Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cancer Gene Ther ; 21(4): 171-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24722357

ABSTRACT

Immune responses against oncolytic adenovirus (Ad) vectors are thought to limit vector anti-tumor efficacy. With Syrian hamsters, which are immunocompetent and whose tumors and normal tissues are permissive for replication of Ad5-based oncolytic Ad vectors, treating with high-dose cyclophosphamide (CP) to suppress the immune system and exert chemotherapeutic effects enhances Ad vector anti-tumor efficacy. However, long-term CP treatment and immunosuppression can lead to anemia and vector spread to normal tissues. Here, we employed three cycles of transient high-dose CP administration plus intratumoral injection of the oncolytic Ad vector VRX-007 followed by withdrawal of CP. Each cycle lasted 4-6 weeks. This protocol allowed the hamsters to remain healthy so the study could be continued for ~100 days. The tumors were very well suppressed throughout the study. With immunocompetent hamsters, the vector retarded tumor growth initially, but after 3-4 weeks the tumors resumed rapid growth and further injections of vector were ineffective. Preimmunization of the hamsters with Ad5 prevented vector spillover from the tumor to the liver yet still allowed for effective long-term anti-tumor efficacy. Our results suggest that a clinical protocol might be developed with cycles of transient chemotherapy plus intratumoral vector injection to achieve significant anti-tumor efficacy while minimizing the side effects of cytostatic treatment.


Subject(s)
Adenoviridae/physiology , Cyclophosphamide/administration & dosage , Genetic Vectors/drug effects , Neoplasms, Experimental/prevention & control , Oncolytic Virotherapy/methods , Oncolytic Viruses/physiology , Adenoviridae/genetics , Adenoviridae/immunology , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Cell Line, Tumor , Cricetinae , Immunosuppressive Agents/administration & dosage , Mesocricetus , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/virology , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology
2.
J Virol ; 88(2): 903-12, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24198418

ABSTRACT

The adenovirus death protein (ADP) is expressed at late times during a lytic infection of species C adenoviruses. ADP promotes the release of progeny virus by accelerating the lysis and death of the host cell. Since some human lymphocytes survive while maintaining a persistent infection with species C adenovirus, we compared ADP expression in these cells with ADP expression in lymphocytes that proceed with a lytic infection. Levels of ADP were low in KE37 and BJAB cells, which support a persistent infection. In contrast, levels of ADP mRNA and protein were higher in Jurkat cells, which proceed with a lytic infection. Epithelial cells infected with an ADP-overexpressing virus died more quickly than epithelial cells infected with an ADP-deleted virus. However, KE37, and BJAB cells remained viable after infection with the ADP-overexpressing virus. Although the levels of ADP mRNA increased in KE37 and BJAB cells infected with the ADP-overexpressing virus, the fraction of cells with detectable ADP was unchanged, suggesting that the control of ADP expression differs between epithelial and lymphocytic cells. When infected with an ADP-deleted adenovirus, Jurkat cells survived and maintained viral DNA for greater than 1 month. These findings are consistent with the notion that the level of ADP expression determines whether lymphocytic cells proceed with a lytic or a persistent adenovirus infection.


Subject(s)
Adenoviridae Infections/virology , Adenovirus E3 Proteins/metabolism , Adenoviruses, Human/metabolism , Lymphocytes/virology , Adenovirus E3 Proteins/genetics , Adenoviruses, Human/genetics , Cell Line , Humans , Virus Release , Virus Replication
3.
Cancer Gene Ther ; 20(9): 531-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23928730

ABSTRACT

We report that radiation enhances the antitumor efficacy of the oncolytic adenovirus vector VRX-007 in Syrian hamster tumors. We used tumor-specific irradiation of subcutaneous tumors and compared treatment options of radiation alone or combined with VRX-007 and cyclophosphamide (CP). Radiation therapy further augmented the VRX-007-mediated inhibition of tumor growth, in both CP-treated and non-CP-treated hamsters, even though radiation did not lead to increased viral replication in tumors when compared with those treated with VRX-007 alone. Moreover, tumor growth inhibition was similar in tumors irradiated either 1 week before or after injection with VRX-007, which suggests that radiation exerts its antitumor effect independently from vector therapy. Thus, our results demonstrate that these two therapies do not have to be provided simultaneously to enhance their combined effectiveness against subcutaneous hamster tumors.


Subject(s)
Adenoviridae/physiology , Genetic Vectors/physiology , Neoplasms , Oncolytic Viruses/physiology , Radiation , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/pharmacology , Cell Line, Tumor , Cricetinae , Cyclophosphamide/administration & dosage , Cyclophosphamide/pharmacology , Female , Genetic Vectors/administration & dosage , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/radiotherapy , Neoplasms/therapy , Oncolytic Virotherapy , Tumor Burden/drug effects , Tumor Burden/radiation effects , Virus Replication/radiation effects
4.
Cancer Gene Ther ; 20(9): 521-30, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23928731

ABSTRACT

We have previously reported that intratumoral injection of VRX-007--an Ad5 (a species C adenovirus)-based vector overexpressing adenovirus death protein--can suppress the growth of subcutaneous HaK (hamster renal cancer) tumors. VRX-007 replication and tumor growth inhibition are enhanced when the hamsters are immunosuppressed by a high dose of cyclophosphamide (CP), an immunosuppressive and chemotherapeutic agent. Here, we report that continuous immunosuppression with CP was not required for increased oncolytic activity of VRX-007 because short-term dosing or continuous dosing with the drug yielded similar antitumor results. Prolonged viral replication was found only in animals on continuous CP treatment. We used 007-Luc, a replication-competent, luciferase-expressing vector similar to VRX-007, to investigate the replication of the vector over time. Tumor growth inhibition was similar in hamsters given CP treatment either 1 week before or 1 week after 007-Luc injection, which suggests that CP exerts its antitumor efficacy independently of vector therapy. 007-Luc did not spread far from the inoculation site, even in immunosuppressed, CP-treated animals. Our results indicate that the enhanced effectiveness that is produced by the combination of VRX-007 and CP therapies is due to their two independent mechanisms and that they do not have to be given simultaneously for the improved outcome.


Subject(s)
Adenoviridae/genetics , Antineoplastic Agents, Alkylating/pharmacology , Cyclophosphamide/pharmacology , Genetic Vectors/genetics , Neoplasms/genetics , Oncolytic Viruses/genetics , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Cell Line, Tumor , Cricetinae , Cyclophosphamide/administration & dosage , Female , Gene Expression/drug effects , Gene Order , Genes, Reporter , Genetic Vectors/administration & dosage , Humans , Neoplasms/drug therapy , Neoplasms/therapy , Oncolytic Virotherapy , Virus Replication/drug effects
5.
Cancer Gene Ther ; 17(11): 761-70, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20596091

ABSTRACT

We have studied the oncolytic efficacy of two adenovirus vectors named KD3 and INGN 007, which differ from each other only in that whereas KD3 has two small deletions in its e1a gene that restrict its replication to rapidly cycling cells, INGN 007 has wild-type e1a gene. Both vectors overexpress the adenovirus death protein (ADP). Both KD3 and INGN 007 effectively suppressed the growth of subcutaneous human A549 and Hep3B tumors in nude mice upon intratumoral injection, and contained the growth of subcutaneous LNCaP tumors after intravenous injection, making some tumors shrink or disappear. However, in a more demanding model, intravenous injections of neither KD3 nor wild-type Ad5 were effective against subcutaneous A549 tumors, whereas INGN 007 increased the mean survival time by 35%. INGN 007 was also effective in suppressing tumor growth in a challenging A549 orthotopic lung cancer model. INGN 007 was superior to dl1520 (ONYX-015) in repressing subcutaneous A549 tumors. Our results suggest that vectors such as INGN 007 might provide better antitumor efficacy in the clinic as well.


Subject(s)
Adenoviridae/physiology , Genetic Vectors/metabolism , Neoplasms, Experimental/therapy , Oncolytic Viruses/genetics , Virus Replication , Adenoviridae/genetics , Animals , Cell Line, Tumor , Cricetinae , Female , Genetic Therapy , Humans , Mice , Mice, Nude , Oncolytic Virotherapy , Xenograft Model Antitumor Assays
6.
Cancer Gene Ther ; 16(12): 912-22, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19478829

ABSTRACT

Syrian hamster is a practical animal model for studying the systemic effects of oncolytic vectors derived from adenovirus serotype 5 (Ad5). Ad5 replicates well in Syrian hamster tissues, and Syrian hamster cell lines are available that are known to support Ad5 replication. In this study, we established four new Syrian hamster cell lines from transplantable pancreatic, renal, hepatic and lung tumors. The pancreatic cell line (SHPC6) and the renal cell line were highly permissive for Ad5 replication. The SHPC6 cell line formed disseminated intraperitoneal tumors when cells were injected into the peritoneal cavity. INGN 007, an oncolytic Ad5-based vector, completely reversed the growth of disseminated intraperitoneal SHPC6 tumor nodules following intraperitoneal injection of the vector, leading to 100% survival of the treated animals. SHPC6 cells also formed subcutaneous tumors, whose growth was suppressed by INGN 007 following intratumoral injection. INGN 007 replicated in both the intraperitoneal and subcutaneous SHPC6 tumors. Following intraperitoneal injection, INGN 007 did not replicate in the livers of hamsters with intraperitoneal SHPC6 tumors, and was not hepatotoxic. These studies suggest that the SHPC6 cell line may be useful as a model for disseminated pancreatic cancer, and that INGN 007 may be a safe and effective vector to treat these tumors.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/virology , Disease Models, Animal , Oncolytic Virotherapy/methods , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/virology , Adenoviridae/physiology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cricetinae , Female , Humans , Mesocricetus , Oncolytic Viruses/physiology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Xenograft Model Antitumor Assays
7.
Cancer Gene Ther ; 16(8): 625-37, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19197322

ABSTRACT

Preclinical biodistribution studies with INGN 007, an oncolytic adenovirus (Ad) vector, supporting an early stage clinical trial were conducted in Syrian hamsters, which are permissive for Ad replication, and mice, which are a standard model for assessing toxicity and biodistribution of replication-defective (RD) Ad vectors. Vector dissemination and pharmacokinetics following intravenous administration were examined by real-time PCR in nine tissues and blood at five time points spanning 1 year. Select organs were also examined for the presence of infectious vector/virus. INGN 007 (VRX-007), wild-type Ad5 and AdCMVpA (an RD vector) were compared in the hamster model, whereas only INGN 007 was examined in mice. DNA of all vectors was widely disseminated early after injection, but decayed rapidly in most organs. In the hamster model, DNA of INGN 007 and Ad5 was more abundant than that of the RD vector AdCMVpA at early times after injection, but similar levels were seen later. An increased level of INGN 007 and Ad5 DNA but not AdCMVpA DNA in certain organs early after injection, and the presence of infectious INGN 007 and Ad5 in lung and liver samples at early times after injection, strongly suggests that replication of INGN 007 and Ad5 occurred in several Syrian hamster organs. There was no evidence of INGN 007 replication in mice. In addition to providing important information about INGN 007, the results underscore the utility of the Syrian hamster as a permissive immunocompetent model for Ad5 pathogenesis and oncolytic Ad vectors.


Subject(s)
Adenoviridae/physiology , Genetic Vectors/pharmacokinetics , Animals , Cricetinae , DNA, Viral/isolation & purification , Female , Genetic Therapy , Genetic Vectors/administration & dosage , Injections, Intravenous , Liver/virology , Lung/virology , Male , Mesocricetus , Mice , Mice, Inbred C57BL , Models, Animal , Neoplasms/therapy , Oncolytic Viruses , Species Specificity , Virus Replication
8.
Cancer Gene Ther ; 16(8): 644-54, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19197324

ABSTRACT

Oncolytic (replication-competent) adenoviruses as anticancer agents provide new, promising tools to fight cancer. In support of a Phase I clinical trial, here we report safety data with INGN 007 (VRX-007), an oncolytic adenovirus with increased anti-tumor efficacy due to overexpression of the adenovirus-encoded ADP protein. Wild-type adenovirus type 5 (Ad5) and a replication-defective version of Ad5 were also studied as controls. A parallel study investigating the biodistribution of these viruses is described elsewhere in this issue. The toxicology experiments were conducted in two species, the Syrian hamster, which is permissive for INGN 007 and Ad5 replication and the poorly permissive mouse. The studies demonstrated that the safety profile of INGN 007 is similar to Ad5. Both viruses caused transient liver damage upon intravenous injection that resolved by 28 days post-infection. The No-Observable-Adverse-Effect-Level (NOAEL) for INGN 007 in hamsters was 3 x 10(10) viral particles per kg. In hamsters, the replication-defective vector caused less toxicity, indicating that replication of Ad vectors in the host is an important factor in pathogenesis. With mice, INGN 007 and Ad5 caused toxicity comparable to the replication-defective adenovirus vector. Partially based on these results, the FDA granted permission to enter into a Phase I clinical trial with INGN 007.


Subject(s)
Adenoviridae/physiology , Genetic Vectors/adverse effects , Adenovirus E3 Proteins/biosynthesis , Animals , Blood Cell Count , Cell Line , Cricetinae , Erythropoiesis , Genetic Vectors/administration & dosage , Humans , Injections, Intravenous , Liver/pathology , Liver/virology , Mesocricetus , Mice , Mice, Inbred C57BL , Oncolytic Viruses , Virus Replication
9.
Cancer Gene Ther ; 15(2): 61-72, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17992200

ABSTRACT

We have previously described oncolytic adenovirus (Ad) vectors KD3 and KD3-interferon (IFN) that were rendered cancer-specific by mutations in the E1A region of Ad; these mutations abolish binding of E1A proteins to p300/CBP and pRB. The antitumor activity of the vectors was enhanced by overexpression of the Adenovirus Death Protein (ADP, E3-11.6K) and by replication-linked expression of IFN-alpha. We hypothesized that the anticancer efficacy of the KD3-IFN vector could be further improved by expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). E1-deleted Ad vectors were constructed carrying reporter genes for enhanced green fluorescent protein or secreted placental alkaline phosphatase (SEAP) and a therapeutic gene for TRAIL under control of the TetON system. Expression of the genes was increased in the presence of a helper virus and the inducer doxycycline such that up to 231-fold activation of expression for the TetON-SEAP vector was obtained. Coinfection with TetON-TRAIL augmented oncolytic activity of KD3 and KD3-IFN in vitro. Induction of TRAIL expression did not reduce the yield of progeny virus. Combination of TetON-TRAIL and KD3-IFN produced superior antitumor activity in vivo as compared with either vector alone demonstrating the efficacy of a four-pronged cancer gene therapy approach, which includes Ad oncolysis, ADP overexpression, IFN-alpha-mediated immunotherapy, and pharmacologically controlled TRAIL activity.


Subject(s)
Adenoviridae/genetics , Adenovirus E3 Proteins/genetics , Genetic Therapy , Genetic Vectors , Interferon-alpha/genetics , Neoplasms/therapy , TNF-Related Apoptosis-Inducing Ligand/biosynthesis , TNF-Related Apoptosis-Inducing Ligand/genetics , Adenoviridae/physiology , Adenovirus E3 Proteins/biosynthesis , Adenovirus E3 Proteins/physiology , Animals , Apoptosis/genetics , Cell Line, Tumor , Doxycycline/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , Interferon-alpha/biosynthesis , Interferon-alpha/physiology , Mice , Mutation , Neoplasms/genetics , Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/physiology , Tetracycline/pharmacology
10.
Gene Ther ; 12(22): 1608-17, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16034456

ABSTRACT

We have constructed a novel oncolytic adenovirus (Ad) vector, named VRX-011, in which the replication of the vector is targeted to cancer cells by the replacement of the wild-type Ad E4 promoter with the human telomerase reverse transcriptase (hTERT) promoter. Genes in the Ad E4 transcription unit are essential for Ad replication; therefore, VRX-011 will grow efficiently only in cells in which the hTERT promoter is active, that is, in a wide range of cancer and immortalized cells but not in most somatic cells. Consistent with these expectations, VRX-011 replicated efficiently in all cancer cell lines examined, while its growth was restricted in various primary and normal cells. VRX-011 overexpresses ADP (also known as E3-11.6K), an Ad protein required for efficient cell lysis and release of virions from cells at late stages of infection. This overexpression enhances cell-to-cell spread and could significantly increase antitumor efficacy. In a xenograft model in nude mice, both intratumoral and intravenous administration of VRX-011 effectively suppressed the growth of subcutaneous Hep3B human liver tumors. Also, intravenous delivery of VRX-011 greatly reduced the number and size of A549 human lung cancer cell nodules in a disseminated lung tumor model in nude mice. Importantly, tail vein administration of different doses of VRX-011 in C57BL/6 mice showed minimal liver toxicity. Considering its broad range of lytic replication in cancer cells, its attenuated phenotype in primary cells, its efficacy in suppressing xenografts, and its low toxicity in mouse liver, VRX-011 is a promising candidate for further evaluation as an anticancer therapeutic.


Subject(s)
Adenoviridae/genetics , Adenovirus E4 Proteins/genetics , DNA-Binding Proteins/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Neoplasms/therapy , Telomerase/genetics , Adenosine Diphosphate/metabolism , Adenoviridae/metabolism , Animals , Cell Line , Cell Line, Tumor , Gene Expression Regulation, Viral , Genetic Engineering , Genetic Vectors/administration & dosage , Humans , Mice , Mice, Inbred Strains , Mice, Nude , Neoplasms/virology , Neoplasms, Experimental , Promoter Regions, Genetic , Transduction, Genetic/methods , Transgenes , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...