Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Ecol Evol ; 13(11): e10746, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38034331

ABSTRACT

Mating systems in angiosperms range from obligate outcrossing to highly self-fertilizing. The belief that obligate selfing does not exist is contradicted by genetic evidence in several populations of L. inflata, in which selfing is enforced by the anthers enclosing the style. However, whether the mating systems of these populations are typical, or an extreme across the species range is unknown. Such trends are hypothesized to result from selection for reproductive assurance under mate limitation at range margins. Here, we use ~7500 iNaturalist community science images, in which stylar exsertion can be observed, to test this hypothesis in L. inflata and, for comparison, in four typical congeneric Lobelias that express a staminate, then a pistillate phase (protandry). Specifically, we analyzed the effects of latitude and range marginality on the frequency of stylar exsertion and number of exserted flowers. Outcrossing capacity in L. inflata increased at low latitudes and near the overall range center, supporting our hypothesis, with exsertion frequencies significantly lower than in congenerics. Interestingly, in outcrossing capable individuals, the number of style-exserted flowers was consistent across the species range and among species, indicating outcrossing capable L. inflata individuals resemble congenerics. These findings suggest that variation in stylar exsertion is expressed among individuals rather than by all individuals within populations. However, whether this is a result of differences in exsertion allele frequencies or of differentiation in the induction of a threshold trait requires further study. Moreover, the trends in outcrossing capability revealed here imply the potential for geographic variation in L. inflata mating system.

2.
Mol Cancer Ther ; 19(5): 1148-1156, 2020 05.
Article in English | MEDLINE | ID: mdl-32156785

ABSTRACT

KRAS mutation is a negative predictive biomarker of anti-EGFR agents in patients with metastatic colorectal cancer (mCRC), and remains an elusive target. Pelareorep, a double-stranded RNA virus selectively replicates in KRAS-mutated cells, and is synergistic with irinotecan. A dose escalation trial of FOLFIRI/bevacizumab [irinotecan (150-180 mg/m2) and pelareorep (1 × 1010 TCID50-3 × 1010 TCID50)] was implemented in adult patients with oxaliplatin refractory/intolerant, KRAS-mutant mCRC. Pelareorep was administered intravenously over 1 hour on days 1-5 every 4 weeks. Additional studies included pharmacokinetics, tumor morphology, and immune responses. Among FOLFIRI-naïve patients, the highest dose of FOLFIRI/bevacizumab (180 mg/m2 irinotecan) and pelareorep (3 × 1010 TCID50) was well tolerated, without a dose-limiting toxicity. At the recommended phase II dose, 3 of 6 patients (50%) had a partial response; the median progression-free and overall survival (PFS, OS) were 65.6 weeks and 25.1 months, respectively. Toxicities included myelosuppression, fatigue, and diarrhea. Transmission electron microscopy revealed viral factories (viral collections forming vesicular structures), at various stages of development. Immunogold staining against viral capsid σ-1 protein demonstrated viral "homing" in the tumor cells. The nucleus displayed sufficient euchromatin regions suggestive of active transcription. Flow cytometry revealed rapid dendritic cell maturation (48 hours) with subsequent activation of cytotoxic T cells (7 days). The combination of pelareorep with FOLFIRI/bevacizumab is safe. The PFS and OS data are encouraging and deserve further exploration. Pelareorep leads to a clear recurrent immune stimulatory response with cytotoxic T-cell activation, and homes and replicates in the tumor.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/therapy , Mutation , Oncolytic Viruses/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Adult , Aged , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Apoptosis , Bevacizumab/administration & dosage , Biomarkers, Tumor/genetics , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Combined Modality Therapy , Female , Humans , Irinotecan/administration & dosage , Male , Maximum Tolerated Dose , Mice , Mice, Nude , Oncolytic Viruses/genetics , Prognosis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
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
4.
Cancers (Basel) ; 10(6)2018 May 25.
Article in English | MEDLINE | ID: mdl-29799479

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis, with 1 and 5-year survival rates of ~18% and 7% respectively. FOLFIRINOX or gemcitabine in combination with nab-paclitaxel are standard treatment options for metastatic disease. However, both regimens are more toxic than gemcitabine alone. Pelareorep (REOLYSIN®), a proprietary isolate of reovirus Type 3 Dearing, has shown antitumor activity in clinical and preclinical models. In addition to direct cytotoxic effects, pelareorep can trigger antitumor immune responses. Due to the high frequency of RAS mutations in PDAC, we hypothesized that pelareorep would promote selective reovirus replication in pancreatic tumors and enhance the anticancer activity of gemcitabine. Chemotherapy-naïve patients with advanced PDAC were eligible for the study. The primary objective was Clinical Benefit Rate (complete response (CR) + partial response (PR) + stable disease (SD) ≥ 12 weeks) and secondary objectives include overall survival (OS), toxicity, and pharmacodynamics (PD) analysis. The study enrolled 34 patients; results included one partial response, 23 stable disease, and 5 progressive disease. The median OS was 10.2 months, with a 1- and 2-year survival rate of 45% and 24%, respectively. The treatment was well tolerated with manageable nonhematological toxicities. PD analysis revealed reovirus replication within pancreatic tumor and associated apoptosis. Upregulation of immune checkpoint marker PD-L1 suggests future consideration of combining oncolytic virus therapy with anti-PD-L1 inhibitors. We conclude that pelareorep complements single agent gemcitabine in PDAC.

5.
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
6.
Gut ; 67(3): 562-573, 2018 03.
Article in English | MEDLINE | ID: mdl-27902444

ABSTRACT

OBJECTIVE: Oncolytic viruses (OVs) represent promising, proinflammatory cancer treatments. Here, we explored whether OV-induced innate immune responses could simultaneously inhibit HCV while suppressing hepatocellular carcinoma (HCC). Furthermore, we extended this exemplar to other models of virus-associated cancer. DESIGN AND RESULTS: Clinical grade oncolytic orthoreovirus (Reo) elicited innate immune activation within primary human liver tissue in the absence of cytotoxicity and independently of viral genome replication. As well as achieving therapy in preclinical models of HCC through the activation of innate degranulating immune cells, Reo-induced cytokine responses efficiently suppressed HCV replication both in vitro and in vivo. Furthermore, Reo-induced innate responses were also effective against models of HBV-associated HCC, as well as an alternative endogenous model of Epstein-Barr virus-associated lymphoma. Interestingly, Reo appeared superior to the majority of OVs in its ability to elicit innate inflammatory responses from primary liver tissue. CONCLUSIONS: We propose that Reo and other select proinflammatory OV may be used in the treatment of multiple cancers associated with oncogenic virus infections, simultaneously reducing both virus-associated oncogenic drive and tumour burden. In the case of HCV-associated HCC (HCV-HCC), Reo should be considered as an alternative agent to supplement and support current HCV-HCC therapies, particularly in those countries where access to new HCV antiviral treatments may be limited.


Subject(s)
Carcinoma, Hepatocellular/therapy , Hepacivirus/physiology , Liver Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/immunology , Reoviridae/immunology , Animals , Burkitt Lymphoma/immunology , Burkitt Lymphoma/therapy , Burkitt Lymphoma/virology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Culture Media, Conditioned/pharmacology , Hepacivirus/immunology , Hepatocytes , Herpesvirus 4, Human , Humans , Immunity, Innate , Interferon-alpha/metabolism , Interferon-beta/metabolism , Interferons , Interleukins/metabolism , Leukocytes, Mononuclear , Liver/immunology , Liver Neoplasms/immunology , Liver Neoplasms/virology , Mice , Mice, SCID , Natural Killer T-Cells/immunology , Virus Replication/drug effects , Xenograft Model Antitumor Assays
7.
Oncotarget ; 8(21): 35138-35153, 2017 May 23.
Article in English | MEDLINE | ID: mdl-28422714

ABSTRACT

New therapeutic interventions are essential for improved management of patients with metastatic colorectal cancer (mCRC). This is especially critical for those patients whose tumors harbor a mutation in the KRAS oncogene (40-45% of all patients). This patient cohort is excluded from receiving anti-EGFR monoclonal antibodies that have added a significant therapeutic benefit for KRAS wild type CRC patients. Reovirus, a double stranded (ds) RNA virus is in clinical development for patients with chemotherapy refractory KRAS mutated tumors. Toll Like Receptor (TLR) 3, a member of the toll like receptor family of the host innate immune system is the pattern recognition motif for dsRNA pathogens. Using TLR3 expressing commercial HEK-BlueTM-hTLR3 cells we confirm that TLR3 is the host pattern recognition motif responsible for the detection of reovirus. Further, our investigation with KRAS mutated HCT116 cell line showed that effective expression of host TLR3 dampens the infection potential of reovirus by mounting a robust innate immune response. Down regulation of TLR3 expression with siRNA improves the anticancer activity of reovirus. In vivo experiments using human CRC cells derived xenografts in athymic mice further demonstrate the beneficial effects of TLR3 knock down by improving tumor response rates to reovirus. Strategies to mitigate the TLR3 response pathway can be utilized as a tool towards improved reovirus efficacy to specifically target the dissemination of KRAS mutated CRC.


Subject(s)
Colorectal Neoplasms/therapy , Orthoreovirus, Mammalian/physiology , Proto-Oncogene Proteins p21(ras)/genetics , Toll-Like Receptor 3/genetics , Animals , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Knockdown Techniques , HCT116 Cells , Humans , Mice , Oncolytic Virotherapy , Orthoreovirus, Mammalian/immunology , Signal Transduction , Xenograft Model Antitumor Assays
8.
Cancer Chemother Pharmacol ; 79(4): 697-703, 2017 04.
Article in English | MEDLINE | ID: mdl-28289863

ABSTRACT

REOLYSIN® (pelareorep) is an investigational new drug, consisting of a live, replication-competent, Reovirus Type 3 Dearing strain in a proprietary formulation. Several preclinical and clinical trials with REOLYSIN® on a wide range of cancer indications have demonstrated antineoplastic activity on cells with activated RAS-signaling pathway. Furthermore, long-term survival benefits were evident in post-treatment patients indicating a potential antitumor immune response triggered by REOLYSIN®. Numerous mono and/or combination therapy studies with the agent showed a consistent safety profile. The current study is a phase II, single-arm, open label trial of REOLYSIN® in combination with carboplatin and paclitaxel for patients with advanced melanoma. Results from the 14 patients enrolled in the study exhibited no grade 4 adverse events or deaths but manageable grade-3 toxicities commonly attributed to REOLYSIN®, including pyrexia, chills, myalgia, pain, fatigue, and nausea. The number of treatment cycles ranged from 2 to 20 with a median of 6 cycles. The study met its treatment and efficacy goal for the first stage with three partial responses (ORR was 21%). No complete responses were noted. The median PFS and OS were 5.2 and 10.9 months, respectively. The 1-year OS was 43% with a disease control rate of 85%. In conclusion, REOLYSIN® combined with carboplatin and paclitaxel is a safe and potentially efficacious therapy for patients with advanced malignant melanoma. Additional combination studies using REOLYSIN® with chemo/immunotherapy drugs may support more favorable outcomes for patients in this indication.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Mammalian orthoreovirus 3 , Melanoma/drug therapy , Oncolytic Virotherapy , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carboplatin/administration & dosage , Disease-Free Survival , Female , Humans , Male , Middle Aged , Paclitaxel/administration & dosage , Treatment Outcome , Young Adult
9.
Cancer ; 122(6): 875-83, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26709987

ABSTRACT

BACKGROUND: The type 3 Dearing reovirus (Reolysin) is a naturally occurring virus that preferentially infects and causes oncolysis in tumor cells with a Ras-activated pathway. It induces host immunity and cell cycle arrest and acts synergistically with cytotoxic agents. METHODS: This study evaluated Reolysin combined with paclitaxel and carboplatin in patients with metastatic/recurrent KRAS-mutated or epidermal growth factor receptor (EGFR)-mutated/amplified non-small cell lung cancer. RESULTS: Thirty-seven patients were treated. Molecular alterations included 20 KRAS mutations, 10 EGFR amplifications, 3 EGFR mutations, and 4 BRAF-V600E mutations. In total, 242 cycles (median, 4; range, 1-47) were completed. The initial doses were area under the curve (AUC) 6 mg/mL/min for carboplatin, 200 mg/m(2) for paclitaxel on day 1, and 3 × 10(10) 50% tissue culture infective dose for Reolysin on days 1 to 5 of each 21-day cycle. Because of diarrhea and febrile neutropenia (in the first 2 patients), subsequent doses were reduced to 175 mg/m(2) for paclitaxel and AUC 5 mg/mL/min for carboplatin. Toxicities included fatigue, diarrhea, nausea/vomiting, neutropenia, arthralgia/myalgia, anorexia, and electrolyte abnormalities. Response Evaluation Criteria in Solid Tumors 1.0 responses included the following: partial response for 11 patients, stable disease (SD) for 20 patients, progressive disease for 4 patients, and not evaluable for 2 patients (objective response rate, 31%; 90% 1-sided lower confidence interval, 21%). Four SD patients had >40% positron emission tomography standardized uptake value reductions. The median progression-free survival, median overall survival, and 12-month overall survival rate were 4 months, 13.1 months, and 57%, respectively. Seven patients were alive after a median follow-up of 34.2 months; they included 2 patients without disease progression at 37 and 50 months. CONCLUSIONS: Reolysin in combination with paclitaxel and carboplatin was well tolerated. The observed response rate suggests a benefit of the reovirus for chemotherapy. A follow-up randomized study is recommended. The proportion of patients surviving longer than 2 years (30%) suggests a second/third-line treatment effect or possibly the triggering of an immune response after tumor reovirus infiltration.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mammalian orthoreovirus 3 , Neoplasm Recurrence, Local/drug therapy , Oncolytic Virotherapy , Proto-Oncogene Proteins p21(ras)/genetics , Aged , Aged, 80 and over , Disease-Free Survival , ErbB Receptors/genetics , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Grading , Oncolytic Virotherapy/methods
10.
Oncotarget ; 5(9): 2807-19, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24798549

ABSTRACT

Reovirus is a double stranded RNA virus, with an intrinsic preference for replication in KRAS mutant cells. As 45% of human colorectal cancers (CRC) harbor KRAS mutations, we sought to investigate its efficacy in KRAS mutant CRC cells, and examine its impact in combination with the topoisimerase-1 inhibitor, irinotecan. Reovirus efficacy was examined in the KRAS mutant HCT116, and the isogenic KRAS WT Hke3 cell line, and in the non-malignant rat intestinal epithelial cell line. Apoptosis was determined by flow cytometry and TUNEL staining. Combination treatment with reovirus and irintoecan was investigated in 15 CRC cell lines, including the HCT116 p21 isogenic cell lines. Reovirus preferentially induced apoptosis in KRAS mutant HCT116 cells compared to its isogenic KRAS WT derivative, and in KRAS mutant IEC cells. Reovirus showed a greater degree of caspase 3 activation with PARP 1 cleavage, and preferential inhibition of p21 protein expression in KRAS mutant cells. Reovirus synergistically induced growth inhibition when combined with irinotecan. This synergy was lost upon p21 gene knock out. Reovirus preferentially induces apoptosis in KRAS mutant colon cancer cells. Reovirus and irinotecan combination therapy is synergistic, p21 mediated, and represents a novel potential treatment for patients with CRC.


Subject(s)
Antineoplastic Agents/therapeutic use , Camptothecin/analogs & derivatives , Colorectal Neoplasms/therapy , Mutation/genetics , Oncolytic Virotherapy , Proto-Oncogene Proteins/genetics , Reoviridae/physiology , ras Proteins/genetics , Animals , Apoptosis , Blotting, Western , Camptothecin/therapeutic use , Cell Cycle , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Combined Modality Therapy , Drug Synergism , Humans , Irinotecan , Microscopy, Electron, Scanning , Proto-Oncogene Proteins p21(ras) , Rats , Tumor Cells, Cultured
11.
Mod Pathol ; 25(10): 1333-44, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22699519

ABSTRACT

We analyzed the in situ molecular correlates of infection from cancer patients treated with reovirus. Melanoma, colorectal, and ovarian cancer samples from such patients showed variable infection of the cancer cells but not the intermingled benign cells. RT in situ PCR showed most cancer cells contained the viral genome with threefold less having productive viral infection as documented by either tubulin or reoviral protein co-expression. Productive infection in the cancer cells was strongly correlated with co-expression of p38 and caspase-3 as well as apoptosis-related death (P<0.001). The cancer cell apoptotic death was due to a marked viral-induced inhibition of microRNA-let-7d that, in turn, upregulated caspase-3 activity. In summary, reovirus shows a striking tropism to cancer cells in clinical samples. A rate-limiting factor of reovirus-induced cancer cell death is productive viral infection that operates via the marked reduction of microRNA-let-7d and concomitant elevated caspase-3 expression.


Subject(s)
Apoptosis , Colorectal Neoplasms/pathology , Melanoma/pathology , MicroRNAs/metabolism , Oncolytic Virotherapy/methods , Orthoreovirus, Mammalian/physiology , Ovarian Neoplasms/pathology , Skin Neoplasms/pathology , Caspase 3/metabolism , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/virology , Female , Humans , Melanoma/metabolism , Melanoma/virology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/virology , Reoviridae Infections/virology , Skin Neoplasms/metabolism , Skin Neoplasms/virology , p38 Mitogen-Activated Protein Kinases/metabolism
12.
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
13.
Invest New Drugs ; 28(5): 641-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-19572105

ABSTRACT

BACKGROUND: Reolysin is reovirus serotype 3-Dearing strain, a double-stranded replication-competent RNA non-enveloped icosahedral virus. It induces cytopathic and anti-cancer effects in cells with an activated ras pathway due to inhibition of the dsRNA-activated protein kinase. METHODS: This was a single center dose escalation trial of Reolysin administered intravenously every 4 weeks in doses ranging from 1 x 10(8) to 3 x 10(10) tissue culture infective dose (TCID)(50). Serum for neutralizing antibody, and serum, stool, saliva, and urine for viral shedding were collected. Tumor samples were analyzed for activating mutations in the ras and braf oncogenes. RESULTS: Eighteen patients received 27 doses of Reolysin in 6 dose cohorts accomplishing a 300 fold dose escalation without a protocol-defined dose limiting toxicity. Drug related grade 2 toxicities included fatigue and fever (1 patient each). All patients developed neutralizing antibody during the course of the study. Viral shedding was observed in 6 patients. One patient with anthracycline and taxane refractory breast cancer experienced a partial response (PR) and her tumor had a ras G12A mutation. Biopsy from her chest wall mass showed evidence of necrosis and viral replication by electron microscopy. Overall clinical benefit (1 PR + 7 stable disease) rate was 45%, and appeared higher in patients with viral shedding (67%) than those without (33%). CONCLUSION: Reolysin administered monthly as a one-hour infusion is safe and well-tolerated even in multiple doses. Reolysin has anti-tumor activity as a single agent warranting further evaluation, including in combination with chemotherapy. Viral shedding may suggest intrapatient replication yielding a benefit and should be studied carefully in future studies.


Subject(s)
Antineoplastic Agents/administration & dosage , Mammalian orthoreovirus 3/physiology , Neoplasms/therapy , Virus Replication/physiology , Adult , Aged , Antibody Formation/immunology , Antineoplastic Agents/adverse effects , DNA Mutational Analysis , Female , Humans , Injections, Intravenous , Male , Mammalian orthoreovirus 3/ultrastructure , Middle Aged , Mutation/genetics , Neoplasms/immunology , RNA, Viral/blood , RNA, Viral/urine
14.
Mol Cancer ; 8: 47, 2009 Jul 14.
Article in English | MEDLINE | ID: mdl-19594950

ABSTRACT

BACKGROUND: Reovirus type 3 Dearing strain (ReoT3D) has an inherent propensity to preferentially infect and destroy cancer cells. The oncolytic activity of ReoT3D as a single agent has been demonstrated in vitro and in vivo against various cancers, including colon, pancreatic, ovarian and breast cancers. Its human safety and potential efficacy are currently being investigated in early clinical trials. In this study, we investigated the in vitro combination effects of ReoT3D and chemotherapeutic agents against human non-small cell lung cancer (NSCLC). RESULTS: ReoT3D alone exerted significant cytolytic activity in 7 of 9 NSCLC cell lines examined, with the 50% effective dose, defined as the initial virus dose to achieve 50% cell killing after 48 hours of infection, ranging from 1.46 +/- 0.12 approximately 2.68 +/- 0.25 (mean +/- SD) log10 pfu/cell. Chou-Talalay analysis of the combination of ReoT3D with cisplatin, gemcitabine, or vinblastine demonstrated strong synergistic effects on cell killing, but only in cell lines that were sensitive to these compounds. In contrast, the combination of ReoT3D and paclitaxel was invariably synergistic in all cell lines tested, regardless of their levels of sensitivity to either agent. Treatment of NSCLC cell lines with the ReoT3D-paclitaxel combination resulted in increased poly (ADP-ribose) polymerase cleavage and caspase activity compared to single therapy, indicating enhanced apoptosis induction in dually treated NSCLC cells. NSCLC cells treated with the ReoT3D-paclitaxel combination showed increased proportions of mitotic and apoptotic cells, and a more pronounced level of caspase-3 activation was demonstrated in mitotically arrested cells. CONCLUSION: These data suggest that the oncolytic activity of ReoT3D can be potentiated by taxanes and other chemotherapeutic agents, and that the ReoT3D-taxane combination most effectively achieves synergy through accelerated apoptosis triggered by prolonged mitotic arrest.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Mammalian orthoreovirus 3/physiology , Oncolytic Virotherapy/methods , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/virology , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Combined Modality Therapy , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Synergism , Enzyme Activation/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/virology , Paclitaxel/administration & dosage , Poly(ADP-ribose) Polymerases/metabolism , Vinblastine/administration & dosage , Virion/physiology , ras Proteins/metabolism , Gemcitabine
15.
Expert Opin Biol Ther ; 9(7): 817-30, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19527106

ABSTRACT

Reovirus is an oncolytic virus that is not associated with significant disease in humans, but is selectively able to replicate in cancer cells through exploitation of abnormal Ras signaling. Pre-clinical studies have demonstrated that treatment with reovirus is associated with significant anticancer activity across a range of tumor types. Reolysin is a proprietary formulation of the human reovirus developed by Oncolytics Biotech. Clinical evaluation of reovirus therapy has shown that it is well tolerated when administered locally or systemically. Encouraging anticancer efficacy has been observed with single-agent treatment and in combination with chemotherapy and radiotherapy. Phase II studies are currently evaluating reovirus alone and in combination with standard therapy in an array of tumor types. While immune sensitization hinders the anticancer efficacy of reovirus, it is important in preventing systemic toxicity. Immunosuppressive strategies are being developed that reduce immune neutralization of the virus to allow for improved tumor penetration, but retain sufficient antibody levels to protect normal tissues. The lack of toxicity and promising efficacy of reovirus has raised hopes that it will become an established anticancer agent.


Subject(s)
Neoplasms/therapy , Oncolytic Virotherapy/methods , Reoviridae , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Combined Modality Therapy , Humans , Neoplasms/immunology , Neoplasms/pathology
16.
Cancer Res ; 63(2): 348-53, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12543787

ABSTRACT

The human reovirus is an oncolytic virus that specifically targets cancer cells with an activated Ras pathway. Because it is replication competent and highly specific for cancer cells, this virus has the potential to be an effective antimetastatic cancer agent through remote site delivery. In this study, we exploited the ability of reovirus to replicate in murine cells to test the efficacy of this virus in eliminating distal and/or metastatic tumors in immune-competent mice. We found that i.v. therapy with reovirus not only inhibited metastatic tumor growth but also led to a significant improvement in animal survival. Combining i.v. reovirus treatment with immune suppression (cyclosporine A or anti-CD4/anti-CD8 antibodies) resulted in further reduction in tumor size and a considerable prolongation in survival, compared with viral therapy alone. Combined therapy was also effective in overcoming a preexisting immunity to reovirus (a common occurrence in humans and thus a potential impediment to oncolytic effectiveness) to induce metastatic tumor regression. This is the first study to use systemic delivery of an oncolytic agent in conjunction with immune-suppressive drugs to effectively prolong animal survival. Altogether, our results suggest that i.v. reovirus therapy may present a feasible, novel alternative in the treatment of metastatic cancer in humans.


Subject(s)
Neoplasms, Experimental/therapy , Neoplasms, Experimental/virology , Retroviridae/physiology , Animals , Carcinoma, Lewis Lung/therapy , Carcinoma, Lewis Lung/virology , Combined Modality Therapy , Cyclosporine/pharmacology , Cytopathogenic Effect, Viral , Disease Models, Animal , Female , Immunosuppressive Agents/pharmacology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Lung Neoplasms/virology , Mammary Neoplasms, Experimental/therapy , Mammary Neoplasms, Experimental/virology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Retroviridae/immunology
17.
Hum Gene Ther ; 13(5): 641-52, 2002 Mar 20.
Article in English | MEDLINE | ID: mdl-11916487

ABSTRACT

We have previously shown that human reovirus replication is restricted to cells with an activated Ras pathway, and that reovirus could be used as an effective oncolytic agent against human glioblastoma xenografts. This study examines in more detail the feasibility of reovirus as a therapeutic for breast cancer, a subset of cancer in which direct activating mutations in the ras proto-oncogene are rare, and yet where unregulated stimulation of Ras signaling pathways is important in the pathogenesis of the disease. We demonstrate herein the efficient lysis of breast tumor-derived cell lines by the virus, whereas normal breast cells resist infection in vitro. In vivo studies of reovirus breast cancer therapy reveal that viral administration could cause tumor regression in an MDA-MB-435S mammary fat pad model in severe combined immunodeficient mice. Reovirus could also effect regression of tumors remote from the injection site in an MDA-MB-468 bilateral tumor model, raising the possibility of systemic therapy of breast cancer by the oncolytic agent. Finally, the ability of reovirus to act against primary breast tumor samples not propagated as cell lines was evaluated; we found that reovirus could indeed replicate in ex vivo surgical specimens. Overall, reovirus shows promise as a potential breast cancer therapeutic.


Subject(s)
Biological Therapy , Breast Neoplasms/therapy , Reoviridae/physiology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Death/genetics , Female , Genes, ras , Genetic Therapy , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Proto-Oncogene Mas , Transplantation, Heterologous , Tumor Cells, Cultured , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...