Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Cancer Biol Ther ; 19(12): 1174-1184, 2018.
Article in English | MEDLINE | ID: mdl-30067431

ABSTRACT

Oncolytic adenoviruses (Ads) are cancer selective tumoricidal agents; however their mechanism of Ad-mediated cancer cell lysis, or oncolysis, remains undefined. This report focuses upon the autophagy mediator c-JUN n-terminal kinase (JNK) and its effects upon Ad oncolysis and replication. Previously, E1b-deleted Ads have been used to treat several hundred cancer patients with limited clinical efficacy. We hypothesize that by studying the potential interactions between E1b and JNK, mechanisms to improve oncolytic Ad design and cancer therapeutic efficacy may be elucidated. To test this hypothesis, E1b was selectively deleted from the Ad genome. These studies indicated that Ads encoding E1b induced JNK phosphorylation predominately occurred via E1b-19K. The expression of another crucial Ad gene E1a was then overexpressed by the CMV promoter via the replication competent Ad vector Adhz69; these data indicated that E1A also induced JNK phosphorylation. To assess the effects of host cell JNK expression upon Ad oncolysis and replication, siRNA targeting JNK1 and JNK2 (JNK1/2) were utilized. The oncolysis and replication of the E1b-19K wild-type Ads Ad5 and Adhz63 were significantly attenuated following JNK1/2 siRNA transfection. However the oncolytic effects and replication of the E1b-19K deleted Ad Adhz60 were not altered by JNK1/2 siRNA transfection, further implicating the crucial role of E1b-19K for Ad oncolysis and replication via JNK phosphorylation. This study has demonstrated for the first time that JNK is an intriguing molecular marker associated with enhanced Ad virotherapy efficacy, influencing future Ad vector design.


Subject(s)
Adenoviridae , Genetic Vectors , JNK Mitogen-Activated Protein Kinases/metabolism , Neoplasms/metabolism , Oncolytic Virotherapy , Oncolytic Viruses , Virus Replication , Adenoviridae/genetics , Adenovirus E1B Proteins/genetics , Adenovirus E1B Proteins/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Expression , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Host-Pathogen Interactions , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Mice , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Phosphorylation , Xenograft Model Antitumor Assays
2.
Viruses ; 8(12)2016 12 20.
Article in English | MEDLINE | ID: mdl-27999391

ABSTRACT

Adenoviruses (Ads) have been extensively manipulated for the development of cancer selective replication, leading to cancer cell death or oncolysis. Clinical studies using E1-modified oncolytic Ads have shown that this therapeutic platform was safe, but with limited efficacy, indicating the necessity of targeting other viral genes for manipulation. To improve the therapeutic efficacy of oncolytic Ads, we treated the entire Ad genome repeatedly with UV-light and have isolated AdUV which efficiently lyses cancer cells as reported previously (Wechman, S. L. et al. Development of an Oncolytic Adenovirus with Enhanced Spread Ability through Repeated UV Irradiation and Cancer Selection. Viruses2016, 8, 6). In this report, we show that no mutations were observed in the early genes (E1 or E4) of AdUV while several mutations were observed within the Ad late genes which have structural or viral DNA packaging functions. This study also reported the increased release of AdUV from cancer cells. In this study, we found that AdUV inhibits tumor growth following intratumoral injection. These results indicate the potentially significant role of the viral late genes, in particular the DNA packaging genes, to enhance Ad oncolysis.


Subject(s)
Adenoviridae/genetics , Adenoviridae/physiology , DNA Packaging , Mutation , Oncolytic Viruses/genetics , Oncolytic Viruses/physiology , Virus Release , Adenoviridae/radiation effects , DNA Mutational Analysis , Oncolytic Viruses/radiation effects , Ultraviolet Rays , Viral Proteins/genetics
3.
Viruses ; 8(6)2016 06 14.
Article in English | MEDLINE | ID: mdl-27314377

ABSTRACT

Oncolytic adenoviruses (Ads) have been shown to be safe and have great potential for the treatment of solid tumors. However, the therapeutic efficacy of Ads is antagonized by limited spread within solid tumors. To develop Ads with enhanced spread, viral particles of an E1-wildtype Ad5 dl309 was repeatedly treated with UV type C irradiation and selected for the efficient replication and release from cancer cells. After 72 cycles of treatment and cancer selection, AdUV was isolated. This vector has displayed many favorable characteristics for oncolytic therapy. AdUV was shown to lyse cancer cells more effectively than both E1-deleted and E1-wildtype Ads. This enhanced cancer cell lysis appeared to be related to increased AdUV replication in and release from infected cancer cells. AdUV-treated A549 cells displayed greater expression of the autophagy marker LC3-II during oncolysis and formed larger viral plaques upon cancer cell monolayers, indicating increased virus spread among cancer cells. This study indicates the potential of this approach of irradiation of entire viral particles for the development of oncolytic viruses with designated therapeutic properties.


Subject(s)
Adenoviridae/growth & development , Adenoviridae/isolation & purification , Oncolytic Viruses/growth & development , Oncolytic Viruses/isolation & purification , Serial Passage , Ultraviolet Rays , Adenoviridae/genetics , Adenoviridae/radiation effects , Cell Line, Tumor , Cell Survival , Humans , Oncolytic Viruses/genetics , Oncolytic Viruses/radiation effects , Viral Plaque Assay
4.
BMC Cancer ; 15: 716, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26475304

ABSTRACT

BACKGROUND: Clinical trials have indicated that preclinical results obtained with human tumor xenografts in mouse models may overstate the potential of adenovirus (Ad)-mediated oncolytic therapies. We have previously demonstrated that the replication of human Ads depends on cyclin E dysregulation or overexpression in cancer cells. ED-1 cell derived from mouse lung adenocarcinomas triggered by transgenic overexpression of human cyclin E may be applied to investigate the antitumor efficacy of oncolytic Ads. METHODS: Ad-cycE was used to target cyclin E overexpression in ED-1 cells and repress tumor growth in a syngeneic mouse model for investigation of oncolytic virotherapies. RESULTS: Murine ED-1 cells were permissive for human Ad replication and Ad-cycE repressed ED-1 tumor growth in immunocompetent FVB mice. ED-1 cells destroyed by oncolytic Ads in tumors were encircled in capsule-like structures, while cells outside the capsules were not infected and survived the treatment. CONCLUSION: Ad-cycE can target cyclin E overexpression in cancer cells and repress tumor growth in syngeneic mouse models. The capsule structures formed after Ad intratumoral injection may prevent viral particles from spreading to the entire tumor.


Subject(s)
Adenocarcinoma/therapy , Cyclin E/biosynthesis , Lung Neoplasms/therapy , Oncolytic Virotherapy , Adenocarcinoma/genetics , Adenoviridae/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin E/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Isografts , Lung Neoplasms/genetics , Mice , Oncolytic Viruses , Xenograft Model Antitumor Assays
5.
J Mol Med (Berl) ; 93(2): 211-23, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25376708

ABSTRACT

Oncolytic virotherapy can selectively destroy cancer cells and is a potential approach in cancer treatment. A strategy to increase tumor-specific selectivity is to control the expression of a key regulatory viral gene with a tumor-specific promoter. We have previously found that cyclin E expression is augmented in cancer cells after adenovirus (Ad) infection. Thus, the cyclin E promoter that is further activated by Ad in cancer cells may have unique properties for enhancing oncolytic viral replication. We have shown that high levels of viral E1a gene expression are achieved in cancer cells infected with Ad-cycE, in which the endogenous Ad E1a promoter was replaced with the cyclin E promoter. Ad-cycE shows markedly selective oncolytic efficacy in vitro and destroys various types of cancer cells, including those resistant to ONYX-015/dl1520. Furthermore, Ad-cycE shows a strong capacity to repress A549 xenograft tumor growth in nude mice and significantly prolongs survival. This study suggests the potential of Ad-cycE in cancer therapy and indicates the advantages of using promoters that can be upregulated by virus infection in cancer cells in development of oncolytic viruses. Key messages: Cyclin E promoter activity is high in cancer cells and enhanced by adenovirus infection. Cyclin E promoter is used to control the E1a gene of a tumor-specific oncolytic adenovirus. Ad-cycE efficiently targets cancer cells and induces oncolysis. Ad-cycE significantly repressed xenograft tumor and prolonged survival.


Subject(s)
Adenoviridae/genetics , Cyclin E/genetics , Genetic Vectors/genetics , Neoplasms/genetics , Oncolytic Viruses/genetics , Promoter Regions, Genetic , Animals , Autophagy/genetics , Cell Line, Tumor , Cell Survival/genetics , Disease Models, Animal , Female , Gene Expression , Genes, Reporter , Humans , Mice , Neoplasms/mortality , Neoplasms/pathology , Neoplasms/therapy , Oncolytic Virotherapy , Tumor Burden , Xenograft Model Antitumor Assays
6.
Cancer Biol Ther ; 15(9): 1256-67, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24972095

ABSTRACT

Epidemiological studies suggest that high intake of cruciferous vegetables is associated with a lower risk of cancer. Experiments have shown that indole-3-carbinol (I3C), a naturally occurring compound derived from cruciferous vegetables, exhibits potent anticarcinogenic properties in a wide range of cancers. In this study, we showed that higher doses of I3C (≥400 µM) induced apoptotic cancer cell death and lower doses of I3C (≤200 µM) repressed cancer cell growth concurrently with suppressed expression of cyclin E and its partner CDK2. Notably, we found that pretreatment with low doses of I3C enhanced Ad-mediated oncolysis and cytotoxicity of human carcinoma cells by synergistic upregulation of apoptosis. Thus, the vegetable compound I3C as a dietary supplement may benefit cancer prevention and improve Ad oncolytic therapies.


Subject(s)
Adenoviridae , Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Indoles/pharmacology , Oncolytic Viruses , Vegetables/chemistry , Cell Cycle Checkpoints , Cell Line, Tumor , Cyclin E/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Drug Synergism , Humans
7.
Virol J ; 10: 293, 2013 Sep 23.
Article in English | MEDLINE | ID: mdl-24059864

ABSTRACT

BACKGROUND: Combination of oncolytic adenoviruses (Ads) and chemotherapy drugs has shown promising therapeutic results and is considered as a potential approach for cancer therapy. We previously have shown that autophagy may generate decomposed cellular molecules that can be used as nutrition to support virus replication in cancer cells. In this study, we evaluated a unique combination of the novel oncolytic Ad-cycE with rapamycin, an autophagy inducer and first-line chemotherapeutic drug. METHODS: The combination of oncolytic Ad-cycE and the autophagy inducer rapamycin was assessed for enhanced antitumor effect. We also evaluated the combined effects of rapamycin and Ad-cycE on cancer cell viability. The interaction between Ad-cycE and rapamycin was analyzed with Calcusyn (Biosoft, Ferguson, MO). RESULTS: We show that rapamycin induces autophagy, enhances Ad E1A expression and increases Ad oncolytic replication. Combination of rapamycin and Ad-cycE elicits stronger cytotoxicity than single treatment alone. The analyzed data indicates that the Ad-cycE and rapamycin combination has a significantly synergistic antitumor effect. CONCLUSIONS: Our study provides a new insight into vector development and demonstrates the novel roles of autophagy in adenovirus replication. The combination of autophagy-induced chemotherapy and oncolytic virotherapy may be a new approach to improve future cancer treatment.


Subject(s)
Adenoviridae/growth & development , Autophagy , Oncolytic Viruses/growth & development , Sirolimus/metabolism , Adenoviridae/physiology , Cell Line , Cell Survival , Humans , Viral Load , Virus Replication
8.
PLoS One ; 8(2): e57340, 2013.
Article in English | MEDLINE | ID: mdl-23437375

ABSTRACT

Adenoviruses (Ads) with deletion of E1b55K preferentially replicate in cancer cells and have been used in cancer therapies. We have previously shown that Ad E1B55K protein is involved in induction of cyclin E for Ad replication, but this E1B55K function is not required in cancer cells in which deregulation of cyclin E is frequently observed. In this study, we investigated the interaction of cyclin E and CDK2 in Ad-infected cells. Ad infection significantly increased the large form of cyclin E (cyclin EL), promoted cyclin E/CDK2 complex formation and increased CDK2 phosphorylation at the T160 site. Activated CDK2 caused pRb phosphorylation at the S612 site. Repression of CDK2 activity with the chemical inhibitor roscovitine or with specific small interfering RNAs significantly decreased pRb phosphorylation, with concomitant repression of viral replication. Our results suggest that Ad-induced cyclin E activates CDK2 that targets the transcriptional repressor pRb to generate a cellular environment for viral productive replication. This study reveals a new molecular basis for oncolytic replication of E1b-deleted Ads and will aid in the development of new strategies for Ad oncolytic virotherapies.


Subject(s)
Adenoviridae/genetics , Cyclin E/genetics , Cyclin-Dependent Kinase 2/genetics , Gene Expression Regulation, Neoplastic/drug effects , Oncogene Proteins/genetics , Retinoblastoma Protein/genetics , Viral Proteins/genetics , Adenoviridae/metabolism , Cell Line, Tumor , Cyclin E/agonists , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/virology , Genes, Reporter , Green Fluorescent Proteins , HEK293 Cells , Host-Pathogen Interactions , Humans , Oncogene Proteins/agonists , Oncogene Proteins/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , RNA, Small Interfering/genetics , Retinoblastoma Protein/antagonists & inhibitors , Retinoblastoma Protein/metabolism , Roscovitine , Viral Proteins/metabolism , Virus Replication/drug effects
9.
Virology ; 433(2): 538-47, 2012 Nov 25.
Article in English | MEDLINE | ID: mdl-23021422

ABSTRACT

Adenovirus-mediated gene transfer into a tumor mass can be improved by combining it with conditionally-replicating adenovirus (CRAd) when both vectors co-infect the same cancer cell. We investigated the efficiency of enhancing transgene expression and effectiveness of cancer killing of two advenoviruses (Ads), one expressing E2F-1 (AdE2F-1) and another expressing a truncated form of E2F-1 that lacks the transactivation domain (AdE2Ftr), when combined with oncolytic Adhz60. We found that AdE2F-1 with Adhz60 actually decreased E2F-1 expression and viral replication through a mechanism apparently involving repression of the cyclin-E promoter and decreased expression of early and late structural proteins necessary for viral replication. In contrast, AdE2Ftr with Adhz60 resulted in increased E2Ftr expression, AdE2Ftr replication, and cancer cell death both in vitro and in vivo. These results indicate that AdE2Ftr coupled with a CRAd enhances AdE2Ftr-mediated cancer cell death.


Subject(s)
Adenoviridae/genetics , E2F1 Transcription Factor/genetics , Neoplasms/therapy , Oncolytic Viruses/genetics , Adenoviridae/physiology , Adenovirus E1A Proteins/metabolism , Animals , Cell Death , Cell Line, Tumor , Cyclin E/genetics , Cyclin E/metabolism , Down-Regulation , Gene Expression , Humans , Mice , Mice, Nude , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Promoter Regions, Genetic , Transduction, Genetic , Virus Replication , Xenograft Model Antitumor Assays
10.
J Cell Mol Med ; 16(3): 605-15, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21564512

ABSTRACT

E2F-1-deleted mutant, 'truncated E2F' (E2Ftr, E2F-1[1-375]), lacking the carboxy-terminal transactivation domain, was shown to be more potent at inducing cancer cell apoptosis than wild-type E2F-1 (wtE2F-1; full-length E2F-1). Mechanisms by which wtE2F-1 and E2Ftr induce apoptosis, however, are not fully elucidated. Our study demonstrates molecular effects of pro-apoptotic BH3-only Bcl-2 family member Harakiri (Hrk) in wtE2F-1- and E2Ftr-induced melanoma cell apoptosis. We found that Hrk mRNA and Harakiri (HRK) protein expression was highly up-regulated in melanoma cells in response to wtE2F-1 and E2Ftr overexpression. HRK up-regulation did not require the E2F-1 transactivation domain. In addition, Hrk gene up-regulation and HRK protein expression did not require p53 in cancer cells. Hrk knockdown by Hrk siRNA was associated with significantly reduced wtE2F-1- and E2Ftr-induced apoptosis. We also found that an upstream factor, 'downstream regulatory element antagonist modulator' (DREAM), may be involved in HRK-mediated apoptosis in response to wtE2F-1 and E2Ftr overexpression. DREAM expression levels increased following wtE2F-1 and E2Ftr overexpression. Western blotting detected increased DREAM primarily in dimeric form. The homodimerization of DREAM resulting from wtE2F-1 and E2Ftr overexpression may contribute to the decreased binding activity of DREAM to the 3'-untranslated region of the Hrk gene as shown by electromobility shift assay. Results showed wtE2F-1- and E2Ftr-induced apoptosis is partially mediated by HRK. HRK function is regulated in response to DREAM. Our findings contribute to understanding the mechanisms that regulate wtE2F-1- and E2Ftr-induced apoptosis and provide insights into the further evaluation of how E2Ftr-induced apoptosis may be used for therapeutic gain.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/genetics , E2F1 Transcription Factor/metabolism , Kv Channel-Interacting Proteins/metabolism , Repressor Proteins/metabolism , 3' Untranslated Regions , Apoptosis Regulatory Proteins/genetics , Binding Sites , Cell Line, Tumor , E2F1 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Kv Channel-Interacting Proteins/genetics , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , RNA, Small Interfering/genetics , Repressor Proteins/genetics , Sequence Deletion , Signal Transduction/genetics , Transfection
11.
Virology ; 416(1-2): 9-15, 2011 Jul 20.
Article in English | MEDLINE | ID: mdl-21575980

ABSTRACT

Adenoviruses with deletion of E1b have been used in clinical trials to treat cancers that are resistant to conventional therapies. The efficacy of viral replication within cancer cells determines the results of oncolytic therapy, which remains poorly understood and requires further improvement. In this report, we show that adenoviruses induce autophagy by increasing the conversion of LC3-I to LC3-II and the formation of the Atg12-Atg5 complex. Inhibition of autophagy with 3-methyladenine (3MA) resulted in a decreased synthesis of adenovirus structural proteins, and thereby a poor viral replication; promotion of autophagy with rapamycin increased adenovirus yield. This study indicates that adenovirus-induced autophagy correlates positively with virus replication and oncolytic cell death, and that autophagy may generate nutrients that can be used for building viral progeny particles. These results further suggest that chemotherapeutic agents that increase cancer cell autophagy may improve the efficacy of oncolytic virotherapy.


Subject(s)
Adenoviridae/physiology , Autophagy/physiology , Virus Replication/physiology , Adenine/analogs & derivatives , Adenine/pharmacology , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Adenovirus E1B Proteins/genetics , Adenovirus E1B Proteins/metabolism , Autophagy-Related Protein 12 , Autophagy-Related Protein 5 , Cell Line, Tumor , Gene Expression Regulation, Viral , Humans , Microtubule-Associated Proteins/metabolism , Mutation , Small Ubiquitin-Related Modifier Proteins/metabolism
12.
Cancer ; 116(18): 4420-32, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20549818

ABSTRACT

BACKGROUND: Adenovirus (Ad)-mediated E2F-1 gene transfer induces apoptosis in cancer cells in vitro and in vivo, but clinical application of E2F-1 in cancer gene therapy remains controversial because of the oncogenic potential of E2F-1. This barrier can be circumvented by using the truncated form of the E2F-1 gene (E2Ftr) (amino acids 1 through 375), which lacks the E2F-1 transactivation domain and cell cycle-promoting effects. METHODS: The authors constructed 3 adenoviral vectors that expressed E2Ftr under regulation of the tetracycline (Tet)-off system (AdTet-E2Ftr1, AdTet-E2Ftr2, and AdTet-E2Ftr3). These vectors were compared for E2Ftr expression and apoptosis induction in cancer cells and normal cells. E2Ftr antitumor activity in vivo also was assessed in a melanoma xenograft model. RESULTS: One of the 3 vectors, AdTet-E2Ftr3, had the highest E2Ftr protein expression levels, which were correlated with the greatest induction of apoptosis and inhibition of cancer cell growth. E2Ftr induced apoptosis in a variety of cancer cell lines independent of p53 status with little cytotoxicity in normal cell lines. In a mouse melanoma xenograft model, AdTet-E2Ftr3 exhibited an approximately 80% decrease in tumor size compared with controls in vivo. CONCLUSIONS: The current results indicated that AdTet-E2Ftr3 is a novel anticancer agent that has significant therapeutic activity in vitro and in vivo.


Subject(s)
Adenoviridae/genetics , E2F1 Transcription Factor/genetics , Genetic Therapy/methods , Genetic Vectors , Neoplasms/therapy , Animals , Apoptosis , Cell Line, Tumor , E2F1 Transcription Factor/chemistry , Genes, p53 , Mice , Mice, Inbred BALB C , Sequence Deletion , Xenograft Model Antitumor Assays
13.
Virology ; 397(2): 337-45, 2010 Feb 20.
Article in English | MEDLINE | ID: mdl-20003994

ABSTRACT

Adenoviral vectors are highly efficient at transferring genes into cells and are broadly used in cancer gene therapy. However, many therapeutic genes are toxic to vector host cells and thus inhibit vector production. The truncated form of E2F-1 (E2Ftr), which lacks the transactivation domain, can significantly induce cancer cell apoptosis, but is also toxic to HEK-293 cells and inhibits adenovirus replication. To overcome this, we have developed binary- and single-vector systems with a modified tetracycline-off inducible promoter to control E2Ftr expression. We compared several vectors and found that the structure of expression cassettes in vectors significantly affects E2Ftr expression. One construct expresses high levels of inducible E2Ftr and efficiently causes apoptotic cancer cell death by activation of caspase-3. The approach developed in this study may be applied in other viral vectors for encoding therapeutic genes that are toxic to their host cells and/or inhibit vector propagation.


Subject(s)
Adenoviridae/genetics , Adenovirus E2 Proteins/genetics , Gene Expression , Genetic Vectors , Recombinant Proteins/genetics , Recombinant Proteins/toxicity , Adenoviridae/growth & development , Cell Culture Techniques/methods , Cell Line , Genetic Therapy/methods , Humans , Transcriptional Activation
14.
J Virol ; 82(7): 3415-27, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18234796

ABSTRACT

Adenoviruses (Ads) with E1B55K mutations can selectively replicate in and destroy cancer cells. However, the mechanism of Ad-selective replication in tumor cells is not well characterized. We have shown previously that expression of several cell cycle-regulating genes is markedly affected by the Ad E1b gene in WI-38 human lung fibroblast cells (X. Rao, et al., Virology 350:418-428, 2006). In the current study, we show that the Ad E1B55K region is required to enhance cyclin E expression and that the failure to induce cyclin E overexpression due to E1B55K mutations prevents viral DNA from undergoing efficient replication in WI-38 cells, especially when the cells are arrested in the G(0) phase of the cell cycle by serum starvation. In contrast, cyclin E induction is less dependent on the function encoded in the E1B55K region in A549 and other cancer cells that are permissive for replication of E1B55K-mutated viruses, whether the cells are in the S phase or G(0) phase. The small interfering RNA that specifically inhibits cyclin E expression partially decreased viral replication. Our study provides evidence suggesting that E1B55K may be involved in cell cycle regulation that is important for efficient viral DNA replication and that cyclin E overexpression in cancer cells may be associated with the oncolytic replication of E1B55K-mutated viruses.


Subject(s)
Adenoviridae/physiology , Adenovirus E1B Proteins/physiology , Cyclin E/biosynthesis , DNA, Viral/biosynthesis , Virus Replication/physiology , Adenoviridae/genetics , Adenovirus E1B Proteins/genetics , Blotting, Western , Cell Line , Fibroblasts/virology , Gene Silencing , Humans , RNA, Small Interfering/genetics , Virus Replication/genetics
15.
Virology ; 350(2): 418-28, 2006 Jul 05.
Article in English | MEDLINE | ID: mdl-16542696

ABSTRACT

Adenoviruses with deletion of E1b gene can selectively replicate in cancer cells. The underlying mechanisms in tumor-selective replication of E1b-deleted adenoviruses are insufficiently understood. Identifying genes with altered expression patterns caused by the E1B proteins in virus-infected cells will further increase our understanding of E1B functions and provide insight into the tumor-selective replication of E1b-mutated adenoviruses on the molecular level. An approach based on large-scale gene array was applied to analyze molecular changes affected by viral E1B. We identified a total of 345 genes with expression changes of two-fold or greater affected by wild-type adenovirus compared with its E1b-deleted counterpart. The gene array data were confirmed by quantitative real-time PCR and Western blot. E1B proteins affect the expression of a diverse range of genes involved in cell cycle regulation, apoptosis, stress responses and angiogenesis. This is the first study of the global profile of gene expression altered by the viral E1B proteins in human lung cells, and the majority of the genes were previously not known to be affected by the viral proteins. The data presented in this study will lead to more detailed analysis of E1B functions and may also lead to development of new agents and approaches for oncolytic therapy.


Subject(s)
Adenovirus E1B Proteins/genetics , Gene Expression Profiling , Lung/virology , Enzymes/genetics , Gene Expression Regulation , Humans , Nucleic Acid Hybridization , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics
16.
Cancer Biol Ther ; 4(11): 1255-62, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16222119

ABSTRACT

One of the promising strategies for targeting replication of oncolytic adenovirus in tumor cells is to regulate the expression of essential viral genes such as E1a by using tumor- or tissue-specific promoters that are preferentially active in cancer cells. However, this approach may lead to some degree of viral replication in normal cells other than in cancer cells if the viral gene also expresses in normal cells. In this study, we investigated the effect of E1a expression levels on the virus replication ability in human cells. Three vectors, all with mutated E1B55K, were created, one without any promoter controlling the E1a gene and two vectors with the E1a gene being controlled by either its endogenous promoter or a strong CMV promoter. We observed that the CMV promoter-mediated high levels of E1A expression could increase virus replication, resulting in the titers of the E1B55K-mutated virus being even higher than the wild-type virus in some cancer cells. However, the strong CMV promoter could not always enhance virus replication, such as in cancer cells OE33 and OsACL. The results suggest that whether increased E1A levels would enhance E1B55K-mutated virus replication may be also depended on cellular factors or pathways in cancer cells. We also observed that the virus without any promoter for the E1a gene could still express leaky levels of E1A which can lead to viral replication in normal and cancer cells. Future efforts in the development of transcription-controlled oncolytic adenoviruses should focus on how to completely block E1a expression in normal cells.


Subject(s)
Adenoviridae/physiology , Adenovirus E1A Proteins/metabolism , Neoplasms/therapy , Oncolytic Viruses/physiology , Virus Replication/genetics , Adenoviridae/genetics , Adenovirus E1A Proteins/genetics , Cell Line , Cell Line, Tumor , Genetic Vectors/genetics , Genetic Vectors/physiology , Humans , Mutation , Neoplasms/classification , Neoplasms/pathology , Promoter Regions, Genetic
17.
Breast Cancer Res ; 7(5): R580-8, 2005.
Article in English | MEDLINE | ID: mdl-16168101

ABSTRACT

INTRODUCTION: Once metastasis has occurred, the possibility of completely curing breast cancer is unlikely, particularly for the 30 to 40% of cancers overexpressing the gene for HER2/neu. A vaccine targeting p185, the protein product of the HER2/neu gene, could have therapeutic application by controlling the growth and metastasis of highly aggressive HER2/neu+ cells. The purpose of this study was to determine the effectiveness of two gene vaccines targeting HER2/neu in preventive and therapeutic tumor models. METHODS: The mouse breast cancer cell line A2L2, which expresses the gene for rat HER2/neu and hence p185, was injected into the mammary fat pad of mice as a model of solid tumor growth or was injected intravenously as a model of lung metastasis. SINCP-neu, a plasmid containing Sindbis virus genes and the gene for rat HER2/neu, and Adeno-neu, an E1,E2a-deleted adenovirus also containing the gene for rat HER2/neu, were tested as preventive and therapeutic vaccines. RESULTS: Vaccination with SINCP-neu or Adeno-neu before tumor challenge with A2L2 cells significantly inhibited the growth of the cells injected into the mammary fat or intravenously. Vaccination 2 days after tumor challenge with either vaccine was ineffective in both tumor models. However, therapeutic vaccination in a prime-boost protocol with SINCP-neu followed by Adeno-neu significantly prolonged the overall survival rate of mice injected intravenously with the tumor cells. Naive mice vaccinated using the same prime-boost protocol demonstrated a strong serum immunoglobulin G response and p185-specific cellular immunity, as shown by the results of ELISPOT (enzyme-linked immunospot) analysis for IFNgamma. CONCLUSION: We report herein that vaccination of mice with a plasmid gene vaccine and an adenovirus gene vaccine, each containing the gene for HER2/neu, prevented growth of a HER2/neu-expressing breast cancer cell line injected into the mammary fat pad or intravenously. Sequential administration of the vaccines in a prime-boost protocol was therapeutically effective when tumor cells were injected intravenously before the vaccination. The vaccines induced high levels of both cellular and humoral immunity as determined by in vitro assessment. These findings indicate that clinical evaluation of these vaccines, particularly when used sequentially in a prime-boost protocol, is justified.


Subject(s)
Cancer Vaccines , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/immunology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Animals , Cell Line, Tumor , Female , Genes, erbB-2/immunology , Immunization, Secondary , Interferon-gamma/biosynthesis , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasm Metastasis/immunology , Neoplasm Metastasis/prevention & control , Rats , Sindbis Virus/immunology , Spleen/immunology
18.
Cancer Gene Ther ; 11(9): 585-93, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15338010

ABSTRACT

Apoptotic pathways are initiated as a cellular defense mechanism to eliminate adenovirus-infected cells. We have investigated how E1A-induced apoptosis interferes with viral replication in cancer cells. We found that E1B19K alone can efficiently suppress E1A-induced apoptosis in cancer cells. Viruses deleted for both E1B19K and E1B55K resulted in cellular DNA degradation. However, less than 20% of human lung cancer cells infected with a virus deleted for both E1B19K and E1B55 K had evidence of chromatin condensation and multiple-micronuclei formation (apoptotic hallmarks); these cells could not produce infectious viral particles. The majority of cancer cells infected with viruses deleted for the entire E1b gene did not undergo extended apoptosis and produced abundant viral progeny. Thus, only a fraction of cancer cells underwent apoptosis and did not allow E1b-deleted viruses to replicate, while the majority of cancer cells were resistant to E1A-induced apoptosis and could support virus-selective replication. The results of this study imply that, in addition to inhibiting E1A-induced apoptosis, E1B proteins may contribute other important roles in the viral life cycle. Our results also suggest that combining virus-induced apoptosis and selective viral replication into one vector will be a novel approach to destroy cancer cells.


Subject(s)
Adenoviridae Infections/virology , Adenovirus E1A Proteins/physiology , Adenovirus E1B Proteins/genetics , Adenoviruses, Human/physiology , Apoptosis , Gene Deletion , Lung Neoplasms/virology , Virus Replication , Adenoviridae Infections/pathology , Blotting, Western , Chromatin/metabolism , DNA, Viral/genetics , DNA, Viral/metabolism , Genetic Vectors , Humans , Lung Neoplasms/pathology , Micronuclei, Chromosome-Defective , Tumor Cells, Cultured
19.
Cancer Res ; 63(12): 3073-8, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12810631

ABSTRACT

The adenovirus E1A proteins are involved in the transcriptional activation of viral and cellular genes needed for controlling cell cycle and virus replication. Undifferentiated embryonic carcinoma cells have the ability to produce an E1A-like activity that can induce the expression of E1A-targeted adenoviral and cellular genes in the absence of the E1A products. Differentiated embryonic carcinoma cells lose the ability to produce the E1A-like activity. In this study, we investigated the E1A-like activity in cancer cells with an adenovirus having a mutated E1a gene. The mutation is generated by the insertion of a large DNA fragment in the E1a gene and interrupts the COOH-terminal region of both the E1A 12S and 13S proteins. The E1a-mutated virus can efficiently replicate in HepG2 and Hep3B liver cancer cells and produce high titers of virus. Replication of the E1a-mutated virus inhibits tumor formation and destroys tumors in vivo. The results obtained in this study imply that cancer cells may produce an E1A-like activity to support the selective replication of mutated virus in cancer cells. In addition, we found that although the E1a-mutated virus could not replicate in Huh1.cl2 liver cells, the viral DNA could amplify in the cells. This result suggests that replication of adenoviral DNA is necessary, but not sufficient, for generating infectious viral progeny and destroying tumor cells.


Subject(s)
Adenovirus E1A Proteins/genetics , Biological Therapy , Carcinoma, Hepatocellular/pathology , Defective Viruses/physiology , Liver Neoplasms/pathology , Mastadenovirus/physiology , Adenovirus E1A Proteins/deficiency , Adenovirus E1A Proteins/physiology , Animals , Carcinoma, Hepatocellular/therapy , Cytopathogenic Effect, Viral , DNA, Viral/genetics , Defective Viruses/genetics , Humans , Liver Neoplasms/therapy , Liver Neoplasms/virology , Mastadenovirus/genetics , Mice , Mice, Nude , Mutagenesis, Insertional , Tumor Cells, Cultured/virology , Virus Integration , Virus Replication , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...