IFNs). These findings confirm a large therapeutic index for JX-594 that is driven by common genetic abnormalities in human solid tumors. This appears to be the first description of multiple selectivity mechanisms, both inherent and engineered, for an oncolytic virus. These findings have implications for oncolytic viruses in general, and suggest that their cancer targeting is a complex and multifactorial process.


Subject(s)
Neoplasms/metabolism , Oncolytic Viruses/physiology , Poxviridae/physiology , Signal Transduction/physiology , Virus Replication/physiology , Animals , Blotting, Western , Cell Line, Tumor , ErbB Receptors/genetics , ErbB Receptors/metabolism , HeLa Cells , Humans , In Vitro Techniques , Leukocytes, Mononuclear , Mice , Mice, Nude , Neoplasms/genetics , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Poxviridae/genetics , Signal Transduction/genetics , Virus Replication/genetics
15.
Curr Pharm Biotechnol ; 13(9): 1768-72, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21740365

ABSTRACT

Oncolytic viruses (OVs) are designed to replicate in, and subsequently lyse cancer cells. Numerous oncolytic virus platforms are currently in development. Here we review preclinical and clinical experience with JX-594, the lead candidate from the targeted and armed oncolytic poxvirus class. JX-594 is derived from a vaccinia vaccine strain that has been engineered for 1) enhanced cancer targeting and 2) has been "armed" with the therapeutic transgene granulocytemacrophage colony stimulating factor (GM-CSF) to stimulate anti-tumoral immunity. Poxviruses have many ideal features for use as oncolytic agents. The development of oncolytic vaccinia viruses is supported by a large safety database accumulated in the smallpox eradication program. In addition, poxviruses have evolved unique capabilities for systemic spread through the blood that can be harnessed for the treatment of metastatic disease. JX-594 demonstrates a high degree of cancer selectivity and systemic efficacy by multiple mechanisms-of-action (MOAs) in preclinical testing. Data from Phase 1 and 2 clinical trials has confirmed that these features result in potent and systemic efficacy in patients with treatment refractory metastatic cancers.


Subject(s)
Neoplasms/therapy , Neoplasms/virology , Oncolytic Virotherapy/methods , Oncolytic Viruses/physiology , Poxviridae/physiology , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Drug Evaluation, Preclinical , Humans , Neoplasms/genetics , Neoplasms/metabolism , Oncolytic Viruses/genetics , Oncolytic Viruses/metabolism , Poxviridae/genetics , Poxviridae/metabolism , Vaccinia virus/genetics , Vaccinia virus/metabolism , Vaccinia virus/physiology
16.
Nature ; 477(7362): 99-102, 2011 Aug 31.
Article in English | MEDLINE | ID: mdl-21886163

ABSTRACT

The efficacy and safety of biological molecules in cancer therapy, such as peptides and small interfering RNAs (siRNAs), could be markedly increased if high concentrations could be achieved and amplified selectively in tumour tissues versus normal tissues after intravenous administration. This has not been achievable so far in humans. We hypothesized that a poxvirus, which evolved for blood-borne systemic spread in mammals, could be engineered for cancer-selective replication and used as a vehicle for the intravenous delivery and expression of transgenes in tumours. JX-594 is an oncolytic poxvirus engineered for replication, transgene expression and amplification in cancer cells harbouring activation of the epidermal growth factor receptor (EGFR)/Ras pathway, followed by cell lysis and anticancer immunity. Here we show in a clinical trial that JX-594 selectively infects, replicates and expresses transgene products in cancer tissue after intravenous infusion, in a dose-related fashion. Normal tissues were not affected clinically. This platform technology opens up the possibility of multifunctional products that selectively express high concentrations of several complementary therapeutic and imaging molecules in metastatic solid tumours in humans.


Subject(s)
Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Poxviridae/physiology , Adult , Aged , Aged, 80 and over , DNA, Viral/blood , Female , Gene Expression Regulation, Enzymologic , Humans , Infusions, Intravenous , Male , Middle Aged , Neoplasms/pathology , Neoplasms/surgery , Neoplasms/virology , Organisms, Genetically Modified/physiology , Transgenes/genetics , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
17.
Mol Ther ; 19(10): 1913-22, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21772252

ABSTRACT

JX-594 is a targeted and granulocyte macrophage-colony stimulating factor (GM-CSF)-expressing oncolytic poxvirus designed to selectively replicate in and destroy cancer cells through viral oncolysis and tumor-specific immunity. In order to study the mechanisms-of-action (MOA) of JX-594 in humans, a mechanistic proof-of-concept clinical trial was performed at a low dose equivalent to ≤10% of the maximum-tolerated dose (MTD) in other clinical trials. Ten patients with previously treated stage IV melanoma were enrolled. Tumors were injected weekly for up to nine total treatments. Blood samples and tumor biopsies were analyzed for evidence of transgene activity, virus replication, and immune stimulation. The ß-galactosidase (ß-gal) transgene was expressed in all patients as evidenced by antibody induction. Six patients had significant induction of GM-CSF-responsive white blood cell (WBC) subsets such as neutrophils (25-300% increase). JX-594 replication and subsequent shedding into blood was detectable in five patients after cycles 1-9. Tumor biopsies demonstrated JX-594 replication, perivascular lymphocytic infiltration, and diffuse tumor necrosis. Mild flu-like symptoms were the most common adverse events. In sum, JX-594 replication, oncolysis, and expression of both transgenes were demonstrated; replication was still evident after multiple cycles. These findings have implications for further clinical development of JX-594 and other transgene-armed oncolytic viruses.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Melanoma/therapy , Oncolytic Virotherapy , Poxviridae/genetics , Adult , Aged , Female , Humans , Male , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Poxviridae/physiology , Transgenes
18.
Mol Ther ; 19(4): 627-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21455205
19.
Mol Ther ; 19(6): 1170-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21427706

ABSTRACT

JX-594 is a targeted and granulocyte-macrophage colony stimulating factor (GM-CSF) expressing oncolytic poxvirus designed to selectively replicate in and destroy cancer cells through viral oncolysis and tumor-specific immunity. In a phase 1 trial, JX-594 injection into hepatocellular carcinoma (HCC) was well-tolerated and associated with viral replication, decreased tumor perfusion, and tumor necrosis. We hypothesized that JX-594 and sorafenib, a small molecule inhibitor of B-raf and vascular endothelial growth factor receptor (VEGFR) approved for HCC, would have clinical benefit in combination given their demonstrated efficacy in HCC patients and their complementary mechanisms-of-action. HCC cell lines were uniformly sensitive to JX-594. Anti-raf kinase effects of concurrent sorafenib inhibited JX-594 replication in vitro, whereas sequential therapy was superior to either agent alone in murine tumor models. We therefore explored pilot safety and efficacy of JX-594 followed by sorafenib in three HCC patients. In all three patients, sequential treatment was (i) well-tolerated, (ii) associated with significantly decreased tumor perfusion, and (iii) associated with objective tumor responses (Choi criteria; up to 100% necrosis). HCC historical control patients on sorafenib alone at the same institutions had no objective tumor responses (0 of 15). Treatment of HCC with JX-594 followed by sorafenib has antitumoral activity, and JX-594 may sensitize tumors to subsequent therapy with VEGF/VEGFR inhibitors.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzenesulfonates/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/therapy , Pyridines/therapeutic use , Vaccinia virus/physiology , Animals , Cell Line, Tumor , Female , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/therapy , Melanoma/drug therapy , Melanoma/therapy , Mice , Mice, SCID , Niacinamide/analogs & derivatives , Oncolytic Virotherapy/methods , Phenylurea Compounds , Sorafenib , Vaccinia virus/genetics , Xenograft Model Antitumor Assays
20.
Cytokine Growth Factor Rev ; 21(2-3): 85-9, 2010.
Article in English | MEDLINE | ID: mdl-20472490

ABSTRACT

Chemotherapy remains a common mode of anticancer treatment even though in most cancer indications the therapeutic approach is not effective and ultimately associated with the onset of chemoresistance. A better understanding of genetic differences in tumors ushered in the era of targeted therapy which has revolutionized the treatment of certain cancer types. However, generally targeted therapies are only cytostatic and a proportion of the patient population may be non-responsive to targeted therapy due to mutations of other genes in the same pathway (e.g. ras mutations in patients with colorectal cancer treated with EGFR targeted therapy). Therefore, there exists a need for a radically new approach to cancer therapy. Oncolytic viruses (OVs) possess many properties of an ideal cancer therapeutic. OVs are cytotoxic and target cancers via multiple mechanisms of action while at the same time exploiting validated genetic pathways known to be dysregulated in many cancers. Indeed, promising safety and efficacy data has emerged from Phase 1 and Phase 2 trials with diverse OVs (e.g. JX-594, a targeted oncolytic poxvirus). Though the field has lagged behind with pivotal, randomized Phase 3 trials, these are currently being initiated for a number of OVs. In addition, the field must ensure a continued clinical development of newly developed OVs; a strategy for the clinical development of novel cancer therapeutics is outlined.


Subject(s)
Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses , Clinical Trials as Topic , Humans , Oncolytic Viruses/genetics , Oncolytic Viruses/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL