1
|
Oncolytic Adenovirus in Cancer Immunotherapy. Cancers (Basel) 2020; 12:cancers12113354. [PMID: 33202717 PMCID: PMC7697649 DOI: 10.3390/cancers12113354] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023] Open
Abstract
Tumor-selective replicating "oncolytic" viruses are novel and promising tools for immunotherapy of cancer. However, despite their first success in clinical trials, previous experience suggests that currently used oncolytic virus monotherapies will not be effective enough to achieve complete tumor responses and long-term cure in a broad spectrum of cancers. Nevertheless, there are reasonable arguments that suggest advanced oncolytic viruses will play an essential role as enablers of multi-stage immunotherapies including established systemic immunotherapies. Oncolytic adenoviruses (oAds) display several features to meet this therapeutic need. oAds potently lyse infected tumor cells and induce a strong immunogenic cell death associated with tumor inflammation and induction of antitumor immune responses. Furthermore, established and versatile platforms of oAds exist, which are well suited for the incorporation of heterologous genes to optimally exploit and amplify the immunostimulatory effect of viral oncolysis. A considerable spectrum of functional genes has already been integrated in oAds to optimize particular aspects of immune stimulation including antigen presentation, T cell priming, engagement of additional effector functions, and interference with immunosuppression. These advanced concepts have the potential to play a promising future role as enablers of multi-stage immunotherapies involving adoptive cell transfer and systemic immunotherapies.
Collapse
|
2
|
In vitro comparison of currently available and investigational antiviral agents against pathogenic human double-stranded DNA viruses: A systematic literature review. Antiviral Res 2019; 163:50-58. [PMID: 30677427 DOI: 10.1016/j.antiviral.2019.01.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/10/2019] [Accepted: 01/16/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Double-stranded (ds) DNA virus infections often occur concomitantly in immunocompromised patients. We performed a systematic search of published in vitro activity for nine approved and investigational antivirals to understand the spectrum of in vitro activity against dsDNA viruses. METHODS A literature search was performed (PubMed and the WoS Core Collection) using keywords related to: 1) targeted approved/developmental antivirals (acyclovir, artesunate, brincidofovir, cidofovir, cyclopropavir (filociclovir), foscarnet, ganciclovir, letermovir, and maribavir); 2) pathogenic dsDNA viruses; 3) in vitro activity. We summarized data from 210 publications. RESULTS Activity against ≤3 viruses was documented for maribavir (cytomegalovirus, Epstein-Barr virus), and letermovir, while activity against > 3 viruses was shown for ganciclovir, cidofovir, acyclovir, foscarnet, cyclopropavir, artesunate, and brincidofovir. The EC50 values of brincidofovir were the lowest, ranging from 0.001 to 0.27 μM, for all viruses except papillomaviruses. The next most potent agents included cidofovir, ganciclovir, foscarnet, and acyclovir with EC50 values between 0.1 μM and >10 μM for cytomegalovirus, herpes simplex virus, and adenovirus. CONCLUSION Most of the identified antivirals had in vitro activity against more than one dsDNA virus. Brincidofovir and cidofovir have broad-spectrum activity, and brincidofovir has the lowest EC50 values. These findings could assist clinical practice and developmental research.
Collapse
|
3
|
An armed, YB-1-dependent oncolytic adenovirus as a candidate for a combinatorial anti-glioma approach of virotherapy, suicide gene therapy and chemotherapeutic treatment. Cancer Gene Ther 2014; 22:30-43. [PMID: 25501992 DOI: 10.1038/cgt.2014.67] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 11/18/2014] [Accepted: 11/18/2014] [Indexed: 12/27/2022]
Abstract
We investigated the novel recombinant oncolytic adenovirus Ad-delo-sr39TK-RGD, armed with a mutant herpes simplex virus type 1 thymidine kinase (HSV1-sr39TK) as a suicide gene, and explored its antitumor efficacy in combination with HSV1-sr39TK/ganciclovir (GCV) gene therapy and temozolomide (TMZ). Ad-delo-sr39TK-RGD is an E1-mutated conditionally replicating adenovirus dependent on the human Y-box binding protein 1 (YB-1). Thus, we utilized the YB-1 dependency of the vector to target human glioma cells in vitro, using two-dimensional cell culture and three-dimensional multicellular spheroids, and demonstrated the strong replication competence and oncolytic potential of the virus. The cytotoxicity mediated by HSV1-sr39TK and its prodrug GCV enhanced the oncolytic effect even at <0.1 μg ml(-1) GCV and induced cell killing of > 95% after adding GCV 0-1 days following infection. An increased bystander effect of viral replication and GCV in co-cultured infected and uninfected cells was observed. Co-administrating Ad-delo-sr39TK-RGD with TMZ and GCV, spheroid growth was reduced drastically. Gamma counting of infected spheroids demonstrated successful accumulation of the radiotracer (18)F-labeled 9-[4-fluoro-3-(hydroxymethyl)butyl]guanine mediated by HSV1-sr39TK. Hence, our results show that the combination of YB-1-dependent virotherapy with suicide genes and TMZ effectively induces glioma cell killing and may allow for in vivo non-invasive imaging within a limited time frame.
Collapse
|
4
|
Ganciclovir inhibits human adenovirus replication and pathogenicity in permissive immunosuppressed Syrian hamsters. Antimicrob Agents Chemother 2014; 58:7171-81. [PMID: 25224011 DOI: 10.1128/aac.03860-14] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Adenovirus infections of immunocompromised patients can develop into deadly multiorgan or systemic disease. The virus is especially threatening for pediatric allogeneic hematopoietic stem cell transplant recipients; according to some studies, 10% or more of these patients succumb to disease resulting from adenovirus infection. At present, there is no drug approved for the treatment or prevention of adenovirus infections. Compounds that are approved to treat other virus infections are used off-label to combat adenovirus, but only anecdotal evidence of the efficacy of these drugs exists. Ganciclovir, a drug approved for the treatment of herpesvirus infection, was previously reported to be effective against human adenoviruses in vitro. To model adenovirus infections in immunocompromised humans, we examined ganciclovir's efficacy in immunosuppressed Syrian hamsters intravenously infected with type 5 human adenovirus (Ad5). This animal model is permissive for Ad5 replication, and the animals develop symptoms similar to those seen in humans. We demonstrate that ganciclovir suppresses Ad5 replication in the liver of infected hamsters and that it mitigates the consequences of Ad5 infections in these animals when administered prophylactically or therapeutically. We show that ganciclovir inhibits Ad5 DNA synthesis and late gene expression. The mechanism of action for the drug is not clear; preliminary data suggest that it exerts its antiadenoviral effect by directly inhibiting the adenoviral DNA polymerase. While more extensive studies are required, we believe that ganciclovir is a promising drug candidate to treat adenovirus infections. Brincidofovir, a drug with proven activity against Ad5, was used as a positive control in the prophylactic experiment.
Collapse
|
5
|
Ibrišimović M, Nagl U, Kneidinger D, Rauch M, Lion T, Klein R. Targeted expression of herpes simplex virus thymidine kinase in adenovirus-infected cells reduces virus titers upon treatment with ganciclovir in vitro. J Gene Med 2012; 14:3-19. [PMID: 22190534 DOI: 10.1002/jgm.1638] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
BACKGROUND Adenoviruses are a frequent cause of life-threatening infections in immunocompromised patients. Available therapeutics still cannot completely prevent fatal outcomes. By contrast, herpes viruses are well treatable with prodrugs such as ganciclovir (GCV), which are selectively activated in virus-infected cells by virus-encoded thymidine kinases. This effective group of prodrugs is not applicable to adenoviruses and other DNA viruses because they lack those kinases. METHODS To render adenoviruses amenable to GCV treatment, we generated an adenoviral vector-based delivery system for targeted expression of herpes simplex virus thymidine kinase (HSV-TK) in wild-type adenovirus 5 (wt Ad5)-infected cells. HSV-TK expression was largely restricted to wt virus-infected cells by transcription of the gene from the Ad5 E4 promoter. Its activity is dependent on the adenoviral E1A gene product which is not produced by the vector but is only provided in cells infected with wt adenovirus. The anti-adenoviral effect of HSV-TK expression and concomitant treatment with GCV was assessed in vitro in four different cell lines or primary cells. RESULTS E4 promoter-mediated HSV-TK background expression was sufficiently low to prevent cytotoxicity in the presence of low-levels GCV in cells not infected with wt Ad5. However, expression was several-fold increased in wt Ad5-infected cells and treatment with low levels of GCV efficiently inhibited wt Ad5 DNA replication. Genome copy numbers and output of infectious particles were reduced by up to > 99.99% and cell viability was greatly increased. CONCLUSIONS We extended the concept of enzyme/prodrug therapy to adenovirus infections by selectively sensitizing adenovirus-infected cells to treatment with GCV.
Collapse
Affiliation(s)
- Mirza Ibrišimović
- Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Department of Pediatrics, Medical University of Vienna, Austria
| | | | | | | | | | | |
Collapse
|
6
|
Jiang G, Zhang L, Xin Y, Pei DS, Wei ZP, Liu YQ, Zheng JN. Conditionally replicating adenoviruses carrying mda-7/IL-24 for cancer therapy. Acta Oncol 2012; 51:285-92. [PMID: 21995527 DOI: 10.3109/0284186x.2011.621447] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Melanoma differentiation associated gene-7/interleukin-24 (mda-7/IL-24) suppresses growth and induces apoptosis in a broad range of human cancers without significant cytotoxicity to normal cells. Conditionally replicating adenoviruses (CRAds) not only have the ability to destroy cancer cells but may also be potential vectors for the expression of therapeutic genes. METHODS This review provides an overview of specifications for a novel anti-tumor approach CRAds carrying IL-24, and discusses recent progress in this field. RESULTS Studies in multiple laboratories report that CRAds carrying IL-24 selectively induced apoptosis in some cancer cells, and enhanced selective toxicity to cancer cells when combined with chemotherapeutic agents. CONCLUSION CRAds carrying IL-24 may prove a novel and effective approach for the treatment of cancers.
Collapse
Affiliation(s)
- Guan Jiang
- Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, China
| | | | | | | | | | | | | |
Collapse
|
7
|
Patil SS, Gentschev I, Nolte I, Ogilvie G, Szalay AA. Oncolytic virotherapy in veterinary medicine: current status and future prospects for canine patients. J Transl Med 2012; 10:3. [PMID: 22216938 PMCID: PMC3398296 DOI: 10.1186/1479-5876-10-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 01/04/2012] [Indexed: 01/14/2023] Open
Abstract
Oncolytic viruses refer to those that are able to eliminate malignancies by direct targeting and lysis of cancer cells, leaving non-cancerous tissues unharmed. Several oncolytic viruses including adenovirus strains, canine distemper virus and vaccinia virus strains have been used for canine cancer therapy in preclinical studies. However, in contrast to human studies, clinical trials with oncolytic viruses for canine cancer patients have not been reported. An 'ideal' virus has yet to be identified. This review is focused on the prospective use of oncolytic viruses in the treatment of canine tumors - a knowledge that will undoubtedly contribute to the development of oncolytic viral agents for canine cancer therapy in the future.
Collapse
Affiliation(s)
- Sandeep S Patil
- Department of Biochemistry, University of Wuerzburg, D-97074 Wuerzburg, Germany
| | | | | | | | | |
Collapse
|
8
|
Mendes G, Soares AR, Sigiliano L, Machado F, Kaiser C, Romeiro N, Gestinari L, Santos N, Romanos MTV. In vitro anti-HMPV activity of meroditerpenoids from marine alga Stypopodium zonale (Dictyotales). Molecules 2011; 16:8437-50. [PMID: 21986522 PMCID: PMC6264370 DOI: 10.3390/molecules16108437] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Revised: 09/09/2011] [Accepted: 09/29/2011] [Indexed: 11/30/2022] Open
Abstract
In this paper, we evaluated the antiviral activity against HMPV replication of crude extract of the marine algae Stypopodium zonale and of two meroditerpenoids obtained from it, atomaric acid and epitaondiol, and a methyl ester derivative of atomaric acid. Their selectivity indexes were 20.78, >56.81, 49.26 and 12.82, respectively. Compared to ribavirin, the substances showed a relatively low cytotoxicity on LLC-MK2 cells, with a significant antiviral activity, inhibiting at least 90% of viral replication in vitro, which demonstrates the potential of these marine natural products to combat infections caused by HMPV in vitro.
Collapse
Affiliation(s)
- Gabriella Mendes
- Laboratório Experimental de Drogas Antivirais e Citotóxicas (LEDAC), Departamento de Virologia do, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), CCS, Bloco I, Caixa Postal 68040, 21941-590 Rio de Janeiro, RJ, Brasil
- Laboratório de Viroses Respiratórias, Entéricas e Oculares, Departamento de Virologia do Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), CCS, Bloco I, Caixa Postal 68040, 21941-590 Rio de Janeiro, RJ, Brasil
| | - Angélica Ribeiro Soares
- Grupo de Produtos Naturais de Organismos Aquáticos (GPNOA), Núcleo em Ecologia e Desenvolvimento Socioambiental de Macaé (NUPEM), Universidade Federal do Rio de Janeiro (UFRJ), CCS, Bloco I, Caixa Postal 68040, 21941-590 Rio de Janeiro, RJ, Brasil
| | - Lorena Sigiliano
- Grupo de Produtos Naturais de Organismos Aquáticos (GPNOA), Núcleo em Ecologia e Desenvolvimento Socioambiental de Macaé (NUPEM), Universidade Federal do Rio de Janeiro (UFRJ), CCS, Bloco I, Caixa Postal 68040, 21941-590 Rio de Janeiro, RJ, Brasil
| | - Fernanda Machado
- Grupo de Produtos Naturais de Organismos Aquáticos (GPNOA), Núcleo em Ecologia e Desenvolvimento Socioambiental de Macaé (NUPEM), Universidade Federal do Rio de Janeiro (UFRJ), CCS, Bloco I, Caixa Postal 68040, 21941-590 Rio de Janeiro, RJ, Brasil
| | - Carlos Kaiser
- Instituto de Química, Universidade Federal do Rio de Janeiro (UFRJ), CCS, Bloco I, Caixa Postal 68040, 21941-590 Rio de Janeiro, RJ, Brasil
| | - Nelilma Romeiro
- Grupo de Produtos Naturais de Organismos Aquáticos (GPNOA), Núcleo em Ecologia e Desenvolvimento Socioambiental de Macaé (NUPEM), Universidade Federal do Rio de Janeiro (UFRJ), CCS, Bloco I, Caixa Postal 68040, 21941-590 Rio de Janeiro, RJ, Brasil
| | - Lísia Gestinari
- Grupo de Produtos Naturais de Organismos Aquáticos (GPNOA), Núcleo em Ecologia e Desenvolvimento Socioambiental de Macaé (NUPEM), Universidade Federal do Rio de Janeiro (UFRJ), CCS, Bloco I, Caixa Postal 68040, 21941-590 Rio de Janeiro, RJ, Brasil
| | - Norma Santos
- Laboratório de Viroses Respiratórias, Entéricas e Oculares, Departamento de Virologia do Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), CCS, Bloco I, Caixa Postal 68040, 21941-590 Rio de Janeiro, RJ, Brasil
| | - Maria Teresa Villela Romanos
- Laboratório Experimental de Drogas Antivirais e Citotóxicas (LEDAC), Departamento de Virologia do, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), CCS, Bloco I, Caixa Postal 68040, 21941-590 Rio de Janeiro, RJ, Brasil
| |
Collapse
|
9
|
Oncolytic viruses: the power of directed evolution. Adv Virol 2011; 2012:586389. [PMID: 22312363 PMCID: PMC3265225 DOI: 10.1155/2012/586389] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 05/26/2011] [Indexed: 12/17/2022] Open
Abstract
Attempts at developing oncolytic viruses have been primarily based on rational design. However, this approach has been met with limited success. An alternative approach employs directed evolution as a means of producing highly selective and potent anticancer viruses. In this method, diverse viruses are grown under conditions that maximize diversity and then passaged under conditions meant to mimic those encountered in the human cancer microenvironment. Viruses which evolve to thrive under this selective pressure are isolated and tested to identify those with increased potency (i.e., ability to replicate and spread) and/or an increased therapeutic window (i.e., differentiated replication and spread on tumor versus normal cells), both of which have potential value but the latter of which defines an oncolytic virus. Using ColoAd1, an oncolytic virus derived by this approach as a prototype, we highlight the benefits of directed evolution, discuss methods to “arm” these novel viruses, and introduce techniques for their genetic modulation and control.
Collapse
|
10
|
5-fluorouracil in lethal mutagenesis of foot-and-mouth disease virus. Future Med Chem 2011; 1:529-39. [PMID: 21426129 DOI: 10.4155/fmc.09.26] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
5-fluorouracil (FU) is a pyrimidine analogue extensively used in cancer chemotherapy. FU can be metabolized into 5-fluorouridine-triphosphate, which can be used as substrate for viral RNA-dependent RNA polymerases. This results in the incorporation of mutations into viral RNA. Accumulation of mutations may lead to loss of virus infectivity, in a process known as lethal mutagenesis. RNA virus pathogens are particularly difficult to control because they are highly mutable, and mutants resistant to antiviral agents are readily selected. Here, we review the basic principles of lethal mutagenesis as an antiviral approach, and the participation of FU in its development. Recent studies with foot-and-mouth disease virus indicate that FU can act both as an inhibitor and as a mutagen during foot-and-mouth disease virus replication. This dual activity renders FU an adequate drug for lethal mutagenesis. We suggest that structural and biochemical studies can contribute to the lead to new design of base or nucleoside analogues targeted specifically to viral polymerases.
Collapse
|
11
|
Jiang G, Xin Y, Zheng JN, Liu YQ. Combining conditionally replicating adenovirus-mediated gene therapy with chemotherapy: a novel antitumor approach. Int J Cancer 2011; 129:263-74. [PMID: 21509783 DOI: 10.1002/ijc.25948] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 01/05/2011] [Indexed: 12/16/2022]
Abstract
Despite significant improvements in diagnosis and innovations in the therapy of specific cancers, effective treatment of neoplastic diseases still presents major challenges. Recent studies have shown that conditionally replicating adenoviruses (CRAds) not only have the ability to destroy cancer cells but may also be potential vectors for the expression of therapeutic genes. Several studies in animal models have demonstrated that the combination of CRAds-mediated gene therapy and chemotherapy has greater therapeutic benefit than either treatment modality alone. In this review, an overview of specifications for a novel antitumor approach combining CRAd-gene therapy and chemotherapy is provided and recent progress in this field is discussed.
Collapse
Affiliation(s)
- Guan Jiang
- Center for Disease Control and Prevention of Xuzhou City, Xuzhou 221006, China
| | | | | | | |
Collapse
|
12
|
Zhang J, Wei F, Wang H, Li H, Qiu W, Ren P, Chen X, Huang Q. A Novel Oncolytic Adenovirus Expressing Escherichia coli Cytosine Deaminase Exhibits Potent Antitumor Effect on Human Solid Tumors. Cancer Biother Radiopharm 2010; 25:487-95. [PMID: 20735209 DOI: 10.1089/cbr.2009.0752] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Jufeng Zhang
- Experimental Center, Shanghai Jiaotong University, First People's Hospital, Shanghai, people's Republic of China
- School of Life Science, Guangdong Pharmaceutical University, Guangzhou, people's Republic of China
| | - Fang Wei
- Experimental Center, Shanghai Jiaotong University, First People's Hospital, Shanghai, people's Republic of China
| | - Huiping Wang
- Experimental Center, Shanghai Jiaotong University, First People's Hospital, Shanghai, people's Republic of China
| | - Huiming Li
- Experimental Center, Shanghai Jiaotong University, First People's Hospital, Shanghai, people's Republic of China
| | - Wei Qiu
- Experimental Center, Shanghai Jiaotong University, First People's Hospital, Shanghai, people's Republic of China
| | - Pengkang Ren
- Experimental Center, Shanghai Jiaotong University, First People's Hospital, Shanghai, people's Republic of China
| | - Xiafang Chen
- Experimental Center, Shanghai Jiaotong University, First People's Hospital, Shanghai, people's Republic of China
| | - Qian Huang
- Experimental Center, Shanghai Jiaotong University, First People's Hospital, Shanghai, people's Republic of China
| |
Collapse
|
13
|
Pei DS, Zheng JN. Oncolytic adenoviruses expressing interleukin: a novel antitumour approach. Expert Opin Biol Ther 2010; 10:917-26. [DOI: 10.1517/14712598.2010.481668] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
14
|
Gainotti R, Ricarte C, Ebekian B, Videla C, Carballal G, Damonte EB, Echavarría M. Real time PCR for rapid determination of susceptibility of adenovirus to antiviral drugs. J Virol Methods 2009; 164:30-4. [PMID: 19941904 DOI: 10.1016/j.jviromet.2009.11.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2009] [Revised: 11/15/2009] [Accepted: 11/17/2009] [Indexed: 11/28/2022]
Abstract
Human adenoviruses (HAdV) are associated with respiratory, ocular and gastrointestinal infections as well as potentially fatal disseminated disease in highly immunocompromised patients. Although there is no specific FDA approved treatment for HAdV infections, some antivirals are used in certain patients. The in vitro antiviral assays for HAdV are not standardized and are usually time consuming. The objective of this study was to evaluate a real time PCR assay for rapid determination of susceptibility of HAdV to antiviral drugs. The nucleoside analogue stavudine (d4T) was used as test drug in A549 cells infected with HAdV5. The antiviral assay measured the reduction of the HAdV DNA levels in culture supernatants by real time PCR using specific primers that amplify a conserved region of the hexon gene. This real time PCR assay demonstrated that stavudine was a selective inhibitor for HAdV5, since the effective concentration 50% (EC(50)) ranged from 0.08 to 0.12 mM at multiplicity of infection between 0.001 and 1. Furthermore, EC(50) showed a high correlation with plaque reduction and virus yield inhibition assays (r(2)=0.9938 and r(2)=0.9468, respectively). In conclusion, the real time PCR-based antiviral assay is rapid, reproducible and could replace classical and more labor-intensive infectivity assays.
Collapse
Affiliation(s)
- Rosana Gainotti
- Laboratorio de Virología Clínica, CEMIC, Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
15
|
Cody JJ, Douglas JT. Armed replicating adenoviruses for cancer virotherapy. Cancer Gene Ther 2009; 16:473-88. [PMID: 19197323 PMCID: PMC2683179 DOI: 10.1038/cgt.2009.3] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Revised: 09/15/2008] [Accepted: 10/15/2008] [Indexed: 12/22/2022]
Abstract
Conditionally replicating adenoviruses (CRAds) have many advantages as agents for cancer virotherapy and have been safely used in human clinical trials. However, replicating adenoviruses have been limited in their ability to eliminate tumors by oncolysis. Thus, the efficacy of these agents must be improved. To this end, CRAds have been engineered to express therapeutic transgenes that exert antitumor effects independent of direct viral oncolysis. These transgenes can be expressed under native gene control elements, in which case placement within the genome determines the expression profile, or they can be controlled by exogenous promoters. The therapeutic transgenes used to arm replicating adenoviruses can be broadly classified into three groups. There are those that mediate killing of the infected cell, those that modulate the tumor microenvironment and those with immunomodulatory functions. Overall, the studies to date in animal models have shown that arming a CRAd with a rationally chosen therapeutic transgene can improve its antitumor efficacy over that of an unarmed CRAd. However, a number of obstacles must be overcome before the full potential of armed CRAds can be realized in the human clinical context. Hence, strategies are being developed to permit intravenous delivery to disseminated cancer cells, overcome the immune response and enable in vivo monitoring of the biodistribution and activity of armed CRAds.
Collapse
Affiliation(s)
- James J. Cody
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics & Gynecology, Pathology and Surgery, and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - Joanne T. Douglas
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics & Gynecology, Pathology and Surgery, and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| |
Collapse
|
16
|
Bauzon M, Jin F, Kretschmer P, Hermiston T. In vitro analysis of cidofovir and genetically engineered TK expression as potential approaches for the intervention of ColoAd1-based treatment of cancer. Gene Ther 2009; 16:1169-74. [PMID: 19458647 DOI: 10.1038/gt.2009.69] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
We have generated a novel oncolytic Adenovirus (Ad), ColoAd1, with significantly increased potency ( approximately 100-fold) relative to its parent viruses, Ad11p and Ad3, or to the clinically tested oncolytic Ad, ONYX-015. Although this agent has a significant increase in its therapeutic window relative to ONYX-015 or its parent viruses, its ability to intervene and control virotherapy in treated patient is an important safety consideration for a novel biological therapy, such as ColoAd1. As there are no approved treatments for Ad infections, we sought to define whether antivirals being used to experimentally treat Ad infections (cidofovir (CDV), ribavirin) had any activity against ColoAd1. In addition, we incorporated a well-described pro-drug converting enzyme, the herpes simplex virus-thymidine kinase (HSV-TK) gene, into the viral genome to test whether the expression of this enzyme directly from the virus could be exploited as a safety valve for arresting the viral infection in the presence of the pro-drug, ganciclovir. Both the antiviral drug, CDV, and the incorporation of the pro-drug-converting TK enzyme were validated as effective approaches to controlling ColoAd1 infection, and this represents an important advancement in the development of ColoAd1 as an anticancer treatment.
Collapse
Affiliation(s)
- M Bauzon
- Bayer Healthcare Pharmaceuticals, Protein Therapeutics, Richmond, CA 94806, USA
| | | | | | | |
Collapse
|
17
|
Morfin F, Dupuis-Girod S, Frobert E, Mundweiler S, Carrington D, Sedlacek P, Bierings M, Cetkovsky P, Kroes ACM, van Tol MJD, Thouvenot D. Differential susceptibility of adenovirus clinical isolates to cidofovir and ribavirin is not related to species alone. Antivir Ther 2009. [DOI: 10.1177/135965350901400110] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background We have previously reported that human adenovirus (HAdV) reference strains clearly show species-dependent resistance to ribavirin, whereas different species of HAdV are equally sensitive to cidofovir. All the serotypes tested were susceptible to cidofovir, whereas only serotypes from species C were sensitive to ribavirin. Here, we aimed to extend these investigations to clinical isolates. Methods In vitro, we tested 126 isolates obtained from 65 patients included in a European survey of HAdV infection. Results Among the 126 isolates tested, all presented cidofovir 50% inhibitory concentration (IC50) in the same range as the HAdV 5 reference strain. Regarding ribavirin, all isolates from species C (79 tested) showed an IC50 comparable with previously reported results for reference strains; however, 24/32, 2/6 and 3/3 tested isolates from species A, B and D, respectively, were shown to have a ribavirin IC50 comparable with the HAdV 5 reference strain (species C), contrary to previous observations for reference strains of the same species. Among patients who were treated with cidofovir for disseminated HAdV infection, ≥4 sequential isolates could be obtained from 9 patients; no variation in cidofovir susceptibility could be detected. Conclusions Cidofovir is active in vitro in all HAdV clinical isolates. Ribavirin was revealed to be active on most HAdV isolates from species A, B and D, and in all isolates from species C. Finally, no resistance to cidofovir became apparent in sequential isolates obtained from treated patients.
Collapse
Affiliation(s)
- Florence Morfin
- Laboratoire de Virologie et Pathogénèse Humaine, Université Lyon 1, CNRS FRE 3011, Lyon, France
- Laboratoire de Virologie, Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Lyon, France
| | - Sophie Dupuis-Girod
- Hematology and Bone Marrow Transplantation Unit, Hôpital Debrousse, Hospices Civils de Lyon, Lyon, France
- Genetics Unit, Hôpital de l'Hôtel Dieu, Hospices Civils de Lyon, Lyon, France
| | - Emilie Frobert
- Laboratoire de Virologie et Pathogénèse Humaine, Université Lyon 1, CNRS FRE 3011, Lyon, France
| | - Stéphanie Mundweiler
- Laboratoire de Virologie et Pathogénèse Humaine, Université Lyon 1, CNRS FRE 3011, Lyon, France
- Laboratoire de Virologie, Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Lyon, France
| | | | - Petr Sedlacek
- Department of Pediatrics, Motol University Hospital, Prague, Czech Republic
| | - Marc Bierings
- Wilhelmina Kinderziekenhuis Hematology, Utrecht, the Netherlands
| | - Petr Cetkovsky
- Institute of Hematology and Blood Transfusion, UHKT, Prague, Czech Republic
| | - Aloys CM Kroes
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Maarten JD van Tol
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Danielle Thouvenot
- Laboratoire de Virologie et Pathogénèse Humaine, Université Lyon 1, CNRS FRE 3011, Lyon, France
- Laboratoire de Virologie, Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
18
|
Lenaerts L, De Clercq E, Naesens L. Clinical features and treatment of adenovirus infections. Rev Med Virol 2008; 18:357-74. [PMID: 18655013 DOI: 10.1002/rmv.589] [Citation(s) in RCA: 168] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Adenoviruses (Ads) are common opportunistic pathogens that are rarely associated with severe clinical symptoms in healthy individuals. In contrast, in patients with compromised immunity, Ad infections often result in disseminated and potentially life-threatening disease. Among these are AIDS patients, individuals with hereditary immunodeficiencies and recipients of solid organ or haematopoietic stem cell transplants (HSCT) who receive immunosuppressive therapy. The latter account for the largest number of severe Ad infections. There is currently no formally approved antiviral therapy for the treatment of severe Ad keratoconjunctivitis and life-threatening Ad infections in immunocompromised patients. Here we update current knowledge on Ad biology, the clinical features observed in different patient groups and specific immune responses towards Ad infections. In addition, we review current and future treatment options, including: (i) the antiviral drugs cidofovir, ribavirin and new investigational compounds, as evaluated in the clinic or in relevant animal models, as well as (ii) novel immunotherapeutic strategies.
Collapse
Affiliation(s)
- Liesbeth Lenaerts
- Division of Virology and Chemotherapy, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | | | | |
Collapse
|
19
|
Hoffmann D, Meyer B, Wildner O. Improved glioblastoma treatment with Ad5/35 fiber chimeric conditionally replicating adenoviruses. J Gene Med 2008; 9:764-78. [PMID: 17640083 DOI: 10.1002/jgm.1076] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Adenovirus type 5 (Ad5)-based vectors have been used in clinical trials for glioblastoma treatment, but the capacity of Ad5 to infect human glioma cells was questioned. Seeking to improve the adenovirus transduction, we tested four Ad5-based vectors differing only in their fiber gene on permanent and short-term cultures of glioblastoma cells. A wild-type fiber Ad5 vector (Ad5.Luc) was compared to an RGD integrin-binding motif-containing fiber adenovirus (AdlucRGD) and the two fiber chimeras Ad5/3 and Ad5/35, with vector binding redirected to the Ad3 or Ad35 receptor, respectively. Compared to Ad5, the transduction of the tested short-term glioblastoma cultures with the vector Ad5/35.Luc, AdlucRGD and Ad5/3.Luc was enhanced by approximately 72%, approximately 13% and approximately 2%, respectively. To limit adenovirus spread, we aimed to develop conditionally replicative Ad5/35 vectors by targeting the expression of the essential E1 and E4 genes; in addition, some vectors had the E1Delta24 deletion. We analyzed eleven promoters for their activity in glioblastoma cells and determined the specificity of eight replicative adenovirus vectors in vitro. We evaluated the most promising vectors with E1/E4 under the control of the GFAP/Ki67 or E2F-1/COX-2 promoters, and the native Ad5 or the chimeric Ad5/35 fiber for their antineoplastic activity in a subcutaneous and intracranial glioblastoma xenograft model. Animals treated with the Ad5/35-based vectors showed significantly smaller tumors and longer survival than those treated with the homologous Ad5 vectors; no significant toxicity was observed in the intracranial model. Our data suggest that Ad5/35-based vectors are promising tools for glioblastoma treatment.
Collapse
Affiliation(s)
- Dennis Hoffmann
- Ruhr-University Bochum, Institute of Microbiology and Hygiene, Department of Molecular and Medical Virology, Bldg MA, Rm 6/40, D-44801, Bochum, Germany
| | | | | |
Collapse
|
20
|
Ungerechts G, Springfeld C, Frenzke ME, Lampe J, Johnston PB, Parker WB, Sorscher EJ, Cattaneo R. Lymphoma chemovirotherapy: CD20-targeted and convertase-armed measles virus can synergize with fludarabine. Cancer Res 2007; 67:10939-47. [PMID: 18006839 DOI: 10.1158/0008-5472.can-07-1252] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Combination chemotherapy regimen incorporating CD20 antibodies are commonly used in the treatment of CD20-positive non-Hodgkin's lymphoma (NHL). Fludarabine phosphate (F-araAMP), cyclophosphamide, and CD20 antibodies (Rituximab) constitute the FCR regimen for treating selected NHL, including aggressive mantle cell lymphoma (MCL). As an alternative to the CD20 antibody, we generated a CD20-targeted measles virus (MV)-based vector. This vector was also armed with the prodrug convertase purine nucleoside phosphorylase (PNP) that locally converts the active metabolite of F-araAMP to a highly diffusible substance capable of efficiently killing bystander cells. We showed in infected cells that early prodrug administration controls vector spread, whereas late administration enhances cell killing. Control of spread by early prodrug administration was also shown in an animal model: F-araAMP protected genetically modified mice susceptible to MV infection from a potentially lethal intracerebral challenge. Enhanced oncolytic potency after extensive infection was shown in a Burkitt's lymphoma xenograft model (Raji cells): After systemic vector inoculation, prodrug administration enhanced the therapeutic effect synergistically. In a MCL xenograft model (Granta 519 cells), intratumoral (i.t.) vector administration alone had high oncolytic efficacy: All mice experienced complete but temporary tumor regression, and survival was two to four times longer than that of untreated mice. Cells from MCL patients were shown to be sensitive to infection. Thus, synergy of F-araAMP with a PNP-armed and CD20-targeted MV was shown in one lymphoma therapy model after systemic vector inoculation.
Collapse
Affiliation(s)
- Guy Ungerechts
- Molecular Medicine Program and Virology and Gene Therapy Track, Mayo Clinic College of Medicine, Rochester, Minnesota 55902, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Hoffmann D, Wildner O. Enhanced killing of pancreatic cancer cells by expression of fusogenic membrane glycoproteins in combination with chemotherapy. Mol Cancer Ther 2006; 5:2013-22. [PMID: 16928822 DOI: 10.1158/1535-7163.mct-06-0128] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pancreatic cancer has a poor prognosis with an annual mortality rate close to the annual incidence rate. We evaluated whether the expression of measles virus fusogenic membrane glycoproteins (FMG) H and F will enhance chemotherapy. Using Chou-Talalay analysis, we showed in vitro in pancreatic cancer cells that the expression of FMG often synergistically enhances clinically relevant chemotherapy. Furthermore, cell fusion in combination with chemotherapy resulted in strongly enhanced Annexin V binding, an early marker for apoptosis, when compared with single treatment. We showed in an i.p. and s.c. pancreatic xenograft model that the administration of a replication-defective adenoviral vector Ad.H/F encoding tumor-restricted FMG in combination with gemcitabine significantly enhanced treatment outcome when compared with treatment with each compound individually. To improve tumor transduction efficiency, the Ad.H/F vector was also transcomplemented with an oncolytic replication-restricted adenovirus (Ad.COX*MK), resulting in significantly improved treatment efficacy. We assessed treatment efficacy by survival analysis or measuring growth, respectively. In the i.p. model, on day 120, three of eight animals treated with this novel triple therapy consisting of Ad.H/F, gemcitabine, and Ad.COX*MK were alive and tumor free. Treatment with Ad.H/F and Ad.COX*MK resulted in one long-term survivor. In all other treatment groups, there were no long-term survivors. The significantly improved therapeutic outcome of animals receiving the triple therapy was attributed to multiple factors, including most likely improved FMG expression throughout the tumor and enhanced sensitivity of the tumor cells to gemcitabine by adenoviral gene products but also FMG expression. Qualitatively similar results were obtained in a s.c. pancreatic xenograft model.
Collapse
Affiliation(s)
- Dennis Hoffmann
- Department of Molecular and Medical Virology, Institute of Microbiology and Hygiene, Ruhr-University Bochum, Room 6/40, Building MA, D-44801 Bochum, Germany
| | | |
Collapse
|
22
|
Hoffmann D, Wildner O. Efficient generation of double heterologous promoter controlled oncolytic adenovirus vectors by a single homologous recombination step in Escherichia coli. BMC Biotechnol 2006; 6:36. [PMID: 16887042 PMCID: PMC1557486 DOI: 10.1186/1472-6750-6-36] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2006] [Accepted: 08/03/2006] [Indexed: 12/05/2022] Open
Abstract
Background Oncolytic adenoviruses are promising agents for the multimodal treatment of cancer. However, tumor-selectivity is crucial for their applicability in patients. Recent studies by several groups demonstrated that oncolytic adenoviruses with tumor-/tissue-specific expression of the E1 and E4 genes, which are pivotal for adenoviral replication, have a specificity profile that is superior to viruses that solely target the expression of E1 or E4 genes. Presently the E1 and E4 regions are modified in a time consuming sequential fashion. Results Based on the widely used adenoviral cloning system AdEasy we generated a novel transfer vector that allows efficient and rapid generation of conditionally replication-competent adenovirus type 5 based vectors with the viral E1 and E4 genes under the transcriptional control of heterologous promoters. For insertion of the promoters of interest our transfer vector has two unique multiple cloning sites. Additionally, our shuttle plasmid allows encoding of a transgene within the E1A transcription unit. The modifications, including E1 mutations, are introduced into the adenoviral genome by a single homologous recombination step in Escherichia coli. Subsequently infectious viruses are rescued from plasmids. As a proof-of-concept we generated two conditionally replication-competent adenoviruses Ad.Ki•COX and Ad.COX•Ki with the promoters of the Ki-67 protein and the cyclooxygenase-2 (COX-2) driving E1 and E4 and vice versa. Conclusion We demonstrated with our cloning system efficient generation of double heterologous promoter controlled oncolytic adenoviral vectors by a single homologous recombination step in bacteria. The generated viruses showed preferential replication in tumor cells and in a subcutaneous HT-29 colon cancer xenograft model the viruses demonstrated significant oncolytic activity comparable with dl327.
Collapse
Affiliation(s)
- Dennis Hoffmann
- Ruhr-University Bochum, Institute of Microbiology and Hygiene, Department of Molecular and Medical Virology, Bldg. MA, Rm. 6/40, D-44801 Bochum, Germany
| | - Oliver Wildner
- Ruhr-University Bochum, Institute of Microbiology and Hygiene, Department of Molecular and Medical Virology, Bldg. MA, Rm. 6/40, D-44801 Bochum, Germany
| |
Collapse
|
23
|
Hoffmann D, Bangen JM, Bayer W, Wildner O. Synergy between expression of fusogenic membrane proteins, chemotherapy and facultative virotherapy in colorectal cancer. Gene Ther 2006; 13:1534-44. [PMID: 16791286 DOI: 10.1038/sj.gt.3302806] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Using Chou-Talalay median effect analysis, we demonstrated in permanent and short-term cultures of colorectal cancer cells that the expression of measles virus fusogenic membrane glycoproteins (FMGs) in combination with chemotherapy often causes over most of the cytotoxic dose range synergistic cell killing. In this combined treatment, we observed strongly enhanced annexin V binding and caspase-3/7 activity when compared to single-agent treatment. Furthermore, we showed increased expression of heat-shock protein (Hsp)70 and Hsp90alpha, but not of Hsp60. In a subcutaneous HT-29 colorectal xenograft model, we demonstrated that the administration of a replication-defective adenoviral or herpes simplex virus (HSV) amplicon vector (Ad.H/F or HSV.H/F) encoding tumor-restricted FMG in combination with FOLFOX significantly enhanced treatment outcome when compared to treatment with each compound individually. To increase the fraction of tumor cells expressing the FMG, we trans-complemented the Ad.H/F and HSV.H/F vector with the respective oncolytic replication-restricted adenovirus Ad.COXDeltaMK or HSV-1 G47Delta vector. At the end of the observation period (day 100), eight out of 10 animals that received G47Delta, HSV.H/F and FOLFOX were alive and tumor free. Administration of the analogous adenovirus-based regimen resulted in four out of 10 long-term survivors. We demonstrated that the expression of FMG in combination with chemotherapy can significantly enhance treatment outcome, which is further enhanced by combination with trans-complementing oncolytic vectors.
Collapse
Affiliation(s)
- D Hoffmann
- Department of Molecular and Medical Virology, Institute of Microbiology and Hygiene, Ruhr-University Bochum, D-44801 Bochum, Germany
| | | | | | | |
Collapse
|
24
|
Hoffmann D, Wildner O. Restriction of adenoviral replication to the transcriptional intersection of two different promoters for colorectal and pancreatic cancer treatment. Mol Cancer Ther 2006; 5:374-81. [PMID: 16505112 DOI: 10.1158/1535-7163.mct-05-0374] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In our current study, we developed oncolytic adenoviruses which preferentially lyse pancreatic and colon cancer cells by replacing viral E1 and/or E4 promoter with the tumor/tissue-specific promoters, cyclooxygenase-2 (COX-2), midkine (MK), or the cell cycle-dependent promoter, E2F1. We generated three sets of recombinant adenoviral vectors. In the first set, only the native E1A promoter was replaced by the COX-2, MK, or E2F1 promoter, respectively. In the second set, the viral E4 promoter was substituted by these heterologous promoters and the viral E1A promoter was substituted by the ubiquitously active cytomegalovirus-IE promoter. In the third set, we substituted the viral E1A and E4 promoters with the COX-2, MK, or E2F1 promoter, respectively. In our system, transcriptional targeting of solitary viral E1A resulted in 50% enhanced restricted vector replication when compared with an unrestricted replication-competent adenovirus. Furthermore, a targeted expression of the viral E1A gene products had a greater effect on restricted adenoviral replication than that of the E4 region. With our vectors, Ad.COX.MK and Ad.MK.COX, using two different heterologous promoters to control E1A and E4 expression, we showed enhanced viral replication specificity when compared with Ad.COX.COX or Ad.MK.MK, respectively. In a s.c. xenograft tumor model, there was no significant difference in the antineoplastic efficacy of the double heterologous promoter-controlled vectors when compared with our unrestricted replication-competent control adenovirus or vectors with only E1A transcriptionally driven by a heterologous promoter.
Collapse
Affiliation(s)
- Dennis Hoffmann
- Ruhr-University Bochum, Institute of Microbiology and Hygiene, Department of Molecular and Medical Virology, Building MA, Room 6/40, D-44801 Bochum, Germany
| | | |
Collapse
|
25
|
Lenaerts L, Naesens L. Antiviral therapy for adenovirus infections. Antiviral Res 2006; 71:172-80. [PMID: 16698093 DOI: 10.1016/j.antiviral.2006.04.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2006] [Revised: 04/06/2006] [Accepted: 04/06/2006] [Indexed: 01/11/2023]
Abstract
The treatment of severe adenovirus keratoconjunctivitis and life-threatening adenovirus infections in immunocompromised patients is still unsatisfactory. We here review the mode of action and antiviral data for cidofovir and ribavirin, obtained in cell culture, animal models or patients. Several nucleoside or nucleotide analogues have been described that target the adenovirus polymerase, whereas other antiviral targets have been poorly investigated. Furthermore, optimal therapeutic response may be achieved by combining antiviral therapy with immunotherapeutic approaches, as currently being explored.
Collapse
Affiliation(s)
- L Lenaerts
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, Minderbroedersstraat 10, B-3000 Leuven, Belgium
| | | |
Collapse
|
26
|
Jogler C, Hoffmann D, Theegarten D, Grunwald T, Uberla K, Wildner O. Replication properties of human adenovirus in vivo and in cultures of primary cells from different animal species. J Virol 2006; 80:3549-58. [PMID: 16537623 PMCID: PMC1440393 DOI: 10.1128/jvi.80.7.3549-3558.2006] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Oncolytic adenoviruses have emerged as a promising approach for the treatment of tumors resistant to other treatment modalities. However, preclinical safety studies are hampered by the lack of a permissive nonhuman host. Screening of a panel of primary cell cultures from seven different animal species revealed that porcine cells support productive replication of human adenovirus type 5 (Ad5) nearly as efficiently as human A549 cells, while release of infectious virus by cells from other animal species tested was diminished by several orders of magnitude. Restriction of productive Ad5 replication in rodent and rabbit cells seems to act primarily at a postentry step. Replication efficiency of adenoviral vectors harboring different E1 deletions or mutations in porcine cells was similar to that in A549 cells. Side-by-side comparison of the viral load kinetics in blood of swine and mice injected with Ad5 or a replication-deficient adenoviral vector failed to provide clear evidence for virus replication in mice. In contrast, evidence suggests that adenovirus replication occurs in swine, since adenoviral late gene expression produced a 13.5-fold increase in viral load in an individual swine from day 3 to day 7 and 100-fold increase in viral DNA levels in the Ad5-infected swine compared to the animal receiving a replication-deficient adenovirus. Lung histology of Ad5-infected swine revealed a severe interstitial pneumonia. Although the results in swine are based on a small number of animals and need to be confirmed, our data strongly suggest that infection of swine with human adenovirus or oncolytic adenoviral vectors is a more appropriate animal model to study adenoviral pathogenicity or pharmacodynamic and toxicity profiles of adenoviral vectors than infection of mice.
Collapse
Affiliation(s)
- Christian Jogler
- Department of Molecular and Medical Virology, Ruhr-University Bochum, D-44801 Bochum, Germany
| | | | | | | | | | | |
Collapse
|
27
|
Esimone CO, Grunwald T, Wildner O, Nchinda G, Tippler B, Proksch P, Uberla K. In vitro pharmacodynamic evaluation of antiviral medicinal plants using a vector-based assay technique. J Appl Microbiol 2006; 99:1346-55. [PMID: 16313407 DOI: 10.1111/j.1365-2672.2005.02732.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
AIMS Medicinal plants are increasingly being projected as suitable alternative sources of antiviral agents. The development of a suitable in vitro pharmacodynamic screening technique could contribute to rapid identification of potential bioactive plants and also to the standardization and/or pharmacokinetic-pharmacodynamic profiling of the bioactive components. METHODS AND RESULTS Recombinant viral vectors (lentiviral, retroviral and adenoviral) transferring the firefly luciferase gene were constructed and the inhibition of viral vector infectivity by various concentrations of plant extracts was evaluated in HeLa or Hep2 cells by measuring the changes in luciferase activity. Cytotoxicity of the extracts was evaluated in parallel on HeLa or Hep2 cells stably expressing luciferase. Amongst the 15 extracts screened, only the methanol (ME) and the ethyl acetate (ET) fractions of the lichen, Ramalina farinacea specifically reduced lentiviral and adenoviral infectivity in a dose-dependent manner. Further, chromatographic fractionation of ET into four fractions (ET1-ET4) revealed only ET4 to be selectively antiviral with an IC50 in the 20 microg ml(-1) range. Preliminary mechanistic studies based on the addition of the extracts at different time points in the viral infection cycle (kinetic studies) revealed that the inhibitory activity was highest if extract and vectors were preincubated prior to infection, suggesting that early steps in the lentiviral or adenoviral replication cycle could be the major target of ET4. Inhibition of wild-type HIV-1 was also observed at a 10-fold lower concentration of the extract. CONCLUSIONS The vector-based assay is a suitable in vitro pharmacodynamic evaluation technique for antiviral medicinal plants. The technique has successfully demonstrated the presence of antiviral principles in R. farinacea. SIGNIFICANCE AND IMPACT OF STUDY Potential anti-HIV medicinal plants could rapidly be evaluated with the reported vector-based technique. The lichen, R. farinacea could represent a lead source of antiviral substances and is thus worthy of further studies.
Collapse
Affiliation(s)
- C O Esimone
- Department of Molecular and Medical Virology, Ruhr-University, Bochum, Germany.
| | | | | | | | | | | | | |
Collapse
|
28
|
Naesens L, Lenaerts L, Andrei G, Snoeck R, Van Beers D, Holy A, Balzarini J, De Clercq E. Antiadenovirus activities of several classes of nucleoside and nucleotide analogues. Antimicrob Agents Chemother 2005; 49:1010-6. [PMID: 15728896 PMCID: PMC549266 DOI: 10.1128/aac.49.3.1010-1016.2005] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The absence of any formally licensed antiadenovirus drugs and the increasing incidence of life-threatening adenovirus infections in immunosuppressed patients warrant the development of effective antiadenovirus compounds. A detailed study was performed on the antiadenovirus activities of several classes of nucleoside and nucleotide analogues in human embryonic lung fibroblast cells. The antiadenovirus activities were evaluated by three methods, viz., evaluating the adenoviral cytopathic effect, monitoring cell viability by a colorimetric assay, and real-time PCR quantitation of viral DNA as a direct parameter for virus replication. The most active and selective compounds were the acyclic nucleoside phosphonate analogues cidofovir, its adenine analogue (S)-9-(3-hydroxy-2-phosphonylmethoxypropyl)adenine [(S)-HPMPA], and the new derivative (S)-2,4-diamino-6-[3-hydroxy-2-(phosphonomethoxy)propoxy]pyrimidine [(S)-HPMPO-DAPy]; the N7-substituted acyclic derivative 2-amino-7-(1,3-dihydroxy-2-propoxymethyl)purine (S-2242); and the 2',3'-dideoxynucleoside analogues zalcitabine and alovudine. No antiadenovirus activity was observed for the antiviral drugs ribavirin, foscarnet, acyclovir, penciclovir, and brivudin, while ganciclovir displayed modest activity. However, in human osteosarcoma cells transfected with herpes simplex virus thymidine kinase, ganciclovir demonstrated highly potent antiadenovirus activity, suggesting that the efficacy of ganciclovir against adenovirus is limited by inefficient phosphorylation in adenovirus-infected cells, rather than by insufficient inhibition at the viral DNA polymerase level. Collectively, our antiviral data show that the adenovirus DNA polymerase exhibits sensitivity to a relatively broad spectrum of inhibitors and should be studied further as an antiviral target in antiadenovirus drug development programs.
Collapse
Affiliation(s)
- L Naesens
- Rega Institute for Medical Research, K.U. Leuven, Minderbroedersstraat 10, B-3000 Leuven, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Morfin F, Dupuis-Girod S, Mundweiler S, Falcon D, Carrington D, Sedlacek P, Bierings M, Cetkovsky P, Kroes ACM, van Tol MJD, Thouvenot D. In Vitro Susceptibility of Adenovirus to Antiviral Drugs is Species-Dependent. Antivir Ther 2005. [DOI: 10.1177/135965350501000204] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Adenovirus infections are a frequent and serious complication following allogeneic haematopoietic stem cell transplantation (HSCT). The antiviral drugs cidofovir and ribavirin have been used as first-line therapy for disseminated infections with variable results. In the present study, in vitro susceptibility to these two drugs was evaluated on HEp-2 cells in adenovirus reference strains representing serotypes of each of the six species and in clinical isolates. Susceptibility to cidofovir was comparable between species with inhibition of replication of all tested serotypes in a narrow dose range (IC50= 17–81 μM). However, susceptibility to ribavirin was highly dependent on the species. Serotypes from species A, B, D, E and F were all resistant to ribavirin (IC50=396 to >500 μM). Only replication of serotypes from species C was inhibited by ribavirin (IC50=48–108 μM). This species-dependent susceptibility of adenovirus to ribavirin was confirmed in clinical isolates. When tested on other cell lines (PLC, A549 and 293), all species were revealed to be resistant to ribavirin. If our in vitro findings are predictive of virological responses in vivo, these results suggest that ribavirin would not be effective for management of non-C species adenovirus infections after HSCT.
Collapse
Affiliation(s)
- Florence Morfin
- Laboratory of Virology, Edouard Herriot Hospital, Lyon, France
- Faculty of Pharmacy, EA3090, Claude Bernard University, Lyon, France
| | - Sophie Dupuis-Girod
- Laboratory of Virology, Edouard Herriot Hospital, Lyon, France
- Haematology and Bone Marrow Transplantation Unit, Debrousse Hospital, Lyon, France
| | | | - Delphine Falcon
- Laboratory of Virology, Edouard Herriot Hospital, Lyon, France
| | | | - Petr Sedlacek
- Department of Paediatrics, Motol University Hospital, Prague, Czech Republic
| | - Marc Bierings
- Wilhelmina Kinderziekenhuis Haematology, Utrecht, The Netherlands
| | - Petr Cetkovsky
- Institute of Haematology and Blood Transfusion, UHKT, Prague, Czech Republic
| | - Aloys CM Kroes
- Department of Medical Microbiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Maarten JD van Tol
- Department of Paediatrics, Leiden University Medical Centre, Leiden, The Netherlands
| | | |
Collapse
|